Consumer medicine information

Xtandi

Enzalutamide

BRAND INFORMATION

Brand name

Xtandi

Active ingredient

Enzalutamide

Schedule

S4

 

Consumer medicine information (CMI) leaflet

Please read this leaflet carefully before you start using Xtandi.

SUMMARY CMI

XTANDI®

Consumer Medicine Information (CMI) summary

The full CMI on the next page has more details. If you are worried about using this medicine, speak to your doctor or pharmacist.

1. Why am I using XTANDI?

XTANDI contains the active ingredient enzalutamide. XTANDI is used to treat adult men with prostate cancer that no longer responds to hormone therapy or surgical treatment to lower testosterone, or has spread to other parts of the body and is considered "hormone sensitive".

For more information, see Section 1. Why am I using XTANDI? in the full CMI.

2. What should I know before I use XTANDI?

Do not use if you have ever had an allergic reaction to XTANDI or any of the ingredients listed at the end of the CMI.

Talk to your doctor if you have any other medical conditions, take any other medicines, or are pregnant or plan to become pregnant, or are breastfeeding. XTANDI is not for use in women.

For more information, see Section 2. What should I know before I use XTANDI? in the full CMI.

3. What if I am taking other medicines?

Some medicines may interfere with XTANDI and affect how it works.

A list of these medicines is in Section 3. What if I am taking other medicines? in the full CMI.

4. How do I use XTANDI?

  • The usual dose is four 40 mg capsules taken at the same time once a day. Your doctor may reduce your dose depending on your medical conditions.

More instructions can be found in Section 4. How do I use XTANDI? in the full CMI.

5. What should I know while using XTANDI?

Things you should do
  • Remind any doctor, dentist or pharmacist you visit that you are using XTANDI.
Things you should not do
  • Do not take XTANDI to treat any other complaints unless your doctor tells you to.
  • Do not give your medicine to anyone else, even if they have the same condition as you.
  • Do not stop taking your medicine or alter the dosage without checking with your doctor.
Driving or using machines
  • Do not drive or operate machinery until you know how XTANDI affects you.
  • XTANDI may have a moderate effect on your ability to drive or use any tools or machinery as the side effects for XTANDI includes seizures.
Looking after your medicine
  • Keep your capsules in the pack until it is time to take them.
  • Keep your capsules in the original packaging in a cool dry place where the temperature stays below 25°C

For more information, see Section 5. What should I know while using XTANDI? in the full CMI.

6. Are there any side effects?

There are a number of side effects associated with this medicine. It is important to be aware of them so that you can identify any symptoms if they occur (see the full CMI for more details). The most common and serious side effects are breathlessness, chest pain, fall, broken bones, cancer as a result of previous treatment with radiation or chemotherapy.

For more information, including what to do if you have any side effects, see Section 6. Are there any side effects? in the full CMI.



FULL CMI

XTANDI® (ex-TAN-dee)

Active ingredient(s): enzalutamide (enza-LOOT-ah-mide)


Consumer Medicine Information (CMI)

This leaflet provides important information about using XTANDI. You should also speak to your doctor or pharmacist if you would like further information or if you have any concerns or questions about using XTANDI.

Where to find information in this leaflet:

1. Why am I using XTANDI?
2. What should I know before I use XTANDI?
3. What if I am taking other medicines?
4. How do I use XTANDI?
5. What should I know while using XTANDI?
6. Are there any side effects?
7. Product details

1. Why am I using XTANDI?

XTANDI contains the active ingredient enzalutamide.

XTANDI is an androgen receptor inhibitor.

XTANDI is used to treat adult men with prostate cancer that no longer responds to hormone therapy or surgical treatment to lower testosterone, or has spread to other parts of the body and is considered "hormone sensitive".

This medicine works by blocking the activity of hormones called androgens (such as testosterone). By blocking androgens, XTANDI stops prostate cancer cells from growing and dividing.

2. What should I know before I use XTANDI?

Warnings

Do not use XTANDI if:

  • you are allergic to enzalutamide, or any of the ingredients listed at the end of this leaflet.
  • Always check the ingredients to make sure you can use this medicine.
  • XTANDI is not for use in women.

Check with your doctor if you:

have any other medical conditions such as:

  • a history of seizures
  • a serious head injury or a history of head trauma
  • a stroke
  • a brain tumour, or cancer which has spread to the brain;
  • drink very large amounts of alcohol either regularly or from time to time;
  • are taking a medicine that can cause seizures or that increases risks for having seizures (see Section 3. What if I am taking other medicines?).
(In some of these situations you may have a higher risk of having a seizure.)

take any medicines for any other condition such as:

  • heart or blood pressure problems
  • kidney problems
  • a partner who is pregnant or is planning to become pregnant.)

During treatment, you may be at risk of developing certain side effects. It is important you understand these risks and how to monitor for them. See additional information under Section 6. Are there any side effects?

Pregnancy and breastfeeding

Check with your doctor if you are pregnant or intend to become pregnant.

Talk to your doctor if you are breastfeeding or intend to breastfeed.

This medicine may cause harm to the unborn child or potential loss of pregnancy if it is taken by women who are pregnant. It must not be taken by women who are pregnant, may become pregnant, or who are breast feeding.

This medicine could possibly have an effect on male fertility.

If you are having sex with a woman who can become pregnant, you must use a condom and another effective birth control method, during treatment and for 3 months after stopping treatment with this medicine. Men who are sexually active with a pregnant woman must use a condom during and for 3 months after stopping treatment with XTANDI to protect the unborn child.

Female caregivers see Section 4. How do I use XTANDI? for handling and use.

Talk with your doctor if you have questions about birth control. Your doctor can discuss with you the risks and benefits involved.

If you are not sure whether you should start taking this medicine, talk to your doctor.

3. What if I am taking other medicines?

Tell your doctor or pharmacist if you are taking any other medicines, including any medicines, vitamins or supplements that you buy without a prescription from your pharmacy, supermarket or health food shop.

When taken at the same time as XTANDI, these medicines may increase the risk of a seizure:

  • certain medicines used to treat asthma and other respiratory diseases (e.g. aminophylline, theophylline)
  • medicines used to treat certain psychiatric disorders such as depression and schizophrenia (e.g. clozapine, olanzapine, risperidone, ziprasidone, bupropion, lithium, chlorpromazine, thioridazine, amitriptyline, desipramine, doxepin, imipramine, mirtazapine)
  • certain medicines for the treatment of pain (e.g. pethidine).

Some medicines and XTANDI may interfere with each other. These include certain medicines used to:

  • treat pain (e.g. fentanyl, tramadol, paracetamol)
  • thin the blood, or to prevent blood clots (e.g. warfarin, clopidogrel)
  • lower cholesterol (e.g. gemfibrozil, atorvastatin, simvastatin)
  • treat cancer (e.g. cabazitaxel)
  • treat epilepsy (e.g. carbamazepine, clonazepam, phenytoin, primidone, valproic acid)
  • treat certain psychiatric disorders such as severe anxiety or schizophrenia (e.g. diazepam, midazolam, haloperidol)
  • treat sleep disorders (e.g. zolpidem)
  • treat heart conditions or lower blood pressure (e.g. bisoprolol, digoxin, diltiazem, felodipine, nicardipine, nifedipine, propanolol, verapamil)
  • treat serious disease related to inflammation (e.g. dexamethasone, prednisolone)
  • lower your immunity (e.g. cyclosporin, tacrolimus)
  • treat HIV infection (e.g. indinavir, ritonavir)
  • treat bacterial infections (e.g. clarithromycin, doxycycline, rifampicin)
  • treat thyroid disorders (e.g. levothyroxine)
  • treat gout (e.g. colchicine)
  • prevent heart conditions or strokes (dabigatran etexilate)
  • treat reflux disease or peptic ulcers (e.g. omeprazole)

These medicines may be affected by XTANDI or may affect how well it works. You may need different amounts of your medicines, or you may need to take different medicines.

Your doctor and pharmacist have more information on medicines to be careful with or avoid while taking this medicine.

Some medicines may interfere with XTANDI and affect how it works.

Medicines that may increase the effect of XTANDI include:

  • gemfibrozil
  • itraconazole

Medicines that may reduce the effect of XTANDI include:

  • rifampicin

Check with your doctor or pharmacist if you are not sure about what medicines, vitamins or supplements you are taking and if these affect XTANDI.

4. How do I use XTANDI?

Follow all directions given to you by your doctor or pharmacist carefully.

They may differ from the information contained in this leaflet.

If you do not understand the instructions on the box, ask your doctor or pharmacist for help.

How much to take

  • The usual dose is four 40 mg capsules taken at the same time once a day. Your doctor may reduce your dose depending on your medical conditions.
  • Reduced dose:
    If you are taking a reduced dose of XTANDI, you may use the remaining capsules in the open dose compartment for your next scheduled dose, provided that the capsules have been otherwise stored under the conditions described below (see Looking after your medicine)
  • Follow the instructions provided and use XTANDI until your doctor tells you to stop.

How to take it

  • Swallow the capsules whole with a full glass of water.
    Do not chew, dissolve or open the capsules before swallowing.
  • You can take XTANDI with or without food.
  • XTANDI should not be handled by persons other than the patient and his caregivers. Women who are or may become pregnant should not handle damaged or opened enzalutamide capsules without protection, e.g., gloves.

When to take XTANDI

  • XTANDI should be taken at about the same time each day. Taking it at the same time each day will have the best effect. It will also help you remember when to take it.

If you forget to use XTANDI

XTANDI should be used regularly at the same time each day. If you miss your dose at the usual time, take it as soon as you remember, and then go back to taking your medicine as you would normally.

If you forget to take XTANDI for the whole day, take your usual dose the following day.

If you forget to take XTANDI for more than one day, talk to your doctor immediately.

If it is almost time for your next dose, skip the dose you missed and take your next dose when you are meant to.

Do not take a double dose to make up for the dose you missed.

This may increase the chance of you getting an unwanted side effect.

If you are not sure what to do, ask your doctor or pharmacist.

If you have trouble remembering to take your medicine, ask your pharmacist for some hints.

If you use too much XTANDI

If you think that you have used too much XTANDI, you may need urgent medical attention.

