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)

KANJINTI contains the active substance trastuzumab, which is a monoclonal antibody. Monoclonal antibodies attach to specific proteins or antigens. Trastuzumab is designed to bind selectively to an antigen called human epidermal growth factor receptor 2 (HER2). HER2 is found in large amounts on the surface of some cancer cells where it stimulates their growth. When trastuzumab binds to HER2 it stops the growth of such cells and causes them to die.

 

Your doctor may prescribe KANJINTI for the treatment of breast and gastric cancer when:

·            You have early breast cancer, with high levels of a protein called HER2.

·            You have metastatic breast cancer (breast cancer that has spread beyond the original tumour) with high levels of HER2. KANJINTI may be prescribed in combination with the chemotherapy medicine paclitaxel or docetaxel as first treatment for metastatic breast cancer or it may be prescribed alone if other treatments have proved unsuccessful. It is also used in combination with medicines called aromatase inhibitors with patients with high levels of HER2 and hormone-receptor positive metastatic breast cancer (cancer that is sensitive to the presence of female sex hormones).

·            You have metastatic gastric cancer with high levels of HER2, when it is in combination with the other cancer medicines capecitabine or 5-fluorouracil and cisplatin.

 


Do not use KANJINTI if:

·            you are allergic to trastuzumab, to murine (mouse) proteins, or to any of the other ingredients of this medicine (listed in section 6).

·            you have severe breathing problems at rest due to your cancer or if you need oxygen treatment.

Warnings and precautions

Your doctor will closely supervise your therapy.

 

Heart checks

Treatment with KANJINTI alone or with a taxane may affect the heart, especially if you have ever used an anthracycline (taxanes and anthracyclines are two other kinds of medicine used to treat cancer). The effects may be moderate to severe and could cause death. Therefore, your heart function will be checked before, during (every three months) and after (up to two to five years) treatment with KANJINTI. If you develop any signs of heart failure (inadequate pumping of blood by the heart), your heart function may be checked more frequently (every six to eight weeks), you may receive treatment for heart failure or you may have to stop KANJINTI treatment.

 

Talk to your doctor, pharmacist or nurse before you are given KANJINTI if:

 

·            you have had heart failure, coronary artery disease, heart valve disease (heart murmurs), high blood pressure, taken any high blood pressure medicine or are currently taking any high blood pressure medicine.

·            you have ever had or are currently using a medicine called doxorubicin or epirubicin (medicines used to treat cancer). These medicines (or any other anthracyclines) can damage heart muscle and increase the risk of heart problems with KANJINTI.

·            you suffer from breathlessness, especially if you are currently using a taxane. KANJINTI can cause breathing difficulties, especially when it is first given. This could be more serious if you are already breathless. Very rarely, patients with severe breathing difficulties before treatment have died when they were given trastuzumab.

·            you have ever had any other treatment for cancer.

 

If you receive KANJINTI with any other medicine to treat cancer, such as paclitaxel, docetaxel, an aromatase inhibitor, capecitabine, 5-fluorouracil, or cisplatin you should also read the patient information leaflets for these products.

 

Children and adolescents

KANJINTI is not recommended for anyone under the age of 18 years.

 

Other medicines and KANJINTI

Tell your doctor, pharmacist or nurse if you are taking, have recently taken or may take any other medicines.

 

It may take up to 7 months for KANJINTI to be removed from the body. Therefore you should tell your doctor, pharmacist or nurse that you have had KANJINTI if you start any new medicine in the 7 months after stopping treatment.

 

Pregnancy

·            If you are pregnant, think you may be pregnant or are planning to have a baby, ask your doctor, pharmacist or nurse for advice before taking this medicine.

·            You should use effective contraception during treatment with KANJINTI and for at least 7 months after KANJINTI treatment has ended.

·            Your doctor will advise you of the risks and benefits of taking KANJINTI during pregnancy. In rare cases, a reduction in the amount of (amniotic) fluid that surrounds the developing baby within the womb has been observed in pregnant women receiving trastuzumab. This condition may be harmful to your baby in the womb and has been associated with the lungs not developing fully resulting in foetal death.

 

Breast-feeding

Do not breast-feed your baby during KANJINTI therapy and for 7 months after the last dose as KANJINTI may pass to your baby through your breast milk.

 

Ask your doctor or pharmacist for advice before taking any medicine.

Driving and using machines

KANJINTI may affect your ability to drive a car or use machines. If during treatment you experience symptoms, such as dizziness, sleepiness, chills or fever, you should not drive or use machines until these symptoms disappear.

 

Sodium

 

This medicine contains less than 1 mmol sodium (23 mg) per dose, that is to say essentially ‘sodium‑free’.

 


Before starting the treatment your doctor will determine the amount of HER2 in your tumour. Only patients with a large amount of HER2 will be treated with KANJINTI. KANJINTI should only be given by a doctor or nurse. Your doctor will prescribe a dose and treatment regimen that is right for you. The dose of KANJINTI depends on your body weight.

 

It is important to check the product labels to ensure that the correct formulation is being given as prescribed. KANJINTI intravenous formulation is not for subcutaneous use and should be given as an intravenous infusion only.

 

KANJINTI intravenous formulation is given as an intravenous infusion (“drip”) directly into your veins. The first dose of your treatment is given over 90 minutes and you will be observed by a health professional while it is being given in case you have any side effects. If the first dose is well tolerated the next doses may be given over 30 minutes (see section 2 under “Warnings and precautions”). The number of infusions you receive will depend on how you respond to the treatment. Your doctor will discuss this with you.

 

In order to prevent medication errors it is important to check the vial labels to ensure that the medicine being prepared and given is KANJINTI (trastuzumab) and not another trastuzumab containing product (e.g. trastuzumab emtansine or trastuzumab deruxtecan).

 

For early breast cancer, metastatic breast cancer and metastatic gastric cancer, KANJINTI is given every 3 weeks. KANJINTI may also be given once a week for metastatic breast cancer.

 

If you stop using KANJINTI

Do not stop using this medicine without talking to your doctor first. All doses should be taken at the right time every week or every three weeks (depending on your dosing schedule). This helps your medicine work as well as it can.

 

It may take up to 7 months for KANJINTI to be removed from your body. Therefore your doctor may decide to continue to check your heart functions, even after you finish treatment.

 

If you have any further questions on the use of this medicine, ask your doctor, pharmacist or nurse.


Like all medicines, KANJINTI can cause side effects, although not everybody gets them. Some of these side effects may be serious and may lead to hospitalisation.

 

During a KANJINTI infusion, chills, fever and other flu like symptoms may occur. These are very common (may affect more than 1 in 10 people). Other infusion-related symptoms are: feeling sick (nausea), vomiting, pain, increased muscle tension and shaking, headache, dizziness, breathing difficulties, high or low blood pressure, heart rhythm disturbances (palpitations, heart fluttering or irregular heart beat), swelling of the face and lips, rash and feeling tired. Some of these symptoms can be serious and some patients have died (see section 2 under “Warnings and precautions”).

 

These effects mainly occur with the first intravenous infusion (“drip” into your vein) and during the first few hours after the start of the infusion. They are usually temporary. You will be observed by a health care professional during the infusion and for at least six hours after the start of the first infusion and for two hours after the start of other infusions. If you develop a reaction, they will slow down or stop the infusion and may give you treatment to counteract the side effects. The infusion may be continued after the symptoms improve.

 

Occasionally, symptoms start later than six hours after the infusion begins. If this happens to you, contact your doctor immediately. Sometimes, symptoms may improve and then get worse later.

 

Serious side effects

 

Other side effects can occur at any time during treatment with trastuzumab, not just related to an infusion. Tell a doctor or nurse straight away, if you notice any of the following side effects:

 

·            Heart problems can sometimes occur during treatment and occasionally after treatment has stopped and can be serious. They include weakening of the heart muscle possibly leading to heart failure, inflammation of the lining around the heart and heart rhythm disturbances. This can lead to symptoms such as breathlessness (including breathlessness at night), cough, fluid retention (swelling) in the legs or arms, palpitations (heart fluttering or irregular heart beat) (see section 2 Heart checks).

 

Your doctor will monitor your heart regularly during and after treatment but you should tell your doctor immediately if you notice any of the above symptoms.

 

·            Tumour lysis syndrome (a group of metabolic complications occurring after cancer treatment characterised by high blood levels of potassium and phosphate, and low blood levels of calcium). Symptoms may include kidney problems (weakness, shortness of breath, fatigue and confusion), heart problems (fluttering of the heart or a faster or slower heartbeat), seizures, vomiting or diarrhoea and tingling in the mouth, hands or feet.

 

If you experience any of the above symptoms when your treatment with KANJINTI has finished, you should see your doctor and tell them that you have previously been treated with KANJINTI.

 

Very common side effects (may affect more than 1 in 10 people):

·            infections

·            diarrhoea

·            constipation

·            heartburn (dyspepsia)

·            fatigue

·            skin rashes

·            chest pain

·            abdominal pain

·            joint pain

·            low counts of red blood cells and white blood cells (which help fight infection) sometimes with fever

·            muscle pain

·            conjunctivitis

·            watery eyes

·            nose bleeds

·            runny nose

·            hair loss

·            tremor

·            hot flush

·            dizziness

·            nail disorders

·            weight loss

·            loss of appetite

·            inability to sleep (insomnia)

·            altered taste

·            low platelet count

·            bruising

·            numbness or tingling of the fingers and toes, which occasionally may extend to the rest of the limb

·            redness, swelling or sores in your mouth and/or throat

·            pain, swelling, redness or tingling of hands and/or feet

·            breathlessness

·            headache

·            cough

·            vomiting

·            nausea

 

Common side effects (may affect up to 1 in 10 people):

·            allergic reactions

·            dry mouth and skin

·            throat infections

·            dry eyes

·            bladder and skin infections

·            inflammation of the breast

·            sweating

·            feeling weak and unwell

·            inflammation of the liver

·            anxiety

·            kidney disorders

·            depression

·            increased muscle tone or tension (hypertonia)

·            asthma

·            infection of lungs

·            pain in the arms and/or legs

·            lung disorders

·            itchy rash

·            back pain

·            sleepiness (somnolence)

·            neck pain

·            haemorrhoids

·            bone pain

·            itchiness

·            acne

 

·            leg cramps

 

Uncommon side effects (may affect up to 1 in 100 people):

·            deafness

·            bumpy rash

·            wheezing

·            inflammation or scarring of the lungs

 

Rare side effects (may affect up to 1 in 1,000 people):

·            jaundice

·            anaphylactic reactions

 

Other side effects that have been reported (frequency cannot be estimated from the available data):

·            abnormal or impaired blood clotting

·            high potassium levels

·            swelling or bleeding at the back of the eyes

·            shock

·            abnormal heart rhythm

·            respiratory distress

·            respiratory failure

·            acute accumulation of fluid in the lungs

·            acute narrowing of the airways

·            abnormally low oxygen levels in the blood

·            difficulty in breathing when lying flat

·            liver damage

·            swelling of the face, lips and throat

·            kidney failure

·            abnormally low levels of fluid around baby in womb

·            failure of the lungs of the baby to develop in the womb

·            abnormal development of the kidneys of the baby in the womb

 

Some of the side effects you experience may be due to your underlying cancer. If you receive KANJINTI in combination with chemotherapy, some of them may also be due to the chemotherapy.

 

If you get any side effects, talk to your doctor, pharmacist or nurse.

 

Reporting of side effects

If you get any side effects, talk to your doctor, pharmacist or nurse. This includes any possible side effects not listed in this leaflet. By reporting side effects you can help provide more information on the safety of this medicine.


KANJINTI will be stored by the health professionals at the hospital or clinic.

 

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 outer carton and on the vial label after EXP. The expiry date refers to the last day of that month.

 

Store in a refrigerator (2°C ‑ 8°C). Do not freeze the reconstituted solution. Store in the original package in order to protect from light.

 

Infusion solutions should be used immediately after dilution. If not used immediately, in-use storage times and conditions prior to use are the responsibility of the user, and would not normally be longer than 24 hours at 2°C ‑ 8°C. Do not use KANJINTI if you notice any particulate matter or discolouration prior to administration.

 

Medicines should not be disposed of via wastewater or household waste. Ask your pharmacist how to dispose of medicines no longer required. These measures will help to protect the environment.

 

 


 

 

 

·            The active substance is trastuzumab. Each vial contains either:

-             150 mg trastuzumab that has to be dissolved in 7.2 mL of water for injection, or

-             420 mg trastuzumab that has to be dissolved in 20 mL of water for injection.

·            The resulting solution contains approximately 21 mg/mL trastuzumab.

·            The other ingredient(s) are:

-             in each 1 vial of 150 mg: histidine (2.25 mg), histidine monohydrochloride (3.49 mg), trehalose dihydrate (142 mg), polysorbate 20 (0.6 mg).

-             in each 1 vial of 420 mg: histidine (6.36 mg), histidine monohydrochloride (9.87 mg), trehalose dihydrate (401 mg), polysorbate 20 (1.76 mg).


KANJINTI is a powder for concentrate for solution for intravenous infusion, which is supplied in a glass vial with a rubber stopper containing either 150 mg or 420 mg of trastuzumab. The powder is a white to pale yellow pellet. Each carton contains 1 vial of powder.

Amgen Europe B.V.

Minervum 7061,

NL-4817 ZK Breda,

The Netherlands


This leaflet was last revised in November 2021.
  نشرة الدواء تحت مراجعة الهيئة العامة للغذاء والدواء (اقرأ هذه النشرة بعناية قبل البدء في استخدام هذا المنتج لأنه يحتوي على معلومات مهمة لك)

 

يحتوي كانجنتي على المادة الفعالة تراستوزوماب، وهو جسم مضاد أحادي النسيلة. ترتبط الأجسام المضادة أحادية النسيلة ببروتينات أو مستضادات محددة. صمم تراستوزوماب للربط الانتقائي لمستضد يسمى مستقبل عامل النمو البشري للبشرة ٢ (HER2). يتواجد عامل النمو HER2 بكميات كبيرة على سطح بعض الخلايا السرطانية حيث يحفز نموها. عندما يرتبط تراستوزوماب بعوامل النمو HER2، فإنه يوقف نمو مثل هذه الخلايا ويسبب موتها.

 

قد يصف الطبيب كانجنتي لعلاج سرطان الثدي والمعدة في حالة:

·            كنت مصابة بسرطان ثدي في مراحله الأولى مع ارتفاع مستويات البروتين المسمى HER2.

·            تعانين من سرطان الثدي النقيلي (سرطان الثدي الذي انتشر خارج موضع الورم الأصلي) مع مستويات عالية من HER2. يمكن وصف كانجنتي بالتزامن مع دواء العلاج الكيميائي باكليتاكسيل أو دوسيتاكسيل كعلاج أول لسرطان الثدي النقيلي أو يمكن وصفه بمفرده إذا ثبت أن بعض العلاجات الأخرى غير ناجحة. يستخدم أيضا بالتزامن مع الأدوية التي يطلق عليها مثبطات الأروماتاز لدى المرضى الذين يعانون من مستويات عالية من HER2 و مستقبل هرمون سرطان الثدي النقيلي الإيجابي (سرطان حساس لوجود الهرمونات الجنسية الأنثوية).

