throbber
Articles
`
`Effi cacy and safety of a microsomal triglyceride
`transfer protein inhibitor in patients with homozygous
`familial hypercholesterolaemia: a single-arm, open-label,
`phase 3 study
`
`Marina Cuchel, Emma A Meagher, Hendrik du Toit Theron, Dirk J Blom, A David Marais, Robert A Hegele, Maurizio R Averna, Cesare R Sirtori,
`Prediman K Shah, Daniel Gaudet, Claudia Stefanutti, Giovanni B Vigna, Anna M E Du Plessis, Kathleen J Propert, William J Sasiela,
`LeAnne T Bloedon, Daniel J Rader, for the Phase 3 HoFH Lomitapide Study investigators
`
`Summary
`Background Patients with homozygous familial hypercholesterolaemia respond inadequately to existing drugs. We
`aimed to assess the effi cacy and safety of the microsomal triglyceride transfer protein inhibitor lomitapide in adults
`with this disease.
`
`Methods We did a single-arm, open-label, phase 3 study of lomitapide for treatment of patients with homozygous
`familial hypercholesterolemia. Current lipid lowering therapy was maintained from 6 weeks before baseline through
`to at least week 26. Lomitapide dose was escalated on the basis of safety and tolerability from 5 mg to a maximum of
`60 mg a day. The primary endpoint was mean percent change in levels of LDL cholesterol from baseline to week 26,
`after which patients remained on lomitapide through to week 78 for safety assessment. Percent change from baseline
`to week 26 was assessed with a mixed linear model.
`
`Findings 29 men and women with homozygous familial hypercholesterolaemia, aged 18 years or older, were recruited
`from 11 centres in four countries (USA, Canada, South Africa, and Italy). 23 of 29 enrolled patients completed both the
`effi cacy phase (26 weeks) and the full study (78 weeks). The median dose of lomitapide was 40 mg a day. LDL cholesterol
`was reduced by 50% (95% CI –62 to –39) from baseline (mean 8·7 mmol/L [SD 2·9]) to week 26 (4·3 mmol/L [2·5];
`p<0·0001). Levels of LDL cholesterol were lower than 2·6 mmol/L in eight patients at 26 weeks. Concentrations of LDL
`cholesterol remained reduced by 44% (95% CI –57 to –31; p<0·0001) at week 56 and 38% (–52 to –24; p<0·0001) at
`week 78. Gastrointestinal symptoms were the most common adverse event. Four patients had aminotransaminase
`levels of more than fi ve times the upper limit of normal, which resolved after dose reduction or temporary interruption
`of lomitapide. No patient permanently discontinued treatment because of liver abnormalities.
`
`Interpretation Our study suggests that treatment with lomitapide could be a valuable drug in the management of
`homozygous familial hypercholesterolaemia.
`
`Funding FDA Offi ce of the Orphan Product Development, Aegerion Pharmaceuticals.
`
`Introduction
`Homozygous familial hypercholesterolaemia is a life-
`threatening disease most commonly caused by loss-of-
`function mutations in both alleles of the LDL receptor
`gene. Mutations in other genes, including APOB,
`PCSK9, and autosomal recessive hypercholesterolaemia
`LDLRAP1, which alter the function of the LDL receptor
`or its ligand ApoB, could also contribute to such a
`phenotype. As a consequence of impaired LDL-receptor
`function, untreated total plasma cholesterol levels are
`typically greater than 13 mmol/L, resulting in premature
`and progressive atherosclerosis often leading to cardio-
`vascular disease before age 20 years and death before age
`30 years.1–3 Early initiation of aggressive treatment for
`these patients is, therefore, essential.4
`Patients with homozygous familial hypercholester-
`olaemia respond inadequately to conventional drug
`therapies,2,5–7 which generally reduce LDL cholesterol
`
`through upregulation of hepatic LDL receptors. There fore,
`the current standard of care for familial hyperchol-
`esterolaemia includes LDL apheresis, which transiently
`reduces LDL cholesterol by more than 50%8,9 and can delay
`the onset and progression of atherosclerosis.7–9 However,
`even with the combined use of available drug therapies
`and apheresis, these patients still have substantially
`elevated levels of LDL cholesterol and persistently high
`risk of cardiovascular disease.10 Liver transplantation has
`also been done in patients with this disease.11,12 In recent
