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)

Valtroza contains the active substance bortezomib, a so-called ‘proteasome inhibitor’. Proteasomes play
an important role in controlling cell function and growth. By interfering with their function, bortezomib
can kill cancer cells.
Valtroza is used for the treatment of multiple myeloma (a cancer of the bone marrow) in patients older
than 18 years:
- alone or together with the medicines pegylated liposomal doxorubicin or dexamethasone, for
patients whose disease is worsening (progressive) after receiving at least one prior treatment and for
whom blood stem cell transplantation was not successful or is unsuitable.
- in combination with the medicines melphalan and prednisone, for patients whose disease has not
been previously treated and are unsuitable for high-dose chemotherapy with blood stem cell
transplantation.
- in combination with the medicines dexamethasone or dexamethasone together with thalidomide, for
patients whose disease has not been previously treated and before receiving high-dose
chemotherapy with blood stem cell transplantation (induction treatment).
Valtroza is used for the treatment of mantle cell lymphoma (a type of cancer affecting the lymph nodes)
in patients 18 years or older in combination with the medicines rituximab, cyclophosphamide,
doxorubicin and prednisone, for patients whose disease has not been previously treated and for whom
blood stem cell transplantation is unsuitable.


Do not use Valtroza
- if you are allergic to bortezomib, boron or any of the other ingredients of this medicine (listed in
section 6).
- if you have certain severe lung or heart problems.
Warnings and precautions
You should tell your doctor if you have any of the following:
• low numbers of red or white blood cells
• bleeding problems and/or low number of platelets in your blood
• diarrhoea, constipation, nausea or vomiting
• fainting, dizziness or light-headedness in the past
• kidney problems
• moderate to severe liver problems
• numbness, tingling, or pain in the hands or feet (neuropathy) in the past
• heart or blood pressure problems
• shortness of breath or cough
• seizures
• shingles (localised including around the eyes or spread across the body)
• symptoms of tumor lysis syndrome such as muscle cramping, muscle weakness, confusion, visual
loss or disturbances and shortness of breath
• memory loss, trouble thinking, difficulty with walking or loss of vision. These may be signs of a
serious brain infection and your doctor may suggest further testing and follow-up.
You will have to take regular blood tests before and during your treatment with Valtroza, to check your
blood cell counts regularly.
If you have mantle cell lymphoma and are given the medicine rituximab with Valtroza you should tell
your doctor:
• if you think you have hepatitis infection now or have had it in the past. In a few cases, patients
who have had hepatitis B might have a repeated attack of hepatitis, which can be fatal. If you have
a history of hepatitis B infection you will be carefully checked by your doctor for signs of active
hepatitis B.
You must read the package leaflets of all medicinal products to be taken in combination with Valtroza
for information related to these medicines before starting treatment with Valtroza. When thalidomide is
used, particular attention to pregnancy testing and prevention requirements are needed (see Pregnancy
and breast-feeding in this section).
Children and adolescents
Valtroza should not be used in children and adolescents because it is not known how the medicine will
affect them.
Other medicines and Valtroza
Please tell your doctor or pharmacist if you are taking, have recently taken or might take any other
medicines.
In particular, tell your doctor if you are using medicines containing any of the following active
substances:

- ketoconazole, used to treat fungal infections
- ritonavir, used to treat HIV infection
- rifampicin, an antibiotic used to treat bacterial infections
- carbamazepine, phenytoin or phenobarbital used to treat epilepsy
- St. John’s Wort (Hypericum perforatum), used for depression or other conditions
- oral antidiabetics
Pregnancy and, breast-feeding
You should not use Valtroza if you are pregnant, unless clearly necessary.
Both men and women receiving Valtroza must use effective contraception during and for up to 3
months after treatment. If, despite these measures, pregnancy occurs, tell your doctor immediately.
You should not breast-feed while using Valtroza. Discuss with your doctor when it is safe to restart
breast-feeding after finishing your treatment.
Thalidomide causes birth defects and foetal death.When Valtroza is given in combination with
thalidomide you must follow the pregnancy prevention programme for thalidomide (see package leaflet
for thalidomide).
Driving and using machines
Valtroza might cause tiredness, dizziness, fainting, or blurred vision. Do not drive or operate tools or
machines if you experience such side effects; even if you do not, you should still be cautious.


Your doctor will work out your dose of Valtroza according to your height and weight (body surface
area). The usual starting dose of Valtroza is 1.3 mg/m2 body surface area twice a week.
Your doctor may change the dose and total number of treatment cycles, depending on your response to
the treatment, on the occurrence of certain side effects and on your underlying conditions (e.g. liver
problems).
Progressive multiple myeloma
When Valtroza is given alone, you will receive 4 doses of Valtroza intravenously or subcutaneously on
days 1, 4, 8 and 11, followed by a 10-day ‘rest period’ without treatment. This 21-day period (3 weeks)
corresponds to one treatment cycle. You might receive up to 8 cycles (24 weeks).
You may also be given Valtroza together with the medicines pegylated liposomal doxorubicin or
dexamethasone.
When Valtroza is given together with pegylated liposomal doxorubicin, you will receive Valtroza
intravenously or subcutaneously as a 21-day treatment cycle and pegylated liposomal doxorubicin 30
mg/m2 is given on day 4 of the Valtroza 21-day treatment cycle as an intravenous infusion after the
Valtroza injection.
You might receive up to 8 cycles (24 weeks).
When Valtroza is given together with dexamethasone, you will receive Valtroza intravenously and
subcutaneously as a 21-day treatment cycle and dexamethasone 20 mg is given orally on days 1, 2, 4, 5,
8, 9, 11, and 12, of the Valtroza , 21-day treatment cycle.
You might receive up to 8 cycles (24 weeks).Previously untreated multiple myeloma
If you have not been treated before for multiple myeloma, and you are not suitable for blood stem cell
transplantation you will receive Valtroza together with two other medicines; melphalan and prednisone.
In this case, the duration of a treatment cycle is 42 days (6 weeks). You will receive 9 cycles (54
weeks).
• In cycles 1 to 4, Valtroza is administered twice weekly on days 1, 4, 8, 11, 22, 25, 29 and 32.
• In cycles 5 to 9, Valtroza is administered once weekly on days 1, 8, 22 and 29.
Melphalan (9 mg/m2) and prednisone (60 mg/m2) are both given orally on days 1, 2, 3 and 4 of the first
week of each cycle.
If you have not been treated before for multiple myeloma, and you are suitable for blood stem cell
transplantation you will receive Valtroza intravenously or subcutaneously together with the medicines
dexamethasone, or dexamethasone and thalidomide, as induction treatment.
When Valtroza is given together with dexamethasone, you will receive Valtroza intravenously or
subcutaneously as a 21-day treatment cycle and dexamethasone 40 mg is given orally on days 1, 2, 3, 4,
8, 9, 10 and 11 of the Valtroza 21-day treatment cycle.
You will receive 4 cycles (12 weeks).
When Valtroza is given together with thalidomide and dexamethasone, the duration of a treatment cycle
is 28 days (4 weeks).
Dexamethasone 40 mg is given orally on days 1, 2, 3, 4, 8, 9, 10 and 11 of the Valtroza 28-day
treatment cycle and thalidomide is given orally daily at 50 mg up to day 14 of the first cycle, and if
tolerated the thalidomide dose is increased to 100 mg on days 15-28 and may be further increased to
200 mg daily from the second cycle onwards.
You might receive up to 6 cycles (24 weeks).

Previously untreated mantle cell lymphoma
If you have not been treated before for mantle cell lymphoma you will receive Valtroza intravenously or
subcutaneously together with the medicines rituximab, cyclophosphamide, doxorubicin and prednisone.
Valtroza is given intravenously or subcutaneously on days 1, 4, 8 and 11, followed by a ‘rest period’
without treatment. The duration of a treatment cycle is 21 days (3 weeks). You might receive up to 8
cycles (24 weeks).
The following medicinal products are given on day 1 of each Valtroza 21-day treatment cycle as
intravenous infusions:
Rituximab at 375 mg/m2, cyclophosphamide at 750 mg/m2 and doxorubicin at 50 mg/m2.
Prednisone is given orally at 100 mg/m2 on days 1, 2, 3, 4 and 5 of the Valtroza treatment cycle.
How Valtroza is given
This medicine is for intravenous or subcutaneous use. Valtroza will be administered by a healthcare
professional experienced in the use of cytotoxic medicines.
Valtroza powder has to be dissolved before administration. This will be done by a healthcare
professional. The resulting solution is then either injected into a vein or under the skin. Injection into a
vein is rapid, taking 3 to 5 seconds. Injection under the skin is in either the thighs or the abdomen.

If you are given too much Valtroza
As this medicine is being given by your doctor or nurse, it is unlikely that you will be given too much.
In the unlikely event of an overdose, your doctor will monitor you for side effects.


Like all medicines, this medicine can cause side effects, although not everybody gets them. Some of
these effects may be serious.
If you are given Valtroza for multiple myeloma or mantle cell lymphoma, tell your doctor straight away
if you notice any of the following symptoms:
− muscle cramping, muscle weakness
− confusion, visual loss or disturbances, blindness, seizures, headaches
− shortness of breath, swelling of your feet or changes in your heart beat, high blood pressure,
tiredness, fainting
− coughing and breathing difficulties or tightness in the chest.
Treatment with Valtroza can very commonly cause a decrease in the numbers of red and white blood
cells and platelets in your blood. Therefore, you will have to take regular blood tests before and during
your treatment with Valtroza, to check your blood cell counts regularly. You may experience a
reduction in the number of:
− platelets, which may make you be more prone to bruising, or to bleeding without obvious injury
(e.g., bleeding from your bowels, stomach, mouth and gum or bleeding in the brain or bleeding
from the liver)
− red blood cells, which can cause anaemia, with symptoms such as tiredness and paleness
− white blood cells may make you more prone to infections or flu-like symptoms.
If you are given Valtroza for the treatment of multiple myeloma the side effects you may get are listed
below:
Very common side effects (may affect more than 1 in 10 people)
• Sensitivity, numbness, tingling or burning sensation of the skin, or pain in the hands or feet, due
to nerve damage
• Reduction in the number of red blood cells and or white blood cells (see above)
• Fever
• Feeling sick (nausea) or vomiting, loss of appetite
• Constipation with or without bloating (can be severe)
• Diarrhoea: if this happens, it is important that you drink more water than usual. Your doctor may
give you another medicine to control diarrhoea
• Tiredness (fatigue), feeling weak
• Muscle pain, bone pain
Common side effects (may affect up to 1 in 10 people)
• Low blood pressure, sudden fall of blood pressure on standing which may lead to fainting
• High blood pressure
• Reduced functioning of your kidneys
• Headache
• General ill feeling, pain, vertigo, light-headedness, a feeling of weakness or loss of consciousness
• Shivering
• Infections, including pneumonia, respiratory infections, bronchitis, fungal infections, coughing
with phlegm, flu like illness

• Shingles (localised including around the eyes or spread across the body)
• Chest pains or shortness of breath with exercise
• Different types of rash
• Itching of the skin, lumps on the skin or dry skin
• Facial blushing or tiny broken capillaries
• Redness of the skin
• Dehydration
• Heartburn, bloating, belching, wind, stomach pain, bleeding from your bowels or stomach
• Alteration of liver functioning
• A sore mouth or lip, dry mouth, mouth ulcers or throat pain
• Weight loss, loss of taste
• Muscle cramps, muscle spasms, muscle weakness, pain in your limbs
• Blurred vision
• Infection of the outermost layer of the eye and the inner surface of the eyelids (conjunctivitis)
• Nose bleeds
• Difficulty or problems in sleeping, sweating, anxiety, mood swings, depressed mood, restlessness
or agitation, changes in your mental status, disorientation
• Swelling of body, to include around eyes and other parts of the body
Uncommon side effects (may affect up to 1 in 100 people)
• Heart failure, heart attack, chest pain, chest discomfort, increased or reduced heart rate
• Failing of your kidneys
• Inflammation of a vein, blood clots in your veins and lungs
• Problems with blood clotting
• Insufficient circulation
• Inflammation of the lining around your heart or fluid around your heart
• Infections including urinary tract infections, the flu, herpes virus infections, ear infection and
cellulitis
• Bloody stools, or bleeding from mucosal membranes, e.g., mouth, vagina
• Cerebrovascular disorders
• Paralysis, seizures, falling, movement disorders, abnormal or change in, or reduced sensation
(feeling, hearing, tasting, smelling), attention disturbance, trembling, twitching
• Arthritis, including inflammation of the joints in the fingers, toes, and the jaw
• Disorders that affect your lungs, preventing your body from getting enough oxygen. Some of these
include difficulty breathing, shortness of breath, shortness of breath without exercise, breathing that
becomes shallow, difficult or stops, wheezing
• Hiccups, speech disorders
• Increased or decreased urine production (due to kidney damage), painful passing of urine or
blood/proteins in the urine, fluid retention
• Altered levels of consciousness, confusion, memory impairment or loss
• Hypersensitivity
• Hearing loss, deafness or ringing in the ears, ear discomfort
• Hormone abnormality which may affect salt and water absorption
• Overactive thyroid gland
• Inability to produce enough insulin or resistance to normal levels of insulin
• Irritated or inflamed eyes, excessively wet eyes, painful eyes, dry eyes, eye infections, lump in the
eyelid (chalazion), red and swollen eyelids, discharge from the eyes, abnormal vision, bleeding of
the eye
• Swelling of your lymph glands
• Joint or muscle stiffness, sense of heaviness, pain in your groin
• Hair loss and abnormal hair texture

• Allergic reactions
• Redness or pain at the injection site
• Mouth pain
• Infections or inflammation of the mouth, mouth ulcers, oesophagus, stomach and intestines,
sometimes associated with pain or bleeding, poor movement of the intestines (including blockage),
abdominal or oesophageal discomfort, difficulty swallowing, vomiting of blood
• Skin infections
• Bacterial and viral infections
• Tooth infection
• Inflammation of the pancreas, obstruction of the bile duct
• Genital pain, problem having an erection
• Weight increase
• Thirst
• Hepatitis
• Injection site or injection device related disorders
• Skin reactions and disorders (which may be severe and life threatening), skin ulcers
• Bruises, falls and injuries
• Inflammation or haemorrhage of the blood vessels that can appear as small red or purple dots
(usually on the legs) to large bruise-like patches under the skin or tissue
• Benign cysts
• A severe reversible brain condition which includes seizures, high blood pressure, headaches,
tiredness, confusion, blindness or other vision problems.
Rare side effects (may affect up to 1 in 1,000 people)
• Heart problems to include heart attack, angina
• Flushing
• Discoloration of the veins
• Inflammation of the spinal nerve
• Problems with your ear, bleeding from your ear
• Underactivity of your thyroid gland
• Budd–Chiari syndrome (the clinical symptoms caused by blockage of the hepatic veins)
• Changes in or abnormal bowel function
• Bleeding in the brain
• Yellow discolouration of eyes and skin (jaundice)
• Serious allergic reaction (anaphylactic shock) signs of which may include difficulty breathing,
chest pain or chest tightness, and/or feeling dizzy/faint, severe itching of the skin or raised lumps
on the skin, swelling of the face, lips, tongue and /or throat, which may cause difficulty in
swallowing, collapse
• Breast disorders
• Vaginal tears
• Genital swelling
• Inability to tolerate alcohol consumption
• Wasting, or loss of body mass
• Increased appetite
• Fistula
• Joint effusion
• Cysts in the lining of joints (synovial cysts)
• Fracture
• Breakdown of muscle fibers leading to other complications
• Swelling of the liver, bleeding from the liver
• Cancer of the kidney

• Psoriasis like skin condition
• Cancer of the skin
• Paleness of the skin
• Increase of platelets or plasma cells (a type of white cell) in the blood
• Blood clot in small blood vessels (thrombotic microangiopathy)
• Abnormal reaction to blood transfusions
• Partial or total loss of vision
• Decreased sex drive
• Drooling
• Bulging eyes
• Sensitivity to light
• Rapid breathing
• Rectal pain
• Gallstones
• Hernia
• Injuries
• Brittle or weak nails
• Abnormal protein deposits in your vital organs
• Coma
• Intestinal ulcers
• Multi-organ failure
• Death
If you are given Valtroza together with other medicines for the treatment of mantle cell lymphoma the
side effects you may get are listed below:
Very common side effects (may affect more than 1 in 10 people)
• Pneumonia
• Loss of appetite
• Sensitivity, numbness, tingling or burning sensation of the skin, or pain in the hands or feet, due
to nerve damage
• Nausea and vomiting
• Diarrhoea
• Mouth ulcers
• Constipation
• Muscle pain, bone pain
• Hair loss and abnormal hair texture
• Tiredness, feeling weak
• Fever
Common side effects (may affect up to 1 in 10 people)
• Shingles (localized including around the eyes or spread across the body)
• Herpes virus infections
• Bacterial and viral infections
• Respiratory infections, bronchitis, coughing with phlegm, flu like illness
• Fungal infections
• Hypersensitivity (allergic reaction)
• Inability to produce enough insulin or resistance to normal levels of insulin
• Fluid retention
• Difficulty or problems in sleeping
• Loss of consciousness
• Altered level of consciousness, confusion

• Feeling dizzy
• Increased heartbeat, high blood pressure, sweating,
• Abnormal vision, blurred vision
• Heart failure, heart attack, chest pain, chest discomfort, increased or reduced heart rate
• High or low blood pressure
• Sudden fall of blood pressure upon standing which may lead to fainting
• Shortness of breath with exercise
• Cough
• Hiccups
• Ringing in the ears, ear discomfort
• Bleeding from your bowels or stomach
• Heartburn
• Stomach pain, bloating
• Difficulty swallowing
• Infection or inflammation of the stomach and intentines
• Stomach pain
• Sore mouth or lip, throat pain
• Alteration of liver function
• Itching of skin
• Redness of skin
• Rash
• Muscle spasms
• Infection of the urinary tract
• Pain in limbs
• Swelling of body, to include eyes and other parts of the body
• Shivering
• Redness and pain at injection site
• General ill feeling
• Weight loss
• Weight increase
Uncommon side effects (may affect up to 1 in 100 people)
• Hepatitis
• Severe allergic reaction (anaphylactic reaction) signs of which may include difficulty breathing,
chest pain or chest tightness, and/or feeling dizzy/faint, severe itching of the skin or raised lumps
on the skin, swelling of the face, lips, tongue and /or throat, which may cause difficulty in
swallowing, collapse
• Movement disorders, paralysis, twitching
• Vertigo
• Hearing loss, deafness
• Disorders that affect your lungs, preventing your body from getting enough oxygen. Some of
these include difficulty breathing, shortness of breath, shortness of breath without exercise,
breathing that becomes shallow, difficult or stops, wheezing
• Blood clots in your lungs
• Yellow discoloration of the eyes and skin (jaundice)
• Lump in the eyelid (chalazion), red and swollen eyelids
Rare side effects (may affect up to 1 in 1,000 people)
• Blood clot in small blood vessels (thrombotic microangiopathy)

If you get any side effects, talk to your doctor or pharmacist. This includes any possible side effects not
listed in this leaflet.

 


Keep this medicine out of the sight and reach of children.
Do not use this medicine after the expiry date stated on the vial and the carton after EXP. The expiry
date refers to the last day of that month.
Do not store above 30°C.
This medicinal product does not require any special storage conditions. Keep the vial in the outer carton
in order to protect from light.
Intravenous administration
The chemical and physical in-use stability of the reconstituted solution at a concentration of 1 mg/ml
has been demonstrated for 8 hours at 20°C-25°C stored in the original vial and/or a syringe. From a
microbiological point of view, unless the method of opening/reconstitution/dilution precludes the risk of
microbial contamination, the reconstituted solution should be used immediately after preparation. If not
used immediately, in-use storage times and conditions prior to use are the responsibility of the user.
Subcutaneous administration
The chemical and physical in -use stability of the reconstituted solution of 2.5 mg/ml has been
demonstrated for 8 hours at 20°C-25°C stored in the original vial and/or a syringe. From a
microbiological point of view, unless the method of opening/reconstitution/dilution precludes the risk of
microbial contamination, the reconstituted solution should be used immediately after preparation. If not
used immediately, in-use storage times and conditions prior to use are the responsibility of the user.
Valtroza is for single use only. Any unused product or waste material should be disposed of in
accordance with local requirements.
Do not throw away any medicines via wastewater or household waste. Your doctor or nurse will throw
away any medicine that is no longer required. These measures will help to protect the environment.


