Search Results
نشرة الممارس الصحي | نشرة معلومات المريض بالعربية | نشرة معلومات المريض بالانجليزية | صور الدواء | بيانات الدواء |
---|
The active substance of WAINUA is eplontersen, which is a type of medicine called an antisense oligonucleotide inhibitor.
What WAINUA is used for
WAINUA is a treatment for adults with hereditary transthyretin amyloidosis (ATTRv), which runs in families. The illness is caused by problems with a protein called transthyretin (TTR). It is made mostly in the liver and carries vitamin A and other substances around the body.
In people with this illness, small fibres of TTR protein clump together to make deposits called ‘amyloid’. Amyloid can build up around or within the nerves, heart and other places in the body, stopping them from working normally. This is what causes the symptoms of the illness.
How WAINUA works
WAINUA works by lowering the amount of TTR protein made by the liver. As a result, there is less TTR protein in the blood to form amyloid deposits. That can help to reduce the effects of the illness.
Warnings and precautions
Lowered vitamin A levels in the blood and vitamin A supplements
Treatment with WAINUA lowers the amount of vitamin A in your blood. Your doctor will measure your vitamin A levels and will ask you to take a daily vitamin A supplement during treatment.
Signs of low vitamin A can include: poor vision especially at night, dry eyes, hazy or cloudy vision.
· Talk to your doctor if you notice a problem with your vision or any other eye problems while using WAINUA. If necessary, your doctor will refer you to an eye specialist for a check-up.
Both too high and too low levels of vitamin A can harm the development of your unborn child. Women of child-bearing age should not be pregnant when starting treatment with WAINUA and should use effective contraception during treatment (see section “Pregnancy, breast-feeding and contraception” below).
· Tell your doctor if you plan to become pregnant. Your doctor might tell you to stop taking WAINUA and vitamin A supplements.
· Tell your doctor if you are pregnant. Your doctor may tell you to stop taking WAINUA and might recommend vitamin A supplements depending on how far in your pregnancy you are.
Children and adolescents
WAINUA is not recommended in children and adolescents under 18 years of age.
Other medicines and WAINUA
Tell your doctor or pharmacist if you are using, have recently used or might use any other medicines.
Pregnancy, breast-feeding and contraception
Before you start using WAINUA, tell your doctor if you are pregnant, think you might be pregnant, or are trying to become pregnant, or you are breast-feeding.
Pregnancy
Do not use WAINUA if you are pregnant unless explicitly advised by your doctor.
Breast-feeding
It is not known whether the ingredients of WAINUA can pass into breast milk. Before you start treatment with WAINUA, tell your doctor if you are breast-feeding or planning to breast-feed. Your doctor may tell you to stop taking WAINUA.
Women of childbearing age
Make sure you’re not pregnant before starting treatment with WAINUA. It will reduce the level of vitamin A in your blood, and vitamin A is important for normal development of your unborn child (see “Warnings and precautions” above).
If you are a woman who is able to become pregnant and intend to use WAINUA, you must use effective contraception. Talk to your doctor or nurse about suitable methods of contraception.
· Tell your doctor if you are planning to become pregnant or if you are pregnant. Your doctor may advise you and might tell you to stop taking WAINUA.
Driving and using machines
It is unlikely that WAINUA will affect your ability to drive and use machines. Your doctor will tell you whether your condition allows you to drive vehicles and use machines safely.
WAINUA contains sodium
This medicine contains less than 1 mmol sodium (23 mg) per dose of 0.8 mL, that is to say essentially ‘sodium-free’.
Always use WAINUA exactly as your doctor, pharmacist, or nurse has told you. Check with your doctor, nurse, or pharmacist if you are not sure.
WAINUA is given as an injection under the skin (subcutaneously). The injection can be done in the stomach (abdomen), thigh or, if given by a carer, in the back of your upper arm.
You and your doctor or nurse should decide if WAINUA should be injected by yourself or by your carer. You or your carer will receive training on the right way to prepare and inject this medicine. Read the ‘Instructions for Use’ carefully before using it.
The recommended dose of WAINUA is one injection of 45 mg every month.
Your doctor will tell you how long you need to receive WAINUA. Do not stop treatment unless your doctor tells you to.
If you use more WAINUA than you should
If you inject too much, get medical advice immediately, or go to a hospital emergency department. Do this even if you have no symptoms.
If you forget to use WAINUA
If you miss your dose of WAINUA, have your next dose as soon as possible and continue your monthly injections from that date on.
If you have any further questions on the use of this medicine, ask your doctor, pharmacist, or nurse.
Like all medicines, WAINUA can have side effects, although not everybody gets them.
Very common (may affect more than 1 in 10 people)
· Effects seen in blood tests: low vitamin A.
Common (may affect up to 1 in 10 people)
· Vomiting.
· Redness, itching, pain where the injection was given.
· Protenuria
· Blurred vision
· Cataracts
Keep this medicine out of the sight and reach of children.
Do not use WAINUA after the expiry date which is stated on the pre-filled pen 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).
WAINUA may be stored unrefrigerated (below 30°C) in the original carton for up to 6 weeks. If not used within 6 weeks, it should be discarded.
Store in the original package to protect from light.
Do not freeze. Do not expose to heat.
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 WAINUA contains
The active substance is eplontersen. Each pre-filled pen contains eplontersen sodium equivalent to 45 mg eplontersen in 0.8 mL solution.
The other ingredients are sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium chloride and water for injection. Hydrochloric acid and sodium hydroxide may be used to adjust the pH (see “WAINUA contains sodium” in section 2).
Marketing Authorisation Holder
AstraZeneca AB
SE-151 85 Södertälje
Sweden
Manufacturer
Vetter Pharma-Fertigung GmbH & Co KG
Schuetzenstrasse 87a
Ravensburg, Baden-Wuerttemberg 88212
Germany
المادة الفعالة لدواء وينوا هي إيبلونتيرسن، وهو نوع من الأدوية يسمّى مثبط قليل النوكليوتيد المضاد.
دواعي استعمال دواء وينوا
دواء وينوا هو علاج للبالغين المصابين بالداء النشواني الوراثي الناتج عن بروتين الترانستيريتين (ATTRv)، والذي يمتد في العائلات. يحدث المرض بسبب مشكلات في بروتين يُسمى الترانستيريتين (TTR). ويُصنع معظمه في الكبد ويحمل فيتامين أ ومواد أخرى حول الجسم.
تتجمع ألياف صغيرة من بروتين TTR معًا لصنع رواسب تسمى "نشَوانيّ" لدى الأشخاص الذين يعانون من هذا المرض. يمكن أن يتراكم النشوانيّ حول الأعصاب والقلب وأماكن أخرى في الجسم أو داخلها، مما يمنعها من العمل بصورة طبيعية. وهذا هو ما يسبب أعراض المرض.
