برجاء الإنتظار ...

Search Results



نشرة الممارس الصحي نشرة معلومات المريض بالعربية نشرة معلومات المريض بالانجليزية صور الدواء بيانات الدواء
  SFDA PIL (Patient Information Leaflet (PIL) are under review by Saudi Food and Drug Authority)

What Praluent is
• Praluent contains the active substance alirocumab.
• Praluent is a monoclonal antibody (a type of specialised protein designed to attach to a target substance in the body). Monoclonal antibodies are proteins that recognise and bind to other unique proteins. Alirocumab binds to PCSK9.
How Praluent works
Praluent helps lower your levels of “bad” cholesterol (also called “LDL cholesterol”). Praluent blocks a protein called PCSK9.
• PCSK9 is a protein secreted by liver cells.
• “Bad” cholesterol is normally removed from your blood by binding to specific “receptors” (docking stations) in your liver.
• PCSK9 lowers the number of these receptors in the liver – this causes your “bad” cholesterol to be higher than it should.
• By blocking PCSK9, Praluent increases the number of receptors available to help remove the “bad” cholesterol – this lowers your “bad” cholesterol levels.
What Praluent is used for
• Adults with high cholesterol levels in their blood (hypercholesterolaemia, heterozygous familial and non-familial, or mixed dyslipidaemia). It is given:
- together with a statin (a commonly used medicine that treats high cholesterol) or other cholesterol lowering medicines, if the maximum dose of a statin does not lower levels of cholesterol sufficiently or,
- alone or together with other cholesterol lowering medicines when statins are not tolerated or cannot be used.
• Continue to follow your cholesterol-lowering diet while taking this medicine.


Do not use Praluent
• if you are allergic to alirocumab or any of the other ingredients of this medicine (listed in section 6).
Warnings and precautions
Talk to your doctor, pharmacist or nurse about all your medical conditions, including allergies, before using Praluent.
If you develop a serious allergic reaction, stop using Praluent, talk to your doctor right away. Sometimes serious allergic reactions such as hypersensitivity (difficulties breathing), nummular eczema (reddish skin spots sometimes with blisters), and hypersensitivity vasculitis (which is a specific form of a hypersensitivity reaction with symptoms such as diarrhoea, with a rash, or purple-coloured skin spots on the skin) have occurred in clinical studies. For allergic reactions that may occur while taking Praluent, see section 4.
Tell your doctor if you have kidney or liver disease before using this medicine, because Praluent has been studied in few patients with severe kidney disease and not in patients with severe liver disease.
Children and adolescents
Praluent is not recommended for children and adolescents under 18 years old. This is because there is no experience of using the medicine in these age groups.
Other medicines and Praluent
Tell your doctor, pharmacist or nurse if you are using, have recently used or might use any other medicines.
Pregnancy and breast-feeding
Praluent is not recommended during pregnancy or breast-feeding.
If you are pregnant or breast-feeding, think you may be pregnant or are planning to have a baby, ask your doctor or pharmacist for advice before using this medicine.
Driving and using machines
This medicine is not expected to have any effect on your ability to drive or use machines.


Always use this medicine exactly as your doctor, pharmacist or nurse has told you. Check with your doctor, pharmacist or nurse if you are not sure.
How much to inject
Your doctor will tell you which dose is right for you and how often to inject (75 mg or 150 mg once every 2 weeks, or 300 mg once every 4 weeks/monthly). Your doctor will check your cholesterol levels and may adjust the dose (up or down) during treatment.
Always check the label of your syringe to make sure you have the right medicine and the right strength.
When to inject
Inject Praluent once every 2 weeks (for the 75 mg or 150 mg dose), or once every 4 weeks/monthly (for the 300 mg dose). To give the 300 mg dose, give two 150 mg injections in a row at two different injection sites.
Before you inject
Read the detailed instructions for use leaflet before you inject Praluent.

Where to inject
Read the detailed instructions for use leaflet on where to inject.
Learning how to use the pre-filled syringe
Before you use the syringe for the first time, your doctor, pharmacist or nurse will show you how to inject Praluent.
• Always read the "Instructions for Use" provided in the box.
• Always use the syringe as described in the “Instructions for Use”.
If you use more Praluent than you should
If you use more Praluent than you should, talk to your doctor, pharmacist or nurse.
If you forget to use Praluent
If you miss a dose of Praluent, inject your missed dose as soon as you can. Then take your next dose at your regular scheduled time. This will keep you on the original schedule. If you are not sure when to inject Praluent, call your doctor, pharmacist or nurse.
If you stop using Praluent
Do not stop using Praluent without talking with your doctor. If you stop using Praluent, your cholesterol levels can increase.
If you have any further questions on the use of this medicine, ask your doctor, pharmacist or nurse.


Like all medicines, this medicine can cause side effects, although not everybody gets them.
If you develop a serious allergic reaction, stop using Praluent , talk to your doctor right away. Sometimes serious allergic reactions such as hypersensitivity (difficulties breathing), nummular eczema (reddish skin spots sometimes with blisters), and hypersensitivity vasculitis (which is a specific form of a hypersensitivity reaction with symptoms such as diarrhoea, with a rash, or purple-coloured skin spots on the skin) have occurred (may affect up to 1 in 1,000 people).
Other side effects are:
Common (may affect up to 1 in 10 people)
• redness, itching, swelling, pain/tenderness where the medicine was injected (local injection site reactions)
• upper respiratory tract signs or symptoms such as sore throat, running nose, sneezing
• itching (pruritus).
Rare (may affect up to 1 in 1,000 people)
• red and itchy raised bumps or hives (urticaria)
Reporting of side effects
The National Pharmacovigilance and Drug Safety Centre (NPC)
Fax: +966-11-205-7662
Call NPC at +966-11-2038222, Exts: 2317-2356-2353-2354-2334-2340.
Toll free phone: 8002490000
E-mail: npc.drug@sfda.gov.sa
Website: www.sfda.gov.sa/npc


Keep this medicine out of the sight and reach of children.
Do not use this medicine after the expiry date which is stated on the label and carton after EXP. The expiry date refers to the last day of that month.
Store in a refrigerator (2°C to 8°C). Do not freeze.
Keep the syringe in the outer carton in order to protect from light.
If needed, individual pre-filled syringes may be kept outside the refrigerator below 25°C for a maximum of 30 days. Protect from light. After removal from the refrigerator, Praluent must be used within 30 days or discarded.
Do not use this medicine if it looks discoloured or cloudy, or if it contains visible flakes or particles.
After use put the syringe into a puncture-resistant container. Always keep the container out of the sight and reach of children. Ask your doctor, pharmacist or nurse how to throw away the container. Do not recycle the container.
Do not throw away any medicines via wastewater or household waste.
Ask your pharmacist how to throw away medicines you no longer use. These measures will help protect the environment.


What Praluent contains
• The active substance is alirocumab. Each single-use syringe contains either 75 milligrams (75 milligrams per ml) or 150 milligrams (150 milligrams per ml) of alirocumab.
• The other ingredients are histidine, sucrose, polysorbate 20 and water for injection.


Praluent is a clear, colourless to pale yellow solution for injection that comes in a pre-filled syringe. Each pre-filled syringe with green plunger contains 1 ml of solution, delivering one single dose of 75 milligrams. It is available in pack size of 1, 2 or 6 pre-filled syringes. Each pre-filled syringe with grey plunger contains 1 ml of solution, delivering one single dose of 150 milligrams. It is available in pack size of 1, 2 or 6 pre-filled syringes. Not all presentations and pack sizes may be marketed.

