throbber
Guidance for Industry
`
`
`Drug Interaction Studies —
`
`Study Design, Data Analysis, Implications
`
`for Dosing, and Labeling
`
`Recommendations
`
`
`
`
`
`
`DRAFT GUIDANCE
`
`This guidance document is being distributed for comment purposes only.
`
`
`
`U.S. Department of Health and Human Services
`
`Food and Drug Administration
`
`Center for Drug Evaluation and Research (CDER)
`
`
`
`
`February 2012
`
`Clinical Pharmacology
`
`
`
`
`
`
`
`
`Comments and suggestions regarding this draft document should be submitted within 90 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 Lei Zhang, 301-796-1635.
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 1 of 79
`
`

`
`
`
`
`
`
`
`Guidance for Industry
`
`Drug Interaction Studies —
`
`Study Design, Data Analysis,
`
`Implications for Dosing, and Labeling
`
`Recommendations
`
`
`
`Additional copies are available from:
`
`
`Office of Communications
`
`Division of Drug Information, WO51, Room 2201
`
`Center for Drug Evaluation and Research
`
`Food and Drug Administration
`
`10903 New Hampshire Avenue
`
`Silver Spring, MD 20993-0002
`
`Phone: 301-796-3400; Fax: 301-847-8714
`
`
`druginfo@fda.hhs.gov
`
`
`
`http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/default.htm
`
`U.S. Department of Health and Human Services
`
`Food and Drug Administration
`
`Center for Drug Evaluation and Research (CDER)
`
`
`
`
`February 2012
`
`Clinical Pharmacology
`
`
`
`
`
`
`
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 2 of 79
`
`

`
`
`
`TABLE OF CONTENTS
`
`
`
`
`
`
`LIST OF FIGURES ................................................................................................................ 2
`
`
`
`
`LIST OF TABLES ..................................................................................................................2
`
`
`
`
` I.
`
`INTRODUCTION.......................................................................................................... 1
`
`
`
`II. SUMMARY OF GUIDANCE....................................................................................... 2
`
`
`
`
`III. BACKGROUND ............................................................................................................ 6
`
`
`
`A. Relevance of Drug Interactions .................................................................................... 6
`
`B. Drug Interaction Considerations for Small Molecule Drugs........................................ 7
`
`C. Drug Interaction Considerations for Therapeutic Proteins......................................... 14
`
`IV. GENERAL STRATEGIES ......................................................................................... 14
`
`
`
`A.
`In Vitro Studies........................................................................................................... 15
`
`B.
`In Vivo Studies ........................................................................................................... 33
`
`1. In Vivo Drug-Drug Interactions ............................................................................. 33
`
`2. In Vivo Drug-Therapeutic Protein (TP) Interactions ............................................. 33
`
`C. Using a Population Pharmacokinetic Approach to Assess Drug-Drug Interactions .. 36
`
`V. DESIGN OF IN VIVO DRUG-DRUG INTERACTION STUDIES ....................... 36
`
`
`
`A. Study Design............................................................................................................... 36
`
`B. Study Population......................................................................................................... 39
`C. Choice of Substrate and Interacting Drugs................................................................. 39
`
`D. Route of Administration ............................................................................................. 55
`
`
`E. Dose Selection ............................................................................................................ 55
`
`F. Endpoints .................................................................................................................... 56
`
`G. Statistical Considerations and Sample Size................................................................ 57
`
`VI. LABELING RECOMMENDATIONS....................................................................... 58
`
`
`
`A. Drug Interactions Section of Labeling........................................................................ 59
`
`B. Clinical Pharmacology Section of Labeling ............................................................... 60
`
`C. Other Labeling Sections.............................................................................................. 62
`
`LIST OF FIGURES IN THE APPENDIX.......................................................................... 63
`
`
`
`
`
`
`APPENDIX............................................................................................................................ 64
`
`
`REFERENCES......................................................................................................................71
`
`
`ABBREVIATIONS............................................................................................................... 75
`
`
`
`
`
`
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 3 of 79
`
`
`
`