كيف يعمل دواء وينوا
يعمل دواء وينوا من خلال تقليل كمية بروتين TTR الذي يصنّعه الكبد. ونتيجة لذلك، هناك بروتين TTR أقل في الدم لتكوين رواسب نشوانيّة. يمكن أن يساعد ذلك في تقليل آثار المرض.
تحذيرات واحتياطات
انخفاض مستويات فيتامين أ في الدم ومكملات فيتامين أ
يقلّل العلاج باستخدام دواء وينوا من كمية فيتامين أ في دمك. سيقيس الطبيب المتابع لك مستويات فيتامين أ لديك وسيطلب منك أخذ مكمل فيتامين أ يوميًا أثناء العلاج.
يمكن أن تشمل علامات انخفاض فيتامين أ: ضعف الرؤية خاصةً في الليل، أو جفاف العينين، أو الرؤية الضبابية أو الغائمة.
· تحدث إلى الطبيب المتابع لك إذا لاحظت مشكلة في الرؤية أو أيّة مشاكل أخرى في العين أثناء استخدام دواء وينوا. إذا لزم الأمر، سيُحيلك الطبيب المتابع لك إلى اختصاصي عيون لإجراء فحص.
يمكن أن تُلحق المستويات المرتفعة جدًا والمنخفضة جدًا من فيتامين أ الضرر بنمو جنينك. ينبغي ألا تكون النساء في سن الإنجاب حوامل عند بدء العلاج بدواء وينوا ويجب أن يستخدمن وسائل منع حمل فعالة أثناء العلاج (انظر قسم "الحمل والرضاعة الطبيعية ووسائل منع الحمل" أدناه).
· أخبري الطبيب المتابع لكِ إذا كنتِ تخططين للحمل. قد يطلب منك الطبيب المتابع لكِ التوقف عن أخذ دواء وينوا ومكملات فيتامين أ.
· أخبري الطبيب المتابع لكِ إذا كنتِ حاملًا. قد يطلب منكِ الطبيب المتابع لكِ التوقف عن أخذ دواء وينوا وقد يوصي بمكملات فيتامين أ بناءً على مدى تقدّم عمر حملكِ.
الأطفال والمراهقون
لا يوصى باستعمال دواء وينوا للأطفال والمراهقين دون سن 18 عامًا.
الأدوية الأخرى ودواء وينوا
أخبر الطبيب المتابع لك أو الصيدلي إذا كنت تستخدم، أو استخدمت مؤخرًا، أو قد تستخدم أيّة أدوية أخرى.
الحمل والرضاعة الطبيعية ومنع الحمل
قبل أن تبدأي في استخدام دواء وينوا، أخبري الطبيب المتابع لكِ إذا كنتِ حاملًا، أو تعتقدين أنكِ قد تكونين حاملًا، أو تحاولين الحمل، أو تُرضعين رضاعة طبيعية.
الحمل
لا تستخدمي دواء وينوا إذا كنتِ حاملًا ما لم ينصحكِ الطبيب المتابع لكِ صراحةً بذلك.
الرضاعة الطبيعية
من غير المعروف ما إذا كانت مكونات دواء وينوا يمكن أن تنتقل إلى حليب الأم. قبل أن تبدأي العلاج باستخدام دواء وينوا، أخبري الطبيب المتابع لكِ إذا كنتِ تُرضعين طبيعيًا أو تخططين للرضاعة الطبيعية. قد يطلب منك الطبيب المتابع لكِ التوقف عن أخذ دواء وينوا.
النساء في سن الإنجاب
تأكّدي من أنكِ لستِ حاملًا قبل بدء العلاج باستخدام دواء وينوا. سيقلّل من مستوى فيتامين أ في دمك، ويُعدّ فيتامين أ مهمًا للنمو الطبيعي للجنين (انظري "التحذيرات والاحتياطات" أعلاه).
إذا كنت امرأة قادرة على الحمل وتنوين استخدام دواء وينوا فينبغي عليكِ استخدام وسيلة منع حمل فعالة. تحدثي إلى الطبيب المتابع لكِ أو ممرضتكِ حول وسائل منع الحمل المناسبة.
· أخبري الطبيب المتابع لكِ إذا كنتِ حاملًا، أو تعتقدين أنك حاملًا، أو تنوين الحمل. قد ينصحك الطبيب المتابع لكِ وقد يطلب منك التوقف عن أخذ دواء وينوا.
القيادة واستخدام الآلات
من غير المحتمل أن يؤثر دواء وينوا على قدرتك على القيادة واستخدام الآلات. سيُخبرك الطبيب المتابع لك بما إذا كانت حالتك تسمح لك بقيادة المركبات واستخدام الآلات بأمان.
يحتوي دواء وينوا على الصوديوم
يحتوي هذا الدواء على أقل من 1 مليمول صوديوم (23 مجم) لكل جرعة تبلغ 0.8 مل، أي أنه "خالٍ من الصوديوم" بصورة أساسية.
استخدم دائمًا دواء وينوا متّبعا بدقة تعليمات الطبيب المتابع لك أو الصيدلي أو الممرضة. استشر الطبيب المتابع لك أو الممرضة أو الصيدلي إذا لم تكن متأكدًا.
يُعطى دواء وينوا في صورة حقن تحت الجلد. يمكن إجراء الحقن في المعدة (البطن)، أو الفخذ، أو في الجزء الخلفي من أعلى ذراعك إذا أعطاه لك مقدم رعاية.
يجب أن تقرر أنت والطبيب المتابع لك أو الممرضة ما إذا كان يجب حقن دواء وينوا بنفسك أو بواسطة مقدم الرعاية المتابع لك. ستتلقى أنت أو مقدم الرعاية المتابع لك تدريبًا على الطريقة الصحيحة لتحضير هذا الدواء وحقنه. اقرأ "تعليمات الاستخدام" بعناية قبل استخدامه.
الجرعة الموصى بها من دواء وينوا هي حقنة واحدة 45 مجم كل شهر.
سيخبرك الطبيب المتابع لك بالمدة التي تحتاج فيها إلى تلقي دواء وينوا. لا تتوقّف عن العلاج إلا إذا أخبرك الطبيب المتابع لك بذلك.
إذا استعملت دواء وينوا بمقدار أكبر مما ينبغي
إذا حقنت مقدار أكثر من اللازم، فاطلب المشورة الطبية على الفور، أو اذهب إلى قسم الطوارئ بالمستشفى. افعل ذلك حتى لو لم تظهر عليك أيّة أعراض.
إذا نسيت استخدام دواء وينوا
إذا فاتتك جرعة دواء وينوا، فخذ جرعتك التالية في أقرب وقت ممكن وواصل حقنك الشهرية من ذلك التاريخ فصاعدًا.
إذا كانت لديك أيّة أسئلة أخرى بشأن استخدام هذا الدواء، فاسأل الطبيب المتابع لك أو الصيدلي أو الممرضة.
