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)

Xyntha is a form of coagulation factor VIII. Coagulation Factor VIII is a protein that is necessary for blood to clot. People who have the hereditary bleeding disease factor VIII deficiency (hemophilia A) lack this clotting factor, causing their blood to take longer to form a clot.

 

Xyntha is used for the control and treatment of bleeding and the routine and surgical prevention of bleeding in people with hemophilia A.

 

Administering Xyntha increases blood levels of factor VIII and helps prevent and control bleeding episodes in patients with hemophilia A. Xyntha has the same clot-promoting effects as factor VIII protein made from human plasma.

 


Do not use Xyntha :

 

-          If you are allergic to moroctocog alfa or any of the other ingredients of this medicine (listed in section 6)

-          You should not take Xyntha unless your doctor confirms you have hemophilia A. Xyntha should not be used for the treatment of other clotting factor deficiencies such as von Willebrand’s disease or hemophilia B.

-          Xyntha is produced in hamster cells and may contain trace amounts of hamster proteins. Patients who have a known history of allergic reactions to hamster proteins should not take Xyntha.   Your doctor will advise you if you have had allergic reactions to hamster proteins.

-          Do not use Xyntha after the expiry date printed on the pack.

-          Do not use Xyntha if the packaging is torn or shows signs of tampering.

 

 Warnings and precautions

 

BEFORE using Xyntha talk to your hemophilia doctor if:

·         You are pregnant or planning to become pregnantIt is not known whether Xyntha can affect your ability to have children or harm your developing baby.

·         You are breastfeeding or planning to breast feed.  It is not known whether Xyntha passes into breast milk.

·         You are taking other medicines. Tell your hemophilia doctor if you are taking any other medicines or naturopathic products, including any that you buy without a prescription from your pharmacy, supermarket, or health food shop.

 

Your hemophilia doctor will advise you whether or not to take Xyntha or if you need to adjust the dose, or adapt your treatment.

 

 

Other medicines and Xyntha :

 

No interactions of Xyntha with other medicinal products are known.

 

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

 

 

Pregnancy, breast-feeding and fertility

 

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

 

Driving and using machines

 

Xyntha has no influence on the ability to drive or use machines.

 

 

 

Xyntha  contains :

 

What the medicinal ingredient is:

Xyntha is a coagulation factor VIII product that is produced by recombinant technology.

 

Xyntha does not contain any human blood or plasma, albumin, preservatives, or added animal or human components in the final product, making it naturally free from the risk of transmission of blood-borne pathogens such as human immunodeficiency virus (HIV), hepatitis viruses, and parvovirus.

 

What the important nonmedicinal ingredients are:

Polysorbate 80 (0.4 mg/vial), Sucrose (12 mg/vial), L-Histidine (6 mg/vial), Calcium Chloride Dihydrate (1 mg/vial), Sodium Chloride (72 mg/vial) [after reconstitution with diluent].

 

What dosage forms it comes in:

Xyntha vial kit:

Xyntha is supplied in kits that include single-use vials that contain nominally 250, 500, 1000, or 2000 IU per vial with pre-filled diluent syringe containing 4 mL 0.9% Sodium Chloride for reconstitution.

 


You should always follow the specific instructions given by your hemophilia doctor.

 

The steps listed below are general guidelines for using Xyntha.   If you are unsure of the procedures, please call your hemophilia doctor or nurse before using.

 

Your hemophilia doctor will decide the dose of Xyntha you will receive.  The dose and duration of Xyntha treatment will depend on your individual need for replacement factor VIII therapy.

 

Your hemophilia doctor may decide to change your dose of Xyntha.  In the presence of an inhibitor, you may need higher doses or specific treatment.  Contact your hemophilia doctor or nurse immediately if bleeding is not controlled after using Xyntha.

 

 

Your doctor may occasionally need to take blood tests to make sure that the level of factor VIII in your blood is high enough to allow normal blood clotting.

 

Each Xyntha vial kit contains:

·         1 vial of Xyntha powder

·         1 syringe pre-filled with 4 mL of sodium chloride diluent

·         1 plunger rod for the syringe

·         1 vial adapter

·         1 sterile infusion set

·         2 alcohol swabs

·         1 bandage

·         1 gauze

·         1 package insert

 

Xyntha is supplied in a sterile, freeze-dried powder form, and it is intended for injection directly into your vein, usually either by yourself, your doctor, your hemophilia nurse, or other trained person.

 

Before it can be injected, the powder must be dissolved with the liquid diluent supplied (0.9% sodium chloride) to make it an injectable liquid.

 

RECONSTITUTION

 

Always wash your hands before performing the following procedures. Use germ-free methods during the preparation procedures.

 

Use only the materials provided in the Xyntha kit for dissolving the Xyntha powder with the sodium chloride diluent.

 

All components used in the mixing and injection of Xyntha should be used as soon as possible after opening their sterile containers to minimize unnecessary exposure to room air.

Xyntha is administered by intravenous injection after dissolving with the supplied diluent (0.9% sodium chloride) in the pre-filled syringe.

 

Additional instructions for using Xyntha are provided after INFUSION (Intravenous Injection) that detail the use of a Xyntha vial or the combined used of Xyntha vial Note: If you use more than one vial of Xyntha per injection, each vial should be dissolved according to the following instructions.  The empty syringe should be removed leaving the vial adapter in place, and a separate large luer lock syringe may be used to draw back the dissolved contents of each vial. Do not detach the diluent syringes or the large luer lock syringe until you are ready to attach the large luer lock syringe to the next vial adapter.

 

  1. Allow the vial of freeze-dried Xyntha powder and the pre-filled diluent syringe to reach room temperature.
  2. Remove the plastic flip-top cap from the Xyntha vial to expose the central portions of the rubber stopper.

 

 

 

  1. Wipe the top of the vial with the alcohol swab provided, or use another antiseptic solution, and allow to dry. After cleaning, do not touch the rubber stopper with your hand or allow it to touch any surface.
  2. Peel back the cover from the clear plastic vial adapter package. Do not remove the adapter from the package.
  3. Place the vial on a flat surface. While holding the adapter in the package, place the vial adapter over the vial. Press down firmly on the package until the adapter snaps into place on top of the vial, with the adapter spike penetrating the vial stopper.

 

 

  1. Grasp the plunger rod as shown in the diagram. Avoid contact with the shaft of the plunger rod. Attach the threaded end of the plunger rod to the diluent syringe by pushing and turning firmly.

  1. Break off the tamper-resistant, plastic-tip cap from the diluent syringe by snapping the perforation of the cap. This is done by bending the cap up and down until the perforation is broken.  Do not touch the inside of the cap or the syringe tip. The diluent syringe may need to be recapped (if the dissolved Xyntha is not used immediately), so place the cap on its top on a clean surface in a spot where it would be least likely to become contaminated.

  1. Lift the package away from the adapter and discard the package.

  1. Place the vial on a flat surface. Connect the diluent syringe to the vial adapter by inserting the tip of the syringe into the adapter opening while firmly pushing and turning the syringe clockwise until the connection is secured.

  1. Slowly depress the plunger rod to inject all the diluent into the Xyntha vial.

  1. With the syringe still connected to the adapter, gently swirl the contents of the vial until the powder is dissolved.

Note: The final solution should be inspected visually for particulate matter before administration.  The solution should be clear to slightly pearly and colorless.  If it is not, the solution should be discarded and a new kit should be used.

  1. Ensuring that the syringe plunger rod is still fully depressed, invert the vial. Slowly draw the solution into the syringe.

Note: If you prepared more than one vial of Xyntha, remove the diluent syringe from the vial adapter, leaving the vial adapter attached to the vial. Quickly attach a separate large luer lock syringe and draw back the dissolved contents as instructed above. Repeat this procedure with each vial in turn. Do not detach the diluent syringes or the large luer lock syringe until you are ready to attach the large luer lock syringe to the next vial

adapter.

 

 

  1. Detach the syringe from the vial adapter by gently pulling and turning the syringe counterclockwise. Discard the vial with the adapter attached.

Note: If the solution is not to be used immediately, the syringe cap should be carefully replaced. Do not touch the syringe tip or the inside of the cap.

Xyntha should be infused within 3 hours after dissolving. The dissolved solution may be stored at room temperature prior to infusion.

 

INFUSION (Intravenous Injection)

 

You should inject Xyntha as instructed by your hemophilia doctor or nurse. Once you learn how to self-infuse, you can follow the instructions in this insert.

 

Always wash your hands before doing the following procedures.  Germ-free methods should be used during injection.

 

Xyntha should be administered using the pre-filled diluent syringe provided or a single sterile disposable plastic luer-lock syringe.  In addition, the solution should be withdrawn from the vial using the vial adapter.

 

1.  Attach the syringe to the luer end of the provided infusion set tubing and perform venipuncture as instructed by your hemophilia doctor or nurse.

2.  Apply a tourniquet and prepare the injection site by wiping the skin well with an alcohol swab provided in the kit.

3.  Insert the needle on the infusion set tubing into the vein, and remove the tourniquet.  Infuse the reconstituted Xyntha product over several minutes.  Your comfort level should determine the rate of infusion.

4.  After injecting Xyntha, remove the infusion set and discard. The amount of drug product left in the infusion set will not affect your treatment. Dispose of all unused solution, the empty vial(s), and the used needles and syringes in an appropriate sharps container used for throwing away waste that might hurt others if not handled properly.

You should record the lot number of the product every time you use Xyntha.  The lot number can be found on the vial label. The peel-off label on the vial may be used to record the lot number.

 

If you use more Xyntha than you should

 

In case of drug overdose, contact a health care practitioner, hospital emergency department or regional Poison Control Centre immediately, even if there are no symptoms.

 

If you stop using Xyntha

 

Do not stop taking Xyntha or lower your dose, without checking with your hemophilia doctor, unless you think you are having an allergic reaction.

 

 

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

 


Like all medicines, this medicine can cause side effects, although not everybody gets them.

 

Xyntha may cause allergic reactions.

 

Some side effects of Xyntha include cold sensation, chest pain, rapid heart beat, sweating, drowsiness, muscle weakness,    loss of appetite, muscle pain, pain, coughing, shortness of breath, diarrhea, stomach cramps, altered taste, injection site pain or inflammation and weakness. 

 

Contact your hemophilia doctor immediately if your bleeding does not stop as expected.

 

Your body can also produce inhibitors, or antibodies, against Xyntha, which may prevent Xyntha from working properly.  If your bleeding is not controlled with the usual dose of Xyntha, call your hemophilia doctor or nurse.  You should be monitored in order to find out whether a factor VIII inhibitor is present.

 

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

 

·         inhibitor development for patients who have never been previously treated with factor VIII products

 

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

 

 

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

 

 

These are not all the possible side effects of Xyntha. Call your hemophilia doctor or nurse right away if any side effect becomes serious, if you notice any side effects not listed in this leaflet, or if there is any other side effect that concerns you.

 

Tell all your doctors, dentists, and pharmacists who are treating you that you are taking Xyntha.

 

If you are about to start taking any new medication, tell your doctor and pharmacist that you are taking Xyntha.

 

If you become pregnant while taking Xyntha, tell your hemophilia doctor and your doctor who will look after you during your pregnancy.

 

SERIOUS SIDE EFFECTS, HOW OFTEN THEY

HAPPEN AND WHAT TO DO ABOUT THEM

Symptom / effect

Talk with your doctor

Stop the infusion and call your doctor

Only if severe

In all cases

Uncommon

Headache

 

 

P

Rare

Fever

 

 

P

Nausea

 

 

P

Vomiting

 

 

P

Very Rare

Bleeding does not stop as expected after infusing Xyntha

 

P

 

Using an increasing amount of Xyntha to control your bleed

 

P

 

Chills

 

 

P

Flushing

 

 

P

Allergic reactions (such as skin rash, itching, chest tightness, wheezing, dizziness, hives, faintness, rapid heartbeat, shortness of breath, and/or a swollen face)

 

 

P

 

This is not a complete list of side effects.  For any unexpected effects while taking Xyntha, contact your doctor or hemophilia treatment center.

 

Reporting of side effects

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

 

 

 

 

To Report any Side effect(s):

 

National Pharmacovigilance Center ( NPC )

·    Fax: +966 11 205 7662

·    Call NPC at 00966 11 2038222, Exts: 2317-2356-2353-2354-2334-2340

·    Toll-Free phone: 8002490000

·    E-mail: npc.drug@sfda.gov.sa

·    Website: www.sfda.gov.sa/npc

 


Keep Xyntha out of the sight and reach of children.

 

Do not use Xyntha after the expiry date which is stated on the outer carton or vial label .The expiry date refers to the last day of that month.

 

Keep the freeze-dried Xyntha vial and the diluent syringe in the refrigerator at a temperature of 2° to 8°C.

 

Xyntha may be stored at room temperature not above 25°C for up to 3 months and not to exceed the expiration date. You should write the date in the space provided on the outer carton when you first store Xyntha at room temperature.

After room temperature storage, the product can be returned to refrigerated storage until the expiration date. Do not store Xyntha at room temperature and return it to refrigerated storage more than once.

 

DO NOT freeze.