`
`
`
`List of Figures
`
`
`Figure 1. Illustration of Examples of Efflux and Uptake Transporters in the Gut Wall (A), Liver
`
`
`(B), and Kidneys (C) that May Be Involved in a Drug’s Absorption, Distribution, Metabolism,
`
`and Excretion. ................................................................................................................................. 9
`
`Figure 2. Metabolism-Based Drug-Drug Interaction Studies — Decision Tree ......................... 16
`
`Figure 3. Evaluation of Investigational Drugs as UGT Substrates.............................................. 20
`
`Figure 4. General Scheme of Model-Based Prediction: The Investigational Drug (and
`
`Metabolite Present at ≥25% of Parent AUC) as an Interacting Drug of CYP Enzymes .............. 23
`
`Figure 5. Using a PBPK Model to Explore Drug-Drug Interaction Potential Between a Substrate
`
`Drug and an Interacting Drug. ...................................................................................................... 29
`
`Figure 6. Evaluation of Investigational Drugs as Substrates for P-gp, BCRP, OATP1B1,
`
`OATP1B3, OAT1, OAT3, and OCT2 Transporters. .................................................................... 31
`
`Figure 7. Summary of The Types of Studies That Have Been Used During Drug Development to
`
`Evaluate Therapeutic Protein (TP)–Small-Molecule Drug (D) Interactions................................ 35
`
`Figure 8. The Effect of Various CYP Inhibitors on a Hyperthetical Drug’s PK as Displayed as
`
`90% Confidence Interval of Geometric Mean AUC and Cmax Ratios.......................................... 61
`
`
`
`
`List of Tables
`
`Table 1. Selected Transporter-Mediated Clinical Significant Drug-Drug Interactions............... 12
`
`Table 2. In Vitro CYP Inducers ................................................................................................... 27
`
`Table 3. Classification of In Vivo Inhibitors of CYP Enzymes .................................................. 41
`
`Table 4. Classification of In Vivo Inducers of CYP Enzymes .................................................... 43
`
`Table 5. Examples of Sensitive In Vivo CYP Substrates and CYP Substrates with Narrow
`
`Therapeutic Range ........................................................................................................................ 44
`
`Table 6. Examples of In Vivo Inhibitors and Inducers of Selected Transporters........................ 49
`
`
`Table 7. Examples of In Vivo Substrates of Selected Transporters ............................................ 51
`
`Table 8. Examples of In Vivo CYP3A and P-gp Inhibitors and Their Relative Potency............ 53
`
`
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 4 of 79
`
`

`
`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
` Guidance for Industry1
`
`Drug Interaction Studies — Study Design, Data Analysis,
`Implications for Dosing, and Labeling Recommendations
`
`
`
`
`
`
`
`
`
`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 biologics regulated by CDER regarding in
`vitro and in vivo studies of drug metabolism, drug transport, and drug-drug or drug-therapeutic
`protein interactions. Drug interactions can result when one drug alters the pharmacokinetics of
`another drug or its metabolites. Drug interactions also can reflect the additive nature of the
`pharmacodynamic effect of either drug when taken with the other drug. The main focus of this
`guidance is pharmacokinetic drug interactions. This guidance reflects the Agency’s view that
`the pharmacokinetic interactions between an investigational new drug and other drugs should be
`defined during drug development, as part of an adequate assessment of the drug’s safety and
`effectiveness. It is important to understand the nature and magnitude of drug-drug interactions
`(DDI) for several reasons. Concomitant medications, dietary supplements, and some foods, such
`as grapefruit juice, may alter metabolism and/or drug transport abruptly in individuals who
`previously had been receiving and tolerating a particular dose of a drug. Such an abrupt
`alteration in metabolism or transport can change the known safety and efficacy of a drug.
`
`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.
`
`
`
`
`
` 1 This guidance has been prepared by the Drug-Drug Interaction Working Group in the Office of Clinical
`
` Pharmacology, Office of Translational Sciences, in the Center for Drug Evaluation and Research (CDER), with
`
`
`input from other offices in CDER.
`
`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
`
`
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 5 of 79
`
`

