You are on page 1of 50

DRUG METABOLISM AND

PHARMACOKINETICS (DMPK)
Lena Gustavsson, H. Lundbeck A/S, LEGU@lundbeck.com
November 2015
DMPK in Drug Discovery and Development
Agenda
Introduction
Optimizing pharmacokinetic properties
Absorption & bioavailability
Distribution
Elimination – Clearance
Understanding clearance mechanisms
Drug metabolism
Drug transporters
Drug drug interactions and interindividual variability
Summary: Drug discovery and development

2
H. Lundbeck A/S – an introduction
A pharmaceutical company with focus on brain diseases
More than 700 million people are affected by brain disease worldwide
Lundbeck is dedicated to address the global burden of brain disease
Psychiatric diseases e.g. bipolar disorder, depression, schizophrenia
Neurologic diseases e.g. Alzheimer´s, Parkinson´s, Huntington´s

A global company with head quaters in Valby, Denmark


Total approximately 5500 employee´s
Approximately 1700 employees in Denmark
Full value chain from research to production

Want to know more? Go to:


www.lundbeck.com/global/about-us/progress-in-mind
www.youtube.com/user/progressinmind

3
Why study Drug Metabolism and
PharmacoKinetics?
DRUG DISCOVERY
Optimise compounds to get...
Good bioavailability – get to its target
Appropriate duration (1-2 doses/day)
Low potential for drug-drug interactions
…predicted to man

Provide basics for understanding of


Toxicology
Pharmacology From Rowland and Tozer, 1995

DRUG DEVELOPMENT
Provide understanding of drug disposition
Preclinical animal species – tox coverage
Human data

Assess the risk for drug drug interactions (DDI)


Decrease risk for drug drug interactions in the clinic
Impact on the design of clinical studies

Comply with guidelines from regulatory authorities

Lena Gustavsson 2015-11-08


Reasons for compound attrition

Kola and Landis 2004

Lena Gustavsson 2015-11-08


Pharmacokinetics – oral administration

Drug concentration in plasma

Cmax

tmax

ADME = Absorption Distribution Metabolism Excretion

Lena Gustavsson 2015-11-08


DMPK in Drug Discovery and Development
Agenda
Introduction
Optimizing pharmacokinetic properties
Absorption & bioavailability
Distribution
Elimination – Clearance
Understanding clearance mechanisms
Drug metabolism
Drug transporters
Drug drug interactions and interindividual variability
Summary: Drug discovery and development

7
Absorption

Lipinski’s rule of 5 to predict poor


permeability/absorption (Lipinski et al, Adv Drug Delivery Rev 23:3-25, 1997)
Mw > 500
Log P > 5
H-bond donors >5
H-bond acceptors > 10
Transporter substrates are exceptions from the rule.

Lena Gustavsson 2015-11-08


Permeability
Caco-2

Is the drug absorbed?


No = low bioavailability !

•Papp: cm/sec x 10-6


correlates to human abs
•Indication of transporter mechanisms

• Automated incubations
• LC-MS/MS analysis

• Alternative to Caco-2:
Caco-2 PAMPA – artificial membrane
• Human colon epithelial cell line
• Differentiates to monolayer
with tight junctions

Lena Gustavsson 2015-11-08


Bioavailability - oral administration

F = Fgut x Fabs x Fhep

Gut
lumen Gut wall

Liver
Fgut Portal
Fabs vein
Fhep F

Fhep = 1 - Ehep

Lena Gustavsson 2015-11-08


Distribution
• Drug distribution is the reversible transfer of drug
to and from the site of measurement (blood/plasma)

• Distribution is influenced by
-perfusion – blood circulation to tissues
-diffusion
-physicochemical properties
-binding to proteins etc

From Rowland and Tozer, 1995

Lena Gustavsson 2015-11-08


Volume of distribution (V)

Not a real volume but a mathematical expression of the


extent to which a drug distributes into tissues
-Low V – drug stays in blood/plasma
-High V – drug distributes extensively into tissues

V relates the concentration at site of measurement to


the total amount of drug in the body (L/kg)

V=Amount drug in body/plasma concentration (L/kg bw)

Lena Gustavsson 2015-11-08


Elimination: The concept of clearance (CL)