- The active substance is bortezomib. Each vial contains 3.5 mg of bortezomib (as a mannitol
boronic ester).
- The other ingredients are: mannitol (E421) and nitrogen.
Intravenous reconstitution:
After reconstitution, 1 ml of solution for intravenous injection contains 1 mg bortezomib.
Subcutaneous reconstitution:
After reconstitution, 1 ml of solution for subcutaneous injection contains 2.5 mg bortezomib.


Valtroza powder for solution for injection is a white to off -white lyophilised powder or plug. Each carton of Valtroza 3.5 mg powder for solution for injection contains 10 ml glass vial, dark grey bromo butyl rubber stopper with light blue colour aluminium seal.

Marketing Authorisation Holder and Manufacturer
Manufactured by Shilpa Medicare Limited for:
SPIMACO
AlQassim pharmaceutical plant
Saudi Pharmaceutical Industries &
Medical Appliance Corporation.


This leaflet was last revised in August 2019
  نشرة الدواء تحت مراجعة الهيئة العامة للغذاء والدواء (اقرأ هذه النشرة بعناية قبل البدء في استخدام هذا المنتج لأنه يحتوي على معلومات مهمة لك)

یحتوي ڤالتروزاعلى المادة الفعالة بورتیزومیب، وھو ما یسمى "مثبط البروتیازوم". یلعب البروتیازوم دورا مھما في التحكم
في وظائف الخلایا ونموھا. بالتدخل في وظیفتھ، یمكن أن یقتل بورتیزومیب الخلایا السرطانیة.
یستخدم ڤالتروزالعلاج الورم النخاعي المتعدد (سرطان نخاع العظم) في المرضى الذین تتجاوز أعمارھم 18 عام ا:ً
- بمفرده أو مع أدویة مثلالدوكسوروبیسین الشحمي أو دیكسامیثازون، للمرضى الذین یتفاقم لدیھم المرض (تدریجی ا)ً بعد تلقي
علاج مسبق واحد على الأقل والذین لم ینجح لدیھم زرع الخلایا الجذعیة بالدم أو یكون غیر مناسباً لھم.
- بالاقتران مع أدویة مثل میلفالان وبریدنیزون، للمرضى الذین لم یتم علاج مرضھم من قبل وغیر مناسبین للعلاج الكیمیائي
بجرعة عالیة مع زرع خلایا جذعیة للدم.
- بالاقتران مع أدویة مثل دیكسامیثازون أو دیكسامیثازون مع ثالیدومید، للمرضى الذین لم یتم علاج مرضھم مسبقا وقبل تلقي
العلاج الكیمیائي بجرعة عالیة مع زرع خلایا جذعیة للدم (العلاج التعریفي).
یستخدم ڤالتروزالعلاج سرطان غلاف الغدد اللیمفاویة (وھو نوع من السرطان یصیب الغدد اللیمفاویة) في المرضى الذین تبلغ
أعمارھم 18 عاما أو أكبر بالاقتران مع أدویة مثل ریتوكسیماب، سیكلوفوسفامید، دوكسوروبیسین وبریدنیزون، وللمرضى
الذین لم یتم علاج مرضھم مسبقا ولمن لا تناسبھ عملیة زرع الخلایا الجذعیة للدم.

لا تقم باستخدام ڤالتروزافي الحالات الآتیة:
- إذا كانت لدیك حساسیة تجاه بورتیزومیب, أو بورون أو تجاه أي من المكونات الأخرى لھذا الدواء (والمذكورة في الفقرة رقم
.(6
- إذا كانت لدیك مشاكل حادة بالرئة أو بالقلب.
تحذیرات واحتیاطات:
یجب أن تخبر طبیبك المعالج إذا كان لدیك أي مما یلي:
• انخفاض في عدد خلایا الدم الحمراء أو البیضاء
• مشاكل النزف و / أو انخفاض عدد الصفائح الدمویة لدیك بالدم.
• إسھال أو إمساك أو غثیان أو قيء.
• إغماء أو دوار أو إحساس بعدم الاتزان في الماضي.
• مشاكل في الكلى.
• مشاكل من معتدلة إلى حادة بالكبد.
• تنمیل أو وخز أو ألم في الیدین أو القدمین (الاعتلال العصبي) في الماضي.
• مشاكل في القلب أو ضغط الدم.
• ضیق في التنفس أو سعال.
• نوبات.
• حزام ناري (موضعیویشمل حول العینین أو منتشر في جمیع أنحاء الجسم).
• أعراض متلازمة تحلل الورم مثل التشنج العضلي وضعف العضلات والارتباك وفقدان أو اضطرابات البصر وضیق
التنفس.
• فقدان الذاكرة، صعوبة في التفكیر، صعوبة في المشي أو فقدان البصر. قد تكون ھذه علامات على وجود عدوى خطیرة
في المخ وقد یقترح طبیبك المعالج المزید من الاختبارات والمتابعة.
سیتعین علیك إجراء اختبارات للدم بشكل منتظم قبل وأثناء علاجك بواسطة فالتروزا، للتحقق من تعداد خلایا الدم لدیك بانتظام.
إذا كان لدیك سرطان غلاف الغدد اللیمفاویة وتم إعطاؤك دواء ریتوكسیماب مع ڤالتروزا، یجب علیك إخبار طبیبك المعالج:
- إذا كنت تعتقد أن لدیك التھاب الكبد الوبائي حالیا أو كنت قد أصبت بھ في الماضي. في حالات قلیلة، قد یتعرض المرضى
الذین أصیبوا بالتھاب الكبد "ب" لھجوم متكرر من التھاب الكبد، والذي قد یكون قاتلا .ً إذا كان لدیك تاریخ من الإصابة
بالتھاب الكبد الوبائي "ب"، فسیتم فحصك بعنایة من قبل طبیبك المعالجبحثا عن علامات التھاب الكبد النشط "ب".
یجب علیك قراءة نشرات العبوات الخاصةبجمیع الأدویة التي یجب استخدامھا بالتزامن مع ڤالتروزاللحصول على معلومات
تتعلق بھذه الأدویة قبل بدء العلاج بواسطةڤالتروزا. عند استخدام ثالیدومید، یلزم إیلاء اھتمام خاص لمتطلبات اختبار الحمل
والوقایة من حدوث الحمل (انظر فقرة الحمل والرضاعة الطبیعیة).
الأطفال والمراھقین
ینبغي عدم استخدامڤالتروزافي الأطفال والمراھقین لأنھ لیس معروفا كیفیةتأثیر ھذا الدواء علیھم.
الأدویة الأخرى و ڤالتروزا
یرجى إخبار طبیبك المعالج أو الصیدلي إذا كنت تتناول حالیاً، أو قد تناولت مؤخراً أو قد تتناول أي أدویة أخرى.
أخبر طبیبك المعالج على وجھ الخصوص إذا كنت تتناول أدویة تحتوي على أي من المواد الفعالة التالیة:
- كیتوكونازول، وھو یستخدم لعلاج الالتھابات الفطریة.
- ریتونافیر، وھو یستخدم لعلاج عدوى فیروس نقص المناعة البشریة.
- ریفامبیسین، وھو مضاد حیوي یستخدم لعلاج العدوى البكتیریة.
- كاربامازیبین، فینیتوین أو فینوباربیتال وھي أدویة تستخدم لعلاج الصرع.
- نبتة سانت جون (ھایبریكوم بیرفوراتوم), وھي تستخدم لعلاج الاكتئاب أو حالات أخرى.
- مضادات مرض السكر التي تؤخذ عن طریق الفم.
الحمل والرضاعة الطبیعیة

یجب عدم استخدام ڤالتروزاإذا كنتِ حاملا،ً إلا إذا كان ذلك ضروریا بشكل واضح.
یجب على كل من الرجال والنساء الذین یستخدمون ڤالتروزااستخدام وسائل فعالة لمنع الحمل أثناء العلاج بواسطة ڤالتروزا
ولمدة تصل إلى 3 أشھر بعد العلاج. إذا حدث الحمل، على الرغم من ھذه الوسائل، یجب إبلاغ طبیبك المعالج فور اً.
یجب علیكِ عدم الرضاعة الطبیعیة أثناء استخدام ڤالتروزا. تواصلي مع طبیبك المعالج لتحدید الوقت اللازم للعودة بأمان مرة
أخرى للرضاعة الطبیعیة بعد الانتھاء من العلاج.
یتسبب ثالیدومید في حدوث عیوب خلقیة ووفاة الجنین. عند إعطاء ڤالتروزامع ثالیدومید، یجب علیك اتباع برنامج الوقایة من
الحمل للثالیدومید (انظر نشرة العبوة الخاصة بالثالیومید).
القیادة واستخدام الآلات
ڤالتروزاقد یسبب التعب، والدوخة، والإغماء، أو عدم وضوح الرؤیة. لا تقم بالقیادة أو تشغیل أدوات أو آلات إذا تعرضتلھذه
الأعراض الجانبیة؛ حتى وإن لم تتعرض لتلك الأعراض, یجب علیك أن تظل حذر اً.

 

https://localhost:44358/Dashboard

سیقوم طبیبك المعالج بتحدید جرعة ڤالتروزا الخاصة بك حسب طولك ووزنك (مساحة سطح الجسم). جرعة البدایة المعتادة من
ڤالتروزاھي 1.3 ملجم/م 2 من مساحة سطح الجسم مرتین في الأسبوع.
قد یغیر طبیبك المعالج الجرعة والعدد الإجمالي لدورات العلاج، وھذا یتوقف على مدى استجابتك للعلاج, وعلى حدوث بعض
الأعراض الجانبیة المعینة وعلى ظروفك الأساسیة (مثل مشاكل الكبد).
في حالة الورم النخاعي المتعددالتقدمیة
، عندما یتم إعطاء ڤالتروزابمفرده، ستتلقى 4 جرعات من ڤالتروزاعن طریق الورید أو تحت الجلد في الأیام 1 و 4 و 8 و 11
ثم تلیھا "فترة راحة" لمدة 10 أیام بدون علاج. ھذه الفترة لمدة 21 یوما ( 3 أسابیع) وذلك یشكل دورة علاج واحدة. قد تتلقى ما
یصل إلى 8 دورات علاجیة ( 24 أسبوع ا)ً.
قد یتم إعطاؤك أیضا ڤالتروزامع الدوكسوروبیسین الشحمي أو دیكسامیثازون.
عندما یتم إعطاء ڤالتروزامع الدوكسوروبیسین الشحمي، سیتم إعطاؤك ڤالتروزاعن طریق الورید أو تحت الجلد كدورة علاج
مدتھا 21 یوما وتعُطى الدوكسوروبیسین الشحمي 30 ملجم/م 2 في الیوم 4 من ھذه الدورة العلاجیةعلى ھیئة تسریب بالورید
بعد حقن ڤالتروزا.
قد تتلقى ما یصل إلى 8 دورات علاجیة ( 24 أسبوع ا)ً.
عندما یتم إعطاء ڤالتروزامع دیكسامیثازون، سوف تتلقى ڤالتروزاعن طریق الورید وتحت الجلد كدورة علاج مدتھا 21 یوما ویتم إعطاء دیكسامیثازون 20 ملجم عن طریق الفم في أیام 1 و 2 و 4 و 5 و 8 و 9 و 11 و 12 من دورة علاج ڤالتروزا
والتي مدتھا 21 یوم ا.ً
قد تتلقى ما یصل إلى 8 دورات علاجیة ( 24 أسبوع ا)ً.
في حالة الورم النخاعي المتعدد الغیر معالجة مسبق اً
إذا لم یتم علاجك مسبقا من حالة الورم النخاعي المتعدد, وكانت عملیة زرع الخلایا الجذعیة للدم غیر مناسبة لك فسوف تتلقى
العلاج بواسطة ڤالتروزا بالتزامن مع دوائین آخرین; میلفالان و بریدنیزون.

في ھذه الحالة, ستكون مدة علاج الدورة الواحدة 42 یوما ( 6 أسابیع). وسوف تتلقى 9 دورات علاجیة ( 54 أسبوع ا)ً.
. • في الدورات العلاجیة من 1 إلى 4، تعُطى ڤالتروزا مرتین أسبوعیا في الأیام 1 و 4 و 8 و 11 و 22 و 25 و 29 و 32
. • في الدورات العلاجیة من 5 إلى 9، تُعطى ڤالتروزا مرة واحدة أسبوعیاً في الأیام 1 و 8 و 22 و 29
3, و 4 من الأسبوع الأول ,2 , یتم إعطاء میلفالان ( 9 ملجم/م 2) مع بریدنیزون ( 60 ملجم/م 2) سویاً عن طریق الفم في الأیام 1
لكل دورة علاجیة.
إذا لم یتم علاجك مسبقا من حالة الورم النخاعي المتعدد, وكانت عملیة زرع الخلایا الجذعیة للدم مناسبة لك فسوف تتلقى
العلاج بواسطة ڤالتروزا بالحقن بالورید أو تحت الجلد بالتزامن مع دیكسامیثازون, أو دیكسامیثازون مع ثالیدومید, كعلاج
تعریفي.
عندما یتم إعطاء ڤالتروزا مع دیكسامیثازون، سوف تتلقى ڤالتروزا عن طریق الحقن بالورید أو تحت الجلد كدورة علاج مدتھا
21 یوما ویتم إعطاء دیكسامیثازون 40 ملجم عن طریق الفم في أیام 1 و 2 و 3 و 4 و 8 و 9 و 10 و 11 من دورة ڤالتروزا
العلاجیة.
سوف تتلقى 4 دورات ( 12 أسبوع ا)ً.
عندما یتم إعطاء ڤالتروزا مع ثالیدومید ودیكسامیثازون، تكون مدة دورة العلاج 28 یوما ( 4 أسابیع).
یتم إعطاء دیكسامیثازون 40 ملجم عن طریق الفم في الأیام 1 و 2 و 3 و 4 و 8 و 9 و 10 و 11 من دورة علاج ڤالتروزا
التي تستغرق 28 یوما،ً ویعُطى الثالیدوماید یومیابًجرعة 50 ملجم حتى الیوم 14 من الدورة الأولى، و إذا تم تحمل جرعة
28 ویمكن زیادتھا إلى 200 ملجمیومیا من الدورة الثانیة فصاعد ا.ً - ثالیدوماید تزداد جرعتھ إلى 100 ملجم في الأیام 15
قد تتلقى ما یصل إلى 6 دورات ( 24 أسبوع ا)ً.
في حالة سرطان الغدد اللیمفاویة التي لم یتم علاجھا مسبق اً
إذا لم تعُالج من قبل من سرطان الغدد اللیمفاویة بالخلایا الوعائیة، فستتلقى ڤالتروزا في الورید أو تحت الجلد مع أدویة
ریتوكسیماب وسیكلوفوسفامید ودوكسوروبیسین وبریدنیزون.
یتم إعطاء ڤالتروزا عن طریق الورید أو تحت الجلد في الأیام 1 و 4 و 8 و 11 ، تلیھا "فترة راحة" بدون علاج. مدة دورة
العلاج الواحدة 21 یوما ( 3 أسابیع). قد تتلقى ما یصل إلى 8 دورات علاجیة ( 24 أسبوع ا)ً.
یتم إعطاء الأدویة التالیة في الیوم الأول من كل دورة علاجیة مدتھا 21 یوما من ڤالتروزا كحقن بالتسریب في الورید:
. ریتوكسیماب بجرعة 375 ملجم/م 2، سیكلوفوسفامید بجرعة 750 ملجم/م 2 ودوكسوروبیسین بجرعة 50 ملجم/م 2
یُعطى بریدنیزون عن طریق الفم بجرعة 100 ملجم/م 2 في الأیام 1 و 2 و 3 و 4 و 5 من دورة علاج ڤالتروزا.
كیفیة إعطاء ڤالتروزا
ھذا الدواء للاستخدام عن طریق الحقن في الورید أو تحت الجلد. سیتم إعطاء ڤالتروزا من قبل أخصائي رعایة صحیة یتمتع
بخبرة واسعة في استخدام الأدویة السامة للخلایا.
یجب حل مسحوق ڤالتروزا قبل الحقن. وسیتم ذلك عن طریق أخصائي الرعایة الصحیة. ثم یتم حقن المحلول الناتج إما في
الورید أو تحت الجلد. یتم الحقن في الورید بسرعة، ویستغرق 3 إلى 5 ثوان. الحقن تحت الجلد یكون إما في الفخذین أو البطن.

إذا تم إعطاؤك الكثیر من ڤالتروزا
نظراً لأنھ یتم إعطاء ھذا الدواء من قبل طبیبك المعالج أو ممرضتك، فمن غیر المرجح أن تحصل على جرعات زائدة. ومع
ذلك, في حالة حدوث ھذا الاحتمال الغیر مرجح من تعاطي جرعة زائدة, سوف یقوم طبیبك المعالج بمراقبتك لرصد الأعراض
الجانبیة.

مثل جمیع الأدویة قد یسبب ھذا الدواء أعراضاً جانبیة وإن لم تكن تحدث لكل من یستخدم ھذا الدواء.
بعض ھذه الأعراض قد یكون خطیر اً.
إذا تم إعطاؤك ڤالتروزا لعلاج الورم النخاعي المتعدد أو سرطان الغدد اللیمفاویة، أخبر طبیبك المعالج على الفور إذا لاحظت
أیا من الأعراض التالیة:
- التشنج العضلي وضعف العضلات.
- الارتباك، فقدان أو اضطرابات البصر، العمى، النوبات، الصداع.
- ضیق في التنفس أو تورم قدمیك أو تغیرات في ضربات قلبك وارتفاع ضغط الدم والإرھاق والإغماء.
- السعال وصعوبات في التنفس أو ضیق في الصدر.
یمكن أن یسبب العلاج بواسطة ڤالتروزا بشكل شائعانخفاضا في أعداد خلایا الدم الحمراء والبیضاء والصفائح الدمویة في الدم
لدیك. لذلك، سیتعین علیك إجراء اختبارات للدم بشكل منتظم قبل وأثناء علاجك بواسطةڤالتروزا، للتحقق من تعداد خلایا الدم
بانتظام. قد تواجھ انخفاضاً في عدد:
- الصفائح الدمویة، والذي قد یجعلك أكثر عرضة للكدمات، أو النزیف دون إصابة واضحة (على سبیل المثال، النزیف من
الأمعاء أو المعدة أو الفم أو اللثة أو النزیف في الدماغ أو النزیف من الكبد).
- خلایا الدم الحمراء، والذي یمكن أن یسبب فقر الدم، مع أعراض مثل التعب والشحوب.
- خلایا الدم البیضاء والذي قد یجعلك أكثر عرضة للعدوى أو أعراض تشبھ الأنفلونزا.
إذا تم إعطاؤك ڤالتروزا لعلاج الورم النخاعي المتعدد، فإن الأعراض الجانبیة التي قد تتعرض لھا مدرجة أدناه:
أعراض جانبیة شائعة جدا (والتي قد تصیب أكثر من شخص واحد من كل 10 أشخاص)
• حساسیة أو تنمیل أو وخز أو إحساس بالحرقة أو ألم في الیدین أو القدمین بسبب تلف الأعصاب.
• انخفاض عدد خلایا الدم الحمراء و / أو خلایا الدم البیضاء (انظر أعلاه).
• حمى.
• شعور بالإعیاء (الغثیان) أو القيء وفقدان الشھیة.
• إمساك مع أو بدون انتفاخ (یمكن أن یكون شدید).
• إسھال: إذا حدث ذلك، فمن المھم أن تشرب كمیة أكبر من الماء أكثر من المعتاد. قد یعطیك طبیبك المعالج دواء آخر
للسیطرة على الإسھال.
• تعب (إرھاق)، وشعور بالضعف.
• آلام بالعضلات وآلام بالعظام.
أعراض جانبیة شائعة (والتي قد تصیب ما یصل إلى شخص واحد من كل 10 أشخاص)
• انخفاض ضغط الدم، والھبوط المفاجئ لضغط الدم عند الوقوف مما قد یؤدي إلى الإغماء.
• ارتفاع ضغط الدم.

