throbber
CENTER FOR DRUG EVALUATION AND
`
`RESEARCH
`
`APPLICA TION NUMBER:
`2 1 -976
`
`MICROBIOLOGY REVIEW
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`NDA: 21—976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`
`NDA#: 21—976
`
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`Serial #: 000
`
`Reviewer's Name:
`
`Lisa K. Naeger, Ph.D.
`
`Sponsor’s Name and Address:
`
`Tibotec—Virco, USA
`
`2505 Meridian Parkway
`Suite 350
`
`Durham, NC 27713
`
`Initial Submission Dates:
`
`Correspondence Date:
`CDER Receipt Date:
`Assigned Date:
`Review Start Date:
`
`December 23, 2005
`December 23, 2005
`December 23, 2005
`October 15, 2005
`
`Review Complete Date:
`PDUFA Date:
`
`June 14, 2006
`June 23, 2005
`
`Amendments:
`
`Related/Supporting Documents: IND62477
`
`Product N ame(s)
`Proprietary: PREZISTA/rtv
`Non-Proprietary/USAN: Darunavir/rtv; darunavir, TMC114
`Code Name/Number:
`'
`
`Empirical formula: C27H37N3O7S C2H5OH
`Chemical Name: {3—[(4—amino-benzenesu1fony1)—isobuty1—amino]— 1 —benzyl—2-hydroxy—
`propyl} —carbamic acidhexahydro—furo-[2,3-b]furan—3—yl
`ester.ethanolate
`
`Molecular mass: Relative molecular mass: 547.656 (active moiety) + 46.068 (ethanol,
`EtOH) = 593.724 (TMC 11 4-ethano late)
`
`Structural Formula:
`
`'
`
`NH2
`
`on (AIR /u\
`(\uu...»
`I’I-O
`
`R O
`I.
`
`s
`
`0
`
`O
`
`\S/O/
`/ \\
`O
`
`N
`H S
`
`R
`
`N
`
`Darunavir
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`NDA: 21—976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`Drug category: antiviral for HIV infection
`Dosage Form(s): Oral; co—administratz'on ofrz'tonavir as 100—mg soft gelatin capsules
`Route(s) of Administration: Oral
`Indication(s): Combination antiretroviral treatment of HIV—1 infected adult subjects with
`. evidence of viral replication who are heavily treatment—experienced or have HIV-1
`strains resistant to multiple protease inhibitors.
`
`Dispensedsz X
`
`OTC
`
`Abbreviations: ABC, abacavir, APV, amprenavir; ATV, atazanavir; AZT, zidovudine;
`Control, comparator Pl arm; ddl, didanosine; d4T, stavudine; DLV, delavirdine; EFV, efavirenz;
`FTC,
`emtricitabine; HAART,
`highly
`active
`antiretroviral
`therapy; HIV-1,
`human
`immunodeficiency virus-1; 1C, inhibitory concentration; IDV, indinavir; LAM, lamivudine; LPV,
`lopinavir; NFV, nelfinavir; NVP, nevirapine; NNRTI, non-nucleoside reverse transcriptase '
`inhibitor; NRTI, nucleoside reverse transcriptase inhibitor; OBT, optimized background therapy;
`PBMC, peripheral blood mononuclear cells; PCR, polymerase chain reaction; Pl, protease
`inhibitor; /rtv, ritonavir-boosted; RT, reverse transcriptase; SQV, saquinavir; ENF, enfuvirtide;
`TNF, tenofovir; TPV, tipranavir
`
`
`
`EXECUTIVE SUMMARY
`
`It selectively inhibits the cleavage of
`Darunavir is an inhibitor of the HIV—1 protease.
`HIV encoded Gag—Pol polyproteins in infected cells, thereby preventing the formation of
`mature Virus particles. Darunavir exhibits activity against laboratory strains and clinical
`isolates of HIV—1 and laboratory strains of HIV—2 in acutely infected T—cell lines, human
`peripheral blood mononuclear cells and human monocytes/macrophages with median
`EC50 values ranging from 1.2 to 8.5 nM (0.7 to 5.0 ng/ml). Darunavir demonstrates
`antiviral activity in cell culture against a broad panel of HIV—1 group M (A, B, C, D, E, F,
`G), and group O'primary isolates with EC50 values ranging from < 0.1 to 4.3 nM. The
`EC5'0 value of darunavir increases by a median factor of 5.4 in the presence of human
`serum. Darunavir did not show antagonism when studied in combination with the
`protease inhibitors amprenavir, atazanavir,
`indinavir,‘ lopinavir, 'nelfinavir, ritonavir,
`saquinavir,or tipranavir, the N(t)RTls abacavir, didanosine, emtricitabine,, lamivudine,
`stavudine, tenofovir, zalcitabine, or zidovudine, the NNRTIs delavirdine, efavirenz , or
`nevirapine, and the fusion inhibitor enfuvirtide.
`
`Resistance
`
`Cell Culture: HIV—1 isolates with a decreased susceptibility to darunavir have been
`selected in cell culture and obtained from subjects treated with darunavir/ritonavir.
`Darunavir—resistant Virus derived in cell culture from wild—type HIV had 6- to 21—fold
`decreased susceptibility to darunavir and harbored 3 to 6 of the following amino acid
`substitutions S37N/D, R41E/S/T, K55Q, K70E, A71T, T74S, V771, or I85V in the
`protease. Selection in cell culture of darunavir resistant HIV—1 from nine HIV—1 strains
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`NDA: 21—976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`harboring multiple protease inhibitor resistance-associated mutations resulted in the
`overall emergence of 22 mutations in the protease gene, including LIOF, V111, 113V,
`115V, G16E, L231, V321, L33F, S37N, M461, 147V, 150V, F53L, L63P, A71V, G73S,
`L76V, V821, 184V, T9lA/S, and Q92R, of which LIOF, V321, L33F, S37N, M461, 147V,
`150V, L63P, A71V, and I84V were the most prevalent. These darunavir—resistant viruses
`had at least eight protease mutations and exhibited 50— to 64] -fold decreases in darunavir
`susceptibility with final EC50 values ranging from 125 nM to 3461 nM
`
`
`In the Phase 2
`Clinical studies of darunavir/ritonavir in treatment-emnced subiects:
`trials Studies C202, C213 and C215, multiple protease inhibitor—resistant HIV-l isolates
`from highly treatment—experienced subjects who received 600/100 mg darunavir/rtv b.i.d.
`and experienced virologic failure, either by rebound, or by never being suppressed,
`developed amino acid substitutions that were associated with a decrease in susceptibility
`to darunavir. The amino acid substitution V321 developed on darunavir/rtv 600/ 100 mg
`b.i.d. in greater than 30% of virologic failure isolates and substitutions at amino acid 154
`developed in greater than 20% of virologic failure isolates. Other substitutions that
`developed in 10% to 20% of darunavir/rtv virologic failure isolates occurred at amino
`acid positions 115, L33, 147, G73 and L89. The median darunavir phenotype (fold
`change from reference) of the virologic failure isolates was 21—fold at baseline and 94-
`fold at failure.
`
`Cross-resistance
`
`Darunavir has a <10—fold decreased susceptibility in cell culture against 90% of 3309
`clinical
`isolates resistant
`to amprenavir, atazanavir,
`indinavir,
`lopinavir, nelfinavir,
`ritonavir, saquinavir and/or tipranavir showing that viruses resistant to most protease
`inhibitors remain susceptible to darunavir.
`In Studies C202 and C213, 60% (24/40) of
`subjects with decreased susceptibility to tipranavir
`(fold change >3) at baseline
`demonstrated a 1
`logo decrease from baseline at week 24 on darunavir/rtv and 45%
`(18/40) achieved <50 copies/mL serum HIV RNA levels. In Study C215, 60% (64/107)
`of subjects with resistance to tipranavir (>3—fold change) at baseline achieved a 1
`loglo
`decrease from baseline at week 24 on darunavir/rtv and 33% (35/107) achieved <50
`copies/mL serum HIV RNA levels.
`
`indinavir,
`Darunavir—resistant Viruses were not susceptible to amprenavir, atazanavir,
`lopinavir, nelfinavir, ritonavir or saquinavir in cell culture. However, six of nine
`darunavir-resistant viruses selected in cell culture from protease inhibitor—resistant
`viruses showed a fold change in EC50 values <3 for tipranavir, indicative of limited cross—
`resistance between darunavir and tipranavir. Of the viruses isolated from subjects
`experiencing virologic failure on darunavir/ritonavir 600/ 100 mg b.i.d., greater than 50%
`were still susceptible to tipranavir while less than 5% were susceptible to other protease
`inhibitors
`(amprenavir,
`atazanavir,
`indinavir,
`lopinavir, nelfinavir,
`ritonavir,
`or
`saquinavir).
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`NDA: 21— 976 SN: 000 DATE REVIEW COMPLETE. 06/19/2006
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`Cross-resistance between darunavir and the nucleoside/nucleotide reverse transcriptase
`inhibitors, the non-nucleoside reverse transcriptase inhibitors or the fusion inhibitor is
`unlikely because the Viral targets are different.
`
`Baseline Genotype/Phenotype and Virologic Outcome Analyses
`Genotypic and/or phenotypic analysis of baseline Virus may aid in determining darunavir
`susceptibility before initiation of 600/100 mg b i. d darunavir/rtv therapy.
`As—treated
`analyses were conducted to evaluate the impact of specific baseline protease inhibitor
`resistance—associated mutations and the number of protease inhibitor
`resistance—
`associated mutations at baseline on virologic response. Both specific mutations and the
`number of baseline mutations as well as susceptible drugs in the optimized background
`regimen and enfuvirtide use affected darunavir/11v response rates in Phase 2 Studies C202
`and C213.
`
`The presence at baseline of the mutations V321, l47V, or 154L or M, was associated with
`a decreased virologic response to darunavir and decreased susceptibility to darunavir. In
`addition, a diminished virologic response was observed in subjects with 27 protease
`inhibitor resistance—associated mutations (any change at amino acid positions 30, 32, 36,
`46, 47, 48, 50, 53, 54, 73, 82, 84, 88, or 90) at baseline. The response rate in all
`subgroups (by type and number of mutations at baseline) was generally higher in the
`darunavir/1W group compared to the control group.
`
`Baseline darunavir phenotype (shift in susceptibility relative to reference) was shown to
`be a predictive factor of virologic outcome. Analyses showed that response rates at
`Week 24 decreased when the baseline darunavir phenotype was >7—fold. Phenotypic
`subgroups of 0—2, >2—7, >7-30 and >30 described responses rates in four tiers of 88%,
`73%, 52% and 43% with a 1 logo decrease from baseline, respectively, and 60%, 47%,
`24% and 19% with <50 copies/mL, respectively.
`
`The number of susceptible drugs in the optimized backgrOund regimen and enfiJViItide
`use affected darunavir/rtv response rates. In Studies C202 and C213, subjects with no
`susceptible NRTIs at baseline had lower response rates (38% with 1 logo decrease and
`13% with <50 copies/mL) than those with at least one susceptible NRTI. In addition, for
`subjects with baseline darunavir phenotypes of >10 in studies C202, C213 and C215,
`response rates were 81% (13/ 16) when ENF was used for the first time concomitantly
`with darunavir while response rates were 36% (27/74) for those who did not use ENF
`concomitantly.
`
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`NDA: 21-—976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`Table of Contents
`
`ExecutiVe Summary
`Recommendations
`
`1.1.Rec0mmendati0ns 0n Approvability
`1.2 Recommendation on Phase 4 Commitments
`
`Summary ofMicrobiology Assessments
`2.1 Non—clinical
`2.2 Clinical
`
`Administrative signatures
`Microbiology Review
`4.1 Important Milestones in Development
`4.2 Methodology
`4.3 State ofantimicrobials usedfor the indication sought
`4.4 Non—clinical Microbiology
`4.5 Clinical Studies
`
`4.6 Clinical Microbiology
`Conclusion
`
`Package Insert
`Appendices
`Appendix A
`Appendix B
`Appendix C
`Appendix D
`Appendix E
`Appendix F
`Appendix G
`
`Page 2
`
`Page 6
`Page 6
`
`Page 7
`Page 8
`Page 10
`
`Pagell
`Pagell
`Pagel3
`Page14
`Page28
`Page30
`Page42
`Page44
`
`Page 48
`Page 50
`. Page 52
`Page 53
`Page 54
`Page 60
`Page 62
`
`AL»
`
`\IG\UI
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`‘
`
`NDA: 21-976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`
`Microbiology Reviewer: Lisa K. Naeger, PhD.
`
`1.
`
`Recommendations
`
`1.1. Recommendation and Conclusion on Approvability
`
`This NDA for is approvable with respect to microbiology for combination
`antiretroviral treatment of HIV—1 infected treatment—experienced adult subjects.
`
`1.2. Recommendation on Phase 4 (Post-Marketing) Commitments, Agreements,
`and/or Risk Management Steps, if Approvable.
`
`We request the following to be submitted with traditional approval (included in the
`approval letter of this NDA):
`.
`1) Determine response rates based upon presence of specific cleavage site mutations
`at baseline and submit this analysis with the PREZISTA traditional approval
`application.
`2) Determine the protease cleavage site mutations that occur most frequently (>10%)
`in virologic failure isolates and submit this analysis with the PREZISTA
`traditional approval application.
`3) Determine if the most frequently occurring protease cleavage site mutations
`contributed to decreases in darunavir susceptibility through site-directed
`mutagenesis and submit this analysis with the PREZISTA traditional approval
`application.
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`NDA: 21-976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`2. Summary of Microbiology Assessments
`
`2.1 Non-clinical Microbiology
`
`Complete study reports describing the mechanism of action, antiviral activity, in vitro
`combination activity, in vitro resistance selection and cross—resistance of darunavir were
`submitted and reviewed in this NDA. Enzymatic assays and time of addition studies
`confirmed that darunavir acts as a protease inhibitor. Darunavir had a K; value of <0.09
`nM when tested in enzymatic assays showing that it has comparable inhibition against
`HIV-1 protease as the currently approved PIs. Crystal structures for darunavir bound to
`WT or mutant HIV-l protease showed that darunavir formed van der Waals interactions
`with protease residues L23, G49, 150, P81, V82 and 184 and interacted with residues
`A28, D29, D30, V32, I47 and I50.
`
`The median ECso values of darunavir against laboratory strains HIV-1“113 and HIV—1ND“3
`ranged from 2.3 to 6.3 nM in MTT and cell-based assays. The median EC50 values were
`1.2 nM, 1.7 nM, and 5 nM against HIV—1"“3, HIV-ls”, and HIVBAL, respectively, in
`PBMCs and macrophages/monocytes using a p24 assay. Darunavir demonstrated
`antiviral activity against HIV—2 and SIV with EC50 values less than 10 nM and
`demonstrated activity against all of the HIV—1 subtype isolates evaluated with ECso
`values ranging from <0.1 nM to 4.28 nM. These results indicate that darunavir had
`antiviral activity against a broad range of virus subtypes. The CC50 value in MT4 cells in
`a 5—day MTT assay was >100 uM giving a therapeutic index of >26,000. The 50% toxic
`concentration of darunavir on cell viability was greater than 200 uM for the cell types
`tested.
`
`'
`
`The antiviral activity of darunavir was decreased by a median 52—fold and 5.4—fold by the
`presence of 1 mg/mL AAG and 50% human serum, respectively. No loss of activity was
`observed for darunaVir in the presence of 45 mg/mL human serum albumin with a 1.6—
`fold median change in antiviral activity. The data from in vitro antiviral activity drug
`combination assays showed that darunavir was not antagonistic with any of the currently
`approved antiretroviral drugs and that synergy was observed in vitro with three PIS:
`amprenavir, nelfinavir and ritonavir
`
`In vitro selection experiments were performed starting from wild-type. Replicating
`viruses could not be selected in the presence of darunavir at concentrations above 220 nM
`out to 738 days. Selection of viruses was slower with darunavir in comparison with other
`PIs tested at micromolar concentrations. Darunavir—resistant virus derived in cell culture
`from wild-type HIV had 6— to 21—fold decreased susceptibility to darunavir and harbored
`3 to 6 of the following amino acid substitutions S37N/D, R41E/S/T, K55Q, K70E, A71T,
`T74S, V771, or I85V in the protease. Selection in cell culture of darunavir resistant HIV—
`1
`from nine HIV-1 strains harboring multiple protease inhibitor resistance—associated
`mutations resulted in the overall emergence of 22 mutations in the protease gene,
`including LIOF, V111, 113V, 115V, G16E, L23I, V321, L33F, S37N, M461, I47V, I50V,
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`NDA: 21-976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`F53L, L63P, A71V, G738, L76V, V821, 184V, T91A/S, and Q92R, of which L10F, V321,
`L33F, S37N, M461, 147V, 150V, L63P, A71V, and 184V were the most prevalent. These
`darunavir—resistant viruses had at least eight protease mutations and exhibited 50- to 641—
`fold decreases in darunavir susceptibility with final EC50 values ranging from 125 nM to
`346] nM.