`years, alternative therapeutic approaches have been
`developed that target either ApoB synthesis13 or the
`production of VLDL, the precursor of LDL.14
`Lomitapide (Aegerion Pharmaceuticals, Cambridge,
`MA, USA) is an inhibitor of the microsomal triglyceride
`transport protein (MTP), a key protein in the assembly and
`secretion of ApoB-containing lipoproteins in the liver and
`intestine.15 The drug substantially reduced levels of LDL
`
`www.thelancet.com Vol 381 January 5, 2013
`
`Lancet 2013; 381: 40–46
`Published Online
`November 2, 2012
`http://dx.doi.org/10.1016/
`S0140-6736(12)61731-0
`See Comment page 7
`Institute for Translational
`Medicine and Therapeutics,
`Cardiovascular Institute, and
`Department of Medicine,
`University of Pennsylvania,
`Philadelphia, PA, USA
`(M Cuchel MD, E A Meagher MD,
`L T Bloedon MS,
`Prof K J Propert ScD,
`Prof D J Rader MD);
`Netcare Private Hospital,
`Bloemfontein, South Africa
`(Prof H du Toit Theron MD);
`Department of Medicine
`(D J Blom PhD) and Department
`of Chemical Pathology
`(Prof A D Marais MD), University
`of Cape Town, Cape Town,
`South Africa; Medical Research
`Council of South Africa, Cape
`Heart Group, Cape Town, South
`Africa (D J Blom, A D Marais);
`Robarts Research Institute and
`Schulich School of Medicine
`and Dentistry, University of
`Western Ontario, London, ON,
`Canada (Prof R A Hegele FRCP);
`Università di Palermo, Palermo,
`Italy (Prof M R Averna MD);
`Ospedale Niguarda, Milano,
`Italy (Prof C R Sirtori MD);
`Division of Cardiology and
`Atherosclerosis Research
`Center, Cedars-Sinai Heart
`Institute, Los Angeles, CA, USA
`(Prof P K Shah MD); Department
`of Medicine, Université de
`Montreal, Chicoutimi, Quebec,
`Canada (D Gaudet MD);
`Extracorporeal Therapeutic
`Techniques Unit,
`Immunohematology and
`Transfusion Medicine,
`Department of Molecular
`Medicine, University of Rome
`‘Sapienza’, Italy
`(C Stefanutti MD); Department
`of Clinical and Experimental
`
`40
`
`Page 1 of 7
`
`PENN EX. 2007
`CFAD V. UPENN
`IPR2015-01835
`
`

`
`Articles
`
`Medicine, Università of Ferrara,
`Italy (G B Vigna MD); Clinical
`Research Unit, University of
`Pretoria, Pretoria, South Africa
`(A M E Du Plessis MMed); and
`Aegerion Pharmaceuticals,
`Cambridge, MA, USA
`(L T Bloedon, W J Sasiela PhD)
`Correspondence to:
`Dr Marina Cuchel, University of
`Pennsylvania, Perelman School
`of Medicine, Institute for
`Translational Medicine and
`Therapeutics, 3600 Spruce
`Street, Maloney Building,
`Room 8039, PA 19104, USA
`mcuchel@mail.med.upenn.edu
`
`cholesterol in the Watanabe Heritable Hyperlipidaemic
`rabbit, an animal model of homozygous familial hyper-
`cholester olaemia.16 We have shown that lomitapide given
`orally for 16 weeks as monotherapy was eff ective in
`reducing LDL cholesterol levels in six patients with homo-
`zygous familial hypercholesterolaemia and that its effi cacy
`was mediated by a reduction in LDL production.14 To assess
`the long-term safety and effi cacy of lomitapide when added
`to currently available lipid-lowering drug therapy with or
`without apheresis (standard of care), we assessed adult
`patients with homozygous familial hyper cholester olaemia
`over a 78 week treatment period. Safety assessments
`included an analysis of the eff ects of chronic MTP
`inhibition on the liver.
`
`Methods
`Study design and patients
`In our phase 3, open-label study, patients were re-
`cruited from 11 centres in four countries (USA, Canada,
`South Africa, and Italy). Diagnostic criteria for homo-
`zygous familial hyperchol esterolaemia were based either
`on clinical criteria (history of untreated total cholesterol
`>13 mmol/L and triglycerides <3·4 mmol/L and both
`parents with history of untreated total cholesterol
`>6·5 mmol/L) or on documented mutation(s) in both
`alleles of the LDL receptor or of other genes known to
`aff ect LDL receptor function. Exclusion criteria included:
`major surgery in the previous 3 months, congestive heart
`failure, history of liver disease or transaminases greater
`than twice the upper limit of normal (ULN), serum
`creatinine >221 μmol/L, recent malignancy, alcohol or
`drug abuse, known bowel disease or malabsorption, or
`chronic lung disease.