 

Avoid exposure of the Xyntha vials to light.

 

Xyntha does not contain a preservative. Use the dissolved solution as soon as possible after mixing.  If the solution is not used right away, it can be stored at room temperature and must be used within 3 hours.

 

DO NOT use Xyntha if the dissolved solution is not clear to slightly pearly and colorless.

 

Do not throw away any medicines via wastewater . Ask your pharmacist how to throw away medicines you no longer use. These measures will help protect the environment.

 


What the medicinal ingredient is:

Xyntha is a coagulation factor VIII product that is produced by recombinant technology.

 

Xyntha does not contain any human blood or plasma, albumin, preservatives, or added animal or human components in the final product, making it naturally free from the risk of transmission of blood-borne pathogens such as human immunodeficiency virus (HIV), hepatitis viruses, and parvovirus.

 

What the important nonmedicinal ingredients are:

Polysorbate 80 (0.4 mg/vial), Sucrose (12 mg/vial), L-Histidine (6 mg/vial), Calcium Chloride Dihydrate (1 mg/vial), Sodium Chloride (72 mg/vial) [after reconstitution with diluent].

 


What dosage forms it comes in: Xyntha vial kit: Xyntha is supplied in kits that include single-use vials that contain nominally 250, 500, 1000, or 2000 IU per vial with pre-filled diluent syringe containing 4 mL 0.9% Sodium Chloride for reconstitution.

Marketing Authorization Holder:

 

WYETH FARMA S.A.

AUTOVIA DEL NORTE A-1, KM. 23. DESVIO ALGETE, KM. 1, 28700 S. SEBASTIAN DE LOS

REYES, MADRID, SPAIN

 

Manufactured, Packed and Released by:

 

WYETH FARMA S.A.

AUTOVIA DEL NORTE A-1, KM. 23. DESVIO ALGETE, KM. 1, 28700 S. SEBASTIAN DE LOS

REYES, MADRID, SPAIN

 


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

زينثا هو أحد أشكال عامل التجلط الثامن. عامل التجلط الثامن هو بروتين ضروري لعملية تجلط الدم. الأشخاص المصابون بمرض نقص العامل الثامن النزفي الوراثي (الهيموفيليا من النوع أ) يكون لديهم نقص في عامل التجلط هذا مما ينتج عنه إطالة المدة التي يحتاجها الدم لتكوين جلطة.

 

يُستخدم زينثا للتحكم في النزيف وعلاجه وللمنع الروتيني والجراحي للنزيف لدى الأشخاص المصابين بالهيموفيليا من النوع أ.

 

يزيد استعمال زينثا من مستويات العامل الثامن بالدم ويساعد في منع نوبات النزيف والتحكم بها لدى المرضى المصابين بالهيموفيليا من النوع أ. زينثا له نفس التأثيرات المحفزة للتجلط مثل بروتين العامل الثامن المصنوع من البلازما البشرية.

 

لا تستخدم زينثا في الحالات التالية :

-          إذا كنت مصابا بالحساسية تجاه موروكتوكوج ألفا  أو أي من المكونات الأخرى لهذا الدواء (المدرجة بالقسم 6).

-          ينبغي ألا تتلقى زينثا إلا إذا أكد طبيبك إصابتك بالهيموفيليا من النوع أ. ينبغي عدم استخدام زينثا لعلاج حالات النقص في عوامل التجلط الأخرى مثل مرض فون فليبراند أو الهيموفيليا من النوع ب.

 

يتم إنتاج زينثا في خلايا الهامستر وقد يحتوي على كميات ضئيلة من بروتينات الهامستر. ينبغي على المرضى الذين لديهم تاريخ معروف من الإصابة بتفاعلات حساسية تجاه بروتينات الهامستر عدم استخدام زينثا.   سيقدم لك طبيبك المشورة إذا ما كان قد سبقت لك الإصابة بتفاعلات حساسية تجاه بروتينات الهامستر.

 

لا تستخدم زينثا بعد مرور تاريخ انتهاء الصلاحية المطبوع على العبوة.

 

لا تستخدم زينثا إذا كانت العبوة ممزقة أو تظهر عليها علامات عبث.

 

 

تحذيرات واحتياطات

 

استشر طبيبك المختص بعلاج الهيموفيليا قبل استخدامك لزينثا إذا:

·         كنتِ حاملًا أو تخططين للحملنظرًا لأنه من غير المعروف إذا ما كان من المحتمل أن يؤثر زينثا على قدرتك على الإنجاب أو أن يسبب أذىً لجنينك أم لا.

·         كنتِ ترضعين رضاعة طبيعية أو تخططين لذلك.  ليس معروفًا إذا ما كان زينثا يُفرز في لبن الثدي أم لا.

·         كنت تتناول أي أدوية أخرى. أخبر طبيبك المختص بعلاج الهيموفيليا إذا كنت تتناول أي أدوية أخرى أو منتجات علاج طبيعية، بما في ذلك أي أدوية يمكنك شراؤها من الصيدلية أو السوبر ماركت أو متجر الأطعمة الصحية بدون وصفة طبية.

 

سيقوم طبيبك المختص بعلاج الهيموفيليا بإخبارك إذا ما كان يمكنك أن تتناول زينثا أم لا أو إذا ما كنت بحاجة لتعديل الجرعة أو تكييف علاجك.

 

 

الأدوية الأخرى وزينثا :

 

لا توجد تفاعلات معروفة بين زينثا وغيره من المنتجات الدوائية.

 

يجب إخطار طبيبك إذا كنت تتناول أو تناولت مؤخرًا أو قد تتناول أي أدوية أخرى.

 

 

الحمل ، الرضاعة الطبيعية والخصوبة

إذا كنتِ حاملًا أو ترضعين رضاعة طبيعية، أو تعتقدين أنكِ ربما تكونين حاملاً أو تخططين للحمل، فاستشيري طبيبكِ قبل تناول هذا الدواء.

 

القيادة واستخدام الآلات

 

لا يوجد تأثير لاستخدام زينثا على القدرة على القيادة أو استخدام الآلات.

 

يحتوي زينثا على:

 

المكونات الدوائية:

زينثا هو منتج يحتوي على عامل التجلط الثامن، يتم إنتاجه بواسطة تكنولوجيا الحمض النووي المأشوب (دمج الجينات).

 

لا يحتوي زينثا على أي دم أو بلازما من النوع البشري، أو الألبومين، أو مواد حافظة، أو مكونات حيوانية أو بشرية مضافة في المنتج النهائي مما يجعله خاليًا بشكل طبيعي من خطر   فيروسات  الدم مثل فيروس نقص المناعة البشرية (HIV)، وفيروسات الالتهاب الكبدي الوبائي، والفيروس الصغير.

 

المكونات غير الدوائية المهمة:

بولي سوربات 80 (0.4 مغ/القارورة)، سكروز (12 مغ/القارورة)، ل-هيستيدين (6 مغ/القارورة)، كلوريد الكالسيوم ثنائي الهيدرات (1 مغ/القارورة)، كلوريد الصوديوم (72 مغ/القارورة) [بعد التحضير باستخدام المحلول المخفِّف].

 

أشكال الجرعات التي يأتي بها:

مجموعة أدوات قارورة زينثا:

يتوفر زينثا في مجموعات أدوات تتضمن قوارير مخصصة للاستخدام مرة واحدة تحتوي على قيم اسمية تساوي 250، أو 500، أو 1000، أو 2000 وحدة دولية للقارورة ومحقنة واحدة مسبقة التعبئة من المحلول المخفِّف تحتوي على 4 مل من كلوريد الصوديوم بتركيز 0.9 % للتحضير.

https://localhost:44358/Dashboard

ينبغي عليك دائمًا اتباع التعليمات المحددة التي يعطيها لك طبيبك المختص بعلاج الهيموفيليا.

 

الخطوات المدرجة أدناه هي توجيهات عامة لاستخدام زينثا.   إذا لم تكن واثقًا من الإجراءات، يُرجى الاتصال بطبيبك أو ممرضك المختص بعلاج الهيموفيليا قبل الاستخدام.

 

سوف يقرر طبيبك المختص بعلاج الهيموفيليا مقدار جرعة زينثا التي ستتلقاها.  ستعتمد جرعة زينثا ومدة العلاج به على حاجتك الفردية للعلاج بالعامل الثامن البديل.

 

قد يقرر طبيبك المختص بعلاج الهيموفيليا تغيير جرعتك من زينثا.  وقد تحتاج لجرعات أعلى أو إلى علاج محدد في حالة وجود أحد المثبطات.  اتصل بطبيبك أو ممرضك المختص بعلاج الهيموفيليا على الفور إذا لم يتم التحكم بالنزيف بعد استخدام زينثا.

 

لا تتوقف عن تناول زينثا أو تقلل جرعتك دون الرجوع لطبيبك المختص بعلاج الهيموفيليا إلا إذا كنت تعتقد أنك تتعرض لتفاعل حساسية.

 

قد يحتاج طبيبك أحيانًا إلى إجراء فحوصات للدم للتأكد من أن مستوى العامل الثامن في دمك مرتفع بما يكفي ليسمح بالتجلط الطبيعي للدم.

 

تحتوي كل مجموعة من مجموعات أدوات قارورة زينثا على:

·         قارورة واحدة من مسحوق زينثا

·         محقنة واحدة مسبقة التعبئة تحتوي على 4 مل من محلول كلوريد الصوديوم المخفِّف

·         مكبس واحد للمحقنة

·         مهايئ واحد للقارورة

·         مجموعة تنقيط وريدي  واحدة معقمة

·         مسحتان كحوليتان

·         ضمادة واحدة

·         قطعة واحدة من الشاش

·         نشرة عبوة واحدة

 

يتوفر زينثا في شكل مسحوق معقم مجفف بالتجميد، وهو مخصص للحقن مباشرةً في وريدك وعادة ما يتم ذلك بواسطتك أو بواسطة طبيبك أو ممرضك المختص بعلاج الهيموفيليا أو شخص مدرب آخر.

 

يجب أن تتم إذابة المسحوق في المحلول المخفِّف السائل المورَّد (كلوريد الصوديوم بتركيز 0.9 %) لجعله سائلًا قابلًا للحقن، قبل أن يصبح من الممكن حقنه.

 

التحضير

 

اغسل يديك دائمًا قبل تنفيذ الإجراءات التالية. استخدم طرقًا خالية من الجراثيم أثناء إجراءات التجهيز.

 

استخدم المواد المرفقة مع مجموعة أدوات زينثا فقط لإذابة مسحوق زينثا بمحلول كلوريد الصوديوم المخفِّف.

 

ينبغي استخدام جميع المكونات المستعملة في عملية خلط وحقن زينثا في أقرب وقت ممكن بعد فتح الحاويات المعقمة الخاصة بها لتقليل تعرضها غير الضروري لهواء الغرفة.

يتم إعطاء زينثا عن طريق الحقن في الوريد بعد إذابته بالمحلول المخفِّف المورَّد (كلوريد الصوديوم بتركيز 0.9 %) بداخل المحقنة مسبقة التعبئة.

 

ترد تعليمات إضافية حول استخدام زينثا بعد العنوان تنقيط وريدي  (الحقن في الوريد) تُفصل كيفية استخدام إحدى قوارير زينثا أو الاستخدام المشترك لقارورة زينثا

 

ملاحظة: إذا استخدمت أكثر من قارورة واحدة من قوارير زينثا في عملية الحقن الواحدة، ينبغي أن تتم إذابة كل واحدة منها وفقًا للتعليمات التالية.  ينبغي نزع المحقنة الفارغة مع ترك مهايئ القارورة في موضعه، ويمكن استخدام محقنة كبيرة منفصلة مزودة بقفل "لور" لسحب المحتوى المذاب من كل قارورة. لا تفصل محاقن المحلول المخفِّف أو المحقنة الكبيرة المزودة بقفل "لور" حتى تكون مستعدًا لتثبيت المحقنة الكبيرة المزودة بقفل "لور" بمهايئ القارورة التالية.

 

  1. اترك قارورة مسحوق زينثا المجفف بالتجميد ومحقنة المحلول المخفِّف مسبقة التعبئة حتى تصلا إلى درجة حرارة الغرفة.
  2. انزع الغطاء البلاستيكي الذي يُفتح لأعلى من على قارورة زينثا لتظهر الأجزاء الوسطية  من السدادة المطاطية.

 

 

 

  1. امسح أعلى القارورة باستخدام المسحة الكحولية المرفقة، أو استخدم محلولاً مطهرًا آخر، واتركه ليجف. وبعد التنظيف، لا تلمس السدادة المطاطية بيدك ولا تدعها تلمس أي سطح.
  2. قم بإزالة الغطاء عن عبوة مهايئ القارورة البلاستيكية الشفافة. لا تقم بإخراج المهايئ من العبوة.

 

  1. ضع القارورة على سطحٍ مستوٍ. ضع مهايئ القارورة أعلى القارورة مع الإمساك بالمهايئ وهو بداخل العبوة. اضغط على العبوة لأسفل بقوة حتى يستقر المهايئ في مكانه أعلى القارورة مصدرًا صوت طقطقة، ويخترق السن المدبب للمهايئ سدادة القارورة.