• انخفاض في وظائف الكلیتین.
• صداع.
• شعور عام بالألم أو الدوار أو عدم الاتزان أو الإحساس بالضعف أو فقدان الوعي.
• رعشات.
• عدوى، بما في ذلك الالتھاب الرئوي وعدوى الجھاز التنفسي والتھاب الشعب الھوائیة والالتھابات الفطریة والسعال مع
وجود بلغم، ومرض یشبھ الأنفلونزا.
• حزام ناري (موضعي ویشمل حول العینین أو منتشر في جمیع أنحاء الجسم).
• آلام في الصدر أو ضیق في التنفس عند ممارسة الریاضة.
• أنواع مختلفة من الطفح الجلدي.
• حكة في الجلد أو كتل على الجلد أو جفاف البشرة.
• احمرار الوجھ أو تكسر الشعیرات الدمویة الصغیرة.
• احمرار الجلد.
• جفاف.
• حرقة بالمعدة، انتفاخ، تجشؤ، ریح، آلام في المعدة، نزیف من الأمعاء أو المعدة.
• تغیر في وظائف الكبد.
• التھاب الفم أو الشفة، جفاف الفم، تقرحات الفم أو ألم الحلق.
• فقدان الوزن، وفقدان التذوق.
• تقلصات و تشنجات و ضعف العضلات، ألم في الأطراف.
• عدم وضوح الرؤیة.
• إصابة الطبقة الخارجیة للعین والسطح الداخلي للجفون (التھاب الملتحمة).
• نزیف الأنف.
• صعوبة أو مشاكل في النوم، تعرق،قلق، تقلب أو اكتئاب المزاج، أرق أو توتر, تغیرات في حالتك العقلیة، ارتباك.
• تورم الجسم، لیشمل حول العینین وأجزاء أخرى من الجسم.
أعراض جانبیة غیر شائعة (والتي قد تصیب ما یصل إلى شخص واحد من كل 100 شخص)
• فشل القلب، أو نوبة قلبیة، أو ألم أو عدم ارتیاح في الصدر، زیادة أو انخفاض معدل ضربات القلب.
• فشل في الكلیتین.
• التھاب الورید والجلطات الدمویة في الأوردة والرئتین.
• مشاكل في تخثر الدم.
• عدم كفایة الدورة الدمویة.
• التھاب في البطانة المحیطة بقلبك أو السائل حول قلبك.
• عدوى بما في ذلك التھابات المسالك البولیة والأنفلونزا وعدوى فیروس الھربس وعدوى الأذن والتھاب النسیج الخلوي.
• براز دموي، أو نزیف من الأغشیة المخاطیة، على سبیل المثال، الفم، المھبل.
• اضطرابات بالأوعیة الدمویة الخاصة بالمخ.
• شلل، أو نوبات، أو سقوط، أو اضطرابات بالحركة، خلل أو تغیر أو نقص في الحواس (الإحساس، السمع، التذوق، الشم)،
اضطراب الانتباه، ارتعاش، رجفات.
• التھاب المفاصل، بما في ذلك التھاب المفاصل في الأصابع وأصابع القدم والفك.
• اضطرابات قد تصیب رئتیك، وتمنع جسمك من الحصول على كمیة كافیة من الأكسجین. بعض ھذه الاضطرابات تشمل
صعوبة في التنفس، وضیق في التنفس، وضیق في التنفس دون ممارسة الریاضة، والتنفس الذي یصبح ضحلا،ً أو صعبا أو یتوقف تماما،ً أو أزیز بالصدر.
• فواق واضطرابات الكلام.
• زیادة أو نقص فیإفراز البول (بسبب تلف الكلى)، أو ألم بالتبول أو وجود دم/بروتینات في البول, احتباس السوائل.

• تغیر في مستویات الوعي مع عدم تركیز وضعف الذاكرة أو فقدانھا.
• فرط الحساسیة.
• فقدان السمع، أو صمم أو رنین في الأذنین، ألم بالأذن.
• خلل بالھرمونات قد یؤثر على امتصاص الماء والملح.
• فرط نشاط الغدة الدرقیة.
• عدم القدرة على إنتاج كمیة كافیة من الإنسولین أو مقاومة لمستویات الإنسولین الطبیعیة.
• تھیج أو التھاب العینین، إفراط في بلل العینین، ألم بالعینین، جفاف العینین، التھابات العین، ورم في الجفن، احمرار
وانتفاخ الجفون، إفرازات من العینین، خلل في الرؤیة، نزف العین.
• تورم الغدد اللمفاویة.
• تصلب المفاصل أو العضلات، شعور بالثقل، ألم في منطقة الفخذ.
• تساقط الشعر وملمس غیر طبیعي للشعر.
• تفاعلات تحسسیة.
• احمرار أو ألم في موقع الحقن.
• ألم في الفم.
• عدوى أو التھاب الفم، تقرحات الفم، أو المريء، أو المعدة والأمعاء، مصحوبةأحیاناًبألم أو نزیف، ضعف في حركة
الأمعاء (بما في ذلك الانسداد المعوي)، ألم في البطن أو المريء، صعوبة في البلع، تقیؤ دم.
• عدوى جلدیة.
• عدوى بكتیریة وفیروسیة.
• عدوى الأسنان.
• التھاب البنكریاس، انسداد القناة الصفراویة.
• ألم تناسلي، مشكلة في حدوث الانتصاب.
• زیادة الوزن.
• عطش.
• التھاب الكبد.
• اضطرابات مرتبطة بموقع الحقن أو بجھاز الحقن.
• تفاعلات و اضطرابات جلدیة (قد تكون شدیدة وتھدد الحیاة)، تقرحات الجلد.
• كدمات وسقوط وإصابات.
• التھاب أو نزیف في الأوعیة الدمویة التي یمكن أن تظھر كنقاط حمراء أو أرجوانیة صغیرة (عادة على الساقین) إلى بقع
كبیرة تشبھ الكدمات تحت الجلد أو الأنسجة.
• تكیسات حمیدة.
• حالة دماغیة شدیدة قابلة للانعكاس تتضمن نوبات أو ارتفاع ضغط الدم أو صداع أو تعب أو ارتباك أو عمى أو مشاكل
أخرى في الرؤیة.
أعراض جانبیة نادرة (والتي قد تصیب ما یصل إلى شخص واحد من كل 1000 شخص)
- مشاكل بالقلب لتشمل النوبة القلبیة، الذبحة الصدریة.
- احمرار الجسم.
- اختفاء تلون الأوردة.
- التھاب العصب الفقري.
- مشاكل في أذنك، أونزیف من أذنك.
- قلة نشاط الغدة الدرقیة.
- متلازمة بود تشیاري (الأعراض السریریة الناجمة عن انسداد الأوردة الكبدیة).
- تغیرات أو خلل في وظیفة الأمعاء.
- نزیف في المخ.

اصفرارالجلد والعینین (الیرقان).
- علامات الحساسیة الخطیرة (صدمة الحساسیة) التي قد تشمل صعوبة في التنفس، أو ألم في الصدر أو ضیق الصدر، و /
أو الشعور بالدوار / الإغماء، أو الحكة الشدیدة في الجلد أو الكتل المرتفعة على الجلد، أو تورم الوجھ أو الشفتین أو اللسان.
و / أو الحلق، والذي قد یسبب صعوبة في البلع، الانھیار.
- اضطرابات الثدي.
- إفرازات مھبلیة.
- تورم الأعضاء التناسلیة.
- عدم القدرة على تحمل شرب الكحول.
- ھدر، أو فقدان كتلة الجسم.
- زیادة الشھیة.
- الناسور.
- ارتشاح المفصل.
- تكیسات في بطانة المفاصل (التكیسات الزلیلیة) .
- كسور.
- انھیار ألیاف العضلات مما یؤدي إلى مضاعفات أخرى.
- تورم الكبد، والنزیف من الكبد.
- سرطان الكلى.
- حالة بالجلد مثل الصدفیة.
- سرطان الجلد.
- شحوب الجلد.
- زیادة الصفائح الدمویة أو خلایا البلازما (نوع من الخلایا البیضاء) في الدم.
- تجلط الدم في الأوعیة الدمویة الصغیرة (اعتلال الأوعیة الدقیقة الخثاري) .
- رد فعل غیر طبیعي لنقل الدم.
- فقدان جزئي أو كلي للرؤیة.
- انخفاض الدافع الجنسي.
- سیلان اللعاب.
- انتفاخ العینین.
- حساسیة تجاه الضوء.
- سرعة التنفس.
- ألم بالمستقیم.
- حصى في المرارة.
- فتق.
- إصابات.
- ھشاشة أو ضعف الأظافر.
- ترسب البروتین بشكل غیر طبیعي في الأعضاء الحیویة الخاصة بك.
- غیبوبة.
- قرحة معویة.
- فشل متعدد بالأعضاء.
- الموت.

إذا تم إعطاؤك ڤالتروزا بالاقتران مع أدویة أخرى لعلاج سرطان الغدد اللیمفاویة، فإن الأعراض الجانبیة التي قد تتعرض لھا
مدرجة أدناه:
أعراض جانبیة شائعة جدا (والتي قد تصیب أكثر من شخص واحد من كل 10 أشخاص)
- التھاب رئوي.
- فقدان الشھیة.
- حساسیة أو تنمیل أو وخز أو إحساس بالحرقة أو ألم في الیدین أو القدمین بسبب تلف الأعصاب.
- غثیان و قئ.
- إسھال.
- قرحة الفم.
- إمساك.
- آلام العضلات وآلام العظام.
- تساقط الشعر وملمس غیر طبیعي للشعر.
- تعب، وشعور بالضعف.
- حمى.
أعراض جانبیة شائعة (والتي قد تصیب ما یصل إلى شخص واحد من كل 10 أشخاص)
- حزام ناري (موضعي ویشمل حول العینین أو منتشر في جمیع أنحاء الجسم).
- عدوى فیروس الھربس.
- التھابات بكتیریة وفیروسیة.
- عدوى الجھاز التنفسي، التھاب الشعب الھوائیة، سعال مع بلغم، مرض مثل الأنفلونزا.
- التھابات فطریة.
- فرط الحساسیة (رد الفعل التحسسي).
- عدم القدرة على إنتاج كمیة كافیة من الإنسولین أو مقاومة لمستویات الإنسولین الطبیعیة.
- احتباس السوائل.
- صعوبة أو مشاكل في النوم.
- فقدان الوعي.
- تغیر مستوى الوعي وعدم التركیز.
- شعور بالدوار.
- زیادة نبضات القلب وارتفاع ضغط الدم والتعرق.
- خلل أو عدم وضوح في الرؤیة.
- فشل القلب، نوبة قلبیة، ألم أو عدم ارتیاح في الصدر، زیادة أو نقص معدل ضربات القلب.
- ارتفاع أو انخفاض ضغط الدم.
- ھبوط مفاجئ لضغط الدم عند الوقوف مما قد یؤدي إلى الإغماء.
- ضیق في التنفس مع ممارسة الریاضة.
- سعال.
- فواق.
- رنین في الأذنین، وعدم ارتیاح في الأذن.
- نزیف من الأمعاء أو المعدة.
- حرقة بالمعدة.

- آلام في المعدة، وانتفاخ.
- صعوبة البلع.
- إصابة أو التھاب المعدة والأمعاء.
- آلام في المعدة.
- التھاب الفم أو الشفة وآلام الحلق.
- تغیر في وظائف الكبد.
- حكة في الجلد.
- احمرار الجلد.
- طفح جلدي.
- تشنجات العضلات.
- التھاب المسالك البولیة.
- ألم في الأطراف.
- تورم في الجسم، لیشمل العینین وأجزاء أخرى من الجسم.
- ارتجاف.
- احمرار وألم في موقع الحقن.
- شعور عام بالإعیاء.
- فقدان الوزن.
- زیادة الوزن.
أعراض جانبیة غیر شائعة (والتي قد تصیب ما یصل إلى شخص واحد من كل 100 شخص)
- التھاب الكبد.
- علامات الحساسیة الشدیدة (تفاعل الحساسیة) قد تشمل صعوبة في التنفس، أو ألم في الصدر أو ضیق في الصدر، و / أو
شعور بالدوار / الإغماء، أو حكة شدیدة في الجلد أو كتل مرتفعة على الجلد، أو تورم في الوجھ أو الشفتین أو اللسان و / أو
الحلق، والذي قد یسبب صعوبة في البلع، الانھیار.
- اضطرابات الحركة، شلل، وخز.
- دوار.
- فقدان السمع، صمم.
- اضطرابات قد تصیب رئتیك، وتمنع جسمك من الحصول على كمیة كافیة من الأكسجین. بعض ھذه الاضطرابات تشمل
صعوبة في التنفس، وضیق في التنفس، وضیق في التنفس دون ممارسة الریاضة، والتنفس الذي یصبح ضحلا،ً أو صعبا أو قد یتوقف، أو أزیز بالصدر.
- حدوث جلطات الدم في رئتیك.
- اصفرارالعینین والجلد (الیرقان).
- تورم في الجفون، انتفاخ أو احمرار الجفون.
أعراض جانبیة نادرة (والتي قد تصیب ما یصل إلى شخص واحد من كل 1000 شخص)
- تجلطالدمفیالأوعیةالدمویةالصغیرة (اعتلالالأوعیةالدقیقةالخثاري).
إذا تعرضت لحدوث أي أعراض جانبیة, تواصل مع طبیبك المعالج أو الصیدلي. یشمل ذلك أي أعراض جانبیة لم یتم ذكرھا
في ھذه النشرة.

 

یحفظ ھذا الدواء بعیدا عن متناول ونظر الأطفال

لا تقم باستخدام ھذا الدواء بعد انتھاء تاریخ الصلاحیة المدون على الزجاجة وعلى العبوة بعد كلمة
الصلاحیة یشیر إلى آخر یوم من الشھر المذكور على العبوة.
لا یحفظ في درجة حرارة أعلى من 30 درجة مئویة.
لا یتطلب ھذا الدواء ظروف خاصة للتخزین. تحفظ الزجاجة داخل العبوة الأصلیة لحمایتھا من الضوء.
في حالة إعطاء الدواء عن طریق الحقن بالورید
تمإثباتثباتالاستخدامالكیمیائیوالفیزیائیللمحلولالمعدللحقنبتركیز 1 ملجم / مللمدة 8 ساعاتعند 20 درجةمئویة - 25
درجةمئویةمخزنةفیالزجاجة الأصلیةو / أوالمحاقنالأصلیة. منالناحیةالمیكروبیولوجیة،مالمتكنطریقةالفتح / إعداد المحلول /
التخفیفتعیق منخطرالتلوثالمیكروبي،یجباستخدامالمحلولالمعدللحقنفورالتحضیر. إذالمیتماستخدامھ
علٮالفور،فإنأوقاتوظروفالتخزینقیدالاستخدامھیمسؤولیةالمستخدم.
في حالة إعطاء الدواء عن طریق الحقن تحتالجلد
تمإثباتثباتالاستخدامالكیمیائیوالفیزیائیللمحلولالمعدللحقنبتركیز 2.5 ملجم / مللمدة 8 ساعاتعنددرجةحرارة 20 درجةمئویة - 25
درجةمئویةمخزنةفیالزجاجة الأصلیةو / أوالمحقنةالأصلیة. منالناحیةالمیكروبیولوجیة،مالمتكنطریقةالفتح / إعداد المحلول/
التخفیفتعیق منخطرالتلوثالمیكروبي،یجباستخدامالمحلولالمعدللحقنفورالتحضیر.
إذالمیتماستخدامھعلٮالفور،فإنأوقاتوظروفالتخزینقیدالاستخدامھیمسؤولیةالمستخدم.
ڤالتروزاھوللاستخدامالفردیفقط. یجبالتخلصمنأیمنتجأونفایاتغیرمستخدمةوفقالًلمتطلباتالمحلیة.
یحظر التخلص من الأدویة عن طریق میاه الصرف الصحى أو نفایات المنزل. سیقوم الطبیب المعالج أو الممرضة بالتخلص
من أي أدویة لم تعد مطلوبة. فسوف تساعد ھذه الإجراءات على حمایة البیئة.

 

محتویات ڤالتروزا
- المادة الفعالة ھي بورتیزومیب. تحتوي كل زجاجةعلى 3.5 ملجم من مادة بورتیزومیب (على ھیئة مانیتولإسترالبورون).
ونیتروجین. (E • المكونات الأخرى: ھى مانیتول ( 421
إعداد المحلول للحقنبالورید:
• بعدإعداد المحلول،یحتوي كل 1 ملمنمحلولالحقنفیالوریدعلى 1 ملجممنبورتیزومیب.
إعداد المحلول للحقنتحتالجلد:
• بعدإعداد المحلول،یحتوي كل 1 ملمنمحلولالحقنتحتالجلدعلى 2.5 ملجممنبورتیزومیب

ڤالتروزا ھي مسحوق لونھ أبیض إلى أبیض فاتح لإعداد محلول للحقن.
كل عبوة من ڤالتروزا 3.5 ملجم مسحوق لإعداد محلول للحق ت نحتوي على زجاجة 10 مل, بھا
سدادةمطاطیةباللونالرمادیالداكنمعختمألومنیومأزرقفاتح.

صُنع بواسطة شیلبا للمستلزمات الطبیة المحدودة لصالح:
الدوائیة
مصنع الأدویة بالقصیم
الشركة السعودیة للصناعات الدوائیة والمستلزمات الطبیة.
المملكة العربیة السعودیة

. تمت مراجعة ھذه النشرة في أغسطس 2019
 Read this leaflet carefully before you start using this product as it contains important information for you

Valtroza powder for solution for injection. Bortezomib 3.5 mg powder for solution for injection.

Each vial contains 3.5 mg bortezomib (as a mannitol boronic ester). After reconstitution with 1.4 ml, 1 ml of solution for subcutaneous injection contains 2.5 mg bortezomib. After reconstitution with 3.5 ml, 1 ml of solution for intravenous injection contains 1 mg bortezomib. For the full list of excipients, see section 6.1

Powder for solution for injection. White to off-white cake or powder.

Bortezomib as monotherapy or in combination with pegylated liposomal doxorubicin or
dexamethasone is indicated for the treatment of adult patients with progressive multiple
myeloma who have received at least 1 prior therapy and who have already undergone or are
unsuitable for haematopoietic stem cell transplantation.
Bortezomib in combination with melphalan and prednisone is indicated for the treatment of
adult patients with previously untreated multiple myeloma who are not eligible for high-dose
chemotherapy with haematopoietic stem cell transplantation.
Bortezomib in combination with dexamethasone, or with dexamethasone and thalidomide, is
indicated for the induction treatment of adult patients with previously untreated multiple
myeloma who are eligible for high-dose chemotherapy with haematopoietic stem cell
transplantation.

Bortezomib in combination with rituximab, cyclophosphamide, doxorubicin and prednisone is
indicated for the treatment of adult patients with previously untreated mantle cell lymphoma
who are unsuitable for haematopoietic stem cell transplantation.

 


Bortezomib treatment must be initiated under supervision of a physician experienced in the
treatment of cancer patients, however Bortezomib may be administered by a healthcare
professional experienced in use of chemotherapeutic agents. Bortezomib must be reconstituted
by a healthcare professional (see section 6.6).
Posology for treatment of progressive multiple myeloma (patients who have received at least
one prior therapy)
Monotherapy
Bortezomib 3.5 mg powder for solution for injection is administered via intravenous or
subcutaneous injection at the recommended dose of 1.3 mg/m2 body surface area twice weekly
for two weeks on days 1, 4, 8, and 11 in a 21-day treatment cycle. This 3-week period is
considered a treatment cycle. It is recommended that patients receive 2 cycles of bortezomib
following a confirmation of a complete response. It is also recommended that responding
patients who do not achieve a complete remission receive a total of 8 cycles of bortezomib

therapy. At least 72 hours should elapse between consecutive doses of bortezomib.
Dose adjustments during treatment and re-initiation of treatment for monotherapy
Bortezomib treatment must be withheld at the onset of any Grade 3 non-haematological or any
Grade 4 haematological toxicities, excluding neuropathy as discussed below (see also section
4.4). Once the symptoms of the toxicity have resolved, bortezomib treatment may be reinitiated
at a 25% reduced dose (1.3 mg/m2 reduced to 1.0 mg/m2; 1.0 mg/m2 reduced to 0.7
mg/m2). If the toxicity is not resolved or if it recurs at the lowest dose, discontinuation of
Bortezomib must be considered unless the benefit of treatment clearly outweighs the risk.

