— ALXN1210 Achieved Non-Inferiority to Soliris® (Eculizumab) on Co-Primary Endpoints of Transfusion Avoidance and Lactate Dehydrogenase Normalization, and All Four Key Secondary Endpoints —

— Safety Profile of ALXN1210 Consistent with That Seen for Soliris®

— Regulatory Submissions Planned in the United States, European Union, and Japan in the Second Half of 2018 —

— Conference Call/Webcast Scheduled for Today, Thursday, March 15, 2018 at 8:30 a.m. EDT —

NEW HAVEN, CT, USA I March 15, 2018 I Alexion Pharmaceuticals, Inc. (NASDAQ: ALXN) announced today that the pivotal Phase 3 study of ALXN1210, the Company’s investigational long-acting C5 complement inhibitor, demonstrated non-inferiority to Soliris® (eculizumab) in complement inhibitor treatment-naïve patients with paroxysmal nocturnal hemoglobinuria (PNH) based on the co-primary endpoints of transfusion avoidance and normalization of lactate dehydrogenase (LDH) levels, a direct marker of complement-mediated hemolysis in PNH. The study also demonstrated non-inferiority on all four key secondary endpoints: percentage change from baseline in LDH levels, change from baseline in quality of life as assessed by the Functional Assessment of Chronic Illness Therapy (FACIT)-Fatigue scale, proportion of patients with breakthrough hemolysis, and proportion of patients with stabilized hemoglobin levels. In addition, numeric results for all six endpoints favored ALXN1210. There were no notable differences in the safety profiles for ALXN1210 and Soliris®.

                     
Endpoint       Treatment effect [95% CI: LB,UB]     Treatment difference
ALXN1210 vs. Soliris®
[95% CI: LB,UB]
    Non-inferiority
      ALXN1210
n=125
    Soliris®
n=121
        Requirement     Achieved*
Primary              
Transfusion avoidance       73.6% [65.9%,81.3%]     66.1% [57.7%,74.6%]     6.8% [-4.7%,18.1%]a     LB > -20%     Yes
LDH normalization       53.6% [45.9%,61.2%]     49.4% [41.7%,57.0%]     1.19 [0.80,1.77]b     LB > 0.39     Yes
Secondary              
Change in LDH levels       -76.8% [-80.0%,-73.7%]     -76.0% [-79.2%,-72.8%]     -0.83% [-5.2%,3.6%]c,†     UB < 20%     Yes
Improvement in FACIT scale       7.1 [5.6,8.6]     6.4 [4.9,8.0]     0.67 [-1.2,2.6]c     LB > -5.0     Yes
Breakthrough hemolysis       4.0% [0.6%,7.4%]     10.7% [5.2%,16.3%]     -6.7% [-14.2%,0.18%]a,†     UB < 20%     Yes
Stabilization of Hb levels       68.0% [59.8%,76.2%]     64.5% [55.9%,73.0%]     2.9% [-8.8%,14.6%]a     LB > -20%     Yes
LDH: lactate dehydrogenase; FACIT: Functional Assessment of Chronic Illness Therapy; Hb: hemoglobin; CI: confidence interval; LB: lower bound; UB: upper bound
*   Non-inferiority is achieved if the LB or UB of the 95% CI of the treatment difference meets the pre-defined requirement. Since non-inferiority was achieved across both co-primary and all four key secondary endpoints, the protocol allowed for superiority testing. Testing for superiority followed a closed-testing procedure, using a 2-sided 0.05 test for each parameter, and followed the pre-specified order per protocol: breakthrough hemolysis, change in LDH levels vs. Baseline, LDH normalization, improvement in FACIT scale, stabilization of Hb levels, transfusion avoidance. As breakthrough hemolysis did not achieve statistical significance (p-value = 0.074), no other endpoints were tested.
a)   Difference in proportion of patients; b) Odds ratio; c) Difference in change vs. Baseline
  Negative value meaning a difference in favor of ALXN1210

Since non-inferiority was achieved across both co-primary and all four key secondary endpoints, the protocol allowed for superiority testing. The hierarchical testing order pre-specified breakthrough hemolysis as the first endpoint tested for superiority. Although ALXN1210 did not achieve superiority, a numeric trend in favor of ALXN1210 was observed for breakthrough hemolysis (4.0% [0.6%,7.4%] vs. 10.7% [5.2%,16.3%] for Soliris®) with a p-value of 0.074. The study also confirmed that ALXN1210 provides immediate and complete (>99%) inhibition of the complement C5 protein that is sustained over the entire 8 week dosing interval. Additionally, treatment with ALXN1210 reduced mean LDH levels to approximately the upper limit of normal (1.0-1.1 times ULN) between months one and six.