You should immediately:

  • phone the Poisons Information Centre
    (by calling 13 11 26), or
  • contact your doctor, or
  • go to the Emergency Department at your nearest hospital.

You should do this even if there are no signs of discomfort or poisoning.

You may need urgent medical attention.

You may be at increased risk of experiencing a seizure.

5. What should I know while using XTANDI?

Things you should do

If you are about to be started on any new medicine, remind your doctor and pharmacist that you are taking XTANDI.

If you are going to have surgery, tell the surgeon or anaesthetist that you are taking this medicine.

Keep all of your doctor's appointments so that your progress can be checked.

Call your doctor straight away if:

  • your partner becomes pregnant while taking this medicine.

Remind any doctor, dentist or pharmacist you visit that you are using XTANDI.

Things you should not do

  • Do not take XTANDI to treat any other complaints unless your doctor tells you to.
  • Do not give your medicine to anyone else, even if they have the same condition as you.
  • Do not stop taking your medicine or alter the dosage without checking with your doctor.

Driving or using machines

Be careful before you drive or use any machines or tools until you know how XTANDI affects you.

XTANDI may have a moderate influence on your ability to drive or use any tools or machinery. Seizures have been reported in patients taking XTANDI. If you are at higher risk of seizures, talk to your doctor.

Looking after your medicine

  • Keep your capsules in the pack until it is time to take them.
  • If you take the capsules out of the pack they may not keep as well.
  • If you are taking a reduced dose of XTANDI, you may store the remaining capsules in the open dose compartment until the next dose, provided that the capsules have been otherwise stored under the conditions described below.
  • Keep your capsules in the original packaging in a cool dry place where the temperature stays below 25°C.
  • Do not take any capsule that is leaking, damaged, or shows signs of tampering.
  • A locked cupboard at least one-and-a-half metres above the ground is a good place to store medicines.

Follow the instructions in the carton on how to take care of your medicine properly.

Store it in a cool dry place away from moisture, heat or sunlight; for example, do not store it:

  • in the bathroom or near a sink, or
  • in the car or on window sills.

Keep it where young children cannot reach it.

When to discard your medicine

If your doctor tells you to stop taking this medicine or the expiry date has passed, ask your pharmacist what to do with any medicine that is left over.

Do not throw away any medicines via wastewater or household waste.

Getting rid of any unwanted medicine

If you no longer need to use this medicine or it is out of date, take it to any pharmacy for safe disposal.

Do not use this medicine after the expiry date.

6. Are there any side effects?

Tell your doctor or pharmacist as soon as possible if you do not feel well while you are taking XTANDI.

All medicines can have side effects. If you do experience any side effects, most of them are minor and temporary. However, some side effects may need medical attention.

See the information below and, if you need to, ask your doctor or pharmacist if you have any further questions about side effects.

Do not be alarmed by the following lists of side effects. You may not experience any of them.

Ask your doctor or pharmacist to answer any questions you may have.

Less serious side effects

Less serious side effectsWhat to do
  • headache
  • weakness
  • tiredness
  • dizziness
  • breathlessness
  • chest pain
  • swelling of the hands, ankles or feet
  • rash
  • pain in back, muscles or joints
  • hot flushes
  • falls
  • broken bones
  • hallucinations
  • feeling anxious
  • dry skin
  • itching
  • high blood pressure
  • constipation
  • diarrhoea
  • feeling sick
  • decreased appetite
  • difficulty remembering things
  • difficulty thinking clearly
  • forgetfulness
  • reduced concentration
  • cancer as a result of previous treatment with radiation or chemotherapy
Speak to your doctor if you have any of these less serious side effects and they worry you.

Serious side effects

Serious side effectsWhat to do
  • Seizures
    Seizures were reported in 5 in every 1,000 people taking XTANDI and in fewer than 3 in every 1,000 people taking placebo. Seizures are more likely if you take more than the recommended dose of this medicine, if you take certain other medicines, or if you are at higher risk of seizure.
    If you have a seizure, see your doctor as soon as possible or go to Accident and Emergency at your nearest hospital immediately. Your doctor may decide that you should stop taking XTANDI.
  • Posterior Reversible Encephalopathy Syndrome (PRES) - There have been rare reports of PRES (may affect up to 1 in 1,000 people), a rare, reversible condition involving the brain, in patients treated with XTANDI. If you have a seizure, worsening headache, confusion, blindness or other vision problems, please contact your doctor as soon as possible.
Call your doctor straight away, or go straight to the Emergency Department at your nearest hospital if you notice any of these serious side effects.

Tell your doctor or pharmacist if you notice anything else that may be making you feel unwell.

Other side effects not listed here may occur in some people.

Reporting side effects

After you have received medical advice for any side effects you experience, you can report side effects to the Therapeutic Goods Administration online at www.tga.gov.au/reporting-problems. By reporting side effects, you can help provide more information on the safety of this medicine.

Always make sure you speak to your doctor or pharmacist before you decide to stop taking any of your medicines.

7. Product details

This medicine is only available with a doctor's prescription.

What XTANDI contains

Active ingredient
(main ingredient)
enzalutamide
Other ingredients
(inactive ingredients)
caprylocaproyl macrogolglycerides
butylated hydroxyanisole
butylated hydroxytoluene
gelatin
sorbitol sorbitan solution
glycerol
titanium dioxide
purified water
OPACODE WB monogramming ink NSP-78-17827 BLACK

Do not take this medicine if you are allergic to any of these ingredients.

What XTANDI looks like

XTANDI capsules are white to off-white, oblong soft gelatin capsules with "ENZ" written on one side in black ink.

XTANDI is available in packs of 112 capsules (in 4 wallets of 28 capsules each).

(AUST R 210494)

Who distributes XTANDI

XTANDI is distributed in Australia by:

Astellas Pharma Australia Pty Ltd
Suite 2.01, 2 Banfield Road
Macquarie Park, NSW 2113

Medical Information:
1800 751 755

® = Registered Trademark

This leaflet was prepared in June 2023.

Published by MIMS August 2023

BRAND INFORMATION

Brand name

Xtandi

Active ingredient

Enzalutamide

Schedule

S4

 

1 Name of Medicine

Enzalutamide.

2 Qualitative and Quantitative Composition

Xtandi is provided as liquid-filled soft gelatin capsules for oral administration. Each soft capsule contains 40 mg of enzalutamide as a solution in caprylocaproyl macrogolglycerides.

Excipient(s) with known effect.

Each soft capsule contains 57.8 mg of sorbitol.
For the full list of excipients, see Section 6.1 List of Excipients.

3 Pharmaceutical Form

Capsule, soft.
Supplied as white to off-white oblong soft gelatin capsules imprinted with "ENZ" in black ink on one side.

4 Clinical Particulars

4.1 Therapeutic Indications

Xtandi is indicated for:
the treatment of patients with metastatic hormone-sensitive prostate cancer;
the treatment of patients with non-metastatic castration-resistant prostate cancer (see Section 5.1, Clinical trials, PROSPER study (patients with non-metastatic CRPC));
the treatment of patients with metastatic castration resistant prostate cancer following failure of androgen deprivation therapy in whom chemotherapy is not yet indicated;
the treatment of patients with metastatic castration-resistant prostate cancer who have previously received docetaxel.

4.2 Dose and Method of Administration

Dosage.

The recommended dose of Xtandi is 160 mg (four 40 mg capsules) as a single oral daily dose. Xtandi can be taken with or without food. Xtandi should be taken at the same time each day.
Medical castration with a luteinising hormone-releasing hormone (LHRH) analogue should be continued during treatment of patients not surgically castrated.
If a patient misses taking Xtandi at the usual time, the prescribed dose should be taken as close as possible to the usual time. If a patient misses a dose for a whole day, treatment should be resumed the following day with the daily dose.
If a patient experiences a ≥ Grade 3 toxicity or an intolerable adverse reaction, withhold dosing for one week or until symptoms improve to ≤ Grade 2, then resume at the same or a reduced dose (120 mg or 80 mg) if warranted.

Method of administration.

Swallow capsules whole with water. Do not chew, dissolve, or open the capsules.
Xtandi should not be handled by persons other than the patient or their caregivers. Based on its mechanism of action and embryo-fetal toxicity observed in mice, Xtandi may harm a developing foetus. Women who are or may become pregnant should not handle damaged or opened enzalutamide capsules without protection, e.g. gloves. (See Section 4.6 Fertility, Pregnancy and Lactation).
Keep out of the reach of children.

Concomitant use with strong CYP2C8 inhibitors.

The concomitant use of strong CYP2C8 inhibitors should be avoided if possible. If patients must be co-administered a strong CYP2C8 inhibitor, reduce the dose of Xtandi to 80 mg once daily. If co-administration of the strong CYP2C8 inhibitor is discontinued, the Xtandi dose should be returned to the dose used prior to initiation of the strong CYP2C8 inhibitor (see Section 4.5 Interactions with Other Medicines and Other Forms of Interactions).

Patients with hepatic impairment.

No dose adjustment is required for patients with mild, moderate or severe hepatic impairment (Child-Pugh Class A, B or C, respectively).

Patients with renal impairment.

No dose adjustment is necessary for patients with mild or moderate renal impairment (see Section 5.2 Pharmacokinetic Properties, Pharmacokinetic characteristics in special populations). Caution is advised in patients with severe renal impairment or end-stage renal disease (see Section 4.4 Special Warnings and Precautions for Use, Use in renal impairment).

4.3 Contraindications

Xtandi is contraindicated in patients with known hypersensitivity to enzalutamide or to any of the excipients in the formulation (see Section 6.1 List of Excipients).
Xtandi is contraindicated in women who are, or may become, pregnant. (See Section 4.2 Dose and Method of Administration; Section 4.6 Fertility, Pregnancy and Lactation, Use in pregnancy.)

4.4 Special Warnings and Precautions for Use

Xtandi capsules should only be prescribed by a medical practitioner who is experienced with the treatment of prostate cancer and the use of antineoplastic endocrine therapies.
The following are clinically significant: seizures (see Risk of seizure below and see Section 4.8 Adverse Effects (Undesirable Effects)), Posterior reversible encephalopathy syndrome (PRES) (see Posterior reversible encephalopathy syndrome below and see Section 4.8 Adverse Effects (Undesirable Effects)) and drug interactions (see Section 4.5 Interactions with Other Medicines and Other Forms of Interactions).

