throbber
Guidance for Industry
`
`Drug Interaction Studies —
`Study Design, Data Analysis, and
`Implications for Dosing and Labeling
`
`
`
`
`DRAFT GUIDANCE
`This guidance document is being distributed for comment purposes only.
`
`
`
`
`
`
`
`
`
`
`Comments and suggestions regarding this draft document should be submitted within 60 days of
`publication in the Federal Register of the notice announcing the availability of the draft
`guidance. Submit comments to the Division of Dockets Management (HFA-305), Food and
`Drug Administration, 5630 Fishers Lane, rm. 1061, Rockville, MD 20852. All comments
`should be identified with the docket number listed in the notice of availability that publishes in
`the Federal Register.
`
`For questions regarding this draft document contact (CDER) Shiew-Mei Huang, 301-796-1541,
`or (CBER) Toni Stifano, 301-827-6190.
`
`
`
`U.S. Department of Health and Human Services
`Food and Drug Administration
`Center for Drug Evaluation and Research (CDER)
`Center for Biologics Evaluation and Research (CBER)
`
`September 2006
`Clinical Pharmacology
`
`1
`
`TEVA1052
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`Guidance for Industry
`Drug Interaction Studies —
`Study Design, Data Analysis, and
`Implications for Dosing and Labeling
`
`
`
`Additional copies are available from:
`
`Office of Training and Communications
`Division of Drug Information, HFD-240
`Center for Drug Evaluation and Research
`Food and Drug Administration
`5600 Fishers Lane
`Rockville, MD 20857
`(Tel) 301-827-4573
`http://www.fda.gov/cder/guidance/index.htm
`
`or
`
` Office of Communication, Training and
`Manufacturers Assistance, HFM-40
`Center for Biologics Evaluation and Research
` Food and Drug Administration
`1401 Rockville Pike, Rockville, MD 20852-1448
` http://www.fda.gov/cber/guidelines.htm
`
`U.S. Department of Health and Human Services
`Food and Drug Administration
`Center for Drug Evaluation and Research (CDER)
`Center for Biologics Evaluation and Research (CBER)
`
`September 2006
`Clinical Pharmacology
`
`G:\6695dft.doc
`09/08/06
`
`2
`
`

`

`
`
`
`
`I.
`
`TABLE OF CONTENTS
`
`INTRODUCTION..............................................................................................................................................1
`
`II. BACKGROUND ................................................................................................................................................2
`A. METABOLISM...................................................................................................................................................2
`B. DRUG-DRUG INTERACTIONS ............................................................................................................................2
`III. GENERAL STRATEGIES ...............................................................................................................................4
`A.
`IN VITRO STUDIES............................................................................................................................................4
`SPECIFIC IN VIVO CLINICAL INVESTIGATIONS..................................................................................................5
`B.
`POPULATION PHARMACOKINETIC SCREENS .....................................................................................................6
`C.
`IV. DESIGN OF IN VIVO DRUG-DRUG INTERACTION STUDIES .............................................................6
`A.
`STUDY DESIGN.................................................................................................................................................6
`STUDY POPULATION.........................................................................................................................................8
`B.
`C. CHOICE OF SUBSTRATE AND INTERACTING DRUGS ..........................................................................................8
`D. ROUTE OF ADMINISTRATION..........................................................................................................................12
`E. DOSE SELECTION ...........................................................................................................................................12
`ENDPOINTS.....................................................................................................................................................13
`F.
`SAMPLE SIZE AND STATISTICAL CONSIDERATIONS ........................................................................................14
`G.
`V. LABELING IMPLICATIONS .......................................................................................................................15
`
`APPENDIX A TABLES............................................................................................................................................17
`
`APPENDIX B FIGURES..........................................................................................................................................24
`
`APPENDIX C-1 IN VITRO DRUG METABOLIZING ENZYME IDENTIFICATION ..................................25
`
`APPENDIX C-2 IN VITRO EVALUATION OF CYP INHIBITORS.................................................................31
`
`APPENDIX C-3 IN VITRO EVALUATION OF CYP INDUCTION..................................................................35
`
`APPENDIX D IN VITRO EVALUATION OF P-GLYCOPROTEIN (P-GP, MDR1) SUBSTRATES AND
`INHIBITORS.............................................................................................................................................................38
`
`REFERENCES ..........................................................................................................................................................51
`
`
`
`
`
`
`G:\6695dft.doc
`09/08/06
`
`3
`
`