 

Refacto 2 of 5

 

  1. أمسك المكبس كما هو موضح في الشكل. تجنب ملامسة المكبس. ثبت الطرف اللولبي للمكبس بمحقنة المحلول المخفِّف عن طريق دفعه وإدارته بقوة.

  1. قم بفصل الغطاء المقاوم للعبث ذي الطرف البلاستيكي عن محقنة المحلول المخفِّف عن طريق كسر الغطاء عند الموضع المثقب. يتم ذلك عن طريق ثني الغطاء لأعلى ولأسفل حتى يتم الكسر عند الموضع المثقب.  لا تلمس الجزء الداخلي من الغطاء أو طرف المحقنة. قد تكون هناك حاجة إلى إعادة تغطية محقنة المحلول المخفِّف (في حالة عدم استخدام زينثا المذاب على الفور)، لذا ضع الغطاء من جهته العلوية على سطح نظيف في موضع توجد به أقل احتمالية لتعرضه للتلوث.

ReFacto 4 of 5

  1. ارفع العبوة عن المهايئ ثم تخلص منها.

ReFacto 3 of 5

  1. ضع القارورة على سطحٍ مستوٍ. قم بتوصيل محقنة المحلول المخفِّف بمهايئ القارورة عن طريق إدخال طرف المحقنة في فتحة المهايئ مع الدفع بقوة وإدارة المحقنة في اتجاه عقارب الساعة حتى يتم التوصيل بإحكام.

  1. اضغط على المكبس ببطء لحقن المحلول المخفِّف بأكمله داخل قارورة زينثا.

  1. والمحقنة لا تزال متصلة بالمهايئ، حرك محتويات القارورة برفق بحركة دائرية إلى أن يذوب المسحوق.

ملاحظة: ينبغي فحص المحلول النهائي بصريًا للتحقق من عدم وجود جسيمات قبل استخدامه.  يُفترض أن يكون المحلول صافيًا أو مائلًا للتعكر قليلًا وعديم اللون.  وإذا لم يكن كذلك، ينبغي التخلص من المحلول واستخدام مجموعة أدوات جديدة.

  1. بعد التأكد من أن مكبس المحقنة لا يزال مضغوطًا بشكل كامل، اقلب القارورة. اسحب المحلول بأكمله إلى المحقنة ببطء.

ملاحظة: إذا قمت بتجهيز أكثر من قارورة زينثا واحدة، انزع محقنة المحلول المخفِّف من مهايئ القارورة، تاركًا المهايئ متصلًا بالقارورة. ثم قم سريعًا بتثبيت محقنة كبيرة منفصلة مزودة بقفل "لور" واسحب المحتويات المذابة وفقًا للتعليمات أعلاه. كرر هذا الإجراء مع كل قارورة بدورها. لا تفصل محاقن المحلول المخفِّف أو المحقنة الكبيرة المزودة بقفل "لور" حتى تكون مستعدًا لتثبيت المحقنة الكبيرة المزودة بقفل "لور" بمهايئ القارورة 

التالية.

  1. افصل المحقنة عن مهايئ القارورة عن طريق سحب المحقنة برفق وإدارتها عكس اتجاه عقارب الساعة. قم بالتخلص من القارورة والمهايئ المثبت بها.

ملاحظة: إذا لم يكن سيتم استخدام المحلول على الفور، ينبغي إعادة غطاء المحقنة إلى مكانه بحذر. لا تلمس طرف المحقنة أو الجزء الداخلي من الغطاء.

ينبغي أن يتم إعطاء زينثا عن طريق التسريب خلال 3 ساعات بعد إذابته. يمكن تخزين المحلول المذاب في درجة حرارة الغرفة قبل التسريب.

 

تنقيط وريدي  (الحقن في الوريد)

 

ينبغي عليك حقن زينثا وفقًا لتعليمات طبيبك أو ممرضك المختص بعلاج الهيموفيليا. بمجرد أن تتعلم كيفية إعطائه لنفسك عن طريق تنقيط وريدي ، يمكنك اتباع التعليمات الموجودة بهذه النشرة.

 

اغسل يديك دائمًا قبل تنفيذ الإجراءات التالية.  ينبغي استخدام طرق خالية من الجراثيم أثناء الحقن.

 

ينبغي إعطاء زينثا باستخدام محقنة المحلول المخفِّف مسبقة التعبئة المرفقة أو محقنة بلاستيكية معقمة معدة للاستخدام مرة واحدة ومزودة بقفل "لور".  بالإضافة إلى ذلك، ينبغي سحب المحلول من القارورة باستخدام مهايئ القارورة.

 

1.  قم بتوصيل المحقنة بالطرف المزود بقفل "لور" الخاص بأنبوب مجموعة التسريب المرفق وقم بثقب الوريد وفقًا لتعليمات طبيبك أو ممرضك المختص بعلاج الهيموفيليا.

2.  ضع مرقأة (عاصبة) وجهِّز موضع الحقن بمسح الجلد جيدًا بإحدى المسحات الكحولية المرفقة في مجموعة الأدوات.

 

3.  أدخل إبرة أنبوب مجموعة التنقيط الوريدي  إلى الوريد، وأزل المرقأة.  قم بإعطاء منتج زينثا الذي تم تحضيره، عن طريق التسريب على مدار عدة دقائق.  من المفترض أن يحدد مستوى الراحة التي تشعر بها معدل التنقيط الوريدي

 .

 

ReFacto AF injection_1

 

4.  بعد حقن زينثا، انزع مجموعة التسريب وتخلص منها. لن تؤثر كمية المنتج الدوائي المتبقية بمجموعة التسريب على علاجك. تخلص من كمية المحلول غير المستخدمة بالكامل، وتخلص كذلك من القارورة (القوارير) الفارغة، والإبر والمحاقن المستعملة في حاوية ملائمة مخصصة للنفايات الحادة تُستخدم للتخلص من المخلفات التي قد تؤذي الآخرين إن لم يتم التخلص منها بصورة سليمة.

ينبغي عليك تسجيل رقم الدفعة الخاص بالمنتج في كل مرة تستخدم فيها زينثا.  يمكن العثور على رقم الدفعة على ملصق القارورة. يمكن استخدام الملصق القابل للنزع الموجود على القارورة لتسجيل رقم الدفعة.

 

في حالة تلقي جرعة مفرطة من العقار، اتصل بأحد ممارسي الرعاية الصحية، أو قسم الطوارئ بإحدى المستشفيات، أو مركز مكافحة السموم الإقليمي على الفور، حتى إن لم تظهر عليك أي أعراض.

 

 

إذا كانت لديك أسئلة أخرى بخصوص استعمال هذا الدواء، فاسأل طبيبك.

 

كما هو الحال بالنسبة لجميع الأدوية، قد يسبب هذا الدواء آثارًا جانبية، غير أنها لا تصيب الجميع.

 

قد يسبب زينثا حدوث تفاعلات حساسية.

 

تتضمن بعض الآثار الجانبية لزينثا الشعور بالبرد، وألم الصدر، وسرعة ضربات القلب، والعرق، والنعاس، وضعف العضلات، وفقدان الشهية، وألم العضلات، والشعور بالألم، والسعال، وضيق التنفس، والإسهال، وتقلصات المعدة، والتغير في حاسة التذوق، والألم أو الالتهاب في موضع الحقن، والضعف. 

 

اتصل بطبيبك المختص بعلاج الهيموفيليا على الفور إذا لم يتوقف نزيفك على النحو المتوقع.

 

قد ينتج جسمك أيضًا مثبطات، أو أجسامًا مضادة لزينثا، مما قد يمنع زينثا من العمل بشكلٍ صحيح.  إذا لم يتم التحكم في نزيفك بتلقي الجرعة المعتادة من زينثا، فاتصل بطبيبك أو ممرضك المختص بعلاج الهيموفيليا.  ينبغي مراقبتك من أجل معرفة إذا ما كان أحد مثبطات العامل الثامن موجودًا أم لا.

 

لا تقتصر الآثار الجانبية المحتملة لزينثا على هذه الآثار. اتصل بطبيبك أو ممرضك المختص بعلاج الهيموفيليا على الفور إذا أصبح أي من الآثار الجانبية خطيرًا، أو إذا لاحظت ظهور أي آثار جانبية غير مدرجة في هذه النشرة، أو إذا ظهرت أي آثار جانبية أخرى تثير قلقك.

 

أخبر جميع من يعالجك من أطباء، وأطباء أسنان، وصيادلة بأنك تتلقى زينثا.

 

إذا كنت على وشك بدء العلاج بأي دواء جديد، فأخبر طبيبك والصيدلي بأنك تتلقى زينثا.

 

إذا أصبحتِ حاملًا أثناء تلقيكِ زينثا، فأخبري طبيبكِ المختص بعلاج الهيموفيليا وطبيبكِ الذي سيتابع حالتكِ خلال فترة الحمل.

 

الآثار الجانبية الخطيرة ومدى تكرار

حدوثها وما ينبغي فعله بشأنها

العرض/الأثر

تحدث مع طبيبك

أوقف التسريب واتصل بطبيبك

فقط إذا كان شديدًا

في جميع الحالات

غير شائع

الصداع

 

 

P

نادر

الحمى

 

 

P

الغثيان

 

 

P

القيء

 

 

P

نادر جدًا

عدم توقف النزيف على النحو المتوقع بعد تسريب زينثا

 

P

 

استخدام كمية متزايدة من زينثا للتحكم في نزيفك

 

P

 

القشعريرة

 

 

P

الاحمرار

 

 

P

تفاعلات الحساسية (مثل الطفح الجلدي، و/أو الحكة، و/أو ضيق الصدر، و/أو الأزيز، و/أو الدوار، و/أو الشرى، و/أو الاغماء ، و/أو سرعة ضربات القلب، و/أو ضيق التنفس، و/أو تورم الوجه)

 

 

P

 

هذه ليست قائمة كاملة بالآثار الجانبية.  بالنسبة لأي آثار غير متوقعة تظهر أثناء تلقي زينثا، اتصل بطبيبك أو مركز علاج الهيموفيليا.

 

 

إذا أصبت بأي آثار جانبية، فتحدث إلى طبيبك، يشمل ذلك أي آثار جانبية محتملة غير مدرجة في هذه النشرة.

عن طريق الإبلاغ عن الآثار الجانبية يمكنك المساعدة في توفير المزيد من المعلومات حول سلامة هذا الدواء.

 

للإبلاغ عن أي أثر (آثار) جانبي(ة)

 

 

المركز الوطني للتيقظ والسلامة الدوائية ( NPC )

·    فاكس: 7662 205 11 966+

·    للاتصال بالمركز الوطني للتيقظ والسلامة الدوائية هاتف: 2038222 11 00966، تحويلة: 2317-2356-2353-2354-2334-2340

·    الهاتف المجاني: 8002490000

·    البريد الإلكتروني: npc.drug@sfda.gov.sa

·    الموقع الإلكتروني: www.sfda.gov.sa/npc

 

 

احفظ زينثا بعيدًا عن مرأى ومتناول الأطفال.

 

لا تستخدم زينثا بعد تاريخ انتهاء الصلاحية المدون على الكرتونة الخارجية أو الملصق على القارورة. يشير تاريخ انتهاء الصلاحية إلى آخر يوم من ذلك الشهر.

 

احفظ قارورة مسحوق زينثا المجفف بالتجميد ومحقنة المحلول المخفِّف في الثلاجة في درجة حرارة تتراوح بين 2 إلى 8 درجات مئوية.

 

يمكن تخزين زينثا في درجة حرارة غرفة لا تتجاوز 25 درجة مئوية لمدة تصل إلى 3 أشهر ولا تتجاوز تاريخ انتهاء الصلاحية. ينبغي عليك كتابة التاريخ في المساحة المتاحة على العبوة الكرتونية الخارجية عند تخزينك زينثا في درجة حرارة الغرفة لأول مرة.

بعد التخزين في درجة حرارة الغرفة، يمكن إعادة المنتج للتخزين في الثلاجة حتى يحين تاريخ انتهاء الصلاحية. لا تقم بتخزين زينثا في درجة حرارة الغرفة وإعادته إلى التخزين في الثلاجة أكثر من مرة.

 

لا تقم بتجميده.

 

تجنب تعريض قوارير زينثا للضوء.

 

لا يحتوى زينثا على مادة حافظة. استخدم المحلول المذاب في أسرع وقت ممكن بعد الخلط.  في حالة عدم استخدام المحلول على الفور، يمكن تخزينه في درجة حرارة الغرفة ويجب استخدامه خلال 3 ساعات.

 

لا تستخدم زينثا إذا لم يكن المحلول المذاب صافيًا أو مائلًا للتعكر قليلًا وعديم اللون.

 

لا تتخلص من أي أدوية عبر مياه الصرف. اسأل الصيدلي عن كيفية التخلص من الأدوية التي لم تعد تستخدمها. ستساعد هذه الإجراءات على حماية البيئة.

 

 

المكونات الدوائية:

زينثا هو منتج يحتوي على عامل التجلط الثامن، يتم إنتاجه بواسطة تكنولوجيا الحمض النووي المأشوب (دمج الجينات).