Marketing Authorisation Holder
sanofi-aventis groupe
54, rue La Boétie
F – 75008 Paris
France
Manufacturer
Sanofi Winthrop Industrie
1051 Boulevard Industriel
76580 Le Trait
France


November 2016
  نشرة الدواء تحت مراجعة الهيئة العامة للغذاء والدواء (اقرأ هذه النشرة بعناية قبل البدء في استخدام هذا المنتج لأنه يحتوي على معلومات مهمة لك)

الفئة الدوائیّة العلاجیةّ
- یحتوي برالوینت على المادة الفاعلة ألیروكوماب.
- برالوینت ھو جسم مضاد أحادي المنشأ( نوع من البروتین الخاصمصمّم لیرتبط بمادة مستھدفة في الجسم ) الأجسام المضادة
PCSK الأحادیّة المنشأ ھي بروتینات تتعرّف إلى بروتینات أخرى فریدة وترتبط بھا .الألیروكوماب یرتبط ببروتین. 9
دواعي الاستعمال العلاجیةّ:
یساعد برالوینت على تخفیضمستویات الكولسترول" الضار( "المسمّى أیضًا" الكولسترول الخفیضالكثافة .)"یحصر برالوینت بروتیناً
PCSK یُسمّى. 9
ھو بروتین تفرزه خلایا الكبد. PCSK9 -
- عادة ما تتمّ إزالة الكولسترول" الضار "من جسمك عبر الارتباط ب"مستقبلات "محددة( محطاّت استقبال )في كبدك.
عدد ھذه المستقبلات في الكبد مما یرفع مستوى الكولسترول الضار إلى مستوى أعلى مما PCSK - یخفضّبروتین 9
یجب أن یكون علیھ.
یزید برالوینت عدد المستقبلات المتوافرة للمساعدة على إزالة الكولسترول" الضار " ، PCSK - عبر حصر بروتین 9
مما یخفضّمستویات الكولسترول" الضار "لدیك.
- لدى البالغین الذین لدیھم مستویات مرتفعة من الكولسترول في الدم( فرط كولسترول الدم، عسر شحمیاّت الدم
المتخالف العائلي أو غیر العائلي أو المختلط)، یُعطى:
مع دواء ستاتین( دواء یسُتعمل بصورة شائعة لعلاج مستوى الكولسترول المرتفع )أو أدویة أخرى تخفضّ o
الكولسترول، إذا كانت الجرعة القصوى من دواء ستاتین لا تخفضّمستویات الكولسترول بما فیھ الكفایة، أو
لوحده أو بالتزامن مع أدویة أخرى مخفضّة للكولسترول عندما لا یتمّ تحمّل أدویة الستاتین أو لا یمكن o
استعمالھا .
واصل اتباع حمیتك الغذائیّة المخفضّة للكولسترول بینما تستعمل ھذا الدواء o

موانع الاستعمال
لا تستعمل برالوینت
.( - إذا كنت مصاباً بحساسیةّ ضدّ الألیروكوماب أو ضدّ أيّ مركّب آخر في ھذا الدواء( المركّبات مذكورة في القسم ٦
محاذیر خاصة للاستعمال؛ تحذیرات خاصة
تحدّث إلى الطبیب أو الصیدليّ أو الممرّضحول مشاكلك الصحیةّ بما فیھا الحساسیاّت التي تعاني منھا، قبل
أن تأخذ برالوینت.
إذا أصبت بارتكاس تحسسي خطیر، توقفّ عن استعمال برالوینت وتحدّث إلى طبیبك على الفور .أحیاناً حدثت في الدراسات السریریّة
ارتكاسات تحسسیةّ خطیرة مثل فرط التحسس( صعوبة في التنفسّ )والأكزیما القرصیّة( بقع جلدیّة ضاربة إلى الحمرة تأتي أحیاناً مع
بثور )والتھاب فرط التحسس الوعائي( وھو شكل محدّد من الارتكاسات التحسسیةّ مع عوارضمثل الإسھال ومع طفح أو بقع أرجوانیةّ
اللون على الجلد.)
للارتكاسات التحسسیةّ التي قد تحصل في خلال فترة أخذ برالوینت، راجع القسم. 4
أعلم الطبیب إذا كنت تعاني من مرضكلوي أو كبدي قبل استعمال ھذا الدواء، لأنھّ تمّت دراسة برالوینت لدى عدد قلیل من المرضى
یعانون من مرضكلوي ولم تتمّ دراستھ لدى مرضى یعانون من مرضكبدي.
الأطفال والمراھقون
لا یوصى باستعمال برالوینت لدى الأطفال والمراھقین ما دون 18 عامًا، بسبب عدم وجود خبرة حول استعمال الدواء لدى الفئات العمریةّ
ھذه.
الأدویة الأخرى وبرالوینت
أعلم الطبیب أو الصیدلي أو الممرّضإذا كنت تستعمل حالیاً أو استعملت مؤخّرًا أو قد تكون تستعمل أدویة أخرى.
الحمل والإرضاع
لا یوصى باستعمال برالوینت في خلال فترتيّ الحمل أو الإرضاع.
إذا كنتِ حاملا أًو مرضعة أو كنت تعتقدین نفسك حاملا أًو كنتِ تنوین الحمل، استشیري الطبیب أو الصیدلي قبل استعمال ھذا الدواء.
قیادة السیّارات واستعمال الآلات
لا یُتوقعّ أن یكون لھذا الدواء أيّ تأثیر على قدرتك على القیادة أو على استعمال الآلات.

https://localhost:44358/Dashboard

استعمل ھذا الدواء دائمًا حسب تعلیمات الطبیب أو الصیدلي أو الممرّضتمامًا .إسأل الطبیب أو الصیدلي أو الممرّضإذا لم تكن متأكّدًا.
ما ھي الكمیةّ الواجب حقنھا
سوف یُعلمك طبیبك بالجرعة المناسبة لك ۷٥ ملغ أو ۱٥۰ ملغ مره كل أسبوعین، أو ۳۰۰ ملغ مرة كل اربع اسابیع شھریا .)
سوف یتحقق طبیبك من مستویات الكولسترول لدیك وقد یضبط الجرعة( زیادة أو نقصاناً )في خلال مدّة العلاج.
تحقق دائمًا من لصاقة محقنتك للتاكّد من أنّ لدیك الدواء الصحیح والعیار الصحیح.
متى یجب حقن الدواء
یحقن مره واحده كل اسبوعین ( لجرعھ ۷٥ ملغ و ۱٥۰ ملغ ) او مره واحده كل اربع اسابیع /شھریا (لجرعة ۳۰۰ ملغ ).
لحقن جرعة ۳۰۰ ملغ یحقن جرعتین من حقنة ۱٥۰ ملغ في مكانین مختلفین.
قبل أن تحقن الدواء
إقرأ نشرة تعلیمات الاستعمال المفصّلة قبل أن تحقن برالوینت.
أین یجب حقن الدواء
إقرأ نشرة تعلیمات الاستعمال المفصّلة حول مكان الحقن.
تعلمّ كیفیّة استعمال المحقنة المعبأّة مسبقًا
قبل أن تستعمل المحقنة للمرّة الأولى، سوف یریك الطبیب أو الصیدلي أو الممرّضكیفیةّ حقن برالوینت.
- إقرأ دائمًا قسم" تعلیمات الاستعمال "الوارد في العلبة.
- استعمل المحقنة دائمًا على النحو الموصوف في" تعلیمات الاستعمال."
إذا استعملت كمیةّ مفرطة من برالوینت
إذا استعملت كمیةّ مفرطة من برالوینت، أعلم الطبیب أو الصیدلي أو الممرّض.
إذا نسیت استعمال برالوینت
إذا فوّت جرعة من برالوینت، احقن الجرعة التي فوّتھا في أقرب وقت ممكن .ومن ثمّ خذ جرعتك التالیة بعد
أسبوعین من تاریخ تفویتك جرعتك .مثلا،ً إذا كنت تحقن الدواء عادة مرّة كلّ أسبوعین یوم الثلاثاء، واصل
الحقن مرّة كلّ أسبوعین یوم الثلاثاء .فھذا سوف یبقیك متبعًا برنامج العلاج الأساسي .إذا لم تكن واثقاً من
الوقت الذي یجب علیك أن تحقن جرعتك فیھ، اتصل بالطبیب أو بالصیدلي أو بالممرّض.
إذا توقفّت عن استعمال برالوینت
لا تتوقفّ عن استعمال برالوینت بدون التحدّث إلى طبیبك .إذا توقفت عن استعمال برالوینت، یمكن أن ترتفع
مستویات الكولسترول لدیك.
إذا كان لدیك أيّ أسئلة أخرى حول استعمال ھذا الدواء، اطرحھا على الطبیب أو الصیدلي أو الممرّض.

