throbber
CENTER FOR DRUG EVALUATION AND
`RESEARCH
`
`
`
`APPLICATION NUMBER:
`
`203858Orig1s000
`
`SUMMARY REVIEW
`
`
`
`Page 1 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`Page 2 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`1. Introduction and Background
`This memo summarizes the basis for the regulatory action for lomitapide. This oral drug is
`being proposed for the orphan indication of the treatment of homozygous familial
`hypercholesteremia (HoFH). In-depth review and analyses of specific issues can be found in
`the primary reviews of the respective disciplines. This memo contains my summary,
`assessments, and conclusions concerning the major issues identified during the review of this
`application.
`
`Lomitapide is a first-in-class small molecule inhibitor of the microsomal triglyceride protein
`(MTP) that transfers lipids to apolipoprotein B to form the apo B-containing lipoprotein
`complex. Inhibition of MTP prevents the assembly and secretion of apo B-containing
`lipoproteins, which include VLDL-C (the precursor of LDL-C), and chylomicrons from the
`liver and intestine, respectively.
`
`Homozygous familial hypercholesteremia (HoFH) results from loss of function mutations in
`both alleles of the LDL receptor (LDL-R). These mutations render the LDL-Rs absent or non-
`functional leading to reduced clearance of LDL particles from circulation, resulting in marked
`elevation in plasma LDL-C levels. Untreated LDL-C levels in individuals with HoFH usually
`range from 500 to 1000 mg/dL. If left untreated, HoFH patients die prematurely from
`accelerated atherosclerotic cardiovascular disease by the second or third decade of life. In the
`U.S., the prevalence of HoFH is approximately 1 per million persons.
`
`Treatment options for HoFH are limited in number and in scope. High potency HMG-CoA
`reductase inhibitors (statins), with or without a cholesterol absorption inhibitor, and LDL
`apheresis are the mainstay of therapy (see Table 1). Statin therapy depends on functional
`LDL-Rs for most of its lipid lowering effects and, therefore, has limited efficacy in HoFH.
`Similar to dialysis, LDL apheresis is an extracorporal procedure that selectively removes apo-
`B containing lipoproteins (VLDL-C, LDL-C, lipoprotein (a), and triglycerides). The
`procedure, however, needs to be performed on a chronic, repetitive basis of every one to two
`weeks, and there are currently only 35 apheresis centers in the U.S. Liver transplantation has
`been employed rarely as a last resort.
`
`Table 1: Non-surgical therapies for HoFH
`Therapy
`Mechanism of action
`
`LDC-C lowering response in
`HoFH
`
`< 10 – 25%
`< 10%
`
`LDLR activity
`HMG-CoA reductase inhibitors
`LDLR activity, inhibits
`Cholesterol absorption
`cholesterol absorption
`inhibitors
`~30 – 40%1
`LDL-C removal
`LDL-apheresis*
`*Response based on time averaged LDL-C levels; acutely, apheresis lowers LDL-C by 50-75%
`
`Drug therapy in combination with LDL apheresis can typically reduce LDL-C by 45% to
`55%.2 Because HoFH patients have such elevated LDL-C levels at baseline (> 500 mg/dL),
`
`1 Pfohl M, Naoumova RP, Klass C, Knisel W, Jakober B, Risler T, Thompson GR. Acute and chronic effects on cholesterol
`biosynthesis of LDL-apheresis with or without concomitant HMG-CoA reductase inhibitor therapy. J Lipid Res. 1994;35(11):1946.
`2 Gilbert R. Thompson, M. Barbir, D. Davies, et al. Efficacy criteria and cholesterol targets for LDL apheresis. Atherosclerosis 208
`(2010) 317–321.
`
`
`
`Reference ID: 3236195
`
`2
`
`Page 3 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`combining multiple treatment modalities still fails to provide adequate control of LDL-C, and
`HoFH patients remain at high-risk for serious adverse cardiovascular events and premature
`death. There is a clear need for additional therapies to help HoFH patients either approach or
`reach LDL-C treatment goals.
