throbber
Journal of Clinical Pharmacy and Therapeutics (2006) 31, 469-476
`
`ORIGINAL ARTICLE
`
`Pharmacokinetic and pharmacodynamic modelling of the
`effects of glimepiride on insulin secretion and glucose
`lowering in healthy humans
`H.-Y. Yun* MS, H.-C. Park* MS, W. Kangft PhD and K.-I. Kwon* PhD
`*College of Pharmacy, Chungnam National University, Daejeon, Korea and +Department of pharmacy,
`Catholic University of Daegu, Daegu, Korea
`
`SUMMARY
`
`glimepiride and its insulin secretion and hypo-
`glycaemic effects in healthy volunteers.
`
`INTRODUCTION
`
`Glimepiride is an oral sulfonylurea antihyper-
`glycaemic agent. We used pharmacokinetic—
`Keywords: glimepiride, pharmacodynamic, phar-
`macokinetic, pharmacokinetic-pharmacodynamic
`pharmacodynamic (PK-PD) modelling to analyse
`the relationship between plasma glimepiride
`modelling
`
`concentration, insulin secretion and_glucose
`lowering to determine the effects of the drug in
`healthy volunteers. A single 2-mg oral dose of
`1-[[p-[2-@-ethyl-4-methyl-2-oxo-3-
`Glimepiride,
`glimepiride was administered to six healthy vol-
`pyrroline-1-carboxamido)ethyl]|phenyl]sulfonyl]-
`unteers. The control group received a placebo. All
`3-(trans-4-methylcyclohexyl) urea, is an oral sulfo-
`subjects consumed 12g of sugar immediately
`nylurea antihyperglycaemic agent that contains a
`after drug administration in order to standardize
`sulfonylurea nucleus and a cyclohexyl ring (1).
`the initial plasma glucose levels. Serial blood
`Glimepiride may be given oncedaily.It has a long-
`sampling was performed for 9 h after oral dosing.
`lasting effect without markedly increasing plasma
`Plasma glimepiride, insulin and glucose levels
`insulin compared with other sulfonylureas (2).
`were determined by validated methods (LC/MS/
`Glimepiride’s majorsite of action is thoughtto be a
`MSassay, hexokinase method and radioimmu-
`membrane receptor on pancreatic B-cells, whereit
`noassay respectively). Time courses of plasma
`acts via ATP-regulated potassium (Karp) channels
`glimepiride concentration, insulin secretion, and
`to cause membrane depolarization and insulin
`glucose lowering effects were analysed by means
`release (3). The association rate of glimepiride is
`of PK-PD modelling with the ADAPT II pro-
`2:5- to 3-fold that of glibenclamide, its dissociation
`gram. The time course of the plasma concentra-
`rate is 8- to 9-fold that of glibenclamide, and its
`tions followed a two-compartmental model with a
`in vitro binding affinity for rat B-cell tumour and
`lag time. The glimepiride concentration peaked at
`insulinomacells is 2:5- to 3-fold lower than that of
`191:5 ng/mL at approximately 4 h after adminis-
`tration. The maximal increase in insulin secretion
`glibenclamide (4). Sulfonylureas interact with dif-
`was 9:98 mIU/L and the maximal decrease in
`ferent sites on the pancreatic f-cell membrane.
`Glimepiride binds to a 65-kDa protein, whereas
`plasma glucose was 19:33 mg/dL. Both peak
`glibenclamide binds to a 140-kDa protein. These
`effects occurred at approximately 2°5 h after drug
`proteins are believed to both be part of the same
`intake. The glucose disappearance model was
`sulfonylurea receptor, as each agent inhibits the
`used to analyse glimepiride’s insulin secretion
`other’s binding to its protein target (5).