Neuropathic pain and/or peripheral neuropathy
Patients who experience bortezomib-related neuropathic pain and/or peripheral neuropathy are
to be managed as presented in Table 1 (see section 4.4). Patients with pre-existing severe
neuropathy may be treated with bortezomib only after careful risk/benefit assessment.

Table 1: Recommended* posology modifications for bortezomib-related neuropathy

Severity of neuropathyPosology modification
Grade 1 (asymptomatic; loss of deep tendon
reflexes or paresthesia) with no pain or loss of
function
None
Grade 1 with pain or Grade 2 (moderate
symptoms; limiting instrumental Activities of
Daily Living (ADL)**)
Reduce bortezomib to 1.0 mg/m2
or Change bortezomib treatment schedule to
1.3 mg/m2 once per week
Grade 2 with pain or Grade 3 (severe
symptoms; limiting self-care ADL***)
Withhold bortezomib treatment until
symptoms of toxicity have resolved. When toxicity resolves re-initiate bortezomib
treatment and reduce dose to 0.7 mg/m2 once
per week
Grade 4 (life-threatening consequences;
urgent
intervention indicated) and/or severe
autonomic neuropathy
Discontinue bortezomib

Based on posology modifications in Phase II and III multiple myeloma studies and postmarketing
experience. Grading based on NCI Common Toxicity Criteria CTCAE v 4.0.
** Instrumental ADL: refers to preparing meals, shopping for groceries or clothes, using
telephone, managing money, etc.
*** Self care ADL: refers to bathing, dressing and undressing, feeding self, using the toilet,
taking medicinal products, and not bedridden.
Combination therapy with pegylated liposomal doxorubicin
Bortezomib 3.5 mg powder for solution for injection is administered via intravenous or
subcutaneous injection at the recommended dose of 1.3 mg/m2 body surface area twice weekly
for two weeks on days 1, 4, 8, and 11 in a 21-day treatment cycle. This 3-week period is
considered a treatment cycle. At least 72 hours should elapse between consecutive doses of
bortezomib.
Pegylated liposomal doxorubicin is administered at 30 mg/m² on day 4 of the bortezomib
treatment cycle as a 1-hour intravenous infusion administered after the bortezomib injection.
Up to 8 cycles of this combination therapy can be administered as long as patients have not
progressed and tolerate treatment. Patients achieving a complete response can continue
treatment for at least 2 cycles after the first evidence of complete response, even if this requires
treatment for more than 8 cycles. Patients whose levels of paraprotein continue to decrease
after 8 cycles can also continue for as long as treatment is tolerated, and they continue to
respond.
For additional information concerning pegylated liposomal doxorubicin, see the corresponding
Summary of Product Characteristics.
Combination with dexamethasone
Bortezomib 3.5 mg powder for solution for injection is administered via intravenous or
subcutaneous injection at the recommended dose of 1.3 mg/m2 body surface area twice weekly
for two weeks on days 1, 4, 8, and 11 in a 21-day treatment cycle. This 3-week period is
considered a treatment cycle. At least 72 hours should elapse between consecutive doses of
bortezomib.
Dexamethasone is administered orally at 20 mg on days 1, 2, 4, 5, 8, 9, 11, and 12 of the
bortezomib treatment cycle.
Patients achieving a response or a stable disease after 4 cycles of this combination therapy can
continue to receive the same combination for a maximum of 4 additional cycles.

For additional information concerning dexamethasone, see the corresponding Summary of
Product Characteristics.
Dose adjustments for combination therapy for patients with progressive multiple myeloma
For bortezomib dosage adjustments for combination therapy follow dose modification
guidelines described under monotherapy above.
Posology for previously untreated multiple myeloma patients not eligible for haematopoietic
stem cell transplantation
Combination therapy with melphalan and prednisone
Bortezomib 3.5 mg powder for solution for injection is administered via intravenous or
subcutaneous injection in combination with oral melphalan and oral prednisone as shown in
Table 2. A 6-week period is considered a treatment cycle. In Cycles 1-4, Bortezomib is
administered twice weekly on days 1, 4, 8, 11, 22, 25, 29 and 32. In Cycles 5-9, bortezomib is
administered once weekly on days 1, 8, 22 and 29. At least 72 hours should elapse between
consecutive doses of bortezomib.
Melphalan and prednisone should both be given orally on days 1, 2, 3 and 4 of the first week of
each bortezomib treatment cycle.
Nine treatment cycles of this combination therapy are administered.

Table 2: Recommended posology for Bortezomib in combination with melphalan and
prednisone

Twice weekly Bortezomib (cycles 1-4)

Week

1

2

3

4

5

6

Vc

(1.3 mg/m2)

Day 1

--

--

Day 4

Day 8

Day 11

rest

period

Day 22

Day 25

Day 29

Day 32

rest

period

M (9 mg/m2)

P (60 mg/m2)

Day 1

Day 2

Day 3

Day 4

--

--

rest

period

--

--

--

--

rest

period

Once weekly Bortezomib (cycles 5-9)

Week

1

2

3

4

5

6

Vc

(1.3 mg/m2)

Day 1

--

--

--

Day 8

rest

period

Day 22

Day 29

rest

period

M (9 mg/m2)

P (60 mg/m2)

Day 1

Day 2

Day 3

Day 4

--

rest

period

--

 

rest

period

 

Vc= Bortezomib; M=melphalan, P=prednisone

 

Dose adjustments during treatment and re-initiation of treatment for combination therapy with melphalan and prednisone

 

Prior to initiating a new cycle of therapy:

  • Platelet counts should be ≥ 70 x 109/l and the absolute neutrophils count should be ≥ 1.0 x 109/l
  • Non-haematological toxicities should have resolved to Grade 1 or baseline

 

 

Table 3: Posology modifications during subsequent cycles of bortezomib therapy in combination with melphalan and prednisone

Toxicity

Posology modification or delay

Haematological toxicity during a cycle

 

·     If prolonged Grade 4 neutropenia or thrombocytopenia, or thrombocytopenia with bleeding is observed in the previous cycle

Consider reduction of the melphalan dose by 25% in the next cycle.

·     If platelet counts ≤ 30 x 109/l or ANC ≤ 0.75 x 109/l on a Bortezomib dosing day (other than day 1)

Bortezomib therapy should be withheld

·     If several bortezomib doses in a cycle are withheld (≥ 3 doses during twice weekly administration or ≥ 2 doses during weekly administration)

Bortezomib dose should be reduced by 1 dose level (from 1.3 mg/m2 to 1 mg/m2, or from 1 mg/m2 to 0.7 mg/m2)

Grade ≥ 3 non-haematological toxicities

Bortezomib therapy should be withheld until symptoms of the toxicity have resolved to Grade 1 or baseline. Then, bortezomib may be reinitiated with one dose level reduction (from 1.3 mg/m2 to 1 mg/m2, or from 1 mg/m2 to 0.7 mg/m2). For bortezomib -related neuropathic pain and/or peripheral neuropathy, hold and/or modify bortezomib as outlined in Table 1.

 

For additional information concerning melphalan and prednisone, see the corresponding Summary of Product Characteristics.

 

Posology for previously untreated multiple myeloma patients eligible for haematopoietic stem cell transplantation (induction therapy)

 

 

Combination therapy with dexamethasone

 

Bortezomib 3.5 mg powder for solution for injection is administered via intravenous or subcutaneous injection at the recommended dose of 1.3 mg/m2 body surface area twice weekly for two weeks on days 1, 4, 8, and 11 in a 21- day treatment cycle. This 3-week period is considered a treatment cycle. At least 72 hours should elapse between consecutive doses of bortezomib.

 

Dexamethasone is administered orally at 40 mg on days 1, 2, 3, 4, 8, 9, 10 and 11 of the bortezomib treatment cycle.

 

Four treatment cycles of this combination therapy are administered.

 

 

Combination therapy with dexamethasone and thalidomide

 

Bortezomib 3.5 mg powder for solution for injection is administered via intravenous or subcutaneous injection at the recommended dose of 1.3 mg/m2 body surface area twice weekly for two weeks on days 1, 4, 8, and 11 in a 28- day treatment cycle. This 4-week period is considered a treatment cycle. At least 72 hours should elapse between consecutive doses of bortezomib.

 

Dexamethasone is administered orally at 40 mg on days 1, 2, 3, 4, 8, 9, 10 and 11 of the bortezomib treatment cycle.

 

Thalidomide is administered orally at 50 mg daily on days 1-14 and if tolerated the dose is increased to 100 mg on days 15-28, and thereafter may be further increased to 200 mg daily from cycle 2 (see Table 4).

 

Four treatment cycles of this combination are administered. It is recommended that patients with at least partial response receive 2 additional cycles.

 

Table 4: Posology for bortezomib combination therapy for patients with previously untreated              multiple myeloma eligible for haematopoietic stem cell transplantation

Vc+Dx

Cycles 1 to 4

Week

1

2

3

Vc

(1.3 mg/m2)

Day 1, 4

Day 8, 11

Rest Period

Dx 40 mg

Day 1, 2, 3, 4

Day 8, 9, 10, 11

--

Vc+Dx+T

Cycle 1

Week

1

2

3

4

Vc

 (1.3 mg/m2)

Day 1, 4

Day 8, 11

Rest Period

Rest Period

T 50 mg

Daily

Daily

-

-

T 100 mga

-

-

Daily

Daily

Dx 40 mg

Day 1,2,3,4

Day 8,9,10,11

-

-

Cycles 2 to 4b

Vc

 (1.3 mg/m2)

Day 1, 4

Day 8, 11

Rest Period

Rest Period

T 200 mga

Daily

Daily

Daily

Daily

Dx 40 mg

Day 1, 2, 3, 4

Day 8, 9, 10, 11

-

-

Vc= Bortezomib; Dx=dexamethasone; T=thalidomide

a Thalidomide dose is increased to 100 mg from week 3 of Cycle 1 only if 50 mg is tolerated and to 200 mg from cycle 2 onwards if 100 mg is tolerated.

b Up to 6 cycles may be given to patients who achieve at least a partial response after 4 cycles.

 

Dosage adjustments for transplant eligible patients

For bortezomib dosage adjustments,dose modification guidelines described for monotherapy should be followed.

In addition, when bortezomib is given in combination with other chemotherapeutic medicinal products, appropriate dose reductions for these products should be considered in the event of toxicities according to the recommendations in the Summary of Product Characteristics.

 

Posology for patients with previously untreated mantle cell lymphoma (MCL)

 

Combination therapy with rituximab, cyclophosphamide, doxorubicin and prednisone (VcR-CAP)

 

Bortezomib 3.5 mg powder for solution for injection is administered via intravenous or subcutaneous injection at the recommended dose of 1.3 mg/m2 body surface area twice weekly for two weeks on days 1, 4, 8, and 11, followed by a 10-day rest period on days 12-21. This 3-week period is considered a treatment cycle. Six bortezomib cycles are recommended, although for patients with a response first documented at cycle 6, two additional bortezomib cycles may be given. At least 72 hours should elapse between consecutive doses of bortezomib.

 

The following medicinal products are administered on day 1 of each bortezomib 3 week treatment cycle as intravenous infusions: rituximab at 375 mg/m2, cyclophosphamide at 750 mg/m2 and doxorubicin at 50 mg/m2.

 

Prednisone is administered orally at 100 mg/m2 on days 1, 2, 3, 4 and 5 of each bortezomib treatment cycle.

 

Dose adjustments during treatment for patients with previously untreated mantle cell lymphoma

 

Prior to initiating a new cycle of therapy:

•       Platelet counts should be ≥ 100,000 cells/μL and the absolute neutrophils count (ANC) should be ≥ 1,500 cells/μL

•       Platelet counts should be ≥ 75,000 cells/μL in patients with bone marrow infiltration or splenic sequestration

•       Haemoglobin ≥ 8 g/dL

•       Non-haematological toxicities should have resolved to Grade 1 or baseline.

 

Bortezomib treatment must be withheld at the onset of any ≥ Grade 3 bortezomib -related non-haematological toxicities (excluding neuropathy) or ≥ Grade 3 haematological toxicities (see also section 4.4). For dose adjustments, see Table 5 below.

 

Granulocyte colony stimulating factors may be administered for haematologic toxicity according to local standard practice. Prophylactic use of granulocyte colony stimulating factors should be considered in case of repeated delays in cycle administration. Platelet transfusion for the treatment of thrombocytopenia should be considered when clinically appropriate.

 

Table 5: Dose adjustments during treatment for patients with previously untreated mantle cell lymphoma

Toxicity

Posology modification or delay

Haematological toxicity

•       ≥ Grade 3 neutropenia with fever, Grade 4 neutropenia lasting more than 7 days, a platelet count < 10,000 cells/μL

Bortezomib therapy should be withheld for up to 2 weeks until the patient has an ANC ≥ 750 cells/μL and a platelet count ≥ 25,000 cells/μL.

•       If, after bortezomib has been held, the toxicity does not resolve, as defined above, then bortezomib must be discontinued.

•       If toxicity resolves i.e. patient has an ANC ≥ 750 cells/μL and a platelet count ≥ 25,000 cells/μL, bortezomib may be reinitiated at a dose reduced by one dose level (from 1.3 mg/m2 to 1 mg/m2, or from 1 mg/m2 to 0.7 mg/m2).

• If platelet counts < 25,000 cells/μL. or ANC < 750 cells/μL on a bortezomib dosing day (other than Day 1 of each cycle)

Bortezomib therapy should be withheld

Grade ≥ 3 non-haematological toxicities considered to be related to bortezomib

Bortezomib therapy should be withheld until symptoms of the toxicity have resolved to Grade 2 or better. Then, bortezomib may be reinitiated at a dose reduced by one dose level (from 1.3 mg/m2 to 1 mg/m2, or from 1 mg/m2to 0.7 mg/m2). For bortezomib -related neuropathic pain and/or peripheral neuropathy, hold and/or modify bortezomib as outlined in Table 1.

 

In addition, when bortezomib is given in combination with other chemotherapeutic medicinal products, appropriate dose reductions for these medicinal products should be considered in the event of toxicities, according to the recommendations in the respective Summary of Product Characteristics.

 

Special populations

 

Elderly

There is no evidence to suggest that dose adjustments are necessary in patients over 65 years of age with multiple myeloma or with mantle cell lymphoma.

 

There are no studies on the use of bortezomib in elderly patients with previously untreated multiple myeloma who are eligible for high-dose chemotherapy with haematopoietic stem cell transplantation. Therefore no dose recommendations can be made in this population.

 

In a study in previously untreated mantle cell lymphoma patients, 42.9% and 10.4% of patients exposed to bortezomib were in the range 65-74 years and ≥ 75 years of age, respectively. In patients aged ≥ 75 years, both regimens, VcR-CAP as well as R-CHOP, were less tolerated (see section 4.8).

 

Hepatic impairment

Patients with mild hepatic impairment do not require a dose adjustment and should be treated per the recommended dose. Patients with moderate or severe hepatic impairment should be started on bortezomib at a reduced dose of 0.7 mg/m2 per injection during the first treatment cycle, and a subsequent dose escalation to 1.0 mg/m2 or further dose reduction to 0.5 mg/m2 may be considered based on patient tolerability (see Table 6 and sections 4.4 and 5.2).

 

 

Table 6: Recommended starting dose modification for Bortezomib in patients with hepatic              impairment

Grade of

hepatic

impairment*

Bilirubin level

SGOT  (AST)

levels

Modification of starting dose

Mild

≤ 1.0 x ULN

> ULN

None

> 1.0 x −1.5 x ULN

Any

None

Moderate

> 1.5 x −3 x ULN

Any

Reduce bortezomib to 0.7 mg/m2 in the first treatment cycle. Consider dose escalation to 1.0 mg/m2 or further dose reduction to 0.5 mg/m2 in subsequent cycles based on patient tolerability.

Severe

> 3 x ULN

Any

Abbreviations: SGOT=serum glutamic oxaloacetic transaminase;

AST=aspartate aminotransferase; ULN=upper limit of the normal range.

* Based on NCI Organ Dysfunction Working Group classification for categorising hepatic impairment (mild, moderate, severe).

 

Renal impairment

 

The pharmacokinetics of bortezomib are not influenced in patients with mild to moderate renal impairment (Creatinine Clearance [CrCL] > 20 ml/min/1.73 m2); therefore, dose adjustments are not necessary for these patients. It is unknown if the pharmacokinetics of bortezomib are influenced in patients with severe renal impairment not undergoing dialysis (CrCL < 20 ml/min/1.73 m2). Since dialysis may reduce Bortezomib concentrations, bortezomib should be administered after the dialysis procedure (see section 5.2).

 

Paediatric population

 

The safety and efficacy of bortezomib in children below 18 years of age have not been established (see sections 5.1 and 5.2). Currently available data are described in section 5.1 but no recommendation on a posology can be made.

 

Method of administration

 

Bortezomib 3.5 mg powder for solution for injection is available for intravenous or subcutaneous administration.

 

Bortezomib 1 mg powder for solution for injection is available for intravenous administration only.

 

Bortezomib should not be given by other routes. Intrathecal administration has resulted in death.

 

Intravenous injection

 

Bortezomib 3.5 mg reconstituted solution is administered as a 3-5 second bolus intravenous injection through a peripheral or central intravenous catheter followed by a flush with sodium chloride 9 mg/ml (0.9%) solution for injection. At least 72 hours should elapse between consecutive doses of bortezomib.

 

Subcutaneous injection

 

Bortezomib 3.5 mg reconstituted solution is administered subcutaneously through the thighs (right or left) or abdomen (right or left). The solution should be injected subcutaneously, at a 45-90° angle. Injection sites should be rotated for successive injections.

 

If local injection site reactions occur following bortezomib subcutaneous injection, either a less concentrated bortezomib solution (bortezomib 3.5 mg to be reconstituted to 1 mg/ml instead of 2.5 mg/ml) may be administered subcutaneously or a switch to intravenous injection is recommended.

 

When bortezomib is given in combination with other medicinal products, refer to the Summary of Product Characteristics of these products for instructions for administration.

 

 


Hypersensitivity to the active substance, to boron or to any of the excipients listed in section 6.1. Acute diffuse infiltrative pulmonary and pericardial disease. When bortezomib is given in combination with other medicinal products, refer to their Summaries of Product Characteristics for additional contraindications.

When bortezomib is given in combination with other medicinal products, the Summary of Product Characteristics of these other medicinal products must be consulted prior to initiation of treatment with bortezomib. When thalidomide is used, particular attention to pregnancy testing and prevention requirements is needed (see section 4.6).

 

Intrathecal administration

 

There have been fatal cases of inadvertent intrathecal administration of bortezomib. Bortezomib 1 mg powder for solution for injection is for intravenous use only, while bortezomib 3.5 mg powder for solution for injection is for intravenous or subcutaneous use. Bortezomib should not be administered intrathecally.

 

Gastrointestinal toxicity

 

Gastrointestinal toxicity, including nausea, diarrhoea, vomiting and constipation are very common with bortezomib treatment. Cases of ileus have been uncommonly reported (see section 4.8). Therefore, patients who experience constipation should be closely monitored.

 

Haematological toxicity

 

Bortezomib treatment is very commonly associated with haematological toxicities (thrombocytopenia, neutropenia and anaemia). In studies in patients with relapsed multiple myeloma treated with bortezomib and in patients with previously untreated MCL treated with bortezomib in combination with rituximab, cyclophosphamide, doxorubicin, and prednisone (VcR-CAP), one of the most common haematologic toxicity was transient thrombocytopenia. Platelets were lowest at Day 11 of each cycle of bortezomib treatment and typically recovered to baseline by the next cycle. There was no evidence of cumulative thrombocytopenia. The mean platelet count nadir measured was approximately 40% of baseline in the single-agent multiple myeloma studies and 50% in the MCL study. In patients with advanced myeloma the severity of thrombocytopenia was related to pre-treatment platelet count: for baseline platelet counts < 75,000/μl, 90% of 21 patients had a count ≤ 25,000/μl during the study, including 14% < 10,000/μl; in contrast, with a baseline platelet count > 75,000/μl, only 14% of 309 patients had a count ≤ 25,000/μl during the study.