`
`This NDA submission supports the use of lomitapide at a starting daily dose of 5 mg titrated to
`a maximum of 60 mg, based on safety and tolerability, as an adjunct to diet and lipid lowering
`therapies to reduce LDL-C in HoFH patients.
`
`2. Recommendations of Review Disciplines regarding Approvability
`This section summarizes key recommendations from the review disciplines.
`
`CMC: In his review signed on October 18, 2012, the primary reviewer (Xavier Ysern)
`recommended approval of lomitapide from a CMC perspective.
`
`Biopharmaceutics (ONDQA): In her review signed October 26, 2012, the primary reviewer
`(Elsbeth Chikhale) recommended approval of lomitapide from a biopharmaceutics perspective.
`A waiver for the requirement to conduct a BA/BE study for the 10 mg capsule strength was
`granted.
`
`Pharmacology Toxicology: In his review signed November 5, 2012, the primary reviewer
`(Brian [Tim] Hummer) recommended approval of lomitapide from a pharmacology-toxicology
`perspective. Safety concerns based on preclinical findings are discussed in Section 4 (Safety).
`The team recommended a juvenile toxicology study, as a postmarket requirement, to be
`conducted prior to evaluating lomitapide in pediatric HoFH patients.
`
`Clinical Pharmacology: In his review signed November 5, 2012, the primary reviewer (Sze
`[Johnny] Lau) recommended approval from a clinical pharmacology perspective.
`Recommended dosing modifications based on drug-drug interactions, food effect, and
`hepatic/renal impairment that will be incorporated into labeling are discussed in Section 4
`(Safety).
`
`Statistics: In her review signed November 30, 2012, the primary reviewer (Cynthia Liu)
`concluded that lomitapide was effective in reducing LDL-C and the pre-specified secondary
`lipid parameters and recommended approval from a statistical perspective.
`
`Clinical: In his review signed November 27, 2012, the primary reviewer (James Smith)
`recommended approval from a clinical perspective. Important clinical findings and
`assessments are discussed in Sections 3 (Efficacy) and 4 (Safety) below.
`
`I concur with the recommendation of approval from the review disciplines.
`
`3. Efficacy
`Efficacy of lomitapide in HoFH patients was demonstrated in one Phase 3 trial (HoFH-pivotal,
`29 HoFH patients); with supportive evidence from one Phase 2 study (HoFH-pilot, 6 HoFH
`
`
`
`Reference ID: 3236195
`
`3
`
`Page 4 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`patients). The primary efficacy endpoint in both trials was the percent change from baseline to
`endpoint in directly measured serum LDL-C; each subject served as his or her own control.
`
`The surrogate endpoint of serum LDL-C has been an accepted primary efficacy measure in
`marketing applications for lipid-lowering therapies in the U.S. The relationship between
`reductions in LDL-C levels and decreased risk of adverse cardiovascular outcomes has been
`well established for statin therapy. Although there are no data correlating LDL-C reduction
`and improved cardiovascular outcomes for MTP inhibitors, there is no reason to believe that
`LDL-C would not be an acceptable efficacy endpoint for HoFH patients treated with
`lomitapide. Moreover, a definitive cardiovascular outcomes trial in HoFH patients would be
`infeasible because of the rarity of the disease, and LDL-C is the most appropriate surrogate
`measure available.
`
`The HoFH-pivotal trial was a multinational, open-label, single-arm trial in 29 HoFH patients
`on stable diet and maximally tolerated LDL lowering drugs, with or without apheresis, at
`baseline. Patients received lomitapide as add-on therapy at an individually defined maximum
`tolerated dose between 5 and 60 mg once daily for 78 weeks (weeks 0 to 26 was the efficacy
`phase, weeks 26 to 78 was the safety phase). Patients could enroll in the extension study
`(HoFH-extension) after completing the 78 weeks of treatment; the extension study is ongoing.