Clearance is the apparent volume of plasma completely


cleared of drug per unit time

Rate of elimination = CL x C
CL = Dose / AUC (iv dose)
Unit: mL/min/kg

Lena Gustavsson 2015-11-08


Hepatic clearance

Hepatic vein

Gall Bladder

Hepatic portal vein


Bile duct Hepatic artery

The liver is the major site


of drug metabolism

Lena Gustavsson 2015-11-08


Drug metabolizing enzymes
Route of elimination of the top 200 most
prescribed drugs in 2002

Enzymes listed in FDA


guidelines
• CYP: 1A2, 2B6, 2C8, 2C9,
2C19, 2D6, 3A
• UGT: 1A1, 1A3, 1A4, 1A6, 1A9,
2B7, 2B15

Weinkers and Health Nat Rev Drug Discov 4:825-833, 2005


How to estimate metabolic clearance from in
vitro studies?

Lena Gustavsson 2015-11-08


Metabolic stability
Microsomes or
hepatocytes

How fast is the drug eliminated by the liver ?


Fast = low bioavailability !
Fast = short duration !
CLint - the intrinsic capacity of a
Phase I +II II system to clear a drug (µL/min/mg
protein or cells)
100

ln Substrate Concentration
metabolites
Vmax 10

CLint = = V0 / [S] 1

Km 0,1
0 20 40 60 80

0,01
Time (minutes)

Lena Gustavsson 2015-11-08


Prediction of in vivo clearance from
in vitro data
In vitro t1/2

CLint= ln 2 /( t1/2 x protein conc)

In vitro CLint

CLint’= CLint x (mg microsomes/g liver) x (g liver/kg bw)

Whole liver CLint

If well-stirred model
CLhep,met,= (Qh x fu x CLint’)/ (Qh + (fu x CLint’)
Hepatic metabolic CL

CL = CL(HepMet)+CL(HepBile)+CL(Renal)+ ....
Whole body CL
Lena Gustavsson 2015-11-08
Interplay between V and CL
Rat pharmacokinetics
10000
Concentration (nmol/L)

1000

Elimination half-life
100
A T1/2 = ln 2 x V / CL
10 B
C
1
0 5 10 15
Time (hours)
CL(mL/min/kg) Vss(L/kg) T½ (h)
A 20 14 10
B 70 12 3
C 80 0.6 0.5
Volume of
Clearance Absorption
distribution

Oral
Half-life
bioavailability

Dosing interval? Dose?

Lena Gustavsson 2015-11-08


DMPK in Drug Discovery and Development
Agenda
Introduction
Optimizing pharmacokinetic properties
Absorption & bioavailability
Distribution
Elimination – Clearance
Understanding clearance mechanisms
Drug metabolism
Drug transporters
Drug drug interactions and interindividual variability
Summary: Drug discovery and development

21
Clearance mechanisms
Total CL = CLMetHep + CLMetBile + CLRenal + .......

Hepatic
• Metabolism – phase I and phase II enzymes
• Bile excretion – sinusoidal and canalicular transporters

Renal
• Passive – glomerular filtration
• Active transport

Extrahepatic metabolism
• Intestinal CYP3A4
• Enzymes in blood
• Other extrahepatic enzymes

Lena Gustavsson 2015-11-08


Hepatic clearance mechanisms
Sinusoidal membrane

Blood Drug

Hepatocyte Drug uptake


Drug transporters

Drug metabolising
Metabolite enzymes

Canalicular Efflux
membrane transporters
Bile canaliculus

Lena Gustavsson 2015-11-08


Uptake & Efflux
Transporters
SLCs ABC series
Solute Carriers ATP Binding Cassette transporters
o OAT
o MDR
Organic Anion Transporter
Multi Drug Resistance proteins
o OCT
o MRP
Organic Cation Transporter
o OATP Multi drug Resistance-like Proteins
Organic Anion Transporting o White family
Polypeptides Drosophila white eye pigment gene

Blood Hepatocyte
Lena Gustavsson 2015-11-08
Drug transporters that influences drug
disposition – clinical evidence

From International Transporter Consortium


Giacomini et al Nature Rev Drug Disc 2010
Modified marking EMA recommended transporters in blue

Lena Gustavsson 2015-11-08


O OChiral
O
Pravastatin O
H
O O

O
3-methylglutaryl-coenzyme A (HMG-CoA) reductase
inhibitor; key enzyme in cholesterol synthesis
Used for the management of hypercholesterolaemia
The target is in the liver
Has short t1/2 (~2h), low F (17%) but successful
Has a good safety profile compared to other statins
WHY?

Lena Gustavsson 2015-11-08


Disposition of Pravastatin

OralOral
tablet Substrate for
tablet
OATP1B1
MRP2

Gut
OATP1B1

Liver
MRP2
Systemic
Circulation
t
Enterohepatic recirculation Kidney
of t
Active secretion

Lena Gustavsson 2015-11-08


Simvastatin-induced myopathy increased due to increased
plasma exposure - OATP1B1 polymorphism

Niemi, Clin.Pharm.Ther. 2010


Pasanen et al, Pharmacogenet. Genomics 2006
Search study N.Engl.J.Med. 2008

Lena Gustavsson 2015-11-08


Prediction of Human PK
In vitro, human

Scaling CL
Species differences Absorption (Caco-2)
Drug-drug interactions

In vitro, animal In vivo, human

Allometric scaling
In vitro/in vivo
Vss (dog, human PPB)
correlation
Absorption (rat)

In vivo, animal

Lena Gustavsson 2015-11-08


DMPK in Drug Discovery and Development
Agenda
Introduction
Optimizing pharmacokinetic properties
Absorption & bioavailability
Distribution
Elimination – Clearance
Understanding clearance mechanisms
Drug metabolism
Drug transporters
Drug drug interactions and interindividual variability
Summary: Drug discovery and development

30
Interindividual variability

Age
Sex
Genetics
Enzyme content
Liver weight
Organ blood flow
….
….
….

Nature Reviews Drug Discovery 6, 140-148 (February 2007) | doi:10.1038/nrd2173


http://www.simcyp.com
Interindividual variation in drug response

Lena Gustavsson 2015-11-08


CYP2D6 phenotypes in a Swedish population

Lena Gustavsson 2015-11-08


Codeine metabolism to morphine is
metabolised by CYP2D6

CYP2D6

Codeine Morphine
Prodrug Active metabolite
CYP2D6
Poor No formation of Lack of
metabolizer morphine analgesia

CYP2D6
Formation of Overdosing
Ultra-rapid
morphine Adverse events
metabolizer
Drug drug interactions - CYP inhibition
Does the drug inhibit Cytochrome P450?
Yes = Potential drug interactions!

P450 P450

metabolite metabolite

Lena Gustavsson 2015-11-08


Metabolism of terfenadine

OH OH

OH
CYP3A4 OH

N N

COOH

Terfenadine Active Metabolite


Almost complete Responsible for
first pass extraction efficacy in man
in man

Lena Gustavsson 2015-11-08


Ketoconazole
• Ketoconazole is an antifungal agent
O
• Potent inhibitor of CYP3A4
N
• IC50 value <1µM
N
• Antifungal dose is high (400mg twice
daily)
O
• Circulating concentrations of
O N
O N ketoconazole exceed IC50 for CYP3A4
Cl inhibition
Cl

Lena Gustavsson 2015-11-08


Ketoconazole – terfenadine interaction

OH OH

OH
CYP3A4 OH

N N

COOH

High circulating Low circulating


concentrations of concentrations of
terfenadine metabolite

Lena Gustavsson 2015-11-08


Implications of terfenadine – ketoconazole
interaction
• High circulating concentrations of terfenadine
• Potential to prolong QT interval of the ECG
• Abnormal heart rhythm
• Small numbers of patients go on to develop fatal
Torsade de Pointes (heart stops)
• Led to withdrawal of terfenadine from the market
• Increased questioning of Regulatory Authorities on QT
and DDIs

Lena Gustavsson 2015-11-08


CYP inhibition
Recombinant enzymes
Human liver microsomes

•CYP1A2, 2B6, 2C8, 2C9, 2C19, 2D6, 3A4


• Human recombinant P450 enzymes
• IC50 = µM
substrate
• If IC50 < 10 µM – potential interaction
• If [I]/Ki >0.1 - need to address in
product clinical study
CYP
* • Discovery: Automated fluorescence based

• Development: LC-MS/MS analysis of


metabolite

Lena Gustavsson 2015-11-08


Induction of P450 enzymes
Transcriptional regulation by nuclear hormone receptors

Aryl hydrocarbon Receptor (AhR)


• Ligands: Polyaromatic hydrocarbons,
dioxins (TCDD), Omeprazol - NUCLEUS -
• Target genes: CYP1A1, CYP1A2, CYP1B1 Arnt

CYP1A
Constitutive Androstane Receptor (CAR) PAH AhR Hsp90 RXR
• Ligands: Phenobarbital, CITCO
RXR
• Target genes: CYP2B6
CYP2B
PB CAR
Pregnane X Receptor (PXR) RXR
• Ligands: Rifampin, Carbamazepine CYP3A
• Target genes: CYP3A4, CYP2C8, CYP2C9, Rif PXR, GR? CYP2C
CYP2C19

• Cross-talk between nuclear hormone


receptors (AhR, CAR, PXR, GR, Hnf4
etc)
DDI Risk Assessment

Victim (substrate) Perpetrator (inhibitor/inducer)


• Enzyme/transporter • Enzyme/transporter IC50/Ki
phenotyping • Concentration plasma, liver,
• Drug disposition e.g. clearance intestine
• Fraction of total elimination • Bound vs unbound
• Mechanistic understanding • Time dependence
• Also includes polymorphism

Complex interactions – how to assess the risk?


• Integration of data my modeling and simulation – PBPK
• Iterative addition of new data
• Other relevant information
- Co-medications
- Biopharmaceutical Classification System etc
Physiology Based Pharmacokinetic (PBPK)
Modelling and Simulation

Jones and Rowland-Yeo 2013

Lena Gustavsson 2015-11-08


PBPK modelling and simulation
A DDI example – compound A
• Compound A is mainly metabolized by CYP3A4
• Assessment of DDI risks with compound A as a
”victim”
• How will the plasma concentration change when co-
dosing a potent CYP3A4 inhibitor
• How will the plasma concentration curve change
when co-dosing with a strong inducer of CYP3A4?
Median % fm and fe in absence of
inhibitor(s)

CYP3A4 Liver
CYP3A5 Liver
Renal

Lena Gustavsson 2015-11-08


Prediction of the effect of a CYP3A4
inhibitor on the AUC of compound A
250E+00
Systemic Concentration (ng/mL)

200E+00

150E+00
200 mg itraconazole QD x 20 days
3 mg compound A on day 12
100E+00

050E+00 AUC ratio = 3.1


0.000E+00
0 45 90 135 180 225 270 315 360 405 450
Time - Substrate (h)

CSys CSys with Interaction

• Co-administration of itraconazole (potent CYP3A4


inhibitor) may result in a 3 fold increase in the AUC of
compound A
• A clinical DDI study is required to investigate the effect
in vivo
45
Simulation of plasma concentration curves –
prediction of the effect of a CYP3A4 inducer
Day 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30
Compound
AF34134
A
3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3 3
rif 600 600 600 600 600 600 600 600 600 600 600

Compound A

• Co-administration of a strong CYP3A4 inducer, rifampicin, with compound


A leads to a decrease in AUC to 20%
• High risk of loosing the pharmacological efficacy of compound A
• Perform a clinical study to assess risk in vivo
DMPK in Drug Discovery and Development
Agenda
Introduction
Optimizing pharmacokinetic properties
Absorption & bioavailability
Distribution
Elimination – Clearance
Understanding clearance mechanisms
Drug metabolism
Drug transporters
Drug drug interactions and interindividual variability
Summary: Drug discovery and development

47
Understanding and predicting drug
disposition – an iterative process of data
integration

Input data

Lena Gustavsson 2015-11-08


Why study Drug Metabolism and
PharmacoKinetics?
DRUG DISCOVERY
Optimise compounds to get...
Good bioavailability – get to its target
Appropriate duration (1-2 doses/day)
Low potential for drug-drug interactions
…predicted to man

Provide basics for understanding of


Toxicology
Pharmacology From Rowland and Tozer, 1995

DRUG DEVELOPMENT
Provide understanding of drug disposition
Preclinical animal species – tox coverage
Human data

Assess the risk for drug drug interactions (DDI)


Decrease risk for drug drug interactions in the clinic
Impact on the design of clinical studies

Comply with guidelines from regulatory authorities

Lena Gustavsson 2015-11-08


THANKS FOR LISTENING!

50

You might also like