`and glucose lowering effects. The PK-PD model
`In single-dose studies with healthy volunteers,
`described well the relationship between plasma
`the peak glimepiride plasma concentration (Cmax)
`and the area under the plasma concentration—time
`curve (AUC) were generally dose-proportional. In
`12 healthy volunteers, the Cymax rose linearly from
`
`Received 24 March 2005, Accepted 20 June 2006
`Correspondence: Kwang-Il Kwon, College of Pharmacy, Chung-
`nam National University, Daejeon 305-764, Korea. Tel.: 82 42 821
`5937; fax: 82 42 823 6781; e-mail: kwon@cnu.ac.kr
`
`© 2006 Blackwell Publishing Ltd
`
`469
`
`MPI EXHIBIT 1046 PAGE1
`
`MPI EXHIBIT 1046 PAGE 1
`
`MPI EXHIBIT 1046 PAGE 1
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1046, p. 1 of 7
`
`

`

`470
`
`=~=4#H.-Y. Yun et al.
`
`103-2 to 550-8 ug/L as the dose increased from 1 to
`8 mg, whereas the AUC increased from 339 to
`2634 pg h/L. The glimepiride Cmax occurred at 0:7—
`2:8 h (Tmax) after the single-dose administration to
`healthy volunteers (6). Glimepiride plasma protein
`binding was 99:-4% (7), and the volumeof distri-
`bution (Vq) was 8:8 L (8).
`From dose-ranging studies in patients with
`type 2 diabetes mellitus, glimepiride appears to
`reduce fasting and postprandial blood glucose
`levels,
`as well
`as glycosylated haemoglobin.
`These effects are dose-dependent over a range of
`1-4 mg daily. For patients receiving the maxi-
`mum daily dose (8 mg), the average reduction in
`glycosylated haemoglobin is 2% in absolute
`units. Age, gender, weight, and race do not affect
`glimepiride’s efficacy (9).
`The main objective of this study was to examine
`the relationship between plasma glimepiride con-
`centration and its insulin secretion and glucose
`lowering effects (i.e. the increase in blood insulin
`and decrease in blood glucose) after
`its oral
`administration to healthy volunteers. This should
`permit prediction of the time course of glimepi-
`ride’s therapeutic and side effect profiles after oral
`dosing. The relationship between the pharmacoki-
`netics of glimepiride andits insulin secretion and
`glucose lowering effects has not yet been analysed.
`Our goal was to assess the usefulness of pharma-
`cokinetic-pharmacodynamic (PK-PD) modelling
`in describing this relationship.
`
`METHODS
`
`Subjects
`
`Six healthy male subjects with a mean age of
`25 years (range = 22-25 years) and a mean weight
`of 69:67 kg (range = 55-83 kg)
`took part
`in this
`study. All subjects underwent a thoroughhistory, a
`complete physical examination, and a battery of
`routine
`laboratory tests
`(haematology,
`serum
`chemistry and urinalysis). None had taken any
`drugs knownto interfere with the study for at least
`10 days beforehand. The exclusion criteria inclu-
`ded health problems, drug or alcohol abuse, and
`abnormalities in laboratory screening tests. All
`subjects were told the full details of the study and
`gave written informed consent. The study was
`approved by the local ethics committee.
`
`Study design
`
`All subjects fasted for at least 10 h before taking
`their study medication. At time zero, an intraven-
`ous cannula was inserted into the forearm vein and
`control blood samples were collected.
`First period, six subjects received a single-oral
`dose Amaryl 2 mg and second period, same sub-
`jects received a placebo. There was a 6-day wash-
`out period between the periods. After baseline
`sampling, the test group took glimepiride (Ama-
`ryl® 2 mg tablet; Handok/Aventis Pharma Co. Ltd,
`Seoul, Korea) with 240 mL of water. The control
`group took a placebo with 240 mL of water. All
`subjects consumed 12 g of sugar cubes immedi-
`ately after drug administration in order to prevent
`hypoglycaemia and maintain standard initial
`plasma glucose level. All subjects were given a
`standardized meal 4h after drug administration.
`They were not allowed to remain supine or to sleep
`until 4 h after drug administration.