·            الإصابة بسرطان المعدة النقيلي مع مستويات عالية من HER2، في حالة التزامن بأدوية السرطان الأخرى مثل كابيسيتابين أو ٥- فلورويوراسيل وسيسبلاتين.

 

لا تستخدم كانجنتي في حالة:

·            لديك حساسية تجاه تراستوزوماب أو بروتينات الفأر أو لأي من مكونات هذا الدواء (المذكورة في القسم ٦).

·            لديك مشاكل شديدة في التنفس عند الراحة بسبب إصابتك بالسرطان أو إذا كنت بحاجة لعلاج بالأوكسجين.

 

التحذيرات والاحتياطات

سيشرف طبيبك عن كثب على علاجك.

 

فحوصات القلب

قد يؤثر العلاج بكانجنتي بمفرده أو مع تاكسين على القلب، وخاصة إذا كنت قد استخدمت أنثراسيكلين من قبل (التاكسينات والإنتراسيكلينات هما نوعان آخران لأدوية تستخدم لعلاج السرطان). قد تكون الآثار متوسطة إلى شديدة ويمكن أن تسبب الموت. لذلك، ستفحص وظيفة قلبك قبل العلاج بكانجنتي وأثنائه (كل ثلاثة أشهر) وبعده (حتى سنتين إلى خمس سنوات). إذا ظهرت أي علامات تشير إلى قصور في القلب (عدم كفاية ضخ القلب للدم)، فيمكن فحص وظائف القلب بشكل أكثر تكرارا (كل ستة إلى ثمانية أسابيع) أو قد تتلقى علاجًا للقصور في القلب أو قد تضطر إلى إيقاف العلاج بكانجنتي.

 

تحدث إلى طبيبك أو الصيدلي أو الممرضة قبل إعطائك كانجنتي في حالة:

 

·            سبق أن عانيت من قصور في القلب أو مرض الشريان التاجي أو داء قلبي صمامي (لغط القلب) أو ارتفاع ضغط الدم أو تناولت أي أدوية لعلاج ضغط الدم المرتفع أو تأخذ حاليا أي أدوية لضغط الدم المرتفع.

·            كنت قد تناولت أو تستخدم حاليا دواء يطلق عليه دوكسوروبيسين أو إبيروبيسين (الأدوية المستخدمة لعلاج السرطان). يمكن لهذه الأدوية (أو أي إنثتراسيكلينات أخرى) أن تضر عضلة القلب وتزيد من مخاطر مشاكل القلب مع كانجنتي.

·            كنت تعاني من ضيق التنفس، وخاصة إذا كنت تستخدم تاكسين حاليا. يمكن أن يسبب كانجنتي مشاكل في التنفس، وخاصة عندما يتم إعطائه لأول مرة. قد يكون ذلك أكثر خطورة إذا كنت تعاني بالفعل من ضيق التنفس. في حالات نادرة للغاية، تعرض المرضى الذين كانوا يعانون من مشاكل شديدة في التنفس قبل العلاج للوفاة عندما تناولوا تراستوزوماب.

·            لم يسبق أن تلقيت أي علاج آخر للسرطان.

 

إذا كنت تتناول كانجنتي مع أي أدوية أخرى تعالج السرطان مثل باكليتاكسيل أو دوسيتاكسيل أو مثبطات الأروماتاز أو كابيسيتابين أو ٥-فلورويوراسيل أو سيسبلاتين، فينبغي عليك أيضا قراءة نشرات معلومات المريض الخاصة بهذه المنتجات.

 

الأطفال والمراهقون

لا يوصى باستخدام كانجنتي لأي شخص يقل عمره عن ١٨ عاما.

 

الأدوية الأخرى وكانجنتي

أخبر طبيبك أو الصيدلي أو الممرضة إذا كنت تتناول أي أدوية أخرى أو تناولتها مؤخرا أو قد تتناولها.

 

قد يستغرق الأمر مدة تصل إلى ٧ أشهر لإزالة كانجنتي من الجسم. ولذلك، يجب أن تخبر طبيبك أو الصيدلي أو الممرضة أنك تناولت كانجنتي إذا بدأت أي دواء جديد خلال ٧ أشهر بعد التوقف عن العلاج.

 

الحمل

·            إذا كنت حاملا، أو كنت تعتقدين أنك حاملا أو تخططين لإنجاب طفل، فاسألي طبيبك أو الصيدلي أو الممرضة للحصول على النصيحة قبل تناول هذا الدواء.

·            يجب عليك استخدام وسائل منع الحمل الفعالة أثناء العلاج بكانجنتي ولمدة ٧ أشهر على الأقل بعد انتهاء العلاج بكانجنتي.

·            سيخبرك طبيبك بمخاطر وفوائد تناول كانجنتي أثناء الحمل. في حالات نادرة، لوحظ وجود انخفاض في كمية السائل (الأمنيوسي) الذي يحيط بالجنين داخل رحم النساء الحوامل اللاتي يتناولن تراستوزوماب. قد تكون هذه الحالة ضارة لطفلك في الرحم، وقد ارتبطت بعدم اكتمال نمو الرئة مما يؤدي إلى موت الجنين.

 

الرضاعة الطبيعية

لا ترضعي طفلك رضاعة طبيعية أثناء العلاج بكانجنتي ولمدة ٧ أشهر بعد آخر جرعة حيث قد ينتقل كانجنتي إلى طفلك عبر لبن الأم.

 

استشيري طبيبك أو الصيدلي قبل تناول أي دواء.

 

القيادة واستخدام الآلات

قد يؤثر كانجنتي على قدرتك على قيادة السيارة أو استخدام الآلات. إذا ظهرت أعراض مثل الدوار أو النعاس أو القشعريرة أو الحمى أثناء العلاج، فينبغي عليك عدم قيادة السيارة أو استخدام الآلات حتى تختفي هذه الأعراض.

 

الصوديوم

يحتوي هذا الدواء على أقل من ١ ملمول (٢٣ مجم) في كل جرعة، أي أنه أساسا "خال من الصوديوم".

https://localhost:44358/Dashboard

 

قبل بدء العلاج، سيحدد طبيبك كمية HER2 في الورم. لا يعالج بكانجنتي إلا المرضى الذين لديهم كميات كبيرة من HER2. ولا ينبغي إعطاء كانجنتي إلا من قبل طبيب أو ممرضة. سيصف طبيبك الجرعة ونظام العلاج المناسب لك. تعتمد جرعة كانجنتي على وزن جسمك.

 

من المهم التحقق من ملصقات المنتج للتأكد من إعطاء التركيبة الصحيحة وفقا لما هو موصوف. التركيبة المعدة للحقن الوريدي من كانجنتي ليست للاستخدام تحت الجلد وينبغي إعطاؤها كتسريب وريدي فقط.

 

تعطى التركيبة المعدة للحقن الوريدي من كانجنتي كتسريب وريدي ("بالتنقيط") مباشرة في أوردتك. تعطى الجرعة الأولى من علاجك على مدى ٩٠ دقيقة وستخضع لمراقبة أخصائي الرعاية الصحية أثناء إعطائها تحسبا لظهور أي آثار جانبية. إذا تحملت الجرعة الأولى بشكل جيد، فيمكن إعطاء الجرعات التالية على مدى ٣٠ دقيقة (انظر القسم ٢ بعنوان "التحذيرات والاحتياطات"). يعتمد عدد مرات التسريب التي تتلقاها على مدى استجابتك للعلاج. سيناقش معك طبيبك ذلك.

 

ولمنع الأخطاء الدوائية، يلزم التحقق من ملصقات القنينة للتأكد من أن الدواء الذي يجري إعداده وإعطاؤه هو كانجنتي (تراستوزوماب) وليس منتجًا آخر يحتوي على التراستوزوماب (على سبيل المثال تراستوزوماب أمتانسين أو تراستوزوماب ديروكستيكان).

 

وفي حالة سرطان الثدي المبكر وسرطان الثدي النقيلي وسرطان المعدة النقيلي، يعطى كانجنتي كل ٣ أسابيع. يمكن أيضا إعطاء كانجنتي مرة واحدة في الأسبوع لعلاج سرطان الثدي النقيلي.

 

إذا توقفت عن استخدام كانجنتي

لا تتوقف عن استخدام هذا الدواء دون التحدث إلى طبيبك أولا. يجب أن تؤخذ جميع الجرعات في الوقت المناسب كل أسبوع أو كل ثلاثة أسابيع (حسب جدول الجرعات الخاص بك). يساعد ذلك على عمل دواءك بشكل جيد.

 

قد يستغرق الأمر ما يصل إلى ٧ أشهر لاختفاء كانجنتي من الجسم. ولذلك قد يقرر الطبيب الاستمرار في فحص وظائف القلب، حتى بعد الانتهاء من العلاج.

 

إذا كان لديك أي أسئلة أخرى حول استخدام هذا الدواء، فاسأل طبيبك أو الصيدلي أو الممرضة.

 

 

كما هو الحال مع جميع الأدوية، يمكن أن يسبب كانجنتي آثارا جانبية على الرغم من عدم إصابة الجميع بها. قد تكون بعض هذه الآثار الجانبية وخيمة وقد تؤدي إلى دخول المستشفى للعلاج.

 

أثناء التسريب الوريدي لكانجنتي، قد تظهر أعراض كالقشعريرة والحمى وأعراض أخرى تتشابه مع أعراض الإنفلونزا. هذا أمر شائع للغاية (قد يؤثر على أكثر من ١ من كل ١٠ أشخاص). الأعراض الأخرى المرتبطة بالتسريب الوريدي هي: الشعور بالمرض (الغثيان) والقيء والألم وزيادة التوتر العضلي والارتجاف والصداع والدوار وصعوبات التنفس وارتفاع أو انخفاض ضغط الدم واضطراب ضربات القلب (خفقان أو رفرفة القلب أو عدم انتظام ضرباته) وتورم الوجه والشفتين والطفح الجلدي والشعور بالتعب. قد تكون بعض هذه الأعراض وخيمة وقد تعرض بعض المرضى للوفاة (انظر القسم ٢ تحت عنوان "التحذيرات والاحتياطات").

 

تحدث هذه الآثار في الغالب مع التسريب الوريدي الأول ("بالتنقيط" في الوريد) وخلال الساعات القليلة الأولى بعد بدء التسريب. وعادة ما تكون مؤقتة. سيراقبك أخصائي الرعاية الصحية أثناء التسريب الوريدي ولمدة ست ساعات على الأقل بعد بدء التسريب الوريدي الأول ولمدة ساعتين بعد بدء عمليات التسريب الأخرى. وإذا حدث رد فعل ما، فسيبطئ أخصائي الرعاية الصحية أو يوقف التسريب الوريدي وقد يعطيك علاجا لمواجهة الآثار الجانبية. وقد يستمر التسريب الوريدي بعد تحسن الأعراض.

 

وفي بعض الأحيان، تبدأ الأعراض بالظهور في وقت لاحق بعد ست ساعات من بدء التسريب الوريدي. إذا حدث لك ذلك، فاتصل بطبيبك على الفور. وفي بعض الأحيان، قد تتحسن الأعراض ثم تزداد سوءا في وقت لاحق.

 

الآثار الجانبية الوخيمة

 

يمكن أن تحدث آثار جانبية أخرى في أي وقت أثناء العلاج بتراستوزوماب، وليست متعلقة بالتسريب فقط. تحدث إلى طبيب أو ممرضة على الفور، إذا لاحظت أي من الآثار الجانبية التالية:

 

·            يمكن أن تحدث مشاكل في القلب في بعض الأحيان أثناء العلاج وأحيانا بعد توقف العلاج ويمكن أن تكون وخيمة. وتشمل ضعف عضلة القلب مما قد يؤدي إلى قصور في القلب أو التهاب في البطانة التي تحيط بالقلب واضطراب ضربات القلب. يمكن أن يؤدي ذلك إلى أعراض مثل ضيق التنفس (بما في ذلك ضيق التنفس في الليل)، السعال، احتباس السوائل (التورم) في الساقين أو الذراعين، الخفقان (رفرفة القلب أو عدم انتظام ضربات القلب) (انظر القسم ٢ "فحوصات القلب").

 

سيراقب طبيبك قلبك بانتظام أثناء العلاج وبعده، ولكن يجب عليك إخبار طبيبك فورا إذا لاحظت أيا من الأعراض المذكورة أعلاه.

 

·            متلازمة انحلال خلايا الورم (مجموعة من المضاعفات الأيضية التي تحدث بعد علاج السرطان، وتتسم بارتفاع مستويات البوتاسيوم والفوسفات في الدم، وانخفاض مستويات الكالسيوم في الدم). وقد تشمل الأعراض مشكلات بالكلى (الضعف وضيق التنفس والإرهاق والارتباك)، ومشكلات بالقلب (رفرفة القلب أو بتسارع نبضاته أو تباطؤها)، والنوبات، والقيء أو الإسهال، والشعور بوخز في الفم أو اليدين أو القدمين.

إذا واجهت أي من الأعراض المذكورة أعلاه عند انتهاء علاجك بكانجنتي، فينبغي أن تزور الطبيب وتخبره بأنك قد عولجت مسبقًا بكانجنتي.