“We are very pleased with these positive data for ALXN1210 in the first and only head-to-head study versus Soliris, and the results reinforce our ambition to establish ALXN1210 as the new standard of care for patients with PNH. The data are also consistent with our hypothesis that immediate, complete, and sustained C5 inhibition is critical for patients with this potentially life-threatening disease,” said John Orloff, M.D., Executive Vice President and Head of Research & Development at Alexion. “Soliris has established a high bar for efficacy. Achieving non-inferiority on both co-primary and all key secondary endpoints, as well as seeing numeric results in favor of ALXN1210, in such a rigorous study met a very high hurdle. We look forward to regulatory submissions of ALXN1210 in PNH in the U.S., EU, and Japan in the second half of 2018.”

“Having a new treatment option that achieves transfusion avoidance, and provides rapid and sustained normalization of LDH levels when administered 6 times a year could be a meaningful improvement for patients with PNH who currently need 26 infusions per year,” said Jong Wook Lee, M.D., Professor of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea, and an investigator in the ALXN1210 study.

ALXN1210 was generally well tolerated with a safety profile that is consistent with that seen for Soliris®. The most frequently observed adverse event was headache. The most frequently observed serious adverse event was pyrexia. One patient in the Soliris® arm died from lung cancer (unrelated to Soliris® treatment) during the extension phase of the study. Two patients withdrew from the Soliris® arm for reasons unrelated to treatment. One anti-drug antibody was observed for ALXN1210 and one for Soliris®. No neutralizing antibodies and no apparent effects on efficacy, safety, pharmacokinetics, or pharmacodynamics were detected. There were no cases of meningococcal infection observed in either the ALXN1210 or Soliris® arms. Meningococcal infections are a known risk with terminal complement inhibition, and specific risk-mitigation plans have been in place for ten years for Soliris® to minimize the risk for patients.

Detailed results from this Phase 3 study will be presented at a future medical congress.

About the ALXN1210-PNH-301 Study

This Phase 3, randomized, open-label, active-controlled, multinational, and multicenter study evaluated the efficacy and safety of ALXN1210 compared to Soliris® administered by intravenous (IV) infusion to adult patients (≥ 18 years of age) with PNH who are naïve to complement inhibitor treatment. The study enrolled 246 adult patients with a confirmed diagnosis of PNH who had never been treated with a complement inhibitor and presented with LDH levels ≥ 1.5 times the upper limit of normal (ULN) at the time of screening, as well as with one or more of the following PNH-related signs or symptoms within 3 months of screening: fatigue, hemoglobinuria, abdominal pain, shortness of breath (dyspnea), anemia (hemoglobin <10 g/dL), history of a major adverse vascular event (MAVE, including thrombosis), dysphagia, or erectile dysfunction; or history of packed red blood cell (pRBC) transfusion due to PNH. Patients in the ALXN1210 arm received a single loading dose of ALXN1210, followed by regular maintenance weight-based dosing every 8 weeks. Patients in the Soliris® arm received 4 weekly induction doses, followed by regular maintenance dosing every 2 weeks. Both arms were treated for 26 weeks. The study was designed to evaluate the non-inferiority of ALXN1210 compared to Soliris®.