`
`The primary efficacy endpoint, % change in LDL-C levels from baseline to Week 26/end of
`treatment, was analyzed using paired t-test performed on the intent to treat population (all 29
`patients) with last-observation-carried-forward (LOCF) imputation of missing data. Primary
`efficacy results are shown in Table 2. It should be noted that the observed LDL-C reduction
`from lomitapide treatment was in addition to the lipid lowering effects of baseline therapies.
`Table 2: Primary Endpoint – Percent change in LDL-C from baseline to Week 26 (HoFH-
`pivotal)
`
`N = 29
`
`Baseline LDL-C
`(mg/dL)
`
`Relative
`Change from
`Baseline (%)
`
`-40 (32)
`-52 to -27
`-50
`-93, +21
`
`P*
`
`<0.001
`
`Absolute
`Week 26/ITT/LOCF
`Change from
`LDL-C
`Baseline
`(mg/dL)
`(mg/dL)
`
`
`
`
`Mean (SD)
`337 (114)
`191 (107)
`-147 (127)
`95% CI
`
`
`
`Median
`357
`169
`-107
`Min, Max
`152, 565
`28, 443
`-351, +49
`Source: Adapted from primary statistical review (Cynthia Liu), Table 2
`* P-value based on paired t-test for mean % change
`
`Maximum LDL-C reduction reached plateau at Week 18 and was maintained at approximately
`45% reduction at Week 56; the mean maximum tolerated dose at Week 26 and at Week 56 was
`approximately 40 mg.
`
`Categorical LDL-C response: 19 of the 29 patients (66%) had LDL-C reductions ≥ 25%, with
`8 (28%) having LDL-C levels < 100 mg/dL. Four of these 8 patients were receiving apheresis.
`The fact that HoFH patients treated with adjunctive lomitapide could attain the National
`Cholesterol Education Program’s target LDL-C treatment goal is noteworthy.
`
`
`
`Reference ID: 3236195
`
`4
`
`Page 5 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`Apheresis: Of the 29 patients, 18 (62%) were receiving apheresis treatment at the beginning of
`the trial. Because the acute reduction and ensuing rebounds in plasma lipid levels could
`confound efficacy measurements, the protocol specified that the timing of on-treatment LDL-C
`measurements relative to the preceding apheresis was to be the same as that established at
`baseline. In general, patients on apheresis experienced a mean LDL-C reduction at Week
`26/LOCF of smaller magnitude compared to patients not on apheresis (-35% versus -49%).
`This observation may be confounded by the fact that a greater proportion of patients on
`apheresis discontinued drug treatment in the efficacy phase and the mean drug dose at Week
`26/LOCF was higher for patients not on apheresis. Taken altogether, there is no compelling
`reason to believe that apheresis negatively impacts the efficacy of lomitapide.
`
`Of the 13 apheresis-treated patients remaining in the HoFH-pivotal study at Week 26, three
`discontinued apheresis and three reduced the frequency of the procedure by Week 78. All but
`one patient either maintained the LDL reduction achieved during the efficacy phase or
`experienced slight rise in LDL-C levels, but these levels remained ≥ 50% below baseline.
`I consider these favorable alterations in apheresis to be significant in reducing the burden of
`the treatment for HoFH patients. Although decreasing the frequency of or discontinuing
`apheresis while on lomitapide may lead to less than optimal LDL-C reduction than if the
`apheresis regimen was not altered, the quality of life benefits of forgoing each apheresis
`procedure can be immediately appreciated, given the avoidance of time, cost, inconvenience,
`and risks that are incurred each time the procedure is performed.
`
`Supportive data from the HoFH-pilot study for the primary endpoint of mean % change in
`LDL-C from baseline to the end of the dosing intervals (approximately 4 weeks each) are
`shown below. This study enrolled 6 adult HoFH patients who received lomitapide once daily
`according to a prespecified dosing scheme that called for dose escalation every 4 weeks
`(starting at 0.03 mg/kg and ending at 1.0 mg/kg). LDL-C levels were measured at the end of
`every 4 weeks at a certain dose. These findings demonstrate a dose response indicating a drug
`effect of lomitapide.