Risk of seizure.

Use of enzalutamide has been associated with seizure. Caution should be used in administering Xtandi to patients with a history of seizures or other predisposing factors including, but not limited to, underlying brain injury, stroke, primary brain tumours or brain metastases, or alcoholism. In addition, the risk of seizure may be increased in patients receiving concomitant medicines that lower the seizure threshold. Permanently discontinue Xtandi in patients who develop a seizure during treatment.
Because of the risk of seizure associated with Xtandi use, patients should be advised of the risk of engaging in any activity where sudden loss of consciousness could cause serious harm to themselves or others.

Posterior reversible encephalopathy syndrome (PRES).

There have been rare reports of PRES in patients receiving Xtandi. PRES is a rare, reversible, neurological disorder which can present with rapidly evolving symptoms including seizure, headache, confusion, blindness, and other visual and neurological disturbances, with or without associated hypertension. A diagnosis of PRES requires confirmation by brain imaging, preferably magnetic resonance imaging (MRI). Discontinuation of Xtandi in patients who develop PRES is recommended.

Falls and fall-related injuries.

In the combined data of four randomised clinical trials, falls or injuries related to falls occurred in 11% of patients treated with Xtandi compared to 4% of patients treated with placebo. Falls were not associated with loss of consciousness or seizure. Fall-related injuries were more severe in patients treated with Xtandi and included non-pathologic fractures, joint injuries, and haematomas. Androgen deprivation therapy has been known to cause bone loss. For patients who fall or have a concomitant recognised risk of falling, it is recommended that the physicians consider additional supportive therapy when appropriate.

Hypersensitivity reactions.

Hypersensitivity reactions manifested by symptoms including, but not limited to, face oedema, tongue oedema, lip oedema, pharyngeal oedema, and rash have been observed with enzalutamide (see Section 4.8 Adverse Effects (Undesirable Effects)). Advise patients who experience any symptoms of hypersensitivity to discontinue enzalutamide and promptly seek medical care.

Androgen deprivation therapy may prolong the QT interval.

In patients with a history of or risk factors for QT prolongation and in patients receiving concomitant medicinal products that might prolong the QT interval, physicians should assess the benefit risk ratio including the potential for Torsade de pointes prior to initiating Xtandi.

Recent cardiovascular disease.

The phase 3 studies excluded patients with recent myocardial infarction (in the past 6 months) or unstable angina (in the past 3 months), New York Heart Association (NYHA) Class III or IV heart failure except if Left Ventricular Ejection Fraction (LVEF) ≥ 45%, bradycardia or uncontrolled hypertension. This should be taken into account if Xtandi is prescribed in these patients.

Hypertension.

In the combined data from four randomised placebo-controlled clinical trials, hypertension was reported in 12% of patients receiving Xtandi and 5% of patients receiving placebo. Medical history of hypertension was balanced between arms. Hypertension led to study discontinuation in < 1% of patients in each arm.

Risk of second malignancy.

Across the clinical trial program, the crude incidence of treatment emergent adverse events (TEAEs) corresponding to second primary malignancies (SPM) in the enzalutamide group and placebo/active surveillance group was 2.4% and 1.3%, and the median duration of treatment for patients with SPM was 21.7 months and 13.9 months, respectively. The incidence rate per 100 patient-years of any event of SPM for patients treated with enzalutamide was similar to those in the placebo/active surveillance group (1.9 vs 1.7) in the clinical trial setting.

Use with chemotherapy.

The safety and efficacy of concomitant use of Xtandi with cytotoxic chemotherapy has not been established.

Contraception in males and females.

As it is not known whether Xtandi or its metabolites are present in semen, and there were severe teratogenic effects observed in the animal studies, a condom is required during and for 3 months after treatment with Xtandi if the patient is engaged in sexual activity with a pregnant woman. If the patient engages in sexual intercourse with a woman of childbearing potential, a condom and another form of birth control must be used during and for 3 months after treatment. Studies in animals have shown reproductive toxicity (see Section 4.6 Fertility, Pregnancy and Lactation).

Use in renal impairment.

Caution is required in patients with severe renal impairment as Xtandi has not been studied in this patient population.

Use in the elderly.

No overall differences in safety or effectiveness were observed between elderly patients and younger patients. No dose adjustment is required for the elderly.

Paediatric use.

The safety and efficacy of Xtandi have not been established in children, and therefore it is not recommended for use in those < 18 years of age.

Effects on laboratory tests.

See Section 4.8 Adverse Effects (Undesirable Effects), Laboratory abnormalities.

4.5 Interactions with Other Medicines and Other Forms of Interactions

Effects of other medicines on Xtandi.

CYP2C8 inhibitors.

CYP2C8 plays an important role in the elimination of enzalutamide and in the formation of its active metabolite. Following oral administration of the strong CYP2C8 inhibitor gemfibrozil (600 mg twice daily) to healthy male subjects, the AUC of the sum of enzalutamide plus the active metabolite increased by 2.17-fold while Cmax decreased by 18%. Strong inhibitors (e.g. gemfibrozil) of CYP2C8 are to be avoided or used with caution during enzalutamide treatment. If patients must be coadministered a strong CYP2C8 inhibitor, the dose of enzalutamide should be reduced to 80 mg once daily (see Section 4.2 Dose and Method of Administration).

CYP3A4 inhibitors.

CYP3A4 plays a minor role in the metabolism of enzalutamide. Following oral administration of the strong CYP3A4 inhibitor itraconazole (200 mg once daily) to healthy male subjects, the AUC of enzalutamide increased by 41% while Cmax was unchanged. For the sum of unbound enzalutamide plus the unbound active metabolite, the AUC increased by 27% while Cmax was again unchanged. No dose adjustment is necessary when enzalutamide is coadministered with inhibitors of CYP3A4.

CYP2C8 and CYP3A4 inducers.

In a drug-drug interaction trial in healthy volunteers, a single 160 mg oral dose of enzalutamide was administered alone or after multiple oral doses of rifampicin (moderate CYP2C8 and strong CYP3A4 inducer). Rifampicin decreased the AUC of enzalutamide plus N-desmethyl enzalutamide by 37% with no effect on Cmax. No dose adjustment is necessary when enzalutamide is co-administered with inducers of CYP2C8 or CYP3A4.

Effects of Xtandi on other medicines.

Enzyme induction.

Enzalutamide is a potent enzyme inducer and increases the synthesis of many enzymes and transporters; therefore, interaction with many common medicinal products that are substrates of enzymes or transporters is expected. The reduction in plasma concentrations can be substantial, and lead to lost or reduced clinical effect. There is also a risk of increased formation of active metabolites. Enzymes that may be induced include CYP3A in the liver and gut, CYP2B6, CYP2C9, CYP2C19 and uridine 5'-diphospho-glucuronosyltransferase (UGTs - glucuronide conjugating enzymes). Some transporters may also be induced, e.g. multidrug resistance-associated protein 2 (MRP2) and the organic anion transporting polypeptide 1B1 (OATP1B1).
In vivo studies have shown that enzalutamide is a strong inducer of CYP3A4 and a moderate inducer of CYP2C9 and CYP2C19. Coadministration of enzalutamide (160 mg once daily) with single oral doses of sensitive CYP substrates in prostate cancer patients resulted in an 86% decrease in the AUC of midazolam (CYP3A4 substrate), a 56% decrease in the AUC of S-warfarin (CYP2C9 substrate), and a 70% decrease in the AUC of omeprazole (CYP2C19 substrate). UGT1A1 may have been induced as well. Enzalutamide did not have a clinically meaningful effect on plasma exposure to intravenously administered docetaxel (CYP3A4 substrate).
Interactions with certain medicinal products that are eliminated through metabolism or active transport are expected. If their therapeutic effect is of large importance to the patient, and dose adjustments are not easily performed based on monitoring of efficacy or plasma concentrations, these medicinal products are to be avoided or used with caution.
The risk for liver injury after paracetamol administration is suspected to be higher in patients concomitantly treated with enzyme inducers.
Groups of medicinal products that can be affected include, but are not limited to:
analgesics (e.g. fentanyl, tramadol);
antibiotics (e.g. clarithromycin, doxycycline);
anticancer agents (e.g. cabazitaxel);
antiepileptics (e.g. carbamazepine, clonazepam, phenytoin, primidone, valproic acid);
antipsychotics (e.g. haloperidol);
antithrombotics (e.g. warfarin, clopidogrel);
beta-blockers (e.g. bisoprolol, propranolol);
calcium channel blockers (e.g. diltiazem, felodipine, nicardipine, nifedipine, verapamil);
cardiac glycosides (e.g. digoxin);
corticosteroids (e.g. dexamethasone, prednisolone);
HIV antivirals (e.g. indinavir, ritonavir);
hypnotics (e.g. diazepam, midazolam, zolpidem);
immunosuppressants (e.g. cyclosporin, tacrolimus);
proton pump inhibitors (e.g. omeprazole);
statins metabolised by CYP3A4 (e.g. atorvastatin, simvastatin);
thyroid agents (e.g. levothyroxine).
The full induction potential of enzalutamide may not occur until approximately 1 month after the start of treatment, when steady-state plasma concentrations of enzalutamide are reached, although some induction effects may be apparent earlier. Patients taking drugs that are substrates of CYP2B6, CYP3A4, CYP2C9, CYP2C19 or UGT1A1 should be evaluated for possible loss of pharmacological effects (or increase in effects in cases where active metabolites are formed) during the first month of Xtandi treatment, and dose adjustment should be considered as appropriate. In consideration of the long half-life of enzalutamide (5.8 days, see Section 5.2 Pharmacokinetic Properties), effects on enzymes may persist for one month or longer after stopping Xtandi. A gradual dose reduction of the concomitant medicinal product may be necessary when stopping Xtandi treatment.

Warfarin and coumarin-like anticoagulants.

Co-administration with warfarin and coumarin-like anticoagulants should be avoided. If Xtandi is co-administered with an anticoagulant metabolised by CYP2C9 (such as warfarin), additional INR monitoring should be conducted.

CYP1A2, CYP2C8 and CYP2D6 substrates.