 

In patients with MCL (study LYM-3002), there was a higher incidence (56.7% versus 5.8%) of Grade ≥ 3 thrombocytopenia in the bortezomib treatment group (VcR-CAP) as compared to the non- bortezomib treatment group (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone [R-CHOP]). The two treatment groups were similar with regard to the overall incidence of all-grade bleeding events (6.3% in the VcR-CAP group and 5.0% in the R-CHOP group) as well as Grade 3 and higher bleeding events (VcR-CAP: 4 patients [1.7%]; R-CHOP: 3 patients [1.2%]). In the VcR-CAP group, 22.5% of patients received platelet transfusions compared to 2.9% of patients in the R-CHOP group.

 

Gastrointestinal and intracerebral haemorrhage, have been reported in association with bortezomib treatment. Therefore, platelet counts should be monitored prior to each dose of bortezomib. Bortezomib therapy should be withheld when the platelet count is < 25,000/μl or, in the case of combination with melphalan and prednisone, when the platelet count is ≤ 30,000/μl (see section 4.2). Potential benefit of the treatment should be carefully weighed against the risks, particularly in case of moderate to severe thrombocytopenia and risk factors for bleeding.

 

Complete blood counts (CBC) with differential and including platelet counts should be frequently monitored throughout treatment with bortezomib. Platelet transfusion should be considered when clinically appropriate (see section 4.2).

 

In patients with MCL, transient neutropenia that was reversible between cycles was observed, with no evidence of cumulative neutropenia. Neutrophils were lowest at Day 11 of each cycle of bortezomib treatment and typically recovered to baseline by the next cycle. In study LYM-3002, colony stimulating factor support was given to 78% of patients in the VcR-CAP arm and 61% of patients in the R-CHOP arm. Since patients with neutropenia are at increased risk of infections, they should be monitored for signs and symptoms of infection and treated promptly. Granulocyte colony stimulating factors may be administered for haematologic toxicity according to local standard practice. Prophylactic use of granulocyte colony stimulating factors should be considered in case of repeated delays in cycle administration (see section 4.2).

 

Herpes zoster virus reactivation

 

Antiviral prophylaxis is recommended in patients being treated with Bortezomib.

In the Phase III study in patients with previously untreated multiple myeloma, the overall incidence of herpes zoster reactivation was more common in patients treated with Bortezomib +Melphalan+Prednisone compared with Melphalan+Prednisone (14% versus 4% respectively).

In patients with MCL (study LYM-3002), the incidence of herpes zoster infection was 6.7% in the VcR-CAP arm and 1.2% in the R-CHOP arm (see section 4.8).

 

Hepatitis B Virus (HBV) reactivation and infection

When rituximab is used in combination with bortezomib, HBV screening must always be performed in patients at risk of infection with HBV before initiation of treatment. Carriers of hepatitis B and patients with a history of hepatitis B must be closely monitored for clinical and laboratory signs of active HBV infection during and following rituximab combination treatment with bortezomib. Antiviral prophylaxis should be considered. Refer to the Summary of Product Characteristics of rituximab for more information.

 

Progressive multifocal leukoencephalopathy (PML)

Very rare cases with unknown causality of John Cunningham (JC) virus infection, resulting in PML and death, have been reported in patients treated with bortezomib. Patients diagnosed with PML had prior or concurrent immunosuppressive therapy. Most cases of PML were diagnosed within 12 months of their first dose of bortezomib. Patients should be monitored at regular intervals for any new or worsening neurological symptoms or signs that may be suggestive of PML as part of the differential diagnosis of CNS problems. If a diagnosis of PML is suspected, patients should be referred to a specialist in PML and appropriate diagnostic measures for PML should be initiated. Discontinue bortezomib if PML is diagnosed.

 

Peripheral neuropathy

Treatment with bortezomib is very commonly associated with peripheral neuropathy, which is predominantly sensory. However, cases of severe motor neuropathy with or without sensory peripheral neuropathy have been reported. The incidence of peripheral neuropathy increases early in the treatment and has been observed to peak during cycle 5.

 

It is recommended that patients be carefully monitored for symptoms of neuropathy such as a burning sensation, hyperesthesia, hypoesthesia, paraesthesia, discomfort, neuropathic pain or weakness.

 

In the Phase III study comparing bortezomib administered intravenously versus subcutaneously, the incidence of Grade ≥ 2 peripheral neuropathy events was 24% for the subcutaneous injection group and 41% for the intravenous injection group (p=0.0124). Grade ≥ 3 peripheral neuropathy occurred in 6% of patients in the subcutaneous treatment group, compared with 16% in the intravenous treatment group (p=0.0264). The incidence of all grade peripheral neuropathy with bortezomib administered intravenously was lower in the historical studies with bortezomib administered intravenously than in study MMY-3021.

 

Patients experiencing new or worsening peripheral neuropathy should undergo neurological evaluation and may require a change in the dose, schedule or route of administration to subcutaneous (see section 4.2). Neuropathy has been managed with supportive care and other therapies.

 

Early and regular monitoring for symptoms of treatment-emergent neuropathy with neurological evaluation should be considered in patients receiving Bortezomib in combination with medicinal products known to be associated with neuropathy (e.g. thalidomide) and appropriate dose reduction or treatment discontinuation should be considered.

 

In addition to peripheral neuropathy, there may be a contribution of autonomic neuropathy to some adverse reactions such as postural hypotension and severe constipation with ileus. Information on autonomic neuropathy and its contribution to these undesirable effects is limited.

 

Seizures

Seizures have been uncommonly reported in patients without previous history of seizures or epilepsy. Special care is required when treating patients with any risk factors for seizures.

 

Hypotension

Bortezomib treatment is commonly associated with orthostatic/postural hypotension. Most adverse reactions are mild to moderate in nature and are observed throughout treatment. Patients who developed orthostatic hypotension on bortezomib (injected  intravenously) did not have evidence of orthostatic hypotension prior to treatment with bortezomib. Most patients required treatment for their orthostatic hypotension. A minority of patients with orthostatic hypotension experienced syncopal events. Orthostatic/postural hypotension was not acutely related to bolus infusion of bortezomib. The mechanism of this event is unknown although a component may be due to autonomic neuropathy. Autonomic neuropathy may be related to bortezomib or bortezomib may aggravate an underlying condition such as diabetic or amyloidotic neuropathy. Caution is advised when treating patients with a history of syncope receiving medicinal products known to be associated with hypotension; or who are dehydrated due to recurrent diarrhoea or vomiting.Management of orthostatic/postural hypotension may include adjustment of antihypertensive medicinal products, rehydration or administration of mineralocorticosteroids and/or  sympathomimetics. Patients should be instructed to seek medical advice if they experience symptoms of dizziness, light-headedness or fainting spells.

 

Posterior Reversible Encephalopathy Syndrome (PRES)

There have been reports of PRES in patients receiving bortezomib. PRES is a rare, often reversible, rapidly evolving neurological condition, which can present with seizure, hypertension, headache, lethargy, confusion, blindness, and other visual and neurological disturbances. Brain imaging, preferably Magnetic Resonance Imaging (MRI), is used to confirm the diagnosis. In patients developing PRES, bortezomib should be discontinued.

 

Heart failure

Acute development or exacerbation of congestive heart failure, and/or new onset of decreased left ventricular ejection fraction has been reported during bortezomib treatment. Fluid retention may be a predisposing factor for signs and symptoms of heart failure. Patients with risk factors for or existing heart disease should be closely monitored.

 

Electrocardiogram investigations

There have been isolated cases of QT-interval prolongation in clinical studies, causality has not been established.

 

Pulmonary disorders

There have been rare reports of acute diffuse infiltrative pulmonary disease of unknown aetiology such as pneumonitis, interstitial pneumonia, lung infiltration, and acute respiratory distress syndrome (ARDS) in patients receiving bortezomib (see section 4.8). Some of these events have been fatal. A pre-treatment chest radiograph is recommended to serve as a baseline for potential post-treatment pulmonary changes.

 

In the event of new or worsening pulmonary symptoms (e.g., cough, dyspnoea), a prompt diagnostic evaluation should be performed and patients treated appropriately. The benefit/risk ratio should be considered prior to continuing bortezomib therapy.

 

In a clinical trial, two patients (out of 2) given high-dose cytarabine (2 g/m2 per day) by continuous infusion over 24 hours with daunorubicin and bortezomib for relapsed acute myelogenous leukaemia died of ARDS early in the course of therapy, and the study was terminated. Therefore, this specific regimen with concomitant administration with high-dose cytarabine (2 g/m2 per day) by continuous infusion over 24 hours is not recommended.

 

Renal impairment

Renal complications are frequent in patients with multiple myeloma. Patients with renal impairment should be monitored closely (see sections 4.2 and 5.2).

 

Hepatic impairment

Bortezomib is metabolised by liver enzymes. Bortezomib exposure is increased in patients with moderate or severe hepatic impairment; these patients should be treated with bortezomib at reduced doses and closely monitored for toxicities (see sections 4.2 and 5.2).

 

Hepatic reactions

Rare cases of hepatic failure have been reported in patients receiving bortezomib and concomitant medicinal products and with serious underlying medical conditions. Other reported hepatic reactions include increases in liver enzymes, hyperbilirubinaemia, and hepatitis. Such changes may be reversible upon discontinuation of bortezomib (see section 4.8).

 

Tumour lysis syndrome

Because bortezomib is a cytotoxic agent and can rapidly kill malignant plasma cells and MCL cells, the complications of tumour lysis syndrome may occur. The patients at risk of tumour lysis syndrome are those with high tumour burden prior to treatment. These patients should be monitored closely and appropriate precautions taken.

 

Concomitant medicinal products

Patients should be closely monitored when given bortezomib in combination with potent CYP3A4-inhibitors. Caution should be exercised when bortezomib is combined with CYP3A4- or CYP2C19 substrates (see section 4.5).

 

Normal liver function should be confirmed and caution should be exercised in patients receiving oral hypoglycemics (see section 4.5).

 

Potentially immunocomplex-mediated reactions

Potentially immunocomplex-mediated reactions, such as serum-sickness-type reaction, polyarthritis with rash and proliferative glomerulonephritis have been reported uncommonly. Bortezomib should be discontinued if serious reactions occur.

 

 

 


In vitro studies indicate that bortezomib is a weak inhibitor of the cytochrome P450 (CYP) isozymes 1A2, 2C9, 2C19, 2D6 and 3A4. Based on the limited contribution (7%) of CYP2D6 to the metabolism of bortezomib, the CYP2D6 poor metaboliser phenotype is not expected to affect the overall disposition of bortezomib.

 

A drug-drug interaction study assessing the effect of ketoconazole, a potent CYP3A4 inhibitor, on the pharmacokinetics of bortezomib (injected intravenously), showed a mean bortezomib AUC increase of 35% (CI90% [1.032 to 1.772]) based on data from 12 patients. Therefore, patients should be closely monitored when given bortezomib in combination with potent CYP3A4 inhibitors (e.g. ketoconazole, ritonavir).

 

In a drug-drug interaction study assessing the effect of omeprazole, a potent CYP2C19 inhibitor, on the pharmacokinetics of bortezomib (injected intravenously), there was no significant effect on the pharmacokinetics of bortezomib based on data from 17 patients.

 

A drug-drug interaction study assessing the effect of rifampicin, a potent CYP3A4 inducer, on the pharmacokinetics of bortezomib (injected intravenously), showed a mean bortezomib AUC reduction of 45% based on data from 6 patients. Therefore, the concomitant use of bortezomib with strong CYP3A4 inducers (e.g., rifampicin, carbamazepine, phenytoin, phenobarbital and St. John's Wort) is not recommended, as efficacy may be reduced.

 

In the same drug-drug interaction study assessing the effect of dexamethasone, a weaker CYP3A4 inducer, on the pharmacokinetics of bortezomib (injected intravenously), there was no significant effect on the pharmacokinetics of bortezomib based on data from 7 patients.

 

A drug-drug interaction study assessing the effect of melphalan-prednisone on the pharmacokinetics of Bortezomib (injected intravenously), showed a mean bortezomib AUC increase of 17% based on data from 21 patients. This is not considered clinically relevant.

 

During clinical trials, hypoglycemia and hyperglycemia were uncommonly and commonly reported in diabetic patients receiving oral hypoglycemics. Patients on oral antidiabetic agents receiving bortezomib treatment may require close monitoring of their blood glucose levels and adjustment of the dose of their antidiabetics.

 


Contraception in males and females

Male and female patients of childbearing potential must use effective contraceptive measures during and for 3 months following treatment.

 

Pregnancy

No clinical data are available for bortezomib with regard to exposure during pregnancy. The teratogenic potential of bortezomib has not been fully investigated.

 

In non-clinical studies, bortezomib had no effects on embryonal/foetal development in rats and rabbits at the highest maternally tolerated doses. Animal studies to determine the effects of bortezomib on parturition and post-natal development were not conducted (see section 5.3). Bortezomib should not be used during pregnancy unless the clinical condition of the woman requires treatment with bortezomib.

If bortezomib is used during pregnancy, or if the patient becomes pregnant while receiving this medicinal product, the patient should be informed of potential for hazard to the foetus.

 

Thalidomide is a known human teratogenic active substance that causes severe life-threatening birth defects. Thalidomide is contraindicated during pregnancy and in women of childbearing potential unless all the conditions of the thalidomide pregnancy prevention programme are met. Patients receiving Bortezomib in combination with thalidomide should adhere to the pregnancy prevention programme of thalidomide. Refer to the Summary of Product Characteristics of thalidomide for additional information.

 

Breast-feeding

It is not known whether bortezomib is excreted in human milk. Because of the potential for serious adverse reactions in breast-fed infants, breast-feeding should be discontinued during treatment with bortezomib.

 

Fertility

Fertility studies were not conducted with bortezomib (see section 5.3).


 

Bortezomib may have a moderate influence on the ability to drive and use machines. Bortezomib may be associated with fatigue very commonly, dizziness commonly, syncope uncommonly and orthostatic/postural hypotension or blurred vision commonly. Therefore, patients must be cautious when driving or using machines and should be advised not to drive or operate machinery if they experience these symptoms (see section 4.8).

 


Summary of the safety profile

 

Serious adverse reactions uncommonly reported during treatment with bortezomib include cardiac failure, tumour lysis syndrome, pulmonary hypertension, posterior reversible encephalopathy syndrome, acute diffuse infiltrative pulmonary disorders and rarely autonomic neuropathy.

The most commonly reported adverse reactions during treatment with bortezomib are nausea, diarrhoea, constipation, vomiting, fatigue, pyrexia, thrombocytopenia, anaemia, neutropenia, peripheral neuropathy (including sensory), headache, paraesthesia, decreased appetite, dyspnoea, rash, herpes zoster and myalgia.

 

Tabulated summary of adverse reactions

 

Multiple Myeloma

Undesirable effects in Table 7 were considered by the investigators to have at least a possible or probable causal relationship to bortezomib. These adverse reactions are based on an integrated data set of 5,476 patients of whom 3,996 were treated with Bortezomib at 1.3 mg/m2 and included in Table 7.

Overall, bortezomib was administered for the treatment of multiple myeloma in 3,974 patients.

 

Adverse reactions are listed below by system organ class and frequency grouping. Frequencies are defined as: Very common (≥ 1/10); common (≥ 1/100 to < 1/10); uncommon (≥ 1/1,000 to < 1/100); rare (≥ 1/10,000 to < 1/1,000); very rare (< 1/10,000), not known (cannot be estimated from the available data). Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness. Table 7 has been generated using Version 14.1 of the MedDRA.

Post-marketing adverse reactions not seen in clinical trials are also included.

 

Table 7: Adverse reactions in patients with Multiple Myeloma treated with Bortezomib  clinical trials, and all post-marketing adverse reactions regardless of indication#

 

System Organ Class

Incidence

Adverse reaction

Infections and infestations

Common

Herpes zoster (inc disseminated & ophthalmic), Pneumonia*, Herpes simplex*, Fungal infection*

Uncommon

Infection*, Bacterial infections*, Viral infections*, Sepsis (inc septic shock)*, Bronchopneumonia, Herpes virus infection*, Meningoencephalitis herpetic#, Bacteraemia (inc staphylococcal), Hordeolum, Influenza, Cellulitis, Device related infection, Skin infection*, Ear infection*, Staphylococcal infection, Tooth infection*

Rare

Meningitis (inc bacterial), Epstein-Barr virus infection, Genital herpes, Tonsillitis, Mastoiditis, Post viral fatigue syndrome

Neoplasms benign, malignant and unspecified (incl cysts and polyps)

Rare

Neoplasm malignant, Leukaemia plasmacytic, Renal cell carcinoma, Mass, Mycosis fungoides, Neoplasm benign*

Blood and lymphatic system disorders

Very Common

Thrombocytopenia*, Neutropenia*, Anaemia*

Common

Leukopenia*, Lymphopenia*

Uncommon

Pancytopenia*, Febrile neutropenia, Coagulopathy*, Leukocytosis*, Lymphadenopathy, Haemolytic anaemia#

Rare

Disseminated intravascular coagulation, Thrombocytosis*, Hyperviscosity syndrome, Platelet disorder NOS, , Thrombotic microangiopathy (inc thrombocytopenic purpura)#, Blood disorder NOS, Haemorrhagic diathesis, Lymphocytic infiltration

Immune system disorders

Uncommon

Angioedema#, Hypersensitivity*

Rare

Anaphylactic shock, Amyloidosis, Type III immune complex mediated reaction

Endocrine disorders

Uncommon

Cushing's syndrome*, Hyperthyroidism*, Inappropriate antidiuretic hormone secretion

Rare

Hypothyroidism

Metabolism and nutrition disorders

Very Common

Decreased appetite

Common

Dehydration, Hypokalaemia*, Hyponatraemia*, Blood glucose abnormal*, Hypocalcaemia*, Enzyme abnormality*

Uncommon

Tumour lysis syndrome, Failure to thrive*, Hypomagnesaemia*, Hypophosphataemia*, Hyperkalaemia*, Hypercalcaemia*, Hypernatraemia*, Uric acid abnormal*, Diabetes mellitus*, Fluid retention

Rare

Hypermagnesaemia*, Acidosis, Electrolyte imbalance*, Fluid overload, Hypochloraemia*, Hypovolaemia, Hyperchloraemia*, Hyperphosphataemia*, Metabolic disorder, Vitamin B complex deficiency, Vitamin B12 deficiency, Gout, Increased appetite, Alcohol intolerance

Psychiatric disorders

Common

Mood disorders and disturbances*, Anxiety disorder*, Sleep disorders and disturbances*

Uncommon

Mental disorder*, Hallucination*, Psychotic disorder*, Confusion*, Restlessness

Rare

Suicidal ideation*, Adjustment disorder, Delirium, Libido decreased

Nervous system disorders

Very Common

Neuropathies*, Peripheral sensory neuropathy, Dysaesthesia*, Neuralgia*

Common

Motor neuropathy*, Loss of consciousness (inc syncope), Dizziness*, Dysgeusia*, Lethargy, Headache*

Uncommon

Tremor, Peripheral sensorimotor neuropathy, Dyskinesia*, Cerebellar coordination and balance disturbances*, Memory loss (exc dementia)*, Encephalopathy*, Posterior Reversible Encephalopathy Syndrome#, Neurotoxicity, Seizure disorders*, Post herpetic neuralgia, Speech disorder*, Restless legs syndrome, Migraine, Sciatica, Disturbance in attention, Reflexes abnormal*, Parosmia

Rare

Cerebral haemorrhage*, Haemorrhage intracranial (inc subarachnoid)*, Brain oedema, Transient ischaemic attack, Coma, Autonomic nervous system imbalance, Autonomic neuropathy, Cranial palsy*, Paralysis*, Paresis*, Presyncope, Brain stem syndrome, Cerebrovascular disorder, Nerve root lesion, Psychomotor hyperactivity, Spinal cord compression, Cognitive disorder NOS, Motor dysfunction, Nervous system disorder NOS, Radiculitis, Drooling, Hypotonia

Eye disorders

Common

Eye swelling*, Vision abnormal*, Conjunctivitis*

Uncommon

Eye haemorrhage*, Eyelid infection*, Chalazion# , Blepharitis#, Eye inflammation*, Diplopia, Dry eye*, Eye irritation*, Eye pain, Lacrimation increased, Eye discharge