`
`Table 3: Supportive Evidence of LDL-C reduction (HoFH-pilot)
`
`
`Results for the secondary endpoints from the HoFH-pivotal trial are shown in Table 4. These
`findings are reassuring in that the changes in these lipid parameters are consistent with LDL-C
`reduction, but it would be premature to conclude that these changes are adequate evidence of
`additional cardiovascular benefits beyond lowering of LDL-C. I agree with Dr. Smith’s
`recommendation that only the secondary endpoints of total cholesterol, apoB, and non-HDL-C
`should be included in the indication for lomitapide. These endpoints are reflected in changes
`
`
`
`
`
`Reference ID: 3236195
`
`5
`
`Page 6 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`in LDL-C and could potentially inform drug benefit in a manner similar to LDL-C.
`Hypertriglyceridemia is not a feature of HoFH disease, and the clinical relevance of
`triglyceride reduction in a population that has normal triglyceride levels is questionable.
`
`
`Table 4: Secondary endpoints (HoFH-pivotal)
`Lipid Parameters Relative Change from Baseline to Week 26/LOCF
`N=29
`Mean change % (SD)
`-36 (28)*
`Total cholesterol
`-40 (30)*
`ApoB
`-29 (56)*
`Triglycerides
`-40 (30)*
`Non-HDL-C
`-29 (58)
`VLDL-C
`-11(34)
`Lp (a)
`Source: Adapted from primary clinical review (James Smith), Table 71
`*P < 0.01 based on paired t-test for mean % change
`
`HDL-C: Serum HDL-C levels declined from Week 0 to Week 26 (mean reduction of 7%
`[using LOCF] or 12% [using completer analysis]) but subsequently returned to baseline levels
`by Week 56. Serum levels of apoA1 followed a similar pattern. The reason for these lipid
`changes is unknown. Given the magnitude of reduction of HDL-C, the spontaneous return of
`HDL levels to baseline with continued treatment, and the uncertain effect of drug-induced
`changes in HDL-C on cardiovascular risk, I do not believe these HDL-C changes would
`significantly offset the benefit anticipated from lomitapide’s favorable effect on LDL-C.
`
`Efficacy conclusion
`Lomitapide was efficacious in reducing serum LDL-C in HoFH patients on a low-fat diet and
`maximally tolerated lipid lowering treatment. Drug benefit was also observed for other lipid
`parameters, such as total cholesterol, apo B, and non-HDL-C. In some patients, lomitapide led
`to beneficial alterations in apheresis regimen or the attainment of NCEP’s LDL-C target
`treatment goal. I consider these drug benefits to be clinically important.
`
`4. Safety
`The clinical safety findings of lomitapide have been thoroughly discussed in Dr. Smith’s
`review. This safety assessment focuses on safety findings in the HoFH population, and is
`limited to significant safety issues of interest identified by the review teams. Although 915
`individuals (most were healthy volunteers, patients with renal/hepatic impairment, or non-
`HoFH patients with hypercholesteremia) received at least one dose of lomitapide in the 24
`studies provided in the NDA, the phase 3 safety database to support the intended use for
`lomitapide only consists of 29 HoFH patients enrolled in the HoFH-pivotal trial and its
`extension. Such a small safety database could only provide assurance that the true incidence
`of an adverse outcome is no greater than 10% when the outcome is not observed in the trial.
`
`Death: One death (myocardial infarction) occurred in the entire drug development program
`(54 year-old man with factor V Leiden, a history of deep vein thrombosis, morbid obesity
`(BMI 42 kg/m2)). Although a potential drug association could not be completely excluded, I
`believe it is unlikely that lomitapide caused the event.
