Join today and have your say! It’s FREE!

Become a member today, It's free!

We will not release or resell your information to third parties without your permission.
Please Try Again
{{ error }}
By providing my email, I consent to receiving investment related electronic messages from Stockhouse.

or

Sign In

Please Try Again
{{ error }}
Password Hint : {{passwordHint}}
Forgot Password?

or

Please Try Again {{ error }}

Send my password

SUCCESS
An email was sent with password retrieval instructions. Please go to the link in the email message to retrieve your password.

Become a member today, It's free!

We will not release or resell your information to third parties without your permission.

Data Published In The New England Journal Of Medicine Demonstrate Naloxegol Improved Opioid-Induced Constipation In Chronic Pain Patients

AZNCF

LONDON, June 19, 2014 /PRNewswire/ -- AstraZeneca today announced that the New England Journal of Medicine (NEJM) has published results of two pivotal Phase III studies – KODIAC-4 and KODIAC-5 of naloxegol, an investigational treatment for opioid-induced constipation (OIC).

Opioids play an important role in chronic pain relief by binding to mu-receptors in the brain, but they also bind to mu-receptors in the bowel. That is why patients taking opioids for chronic pain can develop OIC. In fact, the incidence of OIC varies and has been reported as high as 81%* in patients taking opioids. Naloxegol, which has the potential to be the first FDA approved once-daily oral treatment for patients with OIC, is a peripherally-acting mu-opioid receptor antagonist (PAMORA) studied in adult patients with chronic non-cancer pain experiencing OIC.

**Primary endpoint data from the KODIAC-4 and -5 studies showed that more OIC patients treated with naloxegol 25 mg had a consistent response of increased spontaneous bowel movements (SBMs) through 12 weeks of treatment compared to placebo [44% vs. 29% (p=0.001 KODIAC-4) and 40% vs. 29% (p=0.021 KODIAC-5)]. The 12.5 mg dose in KODIAC-5 did not show statistical significance for the primary endpoint.

The 25 mg dose also demonstrated a higher response rate through 12 weeks of treatment compared to placebo in patients with laxative inadequate response (LIR), a secondary endpoint. Results for an additional secondary endpoint showed that patients taking naloxegol 25 mg in the KODIAC-4 and KODIAC-5 studies were likely to have a first post-dose spontaneous bowel movement 25-30 hours sooner than placebo, respectively (median six and 12 hours for naloxegol 25 mg compared to 36 and 37 hours for placebo, in studies KODIAC-4  and -5, respectively). 

"An estimated 235 million prescriptions for opioids are written in the US each year, of which 20% are for chronic pain. For patients taking prescription opioids for chronic pain, constipation is one of the most common and bothersome side effects, and these patients can experience sub-optimal relief from laxatives," said William Chey, MD, AGAF, FACG, FACP, Professor of Medicine at the University of Michigan Health System. "These results demonstrated the potential for naloxegol, if approved, to be a new treatment option as the studies showed rapid and sustained improvement for these patients, without compromising their pain management over the course of the trials."

The Phase III studies, KODIAC-4 (n=652) and KODIAC-5 (n=700), were 12-week, multicenter, randomised, double blind, placebo-controlled pivotal trials that evaluated 12.5 mg and 25 mg doses of naloxegol, administered once-daily.

Additional results from the KODIAC-4 and KODIAC -5 clinical trials published in NEJM included:

  • The number of SBMs per week increased with naloxegol 25 mg treatment over 12 weeks, with both studies showing an improvement in treatment effect versus placebo
  • Improvements in straining, stool consistency, and frequency of days with complete SBMs were observed with naloxegol 25 mg (both studies)
  • The most commonly reported adverse effects with naloxegol were gastrointestinal in origin (abdominal pain, diarrhea, nausea, vomiting, flatulence) and appeared to be dose-ordered, occurring more commonly in the 25 mg group. Most adverse events were mild to moderate in severity and occurred shortly after initiation of naloxegol
  • There was 1 major adverse cardiovascular event (MACE) in the 25 mg treatment arm, 1 in the 12.5 mg treatment arm and 2 in the placebo arm

A New Drug Application (NDA) for naloxegol was accepted by the US Food and Drug Administration on 19 November 2013. MOVANTIK is the proposed proprietary name for naloxegol. On 11-12 June 2014, the FDA convened the Anesthetic Analgesic Drug Products Advisory Committee (AADPAC) to review the class of peripherally acting opioid receptor antagonists. The Committee voted that the FDA should not require cardiovascular outcomes trials for the PAMORA class of drugs which includes MOVANTIK. Following a clarification of the vote, the majority of the Committee suggested continued post-approval data collection for cardiovascular safety. The Prescription Drug User Fee Act (PDUFA) date set by the FDA for MOVANTIK is 16 September 2014.

MOVANTIK is also under regulatory review with health agencies in the European Union and Canada.

NOTES TO EDITORS
*Published estimates of the incidence of OIC in patients receiving opioids for chronic pain vary due to differences in the studies conducted (eg, study design, definition of constipation, opioids used). Meta-analyses of randomized controlled trials suggest that 15%–41% of these patients develop OIC, while observational and survey based studies suggest that 37%–81% develop OIC.

**The primary endpoint in both trials was percentage of OIC responders, versus placebo, over 12 weeks of treatment.

About naloxegol
Naloxegol is an investigational peripherally-acting mu-opioid receptor antagonist (PAMORA), which has been specifically designed for the treatment of opioid-induced constipation, a condition caused by prescription opioid pain medicines. In the Phase III clinical studies, naloxegol was administered as a once-daily tablet and is designed to block the binding of opioids to the opioid receptors in the gastrointestinal (GI) tract without impacting the opioid receptors in the brain.

Naloxegol is part of the exclusive worldwide license agreement announced on 21 September 2009, between AstraZeneca and Nektar Therapeutics. Naloxegol was developed using Nektar's oral small molecule polymer conjugate technology.

About Opioid-Induced Constipation
Opioids play an important role in chronic pain relief by binding mu-receptors in the brain.  But they also bind mu-receptors in the bowel. That is why patients taking opioids for chronic pain can develop opioid-induced constipation (OIC). In fact, the incidence of OIC varies and has been reported as high as 81%* in patients taking opioids.

About AstraZeneca
AstraZeneca is a global, innovation-driven biopharmaceutical business that focuses on the discovery, development and commercialisation of prescription medicines, primarily for the treatment of cardiovascular, metabolic, respiratory, inflammation, autoimmune, oncology, infection and neuroscience diseases. AstraZeneca operates in over 100 countries and its innovative medicines are used by millions of patients worldwide. For more information please visit: www.astrazeneca.com

CONTACTS

Media Enquiries
Esra Erkal-Paler    +44 20 7604 8030 (UK/Global)
Ayesha Bharmal    +44 20 7604 8034 (UK/Global)
Michele Meixell      +1 302 885 2677 (US)
Jacob Lund            +46 8 553 260 20 (Sweden)

Investor Enquiries
Karl Hård              +44 20 7604 8123 mob: +44 7789 654364
Colleen Proctor    +44 207 604 8128    mob: +1 302 357 4882
Anthony Brown    +44 20 7604 8067    mob: +44 7585 404943
Jens Lindberg      +44 20 7604 8414    mob: +44 7557 319729

SOURCE AstraZeneca