Skip to main content
British Journal of Clinical Pharmacology logoLink to British Journal of Clinical Pharmacology
. 2018 Jan 29;84(4):679–693. doi: 10.1111/bcp.13490

Critical evaluation of causality assessment of herb–drug interactions in patients

Charles Awortwe 1,2,, Memela Makiwane 2, Helmuth Reuter 2, Christo Muller 1, Johan Louw 1, Bernd Rosenkranz 2
PMCID: PMC5867089  PMID: 29363155

Abstract

The aim of this review was to assess the severity of adverse drug reactions (ADRs) due to herb–drug interactions (HDI) in patients taking herbs and prescribed medications based on published evidence. Electronic databases of PubMed, the Cochrane Library, Medline and Scopus were searched for randomized or nonrandomized clinical studies, case–control and case reports of HDI. The data were extracted and the causal relationship of ADRs as consequences of HDI assessed using Horn's drug interaction probability scale or Roussel Uclaf Causality Assessment Method scoring systems. The mechanism of interaction was ascertained using Stockley's herbal medicine interaction companion. Forty‐nine case reports and two observational studies with 15 cases of ADRs were recorded. The majority of the patients were diagnosed with cardiovascular diseases (30.60%), cancer (22.45%) and renal transplants (16.32%) receiving mostly warfarin, alkylating agents and cyclosporine, respectively. HDI occurred in patients resulting in clinical ADRs with different severity. Patients may poorly respond to therapeutic agents or develop toxicity due to severe HDI, which in either scenario may increase the cost of treatment and/or lead to or prolong patient hospitalization. It is warranted to increase patient awareness of the potential interaction between herbs and prescribed medicines and their consequences to curb HDI as a potential health problem.

Keywords: adverse drug reactions, causality assessment, herbal drugs, herb–drug interactions, herb‐induced liver injury, side effects

Introduction

The risk of herb–drug interaction (HDI) is increasingly recognized as a public health problem often accompanied by life‐threatening adverse drug events, prolonged hospitalization and loss of life 1. With a rise in global burden of noncommunicable diseases 2, 3, 4, 5, pain syndromes, anxiety, depression and aging 6, 7, co‐usage of prescribed medications and herbal products will persistently be a potential health problem in both developed and developing nations. For instance, cases of acute rejection episodes have been reported in heart, renal or liver transplant patients stabilized on immunosuppressives including cyclosporine and tacrolimus due to concomitant intake of St John's wort (SJW) known to induce drug metabolizing enzymes 8, 9. In two case reports, patients with a history of generalized anxiety disorder and mild traumatic brain injury experienced serotonin syndrome and hypomania after addition of SJW or Ginkgo biloba to either a buspirone or fluoxetine regimen 10, 11. Patients' deliberate refusal to disclose their use of herbal medicines to clinicians have led to underreporting of clinically relevant HDI cases. In addition, underestimation of the consequences of HDI due to lack of a standardized probability scoring system has contributed to wrong classification of the causality of adverse drug reactions (ADRs). Different experimental models have been used to understand the mechanism of such interactions 12, 13, 14, 15.

In most instances, in vitro and animal studies are conducted to evaluate the effect of herbal extracts and phytochemical constituents on pharmacokinetic and pharmacodynamic properties of probe drugs 16, 17, 18, 19, 20, 21, 22. However, preclinical investigations often do not correlate with findings in human subjects 23, 24, 25, 26, 27. Clinical studies in patients adopting a population pharmacokinetic approach is considered as the gold standard to determine the clinical consequences of potential interaction of herbal medicines with prescribed medications 28. Nevertheless, pharmacokinetic interaction studies are often conducted in healthy subjects 29, 30. Incidences of ADRs are rarely reported in these types of studies due to limited number of co‐administered drugs, small sample size, lack of an appropriate placebo arm and the recruitment of healthy young subjects. Only a few cases recorded HDI‐linked ADRs and the causality of adverse effect was assessed using an appropriate classification tool 31, 32.

Few studies have adapted the Naranjo tool to classify the severity of HDI 33, 34. The Naranjo tool was designed to evaluate ADRs due to a single agent, and it is thus less useful for the categorization of HDI linked ADRs 35. Fugh‐Berman and Ernst developed a 10‐point scoring system to assess the reliability scale of HDI 36. Later, Horn and colleagues proposed a specific tool for causality assessment of drug–drug interactions by adopting the Naranjo scale as a guide. The Horn's drug interaction probability scale (DIPS) consists of 10 questions each with three response options to which a score is assigned 37. In 1985, the French pharmaceutical company Roussel Uclaf organized a consensus meeting for experts in hepatology to define terminologies used in drug induced liver injuries (DILI) assessment and qualitative criteria based on the French causality assessment method (CAM). The meeting outcomes included additional criteria and assigned weight to each criterion leading to the Roussel Uclaf Causality Assessment Method (RUCAM). RUCAM is regarded as a well‐established tool for qualitative assessment of causality in cases of suspected herb‐induced liver injury (HILI) and DILI 38, 39, 40. Nevertheless, only a few HDI studies have applied the Horn's DIPS and RUCAM scores to ascertain the causal relationship of ADRs. In addition, for studies where one or both classification systems were used, the mechanism of HDI was either unclear or not reported. Stockley's herbal medicine interaction companion contains arrays of herbal medicines with documented literature of interaction studies conducted in vitro, in animal models and healthy subjects 41. Observational and survey control studies have adopted this companion to flag herbal medicines with potential to cause adverse effect due to interaction with prescribed medications. Such information could be useful in educating both clinicians and patients about the consequences of consuming herbal medicines with prescribed medications based on established reports. This review, therefore, aims to conduct causality assessment on HDI in clinical trials, case–controls studies, and case reports in in/out‐patients from January 2001 to August 2017 using Horn's DIPS for general HDI and RUCAM for HILI specific injuries, respectively. In addition, the mechanism of HDI was evaluated by using Stockley's herbal medicines interaction companion.

Methods

Databases of PubMed, the Cochrane Library, Medline and Scopus were searched from January 2001 to August 2017. A combination of the following keywords were used for title, abstract and keywords fields: herbal drugs, herbal supplements, phytochemicals, drug interactions, herb–drug interactions, side effect and adverse‐effects. Preliminary assessment was conducted on abstracts retrieved from the databases to identify publications that met the pre‐specified criteria. Articles describing randomized or nonrandomized clinical studies, case–control and case reports in both in‐ and out‐patients where conventional medications and herbal medicines were co‐administered met the inclusion criteria. The review excluded all HDI studies conducted in healthy subjects, in vitro and in vivo models and other sources of natural products. Reference lists from eligible publications were another source for articles included in this review. Full text articles of abstracts meeting the eligibility criteria were extracted, validated and summarized (Table 1). Two independent reviewers (C.A. and M.M.) conducted causality assessment on validated data using the Horn's DIPS for general ADRs or RUCAM for HILI. The Stockley's herbal medicine interaction companion was consulted in cases where the mechanism of interaction was not reported.

Table 1.

Summary of observational studies included

Description Levy et al., 2017 Jeong et al., 2012
Study design, population and duration Cross‐sectional study of 947 patients hospitalized in 12 departments of a tertiary academic medical Centre in Haifa, Israel from 2009 to 2014. A prospective study of 313 inpatients (87 male and 226 female) hospitalized in two Oriental Hospitals of Daejeon University, Daejeon, Korea from August 2008 to October 2010
Inclusion criteria Intake of herbal medicine at least 1 week prior to hospitalization.
Patients at 18 years and above who were able to communicate and provide verbal informed consent.
Intake of herbal medicine at least 10 days during hospitalization.
Exclusion criteria Not mentioned A life expectancy of <12 months (by judgment of the patient's doctor); current or previous liver diseases including carrier status of hepatitis virus, renal disease, or autoimmune disease; and abnormal baseline results on liver or renal function tests on hospitalization.
Sex Male: 152 (33%)
Female: 306 (67%)
Male 87 (27.8%)
Female 226 (72.2%)
Mean age 61.3 years 51 years
Herb users with different comorbidities 458 (100%) 313 (100%)
Metabolic 243 (53%)
Circulatory system 237 (52%) 87 (27.8%)
Haemato‐oncological 95 (21%) 77 (24.6%)
Musculoskeletal system 66 (21.1%)
Neurologic 38 (8%) 40 (12.8%)
Digestive system 39 (8.5%) 17 (5.4%)
Endocrine 132 (29%)
Respiratory 39 (8.5%)
Renal 37 (8%)
Rheumatological 37 (8%)
Pregnancy 30 (7%)
Psychiatric 29 (6%)
Ophthalmological 21 (5%)
Urological 17 (4%)
Hepatobiliary 10 (2%)
HIV 23 (7.3)
Others 3 (1.0)
Mode of causality assessment DIPS RUCAM score
Cases of ADR Total = 17
Herbs/pure compounds = 7
Other supplements = 10
Total = 6
Herb/pure compounds = 6
Other supplements = 0

ADRs classified by RUCAM score for liver injury; no provision of patient history and ADRs classified by Horn's DIPS score for other injuries. ADR = adverse drug reaction; HIV = human immunodeficiency virus; DIPS = drug interaction probability scale; RUCAM = Roussel Uclaf Causality Assessment Method

The results of this review are divided into two parts: 1) characteristics of studies, and 2) tabularized description of studies, probability score and mechanism of interactions.

Results

The initial electronic database search identified 5113 articles, of which 1963 were screened after removal of duplications. In total 5035 publications were excluded based on titles and abstracts. On application of the review inclusion criteria to the 78 full‐text papers, a further 24 articles were excluded. Five additional papers were discarded because full text of two studies were not available, two papers in German and one paper in Turkish could not be translated into English. Thus, 49 full text articles consisting of 47 case reports publications and two observational studies were included in this review. One publication presented three different reports and hence, 49 case reports were reviewed. The majority of the case reports described patients between 51–69 years (n = 12, 24.49%) and older than 70 years (n = 11, 22.45%), respectively. Fifteen of the patients (30.60%) had a history of cardiovascular conditions such as hypertension, dyslipidaemia, myocardial infarction, atrial fibrillation and cardiomyopathy. Thirteen patients (86.67%) with a history of cardiovascular conditions were on warfarin treatment. Eleven cases (22.45%) were cancer patients of whom the majority were on alkylating agents (busulfan, temozolomide and trabectedin) and/or kinase inhibitors (imatinib and gefitinib). There were eight case reports of patients with renal transplant (16.33%) of which five patients (62.5%) received cyclosporine as immunosuppressive agent. Three HIV positive patients received a protease inhibitor (lopinavir), an integrase inhibitor (raltegravir) or a non‐nucleoside reverse transcriptase inhibitor (efavirenz). There were eight patients (16.33%) with central nervous system diseases such as depression, schizophrenia, anxiety disorders and seizures. The patients with central nervous system disorders received anticonvulsants (lamotrigine and phenytoin), atypical antipsychotics (clozapine), a selective serotonin reuptake inhibitor (sertraline) or a norepinephrine reuptake inhibitor (venlafaxine). Concomitant intake of SJW and Panax ginseng were recorded in patients treated with warfarin, cyclosporine or kinase inhibitors (gefitinib and imatinib). HDI cases reported to be probable and highly probable were 25 (51.02%) and four (8.16%), respectively. Eighteen cases (36.73%) were classified as possible whilst two cases (4.08%) were identified to be doubtful. The predominant ADRs were elevated liver enzymes, INR, GIT disturbances and rhabdomyolysis. The detailed descriptions and causality scales of the 49 cases included in this review are presented in Table 2.

Table 2.

Case reports of herb–drug interactions in individual patients

Patient description [reference] Herb supplement Latin name (common use) Affected drug Concomitant medications Signs or symptoms of possible interaction Plausible mode of interaction Probability scale 37
58‐year‐old man with a history of phalangeal fracture 42 Ginkgo biloba (improve cognitive function) Sodium aescinate Cefuroxime Elevated serum creatinine and blood, urea and nitrogen (BUN) levels CYP2C9 and 3A4 inhibition Probable (5)
56‐year‐old man with a history of orthotopic liver transplantation 43 Curcuma longa (anti‐inflammatory and liver protection) Tacrolimus Steroids, IV immunoglobulin, rituximab, sofosbuvir/ribavirin Abdominal distention, scrotal and peripheral oedema, increased creatinine CYP3A4/5 inhibition Probable (7)
50‐year‐old Caucasian man with monophasic synovial sarcoma 44 Diosmin (blood disorders) Trabectedin Epirubicin, ifosfamide Rhabdomyolysis, increase serum myoglobin, creatinine phosphokinase and liver function CYP3A4 inhibition Probable (5)
52‐year‐old white, woman, with a history of major depressive disorder, hypertension and dyslipidaemia 45 Celery root (menopause) Venlafaxine SJW Confusion and speech abnormalities CYP2D6 inhibition Possible (4)
49‐year‐old man without a family history had partial seizures with a secondarily generalization since he was age 27 years. 46 Noni juice (Tahitian Noni Original Bioactive Beverage) Phenytoin Lamotrigine, lorazepam, clobazam Low phenytoin level in blood CYP2C9 induction Highly probable (9)
44‐year‐old white man generalized tonic–clonicseizure disorder 47 Ginseng (improve mental and physical performance) Lamotrigine Deer antler velvet, sildenafil, hydrocortisone cream DRESS syndrome (pruritic rash on more than 50% of his body, eosinophilia, myalgias, and elevated liver enzymes) UGT2B7 inhibition Probable (5)
35‐year‐old woman with a history of depression 48 Centella asiatica and Fucus vesiculosus(rash, eczema, psoriasis and other skin infections) Venlafaxine None Progressive dyspnoea over the previous, New York Heart Association(NYHA) functional class III, myalgia, and dry cough CYP2D6 slow metabolizer or CYP2D6 inhibition by Centella asiatica Possible (3)
56‐year‐old woman Caucasian patient diagnosed witha temporal–parietal glioblastoma WHO IV 49 Bu Zhong Yi Qi Wan (promotes physical strength) Temozolomide Dexamethasone, pantoprazol, levetiracetam, mirtazapine, valaciclovir Grade II thrombopenia and elevated liver enzymes Unknown Probable (5)
78‐year‐old Hispanic, man, renal transplant patient 50 Pneumus boldus(mild dyspepsia and spastic gastrointestinal complaints) Tacrolimus mycophenolate, metoprolol, simvastatin, tamsulosin, aspirin, lisinopril, amlodipine, calcium carbonate, omeprazole, and insulin Asymptomatic Unknown Probable (6)
44‐year‐old obese man (body mass index: 53 kg m –2 ) with stage‐III (ISS) IgG‐κ multiple myeloma 51 Flor‐Essence (anticancer) Busulfan Bortezomib, melphalan GI toxicities, including Grade 3 nausea,vomiting, diarrhoea and oesophagitis Unknown Possible (2)
82‐year‐old woman with a history of hypertension, hypothyroidism, gastritis, atrial fibrillation 52 Artemisia absinthium (sore throat) Warfarin Nebivolol, valsartan hydrochlorothiazide, levothyroxine and esomeprazole abdominal pain and black, tarry stool Unknown Possible (4)
56‐year‐old Caucasian man with a history of progressive abdominal pain due to liposarcoma with a retroperitoneum mass (5 × 8 cm) 53 Chokeberry juice (anticancer) Trabectedin Peg‐granulocyte colony stimulating factor Rhabdomyolysis, G4 pancytopenia, elevated liver enzymes CYP3A4 inhibition Possible (4)
16‐year‐old child with nephrotic syndrome 54 Berberine (diarrhoea) Tacrolimus Prednisone Renal toxicity CYP3A4/5 inhibition Possible (4)
52‐year‐old woman with a history of severe psoriasis 55 Red clover (menopausal flushing) Methotrexate Not mentioned Severe vomiting and epigastric pain Probably OAT3 inhibition Possible (4)
23‐year‐old Japanese woman with multiple sclerosis 56 Lutein and melilot supplements (indigestion and other GI tract problems) Interferon β‐1b Not mentioned Jaundiced palms, elevated alanine transaminase, periventricular and juxtacortical hyperintense signal lesions Unknown Probable (5)
41‐year‐old woman with disorganized schizophrenia 57 SJW (depression) Clozapine Not mentioned Increased disorganization and tension CYP3A4 induction Probable (6)
56‐year‐old white Caucasian man with a history of HIV+ 58 Ginseng‐based oral lozenges (sexual disability) Raltegravir plus lopinavir/ritonavir Aspirin, esomeprazole, trimethoprim/sulfamethoxazole Generalized pruritus, scratching lesions, increased transaminase, visible jaundiced skin and mucous membranes CYP3A4 inhibition Probable (6)
71‐year‐old Ecuadorean‐American woman with a history of complete left knee arthroplasty 33 Himalayan goji juice (cleanse body) Warfarin Ezetimibe, lisinopril, famotidine, meclizine, alprazolam, and diphenhydramine Ecchymosis, epistaxis, and one episode of haematochezia, elevated INR CYP2C9 inhibition Probable (7)
41‐year‐old man diagnosed with HIV 59 Ginkgo biloba (improve cognitive function) Efavirenz Zidovudine, lamivudine Increased viral load at 1350 copies ml–1 CYP3A4 induction Probable (6)
61‐year‐old man with a T3N1M0 (stage IIIA) squamous cell carcinoma of the lung 60 Echinacea (common cold and flu) Etoposide Cisplatin, omeprazole, enalapril, hydrocodone/ paracetamol, prochlorperazine, ondansetron, vitamins B12, E, D, B17 and C Seizure‐like activity, grade 4 thrombocytopenia Unknown Possible (2)
85‐year‐old man with a history of hypertension, old anterior wall myocardial infarctio nand atrial fibrillation 61 SJW (depression) Warfarin Not mentioned Upper gastrointestinal bleeding, increased INR Possibly additive clotting effect Probable (6)
46‐year‐old African American woman with a history of stage 1 sarcoidosis, uterine fibroids, anaemia, cardiomyopathy and depression 62 Cranberry juice (constipation) Warfarin Not mentioned Increased INR Unknown Highly probable (10)
71‐year‐old man with aortic valve and mitral valve replacement 63 Sheng Mai‐yin (improvement of peripheral circulation) Warfarin Not mentioned Consciousness disturbance – right hemiplegia and active pupils, increased INR CYP2C9 induction, downregulates plasminogen inhibitor 1 (PAI‐1), tissue factor pathway inhibitor, coagulation factorXIII, A1, and, coagulation factor II (thrombin) receptor (F2R). Probable (8)
71‐year‐old Caucasian man with a history of atrial flutter, hypertension, hyperlipidemia, diabetes mellitus, erectile dysfunction and hypothyroidism 64 Bee pollen granules (general wellbeing) Warfarin Hydrochlorothiazide, lisinopril, levothyroxine, simvastatin, glyburide, metformin, vardenafil, aspirin, multivitamin and amlodipine Elevated INR CYP2C9 inhibition Probable (5)
58‐year‐old Mexican man with a history of type 2 diabetes mellitus, osteoarthritis, hyperlipidaemia, hypertension, and degenerative disc disease of the spine 65 Prickly pear cactus (diabetes) Glipizide Metformin, rosuvastatin, fenofibrate, aspirin, lisinopril, gabapentin, tramado1, nabumetone
and nitroglycerin
Hypoglycaemic Unknown Probable (8)
26‐year‐old man with chronic myeloid leukaemia 34 Panax ginseng (improve mental and physical performance) Imatinib Not mentioned Right upper quadrant pain, elevated liver enzymes CYP3A4 and P‐glycoprotein inhibition Probable (5)
79‐year‐old man with atrial fibrillation and metastatic bladder carcinoma 66 Grifron‐Pro Maitake D –fraction (immunoinstimulant) Warfarin diltiazem, hydromorphone, tamsulosin, prednisolone ophthalmic suspension, simvastatin and eszopiclone Elevated INR Unknown Possible (4)
59‐year‐old black man with hyperlipidaemia 67 SJW (insomnia) Rosuvastatin Not mentioned Increased total and low‐density lipoprotein cholesterol CYP2C9 and CYP2C19 induction via PXR activation Possible (3)
53‐year‐old Sri Lankan woman with unipolar depression 68 Arthiritis QR, Cholesterol QR, Triphala churna, Yogaraja Guggulu, Mentat, Rumalaya, Decoction‐1, Decoction‐2 (backache) Sertraline Not mentioned Moderate and severe depression Unknown Probable (6)
40‐year‐old man with generalized anxiety disorder and dream disorders 69 Valeriana officinalis L. and Passiflora incarnata L. (anxiety and insomnia) Lorazepam Not mentioned Handshaking, dizziness, throbbing and muscular fatigue Synergistic effect only Possible (3)
52‐year‐old woman with essential hypertension and a minor ischemic stroke 70 Nattokinase (stroke) Aspirin Not mentioned Vertigo and unsteady gait, high blood pressure, cerebral microbleed Unknown Doubtful (0)
47‐year‐old man with HIV‐1 infection 71 Efamol tablets, rheum frangula tablets and colayur syrup (laxative intestinal cleaners) Lopinavir, ritonavir Stavudine, lamivudine and tenofovir Diarrhoea, toxic lopinavir plasma level CYP3A4 and CYP2D6 inhibition Probable (8)
36‐year old woman with stage IV adenocarcinoma of lung 72 Ginseng, Fomes fomentarius, Inonotus obliquus, Phellinus linteus (improve mental and physical performance) Gefitinib Not mentioned Increased shortness of breath CYP3A4/5 induction Probable (5)
61‐year‐old man with a history of primary hypercholesterolemia 73 Green tea (fat and weight loss) Simvastatin Amlodipine Elevated liver enzymes, increase simvastatin lactone levels Unknown Probable (7)
80 year‐old Chinese woman with a history of diabetes mellitus, hypertension, cerebrovascular accident and atrial fibrillation 74 Lycium barbarum L or goji berry (promote longevity) Warfarin Nifedipine, glibenclamide, metformin, lorazepam Increased INR, Probably CYP2C9 inhibition and/or additive anticoagulation Highly probable (9)
77‐year‐old Japanese man with a history of hypertension and hyperuricemia 75 Arejin and Daiokanzo‐to (chronic allergic rhinitis and constipation) Enalapril Nifedipine, famotidine, brotizolam and terazosin Mild anaemia, liver dysfunction, mildly elevated creatine kinase (CK) level, and severehypokalaemia and hypochloraemia. Probably via inhibition of renal 11‐beta‐hydroxysteroid dehydrogenase Probable (7)
70‐year‐old woman with history of a mechanical mitral valve placement and an episode of atrial fibrillation 76 Matricaria chamomilla (pedal oedema) Warfarin Amiodarone, digoxin, synthroid, lendronate, metoprolol and a calcium–vitamin D supplement Elevated INR, dyspnoeic on exertion, bilateralpedal oedema and ecchymoses in her perineal area, across her lower abdomen and over her left hip Probably synergistic anticoagulation Possible (3)
55‐year‐old Indian woman with a node‐positive 4‐cm grade 3 invasive ductal carcinoma 77 Betel quid (CNS stimulant) Doxorubicin, cyclophosphamide, paclitaxel Docetaxel, 5‐fluorouracil and methotrexate Grade IV mucositis, dysuria, mouth pain, and furunculosis. Sensitization of normal tissues to the cytotoxic chemotherapy Probable (5)
57‐year‐old man with prosthetic mitral valve due to rheumatic heart disease 78 Commiphora molmol (acute bronchitis) Warfarin Not mentioned Decreased INR Unknown Possible (3)
48‐year‐old woman with a cadaveric renal allograft 79 Thüringen 9‐ Kräutertee (not indicated) Cyclosporine Mycophenolate mofetil, pravastatin, valsartan and hydrochlorothiazide Decreased cyclosporine level, rhabdomyolysis Unknown Possible (4)
37‐year‐old Armenian man with a cadaveric renal transplant 79 Chamomile Tea (dyspepsia, nausea, vomiting) Cyclosporine Azathioprineand Mycophenolate mofetil Decreased cyclosporine level Unknown Probable (7)
33‐year‐old man with a cadaveric renal transplant 79 Wild fruit tea drink (not indicated) Cyclosporine Mycophenolate mofetil Increased cyclosporine level Unknown Possible (4)
55‐year‐old man with a history of cerebrovascular accident following coronary artery bypass and seizure disorder 80 Ginkgo supplement (improve cognitive function) Valproic acid and phenytoin Not mentioned Seizure disorder leading to death while swimming Indirect inhibition of glutamate decarboxylase and glycine activities and CYP2C19 induction Probable (7)
35‐year‐old woman with a left thigh haemangioma 81 Aloe vera (leg pain) Sevoflurane Propofol, fentanyl, and rocuronium, Cefazolin, morphine Perioperative bleeding Probably additive inhibition of cyclooxygenase activity Possible (3)
57‐year‐old kidney transplant 82 SJW (depression) Cyclosporine Prednisolone Decrease cyclosporine concentration CYP3A4 and P‐gp induction Highly probable (9)
61‐year‐old man with atrial fibrillation and chronic rheumatic heart disease 83 Quilinggao (to quench internal heat) Warfarin Digoxin, simvastatin, furosemide and potassium chloride gum bleeding and epistaxis, elevated INR Inhibition of platelet function Probable (5)
28‐year‐old woman a recipient of a live‐donor allograft due to end‐stage renal disease attributed to hypertensive nephrosclerosis 84 Rice fermented with red yeast, beta‐sitosterol, danshen root (Salvia mitorriza), and garlic bulb (Allium sativum)(to lower cholesterol) Cyclosporine and/or diltiazem azathioprine, prednisone, enalapril and famotidine Increased serum creatine phosphokinase rhabdomyolysis Unknown Possible (3)
61‐year‐old Chinese woman with a history of recurring atrial fibrillation, hypertension, hypercholesterolaemia and tricuspid regurgitation 85 L. barbarum L. (blurred vision) Warfarin Benazepril, atenolol, digoxin and fluvastatin Elevated INR Probably CYP2C9 inhibition and/or additive anticoagulation Probable (6)
67‐year‐old Caucasian woman with a history of hypertension 86 Boldo‐fenugreek (to help liver and stimulate digestion) Warfarin Metoprolol Increased bleeding time Inhibition of thromboxane A2 Probable (5)

CYP, Cytochrome P450; GI, gastrointestinal; HIV, human immunodeficiency virus; INR, international normalized ratio; IV, intravenous; PXR, pregnane X receptor; SJW, St John's wort; UGT, uridine‐5′‐diphospho‐glucuronosyltransferase

The majority of patients in the two observational studies were aged between 51–61 years. Female patients (532 patients, 69.0%) constituted the majority. Most of the patients had a history of metabolic, cardiovascular, endocrine or haemato‐oncological complications (Table 1). In the study by Jeong et al., six cases of ADRs were reported 31. Four ADRs were reported in patients with a history of cerebral infarction taking herbal concoctions with NSAIDs (aspirin and diclofenac), P2Y12 inhibitor (clopidogrel) and analgesic (paracetamol) experiencing probable HDI (Table 3). Levy et al., reported nine cases of ADRs as a consequences of herbal medicine intake 32. The majority of the cases in this study were due to interaction between warfarin and sage or flaxseed (Table 3).

Table 3.

Case reports of herb‐drug interactions in observational studies

Authors [Ref] Patient Herbal medicine/product Reported ADRs Mode of interaction Prescribed drugs (Score)
Jeong et al., 31, a Case 1: 42‐year‐old man diagnosed with cerebral infarction Product A Cholestatic injury Unknown Cefuroxime (6) Paracetamol (6) diclofenac (6)
Case 2: 54‐year‐old man with cerebral infarction. Product B Hepatocellular injury Unknown Sarpogrelate (0) Actobacillus (0)
Case 3: 40‐year‐old man with Bell's palsy Product C Hepatocellular injury Unknown Prednisolone (3)
Case 4: 61‐year ‐old man with subdural hematoma Product D Hepatocellular injury Unknown Paracetamol (7) diclofenac (7) aspirin (7) clopidogrel (7) rebamipide (5)
Case 5: 57‐year‐old woman with cerebral infarction Product E Hepatocellular injury Unknown Roxoprofen (6) baclofen (6) cimetidine (6)
Case 6: 71‐year‐old woman with cerebral infarction Product Hepatocellular injury Unknown Amoxicillin (4) serratiopeptidase (4)
Levy et al., 32, b Case 1 Green tea Lowered digoxin level Unknown Digoxin (3)
Case 2 Turmeric GIT bleeding Additive antiplatelet effect Clopidogrel (5)
Case 3 Sage CO2 narcosis
Respiratory failure
Unknown Methadone (3)
Case 4 Sage/peppermint oil Rhabdomyolysis CYP3A4 inhibition Simvastatin (3)
Case 5 Flaxseed Anaemia
Rectal bleeding
Synergistic effect Aspirin (5)
Case 6 Blond Psyllium Orthostatic hypotension Unknown 3 antihypertensive drugs (3)
Case 7 Flaxseed Melena
INR 4.18
Additive anticoagulation Warfarin (6)
Case 8 Chamomile Melena
INR 4.18
Probably CYP2C9 inhibition or additive anticoagulation Warfarin (3)
Case 9 Sage Melena
INR 4.18
Probably CYP2C9 inhibition or additive anticoagulation Warfarin (6)
a

ADRs classified by RUCAM score for liver injury;

b

No provision of patient history and ADRs classified by DIPS score for other injuries.

ADR = adverse drug reaction; GIT = gastrointestinal tract; INR = international normalized ratio; Product A = Pinellia ternata Breitenbach, Atractylodes japonica koidzumi, Citrus unshiu Markovich, Gastrodia elata Blume, Poria cocos Wolf, Glycyrrhiza glabra Linne, Zingiber officinale Rosco; Product B = Pueraria lobata Ohwil, Scutellaria baicalensis Georgi, Angelica tenuissima Nakai, Raphanus sativus Linne, Platycodon grandiflorum A. De candole, Angelica dahurica Bentham et Hooker.; Product C = Astragalus membranaceus Bunge, Rehmannia glutinosa Liboschitz var. purpurea Makino, Angelica gigas Nakai, Paeonia lactiflora Pallas, Poria cocos Wolf, Atractylodes japonica koidzumi, Panax ginseng C. A. Meyer, Acorus gramineus Solander, Ostericum Koreanum Maxim, Pinellia ternata Breitenbach, Gastrodia elata Blume, Aconitum koreanum Raymond, Glycyrrhiza glabra Linne.; Product D = Scutellaria baicalensis Georgi, Atractylodes lancea D. C, Atracylodes chinensis Koidzumi, Ostericum Koreanum Maxim, Aralia continentalis Kitagawa, Saposhnikovia divaricata Schiskin, Cnidium officinale Makino, Angelica dahurica Bentham et Hooker, Liriope platyphylla Wang et Tang, Vitex rotundifolia Linne fil. Chrysanthemum indicum Linne, Asiasarum sieboldi F. Maekawa, Glycyrrhiza glabra Linne.; Product E = Agastache rugosaO.Kuntze, Perilla frutescens var. acuta Kudo, Angelica dahurica Bentham et Hooker, Areca catechu Linne, Polyporium bellati Polyporaceae, Magnolia ovobata Thunberg, Atractylodes japonica koidzumi, trus unshiu Markovich, Pinellia ternate Breitenbach, Platycodon grandiflorum A. Decandole, Arisaema amurense, Maximowicz Saussurea lappaClarke, Glycyrrhiz aglabra Linne.; Product F = Lindera strichnifolia Villars, ractylodes lancea D.C, Atracylodes chinensis Koidzumi, Ephedra sinica Stapf, Angelica dahurica Bentham et Hooker, Platycodon grandiflorum A. Decandole, Citrus aurantium Linne, Cinnamomum cassia Blume, Glycyrrhiza glabra Linne

Discussion

Concomitant intake of herbal medicines and prescribed medications is a common practice, especially in patients with hypertension, diabetes, cancer, seizures and depression. This is problematic particularly for drugs exhibiting a narrow therapeutic index. Incidences of underreporting and nonstandardized causality estimation of HDI in patients have resulted in life‐threatening ADRs, hospitalization and fatality in some cases 87, 88, 89. Intensification of monitoring and critical appraisal procedures to identify the severity of ADRs linked to concomitant consumption of herbs and prescribed medicines is critical in averting untoward occurrences.

The WHO Collaborating Centre for International Drug Monitoring‐Uppsala Monitoring Centre (WHO‐UMC) has the mandate to co‐ordinate global ADRs data and search this data to identify signals of new ADRs to notify the pharmacovigilance centres of member countries and other organizations concerned with drug safety. In a study conducted by WHO‐UMC the majority of HDI‐linked ADRs reported were from developed countries, including USA and Europe with South Africa as the only contributory developing country 90 and this re‐affirms the concern of underreporting of HDIs in developing countries. Adverse reactions related to HDIs have been documented in observational studies and case reports 70, 71, 83, 84, 91. Nonetheless, few review studies have been conducted to estimate the severity of the ADRs due to HDI in patients. Hence, this review adapted the DIPS and RUCAM scores to estimate the severity of ADRs, whilst the Stockley's herbal medicines interaction companion was consulted in assessing the mechanism of interactions.

The majority of cases recorded in this study showed that patients taking warfarin and/or statins (atorvastatin, simvastatin and rosuvastatin) for the management of cardiovascular complications reported clinically significant interactions after combination with herbal products including sage, flaxseed, SJW, cranberry, goji juice, green tea and chamomile. Warfarin is a racemic mixture of R and S stereoisomers with S‐warfarin being a 3–5 times more potent inhibitor of the vitamin K epoxide reductase complex than the R isomer. Warfarin is used for the management of atrial fibrillation and heart valve replacement. Metabolism of S‐warfarin is predominantly mediated by CYP2C9. Herbal medicines altering the activity of CYP2C9 may cause under‐anticoagulation or bleeding episodes. Potential interaction of warfarin and active constituents of sage, flaxseed, goji juice, cranberry and chamomile led to ADRs such as ecchymosis, epistaxis, haematuria, hemiplegia and elevated INR 33, 63, 74, 76, 85. The active constituents of SJW namely hyperforin, flavonols, flavonol glycosides, biflavones, napthodianthrones, acylphloroglucinols and phenylpropranes are known to reduce plasma concentration of warfarin via induction of CYP3A4 and CYP2C9. However, one case study reported SJW to cause sudden upper GIT bleeding in a sensitive patient 61. This interaction might have been caused by active constituents of SJW potentiating the clotting effect of warfarin. In addition, the Chinese herbal product Sheg Mai‐yin is used to improve peripheral circulation due to its effect on oxidative damage in heart, brain and other tissues. Sheg Mai‐yin contains red ginseng, liriope and Schisandra chinesis. A case of intracranial haematoma has been reported in a patient with a history of aortic valve and mitral valve replacement receiving warfarin and Sheg Mai‐yin 63. Red ginseng enhances the production of interleukin‐1β, which increases the production of tissue plasminogen activators responsible for suppression of thrombin formation during blood coagulation and fibrinolysis processes 92. Furthermore, the saponin‐related active constituents of red ginseng, namely ginsenosides Rg1 and Rg5, downregulate plasminogen inhibitor 1, tissue factor pathway inhibitor, coagulation factors XIII, A1 and coagulation factor II (thrombin) receptor (FR2), which may lead to increase INR 93.

Besides anticoagulants, patients with cardiovascular complications (coronary artery disease) often take statins, including atorvastatin, rosuvastatin, simvastatin and others to lower low‐density lipoprotein and total cholesterol levels. However, many patients on statins complain of muscle pain, which affects quality of life and adherence to treatment. Statin‐induced muscle intolerance could be elicited by its co‐usage with herbal supplements and other prescribed medications. For example, a patient receiving amlodipine (10 mg day–1) and simvastatin (10 mg day–1) complained of intense leg muscle cramps and pain after ingestion of green tea 73. This was attributed to a 2‐fold increase plasma levels of simvastatin lactone due to inhibitory effect of green tea on CYP3A4 94, 95. However, further studies need to be conducted to ascertain other potential mechanisms for statin intolerance due to green tea intake.

In organ transplant patients, tacrolimus and cyclosporine A are principal immunosuppressive agents commonly administered to protect and reduce episodes of organ rejection. Both drugs have a narrow therapeutic index and are metabolized primarily by CYP3A4/5. Herbal medicines that alter the activities of CYP3A4 are likely to affect the protective effect of tacrolimus and cyclosporine in transplant patients. Cases of treatment failure or toxicity have been reported in patients consuming tacrolimus, azathioprine and cyclosporine with herbal preparations such as turmeric or chamomile tea 43, 50, 79, 82. The active phytoconstituents of the herbal medicines inhibit CYP3A4 and P‐glycoprotein to reduce elimination of the immunosuppressive agents 96, 97, 98, 99, 100. Tacrolimus and cyclosporine bind to immunophilins to block calcineurin's mediated T‐lymphocyte activation. In general, increased plasma levels of calcineurins have been reported to trigger calcineurin‐induced vasoconstriction and release of endothelin‐1 (a potent vasoconstrictor), decrease production of nitric oxide and increase expression of transforming growth factor β1 in renal transplantation patients 101. Thus, the active ingredients of turmeric and chamomile induced nephrotoxicity in patients taking tacrolimus and/or cyclosporine via CYP3A4 inhibition. For probe drugs such as azathioprine, metabolic transformation of parent compound to the active moiety 6‐mercaptopurine by glutathione S transferase (GST) and subsequent inactivation to 6‐methylmercaptopurine by thiopurine methyltransferase (TPMT) is necessary 102. Herbal medicines capable of altering the activity of GST and/or TPMT could potentially induce clinically significant ADRs.

Other immunocompromised patients, including those with HIV infections, often take herbs for various reasons, and as immune boosters. A number of antiretroviral medications including protease inhibitors (lopinavir and ritonavir) and non‐nucleoside reverse transcriptase inhibitors (efavirenz and nevirapine) with narrow therapeutic indices and elimination via hepatic metabolism are susceptible to HDI. HIV patients experienced signs of toxicity and detectable levels of viral load while taking raltegravir plus lopinavir/ritonavir 58 or efavirenz 59 with ginseng and G. biloba, respectively. The mechanism of ginseng–raltegravir interaction is unknown, as several conflicting outcomes have been reported on the influence of various active ingredients of ginseng on CYP450 enzymes 103, 104, 105. However, one study reported ginseng‐mediated hepatotoxicity in a chronic myeloid leukaemia patient on imatinib due to inhibition of CYP3A4 34. The inhibitory effect of ginseng on CYP3A4 was thus probably accountable for the observed adverse effect of raltegravir. For G. biloba, a unique active ingredient, bilobalide, has been demonstrated to induce CYP2B6 mRNA in animal models, although such a result was irreproducible in human subjects 106, 107. In a patient taking efavirenz and ginkgo, it is speculated that a breakthrough in viral load occurred because of a decrease in plasma levels of EFV as consequence of the inductive effect of active ingredients in G. biloba.

The current review study also recorded a significant number of HDI cases in patients with depression and seizure disorders. Antidepression medications, including valafaxine and sertraline, are eliminated primarily by hepatic metabolism. Patients taking these medications with herbal drugs including QR and Mentat for arthritis and celery root for menopause developed episodes of worsening depression 45, 68. In addition, cases of seizure induction occurred in a patient taking either phenytoin or valproic acid after consuming G. biloba supplement. In one recorded case, seizure induction led to the demise of a patient while swimming. The autopsy results confirmed a decreased plasma levels of both phenytoin and valproic due to inductive effect of G. biloba on CYP2C19 80. Another study in 18 healthy Chinese subjects showed significant reduction in plasma concentration of omeprazole as a probe substrate of CYP2C19 in the presence of G. biloba compared to the controls 108. Furthermore, Ginkgo contains a potent neurotoxin, 4′‐0‐methoxypyridoxine, which indirectly inhibits glutamate decarboxylase and glycine activities leading to seizure induction 109. The effect of 4′‐0‐methoxypyridoxine on inhibitory neurotransmitters is predicted as the most likely cause of seizure induction leading to his demise since CYP2C19 would have accounted only for a minor proportion of phenytoin and valproic acid elimination.

Finally, the consumption of herbal medicines is a well‐known practice among cancer patients 110, 111, 112, 113. Currently, imatinib is the main drug for the treatment of chronic myeloid leukaemia. It is a first‐ generation inhibitor of bcr‐abl tyrosine kinase enzyme. Imatinib is primarily metabolized by CYP3A4, and hence, any herbal medicine that alters the activity of CYP3A4, may affect the activity of imatinib. Concomitant intake of imatinib and an energy drink containing P. ginseng led to aberrant liver enzyme levels in a patient 34. Previous study showed that ginseng increases plasma concentrations of prescribed medications via CYP3A4 inhibition 105. Thus, the reduced activity of CYP3A4 in the presence of P. ginseng precipitated imatinib‐induced hepatotoxicity characterized by late‐stage acute lobular hepatitis 105. Other chemotherapeuticals such as cisplatin, etoposide and trabectedin have been reported to interact with herbal medicines, including Echinacea purpurea 60 and chokeberry juice (Aronia melanocarpa) 53 in patients presenting with different types of cancer. Chokeberry contains concentrated flavonoids – procyanidin B5, cyaniding‐3‐arabinoside and quercetin – which strongly inhibit CYP3A4 activity in the liver 114. Thus, reduced activity of CYP3A4 due to chokeberry intake caused trabectedin‐induced rhabdomyolysis in this patient 53. Conversely, echinacea has a mild inhibitory effect on CYP3A4 activity, which is likely to potentiate the myelosuppressive effect of etoposide and could elicit neutropenia and thrombocytopenia in patients.

Conclusion

Patients taking herbal medicines containing G. biloba, P. ginseng, SJW, green tea and others affecting the pharmacokinetic and pharmacodynamic properties of prescribed medications are at risk of experiencing different degrees of HDI. Few case reports of potential HDI have been documented in the literature despite the detrimental consequences of such interactions. In addition, even in cases where HDI were reported in patients, inadequate provision of information hindered the utilization of the data to draw clinically meaningful conclusions. Generally, these challenges could be attributed to a number of reasons including: (i) inability to re‐challenge patients with the herbs involved to confirm the causal relationship of the interaction for ethical reasons; (ii) lack of analytical capacity to measure the plasma levels of the affected drug; (iii) difficulty in identification of the phytochemical responsible for the interaction; (iv) nonexistence of adequate genetic information especially for high‐risk drugs, such as clopidogrel, warfarin, codeine, tamoxifen or terbinafine; (v) lack of standardized HDI‐specific causality assessment tool; and (vi) lack of motivation of clinicians to publish HDI case reports. Critical assessment of the causality of ADRs using the recommended scoring systems reported in this study will strengthen the applicability of HDI data in clinical practice. In addition, mechanistic investigations in healthy subjects and in vitro liver models as a follow‐up study on herbs recorded to elicit clinically significant HDI in patients. Thus, we recommend a bench‐to‐bed‐side approach to understand the causal relationship of HDI linked ADRs and the potential mechanism of observed interactions. This approach will inform drug regulatory agencies and pharmaceutical companies about the need to update information in package inserts of medicines to avoid untoward adverse events, based on available data. In conclusion, causality assessment and subsequent mechanistic studies of herbs with clinically relevant HDI must be publicized to alert both clinicians and patients about the need to avoid co‐usage of certain herbal medicines with specific prescribed medications.

Competing Interests

There are no competing interests to declare.

Awortwe, C. , Makiwane, M. , Reuter, H. , Muller, C. , Louw, J. , and Rosenkranz, B. (2018) Critical evaluation of causality assessment of herb–drug interactions in patients. Br J Clin Pharmacol, 84: 679–693. doi: 10.1111/bcp.13490.

References

  • 1. Ekor M. The growing use of herbal medicines: issues relating to adverse reactions and challenges in monitoring safety. Front Pharmacol 2014; 4: 177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. NCD Risk Factor Collaboration (NCD‐RisC) – Africa Working Group . Trends in obesity and diabetes across Africa from 1980 to 2014: an analysis of pooled population‐based studies. Int J Epidemiol 2017; Available from: http://www.ncbi.nlm.nih.gov/pubmed/28582528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. NCD Risk Factor Collaboration (NCD‐RisC) . Worldwide trends in diabetes since 1980: a pooled analysis of 751 population‐based studies with 4.4 million participants. Lancet 2016; 387: 1513–1530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. NCD Risk Factor Collaboration (NCD‐RisC) . Worldwide trends in blood pressure from 1975 to 2015: a pooled analysis of 1479 population‐based measurement studies with 19·1 million participants. Lancet 2017; 389: 37–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Benziger CP, Roth GA, Moran AE. The global burden of disease study and the preventable burden of NCD. Glob Heart 2016; 11: 393–397. [DOI] [PubMed] [Google Scholar]
  • 6. Duan W, Zheng A, Mu X, Li M, Liu C, Huang W, et al How great is the medical burden of disease on the aged? Research based on “System of Health Account 2011”. Health Qual Life Outcomes 2017; 15: 134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Weiss CO. Frailty and chronic diseases in older adults. Clin Geriatr Med 2011; 27: 39–52. [DOI] [PubMed] [Google Scholar]
  • 8. Saraga M, Zullino DF. St. John's Wort, corticosteroids, cocaine, alcohol ... and a first manic episode. Praxis (Bern 1994) 2005; 94: 987–989. [DOI] [PubMed] [Google Scholar]
  • 9. Moschella C, Jaber BL. Interaction between cyclosporine and Hypericum perforatum (St. John's wort) after organ transplantation. Am J Kidney Dis 2001; 38: 1105–1107. [DOI] [PubMed] [Google Scholar]
  • 10. Dannawi M. Possible serotonin syndrome after combination of buspirone and St John's Wort. J Psychopharmacol 2002; 16: 401. [DOI] [PubMed] [Google Scholar]
  • 11. Spinella M, Eaton LA. Hypomania induced by herbal and pharmaceutical psychotropic medicines following mild traumatic brain injury. Brain Inj 2002; 16: 359–367. [DOI] [PubMed] [Google Scholar]
  • 12. Awortwe C, Fasinu PS, Rosenkranz B. Application of Caco‐2 cell line in herb‐drug interaction studies: current approaches and challenges. J Pharm Pharm Sci 2014; 17: 1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Hong M, Li S, Tan HY, Cheung F, Wang N, Huang J, et al A network‐based pharmacology study of the herb‐induced liver injury potential of traditional hepatoprotective Chinese herbal medicines. Molecules 2017; 22: 28420096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Sun D, Zhang C‐Z, Ran R‐X, Cao Y‐F, Du Z, Fu Z‐W, et al In vitro comparative study of the inhibitory effects of mangiferin and its aglycone norathyriol towards UDP‐glucuronosyl transferase (UGT) isoforms. Molecules 2017; 22: 28621744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Jackson JP, Freeman KM, Friley WW, Herman AG, Black CB, Brouwer KR, et al Prediction of clinically relevant herb–drug clearance interactions using sandwich‐cultured human hepatocytes: Schisandra spp. Case Study Drug Metab Dispos 2017; 45: 1019–1026. [DOI] [PubMed] [Google Scholar]
  • 16. Dai G, Jiang Z, Bai Y, Zhang Q, Zhu L, Bai X, et al Pharmacokinetic herb–drug interaction of Xuesaitong dispersible tablet and aspirin after oral administration in blood stasis model rats. Phytomedicine 2017; 26: 62–68. [DOI] [PubMed] [Google Scholar]
  • 17. Ting CT, Cheng YY, Tsai TH. Herb–drug interaction between the traditional hepatoprotective formulation and sorafenib on hepatotoxicity, histopathology and pharmacokinetics in rats. Molecules 2017; 22: 28640225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Thomford NE, Awortwe C, Dzobo K, Adu F, Chopera D, Wonkam A, et al Inhibition of CYP2B6 by medicinal plant extracts: implication for use of efavirenz and nevirapine‐based highly active anti‐retroviral therapy (HAART) in resource‐limited settings. Molecules 2016; 21: 26891286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Patel O, Muller C, Joubert E, Louw J, Rosenkranz B, Awortwe C. Inhibitory interactions of Aspalathus linearis (rooibos) extracts and compounds, aspalathin and z‐2‐(β‐d‐glucopyranosyloxy)‐3‐phenylpropenoic acid, on cytochromes metabolizing hypoglycemic and hypolipidemic drugs. Molecules 2016; 21: 27845750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Awortwe C, Manda VK, Avonto C, Khan SI, Khan IA, Walker LA, et al Echinacea purpurea up‐regulates CYP1A2, CYP3A4 and MDR1 gene expression by activation of pregnane X receptor pathway. Xenobiotica 2015; 45: 218–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Awortwe C, Manda VK, Avonto C, Khan SI, Khan IA, Walker LA, et al In vitro evaluation of reversible and time‐dependent inhibitory effects of Kalanchoe crenata on CYP2C19 and CYP3A4 activities. Drug Metab Lett 2015; 9: 48–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Awortwe C, Bouic PJ, Masimirembwa CM, Rosenkranz B. Inhibition of major drug metabolizing CYPs by common herbal medicines used by HIV/AIDS patients in Africa‐ implications for herb‐drug interactions. Drug Metab Lett 2014; 7: 83–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Kim B‐H, Kim K‐P, Lim KS, Kim J‐R, Yoon SH, Cho J‐Y, et al Influence of Ginkgo biloba extract on the pharmacodynamic effects and pharmacokinetic properties of ticlopidine: an open‐label, randomized, two‐period, two‐treatment, two‐sequence, single‐dose crossover study in healthy Korean male volunteers. Clin Ther 2010; 32: 380–390. [DOI] [PubMed] [Google Scholar]
  • 24. Penzak SR, Robertson SM, Hunt JD, Chairez C, Malati CY, Alfaro RM, et al Echinacea purpurea significantly induces cytochrome P450 3A activity but does not alter lopinavir‐ritonavir exposure in healthy subjects. Pharmacotherapy 2010; 30: 797–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Goey AKL, Meijerman I, Rosing H, Burgers JA, Mergui‐Roelvink M, Keessen M, et al The effect of Echinacea purpurea on the pharmacokinetics of docetaxel. Br J Clin Pharmacol 2013; 76: 467–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Monera‐Penduka TG, Maponga CC, Wolfe AR, Wiesner L, Morse GD, Nhachi CFB. Effect of Moringa oleifera Lam. leaf powder on the pharmacokinetics of nevirapine in HIV‐infected adults: a one sequence cross‐over study. AIDS Res Ther 2017; 14: 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Gwaza L, Aweeka F, Greenblatt R, Lizak P, Huang L, Guglielmo BJ. Co‐administration of a commonly used Zimbabwean herbal treatment (African potato) does not alter the pharmacokinetics of lopinavir/ritonavir. Int J Infect Dis 2013; 17: e857–e861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Sprouse AA, van Breemen RB. Pharmacokinetic interactions between drugs and botanical dietary supplements. Drug Metab Dispos 2016; 44: 162–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Andrén L, Andreasson A, Eggertsen R. Interaction between a commercially available St. John's wort product (Movina) and atorvastatin in patients with hypercholesterolemia. Eur J Clin Pharmacol 2007; 63: 913–916. [DOI] [PubMed] [Google Scholar]
  • 30. Zadoyan G, Rokitta D, Klement S, Dienel A, Hoerr R, Gramatté T, et al Effect of Ginkgo biloba special extract EGb 761 on human cytochrome P450 activity: a cocktail interaction study in healthy volunteers. Eur J Clin Pharmacol 2012; 68: 553–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Jeong TY, Park BK, Cho JH, Kim YI, Ahn YC, Son CG. A prospective study on the safety of herbal medicines, used alone or with conventional medicines. J Ethnopharmacol 2012; 143: 884–888. [DOI] [PubMed] [Google Scholar]
  • 32. Levy I, Attias S, Ben‐Arye E, Goldstein L, Schiff E. Adverse events associated with interactions with dietary and herbal supplements among inpatients. Br J Clin Pharmacol 2017; 83: 836–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Rivera CA, Ferro CL, Bursua AJ, Gerber BS. Probable interaction between Lycium barbarum (goji) and warfarin. Pharmacotherapy 2012; 32: e50–e53. [DOI] [PubMed] [Google Scholar]
  • 34. Bilgi N, Bell K, Ananthakrishnan AN, Atallah E. Imatinib and Panax ginseng: a potential interaction resulting in liver toxicity. Ann Pharmacother 2010; 44: 926–928. [DOI] [PubMed] [Google Scholar]
  • 35. Seger D, Barker K, McNaughton C. Misuse of the Naranjo Adverse Drug Reaction Probability Scale in toxicology. Clin Toxicol (Phila) 2013; 51: 461–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Fugh‐Berman A, Ernst E. Herb–drug interactions: review and assessment of report reliability. Br J Clin Pharmacol 2001; 52: 587–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Horn JR, Hansten PD, Chan LN. Proposal for a new tool to evaluate drug interaction cases. Ann Pharmacother 2007; 41: 674–680. [DOI] [PubMed] [Google Scholar]
  • 38. Danan G, Teschke R. RUCAM in drug and herb induced liver injury: the update. Int J Mol Sci 2015; 17 (1): 1–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Benichou C, Danan G, Flahault A. Causality assessment of adverse reactions to drugs – II. An original model for validation of drug causality assessment methods: case reports with positive rechallenge. J Clin Epidemiol 1993; 46: 1331–1336. [DOI] [PubMed] [Google Scholar]
  • 40. Danan G, Benichou C. Causality assessment of adverse reactions to drugs – I. A novel method based on the conclusions of international consensus meetings: application to drug‐induced liver injuries. J Clin Epidemiol 1993; 46: 1323–1330. [DOI] [PubMed] [Google Scholar]
  • 41. Williamson E, Driver S, Baxter K. Stockly's herbal medicines interactions. London: Pharmaceutical press, 2009; 1–11. [Google Scholar]
  • 42. Ji H, Zhang G, Yue F, Zhou X. Adverse event due to a likely interaction between sodium aescinate and Ginkgo biloba extract: a case report. J Clin Pharm Ther 2017; 42: 237–238. [DOI] [PubMed] [Google Scholar]
  • 43. Nayeri A, Wu S, Adams E, Tanner C, Meshman J, Saini I, et al Acute calcineurin inhibitor nephrotoxicity secondary to turmeric intake: a case report. Transplant Proc 2017; 49: 198–200. [DOI] [PubMed] [Google Scholar]
  • 44. Damato A, Larocca M, Rondini E, Pinto C, Versari A, Menga M. Severe rhabdomylosis during treatment with trabectedin in combination with herbal drug in patient with metastatic synovial sarcoma: a case report. Case Rep Oncol 2017; 10: 258–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Khalid Z, Osuagwu FC, Shah B, Roy N, Dillon JE, Bradley R. Celery root extract as an inducer of mania induction in a patient on venlafaxine and St John's Wort. Postgrad Med 2016; 128: 682–683. [DOI] [PubMed] [Google Scholar]
  • 46. Kang YC, Chen MH, Lai SL. Potentially unsafe herb–drug interactions between a commercial product of noni juice and phenytoin – a case report. Acta Neurol Taiwan 2015; 24: 43–46. [PubMed] [Google Scholar]
  • 47. Myers AP, Watson TA, Strock SB. Drug reaction with eosinophilia and systemic symptoms syndrome probably induced by a lamotrigine–ginseng drug interaction. Pharmacotherapy 2015; 35: e9–12. [DOI] [PubMed] [Google Scholar]
  • 48. Ferreira PG, Costa S, Dias N, Ferreira AJ, Franco F. Simultaneous interstitial pneumonitis and cardiomyopathy induced by venlafaxine. J Bras Pneumol 2014; 40: 313–318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Melchardt T, Magnes T, Weiss L, Grundbichler M, Strasser M, Hufnagl C, et al Liver toxicity during temozolomide chemotherapy caused by Chinese herbs. BMC Complement Altern Med 2014; 14: 115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Carbajal R, Yisfalem A, Pradhan N, Baumstein D, Chaudhari A. Case report: Boldo (Peumus boldus) and tacrolimus interaction in a renal transplant patient. Transplant Proc 2014; 46: 2400–2402. [DOI] [PubMed] [Google Scholar]
  • 51. Carter J, Yeh RF, Braunschweig I, Barta SK. Unreported use of an herbal supplement resulting in decreased clearance of intravenous busulfan in a patient undergoing. Bone Marrow Transplant 2013; 49: 313–314. [DOI] [PubMed] [Google Scholar]
  • 52. Açikgöz SK, Açikgöz E. Gastrointestinal bleeding secondary to interaction of Artemisia absinthium with warfarin. Drug Metabol Drug Interact 2013; 28: 187–189. [DOI] [PubMed] [Google Scholar]
  • 53. Strippoli S, Lorusso V, Albano A, Guida M. Herbal‐drug interaction induced rhabdomyolysis in a liposarcoma patient receiving trabectedin. BMC Complement Altern Med 2013; 13: 199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Hou Q, Han W, Fu X. Pharmacokinetic interaction between tacrolimus and berberine in a child with idiopathic nephrotic syndrome. Eur J Clin Pharmacol 2013; 69: 1861–1862. [DOI] [PubMed] [Google Scholar]
  • 55. Orr A, Parker R. Red clover causing symptoms suggestive of methotrexate toxicity in a patient on high‐dose methotrexate. Menopause Int 2013; 19: 133–134. [DOI] [PubMed] [Google Scholar]
  • 56. Tamura S, Warabi Y, Matsubara S. Severe liver dysfunction possibly caused by the combination of interferon beta‐1b therapy and melilot (sweet clover) supplement. J Clin Pharm Ther 2012; 37: 724–725. [DOI] [PubMed] [Google Scholar]
  • 57. Van Strater ACP, Bogers JPAM. Interaction of St John's wort (Hypericum perforatum) with clozapine. Int Clin Psychopharmacol 2012; 27: 121–124. [DOI] [PubMed] [Google Scholar]
  • 58. Mateo‐Carrasco H, Gálvez‐Contreras MC, Fernández‐Ginés FD, Nguyen TV. Elevated liver enzymes resulting from an interaction between raltegravir and Panax ginseng: a case report and brief review. Drug Metabol Drug Interact 2012; 27: 171–175. [DOI] [PubMed] [Google Scholar]
  • 59. Naccarato M, Yoong D, Gough K. A potential drug‐herbal interaction between Ginkgo biloba and efavirenz. J Int Assoc Physicians AIDS Care 2012; 11: 98–100. [DOI] [PubMed] [Google Scholar]
  • 60. Bossaer JB, Odle BL. Probable etoposide interaction with Echinacea . J Diet Suppl 2012; 9: 90–95. [DOI] [PubMed] [Google Scholar]
  • 61. Uygur Bayramıçlı O, Kalkay MN, Oskay Bozkaya E, Doğan Köse E, Iyıgün O, Görük M, et al St. John's wort (Hypericum perforatum) and warfarin: dangerous liaisons! Turk J Gastroenterol 2011; 22: 115. [PubMed] [Google Scholar]
  • 62. Hamann GL, Campbell JD, George CM. Warfarin–cranberry juice interaction. Ann Pharmacother 2011; 45: e17. [DOI] [PubMed] [Google Scholar]
  • 63. Su Q, Li Y. Interaction between warfarin and the herbal product shengmai‐yin: a case report of intracerebral hematoma. Yonsei Med J 2010; 51: 793–796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Hurren KM, Lewis CL. Probable interaction between warfarin and bee pollen. Am J Health Syst Pharm 2010; 67: 2034–2037. [DOI] [PubMed] [Google Scholar]
  • 65. Sobieraj DM, Freyer CW. Probable hypoglycemic adverse drug reaction associated with prickly pear cactus, glipizide, and metformin in a patient with type 2 diabetes mellitus. Ann Pharmacother 2010; 44: 1334–1337. [DOI] [PubMed] [Google Scholar]
  • 66. Corrigan MA, Atkinson KM, Sha BE, Crank CW. Evaluation of pharmacy‐implemented medication reconciliation directed at antiretroviral therapy in hospitalized HIV/AIDS patients. Ann Pharmacother 2010; 44: 222–223. [DOI] [PubMed] [Google Scholar]
  • 67. Gordon RY, Becker DJ, Rader DJ. Reduced efficacy of rosuvastatin by St. John's wort. Am J Med 2009; 122: e1–e2. [DOI] [PubMed] [Google Scholar]
  • 68. Prasad K, Tharangani PGD, Samaranayake CN. Recurrent relapses of depression in a patient established on sertraline after taking herbal medicinal mixtures – a herb–drug interaction? J Psychopharmacol 2009; 23: 216–219. [DOI] [PubMed] [Google Scholar]
  • 69. Carrasco MC, Vallejo JR, Pardo‐de‐Santayana M, Peral D, Martín MA, Altimiras J. Interactions of Valeriana officinalis L. and Passiflora incarnata L. in a patient treated with lorazepam. Phytother Res 2009; 23: 1795–1796. [DOI] [PubMed] [Google Scholar]
  • 70. Chang Y‐Y, Liu J‐S, Lai S‐L, Wu H‐S, Lan M‐Y. Cerebellar hemorrhage provoked by combined use of nattokinase and aspirin in a patient with cerebral microbleeds. Intern Med 2008; 47: 467–469. [DOI] [PubMed] [Google Scholar]
  • 71. Beukel van den Bout‐van den CJP, Bosch MEW, Burger DM, Koopmans PP, van der Ven AJAM. Toxic lopinavir concentrations in an HIV‐1 infected patient taking herbal medications. AIDS 2008; 22: 1243–1244. [DOI] [PubMed] [Google Scholar]
  • 72. Hwang SW, Han HS, Lim KY, Han JY. Drug interaction between complementary herbal medicines and gefitinib. J Thorac Oncol 2008; 3: 942–943. [DOI] [PubMed] [Google Scholar]
  • 73. Werba JP, Giroli M, Cavalca V, Nava MC, Tremoli E, Dal BL. The effect of green tea on simvastatin tolerability. Ann Intern Med 2008; 149: 286–287. [DOI] [PubMed] [Google Scholar]
  • 74. Leung H, Hung A, Hui ACF, Chan TYK. Warfarin overdose due to the possible effects of Lycium barbarum L. Food Chem Toxicol 2008; 46: 1860–1862. [DOI] [PubMed] [Google Scholar]
  • 75. Iida R, Otsuka Y, Matsumoto K, Kuriyama S, Hosoya T. Pseudoaldosteronism due to the concurrent use of two herbal medicines containing glycyrrhizin: Interaction of glycyrrhizin with angiotensin‐converting enzyme inhibitor. Clin Exp Nephrol 2006; 10: 131–135. [DOI] [PubMed] [Google Scholar]
  • 76. Segal R, Pilote L. Warfarin interaction with Matricaria chamomilla . CMAJ 2006; 174: 1281–1282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Epstein RJ, Leung TWT, Cheung PSY. Panmucositis and chemosensitisation associated with betel quid chewing during dose‐dense adjuvant breast cancer chemotherapy. Cancer Chemother Pharmacol 2006; 58: 835–837. [DOI] [PubMed] [Google Scholar]
  • 78. Al Faraj S. Antagonism of the anticoagulant effect of warfarin caused by the use of Commiphora molmol as a herbal medication: a case report. Ann Trop Med Parasitol 2017; 4983: 10–12. [DOI] [PubMed] [Google Scholar]
  • 79. Nowack R, Nowak B. Case report. Herbal teas interfere with cyclosporin levels in renal transplant patients. Nephrol Dial Transplant 2005; 20: 2554–2556. [DOI] [PubMed] [Google Scholar]
  • 80. Kupiec T, Raj V. Fatal seizures due to potential herb‐drug interactions with Ginkgo biloba . J Anal Toxicol 2005; 29: 755–758. [DOI] [PubMed] [Google Scholar]
  • 81. Lee A, Chui PT, Aun CST, Gin T, Lau ASC. Possible interaction between sevoflurane and Aloe vera . Ann Pharmacother 2004; 38: 1651–1654. [DOI] [PubMed] [Google Scholar]
  • 82. Alscher DM, Klotz U. Drug interaction of herbal tea containing St. John's wort with cyclosporine. Transpl Int 2003; 16: 543–544. [DOI] [PubMed] [Google Scholar]
  • 83. Wong ALN, Chan TYK. Interaction between warfarin and the herbal product Quilinggao . Ann Pharmacother 2003; 37: 836–838. [DOI] [PubMed] [Google Scholar]
  • 84. Rasad GVRAP, Ong TIW, Eliton GALOM. Rhabdomyolysis due to red yeast rice (Monascus purpureus) in a renal transplant recipient. Transplantation 2001; 74: 1200–1201. [DOI] [PubMed] [Google Scholar]
  • 85. Lam AY, Elmer GW, Mohutsky MA. Possible interaction between warfarin and Lycium barbarum L. Ann Pharmacother 2001; 35: 1199–1201. [DOI] [PubMed] [Google Scholar]
  • 86. Lambert JP, Cormier J. Potential interaction between warfarin and boldo‐fenugreek. Pharmacotherapy 2001; 21: 509–512. [DOI] [PubMed] [Google Scholar]
  • 87. Brown AC. Kidney toxicity related to herbs and dietary supplements: online table of case reports. Part 3 of 5 series. Food Chem Toxicol 2017; 107: 502–519. [DOI] [PubMed] [Google Scholar]
  • 88. Alsanad SM, Howard RL, Williamson EM. An assessment of the impact of herb–drug combinations used by cancer patients. BMC Complement Altern Med 2016; 16: 393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Walji R, Boon H, Barnes J, Austin Z, Baker GR, Welsh S. Adverse event reporting for herbal medicines: a result of market forces. Healthc Policy 2009; 4: 77–90. [PMC free article] [PubMed] [Google Scholar]
  • 90. Skalli S, Bencheikh RS. Safety monitoring of herb–drug interactions. Drug Saf 2012; 35: 785–791. [DOI] [PubMed] [Google Scholar]
  • 91. Lee NJ, Pharm D, Fermo JD, Pharm D. Warfarin and royal jelly interaction. Pharmacotherapy 2006; 26: 583–586. [DOI] [PubMed] [Google Scholar]
  • 92. Nakajima S, Uchiyama Y, Yoshida K, Mizukawa H, Haruki E. The effects of ginseng radix rubra on human vascular endothelial cells. Am J Chin Med 1998; 26: 365–373. [DOI] [PubMed] [Google Scholar]
  • 93. Lü J, Ma Z, Yang J, Huang J, Wang S, Wang S. Ginsenoside Rg1‐induced alterations in gene expression in TNF‐alpha stimulated endothelial cells. Chin Med J (Engl) 2004; 117: 871–876. [PubMed] [Google Scholar]
  • 94. Donovan JL, Chavin KD, Devane CL, Taylor RM, Wang JS, Ruan Y, et al Green tea (Camellia sinensis) extract does not alter cytochrome P450 3A4 or 2D6 activity in healthy volunteers. Drug Metab Dispos 2004; 32: 906–908. [DOI] [PubMed] [Google Scholar]
  • 95. Chow H‐HS, Hakim IA, Vining DR, Crowell JA, Cordova CA, Chew WM, et al Effects of repeated green tea catechin administration on human cytochrome P450 activity. Cancer Epidemiol Biomarkers Prev 2006; 15: 2473–2476. [DOI] [PubMed] [Google Scholar]
  • 96. Egashira K, Sasaki H, Higuchi S, Ieiri I. Food‐drug interaction of tacrolimus with pomelo, ginger, and turmeric juice in rats. Drug Metab Pharmacokinet 2012; 27: 242–247. [DOI] [PubMed] [Google Scholar]
  • 97. Zhang W, Tan TMC, Lim LY. Impact of curcumin‐induced changes in P‐glycoprotein and CYP3A expression on the pharmacokinetics of peroral celiprolol and midazolam in rats. Drug Metab Dispos 2007; 35: 110–115. [DOI] [PubMed] [Google Scholar]
  • 98. Zhang W, Lim LY. Effects of spice constituents on P‐glycoprotein‐mediated transport and CYP3A4‐mediated metabolism in vitro . Drug Metab Dispos 2008; 36: 1283–1290. [DOI] [PubMed] [Google Scholar]
  • 99. Ganzera M, Schneider P, Stuppner H. Inhibitory effects of the essential oil of chamomile (Matricaria recutita L.) and its major constituents on human cytochrome P450 enzymes. Life Sci 2006; 78: 856–861. [DOI] [PubMed] [Google Scholar]
  • 100. Budzinski JW, Foster BC, Vandenhoek S, Arnason JT. An in vitro evaluation of human cytochrome P450 3A4 inhibition by selected commercial herbal extracts and tinctures. Phytomedicine 2000; 7: 273–282. [DOI] [PubMed] [Google Scholar]
  • 101. Burdmann EA, Andoh TF, Yu L, Bennett WM. Cyclosporine nephrotoxicity. Semin Nephrol 2003; 23: 465–476. [DOI] [PubMed] [Google Scholar]
  • 102. Cuffari C. A physician's guide to azathioprine metabolite testing. Gastroenterol Hepatol (NY) 2006; 2: 58–63. [PMC free article] [PubMed] [Google Scholar]
  • 103. Ujhelyi MR, Bottorff MB, Schur M, Roll K, Zhang H, Stewart J, et al Aging effects on the organic base transporter and stereoselective renal clearance. Clin Pharmacol Ther 1997; 62: 117–128. [DOI] [PubMed] [Google Scholar]
  • 104. Donovan JL, DeVane CL, Chavin KD, Taylor RM, Markowitz JS. Siberian ginseng (Eleutheroccus senticosus) effects on CYP2D6 and CYP3A4 activity in normal volunteers. Drug Metab Dispos 2003; 31: 519–522. [DOI] [PubMed] [Google Scholar]
  • 105. Gurley BJ, Gardner SF, Hubbard MA, Williams DK, Gentry WB, Cui Y, et al Clinical assessment of effects of botanical supplementation on cytochrome P450 phenotypes in the elderly: St John's wort, garlic oil, Panax ginseng and Ginkgo biloba . Drugs Aging 2005; 22: 525–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Taki Y, Yamazaki Y, Shimura F, Yamada S, Umegaki K. Time‐dependent induction of hepatic cytochrome P450 enzyme activity and mRNA expression by bilobalide in rats. J Pharmacol Sci 2009; 109: 459–462. [DOI] [PubMed] [Google Scholar]
  • 107. Deng Y, Bi H‐C, Zhao L‐Z, He F, Liu Y‐Q, Yu J‐J, et al Induction of cytochrome P450s by terpene trilactones and flavonoids of the Ginkgo biloba extract EGb 761 in rats. Xenobiotica 2008; 38: 465–481. [DOI] [PubMed] [Google Scholar]
  • 108. Yin OQP, Tomlinson B, Waye MMY, Chow AHL, Chow MSS. Pharmacogenetics and herb‐drug interactions: experience with Ginkgo biloba and omeprazole. Pharmacogenetics 2004; 14: 841–850. [DOI] [PubMed] [Google Scholar]
  • 109. Shannon M, McElroy EA, Liebelt EL. Toxic seizures in children: case scenarios and treatment strategies. Pediatr Emerg Care 2003; 19: 206–210. [DOI] [PubMed] [Google Scholar]
  • 110. Kuo YT, Chang TT, Muo CH, Wu MY, Sun MF, Yeh CC, et al Use of complementary traditional Chinese medicines by adult cancer patients in Taiwan: a nationwide population‐based study. Integr Cancer Ther 2017; 1; 1534735417716302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Huet M. Medicinal plants in cancer patients: current practices and evaluation data. Bull Cancer 2013; 100: 485–495. [DOI] [PubMed] [Google Scholar]
  • 112. Poonthananiwatkul B, Howard RL, Williamson EM, Lim RHM. Cancer patients taking herbal medicines: a review of clinical purposes, associated factors, and perceptions of benefit or harm. J Ethnopharmacol 2015; 175: 58–66. [DOI] [PubMed] [Google Scholar]
  • 113. Akpunar D, Bebis H, Yavan T. Use of complementary and alternative medicine in patients with gynecologic cancer: a systematic review. Asian Pac J Cancer Prev 2015; 16: 7847–7852. [DOI] [PubMed] [Google Scholar]
  • 114. Bräunlich M, Christensen H, Johannesen S, Slimestad R, Wangensteen H, Malterud KE, et al In vitro inhibition of cytochrome P450 3A4 by Aronia melanocarpa constituents. Planta Med 2013; 79: 137–141. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Clinical Pharmacology are provided here courtesy of British Pharmacological Society

RESOURCES