The co-primary endpoints were the proportion of patients who remain transfusion-free and do not require a transfusion per protocol-specified guidelines through day 183 and the normalization of LDH levels as directly measured every two weeks by LDH levels ≤ 1 times ULN from day 29 through day 183. Key secondary endpoints included the percentage change from baseline in LDH levels to day 183, change from baseline in quality of life as assessed by the FACIT-Fatigue scale to day 183, proportion of patients with breakthrough hemolysis, and proportion of patients with stabilized hemoglobin levels (defined as avoidance of a ≥ 2 g/dL decrease in hemoglobin level from baseline in the absence of transfusion through day 183). Breakthrough hemolysis was defined as at least one new or worsening symptom or sign of intravascular hemolysis: fatigue, hemoglobinuria, abdominal pain, shortness of breath (dyspnea), anemia [hemoglobin < 10 g/dL], MAVE (including thrombosis), dysphagia, or erectile dysfunction in the presence of an elevated LDH level ≥ 2 times ULN, after prior LDH level reduction to < 1.5 times ULN on therapy. Blood samples for the determination of free and total complement C5 protein were collected before and after administration of study drug once a week during the first 4 weeks and every two weeks after that.

All patients enrolled in an extension study of up to 2 years, during which they will receive ALXN1210 every 8 weeks.

About Paroxysmal Nocturnal Hemoglobinuria (PNH)

Paroxysmal nocturnal hemoglobinuria (PNH) is a chronic, progressive, debilitating, and potentially life-threatening ultra-rare blood disorder that can strike men and women of all races, backgrounds, and ages without warning, with an average age of onset in the early 30s.1,2,3 PNH often goes unrecognized, with delays in diagnosis ranging from one to more than 10 years.2 In patients with PNH, chronic, uncontrolled activation of the complement system, a component of the body’s immune system, results in hemolysis (the destruction of red blood cells)4, which in turn can result in progressive anemia, fatigue, dark urine, and shortness of breath.5,6,7 The most devastating consequence of chronic hemolysis is thrombosis (the formation of blood clots), which can damage vital organs and cause premature death.8 Historically, it had been estimated that one in three patients with PNH did not survive more than five years from the time of diagnosis.2 PNH is more common among patients with disorders of the bone marrow, including aplastic anemia (AA) and myelodysplastic syndromes (MDS).9,10,11 In certain patients with thrombosis of unknown origin, PNH may be an underlying cause.4

About ALXN1210

ALXN1210 is an innovative, long-acting C5 inhibitor discovered and developed by Alexion that works by inhibiting the C5 protein in the terminal complement cascade, a part of the body’s immune system that, when activated in an uncontrolled manner, plays a role in severe ultra-rare disorders like paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and anti-acetylcholine receptor (AchR) antibody-positive myasthenia gravis (MG). In a Phase 3 clinical study in complement inhibitor-naïve patients with PNH, intravenous treatment with ALXN1210 every 8 weeks demonstrated non-inferiority to treatment with Soliris® every 2 weeks. ALXN1210 is also currently being evaluated in a Phase 3 clinical study in patients with PNH who have been treated with Soliris® and in a Phase 3 clinical study in complement inhibitor-naïve patients with aHUS, administered intravenously every eight weeks. In addition, Alexion plans to initiate a single, pharmacokinetics (PK)-based Phase 3 clinical study of ALXN1210 delivered subcutaneously once per week as a potential treatment for patients with PNH and aHUS. Alexion also plans to initiate the development of ALXN1210 as a potential treatment for patients with generalized MG (gMG) and patients with immunoglobulin A nephropathy (IgAN).

ALXN1210 has received Orphan Drug Designation (ODD) for the treatment of patients with PNH in the U.S. and EU, and for the subcutaneous treatment of patients with aHUS in the U.S.

About Soliris® (eculizumab)

Soliris® is a first-in-class complement inhibitor that works by inhibiting the C5 protein in the terminal part of the complement cascade, a part of the immune system that, when activated in an uncontrolled manner, plays a role in severe rare and ultra-rare disorders like paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and anti-acetylcholine receptor (AchR) antibody-positive myasthenia gravis (MG). Soliris® is approved in the U.S., EU, Japan, and other countries as the first and only treatment for patients with PNH and aHUS, in the EU as the first and only treatment of refractory generalized MG (gMG) in adults who are anti-AchR antibody-positive, in the U.S. for the treatment of adult patients with gMG who are anti-AchR antibody-positive, and in Japan for the treatment of patients with gMG who are AChR antibody-positive and whose symptoms are difficult to control with high-dose intravenous immunoglobulin (IVIG) therapy or plasmapheresis (PLEX). Soliris® is not indicated for the treatment of patients with Shiga-toxin E. coli-related hemolytic uremic syndrome (STEC-HUS).