`Patients were screened for eligibility 12 weeks before
`the fi rst dose of lomitapide. Screening procedures
`included medical and drug history, review of current
`lipid-lowering therapies, physical examination, vital
`signs, 12-lead electrocardiograph (ECG), fasting lipid
`panel, safety laboratory assessments, and dietary coun-
`selling. All enrolled patients were required to enter a
`minimum 6-week run-in phase during which concomi-
`tant lipid-lowering therapies, including apheresis, the
`daily dietary supplementation of vitamin E, and essential
`fatty acids were initiated, and the required low-fat diet
`was stabilised. At the end of the run-in phase, patients
`entered a 26-week effi cacy phase, during which they
`received lomitapide in addition to their current lipid-
`lowering therapy. Lomitapide was initiated at a starting
`dose of 5 mg a day for the fi rst 2 weeks and then escalated
`to 10, 20, 40, and 60 mg a day at 4-week intervals or until
`an individually determined maximum dose was achieved
`on the basis of safety and tolerability. Patients remained
`at their maximum dose through to the end of the 26-week
`effi cacy phase. A fasting lipid and safety panel, including
`liver function tests, was obtained at baseline, before each
`dose escalation, and then every 4 weeks through to week
`26 (primary endpoint).
`
`After completion of the effi cacy phase, patients
`continued to receive lomitapide and entered a 52-week
`safety phase (weeks 26–78) during which concomitant
`lipid-lowering therapies, including LDL apheresis, could
`be modifi ed at the investigators discretion. Assessments
`during this phase were done every 5 weeks to 10 weeks
`and at the end of treatment. Total treatment duration was
`78 weeks. Eligible patients completing the treatment
`phase were off ered the option to enter a separate long-
`term study, in which they continued to receive lomitapide.
`Patients who did not enter the long-term study dis-
`continued lomitapide at week 78 and returned for a fi nal
`follow-up visit at week 84.
`If patients had confi rmed alanine transaminase (ALT)
`or aspartate transaminase (AST) elevations between
`5·0 and 9·9 times the ULN, or >100 U/L but <200 U/L
`above the baseline value, the dose of lomitapide was
`reduced to the previously tolerated dose level, with the
`possibility to re-escalate once transaminase elevations
`were resolved. Adverse events were coded with MedDRA
`(version 11.0). These events were
`judged by the
`investigators as: not related, unlikely, possibly, probably,
`or defi nitely related to study drug, and were reviewed
`regularly by an independent data and safety monitoring
`board. The study was approved by each institution’s
`review board or ethics committee and all patients
`provided written, informed consent.
`
`Procedures
`Blood was drawn at baseline and at each visit following a
`12 h fast. Routine testing included a standard metabolic
`panel, a complete blood count, urinalysis, and measure-
`ment of fat soluble vitamins and fatty acids. All testing
`was done at a US Centers for Disease Control and
`Prevention standardised lipid central laboratory (PPD,
`Highland Heights, KY, USA and Brussels, Belgium) or
`referred to a partnering laboratory for the measurement
`of vitamin K and essential fatty acids. In patients
`undergoing apheresis, samples for the fasting lipid
`profi le were obtained shortly before the scheduled
`apheresis treatment. The timing of treatments (eg, every
`14 days) and study blood sampling was maintained
`throughout the study so that lipid assessments would be
`done at the same point on the LDL cholesterol rebound
`curve. Lipid and lipoprotein analyses were done with
`serum. Total cholesterol, directly measured LDL chol-
`esterol and HDL cholesterol, and triglycerides were
`measured enzymatically. Non-HDL cholesterol and
`VLDL cholesterol were calculated. ApoA-I and ApoB were
`measured by immunonephelometry.
`Hepatic lipid content was assessed by nuclear magnetic
`resonance spectroscopy (NMRS) studies at baseline and
`at 6-month intervals. All quantitative measurements
`were done by a single external radiologist who was
`masked to patients’ clinical status and results of liver
`function tests. NMRS was not done in three patients who
`had contraindications to MRI. In these patients a CT
`
`www.thelancet.com Vol 381 January 5, 2013
`
`41
`
`Page 2 of 7
`
`PENN EX. 2007
`CFAD V. UPENN
`IPR2015-01835
`
`

`
`Articles
`
`See Online for appendix
`
`scan or ultrasound was done at the discretion of the local
`physician or if recommended by the data and safety
`monitoring board.