 

آثار جانبية شائعة جدا (قد تؤثر على أكثر من ١ عن كل ١٠ أشخاص):

·            العدوى

·            الإسهال

·            الإمساك

·            حرقان في المعدة (عسر الهضم)

·            الإرهاق

·            الطفح الجلدي

·            ألم في الصدر

·            ألم في البطن

·            ألم في المفاصل

·            انخفاض عدد خلايا الدم الحمراء وخلايا الدم البيضاء (التي تساعد في مكافحة العدوى) أحيانا مع الحمى

·            ألم في العضلات

·            التهاب الملتحمة

·            الدمعان

·            نزيف الأنف

·            سيلان الأنف

·            سقوط الشعر

·            رعاش

·            هبة الحرارة

·            الدوار

·            اضطرابات الأظافر

·            فقدان الوزن

·            فقدان الشهية

·            عدم القدرة على النوم (الأرق)

·            تغير في التذوق

·            انخفاض عدد الصفائح الدموية

·            كدمات

·            خدر أو وخز في أصابع اليدين والقدمين، قد يمتد إلى باقي الطرف في بعض الأحيان

·            إحمرار أو تورم أو تقرحات في الفم و/أو الحلق

·            ألم أو تورم أو احمرار أو وخز في اليدين و/أو القدمين

·            ضيق التنفس

·            الصداع

·            السعال

·            القيء

·            الغثيان

آثار جانبية شائعة (قد تؤثر إلى حدود ١ عن كل ١٠ أشخاص):

·            تفاعلات تحسسية

·            جفاف الفم والجلد

·            التهابات الحلق

·            جفاف العين

·            التهابات المثانة والالتهابات الجلدية

·            التهاب في الثدي

·            التهاب الكبد

·            اضطرابات الكلى

·            زيادة التوتر العضلي (فرط التوتر)

·            ألم في الذراعين و/أو الساقين

·            طفح جلدي مصحوب بحكة

·            النعاس (النيمومة)

·            البواسير

·            الحكة

·            التعرق

·            الشعور بالضعف وعدم الراحة

·            القلق

·            الاكتئاب

·            الربو

·            عدوى الرئتين

·            اضطرابات الرئة

·            ألم في الظهر

·            ألم في الرقبة

·            ألم في العظام

·            حب الشباب

·            تشنجات الساق

 

آثار جانبية غير شائعة (قد تؤثر إلى حدود ١ عن كل ١٠٠ شخص):

·            الصمم

·            طفح جلدي مغطى بالفقاعات

·            الأزيز

·            التهاب أو تقرح في الرئتين

 

آثار جانبية نادرة (قد تؤثر إلى حدود ١ عن كل ١٠٠٠ شخص):

·            اليرقان

·            ردود الأفعال التأقية

 

الآثار الجانبية الأخرى التي تم الإبلاغ عنها (لا يمكن تقدير التكرار من البيانات المتاحة):

·            تخثر الدم غير الطبيعي أو المضطرب

·            ارتفاع مستويات البوتاسيوم

·            تورم أو نزيف في الجزء الخلفي من العين

·            الصدمة

·            ضربات القلب غير الطبيعية

·            الضائقة التنفسية

·            القصور التنفسي

·            تراكم حاد للسوائل في الرئتين

·            تضيق حاد في الشعب الهوائية

·            انخفاض غير طبيعي في مستويات الأوكسجين في الدم

·            صعوبة في التنفس عند الاستلقاء

·            تلف الكبد

·            تورم في الوجه والشفتين والحنجرة

·            االقصور الكلوي

·            مستويات منخفضة بشكل غير طبيعي من السوائل حول الجنين في الرحم

·            قصور في نمو رئتي الجنين في الرحم

·            نمو غير طبيعي لكلى الجنين في الرحم

 

قد تكون بعض الآثار الجانبية التي تواجهها بسبب السرطان الكامن لديك. إذا كنت تتلقى كانجنتي بالتزامن مع العلاج الكيميائي، فقد يكون بعضها أيضا بسبب العلاج الكيميائي.

 

إذا تعرضت لأي آثار جانبية، فتحدث إلى طبيبك أو الصيدلي أو الممرضة.

 

الإبلاغ عن الآثار الجانبية

إذا تعرضت لأي آثار جانبية، فتحدث إلى طبيبك أو الصيدلي أو الممرضة. ويشمل ذلك أي آثار جانبية محتملة غير مدرجة في هذه النشرة. بالإبلاغ عن الآثار الجانبية، يمكنك المساعدة في تقديم مزيد من المعلومات حول سلامة هذا الدواء.

 

 

سيقوم أخصائيو الرعاية الصحية بتخزين كانجنتي في المستشفى أو العيادة

 

يحفظ هذا الدواء بعيدا عن مرأى ومتناول الأطفال.

 

لا تستخدم هذا الدواء بعد تاريخ انتهاء الصلاحية المدون على العلبة الخارجية وعلى ملصق القنينة بعد EXP. يشير تاريخ انتهاء الصلاحية إلى آخر يوم من ذلك الشهر.

 

يحفظ في الثلاجة (٢°م ‑ ٨°م). لا تجمد المحلول المعاد تكوينه. يحفظ في العبوة الأصلية من أجل الحماية من الضوء.

 

ينبغي استخدام محاليل التسريب الوريدي مباشرة بعد التخفيف. وإذا لم تستخدم على الفور، فإن مسؤولية مدة وظروف تخزين المحاليل المخففة قبل الاستخدام تقع على عاتق المستخدم، ويجب ألا تزيد فترة التخزين عادة عن ٢٤ ساعة في درجة حرارة تتراوح بين ٢°م ‑ ٨°م. لا تستخدم كانجنتي إذا لاحظت وجود أي جسيمات أو تغير في لونه قبل الاستخدام.

 

ينبغي عدم التخلص من الأدوية عن طريق إلقائها في الصرف الصحي أو النفايات المنزلية. اسأل الصيدلي عن كيفية التخلص من الأدوية التي لم تعد مطلوبة. وستساعد هذه التدابير على حماية البيئة.

 

 

 

·            المادة الفعالة هي تراستوزوماب. تحتوي كل قنينة على إما:

-             ١٥٠ مجم تراستوزوماب اللازم حله في ٧٫٢ مل من ماء الحقن، أو

-             ٤٢٠ مجم تراستوزوماب اللازم حله في ٢٠ مل من ماء الحقن.

·            يحتوي المحلول الناتج على حوالي ٢١ مجم/مل  من تراستوزوماب.

·            المكونات الأخرى هي:

-        في كل ١ قنينة من ١٥٠ مجم: هيستيدين (٢٫٢٥ مجم)، مونوهيدروكلوريد الهيستدين (٣٫٤٩ مجم)، طرهالوز ثنائي الهيدرات (١٤٢ مجم)، بولي سوربات ٢٠ (٠٫٦ مجم).

-        في كل ١ قنينة من ٤٢٠ مجم: هيستيدين (٦٫٣٦ مجم)، مونوهيدروكلوريد الهيستدين (٩٫٨٧ مجم)، طرهالوز ثنائي الهيدرات (٤٠١ مجم)، بولي سوربات ٢٠(١٫٧٦ مجم).

 

 

كانجنتي هو مسحوق مركز لمحلول التسريب الوريدي، والذي يتوفر في قنينة زجاجية مع سدادة مطاطية تحتوي إما على ١٥٠ مجم أو ٤٢٠ مجم من تراستوزوماب. يميل لون المسحوق الأبيض إلى الأصفر الباهت. تحتوي كل علبة على قنينة واحدة من المسحوق.

 

 

Amgen Europe B.V.

Minervum 7061

 NL- 4817 ZK Breda

هولندا

تم اعتماد هذه النشرة في نوفمبر ٢٠٢١.
 Read this leaflet carefully before you start using this product as it contains important information for you

KANJINTI 150 mg powder for concentrate for solution for infusion KANJINTI 420 mg powder for concentrate for solution for infusion

KANJINTI 150 mg powder for concentrate for solution for infusion One vial contains 150 mg of trastuzumab, a humanised IgG1 monoclonal antibody produced by mammalian (Chinese hamster ovary) cell suspension culture and purified by affinity and ion exchange chromatography including specific viral inactivation and removal procedures. KANJINTI 420 mg powder for concentrate for solution for infusion One vial contains 420 mg of trastuzumab, a humanised IgG1 monoclonal antibody produced by mammalian (Chinese hamster ovary) cell suspension culture and purified by affinity and ion exchange chromatography including specific viral inactivation and removal procedures. The reconstituted KANJINTI solution contains 21 mg/mL of trastuzumab. For the full list of excipients, see section 6.1.

Powder for concentrate for solution for infusion. White to pale yellow lyophilised powder.

 

Breast cancer

 

Metastatic breast cancer

 

KANJINTI is indicated for the treatment of adult patients with HER2 positive metastatic breast cancer (MBC):

 

-             as monotherapy for the treatment of those patients who have received at least two chemotherapy regimens for their metastatic disease. Prior chemotherapy must have included at least an anthracycline and a taxane unless patients are unsuitable for these treatments. Hormone-receptor positive patients must also have failed hormonal therapy, unless patients are unsuitable for these treatments.

 

-             in combination with paclitaxel for the treatment of those patients who have not received chemotherapy for their metastatic disease and for whom an anthracycline is not suitable.

 

-             in combination with docetaxel for the treatment of those patients who have not received chemotherapy for their metastatic disease.

 

-             in combination with an aromatase inhibitor for the treatment of postmenopausal patients with hormone-receptor positive MBC, not previously treated with trastuzumab.

 

Early breast cancer

 

KANJINTI is indicated for the treatment of adult patients with HER2-positive early breast cancer (EBC):

 

-             following surgery, chemotherapy (neoadjuvant or adjuvant) and radiotherapy (if applicable) (see section 5.1).

 

-             following adjuvant chemotherapy with doxorubicin and cyclophosphamide, in combination with paclitaxel or docetaxel.

 

-             in combination with adjuvant chemotherapy consisting of docetaxel and carboplatin.

 

-             in combination with neoadjuvant chemotherapy followed by adjuvant KANJINTI therapy, for locally advanced (including inflammatory) disease or tumours > 2 cm in diameter (see sections 4.4 and 5.1).

 

KANJINTI should only be used in patients with metastatic or early breast cancer whose tumours have either HER2 overexpression or HER2 gene amplification as determined by an accurate and validated assay (see sections 4.4 and 5.1).

 

Metastatic gastric cancer

 

KANJINTI in combination with capecitabine or 5-fluorouracil and cisplatin is indicated for the treatment of adult patients with HER2-positive metastatic adenocarcinoma of the stomach or gastroesophageal junction who have not received prior anti-cancer treatment for their metastatic disease.

 

KANJINTI should only be used in patients with metastatic gastric cancer (MGC) whose tumours have HER2 overexpression as defined by IHC 2+ and a confirmatory SISH or FISH result, or by an IHC 3+ result. Accurate and validated assay methods should be used (see sections 4.4 and 5.1).


HER2 testing is mandatory prior to initiation of therapy (see sections 4.4 and 5.1). KANJINTI treatment should only be initiated by a physician experienced in the administration of cytotoxic chemotherapy (see section 4.4), and should be administered by a healthcare professional only.

 

KANJINTI intravenous formulation is not intended for subcutaneous administration and should be administered via an intravenous infusion only.

 

In order to prevent medication errors it is important to check the vial labels to ensure that the medicinal product being prepared and administered is KANJINTI (trastuzumab) and not another trastuzumab-containing product (e.g. trastuzumab emtansine or trastuzumab deruxtecan).

 

Posology

 

Metastatic breast cancer

 

Three-weekly schedule

The recommended initial loading dose is 8 mg/kg body weight. The recommended maintenance dose at three‑weekly intervals is 6 mg/kg body weight, beginning three weeks after the loading dose.

 

Weekly schedule

The recommended initial loading dose of KANJINTI is 4 mg/kg body weight. The recommended weekly maintenance dose of KANJINTI is 2 mg/kg body weight, beginning one week after the loading dose.

 

Administration in combination with paclitaxel or docetaxel

In the pivotal trials (H0648g, M77001), paclitaxel or docetaxel was administered the day following the first dose of trastuzumab (for dose, see the Summary of Product Characteristics (SmPC) for paclitaxel or docetaxel) and immediately after the subsequent doses of trastuzumab if the preceding dose of trastuzumab was well tolerated.

 

Administration in combination with an aromatase inhibitor

In the pivotal trial (BO16216) trastuzumab and anastrozole were administered from day 1. There were no restrictions on the relative timing of trastuzumab and anastrozole at administration (for dose, see the SmPC for anastrozole or other aromatase inhibitors).

 

Early breast cancer

 

Three‑weekly and weekly schedule

As a three‑weekly regimen the recommended initial loading dose of KANJINTI is 8 mg/kg body weight. The recommended maintenance dose of KANJINTI at three-weekly intervals is 6 mg/kg body weight, beginning three weeks after the loading dose.

 

As a weekly regimen (initial loading dose of 4 mg/kg followed by 2 mg/kg every week) concomitantly with paclitaxel following chemotherapy with doxorubicin and cyclophosphamide.

 

See section 5.1 for chemotherapy combination dosing.

 

Metastatic gastric cancer

 

Three‑weekly schedule

The recommended initial loading dose is 8 mg/kg body weight. The recommended maintenance dose at three-weekly intervals is 6 mg/kg body weight, beginning three weeks after the loading dose.

 

Breast cancer and gastric cancer

 

Duration of treatment

Patients with MBC or MGC should be treated with KANJINTI until progression of disease. Patients with EBC should be treated with KANJINTI for 1 year or until disease recurrence, whichever occurs first; extending treatment in EBC beyond one year is not recommended (see section 5.1).

 

Dose reduction

No reductions in the dose of trastuzumab were made during clinical trials. Patients may continue therapy during periods of reversible, chemotherapy-induced myelosuppression but they should be monitored carefully for complications of neutropenia during this time. Refer to the SmPC for paclitaxel, docetaxel or aromatase inhibitor for information on dose reduction or delays.

 

If left ventricular ejection fraction (LVEF) percentage drops ≥ 10 points from baseline AND to below 50%, treatment should be suspended and a repeat LVEF assessment performed within approximately 3 weeks. If LVEF has not improved, or has declined further, or if symptomatic congestive heart failure (CHF) has developed, discontinuation of KANJINTI should be strongly considered, unless the benefits for the individual patient are deemed to outweigh the risks. All such patients should be referred for assessment by a cardiologist and followed up.

 

Missed doses

If the patient has missed a dose of KANJINTI by one week or less, then the usual maintenance dose (weekly regimen: 2 mg/kg; three‑weekly regimen: 6 mg/kg) should be administered as soon as possible. Do not wait until the next planned cycle. Subsequent maintenance doses should be administered 7 days or 21 days later according to the weekly or three-weekly schedules, respectively.

 

If the patient has missed a dose of KANJINTI by more than one week, a re-loading dose of KANJINTI should be administered over approximately 90 minutes (weekly regimen: 4 mg/kg; three‑weekly regimen: 8 mg/kg) as soon as possible. Subsequent KANJINTI maintenance doses (weekly regimen: 2 mg/kg; three‑weekly regimen 6 mg/kg respectively) should be administered 7 days or 21 days later according to the weekly or three-weekly schedules respectively.

 

Special populations

Dedicated pharmacokinetic studies in the elderly and those with renal or hepatic impairment have not been carried out. In a population pharmacokinetic analysis, age and renal impairment were not shown to affect trastuzumab disposition.

 

Paediatric population

There is no relevant use of trastuzumab in the paediatric population.

 

Method of administration

 

KANJINTI is for intravenous use only. The loading dose should be administered as a 90‑minute intravenous infusion. Do not administer as an intravenous push or bolus. KANJINTI intravenous infusion should be administered by a healthcare provider prepared to manage anaphylaxis and an emergency kit should be available. Patients should be observed for at least six hours after the start of the first infusion and for two hours after the start of the subsequent infusions for symptoms like fever and chills or other infusion‑related symptoms (see sections 4.4 and 4.8). Interruption or slowing the rate of the infusion may help control such symptoms. The infusion may be resumed when symptoms abate.

 

If the initial loading dose was well tolerated, the subsequent doses can be administered as a 30‑minute infusion.

 

For instructions on reconstitution of KANJINTI intravenous formulation before administration, see section 6.6.


• Hypersensitivity to trastuzumab, murine proteins, or to any of the excipients listed in section 6.1 • Severe dyspnoea at rest due to complications of advanced malignancy or requiring supplementary oxygen therapy.