`insulin and
`Samples for plasma glimepiride,
`glucose determinations were taken at 0-5, 1, 1:5, 2,
`2:5, 3, 4,5, 7 and 9 h after drug administration. The
`samples were collected in heparinized tubes,
`immediately centrifuged (10 min at 1650 g), and
`stored at —80 °C for later analysis.
`
`Plasma assay
`
`Plasma glimepiride was assayed by the reported
`LC/MS/MS method, with a slight modification (1).
`Briefly, 50 wL of internal standard (glibenclamide,
`500 ng/mL) and 0:5 mL of 1 mM NaOH were added
`to 0:5 mL of plasma, followed by a 10-min liquid—
`liquid extraction with 5 mL of ethyl ether : ethyl
`acetate (1 : 1, v/v). The organic layer was separated
`and evaporated to dryness at ambient temperature
`in a Speed-Vac (Savant, Holbrook, NY, USA). The
`residue was reconstituted in 100 “L of acetonitrile
`by vortexing for 15 s; then 5 wL of this solution was
`injected onto the column. The mobile phase was a
`mixture of 01% formic acid buffer : acetonitrile
`
`(20: 80, v/v), and the column was eluted at
`0:2 mL/min with an HP 1100 series pump(Agilent,
`Wilmington, DE, USA). The turbo-ion spray inter-
`face was operated in positive ion mode at 5500 V
`and 350 °C. Using flow injection of a mixtureof all
`analyses, the operating conditions were optimized
`to: nebulizing gas flow, 1:04 L/min; auxiliary gas
`
`© 2006 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 31, 469-476
`
`MPI EXHIBIT 1046 PAGE 2
`
`MPI EXHIBIT 1046 PAGE 2
`
`MPI EXHIBIT 1046 PAGE 2
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1046, p. 2 of 7
`
`

`

`PK/PD modeling of glimepiride
`
`471
`
`kinetic model
`to reflect
`the oral administration
`flow, 40 L/min; curtain gas flow, 1-44 L/min; ori-
`fice voltage, 80 V; ring voltage, 400 V and collision
`and flip-flop kinetics. Model development was an
`gas (nitrogen) pressure, 3:58 x 10° Torr. Quantita-
`iterative process with regard to both the under-
`tion was performed by multiple reaction monitor-
`lying data set and the selected model structure.
`Models were constructed as a series of differen-
`ing of the protonated precursor ion and the related
`product
`ion for glimepiride, using the internal
`tial equations that were solved numerically and
`fitted to the data with the ADAPT II software
`standard method with the peak area ratio. The mass
`transitions used for glimepiride and the internal
`(Biomedical Simulation Resource, Los Angeles,
`standard
`were m/z
`4910 — 3524
`and
`CA, USA)
`(11). The fitting with individual data
`
`4940 — 3694 (20 eV_collisionrespectively
`
`was performed by means of maximum likelihood
`energy, 200 ms dwell time). Quadruples Q1 and Q3
`estimation. The following information was used
`wereset on unit resolution. The analytical data were
`to evaluate the goodness of fit and the quality of
`processed using Analyst Software (version 1:2;
`the parameter estimates: coefficient of variation of
`POETsoftware corporation, London, UK).
`parameter estimates, parameter correlation mat-
`rix, sums of squares of residuals, visual exam-
`For all plasma samples, glucose concentrations
`ination of the distribution of residuals and the
`were determined enzymatically by the hexokinase
`method, and insulin concentrations were deter-
`mined by radioimmunoassay (10).