`
`2.2 Clinical Microbiology
`
`For the clinical microbiology assessment, genotypes and phenotypes from 319 subjects
`(1584 total isolates) from study C202 (POWER 2) and 318 subjects (1767 total isolates)
`from study C213 (POWER 1) were submitted and reViewed for this NDA.
`In addition,
`genotypes and phenotypes from 460 subjects (1935 total isolates from study C215
`(POWER 3) were submitted and reviewed. Subjects in studies C202, C213, and C215
`who rebounded or were never suppressed were analyzed for mutations developing on
`treatment in the FDA analysis.
`‘
`
`The FDA analysis of resistance focused on the comparator PI control group and 600 mg
`darunavir/rtv bid group from studies C202 and C213 since 600 mg darunavir/rtv bid is
`the registrational dosage. Baseline genotypic response analyses were performed only on
`the controlled phase 2 studies C202 and C213 using Week 24 data. Baseline phenotypic
`response analyses were performed on the phenotypic data from studies C202, C213 and
`C215 in order to have a largerpool of data to describe baseline darunavir phenotypic
`subgroups. These baseline response analyses were performed on a censored as-treated
`dataset. Subjects who discontinued while suppressed or discontinued at week 2 were
`censored from the analyses. In addition, the virologic failures from studies C202, C213
`and C215 were analyzed for mutations developing on darunavir/11v treatment.
`
`Resistance
`
`1n the Phase 2 trials Studies C202, C213 and C215, multiple protease inhibitor—resistant
`HIV—1 isolates from highly treatment-experienced subjects who received 600/100 mg
`darunavir/rtv b.i.d. and experienced virologic failure (n=164), either by rebound, or by
`never being suppressed, developed amino acid substitutions that were associated with a
`decrease in susceptibility to darunavir. The amino acid substitution V321 developed on
`darunavir/rtv 600/100 mg b.i.d. in 35% of virologic failure isolates and substitutions at
`amino acid 154 developed in 24% of virologic failure isolates. Other substitutions that
`developed in 10% to 20% of darunavir/rtv virologic failure isolates occurred at amino
`acid positions 115, L33, 147, G73 and L89. The median darunavir phenotype (fold
`change from reference) of the virologic failure isolates was 21-fold at baseline and 94-
`fold at failure.
`'
`
`Cross-resistance
`
`Darunavir has a <10-fold decreased susceptibility in cell culture against 90% of 3309
`clinical
`isolates resistant
`to amprenavir, atazanavir,
`indinavir,
`lopinavir, nelfinavir,
`ritonavir, saquinavir and/or tipranavir showing that viruses resistant to most protease
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`'
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`NDA: 21—976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`In studies C202, C213 and C215, 60%
`inhibitors remain susceptible to darunavir.
`(88/147) of subjects with decreased susceptibility to tipranavir (fold change >3) at
`baseline demonstrated a 1 logo decrease from baseline at week 24 on darunavir/rtv and
`36% (53/147) achieved <50 copies/mL serum HIV RNA levels.
`
`indinavir,
`Darunavir—resistant viruses were not susceptible to amprenavir,' atazanavir,
`lopinavir, nelfinavir, ritonavir or saquinavir in cell culture. However, six of nine
`darunavir—resistant viruses selected in cell culture from protease inhibitor—resistant
`Viruses showed a fold change in ECSO values <3 for tipranavir, indicative of limited cross—
`resistance between darunavir and tipranavir. Of the viruses isolated from subjects
`experiencing Virologic failure on darunavir/ritonavir 600/100 mg b.i.d., greater than 50%
`were still susceptible to tipranavir while less than 5% were susceptible to other protease
`inhibitors
`(amprenavir,
`atazanavir,
`indinavir,
`lopinavir, nelfinavir,
`ritonavir,
`or
`saquinavir).
`'
`
`Cross—resistance between darunavir and the nucleoside/nucleotide reverse transcriptase
`inhibitors, the non-nucleoside reverse transcriptase inhibitors or the fusion inhibitor is
`unlikely because the Viral targets are different.
`
`Baseline Genotype/Phenotype and Virologic Outcome Analyses
`Genotypic and/or phenotypic analysis of baseline virus may aid in determining darunavir
`susceptibility before initiation of 600/ 100 mg b.i.d darunavir/rtv therapy. Analyses were
`conducted to evaluate the impact of specific baseline protease inhibitor resistance—
`associated mutations and the number of protease inhibitor resistance—associated mutations
`at baseline on Virologic response. Both specific mutations and the number of baseline
`mutations as well as susceptible drugs in the optimized background regimen and
`enfuviitide use affected darunavir/rtv response rates in Phase 2 Studies C202 and C213.
`
`The presence at baseline of the mutations V321, 147V, or 154L or M, was associated with
`a decreased Virologic response to darunavir and decreased susceptibility to darunavir. In
`addition, a diminished Virologic response was observed in subjects with 27 protease
`inhibitor resistance-associated mutations (any change at amino acid positions 30, 32, 36,
`46,- 47, 48, 50, 53, 54, 73, 82, 84, 88, or 90) at baseline. However, the response rate in all
`subgroups (by type and number of mutations at baseline) was generally higher in the
`darunavir/rtv group compared to the control group.
`
`Baseline darunavir phenotype (shift in susceptibility relative to reference) was shown to
`be a predictive factor of Virologic outcome. Response rates of 340 subjects in studies
`C202, C213, and C215 who received 600/ 100 mg b.i.d darunavir/rtv therapy assessed by
`the baseline darunavir phenotype showed that a baseline darunavir phenotype of greater
`than 7—fold change from reference resulted in decreased response rates at Week 24. The
`subgroup of subjects with baseline darunavir phenotypes of 0-2 had response rates of
`88% (1 logo decrease from baseline) and 60% (<50 copies/mL). The subgroup of
`subjects with baseline darunavir phenotypes of >2—7 had response rates of 73% (1 logo
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW ,
`
`NDA: 21—976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`decrease from baseline) and 47% (<50 copies/mL). The subgroup of subjects with
`baseline darunavir phenotypes >7—30 had response rates of 52% (1 log“) decrease from
`baseline) and 24% (<50 copies/mL). The subgroup of subjects with baseline darunavir
`phenotypes >30 had response rates of 43% (1 logo decrease from baseline) and 18% .
`(<50 copies/mL).
`
`The number of susceptible drugs in the optimized background regimen and enfuvirtide
`use affected darunavir/rtv response rates.
`In Studies C202 and C213, subjects with no
`susceptible NRTIs at baseline had lower response rates (38% with 1 log”) decrease and
`13% with <50 copies/mL) than those with at least one susceptible NRTI. In addition, for
`subjects with baseline darunavir phenotypes of >10 in studies C202, C213 and C215,
`response rates were 81% (13/16) when ENF was used for the first time concomitantly
`with darunavir while response rates were 36% (27/74) for those who did not use ENF
`concomitantly.
`
`3. Administrative
`
`3.]. Reviewer’s Signature(s)
`
`
`
`Lisa K. Naeger, Ph.D.
`Sr. Microbiologist, HFD-530
`
`_
`
`3.2. Concurrence
`
`HFD—5 30/Si gnatory Authority
`
`Signature
`
`8
`
`Date
`
`
`HFD—530/Micro TL .
`'
`
`Signature
`
`Date
`
`10
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`.
`
`NDA: 21—976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`Microbiology Review
`
`4.]
`
`Important Milestones in Product Development
`
`January 2005: Submission of Phase II resistance data in the HIV resistance template
`format (SAS datasets) was discussed with the sponsor and agreed upon.
`
`The NDA was proposed for rolling review submission (IND62477 Serial No. 223)
`and accepted by the Division (FDA facsimile correspondence dated April 5, 2005 and
`email correspondence dated April 7, 2005).
`
`June 2005: Discussions were held regarding NDA submission of'virology reports.
`The sponsor stated that the initial Virology Summary would be provided in the
`September 2005 submission and include the currently available in vitro virology data
`and clinical virology data from trials TMCl l4—C201 and TMC114—C207. The in Vitro
`virology part contains Mechanism of Action, In Vitro Drug Resistance, and
`Combinations with current HIV—1 inhibitors. The clinical virology part contains
`exploratory clinical virology data supporting the efficacy in the controlled clinical
`studies. The updated Virology Summary would be provided in the December 2005
`submission to include the available exploratory clinical virology data from trials
`TMCI l4—C202 and TMCl 14—C2l3 to constitute a comprehensive overview of the
`virology of darunavir.
`’
`
`June 2005: In an effort to get as much resistance data as possible, the Division
`requested that the sponsor provide genotypes and phenotypes from study C215. hi
`addition, we requested that sponsor to provide the Cmin and IQ data in the template
`datasets.
`'
`
`4.2 Methodology
`
`Genotypes and phenotypes were determined by default on plasma samples taken at
`’ predefined timepoints (i.e. screening, Week —2, baseline, Week 2, Week 24, Week 48,
`Week 96 and final/withdrawal visit) when the viral load was > 1000 copies/mL.
`Furthermore, samples at other timepoints, such as confirmed virologic failure, were
`also analyzed upon request of the Protocol Virologist. Isolates from subjects from
`Studies C202, C213, and C215 who started from Day 1 on the recommended dose
`(darunavir/rtv 600 mg b.i.d.), and who were rebounders (i.e.: experienced virologic
`failure by rebound) were analyzed for genotypic and phenotypic changes at virologic
`failure compared to baseline.
`
`Note: In the exploratory resistance analysis, virologic failure by rebound was defined
`as viral load > 0.5 logioHlV-l RNA copies/mL above nadir at 2 consecutive visits,
`following a confirmed virologic response of 2 consecutive viral load measurements >
`1 logro HIV-1 RNA copies/mL below baseline.
`
`11
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`NDA: 21—976 SN : 000 DATE REVIEW COMPLETE: 06/19/2006
`
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`Genotypes and phenotypes from 319 subjects (1584 total isolates) from study C202
`(POWER 2) were submitted for review [63 subjects in 400 mg bid group, 65 subjects
`in 400 mg qd group, 64 subjects from 800 mg qd group 66 subjects from 600 mg bid
`darunavir dosage group and 61 subjects from the control group]. Note: subjects 6603
`and 6604 were mixed up in the treatment arm column, so I assigned 6603 to 4Q and
`6604 to 8Q. Genotypesand phenotypes from 318 subjects (1767 total isolates) from ‘
`study C213 (POWER 1) were submitted for review [63 subjects in 400 mg bid group,
`64 subjects in 400 mg qd group, 63 subjects from 800 mg qd group, 65 subjects from
`600 mg bid darunavir dosage group and 63 subjects from the control group]. In
`addition, genotypes and phenotypes from 460 subjects (1935 total isolates from study
`C215 (POWER 3) were submitted for review. Subjects in study C215 who
`rebounded (n=46) or were never suppressed (n=78) were analyzed in the FDA
`analysis.
`
`The FDA analysis of resistance focused on the control group and 600 mg darunavir
`bid group from studies C202 and C213 since 600 mg darunavir bid is the registration
`dosage. Baseline genotypic response analyses were only done on the controlled
`phase 2 studies C202 and C213, but baseline phenotypic response analyses were done
`on studies C202, C213 and C215. A censored as—treated dataset was used for the
`baseline response analyses. Subjects who discontinued while suppressed or
`discontinued at week 2 were censored. In addition, the virologic failures from studies
`C202, C213 and C215 were analyzed for mutations developing on darunavir
`treatment.
`
`Genotypic Methods
`' Genotypic analyses were performed at Virco by automated population sequencing.
`Individual data were reported as amino acid changes along the PR as compared to the
`HIV—1/HXB2 wild—type reference. Genotypic mixtures (a combination of different
`amino acids at 1 position) were reported. In addition, single genome sequencing
`(SGS) was used to detect mutations that could be missed by population sequencing.
`SGS was performed at Virco on the baseline samples and used to determine whether
`the emerging mutations were selected during the treatment period or were already
`present, at low prevalence, in the baseline species.