Soliris® has received Orphan Drug Designation (ODD) for the treatment of patients with PNH in the U.S., EU, Japan, and many other countries, for the treatment of patients with aHUS in the U.S., EU, and many other countries, for the treatment of patients with MG in the U.S. and EU, and for the treatment of patients with refractory gMG in Japan. Alexion and Soliris® have received some of the pharmaceutical industry’s highest honors for the medical innovation in complement inhibition: the Prix Galien USA (2008, Best Biotechnology Product) and France (2009, Rare Disease Treatment).

For more information on Soliris®, please see full prescribing information for Soliris®, including BOXED WARNING regarding risk of serious meningococcal infection, available at www.soliris.net

About Alexion

Alexion is a global biopharmaceutical company focused on serving patients and families affected by rare diseases through the innovation, development, and commercialization of life-changing therapies. Alexion is the global leader in complement inhibition and has developed and commercializes the first and only approved complement inhibitor to treat patients with paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and anti-acetylcholine receptor (AchR) antibody-positive generalized myasthenia gravis (gMG). In addition, Alexion has two highly innovative enzyme replacement therapies for patients with life-threatening and ultra-rare metabolic disorders, hypophosphatasia (HPP) and lysosomal acid lipase deficiency (LAL-D). As the leader in complement biology for over 20 years, Alexion focuses its research efforts on novel molecules and targets in the complement cascade, and its development efforts on the core therapeutic areas of hematology, nephrology, neurology, and metabolic disorders. This press release and further information about Alexion can be found at: www.alexion.com.

References
___________________
1   Hill A, Richards SJ, Hillmen P. Recent developments in the understanding and management of paroxysmal nocturnal haemoglobinuria. Br J Haematol. 2007 May;137(3):181-92.
2   Hillmen P, Lewis SM, Bessler M, et al. Natural history of paroxysmal nocturnal hemoglobinuria. NEngl J Med. 1995 Nov 9;333(19):1253-8.
3   Socié G, Mary JY, de Gramont A, et al. Paroxysmal nocturnal haemoglobinuria: long-term follow-up and prognostic factors. Lancet. 1996;348:573-577.
4   Hill A, Kelly RJ, Hillmen P. Thrombosis in paroxysmal nocturnal hemoglobinuria. Blood. 2013;121:4985-4996.
5   Nishimura J, Kanakura Y, Ware RE, et al. Clinical course and flow cytometric analysis of paroxysmal nocturnal hemoglobinuria in the United States and Japan. Medicine (Baltimore) 2004 May;83(3):193-207.
6   Weitz I, Meyers G, Lamy T, et al. Cross-sectional validation study of patient-reported outcomes in patients with paroxysmal nocturnal haemoglobinuria. Intern Med J. 2013;43:298-307.
7   Parker C, Omine M, Richards S, et al. Diagnosis and management of paroxysmal nocturnal hemoglobinuria. Blood. 2005 Dec 1;106(12):3699-709.
8   Hillmen P, Muus P, Duhrsen U, et al. Effect of the complement inhibitor eculizumab on thromboembolism in patients with paroxysmal nocturnal hemoglobinuria. Blood. 2007 Dec 1;110(12):4123-8.
9   Wang H, Chuhjo T, Yasue S, et al. Clinical significance of a minor population of paroxysmal nocturnal hemoglobinuria-type cells in bone marrow failure syndrome. Blood. 2002;100 (12):3897-3902.
10   Iwanga M, Furukawa K, Amenomori T, et al. Paroxysmal nocturnal haemoglobinuria clones in patients with myelodysplastic syndromes. Br J Haematol. 1998;102(2):465-474.
11   Maciejewski JP, Rivera C, Kook H, et al. Relationship between bone marrow failure syndromes and the presence of glycophosphatidyl inositol-anchored protein-deficient clones. Br J Haematol. 2001;115:1015-1022.

SOURCE: Alexion Pharmaceuticals