throbber
CENTER FOR DRUG EVALUATION AND
`RESEARCH
`
`APPLICA TION NUMBER:
`
`21-821
`
`CLINICAL PHARMACOLOGY AND
`
`BIOPHARMACEUTICS REVIEW! S 2
`
`
`
`Apotex Exhibit 1026
`Page 1 of 130
`
`Apotex Exhibit 1026
`Page 1 of 130
`
`

`

`CLINICAL PHARMACOLOGY & BIOPHARMACEUTICS REVIEW
`
`
`
`NDA# 21-821
`PRODUCT Tigecycline (Tygacil™)
`FORMULATION Sterile Powder for injection
`DOSAGE STRENGTH 50mg vial
`SUBMISSION DATES December 15, 2004; March 14, 2005; April 12, 2005;
` May 5, 2005; May 16, 2005; May 19, 2005; May 27,
` 2005
`SUBMISSION TYPE New Molecular Entity
`SPONSOR Wyeth Pharmaceuticals
`OCPB DIVISION Division of Pharmaceutical Evaluation III
`MEDICAL DIVISION Division of Anti-Infective Drug Products
`REVIEWER Jeffrey J. Tworzyanski, Pharm.D.
`PM REVIEWER Yaning Wang, Ph.D.
`TEAM LEADER Venkat R. Jarugula, Ph.D.
`PM TEAM LEADER Joga Gobburu, Ph.D.
`
`
` 1
`
` EXECUTIVE SUMMARY
`Wyeth Pharmaceuticals submitted a New Drug Application for Tygacil™ (Tigecycline
`for injection) on December 15, 2004. The FDA granted this submission a priority review
`based upon the sponsor’s studies in the treatment of resistant organisms. Tigecycline
`represents a new class of antimicrobials known as glycylcyclines. The glycylcycline class
`of antimicrobial agents is a synthetic derivative of minocycline. The mechanism of action
`of the tetracyclines is by binding to the 30S ribosomal subunit at the A-site which blocks
`entry of amino-acyl transfer RNA molecules into the ribosome which prevents
`incorporation of amino acid residues into elongating peptide chains. The glycylcyclines
`(tigecycline) also inhibit the 30S ribosomal subunit but bind with substantially higher
`affinity than the tetracyclines. The glycylcyclines interact directly with another region of
`the A-site.
`
`
`Wyeth is requesting approval for two indications for tigecycline. The first indication is
`complicated skin and skin structure infections caused by Escherichia coli, Enterococcus
`faecalis (vancomycin-susceptible strains only), Staphylococcus aureus (methicillin-
`susceptible and –resistant strains), Streptococcus agalactiae, Streptococcus anginosus
`grp. (includes S. anginosis, S. intermedius, and S. constellatus), Streptococcus pyogenes
`and Bacteroides fragilis. The second indication for tigecycline that Wyeth is seeking is
`complicated intra-abdominal infections caused by Citrobacter freundii, Enterobacter
`cloacae, Escherichia coli, Klebsiella oxytoca, Klebsiella pneumonia, Enterococcus
`faecalis (vancomycin-susceptible strains only), Staphylococcus aureus (methicillin-
`susceptible strains only), Streptococcus anginosus grp. (includes S. anginosus, S.
`intermedius, and S. constellatus), Bacteroides fragilis, Bacteroides thetaiotaomicron,
`Bacteroides uniformis, Bacteroides vulgatus, Clostridium perfringens, and
`Peptostreptococcus micros.
`
`
`
`1
`
`Apotex Exhibit 1026
`Page 2 of 130
`
`