 

لا يحتوي زينثا على أي دم أو بلازما من النوع البشري، أو الألبومين، أو مواد حافظة، أو مكونات حيوانية أو بشرية مضافة في المنتج النهائي مما يجعله خاليًا بشكل طبيعي من خطر نقل فيروسات الدم مثل فيروس نقص المناعة البشرية (HIV)، وفيروسات الالتهاب الكبدي الوبائي، والفيروس الصغير.

 

المكونات غير الدوائية المهمة:

بولي سوربات 80 (0.4 مغ/القارورة)، سكروز (12 مغ/القارورة)، ل-هيستيدين (6 مغ/القارورة)، كلوريد الكالسيوم ثنائي الهيدرات (1 مغ/القارورة)، كلوريد الصوديوم (72 مغ/القارورة) [بعد التحضير باستخدام المحلول المخفِّف].

أشكال الجرعات التي يأتي بها: مجموعة أدوات قارورة زينثا: يتوفر زينثا في مجموعات أدوات تتضمن قوارير مخصصة للاستخدام مرة واحدة تحتوي على قيم اسمية تساوي 250، أو 500، أو 1000، أو 2000 وحدة دولية للقارورة ومحقنة واحدة مسبقة التعبئة من المحلول المخفِّف تحتوي على 4 مل من كلوريد الصوديوم بتركيز 0.9 % للتحضير.

 

مالك حق التسويق :

 

وايث فارما

AUTOVIA DEL NORTE A-1, KM. 23. DESVIO ALGETE, KM. 1, 28700 S. SEBASTIAN DE LOS

REYES, MADRID

,

 ، إسبانيا

 

 

 

الجهة المصنع ، تعبئة وتوزيع

 

وايث فارما ،

 

AUTOVIA DEL NORTE A-1, KM. 23. DESVIO ALGETE, KM. 1, 28700 S. SEBASTIAN DE LOS

REYES, MADRID

 ، إسبانيا

 

أبريل 2016
 Read this leaflet carefully before you start using this product as it contains important information for you

Xyntha® 250 IU powder and solvent for solution for injection Xyntha® 500 IU powder and solvent for solution for injection Xyntha® 1000 IU powder and solvent for solution for injection Xyntha® 2000 IU powder and solvent for solution for injection

Xyntha® 250 IU powder and solvent for solution for injection Each vial contains nominally 250 IU* moroctocog alfa**. After reconstitution, each mL of solution contains approximately 62.5 IU moroctocog alfa. Xyntha® 500 IU powder and solvent for solution for injection Each vial contains nominally 500 IU* moroctocog alfa**. After reconstitution, each mL of solution contains approximately 125 IU moroctocog alfa. Xyntha® 1000 IU powder and solvent for solution for injection Each vial contains nominally 1000 IU* moroctocog alfa**. After reconstitution, each mL of solution contains approximately 250 IU moroctocog alfa. Xyntha® 2000 IU powder and solvent for solution for injection Each vial contains nominally 2000 IU* moroctocog alfa**. After reconstitution, each mL of solution contains approximately 500 IU moroctocog alfa. Antihemophilic Factor (Recombinant) [BDDrFVIII] Route of Administration Dosage Form / Strength Clinically Relevant Nonmedicinal Ingredients Intravenous Infusion Available as 250, 500, 1000, or 2000 IU in single-use vials. Polysorbate 80 (0.4 mg/vial), Sucrose (12 mg/vial), L-Histidine (6 mg/vial), Calcium Chloride Dihydrate (1 mg/vial), Sodium Chloride (72 mg/vial) [after reconstitution with diluent]. Xyntha is prepared by a modified process that eliminates any exogenous human- or animal-derived protein in the cell culture process, purification, or final formulation. The purification process uses a series of chromatography steps, one of which is based on affinity chromatography using a synthetic peptide affinity ligand. The process also includes a solvent-detergent viral inactivation step and a virus-retaining nanofiltration step. The labelled potency of Xyntha is based on the European Pharmacopoeial chromogenic substrate assay, in which the Pfizer In-House Recombinant Factor VIII Potency Reference Standard has been calibrated to the WHO International Standard using the one-stage clotting assay. This method of potency assignment is intended to harmonize Xyntha with clinical monitoring using a one-stage clotting assay.

Lyophilized Powder for Reconstitution in a Vial 250, 500, 1000, or 2000 IU in single-use vials and one pre-filled diluent syringe containing 4 mL 0.9% Sodium Chloride for reconstitution*.

Xyntha, Antihemophilic Factor (Recombinant) [BDDrFVIII], indicated for the control and prevention of hemorrhagic episodes and for routine and surgical prophylaxis in patients with hemophilia A (congenital factor VIII deficiency or classic hemophilia).

Xyntha does not contain von Willebrand factor and hence is not indicated in von Willebrand’s disease.

 

Geriatrics (³ 65 years of age):

Clinical studies of Xyntha did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects.  Other reported clinical experience has not identified differences in responses between the elderly and younger patients. As with any patient receiving Xyntha, dose selection for an elderly patient should be individualized.

 

Pediatrics:

Xyntha is appropriate for use in children of all ages, including newborns.


Treatment with Xyntha   should be initiated under the supervision of a physician experienced in the treatment of hemophilia A.

 

Xyntha is appropriate for use in adults and children including newborns.

 

Dosage and duration of treatment depend on the severity of the factor VIII deficiency, the location and extent of bleeding, and the patient’s clinical condition.  Individual patients may vary in their response to factor VIII, achieving different levels of in vivo recovery and demonstrating different half-lives.  Doses administered should be titrated to the patient’s clinical response.  In the presence of an inhibitor, higher doses or appropriate alternative treatment may be required.  Dosage adjustment for patients with renal or hepatic impairment has not been studied in clinical trials.

 

The number of units of factor VIII administered is expressed in International Units (IU), which are related to the current World Health Organization (WHO) international standard for factor VIII activity.  Factor VIII activity in plasma is expressed either as a percentage (relative to normal human plasma) or in IU (relative to an International Standard for factor VIII in plasma).

 

One IU of factor VIII activity corresponds approximately to the quantity of factor VIII in one ml of normal human plasma.  The calculation of the required dosage of factor VIII is based upon the empirical finding that, on average, 1 IU of factor VIII per kg body weight raises the plasma factor VIII activity by 2 IU/dl.  The required dosage is determined using the following formula:

 

Required units = body weight (kg) x desired factor VIII rise (IU/dL or % of normal) x 0.5  (IU/kg per IU/dL)

 

Clinical data support the use of the one-stage clotting assay for monitoring Xyntha therapy.

 

The labeled potency of Xyntha is based on the European Pharmacopoeia chromogenic substrate assay in which the Pfizer In-House Recombinant Factor VIII Potency Reference Standard has been calibrated using a one-stage clotting assay.  This method of potency assignment is intended to harmonize Xyntha with clinical monitoring using a one-stage clotting assay.

 

Precise monitoring of the replacement therapy by means of plasma factor VIII activity assay should be considered, particularly for surgical intervention.

 

Dosing for Bleeding and Surgery:

In the case of the following hemorrhagic events, consideration should be given to maintaining the factor VIII activity at or above the plasma levels (in % of normal or in IU/dL) for the indicated period, as outlined in the following table.

 

Table 2: Maintenance of Factor VIII Activity for Various Hemorrhagic Events

Type of Hemorrhage

Factor VIII

Level Required (% or IU/dl)

Frequency of Doses (h)/

Duration of Therapy (d)

 

Minor

Early hemarthrosis, superficial muscle or soft tissue and oral bleeds

 

 

20-40

Repeat every 12 to 24 hours as necessary until resolved.  At least 1 day, depending upon the severity of the hemorrhage.

 

Moderate

Hemorrhages into muscles.  Mild head trauma capitus.  Minor operations including tooth extraction.  Hemorrhages into the oral cavity.

 

30-60

Repeat infusion every 12 - 24 hours for 3 - 4 days or until adequate hemostasis is achieved.  For tooth extraction a single infusion plus oral antifibrinolytic therapy within 1 hour may be sufficient. 

 

Major

Gastrointestinal bleeding.  Intracranial, intra-abdominal or intrathoracic hemorrhages. Fractures.

Major operations.

 

60-100

Repeat infusion every 8 - 24 hours until threat is resolved or in the case of surgery, until adequate local hemostasis is achieved, then continue therapy for at least another 7 days.

 

Dosage for Prophylaxis

Xyntha has been administered prophylactically in a pivotal clinical trial in adolescent and adult previously treated patients at a dose of 30 + 5 IU/kg given 3 times weekly.

 

Inhibitors

Patients using factor VIII replacement therapy should be monitored for the development of factor VIII inhibitors.  If expected factor VIII activity plasma levels are not attained, or if bleeding is not controlled with an appropriate dose, an assay should be performed to determine if a factor VIII inhibitor is present.  In patients with factor VIII inhibitors, factor VIII therapy may not be effective and other therapeutic options should be considered.  Management of such patients should be directed by physicians with experience in the care of patients with hemophilia.

 

Administration

Patients should follow the specific reconstitution and administration procedures provided by their physicians.  For instructions, patients should follow the recommendations in the below Administration and Reconstitution sections.  The procedures below are provided as general guidelines for the reconstitution and administration of Xyntha.

 

Additional instructions are provided after Infusion section that detail the use of a Xyntha vial

 

Parenteral drug products should be inspected visually for particulate matter and discoloration prior to administration, whenever solution and container permit.

 

Xyntha is administered by IV infusion after reconstitution of the lyophilized powder with the supplied pre-filled diluent (0.9% Sodium Chloride solution) syringe.

 

Reconstitution

Always wash your hands before performing the following procedures.  Use germ-free methods during the preparation procedures.

 

All components used in the mixing and injection of Xyntha should be used as soon as possible after opening their sterile containers to minimize unnecessary exposure to room air.

 

Use only the materials provided in the Xyntha kit for dissolving the Xyntha powder with the sodium chloride diluent.

 

Xyntha is administered by intravenous injection after dissolving with the supplied diluent (0.9% sodium chloride) in the pre-filled syringe.

 

Note: If you use more than one vial of Xyntha per injection, each vial should be dissolved according to the following instructions.  The empty syringe should be removed leaving the vial adapter in place, and a separate large luer lock syringe may be used to draw back the dissolved contents of each vial.  Do not detach the diluent syringes or the large luer lock syringe until you are ready to attach the large luer lock syringe to the next vial adapter.

 

 

  1. Allow the vial of freeze-dried Xyntha powder and the pre-filled diluent syringe to reach room temperature.
  2. Remove the plastic flip-top cap from the Xyntha vial to expose the central portions of the rubber stopper.

 

 

 

  1. Wipe the top of the vial with the alcohol swab provided, or use another antiseptic solution, and allow to dry.  After cleaning, do not touch the rubber stopper with your hand or allow it to touch any surface.
  2. Peel back the cover from the clear plastic vial adapter package.  Do not remove the adapter from the package.
  3. Place the vial on a flat surface.  While holding the adapter in the package, place the vial adapter over the vial.  Press down firmly on the package until the adapter snaps into place on top of the vial, with the adapter spike penetrating the vial stopper.

 

  1. Grasp the plunger rod as shown in the diagram.  Avoid contact with the shaft of the plunger rod.  Attach the threaded end of the plunger rod to the diluent syringe by pushing and turning firmly.

  1. Break off the tamper-resistant, plastic-tip cap from the diluent syringe by snapping the perforation of the cap.  This is done by bending the cap up and down until the perforation is broken.  Do not touch the inside of the cap or the syringe tip.  The diluent syringe may need to be recapped (if the dissolved Xyntha is not used immediately), so place the cap on its top on a clean surface in a spot where it would be least likely to become contaminated.

 

 

  1. Lift the package away from the adapter and discard the package.

 

  1. Place the vial on a flat surface.  Connect the diluent syringe to the vial adapter by inserting the tip of the syringe into the adapter opening while firmly pushing and turning the syringe clockwise until the connection is secured.

 

  1. Slowly depress the plunger rod to inject all the diluent into the Xyntha vial.

 

 

  1. With the syringe still connected to the adapter, gently swirl the contents of the vial until the powder is dissolved.

 

Note: The final solution should be inspected visually for particulate matter before administration.  The solution should be clear to slightly pearly and colorless.  If it is not, the solution should be discarded and a new kit should be used.

 

  1. Ensuring that the syringe plunger rod is still fully depressed, invert the vial.  Slowly draw the solution into the syringe.

 

Note: If you prepared more than one vial of Xyntha, remove the diluent syringe from the vial adapter, leaving the vial adapter attached to the vial.  Quickly attach a separate large luer lock syringe and draw back the dissolved contents as instructed above.  Repeat this procedure with each vial in turn.  Do not detach the diluent syringes or the large luer lock syringe until you are ready to attach the large luer lock syringe to the next vial adapter.

 

 

  1. Detach the syringe from the vial adapter by gently pulling and turning the syringe counterclockwise.  Discard the vial with the adapter attached.