مثل الأدویة كلھّا یمكن أن یسببّ ھذا الدواء تأثیرات جانبیةّ لا تصیب المرضى كلھّم.
إذا أصبت بارتكاس تحسسي خطیر، توقفّ عن استعمال برالوینت وتحدّث إلى طبیبك على الفور .حصلت أحیاناً ارتكاسات تحسسیةّ خطیرة
مثل فرط التحسس( صعوبة في التنفسّ )والأكزیما القرصیّة( بقع جلدیّة ضاربة إلى الحمرة تأتي أحیاناً مع بثور )والتھاب فرط التحسس
الوعائي( وھو شكل محدّد من الارتكاسات التحسسیةّ یأتي مع عوارضمثل الإسھال ومع طفح أو بقع أرجوانیةّ اللون على الجلد) (قد
تصیب حتىّ شخصواحد من أصل 1000 شخص.)
التأثیرات الجانبیةّ الأخرى ھي:
الشائعة( قد تصیب حتىّ شخصواحد من أصل 10 أشخاص)
- احمرار، حكّة، تورّم، ألم/إیلام في موقع حقن الدواء( ارتكاسات موضعیةّ في مكان الحقن)
- علامات أو عوارضفي الجھاز التنفسّي العلوي مثل التھاب الحلق وسیلان الأنف والعطاس
- حكّة( الحكاك.)
النادرة( قد تصیب حتىّ شخصواحد من أصل 1000 شخص)
- نتوءات حمراء تسببّ الحكّة أو شرى
الإبلاغ عن الاعراض الجانبیة :
المركز الوطني للتیقظ والسلامة الدوائیة
+۹٦٦-۱۱-۲۰٥- فاكس ۲٦٦۷
للإتصال بالإدارة التنفیذیة للتیقظ وادارة الأزمات .
۲۳۳٤-۲۳٥٤-۲۳۱۷-۲۳٥٦-۲۳٥۳- ۹٦٦ + تحویلھ ۲۳٤۰ -۱۱-۲۰۳- ھاتف ۲۲۲۸
الھاتف المجاني ۸۰۰۲٤۹۰۰۰۰
npc.drug@sfda.gov.sa البرید الألكتروني
www.sfda.gov.sa/npc الموقع الألكتروني
سانوفي - التیقظ الدوائي:
KSA_Pharmacovigilance@sanofi.com

إحفظ الدواء بعیدًا عن نظر الأطفال ومتناولھم.
EXP . لا تستعمل ھذا الدواء بعد تاریخ انتھاء صلاحیتّھ المدوّن على اللصاقة وعلبة الكرتون بعد كلمة
یشیر تاریخ انتھاء الصلاحیةّ إلى الیوم الأخیر من الشھر المذكور.
إحفظ الدواء في البرّاد( بین درجتین مئویتّین و 8 درجات مئویةّ .)لا تجمّده.
لا تترك برالوینت خارج البرّاد لأكثر من 24 ساعة في حرارة ما دون 25 درجة مئویةّ( لا تحفظ الدواء في حرارة تتخطىّ 25 درجة
مئویةّ.)
احفظ المحقنة في علبة الكرتون الخارجیّة لحمایتھا من الضوء.
لا تستعمل ھذا الدواء إذا تغیرّ لونھ أو بدا عكرًا أو إذا كان یحتوي على رقائق أو جسیمات.
بعد الاستعمال، ضع المحقنة في حاویة مقاومة للثقب .إحفظ الحاویة دائمًا بعیدًا عن نظر الأطفال ومتناولھم.
إسأل الطبیب أو الصیدلي أو الممرّضحول كیفیةّ التخلّصمن الحاویة .لا تعد تدویر الحاویة.
لا ینبغي رمي الأدویة في مجاري الصرف الصحّي أو مع النفایات المنزلیّة العادیّة.
إسأل الصیدلي عن كیفیةّ التخلّصمن الأدویة التي لم تعد بحاجة إلیھا، فمن شأن ھذه الإجراءات حمایة البیئة.

- المادة الفاعلة ھي ألیروكوماب .تحتوي على 75 ملیغرام 75 ( میلیغرام بالمللیلتر )من الألیروكوماب.
- المركّبات الأخرى ھي ھیسیدیتین، سكروز، بولیسوربات 20 وماء للحقن.

برالوینت ھو محلول صافٍ وعدیم اللون إلى أصفر باھت للحقن، یأتي في محقنة معبأّة مسبقاً. تحتوي كلّ محقنة معبأّة مسبقاً مع مكبس أخضر على 1 مل من المحلول، وتعطي جرعة وحیدة من 75 ملیغرام. یأتي الدواء في علبة من محقنة واحدة معبأّة مسبقاً أو من محقنتین معبأّتین مسبقاً أو من 6 محاقن معبأّة مسبقأً. قد لا تكون أحجام العلب مسوّقة في بلدك.

الشركة الحاملة رخصة التسویق
sanofi-aventis groupe
54, rue La Boétie
F – 75008 Paris
France
الشركة المصنّعة
Sanofi Winthrop Industrie
1051 Boulevard Industriel
76580 Le Trait
France

تشرین الثاني ۲۰۱٦
 Read this leaflet carefully before you start using this product as it contains important information for you

Praluent 75 mg solution for injection in pre-filled pen Praluent 150 mg solution for injection in pre-filled pen Praluent 75 mg solution for injection in pre-filled syringe Praluent 150 mg solution for injection in pre-filled syringe

75 mg/ml solution for injection Each single-use pre-filled pen contains 75 mg alirocumab in 1 ml solution. Each single-use pre-filled syringe contains 75 mg alirocumab in 1 ml solution. 150 mg/ml solution for injection: Each single-use pre-filled pen contains 150 mg alirocumab in 1 ml solution. Each single-use pre-filled syringe contains 150 mg alirocumab in 1 ml solution. Alirocumab is a human IgG1 monoclonal antibody produced in Chinese Hamster Ovary cells by recombinant DNA technology. For the full list of excipients, see section 6.1.

Solution for injection (injection) Clear, colourless to pale yellow solution.

Primary hypercholesterolaemia and mixed dyslipidaemia
Praluent is indicated in adults with primary hypercholesterolaemia (heterozygous familial and non-familial)
or mixed dyslipidaemia, as an adjunct to diet:
- in combination with a statin or statin with other lipid lowering therapies in patients unable to reach
LDL-C goals with the maximum tolerated dose of a statin or,
- alone or in combination with other lipid-lowering therapies in patients who are statin-intolerant, or for whom a statin is contraindicated.
Established atherosclerotic cardiovascular disease
Praluent is indicated in adults with established atherosclerotic cardiovascular disease to reduce cardiovascular risk by lowering LDL-C levels, as an adjunct to correction of other risk factors:

- in combination with the maximum tolerated dose of a statin with or without other lipid-lowering therapies or,
- alone or in combination with other lipid-lowering therapies in patients who are statin-intolerant, or for whom a statin is contraindicated.
For study results with respect to effects on LDL-C, cardiovascular events and populations studied see section
5.1.


Posology
Prior to initiating Praluent secondary causes of hyperlipidaemia or mixed dyslipidaemia (e.g., nephrotic syndrome, hypothyroidism) should be excluded.
The usual starting dose for Praluent is 75 mg administered subcutaneously once every 2 weeks. Patients requiring larger LDL-C reduction (>60%) may be started on 150 mg once every 2 weeks, or 300 mg once every 4 weeks (monthly), administered subcutaneously.
The dose of Praluent can be individualised based on patient characteristics such as baseline LDL-C level, goal of therapy, and response. Lipid levels can be assessed 4 to 8 weeks after treatment initiation or titration, and dose adjusted accordingly (up-titration or down-titration). If additional LDL-C reduction is needed in patients treated with 75 mg once every 2 weeks or 300 mg once every 4 weeks (monthly), the dosage may be adjusted to the maximum dosage of 150 mg once every 2 weeks.
If a dose is missed, the patient should administer the injection as soon as possible and thereafter resume treatment on the original schedule.
Special populations
Paediatric population
The safety and efficacy of Praluent in children and adolescents less than 18 years of age have not been established. No data are available.
Elderly
No dose adjustment is needed for elderly patients.
Hepatic impairment
No dose adjustment is needed for patients with mild or moderate hepatic impairment. No data are available in patients with severe hepatic impairment.
Renal impairment
No dose adjustment is needed for patients with mild or moderate renal impairment. Limited data are
available in patients with severe renal impairment (see section 5.2).
Body weight
No dose adjustment is needed in patients based on weight.
Method of administration
Subcutaneous use.
Praluent is injected as a subcutaneous injection into the thigh, abdomen or upper arm.
To administer the 300 mg dose, give two 150 mg injections consecutively at two different injection sites.