`

`1
`2
`3
`4
`5
`6
`7
`8
`9
`10
`11
`12
`13
`14
`
`15
`16
`17
`18
`19
`20
`21
`22
`23
`24
`25
`26
`27
`28
`29
`30
`31
`32
`33
`34
`35
`36
`37
`38
`39
`
`
`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`Guidance for Industry1
`
`Drug Interaction Studies — Study Design, Data Analysis, and
`Implications for Dosing and Labeling
`
`
`
`INTRODUCTION
`
`
`This draft guidance, when finalized, will represent the Food and Drug Administration's (FDA's)
`current thinking on this topic. It does not create or confer any rights for or on any person and does
`not operate to bind FDA or the public. You can use an alternative approach if the approach satisfies
`the requirements of the applicable statutes and regulations. If you want to discuss an alternative
`approach, contact the FDA staff responsible for implementing this guidance. If you cannot identify
`the appropriate FDA staff, call the appropriate number listed on the title page of this guidance.
`
`
`
`I.
`
`This guidance provides recommendations for sponsors of new drug applications (NDAs) and
`biologics license applications (BLAs) for therapeutic biologics2 who are performing in vitro
`and in vivo drug metabolism, drug transport, and drug-drug interaction studies. The
`guidance reflects the Agency’s current view that the metabolism of an investigational new
`drug should be defined during drug development and that its interactions with other drugs
`should be explored as part of an adequate assessment of its safety and effectiveness. For
`drug-drug interactions, the approaches considered in the guidance are offered with the
`understanding that the relevance of a particular study depends on the characteristics and
`proposed indication of the drug under development. Furthermore, not every drug-drug
`interaction is metabolism-based, but may arise from changes in pharmacokinetics caused by
`absorption, distribution, and excretion interactions. Drug-drug interactions related to
`transporters are being documented with increasing frequency and are important to consider in
`drug development. Although less well studied, drug-drug interactions may alter
`pharmacokinetic/pharmacodynamic (PK/PD) relationships. These important areas are not
`considered in detail in this guidance.
`
`Discussion of metabolic and other types of drug-drug interactions is also provided in other
`guidances, including the International Conference on Harmonization (ICH) E7 Studies in
`Support of Special Populations: Geriatrics, and E3 Structure and Content of Clinical Study
`Reports, and FDA guidances for industry on Studying Drugs Likely to be Used in the Elderly
`and Study and Evaluation of Gender Differences in the Clinical Evaluation of Drugs.
`
`1 This guidance has been prepared by the Drug-Drug Interaction Working Group in the Clinical Pharmacology
`Section of the Medical Policy Coordinating Committee in the Center for Drug Evaluation and Research, with
`input from the Center for Biologics Evaluation and Research, at the Food and Drug Administration.
`2 For more information on what constitutes a therapeutic biologic product, please see Internet site
`http://www.fda.gov/cder/biologics/qa.htm.
`
`G:\6695dft.doc
`09/08/06
`
`4
`
`