`

`
`The proposed intravenous dosage regimen for tigecycline is an initial dose of 100mg,
`followed by 50mg every 12 hours. Intravenous infusions of tigecycline should be
`administered over approximately 30 to 60 minutes every 12 hours. The recommended
`duration of treatment with tigecycline for complicated skin and skin structure infections
`or for complicated intra-abdominal infections is 5-14 days depending upon severity and
`site of infection.
`
` A
`
` total of 17 Phase 1 clinical pharmacology studies were conducted to investigate single
`dose and multiple dose pharmacokinetics, metabolic disposition, the effect of special
`populations ( renal and hepatic impairment), drug-drug interactions (digoxin, warfarin),
`and the effect of race, age, and gender on the pharmacokinetics. Eleven phase 2/3 studies
`were submitted of which four Phase 3 and two Phase 2 studies were pertinent to the
`proposed indications. In addition the sponsor performed population PK analysis on the
`sparse sample data collected in Phase 2/3 studies. Thirteen out of 17 submitted Phase 1
`studies were evaluated in this review. Four Phase 1 studies were not reviewed because
`they did not provide relevant or additional information. Sponsor has adequately
`characterized the pharmacokinetics and pharmacodynamics of tigecycline following
`intravenous infusion.
`
`1.1 RECOMMENDATIONS
`The Office of Clinical Pharmacology and Biopharmaceutics/Division of Pharmaceutical
`Evaluation III (OCPB/DPE III) has reviewed NDA 21-821. The submission is acceptable
`from a Clinical Pharmacology point of view. The labeling comments need to be
`communicated to the sponsor.
`
`1.2 PHASE 4 COMMITMENTS
`There are no phase 4 commitments.
`
`1.3 SUMMARY OF CLINICAL PHARMACOLOGY AND
`BIOPHARMACEUTICS FINDINGS
`
`
`Pharmacokinetic characteristics:
`Following intravenous infusion of tigecycline, the drug concentrations in serum declined
`in a polyphasic manner. The initial steep decline in serum concentrations at the end of
`infusion represents the distribution phase during which the drug is distributed out of the
`systemic circulation into various tissues. The terminal elimination phase represents the
`movement of the drug out of tissues into systemic circulation and the subsequent
`elimination from the body. The pharmacokinetics of tigecycline (Cmax and AUC) after
`single dose infusion over 60 minutes are linear over the dose range of 12.5 mg to 300 mg.
`The mean systemic clearance (CL) values were consistent among dose groups and ranged
`from 0.2 L/h/kg to 0.3 L/h/kg. Tigecycline was well distributed into various tissues as
`shown by the mean steady state volume of distribution (Vss), which ranged from 7 to 14
`L/kg. The estimated terminal phase half life varied among dosages (ranges from 18 hrs at
`50 mg dose to 42 hrs at 200 mg dose) because the terminal phase was not adequately
`characterized at lower doses due to assay sensitivity. The steady state plasma
`
`
`
`2
`
`Apotex Exhibit 1026
`Page 3 of 130
`
`

`

`concentrations are achieved by Day 3 with the proposed dosage regimen of 100 mg
`loading dose followed by 50 mg q 12hrs. The mean terminal elimination half-life at
`steady state following the administration of the therapeutic regimen was approximately
`40 hrs.
`
`Distribution:
`In vitro plasma protein binding of tigecycline is concentration dependent and ranges from
`71% at 0.1 µg/ml to 89% at 1 µg/ml to 96% at 15 µ/ml. Following intravenous infusion
`of 100 mg dose, the concentration of tigecycline was 38 times higher in the gallbladder
`than in the serum, two-fold higher in the colon than in the serum and 8.6 times higher in
`the lung compared to serum. The degree of penetration of tigecycline into skin blister
`fluid as measured by the ratio of AUC in blister to AUC in serum was 74%.
`
`Metabolism and Excretion:
`Based on the results of in vitro metabolism studies, tigecycline is not metabolized by the
`cytochrome P450 (CYP 450) enzyme system and it does not inhibit the metabolism of the
`drugs that are metabolized by CYP 450 system. Therefore, tigecycline is not expected to
`cause CYP 450 based metabolic drug interactions.
`
`Following administration of [14C]tigecycline, approximately 33% of the administered
`dose is recovered in urine and 59% of the administered dose is recovered in feces, for a
`total recovery of 92% of the administered dose. Approximately 22% of the total dose is
`excreted as unchanged tigecycline in urine. The biliary excretion of tigecycline and its
`metabolites is the primary route of elimination. The secondary elimination pathways of
`tigecycline and its metabolites are renal excretion, glucuronidation, and amide hydrolysis
`followed by N-acetylation to form N-acetyl-9-aminominocycline; and these secondary
`pathways each account for 15% or less of the total elimination of tigecycline.
`
`Special populations:
`Renal Impairment:
`In study 103-US (Protocol 3074A1) the sponsor evaluated the PK of tigecycline 100mg
`dose in 20 subjects. Approximately one third of the subjects were with normal renal
`function, one third subjects with CrCl < 30 ml/min, and one third subjects with end-stage
`renal disease (ESRD) who were receiving hemodialysis. The renal clearance of
`tigecycline in healthy subjects was similar to creatinine clearance (ClCr), and it
`represented approximately 20% of the total systemic clearance of tigecycline.
`Consequently, the systemic clearance of tigecycline was reduced by approximately 20%
`in subjects with severe renal impairment (ClCr<30 ml/min) or ESRD, and tigecycline area
`under the curve (AUC) increased by approximately 30% in these subjects. Based on the
`results of this study, tigecycline dose does not need to be adjusted in patients with renal
`impairment. Hemodialysis did not remove tigecycline from the systemic circulation of
`the subjects.
`
`Hepatic Impairment:
`In study 105-EU (Protocol 3074A1) the sponsor evaluated the effect of hepatic
`impairment on the pharmacokinetics of tigecycline following the administration of a
`
`
`
`3
`
`Apotex Exhibit 1026
`Page 4 of 130
`
`

