Open access peer-reviewed chapter

Safety Review of Herbs and Supplements in Heart Disease, Diabetes, and COVID-19

Written By

Paula Vieira-Brock

Submitted: 02 February 2021 Reviewed: 24 February 2021 Published: 29 March 2021

DOI: 10.5772/intechopen.96811

From the Edited Volume

Natural Medicinal Plants

Edited by Hany A. El-Shemy

Chapter metrics overview

468 Chapter Downloads

View Full Metrics

Abstract

Usage of supplements has increased dramatically this last decade. From herbs to vitamins and mineral, consumers are interested in improving health, self-treatment and preventing diseases. Often using information from the internet to self-prescribe, many consumers believe that natural products are safe, while many others avoid using these products because of the lack of an approval process by health officials in many countries. Herbs and other supplements including proteins, vitamins and minerals provide significant benefits to health. The lack of guidance from health professionals however can be problematic. When combined with drugs and disease, herbs can interact and cause side effects. Some of the steps to evaluate the safe use of supplements is to know their mechanism of action, clinical effect, and consumers’ medical history. For example, an herb that induces liver enzymes will reduce the effect of a drug that is metabolized by these same enzymes. This can be life threating if the patient depends on this drug for normal function. Based on drug-herb interaction experience and literature review, this book chapter provides insights into safe use of echinacea, licorice, turmeric, and black seed in patients with heart disease, diabetes, and COVID-19.

Keywords

  • herbs
  • supplements
  • drug-herb interaction
  • safety
  • COVID-19
  • heart disease
  • diabetes

1. Introduction

Dietary supplements are defined in the United States as products that contain one or more dietary ingredient such as vitamins, minerals, herbs, botanicals, and amino acids and are intended to supplement the diet [1]. In other countries dietary supplements are named differently including natural health products, complementary medicines, food supplements, and others [2]. Nonetheless, “dietary supplements” is a general term for products that mostly contain herbs, botanicals, proteins, and/or vitamins and minerals that are used with the intention to promote health. Despite the legal framework, dietary ingredients are often used and recommended for treating or preventing diseases. In this chapter, “dietary supplements” will be used as a general term to encompass several dietary ingredients.

Usage of dietary supplements has increased this last two decades [2]. From herbs, proteins, to vitamins and minerals, consumers are interested in self-treatment and preventing diseases [3]. Often using information from the internet to self-prescribe, many consumers believe that natural products are safe, while many others avoid using these products because of the lack of an approval process by health officials in many countries. Many dietary supplements provide significant benefits to health [4]. However, the lack of guidance from health professionals can be problematic.

Dietary supplements are likely safe when used as prescribed [4, 5]. But, when combined with drugs and disease, these products can interact and cause side effects [6, 7]. Some of the steps to evaluate the safe use of dietary ingredients is to know their mechanism of action, clinical effect, and consumers’ medical history. For example, an ingredient that induces liver enzymes will reduce the effect of a drug that is metabolized by these same enzymes. This can be life threating if the patient depends on this drug for normal function.

Due to the benefits that several of these dietary ingredients provide, it is important to evaluate their safety for wide spread recommendation. Particularly due to times of pandemic such as the coronavirus disease 2019 (COVID-19) [8], ways to prevent disease severity and to be used as adjunct treatments are needed. Several dietary ingredients have been reported to be effective against COVID-19 in review articles. For this book chapter, 30 review articles and meta-analysis were evaluated for the selection of the dietary ingredients herein discussed. The selection criterium was based on the number of articles that cited the ingredients as being effective as well as the commonality and accessibility of the ingredients across the globe. Vitamins and minerals were excluded due to their safety being extensively researched. Because COVID-19 severity is worse among patients with diabetes and cardiovascular disease, the safety use of these ingredients in the context of these comorbidities are presented here.

Advertisement

2. Comorbidities and their drug treatments

2.1 COVID-19

COVID-19 is a respiratory infection caused by the virus named “severe acute respiratory syndrome coronavirus 2” (SARS-CoV-2) [8]. COVID-19 is a novel disease officially declared as a pandemic on March 11th, 2020 [9, 10]. SARS-CoV-2 has infected 98.2 million people worldwide and caused 2.1 million deaths as of January 24th, 2021 [11]. COVID-19 is characterized by dry cough, fever, and fatigue symptoms in adults while in children rhinorrhea, abdominal pain, and diarrhea are also present [10]. SARS-CoV-2 binds directly to angiotensin converting enzyme 2 (ACE2) for subsequent entry into cells [10, 12]. Infected cells respond to the virus by generating pro-inflammatory cytokines and chemokines that sometimes lead to a cytokine storm which aggravates the disease [10, 12, 13]. Those with certain underlying health conditions such as respiratory disease, cardiovascular disease, and diabetes as well as older individuals seem to be at a higher risk for developing severe complications from the infection [14, 15]. Because SARS-CoV-2 has approximately 80% genomic homology with SARS-CoV-1, the virus that caused the 2002–2003 epidemic, many research studies have proposed the use of treatments that were effective against SARS-CoV-1 [9]. Current treatments for COVID-19 used in the clinics are ACE2 inhibitors, corticosteroids, chloroquine, anti-inflammatory tocilizumab, comostat, protease inhibitors (lopivavir and ritonavir), and RNA polymerase inhibitors (remdesivir, favipiravir) [16]. Some of the established protocols are: no treatment for mild cases besides acetaminophen for fever; hydroxychloroquine + azithromycin for moderate cases; tocilizumab or sarilumab for worsening respiratory function; and remdesivir, convalescent plasma, corticosteroids for respiratory failure. NSAIDs such as ibuprofen are not recommended due to potential increase in ACE2 expression [17]. Lastly, it has been suggested that reduction in cholesterol decreases viral mRNA [18]. Thus, treatments that reduce cholesterol in addition to antivirals, anti-inflammatories, and respiratory support should be beneficial in managing COVID-19.

2.2 Heart disease and diabetes

As noted above, patients with heart disease and diabetes are more likely to develop severe COVID-19. Thus, many of these patients will be given medications for COVID-19 on top of the current heart/diabetes medications they take. For example, patients continue to take ACE inhibitors or angiotensin II receptor blockers (ARBs) during COVID-19 infection [17]. Furthermore, these are the patients more likely to benefit from dietary ingredients that assist in preventing or treating COVID-19. Due to multiple treatments at once, the likelihood of drug–drug and drug-herb interaction in these patients is high. Drug treatments for heart disease include several types: anticoagulants, antiplatelets, ACE inhibitors, ARBs, beta blockers, calcium channel blockers, cholesterol lowering, diuretics, and vasodilators [19]. For diabetes main medication classes include sulfonylureas, meglitinides, metformin, and glitazones [20]. The metabolism of some commonly prescribed of these medications are listed in Table 1. As noted, the most common cytochrome P450 enzyme involved in the metabolism of these drugs are CYP3A4, followed by CYP2C9, 2D6, and 2C8 [21, 22, 23, 24, 25]. Approximately half of them are primarily excreted via the kidneys.

