nonclinical safety evaluation of biotechnology-derived … 2015_day 4... · 2016. 4. 20. ·...

78
Toxicology for Industrial and Regulatory Scientists Toxicology for Industrial and Regulatory Scientists Nonclinical Safety Evaluation of Biotechnology-Derived Therapeutics Melanie Hartsough Ph D Melanie Hartsough, Ph.D . Senior Consultant Biologics Consulting Group, Inc. April 30, 2015

Upload: others

Post on 19-Feb-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

  • Toxicology for Industrial and Regulatory ScientistsToxicology for Industrial and Regulatory Scientists

    Nonclinical Safety Evaluation of Biotechnology-Derived Therapeutics

    Melanie Hartsough  Ph DMelanie Hartsough, Ph.D.Senior Consultant

    Biologics Consulting Group, Inc.

    April 30, 2015

  • History of biotechnology derived products

    Presentation Overview History of biotechnology-derived products Fundamental differences between small and

    large molecule pharmaceuticals g p Challenges specific to biologics Species specificity

    Immunogenicity Immunogenicity Alternative approaches/models Reproductive/developmental studiesReproductive/developmental studies Studies in Nonclinical Programs Summary

    2

  • Pharmaceutical Products

    Majority are small molecule (traditional) pharmaceuticals Extensive experience and large historical database Industry Regulatory agencies (e g FDA EMA)Regulatory agencies (e.g.,FDA, EMA)

    Scientific and regulatory expectations for nonclinical toxicology studies are well defined

    3

  • Pharmaceutical Products

    Biotechnology-derived products (Biologics) Increased production over the last 25 years for use in

    various clinical indicationsvarious clinical indications Relatively new (compared to small molecules) Less experience (industry; regulatory agencies) Less experience (industry; regulatory agencies) Smaller historical database (clinical and nonclinical)

    4

  • Biologics: DefinitionS b t d i d f li i i Substances derived from living organisms Humans Animals Animals Plants

    Substances produced by biotechnology methodsSubstances produced by biotechnology methods

    5

  • Biopharmaceuticals The definition of biologics encompasses: Vaccines Blood and blood-derived products Anti-toxins

    All i Allergenics Cell, gene and tissue products Monoclonal antibodies Therapeutic proteins Monoclonal antibodies Recombinant proteins Fusion proteins

    Therapeutic proteins,biopharmaceuticals, biotechnology‐derived productsp

    6

  • BiopharmaceuticalsBiologics (using recombinant DNA technology) Biologics (using recombinant DNA technology) initially developed in early 1980’s Revolutionized the treatment of human disease by:

    Mimicking or supplementing a human endogenous protein (growth hormone, erythropoietin)e y opo e )

    Activating (agonistic) or blocking (antagonistic) a signaling pathway through specific receptor or ligand binding (monoclonal antibodyligand binding (monoclonal antibody, fusion protein)

    7

  • Bi h ti l E lBiopharmaceuticals: Examples Humulin® first approved recombinant therapeutic Humulin® - first approved recombinant therapeutic

    protein was recombinant human insulin Produced by genetically modified bacteria Approved in 1982 Diabetes

    Intron® - interferon--2b Approved in 1986 Hairy cell leukemiaOrthoclone OKT 3 ® (muromonab) Orthoclone OKT-3 ® (muromonab) Anti-CD3 murine monoclonal antibody (IgG2a) Approved in 1986

    R l t l t

    8

    Renal transplant

  • Bi h ti l E lBiopharmaceuticals: Examples Enbrel® Fc (IgG ) fusion protein Enbrel® - Fc (IgG1) fusion protein

    Approved in 1998 Specific to human tumor necrosis factor

    Rh t id th iti Rheumatoid arthritis Avastin® (Bevacizumab)

    Anti-vascular endothelial growth factor antibody (IgG1)g y ( g ) Approved in 2004 Colorectal cancer

    Vectibix® (Panitumumab) Vectibix® (Panitumumab) Anti-epidermal growth factor receptor monoclonal antibody

    (IgG2)Approved in 2006

    9

    Approved in 2006 Colorectal cancer

  • Bi h ti l VBiopharmaceuticals Vs. Small Molecules

    10

  • Biopharmaceuticals Vs. Small Molecules

    Fundamental differences exist between small molecules and biopharmaceuticalsp Differences influence the types of nonclinical

    toxicology studies to support the safety assessment Guidance documents support different

    approach for the nonclinical toxicology studies (ICH S6(R1): Preclinical Safety Evaluation of Biotechnology derived Pharmaceuticals)Biotechnology-derived Pharmaceuticals)

    11

  • Biopharmaceuticals Vs Small MoleculesBiopharmaceuticals Vs. Small Molecules Small Molecules

    M f t d b h i l Biopharmaceuticals

    D i d f ti Manufactured by chemical processes

    Derived from genetic manipulation of DNA

    Manufactured by living cells

    Low molecular weight1000 d lt

    High molecular weight ≥ 1000 daltons

    < 1000 daltons Potential for extensive

    distribution within the b d

    ≥ 1000 daltons High molecular weight

    proteins are largely confined initially to thebody confined initially to the vascular space

    More limited distribution within the bodywithin the body

    12

  • S Bi h i l VStructure: Biopharmaceuticals Vs. Small Molecules

    Fab = 1/3 mAb 1 structure Higher order

    Fab = ~1/3 mAb

    Statinstructure Post translational

    modification

    Statin

    modification microheterogeneity

    Statin MW ~400 DaMonoclonal Antibody MW ~ 150 000 Da

    87 AMonoclonal Antibody MW ~ 150,000 Da

    13

  • C f M f i fConsequence of Manufacturing from Living Cells– Comparability Complexity of protein structure results in

    microheterogeneity of final product Glycosylation deamidation oxidation disulfide bonds free thiols Glycosylation, deamidation, oxidation, disulfide bonds, free thiols,

    clipping, aggregates

    Manufacturing of biologics evolves with product d l t (i f t h l t )development (i.e., frequent changes, scale-ups, etc)

    Since manufacturing changes may alter product, comparability assessments are frequently performedcomparability assessments are frequently performed

    Comparability assessment ensures that the manufacturing changes have not affected the safety,

    ff fidentity, purity or efficacy, including immunogenicity, of the product

    14

  • C f M f i fConsequence of Manufacturing from Living Cells– Comparability Analytical and functional assays performed (quality

    attributes/ CMC issues) If product comparability cannot be established with If product comparability cannot be established with

    quality assessments, preclinical studies may be required PK assessments PD assessments Toxicology studies Toxicology studies

    Product used in nonclinical studies not required to be “identical” to that going into the clinic, but does have to be “comparable” in order to extrapolate the preclinical safety data to the clinical scenario

    15

  • Bi h ti l V S ll M l lBiopharmaceuticals Vs. Small Molecules Small Molecules

    Metabolized by Liver Biopharmaceuticals

    Catabolized by body Metabolized by Liver Biotransformation studies Metabolites identified

    Catabolized by body (proteolytic degradation) Amino acids Small peptides

    Potential for active or toxic metabolites

    S a pep des Active or toxic breakdown

    protein products generally not a concern

    Biotransformation studies generally not performed

  • Biopharmaceuticals Vs. Small Molecules

    Small Molecules Biopharmaceuticals Small Molecules Drug-drug interactions

    potential concern Half-life relatively short

    Biopharmaceuticals Drug-drug interactions are

    less of a concern Half-life can be long Half-life relatively short

    Minutes Hours Few days

    Half life can be long mAb-Days to weeks

    y Impurities generally consist

    of: Organic residuals

    Impurities generally consist of host cell molecules: DNA

    Solvents Toxicity studies for

    impurities may be

    Protein Toxicity studies for

    impurities usually not d dnecessary needed

    17

  • Biopharmaceuticals Vs. Small Molecules

    Small Molecules Biopharmaceuticals Small Molecules Less target specificity

    (compared to biologics)