شأنه شأن سائر الأدوية، يمكن أن يُسبب دواء وينوا آثارًا جانبية، على الرغم من عدم تأثر جميع الأشخاص بها.
شائعة جدًا (قد تُصيب أكثر من شخص واحد من كل 10 أشخاص)
· الآثار التي شوهدت في اختبارات الدم: انخفاض فيتامين أ.
شائعة (قد تصيب شخصًا واحدًا على الأكثر من كل 10 أشخاص)
· التقيؤ.
· احمرار، حكة، ألم في موضع الحقن.
· حالة من فرط أو فائض بروتينات الدم في البول (بيلة بروتينية)
· عدم وضوح الرؤية
· ضبابية عدسة العين (الماء الأبيض)
يُحفظ هذا الدواء بعيدًا عن متناول أيدي الأطفال ونظرهم.
لا تستخدم دواء وينوا بعد تاريخ انتهاء الصلاحية المدوّن على ملصق القلم المعبأ مسبقًا والعلبة الكرتونية بعد كلمة ”EXP“. يشير تاريخ انتهاء الصلاحية إلى آخر يوم في ذلك الشهر.
يُحفظ في الثلاجة (في درجة حرارة تتراوح من 2 إلى 8 درجات مئوية).
يمكن تخزين دواء وينوا دون تبريد (أقل من 30 درجة مئوية) في العلبة الكرتونية الأصلية لمدة تصل إلى 6 أسابيع. إذا لم يُستخدم في غضون 6 أسابيع، فيجب التخلص منه.
يُخزّن في العبوة الأصلية لحمايته من الضوء.
لا تجمده. لا تعرّضه للحرارة.
لا تتخلص من أي أدوية عن طريق مياه الصرف الصحي أو النفايات المنزلية. اسأل الصيدلي الذي تتعامل معه عن كيفية التخلص من الأدوية التي لم تعد تستعملها. هذه التدابير من شأنها حماية البيئة.
محتويات دواء وينوا
المادة الفعالة هي إيبلونتيرسن. يحتوي كل قلم معبأ مسبقًا على صوديوم إيبلونتيرسن يعادل 45 مجم إيبلونتيرسن في محلول 0.8 مل.
المكونات الأخرى هي فوسفات ثنائي هيدروجين الصوديوم، وفوسفات هيدروجين ثنائي الصوديوم، وكلوريد الصوديوم، وماء للحقن. يمكن استخدام حمض الهيدروكلوريك وهيدروكسيد الصوديوم لضبط درجة الحموضة (انظر "يحتوي دواء وينوا على الصوديوم" في القسم 2).
شكل دواء وينوا ومحتويات العبوة
دواء وينوا عبارة عن محلول للحقن صافٍ وعديم اللون مائل إلى الأصفر.
يتوفر دواء وينوا في عبوة تحتوي على قلم واحد معبأ مسبقًا مخصّص للاستخدام مرة واحدة.
حامل ترخيص التسويق
AstraZeneca AB
SE-151 85 Södertälje
السويد
جهة التصنيع
Vetter Pharma-Fertigung GmbH & Co KG
Schuetzenstrasse 87a
Ravensburg, Baden-Wuerttemberg 88212
ألمانيا
WAINUA is indicated for the treatment of adult patients with polyneuropathy associated with hereditary transthyretin-mediated amyloidosis (ATTRv).
Treatment should be prescribed and supervised by a treating physician knowledgeable in the management of patients with amyloidosis.
Posology
The recommended dose of WAINUA is 45 mg administered by subcutaneous injection. Doses should be administered monthly.
Missed dose
If a dose of eplontersen is missed, then the next dose should be administered as soon as possible. Resume dosing at monthly intervals from the date of the last dose.
Special populations
Renal impairment
No dose adjustment is necessary in patients with mild to moderate renal impairment (estimated glomerular filtration rate [eGFR] ≥45 to <90 mL/min/1.73 m2) (see section 5.2). WAINUA has not been studied in patients with eGFR<45 mL/min/1.73 m2 or end-stage renal disease.
Hepatic impairment
No dose adjustment is necessary in patients with mild hepatic impairment (see section 5.2). WAINUA has not been studied in patients with moderate or severe hepatic impairment.
Elderly population
No dose adjustment is required in elderly patients (≥65 years of age) (see section 5.2).
Paediatric population
The safety and efficacy of WAINUA in children and adolescents below 18 years of age have not been established. No data are available.
Method of administration
Subcutaneous use only.
The first injection administered by the patient or caregiver should be performed under the guidance of an appropriately qualified health care professional. Patients and/or caregivers should be trained in the subcutaneous administration of WAINUA.
The autoinjector should be removed from refrigerated storage at least 30 minutes before use and allowed to reach room temperature prior to injection. Other warming methods should not be used.
Inspect solution visually before use. The solution should appear colourless to yellow. Do not use if cloudiness, particulate matter or discolouration is observed prior to administration.
If self-administered, inject WAINUA in the abdomen or upper thigh region. If a caregiver administers the injection, the back of the upper arm can also be used.
Comprehensive instructions for administration are provided in the ‘Instructions for Use’.
Reduced Serum Vitamin A Levels and Recommended Supplementation
Based on the mechanism of action, WAINUA is expected to reduce serum vitamin A (retinol) below normal levels (see section 5.1).
Any symptoms or signs related to vitamin A deficiency should be evaluated prior to initiation of treatment with WAINUA.
Patients receiving WAINUA should take oral supplementation of the daily recommended dose of vitamin A to reduce the potential risk of ocular symptoms due to vitamin A deficiency. Referral for ophthalmological assessment is recommended if patients develop ocular symptoms consistent with vitamin A deficiency, including reduced night vision or night blindness, and persistent dry eyes.
It is not known whether vitamin A supplementation in pregnancy will be sufficient to prevent vitamin A deficiency if the pregnant female continues to receive WAINUA (see section 4.6). However, increasing vitamin A supplementation to above the daily recommended dose during pregnancy is unlikely to correct serum retinol levels due to the mechanism of action of eplontersen and may be harmful to the mother and foetus.
No formal clinical drug-drug interaction studies have been performed (see section 5.2).
Women of child-bearing potential
WAINUA will reduce the plasma levels of vitamin A, which is crucial for normal foetal development. It is not known whether vitamin A supplementation will be sufficient to reduce the risk to the foetus. For this reason, pregnancy should be excluded before initiation of WAINUA therapy and women of child-bearing potential should practise effective contraception.
If a woman intends to become pregnant, WAINUA and vitamin A supplementation should be discontinued, and serum vitamin A levels should be monitored and have returned to normal before conception is attempted. Due to the long half-life of eplontersen (see section 5.2), a vitamin A deficit may develop even after cessation of treatment.
Contraception in males and females
Women of child-bearing potential should practise effective contraception.
Pregnancy
There are no data regarding the use of WAINUA in pregnant women.