`
`Akaike Information Criterion (12). The differential
`equations
`that described the changes
`in the
`amounts of glimepiride in the compartments after
`oral administration are given by Eqs (1) to (3):
`
`Pharmacokinetic/pharmacodynamic model and
`data analysis
`
`dx
`Vinaxl2
`(EN x
`1
`a Ge xm
`(1)
`Pharmacokinetics (plasma_glimepiride) and
`
`
`pharmacodynamics (plasma insulin and glucose)
`were modelled sequentially. We developed a
`parsimonious compartmental model that reflects
`the rate of change of glucose as the difference
`between the net hepatic glucose balances. As
`shownin Fig. 1, a two-compartment model with
`a lag time, nonlinear absorption and elimination
`was selected as the most appropriate pharmaco-
`
`2
`
`(2)
`
`(3)
`
`— Ko3
`
`Vimaxl2
`dx2
`es
`dt
`Kmi2 To)
`pense
`— = (SY x ey +h x x
`Vinax2e
`X X — (=) x x
`23
`2 (Gt 2
`dxpo Kas x 32 ~ Kan X83
`
`Fig. 1. The model selected to des-
`cribe the effects of glimepiride on
`insulin secretion and glucose
`lowering in healthy volunteers:1,
`absorption lag time; Vinax and Kyi,
`Michaelis-Menten transport
`parameters; Kj, first-order rate
`constant; K;,, zero-order rate
`constant for insulin production;
`Kour the first-order constant for
`insulin loss.
`
`
`
`2. Central
`
`3. Peripheral
`
`compartment
`compartment
`
`
`
`
`
`4. Insulin
`compartment
`i
`
` 5. Liver
`
`compartment
`
`© 2006 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 31, 469-476
`
`MPI EXHIBIT 1046 PAGE 3
`
`MPI EXHIBIT 1046 PAGE 3
`
`MPI EXHIBIT 1046 PAGE 3
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1046, p. 3 of 7
`
`

`

`472 H.-Y. Yun et al.
`
`First-order rate constants describing intercom-
`partmental transport are denoted by Kj, and the
`active transport with Michaelis-Menten type kin-
`etics is characterized by the apparent maximal
`transport rates Vmax; and the apparent Michaelis
`constants Kyyjj
`(13). After the parameters of the
`pharmacokinetic model were fixed,
`the model
`served as an input function for the pharmacody-
`namic models (14).
`An insulin-dependent glucose disappearance
`model was used to analyse the PK-PDrelationship.
`(15) In our study, the response variables measured
`were the insulin plasma concentration (milli-inter-
`national units per litre, mIU/L) and the glucose
`plasma concentration (mg/mL). The system of
`differential equations shown below describes the
`model:
`
`dt
`
`x4 = Kin x 51 (t) _ Kout xR
`dxaKes X X65 —Kee x So(t) X X53 — KeeS(t) x X5
`dxFr = Kge x S(t) x x5 — KesXxe6
`_
`Smax (x2/V2)
`sil) = Th Scsgy (x2/V2)
`
`So(t) = 14 x4
`
`Kj
`Ro = ZK.
`
`(4)
`
`(5)
`(6)
`
`”)
`
`(8)
`
`(9)
`
`where Ki, represents the zero-order constant for
`production of the insulin response and Ko. defines
`the first-order rate constant for loss of the insulin
`response. In our study, a model
`that assumed
`glimepiride simulated K;, was considered to be
`closest to the pharmacological action of the drug.
`However, both modelling approaches, with glim-
`epiride inhibiting Kou: or stimulating Kj,, were
`investigated to compare the performance of the
`model and the physiologic relevance of
`the
`parameters obtained.
`In this model, the rate of change of plasma glu-
`cose is the difference between the net hepatic glu-
`cose balance and the disappearance of glucose into
`peripheral tissues only (16). We have shown that
`the hepatic glucose balance varies according to the
`relationship shownin Eq. (5). To explain glucose
`disappearance,it is assumed that insulin acts from
`a remote compartment, that insulin increases the
`
`Fig. 2. Plasma glimepiride concentration after a single 2-
`mg oral dose to healthy volunteers (mean + SEM, n = 6).
`Data points are observed values; the solid line is the
`result of maximum likelihood fitting with the ADAPT II
`program.