Rare

Corneal lesion*, Exophthalmos, Retinitis, Scotoma, Eye disorder (inc. eyelid) NOS, Dacryoadenitis acquired, Photophobia, Photopsia, Optic neuropathy#, Different degrees of visual impairment (up to blindness)*

Ear and labyrinth disorders

Common

Vertigo*

Uncommon

Dysacusis (inc tinnitus)*,Hearing impaired (up to and inc deafness), Ear discomfort*

Rare

Ear haemorrhage, Vestibular neuronitis, Ear disorder NOS

Cardiac disorders

Uncommon

Cardiac tamponade#, Cardio-pulmonary arrest*, Cardiac fibrillation (inc atrial), Cardiac failure (inc left and right ventricular)*, Arrhythmia*, Tachycardia*, Palpitations, Angina pectoris, Pericarditis (inc pericardial effusion)*, Cardiomyopathy*, Ventricular dysfunction*, Bradycardia

Rare

Atrial flutter, Myocardial infarction*, Atrioventricular block*, Cardiovascular disorder (inc cardiogenic shock), Torsade de pointes, Angina unstable, Cardiac valve disorders*, Coronary artery insufficiency, Sinus arrest

Vascular disorders

Common

Hypotension*, Orthostatic hypotension, Hypertension*

Uncommon

Cerebrovascular accident#, Deep vein thrombosis*, Haemorrhage*, Thrombophlebitis (inc superficial), Circulatory collapse (inc hypovolaemic shock), Phlebitis, Flushing*, Haematoma (inc perirenal)*, Poor peripheral circulation*, Vasculitis, Hyperaemia (inc ocular)*

Rare

Peripheral embolism, Lymphoedema, Pallor, Erythromelalgia, Vasodilatation, Vein discolouration, Venous insufficiency

Respiratory, thoracic and mediastinal disorders

Common

Dyspnoea*, Epistaxis, Upper/lower respiratory tract infection*, Cough*

Uncommon

Pulmonary embolism, Pleural effusion, Pulmonary oedema (inc acute), Pulmonary alveolar haemorrhage#, Bronchospasm, Chronic obstructive pulmonary disease*, Hypoxaemia*, Respiratory tract congestion*, Hypoxia, Pleurisy*, Hiccups, Rhinorrhoea, Dysphonia, Wheezing

Rare

Respiratory failure, Acute respiratory distress syndrome, Apnoea, Pneumothorax, Atelectasis, Pulmonary hypertension, Haemoptysis, Hyperventilation, Orthopnoea, Pneumonitis, Respiratory alkalosis, Tachypnoea, Pulmonary fibrosis, Bronchial disorder*, Hypocapnia*, Interstitial lung disease, Lung infiltration, Throat tightness, Dry throat, Increased upper airway secretion, Throat irritation, Upper-airway cough syndrome

Gastrointestinal disorders

Very Common

Nausea and vomiting symptoms*, Diarrhoea*, Constipation

Common

Gastrointestinal haemorrhage (inc mucosal)*, Dyspepsia, Stomatitis*, Abdominal distension, Oropharyngeal pain*, Abdominal pain (inc gastrointestinal and splenic pain)*, Oral disorder*, Flatulence

Uncommon

Pancreatitis (inc chronic)*, Haematemesis, Lip swelling*, Gastrointestinal obstruction (inc small intestinal obstruction, ileus)*, Abdominal discomfort, Oral ulceration*, Enteritis*, Gastritis*, Gingival bleeding, Gastrooesophageal reflux disease*, Colitis (inc clostridium difficile)*, Colitis ischaemic#, Gastrointestinal inflammation*, Dysphagia, Irritable bowel syndrome, Gastrointestinal disorder NOS, Tongue coated, Gastrointestinal motility disorder*, Salivary gland disorder*

Rare

Pancreatitis acute, Peritonitis*, Tongue oedema*, Ascites, Oesophagitis, Cheilitis, Faecal incontinence, Anal sphincter atony, Faecaloma*, Gastrointestinal ulceration and perforation*, Gingival hypertrophy, Megacolon, Rectal discharge, Oropharyngeal blistering*, Lip pain, Periodontitis, Anal fissure, Change of bowel habit, Proctalgia, Abnormal faeces

Hepatobiliary disorders

Common

Hepatic enzyme abnormality*

Uncommon

Hepatotoxicity (inc liver disorder), Hepatitis*, Cholestasis

Rare

Hepatic failure, Hepatomegaly, Budd-Chiari syndrome, Cytomegalovirus hepatitis, Hepatic haemorrhage, Cholelithiasis

Skin and subcutaneous tissue disorders

Common

Rash*, Pruritus*, Erythema, Dry skin

Uncommon

Erythema multiforme, Urticaria, Acute febrile neutrophilic dermatosis, Toxic skin eruption, Toxic epidermal necrolysis#, Stevens-Johnson syndrome#, Dermatitis*, Hair disorder*, Petechiae, Ecchymosis, Skin lesion, Purpura, Skin mass*, Psoriasis, Hyperhidrosis, Night sweats, Decubitus ulcer#, Acne*, Blister*, Pigmentation disorder*

Rare

Skin reaction, Jessner's lymphocytic infiltration, Palmar-plantar erythrodysaesthesia syndrome, Haemorrhage subcutaneous, Livedo reticularis, Skin induration, Papule, Photosensitivity reaction, Seborrhoea, Cold sweat, Skin disorder NOS, Erythrosis, Skin ulcer, Nail disorder

Musculoskeletal and connective tissue disorders

Very Common

Musculoskeletal pain*

Common

Muscle spasms*, Pain in extremity, Muscular weakness

Uncommon

Muscle twitching, Joint swelling, Arthritis*, Joint stiffness, Myopathies*,Sensation of heaviness

Rare

Rhabdomyolysis, Temporomandibular joint syndrome, Fistula, Joint effusion, Pain in jaw, Bone disorder, Musculoskeletal and connective tissue infections and inflammations*, Synovial cyst

Renal and urinary disorders

Common

Renal impairment*

Uncommon

Renal failure acute, Renal failure chronic*, Urinary tract infection*, Urinary tract signs and symptoms*, Haematuria*, Urinary retention, Micturition disorder*, Proteinuria, Azotaemia, Oliguria*, Pollakiuria

Rare

Bladder irritation

Reproductive system and breast disorders

Uncommon

Vaginal haemorrhage, Genital pain*, Erectile dysfunction,

Rare

Testicular disorder*, Prostatitis, Breast disorder female, Epididymal tenderness, Epididymitis, Pelvic pain, Vulval ulceration

Congenital, familial and genetic disorders

Rare

Aplasia, Gastrointestinal malformation, Ichthyosis

General disorders and administration site conditions

Very Common

Pyrexia*, Fatigue, Asthenia

Common

Oedema (inc peripheral), Chills, Pain*, Malaise*

Uncommon

General physical health deterioration*, Face oedema*, Injection site reaction*, Mucosal disorder*, Chest pain, Gait disturbance, Feeling cold, Extravasation*, Catheter related complication*, Change in thirst*, Chest discomfort, Feeling of body temperature change*, Injection site pain*

Rare

Death (inc sudden), Multi-organ failure, Injection site haemorrhage*, Hernia(inc hiatus)*, Impaired healing*, Inflammation, Injection site phlebitis*, Tenderness, Ulcer, Irritability, Non-cardiac chest pain, Catheter site pain, Sensation of foreign body

Investigations

Common

Weight decreased

Uncommon

Hyperbilirubinaemia*, Protein analyses abnormal*, Weight increased, Blood test abnormal*,C-reactive protein increased

Rare

Blood gases abnormal*, Electrocardiogram abnormalities (inc QT prolongation)*, International normalised ratio abnormal*, Gastric pH decreased, Platelet aggregation increased, Troponin I increased, Virus identification and serology*, Urine analysis abnormal*

Injury, poisoning and procedural complications

Uncommon

Fall, Contusion

Rare

Transfusion reaction, Fractures*, Rigors*, Face injury, Joint injury*, Burns, Laceration, Procedural pain, Radiation injuries*

Surgical and medical procedures

Rare

Macrophage activation

NOS=not otherwise specified

* Grouping of more than one MedDRA preferred term.

# Postmarketing adverse reaction regardless of indication

 

Mantle Cell Lymphoma (MCL)

 

The safety profile of bortezomib in 240 MCL patients treated with bortezomib at 1.3 mg/m2 in combination with rituximab, cyclophosphamide, doxorubicin, and prednisone (VcR-CAP) versus 242 patients treated with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone [R-CHOP] was relatively consistent to that observed in patients with multiple myeloma with main differences described below. Additional adverse drug reactions identified associated with the use of the combination therapy (VcR-CAP) were hepatitis B infection (< 1%) and myocardial ischaemia (1.3%). The similar incidences of these events in both treatment arms, indicated that these adverse drug reactions are not attributable to bortezomib alone. Notable differences in the MCL patient population as compared to patients in the multiple myeloma studies were a ≥ 5% higher incidence of the haematological adverse reactions (neutropenia, thrombocytopenia, leukopenia, anemia, lymphopenia), peripheral sensory neuropathy, hypertension, pyrexia, pneumonia, stomatitis, and hair disorders.

 

Adverse drug reactions identified as those with a ≥ 1% incidence, similar or higher incidence in the VcR-CAP arm and with at least a possible or probable causal relationship to the components of the VcR-CAP arm, are listed in Table 8 below. Also included are adverse drug reactions identified in the VcR-CAP arm that were considered by investigators to have at least a possible or probable causal relationship to bortezomib based on historical data in the multiple myeloma studies.

 

Adverse reactions are listed below by system organ class and frequency grouping. Frequencies are defined as: Very common (≥ 1/10); common (≥ 1/100 to < 1/10); uncommon (≥ 1/1,000 to < 1/100); rare (≥ 1/10,000 to < 1/1,000); very rare (< 1/10,000), not known (cannot be estimated from the available data). Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness. Table 8 has been generated using Version 16 of the MedDRA.

 

Table 8 Adverse reactions in patients with Mantle Cell Lymphoma treated with VcR-CAP in a clinical trial

System Organ Class

Incidence

Adverse reaction

Infections and infestations

Very Common

Pneumonia*

Common

Sepsis (inc septic shock)*, Herpes zoster (inc disseminated & ophthalmic), Herpes virus infection*, Bacterial infections*, Upper/lower respiratory tract infection*, Fungal infection*, Herpes simplex*

Uncommon

Hepatitis B, Infection*, Bronchopneumonia

Blood and lymphatic system disorders

Very Common

Thrombocytopenia*, Febrile neutropenia, Neutropenia*, Leukopenia*, Anaemia*, Lymphopenia*

Uncommon

Pancytopenia*

Immune system disorders

Common

Hypersensitivity*

Uncommon

Anaphylactic reaction

Metabolism and nutrition disorders

Very Common

Decreased appetite

Common

Hypokalaemia*, Blood glucose abnormal*, Hyponatraemia*, Diabetes mellitus*, Fluid retention

Uncommon

Tumour lysis syndrome

Psychiatric disorders

Common

Sleep disorders and disturbances*

Nervous system disorders

Very Common

Peripheral sensory neuropathy, Dysaesthesia*, Neuralgia*

Common

Neuropathies*, Motor neuropathy*, Loss of consciousness (inc syncope), Encephalopathy*, Peripheral sensorimotor neuropathy, Dizziness*, Dysgeusia*, Autonomic neuropathy

Uncommon

Autonomic nervous system imbalance

Eye disorders

Common

Vision abnormal*

Ear and labyrinth disorders

Common

Dysacusis (inc tinnitus)*

Uncommon

Vertigo*, Hearing impaired (up to and inc deafness)

Cardiac disorders

Common

Cardiac fibrillation (inc atrial), Arrhythmia*, Cardiac failure (inc left and right ventricular)*, Myocardial ischaemia, Ventricular dysfunction*

Uncommon

Cardiovascular disorder (inc cardiogenic shock)

Vascular disorders

Common

Hypertension*, Hypotension*, Orthostatic hypotension

Respiratory, thoracic and mediastinal disorders

Common

Dyspnoea*, Cough*, Hiccups

Uncommon

Acute respiratory distress syndrome, Pulmonary embolism, Pneumonitis, Pulmonary hypertension, Pulmonary oedema (inc acute)

Gastrointestinal disorders

Very Common

Nausea and vomiting symptoms*, Diarrhoea*, Stomatitis*, Constipation

Common

Gastrointestinal haemorrhage (inc mucosal)*, Abdominal distension, Dyspepsia, Oropharyngeal pain*, Gastritis*, Oral ulceration*, Abdominal discomfort, Dysphagia, Gastrointestinal inflammation*, Abdominal pain (inc gastrointestinal and splenic pain)*, Oral disorder*

Uncommon

Colitis (inc clostridium difficile)*

Hepatobiliary disorders

Common

Hepatotoxicity (inc liver disorder)

Uncommon

Hepatic failure

Skin and subcutaneous tissue disorders

Very Common

Hair disorder*

Common

Pruritus*, Dermatitis*, Rash*

Musculoskeletal and connective tissue disorders

Common

Muscle spasms*, Musculoskeletal pain*, Pain in extremity

Renal and urinary disorders

Common

Urinary tract infection*

General disorders and administration site conditions

Very Common

Pyrexia*, Fatigue, Asthenia

Common

Oedema (inc peripheral), Chills, Injection site reaction*, Malaise*

Investigations

Common

Hyperbilirubinaemia*, Protein analyses abnormal*, Weight decreased, Weight increased

* Grouping of more than one MedDRA preferred term.

 

Description of selected adverse reactions

 

Herpes zoster virus reactivation

 

Multiple Myeloma

 

Antiviral prophylaxis was administered to 26% of the patients in the Vc+M+P arm. The incidence of herpes zoster among patients in the Vc+M+P treatment group was 17% for patients not administered antiviral prophylaxis compared to 3% for patients administered antiviral prophylaxis.

 

Mantle cell lymphoma

 

Antiviral prophylaxis was administered to 137 of 240 patients (57%) in the VcR-CAP arm. The incidence of herpes zoster among patients in the VcR-CAP arm was 10.7% for patients not administered antiviral prophylaxis compared to 3.6% for patients administered antiviral prophylaxis (see section 4.4).

 

Hepatitis B Virus (HBV) reactivation and infection

 

Mantle cell lymphoma

 

HBV infection with fatal outcomes occurred in 0.8% (n=2) of patients in the non-bortezomib treatment group (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone; R-CHOP ) and 0.4% (n=1) of patients receiving bortezomib in combination with rituximab, cyclophosphamide, doxorubicin, and prednisone (VcR-CAP). The overall incidence of hepatitis B infections was similar in patients treated with VcR-CAP or with R-CHOP (0.8% vs 1.2% respectively).

 

Peripheral neuropathy in combination regimens

 

Multiple Myeloma

 

In trials in which bortezomib was administered as induction treatment in combination with dexamethasone (study IFM- 2005-01), and dexamethasone-thalidomide (study MMY-3010), the incidence of peripheral neuropathy in the combination regimens is presented in the table below:

Table 9: Incidence of peripheral neuropathy during induction treatment by toxicity and  

              treatment discontinuation due to peripheral neuropathy

 

IFM-2005-01

MMY-3010

 

VDDx

(N=239)

VcDx

(N=239)

TDx

(N=126)

VcTDx

(N=130)

Incidence of PN (%)

 

 

 

 

All GradePN

3

15

12

45

≥ Grade 2 PN

1

10

2

31

≥ Grade 3 PN

<1

5

0

5

Discontinuation due to PN (%)

<1

2

1

5

VDDx=vincristine, doxorubicin, dexamethasone; VcDx= Bortezomib, dexamethasone; TDx=thalidomide, dexamethasone; VcTDx= Bortezomib, thalidomide, dexamethasone; PN=peripheral neuropathy

Note: Peripheral neuropathy included the preferred terms: neuropathy peripheral, peripheral motor neuropathy, peripheral sensory neuropathy, and polyneuropathy.

 

Mantle cell lymphoma

 

In study LYM-3002 in which bortezomib was administered with rituximab, cyclophosphamide, doxorubicin, and prednisone (R-CAP), the incidence of peripheral neuropathy in the combination regimens is presented in the table below:

 

Table 10: Incidence of peripheral neuropathy in study LYM-3002 by toxicity and treatment discontinuation due to peripheral neuropathy

 

VcR-CAP

(N=240)

R-CHOP

(N=242)

Incidence of PN (%)

 

 

All GradePN

30

29

≥ Grade 2 PN

18

9

≥ Grade 3 PN

8

4

Discontinuation due to PN (%)

2

< 1

VcR-CAP= bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone; R-CHOP= rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone; PN=peripheral neuropathy

Peripheral neuropathy included the preferred terms: peripheral sensory neuropathy, neuropathy peripheral, peripheral motor neuropathy, and peripheral sensorimotor neuropathy

 

Elderly MCL patients

42.9% and 10.4% of patients in the VcR-CAP arm were in the range 65-74 years and ≥ 75 years of age, respectively. Although in patients aged ≥ 75 years, both VcR-CAP and R-CHOP were less tolerated, the serious adverse event rate in the VcR-CAP groups was 68%, compared to 42% in the R-CHOP group.

 

Notable differences in the safety profile of bortezomib administered subcutaneously versus intravenously as single agent

 

In the Phase III study patients who received bortezomib subcutaneously compared to intravenous administration had 13% lower overall incidence of treatment emergent adverse reactions that were Grade 3 or higher in toxicity, and a 5% lower incidence of discontinuation of bortezomib. The overall incidence of diarrhoea, gastrointestinal and abdominal pain, asthenic conditions, upper respiratory tract infections and peripheral neuropathies were 12%-15% lower in the subcutaneous group than in the intravenous group. In addition, the incidence of Grade 3 or higher peripheral neuropathies was 10% lower, and the discontinuation rate due to peripheral neuropathies 8% lower for the subcutaneous group as compared to the intravenous group.

 

Six percent of patients had an adverse local reaction to subcutaneous administration, mostly redness. Cases resolved in a median of 6 days, dose modification was required in two patients. Two (1%) of the patients had severe reactions; 1 case of pruritus and 1 case of redness.

 

The incidence of death on treatment was 5% in the subcutaneous treatment group and 7% in the intravenous treatment group. Incidence of death from “Progressive disease” was 18% in the subcutaneous group and 9% in the intravenous group.

 

Retreatment of patients with relapsed multiple myeloma

 

In a study in which bortezomib retreatment was administered in 130 patients with relapsed multiple myeloma, who previously had at least partial response on a bortezomib -containing regimen, the most common all-grade adverse events occurring in at least 25% of patients were thrombocytopenia (55%), neuropathy (40%), anaemia (37%), diarrhoea (35%), and constipation (28%). All grade peripheral neuropathy and grade ≥ 3 peripheral neuropathy were observed in 40% and 8.5% of patients, respectively.

To report any side effect(s):

The National Pharmacovigilance and Drug Safety Centre (NPC)

o Fax: +966-11-205-7662

o Call NPC at +966-11-2038222, Exts: 2317-2356-2340.

o Reporting hotline: 19999

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

o Website: www.sfda.gov.sa/npc


In patients, overdose more than twice the recommended dose has been associated with the acute onset of symptomatic hypotension and thrombocytopenia with fatal outcomes. For preclinical cardiovascular safety pharmacology studies, see section 5.3.

 

There is no known specific antidote for bortezomib overdose. In the event of an overdose, the patient's vital signs should be monitored and appropriate supportive care given to maintain blood pressure (such as fluids, pressors, and/or inotropic agents) and body temperature (see sections 4.2 and 4.4).


Pharmacotherapeutic group: Antineoplastic agents, other antineoplastic agents, ATC code: L01XX32.

 

Mechanism of action

 

Bortezomib is a proteasome inhibitor. It is specifically designed to inhibit the chymotrypsin-like activity of the 26S proteasome in mammalian cells. The 26S proteasome is a large protein complex that degrades ubiquitinated proteins. The ubiquitin-proteasome pathway plays an essential role in regulating the turnover of specific proteins, thereby maintaining homeostasis within cells. Inhibition of the 26S proteasome prevents this targeted proteolysis and affects multiple signalling cascades within the cell, ultimately resulting in cancer cell death.