    Biopharmaceuticals High target specificity Toxicity often related to ( g )

    Toxicities often non-specific/not related to the intended target

    o c y o e e a ed ointended target/ pharmacological action of the drugthe intended target

    “Off-target toxicity”of the drug “On-target toxicity” Exaggerated

    pharmacology

    18

  • Biopharmaceuticals Vs. Small Molecules

    Small Molecules Immunogenicity

    generall not a concern

    Biopharmaceuticals Immunogenicity is often

    a challengegenerally not a concern

    Species limitations

    a challenge

    Species limitations may Species limitations generally not a concern 2 species usually used for

    toxicology studies

    Species limitations may be a challenge Only 1 relevant animal

    species may be availabletoxicology studies (1 rodent; 1 non-rodent)

    species may be available

    19

  • Properties of Biologics that Can Influence Nonclinical Toxicology Programs High degree of target and species specificity High degree of target and species specificity Immunogenicity

    20

  • Species Specificity

    21

  • Properties of Biologics:Properties of Biologics:Species Specificity

    Most biopharmaceuticals are: Human proteins that are highly targeted to a human receptorHuman proteins that are highly targeted to a human receptor Antibodies specific for a human protein or receptor

    Due to high specificity to the human target many Due to high specificity to the human target, many biopharmaceuticals do not recognize the target in all animal species commonly used in nonclinical t i l t ditoxicology studies. Mouse, rat, rabbit, dog, minipig, monkey

    22

  • Species Specificity Toxicology studies for biopharmaceuticals should beToxicology studies for biopharmaceuticals should be

    conducted in a pharmacologically relevant species (ICHS6(R1)).

    Relevant species is one in which the biopharmaceutical is pharmacologically active due to the expression of the intended target (e.g., receptor or epitope).g ( g , p p p )

    Toxicology studies in non-relevant animal species (one that does not express the target) can be misleading and

    di d (ICHS6(R1))are discouraged (ICHS6(R1)). Unreliable safety data Not predictive of potential human toxicity

    23

  • Species Specificity How to demonstrate relevant species: How to demonstrate relevant species: Sequence identity of target Target expression and distributionTarget expression and distribution Binding assays Biacore, flow cytometry, ELISA

    Functional bioassays Cell proliferation, signal transduction modulation, enzyme

    changeschanges In vivo pharmacology

    24

  • S i S ifi itSpecies Specificity

    There are cases when only one relevant animal species can be identified

    M t ft h i t ( l Most often nonhuman primates (e.g., cynomolgus monkeys)

    One species is acceptable with sufficient justificationOne species is acceptable with sufficient justification

    25

  • Species Specificity Nonhuman PrimatesSpecies Specificity – Nonhuman Primates NHP often the only pharmacologically relevant animal species

    for toxicology studies gy Old world NHPs Cynomolgus monkey

    Most common NHP used (larger historical database)( g ) Rhesus monkey

    New world NHPs (marmosets) Limited historical database for toxicity studiesLimited historical database for toxicity studies Very sensitive to disruptions in environment More often used for PK studies

    Great apes Great apes Chimpanzees are NEVER appropriate for toxicology!

    Limited data obtained (endangered species) Small animal number and large variability Small animal number and large variability Difficult data interpretation Experimentally naïve animals not available

    26

  • Ch ll U i N h P i t Various challenges using NHP as the animal model in

    t i l t di

    Challenges Using Nonhuman Primates

    toxicology studies Limited availability (compared to rodents and canines) Contact CROs early to place study

    Limited number of animals/group (compared to rodents/canines) Can make data interpretation difficult

    Cost is high (compared to rodents/canines) Cost is high (compared to rodents/canines) particularly sexually mature animals!

    Background and/or sub-clinical disease ( i b t i l it )(viruses, bacterial, parasites) Hepatitis, malaria, shigella Impact data interpretation

    Particularly if the molecule is an immunosuppressive Particularly if the molecule is an immunosuppressive Are observed changes due to pre-existing infection or molecule?

    27

  • Challenges Using Nonhuman Primates Cont. NHPs of different originsg Indonesian vs. Chinese origin

    NHPs are heterogenousC d t i b d i t Compared to inbred mice or rats

    Increases variability in data

    28

  • Species Specificity There are cases when NO relevant species can be There are cases when NO relevant species can be

    identified Specific for human or human and chimpanzeep p Alternative approaches can be used to identify

    potential safety risks Alternative approaches: Surrogate/analogous proteins

    T i i l Transgenic animals Knockout animals Animal models of disease Animal models of disease

    29

  • Immunogenicity

    30

  • Immunogenicity

    Immunogenicity = immune response against the product, resulting in production of anti-product antibodiesantibodies Biopharmaceuticals are often immunogenic in

    animalsanimals Abbreviations that you may see: ADA = anti-drug antibodies RAHA= rat anti-human antibodies PAHA = primate anti-human antibodies MAHA = monkey anti-human antibodies MAHA = monkey anti-human antibodies

    31

  • Immunogenicity Types of anti-product antibodiesyp p Binding Minimal to no impact on product exposure

    Clearing Increase clearance of product which decrease systemic

    exposure of productexposure of product Can alter distribution of product to tissues

    Sustaining (rare) Decrease clearance of product which prolong half-

    life/exposure of product Potentially lead to or increase toxicityy y

    32

  • Immunogenicity

    Types of anti-product antibodies Neutralizing Interfere with product binding to target Neutralize pharmacological activity of product

    Cross-reactive with endogenous proteinsCross reactive with endogenous proteins Neutralize the pharmacological activity of the product and

    the corresponding endogenous proteinC i f t li ti f ti it f d d t Concern is for neutralization of activity of nonredundant system

    33

  • Immunogenicity Anti product antibodies can affect the outcome of a Anti-product antibodies can affect the outcome of a

    toxicology study No effect Decrease exposure by increasing clearance of the

    product or neutralizing the product’s activity Sustain exposure by decreasing clearance (rare) Cause adverse effects not directly related to the

    product (anaphylaxis immune complex deposition)product (anaphylaxis, immune complex deposition) Cross-react with the endogenous protein

    34

  • Immunogenicity Included as an endpoint in biopharmaceutical Included as an endpoint in biopharmaceutical

    toxicology studies Aid in the interpretation of the study p y

    Primary concern is for the development of clearing and/or neutralizing anti-product antibodies Lower exposure of the target organs to

    biopharmaceutical Generation of misleading toxicity data Generation of misleading toxicity data

    35

  • Immunogenicity

    Mere presence of anti-product antibodies is not a reason to terminate a study (does not invalidate the study)study) Sufficient number of animals may be exposure to

    active productp Studies terminated when complete loss of exposure of

    activity of product and/or severe adverse reactions l d dditi l d ipreclude additional dosing

    36

  • Immunogencity Various factors can influence the immunogenic Various factors can influence the immunogenic

    potential of biopharmaceuticals Route of administration

    Inhalation> SC> IP> IM >IV > topical* Duration of dosing

    Typically repeated dosing is more immunogenic than a single dose

    Product Protein structure Manufacturing processes (glycosylated vs. nonglycosylated) Impurities (DNA, host cell proteins) Aggregates (high order)

    *Braun A et al (1997) Pharm Res. 14(10) 1472-14878

    37

  • Immunogenicity Anti-product antibodies may make long term toxicity

    studies and reproductive and developmental studies difficultdifficult

    Immunogenicity in animals is not necessarily predictive of human response (ICHS6(R1))predictive of human response (ICHS6(R1))

    38

  • Immunogenicity Case Studyg y y

    39

  • I i itImmunogenicity

    St d D i Study Design: Human monoclonal antibody

    Administered to cynomolgus monkeys Administered to cynomolgus monkeys Intravenous injection

    Once weekly for 4 weeksy Development of clearing ADAs resulted in:

    Increase drug clearance Decreased exposure over time

    40

  • Clearing Antibody: Impact on Exposure

    Day 1 Day 281000 1000

    100 100

    n (

    g/m

    l)

    n (

    g/m

    l)

    Antibody negative

    1

    10

    1

    10

    ncen

    trat

    ion

    ncen

    trat

    ion g

    0.1

    1

    0.1

    1

    rug

    X C

    on

    rug

    X C

    on

    0.01 0.01Seru

    m D

    r

    Seru

    m D

    r

    Antibody positive

    41Time (days)

    0 2 4 6 8 10 12 14

    Time (days)

    0 2 4 6 8 10 12 14

    41

  • Alternative Approaches and Models

    42

  • Alt ti A h /M d l In some cases, alternative approaches for the

    Alternative Approaches/Modelspp

    nonclinical toxicology studies for biologics are necessary Product pharmacologically active in humans only Product pharmacologically active in humans only Anti-product antibodies imposed limitations on the ability

    to conduct comprehensive nonclinical safety studies Clearing and/or neutralizing antibodies Unable to maintain exposure/pharmacological activity

    throughout studyg y ICH S6(R1) – allows for flexibility so alternative

    approaches can potentially be used for the nonclinical safety assessment

    43

  • Alternative Approaches/Models Homologous (Surrogate/Analogous)

    proteinsp Transgenic animals Knockout animals Knockout animals Animal models of disease

    44

  • H l P t i Homologous proteins recognize the ortholog

    Homologous Proteing p g g

    of the original human target Animal models

    Mouse (most common)Mouse (most common) Rat Cynomolgus monkey

    Advantage Advantage Good model for molecules that cross-react only with

    human target Most commonly used alternative methodMost commonly used alternative method

    45

  • H l (S t ) P t i

    ICH S6(R1) h l t i h ld

    Homologous (Surrogate) Protein

    ICH S6(R1)- homologous proteins should resemble the clinical candidate as much as possible with regard to: p g Production process Range of impurity and/or contaminant profileN t h t i th h l Necessary to characterize the homologous protein to ensure it is a suitable surrogate for the clinical candidateclinical candidate

    46

  • H l (S t ) P t i Demonstrate surrogate has pharmacological

    Homologous (Surrogate) Protein Demonstrate surrogate has pharmacological mechanism similar to clinical candidate Binding studies

    A /bi k f h l i l ti it Assays/biomarker of pharmacological activity Downmodulation/upmodulation of receptor/target or expression of cytokine(s)

    or proliferation of certain cell populations Necessary to characterize PK and ADA responses of

    surrogate in animal species used in toxicologysurrogate in animal species used in toxicology studies

    47

  • H l (S t ) P t i

    N t d t i th d f th h l

    Homologous (Surrogate) Protein

    Necessary to determine the dose of the homologous protein in your animal species that is pharmacologically equivalent to the human dose g ywith the clinical candidate Based on pharmacology and PK data Data used to determine doses used for toxicologyData used to determine doses used for toxicology

    studies with the homologous protein

    48

  • Homologous (Surrogate) Protein Challenges using an homologous protein:

    Homologous (Surrogate) Protein g g g p

    It will never be your clinical candidate! Differences from clinical candidate

    Manufacturing process Manufacturing process Different impurity profile PK and ADA profile

    Example: lower exposure with surrogate due Example: lower exposure with surrogate due to high rate of ADA responses

    Pharmacological activity Potency equal to clinical candidate?Potency equal to clinical candidate? Mechanism of action similar?

    49

  • Homologous (surrogate) Protein

    Challenges using an homologous protein: No established criteria or regulatory guidance for

    h l t i l t dipharmacology or toxicology studies How rigorous should the assessment of the

    pharmacological/toxicological activities of the homologue be? Aggregates Host-cell proteins

    How rigorous should assessment of the homologous material be?

    Company/regulatory concerns Suitability of homologue to characterize safety of clinical y g y

    candidate when compared to human data

    50

  • Homologous (Surrogate) ProteinHomologous (Surrogate) Protein Challenges using an homologous protein:g g g p Resource intensive! Development & manufacturing resources needed

    (essentially developing a 2nd molecule!!) Generation of material Characterization of material Characterization of material

    Impurities Aggregates Formulation excipients differ from clinical candidate

    Studies to demonstrate comparability to the clinical candidate

    Assay development for PK and ADA 51

  • Alt ti A h /M d l Transgenic animals (mice)

    Alternative Approaches/Models

    Animal model expressing human target Often used in pharmacology studies

    Advantageg Allows for evaluation of clinical candidate (unlike

    surrogate) Limitations

    Expensive to develop and maintain colony Lack of historical data; data interpretation can be difficult Limited lifespan; limited number of animals Spontaneous pathologies can be associated with having

    the gene expressed

    52

    g p May be difficult to produce mice with consistent

    expression and tissue distribution of the human target

  • Alt ti A h /M d l Knockout models

    Alternative Approaches/Models

    Evaluate the pathologies associated with not having the target expressed

    Limitations Expensive to develop and maintain colony

    Lack of historical data Lack of historical data Limited lifespan Limited animal availabilityy Other spontaneous pathologies associated with not

    having gene expressedD l i di t l i l’ lif

    53

    Develop immediately or over animal’s life span Does not mimic therapeutic setting

  • Alt ti A h /M d l Animal models of disease

    Alternative Approaches/Models

    Allows for evaluation of general toxicity and undesirable promotion of disease progression during treatment with drugdrug

    Limitations Lack of historical data Limited lifespan or number of animals Confounding effects of disease

    May interfere with interpretation of the toxicological data GLP compliance may be difficult due to the complexity

    of these studies

    54

    Often available in research facilities that do not have GLP capabilities

  • Reproduction and Development

    Toxicity StudiesToxicity Studies

    55

  • Reproduction/DevelopmentReproduction/Development Studies ICHS6(R1)- Studies performed according to

    ICHS5(R2); however, study design and dosing schedule may be modified based on speciesschedule may be modified based on species specificity, the nature of the product and mechanism of action, immunogenicity and/or PK behavior and , g yembryo-fetal exposure Studies should only use pharmacologically relevant

    species, ideally with clinical product

    56

  • Reproduction/DevelopmentReproduction/Development Studies Studies should be performed in rat and rabbit, if

    pharmacologically relevant species When no relevant species, alternative models

    may be explored If weight of evidence (e.g., mechanism of action,

    phenotypic data from genetically modified animals class effects etc) suggests adverseanimals, class effects, etc) suggests adverse effect on fertility or pregnancy outcome may not need to perform studiesneed to perform studies

    57

  • Reproduction/DevelopmentReproduction/Development Studies If NHP only relevant species: Only developmental studies are necessaryy p y May use alternative model with scientific justification Study may be conducted during Phase 3 and

    submitted at time of licensure, as long as appropriate clinical precautions are taken

    ICHS6(R1) provides a design consideration for ICHS6(R1) provides a design consideration for combined embryo-fetal and pre/post-natal development (PPND) study (referred to as ePPND)

    58

  • Reproduction/DevelopmentReproduction/Development Studies

    Effects on fertility can be assessed in repeat-dose toxicity studies of at least 3 months

    Sexually mature NHPs Sexually mature NHPs evaluation of reproductive tract (organ weights and

    histopathological evaluation) If concern, menstrual cyclicity, sperm count, sperm

    morphology/motility, and reproductive hormone levels can be evaluated

    Not recommended to develop homologous protein or transgenic model to conduct mating studies in rodents

    Studies are used for hazard identificationStudies are used for hazard identification

    59

  • Nonclinical Studies to Support th S f t f Bi h ti lthe Safety of Biopharmaceuticals

    60

  • Biopharmaceutical NonclinicalBiopharmaceutical Nonclinical Programs Pharmacodynamic studies Pharmacodynamic studies

    Mechanism of action, proof of concept

    Safety Pharmacology Studies Many times can be incorporated into the general toxicology

    studies (ICHS7a) hERG assay not performed y p

    Pharmaco-/toxicokinetics Absorption studies

    Ti di t ib ti t di t t i ll f d d t Tissue distribution studies not typically performed due to limitations with data interpretation

    Metabolism/ excretion studies not typically performed ( t b li )(catabolism)

    61

  • Biopharmaceutical NonclinicalBiopharmaceutical Nonclinical Programs General toxicology studies Program designed based on properties of the

    bi h ti lbiopharmaceutical Human Tissue Cross-reactivity study (generally

    mAbs only)mAbs only) Local tolerance studies Incorporated into the general toxicology studiesIncorporated into the general toxicology studies

    Genotoxicity assay Not typically performed, inability of product to enter y y y

    living cells

    62

  • Biopharmaceutical NonclinicalBiopharmaceutical Nonclinical Programs Reproductive and developmental toxicology studies

    Study designs dependent upon relevant species, immunogenicity, mechanism of action embryo-fetal exposuremechanism of action, embryo fetal exposure

    If NHP the only relevant species- studies are more complicated ICHS6(R1) provides design considerations

    C i i it t di Carcinogenicity studies Weight of evidence approach Published data, class effects, product biology and mechanism of p gy

    action, etc Studies may not be required and/or may not be feasible

    63

  • Summary There are fundamental differences between There are fundamental differences between

    biopharmaceuticals and small molecules Properties of biologics can influence the approachProperties of biologics can influence the approach

    and design for the nonclinical toxicology studies Critical to conduct the appropriate study based on

    the properties of the product pharmacologically relevant species

    I i it fil Immunogenicity profile

    64

  • Summary

    Consider using alternative models for the program when appropriate

    E Ab th t t l ith h t t Ex: mAb that cross-reacts only with human target Develop surrogate antibody rather than conducting toxicity

    studies in non-relevant animal species

    There is “no one program fits all” approach for biopharmaceuticals

    65

  • Summary Approach for the nonclinical safety assessment Approach for the nonclinical safety assessment

    strategy for a biopharmaceutical should be based on: Scientific rationale Relevant species, mechanism of action, study feasibility,

    other product propertiesother product properties Indication/patient population Ex: Oncology vs psoriasis Ex: Women of child-bearing potential vs. post-menopausal

    women Relevance of the studies for determining and Relevance of the studies for determining and

    understanding human risk66

  • ReferencesReferences ICH S6 (R1): Preclinical Safety Evaluation of Biotechnology-Derived Products (1997), with addendum (2011)Derived Products (1997), with addendum (2011) ICHS5 (R2):Detection of Toxicity of Reproduction for Medicinal Products and Toxicity to Male Fertility (2000) Points to Consider in the Manufacture and Testing of Points to Consider in the Manufacture and Testing of Monoclonal Antibody Products for Human Use (CBER, 1997) Raptiva®, FDA Review, Application Number 125075/0, approval 10/27/03 Clarke, J. et al (2004). Reg. Tox. Pharm., 219-226 Braun, A. et al (1997). Pharm. Res, 14(10), 1472-1478Braun, A. et al (1997). Pharm. Res, 14(10), 1472 1478 Mounho, B. et al (2003). The Toxicologist #1880 pg 387 Clarke, J. et al (2003). The Toxicologist #361 pg 74 www bio org

    67

    www.bio.org www.fda.gov

  • Extra Slides

    68

  • Homologous Antibody: Case ExampleHomologous Antibody: Case Example

    69

  • C E l Efalizumab (Raptiva®)

    Case ExampleEfalizumab (Raptiva ) Humanized IgG1 monoclonal antibody to human

    CD11a Cross-reacts only with human and chimpanzee CD11aCross reacts only with human and chimpanzee CD11a

    Indication: moderate-severe plaque psoriasis Men & women of childbearing age

    Administered chronically Administered chronically Chronic and reproductive toxicity studies necessary

    Developed surrogate antibody Chimeric rat/mouse anti-mouse CD11a antibody

    muM17 Toxicology studies to support registration were conducted

    i CD 1 i

    70

    in CD-1 mice

    Clarke, J. et al (2004). Reg. Tox. Pharm., 219-226Raptiva, FDA Review, Application Number 125075/0, approval 10/27/03