`

`single dose of 100mg in patients with mild (Child-Pugh-A: score of 5 or 6, n=10),
`moderate (Child-Pugh-B: score of 7 to 9, n=10), and severe hepatic impairment (Child-
`Pugh-C: score of 10 to 13, n=5) in comparison to 23 healthy subjects matching the
`cirrhotic subjects for age, sex, weight, and smoking habit. The results of the study
`indicate that dosage adjustment is not warranted in patients with mild hepatic impairment
`(Child-Pugh A) because of no change in the clearance of drug compared to healthy
`subjects. In moderate and severe hepatic impairment (Child-Pugh B and Child-Pugh C)
`the clearance was approximately 25% and 50% lower, respectively, than the healthy
`subjects. Since the clearance of the drug in the severe hepatic impairment population was
`decreased by 50% and the AUC was increased by 105% these patients should receive an
`initial dose of 100mg followed by a reduced maintenance dose of 25 mg every 12 hours.
`
`Drug Interactions:
`Digoxin: Concomitant administration of the proposed tigecycline regimen (100 mg
`loading dose followed by 50 mg q 12hrs) did not significantly alter the pharmacokinetics
`of digoxin.
`
`Warfarin: Coadministration of the therapeutic regimen of tigecycline significantly
`decreased the oral clearance of both R-warfarin and S-warfarin, thereby increasing the
`Cmax (38% and 43%, respectively) and AUC (68% and 29%, respectively) of both
`compounds. The pharmacodynamic endpoint, the mean INRmax was reduced by 10%.
`Sponsor recommended in the label that prothrombin time or other suitable coagulation
`parameters should be monitored if tigecycline is administered with warfarin.
`
`Oral contraceptives: Sponsor did not conduct a drug interaction study with oral
`contraceptives. However, tetracyclines are reported to reduce the effectiveness of oral
`contraceptives. Tigecycline is eliminated predominantly through biliary excretion and is
`shown to reduce the intestinal microflora. Therefore, the sponsor stated in the label that
`concurrent use of antibiotics with oral contraceptives may render oral contraceptives less
`effective.
`
`Exposure response:
`Animal data:
`Based on the dose fractionation studies in murine neuropenic thigh model, both AUC and
`the time of plasma concentration above MIC (T>MIC) appeared to be best correlated
`with antimicrobial efficacy of tigecycline.
`
`Human data:
`Efficacy:
`Even though good exposure-response relationships were established in animal model (see
`2.2.4.1), the limited exposure levels studied in patients and high success rate in clinical
`trials made the exposure-response analysis quite challenging in human studies. For both
`cSSSI and cIAI indications the exposure-response analysis showed that AUCss(0-
`24)/MIC was a borderline statistically significant predictor of microbiologic and clinical
`response. However, the relationship between exposure and efficacy is so shallow that
`
`
`
`4
`
`Apotex Exhibit 1026
`Page 5 of 130
`
`