 

Bortezomib is highly selective for the proteasome. At 10 μM concentrations, bortezomib does not inhibit any of a wide variety of receptors and proteases screened and is more than 1,500-fold more selective for the proteasome than for its next preferable enzyme. The kinetics of proteasome inhibition were evaluated in vitro, and bortezomib was shown to dissociate from the proteasome with a t½ of 20 minutes, thus demonstrating that proteasome inhibition by bortezomib is reversible.

 

Bortezomib mediated proteasome inhibition affects cancer cells in a number of ways, including, but not limited to, altering regulatory proteins, which control cell cycle progression and nuclear factor kappa B (NF-kB) activation. Inhibition of the proteasome results in cell cycle arrest and apoptosis. NF-kB is a transcription factor whose activation is required for many aspects of tumourigenesis, including cell growth and survival, angiogenesis, cell-cell interactions, and metastasis. In myeloma, bortezomib affects the ability of myeloma cells to interact with the bone marrow microenvironment.

 

Experiments have demonstrated that bortezomib is cytotoxic to a variety of cancer cell types and that cancer cells are more sensitive to the pro-apoptotic effects of proteasome inhibition than normal cells. Bortezomib causes reduction of tumour growth in vivo in many preclinical tumour models, including multiple myeloma.

 

Data from in vitroex-vivo, and animal models with bortezomib suggest that it increases osteoblast differentiation and activity and inhibits osteoclast function. These effects have been observed in patients with multiple myeloma affected by an advanced osteolytic disease and treated with bortezomib.

 

Clinical efficacy in previously untreated multiple myeloma

 

A prospective Phase III, international, randomised (1:1), open-label clinical study (MMY-3002 VISTA) of 682 patients was conducted to determine whether Bortezomib (1.3 mg/m2 injected intravenously) in combination with melphalan (9 mg/m2) and prednisone (60 mg/m2) resulted in improvement in time to progression (TTP) when compared to melphalan (9 mg/m2) and prednisone (60 mg/m2) in patients with previously untreated multiple myeloma. Treatment was administered for a maximum of 9 cycles (approximately 54 weeks) and was discontinued early for disease progression or unacceptable toxicity. The median age of the patients in the study was 71 years, 50% were male, 88% were Caucasian and the median Karnofsky performance status score for the patients was 80. Patients had IgG/IgA/Light chain myeloma in 63%/25%/8% instances, a median hemoglobin of 105 g/l, and a median platelet count of 221.5 x 109/l. Similar proportions of patients had creatinine clearance ≤ 30 ml/min (3% in each arm).

 

At the time of a pre-specified interim analysis, the primary endpoint, time to progression, was met and patients in the M+P arm were offered Vc+M+P treatment. Median follow-up was 16.3 months. The final survival update was performed with a median duration of follow-up of 60.1 months. A statistically significant survival benefit in favour of the Vc+M+P treatment group was observed (HR=0.695; p=0.00043) despite subsequent therapies including Bortezomib-based regimens. Median survival for the Vc+M+P treatment group was 56.4 months compared to 43.1 for the M+P treatment group. Efficacy results are presented in Table 11:

Table 11: Efficacy results following the final survival update to VISTA study

Efficacy endpoint

Vc+M+P

n=344

M+P

n=338

Time to progression

Events n (%)

 

101 (29)

 

152 (45)

Mediana (95% CI)

20.7 mo

(17.6, 24,7)

15.0 mo

(14.1, 17.9)

Hazard ratiob

(95% CI)

0.54

(0.42, 0.70)

p-valuec

0.000002

Progression-free survival

Events n (%)

 

135 (39)

 

190 (56)

Mediana (95% CI)

18.3 mo

(16.6, 21.7)

14.0 mo

(11.1, 15.0)

Hazard ratiob

(95% CI)

0.61

(0.49, 0.76)

p-value c

0.00001

Overall survival*

Events (deaths) n (%)

 

176 (51.2)

 

211 (62.4)

Mediana

(95% CI)

56.4 mo

(52.8, 60.9)

43.1 mo

(35.3, 48.3)

Hazard ratiob

(95% CI)

0.695

(0.567, 0.852)

p-valuec

0.00043

Response rate

populatione n=668

n=337

n=331

CRf n (%)

102 (30)

12 (4)

PRf n (%)

136 (40)

103 (31)

nCR n (%)

5 (1)

0

CR+PRf n (%)

238 (71)

115 (35)

p-valued

< 10-10

Reduction in serum M-protein

populationg n=667

n=336

n=331

≥90% n (%)

151 (45)

34 (10)

Time to first response in CR + PR

 

Median

1.4 mo

4.2 mo

Mediana response duration

 

CRf

24.0 mo

12.8 mo

CR+PRf

19.9 mo

13.1 mo

Time to next therapy

Events n (%)

 

224 (65.1)

 

260 (76.9)

Mediana

(95% CI)

27.0 mo

(24.7, 31.1)

19.2 mo

(17.0, 21.0)

Hazard ratiob

(95% CI)

0.557

(0.462, 0.671)

p-valuec

< 0.000001

a Kaplan-Meier estimate.

b Hazard ratio estimate is based on a Cox proportional-hazard model adjusted for stratification factors: β2-microglobulin, albumin, and region. A hazard ratio less than 1 indicates an advantage for VMP

c Nominal p-value based on the stratified log-rank test adjusted for stratification factors: β2-microglobulin, albumin, and region

d p-value for Response Rate (CR+PR) from the Cochran Mantel-Haenszel chi-square test adjusted for the stratification factors

e Response population includes patients who had measurable disease at baseline

f CR=Complete Response; PR=Partial Response. EBMT criteria

g All randomised patients with secretory disease

* Survival update based on a median duration of follow-up at 60.1 months

mo: months

CI=Confidence Interval

 

Patients eligible for stem cell transplantation

 

Two randomised, open-label, multicenter Phase III trials (IFM-2005-01, MMY-3010) were conducted to demonstrate the safety and efficacy of bortezomib in dual and triple combinations with other chemotherapeutic agents, as induction therapy prior to stem cell transplantation in patients with previously untreated multiple myeloma.

 

In study IFM-2005-01 bortezomib combined with dexamethasone [VcDx, n=240] was compared to vincristine- doxorubicin-dexamethasone [VDDx, n=242]. Patients in the VcDx group received four 21 day cycles, each consisting of bortezomib (1.3 mg/m2 administered intravenously twice weekly on days 1, 4, 8, and 11), and oral dexamethasone (40 mg/day on days 1 to 4 and days 9 to 12, in Cycles 1 and 2, and on days 1 to 4 in Cycles 3 and 4).

 

Autologous stem cell transplants were received by 198 (82%) patients and 208 (87%) patients in the VDDx and VcDx groups respectively; the majority of patients underwent one single transplant procedure. Patient demographic and baseline disease characteristics were similar between the treatment groups. Median age of the patients in the study was 57 years, 55% were male and 48% of patients had high-risk cytogenetics. The median duration of treatment was 13 weeks for the VDDx group and 11 weeks for the VcDx group. The median number of cycles received for both groups was 4 cycles.

 

The primary efficacy endpoint of the study was post-induction response rate (CR+nCR). A statistically significant difference in CR+nCR was observed in favour of the bortezomib combined with dexamethasone group. Secondary efficacy endpoints included post-transplant response rates (CR+nCR, CR+nCR+VGPR+PR), Progression Free Survival and Overall Survival. Main efficacy results are presented in Table 12.

 

Table 12: Efficacy results from study IFM-2005-01

Endpoints

VcDx

VDDx

OR; 95% CI; P valuea

IFM-2005-01

N=240

(ITT population)

N=242

(ITT population)

 

RR (Post-induction)

*CR+nCR

CR+nCR+VGPR+PR % (95% CI)

 

14.6 (10.4, 19.7)

77.1 (71.2, 82.2)

 

6.2 (3.5, 10.0)

60.7 (54.3, 66.9)

 

2.58 (1.37, 4.85); 0.003

2.18 (1.46, 3.24); < 0.001

RR (Post-transplant)b

CR+nCR

CR+nCR+VGPR+PR % (95% CI)

 

37.5 (31.4, 44.0)

79.6 (73.9, 84.5)

 

23.1 (18.0, 29.0)

74.4 (68.4, 79.8)

 

1.98 (1.33, 2.95); 0.001

1.34 (0.87, 2.05); 0.179

CI=confidence interval; CR=complete response; nCR=near complete response; ITT=intent to treat; RR=response rate; Vc=bortezomib; VcDx=bortezomib, dexamethasone; VDDx=vincristine, doxorubicin, dexamethasone; VGPR=very good partial response; PR=partial response; OR=odds ratio.

* Primary endpoint

a OR for response rates based on Mantel-Haenszel estimate of the common odds ratio for stratified tables; p-values by Cochran Mantel-Haenszel test.

b Refers to response rate after second transplant for subjects who received a second transplant (42/240 [18% ] in VcDx group and 52/242 [21%] in VDDx group).

Note: An OR > 1 indicates an advantage for Vc-containing induction therapy.

 

In study MMY-3010 induction treatment with bortezomib combined with thalidomide and dexamethasone [VcTDx, n=130] was compared to thalidomide-dexamethasone [TDx, n=127]. Patients in the VcTDx group received six 4-week cycles, each consisting of bortezomib (1.3 mg/m2 administered twice weekly days 1, 4, 8, and 11, followed by a 17-day rest period from day 12 to day 28), dexamethasone (40 mg administered orally on days 1 to 4 and days 8 to 11), and thalidomide (administered orally at 50 mg daily on days 1-14, increased to 100 mg on days 15-28 and thereafter to 200 mg daily).

 

One single autologous stem cell transplant was received by 105 (81%) patients and 78 (61%) patients in the VcTDx and TDx groups, respectively. Patient demographic and baseline disease characteristics were similar between the treatment groups. Patients in the VcTDx and TDx groups respectively had a median age of 57 versus 56 years, 99% versus 98% patients were Caucasians, and 58% versus 54% were males. In the VcTDx group 12% of patients were cytogenetically classified as high risk versus 16% of patients in the TDx group. The median duration of treatment was 24.0 weeks and the median number of treatment cycles received was 6.0, and was consistent across treatment groups.

 

The primary efficacy endpoints of the study were post-induction and post-transplant response rates (CR+nCR). A statistically significant difference in CR+nCR was observed in favour of the bortezomib combined with dexamethasone and thalidomide group. Secondary efficacy endpoints included Progression Free Survival and Overall Survival. Main efficacy results are presented in Table 13.

 

Table 13: Efficacy results from study MMY-3010

Endpoints

VcTDx

TDx

OR; 95% CI; P valuea

MMY-3010

N=130

(ITT population)

N=127

(ITT population)

 

*RR (Post-induction)

CR+nCR

CR+nCR+PR % (95% CI)

 

49.2 (40.4, 58.1)

84.6 (77.2, 90.3)

 

17.3 (11.2, 25.0)

61.4 (52.4, 69.9)

 

4.63 (2.61, 8.22); < 0.001a

3.46 (1.90, 6.27); < 0.001a

*RR (Post-transplant)

CR+nCR

CR+nCR+PR % (95% CI)

 

55.4 (46.4, 64.1)

77.7 (69.6, 84.5)

 

34.6 (26.4, 43.6)

56.7 (47.6, 65.5)

 

2.34 (1.42, 3.87); 0.001a

2.66 (1.55, 4.57); < 0.001a

CI=confidence interval; CR=complete response; nCR=near complete response; ITT=intent to treat; RR=response rate; Vc=Bortezomib; VcTDx=bortezomib, thalidomide, dexamethasone; TDx=thalidomide, dexamethasone; PR=partial response; OR=odds ratio

* Primary endpoint

a OR for response rates based on Mantel-Haenszel estimate of the common odds ratio for stratified tables; p-values by Cochran Mantel-Haenszel test.

Note: An OR > 1 indicates an advantage for Vc-containing induction therapy

 

Clinical efficacy in relapsed or refractory multiple myeloma

The safety and efficacy of bortezomib (injected intravenously) were evaluated in 2 studies at the recommended dose of 1.3 mg/m2: a Phase III randomised, comparative study (APEX), versus dexamethasone (Dex), of 669 patients with relapsed or refractory multiple myeloma who had received 1-3 prior lines of therapy, and a Phase II single-arm study of 202 patients with relapsed and refractory multiple myeloma, who had received at least 2 prior lines of treatment and who were progressing on their most recent treatment.

 

In the Phase III study, treatment with bortezomib led to a significantly longer time to progression, a significantly prolonged survival and a significantly higher response rate, compared to treatment with dexamethasone (see Table 14), in all patients as well as in patients who have received 1 prior line of therapy. As a result of a pre-planned interim analysis, the dexamethasone arm was halted at the recommendation of the data monitoring committee and all patients randomised to dexamethasone were then offered bortezomib, regardless of disease status. Due to this early crossover, the median duration of follow-up for surviving patients is 8.3 months. Both in patients who were refractory to their last prior therapy and those who were not refractory, overall survival was significantly longer and response rate was significantly higher on the bortezomib arm.

 

Of the 669 patients enrolled, 245 (37%) were 65 years of age or older. Response parameters as well as TTP remained significantly better for bortezomib independently of age. Regardless of β2-microglobulin levels at baseline, all efficacy parameters (time to progression and overall survival, as well as response rate) were significantly improved on the bortezomib arm.

 

In the refractory population of the Phase II study, responses were determined by an independent review committee and the response criteria were those of the European Bone Marrow Transplant Group. The median survival of all patients enrolled was 17 months (range < 1 to 36+ months). This survival was greater than the six-to-nine month median survival anticipated by consultant clinical investigators for a similar patient population. By multivariate analysis, the response rate was independent of myeloma type, performance status, chromosome 13 deletion status, or the number or type of previous therapies. Patients who had received 2 to 3 prior therapeutic regimens had a response rate of 32% (10/32) and patients who received greater than 7 prior therapeutic regimens had a response rate of 31% (21/67).

 

Table 14: Summary of disease outcomes from the Phase III (APEX) and Phase II studies

 

Phase III

Phase III

Phase III

Phase II

 

All patients

1 prior line of therapy

> 1 prior line of therapy

≥ 2 prior lines

Time related events

Vc

n=333a

Dex

n=336a

Vc

n=132a

Dex

n=119a

Vc

n=200a

Dex

n=217a

Vc

n=202a

TTP, days

[95% CI]

189b

[148, 211]

106b

[86, 128]

212d

[188, 267]

169d

[105, 191]

148b

[129, 192]

87b

[84, 107]

210

[154, 281]

1 year survival, %

[95% CI]

80d

[74,85]

66d

[59,72]

89d

[82,95]

72d

[62,83]

73

[64,82]

62

[53,71]

 

60

Best response (%)

Vc

n=315c

Dex

n=312c

Vc

n=128

Dex

n=110

Vc

n=187

Dex

n=202

Vc

n=193

CR

20 (6)b

2 (< 1)b

8 (6)

2 (2)

12 (6)

0 (0)

(4)**

CR+nCR

41 (13)b

5 (2)b

16 (13)

4 (4)

25 (13)

1 (< 1)

(10)**

CR+nCR+PR

121 (38)b

56 (18)b

57 (45)d

29 (26)d

64 (34)b

27 (13)b

(27)**

CR+nCR+ PR+MR

146 (46)

108 (35)

66 (52)

45 (41)

80 (43)

63 (31)

(35)**

Median duration

Days (months)

242 (8.0)

169 (5.6)

246 (8.1)

189 (6.2)

238 (7.8)

126 (4.1)

385*

Time to response

CR+PR (days)

43

43

44

46

41

27

38*

a Intent to Treat (ITT) population

b p-value from the stratified log-rank test; analysis by line of therapy excludes stratification for therapeutic history; p < 0.0001

Response population includes patients who had measurable disease at baseline and received at least 1 dose of study medicinal product.

p-value from the Cochran Mantel-Haenszel chi-square test adjusted for the stratification factors; analysis by line of therapy excludes stratification for therapeutic history

* CR+PR+MR **CR=CR, (IF-); nCR=CR (IF+)

NA=not applicable, NE=not estimated

TTP-Time to Progression

CI=Confidence Interval

Vc=Bortezomib; Dex=dexamethasone

CR=Complete Response; nCR=near Complete response

PR=Partial Response; MR=Minimal response

 

In the Phase II study, patients who did not obtain an optimal response to therapy with bortezomib alone were able to receive high-dose dexamethasone in conjunction with bortezomib. The protocol allowed patients to receive dexamethasone if they had had a less than optimal response to bortezomib alone. A total of 74 evaluable patients were administered dexamethasone in combination with bortezomib. Eighteen percent of patients achieved, or had an improved response [MR (11%) or PR (7%)] with combination treatment.

 

Clinical efficacy with subcutaneous administration of bortezomib in patients with relapsed/refractory multiple myeloma

 

An open label, randomised, Phase III non-inferiority study compared the efficacy and safety of the subcutaneous administration of bortezomib versus the intravenous administration. This study included 222 patients with relapsed/refractory multiple myeloma, who were randomised in a 2:1 ratio to receive 1.3 mg/m2 of bortezomib by either the subcutaneous or intravenous route for 8 cycles. Patients who did not obtain an optimal response (less than Complete Response [CR]) to therapy with bortezomib alone after 4 cycles were allowed to receive dexamethasone 20 mg daily on the day of and after bortezomib administration. Patients with baseline Grade ≥ 2 peripheral neuropathy or platelet counts < 50,000/µl were excluded. A total of 218 patients were evaluable for response.

 

This study met its primary objective of non-inferiority for response rate (CR+PR) after 4 cycles of single agent bortezomib for both the subcutaneous and intravenous routes, 42% in both groups. In addition, secondary response-related and time to event related efficacy endpoints showed consistent results for subcutaneous and intravenous administration (Table 15).

 

Table 15: Summary of efficacy analyses comparing subcutaneous and intravenous administrations of bortezomib

 

Bortezomib intravenous arm

Bortezomib subcutaneous arm

Response Evaluable Population

n=73

n=145

Response Rate at 4 cycles n (%)

 

 

ORR (CR+PR)

31 (42)

61 (42)

p-valuea

0.00201

CR n (%)

6 (8)

9 (6)

PR n (%)

25 (34)

52 (36)

nCR n (%)

4 (5)

9 (6)

Response Rate at 8 cycles n (%)

 

 

ORR (CR+PR)

38 (52)

76 (52)

p-valuea

0.0001

CR n (%)

9 (12)

15 (10)

PR n (%)

29 (40)

61 (42)

nCR n (%)

7 (10)

14 (10)

Intent to Treat Populationb

n=74

n=148

TTP, months

9.4

10.4

(95% CI)

(7.6, 10.6)

(8.5, 11.7)

Hazard ratio (95% CI)c

p-valued

0.839 (0.564, 1.249)

0.38657

Progression Free Survival, months

8.0

10.2

(95% CI)

(6.7, 9.8)

(8.1, 10.8)

Hazard ratio (95% CI)c

p-valued

0.824 (0.574, 1.183)

0.295

1-year Overall Survival (%)e

76.7

72.6

(95% CI)

(64.1, 85.4)

(63.1, 80.0)

a p-value is for the non-inferiority hypothesis that the SC arm retains at least 60% of the response rate in the IV arm.

b 222 subjects were enrolled into the study; 221 subjects were treated with bortezomib

c Hazards ratio estimate is based on a Cox model adjusted for stratification factors: ISS staging and number of prior lines.

d Log rank test adjusted for stratification factors: ISS staging and number of prior lines.

e Median duration of follow up is 11.8 months

 

Bortezomib combination treatment with pegylated liposomal doxorubicin (study DOXIL-MMY-3001)

 

A Phase III randomised, parallel-group, open-label, multicentre study was conducted in 646 patients comparing the safety and efficacy of bortezomib plus pegylated liposomal doxorubicin versus bortezomib monotherapy in patients with multiple myeloma who had received at least 1 prior therapy and who did not progress while receiving anthracycline-based therapy. The primary efficacy endpoint was TTP while the secondary efficacy endpoints were OS and ORR (CR+PR), using the European Group for Blood and Marrow Transplantation (EBMT) criteria.

 

A protocol-defined interim analysis (based on 249 TTP events) triggered early study termination for efficacy. This interim analysis showed a TTP risk reduction of 45% (95% CI; 29-57%, p < 0.0001) for patients treated with combination therapy of bortezomib and pegylated liposomal doxorubicin. The median TTP was 6.5 months for the bortezomib monotherapy patients compared with 9.3 months for the bortezomib plus pegylated liposomal doxorubicin combination therapy patients. These results, though not mature, constituted the protocol defined final analysis.