  • C E l C t Evaluation of muM17 as suitable surrogate antibody

    Case Example Cont. Evaluation of muM17 as suitable surrogate antibody for efalizumab Binding affinity to CD11a

    efalizumab to human CD11a ~ 3.0 nM muM17 to murine CD11a ~ 2.7 nM

    Similar tissue binding propertiesg p p Efalizumab: human leukocytes in blood and spleen muM17: mouse leukocytes in blood and spleen

    Characterize PK/antibody responses of muM17 in mice Characterize PK/antibody responses of muM17 in mice Demonstrate pharmacological activity of muM17 in mice

    similar to efalizumab in humans

    71Clarke, J. et al (2004). Reg. Tox. Pharm., 219-226Raptiva, FDA Review, Application Number 125075/0, approval 10/27/03

    decrease CD11a expression on T lymphocytes

  • C E l C t Determined dose of muM17 in mice that was

    Case Example Cont.

    pharmacologically equivalent to the efalizumab human dose Based on CD11a downmodulation on T lymphocytes Based on CD11a downmodulation on T lymphocytes

    Very sensitive biomarker on pharmacodynamic activity for efalizumab in humans and muM17 in mice

    3 mg/kg/week identified as minimal muM17 dose that maintained maximal CD11a downmodulation Approximately equivalent to efalizumab human dose pp y q

    of 1 mg/kg/week muM17 doses for toxicology studies

    3, 10, and 30 mg/kg (10-fold safety factor based on dose)

    72Clarke, J. et al (2004). Reg. Tox. Pharm., 219-226Raptiva, FDA Review, Application Number 125075/0, approval 10/27/03

  • C E l C t Toxicology program with muM17

    Case Example Cont.

    Repeated-dose toxicity (up to 6 months duration) Reproductive toxicity

    F tilit Fertility Embryo/fetal development Peri/postnatalp

    Immunotoxicology Adult mice

    Off i b t d d i i t d M17 d i Offspring born to dams administered muM17 during gestation and lactation

    73Raptiva, FDA Review, Application Number 125075/0, approval 10/27/03

  • Transgenic Animal Model: Case ExampleTransgenic Animal Model: Case Example

    74

  • T i A i l M d l C St d Keliximab

    H l k hi i Ab

    Transgenic Animal Model: Case Study

    Human-cynomolgus monkey chimeric mAb directed against human CD4

    Cross-reactivity limited to human CD4y Indication

    Rheumatoid arthritisM l d f l f hildb i Males and females of childbearing age

    Chronic treatment

    Developed human CD4 transgenic mouseDeveloped human CD4 transgenic mouse Model (HuCD4/Tg mice)

    Murine CD4 knockout, human CD4 knock-in

    75Bugelski, PJ et al. (2000). Hum. Exp. Toxicol. 19: 230 – 243

  • Case Study Cont Necessary to characterize HuCD4/Tg mice to ensure

    suitable animal model for toxicology studies

    Case Study Cont.

    suitable animal model for toxicology studies Compare distribution of cells expressing human CD4

    transgene in comparison to murine CD4 Demonstrate pharmacological activity of keliximab in

    HuCD4/Tg miceDepletion of HuCD4+ lymphocytes Depletion of HuCD4+ lymphocytes

    Determine suitability of mice for chronic studies with regard to background pathologies and survivalregard to background pathologies and survival

    Ensure immune system of mice was competent Immunotoxicology studies

    76

    Developing/maintaining animal model resource intense!

  • C St d C t Toxicology studies using HuCD4/Tg mice

    Case Study Cont.

    Short term and chronic (6-month) toxicity studies Male/female fertility Embryo-fetal development Immune function in F1 mice

    Host resistance to infection Host resistance to infection Immune function

    Host resistance to infection

    77

  • Please click on the link below to enter your comments on this talk

    https://www.surveymonkey.com/s/VNZC9MQ

    78