Administration of eplontersen or a pharmacologically-active rodent-specific surrogate at doses up to 38‑fold higher than the recommended human dose in a combined fertility and embryo-foetal development toxicity study in mice did not result in effects on male and female fertility or embryo‑foetal development (see section 5.3).
Due to the potential teratogenic risk arising from unbalanced vitamin A levels, WAINUA should not be used during pregnancy. In case of pregnancy, close monitoring of the foetus and Vitamin A status should be carried out, especially during the first trimester.
Breast-feeding
Human or animal lactation studies have not been conducted to assess the presence of eplontersen or it’s metabolites in breast milk, the effects on the breastfed infant, or the effects on milk production for the mother. A risk to the breastfed child cannot be excluded. A decision must be made whether to discontinue breast-feeding or to discontinue/abstain from WAINUA therapy, taking into account the benefit of breast feeding for the child and the benefit of therapy for the woman.
Fertility
There is no information available on the effects of eplontersen on human fertility.
Administration of eplontersen or a pharmacologically-active rodent-specific surrogate in doses up to 38‑fold higher than the recommended human exposure in mice did not indicate any impact of eplontersen on male or female fertility.
WAINUA has no or negligible influence on the ability to drive and use machines.
Overall summary of the safety profile
The safety data described below reflects exposure to WAINUA in 144 patients with polyneuropathy caused by ATTRv (ATTRv-PN) randomised to WAINUA and who received at least one dose of WAINUA . Of these, 141 patients received at least 6 months of treatment and 137 patients received at least 12 months of treatment. The mean duration of treatment was 541 days (range: 57 to 582 days).
The most frequent adverse reactions during treatment with WAINUA observed in ≥5% of patients were vomiting, and vitamin A decreased.
Adverse Drug Reactions
Adverse drug reactions (ADRs) are organised by MedDRA System Organ Class (SOC). Within each SOC, preferred terms are arranged by decreasing frequency and then by decreasing seriousness. Frequencies of occurrence of adverse reactions 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 <1/1000); very rare (<1/10,000) and not known (cannot be estimated from available data).
Table 1: Summary of Adverse Reactions per Frequency Category
System Organ Class | Adverse Reaction | Frequency |
Gastrointestinal disorders | Vomiting | Common |
General disorders and administration site conditions | Injection site erythema | Common |
Injection site pain | Common | |
Injection site pruritus | Common | |
Investigations | Vitamin A decreased | Very Common |
Renal and urinary disorders | Proteninuria | Common |
Eye disorder | Cataracts | Common |
Blurred vision |
Description of selected adverse reaction
Vitamin A decreased
In the clinical study in patients with ATTRv-PN, all patients were instructed to take the recommended daily allowance of vitamin A. All patients treated with WAINUA had normal vitamin A levels at baseline, 96.5% of those developed vitamin A levels below the lower limit of normal (LLN) during the study (see section 5.1).
Injection site reactions
In patients with ATTRv-PN treated with WAINUA, injection site erythema, injection site pain and injection site pruritus were reported in 3.5%, 3.5%, and 2.1% respectively.
To report any side effect(s):
- Saudi Arabia:
- The National Pharmacovigilance Centre (NPC) o Toll free phone: 19999 o E-mail: npc.drug@sfda.gov.sa o Website: https://ade.sfda.gov.sa/
|
· Other GCC States:
- Please contact the relevant competent authority.
There is no specific treatment for an overdose with eplontersen. In the event of an overdose, supportive medical care should be provided including consulting with a healthcare professional.
Pharmacotherapeutic group: other nervous system drugs, ATC code: N07XX21
Mechanism of action
Eplontersen is a GalNAc conjugated 2′-O-2-methoxyethyl (2′-MOE)-modified chimeric gapmer antisense oligonucleotide (ASO) with a mixed backbone of phosphorothioate (PS) and phosphate diester (PO) internucleotide linkages. The GalNAc conjugate enables targeted delivery of the ASO to hepatocytes. The selective binding of eplontersen to the TTR messenger RNA (mRNA) within the hepatocytes causes the degradation of both mutant and wild type (normal) TTR mRNA. This prevents the synthesis of TTR protein in the liver, resulting in significant reductions in the levels of mutated and wild type TTR protein secreted by the liver into the circulation.
TTR is a carrier protein for retinol binding protein 4 (RBP4), which is the principal carrier of vitamin A (retinol). Therefore, a reduction in plasma TTR is expected to result in the reduction of plasma retinol levels to below the lower limit of normal.
Pharmacodynamics
In the clinical study in patients with ATTRv-PN receiving eplontersen, a decrease in serum TTR concentrations was observed at the first assessment (Week 5), and TTR concentrations continued to decrease through Week 35. A sustained reduction of TTR concentration was observed throughout the duration of the treatment (85 weeks). Mean (SD) for serum TTR percent reduction from baseline was 82.1% (11.7) at Week 35, 83.0% (10.4) at Week 65 and 81.8% (13.4) at Week 85 when treated with eplontersen. Similar reduction from baseline in serum TTR concentrations compared to placebo was observed regardless of sex, race, age, region, body weight, cardiomyopathy status, previous treatment, Val30Met mutation status, disease stage, and familial amyloid cardiomyopathy (FAC) clinical diagnosis at baseline (Figures 1a and b).
Figure 1: Forest Plot of Treatment Difference in LSM for Percent Change from Baseline in TTR (g/L) for Key Subgroups (NEURO-TTRansform Study) (full analysis set)
a) at Week 35
* External placebo group from another randomised controlled trial (NEURO-TTR).
[a] Previously treated with tafamidis or diflunisal.
Based on MMRM adjusted by propensity score weights with categorical effects for treatment, time, treatment-by-time interaction, and disease stage, Val30Met mutation, previous treatment, and fixed covariates for the baseline and the baseline-by-time-interaction.
Subgroup models also included treatment-by-subgroup, time-by-subgroup, and treatment-by-time-by-subgroup interactions. Only data up to Week 35 are included in the Week 35 interim analysis.
CM subgroup includes patients with either diagnosis of FAC at study entry or baseline IV septum wall thickness ≥ 13 mm with no hypertension [history or diagnosis during the study].
The Week 35 LSM treatment difference (WAINUA – Placebo) with 95% CI (unadjusted) are presented.
CI = Confidence interval; LSM = Least squares mean; MMRM = Mixed effects model with repeated measures; TTR = Transthyretin, CM = cardiomyopathy, FAC = familial amyloid cardiomyopathy.
b) at Week 65
* External placebo group from another randomized controlled trial (NEURO-TTR).
[a] Previously treated with tafamidis or diflunisal.
Based on MMRM adjusted by propensity score weights with categorical effects for treatment, time, treatment-by-time interaction, and disease stage, Val30Met mutation, previous treatment, and fixed covariates for the baseline and the baseline-by-time-interaction.
Subgroup models also included treatment-by-subgroup, time-by-subgroup, and treatment-by-time-by-subgroup interactions.
CM subgroup includes patients with either diagnosis of FAC at study entry or baseline IV septum wall thickness ≥ 13 mm with no hypertension [history or diagnosis during the study].
The Week 65 LSM treatment difference (WAINUA – Placebo) with 95% CI (unadjusted) are presented.
CI = Confidence interval; LSM = Least squares mean; MMRM = Mixed effects model with repeated measures; TTR = Transthyretin, CM = cardiomyopathy, FAC = familial amyloid cardiomyopathy
Cardiac Electrophysiology
Formal QTc studies have not been conducted with WAINUA . The potential for QTc prolongation with eplontersen was evaluated in a randomised, placebo-controlled trial in healthy volunteers. At a dose 2.7 times the recommended dose of 45 mg eplontersen, no clinically relevant effect on the QT interval was observed.
Immunogenicity
In the clinical study in patients with ATTRv-PN, after an 84‑week treatment period (median treatment duration 561 days (80 weeks), range 57 to 582 days), 58 patients (40.3%) developed treatment-emergent anti-drug antibodies (ADAs).
In the patients who tested positive for anti-eplontersen antibodies, there was no clinically meaningful impact on the efficacy, safety, pharmacokinetics, or pharmacodynamics of WAINUA .
Clinical efficacy
The efficacy and safety of WAINUA was evaluated in a randomised, multicentre, open-label, externally-controlled trial (NEURO-TTRansform) that included a total of 168 patients with ATTRv‑PN. Patients were randomised in a 6:1 ratio to receive subcutaneous injection every 4 weeks with WAINUA 45 mg (N=144) or weekly inotersen 284 mg (N=24) as a reference group. Of the 144 patients randomised to eplontersen, 140 (97.2 %) patients completed treatment through Week 35, 135 (93.8%) completed treatment through Week 65 and 130 (90.3%) completed treatment through Week 85.
An external placebo control consisted of a placebo cohort of patients from the inotersen pivotal study: randomised, double-blind, placebo-controlled, multicentre clinical trial in adult patients with ATTRv-PN (NEURO-TTR). That cohort received subcutaneous injections of placebo once weekly. Both studies employed identical eligibility criteria.
The characteristics of the eplontersen and external placebo groups were generally similar, and potential imbalances in key baseline characteristics (age, Val30Met mutation status, disease stage and previous treatment) were accounted in the prespecified statistical analysis. Baseline demographic and disease characteristics are shown in Table 2.
Table 2 Baseline Demographics and Disease Characteristics in NEURO-TTRansform Study (safety set)
* External placebo group from another randomised controlled trial (NEURO-TTR).
1 Disease stage is defined as stage 1 = does not require assistance with ambulation and stage 2 = requires assistance with ambulation.
2 Includes the genotypes V30M, V50M, V50M MUTATION, VAL50MET, and P.VAL50MET.
3 Based on clinical database. Non Val30Met mutations included: GLU89GLN, LEU58HIS, PHE64LEU, SER50ARG, SER77TYR, THR49ALA, THR60ALA, VAL122LLE and other (including ALA97SER).
4 Familial amyloid cardiomyopathy = Hereditary transthyretin-mediated amyloidosis with cardiomyopathy (ATTRv-CM).
5 Denominator for the percentage calculation is the number of patients diagnosed with FAC.
Only year and months were collected from the informed consent date to calculate disease duration from diagnosis and from onset of symptoms of ATTRv-PN, FAC.
N=number of patients in the safety set; n=number of patients in a subgroup, m=number of patients with non-missing data if different from N, CRF=case report form; NT-proBNP= N-terminal proB-type natriuretic peptide; SD=standard deviation.
Week 35 analyses (interim analysis)
The primary efficacy endpoints were the change from baseline to Week 35 in serum transthyretin (TTR) concentration (see Figure 2) and in the modified Neuropathy Impairment Score + 7 (mNIS+7) composite score. The mNIS+7 composite score is an objective assessment of neuropathy and comprises the NIS and Modified +7 composite scores. In the version of the mNIS+7 composite score used in the trial, the NIS objectively measures deficits in cranial nerve function, muscle strength, reflexes, and sensations, and the Modified +7 assesses heart rate response to deep breathing, quantitative sensory testing (touch-pressure and heat-pain), and peripheral nerve electrophysiology. The validated version of the mNIS+7 composite score used in the trial had a range of ‑22.3 to 346.3 points, with higher scores representing a greater severity of disease.
The secondary endpoint was the change from baseline in the Norfolk Quality of Life – Diabetic Neuropathy (QoL-DN) questionnaire total score. The Norfolk QoL-DN scale is a patient-reported assessment that evaluates the subjective experience of neuropathy in the following domains: physical functioning/large fibre neuropathy, activities of daily living, symptoms, small fibre neuropathy, and autonomic neuropathy. The version of the Norfolk QoL-DN total score that was used in the trial had a range from -4 to 136 points, with higher scores representing greater impairment.
WAINUA showed statistically significant improvement compared to external placebo control at Week 35 in reducing serum TTR with percent change of ‑66.43% (95%CI: ‑71.39%, ‑61.47%; p<0.0001) (see Figure 2). WAINUA showed statistically significant improvement compared to external placebo control at Week 35 for mNIS+7 composite score with LSM difference of ‑9.0 (95%CI: ‑13.5, ‑4.5; p<0.0001) (see Figures 3, 4a, 7a). WAINUA showed statistically significant improvement compared to external placebo control at Week 35 for Norfolk QoL-DN total score with LSM difference of ‑11.8 (95%CI: ‑16.8, ‑6.8; p<0.0001) (Table 3 and Figures 5, 6a, 8a).
Week 65/66 (final analysis)
The co-primary endpoints for the primary objective at the final analysis at Week 66 included percent change from baseline in serum TTR concentration at Week 65, change from baseline in mNIS+7 composite score at Week 66 and change from baseline in Norfolk QoL-DN total score at Week 66. At Week 65, the serum TTR concentration reduction was sustained. In addition, the results at Week 66 for the mNIS+7 composite and Norfolk total scores were all consistent with Week 35 results (see Table 3 and Figures 3, 4b, 5, 6b).
The secondary endpoints were change from baseline in neuropathy symptoms and change (NSC) at Weeks 66 and 35, change from baseline in the physical component score (PCS) score of short form 36 item health survey (version 2) (SF-36) at Week 65, change from baseline in polyneuropathy disability (PND) score at Week 65, and change from baseline in modified body mass index (mBMI) at Week 65.
The NSC was a patient-answered questionnaire to quantify the type, distribution, and severity of muscle weakness, sensory symptoms, pain symptoms, and autonomic symptoms. Higher scores represent worse symptoms.
The SF-36 PCS included 4 scales assessing physical function, role limitations caused by physical problems, bodily pain, and general health. Higher scores represent better physical health.
The PND categorises disability by mobility (e.g., need for stick, crutch, wheelchair, or bed). Higher PND score represents worse disability.
Modified BMI (BMI × serum albumin) is an acceptable method of assessing nutritional status in ATTR. Higher scores represent better nutritional status and is considered to be an indicator of longer survival in ATTRv-PN patients.
All secondary endpoints showed statistically significant superiority to external placebo (see Table 4).
Table 3 Treatment Effect for the Primary and Key Secondary Endpoints (NEURO-TTRansform Study) (full analysis set)
* External placebo group from another randomised controlled trial (NEURO-TTR).
1 Based on a MMRM adjusted by propensity score weights with fixed categorical effects for treatment, time, treatment-by-time interaction, and disease stage, Val30M mutation, previous treatment, and fixed covariates for the baseline value and the baseline-by-time interaction. Only data up to Week 66 are included in the Week 66 analysis.
2 Based on an ANCOVA model adjusted by propensity score with the effects of treatment, disease stage, Val30M mutation, previous treatment, and the baseline value. Only data up to Week 35 are included in the interim analysis.
3 Participants with a missing mNIS+7 or Norfolk QoL-DN at Week 35 had value multiply imputed using an imputation model. Each of 500 imputed data sets was analyzed using simple ANCOVA model and the 500 ANCOVA model results were combined using Rubin’s rules.
4 Not formally tested due to statistically significant results at Week 35.
Analysis based on data collected up to 52 days after last dose of study drug. Week 35 data from interim analysis and Week 65/66 data from Week 66 analysis. In the Full Analysis Set, the eplontersen group included 140 participants at Week 35 and 141 participants at Week 66. One participant did not have a mNIS+7 or Norfolk QoL-DN assessment at Week 35 but did have an assessment for at least one of these at Week 66.
ANCOVA = analysis of covariance; CI = confidence interval; LSM = least squares mean; MMRM = mixed effects model with repeated measures; mNIS+7 = modified Neuropathy Impairment Score +7; N = number of participants in group; Norfolk QoL-DN = Norfolk Quality of Life – Diabetic Neuropathy questionnaire; SD = standard deviation; SE = standard error; TTR = transthyretin.
Table 4: Hierarchical Testing of Secondary Endpoints (NEURO-TTRansform Study)
* External placebo group from another randomised controlled trial (NEURO-TTR).
N=Number of patients in Full Analysis Set at Week 66.
n=Number of patients with non-missing data on baseline covariates and change from baseline at the time point.
Analysis based on data collected up to 28 days after last dose of study drug. Analysis visit window of Week 65 is from Day 419 to Day 479.
Based on a mixed effects model with repeated measures (MMRM) adjusted by propensity score weights with fixed categorical effects for treatment, time, treatment-by-time interaction, and disease stage, Val30M mutation, previous treatment, and fixed covariates for the baseline value and the baseline-by-time interaction. Only data up to Week 65 are included in the Week 66 final analysis.
CI = confidence interval; LSM = least squares mean; mBMI = modified body mass index; NSC = neuropathy symptoms and change; PND = polyneuropathy disability; PCS = physical component score; SF‑36 PCS= short form‑36 health survey questionnaire Physical Component Score.
Figure 2: LSM Percent Change in Serum TTR Concentration from Baseline to Week 65, WAINUA vs. External Placebo* through Week 65 (NEURO-TTRansform Study) (full analysis set)
* External placebo group from another randomised controlled trial (NEURO-TTR).
** Treatment difference presents results from formal Week 35 interim analysis. Only data up to Week 35 are included in the Week 35 interim analysis.
Based on MMRM adjusted by propensity score weights with fixed categorical effects for treatment, time, treatment-by-time interaction, and disease stage, Val30Met mutation, previous treatment, and fixed covariates for the baseline and the base-line-by-time interaction.
Analysis based on data collected up to 28 days after last dose of study treatment. Data up to Week 65 are included. Placebo was assessed at Baseline, Weeks 5, 8, 13 23, 35, 47, 57 and 65, WAINUA assessed at Baseline, Weeks 5, 9, 13, 25, 35, 4, 59 and 65.
The Week 35 and Week 65 LS Mean treatment difference (WAINUA – Placebo) with 95% CI (unadjusted) are presented.
CI = Confidence Interval; LSM = Least squares mean; SEM = standard error of mean, MMRM = Mixed effects model with repeated measures; TTR = Transthyretin.
Figure 3: LSM Change in mNIS+7 Composite Score from Baseline (NEURO-TTRansform Study) (full analysis set)
* External placebo group from another randomised controlled trial (NEURO-TTR).
** Treatment difference presents results from formal Week 35 interim analysis. Based on MI ANCOVA adjusted by propensity score weights with fixed categorical effects for treatment, disease stage, Val30Met mutation, previous treatment, and fixed covariates for the baseline. Only data up to Week 35 are included in the Week 35 interim analysis.
Week 66 analysis based on MMRM adjusted by propensity score weights with categorical effects for treatment, time, treatment-by-time interaction, and disease stage, Val30Met mutation, previous treatment, and fixed covariates for the baseline and the baseline-by-time interaction.
Analysis based on data collected up to 52 days after last dose of study treatment. Data up to Week 66 are included.
The Week 35 and Week 65 LS Mean treatment difference (WAINUA – Placebo) with 95% CI (unadjusted) are presented.
CI = Confidence interval; LS Mean = Least squares mean; SEM = standard error of mean, MI ANCOVA = Multiple imputation Analysis of covariance; MMRM = Mixed effects model with repeated measures.
Figure 4: Histogram of mNIS+7 Composite Score Change from Baseline (NEURO-TTRansform Study) (safety analysis set)
a) at Week 35
mNIS +7 Composite Score Change from Baseline at Week 35
* External placebo group from another randomised controlled trial (NEURO-TTR).
b) at Week 66
mNIS +7 Composite Score Change from Baseline at Week 66
* External placebo group from another randomised controlled trial (NEURO-TTR).
Figure 5: LSM Change in Norfolk QoL-DN Total Score from Baseline (NEURO-TTRansform Study)
* External placebo group from another randomised controlled trial (NEURO-TTR).
** Treatment difference presents results from formal Week 35 interim analysis. Based on MI ANCOVA adjusted by propensity score weights with fixed categorical effects for treatment, disease stage, Val30Met mutation, previous treatment, and fixed covariates for the baseline. Only data up to Week 35 are included in the Week 35 interim analysis.
Week 66 analysis based on MMRM adjusted by propensity score weights with categorical effects for treatment, time, treatment-by-time interaction, and disease stage, Val30Met mutation, previous treatment, and fixed covariates for the baseline and the baseline-by-time interaction.
Analysis based on data collected up to 52 days after last dose of study treatment. Data up to Week 66 are included.
The Week 35 and Week 65 LS Mean treatment difference (WAINUA – Placebo) with 95% CI (unadjusted) are presented.
CI = Confidence interval; LS Mean = Least squares mean; SEM = standard error of mean, MI ANCOVA = Multiple imputation Analysis of covariance; MMRM = Mixed effects model with repeated measures.
Figure 6: Histogram of Norfolk QoL-DN Total Score Change from Baseline (NEURO-TTRansform Study) (safety analysis set)
a) at Week 35
Norfolk QoL-DN Total Score Change from Baseline at Week 35
* External placebo group from another randomised controlled trial (NEURO-TTR).
b) at Week 66
Norfolk QoL-DN Total Score Change from Baseline at Week 66
* External placebo group from another randomised controlled trial (NEURO-TTR).
At both Week 35 and Week 65/66, patients receiving WAINUA experienced similar improvements relative to placebo in the reduction of serum TTR concentration, mNIS+7 composite and Norfolk QoL-DN total scores across all subgroups including age, sex, race, region, baseline NIS score, Val30Met mutation status, cardiomyopathy status, FAC diagnosis at baseline, and disease stage (Figures 1a and b, 7a and b and 8a and b).
Figure 7: Forest Plot of Treatment Difference in LSM for Change from Baseline in mNIS+7 Composite Score for Key Subgroups (NEURO-TTRansform Study) (full analysis set)
a) at Week 35
* External placebo group from another randomised controlled trial (NEURO-TTR).
[a] Previously treated with tafamidis or diflunisal.
CM subgroup includes patients with either diagnosis of FAC at study entry or baseline IV septum wall thickness ≥ 13 mm with no hypertension [history or diagnosis during the study].
Difference in LS means, confidence intervals, and p-values are based on an ANCOVA model adjusted by propensity score with the effects of treatment, subgroup factors, disease stage, Val30Met mutation, previous treatment, treatment-by-subgroup interaction, and the Baseline value. Data up to Week 35 are included in the Week 35 analysis.
b) at Week 66
* External placebo group from another randomised controlled trial (NEURO-TTR).
[a] Previously treated with tafamidis or diflunisal.
Based on MMRM adjusted by propensity score weights with categorical effects for treatment, time, treatment-by-time interaction, and disease stage, Val30Met mutation, previous treatment, and fixed covariates for the baseline and the baseline-by-time-interaction.
Subgroup models also included treatment-by-subgroup, time-by-subgroup, and treatment-by-time-by-subgroup interactions. Data up to Week 66 are included.
CM subgroup includes patients with either diagnosis of FAC at study entry or baseline IV septum wall thickness ≥ 13 mm with no hypertension [history or diagnosis during the study].
The Week 66 LSM treatment difference (WAINUA – Placebo) with 95% CI (unadjusted) are presented.
CI = Confidence interval; LSM = Least squares mean; MMRM = Mixed effects model with repeated measures, CM = cardiomyopathy, FAC = familial amyloid cardiomyopathy.
Figure 8: Forest Plot of Treatment Difference in LSM for Change from Baseline in Norfolk QoL‑DN Total Score for Key Subgroups (NEURO-TTRansform Study) (full analysis set)
a) at Week 35
* External placebo group from another randomised controlled trial (NEURO-TTR).
[a] Previously treated with tafamidis or diflunisal.
CM subgroup includes patients with either diagnosis of FAC at study entry or baseline IV septum wall thickness ≥ 13 mm with no hypertension [history or diagnosis during the study].
Difference in LS means, confidence intervals, and p-values are based on an ANCOVA model adjusted by propensity score with the effects of treatment, subgroup factors, disease stage, Val30Met mutation, previous treatment, treatment-by-subgroup interaction, and the Baseline value. Only data up to Week 35 are included in the Week 35 interim analysis.
b) at Week 66
* External placebo group from another randomised controlled trial (NEURO-TTR).
[a] Previously treated with tafamidis or diflunisal.
Based on MMRM adjusted by propensity score weights with categorical effects for treatment, time, treatment-by-time interaction, and disease stage, Val30Met mutation, previous treatment, and fixed covariates for the baseline and the baseline-by-time-interaction.
Subgroup models also included treatment-by-subgroup, time-by-subgroup, and treatment-by-time-by-subgroup interactions. Data up to Week 66 are included.
CM subgroup includes patients with either diagnosis of FAC at study entry or baseline IV septum wall thickness ≥ 13 mm with no hypertension [history or diagnosis during the study].
The Week 66 LSM treatment difference (WAINUA – Placebo) with 95% CI (unadjusted) are presented.
CI = Confidence interval; LSM = Least squares mean; MMRM = Mixed effects model with repeated measures, CM = cardiomyopathy, FAC = familial amyloid cardiomyopathy.
In an exploratory analysis of cardiac assessments with serial echocardiograms, WAINUA demonstrated improvement in E/e’ ratio (a measure of left ventricular diastolic function) after 65 weeks of treatment in the cardiomyopathy subgroup (adjusted placebo-controlled LS mean difference: ‑3.94 [95% CI ‑6.46, ‑1.42]). Directional changes toward benefit of TRDENAME over placebo at week 66 were also observed for pre‑specified exploratory cardiac endpoints of mean LV wall thickness (LSM difference -0.04 cm, [95% CI -0.12, 0.04]), interventricular septal wall thickness (LSM difference -0.05 cm, [95% CI -0.16, 0.06]), and NT‑proBNP, a prognostic biomarker of cardiac dysfunction, (geometric LSM 0.88, [95% CI 0.68, 1.14]). Despite these observed values a clinical benefit in cardiomyopathy is yet to be confirmed.
Week 85 (end of treatment analysis)
Week 85 data are not available for the external placebo group as the treatment period in NEURO-TTR study was only 66 weeks.
The observed effect in the WAINUA treated group in mNIS+7 composite score was consistent and sustained through the end of treatment at Week 85. The mean (SD) change from baseline in mNIS+7 composite score was -0.04% (16.2) at Week 35, -0.21% (17.6) at Week 66 and ‑2.9% (20.5) at Week 85. The mean Norfolk QoL-DN total score remained stable through Week 85. In the eplontersen group the mean (SD) change from baseline in Norfolk QoL-DN total score was -4.8 (16.5) at Week 35, -7.2 (18.5) at Week 66 and ‑6.2 (18.0) at Week 85.
NSC, PND and mBMI remained stable through Week 85, while SF-36 continued to show trend towards improvement.
The pharmacokinetic (PK) properties of WAINUA were evaluated following subcutaneous administration of single and multiple doses (once every 4 weeks) in healthy subjects and multiple doses (once every 4 weeks) in patients with ATTRv-PN.
Absorption
Following subcutaneous administration, eplontersen is absorbed rapidly into systemic circulation with the time to maximum plasma concentrations of approximately 2 hours, based on population estimates.
Distribution
Based on animal studies (mouse, rat, and monkey), eplontersen distributes primarily to the liver and kidney cortex after subcutaneous dosing. Eplontersen is highly bound to human plasma proteins (>98%). The population estimates for the apparent central volume of distribution is 12.9 L and the apparent peripheral volume of distribution is 11,100 L.
Biotransformation
Eplontersen is metabolised by endo- and exonucleases to short oligonucleotide fragments of varying sizes within the liver. There were no major circulating metabolites in humans. Oligonucleotide therapeutics, including eplontersen, are not typically metabolised by CYP enzymes.
Elimination
Eplontersen is primarily eliminated by metabolism followed by renal excretion of the short oligonucleotide metabolites. The mean fraction of unchanged ASO eliminated in urine was less than 1% of the administered dose within 24 hours. The terminal elimination half-life is approximately 3 weeks based on population estimates.
Linearity/non-linearity
Eplontersen Cmax and AUC showed a slightly greater than dose-proportional increase following single subcutaneous doses ranging from 45 to 120 mg (i.e., 1 to 2.7 times the recommended dose) in healthy volunteers.
Population estimates of steady state maximum concentrations (Cmax), trough concentrations (Ctrough), and area under the curve (AUCτ) were 0.218 μg/mL, 0.000200 μg/mL, and 1.95 μg h/mL, respectively, following 45 mg once every 4 weeks dosing in patients with ATTRv-PN. No accumulation of eplontersen Cmax and AUC was observed in plasma after repeated dosing (once every 4 weeks). Accumulation was observed in Ctrough, and steady-state is reached after approximately 17 weeks.
Special populations
Based on the population pharmacokinetic and pharmacodynamic analysis, body weight, sex, race, and Val30Met mutation status have no clinically meaningful effect on eplontersen exposure or serum TTR reductions at steady-state. Definitive assessments were limited in some cases as covariates were limited by the overall low numbers.
Elderly population
No overall differences in pharmacokinetics were observed between adult and elderly (≥65 years of age) patients.
Renal impairment
No formal clinical studies have been conducted to investigate the effect of renal impairment on eplontersen PK. A population pharmacokinetic and pharmacodynamic analysis showed no clinically meaningful differences in the pharmacokinetics or pharmacodynamics of eplontersen based on mild and moderate renal impairment (eGFR≥45 to <90 mL/min). Eplontersen has not been studied in patients with severe renal impairment or in patients with end-stage renal disease.
Hepatic impairment
No formal clinical studies have been conducted to investigate the effect of hepatic impairment on eplontersen. A population pharmacokinetic and pharmacodynamic analysis showed no clinically meaningful differences in the pharmacokinetics or pharmacodynamics of eplontersen based on mild hepatic impairment (total bilirubin ≤1 x ULN and AST >1 x ULN, or total bilirubin >1.0 to 1.5 x ULN and any AST). Eplontersen has not been studied in patients with moderate or severe hepatic impairment or in patients with prior liver transplant.
Drug-Drug Interaction
No formal clinical drug interaction studies have been conducted. In vitro studies indicate that eplontersen is not a substrate or inhibitor of transporters, does not interact with highly plasma protein bound drugs, and is not an inhibitor or inducer of CYP enzymes. Oligonucleotide therapeutics, including eplontersen, are not typically substrates of CYP enzymes. Therefore, eplontersen is not expected to cause or be affected by drug-drug interactions mediated through drug transporters, plasma protein binding or CYP enzymes.
Non-clinical/Repeat-dose toxicity
Repeated administration of eplontersen or rodent specific surrogate produced reduction in hepatic TTR mRNA levels (up to ~62% and 82% reductions in monkeys and mice, respectively), with subsequent decreases in TTR plasma protein levels (up to 70% reduction in monkeys). There were no toxicologically relevant findings related to this pharmacologic inhibition of TTR expression.
Most of the findings observed after repeated dosing for up to 6 months in mice and 9 months in monkeys were related to the uptake and accumulation of eplontersen and were not considered adverse. Microscopic findings related to uptake of eplontersen was observed by various cell types in multiple organs of all tested animal species including monocytes/macrophages, kidney proximal tubular epithelia, Kupffer cells of the liver, and histiocytic cell infiltrates in lymph nodes and injection sites.
Severely decreased platelet counts associated with spontaneous haemorrhage were observed in a sub-chronic toxicity study in one monkey at the highest dose tested (24 mg/kg/week). Similar findings were not observed in monkeys dosed at a mid‑dose of 6 mg/kg/week which is 73‑fold the human AUC at the recommended therapeutic eplontersen dose.
Mutagenicity and carcinogenicity
Eplontersen did not exhibit genotoxic potential in vitro and in vivo and was not carcinogenic in ras.H2 transgenic mice.
Eplontersen was negative for genotoxicity in in vitro (bacterial mutagenicity, chromosomal aberration in Chinese hamster lung) and in vivo (mouse bone marrow micronucleus) assays.
In a subcutaneous carcinogenicity study in ras.H2 transgenic mice, eplontersen was administered for 26 weeks at doses of 250, 500, and 1500 mg/kg/month. There was no evidence of carcinogenicity for eplontersen following 26 weeks of treatment in mice.
Reproductive toxicity
Embryofoetal/Developmental toxicity/Fertility
Eplontersen had no effects on fertility or embryo-foetal development in the mouse up to 38-fold to the recommended human monthly dose of 45 mg. Eplontersen is not pharmacologically active in mice. However, no effect on fertility or embryo-foetal development was noted with a mouse-specific analogue of eplontersen in mice, which was associated with >90% inhibition of TTR mRNA expression.
List of excipients
Sodium dihydrogen phosphate dihydrate
Disodium hydrogen phosphate anhydrous
Sodium chloride
Hydrochloric acid (for pH adjustment)
Sodium hydroxide (for pH adjustment)
Water for injection
In the absence of compatibility studies, this product must not be mixed with other medicinal products.
Store in a refrigerator (2°C – 8°C).
WAINUA may be stored in original carton unrefrigerated for up to 6 weeks below 30°C. If not used within 6 weeks, it should be discarded.
Store in the original package in order to protect from light.
Do not freeze. Do not expose to heat.
0.8 mL sterile solution in a single-use, type I glass syringe with a staked 27‑gauge ½ inch (12.7 mm) stainless steel needle, rigid needle shield, and siliconised chlorobutyl elastomer stopper in an autoinjector.
Pack size of 1 pre-filled autoinjector.
WAINUA should be inspected visually prior to administration. The solution should be clear and colourless to yellow. Do not use WAINUA if the solution is cloudy, contains visible particulate matter or discoloured.
Single‑use pre-filled autoinjector should be discarded in a puncture-resistant sharps container.
Any unused medicinal product or waste material should be disposed of in accordance with local requirements.
صورة المنتج على الرف
الصورة الاساسية