Enzalutamide (160 mg once daily) did not cause a clinically relevant change in the AUC or Cmax of caffeine (CYP1A2 substrate), pioglitazone (CYP2C8 substrate) or dextromethorphan (CYP2D6 substrate). The AUC of pioglitazone increased by 20% while Cmax decreased by 18%. The AUC and Cmax of caffeine decreased by 11% and 4% respectively. No dose adjustment is indicated when a CYP1A2, CYP2C8 or CYP2D6 substrate is coadministered with Xtandi.

P-gp substrates.

In vitro data indicate that enzalutamide is not a substrate for, but may be an inhibitor of the efflux transporter P-gp. A mild inhibitory effect of enzalutamide, at steady-state, on P-gp was observed in a study in patients with prostate cancer that received a single oral dose of the probe P-gp substrate digoxin before and concomitantly with enzalutamide (concomitant administration followed at least 55 days of once daily dosing of 160 mg enzalutamide). The AUC and Cmax of digoxin increased by 33% and 17%, respectively. Medicinal products with a narrow therapeutic range that are substrates for P-gp (e.g. colchicine, dabigatran etexilate, digoxin) should be used with caution when administered concomitantly with Xtandi and may require dose adjustment to maintain optimal plasma concentrations.

BCRP substrates.

At steady-state, enzalutamide did not cause a clinically meaningful change in exposure to the probe breast cancer resistance protein (BCRP) substrate rosuvastatin in patients with prostate cancer that received a single oral dose of rosuvastatin before and concomitantly with enzalutamide (concomitant administration followed at least 55 days of once daily dosing of 160 mg enzalutamide). The AUC of rosuvastatin decreased by 14% while Cmax increased by 6%. No dose adjustment is necessary when a BCRP substrate is co-administered with Xtandi.

MRP2, OAT1, OAT3, OATP1B1, OATP1B3, OCT1 and OCT2 substrates.

Based on in vitro data, inhibition of MRP2 (in the intestine), as well as organic anion transporter 3 (OAT3), OATP1B1, and OCT1 (systemically) cannot be excluded. Theoretically, induction of OAT3, OATP1B1, OCT1 and MRP2 is also possible, and the net effect is presently unknown. The effects of enzalutamide on these transporters have not been evaluated in vivo. In vitro data indicate that enzalutamide and its major metabolites do not inhibit the following transporters at clinically relevant concentrations: OAT1, OATP1B3, or OCT2.

Effect of food on Xtandi exposure.

Food has no clinically significant effect on the extent of exposure to Xtandi. In clinical trials, Xtandi was administered without regard to food.

4.6 Fertility, Pregnancy and Lactation

Effects on fertility.

Based on its pharmacology and findings in animal studies, male fertility may be impaired by treatment with enzalutamide. Findings in the male reproductive tract of mice, rats and/or dogs treated with enzalutamide included atrophy of the prostate gland, epididymis and seminal vesicles, testicular hypospermia and seminiferous tubule degeneration. These effects were observed at doses below the clinical exposure (based on AUC) and reversed or partially resolved after an 8-week recovery period. (See Section 4.4 Special Warnings and Precautions for Use, Contraception in males and females).
(Category X)
Xtandi is not indicated for use in women. Xtandi has not been shown to be safe for use in women. Xtandi is contraindicated in women who are or may become pregnant (see Section 4.3 Contraindications). There are no human data on the use of Xtandi in pregnancy.
Teratogenicity (cleft palate, cervical rib and decreased anogenital distance) and embryofetal lethality were seen in mouse embryofetal development studies at greater than or equal to 10 mg/kg/day (below the AUC at the maximum recommended human dose). This medicine may cause harm to the unborn child or potential loss of pregnancy if taken by women who are pregnant.
Studies in pregnant rats have shown that enzalutamide and/or its metabolites are transferred to foetuses. After oral administration of radiolabelled 14C-enzalutamide to rats on Day 14 of pregnancy at a dose of 30 mg/kg, the maximum radioactivity in the foetus was reached 4 hours after administration and was lower than that in the maternal plasma with tissue/plasma ratio of 0.27. Radioactivity in the foetus decreased to 0.08 times the maximum concentration at 72 hours after administration.
Xtandi is not indicated for use in females. There is no information available on the presence of Xtandi human milk, the effects of the drug on the breastfed infant, or the effects of the drug on milk production. Xtandi and/or its metabolites are secreted in rat milk. After oral administration of radiolabelled 14C-enzalutamide to lactating rats at a dose of 30 mg/kg, the maximum radioactivity in the milk was reached 4 hours after administration and was up to 3.54-fold higher than that in the maternal plasma. Study results have also shown that enzalutamide and/or its metabolites are transferred to infant rat tissues via milk and subsequently eliminated.

4.7 Effects on Ability to Drive and Use Machines

No formal studies of the effects of Xtandi on the ability to drive or use machines have been conducted. Neurological and psychiatric events (such as seizure, amnesia, fatigue, memory impairment, cognitive disorder, and disturbance in attention) associated with Xtandi may affect some patients' ability to drive or operate machinery. (See Section 4.8 Adverse Effects (Undesirable Effects)). Patients with a history of seizures or other predisposing factors should be advised of the risk of driving or operating machines (see Section 4.4 Special Warnings and Precautions for Use, Risk of seizure).

4.8 Adverse Effects (Undesirable Effects)

Clinical trial experience.

Six randomized, controlled clinical studies enrolled patients with castration-resistant prostate cancer that had progressed on androgen deprivation therapy (LHRH therapy or bilateral orchiectomy). Four studies were placebo-controlled and two studies were bicalutamide-controlled. In the AFFIRM, PREVAIL, Asian PREVAIL, and PROSPER studies, patients received enzalutamide 160 mg or placebo orally once daily. In the TERRAIN and STRIVE studies, patients received enzalutamide 160 mg or bicalutamide 50 mg orally once daily. A randomised, placebo-controlled clinical study was also conducted in patients with metastatic hormone-sensitive prostate cancer (mHSPC) on androgen deprivation therapy (ARCHES). Patients in the ARCHES study received enzalutamide 160 mg or placebo orally once daily. All patients continued androgen deprivation therapy.
The most common adverse reactions (≥ 10%) seen in enzalutamide-treated patients in clinical studies, in decreasing order of frequency are fatigue, nausea, hot flush, diarrhoea, hypertension, fracture, asthenia, and fall. Other important adverse reactions include ischaemic heart disease and seizure.

AFFIRM study: metastatic castration resistant prostate cancer following chemotherapy.

AFFIRM enrolled 1199 patients with metastatic castration-resistant prostate cancer (CRPC) who had previously received docetaxel. The median duration of treatment was 8.3 months with Xtandi and 3.0 months with placebo. During the trial, 48% of patients on the Xtandi arm and 46% of patients on the placebo arm received glucocorticoids. Grade 3 and higher adverse reactions were reported among 47% of Xtandi-treated patients and 53% of placebo-treated patients. Discontinuations due to adverse events were reported for 16% of Xtandi-treated patients and 18% of placebo-treated patients. The most common adverse reaction leading to treatment discontinuation was seizure, which occurred in 0.9% of the Xtandi-treated patients compared to none (0%) of the placebo-treated patients. Table 1 shows adverse reactions reported in AFFIRM that occurred at a ≥ 2% absolute increase in frequency in the Xtandi arm compared to the placebo arm.

PREVAIL study: chemotherapy-naïve metastatic prostate cancer that progressed on androgen deprivation therapy.

PREVAIL enrolled 1717 patients with metastatic prostate cancer that progressed on an LHRH analogue or after bilateral orchiectomy and had not received prior cytotoxic chemotherapy, of whom 1715 received at least one dose of study drug. The median duration of treatment was 16.6 months with Xtandi and 4.6 months with placebo. The most common adverse reaction leading to treatment discontinuation was fatigue, which occurred in 0.2% of the Xtandi-treated patients compared to 0.9% of placebo-treated patients.
Table 2 includes adverse reactions reported in Study 2 that occurred at a ≥ 2% absolute increase in frequency in the Xtandi arm compared to the placebo arm.

TERRAIN study: Xtandi versus bicalutamide in chemotherapy-naïve metastatic CRPC.

TERRAIN enrolled 375 patients with metastatic CRPC who had not received prior cytotoxic chemotherapy, of whom 372 received at least one dose of study drug. The median duration of treatment was 11.6 months with Xtandi and 5.8 months with bicalutamide. Discontinuations with an adverse event as the primary reason were reported for 7.6% of Xtandi-treated patients and 6.3% of bicalutamide-treated patients. The most common adverse reactions leading to treatment discontinuation were back pain and pathological fracture, which occurred in 3.8% of Xtandi-treated patients for each event and in 2.1% and 1.6% of bicalutamide-treated patients, respectively. Table 3 shows overall and common adverse reactions (≥ 10%) in Xtandi-treated patients.

PROSPER study: Xtandi versus placebo in non-metastatic CRPC patients.

PROSPER enrolled 1401 patients with non-metastatic CRPC, of whom 1395 received at least one dose of study drug. Patients were randomized 2:1 and received either Xtandi at a dose of 160 mg once daily (N = 930) or placebo (N = 465). The median duration of treatment at the time of analysis was 18.4 months (range: 0.0 to 42 months) with Xtandi and 11.1 months (range: 0.0 to 43 months) with placebo.
Overall, 32 patients (3.4%) receiving Xtandi died from adverse events. The reasons for death with ≥ 2 patients included coronary artery disorders (n = 7), sudden death (n = 2), cardiac arrhythmias (n = 2), general physical health deterioration (n = 2), stroke (n = 2), and secondary malignancy (n = 5; one each of acute myeloid leukemia, brain neoplasm, mesothelioma, small cell lung cancer, and malignant neoplasm of unknown primary site). Three patients (0.6%) receiving placebo died from adverse events of cardiac arrest (n = 1), left ventricular failure (n = 1), and pancreatic carcinoma (n = 1). Grade 3 or higher adverse reactions were reported among 31% of Xtandi-treated patients and 23% of placebo-treated patients. Discontinuations with an adverse event as the primary reason were reported for 9.4% of Xtandi-treated patients and 6.0% of placebo-treated patients. Of these, the most common adverse event leading to treatment discontinuation was fatigue, which occurred in 1.6% of the Xtandi-treated patients compared to none of the placebo-treated patients. Table 4 shows adverse reactions reported in PROSPER that occurred at a ≥ 2% higher frequency in the Xtandi arm than in the placebo arm.