`
`Statistical analysis
`The sample size calculation was based on an assumption
`of a 25% change from baseline in LDL cholesterol at
`week 26 with a 30% SD and 15% dropout rate. Using an
`alpha of 0·05 with 90% power, 20 patients were needed.
`The statistical analyses were done with SAS software
`(version 9.1). Continuous variables were summarised by
`descriptive statistics (sample size, mean, SD, median,
`minimum and maximum). Categorical variables were
`summarised by frequency (N) and percentages (%).
`Baseline values of lipid parameters were the average of
`two measurements taken 2 weeks apart (after 4 weeks and
`6 weeks of entering the run-in phase). The primary
`effi cacy endpoint measure was the percent change from
`baseline in concentration of LDL cholesterol at the
`maximum tolerated dose after 26 weeks of treatment.
`Prespecifi ed secondary endpoints
`included percent
`changes in other lipid parameters, long-term safety, and
`changes in hepatic-fat content. All patients who received
`at least one dose of the study drug were in the assess-
`ment of the primary and secondary endpoints (intention-
`to-treat analysis) up to the end of the effi cacy phase
`(week 26). Signifi cance of the percent changes in LDL
`cholesterol from baseline to 26 weeks was assessed with a
`mixed linear model, which assumes a missing-at-random
`mechanism. An additional secondary statistical analysis
`was done imputing missing data with the last-observation-
`carried-forward method, because this was the statistical
`approach described in the original statistical analysis
`plan. Further secondary effi cacy and safety analyses were
`done during the safety phase (weeks 26–78); an on-sample
`t test was used to assess percent change from baseline at
`
`week 56 and week 78. Correlations were assessed with
`Spearman’s rank-correlation. Statistical signifi cance was
`defi ned as p≤0·05.
`This study
`is registered with ClinicalTrials.gov
`(NCT00730236).
`
`Role of the funding source
`The sponsor of the study had no role in study design, data
`collection, primary data analysis, data interpretation, or
`initial writing of the report, but was invited to comment
`on the written report. The corresponding author had full
`access to all the data in the study and had fi nal
`responsibility for the fi nal content of the report and the
`decision to submit for publication.
`
`Results
`Of the 32 patients with homozygous familial hyper-
`cholesterolaemia who were screened for eligibility,
`31 entered the run-in period and 29 men and women
`were enrolled in the effi cacy phase. All patients were
`18 years or older and met diagnostic criteria. 23 of
`29 enrolled patients completed both the effi cacy phase
`(26 weeks) and the full study (78 weeks; appendix). Six
`patients discontinued the study during the effi cacy
`phase (the fi rst 4 days after enrolment and the last at
`week 22): four patients discontinued because of adverse
`events (three were gastrointestinal events and one was
`headache); one was withdrawn for non-compliance
`with the protocol; and one withdrew consent for
`personal reasons.