`

`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`
`BACKGROUND
`
`
`FDA’s guidance documents, including this guidance, do not establish legally enforceable
`responsibilities. Instead, guidances describe the Agency’s current thinking on a topic and
`should be viewed only as recommendations, unless specific regulatory or statutory
`requirements are cited. The use of the word should in Agency guidances means that
`something is suggested or recommended, but not required.
`
`
`II.
`
`A. Metabolism
`
`The desirable and undesirable effects of a drug arising from its concentrations at the sites of
`action are usually related either to the amount administered (dose) or to the resulting blood
`concentrations, which are affected by its absorption, distribution, metabolism, and/or
`excretion. Elimination of a drug or its metabolites occurs either by metabolism, usually by
`the liver or gut mucosa, or by excretion, usually by the kidneys and liver. In addition,
`protein therapeutics may be eliminated through a specific interaction with cell surface
`receptors, followed by internalization and lysosomal degradation within the target cell.
`Hepatic elimination occurs primarily by the cytochrome P450 family (CYP) of enzymes
`located in the hepatic endoplasmic reticulum, but may also occur by non-P450 enzyme
`systems, such as N-acetyl and glucuronosyl transferases. Many factors can alter hepatic and
`intestinal drug metabolism, including the presence or absence of disease and/or concomitant
`medications, or even some foods, such as grapefruit juice. While most of these factors are
`usually relatively stable over time, concomitant medications can alter metabolism abruptly
`and are of particular concern. The influence of concomitant medications on hepatic and
`intestinal metabolism becomes more complicated when a drug, including a prodrug, is
`metabolized to one or more active metabolites. In this case, the safety and efficacy of the
`drug/prodrug are determined not only by exposure to the parent drug but by exposure to the
`active metabolites, which in turn is related to their formation, distribution, and elimination.
`Therefore, adequate assessment of the safety and effectiveness of a drug includes a
`description of its metabolism and the contribution of metabolism to overall elimination. For
`this reason, the development of sensitive and specific assays for a drug and its important
`metabolites is critical to the study of metabolism and drug-drug interactions.
`
`B.
`
`
`
`
`
`
`40
`41
`42
`43
`44
`45
`46
`47
`48
`49
`50
`51
`52
`53
`54
`55
`56
`57
`58
`59
`60
`61
`62
`63
`64
`65
`66
`67
`68
`69
`70
`71
`72
`73
`74
`75
`76
`77
`78
`79
`80
`81
`82
`83
`
`Drug-Drug Interactions
`
`1.
`
`Metabolism-Based Drug-Drug Interactions
`
`Many metabolic routes of elimination, including most of those occurring through the
`P450 family of enzymes, can be inhibited or induced by concomitant drug treatment.
`Observed changes arising from metabolic drug-drug interactions can be substantial —
`an order of magnitude or more decrease or increase in the blood and tissue
`concentrations of a drug or metabolite — and can include formation of toxic and/or
`
`G:\6695dft.doc
`09/08/06
`
`2
`
`5
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`84
`85
`86
`87
`88
`89
`90
`91
`92
`93
`94
`95
`96
`97
`98
`99
`100
`101
`102
`103
`104
`105
`106
`107
`108
`109
`110
`111
`112
`113
`114
`115
`116
`117
`118
`119
`120
`121
`122
`123
`124
`125
`126
`127
`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`active metabolites or increased exposure to a toxic parent compound. These large
`changes in exposure can alter the safety and efficacy profile of a drug and/or its
`active metabolites in important ways. This is most obvious and expected for a drug
`with a narrow therapeutic range (NTR), but is also possible for non-NTR drugs as
`well (e.g., HMG CoA reductase inhibitors).
`
`It is important that metabolic drug-drug interaction studies explore whether an
`investigational agent is likely to significantly affect the metabolic elimination of
`drugs already in the marketplace and likely in medical practice to be taken
`concomitantly and, conversely, whether drugs in the marketplace are likely to affect
`the metabolic elimination of the investigational drug. Even drugs that are not
`substantially metabolized can have important effects on the metabolism of
`concomitant drugs. For this reason, metabolic drug-drug interactions should be
`explored, even for an investigational compound that is not eliminated significantly by
`metabolism.
`
`Classical biotransformation studies are not a general requirement for the evaluation of
`therapeutic biologics (ICH guidance S6 Preclinical Safety Evaluation of
`Biotechnology-Derived Pharmaceuticals), although certain protein therapeutics
`modify the metabolism of drugs that are metabolized by the P450 enzymes. Type I
`interferons, for example, inhibit CYP1A2 production at the transcriptional and post-
`translational levels, inhibiting clearance of theophylline. The increased clinical use
`of therapeutic proteins may raise concerns regarding the potential for their impacts on
`drug metabolism. Generally, these interactions cannot be detected by in vitro
`assessment. Consultation with FDA is appropriate before initiating metabolic drug-
`drug interaction studies involving biologics.
`
`Identifying metabolic differences in patient groups based on genetic polymorphism,
`or on other readily identifiable factors, such as age, race, and gender, can aid in
`interpreting results. The extent of interactions may be defined by these variables
`(e.g., CYP2D6 genotypes). Further, in subjects who lack the major clearance
`pathway, remaining pathways become important and should be understood and
`examined.
`
`A specific objective of metabolic drug-drug interaction studies is to determine
`whether the interaction is sufficiently large to necessitate a dosage adjustment of the
`drug itself or the drugs with which it might be used, or whether the interaction would
`require additional therapeutic monitoring.
`
`In some instances, understanding how to adjust dose or dosage regimen in the
`presence of an interacting drug, or how to avoid interactions, may allow marketing of
`a drug that would otherwise have been associated with an unacceptable level of
`toxicity. Sometimes a drug interaction can be used intentionally to increase levels or
`reduce elimination of another drug (e.g., ritonavir and lopinavir). Rarely, the degree
`
`G:\6695dft.doc
`09/08/06
`
`3
`
`6
`
`

