SlideShare une entreprise Scribd logo
1  sur  30
PRESENTATION ON:
TOXICOKINETICS EVALUATION IN PRECLINICAL STUDIED
Presented by: Under the guidence of:
Shivam 18033010 Dr. Phool Chandra
M Pharm 1st Year Associate professor
Pharmacology HOD PharmacologyI F T M
U N I V E R S I T Y
M O R A D A B A D
N A A C A C C R E D I T E D
SCHOOL OF PHARMACEUTICAL SCIENCE
TOXICOKINETICS
Toxicokinetics deals with absorption, distribution
biotransformation (biotransformation) and excretion of
chemicals
A. Toxicokinetics
i) Absorption
Absorption is the process by which the chemical enters the
body. It depends on the route of administration
• Oral route – the GIT is the most important route of
absorption, as most acute poisonings involve ingestions.
• Dermal route – lipid solubility of a substance is an
important factor affecting the degree of absorption through the
skin
• Inhalational route – toxic fumes, particulate and
noxious gases may be absorbed through the lungs.
Bioavailability
• is the fraction of unchanged drug reaching the systemic
circulation following of non-vascular administration.
Therefore, a portion of the chemical fails to reach the
systemic
circulation in original form after oral administration
ii) Distribution
• Distribution-is defined as the apparent volume into
which
a substance is distributed. Volume of distribution (Vd) is
calculated from the dose taken and the resulting plasma
concentration:
Vd = dose /plasma concentration
• The importance of volume of distribution in toxicology is
- Predicting peak blood concentration of the chemical taken
- Calculating the amount of substance in the body to verify
the quantity ingested
- Deciding whether to apply systemic toxin elimination
technique
• Factors determining the rate of distribution of chemicals in
the body are
- Protein binding – chemicals highly bound to protein have
small volume of distribution
• Plasma concentration – when the volume of distribution of
chemicals is small, most of the chemical remains in the plasma
• Physiological barriers – chemicals will not uniformly
distributed to the body due to specialized barriers e.g blood
brain barrier
iii) Biotransformation (metabolism)
• Biotransformation is the biochemical transformation of a
chemical. It is a process by which the body transforms a
chemical and makes it more water soluble so the chemical can
be eliminated more rapidly via the kidney into the urine.
Biotransformation can produce metabolites that are
pharmacologically active and toxic
E.g. parathion→ parathoxon (toxic metabolite). Liver is the major
site of biotransformation for many chemicals & other organs
that are involved are lungs, kidneys, skin &so on. Interactions
during biotransformation include
• There are two phases of biotransformation
• Phase I – the drug is converted into more polar compound
e.g oxidation, reduction, &hydrolysis
• Phase II (conjugation) – a drug or its metabolite is
conjugated with an endogenous substance
e.g glucuronide conjugate
• Enzyme inhibition- by this the biotransformation of drugs is
delayed & is a cause of increased toxicity
• Enzyme induction- by this the biotransformation of
drugs is
accelerated & is a cause of therapeutic failure
• First – pass effect – is the biotransformation of some
chemicals by the liver during the initial pass from the
portal
circulation after oral administration.
• Half life (t ½) –is the time required to reduce the
blood
concentration of the chemical to half.
IV) Excretion
Excretion is the final means of chemical elimination, either as
metabolites or unchanged parent chemical. Excretion through
the lungs is the major route for gaseous substances; and in the
case of non-volatile water – soluble drugs, the kidneys are the
most important routes of excretion. Additional routes include
sweat, saliva, tears, nasal secretions, milk, bile and feces
• Clearance – elimination of chemicals from the body may
be described by the term clearance (CL). It is a quantitative
measure of the volume of blood cleared of drug per unit time,
usually expressed in milliliter per 4 minute
GENERAL PRINCIPLES TO BE CONSIDERED
1. Quantification of exposure .
2. Setting of dose levels
•Low dose levels
•Intermediate dose levels
•High dose levels
3.Extent of exposure assessment in toxicity studies
4.Complicating factors in exposure interpretation
5.Route of administration.
6.Determination of metabolites
7.Statistical evaluation of data.
8.Analytical methods
8.Reporting.
Introduction
•Toxicokinetics (TK) is defined by The International Conference on
Harmonization (ICH) as ‘the generation of pharmacokinetic data, either as
an integral component in the conduct of non-clinical toxicity studies.
• The need for toxicokinetic data and the extent of exposure assessment in
individual toxicity studies should be based on a flexible step-by-step
approach.
• The primary objective of the toxicokinetic studies is to describe the
systemic exposure achieved in animals and its relationship to dose level
and the time course of the toxicity study .
• Choice of species and treatment regimen used in non clinical studies.
Lastly, information on systemic exposure of animals during repeated-dose
toxicity studies is essential for the interpretation of study results.
1. Quantification and extent of exposure :
• The exposure might be represented by plasma (serum or blood) concentrations or
the AUCs of parent compound and/or metabolite(s) and sometimes by tissue
concentrations.
• Quantification of exposure provides an assessment of the burden on the test
species and helps in the interpretation of similarities and differences in toxicity
across species, dose groups and sexes.
• When designing the toxicity studies, the exposure and dose-dependence in
humans at therapeutic dose levels (either expected or established), should be
considered in order to achieve relevant exposure at various dose levels in the
animal toxicity studies.
• Species differences in the pharmacodynamics of the substance (either qualitative
or quantitative) should also be taken into consideration because sometimes it may
have other effects. This information may allow better interspecies comparisons
than simple dose/body weight (or surface area) comparisons.
2. Extent of exposure
•Systemic exposure should be estimated in an appropriate number of
animals and dose groups to provide a basis for risk assessment.
•Concomitant toxicokinetics may be performed either in all or a
representative proportion of the animals used in the main study or in
special satellite groups.
•Both male and female animals are utilized in the main study it is
normal to estimate exposure in animals of both sexes unless some
justification can be made for not so doing.
•Toxicokinetic data is not mandatory for studies of different duration
if the dosing regimen is essentially unchanged.
3. Sampling points
•In concomitant toxicokinetic studies the time points for collecting body
fluids should be as frequent as is necessary
• There are also strict restrictions on blood volume available (no more than
10% of circulating volume can be taken).
•Sample size is typically 0.25–0.50 ml day−1 in rodents and up to1ml day−1
in non-rodents In each study, justification of number of time points should
be made on the basis that they are adequate to estimate exposure.
•The justification should be based on kinetic data gathered from earlier
toxicity studies, from pilot or dose range finding studies, extrapolation.
•Sampling times vary based on the presence (or lack) of pharmacokinetic
data, but are often taken 0.5, 1.0, 2.0, 4.0, 8.0, 12.0 and 24.0 h post-dose, with
only the parent drug generally being measured.
4. Dose level setting
•Dose level for toxicity studies is largely regulated by the
toxicology findings and the pharmacodynamic responses of the
test species.
•Intermediate dose levels should normally represent an
appropriate multiple (or fraction) of the exposure at lower (or
higher) dose levels dependent upon the objectives of the toxicity
study.
•The high dose levels in toxicity studies will normally be
determined by toxicological considerations.
the exposure achieved at the dose levels used should be
assessed.
•This toxicokinetic data indicate that absorption of a compound
limits exposure to parent compound and/or metabolite(s), the
lowest dose level of the substance
• non-linear kinetics should not necessarily result in dose
limitations in toxicity studies.
5. Ratifying factors on study to be considered
•Earlier we discussed the species and sex differences and their
effect on toxicokinetics.
•There are other factors to be considered in this study is protein
binding, tissue uptake, receptor properties and metabolic profile.
•Systemic exposure may be decreased by protein binding and
tissue uptake. I
•Due to the metabolism there will be formation of
pharmacological active metabolites, the toxic metabolites and
antigenic biotechnology products metabolites.
6. Route of administration
•Pharmacokinetics of a substance is greatly affected by the
route of administration.
•For instance orally administered drugs bioavailability time is
more than other routes.
•If the drug is intended to administer through oral route then
oral toxicity should be checked.
•If any drug administering route is already established and new
clinical route of administration is going to establish then it will
be necessary to ascertain whether changing the clinical route
will significantly reduce the safety margin.
•In this case focusing on local toxicity is essential.
7. Metabolite determination
Many of the cases systemic exposure and toxic effect consider
on the basis of parent drug concentration. However, there may
be circumstances when measurement of metabolite
concentrations in plasma or other body fluids is especially
important in the conduct of toxicokinetics. They are
•If it is a 'pro-drug' and the delivered metabolite is
acknowledged to be the primary active entity.
•If the compound is metabolised to one or more
pharmacologically or toxicologically active metabolites which
could make a significant contribution to tissue/organ
responses.
•For the drugs which are extensively metabolized and the
metabolite is only the quantifiable factor.
8. Statistical evaluation of data
•The data should be evaluated statistically which allows
assessment of the exposure.
•Toxicokinetic values are normally calculated as mean SD;
statistical evaluation is not usually performed
•However, because large intra- and inter-individual variation of
kinetic parameters may occur and small numbers of animals are
involved in generating toxicokinetic data,
Ahigh level of precision in terms of statistics is not normally
needed.
•Consideration should be given to the calculation of mean or
median values and estimates of variability,
•If data transformation (e.g. logarithmic) is performed, a rationale
should be provided.
9. Analytical methods
•Regulatory authorities expects that analytical methods used to determine
plasma concentrations of pharmaceuticals are of adequate sensitivity and
precision.
•For evaluation validated analytical methods used and conforms to Good
Laboratory Practice (GLP). Analytical methods used in such studies
include gas chromatography (although this is rarely used), HPLC (UV or
fluorescence), LC, LC–MS, LC-MS-MS, and capillary electrophoresis
(again, rarely used, and more for proteins).
•Generally, toxicity studies use a range of time-points and replicates to
provide toxicokinetic data as we discussed earlier, although staggered and
sparse sampling.
•For replicate designs, toxicokinetic measurements are taken at similar
pre-set timepoints and the mean of the measured values is then taken to
provide an estimate of drug exposure.
•Although staggered designs are less sensitive, they are still used by
various pharmaceutical companies for rodent and/or primate studies.
1. Toxicokinetic studies in Preclinicalstage:
a. Safety assessment
b. Single dose and rising dose studies Repeated-dose
toxicity studies
c. Repeated-dose toxicity studies
d. Genotoxicity studies
e. Reproduction toxicity studies
i. Studies of fertility
ii. In pregnant and lactating animals
e. Carcinogenicity studies:
2. Toxicokinetic studies in clinical phases:
a. Approaches to decrease the animal usage in
toxicokinetics
b. Dried blood spot technology
c. Alternative approaches to animal models
d. Physiologically based pharmacokinetic (PBPK) modeling
1. Toxicokinetic studies in Preclinical stage:
a. Safety assessment
•Generally safety of a molecule can be performed in in-vivo
systems.
•This step is not included in the guidelines but it is very useful
for the researchers to assess the systemic exposure of the
molecule and its effect on it.
•This safety study is integral part in the central nervous
system (CNS), cardio vascular system (CVS) and respiratory
assessments.
b. Single dose and rising dose studies
•These studies are often performed in a very early phase of
drug development before a bioanalytical method has been
developed.
•These studies are usually performed in rodents.
•Plasma samples may be taken in such studies and stored for
later analysis.
c. Repeated-dose toxicity studies
•To give support for phase 1 studies this study is carried out for
four weeks in both rodents as well as non-rodents.
•The treatment regimen (Note 11) and species should be
selected whenever possible with regard to pharmacodynamic
and pharmacokinetic principles.
• This may not be achievable for the very first studies, at a time
when neither animal nor human pharmacokinetic data are
normally available.
d. Genotoxicity studies
•Two in vitro studies and one in vivo study is essential to support
development of drug .
•In vivo investigations usually use a rodent micronucleus (bone
marrow or peripheral erythrocytes) test or chromosome
aberration (bone marrow cells) test.
•These are the well established studies for the genotoxicity
evaluation.
e. Reproduction toxicity studies
Reproduction toxicity measurements are taken in studies of
fertility (rat), embryo-foetal development (rat and rabbit) and
peri- or post-natal development (rat).
a) Studies of fertility
Assessment of fertility toxicity has very important, because most
of the drugs used in fertility conditions so has to strengthen
at that time.
Usually this can be done in rats.
b. In pregnant and lactating animals:
There is a regulatory expectation for toxicokinetic data in
pregnant animals, although no specific guidance is
given1,16. Data from non-pregnant animals is useful to set
dose levels, and the limitation of exposure is usually
governed by maternal toxicity. Toxicokinetics may involve
exposure assessment of dams, embryos, foetuses or newborn
at specified days. Secretion in milk may be assessed to define
its role in the exposure of newborns..
f. Carcinogenicity studies:
•Sometimes drugs are used for longtime for curing purposes, this may
lead to the toxicity or carcinogenicity.
•So lifetime studies in the rodent are needed to support the long-term
clinical use of pharmaceuticals17 and non-rodents can also be used.
•Dose selection is usually determined as the maximum tolerated dose
(MTD
•Selection based on AUC is less common as a 25-fold ratio is often not
feasible.
• It is recommended that monitoring should occur on a few occasions
during the study, although it is not essential for monitoring to occur
beyond six months.
• However, pharmaceutical companies use various strategies for such
monitoring times
2. Toxicokinetic studies in clinical phases:
Regulatory bodies around the world outlining that toxicity studies are
necessary to support human Phase I, II and III studies, and product
license application is available.
•The magnitude of the preclinical toxicokinetic evaluation for each
clinical phase varies significantly among pharmaceutical companies.
•For Phase I investigations the company might only generate.
toxicokinetic data from the four-week repeat-dose toxicitystudies.
•Full pharmacokinetic profile (including in vitro metabolism studies),
and toxicokinetic measurements from four- and 13-week repeat-dose
toxicitystudies prior to Phase I is necessary.
•Toxicity assessments enable the No Observed Effect Level (NOEL) or
No Observed Adverse Effect Level (NOAEL) to be established for a
potential new drug, based on clinical observations, bodyweight, food
consumption, clinical pathology, organ weights, necropsy examination,
and histopathology.
a. Approaches to decrease the animal usage in toxicokinetics:
• To increase the generation of toxicokinetic data it has to increase
the usage of number of rodents through the use of satellite groups.
• As discussed earlier using of animal’s number and sex is restricted
as per OECD-417 guidelines even in blood samplings. For this
purpose many alternative approaches has generated.
b. Dried blood spot technology:
• Recently toxicokinetic studies in dogs by applying dried blood spot
technology published and also it is in improvement stages to use
this model in rodents (bar).
• In Pharmaceutical industry this type of methodology for
toxicokinetics has been developed and combined with high
performance liquid chromatography–mass spectrometry (HPLC–
MS/ MS) to collect and analyse very small amounts of biofluids.
c. Alternative approaches to animal models
• Using alternative models to animal models can reduce the number of
animals in confirmative studies
• Although this is not basis for the use of the chemical entity in clinical
phases, but prior to human usage it should check with the animal
studies.
d. Physiologically based pharmacokinetic (PBPK) modeling:
• PBPK models can scientifically support risk assessment by facilitating
extrapolation between species and exposure routes, and from high to
low doses.
• Loizou et al., 2008 used this approach to understand the relationship
between external and internal exposures, helping in dose response
characterisation and the interpretation of biomarker levels
determined in bio-monitoring studies
References:
1. CPMP/ICH/384/95: Operational June 1995. Toxicokinetics: guidance
for assessing systemic exposure in toxicology studies. Also known asICH
Topic S3A (see http :// www. emea.eu.int)
2. Kate, R., Case, D.E., Hakes, H., Nod, K., Salami, F., Horii,I., Mayahara,
H.,Cayeb n, M.N., Marriott, T.B., Igarashi, T., Toxicokinetics: its
significance and practical problems.J.Toxicol.Sci.18:211-238;( 1993).
3. CPMP/ICH/286/95, modification: operational November 2000.
Nonclinical safety studies for the conduct of human clinical trials for
pharmaceuticals. Also known as ICH Topic M3
(seehttp://www.emea.eu.int)
4. CPMP/SWP/1042 Corr: operational October 2000. Note for guidance
on repeated dose toxicity (see http://www.emea.eu.int)
5. LaDu, B.N. Fundamentals of Drug Metabolism and Drug Disposition,
Robert E Krieger Publishing Company, New York, pp.528 (1979)
6. Igarashi, T. and Sekido, T. Case studies for statistical analysis of
toxicokinetic data. Regul. Toxicol. Pharmacol. 23:193–208; (1996).
Toxicokinetic evaluation in preclinical studies by Shivam Diwaker

Contenu connexe

Tendances

IND Enabling Studies by Kashikant Yadav
IND Enabling Studies by Kashikant YadavIND Enabling Studies by Kashikant Yadav
IND Enabling Studies by Kashikant YadavKashikant Yadav
 
Toxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptxToxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptxARSHIKHANAM4
 
Safety pharmacology (siri)
Safety pharmacology (siri)Safety pharmacology (siri)
Safety pharmacology (siri)Ramavath Aruna
 
Herg assay,Structure, Various screening methods and Advantages
Herg assay,Structure,  Various screening methods and AdvantagesHerg assay,Structure,  Various screening methods and Advantages
Herg assay,Structure, Various screening methods and AdvantagesUrvashi Shakarwal
 
Alternative methods to animal toxicity testing
Alternative methods to        animal toxicity testingAlternative methods to        animal toxicity testing
Alternative methods to animal toxicity testingSachin Sharma
 
Safety pharmacology tier 2
Safety pharmacology  tier 2Safety pharmacology  tier 2
Safety pharmacology tier 2ragini Dani
 
Female reproductive toxicity studies acoording to SECHDULE Y AND ICH S5R3
Female reproductive toxicity studies acoording to SECHDULE Y AND ICH S5R3Female reproductive toxicity studies acoording to SECHDULE Y AND ICH S5R3
Female reproductive toxicity studies acoording to SECHDULE Y AND ICH S5R3SONALPANDE5
 
Dermal Irritation and Dermal Toxicity Studies
Dermal Irritation and Dermal Toxicity Studies Dermal Irritation and Dermal Toxicity Studies
Dermal Irritation and Dermal Toxicity Studies Dinesh Gangoda
 
Regulatory guidelines for conducting toxicity studies
Regulatory guidelines for conducting toxicity studiesRegulatory guidelines for conducting toxicity studies
Regulatory guidelines for conducting toxicity studiesHimikaRathi
 
test item characterization of regulatory of toxicological studies
test item characterization of regulatory of toxicological studies test item characterization of regulatory of toxicological studies
test item characterization of regulatory of toxicological studies SonaliJain736101
 
Safety Pharmacology
Safety PharmacologySafety Pharmacology
Safety PharmacologyPavana K A
 
TEST ITEM CHARACTERIZATION IN REGULATORY TOXICOLOGY STUDIES.pptx
TEST ITEM CHARACTERIZATION IN REGULATORY TOXICOLOGY STUDIES.pptxTEST ITEM CHARACTERIZATION IN REGULATORY TOXICOLOGY STUDIES.pptx
TEST ITEM CHARACTERIZATION IN REGULATORY TOXICOLOGY STUDIES.pptxashharnomani
 
Safety pharmacology ppt
Safety pharmacology pptSafety pharmacology ppt
Safety pharmacology pptShilpa thakur
 
PPT On Female Reproductive Toxicology
PPT On Female Reproductive Toxicology PPT On Female Reproductive Toxicology
PPT On Female Reproductive Toxicology Naveen K L
 
Assignment on Toxicokinetics
Assignment on ToxicokineticsAssignment on Toxicokinetics
Assignment on ToxicokineticsDeepak Kumar
 
Oecd acute,subacte, sub chronic dermal toxicity studies(402, 410, 411).
Oecd acute,subacte, sub chronic dermal toxicity studies(402, 410, 411).Oecd acute,subacte, sub chronic dermal toxicity studies(402, 410, 411).
Oecd acute,subacte, sub chronic dermal toxicity studies(402, 410, 411).helasri gummadi
 
IND (Investigational New Drug) industrial perspective
IND (Investigational New Drug) industrial perspectiveIND (Investigational New Drug) industrial perspective
IND (Investigational New Drug) industrial perspectiveAYESHA NAZEER
 

Tendances (20)

Safety pharmacology
Safety pharmacologySafety pharmacology
Safety pharmacology
 
IND Enabling Studies by Kashikant Yadav
IND Enabling Studies by Kashikant YadavIND Enabling Studies by Kashikant Yadav
IND Enabling Studies by Kashikant Yadav
 
Toxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptxToxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptx
 
Safety pharmacology (siri)
Safety pharmacology (siri)Safety pharmacology (siri)
Safety pharmacology (siri)
 
Herg assay,Structure, Various screening methods and Advantages
Herg assay,Structure,  Various screening methods and AdvantagesHerg assay,Structure,  Various screening methods and Advantages
Herg assay,Structure, Various screening methods and Advantages
 
Safety pharmacology
Safety pharmacologySafety pharmacology
Safety pharmacology
 
Alternative methods to animal toxicity testing
Alternative methods to        animal toxicity testingAlternative methods to        animal toxicity testing
Alternative methods to animal toxicity testing
 
Safety pharmacology tier 2
Safety pharmacology  tier 2Safety pharmacology  tier 2
Safety pharmacology tier 2
 
Female reproductive toxicity studies acoording to SECHDULE Y AND ICH S5R3
Female reproductive toxicity studies acoording to SECHDULE Y AND ICH S5R3Female reproductive toxicity studies acoording to SECHDULE Y AND ICH S5R3
Female reproductive toxicity studies acoording to SECHDULE Y AND ICH S5R3
 
Dermal Irritation and Dermal Toxicity Studies
Dermal Irritation and Dermal Toxicity Studies Dermal Irritation and Dermal Toxicity Studies
Dermal Irritation and Dermal Toxicity Studies
 
Regulatory guidelines for conducting toxicity studies
Regulatory guidelines for conducting toxicity studiesRegulatory guidelines for conducting toxicity studies
Regulatory guidelines for conducting toxicity studies
 
test item characterization of regulatory of toxicological studies
test item characterization of regulatory of toxicological studies test item characterization of regulatory of toxicological studies
test item characterization of regulatory of toxicological studies
 
REPRODUCTIVE TOXICITY STUDIES
REPRODUCTIVE TOXICITY STUDIESREPRODUCTIVE TOXICITY STUDIES
REPRODUCTIVE TOXICITY STUDIES
 
Safety Pharmacology
Safety PharmacologySafety Pharmacology
Safety Pharmacology
 
TEST ITEM CHARACTERIZATION IN REGULATORY TOXICOLOGY STUDIES.pptx
TEST ITEM CHARACTERIZATION IN REGULATORY TOXICOLOGY STUDIES.pptxTEST ITEM CHARACTERIZATION IN REGULATORY TOXICOLOGY STUDIES.pptx
TEST ITEM CHARACTERIZATION IN REGULATORY TOXICOLOGY STUDIES.pptx
 
Safety pharmacology ppt
Safety pharmacology pptSafety pharmacology ppt
Safety pharmacology ppt
 
PPT On Female Reproductive Toxicology
PPT On Female Reproductive Toxicology PPT On Female Reproductive Toxicology
PPT On Female Reproductive Toxicology
 
Assignment on Toxicokinetics
Assignment on ToxicokineticsAssignment on Toxicokinetics
Assignment on Toxicokinetics
 
Oecd acute,subacte, sub chronic dermal toxicity studies(402, 410, 411).
Oecd acute,subacte, sub chronic dermal toxicity studies(402, 410, 411).Oecd acute,subacte, sub chronic dermal toxicity studies(402, 410, 411).
Oecd acute,subacte, sub chronic dermal toxicity studies(402, 410, 411).
 
IND (Investigational New Drug) industrial perspective
IND (Investigational New Drug) industrial perspectiveIND (Investigational New Drug) industrial perspective
IND (Investigational New Drug) industrial perspective
 

Similaire à Toxicokinetic evaluation in preclinical studies by Shivam Diwaker

Toxicokinetics toxicology pre-clinical pharmacology
Toxicokinetics  toxicology  pre-clinical pharmacologyToxicokinetics  toxicology  pre-clinical pharmacology
Toxicokinetics toxicology pre-clinical pharmacologyDilip Kawane
 
Toxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptxToxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptxashharnomani
 
aaGeneral pharmayuuuuuyyjhuucofhjlogy.pptx
aaGeneral pharmayuuuuuyyjhuucofhjlogy.pptxaaGeneral pharmayuuuuuyyjhuucofhjlogy.pptx
aaGeneral pharmayuuuuuyyjhuucofhjlogy.pptxseraphimkassa
 
Microsomal assays toxicokinetics taxicokinetic evaluation in preclinical st...
Microsomal assays  toxicokinetics  taxicokinetic evaluation in preclinical st...Microsomal assays  toxicokinetics  taxicokinetic evaluation in preclinical st...
Microsomal assays toxicokinetics taxicokinetic evaluation in preclinical st...siva ganesh
 
Pharmacology unit I.pptx
Pharmacology unit I.pptxPharmacology unit I.pptx
Pharmacology unit I.pptxAsifAliLashari2
 
understanding-the-fundamental-principles-of-biopharmaceutics-pharmacokinetics...
understanding-the-fundamental-principles-of-biopharmaceutics-pharmacokinetics...understanding-the-fundamental-principles-of-biopharmaceutics-pharmacokinetics...
understanding-the-fundamental-principles-of-biopharmaceutics-pharmacokinetics...MaryRoseFraga
 
TOXICOPHARMACOLOGY.pdf
TOXICOPHARMACOLOGY.pdfTOXICOPHARMACOLOGY.pdf
TOXICOPHARMACOLOGY.pdfRAMDAS BHAT
 
toxicokinetics-1.pptx
toxicokinetics-1.pptxtoxicokinetics-1.pptx
toxicokinetics-1.pptxUnnatikhare1
 
General Pharmacology for Physiotherapists
General Pharmacology for PhysiotherapistsGeneral Pharmacology for Physiotherapists
General Pharmacology for PhysiotherapistsDr. Sanjib Kumar Das
 
S01-Introduction-to-PK.pptx
S01-Introduction-to-PK.pptxS01-Introduction-to-PK.pptx
S01-Introduction-to-PK.pptxWiamKhalil1
 
General Pharmacology part 02.pptx
General Pharmacology part 02.pptxGeneral Pharmacology part 02.pptx
General Pharmacology part 02.pptxMAYUR HARAL
 

Similaire à Toxicokinetic evaluation in preclinical studies by Shivam Diwaker (20)

Toxicokinetics toxicology pre-clinical pharmacology
Toxicokinetics  toxicology  pre-clinical pharmacologyToxicokinetics  toxicology  pre-clinical pharmacology
Toxicokinetics toxicology pre-clinical pharmacology
 
Toxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptxToxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptx
 
Toxicology
ToxicologyToxicology
Toxicology
 
aaGeneral pharmayuuuuuyyjhuucofhjlogy.pptx
aaGeneral pharmayuuuuuyyjhuucofhjlogy.pptxaaGeneral pharmayuuuuuyyjhuucofhjlogy.pptx
aaGeneral pharmayuuuuuyyjhuucofhjlogy.pptx
 
Toxikokinetics
ToxikokineticsToxikokinetics
Toxikokinetics
 
Microsomal assays toxicokinetics taxicokinetic evaluation in preclinical st...
Microsomal assays  toxicokinetics  taxicokinetic evaluation in preclinical st...Microsomal assays  toxicokinetics  taxicokinetic evaluation in preclinical st...
Microsomal assays toxicokinetics taxicokinetic evaluation in preclinical st...
 
Ppt tdm new
Ppt tdm newPpt tdm new
Ppt tdm new
 
Pharmacology unit I.pptx
Pharmacology unit I.pptxPharmacology unit I.pptx
Pharmacology unit I.pptx
 
understanding-the-fundamental-principles-of-biopharmaceutics-pharmacokinetics...
understanding-the-fundamental-principles-of-biopharmaceutics-pharmacokinetics...understanding-the-fundamental-principles-of-biopharmaceutics-pharmacokinetics...
understanding-the-fundamental-principles-of-biopharmaceutics-pharmacokinetics...
 
TOXICOPHARMACOLOGY.pdf
TOXICOPHARMACOLOGY.pdfTOXICOPHARMACOLOGY.pdf
TOXICOPHARMACOLOGY.pdf
 
Biopharm review1
Biopharm review1Biopharm review1
Biopharm review1
 
Pharmacology.pptx
Pharmacology.pptxPharmacology.pptx
Pharmacology.pptx
 
pharmacokinetics ppt
pharmacokinetics pptpharmacokinetics ppt
pharmacokinetics ppt
 
Absorption
AbsorptionAbsorption
Absorption
 
toxicokinetics-1.pptx
toxicokinetics-1.pptxtoxicokinetics-1.pptx
toxicokinetics-1.pptx
 
Toxicokinetics 4042019
Toxicokinetics 4042019Toxicokinetics 4042019
Toxicokinetics 4042019
 
Pharmacokinetics
PharmacokineticsPharmacokinetics
Pharmacokinetics
 
General Pharmacology for Physiotherapists
General Pharmacology for PhysiotherapistsGeneral Pharmacology for Physiotherapists
General Pharmacology for Physiotherapists
 
S01-Introduction-to-PK.pptx
S01-Introduction-to-PK.pptxS01-Introduction-to-PK.pptx
S01-Introduction-to-PK.pptx
 
General Pharmacology part 02.pptx
General Pharmacology part 02.pptxGeneral Pharmacology part 02.pptx
General Pharmacology part 02.pptx
 

Plus de Shivam Diwaker

Anatomy and Physiology of Heart.pdf
Anatomy and Physiology of Heart.pdfAnatomy and Physiology of Heart.pdf
Anatomy and Physiology of Heart.pdfShivam Diwaker
 
Scope of Anatomy and physiology.pptx
Scope of Anatomy and physiology.pptxScope of Anatomy and physiology.pptx
Scope of Anatomy and physiology.pptxShivam Diwaker
 
Screening methods of immunomodulators by shivam diwaker
Screening methods of immunomodulators by shivam diwakerScreening methods of immunomodulators by shivam diwaker
Screening methods of immunomodulators by shivam diwakerShivam Diwaker
 
Oral contraceptives by shivam
Oral contraceptives by shivamOral contraceptives by shivam
Oral contraceptives by shivamShivam Diwaker
 
Cerebral palsy by shivam
Cerebral palsy by shivamCerebral palsy by shivam
Cerebral palsy by shivamShivam Diwaker
 
Pharmacology of free radicals
Pharmacology of free radicalsPharmacology of free radicals
Pharmacology of free radicalsShivam Diwaker
 

Plus de Shivam Diwaker (6)

Anatomy and Physiology of Heart.pdf
Anatomy and Physiology of Heart.pdfAnatomy and Physiology of Heart.pdf
Anatomy and Physiology of Heart.pdf
 
Scope of Anatomy and physiology.pptx
Scope of Anatomy and physiology.pptxScope of Anatomy and physiology.pptx
Scope of Anatomy and physiology.pptx
 
Screening methods of immunomodulators by shivam diwaker
Screening methods of immunomodulators by shivam diwakerScreening methods of immunomodulators by shivam diwaker
Screening methods of immunomodulators by shivam diwaker
 
Oral contraceptives by shivam
Oral contraceptives by shivamOral contraceptives by shivam
Oral contraceptives by shivam
 
Cerebral palsy by shivam
Cerebral palsy by shivamCerebral palsy by shivam
Cerebral palsy by shivam
 
Pharmacology of free radicals
Pharmacology of free radicalsPharmacology of free radicals
Pharmacology of free radicals
 

Dernier

Solving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxSolving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxOH TEIK BIN
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxRoyAbrique
 
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdfssuser54595a
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdfSoniaTolstoy
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxNirmalaLoungPoorunde1
 
microwave assisted reaction. General introduction
microwave assisted reaction. General introductionmicrowave assisted reaction. General introduction
microwave assisted reaction. General introductionMaksud Ahmed
 
Hybridoma Technology ( Production , Purification , and Application )
Hybridoma Technology  ( Production , Purification , and Application  ) Hybridoma Technology  ( Production , Purification , and Application  )
Hybridoma Technology ( Production , Purification , and Application ) Sakshi Ghasle
 
Measures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and ModeMeasures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and ModeThiyagu K
 
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...EduSkills OECD
 
MENTAL STATUS EXAMINATION format.docx
MENTAL     STATUS EXAMINATION format.docxMENTAL     STATUS EXAMINATION format.docx
MENTAL STATUS EXAMINATION format.docxPoojaSen20
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13Steve Thomason
 
mini mental status format.docx
mini    mental       status     format.docxmini    mental       status     format.docx
mini mental status format.docxPoojaSen20
 
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Krashi Coaching
 
Presiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsPresiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsanshu789521
 
Mastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionMastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionSafetyChain Software
 
Arihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdfArihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdfchloefrazer622
 
_Math 4-Q4 Week 5.pptx Steps in Collecting Data
_Math 4-Q4 Week 5.pptx Steps in Collecting Data_Math 4-Q4 Week 5.pptx Steps in Collecting Data
_Math 4-Q4 Week 5.pptx Steps in Collecting DataJhengPantaleon
 
Grant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy ConsultingGrant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy ConsultingTechSoup
 

Dernier (20)

Solving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxSolving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptx
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
 
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
 
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdfBASLIQ CURRENT LOOKBOOK  LOOKBOOK(1) (1).pdf
BASLIQ CURRENT LOOKBOOK LOOKBOOK(1) (1).pdf
 
TataKelola dan KamSiber Kecerdasan Buatan v022.pdf
TataKelola dan KamSiber Kecerdasan Buatan v022.pdfTataKelola dan KamSiber Kecerdasan Buatan v022.pdf
TataKelola dan KamSiber Kecerdasan Buatan v022.pdf
 
Employee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptxEmployee wellbeing at the workplace.pptx
Employee wellbeing at the workplace.pptx
 
microwave assisted reaction. General introduction
microwave assisted reaction. General introductionmicrowave assisted reaction. General introduction
microwave assisted reaction. General introduction
 
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
 
Hybridoma Technology ( Production , Purification , and Application )
Hybridoma Technology  ( Production , Purification , and Application  ) Hybridoma Technology  ( Production , Purification , and Application  )
Hybridoma Technology ( Production , Purification , and Application )
 
Measures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and ModeMeasures of Central Tendency: Mean, Median and Mode
Measures of Central Tendency: Mean, Median and Mode
 
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
 
MENTAL STATUS EXAMINATION format.docx
MENTAL     STATUS EXAMINATION format.docxMENTAL     STATUS EXAMINATION format.docx
MENTAL STATUS EXAMINATION format.docx
 
The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13The Most Excellent Way | 1 Corinthians 13
The Most Excellent Way | 1 Corinthians 13
 
mini mental status format.docx
mini    mental       status     format.docxmini    mental       status     format.docx
mini mental status format.docx
 
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
Kisan Call Centre - To harness potential of ICT in Agriculture by answer farm...
 
Presiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha electionsPresiding Officer Training module 2024 lok sabha elections
Presiding Officer Training module 2024 lok sabha elections
 
Mastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionMastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory Inspection
 
Arihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdfArihant handbook biology for class 11 .pdf
Arihant handbook biology for class 11 .pdf
 
_Math 4-Q4 Week 5.pptx Steps in Collecting Data
_Math 4-Q4 Week 5.pptx Steps in Collecting Data_Math 4-Q4 Week 5.pptx Steps in Collecting Data
_Math 4-Q4 Week 5.pptx Steps in Collecting Data
 
Grant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy ConsultingGrant Readiness 101 TechSoup and Remy Consulting
Grant Readiness 101 TechSoup and Remy Consulting
 

Toxicokinetic evaluation in preclinical studies by Shivam Diwaker

  • 1. PRESENTATION ON: TOXICOKINETICS EVALUATION IN PRECLINICAL STUDIED Presented by: Under the guidence of: Shivam 18033010 Dr. Phool Chandra M Pharm 1st Year Associate professor Pharmacology HOD PharmacologyI F T M U N I V E R S I T Y M O R A D A B A D N A A C A C C R E D I T E D SCHOOL OF PHARMACEUTICAL SCIENCE
  • 2. TOXICOKINETICS Toxicokinetics deals with absorption, distribution biotransformation (biotransformation) and excretion of chemicals A. Toxicokinetics i) Absorption Absorption is the process by which the chemical enters the body. It depends on the route of administration • Oral route – the GIT is the most important route of absorption, as most acute poisonings involve ingestions. • Dermal route – lipid solubility of a substance is an important factor affecting the degree of absorption through the skin
  • 3. • Inhalational route – toxic fumes, particulate and noxious gases may be absorbed through the lungs. Bioavailability • is the fraction of unchanged drug reaching the systemic circulation following of non-vascular administration. Therefore, a portion of the chemical fails to reach the systemic circulation in original form after oral administration ii) Distribution • Distribution-is defined as the apparent volume into which a substance is distributed. Volume of distribution (Vd) is calculated from the dose taken and the resulting plasma concentration: Vd = dose /plasma concentration
  • 4. • The importance of volume of distribution in toxicology is - Predicting peak blood concentration of the chemical taken - Calculating the amount of substance in the body to verify the quantity ingested - Deciding whether to apply systemic toxin elimination technique • Factors determining the rate of distribution of chemicals in the body are - Protein binding – chemicals highly bound to protein have small volume of distribution
  • 5. • Plasma concentration – when the volume of distribution of chemicals is small, most of the chemical remains in the plasma • Physiological barriers – chemicals will not uniformly distributed to the body due to specialized barriers e.g blood brain barrier iii) Biotransformation (metabolism) • Biotransformation is the biochemical transformation of a chemical. It is a process by which the body transforms a chemical and makes it more water soluble so the chemical can be eliminated more rapidly via the kidney into the urine. Biotransformation can produce metabolites that are pharmacologically active and toxic
  • 6.
  • 7. E.g. parathion→ parathoxon (toxic metabolite). Liver is the major site of biotransformation for many chemicals & other organs that are involved are lungs, kidneys, skin &so on. Interactions during biotransformation include • There are two phases of biotransformation • Phase I – the drug is converted into more polar compound e.g oxidation, reduction, &hydrolysis • Phase II (conjugation) – a drug or its metabolite is conjugated with an endogenous substance e.g glucuronide conjugate • Enzyme inhibition- by this the biotransformation of drugs is delayed & is a cause of increased toxicity
  • 8. • Enzyme induction- by this the biotransformation of drugs is accelerated & is a cause of therapeutic failure • First – pass effect – is the biotransformation of some chemicals by the liver during the initial pass from the portal circulation after oral administration. • Half life (t ½) –is the time required to reduce the blood concentration of the chemical to half.
  • 9. IV) Excretion Excretion is the final means of chemical elimination, either as metabolites or unchanged parent chemical. Excretion through the lungs is the major route for gaseous substances; and in the case of non-volatile water – soluble drugs, the kidneys are the most important routes of excretion. Additional routes include sweat, saliva, tears, nasal secretions, milk, bile and feces • Clearance – elimination of chemicals from the body may be described by the term clearance (CL). It is a quantitative measure of the volume of blood cleared of drug per unit time, usually expressed in milliliter per 4 minute
  • 10. GENERAL PRINCIPLES TO BE CONSIDERED 1. Quantification of exposure . 2. Setting of dose levels •Low dose levels •Intermediate dose levels •High dose levels 3.Extent of exposure assessment in toxicity studies 4.Complicating factors in exposure interpretation 5.Route of administration. 6.Determination of metabolites 7.Statistical evaluation of data. 8.Analytical methods 8.Reporting.
  • 11. Introduction •Toxicokinetics (TK) is defined by The International Conference on Harmonization (ICH) as ‘the generation of pharmacokinetic data, either as an integral component in the conduct of non-clinical toxicity studies. • The need for toxicokinetic data and the extent of exposure assessment in individual toxicity studies should be based on a flexible step-by-step approach. • The primary objective of the toxicokinetic studies is to describe the systemic exposure achieved in animals and its relationship to dose level and the time course of the toxicity study . • Choice of species and treatment regimen used in non clinical studies. Lastly, information on systemic exposure of animals during repeated-dose toxicity studies is essential for the interpretation of study results.
  • 12. 1. Quantification and extent of exposure : • The exposure might be represented by plasma (serum or blood) concentrations or the AUCs of parent compound and/or metabolite(s) and sometimes by tissue concentrations. • Quantification of exposure provides an assessment of the burden on the test species and helps in the interpretation of similarities and differences in toxicity across species, dose groups and sexes. • When designing the toxicity studies, the exposure and dose-dependence in humans at therapeutic dose levels (either expected or established), should be considered in order to achieve relevant exposure at various dose levels in the animal toxicity studies. • Species differences in the pharmacodynamics of the substance (either qualitative or quantitative) should also be taken into consideration because sometimes it may have other effects. This information may allow better interspecies comparisons than simple dose/body weight (or surface area) comparisons.
  • 13. 2. Extent of exposure •Systemic exposure should be estimated in an appropriate number of animals and dose groups to provide a basis for risk assessment. •Concomitant toxicokinetics may be performed either in all or a representative proportion of the animals used in the main study or in special satellite groups. •Both male and female animals are utilized in the main study it is normal to estimate exposure in animals of both sexes unless some justification can be made for not so doing. •Toxicokinetic data is not mandatory for studies of different duration if the dosing regimen is essentially unchanged.
  • 14. 3. Sampling points •In concomitant toxicokinetic studies the time points for collecting body fluids should be as frequent as is necessary • There are also strict restrictions on blood volume available (no more than 10% of circulating volume can be taken). •Sample size is typically 0.25–0.50 ml day−1 in rodents and up to1ml day−1 in non-rodents In each study, justification of number of time points should be made on the basis that they are adequate to estimate exposure. •The justification should be based on kinetic data gathered from earlier toxicity studies, from pilot or dose range finding studies, extrapolation. •Sampling times vary based on the presence (or lack) of pharmacokinetic data, but are often taken 0.5, 1.0, 2.0, 4.0, 8.0, 12.0 and 24.0 h post-dose, with only the parent drug generally being measured.
  • 15. 4. Dose level setting •Dose level for toxicity studies is largely regulated by the toxicology findings and the pharmacodynamic responses of the test species. •Intermediate dose levels should normally represent an appropriate multiple (or fraction) of the exposure at lower (or higher) dose levels dependent upon the objectives of the toxicity study. •The high dose levels in toxicity studies will normally be determined by toxicological considerations. the exposure achieved at the dose levels used should be assessed. •This toxicokinetic data indicate that absorption of a compound limits exposure to parent compound and/or metabolite(s), the lowest dose level of the substance • non-linear kinetics should not necessarily result in dose limitations in toxicity studies.
  • 16. 5. Ratifying factors on study to be considered •Earlier we discussed the species and sex differences and their effect on toxicokinetics. •There are other factors to be considered in this study is protein binding, tissue uptake, receptor properties and metabolic profile. •Systemic exposure may be decreased by protein binding and tissue uptake. I •Due to the metabolism there will be formation of pharmacological active metabolites, the toxic metabolites and antigenic biotechnology products metabolites.
  • 17. 6. Route of administration •Pharmacokinetics of a substance is greatly affected by the route of administration. •For instance orally administered drugs bioavailability time is more than other routes. •If the drug is intended to administer through oral route then oral toxicity should be checked. •If any drug administering route is already established and new clinical route of administration is going to establish then it will be necessary to ascertain whether changing the clinical route will significantly reduce the safety margin. •In this case focusing on local toxicity is essential.
  • 18. 7. Metabolite determination Many of the cases systemic exposure and toxic effect consider on the basis of parent drug concentration. However, there may be circumstances when measurement of metabolite concentrations in plasma or other body fluids is especially important in the conduct of toxicokinetics. They are •If it is a 'pro-drug' and the delivered metabolite is acknowledged to be the primary active entity. •If the compound is metabolised to one or more pharmacologically or toxicologically active metabolites which could make a significant contribution to tissue/organ responses. •For the drugs which are extensively metabolized and the metabolite is only the quantifiable factor.
  • 19. 8. Statistical evaluation of data •The data should be evaluated statistically which allows assessment of the exposure. •Toxicokinetic values are normally calculated as mean SD; statistical evaluation is not usually performed •However, because large intra- and inter-individual variation of kinetic parameters may occur and small numbers of animals are involved in generating toxicokinetic data, Ahigh level of precision in terms of statistics is not normally needed. •Consideration should be given to the calculation of mean or median values and estimates of variability, •If data transformation (e.g. logarithmic) is performed, a rationale should be provided.
  • 20. 9. Analytical methods •Regulatory authorities expects that analytical methods used to determine plasma concentrations of pharmaceuticals are of adequate sensitivity and precision. •For evaluation validated analytical methods used and conforms to Good Laboratory Practice (GLP). Analytical methods used in such studies include gas chromatography (although this is rarely used), HPLC (UV or fluorescence), LC, LC–MS, LC-MS-MS, and capillary electrophoresis (again, rarely used, and more for proteins). •Generally, toxicity studies use a range of time-points and replicates to provide toxicokinetic data as we discussed earlier, although staggered and sparse sampling. •For replicate designs, toxicokinetic measurements are taken at similar pre-set timepoints and the mean of the measured values is then taken to provide an estimate of drug exposure. •Although staggered designs are less sensitive, they are still used by various pharmaceutical companies for rodent and/or primate studies.
  • 21. 1. Toxicokinetic studies in Preclinicalstage: a. Safety assessment b. Single dose and rising dose studies Repeated-dose toxicity studies c. Repeated-dose toxicity studies d. Genotoxicity studies e. Reproduction toxicity studies i. Studies of fertility ii. In pregnant and lactating animals e. Carcinogenicity studies: 2. Toxicokinetic studies in clinical phases: a. Approaches to decrease the animal usage in toxicokinetics b. Dried blood spot technology c. Alternative approaches to animal models d. Physiologically based pharmacokinetic (PBPK) modeling
  • 22. 1. Toxicokinetic studies in Preclinical stage: a. Safety assessment •Generally safety of a molecule can be performed in in-vivo systems. •This step is not included in the guidelines but it is very useful for the researchers to assess the systemic exposure of the molecule and its effect on it. •This safety study is integral part in the central nervous system (CNS), cardio vascular system (CVS) and respiratory assessments. b. Single dose and rising dose studies •These studies are often performed in a very early phase of drug development before a bioanalytical method has been developed. •These studies are usually performed in rodents. •Plasma samples may be taken in such studies and stored for later analysis.
  • 23. c. Repeated-dose toxicity studies •To give support for phase 1 studies this study is carried out for four weeks in both rodents as well as non-rodents. •The treatment regimen (Note 11) and species should be selected whenever possible with regard to pharmacodynamic and pharmacokinetic principles. • This may not be achievable for the very first studies, at a time when neither animal nor human pharmacokinetic data are normally available. d. Genotoxicity studies •Two in vitro studies and one in vivo study is essential to support development of drug . •In vivo investigations usually use a rodent micronucleus (bone marrow or peripheral erythrocytes) test or chromosome aberration (bone marrow cells) test. •These are the well established studies for the genotoxicity evaluation.
  • 24. e. Reproduction toxicity studies Reproduction toxicity measurements are taken in studies of fertility (rat), embryo-foetal development (rat and rabbit) and peri- or post-natal development (rat). a) Studies of fertility Assessment of fertility toxicity has very important, because most of the drugs used in fertility conditions so has to strengthen at that time. Usually this can be done in rats. b. In pregnant and lactating animals: There is a regulatory expectation for toxicokinetic data in pregnant animals, although no specific guidance is given1,16. Data from non-pregnant animals is useful to set dose levels, and the limitation of exposure is usually governed by maternal toxicity. Toxicokinetics may involve exposure assessment of dams, embryos, foetuses or newborn at specified days. Secretion in milk may be assessed to define its role in the exposure of newborns..
  • 25. f. Carcinogenicity studies: •Sometimes drugs are used for longtime for curing purposes, this may lead to the toxicity or carcinogenicity. •So lifetime studies in the rodent are needed to support the long-term clinical use of pharmaceuticals17 and non-rodents can also be used. •Dose selection is usually determined as the maximum tolerated dose (MTD •Selection based on AUC is less common as a 25-fold ratio is often not feasible. • It is recommended that monitoring should occur on a few occasions during the study, although it is not essential for monitoring to occur beyond six months. • However, pharmaceutical companies use various strategies for such monitoring times
  • 26. 2. Toxicokinetic studies in clinical phases: Regulatory bodies around the world outlining that toxicity studies are necessary to support human Phase I, II and III studies, and product license application is available. •The magnitude of the preclinical toxicokinetic evaluation for each clinical phase varies significantly among pharmaceutical companies. •For Phase I investigations the company might only generate. toxicokinetic data from the four-week repeat-dose toxicitystudies. •Full pharmacokinetic profile (including in vitro metabolism studies), and toxicokinetic measurements from four- and 13-week repeat-dose toxicitystudies prior to Phase I is necessary. •Toxicity assessments enable the No Observed Effect Level (NOEL) or No Observed Adverse Effect Level (NOAEL) to be established for a potential new drug, based on clinical observations, bodyweight, food consumption, clinical pathology, organ weights, necropsy examination, and histopathology.
  • 27. a. Approaches to decrease the animal usage in toxicokinetics: • To increase the generation of toxicokinetic data it has to increase the usage of number of rodents through the use of satellite groups. • As discussed earlier using of animal’s number and sex is restricted as per OECD-417 guidelines even in blood samplings. For this purpose many alternative approaches has generated. b. Dried blood spot technology: • Recently toxicokinetic studies in dogs by applying dried blood spot technology published and also it is in improvement stages to use this model in rodents (bar). • In Pharmaceutical industry this type of methodology for toxicokinetics has been developed and combined with high performance liquid chromatography–mass spectrometry (HPLC– MS/ MS) to collect and analyse very small amounts of biofluids.
  • 28. c. Alternative approaches to animal models • Using alternative models to animal models can reduce the number of animals in confirmative studies • Although this is not basis for the use of the chemical entity in clinical phases, but prior to human usage it should check with the animal studies. d. Physiologically based pharmacokinetic (PBPK) modeling: • PBPK models can scientifically support risk assessment by facilitating extrapolation between species and exposure routes, and from high to low doses. • Loizou et al., 2008 used this approach to understand the relationship between external and internal exposures, helping in dose response characterisation and the interpretation of biomarker levels determined in bio-monitoring studies
  • 29. References: 1. CPMP/ICH/384/95: Operational June 1995. Toxicokinetics: guidance for assessing systemic exposure in toxicology studies. Also known asICH Topic S3A (see http :// www. emea.eu.int) 2. Kate, R., Case, D.E., Hakes, H., Nod, K., Salami, F., Horii,I., Mayahara, H.,Cayeb n, M.N., Marriott, T.B., Igarashi, T., Toxicokinetics: its significance and practical problems.J.Toxicol.Sci.18:211-238;( 1993). 3. CPMP/ICH/286/95, modification: operational November 2000. Nonclinical safety studies for the conduct of human clinical trials for pharmaceuticals. Also known as ICH Topic M3 (seehttp://www.emea.eu.int) 4. CPMP/SWP/1042 Corr: operational October 2000. Note for guidance on repeated dose toxicity (see http://www.emea.eu.int) 5. LaDu, B.N. Fundamentals of Drug Metabolism and Drug Disposition, Robert E Krieger Publishing Company, New York, pp.528 (1979) 6. Igarashi, T. and Sekido, T. Case studies for statistical analysis of toxicokinetic data. Regul. Toxicol. Pharmacol. 23:193–208; (1996).