 

The final analysis for OS performed after a median follow-up of 8.6 years showed no significant difference in OS between the two treatment arms. The median OS was 30.8 months (95% CI; 25.2-36.5 months) for the Bortezomib monotherapy patients and 33.0 months (95% CI; 28.9-37.1 months) for the Bortezomib plus pegylated liposomal doxorubicin combination therapy patients.

 

Bortezomib combination treatment with dexamethasone

In the absence of any direct comparison between bortezomib and bortezomib in combination with dexamethasone in patients with progressive multiple myeloma, a statistical matched-pair analysis was conducted to compare results from the non randomised arm of bortezomib in combination with dexamethasone (Phase II open-label study MMY-2045), with results obtained in the bortezomib monotherapy arms from different Phase III randomised studies (M34101-039 [APEX] and DOXIL MMY-3001) in the same indication.

 

The matched-pair analysis is a statistical method in which patients in the treatment group (e.g. bortezomib in combination with dexamethasone) and patients in the comparison group (e.g. bortezomib) are made comparable with respect to confounding factors by individually pairing study subjects. This minimises the effects of observed confounders when estimating treatment effects using non-randomised data.

 

One hundred and twenty-seven matched pairs of patients were identified. The analysis demonstrated improved ORR (CR+PR) (odds ratio 3.769; 95% CI 2.045-6.947; p < 0.001), PFS (hazard ratio 0.511; 95% CI 0.309-0.845; p=0.008), TTP (hazard ratio 0.385; 95% CI 0.212-0.698; p=0.001) for bortezomib in combination with dexamethasone over bortezomib monotherapy.

 

Limited information on bortezomib retreatment in relapsed multiple myeloma is available.

Phase II study MMY-2036 (RETRIEVE), single arm, open-label study was conducted to determine the efficacy and safety of retreatment with bortezomib. One hundred and thirty patients (≥ 18 years of age) with multiple myeloma who previously had at least partial response on a bortezomib-containing regimen were retreated upon progression. At least 6 months after prior therapy, bortezomib was started at the last tolerated dose of 1.3 mg/m2 (n=93) or ≤ 1.0 mg/m2 (n=37) and given on days 1, 4, 8 and 11 every 3 weeks for maximum of 8 cycles either as single agent or in combination with dexamethasone in accordance with the standard of care. Dexamethasone was administered in combination with bortezomib to 83 patients in Cycle 1 with an additional 11 patients receiving dexamethasone during the course of bortezomib retreatment cycles.

 

The primary endpoint was best confirmed response to retreatment as assessed by EBMT criteria. The overall best response rate (CR + PR), to retreatment in 130 patients was 38.5% (95% CI: 30.1, 47.4).

 

Clinical efficacy in previously untreated mantle cell lymphoma (MCL)

 

Study LYM-3002 was a Phase III, randomised, open-label study comparing the efficacy and safety of the combination of BORTEZOMIB, rituximab, cyclophosphamide, doxorubicin, and prednisone (VcR-CAP; n=243) to that of rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP; n=244) in adult patients with previously untreated MCL (Stage II, III or IV). Patients in the VcR-CAP treatment arm received bortezomib (1.3 mg/m2; on days 1, 4, 8, 11, rest period days 12-21), rituximab 375 mg/m2 IV on day 1; cyclophosphamide 750 mg/m2 IV on day 1; doxorubicin 50 mg/m2 IV on day 1; and prednisone 100 mg/m2 orally on day 1 through day 5 of the 21 day bortezomib treatment cycle. For patients with a response first documented at cycle 6, two additional treatment cycles were given.

 

The primary efficacy endpoint was progression-free survival based on Independent Review Committee (IRC) assessment. Secondary endpoints included, time to progression (TTP), time to next anti-lymphoma treatment (TNT), duration of treatment free interval (TFI), overall response rate (ORR) and complete response (CR/CRu) rate, overall survival (OS) and response duration.

 

The demographic and baseline disease characteristics were generally well balanced between the two treatment arms: median patient age was 66 years, 74% were male, 66% were Caucasian and 32% Asian, 69% of patients had a positive bone marrow aspirate and/or a positive bone marrow biopsy for MCL, 54% of patients had an International Prognostic Index (IPI) score of ≥ 3, and 76% had Stage IV disease. Treatment duration (median=17 weeks) and duration of follow-up (median=40 months) were comparable in both treatment arms. A median of 6 cycles was received by patients in both treatment arms with 14% of subjects in the VcR-CAP group and 17% of patients in the R-CHOP group receiving 2 additional cycles. The majority of the patients in both groups completed treatment, 80% in the VcR-CAP group and 82% in the R-CHOP group. Efficacy results are presented in Table 16:

 

Table 16: Efficacy results from study LYM-3002

Efficacy endpoint

VcR-CAP

R-CHOP

 

n: ITT patients

243

244

Progression free survival (IRC)a

Events n (%)

133 (54.7%)

165 (67.6%)

HRb (95% CI)=0.63 (0.50; 0.79)

p-value< 0.001

Medianc(95% CI) (months)

24.7 (19.8; 31.8)

14.4 (12; 16.9)

Response rate

n: response-evaluable patients

229

228

 

Overall complete response (CR+CRu)f n(%)

122 (53.3%)

95 (41.7%)

ORe (95% CI)=1.688 (1.148; 2.481)

p-valueg=0.007

Overall response (CR+CRu+PR)h n(%)

211 (92.1%)

204 (89.5%)

OR(95% CI)=1.428 (0.749; 2.722)

p-valueg=0.275

a Based on Independent Review Committee (IRC) assessment (radiological data only).

b Hazard ratio estimate is based on a Cox's model stratified by IPI risk and stage of disease. A hazard ratio < 1 indicates an advantage for VcR-CAP.

c Based on Kaplan-Meier product limit estimates.

d Based on Log rank test stratified with IPI risk and stage of disease.

e Mantel-Haenszel estimate of the common odds ratio for stratified tables is used, with IPI risk and stage of disease as stratification factors. An odds ratio (OR) > 1 indicates an advantage for VcR-CAP.

f Include all CR+CRu, by IRC, bone marrow and LDH.

g P-value from the Cochran Mantel-Haenszel chi-square test, with IPI and stage of disease as stratification factors.

h Include all radiological CR+CRu+PR by IRC regardless the verification by bone marrow and LDH.

CR=Complete Response; CRu=Complete Response unconfirmed; PR=Partial Response; CI=Confidence Interval, HR=Hazard Ratio; OR=Odds Ratio; ITT=Intent to Treat

 

Median PFS by investigator assessment was 30.7 months in the VcR-CAP group and 16.1 months in the R-CHOP group (Hazard Ratio [HR]=0.51; p < 0.001). A statistically significant benefit (p < 0.001) in favour of the VcR-CAP treatment group over the R-CHOP group was observed for TTP (median 30.5 versus 16.1 months), TNT (median 44.5 versus 24.8 months) and TFI (median 40.6 versus 20.5 months). The median duration of complete response was 42.1 months in the VcR-CAP group compared with 18 months in the R-CHOP group. The duration of overall response was 21.4 months longer in the VcR-CAP group (median 36.5 months versus 15.1 months in the R-CHOP group). The final analysis for OS was performed after a median follow-up of 82 months. Median OS was 90.7 months for the VcR-CAP group compared with 55.7 months for the R-CHOP group (HR=0.66; p=0.001). The observed final median difference in the OS between the 2 treatment groups was 35 months.

 

Patients with previously treated light-chain (AL) Amyloidosis

An open label non randomised Phase I/II study was conducted to determine the safety and efficacy of bortezomib in patients with previously treated light-chain (AL) Amyloidosis. No new safety concerns were observed during the study, and in particular bortezomib did not exacerbate target organ damage (heart, kidney and liver). In an exploratory efficacy analysis, a 67.3% response rate (including a 28.6% CR rate) as measured by hematologic response (M-protein) was reported in 49 evaluable patients treated with the maximum allowed doses of 1.6 mg/m2 weekly and 1.3 mg/m2 twice-weekly. For these dose cohorts, the combined 1-year survival rate was 88.1%.

 

Paediatric population

The European Medicines Agency has waived the obligation to submit the results of studies with bortezomib in all subsets of the paediatric population in multiple myeloma and in mantle cell lymphoma (see section 4.2 for information on paediatric use).

A Phase II, single arm activity, safety, and pharmacokinetic trial conducted by the Children’s

Oncology Group assessed the activity of the addition of bortezomib to multi agent re induction chemotherapy in paediatric and young adult patients with lymphoid malignancies (pre-B cell acute lymphoblastic leukemia [ALL], T-cell ALL, and T-cell lymphoblastic lymphoma [LL]). An effective reinduction multiagent chemotherapy regimen was administered in 3 blocks. Bortezomib was administered only in Blocks 1 and 2 to avoid potential overlapping toxicities with coadministered drugs in Block 3.

Complete response (CR) was evaluated at the end of Block 1. In B-ALL patients with relapse within 18 months of diagnosis (n = 27) the CR rate was 67% (95% CI: 46, 84); the 4-month event free survival rate was 44% (95% CI: 26, 62). In B-ALL patients with relapse 18 36 months from diagnosis (n = 33) the CR rate was 79% (95% CI: 61, 91) and the 4-month event free survival rate was 73% (95% CI: 54, 85). The CR rate in first-relapsed T-cell ALL patients (n = 22) was 68% (95% CI: 45, 86) and the 4-month event free survival rate was 67% (95% CI: 42, 83). The reported efficacy data are considered inconclusive (see section 4.2).

 

There were 140 patients with ALL or LL enrolled and evaluated for safety; median age was 10 years (range 1 to 26). No new safety concerns were observed when bortezomib was added to the standard pediatric pre B cell ALL chemotherapy backbone. The following adverse reactions (Grade ≥ 3) were observed at a higher incidence in the bortezomib containing treatment regimen as compared with a historical control study in which the backbone regimen was given alone: in Block 1 peripheral sensory neuropathy (3% versus 0%); ileus (2.1% versus 0%); hypoxia (8% versus 2%). No information on possible sequelae or rates of peripheral neuropathy resolution were available in this study. Higher incidences were also noted for infections with Grade ≥ 3 neutropenia (24% versus 19% in Block 1 and 22% versus 11% in Block 2), increased ALT (17% versus 8% in Block 2), hypokalaemia (18% versus 6% in Block 1 and 21% versus 12% in Block 2) and hyponatraemia (12% versus 5% in Block 1 and 4% versus 0 in Block 2).


Absorption

Following intravenous bolus administration of a 1.0 mg/m2 and 1.3 mg/m2 dose to 11 patients with multiple myeloma and creatinine clearance values greater than 50 ml/min, the mean first-dose maximum plasma concentrations of bortezomib were 57 and 112 ng/ml, respectively. In subsequent doses, mean maximum observed plasma concentrations ranged from 67 to 106 ng/ml for the 1.0 mg/m2 dose and 89 to 120 ng/ml for the 1.3 mg/m2 dose.

 

Following an intravenous bolus or subcutaneous injection of a 1.3 mg/m2 dose to patients with multiple myeloma (n=14 in the intravenous group, n=17 in the subcutaneous group), the total systemic exposure after repeat dose administration (AUClast) was equivalent for subcutaneous and intravenous administrations. The Cmax after subcutaneous administration (20.4 ng/ml) was lower than intravenous (223 ng/ml). The AUClast- geometric mean ratio was 0.99 and 90% confidence intervals were 80.18%-122.80%.

 

Distribution

The mean distribution volume (Vd) of bortezomib ranged from 1,659 l to 3,294 l following single- or repeated-dose intravenous administration of 1.0 mg/m2 or 1.3 mg/m2 to patients with multiple myeloma. This suggests that bortezomib distributes widely to peripheral tissues. Over a bortezomib concentration range of 0.01 to 1.0 μg/ml, the in vitro protein binding averaged 82.9% in human plasma. The fraction of bortezomib bound to plasma proteins was not concentration-dependent.

 

Biotransformation

In vitro studies with human liver microsomes and human cDNA-expressed cytochrome P450 isozymes indicate that bortezomib is primarily oxidatively metabolised via cytochrome P450 enzymes, 3A4, 2C19, and 1A2. The major metabolic pathway is deboronation to form two deboronated metabolites that subsequently undergo hydroxylation to several metabolites. Deboronated-bortezomib metabolites are inactive as 26S proteasome inhibitors.

 

Elimination

The mean elimination half-life (t1/2) of bortezomib upon multiple dosing ranged from 40-193 hours. Bortezomib is eliminated more rapidly following the first dose compared to subsequent doses. Mean total body clearances were 102 and 112 l/h following the first dose for doses of 1.0 mg/m2 and 1.3 mg/m2, respectively, and ranged from 15 to 32 l/h and 18 to 32 l/h following subsequent doses for doses of 1.0 mg/m2 and 1.3 mg/m2, respectively.

 

Special populations

 

Hepatic impairment

The effect of hepatic impairment on the pharmacokinetics of bortezomib was assessed in a Phase I study during the first treatment cycle, including 61 patients primarily with solid tumors and varying degrees of hepatic impairment at bortezomib doses ranging from 0.5 to 1.3 mg/m2.

 

When compared to patients with normal hepatic function, mild hepatic impairment did not alter dose-normalised bortezomib AUC. However, the dose-normalised mean AUC values were increased by approximately 60% in patients with moderate or severe hepatic impairment. A lower starting dose is recommended in patients with moderate or severe hepatic impairment, and those patients should be closely monitored (see section 4.2, Table 6).

 

Renal impairment

A pharmacokinetic study was conducted in patients with various degrees of renal impairment who were classified according to their creatinine clearance values (CrCL) into the following groups: Normal (CrCL ≥ 60 ml/min/1.73 m2, n=12), Mild (CrCL=40-59 ml/min/1.73 m2, n=10), Moderate (CrCL=20-39 ml/min/1.73 m2, n=9), and Severe (CrCL < 20 ml/min/1.73 m2, n=3). A group of dialysis patients who were dosed after dialysis was also included in the study (n=8). Patients were administered intravenous doses of 0.7 to 1.3 mg/m2 of bortezomib twice weekly. Exposure of bortezomib (dose-normalised AUC and Cmax) was comparable among all the groups (see section 4.2).

 

Age:

The pharmacokinetics of bortezomib were characterized following twice weekly intravenous bolus administration of 1.3 mg/m2 doses to 104 paediatric patients (2-16 years old) with acute lymphoblastic leukemia (ALL) or acute myeloid leukemia (AML). Based on a population pharmacokinetic analysis, clearance of bortezomib increased with increasing body surface area (BSA). Geometric mean (%CV) clearance was 7.79 (25%) L/hr/m2, volume of distribution at steady-state was 834 (39%) L/m2, and the elimination half-life was 100 (44%) hours. After correcting for the BSA effect, other demographics such as age, body weight and sex did not have clinically significant effects on bortezomib clearance. BSA-normalized clearance of bortezomib in paediatric patients was similar to that observed in adults.

 


 

 

Bortezomib was positive for clastogenic activity (structural chromosomal aberrations) in the in vitro chromosomal aberration assay using Chinese hamster ovary (CHO) cells at concentrations as low as 3.125 μg/ml, which was the lowest concentration evaluated. Bortezomib was not genotoxic when tested in the in vitro mutagenicity assay (Ames assay) and in vivo micronucleus assay in mice.

 

Developmental toxicity studies in the rat and rabbit have shown embryo-fetal lethality at maternally toxic doses, but no direct embryo-foetal toxicity below maternally toxic doses. Fertility studies were not performed but evaluation of reproductive tissues has been performed in the general toxicity studies. In the 6-month rat study, degenerative effects in both the testes and the ovary have been observed. It is, therefore, likely that bortezomib could have a potential effect on either male or female fertility. Peri- and postnatal development studies were not conducted.

 

In multi-cycle general toxicity studies conducted in the rat and monkey, the principal target organs included the gastrointestinal tract, resulting in vomiting and/or diarrhoea; haematopoietic and lymphatic tissues, resulting in peripheral blood cytopenias, lymphoid tissue atrophy and haematopoietic bone marrow hypocellularity; peripheral neuropathy (observed in monkeys, mice and dogs) involving sensory nerve axons; and mild changes in the kidneys. All these target organs have shown partial to full recovery following discontinuation of treatment.

 

Based on animal studies, the penetration of bortezomib through the blood-brain barrier appears to be limited, if any and the relevance to humans is unknown.

 

Cardiovascular safety pharmacology studies in monkeys and dogs show that intravenous doses approximately two to three times the recommended clinical dose on a mg/m2 basis are associated with increases in heart rate, decreases in contractility, hypotension and death. In dogs, the decreased cardiac contractility and hypotension responded to acute intervention with positive inotropic or pressor agents. Moreover, in dog studies, a slight increase in the corrected QT interval was observed.


Mannitol (E421) and nitrogen.

 


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

 


Unopened vial 2 years Intravenous administration The chemical and physical in-use stability of the reconstituted solution at a concentration of 1 mg/ml has been demonstrated for 8 hours at 20°C-25°C stored in the original vial and/or a syringe. From a microbiological point of view, unless the method of opening/reconstitution/dilution precludes the risk of microbial contamination, the reconstituted solution should be used immediately after preparation. If not used immediately, in-use storage times and conditions prior to use are the responsibility of the user. Subcutaneous administration The chemical and physical in -use stability of the reconstituted solution of 2.5 mg/ml has been demonstrated for 8 hours at 20°C-25°C stored in the original vial and/or a syringe. From a microbiological point of view, unless the method of opening/reconstitution/dilution precludes the risk of microbial contamination, the reconstituted solution should be used immediately after preparation. If not used immediately, in-use storage times and conditions prior to use are the responsibility of the user.

This medicinal product does not require any special storage conditions.

Keep the vial in the outer carton in order to protect from light.

Do not store above 30°C.

For storage conditions after reconstitution of the medicinal product, see section 6.3.


Type 1 glass 10 ml capacity vial with a dark grey bromobutyl stopper and an aluminium seal, with a light blue cap containing 3.5 mg bortezomib.

 

Each pack contains 1 single use vial.


General precautions

Bortezomib is a cytotoxic agent. Therefore, caution should be used during handling and preparation of bortezomib. Use of gloves, goggles and other protective clothing to prevent skin contact is recommended.

 

Aseptic technique must be strictly observed throughout the handling of Bortezomib, since it contains no preservative.

 

There have been fatal cases of inadvertent intrathecal administration of bortezomib. Bortezomib 3.5 mg powder for solution for injection is for intravenous or subcutaneous use. Bortezomib should not be administered intrathecally.

Instructions for reconstitution

Bortezomib must be reconstituted by a healthcare professional.

 

Intravenous injection

Each 10 ml vial of bortezomib must be carefully reconstituted with 3.5 ml of sodium chloride 9 mg/ml (0.9%) solution for injection, by using a syringe of the appropriate size, without removing the vial stopper. Dissolution of the lyophilised powder is completed in less than 2 minutes.

 

After reconstitution, each ml solution contains 1 mg bortezomib. The reconstituted solution is clear and colourless, with a final pH of 4 to 7.

The reconstituted solution must be inspected visually for particulate matter and discolouration prior to administration. If any discolouration or particulate matter is observed, the reconstituted solution must be discarded.

 

Subcutaneous injection

Each 10 ml vial of Bortezomib must be carefully reconstituted with 1.4 ml of sodium chloride 9 mg/ml (0.9%) solution for injection, by using a syringe of the appropriate size, without removing the vial stopper. Dissolution of the lyophilised powder is completed in less than 2 minutes.

 

After reconstitution, each ml solution contains 2.5 mg bortezomib. The reconstituted solution is clear and colourless, with a final pH of 4 to 7. The reconstituted solution must be inspected visually for particulate matter and discolouration prior to administration. If any discolouration or particulate matter is observed, the reconstituted solution must be discarded.

 

Disposal

Bortezomib is for single use only. Any unused medicinal product or waste material should be disposed of in accordance with local requirements.


Manufactured by Shilpa Medicare Limited for: SPIMACO AlQassim pharmaceutical plant Saudi Pharmaceutical Industries & Medical Appliance Corporation.

August 2019.
}

صورة المنتج على الرف

الصورة الاساسية