`
`
`
`Reference ID: 3236195
`
`6
`
`Page 7 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`Non-fatal serious adverse events (SAEs): All narratives were reviewed by Dr. Smith. I
`agree with Dr. Smith’s assessment that lomitapide may have contributed to the SAEs
`experienced by the following 2 patients:
`
`
`1. Subject 11-004: This case of “hepatoxicity” occurred in a 54 year-old man with
`elevated liver enzyme tests at baseline and throughout the phase 3 trial (1.5X to 5X
`ULN). His liver biopsy, obtained at baseline as workup for elevated liver enzymes,
`showed mild steatosis. At the scheduled Week 138 visit, his ALT was 24X ULN, AST
`13X ULN, and alkaline phosphatase 2.5X ULN, with normal bilirubin levels. Prior to
`this visit, he was treated with clarithromycin (a strong CYP3A4 inhibitor) and the
`antidepressant agomelatine (known to increase hepatic aminotransferases), and had
`increased his alcohol intake. He was otherwise asymptomatic. His liver enzymes
`normalized several months after all drugs were discontinued. His follow-up liver
`biopsy showed simple steatosis that had increased from his previous biopsy, without
`histologic evidence of inflammation or fibrosis.
`
` I
`
` concur with Dr. Smith’s assessment that this was a case of (multi) drug-induced
`hepatoxicity, given all the culprits involved. This case illustrates the need to
`emphasize through labeling the potential serious sequelae of drug-drug interactions
`(DDI) of lomitapide with CYP3A4 inhibitors or with hepatotoxic drugs or substances.
`
`2. Subject 01-1003: This patient experienced multiple SAE’s related to over-
`anticoagulation that may have resulted from the now known drug-drug interaction
`between lomitapide and warfarin. Labeling will address this DDI and recommend
`monitoring for INR levels when warfarin is administered concomitantly with
`lomitapide.
`
`
`Drug discontinuation due to adverse events: In the HoFH-pivotal trial, 6 of 29 patients
`(21%) discontinued drug treatment prior to Week 26: 3 discontinued for gastrointestinal
`symptoms; 2 withdrew consent with AEs of headaches, unstable INR, and gastrointestinal
`symptoms; and 1 discontinued because of anxiety about gastrointestinal side effects. In the
`ongoing HoFH-extension study, 2 of 18 patients discontinued due to AE’s (hepatotoxicity,
`aminotransferase elevations).
`
`Common adverse events of interest: Gastrointestinal side effects, such as diarrhea, nausea,
`vomiting, dyspepsia, abdominal pain, were nearly universal, occurring in > 90% of HoFH
`patients. The most common severe AEs were also GI related. A decrease in weight was
`reported as an adverse event in 7 (24%) of the 29 HoFH patients. The largest change in mean
`weight was observed at Week 26 (-5% relative to baseline), with attenuation of weight
`decrease thereafter. It is, however, reassuring that the incidence of these AEs declined after the
`initial 26 weeks, despite the fact that no patients withdrew between Week 26 and Week 78 in
`the pivotal study. This pattern suggests possible tolerance or adaptation to these AEs.
`
`
`
`
`Reference ID: 3236195
`
`7
`
`Page 8 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`Special Safety Issues
`
` •
`
` Liver abnormalities
`
`All HoFH-
`pivotal
`(N=29)
`4 (14%)
`6 (21%)*
`3 (10%)
`1 (3%)
`0
`
`HoFH-
`extension
`(N=18)
`0
`2 (11%)
`1 (6%)
`2 (11%)
`1 (6%)
`
`All HoFH
`(N=29)
`3 (10%)
`4 (14%)*
`4 (14%)
`2 (7%)
`1 (3%)
`
`
`Hepatic transaminase elevations:
`The incidence of peak ALT elevations at various thresholds in the 78-week HoFH-pivotal
`(Week 0 – 26 efficacy phase and Week 26 – 78 safety phase) trial and its ongoing extension
`(HoFH-extension) is shown below:
`
`Table 4: Peak ALT elevations (HoFH)
`Efficacy
`Peak ALT
`Safety Phase
`Phase
`(N=23)
`During Period
`(N=29)
`≥2x, <3x ULN
`3 (10%)
`4 (17%)
`≥3x, <5x ULN
`4 (14%)
`3 (13%)
`≥5x, <10x ULN
`3 (10%)
`2 (9%)
`≥10x, <20x ULN
`1 (3%)
`0
`≥20x ULN
`0
`0
`Source: Primary clinical review (James Smith), Table 86
`*Although 6 patients had peak ALT between 3-5X ULN, two of those experienced higher peak ALT elevations (5-10X ULN, >20X
`ULN) during the extension study, leaving 4 subjects in the 3-5X ULN category when the pivotal and extension studies are
`combined.