`

`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`of interaction caused by a drug, or the degree to which other drugs alter its
`metabolism, can be such that it cannot be marketed safely.
`
`Transporter-Based Drug-Drug Interactions
`2.
`Transporter-based interactions have been increasingly documented. Examples of
`these include the inhibition or induction of transport proteins, such as P-glycoprotein
`(P-gp), organic anion transporter (OAT), organic anion transporting polypeptide
`(OATP), organic cation transporter (OCT), multidrug resistance-associated proteins
`(MRP), and breast cancer resistant protein (BCRP). Examples of transporter-based
`interactions include the interactions between digoxin and quinidine, fexofenadine and
`ketoconazole (or erythromycin), penicillin and probenecid, and dofetilide and
`cimetidine. Of the various transporters, P-gp is the most well understood and may be
`appropriate to evaluate during drug development. Table 1 in Appendix A lists some
`of the major human transporters and known substrates, inhibitors, and inducers.
`
`
`
`III. GENERAL STRATEGIES
`
`To the extent possible, drug development should follow a sequence in which early in vitro
`and in vivo investigations can either fully address a question of interest or provide
`information to guide further studies. Optimally, a sequence of studies could be planned,
`moving from in vitro studies to in vivo human studies, including those employing special
`study designs and methodologies where appropriate. In many cases, negative findings from
`early in vitro and early clinical studies can eliminate the need for later clinical investigations.
`Early investigations should explore whether a drug is eliminated primarily by excretion or
`metabolism, with identification of the principal metabolic routes in the latter case. Using
`suitable in vitro probes and careful selection of interacting drugs for early in vivo studies, the
`potential for drug-drug interactions can be studied early in the development process, with
`further study of observed interactions assessed later in the process, as needed. These early
`studies can also provide information about dose, concentration, and response relationships in
`the general population, specific populations, and individuals, which can be useful in
`interpreting the consequences of a drug-drug interaction. Once potential drug-drug
`interactions have been identified, based on in vitro and/or in vivo studies, sponsors are
`encouraged to design and examine the safety and efficacy databases of larger clinical studies,
`as feasible, to (1) permit confirmation/discovery of the interactions predicted from earlier
`studies and/or (2) verify that dosage adjustments or other prescribing modifications made in
`response to the potential interaction(s) have been adequate to avoid undesired consequences
`of the drug-drug interaction.
`
`A.
`
`A complete understanding of the quantitative relationship between the in vitro findings and
`in vivo results of metabolism/drug-drug interaction studies is still emerging. Nonetheless, in
`vitro studies can frequently serve as a screening mechanism to rule out the importance of a
`
`In Vitro Studies
`
`
`
`
`
`
`128
`129
`130
`131
`132
`133
`134
`135
`136
`137
`138
`139
`140
`141
`142
`143
`144
`145
`146
`147
`148
`149
`150
`151
`152
`153
`154
`155
`156
`157
`158
`159
`160
`161
`162
`163
`164
`165
`166
`167
`168
`169
`170
`171
`
`G:\6695dft.doc
`09/08/06
`
`4
`
`7
`
`