`
`© 2006 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 31, 469-476
`
`MPI EXHIBIT 1046 PAGE 4
`
`mobility of glucose across cell membranes, and that
`glucose mobility potentiates glucose disappear-
`ance. Wheninsulin increases, it stimulates Ks, and
`Keo according to the stimulation function given in
`Eq.(8).
`
`RESULTS
`
`Pharmacokinetic analysis
`
`The mean plasma concentration-vs.-time curve
`after oral administration of 2 mg of glimepirideis
`shownin Fig. 2, where the solid line represents the
`best fit of the pharmacokinetic model to the meas-
`ured concentration, based on the means of the
`individual parameter estimates. Based on the
`maximum likelihood criterion and visual inspec-
`tion of the fits, a two-compartment model with a
`lag time, nonlinear absorption and elimination was
`chosen to describe the data. The estimated phar-
`macokinetic parameters are listed in Table 1: The
`Michaelis-Menten type absorption rate into the
`central compartment Vimaxi2 and the apparent
`Michaelis constant K,,12 equal 1042:58 ng/h and
`70:31 ng respectively. The Michaelis-Menten kin-
`etic analysis of the data indicated the existence of a
`second carrier-mediated transport process and of
`an interaction between sulfonylurea and highly
`protein-bound drugs that govern elimination from
`the
`central
`compartment
`(Vmax= 0-14 ng/h,
`Kye = 0-006 ng) (2). The terminal elimination half-
`
`2 1000,
`“Sp
`&oO
`3a
`z
`“eb
`‘*s
`
`1004
`
`;
`
`10
`
`0
`
`7
`2
`
`r
`4
`Time (h)
`
`7
`6
`
`!
`
`+
`8
`
`
`
`o& So
`
`yg
`
`2wo
`
`a &S
`
`a
`
`MPI EXHIBIT 1046 PAGE 4
`
`MPI EXHIBIT 1046 PAGE 4
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1046, p. 4 of 7
`
`

`

`Table 1. Pharmacokinetic parameters for glimepiride
`after a single 2-mg oral dose in healthy volunteers
`(mean + SEM, 1 = 6)
`
`Value
`Parameter
`
`
`PK/PD modeling of glimepiride
`
`473
`
`1021-43 + 199-55
`191-52 + 29:80
`
`2:58 + 052
`2:26 + 0:33
`7:82 + 1:22
`2:55 + 0:37
`
`Glucose lowering effect
`
`secretion was
`insulin
`the maximal
`group,
`4643 mIU/L at 5h, and secretion returned to
`baseline at 9h.
`In the test group, glimepiride
`caused a further significant
`increase (maximum
`increase of 9:98 mIU/L)
`in plasma insulin at
`between 1 and 5hafter its administration, and the
`Model independent parameter
`insulin concentration returned to baseline by 9 h.
`AUC (ng h/mL)
`Glimepiride produced a statistically significant
`Cmax (ng/mL)
`increasein insulin secretion, relative to placebo, for
`Tmax (h)
`3h (from 1 to 4 h after administration) (P < 0-01).
`CL(inf) /F (L/h)
`Vz(terminal) /F (L)
`ti2
`Model dependent parameter
`70:31 + 30-51
`Kmiz (ng)
`Figure 4 showsthe glucose profiles after drug and
`1042:58 + 161-82
`Vmaxcaz (ng/h)
`placebo administration. The decline in plasma
`0-006 + 0-0004
`Kaze (ng)
`glucose was induced after a lag of approximately
`0-14 + 0-02
`Vmaxze(ng/h)
`15h, and the decrease reached its maximum
`0-21 + 0-11
`Ko3 (/h)
`
`(2100 mg/dL decrease plasma_glucose)in
`
`Kap (/h)
`0-72 + 0:36
`approximately 2 h after dosing. Compared with the
`Tiag (h)
`0-34 + 0-04
`placebo group, glimepiride produced a statistically
`significant decrease in glucoselevels for a period of
`administration
`35h,
`from 15 to 5h after
`(P < 0:01).