`The baseline characteristics of the patients enrolled
`in the study are shown in the appendix. Briefl y, all
`29 patients were either homozygotes or compound
`heterozygotes for mutations in the LDLR gene or
`genes aff ecting LDL-receptor functionality. 27 patients
`were treated with statins, primarily rosuvastatin or
`
`Baseline (n=29) Week 26 (n=23)
`
`Concen trations
`
`Change from
`baseline (%)
`
`Week 56 (n=23)
`
`p value†
`
`Concen trations
`
`Change from
`baseline (%)
`
`Week 78 (n=23)
`
`p value‡
`
`Concen trations
`
`Change from
`baseline (%)
`
`p value‡
`
`11·1 (3·5)
`
`6·1 (2·9)
`
`–46% (–56 to –35) <0·0001
`
`7·1 (3·7)
`
`–39% (–51 to –27) <0·0001
`
`7·3 (3·9)
`
`–35% (–48 to –22) <0·0001
`
`8·7 (2·9)
`
`4·3 (2·5)
`
`–50% (–62 to –39)
`
`<0·0001
`
`5·1 (3·2)
`
`–44% (–57 to –31) <0·0001
`
`5·4 (3·4)
`
`–38% (–52 to –24)
`
`0·0001
`
`0·5 (0·3)
`
`0·3 (0·3)
`
`–45% (–61 to –29) <0·0001
`
`0·4 (0·4)
`
`–28% (–48 to –10)
`
`0·0185
`
`0·4 (0·4)
`
`–31% (–54 to –7)
`
`0·0389
`
`10·0 (3·4)
`
`5·1 (2·8)
`
`–50% (–61 to –39) <0·0001
`
`5·9 (3·6)
`
`–44% (–57 to –31) <0·0001
`
`6·2 (3·8)
`
`–39% (–53 to –25) <0·0001
`
`1·0 (0·4 to 2·9) 0·5 (0·1 to 1·7)
`2·6 (0·8)
`1·3 (0·7)
`2·4 (0·6 to 2·1)
`1·7 (0·3 to 7·1)
`1·1 (0·3)
`1·0 (0·4)
`
`–45% (–61 to –29) <0·0001
`–49% (–60 to –38)
`<0·0001
`–15% (–30 to 0·9)
`0·0003
`–12% (–20 to –4)
`0·0001
`
`0·7 (0·2 to 2·9)
`1·5 (0·8)
`2·0 (0·5 to 8·6)
`1·2 (0·4)
`
`0·0157
`–29% (–47 to –11)
`–45% (–57 to –33) <0·0001
`–19% (–31 to –8)
`0·0111
`1% (–13 to 15)
`0·954
`
`0·7 (0·2 to 4·1)
`1·5 (0·9)
`2·6 (0·6 to 7·0)
`1·1 (0·3)
`
`0·0368
`–31% (–54 to –8)
`–43% (–56 to –29) <0·0001
`–1% (–17 to 6)
`0·5827
`–5% (–13 to 3)
`0·1396
`
`1·2 (0·3)
`
`1·0 (0·2)
`
`–14% (–17 to –4)
`
`0·0003
`
`1·1 (0·3)
`
`1% (–11 to 13)
`
`0·568
`
`1·1 (0·3)
`
`–4% (–10 to 3)
`
`0·1155
`
`Total cholesterol,
`mmol/L
`LDL cholesterol,
`mmol/L
`VLDL cholesterol,
`mmol/L
`Non-HDL cholesterol,
`mmol/L
`Triglycerides, mmol/L
`ApoB, g/L
`Lipoprotein (a), μmol/L
`HDL cholesterol,
`mmol/L
`ApoA-I, g/L
`
`Data are mean (SD), median (range) for triglycerides and lipoprotein (a) at baseline, weeks 26, 56, and 78, or mean (95% CI) for percent change. †p values from mixed model. ‡p values from one-sample t test.
`
`Table: Lipid and lipoprotein concentrations at baseline and weeks 26, 56, and 78 (end of study)
`
`42
`
`www.thelancet.com Vol 381 January 5, 2013
`
`Page 3 of 7
`
`PENN EX. 2007
`CFAD V. UPENN
`IPR2015-01835
`
`

`
`Articles
`
`6
`
`8
`
`10
`
`14
`12
`Study week
`
`16
`
`18
`
`20
`
`22
`
`24
`
`26
`
`LDL cholesterol
`Total cholesterol
`ApoB
`2
`
`4
`
`20
`
`0
`
`–20
`
`–40
`
`–60
`
`–80
`
`Change from baseline (%)
`
`–100
`
`0
`
`Figure 1: Mean percent changes in LDL cholesterol, total cholesterol, and ApoB levels from baseline to
`week 26 (end of effi cacy phase)
`Data available at each time point are expressed as mean (SD).
`
`Alanine transaminase
`Aspartate transaminase
`
`B
`
`2
`
`6 10 14 18 22 26 31 36 41 46 51 56 66 78
`Study week
`
`Baseline
`
`Week 26
`
`Week 56
`
`Week 78
`
`A
`100
`90
`80
`70
`60
`50
`40
`30
`20
`10
`0
`
`B
`25
`
`20
`
`15
`
`10
`
`05
`
`Transaminases (IU/L)
`
`Hepatic fat (%)
`
`Figure 2: Alanine transaminase and aspartate transaminase levels and
`percentage of hepatic fat in the liver
`Data are mean, 95% CI. Laboratory reference ranges for alanine transaminase
`levels were 10–40 U/L in men and 10–33 U/L in women; reference ranges for
`aspartate transaminase levels were 10–43 U/L in men and 10–36 U/L in
`women (A). Percentage of fat in the liver, as measured by nuclear magnetic
`resonance spectroscopy at baseline and 26, 56, and 78 weeks of lomitapide
`treatment (n=20; B).