ARCHES study: Xtandi versus placebo in metastatic HSPC patients.

ARCHES randomized 1150 patients with mHSPC, of whom 1146 received at least one dose of study drug. Patients received either Xtandi at a dose of 160 mg once daily (N=572) or placebo (N=574). At the time of analysis, the median duration of treatment was 12.8 months (range: 0.2 to 26.6 months) with Xtandi and 11.6 months (range: 0.2 to 24.6 months) with placebo. Seventy-six percent (76%) of patients remained on treatment with Xtandi and 58% of patients remained on placebo at the time of analysis.
Grade 3 or higher adverse events were reported in 24% of patients treated with Xtandi and 26% of patients treated with placebo. The primary reason for study drug discontinuation in both treatment groups was disease progression. Disease progression was approximately 3-fold lower in patients in the Xtandi group (11.3%) compared to the placebo group (29.7%). Discontinuation due to adverse events as the primary reason were reported in 4.9% of Xtandi-treated patients and 3.6% of placebo-treated patients. The most common adverse events for the Xtandi group (0.9% each) that were the primary reason leading to study drug discontinuation were anemia (0.5% in the placebo group), decreased appetite (0.7% in the placebo group), diarrhoea (0.3% in the placebo group) and fatigue (0.7% in the placebo group).
Table 5 shows adverse reactions reported in ARCHES that occurred at a ≥ 2% higher frequency in the Xtandi arm than in the placebo arm.

ENZAMET study: Xtandi versus non-steroidal anti-androgen in metastatic HSPC patients.

ENZAMET randomized 1125 patients with mHSPC, of whom 1121 received at least one dose of study drug. Patients received either Xtandi at a dose of 160 mg once daily (N=563) or NSAA (non-steroidal anti-androgen bicalutamide, nilutamide, or flutamide) (N=558). Patients were allowed up to 6 cycles of concomitant docetaxel (at the start of study with up to 2 cycles prior to study). At the time of analysis, the median duration of treatment was 29.5 months (range: 0.1 to 58.4 months) with Xtandi and 22.1 months (range: 0.0 to 58.6 months) with NSAA. Sixty-four percent (64%) of patients remained on treatment with Xtandi and 36% of patients remained on treatment with NSAA at the time of analysis.
Grade 3 or 4 adverse events were reported in a higher percentage of patients who received docetaxel in each arm; in the enzalutamide arm, 60% of patients treated with docetaxel vs 55% without docetaxel, and in the NSAA arm, 52% of patients treated with docetaxel vs 35% without docetaxel. Discontinuations due to serious adverse events were reported in 11% of Xtandi-treated patients and 9% of NSAA-treated patients. The most common serious adverse event leading to study drug discontinuation was seizure which occurred in 0.9% of the Xtandi-treated patients compared to none in the NSAA-treated patients.
Table 6 shows Grade 3 and Grade 4 adverse reactions reported in ENZAMET that occurred at a ≥ 2% higher frequency in the Xtandi arm than in the NSAA arm.

Summary of adverse reactions.

Adverse reactions observed during clinical studies are listed in Table 7 by frequency category in each system organ class. Frequency categories are defined as follows: very common (≥ 1/10); common (≥ 1/100 to < 1/10); uncommon (≥ 1/1,000 to < 1/100); rare (≥ 1/10,000 to < 1/1,000); very rare (< 1/10,000); and not known (cannot be estimated from the available data).

Post marketing experience.

The following adverse events, which are not listed in Table 5, have been reported in association with enzalutamide use during worldwide post-marketing experience:

Musculoskeletal and connective tissue disorders.

Myalgia, muscle spasms, muscular weakness, back pain.

Skin and connective tissue disorders.

Acute generalized exanthematous pustulosis, dermatitis bullous, dermatitis exfoliative generalized, drug reaction with eosinophilia and systemic symptoms, erythema multiforme, exfoliative rash, Stevens-Johnson syndrome (SJS), toxic epidermal necrolysis (TEN), and toxic skin eruption.

Description of selected adverse reactions.

Seizures.

In controlled clinical studies, 24 patients (0.5%) experienced a seizure out of 4403 patients treated with a daily dose of 160 mg enzalutamide, whereas four patients (0.2%) receiving placebo and one patient (0.3%) receiving bicalutamide experienced a seizure. In the patients who experienced seizure when treated with enzalutamide, there was one case of seizure where the patient experienced complications resulting in death. In the controlled clinical studies, patients with prior seizures or other risk factors for seizures were excluded. (See Section 4.4 Special Warnings and Precautions for Use, Risk of seizure).
Patients who are taking concomitant medicines (strong CYP2C8 inhibitors) that increase the blood levels of enzalutamide may be at increased risk of seizure (see Section 4.2 Dose and Method of Administration; Section 4.5 Interactions with Other Medicines and Other Forms of Interactions).
In the single-arm UPWARD study to assess incidence of seizure in patients with predisposing factors for seizure (whereof 1.7% had a history of seizures), and 8 of 366 (2.2%) patients treated with Xtandi experienced a seizure. The median duration of treatment was 9.3 months.

Laboratory abnormalities.

Table 8 shows laboratory abnormalities that occurred in ≥ 5% of patients, and more frequently (> 2%) in the Xtandi arm compared to placebo in the pooled Phase 3 placebo-controlled studies.

Infections.

In the AFFIRM trial, 1% of patients treated with Xtandi compared to 0.3% of patients on placebo died from infections or sepsis. In the PREVAIL trial, 1 patient in each treatment group (0.1%) had an infection resulting in death.

Hallucinations.

In the AFFIRM trial, 1.6% of patients treated with Xtandi were reported to have Grade 1 or 2 hallucinations compared to 0.3% of patients on placebo. In the PREVAIL trial, 1 patient in each treatment group (0.1%) reported an event of Grade 1 hallucination. Of the patients with hallucinations, the majority were on opioid-containing medications at the time of the event. Hallucinations were visual, tactile, or undefined.

Reporting suspected adverse effects.

Reporting suspected adverse reactions after registration of the medicinal product is important. It allows continued monitoring of the benefit-risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions at www.tga.gov.au/reporting-problems.

4.9 Overdose

There is no antidote for Xtandi. In the event of an overdose, treatment with Xtandi should be stopped and general supportive measures initiated taking into consideration the half-life of 5.8 days. Patients may be at increased risk of seizures following an overdose.
For information on the management of overdose, contact the Poisons Information Centre on 13 11 26 (Australia).

5 Pharmacological Properties

5.1 Pharmacodynamic Properties

Mechanism of action.

Enzalutamide is an androgen receptor signalling inhibitor that blocks the androgen receptor signalling pathway. Enzalutamide competitively inhibits androgen binding to androgen receptors, and consequently, inhibits nuclear translocation of these receptors and inhibits the binding of androgen receptor to DNA. In vitro, enzalutamide treatment decreased proliferation and induced prostate cancer cell death. Decreased tumour growth was seen in a mouse prostate cancer xenograft model. In preclinical studies enzalutamide lacked androgen receptor agonist activity against several prostate cancer cell lines. The active metabolite, N-desmethyl enzalutamide, exhibited similar in vitro activity to enzalutamide in the inhibition of testosterone binding to the androgen receptor.

Clinical trials.

Efficacy of enzalutamide was established in three randomised placebo-controlled multicentre phase 3 clinical studies [MDV3100-14 (PROSPER), CRPC2 (AFFIRM), MDV3100-03 (PREVAIL)] of patients with progressive prostate cancer who had disease progression on androgen deprivation therapy [luteinising hormone-releasing hormone (LHRH) analogue or after bilateral orchiectomy]. The PREVAIL study enrolled metastatic CRPC chemotherapy-naïve patients; whereas the AFFIRM study enrolled metastatic CRPC patients who had received prior docetaxel; and the PROSPER study enrolled patients with non-metastatic CRPC. Additionally, efficacy in patients with mHSPC was also established in one randomized, placebo-controlled multicentre phase 3 clinical study (ARCHES). All patients continued on a LHRH analogue or had bilateral orchiectomy.
In the active treatment arm, Xtandi was administered orally at a dose of 160 mg daily. In the four clinical studies (ARCHES, PROSPER, AFFIRM and PREVAIL), patients received placebo in the control arm and patients were allowed, but not required, to take prednisone (maximum daily dose allowed was 10 mg prednisone or equivalent).
Changes in prostate specific antigen (PSA) serum concentration independently do not always predict clinical benefit. Therefore, in the four studies, it was recommended that patients be maintained on their study treatments until discontinuation criteria were met as specified below for each study.

ARCHES study (patients with metastatic HSPC).

The ARCHES study enrolled 1150 patients with mHSPC randomized 1:1 to receive treatment orally once daily with Xtandi 160 mg (N=574) or placebo (N=576). All patients in the trial received a LHRH analog or had a prior bilateral orchiectomy. Patients were stratified by volume of disease (low vs high) and prior docetaxel therapy for prostate cancer (no prior docetaxel, 1-5 cycles, or 6 prior cycles). Treatment with concurrent docetaxel was not allowed. Patients were required to have confirmation of metastatic prostate cancer by positive bone scan or metastatic lesions on CT or MRI scan. Patients continued treatment until radiographic disease progression, initiation of new treatment, unacceptable toxicity, or withdrawal.
The following patient demographics and baseline characteristics were balanced between the two treatment arms. The median age at randomization was 70 years in both treatment groups. Most patients in the total population were Caucasian (80.5%), 13.5% were Asian, and 1.4% were Black. Sixty-seven percent (67%) of patients treated with Xtandi and 65% of patients who received placebo had a Gleason score of ≥ 8. Thirty-seven percent (37%) of patients had a low volume of disease and 63% of patients had a high volume of disease. High volume of disease is defined as metastases involving the viscera or, in the absence of visceral lesions, there must be 4 or more bone lesions, at least 1 of which must be in a bony structure beyond the vertebral column and pelvic bone. Eighty-two percent (82%) of patients had no prior docetaxel treatment; 2% of patients had 1 to 5 cycles of docetaxel and 16% of patients had 6 prior cycles of docetaxel treatment. The Eastern Cooperative Oncology Group Performance Status (ECOG PS) score was 0 for 78% of patients and 1 for 22% of patients at study entry.
Radiographic progression-free survival (rPFS), based on independent central review, was the primary endpoint defined as the time from randomization to the first objective evidence of radiographic disease progression or death (due to any cause from the time of randomization through 24 weeks after study drug discontinuation), whichever occurred first. Xtandi demonstrated a statistically significant 61% reduction in the risk of an rPFS event compared to placebo. Consistent rPFS results were also observed in patients with high or low volume of disease, and patients with and without prior docetaxel therapy. At the pre-specified final analysis for overall survival (OS), conducted when 356 deaths were observed, a statistically significant 34% reduction in the risk of death was demonstrated in the group randomized to receive enzalutamide compared with the group randomized to receive placebo [HR = 0.66, (95% CI: 0.53; 0.81), p < 0.0001]. The median time for overall survival was not reached in either treatment group. The estimated median follow-up time for all patients was 44.6 months. Efficacy results for rPFS and OS from ARCHES are summarized in Table 9, Figure 1 and Figure 2.
The major efficacy outcome was supported by multiple secondary endpoints. A statistically significant 72% reduction in risk of initiation of a new antineoplastic therapy was demonstrated for patients in the Xtandi arm compared to patients in the placebo arm (HR = 0.28 [95% CI: 0.20, 0.40]; p < 0.0001). A statistically significant improvement of 19.3% in objective response rate (percentage of patients with complete or partial response in their soft tissue disease) was demonstrated with an Objective Response Rate (OSS) (calculated as percentage of patients with measurable disease at baseline who achieved a complete or partial response in their soft tissue disease) of 83.1% for patients in the Xtandi arm compared to 63.7% for patients in the placebo arm (95% CI: 10.4, 28.2; p < 0.0001). Time to First Symptomatic Skeletal Event (SSE) (time from randomization to the occurrence of the first symptomatic skeletal event) was associated with a 48% reduction in the risk of a patient experiencing an SSE compared with patients in the placebo arm [HR = 0.52, 95% CI: 0.33, 0.80; nominal p = 0.0026].

ENZAMET study (patients with metastatic HSPC).

The ENZAMET study enrolled 1125 patients with mHSPC randomized 1:1 to receive treatment orally once daily with Xtandi 160 mg (N=563) or NSAA (N=562). All patients in the trial received a LHRH analog or had a prior bilateral orchiectomy. Patients were stratified by volume of disease (low vs high), concomitant antiresorptive therapy (yes vs no), comorbidities (ACE-27: 0 to 1 vs 2 to 3) and planned use of a total of 6 cycles of docetaxel, of which 0-2 cycles were allowed before randomization (yes vs no). Patients were required to have confirmation of metastatic prostate cancer by positive bone scan or metastatic lesions on CT or MRI scan. Patients continued treatment until evidence of clinical progression via CT, MRI or whole body bone scan.
The following patient demographics and baseline characteristics were balanced between the two treatment arms. The median age at randomization was 69 years in the Xtandi group and 68 years in the NSAA group (treated with bicalutamide, nilutamide, or flutamide). The majority of patients had an ECOG performance status score of 0 (72%) and a Gleason score of ≥ 8 (58%). Forty-eight percent (48%) of patients had a low volume of disease and 52% of patients had a high volume of disease. High volume of disease is defined as metastases involving the viscera or, in the absence of visceral lesions, there must be 4 or more bone lesions, at least 1 of which must be in a bony structure beyond the vertebral column and pelvic bone. Ten percent (10%) of patients had concomitant antiresorptive therapy; 75% had no or mild comorbidities (ACE-27 score of 0 to 1) and 45% had a total of 6 cycles of docetaxel, of which 0-2 cycles were allowed before randomization.
At the time of analysis, the median follow-up for OS was 33.8 months. The interim analysis demonstrated a statistically significant 33% reduction in the risk of death for patients treated with Xtandi compared to conventional NSAA treatment [HR of 0.67 (95% CI: 0.52, 0.86; p=0.0018)]. See Figure 3.

PROSPER study (patients with non-metastatic CRPC).

The PROSPER study enrolled 1401 patients with asymptomatic, high-risk non-metastatic CRPC who continued on androgen deprivation therapy (ADT; defined as LHRH analogue or prior bilateral orchiectomy). Patients were required to have a PSA doubling time ≤ 10 months, PSA ≥ 2 nanogram/mL, and confirmation of non-metastatic disease by blinded independent central review (BICR).
Patients with a history of mild to moderate heart failure (NYHA Class I or II), and patients taking medicinal products associated with lowering the seizure threshold were allowed. Patients were excluded with a previous history of seizure, a condition that might predispose them to seizure, or certain prior treatments for prostate cancer (i.e. chemotherapy, ketoconazole, abiraterone acetate, aminoglutethimide and/or enzalutamide).
Patients were randomised 2:1 to receive either enzalutamide at a dose of 160 mg once daily (N = 933) or placebo (N = 468). Patients were stratified by Prostate Specific Antigen (PSA) Doubling Time (PSADT) (< 6 months or ≥ 6 months) and the use of bone-targeting agents (yes or no).
The demographic and baseline characteristics were well-balanced between the two treatment arms. The median age at randomisation was 74 years in the enzalutamide arm and 73 years in the placebo arm. Most patients (approximately 71%) in the study were Caucasian; 16% were Asian and 2% were Black. Eighty-one percent (81%) of patients had an ECOG performance status score of 0 and 19% patients had an ECOG performance status of 1.
Metastasis-free survival (MFS) was the primary endpoint defined as the time from randomisation to radiographic progression or death within 112 days of treatment discontinuation without evidence of radiographic progression, whichever occurred first. Key secondary endpoints assessed in the study were time to PSA progression, time to first use of new antineoplastic therapy (TTA), overall survival (OS). Additional secondary endpoints included time to first use of cytotoxic chemotherapy and chemotherapy-free survival. See results below (Table 10).
Enzalutamide demonstrated a statistically significant 71% reduction in the relative risk of radiographic progression or death compared to placebo [HR = 0.29 (95% CI: 0.24, 0.35), p < 0.0001]. Median MFS was 36.6 months (95% CI: 33.1, NR) on the enzalutamide arm versus 14.7 months (95% CI: 14.2, 15.0) on the placebo arm. Consistent MFS results were also observed in all pre-specified patient sub-groups including PSADT (< 6 months or ≥ 6 months), demographic region (North America, Europe, rest of world), age (< 75 or ≥ 75), use of a prior bone-targeting agent (yes or no). See Figure 4.
At the final analysis for overall survival, conducted when 466 deaths were observed, a statistically significant improvement in overall survival was demonstrated in patients randomised to receive enzalutamide compared with patients randomised to receive placebo with a 26.6% reduction in risk of death [hazard ratio (HR) = 0.734, (95% CI: 0.608; 0.885), p = 0.0011]. The median follow-up time was 48.6 and 47.2 months for the enzalutamide and placebo groups, respectively. Thirty-three percent of enzalutamide-treated and 65% of placebo-treated patients received at least one subsequent antineoplastic therapy that may prolong overall survival. See Figure 5.
Enzalutamide demonstrated a statistically significant 93% reduction in the relative risk of PSA progression compared to placebo [HR = 0.07 (95% CI: 0.05, 0.08), p < 0.0001]. Median time to PSA progression was 37.2 months (95% CI: 33.1, NR) on the enzalutamide arm versus 3.9 months (95% CI: 3.8, 4.0) on the placebo arm.
Enzalutamide demonstrated a statistically significant delay in the time to first use of new antineoplastic therapy compared to placebo [HR = 0.21 (95% CI: 0.17, 0.26), p < 0.0001]. Median time to first use of new antineoplastic therapy was 39.6 months (95% CI: 37.7, NR) on the enzalutamide arm versus 17.7 months (95% CI: 16.2, 19.7) on the placebo arm. See Figure 6.

STRIVE study (chemotherapy-naïve patients with non-metastatic/metastatic CRPC).

The STRIVE study enrolled 396 non-metastatic or metastatic CRPC patients who had serologic or radiographic disease progression despite primary androgen deprivation therapy who were randomized to receive either enzalutamide at a dose of 160 mg once daily (N = 198) or bicalutamide at a dose of 50 mg once daily (N = 198). PFS was the primary endpoint defined as the time from randomization to the earliest objective evidence of PSA progression, radiographic progression, or death on study. Median PFS was 19.4 months (95% CI: 16.5, not reached) in the enzalutamide group versus 5.7 months (95% CI: 5.6, 8.1) in the bicalutamide group [HR = 0.24 (95% CI: 0.18, 0.32), p < 0.0001]. Consistent benefit of enzalutamide over bicalutamide on PFS was observed in all prespecified patient subgroups. See Figure 7.

TERRAIN study (chemotherapy-naïve patients).

TERRAIN enrolled 375 chemotherapy- and anti-androgen-therapy naïve patients who were randomised to receive either enzalutamide orally at a dose of 160 mg once daily (N = 184) or bicalutamide orally at a dose of 50 mg once daily (N = 191). Median progression-free survival (PFS) was 15.7 months for patients on enzalutamide vs. 5.8 months for patients on bicalutamide [HR = 0.44 (95% CI: 0.34, 0.57), P < 0.0001] (Figure 8). PFS was defined as objective evidence of radiographic disease progression by independent central review, skeletal-related events, initiation of new antineoplastic therapy or death by any cause, whichever occurred first. Consistent PFS benefit was observed across all pre-specified patient subgroups.

PREVAIL study (chemotherapy-naïve patients with metastatic CRPC).