 

Note: If the solution is not to be used immediately, the syringe cap should be carefully replaced.  Do not touch the syringe tip or the inside of the cap.

Xyntha should be infused within 3 hours after dissolving.  The dissolved solution may be stored at room temperature prior to infusion.

 

 

Infusion (Intravenous Injection)

Xyntha, when reconstituted, contains polysorbate-80, which is known to increase the rate of di-(2-ethylhexyl) phthalate (DEHP) extraction from polyvinyl chloride (PVC).  This should be considered during the preparation and administration of Xyntha, including storage time elapsed in a PVC container following reconstitution.  It is important that the recommendations in Posology and method of administration section be followed closely. 

 

Note: The tubing of the infusion set included with Xyntha vial kit does not contain DEHP.

 

You should inject Xyntha as instructed by your hemophilia doctor or nurse.  Once you learn how to self-infuse, you can follow the instructions in this insert.

 

Always wash your hands before doing the following procedures.  Germ-free methods should be used during injection.

 

Xyntha should be administered using the pre-filled diluent syringe provided or a single sterile disposable plastic luer-lock syringe.  In addition, the solution should be withdrawn from the vial using the vial adapter.

 

  1. Attach the syringe to the luer end of the provided infusion set tubing and perform venipuncture as instructed by your hemophilia doctor or nurse.
  2. Apply a tourniquet and prepare the injection site by wiping the skin well with an alcohol swab provided in the kit.

 

  1. Insert the needle on the infusion set tubing into the vein, and remove the tourniquet.  Infuse the reconstituted Xyntha product over several minutes.  Your comfort level should determine the rate of infusion.

 

 

  1. After injecting Xyntha, remove the infusion set and discard.  The amount of drug product left in the infusion set will not affect your treatment.  Dispose of all unused solution, the empty vial(s), and the used needles and syringes in an appropriate sharps container used for throwing away waste that might hurt others if not handled properly.

 

You should record the lot number of the product every time you use Xyntha.  The lot number can be found on the vial label.  The peel-off label on the vial may be used to record the lot number.

In the absence of incompatibility studies, reconstituted Xyntha should not be administered in the same tubing or container with other medicinal products.  Infusion kit components supplied in this carton are compatible with Xyntha for administration.

 

The reconstituted Xyntha solution does not contain a preservative and should be used within 3 hours of reconstitution.


Xyntha may be contraindicated in patients with a known hypersensitivity to any of the constituents of the preparation. Xyntha has not been studied in patients with a known history of hypersensitivity to hamster proteins and may be contraindicated in these patients.

Serious Warnings and Precautions

·         Anaphylaxis and severe hypersensitivity reactions are possible as with any intravenous protein product.   Should such reactions occur, treatment with the product should be discontinued and appropriate treatment should be administered.

·         Development of activity-neutralizing antibodies has been detected in patients receiving factor VIII-containing products.   If expected plasma factor VIII activity levels are not attained, or if bleeding is not controlled with an appropriate dose, an assay that measures factor VIII inhibitor concentration should be performed.

 

 

General

It is recommended that, whenever possible, every time that Xyntha is administered to patients the lot number of the product is documented.

 

Carcinogenesis and Mutagenesis

Xyntha has been shown to be nonmutagenic in the mouse micronucleus assay.  No other mutagenicity studies and no investigations on carcinogenesis, impairment of fertility or fetal development have been conducted.

 

Immune

The occurrence of neutralizing antibodies (inhibitors) is well known in patients receiving factor VIII‑containing products.  Inhibitors most commonly occur early in the treatment course of previously untreated patients, but have also been observed in patients who have previously received large amounts of factor VIII products.  All patients using coagulation factor VIII products, including Xyntha, should be monitored periodically for the development of factor VIII inhibitors.  In patients with inhibitors (especially high level inhibitors, above 5 Bethesda units (BU)/mL), factor VIII therapy may not be effective, and other therapeutic options should be considered. In addition, if expected factor VIII activity plasma levels are not attained, or if bleeding is not controlled with an appropriate dose, testing should be performed to determine if a factor VIII inhibitor is present.  Management of such patients should be directed by physicians with experience in the care of patients with hemophilia.

 

Hypersensitivity

As with any intravenous protein product, allergic type hypersensitivity reactions are possible.

Patients should be informed of the early signs or symptoms of hypersensitivity reactions including hives (rash with itching), generalized urticaria, tightness of the chest, wheezing, and hypotension.

 

If allergic or anaphylactic reactions occur, administration of Xyntha should be stopped immediately, and appropriate medical management should be given, which may include treatment for shock.  Patients should be advised to discontinue use of the product and contact their hemophelia physicians and/or seek immediate emergency care, depending on the type and severity of the reaction, if any of these symptoms occur.

 

Special Populations

 

Pediatric:

Xyntha is appropriate for use in children of all ages, including newborns.

Safety and efficacy studies have been performed both in previously treated children and adolescents (N=98, ages 7-18 years) and in previously untreated neonates, infants, and children (N=101, ages 0-52 months).  There are no clinical data of previously untreated patients (PUPs) treated with ReFacto AF or XYNTHA. An additional ongoing clinical study is evaluating the use of Xyntha in previously treated subjects under 6 years of age with moderately severe to severe hemophilia A.  

 

 

 

Geriatrics (³ 65 years of age):

Clinical studies of Xyntha did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects.  Other reported clinical experience with factor VIII products has not identified differences in responses between the elderly and younger patients.  As with any patient receiving Xyntha, dose selection for an elderly patient should be individualized.


No formal drug interaction studies have been conducted with Xyntha.  No interactions with other medicinal products are known.


Pregnancy and Lactation:

No animal reproduction and lactation studies have been conducted with Xyntha.  Based on the rare occurrence of hemophilia A in women, experience regarding the use of factor VIII during pregnancy is not available.  Xyntha should be administered to pregnant and lactating women only if the benefit outweighs the risk.


No studies on the ability to drive and use machines have been performed.


ADVERSE REACTIONS

Summary of Safety Profile

 

The most frequently reported treatment-emergent adverse reaction, on a per infusion basis, was vomiting. Most adverse reactions reported were considered mild or moderate in severity.

 

In addition, as with any intravenous protein product, allergic type hypersensitivity reactions are possible.  Manifestations of hypersensitivity reactions may include hives, generalized urticaria, tightness of the chest, wheezing, hypotension, and anaphylaxis.

 

Patients with hemophilia A may develop neutralizing antibodies (inhibitors) to factor VIII.  As with all coagulation factor VIII products, patients are to be monitored for the development of inhibitors that are quantified in Bethesda Units (BUs) using either the Bethesda assay or Bethesda assay with the Nijmegen modification. If such inhibitors occur, the condition may manifest itself as an insufficient clinical response or an unexpectedly low yield of plasma factor VIII activity.  In such cases, it is recommended that a specialized hemophilia center be contacted.

 

Reports of lack of effect, mainly in prophylaxis patients, have been received during the clinical trials and post-marketing setting.  The lack of effect and/or low factor VIII recovery has been reported in patients with inhibitors but also in patients who had no evidence of inhibitors.  The lack of effect has been described as bleeding into target joints, bleeding into new joints, other bleeding or a subjective feeling by the patient of a new onset bleeding.  In order to ensure an adequate therapeutic response, it is important TO INDIVIDUALLY TITRATE AND MONITOR each patient’s dose of Xyntha, particularly when initiating treatment with Xyntha (see Special warnings and precautions for use and Posology and method of administration).

 

If any reaction takes place that is thought to be related to the administration of Xyntha, the rate of infusion should be decreased or the infusion stopped, as dictated by the response of the patient.

 

In a clinical trial (Study 301), 32 out of 101 (32%) previously untreated patients treated with Xyntha manufactured by the previous process developed inhibitors: 16 out of 101 (16%) with a titer >5 Bethesda Units (BU) and 16 out of 101 (16%) with a titer £5 BU.  The median number of exposure days prior to inhibitor development in these patients was 12 days (range 3 - 49 days).  Of the 16 high responder patients, 15 received immune tolerance (IT) treatment.  Eleven (11) of the high responders had a titer of <0.6 BU at their latest available test after IT.  In addition, IT treatment was started in 10 of the 16 low titer (£5 BU) patients, 9 of whom had titer <0.6 BU for their latest value.  Therefore, IT had an overall efficacy of 80% (20/25), 73% for high-responders and 90% for low-responders.  Five (5) of the 6 remaining low responder patients who did not receive IT also had a titer <0.6 BU for their latest value.

 

In a clinical trial of Xyntha manufactured by the previous process, one of 113 (0.9%) previously heavily treated patients who were evaluated for efficacy in bleeding episodes  developed a high titer inhibitor.  Inhibitor development in this patient occurred in the same time frame as the development of monoclonal gammopathy of uncertain significance.  The patient was noted initially at a local laboratory to have a treatment-emergent low titer inhibitor at 98 exposure days, which was confirmed at 2 BU at the central laboratory at 113 exposure days.  After 18 months on continued treatment with Xyntha, the inhibitor level rose to nearly 13 BU and a bleeding episode failed to respond to Xyntha treatment.

 

In a pivotal phase 3 study, in which previously treated patients (PTPs) with hemophilia A received Xyntha for routine prophylaxis and on-demand treatment, 94 subjects received at least one dose of Xyntha resulting in a total of 6775 infusions.  In this study, the incidence of FVIII inhibitors to Xyntha was the primary safety endpoint.  Two patients with low titer, transient inhibitors were observed in these 94 patients (2.1%).  In a Bayesian statistical analysis, results from this study (two out of 94 subjects developed an inhibitor, 89 had 50 or more exposure days to XYNTHA) were used to update PTP results from prior supporting studies of Xyntha.  This Bayesian analysis indicates that the population (true) inhibitor rate for Xyntha was below a predefined acceptable value of 4.4%; the estimate of the 95% upper limit of the true inhibitor rate was 4.07%.

 

In a pivotal phase 3 study for surgical prophylaxis in patients with hemophilia A (study 311), one low titer persistent inhibitor and one transient false-positive inhibitor were reported.

 

There have been spontaneous postmarketing reports of high titer inhibitors developing in previously treated patients.

 

Laboratory increases in anti-FVIII antibody titers, in the absence of inhibitor development, have been observed in clinical trials.  In a study of PTPs receiving XYNTHA for routine treatment and prevention of bleeding episodes (study 310) and for surgical prophylaxis (study 311), 1 of 94 (1%) patients, and 1 of 30 (3%) patients, respectively, developed anti-FVIII antibodies; these patients did not develop an inhibitor.  The clinical significance of these antibodies, in the absence of an inhibitor, is unclear.

 

In clinical trials of PTPs receiving XYNTHA for routine treatment and prevention of bleeding episodes, 0 of 94 (0%) patients in study 310, and 3 of 110 (3%) patients in study 306/307, developed a lab increase in anti-CHO (Chinese hamster ovary, the cell line which is the source of factor VIII for XYNTHA) antibody titer, without any apparent clinical effect.  In a study of XYNTHA for surgical prophylaxis (study 311) 1 of 30 (3%) patients developed a lab increase for antibody to CHO.  Twenty of 113 (18%) previously treated patients (PTPs) had an increase in anti-CHO (Chinese hamster ovary, the cell line which is the source of factor VIII for Xyntha) antibody titer, without any apparent clinical effect.

 

 

 

Tabulated list of adverse reactions

 

The table presented below is according to the MedDRA system organ classification (SOC and Preferred Term Level). Frequencies have been evaluated according to the following convention: very common (≥ 1/10); common (≥ 1/100 to < 1/10) and uncommon (≥ 1/1,000 to < 1/100).

 

System Organ Class

(disorder)

Very Common

≥10%

Common

≥1%

Uncommon

≥0.1% and <1%

Rare

≥0.01% and <0.1%

Very Rare

<0.01%

Immune system disorders

 

 

 

 

 

Anaphylactoid reaction

Cardiac disorders

 

 

 

 

Angina pectoris, tachycardia, palpitations*

Investigations

 

 

 

Lab increase for antibody to Mouse IgG (ReFacto only),

Lab increase of FVIII antibody,

Lab increase for antibody to CHO protein,

CPK increased,

Increased aspartate aminotransferase,

Increased alanine aminotransferase*

Increased bilirubin

Nervous system

disorders

 

 

 

Headache, Dizziness

Neuropathy*,

Perspiration increased, Somnolence, Taste altered

Metabolism & nutrition disorders

 

 

 

 

Anorexia

Musculoskeletal and connective tissue disorders

 

 

 

Arthralgia

Myalgia

Vascular disorders

 

 

 

Hemorrhage, hematoma

Flushing*,

Thrombophlebitis*

Hypotension, Vasodilation

Respiratory, thoracic & mediastinal disorders

 

 

 

Cough

Dyspnea

Gastrointestinal disorders

 

 

 

Vomiting*, Nausea, Diarrhea, Abdominal pain

 

Skin and subcutaneous tissue disorders

 

 

 

Rash

Pruritis, Urticaria

General disorder & administration site conditions

 

 

 

Pyrexia, Chills, catheter site related reaction

Asthenia

Injection site pain

Injection site reaction, Injection site inflammation*

Factor VIII Inhibition†

FVIII Inhibition in PUPS

FVIII Inhibition in PTPS

 

 

 

Adverse reaction frequencies are calculated on an event per infusion basis

(*) = These adverse reactions were totaled from adverse events and hemophilia events across all studies regardless of relatedness to study drug.