`
`805
`
`604
`
`404
`
`204
`
`
`
`2=g =
`
`§ =
`
`&&o
`
`goS
`
`&2
`
`wo
`
`a 5
`
`Ss
`
`* 04
`:
`:
`2
`4
`6
`8
`
`Time (h)
`
`Fig. 3. Plasma insulin concentration after a single oral
`dose of placebo or 2 mg glimepiride to healthy volun-
`teers (mean + SEM,n = 6). Open circles, plasma insulin
`concentration after placebo. Closed circles, plasma insu-
`lin concentration after 2 mg glimepiride.
`
`life (t1/2p) was 2:548 + 0-901 h and the CLiota/F
`was 2-262 + 0814 L/h. Our pharmacokinetic pro-
`file for glimepiride is similar to that found in other
`studies involving healthy volunteers and the same
`glimepiride dose (10).
`
`Insulin secretion effect
`
`Plasma insulin profiles after drug and placebo
`administration are shown in Fig. 3. In the control
`
`Pharmacokinetic_pharmacodynamic modelling of
`insulin secretion and glucose lowering effects
`
`The corresponding pharmacodynamic parameter
`estimates for plasma insulin and glucose are shown
`in Table 2. Figures 5 and 6 show the plasmainsu-
`lin profile (post-drug level minus placebo level;
`
`
`
`
`
`Plasmaconcentrationofglucose(mg/dL)
`
`160 -
`
`===OQho£OooOoOiii
`
`80 -
`
`60 -
`
`40 -
`
`
`
`20 +
`OQ
`ho
`oO
`4
`6
`8
`
`Time(h)
`
`Fig. 4. Plasma glucose concentration after a single oral
`dose of placebo or 2 mg glimepiride to healthy volun-
`teers (mean + SEM, n = 6). Opencircles, plasma glucose
`concentration after placebo. Closed circles, plasma glu-
`cose concentration after 2 mg glimepiride.
`
`© 2006 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 31, 469-476
`
`MPI EXHIBIT 1046 PAGE 5
`
`MPI EXHIBIT 1046 PAGE 5
`
`MPI EXHIBIT 1046 PAGE 5
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1046, p. 5 of 7
`
`

`

`474 H.-Y. Yun et al.
`
`Table 2. Pharmacodynamic parameters estimated by
`individual fitting of maximum likelihood method with
`ADAPTII software (mean + SEM, 1 = 6)
`
`Parameter
`Value
`
`
`Estimated parameter
`Kin (mIU h/L)
`RO (mIU/L)
`SC50 (ng/mL)
`Sax
`Ke (/h)
`Kse (/h)
`Kes (/h)
`Secondary parameter
`Kout (]Kin/RO) (/h)
`
`4-05 + 1:01
`4-54 + 1-08
`78°89 + 33-66
`21-17 + 10-20
`0-012 + 0-005
`0-007 + 0-002
`0:37 + 0:20
`
`0-89
`
`
`
`|
`
`|
`
`d
`
`a 20,
`
`== 2
`
`3Qa
`
`£= 10
`
`15!
`
`|
`2
`
`54
`
`ol
`0
`
`8B
`
`g
`
`a 2
`
`|
`
`,
`4
`
`J
`
`|
`6
`
`|
`8
`
`This study analysed the effects of a single oral dose
`of glimepiride on insulin secretion and glucose
`lowering as a function of glimepiride concentra-
`tion. This is the first attempt
`to apply PK-PD
`modelling to glimepiride’s effects on these para-
`meters. Our results may help elucidate the rela-
`
`
`
`tionship plasma_concentrationbetween of
`Fig. 5. Time course of insulin secretion after a single
`glimepiride and its physiological effects.