`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`SUMMARY OF GUIDANCE
`
`
`
`
`II.
`
`The key recommendations for sponsors to consider when evaluating drug-drug interactions
`during drug development are listed below. The various sections of this guidance provide more
`details for each recommendation.
`
`
`
`
`Interactions between an investigational new drug and other drugs should be defined during
`drug development, as part of an adequate assessment of the drug’s safety and effectiveness.
`The objective of drug-drug interaction studies is to determine whether potential interactions
`between the investigational drug and other drugs exist and, if so, whether the potential for
`such interactions indicates the need for dosage adjustments, additional therapeutic
`monitoring, a contraindication to concomitant use, or other measures to mitigate risk.
`
`
`
` Development of a drug should include identification of the principal routes of elimination,
`quantitation of the contribution by enzymes and transporters to drug disposition, and
`characterization of the mechanism of drug-drug interactions.
`
`
`
` Sponsors who believe a complete evaluation of the potential for drug-drug interactions is not
`necessary for an investigational drug because of the target population and likely co­
`administered drugs should contact the Office of Clinical Pharmacology and the clinical
`division in the Office of New Drugs.
`
`
`
` This guidance and its appendices include numerous decision trees intended to help sponsors
`determine what types of drug-drug interaction studies may be needed (see Figures 2 through
`7 and Appendix Figures A-1 through A-6).
`
`
`
` The study of drug-drug interaction for a new drug generally begins with in vitro studies to
`determine whether a drug is a substrate, inhibitor, or inducer of metabolizing enzymes. The
`results of in vitro studies will inform the nature and extent of in vivo studies that may be
`
` required to assess potential interactions. Along with clinical pharmacokinetic data, results
`from in vitro studies may serve as a screening mechanism to rule out the need for additional
`in vivo studies, or provide a mechanistic basis for proper design of clinical studies using a
`modeling and simulation approach.
`
`
`
` When testing an investigational drug for the possibility that its metabolism is inhibited or
`induced (i.e., as a substrate), selection of the interacting drugs should be based on in vitro or
`in vivo studies identifying the enzyme systems that metabolize the investigational drug. The
`choice of the interacting drug can then be based on known, important inhibitors and inducers
`of the pathway under investigation. Strong inhibitors and inducers provide the most sensitive
`assessment and should generally be tested first (see section V.C).
`
`
`
`
`
`If potential drug-drug interactions are identified based on in vitro and/or in vivo studies,
`sponsors should design further studies or collect information to determine (1) whether
`
`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
`
`
`
`2
`
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 6 of 79
`
`

`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`
`
`additional studies are needed to better quantify the effect and to examine the effects of
`weaker inhibitors (early studies usually examine strong inhibitors) on the investigational
`drugs as substrates and effects of investigational drugs (as inhibitors) on a range of
`substrates, and (2) whether dosage adjustments or other prescribing modifications (e.g.,
`additional safety monitoring or contraindications) are needed based on the identified
`
`interaction(s) to avoid undesired consequences.
`
`
`
` The potential for drug interactions with metabolites of investigational drugs (metabolites
`present at 25% of parent drug AUC) should be considered (see section IV.A.3).
`
`
`
` Metabolic drug-drug interactions should also be explored for investigational drugs that are
`not eliminated significantly by metabolism because such drugs can inhibit or induce a co­
`administered drug’s metabolic pathway (see section IV.A.1).
`
`
`
` When evaluating a new drug as a potential cytochrome P450 (CYP) enzyme inhibitor,
`sponsors should consider a stepwise, model-based evaluation of metabolism-based
`interactions (from basic model for initial assessment to more mechanistic models including
`physiologically-based pharmacokinetic (PBPK) modeling) (see section IV.A.1). The criteria
`used for assessing “equivalence” (e.g., predicted AUC ratio of 0.8-1.25 using population-
`based PBPK models) may be used as an initial cutoff in deciding whether in vivo studies are
`needed. The criteria discussed in this guidance document are suggested values. We are open
`to discussion based on sponsors’ interpretation.
`
`
`
`-
`
`
`
`PBPK is a useful tool that can help sponsors (1) better design drug-drug interaction
`studies, including dedicated trials and population pharmacokinetic studies, and (2)
`quantitatively predict the magnitude of drug-drug interactions in various clinical
`situations. PBPK models also may offer useful alternatives to dedicated clinical
`studies.
`
`
`
`- When submitting PBPK studies to CDER, sponsors should provide details of model
`assumptions, physiological and biological plausibility, the origin of the parameters,
`and information on uncertainty and variability.
`
`
`
` The evaluation of CYP enzyme induction should begin with studies of CYP1A2, CYP2B6,
`and CYP3A in vitro (Figure 4). If the in vitro induction results are positive according to
`predefined thresholds using basic models, the investigational drug is considered an enzyme
`
`inducer and further in vivo evaluation may be warranted. Alternatively, a sponsor can
`estimate the degree of drug-drug interactions using mechanistic models to determine the need
`for further in vivo evaluation (see section IV.A.1.b-3).
`
`
`
`-
`
`
`
`It should be noted that there may be mechanisms of induction that are presently
`unknown. Therefore, a potential human teratogen needs to be studied in vivo for
`effects on contraceptive steroids if the drug is intended for use in fertile women,
`regardless of in vitro induction study results.
`
`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
`
`
`
`3
`
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 7 of 79
`
`