Drug categoriesLiver metabolismRenal excretionReferences
ACE inhibitors:
  1. Enalapril

  2. Lisinopril

  1. Metabolized to active metabolites

  2. No liver metabolism

  1. ~75% excretion in the urine

  2. Excreted unchanged in urine

[21, 22]
ARBs:
  1. Losartan

  2. Irbesartan

  1. CYP3A4 and CYP2C9

  2. Glucuronide conjugation and CYP2C9 oxidation

  1. Minimal renal excretion with oral administration

  2. ~ 20% excretion in the urine

[22]
HydroxychloroquinePartially metabolizedSlowly excreted by the kidneys[21]
Protease inhibitor, ritonavirCYP3A4 and 2D6 inhibitorMinimal renal excretion[16, 23]
RNA polymerase inhibitor, remdesivirCES1 to form active metabolite~50% renal excretion[22]
Corticosteroids and
anti-inflammatories
  1. Prednisone

  2. Tocilizumab

  1. Metabolized by CYP3A4

  2. Metabolized by CYP3A4

  1. Minimal renal excretion

[21, 24]
Antiplatelet, clopidogrelCYP 2C19 forms active metabolite~50% excreted in urine[22]
Beta blocker, atenololMinimal metabolismMajor renal excretion[21]
Cholesterol lowering:
  1. Atorvastatin

  2. Fluvastatin

  1. Metabolized by CYP3A4

  2. Metabolized by CYP2C9

  1. Minimal renal excretion

[22, 25]
Diuretic, spironolactoneExtensive metabolizedMajor renal excretion[22]
Sulfonylureas
  1. Glyburide

  2. Glipizide

  1. Extensive metabolized

  2. Metabolized by CYP2C9

  1. ~ 50% renal excretion

  2. Minimal renal excretion

[21, 22, 25]
Meglitinides, repaglinideMetabolized by CYP3A4Minimal renal excretion[21]
MetforminMinimal metabolism~90% renal excretion[22]
Glitazones, pioglitazoneMetabolized by CYP2C8~15–30% renal excretion[21]

Table 1.

Metabolism and excretion of some common medications used in COVID-19, heart disease and diabetes.

Advertisement

3. Echinacea spp. (echinacea) – Antiviral and immune support

3.1 Echinacea in COVID-19

Echinacea has antiviral and immunomodulatory effects that seems to be promising against COVID-19 [13, 26]. Several studies have investigated the benefits of echinacea in treating and preventing respiratory tract infections such as the common cold, but not for other health purposes [27]. No studies have yet been completed on echinacea and COVID-19 [28]. A meta-analysis including 17 clinical trials found that echinacea is safe and effective in preventing or treating viral infections. In a separate analysis including 12 clinical trials, echinacea showed to decrease or not change pro-inflammatory cytokines associated with cytokine storm (IL-6, IL-1β, and TNF-α) and increase or not change anti-inflammatory or immune-stimulatory cytokines (IL-10, IL-2, IL-8, IL-3, and IFN-γ). These effects are beneficial during infections since immune stimulatory and anti-inflammatory effects are needed but pro-inflammatory cytokines can aggravate the disease. Adverse events were mild with the most common reported being insomnia, gastrointestinal, and anxiety. One case of serious erythema was reported. Most studies included healthy participants and echinacea dose and method of extraction were quite variable making it difficult to evaluate safety in patients with comorbidities [28].

3.2 Echinacea in diabetes

Not many studies have investigated the effects of echinacea in diabetes. In Wistar rats, 33 days of echinacea root extract showed hypoglycemic activity similar to glibenclamide. No safety parameters were investigated [29]. Echinacea purpurea flower extract and caffeic acid derivatives inhibited α-amylase, α-glucosidase, and ACE activities in a concentration-dependent manner [30].

3.3 Echinacea in heart disease

Almost no studies have evaluated the effects of echinacea on heart conditions such as hypertension and hypercholesterolemia. In one study with 374 elderly, 349 reported to use over-the-counter drugs and 43 reported to use herbal medicine. Echinacea was the most common herbal therapy used while aspirin, acetaminophen, laxatives, antiacids, and vitamins were the most common over-the-counter drugs [31]. This single study suggests the potential for interaction of echinacea with drugs.

3.4 Echinacea toxicity

In a review article, echinacea was considered to have a high or medium evidence for efficacy and safety [32]. Debatable concern of hepatotoxicity with echinacea when used for more than 8 weeks has been raised [6]. On the other hand, echinacea has shown hepatic and renal protection against toxins in rats with no effect by itself on liver and kidney parameters including AST, ALT, ALP, blood urea nitrogen and creatinine [33]. No toxicity was found in rats and mice after oral or intravenous injection of Echinacea purpurea at high doses. No evidence of mutagenicity in vitro and in mice or carcinogenicity in hamster embryo cells [34]. Echinacea is contraindicated in patients with autoimmune disease. Little evidence to evaluate the effect of echinacea in renal impairment [35].

3.5 Echinacea pharmacokinetics

In vivo pharmacokinetics in 12 healthy men and women, Echinacea purpurea root inhibited CYP1A2 and induced CYP3A enzymes [36]. In another similar study with 13 healthy adults, Echinacea purpurea induced CYP3A enzymes but did not change the pharmacokinetics of ritonavir. Ritonavir is an inhibitor of CYP3A thus likely counteracted the induction caused by echinacea. No effect on p-glycoprotein was found [37]. In an in vitro study, Echinacea purpurea showed mixed effects on CYP3A4 and moderate inhibition of CYP2C9 [38]. A review article found echinacea to have high likelihood of drug-herb interaction [39].

3.6 Echinacea safety summary

Echinacea is likely safe when taken short-term, up to 8 weeks, in healthy adults. Unknown safety in patients with diabetes or heart disease. Caution should be taken when combining with medications metabolized by CYP3A, 1A2, and 2C9 enzymes.

Advertisement

4. Glycyrrhiza sp. root (licorice) – Antiviral and respiratory support

4.1 Licorice in COVID-19

Licorice root is used as a flavoring agent in food in many countries. In the United States, anise oil is often used for this purpose. Licorice is promoted as a dietary supplement for digestion, cough, infections, and others [40]. Frequently recommended by herbalists, licorice has recently shown to be the herb most frequently used for COVID-19 treatment [41, 42]. Several review articles have discussed the potential effectiveness of licorice in treating COVID-19 for its antiviral, anti-inflammatory, spasmolytic, and expectorant effects [9, 10, 12, 13, 14]. Some in vitro studies showed that the active component glycyrrhizin inhibits the replication of SARS-coronavirus (SARS-CoV) [43, 44]. Other in vitro studies showed that glycyrrhizin may prevent SARS-CoV-2 entry by binding to ACE2 receptors and other protein targets [45, 46]. Clinical trials of licorice use during COVID-19 are ongoing. Daily doses range from 250 mg 25% extract (62.5 mg glycyrrhizin) for 10 days to 2.28 g 3% extract (70 mg glycyrrhizin) for 7 days [47, 48].

4.2 Licorice in diabetes

Not many studies have investigated the effects of licorice in diabetes. In a clinical trial with 58 overweight and obese but otherwise healthy volunteers, 1.5 g licorice extract (<0.01% glycyrrhizin) for 8 weeks decreased insulin and HOMA-IR without side effects [49]. In cell cultures, de-glycyrrhizinated or regular licorice showed to be a potential therapeutic target in diabetic nephropathy [50]. In diabetic mice, licorice hydrophobic flavonoids demonstrated abdominal fat-lowering and hypoglycemic effects [51].

4.3 Licorice in heart disease

Cases of hypokalemia and hypertension have been reported after daily ingestion of licorice tea or after short-term high dose [52, 53, 54]. In one case patient was combining licorice with the glucocorticoid medication fludrocortisone [55]. The active components of licorice, glycyrrhizinates, inhibit the enzyme responsible for inactivating cortisol and bind to mineralocorticoid receptors resulting in reversible hyper-mineralocorticoid effects [56]. A meta-analysis with 18 clinical trials found that chronic daily intake of 100 mg glycyrrhizin increases systolic and diastolic blood pressure [57]. In another meta-analysis including 26 clinical trials and 985 subjects, mainly healthy and overweight but some with hypercholesterolemia, found licorice to reduce body weight and BMI but increase diastolic blood pressure. Licorice was given as licorice flavonoid oil with a dose range of 300 mg to 1.8 g/day for 2–16 weeks [58]. In a dose–response relationship investigation in healthy men and women, licorice root with 108 or 217 mg of glycyrrhizin per day for 4 weeks caused no adverse events. However, licorice with 380 and 814 mg glycyrrhizin caused headache, arterial hypertension, hyperkalemia, and peripheral edema. One individual had a family history of hypertension [59]. Lastly, a similar study compared adverse events in patients with hypertension versus normotensive individuals during 100 g licorice containing 150 mg glycyrrhetinic acid per day for 4 weeks. Systolic and diastolic blood pressure were slightly increased in normotensive (3.5 and 3.6 mmHg) but significantly greater increase in hypertensive patients (15.3 and 9.3 mmHg). Increase in urinary cortisol correlated with the rise in blood pressure [60]. These data suggest that glycyrrhizin at dose >200 mg/day short-term and > 100 mg/day long-term in patients or healthy individuals can cause reversible hyperkalemia and hypertension.

4.4 Licorice toxicity

Despite licorice being a substance generally recognized as safe (GRAS) in the United States [61] and regarded as having a high safety profile because it is consumed as food [32], licorice can cause hypertension and hypokalemia in a dose-dependent manner [57]. However, safe dose will vary depending on licorice’s composition and the underlying medical conditions. Those with hypertension, heart or kidney disease are more sensitive to licorice toxicity [40]. In a study involving 360 subjects, no clinically significant change in renal function (potassium, blood urea nitrogen, and creatinine levels) were found in 98.3% of the subjects after ~19 days of ~8 g licorice per day taken as dietary supplements that contained other ingredients. The remaining 1.7% of subjects developed hyperkalemia [62]. In a safety and toxicity study with 39 healthy female and male volunteers aged 19–40 years old, glycyrrhizic acid was administered at 1, 2, and 4 mg/kg body weight daily for 8 weeks. A no-effect level of 2 mg/kg was found and applying a 100-safety factor, the acceptable daily intake of 0.2 mg/kg body weight was proposed. This is equivalent to 12 mg glycyrrhizic acid/day for a 60-kg person [63]. Similarly, based on review of in vivo and clinical evidence, an acceptable daily intake has been proposed to be 0.015–0.229 mg glycyrrhizin/kg body weight [64]. The acceptable daily intake without a safety factor is equivalent to 120 mg glycyrrhizic acid. This dose could be considered safe if used short-term in a situation of high benefit versus risk.

4.5 Licorice pharmacokinetics

Glycyrrhiza glabra has shown weak inhibition of CYP3A4 and moderate inhibition of CYP2B6, 2C8, 2C9, and 2C19. Glycyrrhiza uralensis showed strong inhibition of CYP2B6, moderate inhibition of CYP2C8, 2C9, and 2C19, and no inhibition of CYP3A4 and 2D6. Glycyrrhiza inflata strongly inhibited CYP2C enzymes and moderately inhibited of CYP3A4, 1A2, 2B6, and 2D6. None of the three species inhibited CYP2E1 and 2A6. Glycyrrhizin content was highest in G. uralensis suggesting that glycyrrhizin is a weak inhibitor of the major enzymes CYP3A4 and 2D6 [65]. Weak inhibition of CYP3A4 and 2D6 by glycyrrhizin and G. glabra were also found in a different study [66].

4.6 Licorice safety summary

A safe daily dose for short-term use consists of licorice with less than 100 mg glycyrrhizin. For daily long-term use a dose of 12 mg glycyrrhizin has been proposed. COVID-19 studies are using short-term doses of <100 mg glycyrrhizin per day. Caution should be taken when combining licorice with medications. Licorice inhibits several cytochrome P450 enzymes including CYP1A2, 2B6, 2C8, 2C9, and 2C19. Only G. inflata inhibits CYP3A4 and 2D6.

Advertisement

5. Curcuma longa (turmeric) – Antiviral and anti-inflammatory

5.1 Turmeric in COVID-19

Turmeric has antiviral and anti-inflammatory effects that might benefit COVID-19 patients [10, 13]. It has also been hypothesized that the antioxidant effects of turmeric benefit diabetic patients during COVID-19 infection [67]. However, some has expressed concerns that curcumin, the main active component of turmeric, might increase the expression of ACE2 and worsen COVID-19 infection as well as increase pro-inflammatory cytokines and worsen COVID-19 in patients with cytokine storm [26]. In the contrary, curcumin binds to viral S protein and the viral attachment sites of the ACE2 receptor protein to inhibit the entry of SARS-CoV2 [18, 68]. In addition, curcumin has shown to reduce inflammatory cytokines in COVID-19 patients. In a clinical study with 40 COVID-19 patients, curcumin given as nano-curcumin at 160 mg/day for 14 days reduced the inflammatory cytokines IL-6 and IL-1β as well as clinical manifestations (fever, cough, dyspnea, headache, chest radiography, lymphocyte, white blood cells, and platelets count) in comparison to placebo-treated group. Both groups were taking atorvastatin, bromhexine, and betaferon concomitantly with 5–15% of them having diabetes, cardiovascular disease or renal disease. These results suggest the effectiveness and safety of curcumin in COVID-19 patients with underlying medical conditions [69].

5.2 Turmeric in diabetes

In clinical trials with type 2 diabetic patients, curcuminoids from 250 mg/day for 9 months to 1 g/day for 3 months improved glycemic control, β-cell function, insulin resistance, and reduced inflammatory cytokines with no major adverse effects. Minor side effects included diarrhea, constipation, vertigo, and itching. Some clinical and preclinical studies also showed that curcumin improve biomarkers of liver and kidney damage [70]. In a clinical trial on 46 patients with diabetic nephropathy, 1.5 g curcumin for 16 weeks improved 24-h urine analysis for albuminuria with no change in blood urea nitrogen, creatinine, fasting blood sugar, 2-h postprandial blood sugar, lipid profile, serum albumin, and hemoglobin A1C in comparison to placebo and baseline [71].

5.3 Turmeric in heart disease

A recent meta-analysis found that turmeric or curcumin have no effect on diastolic blood pressure and minor effect on systolic blood pressure when taken for longer than 12 weeks [72]. A meta-analysis that included 7 randomized, placebo-controlled clinical trials in patients with cardiovascular risk factors (i.e., non-alcoholic fatty liver disease, metabolic syndrome, type 2 diabetes, prehypertension, and dyslipidemia) found turmeric powder at 2–2.4 g/day for 1–2 months, turmeric extract with 0.6–1.9 g curcuminoids/day for 2–6 months, or curcumin at 70–80 mg/day for 2–3 months were effective in reducing serum LDL-cholesterol and triglycerides levels. Adverse events reported were abdominal pain, nausea, dyspepsia, constipation, and hot flushes. Hot flushes were also reported in the placebo group. In 3 of the trials patients were kept on their medications during the study; however, only one trial disclosed the name of the concomitant drug treatment (metformin) [73].

5.4 Turmeric toxicity

Turmeric has GRAS status in the United States [74]. Through a toxicological assessment, the European Food Safety Authority (EFSA) has recommended curcumin daily intake be ≤3 mg/kg body weight per day (180 mg/day in 60 kg individuals) [75]. In 2–year oral feed studies, turmeric oil at 79–85% curcumin showed no biological significantly differences in hematology, clinical chemistry (liver and kidney function markers), and urinalysis parameters, but showed to potentially cause carcinogenicity in mice and rats especially in females at doses ≥100 mg/kg body weight in rats and 300 mg/kg body weight in mice [76]. However, the EFSA concluded that curcumin is not carcinogenic and studies have demonstrated the benefits of curcumin as an adjunct treatment of cancer [77]. High daily dose of curcumin might cause hepatotoxicity. In rats, 25 and 100 mg/kg body weight for 90 days of curcumin induced liver injury through the generation of reactive oxygen species and pro-inflammatory cytokines as well as reduced antioxidant and detoxifying enzymes SOD and GST [78]. Similarly, 5% turmeric via diet for 90 days in female Wistar rats and 0.2% turmeric via diet in female Swiss mice was hepatotoxic. Human equivalent dose for these rodent studies ranged from 250 mg curcumin/day to 1 g–50 g turmeric/day [79, 80].

5.5 Turmeric pharmacokinetics

Turmeric constituents have shown to inhibit p-glycoprotein in vitro and in vivo models [81]. Inhibition of p-glycoprotein can lead to increased bioavailability of drugs [82]. Curcumin is primarily eliminated in the feces with little renal excretion in a rat study [76]. In a pharmacokinetics study with healthy adults, turmeric reduced the bioavailability of the beta-blocker talinolol [83]. Curcumin was safe and effective when combined with glyburide in patients with type 2 diabetes. Better cholesterol and glycemia control without hypoglycemic side effects were observed. Curcumin increased AUC but did not change Cmax of glyburide [84]. In rats, curcumin increased the Cmax, AUC0-t and half-life of amlodipine – an antihypertensive drug [85]. Amlodipine is metabolized by CYP3A4 in humans [86]. Curcumin inhibits several hepatic CYP enzymes including 3A4, 1A2, 2B6 (competitive type of inhibition), 2D6 and 2C9 (non-competitive inhibition) in human recombinant cytochrome P450s [87]. However, it is been suggested that these effects are not clinically significant due to poor bioavailability of curcumin. In fact, in a pharmacokinetics study in healthy volunteers, 4 g curcuminoids +24 mg piperine to enhance bioavailability did not affect Cmax, AUC, clearance, or half-life of drugs metabolized by CYP3A, CYP2C9, and UGT, SULT conjugation enzymes [88].

5.6 Turmeric safety summary

Turmeric is safe and effective at doses ≤250 mg curcumin/day. Higher doses are associated with hepatotoxicity and potentially carcinogenicity. Doses as low as 70–250 mg curcuminoids/day has shown to be effective in metabolic disorders and COVID-19. Although turmeric inhibits cytochrome P450 enzymes, these effects seem to be clinically negligible. Caution when taken with drugs that are substrates of p-glycoprotein in order to avoid drug overdose. Although turmeric has hypoglycemic effects and might cause side effects such as fainting when combined with antidiabetic medications, this combination has shown to be safe in clinical trials.

Advertisement

6. Nigella sativa (black seed) – Anti-inflammatory and respiratory support

6.1 Nigella sativa in COVID-19

N. sativa is a plant native to South East Asia with several pharmacological effects including bronchodilation, antitussive, and anti-inflammatory and used as treatments of respiratory conditions, diabetes, cardiovascular diseases, among others [89, 90]. For example, in a clinical trial with 90 obese women, 3 g/day of N. sativa oil for 8 weeks reduced serum levels of TNFα and hsCRP in comparison to placebo with no adverse events reported [91]. In patients with asthma, 1 g N. sativa oil per day for 4 weeks reduced several inflammatory markers and improved pulmonary function [92]. Preclinical studies have shown that constituents in the methanolic extract of N. sativa seeds are responsible for the bronchodilator effect [93]. Recently, N. sativa has been regarded as a potential therapy for COVID-19 [13, 18, 94, 95]. For example, in a molecular docking-based study N. sativa inhibited SARS-CoV2 [94].

6.2 Nigella sativa in diabetes

Several clinical trials have been conducted to evaluate N. sativa in patients with type 2 diabetes [96]. For example, three controlled studies investigated the adjuvant use of 1–3 g/day N. sativa seeds powder or 2.5 ml/day N. sativa oil for 12 weeks in patients with type-2 diabetes. Significant and similar effects were observed with doses of 2 and 3 g/d on the reductions in fasting blood glucose, 2-hour postprandial glucose, HbA1C levels, and insulin resistance. Treatments were not associated with any adverse renal or hepatic functions throughout the study period. Patients were concomitantly taking oral hypoglycemic drugs (glibenclamide, metformin, rosiglitazone) but not insulin. Patients with coronary artery disease, valvular heart disease, heart failure, uncontrolled hypertension, renal failure and hepatic failure were excluded [97, 98, 99].

6.3 Nigella sativa in heart disease

In a meta-analysis including 11 randomized clinical trials with 860 hypertensive or normotensive individuals, N. sativa seeds versus placebo and one versus standard treatment significantly reduced systolic blood pressure by −3.60 mmHg and diastolic blood pressure by −2.80 mmHg. [100]. A similar meta-analysis including 17 randomized clinical trials with 1185 individuals with hyperlipidemia, obesity, hypertension, type 2 diabetes, or others, found that N. sativa seed powder or oil 1–3 g/day for up to 3 months reduces total cholesterol, LDL-cholesterol, and triglycerides [101]. No adverse events were reported by the subjects [102, 103]. One study in elderly with hypertension reported mild adverse events including dyspepsia in 6 subjects (15.7%), nausea in 3 subjects (7.8%), and constipation in 2 subjects (5.2%). No electrolyte abnormalities, liver and renal toxicities, or orthostatic hypotension were observed [104].

6.4 Nigella sativa toxicity

N. sativa has GRAS status in the United States [74]. A randomized, placebo-controlled study with 40 healthy elderly investigated the safety profile after daily intake of 1 g N. sativa seed powder for 9 weeks. Results found no statistical changes in any of the biochemical markers of cardiac, liver, and kidney function [105]. In 70 patients with chronic renal disease, 2.5 ml/day N. sativa oil for 12 weeks was safe and effective in improving clinical and biochemical parameters of kidney function without adverse events [106]. In a clinical trial with obese women, N. sativa oil (which is present in whole seeds and polar seed extracts), reduced body weight, VLDL cholesterol and triglycerides [107]. Traditional toxicity studies in rodents have been performed. In mice, hepatotoxicity was observed after 14-days of oral dosing at 6–21 g/kg body weight of N. sativa seeds water extract. No signs of hepatotoxicity were observed with methanolic and chloroform extracts. Body weight reductions were seen in methanolic extracts [108]. Similar findings were observed with the water extract in rats with increases in serum gamma-glutamyl transferase and alanine aminotransferase, but no changes in alkaline phosphatase and degeneration of hepatocytes [109]. In another rat study, 1 g/day for 6 weeks of whole N. sativa seeds were protective against hyperlipidemia to a similar extent as simvastatin without adverse effects to liver markers [110]. Human equivalent doses are 30–100 g N. sativa extracts per day for the mice study, and 10 g whole seeds per day for the rat study [80].

6.5 Nigella sativa pharmacokinetics

One of the main active constituents in N. sativa seeds and oil is thymoquinone. Thymoquinone has shown to bind to human α(1)-acid glycoprotein in the plasma [111] and inhibit CYP2C9 > 2D6 > 1A2 > 3A4 liver enzymes [112]. In hypertensive rats, N. sativa + alompidine showed greater reduction in blood pressure and heart rate than N. sativa alone, but no effect on alompidine pharmacokinetics (Cmax, AUC0-t, Kel, and terminal half-life) [113]. In another study in hypertensive rats, N. sativa + losartan showed greater reduction in blood pressure than N. sativa alone. N. sativa slightly reduced losartan Cmax and AUC0-t [114]. Alompidine is metabolized by CYP3A4 and losartan by CYP2C9 and 3A4 in humans. These data suggest that N. sativa has minimal effect on CYP3A4 but inhibits CYP2C9. In other words, N. sativa has antihypertensive effects on its own but potentiates the effect of drugs metabolized by CYP2C9 which can cause further drop in blood pressure and lead to side effects such as fainting.

6.6 Nigella sativa safety summary

N. sativa whole seeds, oil or polar extracts (i.e., non-aqueous) at human doses up to 3 g/day for 12 weeks beneficially affect inflammatory and metabolic markers without adverse effects on heart, liver, or kidneys in healthy adults as well as in patients with heart disease and diabetes. N. sativa reduces blood glucose and blood pressure. Thus, caution when combining with hypoglycemic and antihypertensive drugs to avoid side effects. N. sativa can increase the bioavailability of drugs metabolized by CYP2C9 leading to higher risks of their side effects. Some diabetes and heart medications metabolized by CYP2C9 are losartan, fluvastatin, glipizide [25].

Advertisement

7. Interactions summary

The combination of several dietary ingredients might be desirable when their main mechanisms of action and clinical effects differ. For example, combination of an anti-inflammatory, antiviral, immunostimulant, and bronchodilator herbs might be recommended. Safety combination of black seed and turmeric has been demonstrated in a clinical study. N. sativa seed (1.5 g/d) and turmeric (2.4 g/d) in patients with metabolic syndrome for 4 weeks was safe and effective in reducing blood glucose, cholesterol, and blood pressure despite both ingredients having hypoglycemic and antihypertensive effects alone [115]. Any effect of echinacea on blood glucose and blood pressure is insufficient to evaluate. Licorice can increase blood pressure depending on the dose. Since turmeric and N. sativa have anti-hypertensive effects, the addition of licorice might be safe. All of the four dietary ingredients described here inhibit CYP2C9. All except N. sativa also inhibit CYP1A2. Turmeric and licorice also inhibit CYP2B6. Turmeric inhibits CYP3A4 and echinacea induces it. Thus, caution should be taken when combining these dietary ingredients with drugs metabolized by CYP3A4, 2C9, 1A2 and 2B6. As presented in Table 1, many drugs used in COVID-19, diabetes, and heart disease are metabolized by CYP3A4 and 2C9. Caution should be taken with echinacea and turmeric because they induce or inhibit CYP3A4, respectively. Lastly, many drug examples presented in Table 2 are excreted via urine. Turmeric and black seed are likely safe when combined with medications that are excreted by the kidneys. Caution when combining with licorice due to its potential to cause hyperkalemia. No sufficient evidence to evaluate echinacea’s effect on kidney function.

Dietary ingredientLevel of evidence in COVID-19Level of evidence in heart diseaseLevel of evidence in diabetesMain interactionsReferences
EchinaceaNoneNoneScarce: positive effect in 1 preclinical studyInduces CYP3A, inhibits CYP1A2, and CYP2C9[27, 28, 29, 36, 38]
LicoricePositive effects in vitro and 2 ongoing clinical trialsNegative effects in several clinical trials showing hypertension and hyperkalemiaScarce: positive effects in 1 clinical, 1 preclinical, and 1 in vitro studySafe dose <100 mg glycyrrhizin.
Inhibits CYP1A2, 2B6, 2C8, 2C9, and 2C19. Only G. inflata inhibits CYP3A4 and 2D6
[45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 63, 64, 65, 66]
TurmericPositive effects in vitro and 1 completed clinical trialPositive effects in several clinical trialsPositive effects in several clinical trialsSafe dose ≤250 mg. Inhibits p-glycoprotein and not clinically significant inhibition of P450 enzymes[68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 88]
Black seedPositive effects in vitroPositive effects in several clinical trialsPositive effects in several clinical trialsInhibits CYP2C9[94, 96, 97, 98, 99, 100, 101, 102, 103, 104, 111, 112, 113, 114]

Table 2.

Summary of level of evidence for efficacy and safety of echinacea, licorice, turmeric, and black seed in COVID-19, heart disease, and diabetes.

Advertisement

8. Conclusions

All the four dietary ingredients discussed herein are safe for use short-term as in a setting of treating a disease. However, some might not be safe when taken long-term. For example, no safety data was found for echinacea in heart disease and diabetes. Long-term use of low dose or short-term use of high dose licorice can cause reversible hypertension. Hepatotoxicity might occur with long-term use of turmeric >250 mg/day. Lastly, all of these four dietary ingredients are metabolized by cytochrome P450 enzymes to some extent. Mostly they inhibit CYP2C9, 1A2 and 2B6. Caution with echinacea because it induces CYP3A4 and turmeric because it inhibits it.

Advertisement

Conflict of interest

The author declares no conflict of interest.

References

  1. 1. Congress, Dietary Supplement Health and Education Act of1994, in S.784, U.S.o. America, Editor. 1994: https://www.congress.gov/bill/103rd-congress/senate-bill/784/text
  2. 2. Dwyer, J.T., P.M. Coates, and M.J. Smith, Dietary Supplements: Regulatory Challenges and Research Resources. Nutrients, 2018. 10(1)
  3. 3. Bailey, R.L., et al., Why US adults use dietary supplements. JAMA Intern Med, 2013. 173(5): p. 355-361
  4. 4. Fields, J.M., Dangers of scientific bias against herbal drugs for coronavirus disease 2019. J Integr Med, 2020. 18(6): p. 459-461
  5. 5. Posadzki, P., L.K. Watson, and E. Ernst, Adverse effects of herbal medicines: an overview of systematic reviews. Clin Med (Lond), 2013. 13(1): p. 7-12
  6. 6. Miller, L.G., Herbal medicinals: selected clinical considerations focusing on known or potential drug-herb interactions. Arch Intern Med, 1998. 158(20): p. 2200-2211
  7. 7. Liu, M.Z., et al., Pharmacogenomics and herb-drug interactions: merge of future and tradition. Evid Based Complement Alternat Med, 2015. 2015: p. 321091
  8. 8. WHO. Naming the Coronavirus Disease (COVID-19) and the Virus that Causes it. 2020 January 31st.]; Available from: https://www.who.int/emergencies/diseases/novel-coronavirus-2019/technical-guidance/naming-the-coronavirus-disease-(covid-2019)-and-the-virus-that-causes-it
  9. 9. Fuzimoto, A.D. and C. Isidoro, The antiviral and coronavirus-host protein pathways inhibiting properties of herbs and natural compounds - Additional weapons in the fight against the COVID-19 pandemic? J Tradit Complement Med, 2020. 10(4): p. 405-419
  10. 10. Jalali, A., et al., A pharmacology-based comprehensive review on medicinal plants and phytoactive constituents possibly effective in the management of COVID-19. Phytother Res, 2020
  11. 11. WHO. Weekly epidemiological update - 27 January 2021. 2021 January, 31st. ]; Available from: https://www.who.int/publications/m/item/weekly-epidemiological-update---27-january-2021
  12. 12. Wyganowska-Swiatkowska, M., et al., Influence of Herbal Medicines on HMGB1 Release, SARS-CoV-2 Viral Attachment, Acute Respiratory Failure, and Sepsis. A Literature Review. Int J Mol Sci, 2020. 21(13)
  13. 13. Brendler, T., et al., Botanical drugs and supplements affecting the immune response in the time of COVID-19: Implications for research and clinical practice. Phytother Res, 2020
  14. 14. Boukhatem, M.N. and W.N. Setzer, AromaticHerbs,Medicinal Plant-Derived EssentialOils, and Phytochemical Extracts as Potential Therapies for Coronaviruses: Future Perspectives. Plants (Basel), 2020. 9(6)
  15. 15. Fang, L., G. Karakiulakis, and M. Roth, Are patients with hypertension and diabetes mellitus at increased risk for COVID-19 infection? Lancet Respir Med, 2020. 8(4): p. e21
  16. 16. Hushmandi, K., et al., A review of medications used to control and improve the signs and symptoms of COVID-19 patients. Eur J Pharmacol, 2020. 887: p. 173568
  17. 17. Mehta, N., et al., Pharmacotherapy in COVID-19; A narrative review for emergency providers. Am J Emerg Med, 2020. 38(7): p. 1488-1493
  18. 18. Ho, P., et al., Perspective Adjunctive Therapies for COVID-19: Beyond Antiviral Therapy. Int J Med Sci, 2021. 18(2): p. 314-324
  19. 19. AHA. Types of Heart Medications. 2020 January 31st ]; Available from: https://www.heart.org/en/health-topics/heart-attack/treatment-of-a-heart-attack/cardiac-medications#ARB
  20. 20. ADA. Oral medications. 2021 January 31st]; Available from: https://www.diabetes.org/healthy-living/medication-treatments/oral-medication
  21. 21. ASHP. AHFS Drug Information Monographs. 2021 January 31st]; Available from: https://www.drugs.com/monograph/
  22. 22. FDA. Professional Drug Information. 2021 January 31st]; Available from: https://www.drugs.com/pro/
  23. 23. Hsu, A., G.R. Granneman, and R.J. Bertz, Ritonavir. Clinical pharmacokinetics and interactions with other anti-HIV agents. Clin Pharmacokinet, 1998. 35(4): p. 275-291
  24. 24. FDA. Full prescribing information for Actemra. 2010 January 31st]; Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/125276s107_125472s018lbl.pdf
  25. 25. Van Booven, D., et al., Cytochrome P450 2C9-CYP2C9. Pharmacogenet Genomics, 2010. 20(4): p. 277-281
  26. 26. Nugraha, R.V., et al., Traditional Herbal Medicine Candidates as Complementary Treatments for COVID-19: A Review of Their Mechanisms,Pros and Cons. Evid Based Complement Alternat Med, 2020. 2020: p. 2560645
  27. 27. FDA, Echinacea, U.S.D.o.H.a.H. Services, Editor. 2020: https://www.nccih.nih.gov/health/echinacea
  28. 28. Aucoin, M., et al., The effect of Echinacea spp. on the prevention or treatment of COVID-19 and other respiratory tract infections in humans: A rapid review. Adv Integr Med, 2020. 7(4): p. 203-217
  29. 29. Aarland, R.C., et al., Studies on phytochemical, antioxidant, anti-inflammatory, hypoglycaemic and antiproliferative activities of Echinacea purpurea and Echinacea angustifolia extracts. Pharm Biol, 2017. 55(1): p. 649-656
  30. 30. Chiou, S.Y., et al., Antioxidant, Antidiabetic, and Antihypertensive Properties of Echinacea purpurea Flower Extract and Caffeic Acid Derivatives Using In Vitro Models. J Med Food, 2017. 20(2): p. 171-179
  31. 31. Albert, N.M., et al., Predictors of over-the-counter drug and herbal therapies use in elderly patients with heart failure. J Card Fail, 2009. 15(7): p. 600-606
  32. 32. Silveira, D., et al., COVID-19: Is There Evidence for the Use of Herbal Medicines as Adjuvant Symptomatic Therapy? Front Pharmacol, 2020. 11: p. 581840
  33. 33. Rezaie, A., et al., Effects of Echinacea purpurea on Hepatic and Renal Toxicity Induced by Diethylnitrosamine in Rats. Jundishapur J Nat Pharm Prod, 2013. 8(2): p. 60-64
  34. 34. Mengs, U., C.B. Clare, and J.A. Poiley, Toxicity of Echinacea purpurea. Acute, subacute and genotoxicity studies. Arzneimittelforschung, 1991. 41(10): p. 1076-1081
  35. 35. EMA, Assessment report on Echinacea purpurea (L.) Moench., herba recens, E.M. Agency, Editor. 2014: https://www.ema.europa.eu/en/documents/herbal-report/final-assessment-report-echinacea-purpurea-l-moench-herba-recens_en.pdf
  36. 36. Gorski, J.C., et al., The effect of echinacea (Echinacea purpurea root) on cytochrome P450 activity in vivo. Clin Pharmacol Ther, 2004. 75(1): p. 89-100
  37. 37. Penzak, S.R., et al., Echinacea purpurea significantly induces cytochrome P450 3A activity but does not alter lopinavir-ritonavir exposure in healthy subjects. Pharmacotherapy, 2010. 30(8): p. 797-805
  38. 38. Yale, S.H. and I. Glurich, 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(3): p. 433-439
  39. 39. Tsai, H.H., et al., Evaluation of documented drug interactions and contraindications associated with herbs and dietary supplements: a systematic literature review. Int J Clin Pract, 2012. 66(11): p. 1056-1078
  40. 40. FDA, Licorice Root, U.S.D.o.H.a.H. Services, Editor. 2020: www.nccih.nih.gov/health/licorice-root
  41. 41. Ang, L., et al., Herbal medicine for treatment of children diagnosed with COVID-19: A review of guidelines. Complement Ther Clin Pract, 2020. 39: p. 101174
  42. 42. Ang, L., et al., Herbal medicine for the management of COVID-19 during the medical observation period: A review of guidelines. Integr Med Res, 2020. 9(3): p. 100465
  43. 43. Cinatl, J., et al., Glycyrrhizin, an active component of liquorice roots, and replication of SARS-associated coronavirus. Lancet, 2003. 361(9374): p. 2045-2046
  44. 44. Hoever, G., et al., Antiviral activity of glycyrrhizic acid derivatives against SARS-coronavirus. J Med Chem, 2005. 48(4): p. 1256-1259
  45. 45. Chen, H. and Q . Du, Potential natural compounds for preventing SARS-CoV-2 (2019-nCoV) infection. Preprints, 2020. 2020010358
  46. 46. Vardhan, S. and S.K. Sahoo, In silico ADMET and molecular docking study on searching potential inhibitors from limonoids and triterpenoids for COVID-19. Comput Biol Med, 2020. 124: p. 103936
  47. 47. Network, E.B.R., Evaluation of The Potential Therapeutic Effects of Licorice and Boswellia Serrata Gum in Egyptian Patients With COVID-19 as a Complementary Medicine. Identifier: NCT04487964, 2020, January - 2021, January. https://clinicaltrials.gov/ct2/show/NCT04487964
  48. 48. Safa, O., et al., Effects of Licorice on clinical symptoms and laboratory signs in moderately ill patients with pneumonia from COVID-19: A structured summary of a study protocol for a randomized controlled trial. Trials, 2020. 21(1): p. 790
  49. 49. Alizadeh, M., et al., Changes of Insulin Resistance and Adipokines Following Supplementation with Glycyrrhiza Glabra L. Extract in Combination with a Low-Calorie Diet in Overweight and Obese Subjects: a Randomized Double Blind Clinical Trial. Adv Pharm Bull, 2018. 8(1): p. 123-130
  50. 50. Hsu, Y.C., et al., De-Glycyrrhizinated Licorice Extract Attenuates High Glucose-Stimulated Renal Tubular Epithelial-Mesenchymal Transition via Suppressing the Notch2 Signaling Pathway. Cells, 2020. 9(1)
  51. 51. Nakagawa, K., et al., Licorice flavonoids suppress abdominal fat accumulation and increase in blood glucose level in obese diabetic KK-A(y) mice. Biol Pharm Bull, 2004. 27(11): p. 1775-1778
  52. 52. Kwon, Y.E., D.J. Oh, and H.M. Choi, Severe asymptomatic hypokalemia associated with prolonged licorice ingestion: A case report. Medicine (Baltimore), 2020. 99(30): p. e21094
  53. 53. Yang, L.Y., et al., Liquorice-induced severe hypokalemic rhabdomyolysis with Gitelman syndrome and diabetes: A case report. World J Clin Cases, 2019. 7(10): p. 1200-1205
  54. 54. Smedegaard, S.B. and M.V. Svart, Licorice induced pseudohyperaldosteronism, severe hypertension, and long QT. Endocrinol Diabetes Metab Case Rep, 2019. 2019
  55. 55. Benge, E., et al., Trick or Treat?Licorice-Induced Hypokalemia: A Case Report. Cureus, 2020. 12(11): p. e11656
  56. 56. Kwon, Y.J., et al., A Review of the Pharmacological Efficacy and Safety of Licorice Root from Corroborative Clinical Trial Findings. J Med Food, 2020. 23(1): p. 12-20
  57. 57. Penninkilampi, R., E.M. Eslick, and G.D. Eslick, The association between consistent licorice ingestion, hypertension and hypokalaemia: a systematic review and meta-analysis. J Hum Hypertens, 2017. 31(11): p. 699-707
  58. 58. Luís, Â., F. Domingues, and L. Pereira, Metabolic changes after licorice consumption: A systematic review with meta-analysis and trial sequential analysis of clinical trials. Phytomedicine, 2018. 39: p. 17-24
  59. 59. Bernardi, M., et al., Effects of prolonged ingestion of graded doses of licorice by healthy volunteers. Life Sci, 1994. 55(11): p. 863-872
  60. 60. Sigurjonsdottir, H.A., et al., Subjects with essential hypertension are more sensitive to the inhibition of 11 beta-HSD by liquorice. J Hum Hypertens, 2003. 17(2): p. 125-131
  61. 61. CFR, Listing of Specific Substances Affirmed as GRAS: Licorice and licorice derivatives, in 21 Code of Federal Regulations 184.1408, U.S.o. America, Editor. 2020
  62. 62. Jung, W., et al., Influence of herbal complexes containing licorice on potassium levels: a retrospective study. Evid Based Complement Alternat Med, 2014. 2014: p. 970385
  63. 63. van Gelderen, C.E., et al., Glycyrrhizic acid: the assessment of a no effect level. Hum Exp Toxicol, 2000. 19(8): p. 434-439
  64. 64. Isbrucker, R.A. and G.A. Burdock, Risk and safety assessment on the consumption of Licorice root (Glycyrrhiza sp.), its extract and powder as a food ingredient, with emphasis on the pharmacology and toxicology of glycyrrhizin. Regul Toxicol Pharmacol, 2006. 46(3): p. 167-192
  65. 65. Li, G., et al., Cytochrome P450 inhibition by three licorice species and fourteen licorice constituents. Eur J Pharm Sci, 2017. 109: p. 182-190
  66. 66. Pandit, S., et al., Exploring the possible metabolism mediated interaction of Glycyrrhiza glabra extract with CYP3A4 and CYP2D6. Phytother Res, 2011. 25(10): p. 1429-1434
  67. 67. Tabatabaei-Malazy, O., M. Abdollahi, and B. Larijani, Beneficial Effects of Anti-Oxidative Herbal Medicines in Diabetic Patients Infected with COVID-19: A Hypothesis. Diabetes Metab Syndr Obes, 2020. 13: p. 3113-3116
  68. 68. Noor, H., et al., Immunomodulatory and anti-cytokine therapeutic potential of curcumin and its derivatives for treating COVID-19 - a computational modeling. J Biomol Struct Dyn, 2021: p. 1-16
  69. 69. Valizadeh, H., et al., Nano-curcumin therapy, a promising method in modulating inflammatory cytokines in COVID-19 patients. Int Immunopharmacol, 2020. 89(Pt B): p. 107088
  70. 70. Pivari, F., et al., Curcumin and Type 2 Diabetes Mellitus: Prevention and Treatment. Nutrients, 2019. 11(8)
  71. 71. Vanaie, A., et al., Curcumin as a major active component of turmeric attenuates proteinuria in patients with overt diabetic nephropathy. J Res Med Sci, 2019. 24: p. 77
  72. 72. Hadi, A., et al., The effect of Curcumin/Turmeric on blood pressure modulation: A systematic review and meta-analysis. Pharmacol Res, 2019. 150: p. 104505
  73. 73. Qin, S., et al., Efficacy and safety of turmeric and curcumin in lowering blood lipid levels in patients with cardiovascular risk factors: a meta-analysis of randomized controlled trials. Nutr J, 2017. 16(1): p. 68
  74. 74. CFR, Substances generally recognized as safe: Spices and other natural seasonings and flavorings., in 21 Code of Federal Regulations 182.10, U.S. government, Editor. 2021
  75. 75. EFSA, Scientific Opinion on the re-evaluation of curcumin (E 100) as a food additive. European Food Safety Authority Journal, 2010. 8(9): p. 1679
  76. 76. NTP, NTPToxicology and Carcinogenesis Studies of Turmeric Oleoresin (CAS No. 8024-37-1) (Major Component 79%-85% Curcumin, CAS No. 458-37-7) in F344/N Rats and B6C3F1 Mice (Feed Studies). Natl Toxicol Program Tech Rep Ser, 1993. 427: p. 1-275
  77. 77. Adiwidjaja, J., A.J. McLachlan, and A.V. Boddy, Curcumin as a clinically-promising anti-cancer agent: pharmacokinetics and drug interactions. Expert Opin Drug Metab Toxicol, 2017. 13(9): p. 953-972
  78. 78. Qiu, P., et al., Overdose Intake of Curcumin Initiates the Unbalanced State of Bodies. J Agric Food Chem, 2016. 64(13): p. 2765-2771
  79. 79. EPA, Recommendations for and Documentation of Biological Values for Use in Risk Assessment., U.S.E.P. Agency, Editor. 1988: https://cfpub.epa.gov/ncea/risk/recordisplay.cfm?deid=34855
  80. 80. Reagan-Shaw, S., M. Nihal, and N. Ahmad, Dose translation from animal to human studies revisited. Faseb j, 2008. 22(3): p. 659-661
  81. 81. Lopes-Rodrigues, V., E. Sousa, and M.H. Vasconcelos, Curcumin as a Modulator of P-Glycoprotein in Cancer: Challenges and Perspectives. Pharmaceuticals (Basel), 2016. 9(4)
  82. 82. Lin, J.H. and M. Yamazaki, Role of P-glycoprotein in pharmacokinetics: clinical implications. Clin Pharmacokinet, 2003. 42(1): p. 59-98
  83. 83. Juan, H., et al., Unexpected effect of concomitantly administered curcumin on the pharmacokinetics of talinolol in healthy Chinese volunteers. Eur J Clin Pharmacol, 2007. 63(7): p. 663-668
  84. 84. Neerati, P., R. Devde, and A.K. Gangi, Evaluation of the effect of curcumin capsules on glyburide therapy in patients with type-2 diabetes mellitus. Phytother Res, 2014. 28(12): p. 1796-1800
  85. 85. Jiang, N., et al., Effects of curcumin on the pharmacokinetics of amlodipine in rats and its potential mechanism. Pharm Biol, 2020. 58(1): p. 465-468
  86. 86. Zhu, Y., et al., Amlodipine metabolism in human liver microsomes and roles of CYP3A4/5 in the dihydropyridine dehydrogenation. Drug Metab Dispos, 2014. 42(2): p. 245-249
  87. 87. Appiah-Opong, R., et al., Inhibition of human recombinant cytochrome P450s by curcumin and curcumin decomposition products. Toxicology, 2007. 235(1-2): p. 83-91
  88. 88. Volak, L.P., et al., Effect of a herbal extract containing curcumin and piperine on midazolam, flurbiprofen and paracetamol (acetaminophen) pharmacokinetics in healthy volunteers. Br J Clin Pharmacol, 2013. 75(2): p. 450-462
  89. 89. Clarke, R., F. Lundy, and L. McGarvey, Herbal treatments in asthma and COPD - current evidence. Clinical Phytoscience, 2015. 1(4)
  90. 90. Dajani, E.Z., T.G. Shahwan, and N.E. Dajani, Overview of the preclinical pharmacological properties of Nigella sativa (black seeds): a complementary drug with historical and clinical significance. J Physiol Pharmacol, 2016. 67(6): p. 801-817
  91. 91. Mahdavi, R., et al., Nigella sativa oil with a calorie-restricted diet can improve biomarkers of systemic inflammation in obese women: A randomized double-blind, placebo-controlled clinical trial. J Clin Lipidol, 2016. 10(5): p. 1203-1211
  92. 92. Koshak, A., et al., Nigella sativa Supplementation Improves Asthma Control and Biomarkers: A Randomized, Double-Blind,Placebo-Controlled Trial. Phytother Res, 2017. 31(3): p. 403-409
  93. 93. Keyhanmanesh, R., et al., The Relaxant Effects of Different Methanolic Fractions of Nigella sativa on Guinea Pig Tracheal Chains. Iran J Basic Med Sci, 2013. 16(2): p. 123-128
  94. 94. Ahmad, S., et al., Molecular docking, simulation and MM-PBSA studies of Nigella sativa compounds: a computational quest to identify potential natural antiviral for COVID-19 treatment. J Biomol Struct Dyn, 2020: p. 1-9
  95. 95. Islam, M.N., et al., Revisiting pharmacological potentials of Nigella sativa seed: A promising option for COVID-19 prevention and cure. Phytother Res, 2020
  96. 96. Hamdan, A., R. Haji Idrus, and M.H. Mokhtar, Effects of Nigella sativa on Type-2 Diabetes Mellitus: A Systematic Review. Int J Environ Res Public Health, 2019. 16(24)
  97. 97. Bamosa, A.O., et al., Effect of Nigella sativa seeds on the glycemic control of patients with type 2 diabetes mellitus. Indian J Physiol Pharmacol, 2010. 54(4): p. 344-354
  98. 98. Hosseini, M., et al., Effects of Nigella sativa L. Seed Oil in Type II Diabetic Patients: a Randomized, Double-Blind, Placebo - Controlled Clinical Trial. Journal of Medicinal Plants, 2013. 12(47): p. 93-99
  99. 99. Kaatabi, H., et al., Nigella sativa improves glycemic control and ameliorates oxidative stress in patients with type 2 diabetes mellitus: placebo controlled participant blinded clinical trial. PLoS One, 2015. 10(2): p. e0113486
  100. 100. Sahebkar, A., et al., A systematic review and meta-analysis of randomized controlled trials investigating the effects of supplementation with Nigella sativa (black seed) on blood pressure. J Hypertens, 2016. 34(11): p. 2127-2135
  101. 101. Sahebkar, A., et al., Nigella sativa (black seed) effects on plasma lipid concentrations in humans: A systematic review and meta-analysis of randomized placebo-controlled trials. Pharmacol Res, 2016. 106: p. 37-50
  102. 102. Sabzghabaee, A.M., et al., Clinical evaluation of Nigella sativa seeds for the treatment of hyperlipidemia: a randomized, placebo controlled clinical trial. Med Arch, 2012. 66(3): p. 198-200
  103. 103. Dehkordi, F.R. and A.F. Kamkhah, Antihypertensive effect of Nigella sativa seed extract in patients with mild hypertension. Fundam Clin Pharmacol, 2008. 22(4): p. 447-452
  104. 104. Rizka, A., et al., Effect of Nigella sativa Seed Extract for Hypertension in Elderly: a Double-blind,Randomized Controlled Trial. Acta Med Indones, 2017. 49(4): p. 307-313
  105. 105. Bin Sayeed, M.S., et al., The effect of Nigella sativa Linn. seed on memory, attention and cognition in healthy human volunteers. J Ethnopharmacol, 2013. 148(3): p. 780-786
  106. 106. Alam, M.A., et al., Evaluation of safety and efficacy profile of Nigella sativa oil as an add-on therapy, in addition to alpha-keto analogue of essential amino acids in patients with chronic kidney disease. Saudi J Kidney Dis Transpl, 2020. 31(1): p. 21-31
  107. 107. Mahdavi, R., et al., Effects of Nigella sativa oil with a low-calorie diet on cardiometabolic risk factors in obese women: a randomized controlled clinical trial. Food Funct, 2015. 6(6): p. 2041-2048
  108. 108. Vahdati-Mashhadian, N., H. Rakhshandeh, and A. Omidi, An investigation on LD50 and subacute hepatic toxicity of Nigella sativa seed extracts in mice. Pharmazie, 2005. 60(7): p. 544-7
  109. 109. Tennekoon, K.H., et al., Possible hepatotoxicity of Nigella sativa seeds and Dregea volubilis leaves. J Ethnopharmacol, 1991. 31(3): p. 283-289
  110. 110. Muneera, K.E., A. Majeed, and A.K. Naveed, Comparative evaluation of Nigella sativa (Kalonji) and simvastatin for the treatment of hyperlipidemia and in the induction of hepatotoxicity. Pak J Pharm Sci, 2015. 28(2): p. 493-498
  111. 111. Lupidi, G., et al., Characterization of thymoquinone binding to human α₁-acid glycoprotein. J Pharm Sci, 2012. 101(7): p. 2564-2573
  112. 112. Albassam, A.A., et al., Inhibition of cytochrome P450 enzymes by thymoquinone in human liver microsomes. Saudi Pharm J, 2018. 26(5): p. 673-677
  113. 113. Alam, M.A., et al., Effect of Nigella sativa and Fenugreek on the Pharmacokinetics and Pharmacodynamics of Amlodipine in Hypertensive Rats. Curr Drug Metab, 2020. 21(4): p. 318-325
  114. 114. Ahad, A., et al., Potential pharmacodynamic and pharmacokinetic interactions of Nigella Sativa and Trigonella Foenum-graecum with losartan in L-NAME induced hypertensive rats. Saudi J Biol Sci, 2020. 27(10): p. 2544-2550
  115. 115. Amin, F., et al., Clinical efficacy of the co-administration of Turmeric and Black seeds (Kalongi) in metabolic syndrome - a double blind randomized controlled trial - TAK-MetS trial. Complement Ther Med, 2015. 23(2): p. 165-174

Written By

Paula Vieira-Brock

Submitted: 02 February 2021 Reviewed: 24 February 2021 Published: 29 March 2021