`

`under the proposed dosage regimen (100mg loading dose and 50mg maintenance dose),
`the exposure change did not cause clinically relevant difference in efficacy.
`
`Safety:
`First occurrence of nausea and vomiting was selected as the endpoint for tigecycline
`safety evaluation. No exposure-response relationship was established within the observed
`exposure levels in Phase 2/3 studies for safety. The analysis based on phase 1 studies (84
`subjects from Study 3074A1-100, 46 subjects from Study 3074A1-102, and 6 subjects
`from Study 3074A1-103) showed AUC0-inf and Cmax were significant predictors for
`both first nausea and first vomiting occurrence due to the wide range of doses studied.
`The lack of exposure-response relationship for the first nausea and first vomiting
`occurrence in phase 2/3 studies was due to the narrow range of exposure under the two
`doses, 25mg and 50mg.
`
`
`
`
`
`
` __________________________________________________
` Jeffrey J. Tworzyanski, Pharm.D.
` Office of Clinical Pharmacology and Biopharmaceutics
` Division of Pharmaceutical Evaluation III
`
`
`
`
`RD/FT Initialed by Venkat R. Jarugula, Ph.D.,__________________________________
`Team Leader
`
`cc:
`Division File: NDA 21-821
`HFD-520 (CSO/Milstein)
`HFD-520 (MO/Cooper)
`HFD-520 (Microbiology/Marsik)
`HFD-880 (Division File, Lazor, Selen, Jarugula, Tworzyanski)
`CDR (Clin. Pharm./Biopharm.)
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`5
`
`Apotex Exhibit 1026
`Page 6 of 130
`
`

`

`
`
`
`Table of Contents
`
` 1
`
` Executive Summary………………………………………………………………….1
` 1.1 Recommendations……………………………………………………………….2
` 1.3 Summary of Clinical Pharmacology and Biopharmaceutics Findings………….2
`
` 2
`
` Question Based Review
` 2.1 General Attributes of the Drug…………………………………………………..7
` 2.2 General Clinical Pharmacology………………………………………………….9
` 2.3 Intrinsic Factors………………………………………………………………….30
` 2.4 Extrinsic Factors…………………………………………………………………35
` 2.5 General Biopharmaceutics……………………………………………………....39
` 2.6 Analytical Section……………………………………………………………….39
`
` 3
`
` 4
`
` Detailed Labeling Recommendations………………………………………………42
`
` Appendices
` 4.2 Individual Study Reports
` 4.2.1 Study 3074A1-104-US………………………………………………63
` 4.2.2 Study 3074A1-100-EU………………………………………………69
` 4.2.3 Study 3074A1-101-US………………………………………………77
` 4.2.4 Study 3074A1-109-US………………………………………………85
` 4.2.5 Study 3074A1-113-US………………………………………………91
` 4.2.6 Study 3074A1-117-US………………………………………………95
` 4.2.7 Study 3074A1-116-EU……………………………………………..102
` 4.2.8 Study 3074A1-103-US……………………………………………..107
` 4.2.9 Study 3074A1-105-EU……………………………………………..112
` 4.2.10 Study 3074A1-115-US……………………………………………..121
` 4.2.11 Study 3074A1-111-US……………………………………………..128
` 4.2.12 Study 3074A1-102-US……………………………………………..135
` 4.2.13 Study 3074A1-112-US……………………………………………..141
`
` 4.3 Pharmacometric Consult Request Form……………………………………......146
` 4.4 Cover Sheet and OCPB Filing/Review Form…………………………………..148
`
`
`
`
`
`
`
`
`
`
`
`
`6
`
`Apotex Exhibit 1026
`Page 7 of 130
`
`

`

`
`
`
` 2
`
` QUESTION BASED REVIEW
`
`
`2.1 General Attributes of the Drug
`
`2.1.1 What are the highlights of the chemistry and physical-chemical properties of
`the drug substance and the formulation of the drug product as they relate to clinical
`pharmacology and biopharmaceutics review?
`
`
`Tigecycline is a glycylcycline antibacterial for intravenous infusion. The chemical name
`of tigecycline is (4S, 4aS, 5aR, 12aS)-9-[2-(tert-butylamino)acetamido]-4,7-
`bis(dimethylamino)-1, 4, 4a, 5, 5a, 6, 11, 12a-octahydro-3, 10, 12, 12a-tetrahydoxyl-1,
`11-dioxo-2-naphthacenecarboxamide. The empirical formula is C29H39N5O8 and the
`molecular weight is 585.65. The chemical structure of tigecycline is shown below:
`
`
`
`
`
`
`Tigecycline for injection is supplied as an orange lypholized powder or cake. Each
`Tygacil vial contains 50mg lypholized powder for intravenous infusion. The product does
`not contain excipients or preservatives.
`
`
` when titrated with
`Tigecycline is freely soluble
` Tigecyline has five ionizable groups, three basic
` in a wide pH range of
`nitrogens and two acidic hydroxyl groups. The pKa’s are expected to overlap and
`assignment of each pKa to its respective functional group is difficult. The NMR method
`was employed by the sponsor because it can determine the overlapping pKa values as
`well as allow the assignment of each pKa to its respective functional group. Figure 1
`shows the chemical structure with pKa values determined by NMR.
`
`
`
`
`
`
`
`
`7
`
`(b) (4)
`
`(b) (4)
`
`(b) (4)
`
`(b) (4)
`
`Apotex Exhibit 1026
`Page 8 of 130
`
`