All other adverse reactions were totaled across all studies from study drug-related adverse events and hemophilia events ONLY

For the adverse reaction frequencies, surgical patients receiving continuous infusion (CI), any day CI is administered is considered one infusion.

(†) = Frequency for the Adverse Reaction Factor VIII inhibition is expressed on a per patient basis

 

 

Reporting of suspected adverse reactions

Reporting suspected adverse reactions after marketing 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 according to their local country requirements.

 

To Report any Side effect(s):

 

National Pharmacovigilance Center ( NPC )

·    Fax: +966 11 205 7662

·    Call NPC at 00966 11 2038222, Exts: 2317-2356-2353-2354-2334-2340

·    Toll-Free phone: 8002490000

·    E-mail: npc.drug@sfda.gov.sa

·    Website: www.sfda.gov.sa/npc


No symptoms case of overdose have been reported with recombinant coagulation factor VIII products.

 


Drug Substance

 

Proper name:    Antihemophilic Factor (Recombinant) [BDDrFVIII]

 

Chemical name: BDDrFVIII (B domain deleted recombinant factor VIII)

 

Molecular Formula and Molecular mass: Moroctocog alfa is expressed as a single‑chain polypeptide containing 1438 amino acid residues, which is cleaved during processing to the mature, 2-chain form.  The total molecular mass including glycosylation is approximately 173 kDa.

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Structural formula: Amino Acid Sequence of Moroctocog Alfa

 
 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Product Characteristics

 

Antihemophilic Factor (Recombinant) the active ingredient in Xyntha, is a recombinant coagulation factor VIII produced by recombinant DNA technology for use in therapy of factor VIII deficiency.  Antihemophilic Factor (Recombinant) is a purified glycoprotein with an approximate molecular mass of 170 kDa consisting of 1438 amino acids.  It does not contain the B-domain, which has no known function in the circulating form of factor VIII.  The amino acid sequence of moroctocog alfa is comparable to the 90 + 80 kDa form of factor VIII.  The post-translational modifications and in vitro functional characteristics of moroctocog alfa are comparable to those of endogenous factor VIII.

 

Antihemophilic Factor (Recombinant) is secreted by a genetically engineered Chinese hamster ovary (CHO) cell line.  The CHO cell line has been extensively studied and found to be free of detectable viruses.  The cell line is grown in a chemically-defined cell culture medium that does not contain any materials derived from human or animal sources.  The purification process has been refined to affinity purify moroctocog alfa using a column chromatography method that employs a chemically synthesized affinity ligand, replacing the murine monoclonal antibody Sepharose resin and eliminating a potential risk of viral contamination associated with the murine monoclonal antibody and its manufacture, and of hypersensitivity reactions to murine protein.

 

Antihemophilic Factor (Recombinant) is inherently free from the risk of transmission of human blood-borne pathogens, such as human immunodeficiency virus (HIV), hepatitis viruses and parvovirus, because it is not purified from human blood and is manufactured from a well-characterized cell line in the absence of human- or animal-derived materials.  To further enhance the viral safety profile and provide additional assurance to the hemophilia A community, a solvent-detergent viral inactivation step and a virus-retaining nanofiltration step have been included during purification.

 

The protein is purified by a chromatography purification process that yields a high-purity, active product.  The potency expressed in international units (IU) is determined using the chromogenic assay of the European Pharmacopoeia.  The Pfizer In-House Recombinent Factor VIII Potency Reference Standard has been calibrated against the World Health Organization (WHO) International Standard for factor VIII activity using the one-stage clotting assay.  This method of potency assignment is intended to harmonize Xyntha with clinical monitoring using a one-stage clotting assay.  The specific activity of Xyntha is 5500 to 9900 IU per milligram of protein.

 

Mechanism of Action

Xyntha is a glycoprotein with an approximate molecular mass of 170 000 Da consisting of 1438 amino acids.  Xyntha is a recombinant DNA-based substance which has functional characteristics comparable to those of endogenous factor VIII.  Activated factor VIII acts as a cofactor for activated factor IX accelerating the conversion of factor X to activated factor X.  Activated factor X converts prothrombin into thrombin.  Thrombin then converts fibrinogen into fibrin which forms an insoluble clot.

 

Pharmacodynamics

 

Factor VIII is the specific clotting factor deficient in patients with hemophilia A (classical hemophilia).  The administration of Xyntha, Antihemophilic Factor (Recombinant)[BDDrFVIII] increases plasma levels of factor VIII and can temporarily correct the coagulation defect in these patients. 

 

DETAILED PHARMACOLOGY

 

Pharmacology studies of BDDrFVIII in a canine model of hemophilia A indicate that it can correct genetically deficient hemostasis.  BDDrFVIII exhibits similar interactions similar to those of plasma-derived factor VIII with von Willebrand Factor (vWF), similar cofactor activity in the activation of factor X to Xa as well as similar activation and inactivation profiles with thrombin and Protein C.

 

In a pharmacokinetic study in hemophilic dogs, elimination and half-life were similar for BDDrFVIII and plasma-derived factor VIII product (Octonativ-M).  Clearance and volume of distribution at steady state of BDDrFVIII were lower than for Octonativ-M; however, these differences were not considered to be clinically relevant.  In a pharmacokinetic study in cynomologus monkeys, the activity time profiles and pharmacokinetic parameter estimates of  [BDDrFVIII] and a single-polypeptide-chain 170 kDa form of the product were similar, indicating that any unprocessed 170 kDa protein remaining in the final product should not have any effect on the pharmacokinetics of BDDrFVIII.

 

Human

Factor VIII is the specific clotting factor deficient in patients with hemophilia A.

 

Clinical studies involving a total of 457 treated patients (317 PTPs, 101 PUPs, 5 patients participating only in PK studies and 34 patients participating only in the surgical studies) demonstrate that Xyntha can be used safely and effectively in the treatment or prevention of hemorrhage, including hemarthroses in hemophilia A patients.  Xyntha has been shown to be effective in routine prophylaxis.  In clinical trials, an average dose of 27 IU/kg (n= 85) or 30 IU/kg (n=94) in PTPs and an average dose of 50 IU/kg in PUPs (n= 45) given regularly two or more times a week prevented or reduced breakthrough spontaneous musculoskeletal bleeding episodes.  Management of hemostasis was evaluated in the surgical setting (75 surgical procedures, 64 patients).  Circulatory factor VIII levels targeted to restore and maintain hemostasis were achieved.

 

 

CLINICAL TRIALS

 

In a pivotal phase 3 study, the efficacy of Xyntha was evaluated in routine prophylaxis and on-demand treatment.  Prophylaxis was to be initiated at a dose of 30 IU/kg given 3 times per week.  The on-demand treatment dosing regimen was to be determined by the investigator.  Ninety-four (94) PTPs with moderately severe or severe hemophilia A (FVIII:C £2%) received at least 1 dose of Xyntha and were included in the intent-to-treat (ITT) population.  Eighty-nine (89) patients accrued at least 50 exposure days (EDs) to Xyntha in the study.

 

Of the 94 patients in the ITT population, 30 patients with FVIII:C £1% also participated in the double-blind, randomized, crossover PK period of the study and were included in the per‑protocol population for analyses of bioequivalence versus another rFVIII product Advate® and full pharmacokinetic characterization.  Both endpoints were surrogate markers for clinical efficacy.  The results of these analyses showed that Xyntha is bioequivalent to Advate® and the pharmacokinetic profile of Xyntha remained stable after 6 months of repeated use.

 

Intent-to-treat analysis of clinical efficacy variables in the open-label safety and efficacy period yielded similarly positive outcomes.  All 94 patients received Xyntha for routine prophylaxis; the median dose administered was 30.2 IU/kg (range, 6.8 to 76.9 IU/kg).  Most patients (57/94; 60.6%) reported no spontaneous bleeding while on routine prophylaxis.  The median annualized bleeding rate (ABR) for all bleeding episodes was 1.9 (mean 3.9, range 0 to 42.1), indicating effective prevention of bleeding in the study population.  Fifty-three (53) of 94 patients received Xyntha for on-demand treatment; the median dose administered was 30.6 IU/kg (range, 6.4 to 74.4 IU/kg).  The majority of bleeding episodes (173/187; 92.5%) resolved with 1 or 2 infusions.  This outcome was not restricted to any particular bleeding location as similar efficacy was seen in bleeding occurring in joints, soft tissues/muscles, and other sites.  A wide range of doses was used to initiate treatment of bleeding; however, the distribution of doses used to initiate treatment of bleeding was similar regardless of location of bleeding.  Patients rated the majority of infusions used to initiate treatment of bleeding as either excellent or good (132/187; 70.6%).  The incidence of less than expected therapeutic effect (LETE) occurred at a rate of 0.4% (25/6404 prophylactic infusions) when Xyntha was administered for prophylaxis and 0.5% (1/187 bleeding episodes) when administered for on-demand treatment.

 

In another series of clinical trials, the efficacy of Xyntha manufactured by the previous process was evaluated in uncontrolled phase 3 studies of 113 PTPs and 101 PUPs who received Xyntha manufactured by the previous process for on-demand treatment, routine prophylaxis, and/or surgical prophylaxis and were followed for up to 6 years.  Hemostatic efficacy was rated on an ordinal scale of excellent, good, fair, and none.

 

In 112 of 113 PTPs treated on demand, a total of 10,882 bleeding episodes were reported, with a median of 77.5 bleeding episodes per study subject.  Of these, the hemostatic efficacy of Xyntha manufactured by the previous process was assessed following the first infusion for treatment of 10,445 bleeding episodes: 9944 (95%) were rated excellent or good in their response to treatment, 429 (4%) were rated fair, and 72 (0.7%) were rated as having no response; 4% (437/10,882) of the bleeding episodes were not rated.  Of the 10,882 bleeding episodes, 7981 (73%) were managed with a single infusion, 1612 (15%) required 2 infusions, 623 (6%) required 3 infusions, and 666 (6%) required 4 or more infusions for satisfactory resolution.  The mean dose per infusion was 31 IU/kg.

 

In 100 of 101 PUPs treated on demand, a total of 2715 bleeding episodes were reported with a median of 19.5 bleeding episodes per study subject.  Of these, the hemostatic efficacy of Xyntha manufactured by the previous process was assessed following the first infusion for treatment of 2604 bleeding episodes: 2459 (94%) were rated excellent or good in their response to treatment, 142 (5%) were rated fair, and 3 (0.1%) were rated as having no response; 4% (111/2,715) of the bleeding episodes were not rated.  Of the 2715 bleeding episodes, 1794 (66%) were managed with a single infusion, 502 (19%) required 2 infusions, 229 (8%) required 3 infusions, and 190 (7%) required 4 or more infusions for satisfactory resolution.  The mean dose per infusion was 51 IU/kg.

 

Xyntha manufactured by the previous process has been studied in short-term routine prophylaxis. In uncontrolled phase 3 clinical trials, a mean dose of 27 ± 11 IU/kg per infusion in PTPs (n=85) and a mean dose of 49 ± 17 IU/kg per infusion in PUPs (n=45) was given repeatedly at variable intervals (for PTPs: median 94 weeks, range 3-296 weeks; for PUPs: median 61 weeks, range 2‑222 weeks).  In PTPs and PUPs, the mean rate of spontaneous musculoskeletal bleeding episodes was lower during periods of routine prophylaxis.  PTPs (n=85) had a mean of 10 bleeding episodes (spontaneous and injury-related) per year during the prophylactic periods compared to a mean of 25 bleeding episodes per year during on-demand periods.  PUPs (n=45) had a mean of 6 bleeding episodes (spontaneous and injury-related) per year during the prophylactic periods compared to a mean of 11 bleeding episodes per year during the on-demand periods.  These non‑randomized trial results should be interpreted with caution, as the investigators exercised their own discretion in deciding when and in whom prophylaxis was to be initiated and terminated.

 

A pivotal phase 3 study (Study 311) for surgical prophylaxis in patients with hemophilia A included PTPs with severe or moderately severe (FVIII:C≤2%) hemophilia A undergoing major surgical procedures who received XYNTHA. Thirty (30) patients were treated with XYNTHA or and comprised the ITT population; 29 patients underwent major surgery and completed the study. Thirty (30) subjects were assigned to receive XYNTHA by bolus injection (BI; 22 patients) or by continuous infusion (CI; 8 patients) at the physician’s discretion to support surgical hemostasis followed by inpatient and outpatient postoperative care.  One subject assigned to CI received XYNTHA for a pre-surgery pharmacokinetic assessment only and subsequently elected not to undergo surgery. The 22 patients treated by BI received a total of 942 infusions (ranging from 16 to 72 infusions per patient) for a cumulative total dose of 2,037,386 IU of XYNTHA over 682 cumulative total exposure days (EDs) (ranging from 15 to 40 EDs per patient).  The 8 patients assigned to treatment by CI, including 1 patient who received only 1 dose for PK assessment, received a total dose of 529,977 IU of XYNTHA over 204 total EDs (range 1 to 37 EDs per patient).  