`
`In the combined HoFH-pivotal and extension trials, 38% (11 of 29) of HoFH patients
`experienced at least one ALT value ≥ 3X ULN. These changes were not accompanied by
`laboratory changes indicating liver dysfunction, such as elevations in total bilirubin or INR.
`The median time to first ALT elevation was 126 days ([IQR 43, 155] for the 10 patients who
`had a peak ALT value ≥ 3X ULN during HoFH-pivotal. The remaining patient had her first
`ALT elevation during the HoFH-extension when she was found to have ALT elevations 10X
`ULN found on routine testing. She had been taking cold medications for the preceding several
`weeks. Her transaminase values decreased to near normal levels with dose interruption, and
`lomitapide was restarted at a lower dose 5 weeks later.
`
`The single patient with ALT elevation > 20X ULN in the HoFH-extension trial was discussed
`earlier in the “Non-fatal SAE” section. Increased ALT ≥ 5X ULN was observed in 4 patients
`during the HoFH-pivotal trial and occurred across a wide range of lomitapide doses (10 mg, 10
`mg, 20 mg, and 60 mg). Liver enzyme levels improved with dose modifications, and all four
`patients completed Week 78, with three of them ultimately tolerating doses equal to or
`exceeding the dose being taken at the time of their first ALT exceeding 5x ULN. One of the
`four patients subsequently experienced an ALT elevation > 10X ULN in the HoFH-extension
`trial and permanently discontinued lomitapide.
`
`Of the six patients with peak ALT ≥ 3X ULN < 5X ULN, five had resolution to <3x ULN
`without dose reduction or interruption.
`
`The temporal trend in median ALT and AST values up to Week 56 in the HoFH-pivotal trial is
`shown below. The shifts in aminotransferases are evident but there were no cases of “Hy’s
`Law.” One must be mindful, however, of the very small safety database when interpreting
`these findings.
`
`
`
`Reference ID: 3236195
`
`8
`
`Page 9 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`Figure 1: Median ALT and AST over time (HoFH-pivotal)
`
`
`
`Source: Primary clinical review (James Smith), Figure 34
`
`In the 16-week HoFH-pilot trial, 3 of 6 patients experienced on-treatment ALT/AST elevations
`≥ 5X ULN. Liver enzymes declined between dose titration, and all patients eventually received
`the highest daily dose of 1.0 mg/kg. All aminotransferase elevations normalized after drug
`discontinuation. There were no changes in other liver laboratory parameters, including total
`bilirubin, outside the normal range.
`
`In summary, lomitapide increases aminotransferases at a high frequency. The mechanism of
`action responsible for this adverse outcome is not well understood and may or may not be
`entirely related to hepatic fat accumulation; however,
`• Extreme transaminase elevations > 10X ULN occurred in the presence of other factors
`known to increase the risk of hepatic injury (drug-drug interactions, exposure to other
`hepatotoxins).
`• Hepatic enzyme elevations, even those of substantial magnitude, were reversible within
`weeks of drug interruption or dose reduction. Rechallenges were usually successful in
`patients who required drug interruption.
`• More modest elevations of < 5X ULN can resolve despite continued dosing, suggesting
`possible adaptation.
`• The serum aminotransferase elevations were not accompanied by evidence of impaired
`liver function, and there were no cases of Hy’s law, albeit the available safety data are very
`limited.
`
`
`For the reasons listed above, I believe that this significant risk of lomitapide can be
`appropriately managed in clinical practice, with available risk management strategies of
`labeling and REMS. That said, the risk of serious liver injury with lomitapide is undefined at
`this time due to a very small safety database, and will need to be further characterized in the
`postmarket long-term observational study.