A total of 1717 asymptomatic or mildly symptomatic chemotherapy-naïve patients were randomised 1:1 to receive either enzalutamide orally at a dose of 160 mg once daily (N = 872) or placebo orally once daily (N = 845). Patients with visceral disease, patients with a history of mild to moderate heart failure (NYHA Class I or II), and patients taking medications associated with lowering the seizure threshold were allowed. Patients with a previous history of seizure or a condition that might predispose to seizure and patients with moderate or severe pain from prostate cancer were excluded. Study treatment continued until disease progression (evidence of radiographic progression, a skeletal related event, or clinical progression) and the initiation of either a cytotoxic chemotherapy or an investigational agent, or until unacceptable toxicity.
Patient demographics and baseline disease characteristics were balanced between the treatment arms. The median age was 71 years (range 42 - 93) and the racial distribution was 77% Caucasian, 10% Asian, 2% Black and 11% other or unknown races. Sixty-eight percent (68%) of patients had an ECOG performance status score of 0 and 32% of patients had an ECOG performance status of 1. Baseline pain assessment was 0 - 1 (asymptomatic) in 67% of patients and 2 - 3 (mildly symptomatic) in 32% of patients as defined by the Brief Pain Inventory Short Form (worst pain over past 24 hours on a scale of 0 to 10). Approximately 45% of patients had measurable soft tissue disease at study entry, and 12% of patients had visceral (lung and/or liver) metastases.
Co-primary efficacy endpoints were overall survival and radiographic progression-free survival (rPFS). In addition to the co-primary endpoints, benefit was also assessed using time to initiation of cytotoxic chemotherapy, best overall soft tissue response, time to first skeletal-related event, PSA response (≥ 50% decrease from baseline), time to PSA progression, and time to FACT-P total score degradation.
Radiographic progression was assessed with the use of sequential imaging studies as defined by Prostate Cancer Clinical Trials Working Group 2 (PCWG2) criteria (for bone lesions) and/or Response Evaluation Criteria in Solid Tumours (RECIST v 1.1) criteria (for soft tissue lesions). Analysis of rPFS utilised centrally reviewed radiographic assessment of progression.
At the pre-specified interim analysis for overall survival, when 540 deaths were observed, treatment with enzalutamide demonstrated a statistically significant improvement in overall survival compared to treatment with placebo with a 29.4% reduction in risk of death [HR = 0.71 (95% CI: 0.60; 0.84), p < 0.0001] (Table 11). Of note, 40.4% of enzalutamide-treated patients and 70.5% of placebo-treated patients received subsequent therapies with a demonstrated survival benefit.
An updated overall survival analysis was conducted when 784 deaths were observed. Results from this analysis were consistent with those from the pre-specified interim analysis (see Table 11). At the updated analysis, 52% of enzalutamide-treated and 81% of placebo-treated patients had received subsequent therapies for metastatic castration resistant prostate cancer which may prolong overall survival.
A final analysis of 5-year PREVAIL data showed a statistically significant increase in overall survival was maintained in patients treated with enzalutamide compared to placebo [HR = 0.835, (95% CI: 0.75, 0.93); p-value = 0.0008] despite 28% of patients on placebo crossing over to enzalutamide. (Table 11 and Figures 9 and 10). The 5-year OS rate was 26% for the enzalutamide arm compared to 21% for the placebo arm.
At the pre-specified rPFS analysis, a statistically significant improvement was demonstrated between the treatment groups with an 81.4% reduction in risk of radiographic progression or death [HR = 0.19 (95% CI: 0.15, 0.23), p < 0.0001]. One hundred and eighteen (14%) enzalutamide-treated patients and 321 (40%) of placebo-treated patients had an event. The median rPFS was not reached (95% CI: 13.8, not reached) in the enzalutamide-treated group and was 3.9 months (95% CI: 3.7, 5.4) in the placebo-treated group (see Figure 11). Consistent rPFS benefit was observed across all pre-specified patient subgroups (e.g. age, baseline ECOG performance, baseline PSA and LDH, Gleason score at diagnosis, and visceral disease at screening). A pre-specified follow-up rPFS analysis based on the investigator assessment of radiographic progression demonstrated a statistically significant improvement between the treatment groups with a 69.3% reduction in risk of radiographic progression or death [HR = 0.31 (95% CI: 0.27, 0.35), p < 0.0001]. The median rPFS was 19.7 months in the enzalutamide group and 5.4 months in the placebo group.
At the time of the primary analysis there were 1633 patients randomised.
In addition to the co-primary efficacy endpoints, statistically significant improvements were also demonstrated in the following prospectively defined endpoints.
The median time to initiation of cytotoxic chemotherapy was 28.0 months for patients receiving enzalutamide and 10.8 months for patients receiving placebo (HR = 0.35, 95% CI: [0.30, 0.40], p < 0.0001).
The proportion of enzalutamide-treated patients with measurable disease at baseline who had an objective soft tissue response was 58.8% (95% CI: 53.8, 63.7) compared with 5.0% (95% CI: 3.0, 7.7) of patients receiving placebo. The absolute difference in objective soft tissue response between enzalutamide and placebo arms was 53.9% (95% CI: 48.5, 59.1, p < 0.0001). Complete responses were reported in 19.7% of enzalutamide-treated patients compared with 1.0% of placebo-treated patients, and partial responses were reported in 39.1% of enzalutamide-treated patients versus 3.9% of placebo-treated patients.
Enzalutamide significantly decreased the risk of the first skeletal-related event by 27% [HR = 0.526, (95% CI: 0.421, 0.656), p < 0.0001], 21.6% of enzalutamide-treated patients reported a skeletal-related event versus 18.5% of placebo-treated patients, an absolute difference of 3.1% events. A skeletal-related event was defined as radiation therapy or surgery to bone for prostate cancer, pathologic bone fracture, spinal cord compression, or change of antineoplastic therapy to treat bone pain.
The median time to PSA progression per PCWG2 criteria was 11.2 months for patients treated with enzalutamide and 2.8 months for patients who received placebo [HR = 0.17, (95% CI: 0.15, 0.20), p < 0.0001].
Treatment with enzalutamide decreased the risk of FACT-P degradation by 37.5% compared with placebo (p < 0.0001). The median time to degradation in FACT-P was 11.3 months in the enzalutamide group and 5.6 months in the placebo group.

AFFIRM study (patients with metastatic CRPC who previously received chemotherapy).

The efficacy and safety of Xtandi in patients with metastatic castration resistant prostate cancer who had received docetaxel and were using a LHRH analogue or had undergone orchiectomy were assessed in a randomised, placebo controlled, multicentre phase 3 clinical trial (AFFIRM). A total of 1199 patients were randomised 2:1 to receive either Xtandi orally at a dose of 160 mg once daily (N = 800) or placebo once daily (N = 399). Patients were allowed but not required to take prednisone (maximum daily dose allowed was 10 mg prednisone or equivalent). Patients randomised to either arm were to continue treatment until disease progression (defined as confirmed radiographic progression or the occurrence of a skeletal-related event) and initiation of new systemic antineoplastic treatment, unacceptable toxicity, or withdrawal. Following progression, 41% of study drug arm and 61.7% of placebo arm received ≥ 1 further systemic treatment; therefore, the observed survival data and Kaplan-Meier curve reflect a median duration of treatment of 8 months of enzalutamide vs. 3 months of placebo followed by additional treatments.
The following patient demographics and baseline disease characteristics were balanced between the treatment arms. The median age was 69 years (range 41 - 92) and the racial distribution was 93% Caucasian, 4% Black, 1% Asian, and 2% Other. The ECOG performance score was 0 - 1 in 91.5% of patients and 2 in 8.5% of patients; 28% had a mean Brief Pain Inventory score of ≥ 4 (mean of patient's reported worst pain over the previous 24 hours calculated for seven days prior to randomisation). Most (91%) patients had metastases in bone and 23% had visceral lung and/or liver involvement. At study entry, 41% of randomised patients had PSA progression only, whereas 59% of patients had radiographic progression. Fifty-one percent (51%) of patients were on bisphosphonates at baseline.
The AFFIRM study excluded patients with medical conditions that may predispose them to seizures and medications known to decrease the seizure threshold, as well as clinically significant cardiovascular disease such as uncontrolled hypertension, recent history of myocardial infarction or unstable angina, New York Heart Association class III or IV heart failure (unless ejection fraction was ≥ 45%), clinically significant ventricular arrhythmias or AV block (without permanent pacemaker).
The protocol pre-specified interim analysis after 520 deaths showed a statistically significant superiority in overall survival in patients treated with Xtandi compared to placebo (see Table 12 and Figures 12 and 13).
In addition to the observed improvement in overall survival, key secondary endpoints (radiographic progression-free survival, and time to first skeletal-related event) favoured Xtandi and were statistically significant after adjusting for multiple testing.
Radiographic progression-free survival as assessed by the investigator using RECIST v1.1 for soft tissue and appearance of 2 or more bone lesions in bone scan was 8.3 months for patients treated with Xtandi and 2.9 months for patients who received placebo [HR = 0.40, (95% CI: 0.35, 0.47)]; p < 0.0001). The analysis involved 216 deaths without documented progression and 645 documented progression events, of which 303 (47%) were due to soft tissue progression, 268 (42%) were due to bone lesion progression and 74 (11%) were due to both soft tissue and bone lesions.
The median time to first skeletal-related event was 16.7 months for patients treated with Xtandi and 13.3 months for patients who received placebo [HR = 0.69, (95% CI: 0.57, 0.84); p < 0.0001]. A skeletal-related event was defined as radiation therapy or surgery to bone, pathologic bone fracture, spinal cord compression, or change of antineoplastic therapy to treat bone pain. The analysis involved 448 skeletal-related events, of which 277 events (62%) were radiation to bone, 95 events (21%) were spinal cord compression, 47 events (10%) were pathologic bone fracture, 36 events (8%) were change in antineoplastic therapy to treat bone pain and 7 events (2%) were surgery to bone.

9785-CL-0410 study (enzalutamide post abiraterone in patients with metastatic CRPC).

The study was a single-arm study in 214 patients with progressing metastatic CRPC who received enzalutamide (160 mg once daily) after at least 24 weeks of treatment with abiraterone acetate plus prednisone. Median rPFS (radiologic progression free survival, the study's primary endpoint) was 8.1 months (95% CI: 6.1, 8.3). Median OS was not reached. PSA Response (defined as ≥ 50% decrease from baseline) was 22.4% (95% CI: 17.0, 28.6).
For the 69 patients who previously received chemotherapy, median rPFS was 7.9 months (95% CI: 5.5, 10.8). PSA Response was 23.2% (95% CI: 13.9, 34.9).
For the 145 patients who had no previous chemotherapy, median rPFS was 8.1 months (95% CI: 5.7, 8.3). PSA Response was 22.1% (95% CI: 15.6, 29.7).
Although there was a limited response in some patients from treatment with enzalutamide after abiraterone, the reason for this finding is currently unknown. The study design could neither identify the patients who are likely to benefit, nor the order in which enzalutamide and abiraterone should be optimally sequenced.