`

`
`
`
`Figure 1. Tigecycline pKa Values
`
`
`
`
`
`Tigecycline’s solubility is independent of pH because no changes in solubility were seen
`in a wide pH range.
`
`2.1.2 What are the proposed mechanism(s) of action and therapeutic indication(s)?
`The bacterial ribosome is the target for the tetracyclines. Binding to the 30S ribosomal
`subunit at the A-site blocks entry of amino-acyl transfer RNA molecules into the
`ribosome, preventing incorporation of amino acid residues into elongating peptide chains.
`Tigecycline has in its chemical structure a 9-glycyl amido residue substitution that allows
`for novel drug-amino acid interactions at binding sites. The sponsor’s in vitro data
`suggest that tigecycline has a ribosomal binding site affinity that is at least 10-fold greater
`than tetracycline-class antibiotics. Ribosomal protection and efflux are the two major
`resistance mechanisms for tetracyclines. Tigecycline is active against strains with either
`mechanism of tetracycline resistance. In general, tigecycline is considered bacteriostatic.
`
`The sponsor is seeking an indication for the treatment of complicated skin and skin
`structure infections caused by Escherichia coli, Enterococcus faecalis (vancomycin-
`susceptible strains only), Staphylococcus aureus (methicillin-susceptible and –resistant
`strains), Streptococcus agalactiae, Streptococcus anginosus grp. (includes S. anginosus,
`S. intermedius, and S. constellatus), Streptococcus pyogenes and Bacteroides fragilis.
`The sponsor is also seeking an indication for complicated intra-abdominal infections
`caused by Citrobacter freundii, Enterobacter cloacae, Escherichia coli, Klebsiella
`oxytoca, Klebsiella pneumoniae, Enterococcus faecalis (vancomycin-susceptible strains
`only), Staphylococcus aureus (methicillin-susceptible strains only), Streptococcus
`anginosus grp. (includes S. anginosus, S. intermedius, and S. constellatus), Bacteroides
`fragilis, Bacteroides thetaiotaomicron, Bacteroides uniformis, Bacteroides vulgatus,
`Clostridium perfringens, and Peptostreptococcus micros.
`
`
`
`
`8
`
`Apotex Exhibit 1026
`Page 9 of 130
`
`

`

`2.1.3 What are the proposed dosage(s) and route(s) of administration?
`The proposed dosage regimen for intravenous tigecycline is an initial dose of 100mg,
`followed by a dose of 50mg administered every 12 hours following the initial dose. These
`doses are administered over approximately 30 to 60 minutes. The proposed duration of
`treatment with tigecycline for complicated skin and skin structure infections or for
`complicated intra-abdominal infections is 5-14 days, depending upon severity and site of
`infection.
`
`2.2 General Clinical Pharmacology
`
`2.2.1 What are the design features of the clinical pharmacology and clinical studies
`used to support dosing or claims?
`The sponsor performed 17 Phase I studies to assess the single dose and multiple dose
`effects of safety, tolerability, pharmacokinetics, metabolic disposition, renally impaired
`patients, hepatically impaired patients, and drug interactions (digoxin, warfarin) with
`tigecycline.
`
`
`The sponsor performed two Phase 2 studies (3074A1-200-Us and 3074A1-202-US)
`evaluating the safety and efficacy of tigecycline in subjects with cSSSI and cIAI.
`In study 3074A1-200-US subjects were randomized to receive either an initial 100 mg IV
`tigecycline load dose, followed by 50 mg tigecycline IV q12h (n=54), or 50mg IV
`tigecycline load dose, followed by 25 mg tigecycline IV q12h (n=55) for the treatment of
`cSSSI for seven to 14 days. In study 3074A1-202-US subjects received an initial 100mg
`IV tigecycline load followed by 50 mg IV tigecycline q12h for the treatment of cIAI
`(n=66) for at least five days but not more than 14 days of therapy.
`
`The sponsor performed four Phase 3 studies evaluating the safety and efficacy of
`tigecycline for the treatment of cSSSI and cIAI. Table 1 shows the features of these
`studies.
`
`Table 1. Phase 3 Study Features
`Study
`Indications
`
`Treatment of known
`or suspected
`diagnosis of cSSSI
`Treatment of
`hospitalized subjects
`with cSSSI
`Treatment of cIAI
`
`Study Drug
`(number of subjects)
`Tigecycline 100mg load
`followed by 50mg IV q12h
`x 14days (n=292)
`Tigecycline 100mg load
`followed by 50mg IV q12h
`x 14days (n=281)
`Tigecycline 100mg load
`followed by 50mg IV q12h
`x 14 days (n=413)
`
`3074A1-
`300-US/CA
`
`3074A1-
`305-WW
`
`3074A1-
`301-WW
`
`3074A1-
`306-WW
`
`Comparator Drug
`(number of subjects)
`Vancomycin 1 g IV
`followed by Aztreonam
`2g IV q12h (n=281)
`Vancomycin 1g IV
`followed byAztreonam 2
`g IV q12h (n=269)
`Imipenem/cilistatin IV
`q6h dose adjusted based
`upon weight, CrCl
`(n=412)
`Imipenem/cilistatin
`IV q6h dose adjusted
`upon weight, CrCl
`
`Treatment of cIAI
`
`Tigecycline 100mg load
`followed by 50mg IV q12h
`x 14days (n=404)
`
`
`
`9
`
`Apotex Exhibit 1026
`Page 10 of 130
`
`

