Skip to main content
Sage Choice logoLink to Sage Choice
. 2024 May 8;30(6):1057–1072. doi: 10.1177/10781552241238198

Review of drug-drug interactions in patients with prostate cancer

Andrew Ruplin 1, Eve Segal 1,, Tom McFarlane 2
PMCID: PMC11476483  PMID: 38720547

Abstract

Objective

The objective of this review is to provide an overview of common drug-drug interactions (DDIs) associated with prostate cancer treatments and outline recommendations for managing polypharmacy.

Data sources

A literature search of PubMed, Embase, and CINAHL was carried out to identify pharmacokinetic and pharmacodynamic changes caused by DDIs that are relevant for prostate cancer patients, DDIs between prostate cancer therapies and co-administered medications (both prescription and over-the-counter), and measures to prevent DDIs. Medication package inserts were used to identify the impact of DDI on the prostate cancer therapy and suggested interventions.

Data summary

No DDIs are expected for the LHRH agonists leuprolide acetate, histrelin, goserelin, or leuprolide mesylate. However, DDIs have been reported for GnRH antagonists, anti-androgens, PARP inhibitors, and taxanes. Although there are no confirmed DDIs for sipuleucel-T to date, it is not generally recommended to use sipuleucel-T concurrently with immunosuppressive medications. Interventions to prevent DDIs include the use of software that can detect clinically significant DDIs, up-to-date medication reconciliation, the inclusion of dedicated clinical pharmacists in cancer treatment teams, and patient/caregiver education.

Conclusions

Prostate cancer patients have a high risk of potential DDIs due to numerous new anti-cancer therapies, the increased use of treatment combinations, and the likelihood of comorbid conditions also requiring drug therapy. Drug-drug interaction screening software, up-to-date medication reconciliation, inclusion of oncology pharmacists on healthcare teams, and patient/caregiver education will aid the development of treatment plans that focus on achieving an optimal risk-benefit profile whilst reducing the risk of DDIs.

Keywords: drug-drug interactions, prostate cancer, LHRH agonists, polypharmacy, pharmacokinetics

Introduction

The prostate cancer (PCa) treatment landscape has expanded significantly over the past ten years and continues to evolve, with the approval of many new agents to treat this disease. Medications used to treat PCa include antineoplastic chemotherapies, hormonal therapies, poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors, radiopharmaceuticals, cellular immunotherapy, and immune checkpoint inhibitors. These treatment options have greatly reduced mortality rates for PCa patients.1,2 Of the systemic PCa treatments, hormonal agents and PARP inhibitors can be taken orally. Although this route of administration may afford patients greater convenience and improved quality of life compared to intravenous administration, these oral anticancer treatments are associated with the risk of nonadherence and many possible drug-drug interactions (DDIs).

Prostate cancer patients are at a high risk of DDIs due to the use of multiple new and existing therapies, including treatment combinations, comorbidities that require additional medication management, and increasing age. First, treatment combinations (vs. monotherapy) to achieve improved efficacy are now standard of care,3,4 which can result in more DDIs. Comorbidities requiring additional medications are common in PCa patients, 5 adding another layer of complexity to treatment management. Increasing age is another factor that increases DDI risk, as age-related physiological changes can alter drug pharmacokinetics (PK). For example, moderately or severely decreased kidney function is strongly associated with increasing age, 6 with decreased glomerular filtration rates impacting the excretion of some drugs. A study of drug combinations that are commonly problematic in older adults (≥65 years) listed 66 interactions deemed to be moderate (moderate injury requiring intervention), major (major injury requiring intervention), or catastrophic (leading to death, multiple permanent injuries, or irreversible health effects). 7 Most DDIs on this list concern antithrombotic agents, cardiovascular system drugs, central nervous system drugs, and drugs with a narrow therapeutic index (e.g., digoxin). However, their negative consequences can be avoided by treatment modification or monitoring. 7

The high likelihood of PCa patients experiencing DDIs is a significant clinical concern because unwanted effects may occur (e.g., loss of efficacy and/or increased adverse events). The objective of this review is to provide an overview of common DDIs associated with PCa treatments and outline recommendations for managing polypharmacy.

Patient vignettes of serious drug-drug interactions with prostate cancer therapies: introduction of cases

Four hypothetical cases of potential DDIs in PCa patients are presented in this review. These cases are introduced below and resolved at the end of the manuscript.

Case 1: A 75-year-old man with a history of myocardial infarction, psoriasis, and a recent diagnosis of metastatic castration-resistant PCa (mCRPC) is treated with atorvastatin, cyclosporine, enzalutamide, and relugolix.

Case 2: A 79-year-old man with CRPC and concurrent Type-2 diabetes is prescribed leuprolide acetate, abiraterone, prednisone, and repaglinide.

Case 3: A 77-year-old man with atrial fibrillation, who is on warfarin (for anticoagulation) and diltiazem (for rate control), is prescribed olaparib at 300 mg by mouth twice daily in combination with goserelin for mCRPC showing Ataxia-Telangiectasia Mutated (ATM) mutation.

Case 4: A 79-year-old man with non-metastatic CRPC (nmCRPC) and a history of venous thromboembolism necessitating long-term use of the anticoagulant apixaban is prescribed apalutamide and triptorelin.

Mechanisms of possible drug-drug interactions in patients with prostate cancer

Pharmacokinetics

Drug-drug interactions occur when the PK of one drug is modified by the administration of another. The four main PK parameters are absorption, distribution, metabolism, and excretion (ADME).

Causes for absorption-related DDIs include the inhibition or induction of drug transporters responsible for absorption within the enterocyte and changes in gastrointestinal (GI) bioavailability. Transporters linked to DDIs include P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and organic anion-transporting polypeptide 1B1 (OATP1B1). 8 Found in the liver, pancreas, kidneys, and GI tract, P-gp regulates intestinal drug absorption, promotes drug excretion, and limits drug tissue distribution. BCRP is expressed in many tissues, including the intestines and liver, and limits the intestinal absorption of some drugs. Organic anion-transporting polypeptide 1B1 is highly expressed in the sinusoidal membrane of hepatocytes and mediates the uptake of drugs from many classes. 8 Additionally, drug-induced changes in gastric pH or GI motility can alter GI drug absorption.9,10 Although not strictly DDIs, food-related changes in drug absorption are important, for example, grapefruit juice can alter the plasma concentration of many drugs 11 and food should not be consumed for at least two hours before and one hour after taking non-micronized abiraterone to avoid increased exposure (up to 10-fold). 12 This is further complicated by the requirement for taking prednisone concomitantly with abiraterone, 12 as steroids should be taken with food to reduce GI upset. Absorption of micronized abiraterone is unaffected by food. 13 Clinicians should ensure patients are aware of these types of issues and counsel them on how to safely incorporate the medications into a routine.

Distribution-related DDIs may occur when one drug is displaced by another with a stronger affinity for the same binding site. 10 Drugs are generally distributed via binding to plasma and tissue proteins, and as only free drug can be delivered to extravascular or tissue sites, the concentration of unbound drug is most relevant for efficacy. Important plasma proteins that bind drugs include albumin, α1-acid glycoprotein, and lipoproteins. 10 Acidic drugs tend to bind more extensively to albumin and basic drugs to α1-acid glycoprotein, lipoproteins, or both. 10 Highly protein-bound drugs, such as anticonvulsants, are very prone to DDIs mediated by effects on distribution. 14 To the best of our knowledge, there are no distribution-related DDIs for prostate cancer drugs. However, although not a DDI itself, drug-induced hepatotoxicity may indirectly lead to distribution-related DDIs by, for instance, causing hypoalbuminemia that would impact the binding of concomitant therapy.15,16 Hepatotoxicity due to chemotherapy drugs (e.g., docetaxel, cabazitaxel) occurs frequently in an unpredictable fashion, and pre-existing liver disease increases this risk; hence, clinicians should closely monitor patients receiving chemotherapy. 15

Drugs that impact the activity of cytochrome P450 (CYP450), the class of enzymes that plays a role in metabolizing 90% of drugs, 17 can alter the metabolism of concurrently administered drugs, leading to DDIs. 18 Drugs that impact CYP450 activity can be inhibitors, inducers, substrates, or both inhibitors/inducers and substrates for a specific enzymatic pathway.1820 The most significant CYP450 enzymes involved in DDIs are 3A4 and 2D6. 17 A common cause of severe DDIs is the inhibition of CYP450 enzymes by the co-administered drug. 21 When competing for the same enzyme, the elimination of one of the co-administered drugs will be reduced, leading to its accumulation. Enzyme induction, or an increase of activity through increased enzyme synthesis, 17 can also result in DDIs. With the increase in enzyme expression levels, drug exposure and consequently efficacy will decrease in a time-dependent manner. 10 Substrates are drugs that bind to an enzyme and are transformed into metabolites. 19 The strength of binding is measured as “binding affinity,” and substrate affinities can be classified as being weak, intermediate, or strong. 19 Drug-drug interactions can lead to changes in the binding affinity of a substrate, thus affecting its metabolism. 19 Not all CYP-mediated DDIs have clinically important consequences, but those impacting drugs with narrow therapeutic windows are more likely to be clinically significant. For example, concomitant use of enzalutamide (a strong 3A4 inducer and a moderate CYP2C9 and CYP2C19 inducer) with narrow therapeutic index drugs metabolized by CYP3A4 (e.g., cyclosporine), CYP2C9 (e.g., warfarin), and CYP2C19 (e.g., S-mephenytoin) should be avoided as exposure of the narrow therapeutic index drugs may decrease, 22 which could potentially reduce efficacy with unwanted clinical consequences. Additionally, strong inhibitors (e.g., clarithromycin) or inducers (e.g., rifampin) of CYP enzymes have a greater impact on exposure to the concomitant drug than moderate or weak inhibitors or inducers, so DDIs involving strong inhibitors/inducers are more clinically relevant.

Changes in drug excretion, which impact plasma drug concentration, can also cause DDIs. Renal drug excretion, the most common route, is primarily mediated by drug transporters within renal tubular cells. Potential mechanisms for such interactions include competition at a tubular reabsorption site leading to an increase in drug excretion, change in urinary pH and/or flow that can increase or decrease drug excretion, and inhibition of drug metabolism within the kidney. 23 Biliary excretion through the liver is another important route for drug elimination, 24 and drug-induced changes (e.g., through inhibition of efflux transporters) may change the PK of a concomitant drug, resulting in a DDI. 25

In conclusion, only absorption- and metabolism-related clinically significant DDIs for PCa drugs have been reported. However, clinicians should be mindful of DDIs related to all four PK parameters as not all possible DDIs have been reported, and DDIs may still occur with non-cancer drugs that patients with PCa are prescribed.

Pharmacodynamics

In addition to the PK interactions discussed above that alter a drug's pharmacological effect by changing absorption, distribution, metabolism, and/or excretion, pharmacodynamic (PD) interactions also require monitoring. A clinically important example is QT prolongation, which is when two or more drugs that prolong QT interval are co-prescribed, resulting in an additive or potentiating effect. 26 Androgen deprivation therapy (ADT), including luteinizing hormone-releasing hormone (LHRH) agonists and gonadotropin-releasing hormone (GnRH) antagonists, has been associated with significant prolongation of QT interval, 27 and PCa patients may be prescribed many concomitant therapies that can compound QT prolongation while receiving ADT. For example, some antidepressants (e.g., citalopram) are associated with QT prolongation. 28 Significant QT prolongation carries a risk of sudden cardiac death due to polymorphic tachycardia, also known as Torsades de Pointes. 29 The risk-benefit profile of using drugs that prolong QT should be considered on an individualized basis, taking into account the medical history, baseline electrocardiogram, and blood tests. 29 Labelling for ADT drugs recommends periodic monitoring of electrolytes and electrocardiograms to identify increases in the QT interval.3032 Monitoring levels of potassium, magnesium, and calcium is particularly important if there are additional clinical reasons for concern, e.g., GI disturbances.3032 It is vital that pharmacists and clinicians review all patients’ treatment plans to avoid DDIs that may increase cardiovascular risk.

Drug-drug interactions with common concomitant therapies in patients with PCa by drug class

As men with PCa frequently have comorbidities that require medications in addition to their cancer treatments, clinicians should be vigilant about potential DDIs when creating or adjusting treatment plans. Although DDI precautions are often described in the prescribing information,3335 clinicians may not always know where to find specific information or be aware of these precautions, and the DDIs listed may not be all-inclusive. A comprehensive list of DDIs for PCa therapies can be found in Table 1.

Table 1.

Drug-drug interactions of prostate cancer therapies.

a. LHRH Agonists

Drug Class LHRH Agonists
Therapy Leuprolide Acetate (IM:LUPRON DEPOT®) [1989] (SQ: ELIGARD®) [2002] Triptorelin Pamoate (TRELSTAR®) [2000] Goserelin Acetate (ZOLADEX®) [1989] Histrelin Acetate (VANTAS®) [2004] Leuprolide Mesylate (CAMCEVI™) [2021]
Avoid None listed30,31 None listed 36 None listed 37 None listed 38 None listed 39
Impact on Exposure of PCa Therapy or Co-Administered Drug N/A N/A N/A N/A N/A
Examples N/A N/A N/A N/A N/A
Suggested Intervention N/A N/A N/A N/A N/A

b.

LHRH Antagonist, and Combination PARP Inhibitor and Androgen Synthesis Inhibitor

Drug Class LHRH Antagonist Combination PARP Inhibitor and Androgen Synthesis Inhibitor
Therapy Relugolix (ORGOVYX™)
[2020]
Niraparib and Abiraterone Acetate (AKEEGA™)
[2023]
Avoid P-gp inhibitors 32 Combined P-gp and strong CYP3A inducers 32 Strong CYP3A4 inducers 40 Sensitive CYP2D6 or 2C8 substrates 40
Impact on Exposure of PCa Therapy or Co-Administered Drug Increased exposure of PCa therapy 32 Decreased exposure of PCa therapy 32 3A4 inducers: decreased exposure of abiraterone component of treatment 40 2D6/2C8 substrates: increased exposure to these substrates 40
Examples Antibiotics: erythromycin, cyclosporine
Antifungals: clotrimazole, ketoconazole
Antihypertensives: amlodipine, verapamil, reserpine, nifedipine, nicardipine, diltiazem
Anti-neoplastic agents: daunorubicin
Corticosteroids: dexamethasone
Anti-androgens: apalutamide, enzalutamide
Antibiotics: rifampicin
Antifungals: clotrimazole
Corticosteroids: rimexolone, dexamethasone
Herbal anti-depressants: St John's wort
Antibiotics: rifampicin
Anticonvulsants:
Phenytoin, carbamazepine
Antidepressants: desipramine
Antidiabetics: repaglinide
Antihypertensives: nebivolol
Antitussives: dextromethorphan
Suggested Intervention > Avoid P-gp inhibitors if possible. If concurrent therapy is necessary, take relugolix first, separate dosing by at least 6 h and monitor patients closely for toxicity
> Interrupting relugolix for up to 2 weeks may be appropriate for short courses of certain P-gp inhibitors 32
> Consider a switch to an injectable LHRH agonist (e.g., leuprolide acetate, goserelin acetate) or antagonist (degarelix)
> Avoid co-administration if possible. If unavoidable, increase the relugolix dose to 240 mg once daily
> After discontinuation of the combined P-gp and strong CYP3A inducer, resume the recommended relugolix dose of 120 mg once daily 32
> Consider a switch to an injectable LHRH agonist (e.g., leuprolide acetate, goserelin acetate) or antagonist (degarelix)
> Avoid strong CYP3A4 inducers with this medication
> There is no dose modification recommendation available 40
> Monitor for toxicity from CYP2D6 or 2C8 substrates
> Consider dose reduction of these substrates if there is a narrow therapeutic index 40

c.

Anti-androgens (1 of 2)

Drug Class Anti-androgens (1 of 2)
Therapy Abiraterone (ZYTIGA®, YONSA®)
[2011]
Enzalutamide (XTANDI®)
[2012]
Avoid CYP2D6 substrates Strong CYP3A4 inducers Strong CYP3A4 inhibitors Strong CYP2C8 inhibitors Moderate CYP3A4 or CYP2C8 inducers CYP3A4, CYP2C9, and CYP2C19 substrates
Impact on Exposure of PCa Therapy or Co-Administered Drug Increased exposure of co-administered drug(s) 12 Strong CYP3A4 inducers reduce the AUC by approximately half 13 Strong CYP3A4 inhibitors seem to have no meaningful pharmacokinetic effect on abiraterone 13 Increased exposure of PCa therapy 22 Decreased exposure of PCa therapy 22 Decreased exposure of co-administered drug(s) 22
Examples Antihypertensives: metoprolol, flecainide, carvedilol
Opioid analgesics: tramadol, codeine, oxycodone, methadone
Psychotropics: duloxetine, haloperidol, dextromethorphan, paroxetine, risperidone, clomipramine, amitriptyline, aripiprazole, fluoxetine, nortriptyline
Anticonvulsants: phenytoin, carbamazepine
Antibiotics: rifampin, rifabutin, rifapentine
Sedative: phenobarbital
Antiinfectives: ritonavir, erythromycin, ketoconazole, itraconazole, clarithromycin, atazanavir, saquinavir, telithromycin, indinavir, nelfinavir, voriconazole
Antidepressant: nefazodone
Calcium channel blocker: verapamil
Antihypertensive: felodipine
Lipid-regulator: gemfibrozil
Antiplatelet: clopidogrel
Antifungals: clotrimazole, ketoconazole
Anti-asthmatics: salmeterol, fluticasone
Cardiovascular modifying agents: bosentan, irbesartan, nicardipine, diltiazem, amlodipine, lovastatin
Antibiotic: nafcillin
Corticosteroids: dexamethasone
Antifungals: ketoconazole, clotrimazole
Erectile dysfunction treatments: tadalafil, vardenafil
Antihypertensives: lovastatin, diltiazem, amlodipine, atorvastatin, felodipine, nifedipine, losartan
Oral anticoagulants: warfarin, apixaban
Lipid-regulator: fluvastatin
Antidiabetics: rosiglitazone, glyburide, chlorpropamide, gliclazide
Nonsteroidal anti-inflammatory (NSAID): celecoxib, diclofenac, ibuprofen, naproxen
Antihistamine: chlorpheniramine
Suggested Intervention Monitor for increased toxicity of CYP2D6 substrates 12 >If a strong CYP3A4 inducer must be given concurrently, increase the frequency of abiraterone to twice a day
>Resume once daily frequency upon stopping the strong CYP3A4 inducer
>There is no recommended change to the milligram dose; only frequency
>Ex. Increase abiraterone 1000 mg once daily to 1000 mg twice a day 13
Monitor for abiraterone toxicity 12 Reduce the dose of enzalutamide to 80 mg once daily during treatment with strong CYP2C8 inhibitors 22 Increase the dose of enzalutamide to 240 mg once daily during treatment with strong CYP3A4 inducers 22 Monitor for decreased efficacy of CYP3A4, CYP2C9, and CYP2C19 substrates 22

c.

Anti-androgens (2 of 2)

Drug Class Anti-androgens (2 of 2)
Therapy Apalutamide (ERLEADA®)
[2018]
Darolutamide (NUBEQA™)
[2019]
Avoid Strong CYP2C8 or CYP3A4 inhibitors CYP3A4, CYP2C19, CYP2C9, UGT, P-gp, BCRP, or OATP1B1 substrates Combined P-gp and strong or moderate CYP3A inducers Combined P-gp and strong CYP3A inhibitors BCRP substrates
Impact on Exposure of PCa Therapy or Co-Administered Drug Increased exposure of PCa therapy 41 Decreased exposure of co-administered drug(s) 41 Decreased exposure of PCa therapy 42 Increased exposure of PCa therapy 42 Increased exposure of co-administered drug(s) 42
Examples Antihypertensive: felodipine
Lipid-regulator: gemfibrozil
Antiplatelet: clopidogrel
Antiasthmatics: salmeterol, fluticasone
Antiinfectives: ketoconazole, clotrimazole itraconazole, ritonavir, saquinavir, atazanavir, indinavir, nelfinavir, voriconazole, clarithromycin
Antidepressant: nefazodone
Erectile dysfunction treatments: tadalafil, vardenafil
Antihypertensives: amlodipine, diltiazem, losartan, olmesartan
Oral anticoagulant: warfarin
Lipid-regulators: lovastatin, fluvastatin, simvastatin
Nonsteroidal anti-inflammatory (NSAID): ibuprofen
Corticosteroid: dexamethasone
Antifungal: ketoconazole
Analgesic: acetaminophen
Antibiotic: rifampicin
Corticosteroids: dexamethasone, rimexolone
Antifungal: clotrimazole
Herbal supplement: St John's wort
Antibiotics: clarithromycin, troleandomycin,
Antifungals: ketoconazole, itraconazole
Antihypertensive: prazosin
Lipid-regulator: pravastatin
Antidiabetic: glyburide
Suggested Intervention >No initial dose changes to apalutamide are recommended
>Monitor and reduce the dose based on tolerability 41
Monitor for decreased efficacy of CYP3A4, CYP2C19, CYP2C9, UGT, P-gp, BCRP, or OATP1B1 substrates 41 >Avoid co-administration of these agents with darolutamide.
>There is no dose modification recommendation available 42
>Monitor for darolutamide toxicity more frequently.
>No dose adjustment recommended initially 42
Monitor patients more frequently for adverse reactions, and consider dose reduction of the BCRP substrate drug 42

d.

PARP Inhibitors

Drug Class PARP Inhibitors
Therapy Olaparib (LYNPARZA®)
[2020]
Rucaparib (RUBRACA®) [2020] Talazoparib (TALZENNA®)
[2023]
Avoid Strong or moderate CYP3A inducers 43 Strong or moderate CYP3A inhibitors 43 CYP2C9, 1A2, and/or 3A4 substrates 43 Sensitive CYP1A2, CYP3A, CYP2C9, or CYP2C19 substrates 44 P-gp inhibitors 2 BCRP inhibitors 2
Impact on Exposure of PCa Therapy or Co-Administered Drug Decreased exposure of PCa therapy 43 Increased exposure of PCa therapy 43 Increased exposure of co-administered drug(s) Increased exposure of co-administered drug(s) 44 Increased exposure of PCa therapy 2 Increased exposure of PCa therapy 2
Examples Antibiotics: rifampicin, nafcillin
Anticonvulsants: phenytoin, carbamazepine
Endothelin receptor antagonists:
bosentan
Herbal supplement: St John's Wort
HIV antivirals: efavirenz, etravirine
Stimulants: modafinil
Antibiotics: telithromycin, clarithromycin, ciprofloxacin, erythromycin
Antidepressants: nefazodone
Antiemetics: aprepitant
Antifungals: itraconazole, ketoconazole, voriconazole, posaconazole, fluconazole
Antihypertensives: diltiazem, imatinib, verapamil
Chemotherapies: crizotinib
HIV antivirals: amprenavir, atazanavir, darunavir/ritonavir, fosamprenavir
Anticoagulants: Warfarin Antiarrhythmic and blood pressure support: digoxin
Oral anticoagulant: warfarin
Proton-pump inhibitors: omeprazole
Sedatives: midazolam
Specific P-gp inhibitors requiring talazoparib dose adjustment for example:
Antiarrhythmics: amiodarone
Antibiotics: clarithromycin
Antifungals: itraconazole
Antihypertensives: verapamil, carvedilol
Antifungals: ketoconazole
HIV antivirals: nelfinavir
Suggested Intervention > Avoid strong or moderate inducers. There is no available recommended dose adjustment for olaparib 43 > Avoid strong or moderate inhibitors when possible.
> If unavoidable, reduce olaparib dose to 100 mg twice daily with a strong inhibitor or 150 mg twice daily with a moderate inhibitor.
> After the inhibitor is discontinued for 3–5 half-lives, resume original olaparib dose 43
> Monitor for increased toxicity of CYP2C9, 1A2, and/or 3A4 substrates > Monitor for increased toxicity of CYP1A2, CYP3A, CYP2C9, or CYP2C19 substrates.
> Adjust dosage of CYP1A2, CYP3A, CYP2C9, or CYP2C19 substrates, if clinically indicated 44
> Avoid listed P-gp inhibitors when possible. If unavoidable, reduce the dose of talazoparib to 0.75 mg once daily.
> When the P-gp inhibitor is discontinued for 3 to 5 half-lives, resume the original talazoparib dose 2
> Monitor for increased talazoparib toxicity 2

e.

Autologous Cellular Immunotherapy and Herbal Supplements

Drug Class Autologous Cellular Immunotherapy Herbal Supplements
Therapy Sipuleucel-T (PROVENGE®)
[2010]
Saw Palmetto Black Cohosh
Avoid None listed 45 CYP3A4 substrates 46 CYP2D6 substrates 46 CYP2C9 substrates 46 CYP2D6 substrates 47
Impact on Exposure of PCa Therapy or Co-Administered Drug N/A Increased exposure of co-administered drug(s) Increased exposure of co-administered drug(s) Increased exposure of co-administered drug(s) Increased exposure of co-administered drug(s)
Examples N/A Anticoagulants: rivaroxaban Anticoagulants: Warfarin
Antihypertensives:
metoprolol, flecainide, carvedilol
Opioid analgesics:
tramadol, codeine, oxycodone, methadone
Psychotropics:
duloxetine, haloperidol, dextromethorphan, paroxetine, risperidone, clomipramine, amitriptyline, aripiprazole, fluoxetine, nortriptyline
Anticoagulants: warfarin
Antidiabetics: tolbutamide
Antidepressants: fluoxetine
Antiepileptics: phenytoin
Antihypertensives: losartan
Diuretics: torsemide
Anticoagulants: Warfarin
Antidepressants: Desipramine
Antihypertensives:
metoprolol, flecainide, carvedilol
Opioid analgesics:
tramadol, codeine, oxycodone, methadone
Psychotropics:
duloxetine, haloperidol, dextromethorphan, paroxetine, risperidone, clomipramine, amitriptyline, aripiprazole, fluoxetine, nortriptyline
Suggested Intervention N/A N/A N/A N/A

f.

Taxane Chemotherapies

Drug Class Taxane Chemotherapies
Therapy Doxetaxel (TAXOTERE®) [2004] Cabazitaxel (JEVTANA®) [2010] Paclitaxel (ABRAXANE®)
[2005]
Avoid Strong CYP3A inhibitors 48 Strong CYP3A inhibitors 49 CYP3A4 inhibitors 50 CYP3A4 substrates 50 CYP2C8 inhibitors 50 CYP2C8 substrates 50
Impact on Exposure of PCa Therapy or Co-Administered Drug Increased exposure of PCa therapy 48 Increased exposure of PCa therapy 49 Increased exposure of PCa therapy 50 Increased exposure of co-administered drug(s) 50 Increased exposure of PCa therapy 50 Increased exposure of co-administered drug(s) 50
Examples Antibiotics: clarithromycin, telithromycin
Antidepressants: nefazodone
Antifungals: ketoconazole, itraconazole, voriconazole
HIV antiviral: ritonavir, saquinavir, atazanavir, indinavir, nelfinavir
Antibiotics: clarithromycin, telithromycin
Antidepressants: nefazodone
Antifungals: ketoconazole, itraconazole, voriconazole
Herbal supplements: St John's wort
HIV antivirals: ritonavir, saquinavir, atazanavir, indinavir, nelfinavir
Antibiotics: clarithromycin, telithromycin
Antidepressants: nefazodone
Antifungals: ketoconazole, itraconazole, voriconazole
HIV antivirals: ritonavir, saquinavir, atazanavir, indinavir, nelfinavir
Antibiotics: cyclosporin, erythromycin
Sedatives: diazepam
Anti-asthmatics: salmeterol, fluticasone
Antifungals: clotrimazole, ketoconazole
Antihypertensive: felodipine
Antiplatelet: clopidogrel
Lipid-regulator: gemfibrozil
Antidiabetics: repaglinide
Antidepressants: desipramine
Antihypertensives: nebivolol
Antitussives: dextromethorphan
Antivirals: dasabuvir
Suggested Intervention > Monitor for increased side effects of docetaxel.
> Consider 50% dose reduction of docetaxel 48
> Monitor for increased side effects of cabazitaxel
> Consider 25% dose reduction of cabazitaxel 49
N/A N/A N/A N/A

Luteinizing hormone-releasing hormone agonists and gonadotropin-releasing hormone antagonists

Luteinizing hormone-releasing hormone agonists (e.g., leuprolide acetate, triptorelin pamoate, goserelin acetate, histrelin acetate, leuprolide mesylate) and GnRH antagonists (e.g., relugolix, degarelix) are options for ADT, the foundational systemic therapy for men with advanced PCa. Although no PK DDI studies have been conducted with leuprolide acetate, DDIs are not expected as it is a peptide that is primarily degraded by peptidase, not CYP450 enzymes as noted in specific studies, and is not highly protein-bound.30,31,51 Similar to leuprolide acetate, no potential DDIs or precautions are listed for histrelin, 38 goserelin, 37 or leuprolide mesylate.3739 The triptorelin label recommends that it should not be used concomitantly with hyperprolactinemic drugs because hyperprolactinemia reduces the number of pituitary GnRH receptors, although no DDI studies have been conducted. 36 However, DDIs have been noted for GnRH antagonists. Relugolix should not be co-administered with P-gp inhibitors 32 or combined P-gp and strong CYP3A inducers. 32 Co-administration of relugolix with P-gp inhibitors leads to increased relugolix exposure, which may increase the risk of adverse reactions. 32 If co-administration is unavoidable, relugolix should be taken first, dosing should be separated by at least six hours, and patients should be monitored frequently for adverse reactions. 32 Co-administration of relugolix with combined P-gp and strong CYP3A inducers should be avoided because the DDI leads to decreased relugolix exposure, which may decrease efficacy. 32 However, if co-administration is unavoidable, the relugolix dose should be increased to 240 mg once daily. Although not included in the label, a 2022 review recommended that relugolix should not be administered concomitantly with many androgen-receptor-axis targeted therapies (e.g., abiraterone, apalutamide), cabazitaxel, or lutetium Lu 177 vipivotide tetraxetan as the safety of these combinations have not been established. 52 The National Comprehensive Cancer Network (NCCN) guidelines for the treatment of PCa have similar recommendations on combining novel hormonal therapies with relugolix and note that further studies are needed to evaluate the safety of these combinations. 53 The degarelix label states that clinically significant CYP450 interactions are unlikely, but a study found that its combination with enzalutamide resulted in significant QT prolongation compared to degarelix monotherapy. 54 Hence, clinicians should be aware that clinically significant DDIs are possible when prescribing GnRH antagonists concurrently with other frequently used drugs. This is a relevant factor to be considered when selecting the most appropriate drug for ADT.

Anti-androgens

First-generation anti-androgens (bicalutamide, flutamide, nilutamide) are androgen receptor inhibitors used in combination with ADT. Co-administration of bicalutamide, a CYP3A4 inhibitor, with a CYP3A4 substrate such as midazolam increased mean substrate concentrations 1.5-fold, potentially increasing risk of adverse events. 55 Bicalutamide can also displace anticoagulants from binding sites, causing loss of efficacy; there have been post-marketing reports of serious bleeding in patients taking both bicalutamide and an anticoagulant e.g., warfarin. 55 Flutamide prescribing information also warns that when administered concomitantly with warfarin, close monitoring and anticoagulant dose adjustment may be necessary. 56 Nilutamide inhibits the activity of CYP450 isoenzymes and may reduce the metabolism of drugs requiring these systems, especially those with a low therapeutic margin (e.g., vitamin K, phenytoin), causing delayed elimination and increased half-life leading to toxicity. 41 Dose reduction of the co-administered drug may need to be required. 41

Second generation anti-androgens (e.g., enzalutamide, apalutamide, darolutamide, abiraterone), which inhibit binding of androgen-to-androgen receptors, are hormone therapies that are administered concurrently with ADT. Enzalutamide is indicated for metastatic castration-sensitive PCa (mCSPC), non-metastatic castration-sensitive PCa (nmCSPC), and CRPC 22 ; apalutamide is indicated for mCSPC and nmCRPC 42 ; darolutamide is indicated for mCSPC (with docetaxel) and nmCRPC 57 ; and abiraterone is indicated (with prednisone) for mCSPC and mCRPC. 12 Co-administration of enzalutamide with the CYP inhibitors gemfibrozil and itraconazole has been shown to increase the composite area under the curve of enzalutamide by 220% and 130%, respectively. 58 Similarly, co-administration of a single dose of apalutamide with the CYP inhibitor gemfibrozil increases systemic exposure of apalutamide by 68%. 42 These increases in exposure of enzalutamide and apalutamide may increase the risk of toxicity. Conversely, strong CYP3A4 inducers like rifampin, which is also a CYP2C8 inducer, have been shown to decrease plasma concentrations of enzalutamide. 22 Although rifampin, a strong CYP3A4 and CYP2C8 inducer, decreases apalutamide active moieties, these changes are not significant enough to require dose modification. 59 A phase 1 study evaluating co-administration of a P-gp and CYP3A4 inducer with darolutamide in healthy volunteers demonstrated a 72% decrease in darolutamide exposure, potentially decreasing efficacy. 60 Additionally, preclinical data indicated that darolutamide may inhibit substrates of BCRP, OATP1B1, OAT3, and OATP1B3. 60 Abiraterone should not be co-administered with strong CYP3A4 inducers or CYP2D6 substrates. 12 A DDI between abiraterone and a strong CYP3A4 inducer (e.g., rifampin) results in decreased abiraterone exposure, which could decrease drug efficacy and negatively impact clinical outcomes. 12 If co-administration is unavoidable, clinicians should increase abiraterone dosing frequency. 12 Additionally, co-administration of abiraterone and CYP2D6 substrates with a narrow therapeutic index (e.g., thioridazine) leads to a DDI that increases the exposure of the substrate. 12 If an alternative treatment cannot be used, clinicians should exercise caution and consider a dose reduction of the concomitant CYP2D6 substrate. 12

Given that many patients with PCa are at high risk of cardiovascular adverse events, 61 DDIs with direct oral anticoagulants (DOACs), such as dabigatran, rivaroxaban, and apixaban, are important to consider. For example, co-administration of the strong CYP3A4 inducer enzalutamide with apixaban, which is metabolized by CYP3A4, reduces apixaban exposure, potentially decreasing efficacy for treatment and prevention of blood clots. 43 Thus, DDIs with these second-generation anti-androgens are very likely, particularly as they are often administered as part of combination treatments in PCa management and with other drugs for concurrent illness.

PARP inhibitors

PARP inhibitors (olaparib, talazoparib, rucaparib, niraparib), which are indicated for mCRPC, are at high risk for DDIs. First, clinicians must consider DDIs with PARP inhibitors themselves. Olaparib should not be co-administered with strong or moderate CYP3A inhibitors or inducers. 44 If talazoparib is co-administered with some P-gp inhibitors (itraconazole, amiodarone, carvedilol, clarithromycin, itraconazole, and verapamil), the talazoparib dose should be reduced to 0.75 mg once daily and patients should be monitored for increased adverse reactions. 2 Patients taking both talazoparib and BCRP inhibitors should also be monitored for potential increased toxicity. 2 Concomitant administration of rucaparib with some CYP1A2, CYP3A, CYP2C9, or CYP2C19 substrates can increase the systemic exposure of these substrates, potentially increasing the frequency or severity of adverse effects. 40 More recently, PARP inhibitors can be administered in combination with other PCa therapies (for example, olaparib with abiraterone, prednisone or prednisolone, 44 and ADT; talazoparib with enzalutamide and ADT 2 ; rucaparib with ADT), 40 so DDIs with these other therapies must be considered as well. The tablet combining niraparib and abiraterone acetate should not be administered with strong CYP3A4 inducers, or CYP2D6 or 2C8 substrates. 45 Currently, there are more PARP inhibitor combination therapies under clinical investigation and undergoing FDA review. Therefore, new DDIs may emerge, requiring increased vigilance in PCa DDIs.

Sipuleucel-T

Sipuleucel-T is an immunotherapy derived from patients’ own cells approved only for treatment of mCRPC that is asymptomatic or minimally symptomatic. 62 To date, there are no confirmed DDIs that influence PK or PD of sipuleucel-T. 62 However, based on the presumed effect that immunosuppressive medications, such as chemotherapy or corticosteroids, may diminish patients’ immune response to sipuleucel-T or impede the harvesting of cells during leukapheresis, it is not generally recommended to use these treatments concurrently. 62 Of note, patients on immunosuppressive medications were excluded from clinical trials. 62 Patients who are receiving steroids as part of their treatment with docetaxel and/or abiraterone, for example, need to be appropriately counseled to also stop the steroid when they discontinue docetaxel/abiraterone in anticipation of receiving sipuleucel-T.

Taxane chemotherapy

Taxanes (docetaxel, paclitaxel, and cabazitaxel) have historically been used in metastatic castration-resistant disease upon failure of other therapies, but earlier use in the course of disease may become more frequent given the demonstrated survival benefit in combination with ADT, 63 or, more recently, as part of triplet therapy in combination with abiraterone or darolutamide and ADT.48,64 Clinically significant DDIs may occur with this class of drugs, as well as overlap when used in combination with anti-androgens due to commonalities in CYP450 enzymes. For example, the metabolism of docetaxel may be altered if it is co-administered with CYP450 3A4 inducers, inhibitors, or substrates. 49 A 50% dose reduction of docetaxel should be considered if co-administration with a strong CYP3A4 inhibitor is unavoidable. 49 If co-administered with a strong CYP3A inhibitor, cabazitaxel exposure may increase, thereby increasing the risk of adverse effects. 50 Additionally, co-administration of cabazitaxel with a CYP3A inducer is expected to decrease cabazitaxel exposure, potentially decreasing efficacy. 50 Therefore, co-administration of cabazitaxel with strong CYP3A inhibitors and inducers should be avoided. 50 Cabazitaxel prescribing information also advises that patients should refrain from taking St John's wort. 50 Paclitaxel prescribing information warns that caution should also be exercised when administering paclitaxel concomitantly with CYP3A4 and CTP2C8 substrates or inhibitors, 65 but does not list suggested interventions if co-administration is unavoidable.

Strong patient/provider partnerships reduce risk of drug-drug interactions

In addition to healthcare providers working together in inter-disciplinary teams (e.g., primary care physicians [PCP], oncologists, urologists, pharmacists) to identify and prevent potential DDIs, a member of the healthcare team should proactively engage patients early in the treatment process to establish a strong patient/provider partnership. Patients who feel that they have a strong relationship with members of their healthcare team may, for instance, be more likely to fully disclose all over-the-counter and herbal supplements they take, which when administered with prescription medicines can have serious clinical consequences due to DDIs.66,67 Effective medication reconciliation is crucial to ensure medication safety and appropriate, informed clinical decision-making. 68

Drug-drug interactions may occur with herbal supplements commonly taken by PCa patients, including saw palmetto and black cohosh. Saw palmetto is an over-the-counter product some patients may use to treat symptoms of benign prostatic hyperplasia.69,70 There is evidence that saw palmetto may decrease the level of testosterone in the body, 71 inhibit an enzyme that converts testosterone to DHT (5α-reductase), 72 and have an anti-inflammatory effect on the prostate. 46 An in vitro study found potent inhibition of CYP3A4, 2D6 and 2C9 by saw palmetto, and the authors concluded that there is potential for strong adverse DDIs. 73 A separate publication suggested that inhibition of CYP2C9 by saw palmetto may account for interactions between the supplement and warfarin. 74 It also may reduce the number of estrogen and androgen receptors and thus have hormone-like effects. 46 Additionally, a potential DDI with the anticoagulant rivaroxaban, in which saw palmetto may have contributed to hemopericardium by increasing rivaroxaban activity, has been reported. 75 Although not a direct DDI, a laboratory study found that saw palmetto can increase the sensitivity of normal prostate cells to radiation. 76 As this can increase the risk of complications, patients with PCa should consult their physician before using saw palmetto supplements during radiation therapy. 76 Black cohosh root is commonly taken by PCa patients on hormonal therapy to reduce hot flashes. 47 Drug-drug interactions may occur between black cohosh and CYP2D6 substrates (e.g., desipramine), 77 which could decrease the efficacy of the co-administered drug or increase the risk of adverse effects.

In addition to non-prescription medications that may be selected by the patient or a caregiver without informing their healthcare team, use of medications like a testosterone replacement therapy, which could negate the impact of ADT, may not be disclosed to the healthcare team if the patient does not understand what medications they are using and why. Thus, a good partnership between the provider and patient and/or caregiver, in which the patient fully discloses their medication regimen and the provider educates the patient on each medication, is an important element in preventing adverse DDIs.

Patient vignettes of serious drug-drug interactions with prostate cancer therapies continued: resolution of cases

The following are the continuation of the four illustrative patient case reports: this section presents how the potential DDIs could be prevented to avoid negative clinical consequences.

Case 1: Two potential clinically significant DDIs were flagged by DDI screening software: enzalutamide and atorvastatin, 22 and relugolix and cyclosporine. 32 Clinicians amended the treatment plan by replacing atorvastatin with rosuvastatin and instructed the patient to take relugolix at least six hours before cyclosporine. If these DDIs had not been flagged by the software or a pharmacist, the patient may have suffered hypercholesterolemia, potentially recurrent myocardial infarctions due to decreased atorvastatin exposure, and/or adverse reactions associated with relugolix.

Case 2: A potential clinically significant DDI between abiraterone and repaglinide was flagged by DDI screening software, allowing clinicians to amend the treatment plan. If this DDI had not been identified by the software or a pharmacist, inhibition of CYP2C8 by abiraterone could have led to increased repaglinide exposure, 12 causing the patient to suffer hypoglycemic episodes with blood glucose levels less than 30 mg/dL and possibly require hospitalization.

Case 3: Two potential clinically significant DDIs were flagged by DDI screening software and the treating clinician consulted the clinical pharmacist for input. The first DDI was between olaparib and warfarin. The clinical pharmacist recommended replacing warfarin with a direct oral anticoagulant to avoid increased serum concentration of warfarin and consequent increased risk of warfarin-associated adverse events. The second DDI was between olaparib and diltiazem. Diltiazem is a known CYP3A4 inhibitor and is categorized as “moderate.” 78 The FDA-approved prescribing information for olaparib recommends against concurrent treatment with moderate or strong CYP3A4 inhibitors. 44 However, the prescribing information also suggests the dose of olaparib can be reduced to 150 mg twice daily if moderate inhibitors must be used concurrently. 44 If this DDI had not been identified, inhibition of CY3PA4 by diltiazem could have led to increased olaparib exposure, 44 thus increasing the risk for adverse reactions.

Case 4: A potential clinically significant DDI between apalutamide and apixaban was flagged by a clinical pharmacist and the treatment plan was discussed. Apalutamide is a known “strong” inducer of CYP3A4 as well as an inducer of P-glycoprotein (P-gp)/ABCB1. Both apixaban 79 and rivaroxaban 80 are not recommended to be used concurrently with any strong inducer of both CYP3A4 and P-gp. Among direct oral anticoagulants, edoxaban is still potentially sensitive to P-gp induction-mediated decreased efficacy based on data when used with strong CYP3A4 inducer and P-gp inducer rifampin 81 but, from a risk-benefit standpoint, represents the least amount of overall interaction potential. Given the lack of efficacy monitoring for DOACs 82 and adherence concerns with enoxaparin, 83 in discussion with the patient and treating physician the decision was made to switch to edoxaban. If this DDI had not been identified, decreased apixaban exposure caused by the strong CYP3A4 inducer apalutamide could have reduced apixaban efficacy and increased risk of recurrent venous thromboembolism.

Discussion of measures to prevent drug-drug interactions

In addition to vigilance by clinicians and pharmacists when formulating treatment plans and developing strong patient-provider partnerships, further measures are important to prevent DDIs. These interventions include the use of software that can detect clinically significant DDIs, up-to-date medication reconciliation, the inclusion of dedicated clinical pharmacists in cancer treatment teams, and patient/caregiver education.

As clinicians may not have the resources to identify and manage drug toxicities, DDI screening software can help reduce the risk of preventable harm. An analysis of data from 775,186 patients across 572 pharmacies found that computerized prescription entry and drug interaction screening reduced the dispensing of prescriptions with potentially severe interactions by 63%. 84 There are various options for DDI screening software. A random-sampling-based statistical algorithm aimed at identifying DDIs based on MEDLINE records was found to have high accuracy, based on case studies of four commonly prescribed drugs (warfarin, ibuprofen, furosemide, and sertraline). A separate study comparing five common DDI programs (Lexi-Interact, Micromedex Drug Interactions, iFacts, Medscape, and Epocrates) found that Lexi-Interact identified DDIs with the greatest accuracy and comprehensiveness. 85 Although such programs cannot replace clinician evaluation of medical histories, monitoring, and evidence-based judgement, they represent powerful support tools.

However, it should be noted that DDI screening software and pharmacists can only identify potential DDIs if patients’ electronic medical records are accurate. Given that elderly PCa patients are likely to receive medicines prescribed by multiple specialists (e.g., urologists, cardiologists, oncologists, endocrinologists, PCP, etc.), accurate and up-to-date medication reconciliation may be a challenge. A study reporting an increased risk of potentially inappropriate drug combinations with greater numbers of treating physicians concluded that a single PCP and single dispensing pharmacy may be protective factors against DDIs. Healthcare providers across disciplines should work together to ensure that patient records are accurate and up-to-date to help in the prevention of DDIs. If a PCa patient does not have a PCP, the treating urologist or oncologist should ensure that the patient has one before starting advanced care and maintain open communication with the PCP. The PCP can manage the patient's non-oncology-related issues, manage relevant DDIs, care for the “whole” patient, and oversee all potential DDIs, allowing the oncologist or urologist to focus on treating the cancer.

Oncology pharmacy is a field that combines the expertise of pharmacy practice with a focus on hematology and oncology, 86 and the inclusion of oncology pharmacists in cancer treatment teams can help prevent DDIs. A position statement from the International Society of Oncology Pharmacy Practitioners states that the primary role of an oncology pharmacist is to ensure safe and appropriate medication use for individuals with cancer. 86 This includes assessing medication safety, adjusting drug dose and verifying that prescriptions are accurate. 86 An analysis of clinical interventions by an oncology pharmacist found that, out of 346 total interventions, one was classified as potentially lethal, 114 as serious and 140 as significant, 87 indicating that the presence of an oncologist pharmacist on inter-disciplinary teams is highly likely to improve patient safety.

Educating patients and caregivers on how to identify and prevent DDIs may also be beneficial and should increase feelings of empowerment. The Food and Drug Administration has published a guide to help patients avoid DDIs, which includes how to read drug labels and what questions to ask their doctor or pharmacist such as “Can I take it with other drugs? Should I avoid certain foods, beverages, or other products? What are possible drug interaction signs I should know about?” 88 A member of the healthcare team could direct patients and/or caregivers to this guide before starting new medications and answer any questions their patients may have. There is some evidence of a gap between the amount of information about DDIs that patients would like to receive from pharmacists and what they actually receive, 89 indicating a need to improve patient-centered communication in pharmacies. To the best of our knowledge, the impact of patient and/or caregiver education on reducing DDI's has not been assessed in a trial.

Conclusions

In general, prevention of DDIs has become an increasingly important consideration in the development of treatment plans, particularly for an aging population with associated comorbidities and polypharmacy. Patients with PCa have a high risk of potential DDIs due to numerous new anti-cancer therapies, the increased use of treatment combinations, and the likelihood of comorbid conditions also requiring drug therapy. Drug-drug interactions can result in adverse PK or PD effects that can impact efficacy, toxicity, and/or pharmacological activity of one or more of the medicines being taken. Although most DDIs are listed on drug labels, these listings are not comprehensive, and clinicians may not always be aware of appropriate precautions. An especially important PD DDI to consider for PCa patients is QT prolongation, which can increase the risk of arrhythmia and cardiac arrest. Drug-drug interaction screening software, up-to-date medication reconciliation, inclusion of oncology pharmacists on healthcare teams, patient/caregiver education, and strong patient/provider partnerships will assist development of treatment plans that focus on achieving an optimal risk-benefit profile whilst reducing the risk of DDIs.

Acknowledgment

Editorial support was provided by Xelay Acumen Group, Inc. (funded by Tolmar, Inc.). All the authors have authorized the submission of their manuscript via Xelay Acumen Group, Inc. and have approved all statements and declarations, including conflicting interests and funding.

Footnotes

Author contributions: AR, EV, and TM made substantial contributions to the acquisition and interpretation of literature, revised the draft critically for important intellectual content, contributed to writing and formatting throughout the manuscript, and approved the final version of the manuscript.

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article. Editorial support was funded by Tolmar, Inc.

Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: Editorial support was supported by the Tolmar, Inc.,

References

  • 1.Cancer Stat Facts: Prostate Cancer. National Cancer Insitute. Surveillance, epidemiology, and end results program, https://seer.cancer.gov/statfacts/html/prost.html (accessed 26 April 2023).
  • 2.TALZENNA® (talazoparib) capsules, for oral use [prescribing information]. New York, NY: Pfizer Inc., 2018. [Google Scholar]
  • 3.Mottet N, Cornford P, van den Bergh RCN, et al. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on Prostate Cancer. The Netherlands: European Association of Urology, 2023. [Google Scholar]
  • 4.Lowrance WT, Breau RH, Chou R, et al. Advanced prostate cancer: AUA/ASTRO/SUO guideline. J Urol 2021; 205: 14–21. [DOI] [PubMed] [Google Scholar]
  • 5.Jefferson M, Drake RR, Lilly M, et al. Co-morbidities in a retrospective cohort of prostate cancer patients. Ethn Dis 2020; 30: 185–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Coresh J, Astor BC, Greene T, et al. Prevalence of chronic kidney disease and decreased kidney function in the adult US population: third national health and nutrition examination survey. Am J Kidney Dis 2003; 41: 1–12. [DOI] [PubMed] [Google Scholar]
  • 7.Anrys P, Petit AE, Thevelin S, et al. An international consensus list of potentially clinically significant drug-drug interactions in older people. J Am Med Dir Assoc 2021; 22: 2121–33.e24. [DOI] [PubMed] [Google Scholar]
  • 8.Giacomini KM, Balimane PV, Cho SK, et al. International transporter consortium commentary on clinically important transporter polymorphisms. Clin Pharmacol Ther 2013; 94: 23–26. [DOI] [PubMed] [Google Scholar]
  • 9.Lee HT, Lee YJ, Chung SJet al. et al. Effect of prokinetic agents, cisapride and metoclopramide, on the bioavailability in humans and intestinal permeability in rats of ranitidine, and intestinal charcoal transit in rats. Res Commun Mol Pathol Pharmacol 2000; 108: 311–323. [PubMed] [Google Scholar]
  • 10.Palleria C, Di Paolo A, Giofrè C, et al. Pharmacokinetic drug-drug interaction and their implication in clinical management. J Res Med Sci 2013; 18: 601–610. [PMC free article] [PubMed] [Google Scholar]
  • 11.Bressler R. Grapefruit juice and drug interactions. Exploring mechanisms of this interaction and potential toxicity for certain drugs. Geriatrics 2006; 61: 12–18. [PubMed] [Google Scholar]
  • 12.ZYTIGA® (Abiraterone acetate) [prescribing information]. Horsham, PA: Janssen Biotech, Inc., 2011. [Google Scholar]
  • 13.YONSA® (Abiraterone acetate) tablets, for oral use [prescribing information]. Cranbury, New Jersey: Sun Pharmaceuticals, Inc., 2011. [Google Scholar]
  • 14.Chadwick B, Waller DG, Edwards JG. Potentially hazardous drug interactions with psychotropics. Adv Psychiatr Treat 2005; 11: 440–449. [Google Scholar]
  • 15.Bernardi M, Maggioli C, Zaccherini G. Human albumin in the management of complications of liver cirrhosis. Crit Care 2012; 16: 211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fisher K, Vuppalanchi R, Saxena R. Drug-Induced liver injury. Arch Pathol Lab Med 2015; 139: 876–887. [DOI] [PubMed] [Google Scholar]
  • 17.Lynch T, Price A. The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. Am Fam Physician 2007; 76: 391–396. [PubMed] [Google Scholar]
  • 18.McDonnell AM, Dang CH. Basic review of the cytochrome p450 system. J Adv Pract Oncol 2013; 4: 263–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Deodhar M, Rihani A, Arwood SB, et al. Mechanisms of CYP450 inhibition: understanding drug-drug interactions due to mechanism-based inhibition in clinical practice. Pharmaceutics 2020; 12(9): 846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jacobs TG, Marzolini C, Back DJet al. et al. Dexamethasone is a dose-dependent perpetrator of drug-drug interactions: implications for use in people living with HIV. J Antimicrob Chemother 2022; 77: 568–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Guengerich FP. Inhibition of cytochrome P450 enzymes by drugs-molecular basis and practical applications. Biomol Ther (Seoul) 2022; 30: 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.XTANDI® (enzalutamide) [prescribing information]. Tokyo: Astellas Pharma Inc., 2012. [Google Scholar]
  • 23.Bonate PL, Reith K, Weir S. Drug interactions at the renal level. Implications for drug development. Clin Pharmacokinet 1998; 34: 375–404. [DOI] [PubMed] [Google Scholar]
  • 24.Rollins DE, Klaassen CD. Biliary excretion of drugs in man. Clin Pharmacokinet 1979; 4: 368–379. [DOI] [PubMed] [Google Scholar]
  • 25.Kong LL, Shen GL, Wang ZY, et al. Inhibition of P-glycoprotein and multidrug resistance-associated protein 2 regulates the hepatobiliary excretion and plasma exposure of thienorphine and its glucuronide conjugate. Front Pharmacol 2016; 7: 242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Khatib R, Sabir FRN, Omari C, et al. Managing drug-induced QT prolongation in clinical practice. Postgrad Med J 2021; 97: 452–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Gagliano-Jucá T, Travison TG, Kantoff PW, et al. Androgen deprivation therapy is associated with prolongation of QTc interval in men with prostate cancer. J Endocr Soc 2018; 2: 485–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Castro VM, Clements CC, Murphy SN, et al. QT Interval and antidepressant use: a cross sectional study of electronic health records. Br Med J 2013; 346: f288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Nachimuthu S, Assar MD, Schussler JM. Drug-induced QT interval prolongation: mechanisms and clinical management. Ther Adv Drug Saf 2012; 3: 241–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.ELIGARD® (leuprolide acetate for injectable suspension) [prescribing information]. Fort Collins, CO: Tolmar Pharmaceuticals, Inc., 2002. [Google Scholar]
  • 31.LUPRON DEPOT® (leuprolide acetate for depot suspension) [prescribing information]. North Chicago, IL: AbbVie Inc., 1989. [Google Scholar]
  • 32.ORGOVYX (relugolix) tablets, for oral use [prescribing information]. Myovant Sciences, Inc., 2020. [Google Scholar]
  • 33.FDA. Guidance for Industry: Drug Interaction Studies—Study Design, Data Analysis, Implications for Dosing, and Labeling Recommendations. Silver Spring, MD: Division of Drug Information at Center for Drug Evaluation and Research, 2012. [Google Scholar]
  • 34.Administration FaD. Guidance for Industry: Drug Interaction Studies—Study Design, Data Analysis, Implications for Dosing, and Labeling Recommendations. Silver Spring, MD: Division of Drug Information at Center for Drug Evaluation and Research, 2012. [Google Scholar]
  • 35.Porcalla A, Barshteyn N, Snyder Set al. et al. An innovative, collaborative, and strategic approach to proactively evaluate and update drug interactions based on prescribing information of newly approved medicinal products. Ther Innov Regul Sci 2017; 51: 780–786. [DOI] [PubMed] [Google Scholar]
  • 36.TRELSTAR® (triptorelin pamoate for injectable suspension), for intramuscular use [prescribing information]. Mississauga, Ontario: Verity Pharma, 2000. [Google Scholar]
  • 37.ZOLADEX® (goserelin acetate implant) [prescribing information]. Cambridge: AstraZeneca, 1989. [Google Scholar]
  • 38.VANTAS® (histrelin acetate) subcutaneous implant [prescribing information]. Endo Pharmaceuticals Solutions Inc., 2004. [Google Scholar]
  • 39.CAMCEVI® (leuprolide) injectable emulsion, for subcutaneous use [prescribing information]. Taipei City: Foresee Pharmaceuticals, 2021. [Google Scholar]
  • 40.RUBRACA® (rucaparib) tablets, for oral use [prescribing information]. Boulder, CO: Clovis Oncology, Inc., 2016. [Google Scholar]
  • 41.NILANDRON® (nilutamide) Tablets [prescribing information]. St. Michael, Barbados: Concordia Pharmaceuticals Inc., 2015. [Google Scholar]
  • 42.ERLEADA (apalutamide) tablets, for oral use [prescribing information]. Beerse: Janssen Products, LP, 2018. [Google Scholar]
  • 43.Shatzel JJ, Daughety MM, Olson SR, et al. Management of anticoagulation in patients with prostate cancer receiving enzalutamide. J Oncol Pract 2017; 13: 720–727. [DOI] [PubMed] [Google Scholar]
  • 44.LYNPARZA® (olaparib) tablets, for oral use [prescribing information]. Wilmington, DE: AstraZeneca, 2014. [Google Scholar]
  • 45.AKEEGA™ (niraparib and Abiraterone acetate) tablets, for oral use. Horsham, PA: Janssen Biotech, Inc., 2023. [Google Scholar]
  • 46.Gordan A, Shaughnessy A. Saw Palmetto for prostate disorders. Am Fam Physician 2003; 67: 1281–1283. [PubMed] [Google Scholar]
  • 47.Black Cohosh Root. European Medical, https://www.europeanmedical.info/prostate-cancer-2/black-cohosh-root.html (2022, accessed 10 May 2023).
  • 48.Smith MR, Hussain M, Saad F, et al. Darolutamide and survival in metastatic, hormone-sensitive prostate cancer. N Engl J Med 2022; 386: 1132–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.TAXOTERE® (docetaxel) injection, for intravenous use [prescribing information]. Paris: Sanofi-aventis U.S. LLC, 1996. [Google Scholar]
  • 50.JEVTANA® (cabazitaxel) injection, for intravenous use [prescribing information]. Bridgewater, NJ: sanofi-aventis U.S. LLC, 2010. [Google Scholar]
  • 51.Chi D, Berchuck A, Dizon DSet al. et al. Principles and Practice of Gynecologic Oncology. Philadelphia, PA: Lippincott Williams & Wilkins, 2017. [Google Scholar]
  • 52.Sahu KK, Tripathi N, Agarwal Net al. et al. Relugolix in the management of prostate cancer. Expert Rev Anticancer Ther 2022; 22: 891–902. [DOI] [PubMed] [Google Scholar]
  • 53.Mohler JL, Antonarakis ES, Armstrong AJ, et al. Prostate cancer, version 2.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2019; 17: 479–505. [DOI] [PubMed] [Google Scholar]
  • 54.Gheorghe ACD, Ciobanu A, Hodorogea AS, et al. Evolution of electrocardiographic repolarization parameters during antiandrogen therapy in patients with prostate cancer and hypogonadism. Cardiovasc Toxicol 2020; 20: 390–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.CASODEX® (bicalutamide) tablet, for oral use. Wilmington, DE: AstraZeneca Pharmaceuticals LP, 1995. [Google Scholar]
  • 56.EULEXIN® (flutamide) capsules, USP [prescribing information]. Kenilworth, NJ: Schering Corporation, 2001. [Google Scholar]
  • 57.NUBEQA (darolutamide) tablets, for oral use [prescribing information]. Berlin: Bayer, 2019. [Google Scholar]
  • 58.Gibbons JA, de Vries M, Krauwinkel W, et al. Pharmacokinetic drug interaction studies with enzalutamide. Clin Pharmacokinet 2015; 54: 1057–1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Van den Bergh A, Snoeys J, De Zwart L, et al. Pharmacokinetic drug-drug interaction of apalutamide, part 2: investigating interaction potential using a physiologically based pharmacokinetic model. Clin Pharmacokinet 2020; 59: 1149–1160. [DOI] [PubMed] [Google Scholar]
  • 60.Zurth C, Koskinen M, Fricke R, et al. Drug-Drug interaction potential of darolutamide: in vitro and clinical studies. Eur J Drug Metab Pharmacokinet 2019; 44: 747–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Leong DP, Fradet V, Shayegan B, et al. Cardiovascular risk in men with prostate cancer: insights from the RADICAL PC study. J Urol 2020; 203: 1109–1116. [DOI] [PubMed] [Google Scholar]
  • 62.PROVENGE® (sipuleucel-T) suspension for intravenous infusion [prescribing information]. Dendreon Corporation, 2010. [Google Scholar]
  • 63.Sweeney CJ, Chen YH, Carducci M, et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. N Engl J Med 2015; 373: 737–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Fizazi K, Foulon S, Carles J, et al. Abiraterone Plus prednisone added to androgen deprivation therapy and docetaxel in de novo metastatic castration-sensitive prostate cancer (PEACE-1): a multicentre, open-label, randomised, phase 3 study with a 2×2 factorial design. Lancet 2022; 399: 1695–1707. [DOI] [PubMed] [Google Scholar]
  • 65.ABRAXANE® (paclitaxel) INJECTION [prescribing information]. Summit, New Jersey: Abraxis BioScience, LLC., 2005. [Google Scholar]
  • 66.Izzo AA, Ernst E. Interactions between herbal medicines and prescribed drugs: a systematic review. Drugs 2001; 61: 2163–2175. [DOI] [PubMed] [Google Scholar]
  • 67.Honig PK, Gillespie BK. Drug interactions between prescribed and over-the-counter medication. Drug Saf 1995; 13: 296–303. [DOI] [PubMed] [Google Scholar]
  • 68.Barnsteiner J. Medication reconciliation. In: Hughes R. (ed.) Patient Safety and Quality: An Evidence-Based Handbook for Nurses. Rockville, MD: Agency for Healthcare Research and Quality, 2008, pp. 459–470. [PubMed] [Google Scholar]
  • 69.Saw palmetto. Mount Sinai, https://www.mountsinai.org/health-library/herb/saw-palmetto (accessed 11 April 2023).
  • 70.Opoku-Acheampong AB, Penugonda K, Lindshield BL. Effect of saw Palmetto supplements on androgen-sensitive LNCaP human prostate cancer cell number and Syrian hamster flank organ growth. Evid Based Complement Alternat Med 2016; 2016: 8135135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Sudeep HV, Thomas JV, Shyamprasad K. A double blind, placebo-controlled randomized comparative study on the efficacy of phytosterol-enriched and conventional saw palmetto oil in mitigating benign prostate hyperplasia and androgen deficiency. BMC Urol 2020; 20: 86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Suzuki M, Ito Y, Fujino T, et al. Pharmacological effects of saw palmetto extract in the lower urinary tract. Acta Pharmacol Sin 2009; 30: 227–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Yale SH, Glurich I. Analysis of the inhibitory potential of Ginkgo biloba, Echinacea purpurea, and Serenoa repens on the metabolic activity of cytochrome P450 3A4, 2D6, and 2C9. J Altern Complement Med 2005; 11: 433–439. [DOI] [PubMed] [Google Scholar]
  • 74.Ge B, Zhang Z, Zuo Z. Updates on the clinical evidenced herb-warfarin interactions. Evid Based Complement Alternat Med 2014; 2014: 957362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Shivamurthy P, Brar N, Therrien ML. Isolated hemopericardium associated with rivaroxaban: first case report. Pharmacotherapy 2014; 34: e169–e172. [DOI] [PubMed] [Google Scholar]
  • 76.Saw Palmetto. Memorial Sloan Kettering Cancer Center, https://www.mskcc.org/cancer-care/integrative-medicine/herbs/saw-palmetto (2020, accessed 11 April 2023).
  • 77.Gurley BJ, Gardner SF, Hubbard MA, et al. In vivo effects of goldenseal, kava kava, black cohosh, and valerian on human cytochrome P450 1A2, 2D6, 2E1, and 3A4/5 phenotypes. Clin Pharmacol Ther 2005; 77: 415–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.CARDIZEM® LA (diltiazem hydrochloride) extended-release tablets [prescribing information]. North Chicago, Illinois: Abbott Laboratories, 1982. [Google Scholar]
  • 79.ELIQUIS (apixaban) tablets for oral use [prescribing information]. Princeton, New Jersey: Bristol-Myers Squibb, 2012. [Google Scholar]
  • 80.XARELTO® (rivaroxaban) tablets, for oral use [prescribing information]. Titusville, New Jersey: Janssen Pharmaceuticals, Inc., 2011. [Google Scholar]
  • 81.SAVAYSA (edoxaban) tablets for oral use. Tokyo, Japan: Daiichi Sankyo, Inc., 2015. [Google Scholar]
  • 82.Dunois C. Laboratory monitoring of direct oral anticoagulants (DOACs). Biomedicines 2021; 9: 445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sakon C, Nikirk M, O’Brien ARW. Enoxaparin adherence for venous thromboembolism prophylaxis in hospitalized patients with sickle cell disease. Expert Rev Hematol 2023; 16: 147–150. [DOI] [PubMed] [Google Scholar]
  • 84.Halkin H, Katzir I, Kurman I, et al. Preventing drug interactions by online prescription screening in community pharmacies and medical practices. Clin Pharmacol Ther 2001; 69: 260–265. [DOI] [PubMed] [Google Scholar]
  • 85.Kheshti R, Aalipour M, Namazi S. A comparison of five common drug-drug interaction software programs regarding accuracy and comprehensiveness. J Res Pharm Pract 2016; 5: 257–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Holle LM, Bilse T, Alabelewe RM, et al. International society of oncology pharmacy practitioners (ISOPP) position statement: role of the oncology pharmacy team in cancer care. J Oncol Pharm Pract 2021; 27: 785–801. [DOI] [PubMed] [Google Scholar]
  • 87.Duarte NC, Barbosa CR, Tavares MG, et al. Clinical oncology pharmacist: effective contribution to patient safety. J Oncol Pharm Pract 2019; 25: 1665–1674. [DOI] [PubMed] [Google Scholar]
  • 88.Drug Interactions: What You Should Know. US Food and Drug Administration, https://www.fda.gov/drugs/resources-you-drugs/drug-interactions-what-you-should-know (2013, accessed 29 September 2021).
  • 89.Kim MG, Lee NE, Sohn HS. Gap between patient expectation and perception during pharmacist-patient communication at community pharmacy. Int J Clin Pharm 2020; 42: 677–684. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Oncology Pharmacy Practice are provided here courtesy of SAGE Publications

RESOURCES