`
`treatment with lomitapide were gastro intestinal in nature
`(27 patients during the effi cacy phase, and 17 during the
`safety phase). The three patients who discontinued the
`study because of gastrointestinal disorders permanently
`stopped lomitapide by week 12 (appendix). No patients
`died during the study. Three (10%) of 29 patients had
`serious adverse events: one had acute coronary syndrome
`and angina pectoris and lower respiratory tract infection,
`one had elective hysterectomy for menorrhagia, and one
`had chest pain. All serious adverse events were assessed as
`
`atorvastatin, 22 with ezetimibe (all in combination with a
`statin), three with niacin, one with a fi brate, and one with
`a bile acid sequestrant. 18 patients regularly underwent
`apheresis with a frequency that ranged from weekly to
`every 6 weeks. Despite aggressive lipid lowering treat-
`ment, total cholesterol, LDL cholesterol, and ApoB were
`substantially elevated at baseline (table).
`Compliance with study drug dosing, defi ned as more
`than 80% of capsules taken, was 28 (93%) during the
`effi cacy phase and 22 (95%) during the safety phase. Of
`the six patients who discontinued lomitapide treatment,
`two were receiving 5 mg, two were receiving 10 mg, one
`was receiving 20 mg, and one was receiving 40 mg.
`Among the 23 patients who completed the study, the
`maximum dose was 5 mg in one patient; 20 mg in fi ve;
`40 mg in six, and 60 mg in 11 at the end of the effi cacy
`phase. The dose distribution remained similar at week 78.
`Mean levels of LDL cholesterol remained stable during
`the run-in phase, as shown by a mean percent change
`from screening in LDL cholesterol of –1·20% (95% CI
`–15·66 to 13·18) at week 0. Mean percent changes in LDL
`cholesterol during the effi cacy phase are shown in
`fi gure 1. Mean LDL cholesterol signifi cantly decreased by
`50% from baseline to the end of the effi cacy phase
`(week 26; table). Percent changes from baseline for key
`secondary endpoints (total cholesterol, ApoB, and tri-
`glycerides) were consistent with those for LDL cholesterol
`at week 26 (table). Analysis done with the last observation
`carried forward gave similar results.
`Overall, 19 of 23 patients with data at week 26 had
`decreased concentrations of LDL cholesterol of more
`than 25% with 12 having more than a 50% reduction.
`Eight patients had LDL cholesterol levels lower than
`2·6 mmol/L at week 26, with one having levels lower
`than 1·8 mmol/L. On the basis of LDL cholesterol
`response, three patients permanently discontinued LDL
`apheresis and three permanently increased the time
`interval between apheresis treatments at some point
`during the safety phase (weeks 26–78). Lomitapide
`signifi cantly reduced LDL cholesterol at week 78, despite
`changes in concomitant lipid lowering therapy or any
`adjustment in lomitapide dose (table). Similar effi cacy
`results were reported for total cholesterol, ApoB, and
`triglycerides (table). Lipoprotein (a) levels were sig nifi -
`cantly reduced from baseline at week 26 and 56, but were
`not sig nifi cantly diff erent at week 78 (table).
`Concentrations of HDL cholesterol were signifi cantly
`reduced at week 26, and mirrored the reduction in the
`levels of ApoA-I (table). HDL cholesterol and ApoA-I
`returned to levels similar to those at baseline by week 78
`(table).
`A summary of adverse events reported during the
`effi cacy and safety phase is shown in the appendix.
`Most patients had at least one adverse event during both
`the effi cacy (27 of 29 patients) and safety (21 of 23) phases.
`Most adverse events were assessed as mild to moderate in
`intensity. The most commonly reported events during
`
`www.thelancet.com Vol 381 January 5, 2013
`
`43
`
`Page 4 of 7
`
`PENN EX. 2007
`CFAD V. UPENN
`IPR2015-01835
`
`

`
`Articles
`
`Panel: Research in context
`
`Systematic review
`We searched PubMed for intervention studies of homozygous familial
`hypercholesterolaemia between January, 1980, and August, 2012. Patients with this rare
`disease have untreated cholesterol levels greater than 13 mmol/L. Drug-based treatments
`were scarcely eff ective until the introduction of HMG-CoA reductase inhibitors (statins).