`

`(n=413)
`
`
`2.2.2 What is the basis for selecting the response endpoints (i.e., clinical or surrogate
`endpoints) or biomarkers (collectively called pharmacodynamics (PD)) and how are
`they measured in clinical pharmacology and clinical studies?
`
`
`cSSSI:
`The primary efficacy endpoint was the assessment of clinical response at the test of cure
`(TOC) visit. The TOC visit was at least 12 but not more than 92 days after the last dose
`of study drug. Clinical response was measured according to predefined criteria. The
`secondary efficacy endpoints were microbiological response from specimens obtained
`from 2 sets of blood cultures, aerobic and anaerobic cultures from the primary site of
`infection.
`
`cIAI:
`The primary efficacy endpoint was the TOC assessment. The TOC visit was at least 12
`but not more than 44 days after the last dose of study drug. Clinical response was
`measured according to predefined criteria. Microbiologic efficacy was evaluated by two
`sets of blood cultures, aerobic and anaerobic cultures from the primary intra-abdominal
`site of infection at baseline. The outcome of the microbiologic response at the subject
`level and pathogen level was described according to predefined criteria.
`
`2.2.3 Are the active moieties in the plasma (or other biological fluid) appropriately
`identified and measured to assess pharmacokinetic parameters and exposure
`response relationships?
` Yes, refer to 2.6, Analytical Section
`
`2.2.4.1 What are the characteristics of the exposure-response relationships (dose-
`response, concentration-response) for efficacy?
`
`
`Animal studies:
`The exposure-response relationship of tigecycline has been evaluated using in vivo
`animal models of infection. A murine thigh model study was performed by
`Six week old, specific-pathogen-free female ICR/Swiss mice (weight 23 to 25 g) from
`Sprague-Dawley were used. The mice were rendered neutropenic (<100 neutrophils/µl)
`by injecting a dose of cyclophosphamide intraperitoneally at four days (150mg/kg) and
`one day (100 mg/kg) before the infection experiment. After a 1:10 dilution of
`pneumococci in fresh broth, 0.1ml (~106 CFU) was injected into the thighs of ether-
`anesthetized mice. Mice were treated for 24 hours with total doses in the range of 0.19 to
`24 mg/kg body weight/day for the experiments with the strains of S. pneumoniae.
`Antibiotics were administered subcutaneously in 0.2ml volumes beginning 2 hours after
`thigh inoculation. Mouse thighs were removed and homogenized and cultured
`quantitatively. The level of detection of this assay was
`/thigh. Single-dose
`pharmacokinetic studies were performed with sera from thigh-infected mice with the
`following doses of tigecycline: 3, 12, and 48 mg/kg. The pharmacokinetics appeared to
`
`.
`
`
`
`10
`
`(b) (4)
`
`(b) (4)
`
`Apotex Exhibit 1026
`Page 11 of 130
`
`