`
`
`
`
`
`Reference ID: 3236195
`
`9
`
`Page 10 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`Hepatic steatosis
`Hepatic fat accumulation is an expected pharmacodynamic effect of lomitapide by virtue of its
`mechanism of action. Measurement of hepatic fat was performed using nuclear magnetic
`resonance spectroscopy (NMRS)/MRI in the HoFH-pivotal trial (at weeks 0, 26, 56, and 78)
`and in the ongoing HoFH-extension trial (every 6 months). The table below shows the
`temporal trend of hepatic fat accumulation observed in the HoFH-pivotal and HoFH-extension
`trials. In general, hepatic steatosis persisted, or increased modestly, with continued lomitapide
`treatment.
`
`
`Table 5: Hepatic fat accumulation over time (HoFH-pivotal, HoFH-extension)
`
`
`
`
`
`Source: Primary clinical review (James Smith), Table 89
`
`Categorical maximum changes in hepatic fat observed in the HoFH-pivotal and HoFH-
`extension trials are shown below. Overall, 11 (48%) of 23 patients with hepatic fat data had a
`peak increase in hepatic fat content > 10%.
`
`Table 6: Categorical maximum hepatic fat changes (HoFH-pivotal, HoFH-extension)
`
`Source: Primary clinical review (James Smith), Table 90
`
`Multiple analyses conducted by Dr. Smith indicated that neither the magnitude of LDL-C
`reduction nor aminotransferase elevations could reliably predict the presence or severity of
`hepatic steatosis. Dr. Smith’s analysis of the temporal trend of hepatic fat accumulation in
`affected individuals showed a general trend of initial increase in the first 26 weeks with
`stabilization thereafter in most, but not all, patients. Dr. Smith also concluded that the
`“plateaus or observed reductions in hepatic fat cannot often be explained by a decrease in
`dose.” Off-treatment imaging data available for the 7 HoFH patients who discontinued
`lomitapide indicate reversibility of fat accumulation on imaging weeks to months after drug
`discontinuation.
`
`
`
`
`Reference ID: 3236195
`
`10
`
`Page 11 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`All 6 HoFH patients enrolled in the 16-week HoFH-pilot trial had increased hepatic fat on
`imaging, with peak values ranging from 9% to 44%. These imaging changes returned to
`baseline 4 to 14 weeks after drug discontinuation. In a non-HoFH phase 2 trial evaluating the
`reversibility of fat accumulation, lomitapide treatment (25 mg daily) resulted in a placebo-
`adjusted mean ~20% increase in hepatic fat after 4 weeks of treatment. These imaging
`changes were reversible 6 weeks after drug discontinuation. There were no data on
`reversibility based on histopathology in the lomitapide safety database.
`
`Liver biopsies were not protocol-mandated in lomitapide trials. In the entire clinical safety
`database, “for-cause” liver biopsy results from two patients treated with lomitapide for an
`extended duration were available for review. Biopsy from the patient with the SAE of
`“hepatoxicity” at Week 138 in the HoFH-extension trial showed worsening of simple steatosis,
`but without evidence of inflammation or fibrosis, compared to his biopsy obtained at baseline.
`A 2012 biopsy from a patient with familial chylomicronemia treated with lomitapide for 13
`years in a compassionate care program showed progression of inflammation and fibrosis
`compared to biopsy results obtained in 2008. The role of lomitapide in the patient’s worsening
`liver histopathology could not be discerned in light of her underlying disease.
`
`Our understanding of non-alcoholic fatty liver disease (NAFLD), a primary disease not
`secondary to other known causes of hepatic fat accumulation, sheds some light on the natural
`history of chronic liver injury from hepatic fat accumulation. Simple steatosis of NAFLD
`generally has a benign course, whereas steatosis associated with inflammation and necrosis
`(non-alcoholic steatohepatitis, or NASH) can progress to cirrhosis in up to 10 to 20% of the
`cases. There are no non-invasive biomarkers that could reliably predict the progression from
`simple steatosis to NASH or allow for early detection of NASH.
`
`The risk of chronic liver injury, including steatohepatitis, from lomitapide-induced hepatic
`steatosis is unknown at this time. Whether the clinical course of hepatic steatosis caused by
`lomitapide follows a similar path as NAFLD is uncertain. Published literature on marketed
`drugs implicated in chronic steatosis and steatohepatitis (e.g., amiodarone) is not likely to be
`generalizable, as the risks may be drug-, patient-, or disease-specific. No evidence currently
`exists to inform the routine use of biomarkers or imaging studies to screen for drug-induced
`steatohepatitis or to guide treatment decisions, such as when to contraindicate or discontinue
`treatment.
`
`Regardless of the aforementioned informational gaps, for lomitapide-treated patients, hepatic
`fat could be monitored by imaging studies, and a liver biopsy could be performed to inform
`histopathological changes and provide an opportunity to discontinue treatment prior to the
`development of more serious liver injury. Lomitapide-induced fat accumulation measured by
`imaging study appears to be reversible with drug discontinuation.
`
`• Dietary fat malabsorption
`
`Lomitapide interferes with dietary fat absorption from the intestine by virtue of its mechanism
`of action. The HoFH-pilot trial showed statistically significant reduction in systemic levels of
`fatty acid nutrients at lomitapide doses ≥ 0.3 mg/kg. Subsequently, all patients in the HoFH-
`
`
`
`Reference ID: 3236195
`
`11
`
`Page 12 of 18
`
`PENN EX. 2016
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`NDA 203858/S000 (lomitapide)
`Summary Basis for Regulatory Action
`
`
`
`pivotal trial received dietary supplements containing vitamin E (400 IU), linoleic acid (200
`mg), alpha-linolenic acid (220 mg), EPA (110 mg), and DHA (80 mg). With supplementation,
`the median levels of serum vitamin E, beta-carotene, ALA, linoleic acid, EPA, DHA, and
`arachidonic acid decreased from baseline to Week 26, but remained above the lower limit of
`the reference range, with subsequent stabilization or trends toward baseline.
`
`It appears that dietary supplements, which are available over the counter, provided adequate
`coverage for lomitapide-induced reduced absorption of fat-soluble nutrients in adults, although
`no information is available in pediatric HoFH patients.
`
` •
`
` Preclinical safety signals
`
`
`Teratogenicity: Embryo-fetal developmental studies in rats and ferrets showed major
`teratogenic effects during the organogenesis period at clinically relevant exposures. Fetal
`malformations affecting the eye, brain, and limb, and increased perinatal mortality were also
`observed at relevant human exposure in the peri-and post-natal development toxicity study in
`the rat. The review teams, OSE, and the Pediatric and Maternal Health Team all concluded
`that this risk could be managed with labeling (Pregnancy Category X, and Medication Guide
`to advise patients of this risk with an emphasis on the use of effective contraception) and the
`recommendation that pregnancy should be excluded prior to initiating lomitapide treatment.
`
` I
`
` agree with this approach. The most important risk management strategy lies in the
`prevention of an unintended pregnancy, as 50% of pregnancies in the US are unplanned and
`the teratogenic effects in the preclinical studies occurred during organogenesis, a period where
`most women are often not aware that they are pregnant. Because lomitapide will be used in a
`small at-risk population managed primarily by healthcare providers familiar with prescribing
`products with similar teratogenic profile, requirements such as ongoing pregnancy testing or a
`patient’s attestation of compliance with effective contraception are not warranted at this time.
`
`Neoplasms: A two-year carcinogenicity study in mice showed a statistically significant
`increase in hepatocellular and small intestinal neoplasms at clinically relevant exposures. A
`two-year carcinogenicity study in rats did not demonstrate any drug-related neoplastic
`findings. No malignancies were reported in the small clinical safety database of lomitapide.
`
`Important Clinical Pharmacology findings affectin

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