`

`
`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`metabolic pathway and the drug-drug interactions that occur through this pathway so that
`subsequent in vivo testing is unnecessary. This opportunity should be based on appropriately
`validated experimental methods and rational selection of substrate/interacting drug
`concentrations.
`
`For example, if suitable in vitro studies at therapeutic concentrations indicate that CYP1A2,
`CYP2C8, CYP2C9, CYP2C19, CYP2D6, or CYP3A enzyme systems do not metabolize an
`investigational drug, then clinical studies to evaluate the effect of CYP2D6 inhibitors or
`CYP1A2, CYP2C8, CYP2C9, CYP2C19, or CYP3A inhibitors/inducers on the elimination
`of the investigational drug will not be needed.
`
`Similarly, if in vitro studies indicate that an investigational drug does not inhibit CYP1A2,
`CYP2C8, CYP2C9, CYP2C19, CYP2D6, or CYP3A metabolism, then corresponding in vivo
`inhibition-based interaction studies of the investigational drug and concomitant medications
`eliminated by these pathways are not needed. Figure 1 in Appendix B shows a decision tree
`on when in vivo interaction studies are indicated based on in vitro metabolism, inhibition,
`and induction and in vivo metabolism data.
`
`The CYP2D6 enzyme has not been shown to be inducible. Recent data have shown co-
`induction of CYP2C, CYP2B and ABCB1 (P-gp) transporter with CYP3A. CYP3A appears
`to be sensitive to all known co-inducers. Therefore, to evaluate whether an investigational
`drug induces CYP1A2, CYP2C8, CYP2C9, CYP2C19, or CYP3A, the initial in vitro
`induction evaluation may include only CYP1A2 and CYP3A. If in vitro studies indicate that
`an investigational drug does not induce CYP3A metabolism, then in vivo induction-based
`interaction studies of the investigational drug and concomitant medications eliminated by
`CYP2C/CYP2B and CYP3A may not be needed.
`
`Drug interactions based on CYP2B6 are emerging as important interactions. When
`appropriate, in vitro evaluations based on this enzyme can be conducted. Other CYP
`enzymes, including CYP2A6 and CYP2E1, are less likely to be involved in clinically
`important drug interactions, but should be considered when appropriate.
`
`Appendix C describes general considerations in the in vitro evaluation of CYP-related
`metabolism and interactions. Appendices C-1, C-2, and C-3 provide considerations in the
`experimental design, data analysis, and data interpretation in drug metabolizing enzyme
`identification, including CYP enzymes (new drug as a substrate), CYP inhibition (new drug
`as an inhibitor), and CYP induction (new drug as an inducer), respectively. Appendix D
`describes general considerations in the in vitro evaluation of P-gp substrates and inhibitors.
`Figures 1 and 2 in Appendix D provide decision trees on when in vivo P-gp based interaction
`studies are indicated based on in vitro evaluation.
`
`B.
`
`In addition to in vitro metabolism and drug-drug interaction studies, appropriately designed
`
`Specific In Vivo Clinical Investigations
`
`172
`173
`174
`175
`176
`177
`178
`179
`180
`181
`182
`183
`184
`185
`186
`187
`188
`189
`190
`191
`192
`193
`194
`195
`196
`197
`198
`199
`200
`201
`202
`203
`204
`205
`206
`207
`208
`209
`210
`211
`212
`213
`214
`215
`
`G:\6695dft.doc
`09/08/06
`
`5
`
`8
`
`