`

`be nonlinear, resulting in a higher elimination t½ at a dose of 48mg/kg. The results of the
`pharmacokinetic-pharmacodynamic parameters for tigecycline are shown in figure 2.
`
`Figure 2. Relationship between pharmacokinetic-pharmacodynamic parameters
`and therapeutic efficacy of tigecycline (free drug) against S. pneumnoniae 1199 in
`the neutropenic mouse thigh muscle infection model
`
`
`
`
`
`(R2 = 0.82, 0.83, and 0.54 for panels a, b, and c, respectively)
`(a) time above the 0.5 x MIC versus effect
`(b) Log AUC versus effect
`(c) Log Cmax versus effect
`
`The pharmacokinetic parameter that correlated best with efficacy was the time above a
`certain factor times the MIC for five of the six organism-drug combinations studies. The
`magnitude of this factor varied from 0.5 to 4. The only exception to this observation was
`the combination of tigecycline and S. pneumoniae 1199, for which both AUC and time
`above MIC were important in predicting outcome (R2 = 0.83 and 0.82, respectively).
`The results of the study indicated that tigecycline exhibited time-dependent antimicrobial
`activity in vivo. However, due to the relatively long t½ and the long post-antibiotic effect
`(PAE), the AUC was also reasonably predictive, with slightly lower R2 values.
`
` murine intraperitoneal model was used for tigecycline to determine PK in CD-1 mice
`after 0.15, 1, 4, 10 and 20mg/kg doses. Tigecycline concentrations in blood were assayed
`by HPLC methods at various times after dosing. The pharmacokinetic analysis indicates
`that AUC is linear over the dose range tested. Table 2 presents the range of AUCs and
`AUC/MIC from the murine intraperitoneal test performed at Wyeth (MIRACL-26501;
`MIRACL-26884; GTR-28013; MIRACL-25770).
`
`
`
`
`
`
`
`
`
`
` A
`
`
`
`11
`
`Apotex Exhibit 1026
`Page 12 of 130
`
`

`

`Table 2. Range of AUCs and AUC/MICs in the mouse Intraperitoneal Infection
`Model
`
`
`
`
`Human studies
`Dose selection:
`Animal studies have shown that both AUC and the T>MIC ratio as important parameters
`for efficacy and tigecycline was efficacious in animals at AUC exposures comparable to
`those observed in humans with a 50 to 100mg total daily dose. Based on this data, doses
`of 25mg and 50mg every 12 hours were studied in Phase 2. A Phase 2 study for cSSSI
`showed that the cure rate was higher in the 50mg q12h group (75%) than the 25mg q12h
`dose group (67%). Phase 1 studies showed tigecycline was tolerable up to 50mg q12 and
`75mg q12 was not well tolerated because of high incidence of nausea and vomiting.
`Therefore, the sponsor selected the current dose regimen of 100mg load followed by
`50mg q12h for Phase 3 studies.
`
`Efficacy:
`Even though good exposure-response relationships were established in animal model (see
`2.2.4.1), the limited exposure levels studied in patients and high success rate in clinical
`trials made the exposure-response analysis quite challenging in human studies. For cIAI
`microbiological efficacy, 1 phase 2 study (3074A1-202: 19 patients, 32 observations) and
`2 phase 3 studies (3074A1-301: 12 patients, 17 observations; 3074A1-306: 40 patients,
`57 observations) were combined for exposure-response analysis at the pathogen level
`(Table 3). Formal statistical analysis, however, could not be applied unless cohorts 1, 2
`and 3 are combined due to the low failure rate in each single cohort. The sponsor found
`that AUCss(0-24)/MIC, as a continuous covariate, was a borderline statistically
`significant predictor of microbiologic response for cohorts 1, 2 and 3 combined. Under
`the proposed dosage regimen (100mg loading dose and 50mg maintenance dose), the
`exposure change did not cause clinically relevant difference on microbiological efficacy
`as indicated by Figure 3.
`
`Table 3: Baseline Pathogen Classification Used in Exposure-Response Analysis for cIAI Efficacy
`Cohort
`Baseline Pathogen(s) Included
`Monomicrobial E. coliinfections
`1
`2
`Other monomicrobial or polymicrobial Gram-negative infections
`(Klebsiellaspp., Enterobacterspp. and/or Citrobacterspp. plus or
`minus E. coli)
`Infections with at least one Gram-negative pathogen plus at least
`one anaerobic pathogen
`
`3
`
`
`
`12
`
`Apotex Exhibit 1026
`Page 13 of 130
`
`