It is recommended to rotate the injection site with each injection.
Praluent should not be injected into areas of active skin disease or injury such as sunburns, skin rashes, inflammation, or skin infections.
Praluent must not be co-administered with other injectable medicinal products at the same injection site. The patient may either self-inject Praluent, or a caregiver may administer Praluent, after guidance has been
provided by a healthcare professional on proper subcutaneous injection technique.
Precautions to be taken before handling
Praluent should be allowed to warm to room temperature prior to use. (See section 6.6) Each pre-filled pen or pre-filled syringe is for single use only.


Hypersensitivity to the active substance or to any of the excipients listed in section 6.1.

Allergic reactions
General allergic reactions, including pruritus, as well as rare and sometimes serious allergic reactions such as hypersensitivity, nummular eczema, urticaria, and hypersensitivity vasculitis have been reported in clinical studies (see section 4.8). If signs or symptoms of serious allergic reactions occur, treatment with Praluent must be discontinued and appropriate symptomatic treatment initiated (see section 4.3).
Renal impairment
In clinical studies, there was limited representation of patients with severe renal impairment (defined as eGFR < 30 mL/min/1.73 m2) (see section 5.2). Praluent should be used with caution in patients with severe renal impairment.
Hepatic impairment
Patients with severe hepatic impairment (Child-Pugh C) have not been studied (see section 5.2). Praluent should be used with caution in patients with severe hepatic impairment.


Effects of alirocumab on other medicinal products
Since alirocumab is a biological medicinal product, no pharmacokinetic effects of alirocumab on other medicinal products and no effect on cytochrome P450 enzymes are anticipated.
Effects of other medicinal products on alirocumab
Statins and other lipid-modifying therapy are known to increase production of PCSK9, the protein targeted by alirocumab. This leads to the increased target-mediated clearance and reduced systemic exposure of alirocumab. Compared to alirocumab monotherapy, the exposure to alirocumab is about 40%, 15%, and 35% lower when used concomitantly with statins, ezetimibe, and fenofibrate, respectively. However, reduction of LDL-C is maintained during the dosing interval when alirocumab is administered every two weeks.


Pregnancy
There are no data from the use of Praluent in pregnant women. Alirocumab is a recombinant IgG1 antibody, therefore it is expected to cross the placental barrier (see section 5.3). Animal studies do not indicate direct or indirect harmful effects with respect to maintenance of pregnancy or embryo-fetal development; maternal toxicity was noted in rats, but not in monkeys at doses in excess of the human dose, and a weaker secondary immune response to antigen challenge was observed in the offspring of monkeys (see section 5.3). The use of Praluent is not recommended during pregnancy unless the clinical condition of the woman requires treatment with alirocumab.
Breast-feeding
It is not known whether alirocumab is excreted in human milk. Human immunoglobulin G (IgG) is excreted in human milk, in particular in colostrum; the use of Praluent is not recommended in breast-feeding women during this period. For the remaining duration of breast-feeding, exposure is expected to be low. Since the effects of alirocumab on the breast-fed infant are unknown, a decision should be made whether to discontinue nursing or to discontinue Praluent during this period.
Fertility
In animal studies, there were no adverse effects on surrogate markers of fertility (see section 5.3). There are no data on adverse effects on fertility in humans.


Praluent has no or negligible influence on the ability to drive and use machines.


Summary of the safety profile
In ten phase 3 controlled trials, involving patients with primary hypercholesterolemia and mixed dyslipidaemia, the most common adverse reactions were local injection site reactions, upper respiratory tract signs and symptoms, and pruritus. Most common adverse reactions leading to treatment discontinuation in patients treated with Praluent were local injection site reactions.
The only adverse reaction identified in ODYSSEY OUTCOMES (a long-term cardiovascular outcome trial)
was injection site reaction.
The safety profile in ODYSSEY OUTCOMES was consistent with the overall safety profile described in the phase 3 controlled trials.
No difference in the safety profile was observed between the two doses (75 mg and 150 mg) used in the phase 3 program.
Tabulated list of adverse reactions
The following adverse reactions were reported in patients treated with alirocumab in pooled controlled studies and/or post-marketing use (see Table 1).
Frequencies for all adverse reactions identified from clinical trials have been calculated based on their incidence in pooled phase 3 clinical trials. Adverse reactions are presented by system organ class. Frequency categories are defined as: Very common (≥1/10); common (≥1/100 to <1/10); uncommon (≥1/1,000 to <1/100); rare (≥1/10,000 to <1/1,000); very rare (<1/10,000) and not known (cannot be estimated from the available data).
The frequency of adverse reactions reported during post-marketing use cannot be determined as they are derived from spontaneous reports. Consequently, the frequency of these adverse events is qualified as "not known".

Description of selected adverse reactions
Local injection site reactions
Local injection site reactions, including erythema/redness, itching, swelling, and pain/tenderness, were reported in 6.1% of patients treated with alirocumab versus 4.1% in the control group (receiving placebo
injections). Most injection site reactions were transient and of mild intensity. The discontinuation rate due to
local injection site reactions was comparable between the two groups (0.2% in the alirocumab group versus
0.3% in the control group). In the cardiovascular outcomes study (ODYSSEY OUTCOMES), injection site reactions also occurred more frequently in alirocumab-treated patients than in placebo-treated patients (3.8%
alirocumab versus 2.1% placebo).
General allergic reactions
General allergic reactions were reported more frequently in the alirocumab group (8.1% of patients) than in the control group (7.0% of patients), mainly due to a difference in the incidence of pruritus. The observed
cases of pruritus were typically mild and transient. In addition, rare and sometimes serious allergic reactions such as hypersensitivity, nummular eczema, urticaria, and hypersensitivity vasculitis have been reported in
controlled clinical studies (See section 4.4). In the cardiovascular outcomes study (ODYSSEY OUTCOMES), general allergic reactions were similar in alirocumab-treated patients and placebo-treated
patients (7.9% alirocumab, 7.8% placebo). No difference was seen in the incidence of pruritus.
Special populations
Elderly
Although no safety issues were observed in patients over 75 years of age, data are limited in this age group. In the phase 3 primary hypercholesterolemia and mixed dyslipidaemia controlled studies, 1158 patients (34.7%) treated with Praluent were ≥65 years of age and 241 patients (7.2%) treated with Praluent were ≥75 years of age. In the cardiovascular outcomes controlled study, 2505 patients (26.5%) treated with Praluent

were ≥65 years of age and 493 patients (5.2%) treated with Praluent were ≥75 years of age. There were no significant differences observed in safety and efficacy with increasing age.
Every 4 week dosing study
The safety profile in patients treated with a 300 mg once every 4 week (monthly) dosing regimen, was similar to the safety profile as described for the clinical studies program using a 2 week dosing regimen, except for a higher rate of local injection site reactions. Local injection site reactions were reported overall at a frequency of 16.6% in the 300 mg once every 4 weeks treatment group and 7.9% in the placebo group. Patients in the alirocumab 300 mg every 4 weeks treatment group received alternating placebo injections to maintain blinding in regard to injection frequency. Excluding injection site reactions (ISRs) that occurred after these placebo injections, the frequency of ISRs was 11.8%. The discontinuation rate due to injection
site reactions was 0.7% in the 300 mg once every 4 weeks treatment group and 0% in the placebo group.
LDL-C values <25 mg/dL (<0.65 mmol/L)
In all clinical studies background lipid lowering therapies could not be adjusted by trial design. The percentage of patients who reached LDL-C values <25 mg/dL (<0.65 mmol/L) depended both on the baseline LDL-C and the dose of alirocumab.
In a pool of controlled studies using a 75 mg every 2 week (Q2W) starting dose and in which the dose was increased to 150 mg Q2W if the patient’s LDL-C was not <70 mg/dL or < 100 mg/dL (1.81 mmol/L or 2.59 mmol/L), 29.3% of patients with baseline LDL-C <100 mg/dL and 5.0% of patients with baseline LDL-C
≥100 mg/dL treated with alirocumab had two consecutive values of LDL-C <25 mg/dL (<0.65 mmol/L). In the ODYSSEY OUTCOMES study, in which the starting alirocumab dose was 75 mg Q2W and the dose
was increased to 150 mg Q2W if the patient’s LDL-C was not <50 mg/dL (1.29 mmol/L), 54.8% of patients with baseline LDL-C <100 mg/dL and 24.2% of patients with baseline LDL-C ≥100 mg/dL treated with
alirocumab had two consecutive values of LDL-C <25 mg/dL (<0.65 mmol/L).
Although adverse consequences of very low LDL-C were not identified in alirocumab trials, the long-term effects of very low levels of LDL-C are unknown. In published genetic studies as well as clinical and observational trials with lipid lowering therapies an increased risk of new onset of diabetes has been associated with lower levels of LDL-C.
Immunogenicity/ Anti-drug-antibodies (ADA)
In the ODYSSEY OUTCOMES trial, 5.5% of patients treated with alirocumab 75 mg and/or 150 mg every
2 weeks (Q2W) had anti-drug antibodies (ADA) detected after initiating treatment compared with 1.6% of patients treated with placebo, most of these were transient responses. Persistent ADA responses were observed in 0.7% of patients treated with alirocumab and 0.4% of patients treated with placebo. Neutralising antibody (NAb) responses were observed in 0.5% of patients treated with alirocumab and in <0.1% of patients treated with placebo.
Anti-drug antibody responses, including NAb, were low titer and did not appear to have a clinically meaningful impact on the efficacy or safety of alirocumab, except for a higher rate of injection site reactions in patients with treatment emergent ADA compared to patients who were ADA negative (7.5% vs 3.6%). The long-term consequences of continuing alirocumab treatment in the presence of ADA are unknown.
In a pool of ten placebo-controlled and active-controlled trials of patients treated with alirocumab 75 mg and/or 150 mg Q2W as well as in a separate clinical study of patients treated with alirocumab 75 mg Q2W or
300 mg every 4 weeks (including some patients with dose adjustment to 150 mg Q2W), the incidence of
detecting ADA and NAb was similar to the results from the ODYSSEY OUTCOMES trial described above.
Immunogenicity data are highly dependent on the sensitivity and specificity of the ADA assay.
Reporting of suspected adverse reactions:

The National Pharmacovigilance and Drug Safety Centre (NPC)
Fax: +966‐11‐205‐7662
Call NPC at +966‐11‐2038222, Exts: 2317‐2356‐2353‐2354‐2334‐2340.
Toll free phone: 8002490000
E‐mail: npc.drug@sfda.gov.sa
Website: www.sfda.gov.sa/npc


In controlled clinical studies, no safety issues were identified with more frequent dosing than the recommended every 2 week dosing schedule. There is no specific treatment for Praluent overdose. In the event of an overdose, the patient should be treated symptomatically, and supportive measures instituted as required.


Pharmacotherapeutic group: Other lipid modifying agents, ATC code: C10AX14. Mechanism of action
Alirocumab is a fully human IgG1 monoclonal antibody that binds with high affinity and specificity to proprotein convertase subtilisin kexin type 9 (PCSK9). PCSK9 binds to the low-density lipoprotein receptors (LDLR) on the surface of hepatocytes to promote LDLR degradation within the liver. LDLR is the primary receptor that clears circulating LDL, therefore the decrease in LDLR levels by PCSK9 results in higher blood levels of LDL-C. By inhibiting the binding of PCSK9 to LDLR, alirocumab increases the number of LDLRs available to clear LDL, thereby lowering LDL-C levels.
The LDLR also binds triglyceride-rich VLDL remnant lipoproteins and intermediate-density lipoprotein (IDL). Therefore, alirocumab treatment can produce reductions in these remnant lipoproteins as evidenced by its reductions in apolipoprotein B (Apo B), non-high-density lipoprotein cholesterol (non-HDL-C) and triglycerides (TG). Alirocumab also results in reductions in lipoprotein (a) [Lp(a)], which is a form of LDL that is bound to apolipoprotein (a). However, the LDLR has been shown to have a low affinity for Lp(a), therefore the exact mechanism by which alirocumab lowers Lp(a) is not fully understood.
In genetic studies in humans, PCSK9 variants with either loss-of-function or gain-of-function mutations have been identified. Individuals with single allele PCSK9 loss-of-function mutation have lower levels of LDL-C, which correlated with a significantly lower incidence of coronary heart disease. A few individuals have been reported, who carry PCSK9 loss-of-function mutations in two alleles and have profoundly low LDL-C
levels, with HDL-C and TG levels in the normal range. Conversely, gain-of-function mutations in the
PCSK9 gene have been identified in patients with increased LDL-C levels and a clinical diagnosis of familial hypercholesterolaemia.
In a multicenter, double-blind, placebo-controlled, 14 week study, 13 patients with heterozygous familial hypercholesterolaemia (heFH) due to gain-of-function mutations in the PCSK9 gene were randomised to receive either alirocumab 150 mg Q2W or placebo. Mean baseline LDL-C was 151.5 mg/dL (3.90 mmol/L). At week 2, the mean reduction from baseline in LDL-C was 62.5% in the alirocumab-treated patients as compared to 8.8% in the placebo patients. At week 8, the mean reduction in LDL-C from baseline with all patients treated with alirocumab was 72.4%.
Pharmacodynamic effects
In in vitro assays, alirocumab did not induce Fc-mediated effector function activity (antibody-dependent
cell-mediated toxicity and complement-dependent cytotoxicity) either in the presence or absence of PCSK9 and no soluble immune complexes capable of binding complement proteins were observed for alirocumab
when bound to PCSK9.

Clinical efficacy and safety in primary hypercholesterolaemia and mixed dyslipidaemia
Summary of the Phase 3 Clinical Trials Program - 75 mg and/or 150 mg every 2 weeks (Q2W) dosing regimen
The efficacy of alirocumab was investigated in ten phase 3 trials (five placebo-controlled and five
ezetimibe-controlled studies), involving 5296 randomised patients with hypercholesterolaemia (heterozygous familial and non-familial) or mixed dyslipidaemia, with 3188 patients randomised to alirocumab. In the
phase 3 studies, 31% of patients had type 2 diabetes mellitus, and 64% of patients had a history of coronary heart disease. Three of the ten studies were conducted exclusively in patients with heterozygous familial hypercholesterolaemia (heFH). The majority of patients in the phase 3 program were taking background
lipid-modifying therapy consisting of a maximally tolerated dose of statin, with or without other lipid- modifying therapies, and were at high or very high cardiovascular (CV) risk. Two studies were conducted in
patients who were not concomitantly treated with a statin, including one study in patients with documented statin intolerance.
Two studies (LONG TERM and HIGH FH), involving a total of 2416 patients, were performed with a
150 mg every 2 weeks (Q2W) dose only. Eight studies were performed with a dose of 75 mg Q2W, and criteria-based up-titration to 150 mg Q2W at week 12 in patients who did not achieve their pre-defined target
LDL-C based on their level of CV risk at week 8.
The primary efficacy endpoint in all of the phase 3 studies was the mean percent reduction from baseline in LDL-C at week 24 as compared to placebo or ezetimibe. All of the studies met their primary endpoint. In general, administration of alirocumab also resulted in a statistically significant greater percent reduction in total cholesterol (Total-C), non-high-density lipoprotein cholesterol (non-HDL-C), apolipoprotein B (Apo B), and lipoprotein (a) [Lp(a)] as compared to placebo/ ezetimibe, whether or not patients were concomitantly being treated with a statin. Alirocumab also reduced triglycerides (TG), and increased
high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A-1 (Apo A-1) as compared to placebo.
For detailed results see Table 2 below. Reduction in LDL-C was seen across age, gender, body mass index (BMI), race, baseline LDL-C levels, patients with heFH and non-heFH, patients with mixed dyslipidaemia, and diabetic patients. Although similar efficacy was observed in patients over 75 years, data are limited in this age group. LDL-C reduction was consistent regardless of concomitantly used statins and doses. A significantly higher proportion of patients achieved an LDL-C of ˂70 mg/dL (˂1.81 mmol/L) in the alirocumab group as compared to placebo or ezetimibe at week 12 and week 24. In studies using the criteria- based up-titration regimen, a majority of patients achieved the pre-defined target LDL-C (based on their
level of CV risk) on the 75 mg Q2W dose, and a majority of patients maintained treatment on the 75 mg Q2W dose. The lipid-lowering effect of alirocumab was observed within 15 days after the first dose reaching maximum effect at approximately 4 weeks. With long-term treatment, efficacy was sustained over the duration of the studies (up to 2 years). Following discontinuation of alirocumab, no rebound in LDL-C was observed, and LDL-C levels gradually returned to baseline levels.
In pre-specified analyses before possible up-titration at week 12 in the 8 studies in which patients started with the 75 mg every 2 weeks dosing regimen, mean reductions in LDL-C ranging from 44.5% to 49.2% were achieved. In the 2 studies in which patients were started and maintained on 150 mg every 2 weeks, the achieved mean reduction of LDL-C at week 12 was 62.6%. In analyses of pooled phase 3 studies that allowed up-titration, among the subgroup of patients up-titrated, an increase from 75 mg Q2W to 150 mg Q2W alirocumab at week 12 resulted in an additional 14% mean reduction in LDL-C in patients on a background statin. In patients not on a background statin, up-titration of alirocumab resulted in an additional
3% mean reduction in LDL-C, with the majority of the effect seen in approximately 25% of patients who achieved at least an additional 10% LDL-C lowering after up-titration. Patients up-titrated to 150 mg Q2W
had a higher mean baseline LDL-C.
Evaluation of Cardiovascular (CV) events
In pre-specified analyses of pooled phase 3 studies, treatment-emergent CV events confirmed by adjudication, consisting of coronary heart disease (CHD) death, myocardial infarction, ischemic stroke,

unstable angina requiring hospitalisation, congestive heart failure hospitalisation, and revascularisation, were reported in 110 (3.5%) patients in the alirocumab group and 53 (3.0%) patients in the control group (placebo or active control) with HR=1.08 (95% CI, 0.78 to 1.50). Major adverse cardiovascular events (“MACE- plus”, i.e.: CHD death, myocardial infarction, ischemic stroke, and unstable angina requiring hospitalisation) confirmed by adjudication were reported in 52 of 3182 (1.6%) patients in the alirocumab group and 33 of
1792 (1.8%) patients in the control group (placebo or active control); HR=0.81 (95% CI, 0.52 to 1.25).
In pre-specified final analyses of the LONG TERM study, treatment-emergent CV events confirmed by adjudication occurred in 72 of 1550 (4.6%) patients in the alirocumab group and in 40 of 788 (5.1%) patients in the placebo group; MACE-plus confirmed by adjudication were reported in 27 of 1550 (1.7%) patients in the alirocumab group and 26 of 788 (3.3%) patients in the placebo group. Hazard ratios were calculated post- hoc; for all CV events, HR=0.91 (95% CI, 0.62 to 1.34); for MACE-plus, HR=0.52 (95% CI, 0.31 to 0.90).
All-cause mortality
All-cause mortality in phase 3 studies was 0.6% (20 of 3182 patients) in the alirocumab group and 0.9% (17 of 1792 patients) in the control group. The primary cause of death in the majority of these patients was CV
events.
Combination therapy with a statin
Placebo-controlled phase 3 studies (on background statin) in patients with primary hypercholesterolaemia or mixed dyslipidaemia
LONG TERM study
This multicenter, double-blind, placebo-controlled, 18-month study included 2310 patients with primary hypercholesterolaemia at high or very high CV risk and on a maximally tolerated dose of statin, with or without other lipid-modifying therapy. Patients received either alirocumab at a dose of 150 mg Q2W or placebo in addition to their existing lipid-modifying therapy. The LONG TERM study included 17.7% heFH patients, 34.6% with type 2 diabetes mellitus, and 68.6% with a history of coronary heart disease. At week
24, the mean treatment difference from placebo in LDL-C percent change from baseline was -61.9% (95% CI: -64.3%, -59.4%; p-value: ˂0.0001). For detailed results see Table 2. At week 12, 82.1% of patients in the alirocumab group reached an LDL-C ˂70 mg/dL (˂1.81 mmol/L) compared to 7.2% of patients in the placebo group. Difference versus placebo was statistically significant at week 24 for all lipids/ lipoproteins.
COMBO I study
A multicenter, double-blind, placebo-controlled, 52 week study included 311 patients categorised as very high CV risk and not at their pre-defined target LDL-C on a maximally tolerated dose of statin, with or
without other lipid-modifying therapy. Patients received either 75 mg alirocumab Q2W or placebo in
addition to their existing lipid-modifying therapy. Dose up-titration of alirocumab to 150 mg Q2W occurred at week 12 in patients with LDL-C ≥70 mg/dL (≥1.81 mmol/L). At week 24, the mean treatment difference
from placebo in LDL-C percent change from baseline was -45.9% (95% CI: -52.5%, -39.3%; p-value:
˂0.0001). For detailed results see Table 4. At week 12 (before up-titration), 76.0% of patients in the alirocumab group reached an LDL-C of ˂70 mg/dL (˂ 1.81 mmol/L) as compared to 11.3% in the placebo group. The dose was up-titrated to 150 mg Q2W in 32 (16.8%) patients treated beyond 12 weeks. Among the subgroup of patients up-titrated at week 12, an additional 22.8% mean reduction in LDL-C was achieved at week 24. The difference versus placebo was statistically significant at week 24 for all lipids/ lipoproteins except TG and Apo A-1.
Placebo-controlled phase 3 studies (on background statin) in patients with heterozygous familial hypercholesterolaemia (heFH)
FH I and FH II studies
Two multicenter, placebo-controlled, double-blind 18-month studies included 732 patients with heFH receiving a maximally tolerated dose of statin, with or without other lipid-modifying therapy. Patients received either alirocumab 75 mg Q2W or placebo in addition to their existing lipid-modifying therapy. Dose up-titration of alirocumab to 150 mg Q2W occurred at week 12 in patients with LDL-C
≥70 mg/dL (≥1.81 mmol/L). At week 24, the mean treatment difference from placebo in LDL-C percent change from baseline was -55.8% (95% CI: -60.0%, -51.6%; p-value: ˂ 0.0001). For detailed results see
Table 2. At week 12 (before up-titration), 50.2% of patients reached an LDL-C of
˂70 mg/dL (˂1.81 mmol/L) as compared to 0.6% in the placebo group. Among the subgroup of patients
up-titrated at week 12, an additional 15.7% mean reduction in LDL-C was achieved at week 24. Difference versus placebo was statistically significant at week 24 for all lipids/ lipoproteins.
HIGH FH study
A third multicenter, double-blind, placebo-controlled 18-month study included 106 heFH patients on a maximally tolerated dose of statin, with or without other lipid-modifying therapies, and a baseline LDL-C
≥160 mg/dL (≥4.14 mmol/L). Patients received either alirocumab at a dose of 150 mg Q2W or placebo in
addition to their existing lipid-modifying therapy. At week 24, the mean treatment difference from placebo in LDL-C percent change from baseline was -39.1% (95% CI: -51.1%, -27.1%; p-value: ˂0.0001). For detailed results see Table 2. Mean changes for all other lipids/ lipoproteins were similar to the FH I and FH II studies, however statistical significance was not reached for TG, HDL-C and Apo A-1.
Ezetimibe-controlled phase 3 study (on background statin) in patients with primary hypercholesterolaemia or mixed dyslipidaemia
COMBO II study
A multicenter, double-blind, ezetimibe-controlled 2 year study included 707 patients categorised as very high
CV risk and not at their pre-defined target LDL-C on a maximally tolerated dose of statin. Patients received either alirocumab 75 mg Q2W or ezetimibe 10 mg once daily in addition to their existing statin therapy.
Dose up-titration of alirocumab to 150 mg Q2W occurred at week 12 in patients with LDL-C
≥70 mg/dL (≥1.81 mmol/L). At week 24, the mean treatment difference from ezetimibe in LDL-C percent change from baseline was -29.8% (95% CI: -34.4%, -25.3%; p-value: ˂0.0001). For detailed results see Table 2. At week 12 (before up-titration), 77.2% of patients reached an LDL-C of
˂70 mg/dL (˂1.81 mmol/L) as compared to 46.2% in the ezetimibe group. Among the subgroup of patients up-titrated at week 12, an additional 10.5% mean reduction in LDL-C was achieved at week 24. Difference
versus ezetimibe was statistically significant at week 24 for all lipids/ lipoproteins except for TG, and Apo
A-1.
Monotherapy or as add-on to non-statin lipid-modifying therapy
Ezetimibe-controlled phase 3 trials in patients with primary hypercholesterolaemia (without a background statin)
ALTERNATIVE study
A multicenter, double-blind, ezetimibe-controlled, 24 week study included 248 patients with documented statin intolerance due to skeletal muscle-related symptoms. Patients received either alirocumab 75 mg Q2W or ezetimibe 10 mg once daily, or atorvastatin 20 mg once daily (as a re-challenge arm). Dose up-titration of alirocumab to 150 mg Q2W occurred at week 12 in patients with LDL-C ≥70 mg/dL (≥1.81 mmol/L) or
≥100 mg/dL (≥2.59 mmol/L), depending on their level of CV risk. At week 24, the mean treatment
difference from ezetimibe in LDL-C percent change from baseline was -30.4% (95% CI: -36.6%, -24.2%; p- value: ˂0.0001). For detailed results see Table 2. At week 12 (before up-titration), 34.9% of patients reached an LDL-C of ˂70 mg/dL (˂1.81 mmol/L) as compared to 0% in the ezetimibe group. Among the subgroup of patients up-titrated at week 12, an additional 3.6% mean reduction in LDL-C was achieved at week 24. Difference versus ezetimibe was statistically significant at week 24 for LDL-C, Total-C, Non-HDL-C, Apo
B, and Lp(a).
This trial evaluated patients who did not tolerate at least two statins (at least one at the lowest approved dose), In these patients, musculo-skeletal adverse events occurred at a lower rate in the alirocumab group (32.5%)
as compared to the atorvastatin group (46.0%) (HR= 0.61 [95% CI, 0.38 to 0.99]), and a lower percentage of patients in the alirocumab group (15.9%) discontinued study treatment due to musculo-skeletal adverse events as compared to the atorvastatin group (22.2%). In the five placebo-controlled trials in patients on a maximally tolerated dose of statin (n=3752), the discontinuation rate due to musculo-skeletal adverse events was 0.4% in the alirocumab group and 0.5% in the placebo group. MONO study
A multicenter, double-blind, ezetimibe-controlled, 24 week study included 103 patients with a moderate CV
risk, not taking statins or other lipid-modifying therapies, and a baseline LDL-C between
100 mg/dL (2.59 mmol/L) to 190 mg/dL (4.91 mmol/L). Patients received either alirocumab 75 mg Q2W or ezetimibe 10 mg once daily. Dose up-titration of alirocumab to 150 mg Q2W occurred at week 12 in patients with LDL-C ≥70 mg/dL (≥1.81 mmol/L). At week 24, the mean treatment difference from ezetimibe in
LDL-C percent change from baseline was -31.6% (95% CI: -40.2%, -23.0%; p-value: ˂0.0001). For detailed results see Table 2. At week 12 (before up-titration), 57.7% of patients reached an LDL-C of
˂70 mg/dL (˂1.81 mmol/L) as compared to 0% in the ezetimibe group. The dose was up-titrated to 150 mg
Q2W in 14 (30.4%) patients treated beyond 12 weeks. Among the subgroup of patients up-titrated at week
12, an additional 1.4 % mean reduction in LDL-C was achieved at week 24. The difference versus ezetimibe was statistically significant at week 24 for LDL-C, Total-C, Non-HDL-C and Apo B.

Every 4 Week (Q4W) Dosing Regimen
CHOICE I study
A multicenter, double-blind, placebo-controlled, 48 week study included 540 patients on a maximally tolerated dose of a statin, with or without other lipid-modifying therapy (308 in the alirocumab 300 mg Q4W group, 76 in the alirocumab 75 mg Q2W group, and 156 in the placebo group), and 252 patients not treated with a statin (144 in the alirocumab 300 mg Q4W group, 37 in the alirocumab 75 mg Q2W group, and 71 in the placebo group). Patients received either alirocumab 300 mg Q4W, alirocumab 75 mg Q2W, or placebo in addition to their existing lipid-modifying therapy (statin, non-statin therapy or diet alone). Patients in the alirocumab 300 mg every 4 weeks treatment group received alternating placebo injections to maintain blinding in regard to injection frequency. Overall, 71.6% of patients were categorized at high or very high
CV risk and not at their LDL-C target. Dose adjustment in the alirocumab groups to 150 mg Q2W occurred
at week 12 in patients with LDL-C ≥70 mg/dL or ≥100 mg/dL, depending on their level of CV risk, or in patients who did not have at least a 30% reduction of LDL-C from baseline.
In the cohort of patients on background statin, the mean baseline LDL-C was 112.7 mg/dL. At week 12, the mean percent change from baseline with alirocumab 300 mg Q4W in LDL-C (ITT analysis) was -55.3% compared to +1.1% for placebo. At week 12 (before dose adjustment), 77.3% of patients treated with alirocumab 300 mg Q4W reached an LDL-C of ˂70 mg/dL as compared to 9.3% in the placebo group. At week 24, the mean percent change from baseline with alirocumab 300 mg Q4W/150 mg Q2W in LDL-C (ITT analysis) was -58.8% compared to -0.1% for placebo. At week 24, the mean treatment difference for alirocumab 300 mg Q4W/150 mg Q2W from placebo in LDL-C percent change from baseline was -58.7% (97.5% CI: -65.0%, -52.4%; p-value: ˂ 0.0001). In patients treated beyond 12 weeks, the dose was adjusted to 150 mg Q2W in 56 (19.3%) of 290 patients in the alirocumab 300 mg Q4W arm. Among the subgroup of patients dose adjusted to 150 mg Q2W at week 12, an additional 25.4% reduction in LDL-C was achieved at week 24.
In the cohort of patients not treated with a concomitant statin, the mean baseline LDL-C was 142.1 mg/dL. At week 12, the mean percent change from baseline with alirocumab 300 mg Q4W in LDL-C (ITT analysis) was -58.4% compared to +0.3% for placebo. At week 12 (before dose adjustment), 65.2% of patients treated with alirocumab 300 mg Q4W reached an LDL-C of ˂70 mg/dL as compared to 2.8% in the placebo group. At week 24, the mean percent change from baseline with alirocumab 300 mg Q4W/150 mg Q2W in LDL-C (ITT analysis) was -52.7% compared to -0.3% for placebo. At week 24, the mean treatment difference for alirocumab 300 mg Q4W/150 mg Q2W from placebo in LDL-C percent change from baseline was -52.4% (97.5% CI: -59.8%, -45.0%; p-value: ˂ 0.0001). In patients treated beyond 12 weeks, the dose was adjusted to 150 mg Q2W in 19 (14.7%) of 129 patients in the alirocumab 300 mg Q4W arm. Among the subgroup of patients dose adjusted to 150 mg Q2W at week 12, an additional 7.3% mean reduction in LDL-C was achieved at week 24.
In both cohorts, the difference vs placebo was statistically significant at week 24 for all lipid parameters, except for Apo A-1 in the subgroup of patients on background statin. Clinical efficacy and safety in prevention of cardiovascular events
ODYSSEY OUTCOMES study
A multicentre, double-blind, placebo-controlled trial included 18,924 adult patients (9462 alirocumab; 9462 placebo) followed for up to 5 years. Patients had experienced an acute coronary syndrome (ACS) event 4 to
52 weeks prior to randomization and were treated with a lipid-modifying-therapy (LMT) regimen that was statin-intensive (defined as atorvastatin 40 or 80 mg, or rosuvastatin 20 or 40 mg) or at maximally tolerated dose of those statins, with or without other LMT. Patients were randomized 1:1 to receive either alirocumab
75 mg once every two weeks (Q2W) or placebo Q2W. At month 2, if additional LDL-C lowering was
required based on pre-specified LDL-C criteria (LDL-C ≥50 mg/dL or 1.29 mmol/dL), alirocumab was adjusted to 150 mg Q2W. For patients who had their dose adjusted to 150 mg Q2W and who had two consecutive LDL-C values below 25 mg/dL (0.65 mmol/L), down-titration from 150 mg Q2W to 75 mg Q2W was performed. Patients on 75 mg Q2W who had two consecutive LDL-C values below 15 mg/dL (0.39 mmol/L) were switched to placebo in a blinded fashion. Approximately 2615 (27.7%) of 9451 patients
treated with alirocumab required dose adjustment to 150 mg Q2W. Of these 2615 patients, 805 (30.8%) were
down-titrated to 75 mg Q2W. Overall, 730 (7.7%) of 9451 patients switched to placebo. A total of 99.5% of patients were followed for survival until the end of the trial. The median follow-up duration was 33 months.
The index ACS event was a myocardial infarction in 83.2% of patients (34.6% STEMI, 48.6% NSTEMI) and an episode of unstable angina in 16.8% of patients. Most patients (88.8%) were receiving high intensity statin therapy with or without other LMT at randomization. The mean LDL-C value at baseline was 92.4 mg/dL (2.39 mmol/L).

Alirocumab significantly reduced the risk for the primary composite endpoint of the time to first occurrence of Major Adverse Cardiovascular Events (MACE-plus) consisting of coronary heart disease (CHD) death, non-fatal myocardial infarction (MI), fatal and non-fatal ischemic stroke, or unstable angina (UA) requiring hospitalization (HR 0.85, 95% CI: 0.78, 0.93; p-value=0.0003). Alirocumab also significantly reduced the following composite endpoints: risk of CHD event; major CHD event; cardiovascular event; and the composite of all-cause mortality, non-fatal MI, and non-fatal ischemic stroke. A reduction of all-cause mortality was also observed, with only nominal statistical significance by hierarchical testing (HR 0.85, 95% CI: 0.73, 0.98). The results are presented in Table 3.

Paediatric population
The European Medicines Agency has deferred the obligation to submit the results of studies with Praluent in one or more subsets of the paediatric population in the treatment of elevated cholesterol (see section 4.2 for information on paediatric use).
The European Medicines Agency has waived the obligation to submit the results of studies with Praluent in all subsets of the paediatric population in the treatment of mixed dyslipidaemia (see section 4.2 for information on paediatric use).


Absorption
After subcutaneous administration of 50 mg to 300 mg alirocumab, median times to maximum serum concentration (tmax) were 3-7 days. The pharmacokinetics of alirocumab after single subcutaneous administration of 75 mg into the abdomen, upper arm or thigh were similar. The absolute bioavailability of alirocumab after subcutaneous administration was about 85% as determined by population pharmacokinetic analysis. Monthly exposure with 300 mg every 4 weeks treatment was similar to that of 150 mg every
2 weeks. The fluctuations between Cmax and Ctrough were higher for the every 4 weeks dosage regimen. Steady state was reached after 2 to 3 doses with an accumulation ratio up to a maximum of about 2-fold.
Distribution
Following intravenous administration, the volume of distribution was about 0.04 to 0.05 L/kg indicating that alirocumab is distributed primarily in the circulatory system.
Biotransformation
Specific metabolism studies were not conducted, because alirocumab is a protein. Alirocumab is expected to degrade to small peptides and individual amino acids.
Elimination
Two elimination phases were observed for alirocumab. At low concentrations, the elimination is predominately through saturable binding to target (PCSK9), while at higher concentrations the elimination of alirocumab is largely through a non-saturable proteolytic pathway. Based on a population pharmacokinetic analysis, the median apparent half-life of alirocumab at steady state was 17 to 20 days in patients receiving alirocumab as monotherapy at subcutaneous doses of either 75 mg Q2W or 150 mg Q2W. When co-administered with a statin, the median apparent half-life of alirocumab was
12 days.
Linearity/non-linearity
A slightly greater than dose proportional increase was observed, with a 2.1- to 2.7-fold increase in total alirocumab concentrations for a 2-fold increase in dose from 75 mg to 150 mg Q2W.
Special populations
Elderly
Based on a population pharmacokinetic analysis, age was associated with a small difference in alirocumab
exposure at steady state, with no impact on efficacy or safety.
Gender
Based on a population pharmacokinetic analysis, gender has no impact on alirocumab pharmacokinetics.
Race
Based on a population pharmacokinetic analysis, race had no impact on alirocumab pharmacokinetics. Following single-dose subcutaneous administration of 100 mg to 300 mg alirocumab, there was no meaningful difference in exposure between Japanese and Caucasian healthy subjects.
Body weight
Body weight was identified as one significant covariate in the final population PK model impacting
alirocumab pharmacokinetics. Alirocumab exposure (AUC0-14d) at steady state at both the 75 and 150 mg Q2W dosing regimen was decreased by 29% and 36% in patients weighing more than 100 kg as compared to patients weighing between 50 kg and 100 kg. This did not translate into a clinically meaningful difference in LDL-C lowering.
Hepatic impairment
In a phase 1 study, after administration of a single 75 mg subcutaneous dose, alirocumab pharmacokinetic profiles in subjects with mild and moderate hepatic impairment were similar as compared to subjects with normal hepatic function. No data are available in patients with severe hepatic impairment.
Renal impairment
Since monoclonal antibodies are not known to be eliminated via renal pathways, renal function is not expected to impact the pharmacokinetics of alirocumab. Population pharmacokinetic analyses showed that alirocumab exposure (AUC0-14d) at steady state at both the 75 and 150 mg Q2W dosing regimen was increased by 22%-35%, and 49%-50% in patients with mild and moderate renal impairment, respectively, compared to patients with normal renal function. The distribution of body weight and age, two covariates impacting alirocumab exposure, were different among renal function categories and most likely explain the observed pharmacokinetic differences. Limited data are available in patients with severe renal impairment; in these patients the exposure to alirocumab was approximately 2-fold higher compared with subjects with normal renal function.
Pharmacokinetic/pharmacodynamic relationship(s)
The pharmacodynamic effect of alirocumab in lowering LDL-C is indirect, and mediated through the binding to PCSK9. A concentration-dependent reduction in free PCSK9 and LDL-C is observed until target
saturation is achieved. Upon saturation of PCSK9 binding, further increases in alirocumab concentrations do not result in a further LDL-C reduction, however an extended duration of the LDL-C lowering effect is observed.


Non-clinical data reveal no special hazard for humans based on evaluations of safety pharmacology, and repeated dose toxicity.
Reproductive toxicology studies in rats and monkeys indicated that alirocumab, like other IgG antibodies, crosses the placental barrier.
There were no adverse effects on surrogate markers of fertility (e.g. estrous cyclicity, testicular volume, ejaculate volume, sperm motility, or total sperm count per ejaculate) in monkeys, and no alirocumab-related anatomic pathology or histopathology findings in reproductive tissues in any rat or monkey toxicology study.
There were no adverse effects on fetal growth or development in rats or monkeys. Maternal toxicity was not evident in pregnant monkeys at systemic exposures that were 81 times the human exposure at the 150 mg Q2W dose. However, maternal toxicity was noted in pregnant rats at systemic exposures estimated to be approximately 5.3 times greater than the human exposure at the 150 mg Q2W dose (based on exposure measured in non-pregnant rats during a 5-week toxicology study).
The offspring of monkeys that received high doses of alirocumab weekly throughout pregnancy had a
weaker secondary immune response to antigen challenge than did the offspring of control animals. There was no other evidence of alirocumab-related immune dysfunction in the offspring.


Histidine

Sucrose

Polysorbate 20
Water for injections


In the absence of compatibility studies, this medicinal product must not be mixed with other medicinal products.


2 years

Store in a refrigerator (2°C to 8°C). Do not freeze.
Praluent can be stored outside the refrigerator (below 25 °C) protected from light for a single period not exceeding 30 days. After removal from the refrigerator, the medicinal product must be used within 30 days or discarded.
Keep the pen or syringe in the outer carton in order to protect from light


1 ml solution in a siliconised Type 1 clear glass syringe, equipped with a stainless steel staked needle, a styrene-butadiene rubber soft needle shield, and an ethylene tetrafluoroethylene -coated bromobutyl rubber plunger stopper.
Pre-filled pen 75 mg:

The syringe components are assembled into a single-use pre-filled pen with a blue cap and a light green activation button.
Pre-filled pen 150 mg:
The syringe components are assembled into a single-use pre-filled pen with a blue cap and a dark grey activation button.
Pre-filled syringe 75 mg:
The syringe is equipped with a light green polypropylene plunger rod. Pre-filled syringe 150 mg:
The syringe is equipped with a dark grey polypropylene plunger rod. Pack size:
1, 2, or 6 pre-filled pens.
1, 2, or 6 pre-filled syringes.
Not all presentations and pack sizes may be marketed.


The solution should be clear, colourless to pale yellow. If the solution is discoloured or contains visible particulate matter, the solution should not be used.
After use, place the pre-filled pen/ pre-filled syringe into a puncture resistant container and discard as required by local regulations. Do not recycle the container. Always keep the container out of the sight and reach of children. Any unused medicinal product or waste material should be disposed of in accordance with local requirements.


sanofi-aventis groupe 54, rue La Boétie F – 75008 Paris France

December 2017
}

صورة المنتج على الرف

الصورة الاساسية