Traceability

 

In order to improve the traceability of biological medicinal products, the trade name and the batch number of the administered product should be clearly recorded.

 

HER2 testing must be performed in a specialised laboratory which can ensure adequate validation of the testing procedures (see section 5.1).

Currently no data from clinical trials are available on re-treatment of patients with previous exposure to trastuzumab in the adjuvant setting.

 

Cardiac dysfunction

 

General considerations

 

Patients treated with KANJINTI are at increased risk for developing CHF (New York Heart Association [NYHA] class II‑IV) or asymptomatic cardiac dysfunction. These events have been observed in patients receiving trastuzumab therapy alone or in combination with paclitaxel or docetaxel, particularly following anthracycline (doxorubicin or epirubicin) containing chemotherapy. These may be moderate to severe and have been associated with death (see section 4.8). In addition, caution should be exercised in treating patients with increased cardiac risk, e.g. hypertension, documented coronary artery disease, CHF, LVEF of < 55%, older age.

 

All candidates for treatment with KANJINTI, but especially those with prior anthracycline and cyclophosphamide (AC) exposure, should undergo baseline cardiac assessment including history and physical examination, electrocardiogram (ECG), echocardiogram, and/or multigated acquisition (MUGA) scan or magnetic resonance imaging. Monitoring may help to identify patients who develop cardiac dysfunction. Cardiac assessments, as performed at baseline, should be repeated every 3 months during treatment and every 6 months following discontinuation of treatment until 24 months from the last administration of KANJINTI. A careful risk-benefit assessment should be made before deciding to treat with KANJINTI.

 

Trastuzumab may persist in the circulation for up to 7 months after stopping KANJINTI treatment based on population pharmacokinetic analysis of all available data (see section 5.2). Patients who receive anthracyclines after stopping KANJINTI may possibly be at increased risk of cardiac dysfunction. If possible, physicians should avoid anthracycline-based therapy for up to 7 months after stopping KANJINTI. If anthracyclines are used, the patient’s cardiac function should be monitored carefully.

 

Formal cardiological assessment should be considered in patients in whom there are cardiovascular concerns following baseline screening. In all patients cardiac function should be monitored during treatment (e.g. every 12 weeks). Monitoring may help to identify patients who develop cardiac dysfunction. Patients who develop asymptomatic cardiac dysfunction may benefit from more frequent monitoring (e.g. every 6 ‑ 8 weeks). If patients have a continued decrease in left ventricular function, but remain asymptomatic, the physician should consider discontinuing therapy if no clinical benefit of KANJINTI therapy has been seen.

 

The safety of continuation or resumption of trastuzumab in patients who experience cardiac dysfunction has not been prospectively studied. If LVEF percentage drops ≥ 10 points from baseline AND to below 50%, treatment should be suspended and a repeat LVEF assessment performed within approximately 3 weeks. If LVEF has not improved, or declined further, or symptomatic CHF has developed, discontinuation of KANJINTI should be strongly considered, unless the benefits for the individual patient are deemed to outweigh the risks. All such patients should be referred for assessment by a cardiologist and followed up.

 

If symptomatic cardiac failure develops during KANJINTI therapy, it should be treated with standard medicinal products for CHF. Most patients who developed CHF or asymptomatic cardiac dysfunction in pivotal trials improved with standard CHF treatment consisting of an angiotensin‑converting enzyme (ACE) inhibitor or angiotensin receptor blocker (ARB) and a beta-blocker. The majority of patients with cardiac symptoms and evidence of a clinical benefit of trastuzumab treatment continued on therapy without additional clinical cardiac events.

 

Metastatic breast cancer

 

KANJINTI and anthracyclines should not be given concurrently in combination in the MBC setting.

Patients with MBC who have previously received anthracyclines are also at risk of cardiac dysfunction with KANJINTI treatment, although the risk is lower than with concurrent use of KANJINTI and anthracyclines.

 

Early breast cancer

 

For patients with EBC, cardiac assessments, as performed at baseline, should be repeated every 3 months during treatment and every 6 months following discontinuation of treatment until 24 months from the last administration of KANJINTI. In patients who receive anthracycline‑containing chemotherapy further monitoring is recommended, and should occur yearly up to 5 years from the last administration of KANJINTI, or longer if a continuous decrease of LVEF is observed.

 

Patients with history of myocardial infarction (MI), angina pectoris requiring medical treatment, history of or existing CHF (NYHA class II–IV), LVEF of < 55%, other cardiomyopathy, cardiac arrhythmia requiring medical treatment, clinically significant cardiac valvular disease, poorly controlled hypertension (hypertension controlled by standard medical treatment eligible), and haemodynamic effective pericardial effusion were excluded from adjuvant and neoadjuvant EBC pivotal trials with trastuzumab and therefore treatment cannot be recommended in such patients.

 

Adjuvant treatment

 

KANJINTI and anthracyclines should not be given concurrently in combination in the adjuvant treatment setting.

 

In patients with EBC an increase in the incidence of symptomatic and asymptomatic cardiac events was observed when trastuzumab was administered after anthracycline‑containing chemotherapy compared to administration with a non‑anthracycline regimen of docetaxel and carboplatin and was more marked when trastuzumab was administered concurrently with taxanes than when administered sequentially to taxanes. Regardless of the regimen used, most symptomatic cardiac events occurred within the first 18 months. In one of the 3 pivotal studies conducted in which a median follow‑up of 5.5 years was available (BCIRG 006) a continuous increase in the cumulative rate of symptomatic cardiac or LVEF events was observed in patients who were administered trastuzumab concurrently with a taxane following anthracycline therapy up to 2.37% compared to approximately 1% in the two comparator arms (anthracycline plus cyclophosphamide followed by taxane and taxane, carboplatin and trastuzumab).

 

Risk factors for a cardiac event identified in four large adjuvant studies included advanced age (> 50 years), low LVEF (< 55%) at baseline, prior to or following the initiation of paclitaxel treatment, decline in LVEF by 10‑15 points, and prior or concurrent use of anti-hypertensive medicinal products. In patients receiving trastuzumab after completion of adjuvant chemotherapy, the risk of cardiac dysfunction was associated with a higher cumulative dose of anthracycline given prior to initiation of trastuzumab and a body mass index (BMI) > 25 kg/m2.

 

Neoadjuvant‑adjuvant treatment

 

In patients with EBC eligible for neoadjuvant‑adjuvant treatment, KANJINTI should be used concurrently with anthracyclines only in chemotherapy‑naive patients and only with low‑dose anthracycline regimens i.e. maximum cumulative doses of doxorubicin 180 mg/m2 or epirubicin 360 mg/m2.

 

If patients have been treated concurrently with a full course of low-dose anthracyclines and KANJINTI in the neoadjuvant setting, no additional cytotoxic chemotherapy should be given after surgery. In other situations, the decision on the need for additional cytotoxic chemotherapy is determined based on individual factors.

 

Experience of concurrent administration of trastuzumab with low-dose anthracycline regimens is currently limited to two trials (MO16432 and BO22227).

In the pivotal trial MO16432, trastuzumab was administered concurrently with neoadjuvant chemotherapy containing three cycles of doxorubicin (cumulative dose 180 mg/m2).

 

The incidence of symptomatic cardiac dysfunction was 1.7% in the trastuzumab arm.

 

In the pivotal trial BO22227, trastuzumab was administered concurrently with neoadjuvant chemotherapy that contained four cycles of epirubicin (cumulative dose 300 mg/m2); at a median follow-up exceeding 70 months, the incidence of cardiac failure/congestive cardiac failure was 0.3% in the trastuzumab intravenous arm.

 

Clinical experience is limited in patients above 65 years of age.

 

Infusion‑related reactions (IRRs) and hypersensitivity

 

Serious IRRs to trastuzumab infusion including dyspnoea, hypotension, wheezing, hypertension, bronchospasm, supraventricular tachyarrhythmia, reduced oxygen saturation, anaphylaxis, respiratory distress, urticaria and angioedema have been reported (see section 4.8). Pre‑medication may be used to reduce risk of occurrence of these events. The majority of these events occur during or within 2.5 hours of the start of the first infusion. Should an infusion reaction occur the infusion should be discontinued or the rate of infusion slowed and the patient should be monitored until resolution of all observed symptoms (see section 4.2). These symptoms can be treated with an analgesic/antipyretic such as meperidine or paracetamol, or an antihistamine such as diphenhydramine. The majority of patients experienced resolution of symptoms and subsequently received further infusions of trastuzumab. Serious reactions have been treated successfully with supportive therapy such as oxygen, beta-agonists, and corticosteroids. In rare cases, these reactions are associated with a clinical course culminating in a fatal outcome. Patients experiencing dyspnoea at rest due to complications of advanced malignancy and comorbidities may be at increased risk of a fatal infusion reaction. Therefore, these patients should not be treated with KANJINTI (see section 4.3).

 

Initial improvement followed by clinical deterioration and delayed reactions with rapid clinical deterioration have also been reported. Fatalities have occurred within hours and up to one week following infusion. On very rare occasions, patients have experienced the onset of infusion symptoms and pulmonary symptoms more than six hours after the start of the trastuzumab infusion. Patients should be warned of the possibility of such a late onset and should be instructed to contact their physician if these symptoms occur.

 

Pulmonary events

 

Severe pulmonary events have been reported with the use of trastuzumab in the post‑marketing setting (see section 4.8). These events have occasionally been fatal. In addition, cases of interstitial lung disease including lung infiltrates, acute respiratory distress syndrome, pneumonia, pneumonitis, pleural effusion, respiratory distress, acute pulmonary oedema and respiratory insufficiency have been reported. Risk factors associated with interstitial lung disease include prior or concomitant therapy with other antineoplastic therapies known to be associated with it such as taxanes, gemcitabine, vinorelbine and radiation therapy. These events may occur as part of an infusion‑related reaction or with a delayed onset. Patients experiencing dyspnoea at rest due to complications of advanced malignancy and comorbidities may be at increased risk of pulmonary events. Therefore, these patients should not be treated with KANJINTI (see section 4.3). Caution should be exercised for pneumonitis, especially in patients being treated concomitantly with taxanes.

 

Sodium

 

This medicinal product contains less than 1 mmol sodium (23 mg) per dose, that is to say essentially ‘sodium‑free’.

 


No formal drug interaction studies have been performed. Clinically significant interactions between trastuzumab and the concomitant medicinal products used in clinical trials have not been observed.

 

Effect of trastuzumab on the pharmacokinetics of other antineoplastic agents

 

Pharmacokinetic (PK) data from studies BO15935 and M77004 in women with HER2‑positive MBC suggested that exposure to paclitaxel and doxorubicin (and their major metabolites 6‑α hydroxylpaclitaxel, POH, and doxorubicinol, DOL) was not altered in the presence of trastuzumab (8 mg/kg or 4 mg/kg intravenous loading dose followed by 6 mg/kg q3w or 2 mg/kg q1w intravenous, respectively). However, trastuzumab may elevate the overall exposure of one doxorubicin metabolite, (7‑deoxy‑13 dihydro-doxorubicinone, D7D). The bioactivity of D7D and the clinical impact of the elevation of this metabolite was unclear.

 

Data from study JP16003, a single‑arm study of trastuzumab (4 mg/kg intravenous loading dose and 2 mg/kg intravenous weekly) and docetaxel (60 mg/m2 intravenous) in Japanese women with HER2‑positive MBC, suggested that concomitant administration of trastuzumab had no effect on the single-dose pharmacokinetics of docetaxel. Study JP19959 was a substudy of BO18255 (ToGA) performed in male and female Japanese patients with advanced gastric cancer to study the pharmacokinetics of capecitabine and cisplatin when used with or without trastuzumab. The results of this substudy suggested that the exposure to the bioactive metabolites (e.g. 5‑FU) of capecitabine was not affected by concurrent use of cisplatin or by concurrent use of cisplatin plus trastuzumab. However, capecitabine itself showed higher concentrations and a longer half‑life when combined with trastuzumab. The data also suggested that the pharmacokinetics of cisplatin were not affected by concurrent use of capecitabine or by concurrent use of capecitabine plus trastuzumab.

 

Pharmacokinetic data from study H4613g/GO01305 in patients with metastatic or locally advanced inoperable HER2‑positive cancer suggested that trastuzumab had no impact on the PK of carboplatin.

 

Effect of antineoplastic agents on trastuzumab pharmacokinetics

 

By comparison of simulated serum trastuzumab concentrations after trastuzumab monotherapy (4 mg/kg loading/2 mg/kg q1w intravenous) and observed serum concentrations in Japanese women with HER2‑positive MBC (study JP16003) no evidence of a PK effect of concurrent administration of docetaxel on the pharmacokinetics of trastuzumab was found.

 

Comparison of PK results from two Phase II studies (BO15935 and M77004) and one Phase III study (H0648g) in which patients were treated concomitantly with trastuzumab and paclitaxel and two Phase II studies in which trastuzumab was administered as monotherapy (WO16229 and MO16982), in women with HER2‑positive MBC indicates that individual and mean trastuzumab trough serum concentrations varied within and across studies but there was no clear effect of the concomitant administration of paclitaxel on the pharmacokinetics of trastuzumab. Comparison of trastuzumab PK data from study M77004 in which women with HER2‑positive MBC were treated concomitantly with trastuzumab, paclitaxel and doxorubicin to trastuzumab PK data in studies where trastuzumab was administered as monotherapy (H0649g) or in combination with anthracycline plus cyclophosphamide or paclitaxel (study H0648g), suggested no effect of doxorubicin and paclitaxel on the pharmacokinetics of trastuzumab.

 

Pharmacokinetic data from study H4613g/GO01305 suggested that carboplatin had no impact on the PK of trastuzumab.

 

The administration of concomitant anastrozole did not appear to influence the pharmacokinetics of trastuzumab.

 

 


Women of childbearing potential

 

Women of childbearing potential should be advised to use effective contraception during treatment with KANJINTI and for 7 months after treatment has concluded (see section 5.2).

 

Pregnancy

 

Reproduction studies have been conducted in cynomolgus monkeys at doses up to 25 times that of the weekly human maintenance dose of 2 mg/kg trastuzumab intravenous formulation and have revealed no evidence of impaired fertility or harm to the foetus. Placental transfer of trastuzumab during the early (days 20‑50 of gestation) and late (days 120‑150 of gestation) foetal development period was observed. It is not known whether trastuzumab can affect reproductive capacity. As animal reproduction studies are not always predictive of human response, KANJINTI should be avoided during pregnancy unless the potential benefit for the mother outweighs the potential risk to the foetus.

 

In the post‑marketing setting, cases of foetal renal growth and/or function impairment in association with oligohydramnios, some associated with fatal pulmonary hypoplasia of the foetus, have been reported in pregnant women receiving trastuzumab. Women who become pregnant should be advised of the possibility of harm to the foetus. If a pregnant woman is treated with KANJINTI, or if a patient becomes pregnant while receiving KANJINTI or within 7 months following the last dose of KANJINTI, close monitoring by a multidisciplinary team is desirable.

 

Breast‑feeding

 

A study conducted in cynomolgus monkeys at doses 25 times that of the weekly human maintenance dose of 2 mg/kg trastuzumab intravenous formulation from days 120 to 150 of pregnancy demonstrated that trastuzumab is secreted in the milk postpartum. The exposure to trastuzumab in utero and the presence of trastuzumab in the serum of infant monkeys was not associated with any adverse effects on their growth or development from birth to 1 month of age. It is not known whether trastuzumab is secreted in human milk. As human IgG1 is secreted into human milk, and the potential for harm to the infant is unknown, women should not breast-feed during KANJINTI therapy and for 7 months after the last dose.

 

Fertility

 

There is no fertility data available.


Trastuzumab has a minor influence on the ability to drive or use machines (see section 4.8). Dizziness and somnolence may occur during treatment with KANJINTI (see section 4.8). Patients experiencing infusion‑related symptoms (see section 4.4) should be advised not to drive and use machines until symptoms abate.


Summary of the safety profile

 

Amongst the most serious and/or common adverse reactions reported in trastuzumab usage to date are cardiac dysfunction, infusion‑related reactions, haematotoxicity (in particular neutropenia), infections and pulmonary adverse reactions.

 

Tabulated list of adverse reactions

 

In this section, the following categories of frequency have been used: 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), not known (cannot be estimated from the available data). Within each frequency grouping, adverse reactions are presented in order of decreasing seriousness.

 

Presented in table 1 are adverse reactions that have been reported in association with the use of intravenous trastuzumab alone or in combination with chemotherapy in pivotal clinical trials and in the post‑marketing setting.

 

All the terms included are based on the highest percentage seen in pivotal clinical trials. In addition, terms reported in the post-marketing setting are included in table 1.

 

Table 1 Undesirable effects reported with intravenous trastuzumab monotherapy or in combination with chemotherapy in pivotal clinical trials (N = 8,386) and in post‑marketing

 

System organ class

Adverse reaction

Frequency

Infections and infestations

Infection

Very common

Nasopharyngitis

Very common

Neutropenic sepsis

Common

Cystitis

Common

Influenza

Common

Sinusitis

Common

Skin infection

Common

Rhinitis

Common

Upper respiratory tract infection

Common

Urinary tract infection

Common

Pharyngitis

Common

Neoplasms benign, malignant and unspecified (incl. Cysts and polyps)

Malignant neoplasm progression

Not known

Neoplasm progression

Not known

Blood and lymphatic system disorders

Febrile neutropenia

Very common

Anaemia

Very common

Neutropenia

Very common

White blood cell count decreased/leukopenia

Very common

Thrombocytopenia

Very common

Hypoprothrombinaemia

Not known

Immune thrombocytopenia

Not known

Immune system disorders

Hypersensitivity

Common

+Anaphylactic reaction

Rare

+Anaphylactic shock

Rare

Metabolism and nutrition disorders

Weight decreased/Weight loss

Very common

Anorexia

Very common

Tumour lysis syndrome

Not known

Hyperkalaemia

Not known

Psychiatric disorders

Insomnia

Very common

Anxiety

Common

Depression

Common

Nervous system disorders

1Tremor

Very common

Dizziness

Very common

Headache

Very common

Paraesthesia

Very common

Dysgeusia

Very common

Peripheral neuropathy

Common

Hypertonia

Common

Somnolence

Common

Eye disorders

Conjunctivitis

Very common

Lacrimation increased

Very common

Dry eye

Common

Papilloedema

Not known

Retinal haemorrhage

Not known

Ear and labyrinth disorders

Deafness

Uncommon

Cardiac disorders

1Blood pressure decreased

Very common

1Blood pressure increased

Very common

1Heart beat irregular

Very common

1Cardiac flutter

Very common

Ejection fraction decreased*

Very common

+Cardiac failure (congestive)

Common

+1Supraventricular tachyarrhythmia

Common

Cardiomyopathy

Common

1Palpitation

Common

Pericardial effusion

Uncommon

Cardiogenic shock

Not known

Gallop rhythm present

Not known

Vascular disorders

Hot flush

Very common

+1Hypotension

Common

Vasodilatation

Common

Respiratory, thoracic and mediastinal disorders

+Dyspnoea

Very common

Cough

Very common

Epistaxis

Very common

Rhinorrhoea

Very common

+Pneumonia

Common

Asthma

Common

Lung disorder

Common

+Pleural effusion

Common

+1Wheezing

Uncommon

Pneumonitis

Uncommon

+Pulmonary fibrosis

Not known

+Respiratory distress

Not known

+Respiratory failure

Not known

+Lung infiltration

Not known

+Acute pulmonary oedema

Not known

+Acute respiratory distress syndrome

Not known

+Bronchospasm

Not known

+Hypoxia

Not known

+Oxygen saturation decreased

Not known

Laryngeal oedema

Not known

Orthopnoea

Not known

Pulmonary oedema

Not known

Interstitial lung disease

Not known

Gastrointestinal disorders

Diarrhoea

Very common

Vomiting

Very common

Nausea

Very common

1Lip swelling

Very common

Abdominal pain

Very common

Dyspepsia

Very common

Constipation

Very common

Stomatitis

Very common

Haemorrhoids

Common

Dry mouth

Common

Hepatobiliary disorders

Hepatocellular injury

Common

Hepatitis

Common

Liver tenderness

Common

Jaundice

Rare

Skin and subcutaneous tissue disorders

Erythema

Very common

Rash

Very common

1Swelling face

Very common

Alopecia

Very common

Nail disorder

Very common

Palmar-plantar erythrodysaesthesia syndrome

Very common

Acne

Common

Dry skin

Common

Ecchymosis

Common

Hyperhydrosis

Common

Maculopapular rash

Common

Pruritus

Common

Onychoclasis

Common

Dermatitis

Common

Urticaria

Uncommon

Angioedema

Not known

Musculoskeletal and connective tissue disorders

Arthralgia

Very common

1Muscle tightness

Very common

Myalgia

Very common

Arthritis

Common

Back pain

Common

Bone pain

Common

Muscle spasms

Common

Neck pain

Common

Pain in extremity

Common

Renal and urinary disorders

Renal disorder

Common

Glomerulonephritis membranous

Not known

Glomerulonephropathy

Not known

Renal failure

Not known

Pregnancy, puerperium and perinatal conditions

Oligohydramnios

Not known

Renal hypoplasia

Not known

Pulmonary hypoplasia

Not known

Reproductive system and breast disorders

Breast inflammation/mastitis

Common

General disorders and administration site conditions

Asthenia

Very common

Chest pain

Very common

Chills

Very common

Fatigue

Very common

Influenza-like symptoms

Very common

Infusion-related reaction

Very common

Pain

Very common

Pyrexia

Very common

Mucosal inflammation

Very common

Peripheral oedema

Very common

Malaise

Common

Oedema

Common

Injury, poisoning and procedural complications

Contusion

Common

+ Denotes adverse reactions that have been reported in association with a fatal outcome.

1 Denotes adverse reactions that are reported largely in association with Infusion-related reactions. Specific percentages for these are not available.

* Observed with combination therapy following anthracyclines and combined with taxanes.

 

Description of selected adverse reactions

 

Cardiac dysfunction

 

Congestive heart failure (NYHA class II–IV) is a common adverse reaction associated with the use of trastuzumab and has been associated with a fatal outcome (see section 4.4). Signs and symptoms of cardiac dysfunction such as dyspnoea, orthopnoea, increased cough, pulmonary oedema, S3 gallop, or reduced ventricular ejection fraction, have been observed in patients treated with trastuzumab (see section 4.4).

 

In 3 pivotal clinical trials of adjuvant trastuzumab given in combination with chemotherapy, the incidence of grade 3/4 cardiac dysfunction (specifically symptomatic congestive heart failure) was similar in patients who were administered chemotherapy alone (i.e. did not receive trastuzumab) and in patients who were administered trastuzumab sequentially to a taxane (0.3‑0.4%). The rate was highest in patients who were administered trastuzumab concurrently with a taxane (2.0%). In the neoadjuvant setting, the experience of concurrent administration of trastuzumab and low‑dose anthracycline regimen is limited (see section 4.4).

 

When trastuzumab was administered after completion of adjuvant chemotherapy NYHA class III‑IV heart failure was observed in 0.6% of patients in the one year arm after a median follow-up of 12 months. In study BO16348, after a median follow‑up of 8 years the incidence of severe CHF (NYHA class III & IV) in the trastuzumab 1 year treatment arm was 0.8%, and the rate of mild symptomatic and asymptomatic left ventricular dysfunction was 4.6%.

 

Reversibility of severe CHF (defined as a sequence of at least two consecutive LVEF values ≥ 50% after the event) was evident for 71.4% of trastuzumab-treated patients. Reversibility of mild symptomatic and asymptomatic left ventricular dysfunction was demonstrated for 79.5% of patients. Approximately 17% of cardiac dysfunction related events occurred after completion of trastuzumab.

 

In the pivotal metastatic trials of intravenous trastuzumab, the incidence of cardiac dysfunction varied between 9% and 12% when it was combined with paclitaxel compared with 1%‑4% for paclitaxel alone. For monotherapy, the rate was 6%‑9%. The highest rate of cardiac dysfunction was seen in patients receiving trastuzumab concurrently with anthracycline/cyclophosphamide (27%), and was significantly higher than for anthracycline/cyclophosphamide alone (7%‑10%). In a subsequent trial with prospective monitoring of cardiac function, the incidence of symptomatic CHF was 2.2% in patients receiving trastuzumab and docetaxel, compared with 0% in patients receiving docetaxel alone. Most of the patients (79%) who developed cardiac dysfunction in these trials experienced an improvement after receiving standard treatment for CHF.

 

Infusion reactions, allergic-like reactions and hypersensitivity

 

It is estimated that approximately 40% of patients who are treated with trastuzumab will experience some form of infusion-related reaction. However, the majority of infusion‑related reactions are mild to moderate in intensity (NCI‑CTC grading system) and tend to occur earlier in treatment, i.e. during infusions one, two and three and lessen in frequency in subsequent infusions. Reactions include chills, fever, dyspnoea, hypotension, wheezing, bronchospasm, tachycardia, reduced oxygen saturation, respiratory distress, rash, nausea, vomiting and headache (see section 4.4). The rate of infusion‑related reactions of all grades varied between studies depending on the indication, the data collection methodology, and whether trastuzumab was given concurrently with chemotherapy or as monotherapy.

 

Severe anaphylactic reactions requiring immediate additional intervention can occur usually during either the first or second infusion of trastuzumab (see section 4.4) and have been associated with a fatal outcome. Anaphylactoid reactions have been observed in isolated cases.

 

Haematotoxicity

 

Febrile neutropenia, leukopenia, anaemia, thrombocytopenia and neutropenia occurred very commonly. The frequency of occurrence of hypoprothrombinaemia is not known. The risk of neutropenia may be slightly increased when trastuzumab is administered with docetaxel following anthracycline therapy.

 

Pulmonary events

 

Severe pulmonary adverse reactions occur in association with the use of trastuzumab and have been associated with a fatal outcome. These include, but are not limited to, pulmonary infiltrates, acute respiratory distress syndrome, pneumonia, pneumonitis, pleural effusion, respiratory distress, acute pulmonary oedema and respiratory insufficiency (see section 4.4).

 

Details of risk minimisation measures that are consistent with the EU Risk Management Plan are presented in (see section 4.4) warnings and precautions.

 

Immunogenicity

 

In the neoadjuvant-adjuvant EBC study (BO22227), at a median follow-up exceeding 70 months, 10.1% (30/296) of patients treated with trastuzumab intravenous developed antibodies against trastuzumab. Neutralising anti-trastuzumab antibodies were detected in post-baseline samples in 2 of 30 patients in the trastuzumab intravenous arm.

 

The clinical relevance of these antibodies is not known. The presence of anti-trastuzumab antibodies had no impact on pharmacokinetics, efficacy (determined by pathological complete response [pCR] and event free survival [EFS]) and safety determined by occurrence of administration related reactions (ARRs) of trastuzumab intravenous.

 

There are no immunogenicity data available for trastuzumab in gastric cancer.

 

Reporting of suspected adverse reactions

 

Reporting suspected adverse reactions after authorisation of the medicinal product is important. It allows continued monitoring of the benefit-risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions to their local representative.

To report any side effects:

Saudi Arabia:

 

·       The National Pharmacovigilance Centre (NPC):

 

-          SFDA Call Center: 19999

-          E-mail: npc.drug@sfda.gov.sa

-          Website: https://ade.sfda.gov.sa/

 


 

There is no experience with overdose in human clinical trials. Single doses of trastuzumab alone greater than 10 mg/kg have not been administered in the clinical trials; a maintenance dose of 10 mg/kg q3w following a loading dose of 8 mg/kg has been studied in a clinical trial with metastatic gastric cancer patients. Doses up to this level were well tolerated.

 


Pharmacotherapeutic group: Antineoplastic agents, monoclonal antibodies, ATC code: L01XC03

 

KANJINTI is a biosimilar medicinal product.

 

Trastuzumab is a recombinant humanised IgG1 monoclonal antibody against the human epidermal growth factor receptor 2 (HER2). Overexpression of HER2 is observed in 20%‑30% of primary breast cancers. Studies of HER2‑positivity rates in gastric cancer (GC) using immunohistochemistry (IHC) and fluorescence in situ hybridisation (FISH) or chromogenic in situ hybridisation (CISH) have shown that there is a broad variation of HER2‑positivity ranging from 6.8% to 34.0% for IHC and 7.1% to 42.6% for FISH. Studies indicate that breast cancer patients whose tumours overexpress HER2 have a shortened disease‑free survival compared to patients whose tumours do not overexpress HER2. The extracellular domain of the receptor (ECD, p105) can be shed into the blood stream and measured in serum samples.

 

Mechanism of action

 

Trastuzumab binds with high affinity and specificity to sub‑domain IV, a juxta-membrane region of HER2’s extracellular domain. Binding of trastuzumab to HER2 inhibits ligand‑independent HER2 signalling and prevents the proteolytic cleavage of its extracellular domain, an activation mechanism of HER2. As a result, trastuzumab has been shown, in both in vitro assays and in animals, to inhibit the proliferation of human tumour cells that overexpress HER2. Additionally, trastuzumab is a potent mediator of antibody‑dependent cell‑mediated cytotoxicity (ADCC). In vitro, trastuzumab-mediated ADCC has been shown to be preferentially exerted on HER2 overexpressing cancer cells compared with cancer cells that do not overexpress HER2.

 

Detection of HER2 overexpression or HER2 gene amplification

 

Detection of HER2 overexpression or HER2 gene amplification in breast cancer

KANJINTI should only be used in patients whose tumours have HER2 overexpression or HER2 gene amplification as determined by an accurate and validated assay. HER2 overexpression should be detected using an immunohistochemistry (IHC)-based assessment of fixed tumour blocks (see section 4.4). HER2 gene amplification should be detected using fluorescence in situ hybridisation (FISH) or chromogenic in situ hybridisation (CISH) of fixed tumour blocks. Patients are eligible for KANJINTI treatment if they show strong HER2 overexpression as described by a 3+ score by IHC or a positive FISH or CISH result.

To ensure accurate and reproducible results, the testing must be performed in a specialised laboratory, which can ensure validation of the testing procedures.

 

The recommended scoring system to evaluate the IHC staining patterns is as stated in table 2:

 

Table 2 Recommended scoring system to evaluate the IHC staining patterns in breast cancer

 

Score

Staining pattern

HER2 overexpression assessment

0

No staining is observed or membrane staining is observed in < 10% of the tumour cells.

Negative

1+

A faint/barely perceptible membrane staining is detected in > 10% of the tumour cells. The cells are only stained in part of their membrane.

Negative

2+

A weak to moderate complete membrane staining is detected in > 10% of the tumour cells.

Equivocal

3+

Strong complete membrane staining is detected in > 10% of the tumour cells.

Positive

 

In general, FISH is considered positive if the ratio of the HER2 gene copy number per tumour cell to the chromosome 17 copy number is greater than or equal to 2, or if there are more than 4 copies of the HER2 gene per tumour cell if no chromosome 17 control is used.

 

In general, CISH is considered positive if there are more than 5 copies of the HER2 gene per nucleus in greater than 50% of tumour cells.

 

For full instructions on assay performance and interpretation please refer to the package inserts of validated FISH and CISH assays. Official recommendations on HER2 testing may also apply.

 

For any other method that may be used for the assessment of HER2 protein or gene expression, the analyses should only be performed by laboratories that provide adequate state‑of‑the‑art performance of validated methods. Such methods must clearly be precise and accurate enough to demonstrate overexpression of HER2 and must be able to distinguish between moderate (congruent with 2+) and strong (congruent with 3+) overexpression of HER2.

 

Detection of HER2 over expression or HER2 gene amplification in gastric cancer

Only an accurate and validated assay should be used to detect HER2 over expression or HER2 gene amplification. IHC is recommended as the first testing modality and in cases where HER2 gene amplification status is also required, either a silver‑enhanced in situ hybridisation (SISH) or a FISH technique must be applied. SISH technology is however, recommended to allow for the parallel evaluation of tumour histology and morphology. To ensure validation of testing procedures and the generation of accurate and reproducible results, HER2 testing must be performed in a laboratory staffed by trained personnel. Full instructions on assay performance and results interpretation should be taken from the product information leaflet provided with the HER2 testing assays used.

 

In the ToGA (BO18255) trial, patients whose tumours were either IHC3+ or FISH positive were defined as HER2-positive and thus included in the trial. Based on the clinical trial results, the beneficial effects were limited to patients with the highest level of HER2 protein overexpression, defined by a 3+ score by IHC, or a 2+ score by IHC and a positive FISH result.

 

In a method comparison study (study D008548) a high degree of concordance (> 95%) was observed for SISH and FISH techniques for the detection of HER2 gene amplification in gastric cancer patients.

 

HER2 over expression should be detected using an immunohistochemistry (IHC)‑based assessment of fixed tumour blocks; HER2 gene amplification should be detected using in situ hybridisation using either SISH or FISH on fixed tumour blocks.

 

The recommended scoring system to evaluate the IHC staining patterns is as stated in table 3:

 

Table 3 Recommended scoring system to evaluate the IHC staining patterns in gastric cancer

 

Score

Surgical specimen-staining pattern

Biopsy specimen-staining pattern

HER2 overexpression assessment

0

No reactivity or membranous reactivity in < 10% of tumour cells

No reactivity or membranous reactivity in any tumour cell

Negative

1+

Faint/barely perceptible membranous reactivity in ≥ 10% of tumour cells; cells are reactive only in part of their membrane

Tumour cell cluster with a faint/barely perceptible membranous reactivity irrespective of percentage of tumour cells stained

Negative

2+

Weak to moderate complete, basolateral or lateral membranous reactivity in ≥ 10% of tumour cells

Tumour cell cluster with a weak to moderate complete, basolateral or lateral membranous reactivity irrespective of percentage of tumour cells stained

Equivocal

3+

Strong complete, basolateral or lateral membranous reactivity in ≥ 10% of tumour cells

Tumour cell cluster with a strong complete, basolateral or lateral membranous reactivity irrespective of percentage of tumour cells stained

Positive

 

In general, SISH or FISH is considered positive if the ratio of the HER2 gene copy number per tumour cell to the chromosome 17 copy number is greater than or equal to 2.

 

Clinical efficacy and safety

 

Metastatic breast cancer

 

Trastuzumab has been used in clinical trials as monotherapy for patients with MBC who have tumours that overexpress HER2 and who have failed one or more chemotherapy regimens for their metastatic disease (trastuzumab alone).

 

Trastuzumab has also been used in combination with paclitaxel or docetaxel for the treatment of patients who have not received chemotherapy for their metastatic disease. Patients who had previously received anthracycline‑based adjuvant chemotherapy were treated with paclitaxel (175 mg/m2 infused over 3 hours) with or without trastuzumab. In the pivotal trial of docetaxel (100 mg/m2 infused over 1 hour) with or without trastuzumab, 60% of the patients had received prior anthracycline-based adjuvant chemotherapy. Patients were treated with trastuzumab until progression of disease.

 

The efficacy of trastuzumab in combination with paclitaxel in patients who did not receive prior adjuvant anthracyclines has not been studied. However, trastuzumab plus docetaxel was efficacious in patients whether or not they had received prior adjuvant anthracyclines.

 

The test method for HER2 overexpression used to determine eligibility of patients in the pivotal trastuzumab monotherapy and trastuzumab plus paclitaxel clinical trials employed immunohistochemical staining for HER2 of fixed material from breast tumours using the murine monoclonal antibodies CB11 and 4D5. These tissues were fixed in formalin or Bouin’s fixative. This investigative clinical trial assay performed in a central laboratory utilised a 0 to 3+ scale. Patients classified as staining 2+ or 3+ were included, while those staining 0 or 1+ were excluded. Greater than 70% of patients enrolled exhibited 3+ overexpression. The data suggest that beneficial effects were greater among those patients with higher levels of overexpression of HER2 (3+).

 

The main test method used to determine HER2 positivity in the pivotal trial of docetaxel, with or without trastuzumab, was immunohistochemistry. A minority of patients was tested using fluorescence in situ hybridisation (FISH). In this trial, 87% of patients entered had disease that was IHC3+, and 95% of patients entered had disease that was IHC3+ and/or FISH‑positive.

 

Weekly dosing in metastatic breast cancer

The efficacy results from the monotherapy and combination therapy studies are summarised in table 4:

 

Table 4 Efficacy results from the monotherapy and combination therapy studies

 

Parameter

Monotherapy

Combination therapy

 

Trastuzumab1

N = 172

Trastuzumab

plus

paclitaxel2

N = 68

Paclitaxel2

N = 77

Trastuzumab plus docetaxel3 N = 92

Docetaxel3

N = 94

Response rate (95% CI)

18%

(13‑25)

49%

(36‑61)

17%

(9‑27)

61%

(50-71)

34%

(25-45)

Median duration of response (months)

(95% CI)

9.1

(5.6-10.3)

8.3

(7.3-8.8)

4.6

(3.7-7.4)

11.7

(9.3-15.0)

5.7

(4.6-7.6)

Median TTP (months) (95% CI)

3.2

(2.6-3.5)

7.1

(6.2-12.0)

3.0

(2.0-4.4)

11.7

(9.2-13.5)

6.1

(5.4-7.2)

Median survival (months) (95% CI)

16.4

(12.3-ne)

24.8

(18.6-33.7)

17.9

(11.2-23.8)

31.2

(27.3-40.8)

22.74

(19.1-30.8)

CI = confidence interval, TTP = time to progression; "ne" indicates that it could not be estimated or it was not yet reached.

1. Study H0649g: IHC3+ patient subset

2. Study H0648g: IHC3+ patient subset

3. Study M77001: Full analysis set (intent-to-treat), 24 months results

 

Combination treatment with trastuzumab and anastrozole

Trastuzumab has been studied in combination with anastrozole for first line treatment of MBC in HER2 overexpressing, hormone‑receptor (i.e. estrogen‑receptor (ER) and/or progesterone-receptor (PR)) positive postmenopausal patients. Progression free survival was doubled in the trastuzumab plus anastrozole arm compared to anastrozole (4.8 months versus 2.4 months). For the other parameters the improvements seen for the combination were for overall response (16.5% versus 6.7%); clinical benefit rate (42.7% versus 27.9%); time to progression (4.8 months versus 2.4 months). For time to response and duration of response no difference could be recorded between the arms. The median overall survival (OS) was extended by 4.6 months for patients in the combination arm. The difference was not statistically significant, however more than half of the patients in the anastrozole alone arm crossed over to a trastuzumab containing regimen after progression of disease.

 

Three‑weekly dosing in metastatic breast cancer

The efficacy results from the non‑comparative monotherapy and combination therapy studies are summarised in table 5:

 

Table 5 Efficacy results from the non-comparative monotherapy and combination therapy studies

 

Parameter

Monotherapy

Combination therapy

 

Trastuzumab1

N = 105

Trastuzumab2

N = 72

Trastuzumab plus paclitaxel3 N = 32

Trastuzumab plus docetaxel4

N = 110

Response rate

(95% CI)

24%

(15-35)

27%

(14-43)

59%

(41-76)

73%

(63-81)

Median duration of response (months) (range)

10.1

(2.8-35.6)

7.9

(2.1-18.8)

10.5

(1.8-21)

13.4

(2.1-55.1)

Median TTP (months) (95% CI)

3.4

(2.8-4.1)

7.7

(4.2-8.3)

12.2

(6.2-ne)

13.6

(11-16)

Median survival (months) (95% CI)

ne

ne

ne

47.3

(32-ne)

CI = confidence interval; TTP = time to progression; "ne" indicates that it could not be estimated or it was not yet reached.

1. Study WO16229: loading dose 8 mg/kg, followed by 6 mg/kg 3-weekly schedule

2. Study MO16982: loading dose 6 mg/kg weekly × 3; followed by 6 mg/kg 3‑weekly schedule

3. Study BO15935

4. Study MO16419

 

Sites of progression

The frequency of progression in the liver was significantly reduced in patients treated with the combination of trastuzumab and paclitaxel, compared to paclitaxel alone (21.8% versus 45.7%; p = 0.004). More patients treated with trastuzumab and paclitaxel progressed in the central nervous system than those treated with paclitaxel alone (12.6% versus 6.5%; p = 0.377).

 

Early breast cancer (adjuvant setting)

 

Early breast cancer is defined as non‑metastatic primary invasive carcinoma of the breast.

 

In the adjuvant treatment setting, trastuzumab was investigated in 4 large multicentre, randomised, trials.

-             Study BO16348 was designed to compare one and two years of three‑weekly trastuzumab treatment versus observation in patients with HER2-positive EBC following surgery, established chemotherapy and radiotherapy (if applicable). In addition, comparison of two years of trastuzumab treatment versus one year of trastuzumab treatment was performed. Patients assigned to receive trastuzumab were given an initial loading dose of 8 mg/kg, followed by 6 mg/kg every three weeks for either one or two years.

-             The NSABP B‑31 and NCCTG N9831 studies that comprise the joint analysis (JA) were designed to investigate the clinical utility of combining trastuzumab treatment with paclitaxel following AC chemotherapy, additionally the NCCTG N9831 study also investigated adding trastuzumab sequentially to AC→P chemotherapy in patients with HER2-positive EBC following surgery.

-             The BCIRG 006 study was designed to investigate combining trastuzumab treatment with docetaxel either following AC chemotherapy or in combination with docetaxel and carboplatin in patients with HER2-positive EBC following surgery.

 

Early breast cancer in the HERA trial was limited to operable, primary, invasive adenocarcinoma of the breast, with axillary nodes positive or axillary nodes negative if tumours at least 1 cm in diameter.

 

In the joint analysis of the NSABP B‑31 and NCCTG N9831 studies, EBC was limited to women with operable breast cancer at high risk, defined as HER2‑positive and axillary lymph node positive or HER2-positive and lymph node negative with high risk features (tumour size > 1 cm and ER negative or tumour size > 2 cm, regardless of hormonal status).

 

In the BCIRG 006 study HER2-positive, EBC was defined as either lymph node positive or high risk node negative patients with no (pN0) lymph node involvement, and at least 1 of the following factors: tumour size greater than 2 cm, estrogen-receptor and progesterone-receptor negative, histological and/or nuclear grade 2‑3, or age < 35 years).

 

The efficacy results from the BO16348 trial following 12 months* and 8 years** median follow-up are summarised in table 6:

 

Table 6 Efficacy results from study BO16348

 

Median follow-up

12 months*

Median follow-up

8 years**

Parameter

Observation

N = 1,693

Trastuzumab

1 Year

N = 1,693

Observation

N = 1,697***

Trastuzumab

1 Year

N = 1,702***

Disease-free survival

- No. patients with event

- No. patients without event

p‑value versus Observation

Hazard ratio versus Observation

 

219 (12.9%)

1,474 (87.1%)

 

127 (7.5%)

1,566 (92.5%)

 

570 (33.6%)

1,127 (66.4%)

 

471 (27.7%)

1,231 (72.3%)

< 0.0001

0.54

< 0.0001

0.76

Recurrence-free survival

- No. patients with event

- No. patients without event

p‑value versus Observation

Hazard ratio versus Observation

 

208 (12.3%)

1,485 (87.7%)

 

113 (6.7%)

1,580 (93.3%)

 

506 (29.8%)

1,191 (70.2%)

 

399 (23.4%)

1,303 (76.6%)

< 0.0001

0.51

< 0.0001

0.73

Distant disease-free survival

- No. patients with event

- No. patients without event

p‑value versus Observation

Hazard ratio versus Observation

 

184 (10.9%)

1,508 (89.1%)

 

99 (5.8%)

1,594 (94.6%)

 

488 (28.8%)

1,209 (71.2%)

 

399 (23.4%)

1,303 (76.6%)

< 0.0001

0.50

< 0.0001

0.76

Overall survival (death)

- No. patients with event

- No. patients without event

p‑value versus Observation

Hazard ratio versus Observation

 

40 (2.4%)

1,653 (97.6%)

 

31 (1.8%)

1,662 (98.2%)

 

350 (20.6%)

1,347 (79.4%)

 

278 (16.3%)

1,424 (83.7%)

0.24

0.75

0.0005

0.76

*Co‑primary endpoint of DFS of 1 year versus observation met the pre‑defined statistical boundary

**Final analysis (including cross-over of 52% of patients from the observation arm to trastuzumab)

***There is a discrepancy in the overall sample size due to a small number of patients who were randomised after the cut‑off date for the 12‑month median follow-up analysis

 

The efficacy results from the interim efficacy analysis crossed the protocol pre-specified statistical boundary for the comparison of 1‑year of trastuzumab versus observation. After a median follow-up of 12 months, the hazard ratio (HR) for disease-free survival (DFS) was 0.54 (95% CI: 0.44, 0.67) which translates into an absolute benefit, in terms of a 2‑year disease-free survival rate, of 7.6 percentage points (85.8% versus 78.2%) in favour of the trastuzumab arm.

 

A final analysis was performed after a median follow‑up of 8 years, which showed that 1 year trastuzumab treatment is associated with a 24% risk reduction compared to observation only (HR = 0.76, 95% CI: 0.67, 0.86). This translates into an absolute benefit in terms of an 8 year disease‑free survival rate of 6.4 percentage points in favour of 1 year trastuzumab treatment.

 

In this final analysis, extending trastuzumab treatment for a duration of two years did not show additional benefit over treatment for 1 year [DFS HR in the intent to treat (ITT) population of 2 years versus 1 year = 0.99 (95% CI: 0.87, 1.13), p‑value = 0.90 and OS HR = 0.98 (0.83, 1.15); p‑value = 0.78]. The rate of asymptomatic cardiac dysfunction was increased in the 2‑year treatment arm (8.1% versus 4.6% in the 1‑year treatment arm). More patients experienced at least one grade 3 or 4 adverse event in the 2‑year treatment arm (20.4%) compared with the 1‑year treatment arm (16.3%).

 

In the NSABP B‑31 and NCCTG N9831 studies trastuzumab was administered in combination with paclitaxel, following AC chemotherapy.

Doxorubicin and cyclophosphamide were administered concurrently as follows:

 

-             intravenous push doxorubicin, at 60 mg/m2, given every 3 weeks for 4 cycles.

 

-             intravenous cyclophosphamide, at 600 mg/m2 over 30 minutes, given every 3 weeks for 4 cycles.

 

Paclitaxel, in combination with trastuzumab, was administered as follows:

 

-             intravenous paclitaxel – 80 mg/m2 as a continuous intravenous infusion, given every week for 12 weeks.

or

-             intravenous paclitaxel – 175 mg/m2 as a continuous intravenous infusion, given every 3 weeks for 4 cycles (day 1 of each cycle).

 

The efficacy results from the joint analysis of the NSABP B‑31 and NCCTG N9831 trials at the time of the definitive analysis of DFS* are summarised in table 7. The median duration of follow-up was 1.8 years for the patients in the AC→P arm and 2.0 years for patients in the AC→PH arm.

 

Table 7 Summary of efficacy results from the joint analysis of the NSABP B-31 and NCCTG N9831 trials at the time of the definitive DFS analysis*

 

Parameter

ACP

(n = 1,679)

ACPH

(n = 1,672)

Hazard Ratio vs

ACP

(95% CI)

p-value

Disease-free survival

No. patients with event (%)

 

Distant recurrence

No. patients with event

 

Death (OS event)

No. patients with event

 

261 (15.5)

 

 

193 (11.5)

 

 

92 (5.5)

 

133 (8.0)

 

 

96 (5.7)

 

 

62 (3.7)

 

0.48 (0.39, 0.59)

p < 0.0001

 

0.47 (0.37, 0.60)

p < 0.0001

 

0.67 (0.48, 0.92)

p = 0.014**

A: doxorubicin; C: cyclophosphamide; P: paclitaxel; H: trastuzumab

* At median duration of follow-up of 1.8 years for the patients in the AC→P arm and 2.0 years for patients in the AC→PH arm

** p-value for OS did not cross the pre‑specified statistical boundary for comparison of AC→PH vs AC→P

 

For the primary endpoint, DFS, the addition of trastuzumab to paclitaxel chemotherapy resulted in a 52% decrease in the risk of disease recurrence. The hazard ratio translates into an absolute benefit, in terms of 3‑year disease‑free survival rate estimates of 11.8 percentage points (87.2% versus 75.4%) in favour of the AC→PH (trastuzumab) arm.

 

At the time of a safety update after a median of 3.5‑3.8 years follow-up, an analysis of DFS reconfirms the magnitude of the benefit shown in the definitive analysis of DFS. Despite the cross-over to trastuzumab in the control arm, the addition of trastuzumab to paclitaxel chemotherapy resulted in a 52% decrease in the risk of disease recurrence. The addition of trastuzumab to paclitaxel chemotherapy also resulted in a 37% decrease in the risk of death.

 

The pre‑planned final analysis of OS from the joint analysis of studies NSABP B-31 and NCCTG N9831 was performed when 707 deaths had occurred (median follow-up 8.3 years in the AC→PH group). Treatment with AC→PH resulted in a statistically significant improvement in OS compared with AC→P (stratified HR = 0.64; 95% CI [0.55, 0.74]; log-rank p-value < 0.0001). At 8 years, the survival rate was estimated to be 86.9% in the AC→PH arm and 79.4% in the AC→P arm, an absolute benefit of 7.4% (95% CI: 4.9%, 10.0%).

The final OS results from the joint analysis of studies NSABP B‑31 and NCCTG N9831 are summarised in table 8:

 

Table 8 Final overall survival analysis from the joint analysis of trials NSABP B-31 and NCCTG N9831

 

Parameter

ACP

(N = 2,032)

ACPH

(N = 2,031)

p‑value versus

ACP

Hazard Ratio

versus

ACP

(95% CI)

Death (OS event):

No. patients with event (%)

 

418 (20.6%)

 

289 (14.2%)

 

< 0.0001

 

0.64

(0.55, 0.74)

CI = confidence interval; OS = overall survival; A: doxorubicin; C: cyclophosphamide; P: paclitaxel; H: trastuzumab

 

DFS analysis was also performed at the final analysis of OS from the joint analysis of studies NSABP B-31 and NCCTG N9831. The updated DFS analysis results (stratified HR = 0.61; 95% CI [0.54, 0.69]) showed a similar DFS benefit compared to the definitive primary DFS analysis, despite 24.8% patients in the AC→P arm who crossed over to receive trastuzumab. At 8 years, the disease‑free survival rate was estimated to be 77.2% (95% CI: 75.4, 79.1) in the AC→PH arm, an absolute benefit of 11.8% compared with the AC→P arm.

 

In the BCIRG 006 study trastuzumab was administered either in combination with docetaxel, following AC chemotherapy (AC→DH) or in combination with docetaxel and carboplatin (DCarbH).

 

Docetaxel was administered as follows:

 

-             intravenous docetaxel – 100 mg/m2 as an intravenous infusion over 1 hour, given every 3 weeks for 4 cycles (day 2 of first docetaxel cycle, then day 1 of each subsequent cycle)

or

-             intravenous docetaxel – 75 mg/m2 as an intravenous infusion over 1 hour, given every 3 weeks for 6 cycles (day 2 of cycle 1, then day 1 of each subsequent cycle)

which was followed by:

-             carboplatin – at target AUC = 6 mg/mL/min administered by intravenous infusion over 30‑60 minutes repeated every 3 weeks for a total of six cycles

 

Trastuzumab was administered weekly with chemotherapy and 3-weekly thereafter for a total of 52 weeks.

 

The efficacy results from the BCIRG 006 are summarised in tables 9 and 10. The median duration of follow-up was 2.9 years in the AC→D arm and 3.0 years in each of the AC→DH and DCarbH arms.

 

Table 9 Overview of efficacy analyses BCIRG 006 ACD versus ACDH

 

Parameter

ACD

(n = 1,073)

ACDH (n = 1,074)

Hazard Ratio vs

ACD

(95% CI)
p-value

Disease-free survival

No. patients with event

 

195

 

134

 

0.61 (0.49, 0.77)

p < 0.0001

Distant recurrence

No. patients with event

 

144

 

95

 

0.59 (0.46, 0.77)

p < 0.0001

Death (OS event)

No. patients with event

 

80

 

49

 

0.58 (0.40, 0.83)

p = 0.0024

AC→D = doxorubicin plus cyclophosphamide, followed by docetaxel; AC→DH = doxorubicin plus cyclophosphamide, followed by docetaxel plus trastuzumab; CI = confidence interval

 

Table 10 Overview of efficacy analyses BCIRG 006 ACD versus DCarbH

 

Parameter

ACD

(n = 1,073)

DCarbH

(n = 1,074)

Hazard Ratio vs

ACD

(95% CI)

Disease-free survival

No. patients with event

 

195

 

145

 

0.67 (0.54, 0.83)

p = 0.0003

Distant recurrence

No. patients with event

 

144

 

103

 

0.65 (0.50, 0.84)

p = 0.0008

Death (OS event)

No. patients with event

 

80

 

56

 

0.66 (0.47, 0.93)

p = 0.0182

AC→D = doxorubicin plus cyclophosphamide, followed by docetaxel; DCarbH = docetaxel, carboplatin and trastuzumab; CI = confidence interval

 

In the BCIRG 006 study for the primary endpoint, DFS, the hazard ratio translates into an absolute benefit, in terms of 3‑year disease-free survival rate estimates of 5.8 percentage points (86.7% versus 80.9%) in favour of the AC→DH (trastuzumab) arm and 4.6 percentage points (85.5% versus 80.9%) in favour of the DCarbH (trastuzumab) arm compared to AC→D.

 

In study BCIRG 006, 213/1,075 patients in the DCarbH (TCH) arm, 221/1,074 patients in the AC→DH (AC→TH) arm, and 217/1,073 in the AC→D (AC→T) arm had a Karnofsky performance status ≤ 90 (either 80 or 90). No disease-free survival (DFS) benefit was noticed in this subgroup of patients (hazard ratio = 1.16, 95% CI [0.73, 1.83] for DCarbH (TCH) versus AC→D (AC→T); hazard ratio 0.97, 95% CI [0.60, 1.55] for AC→DH (AC→TH) versus AC→D).

In addition a post‑hoc exploratory analysis was performed on the data sets from the joint analysis (JA) NSABP B-31/NCCTG N9831* and BCIRG 006 clinical studies combining DFS events and symptomatic cardiac events and summarised in table 11:

 

Table 11 Post-hoc exploratory analysis results from the joint analysis NSABP B‑31/NCCTG N9831* and BCIRG 006 clinical studies combining DFS events and symptomatic cardiac events

 

 

ACPH

(vs. ACP) (NSABP B‑31 and NCCTG N9831)*

ACDH

(vs. ACD) (BCIRG 006)

DCarbH

(vs. ACD)

(BCIRG 006)

Primary efficacy analysis

DFS Hazard ratios

(95% CI)

p‑value

 

0.48

(0.39, 0.59)

p < 0.0001

 

0.61

(0.49, 0.77)

p < 0.0001

 

0.67

(0.54, 0.83)

p = 0.0003

Long term follow-up efficacy

analysis**

DFS Hazard ratios

(95% CI)

p-value

 

 

0.61

(0.54, 0.69)

p < 0.0001

 

 

0.72

(0.61, 0.85)

p < 0.0001

 

 

0.77

(0.65, 0.90)

p = 0.0011

Post‑hoc exploratory analysis with DFS and symptomatic cardiac events

Long term follow-up**

Hazard ratios

(95% CI)

 

 

 

 

0.67

(0.60, 0.75)

 

 

 

 

0.77

(0.66, 0.90)

 

 

 

 

0.77

(0.66, 0.90)

A: doxorubicin; C: cyclophosphamide; P: paclitaxel; D: docetaxel; Carb: carboplatin; H: trastuzumab CI = confidence interval

* At the time of the definitive analysis of DFS. Median duration of follow-up was 1.8 years in the AC→P arm and 2.0 years in the AC→PH arm

** Median duration of long term follow-up for the Joint Analysis clinical studies was 8.3 years (range: 0.1 to 12.1) for the AC→PH arm and 7.9 years (range: 0.0 to 12.2) for the AC→P arm; Median duration of long term follow-up for the BCIRG 006 study was 10.3 years in both the AC→D arm (range: 0.0 to 12.6) arm and the DCarbH arm (range: 0.0 to 13.1), and was 10.4 years (range: 0.0 to 12.7) in the AC→DH arm

 

Early breast cancer (neoadjuvant-adjuvant setting)

 

So far, no results are available which compare the efficacy of trastuzumab administered with chemotherapy in the adjuvant setting with that obtained in the neoadjuvant-adjuvant setting.

 

In the neoadjuvant‑adjuvant treatment setting, study MO16432, a multicentre randomised trial, was designed to investigate the clinical efficacy of concurrent administration of trastuzumab with neoadjuvant chemotherapy including both an anthracycline and a taxane, followed by adjuvant trastuzumab, up to a total treatment duration of 1 year. The study recruited patients with newly diagnosed locally advanced (Stage III) or inflammatory EBC. Patients with HER2+ tumours were randomised to receive either neoadjuvant chemotherapy concurrently with neoadjuvant‑adjuvant trastuzumab, or neoadjuvant chemotherapy alone.

 

In study MO16432, trastuzumab (8 mg/kg loading dose, followed by 6 mg/kg maintenance every 3 weeks) was administered concurrently with 10 cycles of neoadjuvant chemotherapy as follows:

 

-             Doxorubicin 60 mg/m2 and paclitaxel 150 mg/m2, administered 3‑weekly for 3 cycles,

 

which was followed by

-             Paclitaxel 175 mg/m2 administered 3‑weekly for 4 cycles,

 

which was followed by

-             CMF on day 1 and 8 every 4 weeks for 3 cycles,

 

which was followed after surgery by

-             additional cycles of adjuvant trastuzumab (to complete 1 year of treatment).

 

The efficacy results from study MO16432 are summarised in table 12. The median duration of follow‑up in the trastuzumab arm was 3.8 years.

 

Table 12 Efficacy results from MO16432

 

Parameter

Chemo + trastuzumab

(n = 115)

Chemo only (n = 116)

 

Event‑free survival

 

No. patients with event

 

 

46

 

 

59

Hazard Ratio

(95% CI)

0.65 (0.44, 0.96) p = 0.0275

Total pathological complete response* (95% CI)

40%

(31.0, 49.6)

20.7%

(13.7, 29.2)

p = 0.0014

Overall survival

 

No. patients with event

 

 

22

 

 

33

Hazard Ratio

(95% CI)

0.59 (0.35, 1.02) p = 0.0555

*defined as absence of any invasive cancer both in the breast and axillary nodes

 

An absolute benefit of 13 percentage points in favour of the trastuzumab arm was estimated in terms of 3 year event-free survival rate (65% versus 52%).

 

Metastatic gastric cancer

 

Trastuzumab has been investigated in one randomised, open‑label Phase III trial ToGA (BO18255) in combination with chemotherapy versus chemotherapy alone.

 

Chemotherapy was administered as follows:

 

-             capecitabine – 1,000 mg/m2 orally twice daily for 14 days every 3 weeks for 6 cycles (evening of day 1 to morning of day 15 of each cycle)

or

-             intravenous 5‑fluorouracil-800 mg/m2/day as a continuous intravenous infusion over 5 days, given every 3 weeks for 6 cycles (days 1 to 5 of each cycle)

 

Either of which was administered with:

 

-             cisplatin – 80 mg/m2 every 3 weeks for 6 cycles on day 1 of each cycle.

 

The efficacy results from study BO18225 are summarised in table 13:

 

Table 13 Efficacy results from BO18225

 

Parameter

FP

N = 290

FP + H

N = 294

HR (95% CI)

p‑value

Overall survival, median months

11.1

13.8

0.74 (0.60-0.91)

0.0046

Progression‑free survival, median months

5.5

6.7

0.71 (0.59-0.85)

0.0002

Time to disease progression, median months

5.6

7.1

0.70 (0.58-0.85)

0.0003

Overall response rate, %

34.5%

47.3%

1.70a (1.22-2.38)

0.0017

Duration of response, median months

4.8

6.9

0.54 (0.40-0.73)

< 0.0001

FP + H: Fluoropyrimidine/cisplatin + trastuzumab

FP: Fluoropyrimidine/cisplatin

a Odds ratio

 

Patients were recruited to the trial who were previously untreated for HER2‑positive inoperable locally advanced or recurrent and/or metastatic adenocarcinoma of the stomach or gastro-oesophageal junction not amenable to curative therapy. The primary endpoint was overall survival which was defined as the time from the date of randomisation to the date of death from any cause. At the time of the analysis a total of 349 randomised patients had died: 182 patients (62.8%) in the control arm and 167 patients (56.8%) in the treatment arm. The majority of the deaths were due to events related to the underlying cancer.

 

Post‑hoc subgroup analyses indicate that positive treatment effects are limited to targeting tumours with higher levels of HER2 protein (IHC 2+/FISH+ or IHC 3+). The median overall survival for the high HER2 expressing group was 11.8 months versus 16 months, HR 0.65 (95% CI: 0.51-0.83) and the median progression free survival was 5.5 months versus 7.6 months, HR 0.64 (95% CI: 0.51-0.79) for FP versus FP + H, respectively. For overall survival, the HR was 0.75 (95% CI: 0.51-1.11) in the IHC 2+/FISH+ group and the HR was 0.58 (95% CI: 0.41‑0.81) in the IHC 3+/FISH+ group.

 

In an exploratory subgroup analysis performed in the ToGA (BO18255) trial there was no apparent benefit on overall survival with the addition of trastuzumab in patients with ECOG PS 2 at baseline [HR 0.96 (95% CI: 0.51-1.79)], non measurable [HR 1.78 (95% CI: 0.87-3.66)] and locally advanced disease [HR 1.20 (95% CI: 0.29-4.97)].

 

Paediatric population

 

The European Medicines Agency has waived the obligation to submit the results of studies with trastuzumab in all subsets of the paediatric population for breast and gastric cancer (see section 4.2 for information on paediatric use).


The pharmacokinetics of trastuzumab were evaluated in a population pharmacokinetic model analysis using pooled data from 1,582 subjects, including patients with HER2-positive MBC, EBC, AGC or other tumour types, and healthy volunteers, in 18 Phase I, II and III trials receiving trastuzumab intravenously. A two‑compartment model with parallel linear and non‑linear elimination from the central compartment described the trastuzumab concentration‑time profile. Due to non‑linear elimination, total clearance increased with decreasing concentration. Therefore, no constant value for half‑life of trastuzumab can be deduced. The t1/2 decreases with decreasing concentrations within a dosing interval (see table 16). MBC and EBC patients had similar PK parameters (e.g. clearance (CL), the central compartment volume (Vc)) and population predicted steady‑state exposures (Cmin, Cmax and AUC). Linear clearance was 0.136 L/day for MBC, 0.112 L/day for EBC and 0.176 L/day for AGC. The non‑linear elimination parameter values were 8.81 mg/day for the maximum elimination rate (Vmax) and 8.92 µg/mL for the Michaelis‑Menten constant (Km) for the MBC, EBC, and AGC patients. The central compartment volume was 2.62 L for patients with MBC and EBC and 3.63 L for patients with AGC. In the final population PK model, in addition to primary tumour type, body weight, serum aspartate aminotransferase and albumin were identified as a statistically significant covariates affecting the exposure of trastuzumab. However, the magnitude of effect of these covariates on trastuzumab exposure suggests that these covariates are unlikely to have a clinically meaningful effect on trastuzumab concentrations.

 

The population predicted PK exposure values (median with 5th‑95th percentiles) and PK parameter values at clinically relevant concentrations (Cmax and Cmin) for MBC, EBC and AGC patients treated with the approved q1w and q3w dosing regimens are shown in table 14 (cycle 1), table 15 (steady‑state), and table 16 (PK parameters).

 

Table 14 Population predicted cycle 1 PK exposure values (median with 5th‑95th percentiles) for trastuzumab intravenous dosing regimens in MBC, EBC and AGC patients

 

Regimen

Primary tumour type

N

Cmin

(µg/mL)

Cmax

(µg/mL)

AUC0-21days (µg.day/mL)

8 mg/kg +

6 mg/kg q3w

MBC

805

28.7

(2.9‑46.3)

182

(134‑280)

1,376

(728 ‑1,998)

EBC

390

30.9

(18.7‑45.5)

176

(127‑227)

1,390

(1,039‑1,895)

AGC

274

23.1

(6.1‑50.3)

132

(84.2‑225)

1,109

(588‑1,938)

4 mg/kg + 2 mg/kg qw

MBC

805

37.4

(8.7‑58.9)

76.5

(49.4‑114)

1,073

(597‑1,584)

EBC

390

38.9

(25.3‑58.8)

76.0

(54.7‑104)

1,074

(783‑1,502)

 

Table 15 Population predicted steady-state PK exposure values (median with 5th‑95th percentiles) for trastuzumab intravenous dosing regimens in MBC, EBC and AGC patients

 

Regimen

Primary tumour type

N

Cmin,ss*

(µg/mL)

Cmax,ss** (µg/mL)

AUCss, 0-21days (µg.day/mL)

Time to steady-state*** (week)

8 mg/kg + 6 mg/kg q3w

MBC

805

44.2

(1.8‑85.4)

179

(123‑266)

1,736

(618‑2,756)

12

EBC

390

53.8

(28.7‑85.8)

184

(134‑247)

1,927

(1,332‑2,771)

15

AGC

274

32.9

(6.1‑88.9)

131

(72.5‑251)

1,338

(557‑2,875)

9

4 mg/kg + 2 mg/kg qw

MBC

805

63.1

(11.7‑107)

107

(54.2‑164)

1,710

(581‑2,715)

12

EBC

390

72.6

(46‑109)

115

(82.6‑160)

1,893

(1,309‑2,734)

14

*Cmin,ss = Cmin at steady‑state

**Cmax,ss = Cmax at steady‑state

***time to 90% of steady‑state

 

Table 16 Population predicted PK parameter values at steady-state for trastuzumab intravenous dosing regimens in MBC, EBC and AGC patients

 

Regimen

Primary tumour type

N

Total CL range

from Cmax,ss to Cmin,ss

(L/day)

t1/2 range from Cmax,ss to Cmin,ss

(day)

8 mg/kg + 6 mg/kg q3w

MBC

805

0.183‑0.302

15.1‑23.3

EBC

390

0.158‑0.253

17.5‑26.6

AGC

274

0.189‑0.337

12.6‑20.6

4 mg/kg + 2 mg/kg qw

MBC

805

0.213‑0.259

17.2‑20.4

EBC

390

0.184‑0.221

19.7‑23.2

 

Trastuzumab washout

 

Trastuzumab washout period was assessed following q1w or q3w intravenous administration using the population PK model. The results of these simulations indicate that at least 95% of patients will reach concentrations that are < 1 µg/mL (approximately 3% of the population predicted Cmin,ss, or about 97% washout) by 7 months.

 

Circulating shed HER2-ECD

 

The exploratory analyses of covariates with information in only a subset of patients suggested that patients with greater shed HER2-ECD level had faster non-linear clearance (lower Km) (p < 0.001). There was a correlation between shed antigen and SGOT/AST levels; part of the impact of shed antigen on clearance may have been explained by SGOT/AST levels.

 

Baseline levels of the shed HER2-ECD observed in MGC patients were comparable to those in MBC and EBC patients and no apparent impact on trastuzumab clearance was observed.


There was no evidence of acute or multiple dose-related toxicity in studies of up to 6 months, or reproductive toxicity in teratology, female fertility or late gestational toxicity/placental transfer studies. Trastuzumab is not genotoxic. A study of trehalose, a major formulation excipient did not reveal any toxicities.

 

No long-term animal studies have been performed to establish the carcinogenic potential of trastuzumab, or to determine its effects on fertility in males.


1 vial of 150 mg contains:

Histidine: 2.25 mg

Histidine monohydrochloride: 3.49 mg

Trehalose dihydrate: 142 mg

Polysorbate 20: 0.6 mg

 

1 vial of 420 mg contains:

Histidine: 6.36 mg

Histidine monohydrochloride: 9.87 mg

Trehalose dihydrate: 401 mg

Polysorbate 20: 1.76 mg

 


This medicinal product must not be mixed or diluted with other medicinal products except those mentioned under section 6.6.

 

KANJINTI should not be diluted with glucose solutions since these cause aggregation of the protein.


Unopened vial 3 years. Aseptic reconstitution and dilution: After aseptic reconstitution with sterile water for injections, chemical and physical stability of the reconstituted solution has been demonstrated for 48 hours at 2°C 8°C. After aseptic dilution in polyvinylchloride, polyethylene or polypropylene bags containing sodium chloride 9 mg/mL (0.9%) solution for injection, chemical and physical stability of KANJINTI has been demonstrated for up to 30 days at 2°C 8°C, and subsequently for 24 hours at temperatures not exceeding 30°C. From a microbiological point of view, the reconstituted solution and KANJINTI infusion solution should be used immediately. If not used immediately, in use storage times and conditions prior to use are the responsibility of the user, and would not normally be longer than 24 hours at 2°C 8°C, unless reconstitution and dilution have taken place under controlled and validated aseptic conditions.

Store in a refrigerator (2°C ‑ 8°C).

Do not freeze the reconstituted solution.

Store in the original package in order to protect from light.

 

For storage conditions of the reconstituted medicinal product, see sections 6.3 and 6.6.


KANJINTI 150 mg powder for concentrate for solution for infusion

 

20 mL clear glass type I vial with butyl rubber stopper laminated with a fluoro‑resin film and aluminium seal flip-off dust cover containing 150 mg of trastuzumab.

 

Each carton contains one vial.

 

KANJINTI 420 mg powder for concentrate for solution for infusion

 

50 mL clear glass type I vial with butyl rubber stopper laminated with a fluoro‑resin film and aluminium seal flip-off dust cover containing 420 mg of trastuzumab.

 

Each carton contains one vial.


Appropriate aseptic technique should be used for reconstitution and dilution procedures. Care must be taken to ensure the sterility of prepared solutions. Since the medicinal product does not contain any anti-microbial preservative or bacteriostatic agents, aseptic technique must be observed.

Aseptic preparation, handling and storage:

 

Aseptic handling must be ensured when preparing the infusion. Preparation should be:

·            performed under aseptic conditions by trained personnel in accordance with good practice rules especially with respect to the aseptic preparation of parenteral products.

·            prepared in a laminar flow hood or biological safety cabinet using standard precautions for the safe handling of intravenous agents.

·            followed by adequate storage of the prepared solution for intravenous infusion to ensure maintenance of the aseptic conditions.

 

KANJINTI should be carefully handled during reconstitution. Causing excessive foaming during reconstitution or shaking the reconstituted solution may result in problems with the amount of KANJINTI that can be withdrawn from the vial.

 

The reconstituted solution should not be frozen.

 

KANJINTI 150 mg powder for concentrate for solution for infusion

 

Each 150 mg vial of KANJINTI is reconstituted with 7.2 mL of sterile water for injections (not supplied). Use of other reconstitution solvents should be avoided.

 

This yields a 7.4 mL solution for single‑dose use, containing approximately 21 mg/mL trastuzumab, at a pH of approximately 6.1. A volume overage of 4% ensures that the labelled dose of 150 mg can be withdrawn from each vial.

 

KANJINTI 420 mg powder for concentrate for solution for infusion

 

Each 420 mg vial of KANJINTI is reconstituted with 20 mL of sterile water for injections (not supplied). Use of other reconstitution solvents should be avoided.

 

This yields a 21 mL solution for single‑dose use, containing approximately 21 mg/mL trastuzumab, at a pH of approximately 6.1. A volume overage of 5% ensures that the labelled dose of 420 mg can be withdrawn from each vial.

 

KANJINTI vial

 

Volume of sterile water for injections

 

Final concentration

150 mg vial

+

7.2 mL

=

21 mg/mL

420 mg vial

+

20 mL

=

21 mg/mL

 

Instructions for aseptic reconstitution

 

1) Using a sterile syringe, slowly inject the appropriate volume (as noted above) of sterile water for injections in the vial containing the lyophilised KANJINTI, directing the stream into the lyophilised cake.

 

2) Swirl the vial gently to aid reconstitution. DO NOT SHAKE.

 

Slight foaming of the product upon reconstitution is not unusual. Allow the vial to stand undisturbed for approximately 5 minutes. The reconstituted KANJINTI results in a colourless to pale yellow transparent solution and should be essentially free of visible particulates.

 

Instructions for aseptic dilution of the reconstituted solution

 

Determine the volume of the solution required:

·            based on a loading dose of 4 mg trastuzumab/kg body weight, or a subsequent weekly dose of 2 mg trastuzumab/kg body weight:

 

Volume (mL) = Body weight (kg) × dose (mg/kg for loading or mg/kg for maintenance)

21 (mg/mL, concentration of reconstituted solution)

 

·            based on a loading dose of 8 mg trastuzumab/kg body weight, or a subsequent 3‑weekly dose of 6 mg trastuzumab/kg body weight:

 

Volume (mL) = Body weight (kg) × dose (mg/kg for loading or mg/kg for maintenance)

21 (mg/mL, concentration of reconstituted solution)

 

The appropriate amount of solution should be withdrawn from the vial using a sterile needle and syringe and added to an infusion bag containing 250 mL of sodium chloride 9 mg/mL (0.9%) solution for injection. Do not use with glucose‑containing solutions (see section 6.2). The bag should be gently inverted to mix the solution in order to avoid foaming.

 

Parenteral medicinal products should be inspected visually for particulate matter and discolouration prior to administration.

 

No incompatibilities between KANJINTI and polyvinylchloride, polyethylene or polypropylene bags have been observed.

 

KANJINTI is for single‑use only, as the product contains no preservatives. Any unused medicinal product or waste material should be disposed of in accordance with local requirements.


Amgen Europe B.V. Minervum 7061 NL 4817 ZK Breda The Netherlands

November 2021
}

صورة المنتج على الرف

الصورة الاساسية