`Patients with homozygous familial hypercholesterolaemia have an inadequate response to
`existing lipid-lowering drug therapies such as statins and ezetimibe7,17–19 and remain at very
`high risk for cardiovascular events and mortality. Treatment at high doses of atorvastatin
`and rosuvastatin results in about 27% reduction in LDL cholesterol.19 Addition of ezetimibe
`to statin treatment can result in an additional 20% reduction in LDLcholesterol.7 Apheresis
`treatment can acutely lower LDL cholesterol levels by 70–80% and result in a time-average
`reduction by 40–50% when done regularly.20 A phase 3, randomised study assessing the
`effi cacy of an anti-ApoB antisense oligonucleotide, mipomersen, showed a reduction in
`LDL cholesterol of about 25% in patients with homozygous familial hypercholesterolaemia
`treated with maximum-tolerated lipid-lowering drug therapy.13
`
`Interpretation
`Our study expands the results obtained in a previous phase 2 study.14 We report that
`lomitapide, when given in addition to currently available lipid-lowering therapy, results in
`an additional 50% reduction in LDL cholesterol, potentially bringing these high-risk
`patients closer to target levels. The limitations due to the single-arm, open-label design
`and the safety considerations of potential dose-related transaminase elevations, and
`liver-fat accumulation are counterbalanced and outweighed by the signifi cant LDL
`cholesterol-lowering eff ects of lomitapide in this severe disorder of unmet medical need.
`Our study suggests that treatment with lomitapide could be a valuable drug in the
`management of homozygous familial hypercholesterolaemia.
`
`unrelated or unlikely related to study treatment. No serious
`adverse events were reported between weeks 26 and 78.
`Ten patients had elevated levels of ALT, AST, or both of
`more than three times the ULN at least once during the
`study (fi gure 2). Four of these patients had ALT increases
`more than fi ve times the ULN and one patient had a
`similar elevation in AST; these elevations occurred at
`lomitapide doses of 10 mg, 20 mg, 40 mg, and 60 mg. No
`patient discontinued treatment permanently because
`of elevations in liver-function-test parameters and all
`elevations were managed either by dose reduction or
`temporary inter ruption of lomitapide as per protocol. Of
`note, three of four patients with elevations of more than
`fi ve times the ULN in liver-function-test parameters
`reported consuming quantities of alcohol higher than
`those allowed per protocol. No patient had elevations in
`bilirubin or alkaline phosphatase levels.
`Hepatic fat was measured non-invasively with NMRS.
`Mean hepatic fat in the 20 patients with evaluable NMRS
`scans was 1·0% (range 0–5·0) at baseline, 8·6% (0–33·6)
`at week 26, 5·8% (0–16·5%) at week 56, and 8·3%
`(0–19·0%) at week 78 (fi gure 2). Percent change in
`hepatic fat was negatively associated with change in LDL
`cholesterol. This association was signifi cant at week 26
`(r=–0·50, 95% CI –0·76 to –0·09; p=0·0161) and week 56
`(r=–0·55, –0·79 to –0·15; p=0·0083), but was not signifi -
`cant at week 78 (r=–0·21, –0·59 to 0·25; p=0·3618).
`
`Discussion
`Our open-label study shows that lomitapide, admin-
`istered concurrently with background lipid-lowering
`therapies including LDL apheresis, sig nifi cantly reduced
`LDL cholesterol in patients with homozygous familial
`hypercholesterolaemia. This reduction is similar to that
`reported during lomitapide monotherapy in patients
`with the disorder,14 and shows that lomitapide had
`similar effi cacy when added to existing concomitant
`treatment (panel).
`While studies of cardiovascular outcome are not
`feasible in view of the rarity of homozygous familial
`hypercholesterolaemia, retrospective studies show that
`even a modest reduction in LDL cholesterol, either by
`pharmacological intervention or LDL apheresis, results
`in apparent improvement in morbidity and mortality.6,8,9,21
`Furthermore, observa tional studies clearly show that
`patients with homozygous familial hyper cholesterol-
`aemia and some LDL-receptor function (receptor-
`defective) have lower levels of LDL cholesterol and better
`prognosis than those with no LDL-receptor function
`(receptor-negative).4 Thus, although we are unable to
`provide direct evidence,
`the magnitude of LDL
`cholesterol reduction with lomitapide would be expected
`to reduce cardiovascular risk and improve survival.