Elderly.

Of the 4403 patients in the controlled clinical trials who received enzalutamide, 3451 patients (78%) were 65 years and over and 1540 patients (35%) were 75 years and over. No overall differences in safety or effectiveness were observed between these older patients and younger patients.

5.2 Pharmacokinetic Properties

The pharmacokinetics of enzalutamide have been evaluated in prostate cancer patients and in healthy male subjects. The mean terminal half-life (t1/2) for enzalutamide in patients after a single oral dose is 5.8 days (range 2.8 to 10.2 days), and steady state is achieved in approximately one month. With daily oral administration, enzalutamide accumulates approximately 8.3-fold relative to a single dose. Daily fluctuations in plasma concentrations are low (peak-to-trough ratio of 1.25). Clearance of enzalutamide is primarily via hepatic metabolism, producing an active metabolite that circulates at approximately the same plasma concentration as enzalutamide.

Absorption.

Maximum plasma concentrations (Cmax) of enzalutamide in patients are observed 1 to 2 hours after administration. Based on a mass balance study in humans, oral absorption of enzalutamide is estimated to be at least 84.2%. Enzalutamide is not a substrate of the efflux transporters P-gp or BCRP. At steady state, the mean Cmax values for enzalutamide and its active metabolite are 16.6 microgram/mL (23% coefficient of variation [CV]) and 12.7 microgram/mL (30% CV), respectively.
Food has no clinically significant effect on the extent of absorption. In clinical trials, Xtandi was administered without regard to food.

Distribution.

The mean apparent volume of distribution (V/F) of enzalutamide in patients after a single oral dose is 110 L (29% CV). The volume of distribution of enzalutamide is greater than the volume of total body water, indicative of extensive extravascular distribution. Studies in rodents indicate that enzalutamide and its active metabolite can cross the blood brain barrier.
Enzalutamide is 97% to 98% bound to plasma proteins, primarily albumin. The active metabolite is 95% bound to plasma proteins. There was no protein binding displacement between enzalutamide and other highly bound medicinal products (warfarin, ibuprofen and salicylic acid) in vitro.

Metabolism.

Enzalutamide is extensively metabolised. There are two major metabolites in human plasma: N-desmethyl enzalutamide (active) and a carboxylic acid derivative (inactive). Enzalutamide is metabolised by CYP2C8 and to a lesser extent by CYP3A4/5 (see Section 4.5 Interactions with Other Medicines and Other Forms of Interactions), both of which play a role in the formation of the active metabolite. N-desmethyl enzalutamide was not metabolised by CYPs in vitro.
Under conditions of clinical use, enzalutamide is a strong inducer of CYP3A4, a moderate inducer of CYP2C9 and CYP2C19, and has no clinically relevant effect on CYP2C8 (see Section 4.5 Interactions with Other Medicines and Other Forms of Interactions).
In vitro, small amounts of N-desmethyl enzalutamide are metabolised to the carboxylic acid metabolite by carboxylesterase 1 which also plays a minor role in the metabolism of enzalutamide to the inactive carboxylic acid metabolite. Carboxylesterase 2 does not appear to play a role in the metabolism of either enzalutamide or N-desmethyl enzalutamide.

Excretion.

The mean apparent clearance (CL/F) of enzalutamide in patients ranges from 0.520 and 0.564 L/h.
Following oral administration of 14C-enzalutamide, 84.6% of the radioactivity is recovered by 77 days post dose: 71.0% is recovered in urine (primarily as the inactive metabolite, with trace amounts of enzalutamide and the active metabolite), and 13.6% is recovered in faeces (0.39% of dose as unchanged enzalutamide).

Linearity.

No major deviations from dose proportionality are observed over the dose range 40 to 160 mg. The steady-state Cmin values of enzalutamide and the active metabolite in individual patients remained constant during more than one year of chronic therapy, demonstrating time linear pharmacokinetics once steady state is achieved.

Pharmacokinetic characteristics in special populations.

Patients with hepatic impairment.

The pharmacokinetics of enzalutamide were examined in subjects with baseline mild (N = 6), moderate (N = 8) or severe (N = 8) hepatic impairment (Child-Pugh Class A, B and C respectively) and in 22 matched control subjects with normal hepatic function.
Following a single oral 160 mg dose of enzalutamide, the AUC and Cmax for enzalutamide in subjects with mild impairment increased by 5% and 24%, respectively, the AUC and Cmax of enzalutamide in subjects with moderate impairment increased by 29% and decreased by 11%, respectively and the AUC and Cmax of enzalutamide in subjects with severe impairment increased by 5% and decreased by 41%, respectively, compared to healthy control subjects.
For the sum of unbound enzalutamide plus the unbound active metabolite, the AUC and Cmax in subjects with mild impairment increased by 14% and 19%, respectively, the AUC and Cmax in subjects with moderate impairment increased by 14% and decreased by 17%, respectively, and the AUC and Cmax of enzalutamide in subjects with severe hepatic impairment increased by 34% and decreased by 27%, respectively, compared to healthy control subjects.

Patients with renal impairment.

No formal renal impairment study for enzalutamide has been completed. Patients with serum creatinine > 177 micromol/L (2 mg/dL) were excluded from clinical studies. Based on a population pharmacokinetic analysis, no dose adjustment is necessary for patients with calculated creatinine clearance (CrCL) values ≥ 30 mL/min (estimated by the Cockcroft and Gault formula). Enzalutamide has not been evaluated in patients with severe renal impairment (CrCL < 30 mL/min) or end-stage renal disease, and caution is advised when treating these patients. It is unlikely that enzalutamide will be significantly removed by intermittent haemodialysis or continuous ambulatory peritoneal dialysis.

Elderly.

No clinically relevant effect of age on enzalutamide pharmacokinetics was seen in the elderly population pharmacokinetic analysis.

Paediatric use.

The pharmacokinetics of enzalutamide in paediatric patients have not been established.

Gender and race.

The effect of gender on the pharmacokinetics of enzalutamide has not been evaluated. Most patients in the randomised clinical studies (> 75%) were Caucasian. Based on pharmacokinetic data from studies in Japanese and Chinese patients with prostate cancer, there were no clinically relevant differences in exposure among the populations. There are insufficient data to evaluate potential differences in the pharmacokinetics of enzalutamide in other races.

5.3 Preclinical Safety Data

Genotoxicity.

Enzalutamide did not induce mutations in the microbial mutagenesis (Ames) assay and was not clastogenic in either the in vitro cytogenetic assay with mouse lymphoma cells or the in vivo mouse micronucleus assay.

Carcinogenicity.

In a 6-month study in transgenic rasH2 mice, enzalutamide did not show carcinogenic potential (absence of neoplastic findings) at doses up to 20 mg/kg/day, which resulted in combined exposure levels (based on AUC), for enzalutamide plus its active metabolite M2, approximately half the clinical exposure in metastatic CRPC patients receiving 160 mg daily.
Daily oral dosing of rats for two years with enzalutamide at 10-100 mg/kg/day produced an increased incidence of neoplastic findings (compared to control) that were considered related to the primary pharmacology of enzalutamide. These included benign thymoma, fibroadenoma in the mammary glands, and benign Leydig cell tumours in the testes in males; benign granulosa cell tumour in the of ovaries in females; and adenoma in the pars distalis of the pituitary in both sexes.
Benign Leydig cell tumours are generally not considered relevant to humans based on experience with other anti-androgens. The human relevance of thymoma, pituitary adenoma and fibroadenoma in rats is unclear, but a potential relevance cannot be ruled out.
The urothelial papilloma and carcinoma of the urinary bladder observed at the 100 mg/kg/day dose in male rats are considered secondary to the continuous irritation caused by the increased accumulation of urinary crystal/calculi in the rat urinary bladder possibly due to its horizontal structure. The finding is not expected to occur in humans due to the upright positioning of the bladder. Prolonged irritation by urinary crystalluria or calculi may dispose rats to urothelial hyperplasia and/or tumour formation, however the incidence of bladder calculi reported in the clinical trials of enzalutamide was comparable between enzalutamide and the placebo groups.
The combined exposure levels (based on AUC) achieved in this study, for enzalutamide and its active metabolite M2 in rats were less than or similar to those in prostate cancer patients receiving 160 mg daily.

6 Pharmaceutical Particulars

6.1 List of Excipients

Xtandi contains the following inactive ingredients: caprylocaproyl macrogolglycerides, butylated hydroxyanisole, butylated hydroxytoluene, gelatin, sorbitol sorbitan solution, glycerol, purified water, titanium dioxide. The soft capsules also contain Opacode WB monogramming ink NSP-78-17827 Black.

6.2 Incompatibilities

Incompatibilities were either not assessed or not identified as part of the registration of this medicine.
Also see Section 4.5 Interactions with Other Medicines and Other Forms of Interactions.

6.3 Shelf Life

In Australia, information on the shelf life can be found on the public summary of the Australian Register of Therapeutic Goods (ARTG). The expiry date can be found on the packaging.

6.4 Special Precautions for Storage

Store below 25°C.

6.5 Nature and Contents of Container

Xtandi 40 mg capsules are supplied in a cardboard wallet incorporating a PVC/PCTFE/aluminium blister of 28 soft capsules. Each carton contains 4 wallets (112 soft capsules).

6.6 Special Precautions for Disposal

In Australia, any unused medicine or waste material should be disposed of by taking to your local pharmacy.

6.7 Physicochemical Properties

Enzalutamide is a white crystalline non-hygroscopic solid. It is practically insoluble in water.

Chemical structure.


Chemical name: 4-{3-[4-cyano-3-(trifluoromethyl)phenyl]- 5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl}-2-fluoro- N-methylbenzamide.
Molecular formula: C21H16F4N4O2S.

CAS number.

915087-33-1.

7 Medicine Schedule (Poisons Standard)

Schedule 4 - Prescription Only Medicine.

Summary Table of Changes