`
`Briefly, the HIV-1 complementary DNA (cDNA) region encompassing the PR and
`the first 400 codons of the RT was derived from plasma RNA by reverse transcription
`and amplified by polymerase chain reaction (PCR). The corresponding 2.2 kilobases
`PR—RT fragments were run and extracted from a 1% agarose gel. Ligations and
`
`transformations of these fragments were performed using the
`.
`——-—A—-————————~.
`Sixty—four single colonies were picked
`and resuspended individually in the PCR reaction mix. The cloned HIV—l PR-RT
`segments were then reamplified by PCR and sequenced. For analysis all clones
`mutation profiles were recorded per unique sequences, and a composite profile of the
`overall detected mutations was generated.
`
`12
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`'
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`NDA: 21-976 SN: 000 DATE REVIEW COMPLETE: 06/19/2006
`
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`Phenotygic Methods ‘
`Phenotypic analyses were performed at Vireo. Recombinant clinical isolates were
`constructed according to the Antivirogram® method. Briefly, protease (PR) and
`reverse transcriptase (RT) coding sequences were amplified from patient—derived
`Viral RNA with HIV—1 specific primers. After homologous recombination of
`amplicons into a PR—RT deleted proviral clone, the resulting recombinant viruses
`were harvested and used for in vitro susceptibility testing. Cut—offs used based on the
`Antivirogram assay to- determine resistance are shown in Table].
`
`Table 1. Resistance Call Determination Using Biological or Clinical Cut-off (FC)
`based on the Antivirogram
`
`- Biolo ital Cut-off (FC)
`
`Clinical Cut-ofi’fFC)
`
`NNRTI
`
`
`
`
`
`4.3 State of antimicrobials used for the indication (s) sought:
`
`An estimated 40 million people worldwide were infected with HIV in 2001 and 3
`million died from AIDS. Since HAART regimens have been introduced, the number of
`AIDS cases has decreased dramatically. HAART does not eradicate HIV from subjects
`completely and even though the number of HIV RNA copies is reduced to undetectable
`levels, HIV re—emerges quickly after discontinuation of HAART. Therefore, with the
`currently available regimens, it is likely that most HIV—infected subjects will require
`antiretroviral therapy throughout their lives.
`
`There are currently over 20 FDA—approved anti—HIV drugs including seven Pls
`(amprenavir/fosamprenavir, atazanavir, indinavir, lopinavir, nelfinavir, ritonavir,
`saquinavir, tipranavir), eight NRTIs (abacavir, didanosine, emtn'citabine, lamivudine,
`
`l3
`
`

`

`DIVISION OF ANTIVIRAL PRODUCTS
`OFFICE OF NEW DRUGS
`MICROBIOLOGY REVIEW
`
`NDA: 21-976 SN : 000 DATE REVIEW COMPLETE: 06/19/2006
`
`Microbiology Reviewer: Lisa K. Naeger, Ph.D.
`
`stavudine, tenofovir, zalcitabine, zidovudine), three NNRTIs (delavirdine, efavirenz,
`nevirapine) and the fusion inhibitor enfuvirtide.‘ PIS work at the late stage of viral
`replication to prevent virus production from infected cells. They block the HIV
`protease enzyme, which is necessary for the production of mature virions, resulting in
`defective particles that are unable to infect new cells. NRTls mimic nucleosides and
`target HIV-1 RT by competing with natural deoxynucleoside triphosphates for binding
`to RT and by incorporating into newly synthesized Viral DNA resulting in chain—
`terrnination. NNRTls inhibit HIV—l RT by binding near the catalytic site of RT and
`. acting as noncompetitive inhibitors. Enfuvirtide (T—20) is a gp4l fusion inhibitor
`preventing the joining of the viral and cellular membranes necessary for virus entry.
`
`Unfortunately, HIV develops resistance to antiretroviral drugs over time usually from
`the accumulation of multiple mutations. HAART regimens are also associated with
`. acute toxicities such as diarrhea, kidney stones, rash, CNS toxicities and hepatotoxicity.
`Long—term toxicities from antiretroviral therapies include mitochondrial toxicities
`associated with NRTls (lactic acidosis, myopathy, neuropathy, pancreatitis), and
`disorders of lipid metabolism (dyslipidemia) and glucose metabolism (lipodystophy,
`hypercholesterolemia, hypertriglyceridemia)

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