`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`In addition to CYPs, other metabolizing enzymes (e.g., uridine diphosphate (UDP)­
`glucuronosyl transferases (UGTs)) that may be important for the drugs under evaluation
`should also be considered (see section IV.A.1).
`
`
`
` A number of transporter-based interactions have been documented in recent years (see Table
`1, section III.B.2).
`
`
`
`
`
`
`
`
`
`
`
`- All investigational drugs should be evaluated in vitro to determine whether they are a
`potential substrate of P-glycoprotein (P-gp) or Breast Cancer Resistance Protein
`(BCRP) (see Figure 6, left panel, section IV.A.2). Investigational drugs should be
`evaluated in vitro to determine whether they are a substrate of hepatic uptake
`transporters Organic Anion Transporting Polypeptide 1B1(OATP1B1) or OATP1B3
`when their hepatic pathway is significant (see Figure 6, middle panel, section
`IV.A.2). Similarly, investigational drugs should be evaluated in vitro to determine
`whether they are a substrate of Organic Aniton Transporter 1 (OAT1) or OAT3 or
`Organic Cation Transporter 2 (OCT2) when renal active secretion is important
`(Figure 6, right panel, section IV.A.2).
`
`
`
`- Because there have been clinically significant interactions demonstrated for critical
`drugs that are known substrates for P-gp (e.g., digoxin), BCRP (e.g., rosuvastatin),
`OATP1B1/OATP1B3 (e.g., statin drugs), OAT1/OAT3 (e.g., methotrexate, tenofovir)
`and OCT2 (e.g., metformin), evaluation of investigational drugs as inhibitors for
`these transporters should be conducted (see section IV.A.2).
`
`
`- The need for further in vivo drug interaction studies based on in vitro evaluation will
`be based on the criteria described in the decision trees in Figures A1-A6 in the
`Appendix.
`
`
`
` Because of the lack of a validated in vitro system to study transporter induction, the
`definitive determination of induction potential of an investigational drug on transporters is
`based on in vivo induction studies. The sponsor should consult with CDER about studying
`induction of transporters in vivo.
`
`
`
` Human clinical studies to assess drug-drug interactions may include simultaneous
`administration of a mixture of substrates of multiple CYP enzymes and transporters in one
`study (i.e., a “cocktail approach”) to evaluate a drug’s inhibition or induction potential (see
`section V.C.3). Negative results from a well-conducted cocktail study may eliminate the
`need for further evaluation of particular CYP enzymes and transporters. However, positive
`results may indicate that further in vivo evaluation should be conducted.
`
`
`
`
` The potential for interactions with drug products should be considered for certain classes of
`therapeutic proteins (TPs) (see Figure 7, section IV.B.2).
`
`
`
`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
`
`
`
`4
`
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 8 of 79
`
`

`
`
`
`-
`
`
`
`
`-
`
`
`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`If an investigational TP is a cytokine or cytokine modulator, studies should be
`conducted to determine the TP’s effects on CYP enzymes or transporters. The in
`vivo evaluations of TPs in targeted patient populations can be conducted with
`
`individual substrates for specific CYP enzymes and transporters, or studies can be
`
`conducted using a “cocktail approach” (see section V.C.3).
`
`For TPs that will be used in combination with other drug products (small molecule or
`TP) as a combination therapy, studies should evaluate the effect of each product on
`the other. This evaluation is particularly important when the drug used in
`combination has a narrow therapeutic range.
`
`
`
`
`- When there are known mechanisms or prior experience with certain PK or PD
`interactions for other similar TPs, appropriate in vitro or in vivo assessments for
`possible interactions should be conducted.
`
`
`
`
` Refer to section V for information regarding in vivo drug interaction study design. The
`section also contains tables on classification of in vivo inhibitors (Table 3) or inducers for
`CYP enzymes (Table 4), examples of sensitive in vivo CYP substrates and CYP substrates
`with narrow therapeutic ranges (Table 5), examples of in vivo inhibitors and inducers of
`selected transporters (Table 6), examples of in vivo substrates of selected transporters (Table
`7) and examples of in vivo CYP3A and P-gp inhibitors and their relative potency (Table 8).
`
`
`
`
`
`-
`
`
`
`Simulations (e.g., by population-based PBPK models) can provide valuable insight
`into optimizing the study design (see section IV.A.1).
`
`
`
`- Detailed information on the dose given and time of administration should be
`documented for the co-administered drugs. When relevant for the specific drug, the
`time of food consumption should be documented.
`
`-
`
`
`
`Population pharmacokinetic (PopPK) analyses of data obtained from large-scale
`clinical studies that include sparse or intensive blood sampling can help characterize
`the clinical impact of known or newly identified interactions and determine
`recommendations for dosage modifications for the investigational drug as a substrate
`(section V.B). DDI analyses using a population PK approach should focus on
`excluding a specific clinically meaningful PK change. Because exposure of co­
`administered drugs is not monitored in most PopPK studies, the PopPK approach may
`not be useful to assess the effect of the investigational drugs on other drugs.
`
`
`
` The likelihood of drug interactions in specific populations (e.g., patients with organ
`impairment, and pediatric and geriatric patients) should be considered on a case-by-case
`basis. PBPK modeling (if well verified for intended purposes) can be helpful to guide the
`determination of the need to conduct population-specific studies (see “Populations” in
`section V.B and “Complex Drug Interactions” section V.C.4).
`
`
`
`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
`
`
`
`5
`
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 9 of 79
`
`

`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`
`
` Additional study design issues are discussed throughout the guidance (e.g., route of
`administration (section V.D), dose selection (section V.E), defining endpoints (section V.F),
`and statistical considerations (section V.G)).
`
`
`
` Labeling recommendations with regard to drug interactions are described in section VI.
`
`
`
`- A forest plot is considered a useful tool for presenting changes in pharmacokinetic
`exposure measures by various intrinsic and extrinsic factors including drug
`interactions in the PHARMACOKINETIC subsection of the labeling (see Figure 8,
`section VI).
`
`
`
`-
`
`
`
`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
`
`III. BACKGROUND
`247
`
`248
`
`249 A.
`
`250
`The desirable and undesirable effects of a drug are related to its concentration at various sites of
`251
`action, which is usually related to the blood or tissue concentration of the drug. The blood or
`252
`tissue concentrations resulting from a dose are determined by the drug’s absorption, distribution,
`253
`254 metabolism, and excretion (ADME). Elimination of a drug or its active metabolites occurs either
`by metabolism to an inactive metabolite that is excreted, or by direct excretion of the drug or
`255
`active metabolites. The kidneys and liver are responsible for most drug excretion. Drug
`256
`interactions related to metabolism and excretion are well-recognized, but effects related to
`257
`transporters are being documented with increasing frequency and are, therefore, important to
`258
`consider in drug development. Therapeutic proteins may be eliminated through a specific
`259
`
`If the sponsor wishes to include a statement in the labeling that no known drug-drug
`interaction of clinical significance exists, the sponsor should recommend specific no
`effect boundaries, or clinical equivalence intervals, for a drug-drug interaction and
`should provide the scientific justification for the recommendations. No effect
`boundaries represent the interval within which a change in a systemic exposure
`measure is considered not clinically meaningful. These conclusions can be based on
`exposure-response or dose-response data.
`
`
`
` Sponsors are encouraged to communicate with the Office of Clinical Pharmacology or the
`appropriate clinical review divisions within CDER regarding questions about drug
`interactions, in particular when
`
`
`
`- Using mechanistic or PBPK models for the prediction of drug-drug interactions
`including evaluation of complex drug-drug interactions
`- Determining the need to evaluate drug interactions with non-CYP enzymes or
`additional transporters that are not included in the decision trees
`- Determining drug-drug interaction studies involving TPs.
`
`Relevance of Drug Interactions
`
`
`
`
`
`
`
`6
`
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 10 of 79
`
`

`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`The overall objective of interaction studies for a new drug is to determine:
`
`
`interaction with cell surface receptors, followed by internalization and lysosomal degradation
`260
`261 within the target cell.
`262
`263
`264
`265
`266
`267
`268
`269
`270
`In some instances, understanding how to adjust a dose or dosage regimen in the presence of an
`271
`interacting drug, or how to avoid drug-drug interactions, may allow marketing of a drug that
`272
`273 would otherwise have an unacceptable level of risk. In a few cases, consequences of an
`interaction have led to the conclusion that the drug could not be marketed safely. In almost all of
`274
`these cases, that conclusion was strengthened by the availability of alternative drugs with lower
`275
`risks for interactions. Several drugs have been withdrawn from the market because of significant
`276
`drug interactions that led to QT prolongation and Torsades de Pointes (TdP) arrhythmias, after
`277
`278 warnings in drug labels did not adequately manage the risk of drug interactions. For example,
`terfenadine and astemizole, two early nonsedating antihistamines metabolized by CYP3A, were
`279
`280 withdrawn after labeling failed to reduce cases of TdP sufficiently, because fexofenadine and
`loratadine fulfilled the need for nonsedating antihistamines that had no risk of TdP. Cisapride, a
`281
`282 CYP3A metabolized drug, was withdrawn because its gastrointestinal benefits were not felt to
`outweigh its TdP risk. A fourth drug, mibefradil (a calcium channel blocker similar to verapamil
`283
`and diltiazem) was a strong CYP3A inhibitor and, when used with simvastatin, caused
`284
`rhabdomyolysis because of markedly increased simvastatin exposure.
`285
`286
`287
`
`288 B.
`Drug Interaction Considerations for Small Molecule Drugs
`
`289
`The main focus of this guidance is pharmacokinetic drug interactions. The drug development
`290
`process should include evaluation of a new drug’s potential to affect the metabolism or transport
`291
`of other drugs and the potential for the new drug’s metabolism or transport to be affected by
`292
`other drugs. Use of in vitro tools to determine whether a drug is a substrate, inhibitor, or inducer
`293
`of metabolizing enzymes, followed by in vivo interaction studies to assess potential interactions,
`294
`has become an integral part of drug development and regulatory review. In addition to the
`295
`evaluation of metabolic drug interactions, the role of transporters in drug interactions should be
`296
`evaluated. This section will separately discuss drug-drug interactions at the levels of
`
`297
`298 metabolizing enzymes and transporters, and also consider situations when multiple drug-drug
`interaction mechanisms are present.
`299
`
`300
`1.
`301
`302
`
`
`
` whether any interactions are sufficiently large to necessitate a dosage adjustment of the
`drug itself or of the drugs with which it might be used,
` whether any interactions calls for additional therapeutic monitoring, or
` whether there should be a contraindication to concomitant use when lesser measures
`cannot mitigate risk.
`
`Metabolism-Based Drug-Drug Interactions
`
`
`
`
`
`
`
`
`
`
`
`
`7
`
`
`PAR1011
`IPR of U.S. Patent No. 8,772,306
`Page 11 of 79
`
`

`
`Contains Nonbinding Recommendations
`
`Draft – Not for Implementation
`
`Hepatic metabolism occurs primarily through the cytochrome P450 family (CYP) of
`enzymes located in the hepatic endoplasmic reticulum, but may also occur through non­
`CYP enzyme systems, such as glucuronosyl- and sulfo-transferases, which can, in
`general, inactivate a drug and increase its renal elimination. Some drug metabolizing
`enzymes are present in the gut wall and other extrahepatic tissues, in addition to the liver.
`
`Many metabolic routes of elimination can be inhibited or induced by concomitant drug
`treatment. Metabolic drug-drug interactions can cause substantial changes — an order of
`magnitude or more decrease or increase in the blood and tissue concentrations of a drug
`or metabolite — and can affect the extent to which toxic or active metabolites are
`formed. These large changes in exposure can alter the safety and efficacy profile of a
`drug and its active metabolites, regardless of whether the drug has a narrow therapeutic
`range (NTR). For example, certain HMG-CoA reductase inhibitors (e.g., lovastatin,
`simvastatin) that are extensively metabolized by CYP3A can have a 10-fold or more
`increase in blood levels when their metabolism is inhibited by co-administration with
`strong CYP3A inhibitors such as mibefradil or ketoconazole, or even moderate inhibitors
`such as erythromycin. Although the HMG-CoA reductase inhibitors are not NTR drugs,
`the blood level increases caused by interactions between HMG-CoA reductase inhibitors
`and CYP3A inhibitors can cause myopathy and in some cases rare and life-threatening
`
`rhabdomyolysis.
`
`In addition to evaluating a drug as a substrate of an enzyme that another drug may inhibit
`or induce, it is important to determine whether an investigational drug significantly
`affects the metabolic elimination of

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