 

Of the 29 patients who underwent surgery, 25 were included in the efficacy evaluable population. Major surgical procedures for the 25 efficacy evaluable subjects were 11 total knee replacements, 1 hip replacement, 5 synovectomies, 1 left ulnar nerve transposition release, 1 ventral hernia repair/scar revision, 1 knee arthroscopy, 1 revision and debridement of the knee after a total knee replacement, 1 hip arthroploasty revision, 1 stapes replacement, 1 ankle arthrodesis, and 1 pseudotumor excision. For the 25 surgical subjects, investigator’s ratings of the efficacy at the end of surgery were excellent for 72% (18/25) and good for 28% (7/25) of patients and at the end of the initial postoperative period were excellent for 92% (23/25) and good for 8% (2/25) of patients. Intraoperative blood loss was reported as normal or absent for all procedures. Thirteen of the 25 evaluable patients had blood loss in the postoperative period, and in 10 cases the postoperative blood loss was rated normal. In 3 cases, the postoperative blood loss was rated abnormal: 1 due to hemorrhage following surgical trauma to the epigastric artery, 1 due to an 800 mL blood loss after hip replacement surgery, and 1 after an elbow synovectomy where the blood loss could not be measured by the investigator.  

 

In a supporting study of Xyntha, there were 6 surgical procedures that were classified as major per the definitions of the pivotal surgery study.  In all cases, hemostatic efficacy was effectively managed with Xyntha.  No patient had blood loss greater than 50 mL, and no blood transfusions were given. 

 


One Stage Assay

 

In a pivotal cross-over pharmacokinetic study, Xyntha was shown to be bioequivalent to another recombinant factor VIII product (rFVIII, Advate®) in 30 previously treated patients (PTPs) (³ 12 years) using the one-stage clotting assay.  The ratios of geometric least square means of Xyntha -to-Advate® were 100%, 89.8% and 88.0% for K-value, AUCt and AUC¥, respectively.  The corresponding 90% confidence intervals about the ratios of Xyntha to Advate® geometric means were within the bioequivalence window of 80% to 125%, demonstrating bioequivalence of Xyntha to Advate®.

 

Figure -1: Xyntha vs. Advate

Xyntha

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

In the same study, the pharmacokinetic parameters for Xyntha were determined at baseline and followed-up in 25 PTPs (≥12 years) after repeated administration of Xyntha for six months.  At baseline, following a single 2-minute intravenous infusion of 50 IU/kg dose of Xyntha, plasma FVIII:C increased sharply with a mean (±SD) Cmax of 1.12 (±0.19) IU/mL.  Thereafter, the decline of FVIII:C exhibited biphasic disposition characteristics.  In the initial phase, the activity dropped at a rate consistent with relatively rapid but limited distribution into extravascular space.  The mean (±SD) steady-state volume of distribution was 65.1 (± 35.1) mL/kg.  During the terminal phase, the rate of decline in FVIII:C was slower than the initial phase with a mean (±SD) terminal elimination half-life of 11.8 (± 5.1) hours.  A comparable pharmacokinetic profile was obtained after repeated use for six months.  The ratios of geometric least square means of month 6-to-baseline pharmacokinetic were 107%, 100% and 104% for recovery, AUCt and AUC¥, respectively. No time-dependent changes in the pharmacokinetic properties of Xyntha were observed (Table 3).

                                                                                                                                          

TABLE 3 MEAN FACTOR VIII PHARMACOKINETIC PARAMETERS FOR 25 PTPS FOLLOWING A RAPID INFUSION OF XYNTHA AT A DOSE OF 50 IU/KG

Parameter

Cmax (IU/ml)

AUCT (hr*IU/ml)

Half-life (hr)

AUC¥ (hr*IU/ml)

Clearance (ml/hr/kg)

Mean Residence Time

(hr)

Vss

(ml/kg)

Recovery (IU/dl/IU/kg)

Baseline

 

 

 

 

 

 

 

 

   Mean

1.12

13.3

11.8

14.2

4.21

16.3

65.1

2.23

   SD

0.19

5.2

5.1

5.5

2.08

5.9

35.1

0.39

   Min

0.59

4.1

6.4

4.7

2.00

7.9

34.8

1.19

   Max

1.41

23.6

33.9

25.0

10.63

40.0

195.1

2.83

 

 

 

 

 

 

 

 

 

Month 6

 

 

 

 

 

 

 

 

   Mean

1.24

13.3

11.8

15.0

4.04

19.5

67.4

2.47

   SD

0.42

6.7

6.2

7.5

1.87

16.1

32.6

0.84

   Min

0.65

5.0

5.8

5.3

1.19

7.6

18.5

1.29

   Max

2.60

41.0

32.6

14.8

9.45

89.2

168.8

5.20

Abbreviations: AUC¥ = area under the plasma concentration-time curve from time zero to infinity; AUCt = area under the plasma concentration-time curve from zero to the last measurable concentration; Cmax = peak concentration; SD=standard deviation; Vss=volume of distribution at steady-state

 

In a pivotal phase III  study (Study 311) for surgical prophylaxis, XYNTHA pharmacokinetics were evaluated during the perioperative management of patients with hemophilia A who were undergoing major surgery.  At the baseline visit, all patients received a single dose of XYNTHA of approximately 50 IU/kg. Plasma samples were analyzed for FVIII activity using a validated one-stage (OS) clotting method.   Recovery data are available for a total of 30 patients; the mean (± standard deviation [SD]) K-value was 2.11(± 0.43) IU/dL per IU/kg, and the mean (±SD) in vivo recovery value was 101.0% (± 20%).

 

Chromogenic Assay

 

The labeled potency of Xyntha manufactured by the previous process is based on the European Pharmacopoeia chromogenic substrate assay in which the Pfizer In-House Recombinant Factor VIII Potency Reference Standard has been calibrated to the WHO International Standard using the chromogenic substrate assay.

 

In a crossover pharmacokinetic study of eighteen (18) previously treated patients using the chromogenic assay, the circulating mean half-life for Xyntha manufactured by the previous process was 14.8 ± 5.6 hours (ranged from 7.6 - 28.5 hours), which was not statistically significantly different from plasma-derived Antihemophilic Factor (Human), (pdAHF), which had a mean half-life of 13.7 ± 3.7 hours  (ranged from 8.8 - 25.1 hours).  Mean incremental recovery (K-value) of Xyntha manufactured by the previous process in plasma was 2.4 ± 0.4 IU/dL per IU/kg (ranged from 1.9 - 3.3 IU/dL per IU/kg).  This was comparable to the mean incremental recovery observed in plasma for pdAHF which was 2.3 ± 0.3 IU/dL per IU/kg (ranged from 1.7 - 2.9 IU/dL per IU/kg).

 

In additional clinical studies using Xyntha manufactured by the previous process, pharmacokinetic parameters measured using the chromogenic assay were determined for previously treated patients (PTPs) and previously untreated patients (PUPs).  In PTPs (n=101; median age 26 ± 12 years), Xyntha manufactured by the previous process had a recovery at Week 0 of 2.4 ± 0.4 IU/dl per IU/kg (range 1.1 to 3.8 IU/dl per IU/kg).  In measurements over 4 years of use (Month 3 [n=90], Month 6 [n=87], Month 12 [n=88], Month 24 [n=70], Month 36 [n=64] and Month 48 [n=52]), the mean incremental recovery was reproducible and ranged from 2.3 to 2.5 IU/dl per IU/kg.  A subset of 37 study subjects had evaluable pharmacokinetic profiles at both baseline and Month 12.  The 90% confidence intervals for the ratios of the mean values of Month 12-to-baseline AUCT, AUC¥, and K-value were well within the bioequivalence window of 80% to 125%, demonstrating the stability of these pharmacokinetic parameters over 1 year.  In PUPs (n=59; median age 10 ± 8.3 months), Xyntha manufactured by the previous process had a mean recovery at Week 0 of 1.5 ± 0.6 IU/dl per IU/kg (range 0.2 to 2.8 IU/dl per IU/kg).  The mean incremental recovery for PUPs was stable over time (5 visits during a 2-year period) and ranged from 1.5 to 1.8 IU/dL per IU/kg of Xyntha manufactured by the previous process.  Population pharmacokinetic modeling using data from 44 PUPs led to a mean estimated half-life of Xyntha manufactured by the previous process in PUPs of 8.0 ± 2.2 hours.

 

Table 4: Mean Factor VIII Pharmacokinetic Parameters for 37 PTPS with Both Baseline and Month 12 Pharmacokinetic Profiles Following A Rapid Infusion of Xyntha manufactured by the previous process at a Dose of 50 IU/KG

Parameter

Cmax (IU/ml)

AUCT (hr*IU/ml)

Half-life (hr)

AUC¥ (hr*IU/ml)

Clearance (ml/hr/kg)

Mean Residence Time

(hr)

Vss

(ml/kg)

K-value (IU/dl/IU/kg)

Baseline

 

 

 

 

 

 

 

 

   Mean

1.17

13.6

10.6

15.4

3.53

15.0

50.9

2.34

   SD

0.24

3.4

2.5

4.5

1.03

3.4

13.0

0.49

   Min

0.55

6.0

6.8

7.6

1.78

9.8

36.9

1.10

   Max

1.90

21.1

17.2

28.1

6.60

24.7

99.0

3.80

Parameter

Cmax (IU/ml)

AUCT (hr*IU/ml)

Half-life (hr)

AUC¥ (hr*IU/ml)

Clearance (ml/hr/kg)

Mean Residence Time

(hr)

Vss

(ml/kg)

K-value (IU/dl/IU/kg)

Month 12

 

 

 

 

 

 

 

 

  Mean

1.20

14.0

11.4

16.5

3.37

16.1

51.1

2.40

  SD

0.29

4.7

3.5

5.7

1.08

4.6

11.4

0.58

  Min

0.84

7.8

6.6

8.8

1.49

9.7

21.3

1.67

  Max

2.31

32.4

20.1

33.5

5.66

27.8

83.2

4.61

 

 


TOXICOLOGY

 

Single Dose Toxicity

BDDrFVIII was tested for potential toxicity of a single intravenous (IV) dose in Sprague-Dawley rats and cynomologus monkeys.  

 

Sprague-Dawley rats (5/sex/group) were administered single doses of 0 (saline), 2500 or 10000 IU/kg of BDDrFVIII intravenously.  Animals were observed for 14 days and sacrificed for gross pathology and histopathology.  There were no adverse effects related to BDDrFVIII administration; therefore, the no-observed-adverse-effect-level (NoAEL) was ≥ 10000 IU/kg.

 

Two groups of cynomologus monkeys (1/sex/group) were administered alternate escalating single doses of 200, 400, 800, or 1600 IU/kg BDDrFVIII intravenously every third day in a dose-range finding toxicology study.  The animals were subsequently allowed a 14-day washout period and used for a 7-day repeat dose study described below.  As expected, there were transient dose-dependent increases in plasma factor VIII activity.  During the single dose escalation study there were no adverse effects associated with BDDrFVIII administration; therefore, the NoAEL was ≥ 1600 IU/kg. 

 

Repeated Dose Toxicity

BDDrFVIII was tested for potential toxicity of repeated IV doses in Sprague-Dawley rats and cynomologus monkeys.

 

Rat Studies

Sprague-Dawley rats (4/sex/group) were administered doses of 0 (vehicle), 200, 400, 800, or 1600 IU/kg/day BDDrFVIII intravenously once daily for 10 or 11 days.  Satellite groups of 10 males each were included at dose levels of 200 and 800 IU/kg/day to monitor for antibodies against BDDrFVIII at 4 and 19 days after the last dose administration.  There were significant BDDrFVIII antibody responses in all animals from the satellite treatment groups as expected when administering a human protein to animals.  There were no adverse effects associated with BDDrFVIII administration; therefore, the NoAEL was ≥ 1600 IU/kg/day.

 

The general toxicity of BDDrFVIII was evaluated in a 4-week study in Sprague-Dawley rats.  Animals (10/sex/group) were administered 0 (vehicle), 50, 250, or 1250 IU/kg/day BDDrFVIII intravenously once daily for 4 weeks.  In addition to standard comprehensive assessments of clinical, clinical laboratory, and anatomic pathology,  BDDrFVIII antibody responses were assessed at the end of the treatment period in a subset of the animals.  There was a dose-related induction of antibodies reactive to BDDrFVIII in the treated animals.  There were slight increases in APTT in a small number of animals from the 250 and 1250 IU/kg/day groups.  These minor elevations in APTT were considered secondary to the anti- BDDrFVIII antibody responses which apparently have the potential for neutralization of exogenous human recombinant and endogenous rat factor VIII activity.  This neutralization results in prolongation of APTT which is an ex vivo measure of the intrinsic and common pathways of coagulation.  There were no adverse effects associated with BDDrFVIII administration; therefore, the NoAEL was ≥ 1250 IU/kg/day. 

 

Cynomologus Monkey Studies

A 7-day repeat dose toxicity study in cynomologus monkeys was performed using the same animals as described in the single-dose escalation study with 4 additional naive animals.  After a 14-day washout period, 0 (vehicle) (1/sex), 800 (2/sex), or 1250 (2/sex) IU/kg/day BDDrFVIII was administered intravenously once daily for 7 days.  In addition to standard clinical laboratory and anatomic pathology assessments, animals were monitored for plasma factor VIII activity. Three of four previously treated animals had decreased plasma factor VIII activity on Day 7 compared to previously untreated animals suggesting the appearance of antibody responses resulting in neutralization of both exogenous recombinant human and endogenous factor VIII activity.  Antibodies to BDDrFVIII were not measured in this study.  There were no adverse effects associated with BDDrFVIII administration; therefore, the NoAEL was ≥ 1250 IU/kg/day. 

 

The general toxicity of BDDrFVIII was evaluated in a 4-week study in cynomologus monkeys (3/sex/group) at doses of 0 (vehicle), 50, 250, or 1250 IU/kg/day BDDrFVIII intravenously.  In addition to comprehensive clinical, clinical laboratory, and postmortem examinations, plasma factor VIII activity was assessed prestudy and on Days 13, 20, and 28, and anti-BDDrFVIII antibodies and neutralizing activity were determined prestudy and on Day 28.  One animal was found dead and three sacrificed when moribund.  These deaths occurred on or shortly after days of scheduled bleeds (Days 20 and 28/29), and in each case the predominant signs were hemorrhage at sites of venipuncture and/or marked anemia.  Clinical signs related to treatment were extensive hemorrhage and bruising at sites of venipuncture.  Body weight losses were observed in two moribund animals, and reduced food intake was observed in one moribund animal.  Dose and time-related appearance of anti-BDDrFVIII antibodies were associated with prolongation of APTT, decreases in red blood cell indices (PCV, Hb, and RBC count) and decreased plasma factor VIII activity in animals from the 250 and 1250 IU/kg/day groups. 

 

Treatment-related gross lesions were limited to hemorrhage at sites of venipuncture and at scattered other locations.  These changes were observed predominantly in animals from the 250 and 1250 IU/kg/day groups.  Histologic changes related to treatment included hemorrhage at sites of venipuncture and in various other organs including the heart, subcutaneous tissues, urinary bladder, spinal canal, skeletal muscle, gastrointestinal tract, and connective tissues.  The heart was apparently a predisposition site for histologic lesions of hemorrhage and edema with secondary inflammation and early fibrosis and, sometimes, associated regions of myocardial degeneration. 

 

All adverse effects observed in this study were considered related to the neutralizing antibody response to both administered recombinant human and endogenous monkey factor VIII.  Neutralizing antibody responses resulted in an acquired hemophilia syndrome and predisposition to multiorgan hemorrhage and the sequelae of death, moribundity, anemia, and hemorrhage-induced inflammation.  The NoAEL for changes related to this syndrome was 50 IU/kg/day, but no adverse effects unrelated to the immunogenicity of BDDrFVIII were observed at any dose level.

 

After completing the 4-week IV toxicity study with BDDrFVIII in cynomologus monkeys, it was considered appropriate to conduct a similar study with a plasma-derived human factor VIII product, Octonativ-M®, in order to demonstrate the comparability of the changes observed with BDDrFVIII to those with a plasma-derived factor VIII product.  Cynomologus monkeys were administered daily doses of 0 (vehicle) (2/sex), 250 or 1250 (3/sex/group) IU/kg/day of Octonativ-M® intravenously for 5 weeks.  Parameters examined were similar to those in the BDDrFVIII 4-week study described above. 

 

One animal from the 250 IU/kg/day group was sacrificed moribund on Day 29.  Clinical signs related to treatment were hemorrhage at sites of venipuncture.  Anti-factor VIII antibodies were detected in all animals treated with Octonativ-M®.  Clinical laboratory changes included dose- and time-related increases in APTT, decreased red cell indices, increased fibrinogen, and decreased erythrocyte sedimentation rates.  The longitudinal kinetics of onset of these changes correlated with dose- and time-related appearance of decreased plasma factor VIII activity and the appearance of plasma factor VIII inhibitors.  Treatment-related postmortem findings in animals from both treatment groups included gross hemorrhage at sites of venipuncture and in the heart.  These changes correlated with microscopic lesions of hemorrhage and edema, with associated inflammation and early fibrosis and myocyte degeneration.

 

Overall, changes observed in this study with a plasma-derived human factor VIII product were analogous to those seen with BDDrFVIII.  All adverse effects were considered secondary to the immunogenicity of the test molecule and the acquired hemophilic state of the animals.  These effects occurred at IV doses of 250 and 1250 IU/kg/day.  There was no NoAEL identified in this study of Octonativ-M®.

 

A 6-week multidose subcutaneous (SC) and IV antigenicity study of BDDrFVIII, which included limited toxicology endpoints, was performed in cynomologus monkeys.  In this study, animals were administered 0 (vehicle) (3/SC route only), 50 (6/route), or 250 IU/kg (4/route) BDDrFVIII either IV or SC every other day for 6 weeks.  Limited clinical,

 

hematologic, clinical chemistry, plasma factor VIII activity, factor VIII antibody, and plasma inhibitor determinations were performed.  Both SC and IV administration resulted in antibody formation (2/6 animals in the low dose IV treatment group and 100% of the animals in all other groups).  Inhibitor antibodies were detected in some animals after treatment by either route of administration (2/6 animals in the low dose IV-treated group, 4/4 animals in the high dose IV group, 1/6 animals in the low dose SC-treated group, and 2/4 animals in the high dose SC group).  Gross necropsy and limited histopathology evaluations were performed.  Two deaths were observed in the high dose IV administration group.  These were considered secondary to hemorrhage and severe anemia.  There were dose- and time-related increases in APTT and decreases in red blood cell indices by both routes.  There were clinical and postmortem observations of hemorrhage at venipuncture sites.  Histologic changes of ischemic degeneration in the heart were seen in one animal from the 50 IU/kg IV group and one animal from the 250 IU/kg IV group.  These changes were all considered related to the immunogenicity of BDDrFVIII in cynomologus monkeys, the acquired hemophilic syndrome, and resultant hemorrhagic anemia and myocardial ischemia.  There was no NoAEL identified in this study.

 

Reproductive Toxicity

No specific studies to investigate the potential effects of BDDrFVIII on reproductive or developmental function have been conducted.  In the 4-week repeat dose toxicity studies, gonadal and secondary sex organs were examined by gross and histopathology, and there were no apparent effects of  BDDrFVIII on these tissues.

 

Mutagenic Potential

BDDrFVIII was assessed for the potential to induce bone marrow micronuclei in vivo in the CD-1 mouse.  Animals (10/sex/group) were administered 0 (vehicle), 2490, 4980, or 9960 IU/kg/day BDDrFVIII IV for two consecutive days (Tabular Format and Kabi Pharmacia Report 9296824).  Five animals/sex/group were sacrificed 24 and 48 hours after the second dose administration.  It was concluded that BDDrFVIII did not induce micronuclei in the polychromatic erythrocytes of the bone marrow of mice at any dose level tested.

 

Repeat Dose

As part of the nonclinical evaluation of Xyntha, a 4-week toxicity study was conducted in cynomologus monkeys with Xyntha administered by intravenous (IV) injection at dosages of 0, 50, or 1250 IU/kg/day once daily for 29 or 30 days.  The toxicity profile from this study was compared with that obtained in a previously conducted 4-week toxicity study in cynomologus monkeys with original BDDrFVIII administered by IV injection at dosages of 0, 50, 250, and 1250 IU/kg/day. 

 

The administration of Xyntha to monkeys at an IV dosage of 1250 IU/kg/day for 29 or 30 days was generally well tolerated prior to the formation of anti‑Xyntha antibodies.  The immunologic response against Xyntha consisted of anti- Xyntha antibody production and increased FVIII inhibitors, which resulted in decreased FVIII activity that caused impairment of the coagulation pathway.  These findings correlated with clinical pathology changes, hemorrhage, and tissue changes secondary to hemorrhage and were also observed in a previously conducted 4-week toxicity study in cynomologus monkeys with BDDrFVIII.

 

The most significant hematologic alteration was a dose‑dependent increase in activated partial thromboplastin time (APTT) that was also observed in monkeys at 250 and 1250 IU/kg/day in the previous 4-week toxicity study with original BDDrFVIII.  Absolute and relative (to body and brain) mean liver weights were increased (21% to 25%, compared to controls) in male monkeys given 1250 IU/kg/day.  Individual liver weights were 74.9, 72.4, 57.6 G (absolute), 99.5, 102.0, 88.2 (% relative to brain weight) and 1.9, 2.1, 1.6 (% relative to body weight) in control males and 80.8, 79.6, 88.5 G (absolute), 123.2, 121.9, 116.8 (% relative to brain weight) and 2.4, 2.3, 2.3 (% relative to body weight) in males given 1250 IU/kg/day.  The weight increase was not considered to be toxicologically significant due to the low magnitude of change and the lack of macroscopic or microscopic correlates.

 

Local Tolerance Studies

A separate local tolerance study was not conducted with Xyntha.  Instead, local injection sites were evaluated macroscopically and microscopically in the repeat-dose study conducted in monkeys.  Macroscopically, red discoloration occurred at the injection sites of all monkeys regardless of treatment (including controls).  Microscopically, slight to moderate perivascular and vascular neutrophilic inflammation and/or fibrosis often accompanied hemorrhage at the injection sites in animals given Xyntha, as well as in controls.  Hemorrhage was more severe in monkeys given Xyntha (slight to marked at 50 IU/kg/day and mild to severe at 1250 IU/kg/day) when compared with controls (mild to moderate).  The macroscopic and microscopic observations with Xyntha were similar to findings in the previous 4‑week toxicity study in cynomologus monkeys conducted with original BDDrFVIII. 

 

Other Toxicity Studies

Because a novel affinity ligand (TN8.2) is used in the purification process for Xyntha, a non‑GLP-compliant, single‑dose toxicity study was conducted in rats to assess the acute toxicity of TN8.2 in the event that the peptide leached from the chromatographic resin into the product stream during purification.  Because TN8.2 binds to FVIII, in vitro studies were also conducted to evaluate the possible effects of TN8.2 on the clotting activity of rat plasma, human plasma, or Xyntha.  TN8.2 has not been detected in any batch of drug substance tested to date.

 

Single-dose administration of the TN8.2 affinity ligand in rats at 0.6 mg/kg was well tolerated and did not induce acute toxicity.


Clinically Relevant Nonmedicinal Ingredients

 

Polysorbate 80 (0.4 mg/vial), Sucrose (12 mg/vial), L-Histidine (6 mg/vial), Calcium Chloride Dihydrate (1 mg/vial), Sodium Chloride (72 mg/vial) [after reconstitution with diluent].


In the absence of incompatibility studies, reconstituted Xyntha should not be administered in the same tubing or container with other medicinal products.  Infusion kit components supplied in this carton are compatible with Xyntha for administration.

 


3 years

Keep out of the sight and reach of children.

 

 

Xyntha Antihemophilic Factor (Recombinant) should be stored under refrigeration at a temperature of 2° to 8°C.  Xyntha vial may also be stored at room temperature not to exceed 25°C for up to 3 months.  The diluent syringe should be stored at 2° to 25°C and should not be used subsequent to expiration of the Xyntha drug product.  The patient should write in the space provided on the outer carton the date the product was placed at room temperature.  After room temperature storage, the product can be returned to refrigerated storage until the expiration date. Do not store Xyntha vial at room temperature and return it to refrigerated storage more than once.  Do not use Xyntha vial after the expiry date on the label.

 

Product after reconstitution:  The reconstituted solution may be stored at room temperature prior to administration.  The product does not contain a preservative and should be used within 3 hours.


Xyntha, Antihemophilic Factor (Recombinant) freeze-dried is supplied in kits that include single-use vials that contain nominally 250, 500, 1000, or 2000 IU per vial.  Actual factor VIII activity in IU is stated on the label of each Xyntha Antihemophilic Factor (Recombinant) vial.

 

In addition, each Xyntha Antihemophilic Factor (Recombinant) kit contains: one pre-filled diluent syringe containing 4 mL 0.9% Sodium Chloride with plunger rod for assembly, one vial adapter, one sterile infusion set, two alcohol swabs, one bandage, one gauze, and one package insert.


Freezing should be avoided to prevent damage to the pre-filled diluent syringe.  During storage, avoid prolonged exposure of Xyntha vial to light.

 

 

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.

 


MARKET AUTHORIZATION HOLDER: WYETH FARMA S.A. AUTOVIA DEL NORTE A-1, KM. 23. DESVIO ALGETE, KM. 1, 28700 S. SEBASTIAN DE LOS REYES, MADRID, SPAIN MANUFACUTRED, PACKED & RELEASED BY WYETH FARMA S.A. AUTOVIA DEL NORTE A-1, KM. 23. DESVIO ALGETE, KM. 1, 28700 S. SEBASTIAN DE LOS REYES, MADRID SPAIN

April 2016
}

صورة المنتج على الرف

الصورة الاساسية