`

`
`
`Contains Nonbinding Recommendations
`
`Population Pharmacokinetic Screens
`
`Draft – Not for Implementation
`
`pharmacokinetic studies, usually performed in the early phases of drug development, can
`provide important information about metabolic routes of elimination, their contribution to
`overall elimination, and metabolic drug-drug interactions. Together with information from in
`vitro studies, these in vivo investigations can be a primary basis of labeling statements and
`can often help avoid the need for further investigations. Further recommendations about
`these types of studies appear in section IV of this guidance.
`
`C.
`
`Population pharmacokinetic analyses of data obtained from large-scale clinical studies with
`sparse or intensive blood sampling can be valuable in characterizing the clinical impact of
`known or newly identified interactions, and in making recommendations for dosage
`modifications. The results from such analyses can be informative and sometimes conclusive
`when the clinical studies are adequately designed to detect significant changes in drug
`exposure due to drug-drug interactions. Simulations can provide valuable insights into
`optimizing the study design. Population pharmacokinetic evaluations may detect
`unsuspected drug-drug interactions. Population analysis can also provide further evidence of
`the absence of a drug-drug interaction when this is supported by prior evidence and
`mechanistic data. However, it is unlikely that population analysis can be used to prove the
`absence of an interaction that is strongly suggested by information arising from in vivo
`studies specifically designed to assess a drug-drug interaction. To be optimally informative,
`population pharmacokinetic studies should have carefully designed study procedures and
`sample collections. A guidance for industry on population pharmacokinetics is available
`(Ref. 11).
`
`IV. DESIGN OF IN VIVO DRUG-DRUG INTERACTION STUDIES
`
`If in vitro studies and other information suggest that in vivo drug-drug interaction studies
`would be helpful (e.g., based on Figure 1 in Appendix B), the following general issues and
`approaches should be considered. Consultation with FDA regarding study protocols is
`recommended. In the following discussion, the term substrate (S) is used to indicate the
`drug studied to determine whether its exposure is changed by another drug, termed the
`interacting drug (I). Depending on the study objectives, the substrate and the interacting
`drug can be the investigational agents or approved products.
`
`A.
`
`In vivo drug-drug interaction studies generally are designed to compare substrate
`concentrations with and without the interacting drug. Because a specific study can consider
`a number of questions and clinical objectives, many study designs for studying drug-drug
`interactions can be considered. A study can use a randomized crossover (e.g., S followed by
`S+I, S+I followed by S), a one-sequence crossover (e.g., S always followed by S+I or the
`reverse), or a parallel design (S in one group of subjects and S+I in another). The following
`possible dosing regimen combinations for a substrate and interacting drug can also be used:
`
`Study Design
`
`216
`217
`218
`219
`220
`221
`222
`223
`224
`225
`226
`227
`228
`229
`230
`231
`232
`233
`234
`235
`236
`237
`238
`239
`240
`241
`242
`243
`244
`245
`246
`247
`248
`249
`250
`251
`252
`253
`254
`255
`256
`257
`258
`259
`
`G:\6695dft.doc
`09/08/06
`
`6
`
`9
`
`

`

`Contains Nonbinding Recommendations
`
`
`
`
`•
`
`
`•
`
`
`•
`
`
`•
`
`260
`261
`262
`263
`264
`265
`266
`267
`268
`269
`270
`271
`272
`273
`274
`275
`276
`277
`278
`279
`280
`281
`282
`283
`284
`285
`286
`287
`288
`289
`290
`291
`292
`293
`294
`295
`296
`297
`298
`299
`300
`301
`302
`303
`
`Draft – Not for Implementation
`
`single dose/single dose, single dose/multiple dose, multiple dose/single dose, and multiple
`dose/multiple dose. The selection of one of these or another study design depends on a
`number of factors for both the substrate and interacting drug, including (1) acute or chronic
`use of the substrate and/or interacting drug; (2) safety considerations, including whether a
`drug is likely to be an NTR (narrow therapeutic range) or non-NTR drug; (3)
`pharmacokinetic and pharmacodynamic characteristics of the substrate and interacting drugs;
`and (4) assessment of induction as well as inhibition. The inhibiting/inducing drugs and the
`substrates should be dosed so that the exposures of both drugs are relevant to their clinical
`use, including the highest doses likely to be used. Simulations can be helpful in selecting an
`appropriate study design. The following considerations may be useful:
`
`•
`
`When attainment of steady state is important and either the substrate or interacting
`drugs and/or their metabolites have long half-lives and a loading dose to reach steady
`state promptly cannot be used, special approaches may be needed. These include the
`selection of a one-sequence crossover or a parallel design, rather than a randomized
`crossover study design.
`
`When it is important that a substrate and/or an interacting drug be studied at steady
`state because the effect of an interacting drug is delayed, as is the case for inducers
`and certain inhibitors, documentation that near steady state has been attained for the
`pertinent drug and metabolites of interest is critical. This documentation can be
`accomplished by sampling over several days prior to the periods when test samples
`are collected. This is important for both metabolites and the parent drug, particularly
`when the half-life of the metabolite is longer than the parent, and is especially
`important if both parent drug and metabolites are metabolic inhibitors or inducers.
`
`Studies can usually be open label (unblinded), unless pharmacodynamic endpoints
`(e.g., adverse events that are subject to bias) are critical to the assessment of the
`interaction.
`
`For a rapidly reversible inhibitor, administration of the interacting drug either just
`before or simultaneously with the substrate on the test day might increase sensitivity.
`For a mechanism-based inhibitor (a drug that requires metabolism prior to its
`inactivation of the enzyme; examples include erythromycin), administration of the
`inhibitor prior to the administration of the substrate drug can maximize the effect. If
`the absorption of an interacting drug (e.g., an inhibitor or an inducer) may be affected
`by other factors (e.g., the gastric pH), it may be appropriate to control the variables
`and confirm the absorption through plasma level measurements of the interacting
`drug.
`
`When the effects of two drugs on one another are of interest, the potential for
`interactions can be evaluated in a single study or two separate studies. Some design
`options are randomized three-period crossover, parallel group, and one-sequence
`crossover.
`
`
`G:\6695dft.doc
`09/08/06
`
`7
`
`10
`
`