`Reduction of LDL cholesterol levels was somewhat
`attenuated at the end of the study. This eff ect could be
`explained by the changes during the safety phase that
`were made in apheresis treatment or in concomitant
`lipid lowering therapy in some of the better responders,
`as well as reductions in lomitapide dose in some of the
`patients that had elevated liver enzymes or gastro-
`intestinal tolerability issues.
`We noted a signifi cant decrease in lipoprotein (a) levels
`at week 26, that persisted up to week 56. The mechanism
`underlying this eff ect is not known, but a similar fi nding
`has been reported with other drugs aff ecting the
`secretion of ApoB-containing lipoproteins by the liver.13
`The reason for loss of signifi cance in lipoprotein (a)
`reduction at week 78 is not clear. Lipoprotein (a) levels
`are substantially aff ected by apheresis treatment,22,23 thus
`changes in apheresis treatment that were allowed during
`the safety phase could have confounded the eff ect on
`lipoprotein (a). Further studies are needed to test this
`hypothesis and clarify these fi ndings.
`HDL cholesterol and ApoA-I levels were transiently
`decreased during the effi cacy phase, a fi nding reported in
`previous studies with lomitapide.14,24 The mechanism(s)
`underlying these changes are not known and further
`studies will be necessary to explain this eff ect. Possible
`reasons might include the low-fat diet or the inhibitory
`eff ects of lomitapide on dietary fat absorption; the
`reduced secretion of triglyceride-rich lipoproteins, which
`carry ApoA-I, from the gut or liver, as a direct consequence
`of MTP inhibition; or a reduction in ApoA-I production.
`The decrease in levels of HDL cholesterol occurred
`during the titration period, when the dose was gradually
`
`44
`
`www.thelancet.com Vol 381 January 5, 2013
`
`Page 5 of 7
`
`PENN EX. 2007
`CFAD V. UPENN
`IPR2015-01835
`
`

`
`Articles
`
`increased, and subsequently returned to levels approach-
`ing those at baseline once the dose was stabilised,
`suggesting the existence of a compensatory mechanism.
`The clinical implications of this temporary reduction in
`levels of HDL cholesterol are unknown.
`Our study was the fi rst long-term study of any MTP
`inhibitor in human beings, and safety and tolerability
`were carefully assessed. Lomitapide, initiated at a low
`dose and escalated to an individualised maximum dose in
`the presence of a low-fat diet, was generally well tolerated.
`All three discontinuations due to gastrointestinal events
`occurred during the titration phase. The incidence and
`the number of patients who experienced gastrointestinal
`events improved during the safety phase suggesting that
`patients become more tolerant or learnt to control their
`diet better, similar to patients with abetalipoproteinaemia.15
`Indeed, of the 23 patients who completed the effi cacy
`phase, all 23 remained on lomitapide for another
`12 months and completed the entire protocol. Investi-
`gators and patients were aware of the lomitapide dose and
`the lipid response because of the open-label design of the
`study, therefore, we cannot exclude the possibility that
`this factor infl uenced the reporting and assessment of
`adverse events.
`Accumulation of liver fat is intrinsically linked to the
`mechanism of action of MTP inhibitors, and has been
`the basis of concerns regarding the clinical use of this
`class of agents. The 18-month duration of this study
`aff orded the fi rst opportunity to assess the eff ect of
`chronic MTP inhibition on liver safety and liver fat.
`While ALT levels more than three times the ULN were
`seen in ten of 29 patients, these changes were generally
`transient or resolved with dose reduction and were not
`associated with elevated bilirubin or alkaline phosphatase
`or evidence of impaired synthetic function.
`As expected, mean hepatic fat increased from 1·0% to
`8·6% at week 26, but no further increase was reported
`for the remainder of the study. Since the clinical
`signifi cance and long-term implications of the increase
`in hepatic fat as a result of lomitapide therapy is not
`clearly understood, rigorous and standardised long-term
`monitoring will be necessary.
`Our study has several limitations that need to be taken
`into account when interpreting the results. The study was
`a non-randomised, open-label study. Since homo zygous
`familial hypercholesterolaemia is a rare disease, our
`intent was to expose the maximum number of patients to
`treatment for the duration of the study so that safety
`(especially the potential liver adverse events) could be
`assessed fully. Furthermore, in view of the striking
`changes in LDL cholesterol and ApoB that were reported
`in the phase 2 study14 we expected to be able to easily
`discern the eff ect of lomitap

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