`2-mg oral dose of glimepiride to healthy volunteers
`Glimepiride absorption kinetics have previously
`(mean + SEM, n= 6). Curve, pharmacodynamic fit
`to
`been studied in vitro and in vivo. Glimepiride dis-
`measure increases relative to placebo (circles).
`solves very rapidly in vitro (>80% after 15 min). In
`contrast,
`in vivo dissolution is delayed (>80%in
`approximately 4h) because of
`the low, pH-
`dependentsolubility of the drug (17). Poor aqueous
`solubility and slow dissolution are factors in non-
`linear absorption kinetics (18). Thus, first-order
`absorption kinetics were unsuccessful
`in repre-
`senting the behaviour of glimepiride, as evidenced
`by the poor correlation between observed andcal-
`culated data.
`Depending on the knowledge of a drug’s
`mechanism of action, different types of indirect
`response models may be used. In our study, we
`investigated pharmacokinetic and pharmacody-
`namic modelling approaches to link glucose dis-
`appearance and glimepiride levels. Given that
`
`
`
`
`
`
`
` 30- Glucosedisappearancerelativetoplacebo(mg//dL)
`
`Time (h)
`
`Fig. 6. Time course of glucose disappearance after a
`single 2-mg oral dose of glimepiride to healthy volun-
`teers (mean + SEM, n = 6). Curve, Pharmacodynamicfit
`to measure decreases relative to placebo (circles).
`
`consistent with the known physiology of glucose
`metabolism.
`
`DISCUSSION
`
`Time(h)
`
`mIU/L) and the plasma glucose profile (placebo
`level minus post-drug level; mg/dL), respectively,
`after a single 2-mg dose of glimepiride. The solid
`lines in Figs 5 and 6 representthe best fit of the PK—
`PD model. Pharmacokinetic and pharmacody-
`namic parameters for insulin secretion and glucose
`lowering effects after glimepiride dosing were
`estimated for each subject. Glimepiride plasma
`concentrations could be linked to the observed
`insulin secretion effects by means of an indirect-
`response model, and the change in plasma glucose
`was explained by a glucose disappearance model.
`Weselected this model because it
`is acceptable
`from the identification point of view and is broadly
`
`© 2006 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 31, 469-476
`
`MPI EXHIBIT 1046 PAGE 6
`
`MPI EXHIBIT 1046 PAGE 6
`
`MPI EXHIBIT 1046 PAGE 6
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1046, p. 6 of 7
`
`

`

`glimepiride stimulates insulin secretion, a model
`that assumes the stimulation of Kj, may better
`describe the drug’s pharmacological action.
`A nonlinear relationship between plasma glim-
`epiride levels and the stimulation of insulin secre-
`tion was observed. We therefore assume that
`insulin was secreted in accordance with drug-
`receptor binding via a sigmoid E,,,, model (4). The
`adaptation of other (e.g. linear and exponential)
`models was also explored, but only the sigmoid
`Emax model
`fit
`the observed plasma insulin
`dynamics. We explain the direct response between
`plasma insulin and the stimulation of glucose dis-
`appearance by assuming that the unit of plasma
`insulin Gin mIU/L) reflects the glucose lowering
`effect directly. Similar multi-compartments have
`been demonstrated by Ole et al. (14).
`Our model might more closely simulate insulin
`and glucose regulation if other factors, such as the
`stimulation of Ko, by plasma glucose concentra-
`tion, were considered. However, the insulin secre-
`tion
`and
`glucose
`lowering
`effects
`of
`oral
`glimepiride fitted sufficiently well, as shown in
`Figs 5 and 6. This indicated that our present model
`appropriately describes the PK-PDrelationships of
`oral glimepiride. As the onset of glimepiride’s
`pharmacological effect
`is
`fundamentally inde-
`pendent of dose and route, this quantified model
`for the insulin-glucose regulation system could
`describe plasma levels after another different dose
`or route of administration.
`In conclusion,
`the use of the glucose disap-
`pearance model in the context of PK-PD model-
`ling
`showed
`that
`the
`two measures
`of
`pharmacodynamic response (insulin secretion and
`glucose lowering effects) are valid and clinically
`relevant ways to investigate the effects of glim-
`epiride. By using an indirect response model, we
`could describe the stimulatory effect of glimepi-
`ride on insulin secretion. The glucose disappear-
`ance model, regulated by insulin, also fit well
`with the relation between glimepiride-induced
`changes in insulin secretion and glucose reducing
`effects.
`The established PK-PD model may be used to
`predict the plasma level of glimepiride, glucose
`and insulin under new conditions, for example
`following multiple dosing, and increasing or
`decreasing dose. This study involved healthy vol-
`unteers. By definition in clinical practice glime-
`
`PK/PD modeling of glimepiride
`
`475
`
`pride is going to be used in patients with diabetes.
`To overcome the limitation of this PK/PD model,
`we need more pharmacokinetics and pharmaco-
`dynamics data from such patients.
`
`ACKNOWLEDGEMENTS
`
`This study was supported by the contract, ‘PK/PD
`model development of glimepiride 2 mg after
`administering a single oral dose to healthy Korean
`volunteers’,
`from SAM-NAM Pharm Co., Ltd,
`Chungnam, Korea.
`
`REFERENCES
`
`1. Salem I, Idress J, Al Tamimi JI (2004) Determination
`of glimepiride in human plasma by liquid chroma-
`tography-electrospray
`ionization
`tandem mass
`spectrometry. Journal of Chromatography B, 79, 103-
`109.
`
`2. Langtry HD, Balfour JA (1998) Glimepiride - a re-
`view ofits use in the managementof type 2 diabetes
`mellitus. Drugs, 55, 563-584.
`3. Kramer W, Muller G, Geisen K (1996) Characteriza-
`tion of the molecular mode of action of the sulfo-
`
`and
`
`f-cells. Hormone
`at
`nylurea, glimepiride,
`Metabolic Research, 28, 464-468.
`4. Schwanstecher M, Manner K, Panten U (1994) Inhi-
`bition of K* channels and stimulation of insulin
`secretion by the sulfonylurea, glimepiride, in relation
`to its membrane binding in pancreatic islets. Phar-
`macology, 49, 105-111.
`5. Malerczyk V, Babian M, Korn A (1994) Doselinearity
`assessment of glimepiride (Amaryl®)
`tablets
`in
`healthy volunteers. Drug Metabolism and Drug Inter-
`actions, 11, 341-357.
`6. Profozic V, Mrzljak V, Rosenkranz B, Lehr KH,
`Waldhausl W (1991) Pharmacokinetics of glimepi-
`ride in kidney disease. Diabetes, 40, 343-347.
`7. Rosekranz B (1996) Pharmacokinetic basis for the
`safety of glimepiride in risk groups of NIDDM pa-
`tients. Hormone and Metabolic Research, 28, 434-439.
`8. Campbell RK (1998) Glimepiride: role of a new sul-
`fonylurea in the treatment of type 2 diabetes melli-
`tus. Annals of Pharmacotherapy, 32, 1044-1052.
`9. McEvoy GK, (2002) AHFS drug information. Wiscon-
`sin: American Society of Health-Systems Pharma-
`cists Inc., 3025-3028.
`10. Badian M, Korn A, Lehr K-H, Malerczyk V, Wald-
`hausal W (1996) Pharmacokinetics and pharmaco-
`dynamics of the hydroxyl metabolite of glimepiride
`(Amaryl®) after intravenous administration. Drug
`Metabolism and Drug Interactions, 13, 69-85.
`
`© 2006 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 31, 469-476
`
`MPI EXHIBIT 1046 PAGE 7
`
`MPI EXHIBIT 1046 PAGE 7
`
`MPI EXHIBIT 1046 PAGE 7
`
`DR. REDDY’S LABORATORIES, INC.
`IPR2024-00009
`Ex. 1046, p. 7 of 7
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket