SlideShare a Scribd company logo
1 of 46
786yam
FIRST IN HUMAN DOSE
INTRODUCTION
• Estimating the first in human (FIH) dose is one of the initial steps in the
clinical development of any molecule that has successfully gone through
all of the hurdles in preclinical evaluations.
• It is an important parameter in the FIH clinical trials, since a high starting
dose may cause serious toxicity in volunteers, while a low starting dose
could prolong the dose escalation/optimisation, leading to unnecessary
delay in the clinical programs.
• In 2005, the US Food and Drug Administration (FDA) issued guidance on
estimating the maximum safe starting dose in initial clinical trials for
therapeutics in adult healthy volunteers, which provided a framework to
carry out the estimation.
• The process of calculating the MRSD should begin after the toxicity data
have been analyzed.
DEFINITIONS
1) MRSD: MAXIMUM RECOMMENDED STARTING DOSE
2) LEVEL: Refers to the dose or dosage, generally expressed as mg/kg or
mg/kg/day.
3) NOAEL: NO OBSERVED ADVERSE EFFECT LEVEL (NOAEL) : The highest dose
level that does not produce a significant increase in adverse effects.
4) NOEL: NO OBSERVED EFFECT LEVEL: Refers to any effect, not just adverse
ones, although in some cases the two might be identical.
5) LOWEST OBSERVED ADVERSE EFFECT LEVEL (LOAEL): The lowest dose that
produces adverse effects.
6) HED: HUMAN EQUIVALENT DOSE: The quantity of a chemical that, when
administered to humans, produces an effect equal to that produced in test
animals by a smaller dose.
7) MABEL: MINIMAL ANTICIPATED BIOLOGICAL EFFECT LEVELS : The lowest
dose that is associated with any biological effect, whether it be toxicity or a
desired pharmacological effect
MAXIMUM RECOMMENDED STARTING DOSE
(MRSD)
Aim of MSRD:
• Avoid toxicity at initial dose
• Dose needs to be high enough to allow reasonably rapid attainment of phase
I trial objectives.
Not applicable to:
• Endogenous hormones and proteins (i.e. recombinant clotting factors) used
at physiological concentrations
• Vaccines
Limitations:
• Does not address dose escalation or maximum allowable doses in clinical
trials
ESTIMATING THE MRSD-METHODS
1) NOAEL Method
2) MABEL Method
3) Similar Drug Comparison Method
4) Pharmacokinetic Guided Approach
5) PK/PD Modelling Guided Approach
Calculations based on:
1) Animal pharmacokinetic data
2) Administered doses
3) Observed toxicities
4) Algorithmic calculation
NOAEL METHOD
• The NOAEL method is based on selecting a dose with minimal risk of toxicity,
rather than selecting one with minimal pharmacologic activity in humans.
• This approach works well with new molecules that act on established targets
and/or have the pharmacology that is more or less understood.
• 5 Steps using animal toxicology data:
1) Determine No Observed Adverse Effect Level (NOAEL) ‰
2) Convert NOAEL to Human Equivalent Dose (HED) ‰
3) Select most appropriate species ‰
4) Apply Safety Factor ‰
5) Consider Pharmacologically Active Dose
STEP 1: NO OBSERVED ADVERSE
EFFECT LEVEL DETERMINATION
 The NOAEL is a generally accepted benchmark for safety when derived
from appropriate animal studies.
 The available animal toxicology data is reviewed and evaluated so that a
NOAEL can be determined for each study.
 While reviewing the animal toxicology data, the adverse effects that are
statistically significant and adverse effects that may be clinically
significant (even if they are not statistically significant) should be
considered in the determination of the NOAEL.
TYPES OF FINDINGS IN NONCLINICAL TOXICOLOGY STUDIES THAT
CAN BE USED TO DETERMINE THE NOAEL
 Overt toxicity (e.g., clinical signs, macro- and microscopic lesions)
 Surrogate markers of toxicity (e.g., serum liver enzyme levels) and
 Exaggerated pharmacodynamic effects.
 As a general rule, an adverse effect observed in nonclinical toxicology
studies used to define a NOAEL for the purpose of dose-setting should be
based on an effect that would be unacceptable if produced by the initial
dose of a therapeutic in a phase 1 clinical trial conducted in adult healthy
volunteers.
PK INFLUENCE ON NOAEL:
 Bioavailability, metabolite profile, and plasma drug levels associated with
toxicity may influence the choice of the NOAEL.
 Ex: Saturation of drug absorption at a dose that produces no toxicity. In
this case, the lowest saturating dose, not the highest (non-toxic) dose,
should be used for calculating the HED.
STEP 2: HUMAN EQUIVALENT DOSE CALCULATION
Conversion Based on Body Surface Area
• After the NOAELs in the relevant animal studies have been determined, they
are converted to human equivalent doses (HEDs) using appropriate scaling
factors.
• The most appropriate method for extrapolating the animal dose to the
equivalent human dose should be decided.
• Toxic endpoints for therapeutics administered systemically to animals, such as
the MTD or NOAEL, are usually assumed to scale well (doses scaled 1:1)
between species when doses are normalized to body surface area (mg/m2).
• The basis for this assumption: Doses lethal to 10% of rodents (LD10) and MTDs
in non-rodents both correlated with the human MTD when the doses were
normalized to the same administration schedule and expressed as mg/m2.
STEP 2: HUMAN EQUIVALENT DOSE CALCULATION
• Correcting for body surface area increases clinical trial safety by resulting in a
more conservative starting dose estimate.
• Hence, it was concluded that the approach of converting NOAEL doses to an
HED based on body surface area correction factors (i.e., W0.67) should be
maintained for selecting starting doses for initial studies in adult healthy
volunteers.
CONVERSION FACTORS:
• These are recommended as the standard values to be used for interspecies
dose conversions for NOAELs.
• Since surface area varies with W0.67, the conversion factors are therefore
dependent on the weight of the animals in the studies.
• These factors may also be applied when comparing safety margins for other
toxicity endpoints (e.g., reproductive toxicity and carcinogenicity) when data
for comparison, (i.e., AUCs) are unavailable or are otherwise inappropriate for
comparison.
STEP 2: HUMAN EQUIVALENT DOSE CALCULATION
 HED = Animal NOAEL x (W animal/W human)(1-b)
 Conversion factors = (W animal/W human)(1-b)
 Conventionally, for a mg/m2 normalization b would be 0.67, but studies have
shown that MTDs scale best across species when b=0.75.
 Conversion factors are calculated over a range of animal and human weights
using (Wanimal/Whuman)0.33 or (Wanimal/Whuman)0.25 to assess the effect on starting
dose selection of using b = 0.75 instead of b = 0.67.
 However, converting doses based on an exponent of 0.75 would lead to
higher, more aggressive and potentially more dangerous starting doses.
• Nonetheless, use of a different dose normalization approach, such as directly
equating the human dose to the NOAEL in mg/kg, may be appropriate in some
circumstances.
STEP 2: HUMAN EQUIVALENT DOSE
CALCULATION
BASIS FOR USING Mg/Kg CONVERSIONS
 The “mg/kg” scaling will give a 12-,6- & 2- fold higher HED than the default
mg/m2 approach for mice, rats, and dogs, respectively.
 Circumstances for using mg/kg for calculating HED
1. NOAELs occur at a similar mg/kg dose across test species (for the studies
with a given dosing regimen relevant to the proposed initial clinical trial).
2. When toxicity in humans (for a particular class) is dependent on an
exposure parameter that is highly correlated across species with dose on a
mg/kg basis.
Ex: Complement activation by systemically administered antisense
oligonucleotides in humans is believed to be dependent upon Cmax. For
some antisense drugs, the Cmax correlates across nonclinical species with
mg/kg dose and in such instances mg/kg scaling would be justified.
Also there is a robust correlation between plasma drug levels (Cmax and
AUC) and dose in mg/kg.
3. Other pharmacologic and toxicologic endpoints also scale between species
by mg/kg for the therapeutic. Examples of such endpoints include the
MTD, lowest lethal dose, and the pharmacologically active dose.
BASIS FOR USING MG/KG CONVERSIONS
4. Therapeutics administered by alternative routes (e.g., topical, intranasal,
subcutaneous, intramuscular) for which the dose is limited by local toxicities.
Such therapeutics should be normalized to concentration (e.g., mg/area of
application) or amount of drug (mg) at the application site.
5. Therapeutics administered into anatomical compartments that have little
subsequent distribution outside of the compartment.
Ex: Intrathecal, Intravesical, Intraocular, or Intrapleural administration.
Such therapeutics should be normalized between species according to the
compartmental volumes and concentrations of the therapeutic.
6. Biological products administered intravascularly with MW > 100,000 daltons.
Such therapeutics should be normalized to mg/kg.
mg/m2 = km × mg/kg
where km = 100/K × W0.33 where K is a value unique to each species
(Freireich et al. 1966)
CONVERSION OF ANIMAL DOSES TO HUMAN
EQUIVALENT DOSES BASED ON BODY SURFACE
AREA
STEP 3: MOST APPROPRIATE SPECIES
SELECTION
 HED should be chosen from the most appropriate species.
 Most sensitive species (i.e., the species in which the lowest HED can be
identified).
 Factors that could influence the choice of the most appropriate species:
 (1) Differences in the absorption, distribution, metabolism, and excretion
(ADME) of the therapeutic between the species.
When determining the MRSD for the first dose of a new therapeutic in
humans, the ADME parameters will not be known. Comparative
metabolism data, however, might be available based on in vitro studies.
These data are particularly relevant when there are marked differences in
both the in vivo metabolite profiles and HEDs in animals.
MOST APPROPRIATE SPECIES
SELECTION
 (2) Class experience that may indicate a particular animal model is more
predictive of human toxicity.
Class experience implies that previous studies have demonstrated that a
particular animal model is more appropriate for the assessment of safety
for a particular class of therapeutics.
Selection of the most appropriate species for certain biological products
involves consideration of various factors unique to these products.
(a) Factors such as whether an animal species expresses relevant receptors
or epitopes may affect species selection.
Ex: In the nonclinical safety assessment of the phosphorothioate antisense
drugs, the monkey is considered the most appropriate species because
monkeys experience the same dose limiting toxicity as humans (e.g.,
complement activation) whereas rodents do not.
MOST APPROPRIATE SPECIES
SELECTION
 For this class of therapeutics, the MRSD would be based on the HED for the
NOAEL in monkeys regardless of whether it was lower than that in rodents,
unless unique dose limiting toxicities were observed with the new antisense
compound in the rodent species.
(b) Similarities of biochemistry and physiology between the species and
humans that are relevant to the limiting toxicities of the therapeutic should
also be considered under class experience.
 If a species is the most sensitive but has differences in physiology compared
to humans, it may not be the most appropriate species for selecting the
MRSD.
 (3) Limited biological cross-species pharmacologic reactivity of the
therapeutic. This is especially important for biological therapeutics as many
are human proteins that bind to human or non-human primate targets.
STEP 4: APPLICATION OF SAFETY FACTOR
 A safety factor is applied in order to provide a margin of safety for
protection of human subjects receiving the initial clinical dose.
 This safety factor allows for variability in extrapolating from animal toxicity
studies to studies in humans resulting from:
(1) Uncertainties due to enhanced sensitivity to pharmacologic activity in
humans versus animals
(2) Difficulties in detecting certain toxicities in animals (e.g., headache,
myalgias, mental disturbances)
(3) Differences in receptor densities or affinities
(4) Unexpected toxicities and
(5) Interspecies differences in ADME of the therapeutic.
 These differences can be accommodated by lowering the human starting
dose from the HED of the selected species NOAEL.
APPLYING SAFETY FACTOR
 In practice, the MRSD for the clinical trial is determined by dividing the
HED derived from the animal NOAEL by the default safety factor of “10”.
 While a safety factor of 10 can generally be considered adequate for
protection of human subjects participating in initial clinical trials, this
safety factor may not be appropriate for all cases.
 The safety factor should be raised when there is reason for increased
concern, and lowered when concern is reduced because of available data
that provide added assurance of safety.
INCREASING THE SAFETY FACTOR
 Steep dose response curve.
 Severe toxicities.
 Nonmonitorable toxicity
 Toxicities without premonitory signs.
 Variable bioavailability.
 Irreversible toxicity
 Unexplained mortality
Large variability in doses or AUC
levels eliciting effect.
Nonlinear pharmacokinetics.
Questionable study design or
conduct
Inadequate dose-response data.
Novel therapeutic targets.
Animal models with limited utility.
DECREASING THE SAFETY FACTOR
 When the toxicologic testing in these cases is of the highest caliber in both
conduct and design.
 When the candidate therapeutics are members of a well-characterized
class.
 When within the class, the therapeutics are administered by the same
route, schedule, and duration of administration; have a similar metabolic
profile and bioavailability; and have similar toxicity profiles across all the
species tested including humans.
 When toxicities produced by the therapeutic are easily monitored,
reversible, predictable, and exhibit a moderate-to-shallow dose-response
relationship with toxicities that are consistent across the tested species
(both qualitatively and with respect to appropriately scaled dose and
exposure).
 When the NOAEL was determined based on toxicity studies of longer
duration compared to the proposed clinical schedule in healthy volunteers.
STEP 5: CONSIDERATION OF THE
PHARMACOLOGICALLY ACTIVE DOSE(PAD)
 Selection of a PAD depends upon many factors and differs markedly among
pharmacological drug classes and clinical indications.
 Once the MRSD has been determined, it may be of value to compare it to
the PAD derived from appropriate pharmacodynamic models.
 If the PAD is from an in vivo study, an HED can be derived from a PAD
estimate by using a body surface area conversion factor (BSA-CF). This
HED value should be compared directly to the MRSD.
 If this pharmacologic HED is lower than the MRSD, it may be appropriate
to decrease the clinical starting dose for scientific reasons.
 Additionally, for certain classes of drugs or biologics, toxicity may arise
from exaggerated pharmacologic effects. The PAD in these cases may be a
more sensitive indicator of potential toxicity than the NOAEL and might
therefore warrant lowering the MRSD.
STARTING DOSE CALCULATION
IT IS ALWAYS ACCEPTABLE (FROM
A SAFETY PERSPECTIVE) TO USE A
CLINICAL START DOSE LOWER
THAN THE MRSD.
IS NOAEL APPROACH THE FOOLPROOF
METHOD FOR CALCULATING FIH DOSE?
• NOAEL approach= safety window based on
toxicological threshold
• TRAGIC TRIAL OF TGN1412
• TGN1412 : A novel CD28 super-agonist antibody immunomodulator
• Developed by TeGenero Immuno Therapeutics,London, tested by
Parexel and manufactured by Boehringer-Ingelheim.
• Intended for the treatment of B cell chronic lymphocytic leukemia (B-CLL)
and rheumatoid arthritis.
• Caused cytokine storm.
• Dose in humans was 1/160 of the NOAEL in monkeys, so it was well within
accepted safety margins.
COULD THIS TOXICITY HAVE BEEN
PREDICTED?
• No evidence of contamination of the product.
• Conduct of the trial appeared to have followed the protocol, e.g., no
dosing errors.
• Nothing in the preclinical data that predicted the overwhelming systemic
reaction to the antibody.
 Initial dose 0.1 mg/kg would achieve plasma level of ~16nM compared
with binding affinity of 2nM. 8X the binding affinity of “super agonist”
might result in 90% CD28 receptor binding on first dose.
 This occupancy is considered sufficient for an antagonist but too high for
FIH with agonist. If starting dose was established using pharmacological
principles, the dose might have been started 100X lower.
DUFF REPORT RECOMMENDATIONS
SELECTION OF THE FIH DOSE FOR CLINICAL TRIAL
 The EMEA proposed for micro dosing a safety factor of 1/100th of the dose
calculated to yield a pharmacologic effect, on a mg/m2 basis, and suggested
that such a dose should be 1/1000th of the minimal toxic effect dose (MTED)
from a single-dose toxicology study.
 Special consideration should be given to new agents for which the primary
pharmacological action, for the proposed therapeutic effect, cannot be
demonstrated in an animal model.
 A broader approach to dose calculation, beyond reliance on NOEL or NOAEL in
animal studies, should be taken.
DUFF REPORT FOR FIH DOSE SELECTION
 The calculation of starting dose should utilize all relevant information. Factors
to be taken into account include
 The novelty of the agent
 Biological potency and its mechanism of action
 Degree of species-specificity of the agent
 Dose-response curves of biological effects in human and animal cells
 Dose-response data from in vivo animal studies
 Pharmacokinetic and pharmacodynamic modeling
 Calculation of target occupancy versus concentration
 Calculated exposure of targets or target cells in humans in vivo.
 If different methods give different estimates of a safe dose in humans, the
lowest value should be taken as the starting point in first-in-man trials and a
margin of safety introduced in calculation of the actual starting doses in man.
THE MABEL APPROACH FOR DOSE SELECTION
• This is an improved approach to dose selection through the combined
analysis of all of the pharmacology, safety and efficacy preclinical data.
• The main objective of the new process is to define a starting dose that is
expected to result in a minimum anticipated biological effect level
(MABEL), particularly for investigational medicinal products where risk
factors were identified in
(1) Mode of action
(2) Nature of the target and
(3) Relevance of animal species and models.
THE MABEL APPROACH FOR DOSE SELECTION
(1) The mode of action :
• This refers to the knowledge on the nature and intensity of the effect of the
medicinal product on the specific target and non-targets and subsequent
mechanisms.
• Certain modes of action have been identified to require special attention:
 Targets that have pleiotropic effects or are ubiquitously expressed (e.g., as
often occur in the immune system)
 Targets that have a biological cascade or cytokine release, including those
leading to an amplification of an effect that might not be sufficiently
controlled by a physiological feedback mechanisms (e.g., Monoclonal
antibodies against the T cell targets).
(2) The nature of the target
• This may include information on the structure, tissue distribution, cell
specificity, disease specificity, regulation, polymorphisms, level of expression,
and biological function of the human target, including downstream effects,
and how it might vary between individuals in different populations of healthy
subjects and patients.
(3) The relevance of animal models.
• The available animal species should be compared to humans taking into
account the target, its structural homology, distribution, signal transduction
pathways and the nature of pharmacological effects.
• Where the available animal species/models or surrogates are perceived to be
of questionable relevance for thorough investigation of the pharmacological
and toxicological effects of the medicinal product, this should be considered as
adding to the risk.
• The risk assessment should be performed on a ‘‘case-by-case’’ basis and that
a ‘‘weight-of-evidence’’ approach should be used.
• The calculation of MABEL should use all in vitro and in vivo information
available from PK/PD, which may include the following:
(1) Target binding and receptor occupancy studies in vitro in target cells from
human and the relevant animal species
(2) Concentration-response curves in vitro in target cells from human and the
relevant animal species and dose/exposure-response in vivo in the relevant
animal species
(3) Exposures at pharmacological doses in the relevant animal species
• To further limit the potential for adverse reactions in humans, a safety
factor may be applied in the calculation of the first dose in humans from
MABEL.
• In cases where the methods of calculation (e.g. NOAEL, MABEL) give
different estimations of the first dose in man, the lowest value should be
used.
SIMILAR DRUG COMPARISON APPROACH
• This may be used when human PK/PD data are available for a drug similar
to the one under investigation.
• The dose of the investigated drug can be calculated from the dose of the
reference drug:
Dosei = Doser NOAELi / NOAELr
• The dose obtained is usually corrected by an arbitrary safety factor to
accommodate uncertainty.
PHARMACOKINETIC-GUIDED APPROACH
• In this approach, the NOAEL and corresponding AUC in several animal
species are determined.
• The species that results the lowest NOAEL is used as the index species for
scaling.
• The starting oral dose can be calculated as follows:
Dose = CL AUC / F
Dose = Clhuman Css Dosing Interval /F
Dose = {AUCanimal CLhuman} / Correction factor
A Correction factor
is obtained by
dividing the
clearance of the
chosen species
by the predicted
human clearance.
• A few assumptions are made with pharmacokinetic-guided approaches:
(1) Only the parent compound is active, and
(2) The drug shows equal pharmacological activity or toxicity in human and
nonhuman animal species at equal plasma concentrations.
• This results in the inability to account for interspecies differences in
pharmacodynamics, which are extremely important to identify prior to
phase I trials.
PK/PD MODEL-GUIDED APPROACH
• To avoid inaccuracy caused by interspecies differences in exposure–
response relationships, PK/PD modeling has been utilized to estimate the
FIH dose.
• The human PK/PD profile could be projected using animal data from a
single species.
Estimation of human doses using a four-step approach (Lowe et al):
1) First step: Set up the concentration–effect relationships to identify
biomarkers, determined PD parameters such as EC50, and develop the
PD models.
2) Second step: Identify the interspecies differences in the concentration–
response profiles, for both desired and adverse effects. This includes
differences in tissue distribution, tissue and plasma protein binding,
blood cell binding, and target receptor occupancy.
3) Third step: Human PK parameters, such as CL, bioavailability, and plasma
concentration–time profiles are predicted.
4) Fourth step: Integrate human PK and PD models to predict human dose–
response relationships, which involves further two approaches.
A) Threshold model: A dosing regimen is designed to keep drug
concentrations above the threshold of efficacy (e.g.,EC50) but below
the threshold of adverse effects.
B) PK/PD model: To simulate the dose–exposure–response–time profiles,
which could incorporate the complex PD models.
 The PK/PD model- based approach is successful for monoclonal antibodies,
whose PK properties are usually uniform based on their isotype.
• The receptor occupancy (RO), which can be measured by in vitro
experiments, may be used as the biomarker for the human PK/PD model.
• When estimating the starting dose the RO value is recommended to be low
for an agonist and high for an antagonist.
METHOD APPLICABILITY ADVANTAGES DISADVANTAGES
NOAEL To drugs administered
orally, intranasally, SC, or
IM and the dose is
limited by local toxicities.
Proteins administered
intravascularly with MW
>100 kDa
Good safety record for
small molecule drugs;
easy to use
Arbitrary safety factor
makes the approach
very conservative.
Neglect the
interspecies
differences in
pharmacology, such as
binding affinity and
potency
MABEL To both biotherapeutics
and small molecule
chemical entities
Based on pharmacologic
knowledge rather than
an empirical factor
Requires extensive
mechanistic data
SIMILAR DRUG
COMPARISON
Human PK data are
available for a drug
similar to the one under
investigation
Easy to use; very limited
data are required
Only applicable to
very limited drug
candidates
METHOD APPLICABILITY ADVANTAGES DISADVANTAGES
PK-GUIDED
APPROACH
Human PK parameters
predicted from
preclinical data with a
reasonable accuracy.
Based on pharmacokinetic
properties rather than
an empirical factor
Neglect interspecies
differences in
pharmacodynamics;
depends on the
prediction
accuracy of human PK
parameters
PK/PD
MODELING-
GUIDED
APPROACH
Human dose–exposure–
response relationships are
simulated by a mathematic
model
Interspecies differences in
both PK and PD are
considered; reduce the
reliance on empirical
safety factors
A lot of efforts are
required
to establish and
validate
PK/PD models
 Adequate preclinical in vivo toxicological testing may not be possible due
to lack of cross-reactivity in commonly accepted toxicological test species
such as rats and dogs.
 Even for cross-reactive MABs, due to differences in the pharmacology
between test species and humans, the NOAEL obtained in test species may
not be relevant to human testing in some cases.
 Furthermore, toxicity for many biologics is typically due to exaggerated
pharmacology
 Predicting human pharmacological response from preclinical data also
presents unique challenges in the case of biologics compared with small
molecules.
 Therefore, characterizing the preclinical pharmacological response is
critical to understanding potential clinical safety implications for these
compounds.
POINTS TO CONSIDER IN THE SELECTION
OF FIH DOSES FOR BIOLOGICS
INTERACTION OF MABs WITH THEIR TARGET IS, DIFFERS FROM
THAT OF SMALL MOLECULES:
 Because of their high affinity, MABs are typically dosed at equal molar
ratios to their targets.
 The on- and off-rates of MABs at their receptors are, slower than those of
small molecules.
 Binding of target by MAB may change the natural kinetics of the receptor,
e.g. trigger internalization or stabilization of the receptor.
 Due to the relatively slow distribution to the site of action and target
mediated elimination of MABs, unbound MAB concentrations at the
biophase after single doses and at steady state may be one to three orders
of magnitude below unbound MAB concentrations in plasma.
 Because of these characteristics, simple equilibrium calculations of in vivo
RO based on affinity that are applied for many small molecules may not be
applicable for MAB therapeutics.
 Toxic effects in humans are likely to be sustained for long durations due to
the long half-lives
 Selection of too low a starting dose could substantially increase the
duration of clinical studies, increasing the cost and duration of
development with limited additional safety benefit.
 Rational selection of starting doses is of added importance for MAB
programmes
PROPOSED APPROACH BY REGULATORS
(a) Obtain the predicted human dose–
response for all measured biological
effects. in vivo biomarker studies,
receptor binding affinity and occupancy
experiments, antibody-dependant cell
cytotoxicity (ADCC) assay, PK–PD
experiments
(b) Select the dose that would result in minimal biological activity – MABEL dose.
b-agonists produce maximal bronchodilation at <5% RO, and ERAs produce maximum
effect at 20–30% RO, antagonists require generally higher RO to exert their effect: anti-IgE
and anti-CCR5 therapeutics are clinically effective at >90% and >99% RO, respectively.
Therefore, based on target-mediated effects, a starting dose targeting 50% RO could be
considered a MABEL dose for a CCR5 antagonist, whereas <3% RO is more appropriate for an
ERA.
Hence, the choice of starting dose should take into consideration all available information
on the relationship between RO and biological effect for the relevant class of compounds.
(c) Obtain the NOAEL from toxicological testing

More Related Content

What's hot

alternative methods of animal toxicity.pptx
alternative methods of animal toxicity.pptxalternative methods of animal toxicity.pptx
alternative methods of animal toxicity.pptx
ashharnomani
 

What's hot (20)

Oecd guidelines for toxicology studies
Oecd guidelines for toxicology studiesOecd guidelines for toxicology studies
Oecd guidelines for toxicology studies
 
Extrapolation of in vitro data to preclinical and.pptx
Extrapolation of in vitro data to preclinical and.pptxExtrapolation of in vitro data to preclinical and.pptx
Extrapolation of in vitro data to preclinical and.pptx
 
Safety monitoring in clinical trails
Safety monitoring in clinical trailsSafety monitoring in clinical trails
Safety monitoring in clinical trails
 
clinical trials types and design
clinical trials types and designclinical trials types and design
clinical trials types and design
 
Alternative methods to animal toxicity testing
Alternative methods to animal toxicity testingAlternative methods to animal toxicity testing
Alternative methods to animal toxicity testing
 
Pharmacology of free radicals
Pharmacology of free radicalsPharmacology of free radicals
Pharmacology of free radicals
 
Pharmacoepidemiology
PharmacoepidemiologyPharmacoepidemiology
Pharmacoepidemiology
 
Clinical trial study team
Clinical trial study teamClinical trial study team
Clinical trial study team
 
Drug Discovery Process by Kashikant Yadav
Drug Discovery Process by Kashikant YadavDrug Discovery Process by Kashikant Yadav
Drug Discovery Process by Kashikant Yadav
 
alternative methods of animal toxicity.pptx
alternative methods of animal toxicity.pptxalternative methods of animal toxicity.pptx
alternative methods of animal toxicity.pptx
 
Clinical trials its types and designs
Clinical trials  its types and designsClinical trials  its types and designs
Clinical trials its types and designs
 
Safety pharmacology
Safety pharmacologySafety pharmacology
Safety pharmacology
 
OECD Guideline 420: Acute oral Toxicity - Fixed Dose Procedure
OECD Guideline 420: Acute oral Toxicity - Fixed Dose ProcedureOECD Guideline 420: Acute oral Toxicity - Fixed Dose Procedure
OECD Guideline 420: Acute oral Toxicity - Fixed Dose Procedure
 
Target discovery and validation
Target discovery and validation Target discovery and validation
Target discovery and validation
 
toxicokinetics and saturation kinetics
toxicokinetics and saturation kineticstoxicokinetics and saturation kinetics
toxicokinetics and saturation kinetics
 
Target Validation
Target ValidationTarget Validation
Target Validation
 
economics of drug discovery.pptx
economics of drug discovery.pptxeconomics of drug discovery.pptx
economics of drug discovery.pptx
 
Pharmacoeconomics & pharmacoepidemioology
Pharmacoeconomics &  pharmacoepidemioologyPharmacoeconomics &  pharmacoepidemioology
Pharmacoeconomics & pharmacoepidemioology
 
OECD Test Guideline 420: Acute Oral Toxicity - Fixed Dose
OECD Test Guideline 420: Acute Oral Toxicity - Fixed DoseOECD Test Guideline 420: Acute Oral Toxicity - Fixed Dose
OECD Test Guideline 420: Acute Oral Toxicity - Fixed Dose
 
Roles and responsibilities in clinical trials
Roles and responsibilities in clinical trialsRoles and responsibilities in clinical trials
Roles and responsibilities in clinical trials
 

Similar to First in Human dose

EXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICALEXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
TMU
 

Similar to First in Human dose (20)

MMM PPT.pptx
MMM PPT.pptxMMM PPT.pptx
MMM PPT.pptx
 
Extrapolation.pptx
Extrapolation.pptxExtrapolation.pptx
Extrapolation.pptx
 
First dose size in humans and non linear pharmacokinetics.pptx
First dose size in humans and non linear pharmacokinetics.pptxFirst dose size in humans and non linear pharmacokinetics.pptx
First dose size in humans and non linear pharmacokinetics.pptx
 
Dose determination in preclinical and clinical studies
Dose determination in preclinical and clinical studiesDose determination in preclinical and clinical studies
Dose determination in preclinical and clinical studies
 
phase 1 clinical trial
phase 1 clinical trialphase 1 clinical trial
phase 1 clinical trial
 
EPA Guidelines for Carcinogen Risk Assessment.pptx
EPA Guidelines for Carcinogen Risk Assessment.pptxEPA Guidelines for Carcinogen Risk Assessment.pptx
EPA Guidelines for Carcinogen Risk Assessment.pptx
 
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICALEXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
EXTRAPOLATION OF IN VITRO DATA TO PRECLINICAL
 
UPDATED-Early Phase Drug Developmetn and Population PK and Its' Value
UPDATED-Early Phase Drug Developmetn and  Population PK and Its' ValueUPDATED-Early Phase Drug Developmetn and  Population PK and Its' Value
UPDATED-Early Phase Drug Developmetn and Population PK and Its' Value
 
Acute and chronic toxicity studies in animals
Acute and chronic toxicity studies in animalsAcute and chronic toxicity studies in animals
Acute and chronic toxicity studies in animals
 
1 Phases of clinical trial.pptx
1 Phases of clinical trial.pptx1 Phases of clinical trial.pptx
1 Phases of clinical trial.pptx
 
Microdosing (Phase 0) studies
Microdosing (Phase 0) studiesMicrodosing (Phase 0) studies
Microdosing (Phase 0) studies
 
Microdosing
MicrodosingMicrodosing
Microdosing
 
Dose selection
Dose selectionDose selection
Dose selection
 
Pharmacokinetics & BA - Part-2.pdf
Pharmacokinetics & BA - Part-2.pdfPharmacokinetics & BA - Part-2.pdf
Pharmacokinetics & BA - Part-2.pdf
 
Pharmacokinetics & BA - Part-2.pdf
Pharmacokinetics & BA - Part-2.pdfPharmacokinetics & BA - Part-2.pdf
Pharmacokinetics & BA - Part-2.pdf
 
Phase 0 clinical trials
Phase 0 clinical trialsPhase 0 clinical trials
Phase 0 clinical trials
 
Ppt tdm new
Ppt tdm newPpt tdm new
Ppt tdm new
 
Toxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptxToxicokinetic evaluation in preclinical studies.pptx
Toxicokinetic evaluation in preclinical studies.pptx
 
Population pharmacokinetics
Population pharmacokineticsPopulation pharmacokinetics
Population pharmacokinetics
 
Introduction to dosage regimen and Individualization of dosage regimen
Introduction to dosage regimen and Individualization of dosage regimenIntroduction to dosage regimen and Individualization of dosage regimen
Introduction to dosage regimen and Individualization of dosage regimen
 

Recently uploaded

Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls * UPA...
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls  * UPA...Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls  * UPA...
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls * UPA...
mahaiklolahd
 

Recently uploaded (20)

Call Girls Visakhapatnam Just Call 8250077686 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 8250077686 Top Class Call Girl Service Ava...Call Girls Visakhapatnam Just Call 8250077686 Top Class Call Girl Service Ava...
Call Girls Visakhapatnam Just Call 8250077686 Top Class Call Girl Service Ava...
 
Model Call Girls In Chennai WhatsApp Booking 7427069034 call girl service 24 ...
Model Call Girls In Chennai WhatsApp Booking 7427069034 call girl service 24 ...Model Call Girls In Chennai WhatsApp Booking 7427069034 call girl service 24 ...
Model Call Girls In Chennai WhatsApp Booking 7427069034 call girl service 24 ...
 
VIP Service Call Girls Sindhi Colony 📳 7877925207 For 18+ VIP Call Girl At Th...
VIP Service Call Girls Sindhi Colony 📳 7877925207 For 18+ VIP Call Girl At Th...VIP Service Call Girls Sindhi Colony 📳 7877925207 For 18+ VIP Call Girl At Th...
VIP Service Call Girls Sindhi Colony 📳 7877925207 For 18+ VIP Call Girl At Th...
 
Call Girls Tirupati Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Tirupati Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Tirupati Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Tirupati Just Call 8250077686 Top Class Call Girl Service Available
 
Call Girls Raipur Just Call 9630942363 Top Class Call Girl Service Available
Call Girls Raipur Just Call 9630942363 Top Class Call Girl Service AvailableCall Girls Raipur Just Call 9630942363 Top Class Call Girl Service Available
Call Girls Raipur Just Call 9630942363 Top Class Call Girl Service Available
 
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
Manyata Tech Park ( Call Girls ) Bangalore ✔ 6297143586 ✔ Hot Model With Sexy...
 
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service AvailableCall Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Gwalior Just Call 8617370543 Top Class Call Girl Service Available
 
Premium Call Girls In Jaipur {8445551418} ❤️VVIP SEEMA Call Girl in Jaipur Ra...
Premium Call Girls In Jaipur {8445551418} ❤️VVIP SEEMA Call Girl in Jaipur Ra...Premium Call Girls In Jaipur {8445551418} ❤️VVIP SEEMA Call Girl in Jaipur Ra...
Premium Call Girls In Jaipur {8445551418} ❤️VVIP SEEMA Call Girl in Jaipur Ra...
 
Call Girls Vadodara Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Vadodara Just Call 8617370543 Top Class Call Girl Service AvailableCall Girls Vadodara Just Call 8617370543 Top Class Call Girl Service Available
Call Girls Vadodara Just Call 8617370543 Top Class Call Girl Service Available
 
8980367676 Call Girls In Ahmedabad Escort Service Available 24×7 In Ahmedabad
8980367676 Call Girls In Ahmedabad Escort Service Available 24×7 In Ahmedabad8980367676 Call Girls In Ahmedabad Escort Service Available 24×7 In Ahmedabad
8980367676 Call Girls In Ahmedabad Escort Service Available 24×7 In Ahmedabad
 
Call Girls Vasai Virar Just Call 9630942363 Top Class Call Girl Service Avail...
Call Girls Vasai Virar Just Call 9630942363 Top Class Call Girl Service Avail...Call Girls Vasai Virar Just Call 9630942363 Top Class Call Girl Service Avail...
Call Girls Vasai Virar Just Call 9630942363 Top Class Call Girl Service Avail...
 
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
Pondicherry Call Girls Book Now 9630942363 Top Class Pondicherry Escort Servi...
 
Call Girls Hosur Just Call 9630942363 Top Class Call Girl Service Available
Call Girls Hosur Just Call 9630942363 Top Class Call Girl Service AvailableCall Girls Hosur Just Call 9630942363 Top Class Call Girl Service Available
Call Girls Hosur Just Call 9630942363 Top Class Call Girl Service Available
 
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...Top Rated Bangalore Call Girls Mg Road ⟟   9332606886 ⟟ Call Me For Genuine S...
Top Rated Bangalore Call Girls Mg Road ⟟ 9332606886 ⟟ Call Me For Genuine S...
 
💕SONAM KUMAR💕Premium Call Girls Jaipur ↘️9257276172 ↙️One Night Stand With Lo...
💕SONAM KUMAR💕Premium Call Girls Jaipur ↘️9257276172 ↙️One Night Stand With Lo...💕SONAM KUMAR💕Premium Call Girls Jaipur ↘️9257276172 ↙️One Night Stand With Lo...
💕SONAM KUMAR💕Premium Call Girls Jaipur ↘️9257276172 ↙️One Night Stand With Lo...
 
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls * UPA...
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls  * UPA...Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls  * UPA...
Call Girl in Indore 8827247818 {LowPrice} ❤️ (ahana) Indore Call Girls * UPA...
 
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
Mumbai ] (Call Girls) in Mumbai 10k @ I'm VIP Independent Escorts Girls 98333...
 
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
Night 7k to 12k Chennai City Center Call Girls 👉👉 7427069034⭐⭐ 100% Genuine E...
 
Top Rated Bangalore Call Girls Richmond Circle ⟟ 9332606886 ⟟ Call Me For Ge...
Top Rated Bangalore Call Girls Richmond Circle ⟟  9332606886 ⟟ Call Me For Ge...Top Rated Bangalore Call Girls Richmond Circle ⟟  9332606886 ⟟ Call Me For Ge...
Top Rated Bangalore Call Girls Richmond Circle ⟟ 9332606886 ⟟ Call Me For Ge...
 
Call Girls Kurnool Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Kurnool Just Call 8250077686 Top Class Call Girl Service AvailableCall Girls Kurnool Just Call 8250077686 Top Class Call Girl Service Available
Call Girls Kurnool Just Call 8250077686 Top Class Call Girl Service Available
 

First in Human dose

  • 2. INTRODUCTION • Estimating the first in human (FIH) dose is one of the initial steps in the clinical development of any molecule that has successfully gone through all of the hurdles in preclinical evaluations. • It is an important parameter in the FIH clinical trials, since a high starting dose may cause serious toxicity in volunteers, while a low starting dose could prolong the dose escalation/optimisation, leading to unnecessary delay in the clinical programs. • In 2005, the US Food and Drug Administration (FDA) issued guidance on estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers, which provided a framework to carry out the estimation. • The process of calculating the MRSD should begin after the toxicity data have been analyzed.
  • 3. DEFINITIONS 1) MRSD: MAXIMUM RECOMMENDED STARTING DOSE 2) LEVEL: Refers to the dose or dosage, generally expressed as mg/kg or mg/kg/day. 3) NOAEL: NO OBSERVED ADVERSE EFFECT LEVEL (NOAEL) : The highest dose level that does not produce a significant increase in adverse effects. 4) NOEL: NO OBSERVED EFFECT LEVEL: Refers to any effect, not just adverse ones, although in some cases the two might be identical. 5) LOWEST OBSERVED ADVERSE EFFECT LEVEL (LOAEL): The lowest dose that produces adverse effects. 6) HED: HUMAN EQUIVALENT DOSE: The quantity of a chemical that, when administered to humans, produces an effect equal to that produced in test animals by a smaller dose. 7) MABEL: MINIMAL ANTICIPATED BIOLOGICAL EFFECT LEVELS : The lowest dose that is associated with any biological effect, whether it be toxicity or a desired pharmacological effect
  • 4.
  • 5.
  • 6. MAXIMUM RECOMMENDED STARTING DOSE (MRSD) Aim of MSRD: • Avoid toxicity at initial dose • Dose needs to be high enough to allow reasonably rapid attainment of phase I trial objectives. Not applicable to: • Endogenous hormones and proteins (i.e. recombinant clotting factors) used at physiological concentrations • Vaccines Limitations: • Does not address dose escalation or maximum allowable doses in clinical trials
  • 7. ESTIMATING THE MRSD-METHODS 1) NOAEL Method 2) MABEL Method 3) Similar Drug Comparison Method 4) Pharmacokinetic Guided Approach 5) PK/PD Modelling Guided Approach Calculations based on: 1) Animal pharmacokinetic data 2) Administered doses 3) Observed toxicities 4) Algorithmic calculation
  • 8. NOAEL METHOD • The NOAEL method is based on selecting a dose with minimal risk of toxicity, rather than selecting one with minimal pharmacologic activity in humans. • This approach works well with new molecules that act on established targets and/or have the pharmacology that is more or less understood. • 5 Steps using animal toxicology data: 1) Determine No Observed Adverse Effect Level (NOAEL) ‰ 2) Convert NOAEL to Human Equivalent Dose (HED) ‰ 3) Select most appropriate species ‰ 4) Apply Safety Factor ‰ 5) Consider Pharmacologically Active Dose
  • 9. STEP 1: NO OBSERVED ADVERSE EFFECT LEVEL DETERMINATION  The NOAEL is a generally accepted benchmark for safety when derived from appropriate animal studies.  The available animal toxicology data is reviewed and evaluated so that a NOAEL can be determined for each study.  While reviewing the animal toxicology data, the adverse effects that are statistically significant and adverse effects that may be clinically significant (even if they are not statistically significant) should be considered in the determination of the NOAEL.
  • 10. TYPES OF FINDINGS IN NONCLINICAL TOXICOLOGY STUDIES THAT CAN BE USED TO DETERMINE THE NOAEL  Overt toxicity (e.g., clinical signs, macro- and microscopic lesions)  Surrogate markers of toxicity (e.g., serum liver enzyme levels) and  Exaggerated pharmacodynamic effects.  As a general rule, an adverse effect observed in nonclinical toxicology studies used to define a NOAEL for the purpose of dose-setting should be based on an effect that would be unacceptable if produced by the initial dose of a therapeutic in a phase 1 clinical trial conducted in adult healthy volunteers. PK INFLUENCE ON NOAEL:  Bioavailability, metabolite profile, and plasma drug levels associated with toxicity may influence the choice of the NOAEL.  Ex: Saturation of drug absorption at a dose that produces no toxicity. In this case, the lowest saturating dose, not the highest (non-toxic) dose, should be used for calculating the HED.
  • 11. STEP 2: HUMAN EQUIVALENT DOSE CALCULATION Conversion Based on Body Surface Area • After the NOAELs in the relevant animal studies have been determined, they are converted to human equivalent doses (HEDs) using appropriate scaling factors. • The most appropriate method for extrapolating the animal dose to the equivalent human dose should be decided. • Toxic endpoints for therapeutics administered systemically to animals, such as the MTD or NOAEL, are usually assumed to scale well (doses scaled 1:1) between species when doses are normalized to body surface area (mg/m2). • The basis for this assumption: Doses lethal to 10% of rodents (LD10) and MTDs in non-rodents both correlated with the human MTD when the doses were normalized to the same administration schedule and expressed as mg/m2.
  • 12. STEP 2: HUMAN EQUIVALENT DOSE CALCULATION • Correcting for body surface area increases clinical trial safety by resulting in a more conservative starting dose estimate. • Hence, it was concluded that the approach of converting NOAEL doses to an HED based on body surface area correction factors (i.e., W0.67) should be maintained for selecting starting doses for initial studies in adult healthy volunteers. CONVERSION FACTORS: • These are recommended as the standard values to be used for interspecies dose conversions for NOAELs. • Since surface area varies with W0.67, the conversion factors are therefore dependent on the weight of the animals in the studies. • These factors may also be applied when comparing safety margins for other toxicity endpoints (e.g., reproductive toxicity and carcinogenicity) when data for comparison, (i.e., AUCs) are unavailable or are otherwise inappropriate for comparison.
  • 13. STEP 2: HUMAN EQUIVALENT DOSE CALCULATION  HED = Animal NOAEL x (W animal/W human)(1-b)  Conversion factors = (W animal/W human)(1-b)  Conventionally, for a mg/m2 normalization b would be 0.67, but studies have shown that MTDs scale best across species when b=0.75.  Conversion factors are calculated over a range of animal and human weights using (Wanimal/Whuman)0.33 or (Wanimal/Whuman)0.25 to assess the effect on starting dose selection of using b = 0.75 instead of b = 0.67.  However, converting doses based on an exponent of 0.75 would lead to higher, more aggressive and potentially more dangerous starting doses. • Nonetheless, use of a different dose normalization approach, such as directly equating the human dose to the NOAEL in mg/kg, may be appropriate in some circumstances.
  • 14. STEP 2: HUMAN EQUIVALENT DOSE CALCULATION
  • 15. BASIS FOR USING Mg/Kg CONVERSIONS  The “mg/kg” scaling will give a 12-,6- & 2- fold higher HED than the default mg/m2 approach for mice, rats, and dogs, respectively.  Circumstances for using mg/kg for calculating HED 1. NOAELs occur at a similar mg/kg dose across test species (for the studies with a given dosing regimen relevant to the proposed initial clinical trial). 2. When toxicity in humans (for a particular class) is dependent on an exposure parameter that is highly correlated across species with dose on a mg/kg basis. Ex: Complement activation by systemically administered antisense oligonucleotides in humans is believed to be dependent upon Cmax. For some antisense drugs, the Cmax correlates across nonclinical species with mg/kg dose and in such instances mg/kg scaling would be justified. Also there is a robust correlation between plasma drug levels (Cmax and AUC) and dose in mg/kg. 3. Other pharmacologic and toxicologic endpoints also scale between species by mg/kg for the therapeutic. Examples of such endpoints include the MTD, lowest lethal dose, and the pharmacologically active dose.
  • 16. BASIS FOR USING MG/KG CONVERSIONS 4. Therapeutics administered by alternative routes (e.g., topical, intranasal, subcutaneous, intramuscular) for which the dose is limited by local toxicities. Such therapeutics should be normalized to concentration (e.g., mg/area of application) or amount of drug (mg) at the application site. 5. Therapeutics administered into anatomical compartments that have little subsequent distribution outside of the compartment. Ex: Intrathecal, Intravesical, Intraocular, or Intrapleural administration. Such therapeutics should be normalized between species according to the compartmental volumes and concentrations of the therapeutic. 6. Biological products administered intravascularly with MW > 100,000 daltons. Such therapeutics should be normalized to mg/kg. mg/m2 = km × mg/kg where km = 100/K × W0.33 where K is a value unique to each species (Freireich et al. 1966)
  • 17. CONVERSION OF ANIMAL DOSES TO HUMAN EQUIVALENT DOSES BASED ON BODY SURFACE AREA
  • 18. STEP 3: MOST APPROPRIATE SPECIES SELECTION  HED should be chosen from the most appropriate species.  Most sensitive species (i.e., the species in which the lowest HED can be identified).  Factors that could influence the choice of the most appropriate species:  (1) Differences in the absorption, distribution, metabolism, and excretion (ADME) of the therapeutic between the species. When determining the MRSD for the first dose of a new therapeutic in humans, the ADME parameters will not be known. Comparative metabolism data, however, might be available based on in vitro studies. These data are particularly relevant when there are marked differences in both the in vivo metabolite profiles and HEDs in animals.
  • 19. MOST APPROPRIATE SPECIES SELECTION  (2) Class experience that may indicate a particular animal model is more predictive of human toxicity. Class experience implies that previous studies have demonstrated that a particular animal model is more appropriate for the assessment of safety for a particular class of therapeutics. Selection of the most appropriate species for certain biological products involves consideration of various factors unique to these products. (a) Factors such as whether an animal species expresses relevant receptors or epitopes may affect species selection. Ex: In the nonclinical safety assessment of the phosphorothioate antisense drugs, the monkey is considered the most appropriate species because monkeys experience the same dose limiting toxicity as humans (e.g., complement activation) whereas rodents do not.
  • 20. MOST APPROPRIATE SPECIES SELECTION  For this class of therapeutics, the MRSD would be based on the HED for the NOAEL in monkeys regardless of whether it was lower than that in rodents, unless unique dose limiting toxicities were observed with the new antisense compound in the rodent species. (b) Similarities of biochemistry and physiology between the species and humans that are relevant to the limiting toxicities of the therapeutic should also be considered under class experience.  If a species is the most sensitive but has differences in physiology compared to humans, it may not be the most appropriate species for selecting the MRSD.  (3) Limited biological cross-species pharmacologic reactivity of the therapeutic. This is especially important for biological therapeutics as many are human proteins that bind to human or non-human primate targets.
  • 21. STEP 4: APPLICATION OF SAFETY FACTOR  A safety factor is applied in order to provide a margin of safety for protection of human subjects receiving the initial clinical dose.  This safety factor allows for variability in extrapolating from animal toxicity studies to studies in humans resulting from: (1) Uncertainties due to enhanced sensitivity to pharmacologic activity in humans versus animals (2) Difficulties in detecting certain toxicities in animals (e.g., headache, myalgias, mental disturbances) (3) Differences in receptor densities or affinities (4) Unexpected toxicities and (5) Interspecies differences in ADME of the therapeutic.  These differences can be accommodated by lowering the human starting dose from the HED of the selected species NOAEL.
  • 22. APPLYING SAFETY FACTOR  In practice, the MRSD for the clinical trial is determined by dividing the HED derived from the animal NOAEL by the default safety factor of “10”.  While a safety factor of 10 can generally be considered adequate for protection of human subjects participating in initial clinical trials, this safety factor may not be appropriate for all cases.  The safety factor should be raised when there is reason for increased concern, and lowered when concern is reduced because of available data that provide added assurance of safety.
  • 23. INCREASING THE SAFETY FACTOR  Steep dose response curve.  Severe toxicities.  Nonmonitorable toxicity  Toxicities without premonitory signs.  Variable bioavailability.  Irreversible toxicity  Unexplained mortality Large variability in doses or AUC levels eliciting effect. Nonlinear pharmacokinetics. Questionable study design or conduct Inadequate dose-response data. Novel therapeutic targets. Animal models with limited utility.
  • 24. DECREASING THE SAFETY FACTOR  When the toxicologic testing in these cases is of the highest caliber in both conduct and design.  When the candidate therapeutics are members of a well-characterized class.  When within the class, the therapeutics are administered by the same route, schedule, and duration of administration; have a similar metabolic profile and bioavailability; and have similar toxicity profiles across all the species tested including humans.  When toxicities produced by the therapeutic are easily monitored, reversible, predictable, and exhibit a moderate-to-shallow dose-response relationship with toxicities that are consistent across the tested species (both qualitatively and with respect to appropriately scaled dose and exposure).  When the NOAEL was determined based on toxicity studies of longer duration compared to the proposed clinical schedule in healthy volunteers.
  • 25. STEP 5: CONSIDERATION OF THE PHARMACOLOGICALLY ACTIVE DOSE(PAD)  Selection of a PAD depends upon many factors and differs markedly among pharmacological drug classes and clinical indications.  Once the MRSD has been determined, it may be of value to compare it to the PAD derived from appropriate pharmacodynamic models.  If the PAD is from an in vivo study, an HED can be derived from a PAD estimate by using a body surface area conversion factor (BSA-CF). This HED value should be compared directly to the MRSD.  If this pharmacologic HED is lower than the MRSD, it may be appropriate to decrease the clinical starting dose for scientific reasons.  Additionally, for certain classes of drugs or biologics, toxicity may arise from exaggerated pharmacologic effects. The PAD in these cases may be a more sensitive indicator of potential toxicity than the NOAEL and might therefore warrant lowering the MRSD.
  • 26. STARTING DOSE CALCULATION IT IS ALWAYS ACCEPTABLE (FROM A SAFETY PERSPECTIVE) TO USE A CLINICAL START DOSE LOWER THAN THE MRSD.
  • 27. IS NOAEL APPROACH THE FOOLPROOF METHOD FOR CALCULATING FIH DOSE? • NOAEL approach= safety window based on toxicological threshold • TRAGIC TRIAL OF TGN1412 • TGN1412 : A novel CD28 super-agonist antibody immunomodulator • Developed by TeGenero Immuno Therapeutics,London, tested by Parexel and manufactured by Boehringer-Ingelheim. • Intended for the treatment of B cell chronic lymphocytic leukemia (B-CLL) and rheumatoid arthritis. • Caused cytokine storm. • Dose in humans was 1/160 of the NOAEL in monkeys, so it was well within accepted safety margins.
  • 28. COULD THIS TOXICITY HAVE BEEN PREDICTED? • No evidence of contamination of the product. • Conduct of the trial appeared to have followed the protocol, e.g., no dosing errors. • Nothing in the preclinical data that predicted the overwhelming systemic reaction to the antibody.  Initial dose 0.1 mg/kg would achieve plasma level of ~16nM compared with binding affinity of 2nM. 8X the binding affinity of “super agonist” might result in 90% CD28 receptor binding on first dose.  This occupancy is considered sufficient for an antagonist but too high for FIH with agonist. If starting dose was established using pharmacological principles, the dose might have been started 100X lower.
  • 29. DUFF REPORT RECOMMENDATIONS SELECTION OF THE FIH DOSE FOR CLINICAL TRIAL  The EMEA proposed for micro dosing a safety factor of 1/100th of the dose calculated to yield a pharmacologic effect, on a mg/m2 basis, and suggested that such a dose should be 1/1000th of the minimal toxic effect dose (MTED) from a single-dose toxicology study.  Special consideration should be given to new agents for which the primary pharmacological action, for the proposed therapeutic effect, cannot be demonstrated in an animal model.  A broader approach to dose calculation, beyond reliance on NOEL or NOAEL in animal studies, should be taken.
  • 30. DUFF REPORT FOR FIH DOSE SELECTION  The calculation of starting dose should utilize all relevant information. Factors to be taken into account include  The novelty of the agent  Biological potency and its mechanism of action  Degree of species-specificity of the agent  Dose-response curves of biological effects in human and animal cells  Dose-response data from in vivo animal studies  Pharmacokinetic and pharmacodynamic modeling  Calculation of target occupancy versus concentration  Calculated exposure of targets or target cells in humans in vivo.  If different methods give different estimates of a safe dose in humans, the lowest value should be taken as the starting point in first-in-man trials and a margin of safety introduced in calculation of the actual starting doses in man.
  • 31. THE MABEL APPROACH FOR DOSE SELECTION • This is an improved approach to dose selection through the combined analysis of all of the pharmacology, safety and efficacy preclinical data. • The main objective of the new process is to define a starting dose that is expected to result in a minimum anticipated biological effect level (MABEL), particularly for investigational medicinal products where risk factors were identified in (1) Mode of action (2) Nature of the target and (3) Relevance of animal species and models.
  • 32. THE MABEL APPROACH FOR DOSE SELECTION (1) The mode of action : • This refers to the knowledge on the nature and intensity of the effect of the medicinal product on the specific target and non-targets and subsequent mechanisms. • Certain modes of action have been identified to require special attention:  Targets that have pleiotropic effects or are ubiquitously expressed (e.g., as often occur in the immune system)  Targets that have a biological cascade or cytokine release, including those leading to an amplification of an effect that might not be sufficiently controlled by a physiological feedback mechanisms (e.g., Monoclonal antibodies against the T cell targets).
  • 33. (2) The nature of the target • This may include information on the structure, tissue distribution, cell specificity, disease specificity, regulation, polymorphisms, level of expression, and biological function of the human target, including downstream effects, and how it might vary between individuals in different populations of healthy subjects and patients. (3) The relevance of animal models. • The available animal species should be compared to humans taking into account the target, its structural homology, distribution, signal transduction pathways and the nature of pharmacological effects. • Where the available animal species/models or surrogates are perceived to be of questionable relevance for thorough investigation of the pharmacological and toxicological effects of the medicinal product, this should be considered as adding to the risk. • The risk assessment should be performed on a ‘‘case-by-case’’ basis and that a ‘‘weight-of-evidence’’ approach should be used.
  • 34. • The calculation of MABEL should use all in vitro and in vivo information available from PK/PD, which may include the following: (1) Target binding and receptor occupancy studies in vitro in target cells from human and the relevant animal species (2) Concentration-response curves in vitro in target cells from human and the relevant animal species and dose/exposure-response in vivo in the relevant animal species (3) Exposures at pharmacological doses in the relevant animal species • To further limit the potential for adverse reactions in humans, a safety factor may be applied in the calculation of the first dose in humans from MABEL. • In cases where the methods of calculation (e.g. NOAEL, MABEL) give different estimations of the first dose in man, the lowest value should be used.
  • 35. SIMILAR DRUG COMPARISON APPROACH • This may be used when human PK/PD data are available for a drug similar to the one under investigation. • The dose of the investigated drug can be calculated from the dose of the reference drug: Dosei = Doser NOAELi / NOAELr • The dose obtained is usually corrected by an arbitrary safety factor to accommodate uncertainty.
  • 36. PHARMACOKINETIC-GUIDED APPROACH • In this approach, the NOAEL and corresponding AUC in several animal species are determined. • The species that results the lowest NOAEL is used as the index species for scaling. • The starting oral dose can be calculated as follows: Dose = CL AUC / F Dose = Clhuman Css Dosing Interval /F Dose = {AUCanimal CLhuman} / Correction factor A Correction factor is obtained by dividing the clearance of the chosen species by the predicted human clearance.
  • 37. • A few assumptions are made with pharmacokinetic-guided approaches: (1) Only the parent compound is active, and (2) The drug shows equal pharmacological activity or toxicity in human and nonhuman animal species at equal plasma concentrations. • This results in the inability to account for interspecies differences in pharmacodynamics, which are extremely important to identify prior to phase I trials.
  • 38. PK/PD MODEL-GUIDED APPROACH • To avoid inaccuracy caused by interspecies differences in exposure– response relationships, PK/PD modeling has been utilized to estimate the FIH dose. • The human PK/PD profile could be projected using animal data from a single species. Estimation of human doses using a four-step approach (Lowe et al): 1) First step: Set up the concentration–effect relationships to identify biomarkers, determined PD parameters such as EC50, and develop the PD models. 2) Second step: Identify the interspecies differences in the concentration– response profiles, for both desired and adverse effects. This includes differences in tissue distribution, tissue and plasma protein binding, blood cell binding, and target receptor occupancy.
  • 39. 3) Third step: Human PK parameters, such as CL, bioavailability, and plasma concentration–time profiles are predicted. 4) Fourth step: Integrate human PK and PD models to predict human dose– response relationships, which involves further two approaches. A) Threshold model: A dosing regimen is designed to keep drug concentrations above the threshold of efficacy (e.g.,EC50) but below the threshold of adverse effects. B) PK/PD model: To simulate the dose–exposure–response–time profiles, which could incorporate the complex PD models.  The PK/PD model- based approach is successful for monoclonal antibodies, whose PK properties are usually uniform based on their isotype.
  • 40. • The receptor occupancy (RO), which can be measured by in vitro experiments, may be used as the biomarker for the human PK/PD model. • When estimating the starting dose the RO value is recommended to be low for an agonist and high for an antagonist.
  • 41. METHOD APPLICABILITY ADVANTAGES DISADVANTAGES NOAEL To drugs administered orally, intranasally, SC, or IM and the dose is limited by local toxicities. Proteins administered intravascularly with MW >100 kDa Good safety record for small molecule drugs; easy to use Arbitrary safety factor makes the approach very conservative. Neglect the interspecies differences in pharmacology, such as binding affinity and potency MABEL To both biotherapeutics and small molecule chemical entities Based on pharmacologic knowledge rather than an empirical factor Requires extensive mechanistic data SIMILAR DRUG COMPARISON Human PK data are available for a drug similar to the one under investigation Easy to use; very limited data are required Only applicable to very limited drug candidates
  • 42. METHOD APPLICABILITY ADVANTAGES DISADVANTAGES PK-GUIDED APPROACH Human PK parameters predicted from preclinical data with a reasonable accuracy. Based on pharmacokinetic properties rather than an empirical factor Neglect interspecies differences in pharmacodynamics; depends on the prediction accuracy of human PK parameters PK/PD MODELING- GUIDED APPROACH Human dose–exposure– response relationships are simulated by a mathematic model Interspecies differences in both PK and PD are considered; reduce the reliance on empirical safety factors A lot of efforts are required to establish and validate PK/PD models
  • 43.  Adequate preclinical in vivo toxicological testing may not be possible due to lack of cross-reactivity in commonly accepted toxicological test species such as rats and dogs.  Even for cross-reactive MABs, due to differences in the pharmacology between test species and humans, the NOAEL obtained in test species may not be relevant to human testing in some cases.  Furthermore, toxicity for many biologics is typically due to exaggerated pharmacology  Predicting human pharmacological response from preclinical data also presents unique challenges in the case of biologics compared with small molecules.  Therefore, characterizing the preclinical pharmacological response is critical to understanding potential clinical safety implications for these compounds. POINTS TO CONSIDER IN THE SELECTION OF FIH DOSES FOR BIOLOGICS
  • 44. INTERACTION OF MABs WITH THEIR TARGET IS, DIFFERS FROM THAT OF SMALL MOLECULES:  Because of their high affinity, MABs are typically dosed at equal molar ratios to their targets.  The on- and off-rates of MABs at their receptors are, slower than those of small molecules.  Binding of target by MAB may change the natural kinetics of the receptor, e.g. trigger internalization or stabilization of the receptor.  Due to the relatively slow distribution to the site of action and target mediated elimination of MABs, unbound MAB concentrations at the biophase after single doses and at steady state may be one to three orders of magnitude below unbound MAB concentrations in plasma.
  • 45.  Because of these characteristics, simple equilibrium calculations of in vivo RO based on affinity that are applied for many small molecules may not be applicable for MAB therapeutics.  Toxic effects in humans are likely to be sustained for long durations due to the long half-lives  Selection of too low a starting dose could substantially increase the duration of clinical studies, increasing the cost and duration of development with limited additional safety benefit.  Rational selection of starting doses is of added importance for MAB programmes
  • 46. PROPOSED APPROACH BY REGULATORS (a) Obtain the predicted human dose– response for all measured biological effects. in vivo biomarker studies, receptor binding affinity and occupancy experiments, antibody-dependant cell cytotoxicity (ADCC) assay, PK–PD experiments (b) Select the dose that would result in minimal biological activity – MABEL dose. b-agonists produce maximal bronchodilation at <5% RO, and ERAs produce maximum effect at 20–30% RO, antagonists require generally higher RO to exert their effect: anti-IgE and anti-CCR5 therapeutics are clinically effective at >90% and >99% RO, respectively. Therefore, based on target-mediated effects, a starting dose targeting 50% RO could be considered a MABEL dose for a CCR5 antagonist, whereas <3% RO is more appropriate for an ERA. Hence, the choice of starting dose should take into consideration all available information on the relationship between RO and biological effect for the relevant class of compounds. (c) Obtain the NOAEL from toxicological testing