`

`4
`
`5
`
`Infections with at least one Gram-negative pathogen plus at least
`one Gram-positive pathogen
`All other monomicrobial or polymicrobial infections
`
`
`For cIAI clinical efficacy, similar results were obtained. Both baseline APACHE II score
`and AUCss(0-24)/MIC were identified as statistically significant predictors of clinical
`response based on univariate logistic analysis. Under the proposed dosage regimen
`(100mg loading dose and 50mg maintenance dose), the exposure change did not cause
`clinically relevant difference on clinical efficacy as indicated by Figure 4.
`
`Figure 3. Exposure-Response Analysis of cIAI Efficacy - Final Logistic Regression
`Model of Microbiological Response versus AUCSS(0-24)/MIC Ratio with Histogram of
`Observed Data (71 patients with 106 observations.)
`
`=26 n=26
`
`n=27
`
`n=27
`
`0
`
`20
`
`40
`
`60
`80
`AUCss(0-24)/MIC Ratio
`
`100
`
`120
`
`140
`
`
`
`13
`
`1.0
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Probability of Bacteriologic Cure
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Apotex Exhibit 1026
`Page 14 of 130
`
`

`

`Figure 4. Exposure-Response Analysis of cIAI Efficacy - Final Logistic Regression
`Model of Clinical Response versus AUCSS(0-24)/MIC with Histogram of Observed Data
`(71 patients)
`
`n=17
`
`n=18
`
`n=18
`
`n=18
`
`0
`
`20
`
`40
`
`60
`
`1.0
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Probability of Clinical Cure
`
`AUCss(0-24)/MIC Ratio
`
`
`
`For cSSSI microbiological efficacy, 1 phase 2 study (3074A1-200: 25 patients) and 2
`phase 3 studies (3074A1-300: 2 patients; 3074A1-305: 9 patients) were combined for
`exposure-response analysis at the patient level (Table 4). Formal statistical analysis,
`however, could not be applied unless cohorts 2 and 3 are combined due to the low failure
`rate in each single cohort. The sponsor found that AUCss(0-24)/MIC, as a continuous
`covariate, was a borderline statistically significant (p=0.113) predictor of microbiologic
`response for cohorts 2 and 3 combined. Under the proposed dosage regimen (100mg
`loading dose and 50mg maintenance dose), the exposure change did not cause clinically
`relevant difference on microbiological efficacy as indicated by Figure5.
`
`Table 4: Baseline Pathogen Classification Used in Exposure-Response Analysis for cSSSI
`Efficacy
`Cohort
`1
`2
`3
`4
`5
`
`Baseline Pathogen(s) Included
`Monomicrobial S. aureus
`Monomicrobial S. aureus or Streptococcus spp.
`Polymicrobial S. aureus plus Streptococcus spp. or two Strep. spp.
`Other polymicrobial Gram-positive and/or Gram-negative pathogens
`Gram-negative or anaerobic monomicrobial pathogens
`
`
`For cSSSI clinical efficacy, similar results were obtained. Both AUCss(0-24) (p=0.065)
`and AUCss(0-24)/MIC (p=0.1723) were identified as marginally significant predictors of
`clinical response based on univariate logistic analysis. Under the proposed dosage
`regimen (100mg loading dose and 50mg maintenance dose), the exposure change did not
`cause clinically relevant difference on clinical efficacy as indicated by Figure 6.
`
`
`
`14
`
`Apotex Exhibit 1026
`Page 15 of 130
`
`

`

`
`Figure 5. Exposure-Response Analysis of cSSSI Efficacy - Final Logistic Regression Model of
`Microbiological Response versus AUCSS(0-24)/MIC Ratio with Histogram of Observed Data in
`Cohorts 2 and 3 and AUCSS(0-24)/MIC Ratio Range under Each Dose (36 subjects)
`dose50mg
`
`1.0
`
`n=9
`
`n=9
`
`n=9
`
`dose25mg
`
`n=9
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Probability of Bacteriologic Cure
`
`20
`
`40
`
`60
`
`80
`
`100
`
`AUCss(0-24)/MIC Ratio
`
`
`Figure 6. Exposure-Response Analysis of cSSSI Efficacy - Final Logistic Regression Model of
`Clinical Response versus AUCSS(0-24)/MIC Ratio with Hi

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