`

`
`
`•
`
`
`
`
`
`304
`305
`306
`307
`308
`309
`310
`311
`312
`313
`314
`315
`316
`317
`318
`319
`320
`321
`322
`323
`324
`325
`326
`327
`328
`329
`330
`331
`332
`333
`334
`335
`336
`337
`338
`339
`340
`341
`342
`343
`344
`345
`346
`347
`
`
`B.
`
`Clinical drug-drug interaction studies can generally be performed using healthy volunteers.
`Findings in this population should predict findings in the patient population for which the
`drug is intended. Safety considerations may preclude the use of healthy subjects, however,
`and in certain circumstances, subjects drawn from the population of patients for whom the
`investigational drug is intended offer advantages, including the opportunity to study
`pharmacodynamic endpoints not present in healthy subjects. Performance of phenotype or
`genotype determinations to identify genetically determined metabolic polymorphisms is
`important in evaluating effects on enzymes with polymorphisms, notably CYP2D6,
`CYP2C19, and CYP2C9. The extent of drug interactions (inhibition or induction) may be
`different depending on the subjects’ genotype for the specific enzyme being evaluated.
`Subjects lacking the major clearance pathway, for example, cannot show metabolism and
`remaining pathways can become important and should be understood and examined.
`
`C.
`
`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`
`
`To avoid variable study results because of uncontrolled use of dietary supplements,
`juices, or other foods that may affect various metabolizing enzymes and transporters
`during in vivo studies, it is important to exclude their use when appropriate.
`
`Examples of statements in a study protocol could include “Participants will be
`excluded for the following reasons: Use of prescription or over-the-counter
`medications, including herbal products, or alcohol within two weeks prior to
`enrollment,” “For at least two weeks prior to the start of the study until its conclusion,
`volunteers will not be allowed to eat any food or drink any beverage containing
`alcohol, grapefruit or grapefruit juice, apple or orange juice, vegetables from the
`mustard green family (e.g., kale, broccoli, watercress, collard greens, kohlrabi,
`brussels sprouts, mustard) and charbroiled meats.”
`
`Study Population
`
`Choice of Substrate and Interacting Drugs
`
`1.
`
`Investigational Drug as an Inhibitor or an Inducer of CYP Enzymes
`
`In contrast to earlier approaches that focused mainly on a specific group of approved
`drugs (digoxin, hydrochlorothiazide) where co-administration was likely or the
`clinical consequences of an interaction were of concern, improved understanding of
`the mechanistic basis of metabolic drug-drug interactions enables more general
`approaches to and conclusions from specific drug-drug interaction studies. In
`studying an investigational drug as the interacting drug, the choice of substrates
`(approved drugs) for initial in vivo studies depends on the P450 enzymes affected by
`the interacting drug. In testing inhibition, the substrate selected should generally be
`one whose pharmacokinetics are markedly altered by co-administration of known
`specific inhibitors of the enzyme systems to assess the impact of the interacting
`investigational drug. Examples of substrates include (1) midazolam for CYP3A; (2)
`
`G:\6695dft.doc
`0

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


Or .

Accessing this document will incur an additional charge of $.

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

Accept $ Charge
throbber

Still Working On It

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

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

throbber

A few More Minutes ... Still Working

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

Thank you for your continued patience.

This document could not be displayed.

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

Your account does not support viewing this document.

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

Your account does not support viewing this document.

Set your membership status to view this document.

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

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

Become a Member

One Moment Please

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

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

Your document is on its way!

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

Sealed Document

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

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


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket