Hostname: page-component-76fb5796d-vvkck Total loading time: 0 Render date: 2024-04-29T15:12:20.506Z Has data issue: false hasContentIssue false

Review: bioavailability and efficacy of ‘free’ curcuminoids from curcumagalactomannoside (CGM) curcumin formulation

Published online by Cambridge University Press:  19 January 2023

Cristina Matthewman
Affiliation:
Life Extension, Inc., Ft. Lauderdale, Florida, USA
I.M. Krishnakumar
Affiliation:
Akay Natural Ingredients, Cochin, India
Andrew G. Swick*
Affiliation:
Life Extension, Inc., Ft. Lauderdale, Florida, USA
*
*Corresponding author: Andrew G. Swick, email: aswick@lifeextension.com
Rights & Permissions [Opens in a new window]

Abstract

The golden spice turmeric with its main bioactive component curcumin is one of the most popular and extensively studied nutraceuticals. Despite numerous pre-clinical studies reporting positive pharmacodynamics of turmeric extracts and curcumin, the main issues in translating the pharmacological effects to clinical efficacy have been to overcome its poor pharmacokinetics and to deliver significant amounts of the biologically relevant forms of the actives to various tissues. This review is aimed at providing a first critical evaluation of the current published literature with the novel curcumagalactomannoside (CGM) formulation of curcumin using fenugreek galactomannan dietary fibre, specifically designed to address curcumin poor pharmacokinetics. We describe CGM and its technology as a food-grade formulation to deliver ‘free’ unconjugated curcuminoids with enhanced bioavailability and improved pharmacokinetic properties. The therapeutic relevance of improving bioavailability of ‘free’ curcuminoids and some of the technical challenges in the measurement of the ‘free’ form of curcuminoids in plasma and tissues are also discussed. A total of twenty-six manuscripts are reviewed here, including fourteen pre-clinical and twelve clinical studies that have investigated CGM pharmacokinetics, safety and efficacy in various animal models and human conditions. Overall current scientific evidence suggests CGM formulation has improved bioavailability and tissue distribution of the biologically relevant unconjugated forms of turmeric actives called ‘free’ curcuminoids that may be responsible for the superior clinical outcomes reported with CGM treatments in comparison with unformulated standard curcumin across multiple studies.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
© The Author(s), 2023. Published by Cambridge University Press on behalf of The Nutrition Society

Introduction

Scientific research has unravelled several biologically active components from the age-old Ayurvedic medicine and kitchen spice turmeric (Curcuma longa L.). Curcumin, the yellow polyphenolic pigment in turmeric, has received much scientific scrutiny over the years and has been proven to be the major active component responsible for the pharmacological effects of turmeric. Turmeric is considered ‘Generally Recognized As Safe’ (GRAS) by the US Food and Drug Administration, and curcumin is safe and well tolerated even when administered at high doses of 10–12 g in healthy volunteers(Reference Lao1).

Today there are thousands of publications reporting on the functional and therapeutic benefits of curcumin mediated through its pleiotropic mechanisms of action in the context of many chronic diseases. Its distinct chemical structure as α,β-unsaturated diketone moiety flanked by two phenolic groups, [1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione], makes curcumin a unique pharmacophore with high reactivity and great binding affinity towards several molecular targets such as transcription factors, enzymes and receptors involved in oxidative stress, apoptosis and inflammatory pathways which further translate to the observed neuroprotective, cardioprotective, gastroprotective, hepatoprotective and anticancer effects when a proper dosage of the bioactive form is delivered(Reference Salehi2Reference Sharifi-Rad13).

Despite the interesting pharmacodynamics, curcumin has been notorious for its poor pharmacokinetics. Curcumin is a class IV Biopharmaceutics Classification System molecule with low solubility (<10 ng/ml), low permeability and low oral bioavailability(Reference Wahlang, Pawar and Bansal14). Curcumin’s H-bond donating and accepting capacity, Michael acceptor type ligand character and metal-chelating properties also contribute to its in vivo instability(Reference Heger15). Moreover, curcumin undergoes rapid intestinal biotransformation into inactive metabolites, mainly the glucuronide and sulphate conjugates, which contribute to the major limitations of curcumin in translation to therapeutic benefits (Fig. 1)(Reference Nelson16Reference Kurita and Makino19). A key role in mediating the efficacy of any substance that undergoes rapid biotransformation is the absorption of the bioactive and permeable form; thus, drug delivery systems capable of providing the unconjugated or ‘free’ curcuminoids have major therapeutic implications.

Fig. 1. Oral administration of curcumin results mainly in conjugated curcumin detected in plasma, and intravenous or intraperitoneal administration results mainly in reduced curcumin metabolites, while there are minor levels of free curcumin detected in plasma following any administration method.

To our knowledge, this review is the first critical overview of the current pre-clinical and clinical research conducted with curcumagalactomannoside (CGM), a novel curcumin formulation designed to overcome the pharmacokinetic challenges of curcumin and improve the bioavailability and tissue distribution of the unconjugated ‘free’ curcuminoids. Running a search across for the terms ‘CGM’, ‘curcumin’, ‘CurQfen’ and ‘curcumagalactomannoside’ in PubMed, MEDLINE and EMBASE databases along with Google Scholar yielded thirty-two results (5 May 2022). We have excluded duplicates and manuscripts that have not explicitly evaluated CGM formulation, which resulted in twenty-six study-specific manuscripts (two in vitro, twelve in vivo and twelve clinical studies) included in this review.

The first section describes CGM formulation technology, highlighting the therapeutic relevance of improving bioavailability of the bioactive ‘free’ curcuminoids as well as some of the technical challenges in quantifying these actives. The following sections summarise current published literature with CGM evaluating pharmacokinetics, safety and efficacy of CGM in various models and health/disease conditions.

CGM curcumin formulation

CGM is a patented formulation of curcumin with galactomannan biopolymer, a soluble dietary fibre component of another GRAS-listed traditional Indian medicinal spice, fenugreek (Trigonella foenum graecum). CGM contains 40% turmeric root extract standardised to 35% total curcuminoids (sum of curcumin, demethoxycurcumin (DMC) and bisdemethoxycurcumin (BDMC)) and 60% fenugreek galactomannan dietary fibre, without any other phytochemicals found in fenugreek (M/s Akay Natural Ingredients Private Limited, Cochin, India, trademarked as CurQfen®).

FENUMAT® patented technology

In the manufacturing process, curcuminoid molecules are allowed to interact and uniformly impregnate the homogeneous gel phase of fenugreek galactomannans. Curcuminoids are effectively encapsulated in the hydrophobic pockets created by the conformationally restricted molecular chains of galactose and mannose (1:1 ratio) that behave like a soft hydrogel scaffold. The result is a self-emulsifying non-covalent molecular complex of curcumin and fenugreek galactomannans that converts the insoluble crystalline curcumin to amorphous, water-dispersible and stable ‘curcumagalactomannoside’. The microparticle with a size of less than 150 µm renders it a reversible hydrogel with high swelling index, optimum hydrophobic–hydrophilic balance and amphiphilicity that confers both protection from intestinal conjugation and an increase in the absorption of ‘free’ curcuminoids into bloodstream(Reference Krishnakumar, Dinesh Kumar, Kuttan and Maliakel20,Reference Song21) . As a water-only-based process, FENUMAT® formulation technology is considered the first ‘green’ approach in phytonutrient delivery. This technology also enabled the co-delivery and enhanced bioavailability of low-dose curcumin with other known lipophilic phytonutrients such as boswellic acids(Reference Abhilash22). Subsequently, FENUMAT® technology was successfully employed for gastric irritation prevention and sustained intestinal delivery of the pungent capsaicinoids from red chili pepper (Capsicum annum L.)(Reference Joseph23). FENUMAT® could also deliver gummy- and essential-oil-rich components of Ferula asafoetida oleo gum resin, an Ayurvedic remedy for functional dyspepsia(Reference Vijayasteltar24), and fixed oils such as vitamin-E-rich sunflower oil in free-flowing water-soluble granular powders(Reference Vismaya25).

Therapeutic relevance of biologically active ‘free’ curcuminoids

Several studies have highlighted the significance of the native unconjugated or ‘free’ forms of curcuminoids, owing to their ability to cross cellular membranes, blood–brain barrier permeability and potential in vivo antioxidant, anti-inflammatory, anti-proliferative, neuroprotective, anti-amyloid and neurogenesis effects(Reference Begum26Reference Ireson29). Although some nano formulations provide ‘free’ curcuminoids upon intravenous administrations, most of the oral curcumin formulations yield only the conjugated metabolites in plasma(Reference Kumar30). Oral administration of native unformulated curcumin with 95% purity at high doses of 10–12 g could detect only less than 10 ng/g ‘free’ curcuminoids in some subjects(Reference Lao1). Garcea et al. mentioned that doses of (unformulated) curcumin required to provide sufficient tissue levels to exert pharmacological activity are probably not feasible in humans(Reference Garcea31). So, formulations that can deliver high levels of native unconjugated ‘free’ curcuminoids into systemic circulation and further into tissues at convenient doses have great relevance for functional benefits.

The main active constituents in turmeric roots are curcuminoids, consisting of curcumin, DMC and BDMC. When isolated from dried turmeric rhizomes by typical solvent extraction process, these molecules are generally obtained as a complex (commonly referred to as ‘curcumin’) of no less than 95% purity, with 70–80% (w/w) curcumin as most abundant, 12–15% (w/w) DMC and 2–5% (w/w) BDMC(Reference Pancholi32). Following oral administration of unformulated curcumin, most of the ingested curcumin remains unabsorbed due to its hydrophobicity and insolubility as evidenced by 1% bioavailability compared with intraperitoneal administration, with the remaining fraction undergoing rapid intestinal/hepatic metabolism to conjugated metabolites such as glucuronides and sulphates (Fig. 1 (Reference Kurita and Makino19,Reference Fança-Berthon33,Reference Prasad34) ). Intravenous or intraperitoneal curcumin administration leads mainly to enzymatically reduced curcumin metabolites such as di-, tetra-, hexa- and octa-hydrocurcumin(Reference Xu11,Reference Prasad, Tyagi and Aggarwal35) . Curcumin metabolites have a limited contribution to the observed biological effects of curcumin, especially the conjugated metabolites. Glucuronides and sulphates are bulky water-soluble molecules which undergo rapid renal elimination and possess low cellular permeability. While some of the enzymatic metabolites and rapid degradation products of curcumin (tetrahydrocurcumin, ferulic acid or vanillin) may partially account for the observed activity of curcumin(Reference Pandey36Reference Shoji38), the majority of its conjugated metabolites have shown significantly weaker relative absorption, tissue permeability, blood–brain barrier permeability and anti-inflammatory, antioxidant and anti-proliferative activities(Reference Sandur27,Reference Pal28,Reference Prasad34,Reference Shoji38) . For example, curcumin monoglucuronide, a major conjugated metabolite of curcumin, has shown ten-fold lower antioxidant activity(Reference Choudhury39), anti-inflammatory(Reference Sandur27) properties and no anti-proliferative effects in comparison with ‘free’ curcumin(Reference Pal28,Reference Pal40) .

First-generation techniques to overcome curcumin metabolism and glucuronidation issues to enhance its oral bioavailability included the use of inhibitors such as piperine (black pepper extract) and permeability enhancers such as turmeric oils or turmerones(Reference Pancholi32). Both piperine and turmeric oil have been shown to boost curcumin bioavailability primarily by inhibiting UDP-glucuronyltransferase, glucuronidase/sulfatase, cytochrome P450, hepatic aryl hydrocarbon hydroxylase and mixed-function oxygenases(Reference Volak41,Reference Srinivasan42) . These are essential endogenous drug metabolism and detoxification enzymes, and blocking their activity may compromise the body’s defence and immunity. Yet another mechanism of these adjuvants was an expected increase in the intestinal permeability of drugs, which also renders toxins into systemic circulation. Furthermore, piperine has been shown to have reproductive toxicity and embryotoxic effects when administered as a high bolus dose in animals(Reference Ziegenhagen43). Subsequent approaches to enhance the curcuminoids solubility included the use of synthetic emulsifiers in nano forms. These second-generation formulations were reported to enhance the plasma curcuminoids levels, but mainly as their conjugated metabolites(Reference Pancholi32). Thus, there were safety concerns with the adjuvant-based methods to enhance bioavailability, and the existing methods to enhance solubility were only increasing the levels of conjugated metabolites without increasing the bioactive ‘free’ forms and would not guarantee an improvement in clinical efficacy. Regarded as a third-generation bioavailable curcumin, CGM formulation reportedly addresses both the bioavailability and cellular uptake issues of ‘free’ curcuminoids.

Technical challenges in the assessment of ‘free’ curcumin bioavailability

Aside from differences in study populations, analytical instruments and dosage regimens, some of the key issues contributing to the high heterogeneity of curcumin bioavailability reported in various studies stem from the different methodologies used to assess pharmacokinetics. A common practice in most pharmacokinetic studies is that the blood samples are subjected to hydrolysis with β-glucuronidase and sulfatase enzymes that convert the predominantly circulating forms of conjugated curcumin glucuronides and sulphates into ‘free’ curcuminoids prior to quantification of plasma curcuminoids. Thus, the true amount of ‘free’ bioactive curcumin cannot be quantified and the observed bioavailability is mainly that of the artificially un-conjugated curcumin metabolites, which may lead to misrepresentation of the formulation biological effects since there are significant differences in the activity of ‘free’ curcuminoids versus conjugated metabolites(Reference Stohs44,Reference Liju45) .

Another key issue is reporting enhanced bioavailability of a curcumin formulation by calculating the cumulative area under the curve (AUC) of all native curcuminoids: curcumin, DMC and BDMC in the unformulated and in the formulation following enzymatic hydrolysis. The standard unformulated curcumin has significantly lower amounts of DMC and BDMC, possibly even undetectable in plasma, while in formulations with enhanced absorption these additional curcuminoids can be detected as their glucuronide and sulphate conjugated forms. Thus, enzymatic hydrolysis of plasma samples results in an increase in total curcuminoids concentration or number of folds of bioavailability but does not typically represent the true bioavailability of the bioactive ‘free’ curcuminoids(Reference Szymusiak46,Reference Krishnakumar, Maliakel, Gopakumar, Kumar, Maliakel and Kuttan47) .

Curcumin bioavailability is also estimated by calculating maximum plasma concentration per mg (C max/mg) and total area under plasma concentration curve per mg (AUC/mg) of administered formulation after enzymatic hydrolysis(Reference Stohs44). A higher ratio is regarded as better bioavailability but is not a good estimation of the ‘true’ bioavailability, since formulations containing low curcumin content usually yield high ratios(Reference Douglass and Clouatre48). Furthermore, because these calculations use total plasma concentration of the sum of both free and conjugated curcumin metabolites, they cannot determine the bioavailability of ‘free’ curcuminoids.

Another potential source of errors is the use of the more limited and less sensitive HPLC techniques with ultraviolet/visible or fluorescence detectors instead of more sensitive techniques such as ultraperformance liquid chromatography electrospray ionisation triple quadrupole tandem mass spectrometry (UPLC–QQQ–ESI–MS/MS). To address these issues, in their pharmacokinetic study evaluating the bioavailability of CGM, Kumar et al. employed the gold standard UPLC–QQQ–ESI–MS/MS technique and measured the ‘free curcuminoids ratio’ (FCR)(Reference Kumar30). FCR is a direct measure of the ‘free’ curcuminoids in circulation versus curcumin conjugates and gives the fraction of the absorbed curcumin in the bioactive ‘free’ form and the fraction absorbed as conjugated metabolites after a single dose. Higher FCR ratio is correlated with better bioavailability of the ‘free’ curcuminoids.

It is not possible to compare the ‘free’ curcuminoids bioavalability of CGM with other bioavailable curcumin formulations because to our knowledge, a pharmacokinetic study with such parallel testing has not been reported. However, several studies that have investigated oral administration of unformulated curcumin and other bioavailable curcumin formulations, yielded insignificant plasma levels and tissue distribution of the physiologically relevant ‘free’ curcuminoids, or employed the enzymatic treatment of samples so the observed bioavailability enhancement would be mainly that of the conjugated metabolites, such as glucuronides and sulphates (Reference Lao1,Reference Garcea31,Reference Jager49Reference Schiborr51) . Overall, CGM is distinct from other bioavailable curcumin formulations due to its all-natural food-grade status and water-based manufacturing process without synthetic emulsifiers, excipients or processing aids.

CGM studies

This section discusses the fourteen pre-clinical and twelve human clinical trials that have evaluated the bioavailability, safety and the effects of CGM formulation on cognitive, cardiovascular and hepatic function, as well as in various inflammation-driven conditions (Fig. 2). Tables 1 and 2 summarise the pre-clinical and clinical studies with CGM that are discussed in detail below.

Fig. 2. CGM results in higher bioavailability and tissue distribution of free curcuminoids with improved effects on inflammatory, oxidative stress and endogenous antioxidant pathways in multiple organ systems. AChE, acetylcholine esterase; CAT, catalase; CRP, C-reactive protein; Dopa, dopamine; Glu, glutamate; GPx, glutathione peroxidase; GSH, Reduced Glutathione; HDL, High density lipoprotein; IL-1β, interleukin-1β; IL-6, interleukin-6; i-NOS, inducible nitric oxide synthase; MMP-2 & 9, matrix metalloproteinase-2 & 9; NF-κB, nuclear factor kappa B; Ser, serotonin; sVCAM, solublevascular cell adhesion molecule; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor α.

Table 1. Pre-clinical studies

* Abbreviations: AA, adjuvant induced arthritic control; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; Cat, catalase; CGM, curcumaglactomannosides; GGT, gamma-glutamyl transferase; CHN, chondroitin sulphate; GLN, glucosamine hydrochloride; GGT, gamma-glutamyl transferase; GSH, reduced glutathione; LPS, lipopolysaccharide; MS, multiple sclerosis; RC, radiation control; PP, pulse pressure; SOD, superoxide dismutase; SS, sulfasalazine; UC, ulcerative colitis; USC, unformulated standard curcumin.

Table 2. Clinical studies

* Abbreviations: AC, active-controlled; ALP, alkaline phosphatase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; BBB, blood–brain barrier; BID, two times per day; BMI, body mass index; BP, blood pressure; BPP, brachial pulse pressure determined by subtracting diastolic BP from systolic; BUN, blood urea nitrogen; Cat, catalase; CGM, curcumaglactomannosides; GSH, reduced glutathione; CHN, chondroitin sulphate; CO, crossover; DB, double blinded; EEEG, electroencephalography; FEN, fenugreek galactomannan fibre; GGT, gamma-glutamyl transferase; GLN, glucosamine hydrochloride; KPS, Karnofsky Performance Scale; LDL, low-density lipoproteins; ND, not detected; OA, osteoarthritis; OL, open label; PC, placebo controlled; PP, pulse pressure; RC, radiation control; RCT, randomised clinical trial; SA, single arm; SOD, superoxide dismutase; USC, unformulated standard curcumin; QoL, quality of life; VAS, visual analogue scale; WOMAC, Western Ontario and McMaster Universities Arthritis Index; TC, total cholesterol; UC, ulcerative colitis; SS, sulfasalazine.

Pharmacokinetics

Absorption and tissue distribution of significant amounts of bioactive forms at the target site are crucial for functional benefits. Begum et al. indicated an optimal tissue concentration of 1–2 µM of ‘free’ curcumin to elicit any favourable biochemical transformations in vivo (Reference Begum26). Krishnakumar et al. showed a 20-fold increase in curcumin bioavailability in rats administered with 250 mg/kg body weight (b.w.) CGM by oral gavage and a 12·9-fold and 15·8-fold increase in bioavailability at the 250 mg and 1500 mg CGM dose in humans compared with unformulated curcumin dosage of 1000 mg(Reference Krishnakumar, Dinesh Kumar, Kuttan and Maliakel20). HPLC method of estimation of plasma curcumin content, the small sample size of only eight male subjects, and the different equivalent curcuminoids content in the administered doses were the major limitations of this pilot study.

In a subsequent study using triple quadruple tandem mass spectrometry, Krishmakumar et al. showed that rats administered CGM at 200 mg/kg body weight (b.w.) curcumin content by oral gavage had twenty-five-fold increased plasma bioavailability of ‘free’ curcuminoids with a C max of 341·57 ± 30·88 ng/ml and an elimination half-life of 3·7 h compared with the equivalent amount of unformulated standard curcumin(Reference Krishnakumar, Maliakel, Gopakumar, Kumar, Maliakel and Kuttan47). CGM led to significant uptake of ‘free’ curcuminoids into various tissues, with 12- to 347-fold higher levels of the native unconjugated curcuminoids (sum of curcumin, DMC and BDMC) in the brain, liver, kidney, heart and spleen tissue for a longer duration of time (5 h versus 30 min) compared with standard curcumin, which showed curcumin at a concentration of only 1·4 ± 0·8 ng/g of brain tissues(Reference Krishnakumar, Maliakel, Gopakumar, Kumar, Maliakel and Kuttan47). However, this study did not attempt to investigate the regional distribution of ‘free’ curcuminoids in brain and further the improved brain tissue uptake was not correlated with any functional assessment. Another caveat was that only male rats were used in this single dose intake study.

Subsequently Kannan and colleagues addressed some of the caveats from these previous studies by evaluating the blood–brain barrier permeability, brain regional distribution of ‘free’ curcuminoids using UPLC–ESI–QQQ–MS/MS method concomitant with cognitive testing with single and repeated doses of CGM in rats. The authors showed that both single and 28 d repeated CGM administration at 100 mg/kg b.w. curcuminoid content yielded significant amounts of ‘free’ curcuminoids in brain, especially in the hippocampus. Correlated with the increased levels of ‘free’ curcuminoids measured in the brain, the animals in the CGM group also showed improvements in behavioural tests, locomotor activity and spatial memory errors that were significantly better compared with the unformulated curcumin group administered equivalent amount of curcuminoids for 28 d, p < 0·05(Reference Kannan52). A limitation of this study was that the authors did not measure regional brain tissue distribution of ‘free’ curcumioids with unformulated curcumin intake but only used equivalent curcuminoids intake in the functional behavioural testing part of the study.

Several studies report better pharmacological effects and enhanced permeability of ‘free’ curcuminoids as compared with their metabolites(Reference Begum26Reference Ireson29,Reference Szymusiak46,Reference Jager49,Reference Pan, Huang and Lin53) . Significantly increased bioavailability of ‘free’ curcuminoids with CGM administration has also been demonstrated in clinical studies. Pharmacokinetics of ‘free’ curcuminoids following the oral administration of high (1000 mg) and low (250 mg) doses of CGM and an equivalent dose of unformulated curcuminoids was assessed in a large, randomised, double-blinded human trial with fifty healthy volunteers using UPLC–ESI–QQQ–MS/MS. In this study, plasma levels of ‘free’ curcuminoids were measured without enzymatic pre-treatment with glucuronidase/sulfatase that converts conjugated curcumin metabolites to ‘free’ curcumin(Reference Kumar30). The high dose of CGM provided over 45·5-fold increase in ‘free’ curcuminoid bioavailability (24·8-fold for the low-dose), with significant improvement in all the pharmacokinetic parameters compared with unformulated standard curcumin which did not yield detectable levels of plasma curcuminoids at the low dose. Further comparison of plasma with and without enzymatic treatment revealed significantly higher levels of ‘free’ curcuminoids in plasma (74 ± 8%) as compared with conjugated forms of curcuminoids (26 ± 12%). The improved pharmacokinetic parameters, such as 440 ng/ml C max versus 13 ng/ml with unformulated curcumin and more than 3 h of elimination half-life, can lead to increased absorption and in vivo stability for cellular uptake, as well as favourable pharmacological effects via interaction with intracellular components(Reference Kumar30). A weakness of this study was that the relative distribution of ‘free’ and conjugated curcuminoids in plasma following the oral administration of CGM was not compared in parallel with administration of equivalent amounts of unformulated curcumin.

Pandaran et al. investigated the pharmacokinetics of ‘free’ curcuminoids following CGM administration of 500 mg as a single dose and 500 mg twice per day as a repeated dose for 30 d in healthy volunteers(Reference Pandaran Sudheeran54). Significantly higher relative absorptions and improved pharmacokinetics of total ‘free’ curcuminoids were observed with administration of CGM both as single dose (30·7-fold, p < 0·001) and repeated dose (39·1-fold, p < 0·001) compared with the unformulated curcumin. There was a significantly higher absorption (C max) and bioavailability with the repeated-dose regimen but no accumulation of plasma curcuminoids. The plasma concentration timeline followed the absorption, distribution and elimination phases, indicating its safe pharmacokinetics(Reference Pandaran Sudheeran54).

Liju et al. also reported the improved bioavailability of CGM from various food matrices (honey, soups, chocolate, yogurt) in healthy volunteers(Reference Liju45). In their study CGM was seven to ten times more bioavailable when incorporated into various foods or beverages at 100 mg CGM per serving size, as compared with unformulated curcumin. There was a good compatibility of CGM and no sensory issues in the various foods tested(Reference Liju45).

Safety

Recently, a few cases of hepatotoxicity have been reported among some long-term consumers of curcumin. Pancholi et al. reviewed the various related adverse events reported so far and speculated that added adjuvants known to inhibit essential detoxification pathways, adulteration with synthetic curcumin, or contaminations with heavy metals, chromate, illegal dyes, mycotoxins, non-steroidal anti-inflammatory agents, polyaromatic hydrocarbons and pyrrole alkaloids could be the main potential causes of the observed hepatotoxicity(Reference Pancholi32). Interestingly, among the reported cases of hepatotoxicity, twelve were women and only three were men. Moreover, the subjects reported with toxic manifestations were older (mean 55+ years) individuals under polypharmacy. Currently there are only a few safe, all-natural, food-grade curcumin formulations that have been shown to provide significant levels of bioactive ‘free’ curcuminoids in plasma and tissues(Reference Jamwal55,Reference Dei Cas and Ghidoni56) .

Pancholi and colleagues evaluated the safety and tolerance of long-term oral administration of CGM in healthy volunteers. Daily supplementation of 1000 mg CGM for 90 d did not cause any adverse effects or any clinically meaningful variations in the vital signs, haematological parameters, lipid profile and renal function markers of the volunteers. Liver function enzymes and bilirubin also stayed within the normal ranges after 90-d CGM supplementation(Reference Pancholi32). Some caveats with this study were the relatively younger study population (mean 31·32 years) that were healthy and not undergoing any pharmacological treatments. Considering the reported hepatotoxicity studies, it would be important to investigate the safety of long-term CGM intake in older subjects.

In animals, acute and chronic administration of CGM showed a no-observed-adverse-effect level (NOAEL) of 2000 mg/kg b.w. Furthermore, Ames test indicated no mutagenic character for CGM in concentrations of up to 5 mg per plate(Reference Liju45). Toxicology studies together with the pharmacokinetic and several clinical trials support CGM formulation safety for regular human consumption in doses of up to 1000 mg/d.

Neuroprotection

Neuroinflammation and abnormal protein aggregates are characteristic features of many neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease. Multiple studies report that curcumin administration can protect from neuroinflammation, β-amyloid and tau aggregates(Reference Maiti and Dunbar57Reference Reddy61).

On the basis of the observed improved blood–brain barrier permeability and brain distribution of bioactive ‘free’ curcuminoids, CGM has been tested for neuroprotective and cognitive benefits in animal models(Reference Krishnakumar, Maliakel, Gopakumar, Kumar, Maliakel and Kuttan47). Sunny et al. investigated the neuroprotective efficacy of CGM versus unformulated standard curcumin (USC) in a rat model of lipopolysaccharide (LPS)-induced neuroinflammation and neurotoxicity(Reference Sunny, Ramalingam, Das, Maliakel, Krishnakumar and Ittiyavirah62). LPS administration causes a detrimental increase in neuro-inflammation, acetylcholinesterase (AChE) activity, stimulation of serotonergic and glutamatergic neurotransmission, and reduced levels of dopamine in the brain leading to cognitive impairments as seen in neurodegenerative disorders. Oral administration of 200 mg/kg b.w. CGM for 28 d showed significant cognitive improvement and reduced inflammation in the brain compared with unformulated curcumin: 80% increase in number of entries in EPM test and 62·71% increase in the time spent (from 4·5% to 12·07%) in CGM compared with LPS group (p < 0·001) and 93·3% improved Y-maze behaviour in CGM versus LPS group. Unformulated standard curcumin (USC) group also showed improvements in the cognitive behavioural studies, but the percentage difference in reference, working memory errors and Y-maze test were 33·39%, 39·63% and 23·3% less respectively, as compared with CGM. CGM and USC significantly reduced AChE activity by 25% and 14%, respectively CGM reduced brain levels of glutamate from 3·17 ± 0·11 to and 2·35 ± 0·087, p < 0·001. CGM showed a 50% increase tissue levels of serotonin and dopamine that were depleted by LPS treatment, while the USC group showed a 10% relative increase. Furthermore, LPS-induced up-regulation of NF-κB, a key transcription factor regulating immunity and inflammation, was significantly down-regulated with CGM treatment. Histopathological analysis of the brain tissue showed significant reduction in inflammation, normal astrocyte morphology and minimal oedema and necrosis in CGM group, while USC also showed reduced oedema but enlarged astrocytes still present(Reference Sunny, Ramalingam, Das, Maliakel, Krishnakumar and Ittiyavirah62). It was unclear from the study design whether animals were administered multiple doses of LPS (after 7 and 14 d supplementation with CGM or USC) before the behavioural testing and whether the animals continued treatment for 28 d. With repeated injection of LPS there is also an increase in β-amyloid toxicity which is a hallmark of Alzheimer’s disease(Reference Lee63). Future studies are needed to determine CGM’s effect on Aβ levels or α/β-secretase activity. Another caveat of the study may be the lack of analysis of brain tissue lysates from different cortical and hippocampal regions.

In a mouse model of cuprizone-induced multiple sclerosis, cuprizone treatment leads to decrease in body weight, oligodendrocyte death and subsequent reversible demyelination(Reference Ittiyavirah64,Reference Torkildsen65) . Oral administration of 200 and 400 mg/kg b.w. CGM or unformulated curcumin significantly prevented the body weight loss induced by cuprizone treatment, in such a way that the low-dose CGM was similarly effective as the high-dose USC: average body weight 19·76 ± 0·2 g in 200 mg/kg in CGM group compared with 18·11 ± 0·2 g in 400 mg/kg USC group, and 11·35 ± 0·17 g in cuprizone-treated animals, (p < 0·01)(Reference Ittiyavirah64). Histopathology analysis showed less demyelinated lesions in the high-dose CGM group compared with unformulated curcumin- and cuprizone-treated groups. A major limitation of this study was the lack of quantification of demyelination and specific immunohistochemical myelin protein staining in the histology analysis.

Another recent publication compared the effect of CGM and unformulated curcumin on carbofuran (CF)-induced neurotoxicity in rats. CF is a carbamate pesticide toxic to neurons involved in locomotor function(Reference Sindhu, Binitha, Nair, Maliakel, Ramadasan and Krishnakumar66). Rotarod experiments show significant decline in muscle coordination and strength in CF-treated animals including 19 s reduction in retention time on the rotarod (p < 0·01), 22 s reduction in grip strength (p < 0·05) and forty-fold increase in time to elicit pain response (p < 0·001). Both unformulated curcumin and CGM showed improvement in muscle strength; however, the CGM effect was significantly higher and almost reversed the motor deficit to the normal control level: CGM reverted grip strength similar to normal levels and decreased the pain threshold by 26 s (p < 0·001) compared with 5 s improved grip strength and maximum 10 s pain threshold decrease with the same dose of unformulated curcumin. Similarly, there was a significant improvement in CF-induced increase in the time to elicit pain response in the CGM-treated group compared with unformulated curcumin(Reference Sindhu, Binitha, Nair, Maliakel, Ramadasan and Krishnakumar66). Interestingly, CGM treatment also improved other aspects of CF-induced toxicity, such as reduced oxidative stress parameters and enhanced mitochondrial function with low dose of CGM comparable to high-dose USC and high dose of 250 mg CGM closer to normal controls (p > 0·05). Some limitations with this study include the use of only male rats and lack of assessment of the number and morphology of mitochondria in the brain(Reference Sindhu, Binitha, Nair, Maliakel, Ramadasan and Krishnakumar66). In summary, three different chemically induced neurotoxic animal models CGM showed superior functional benefits, as well as higher improvement in neurotransmitter levels, inflammatory markers, mitochondrial function and tissue histopathology as compared with unformulated curcumin. The observed benefits can be attributed to the better brain bioavailability and pharmacokinetics of ‘free’ curcuminoids with CGM(Reference Sandur27,Reference Krishnakumar, Maliakel, Gopakumar, Kumar, Maliakel and Kuttan47) .

In a randomised placebo-controlled clinical trial, Pandaran et al. investigated the effect of CGM or unformulated curcumin supplementation on endogenous oxidative stress biomarkers and quality of life in healthy subjects experiencing occupational stress-related anxiety and fatigue(Reference Pandaran Sudheeran54). Occupational stress and anxiety have been shown to increase formation of highly reactive free radicals and deplete levels of endogenous antioxidants with a negative impact on health(Reference Fedoce67,Reference Lobo68) . Repeated supplementation with 500 mg CGM twice per day for 30 d significantly increased plasma levels of antioxidant enzymes (superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase (CAT), reduced glutathione (GSH)) and reduced lipid peroxidation. Changes in serum biomarkers correlated with significant improvements among 78% of subjects in perceived stress, anxiety and physical/mental fatigue scores and in quality of life. The enhanced efficacy with CGM supplementation versus unformulated curcumin was attributed to better antioxidant status and hence the modulation of oxidative stress with higher amounts of ‘free’ curcuminoids such as 30·7-fold higher bioavailability of ‘free’ curcuminoids with single dose of 500 mg CGM and 39·1-fold increase with 30-d repeated-dose CGM(Reference Pandaran Sudheeran54). The study did not show any adverse events, side effects or deviations in clinical laboratory parameters indicating its safety.

In another randomised, double-blinded, placebo-controlled clinical trial, Khanna and colleagues investigated the effects of CGM administration on brain waves in healthy volunteers(Reference Khanna69). Changes in brain waves measured by electroencephalogram have been used as an index of blood–brain barrier permeability and bioavailability of drugs or nutrients(Reference Bewernitz and Derendorf70). In this study, 500 mg of CGM twice daily for 30 d showed significant increases in α- and β-waves as compared with unformulated curcumin and placebo groups. Previous clinical studies have shown that increases in α-waves correlate with better cognitive performance(Reference Klimesch, Schimke and Pfurtscheller71), while increases in β-waves were associated with higher alertness/arousal and arithmetic calculation ability(Reference Fernandez72,Reference Lal and Craig73) . Studies show that α- and β-wave generation reflects cholinergic modulation in various areas of the brain. CGM’s effect on these waves together with modulation of the cholinergic system via AChE indicates CGM’s capacity to influence attention, learning and memory(Reference Sarlak, Oryan and Moghaddasi74,Reference Roopun75) . CGM group showed a 36% improvement in choice-based visual-reaction time compared with 15·36% in unformulated curcumin and 5·2% in placebo groups. The CGM-supplemented group also showed significant (29·8%) reduction versus placebo in α/β ratio, a validated indicator of fatigue/tiredness(Reference Tiago-Costa, Quelhas-Costa and Santos-Baptista76). A major limitation of the study was the relatively small number of just eighteen subjects, though the results were consistent with a previous larger clinical study in sixty subjects with occupational stress, indicating CGM’s ability to influence brain waves is consistent with brain penetration and several potential cognitive benefits including audio-visual and working memory improvements, as well as stress and fatigue reduction(Reference Pandaran Sudheeran54).

Promising results from an unpublished, randomised, placebo-controlled clinical trial in forty-eight patients with Alzheimer’s disease suffering from moderate dementia, indicate that 400 mg CGM supplementation twice daily for 180 d significantly improved cognitive and locomotor function (assessed by Mini-Mental State Examination and Geriatric Locomotive Function Scale score) compared with unformulated curcumin and placebo groups. Functional benefits were correlated with improvements in plasma levels of β-amyloid 42, Tau proteins, antioxidant markers SOD and GSH, and inflammatory markers IL-6, IL-1β, TNF-α and BDNF (unpublished results, R&D Internal Report 06/2021, Akay Natural Ingredients, Cochin, India). As in previous studies, the authors attribute the benefits observed with CGM to the improved blood–brain barrier permeability, brain bioavailability and brain tissue distribution of ‘free’ curcuminoids in this formulation. However, further larger clinical studies are warranted to confirm these results and to investigate the effects of long-term supplementation in various populations, including paediatrics and geriatrics.

Cardiovascular effects

Oxidative stress-induced intracellular redox imbalance plays a pivotal role in the pathogenesis of various cardiometabolic risk factors (hyperlipidaemia, hypertriglyceridemia, hyperglycemia, hypertension, obesity) leading to severe cardiovascular diseases. Enhancing the endogenous antioxidant defence mechanisms (enzymatic and non-enzymatic) with antioxidant supplementation has been shown to significantly reduce cardiometabolic risk factors(Reference Yin77). Due to its specific chemical structure, when provided at an effective dose and not compromised by its poor bioavailability, curcumin has substantial free radical scavenging and anti-atherosclerosis activities, improving several cardiovascular biomarkers such as homocysteine, C-reactive proteins, inflammatory cytokines, cholesterol, triglycerides, blood pressure and glycemia(Reference Li78Reference Jin81). CGM also demonstrated a strong anti-inflammatory effect against oxidised LDL (ox-LDL)-induced inflammatory responses in vitro in human peripheral blood mononuclear cells with 85% inhibition of COX activity (12·5 μg/ml CGM), down-regulated reactive oxygen species, TNF-α, IL-6, INOS and VCAM-1 mRNA, and inhibited nuclear translocation of NF-kB and lipid peroxidation (p < 0·05)(Reference Saji82).

High levels of homocysteine and altered lipid metabolism are correlated with an increased risk of cardiovascular disease, heart attacks and strokes(Reference Baszczuk83). Lipid peroxidation is another important step in the pathogenesis of atherosclerosis(Reference Esterbauer, Wag and Puhl84). In two clinical trials, CGM supplementation has significantly reduced lipid peroxidation and enhanced levels of glutathione and superoxide dismutase(Reference Pandaran Sudheeran54,Reference Krishnareddy85) . The first study in young obese men receiving daily supplementation with 500 mg CGM for 12 weeks, without any dietary restrictions and exercise, showed a significant increase in HDL-cholesterol by 34% and reduction in homocysteine levels by 29% from 12·22 ± 2·29 µg/ml before to 8·62 ± 1·02 µg/ml compared with the placebo group, who saw an increase from 9·45 ± 0·84 mg/ml to 11·84 ± 1·63 mg/ml at the end of study, p = 0·04(Reference Campbell86).

In another clinical study in a similar population of healthy obese men, daily supplementation of 500 mg CGM with no exercise or dietary modifications led to significant improvement in arterial stiffness in the ‘responder’ subgroup with higher aortic stiffness at baseline. The CGM responder group (6/11 subjects) showed a higher effect on carotid–femoral pulse wave velocity compared with non-responders with lower levels at baseline (6·81 ± 0·83 m/s versus 5·84 ± 0·41 m/s, p = 0·045)(Reference Campbell87). Even small increases in aortic stiffness, as measured by 1·0 m/s increase in carotid–femoral pulse wave velocity, has been associated with a 14% increase in cardiovascular events, 15% increase in cardiovascular mortality and 15% increase in all-cause mortality(Reference Vlachopoulos, Aznaouridis and Stefanadis88). Thus, the 1 m/s relaxation observed in the CGM responder subgroup is a clinically meaningful improvement. The authors suggest that the change in brachial pulse pressure observed in the CGM group was likely due to functional rather than structural changes in the artery, as structural changes would have been expected to cause an overall change in the carotid–femoral pulse wave velocity. The CGM responders also had a significant increase in the plasma levels of the anti-inflammatory cytokine IL-13. Previous work suggested these cytokines may be protective against cardiovascular pathologies via vasoactive properties and modulation of collagen(Reference Tang89,Reference Cardilo-Reis90) . The observed beneficial effect of CGM on several cardiovascular markers indicates its potential in the maintenance or improvement of cardiovascular health in younger obese individuals; however, further larger and longer studies in older populations are warranted. Studies in combination with dietary restrictions and exercise may provide a better understanding of the possible role of CGM to maintain healthy arteries or ‘de-stiffen’ arteries.

An unpublished clinical study showed promising results of CGM supplementation in diabetic patients on metformin treatment. In this study, 400 mg CGM supplementation daily led to a significant decrease in the postprandial blood glucose spike, significant reduction in diabetes-related side effects such as thirst, dry mouth and fatigue, and improvement in quality of life (unpublished results, R&D Internal Report 08/2021, Akay Natural Ingredients, Cochin, India).

Hepatoprotection

Many natural treatments for liver dysfunction target the overactivated inflammatory and oxidative stress cascade(Reference Hong91). Curcumin has multiple therapeutically relevant molecular targets in the liver, serving as a powerful antioxidant against free-radical damage(Reference Bielak-Zmijewska92) as well as anti-inflammatory agent via inhibition of NF-κB(Reference Saji82,Reference Sandur93Reference Deguchi95) and reduction of pro-inflammatory cytokines(Reference Tasneem96,Reference Chin97) . In a previously mentioned clinical trial, CGM treatment significantly increased endogenous antioxidant enzymes and hence the detoxification capacity in subjects with occupational stress(Reference Pandaran Sudheeran54).

In an animal model of alcohol-induced liver damage, 250 mg/kg b.w. CGM significantly reversed the detrimental effects of ethanol on liver enzymes, lipid peroxidation, inflammation and endogenous antioxidants(Reference Mohan98). Chronic alcohol consumption has been shown to induce oxidative stress-mediated inflammation along with up-regulation of NF-kB, TNF-α, IL-6, IL-1β and CRP and activation of matrix metalloproteases (MMP-2 and MMP-9) and toll-like receptors (TLR4), which are involved in the pathogenesis of alcoholic liver disease via degradation of extracellular matrix and activation of immune responses, respectively(Reference Banerjee99,Reference Gustot100) . Both CGM and standard curcumin treatment significantly down-regulated the ethanol-induced increased levels of TNF-α and IL-6, supporting the anti-inflammatory role of curcumin. Ethanol-induced up-regulation of TLR4 and MMP-2 and MMP-9 were also significantly counterbalanced in both curcumin treatment groups. However, CGM formulation was superior to standard curcumin in regulating oxidative stress, liver function and inflammatory markers, as well as TLRs and MMPs expression almost to control levels, highlighting the therapeutic importance of higher bioavailability of ‘free’ curcuminoids with CGM. The enhanced protective effect of CGM was also evident from the improved histopathology observed of liver tissues of CGM-treated animals(Reference Krishnakumar, Maliakel, Gopakumar, Kumar, Maliakel and Kuttan47,Reference Mohan98) .

In a double-blinded placebo-controlled clinical trial, Krishnareddy et al. showed that 250 mg CGM supplemented twice per day for 56 d improved hepatic function in subjects with chronic alcohol consumption of more than six alcohol units per week (one unit is equivalent to 150 ml of wine or 360 ml of beer or 45 ml of 40% (v/v) alcohol) and elevated levels of two biomarkers of liver disease, serum transaminases and gamma-glutamyl transferase (GGT). Benefits were apparent from 28 d of treatment and led to an average 36% and 29% decrease in ALT and GGT transaminases, respectively, from baseline by the end of the study. There was also up to 25% increase in levels of endogenous antioxidant enzymes (GSH, SOD, and GPx) and a significant reduction in markers of systemic inflammation (IL-6 and CRP) as compared with both baseline and placebo groups(Reference Krishnareddy85). Interestingly, the subjects were not allowed to consume more than three units of alcohol per week for the duration of the study, but this 50% reduction in alcohol intake did not elicit improvements in liver function markers in the placebo group. Taken together, both pre-clinical and clinical studies show that CGM can potentially attenuate the alcohol-induced alterations in several biomarkers of liver function and offer hepatoprotective effects in younger males. It would be important to confirm and further investigate the effect of CGM supplementation in larger studies including women, as well as subjects with other chronic liver diseases such as non-alcoholic liver diseases.

Anti-inflammatory effects

The anti-inflammatory activity of curcumin has been well documented. The molecular mechanisms underlying this effect include inhibition of enzymes and factors that mediate pro-inflammatory pathways such as cyclooxygenases, lipoxygenases, inducible nitric oxide synthase and NF-κB(Reference Jurenka101Reference Menon and Sudheer103).

In an in vitro study, Saji et al. explored the anti-inflammatory effect of CGM against ox-LDL-induced inflammatory responses in human peripheral blood mononuclear cells (hPBMCs). Gene expression analysis showed that ox-LDL treatment increased expression of several pro-inflammatory biomarkers such as iNOS, TNF-α, IL-6, VCAM-1, LOX, PGE2, total COX and lipid peroxidation along with nuclear translocation of NF-κB. CGM treatment down-regulated the activation of these cell adhesion molecules and pro-inflammatory cytokines, indicating its potential anti-inflammatory effect via the NF-κB signalling pathway(Reference Saji82).

Sheethal et al. investigated the effect of CGM in an acetic-acid-induced animal model which mimics some of the features of ulcerative colitis disease(Reference Sheethal104). Ulcerative colitis is a chronic inflammatory bowel disease characterised by reduced antioxidant capacity and increased inflammation. In this experimental model CGM significantly increased endogenous antioxidant enzymes (SOD, GPx, CAT, normalised GSH and reduced myeloperoxidase (MPO) activity, p < 0·05) and decreased oxidative damage thiobarbituric acid reactive substances (TBARS) and the level of pro-inflammatory markers (total COX, iNOS, TLR4, TNF-a, IL-6, CRP) as compared with both sulfasalazine standard therapy and unformulated curcumin groups, restoring the heavily inflamed and damaged colon to almost normal levels with mild inflammation(Reference Sheethal104). A limitation of this study was that the experimental model does not provide the opportunity to evaluate both the acute and chronic mucosa repair mechanisms, so further studies with chronic exposure to different chemical agents such as dextran sulphate sodium or 2,4,6-trinitrobenzene sulfonic acid 34 may be more relevant to the complex human pathological phenotype of ulcerative colitis.

Saji et al. demonstrated the acute and chronic anti-inflammatory effects of CGM using a carrageenan-induced paw oedema acute model and an adjuvant induced arthritic model of chronic inflammation(Reference Sangeeth Saji, Jose, Ratheesh, Sandya and Asha105). In the carragenaan model of acute inflammation, 100 mg/kg b.w. CGM showed paw oedema inhibition of 82% compared with 72% with the unformulated standard curcumin. In the adjuvant induced chronic arthritic model, 30 d supplementation with 100 mg/kg b.w. CGM significantly increased the levels of antioxidant enzymes SOD and GPx, CAT and GSH compared with indomethacin, down-regulated several inflammatory biomarkers such as plasma CRP and COX2, lowered TNF-α and IL-6, and reduced the massive infiltration of polymorphonuclear cells contributing to soft tissue oedema. Further radiological analysis showed that the CGM-treated arthritic rats had significant improvement in the degenerative joint changes such as sub-chondral erosion and joint space narrowing(Reference Sangeeth Saji, Jose, Ratheesh, Sandya and Asha105).

Two clinical trials evaluated the anti-inflammatory potential of CGM treatment in subjects with symptomatic knee osteoarthritis and swelling in the knee joint(Reference Khanna106,Reference Thomas107) . Osteoarthritis is a degenerative disease that involves deterioration of the cartilage which acts as a cushion between the bones in a joint. This tissue damage triggers the inflammatory signalling cascade and overproduction of pro-inflammatory cytokines that cause swelling, pain and stiffness, and functional impairment of joints. Acetaminophen and non-steroidal anti-inflammatory drugs (NSAIDs) are part of the conventional therapy but are also typically associated with adverse gastrointestinal, renal and cardiovascular effects upon prolonged use or high doses. Natural components of cartilage, glucosamine and chondroitin are marketed as dietary supplements for joint support, but there are conflicting reports on their efficacy, including joint pain relief mainly due to their lack of anti-inflammatory efficacy, and typically require longer duration of treatment for the beneficial effect to be felt(Reference Zhu108Reference Clegg110).

In one of the randomised, double-blinded, active-controlled studies, Khanna et al. compared the efficacy of standard 500 mg glucosamine hydrochloride (GLN) plus 415 mg chondroitin sulphate (CHN) with 400 mg CGM plus 500 mg GLN administered twice daily in improving pain and quality of life in patients with confirmed osteoarthritis. The study used multiple validated questionnaires Visual Analog Scale (VAS), Karnofsky Performance Scale (KPS) and Western Ontario and McMaster Universities Arthritis Index (WOMAC) to evaluate pain, stiffness and physical activities along with the measurement of anti-inflammatory markers. After 4 weeks of supplementation (day 28) the CGM–GLN-treated group showed an 81% increase in treadmill performance, 32% reduction in WOMAC pain score, 17% reduction in stiffness score and 30% reduction in physical activity difficulty score compared with the CHN–GLN group, indicating relatively faster action of CGM. After 12 weeks at the end of the study, the CGM–GLN-treated group showed almost double the efficacy compared with CHN–GLN, with significant four-fold improvement in treadmill walking score (p < 0·001) versus CHN–GLN group (mean difference 301·35 ± 74·46 m from baseline, p < 0·001 in CGM–GLN versus mean difference of 89·78 ± 19·25 m from baseline in CHN–GLN), significant improvements in walking performance (mean difference of 24·19 ± 6·20 in CGM–GLN compared with 16·56 ± 7·45 in CHN–GLN group), pain intensity (60·46% reduction VAS in CGM–GLN, p < 0·001 and 21·25% in CHN–GLN from baseline), stiffness and physical function. There was also significant 54·52%, 59·08% and 22·03% reduction in serum inflammatory markers IL-1β, IL-6 and soluble vascular cell adhesion molecule-1 (sVCAM) from baseline compared with 23·17%, 21·38% and 6·82%, respectively, in the active-control group. Interestingly, the subjects in the CGM–GLN-supplemented group also showed a decrease in body weight which may be beneficial for reduction in pain(Reference Khanna106). Lack of a placebo group and information on dropout and responder rates were some of the major limitations of the study. Moreover, co-administration of CGM with GLN did not allow for estimation of the effect due to CGM supplementation.

In the second study in patients with osteoarthritis, Thomas et al. compared the safety and efficacy of CGM supplementation alone versus the standard GLN–CHN combination. The group administered 400 mg CGM per day for 42 d showed superior improvements compared with the active-control group in VAS, KPS and walking performance (47·02%, 21·43% and 206% in CGM compared with 24·67%, 23·84% and 85·69% in CHN–GLN, respectively) as well as pain, stiffness, physical function and WOMAC scores (31·17%, 32·93%, 46·44% and 35% in CGM compared with 26·62%, 25·97%, 12·56% and 16·46% in CHN–GLN, respectively) and reduction in IL-1b, IL-6 and sVCAM (54·52%, 59·08% and 22·03% in CGM compared with 23·17%, 21·38% and 6·82% in CHN–GLN, respectively). Furthermore, the number of subjects that required ‘rescue’ NSAIDs/analgesics during the study period was 31·43% in CGM compared with 52·35% in the CHN–GLN group. Again, a significant beneficial effect was observed from day 28 onwards. CGM supplementation was considered safe and did not cause any adverse effects in the subjects who completed the study(Reference Thomas107). While the study included several validated questionnaires to compare the pain, stiffness, physical activity and quality of life along with anti-inflammatory markers, the reported 15% dropout rate that was not further described in detail in the manuscript limits the enthusiasm. Another caveat was the relatively short 6-week duration of the study and the lack of a placebo group and double-blinded study design due to the different dosage levels in the two groups (400 mg CGM once daily versus 900 mg CHN–GLN twice per day).

It is worth mentioning that both of the above studies reported a reduction in body mass index (BMI) in CGM-treated obese individuals that was attributed mainly to the fenugreek fibre provided in the CGM formulation(Reference Khanna106,Reference Thomas107) . In addition, most clinical studies for the treatment of osteoarthritis use high doses of at least 1000 mg of curcumin per day(Reference Daily, Yang and Park111). Taking into account the negative side effects associated with chronic use of acetaminophen and NSAIDs for the management of pain and inflammation, a low daily dose of 400 mg of CGM warrants further investigations in larger double-blinded, placebo-controlled clinical trials as a potentially safer alternative in osteoarthritis and body weight management.

Aerobic performance

In a randomised placebo-controlled study, Goh et al. examined the effects of CGM on aerobic performance in healthy untrained young volunteers with no regular exercise or dietary change. Participants completed a maximal graded exercise on a cycle ergometer before and after 28 d of daily supplementation with either 500 mg CGM, equivalent 300 mg fenugreek soluble fibre (FEN), or placebo. This test is commonly used to determine submaximal endurance performance and maximal endurance performance as measured by the ventilatory threshold (VT) and peak oxygen consumption (VO2 peak), respectively. VT increased 6·2% (increase 0·094 litres/min) and 6·7% (increase 0·099 litres/min) from baseline for CGM and FEN, respectively, and decreased in the placebo group (−2·2%, decrease 0·034 litres/min). Only four of the eighteen participants in the FEN group and two of the fourteen participants in the CGM group showed a real change in the mean VT response. These results suggest that fenugreek galactomannan fibre is biologically active and can provide significant benefit even at a dose of 300 mg/d. The authors attributed these results to galactomannan mucilage fibre ability to improve insulin sensitivity and increase NEFA availability(Reference Goh112). Interestingly, increases of 6·2–6·7% in the VT in this study are consistent with 4·1–5·4% increases previously reported for the gas exchange threshold after 28 d of supplementation with 3 g/d arginine(Reference Camic113), which were also not accompanied by changes in maximal endurance. A limitation of the study was that authors did not measure any physiological markers aside from resting blood pressure, heart rate and patient self-reported medical history to confirm the metabolic status of the participants. Further, longer studies in trained athletes or including exercise and a higher-dose treatment arm are recommended to better understand the influence of CGM and fenugreek galactomannan fibre on peak oxygen consumption.

Other cellular effects

Antioxidant, anti-inflammatory and epigenetic regulation mechanisms modulated by curcumin are well documented(Reference Hassan114,Reference Sharifi-Rad115) . The potential effect of CGM as an epigenetic modulator has been recently evaluated in vivo. Using whole transcriptome analysis (RNA-seq), Banik et al. identified 559 differentially expressed genes in the liver tissue of mice with LPS-induced inflammation treated with CGM for 14 d versus untreated. Subsequent gene expression and pathway analysis showed that thirty-three genes which were highly up-regulated in disease conditions were significantly down-regulated by CGM, and thirty-two genes highly down-regulated in various disease conditions were significantly up-regulated following CGM treatment(Reference Banik116). These results suggest that CGM could restore expression patterns of genes up-regulated or down-regulated in many chronic disease conditions. As uncontrolled inflammation plays a major role in the development of various chronic diseases and most chronic diseases are caused by perturbations in multiple molecular pathways, the authors suggest that targeting a single gene/protein/pathway might not be sufficient in tackling these diseases. Interestingly, this study highlights the pleiotropic action of CGM and potential to modulate multiple cellular targets such as nucleotides and transcription factors that modulate gene expression and pathways known to be involved in the development of several chronic diseases(Reference Banik116). Some limitations of this study were the use of only female mice, single intraperitoneal injection of LPS that can cause an acute inflammatory response, and the examination of solely liver tissue. Further studies with repeated exposure to LPS to produce chronic systemic inflammation, evaluation of multiple organs and including male animals would provide better information on the systemic gene modulatory effects of CGM in chronic diseases.

Radiation therapy is an important treatment against various types of cancer that has unfortunately deleterious effects on both tumour as well as surrounding healthy cells. There are opposing views regarding the use of supplementation with antioxidants during radiation therapy, with some radiation oncologists advising against it for fear that cancer cells may also benefit and render radiation less effective, but antioxidants can reduce the side effects of radiation therapy in normal cells(Reference Prasad117). A recent study evaluated the radioprotective effects of CGM in an animal model of radiation-induced damage(Reference Liju118). Ionising gamma radiation significantly reduces leucocytes and bone marrow cells and increases formation of cytotoxic reactive oxygen and nitrogen species as well as genetic instability. CGM showed better protection against radiation-induced bone marrow damage compared with standard curcumin treatment by restoring monocyte alpha esterase activity and haemoglobin levels (50% reduction in percentage of radiation-induced micronuclei increase in bone marrow in CGM compared with only 34% in USC group and a 60% reduction in polychromatic to normochromic (P/N) erythrocyte ratio upon radiation recovered by 2·1 times in CGM group, p > 0·01 compared with 1·2 times in USC, p > 0·05). The CGM group also showed higher levels of antioxidant enzymes (21% reduced hepatic GPx activity compared with 25% in unformulated curcumin and 42% in radiation control group) and prevented the radiation-induced increase in lipid peroxidation and infiltration of inflammatory exudates in intestinal mucosa. Furthermore, CGM treatment reduced the degree of chromosomal defects and DNA damage and yielded a better survival rate compared with unformulated curcumin (4-fold reduction in DNA damage in CGM, p < 0·05 compared with 2·5-fold in USC group, p < 0·01 and 83% reduction in radiation-induced chromosomal aberration in CGM vs 66% in USC group, p < 0·05)(Reference Liju118). Depending on the type, size, location of cancer and/or goal of the radiation therapy, multiple consecutive treatments may be required (‘Targeted Therapy to Treat Cancer’, originally published by the National Cancer Institute, 30 March 2022); thus, the single exposure to radiation used in this study may be considered a limitation of its translatability to clinical trials.

A recent study shows the anti-tumour potential of CGM via effects on cell cycle and apoptosis in vitro. Exposure to 25 μg/ml CGM showed higher cytotoxic activity and inhibition of cancer cell survival in HeLa cells compared with the drug doxorubicin. CGM induced apoptosis and cell cycle arrest as shown by increased levels of Bax and cleaved caspase-8 protein in cancer cells(Reference Ratheesh119). Previous studies have shown that curcumin can selectively induce apoptosis and inhibit cell growth and invasion in cancer cells but not in normal cells, highlighting the potential of exploring CGM’s anti-cancer potential in human interventions(Reference Jiang120Reference Borek123).

Concluding remarks

CGM curcumin formulation with a proprietary fenugreek galactomannan dietary fibre called FENUMAT has consistently shown significantly higher bioavailability of ‘free’ curcuminoids in multiple animal models and human clinical studies when compared with equivalent doses of unformulated standard curcumin. In agreement with the pharmacokinetic studies, all the pre-clinical and clinical studies with CGM (brain, liver, heart and joint health) have also shown superior efficacy when compared with the unformulated standard curcumin, highlighting the therapeutic relevance of the improved bioavailability and tissue uptake of the ‘free’ curcuminoids. Furthermore, the whole genome transcriptome study provided the mechanistic evidence for CGM’s ability to carry out the pleiotropic effects by revealing the interaction of ‘free’ curcuminoids with various cellular targets leading to the regulation of several genes, including ones involved in the pathogenesis of cancers.

It has also been suggested that curcumin’s overall health benefits derive from its direct influence on the gastrointestinal microbiome(Reference Lopresti124,Reference Jabczyk125) . Interestingly, the fenugreek galactomannan used in the formulation of CGM is a soluble dietary fibre with significant prebiotic potential. It is also a functional dietary fibre with hypolipidaemic, hypoglycaemic and gastroprotective properties(Reference Vijayasteltar24). So, CGM would be expected to have higher impact on the microbiome and may bring additional benefits from galactomannan fibre compared with standard unformulated curcumin. Further clinical studies evaluating specific changes in gut microbiome and metabolites with long-term supplementation are required to confirm the effects of CGM on the microbiome.

Overall, scientific evidence on CGM curcumin addresses the issues of poor bioavailability and tissue distribution of the bioactive ‘free’ curcuminoids and demonstrates superior efficacy compared with standard unformulated curcumin. Clinical studies with the all-natural CGM curcumin formulation demonstrate efficacy at daily doses as low as 400 mg and safety at doses of up to 1000 mg/d, highlighting its potential in prevention and long-term management of different chronic diseases.

Acknowledgements

The authors would like to acknowledge Dr Balu Maliakel and Dr Shayna Sandhaus for manuscript review and valuable suggestions.

This research received no external funding.

C.M. and A.G.S. are employed by Life Extension company that sells CGM ingredient in dietary formulations, and I.M.K. works for Akay, which is a CGM ingredient manufacturer and supplier.

Author contributions

C.M. drafted the manuscript and created figures and the graphical abstract; C.M., I.M.K. and A.G.S. performed critical revision of the manuscript. All authors have read and agreed to the published version of the manuscript.

Author information

Cristina Matthewman is a Research Scientist at Life Extension Inc., Fort Lauderdale, Florida. She earned an MS in Biophysics and Medical Physics from University of Physics, Bucharest, Romania and a PhD in Neuroscience from University of Miami Miller School of Medicine, FL. Her current research interests are natural ingredients with health benefits. I. M. Krishnakumar is Chief Research Officer at Akay Natural Ingredients Ltd, Kerala, India. He earned a PhD in Chemistry from Mahathma Gandhi University, Kerala, India and received postdoctoral research fellowships from Houston Medical School, University of Texas, Houston and Burnham Institute of Medical Research, San Diego, CA. He is a bio-organic chemist interested in natural bioactive molecules, delivery forms and pharmacological effects. Andrew G. Swick is Chief Scientific Officer at Life Extension Inc., Fort Lauderdale, Florida. He earned an MS from the University of Nebraska, and a PhD from the University of Wisconsin and received postdoctoral fellowship from the University of North Carolina Lineberger Cancer Research Center and Johns Hopkins University School of Medicine. His current research interests focus on the use of nutraceuticals for disease prevention and maximisation of health span.

Institutional review board statement

Not applicable.

Informed consent statement

Not applicable.

Data availability statement

Not applicable.

References

Lao, CD, et al. (2006) Dose escalation of a curcuminoid formulation. BMC Complement Altern Med 6, 10.CrossRefGoogle ScholarPubMed
Salehi, B, et al. (2019) The therapeutic potential of curcumin: a review of clinical trials. Eur J Med Chem 163, 527545.CrossRefGoogle ScholarPubMed
Pagano, E, et al. (2018) The clinical efficacy of curcumin-containing nutraceuticals: an overview of systematic reviews. Pharmacol Res 134, 7991.CrossRefGoogle ScholarPubMed
Panahi, Y, et al. (2016) Mitigation of systemic oxidative stress by curcuminoids in osteoarthritis: results of a randomized controlled trial. J Diet Suppl 13, 209220.CrossRefGoogle ScholarPubMed
Al-Karawi, D, Al Mamoori, DA & Tayyar, Y (2016) The role of curcumin administration in patients with major depressive disorder: mini meta-analysis of clinical trials. Phytother Res 30, 175183.CrossRefGoogle ScholarPubMed
Panahi, Y, et al. (2017) Antioxidant effects of curcuminoids in patients with type 2 diabetes mellitus: a randomized controlled trial. Inflammopharmacology 25, 2531.CrossRefGoogle ScholarPubMed
Sahebkar, A & Henrotin, Y (2016) Analgesic efficacy and safety of curcuminoids in clinical practice: a systematic review and meta-analysis of randomized controlled trials. Pain Med 17, 11921202.Google ScholarPubMed
Qin, S, et al. (2017) 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 16, 68.CrossRefGoogle ScholarPubMed
Haroyan, A, et al. (2018) Efficacy and safety of curcumin and its combination with boswellic acid in osteoarthritis: a comparative, randomized, double-blind, placebo-controlled study. BMC Complement Altern Med 18, 7.CrossRefGoogle Scholar
Bahadori, F & Demiray, M (2017) A realistic view on “The Essential Medicinal Chemistry of Curcumin”. ACS Med Chem Lett 8, 893896.CrossRefGoogle Scholar
Xu, X-Y, et al. (2018) Bioactivity, health benefits, and related molecular mechanisms of curcumin: current progress, challenges, and perspectives. Nutrients 10, 1553.CrossRefGoogle ScholarPubMed
Goel, A, Kunnumakkara, AB & Aggarwal, BB (2008) Curcumin as “Curecumin”: from kitchen to clinic. Biochem Pharmacol 75, 787809.CrossRefGoogle ScholarPubMed
Sharifi-Rad, J, et al., (2020) Turmeric and its major compound curcumin on health: bioactive effects and safety profiles for food, pharmaceutical, biotechnological and medicinal applications. Front Pharmacol 11, 01021.CrossRefGoogle ScholarPubMed
Wahlang, B, Pawar, YB & Bansal, AK (2011) Identification of permeability-related hurdles in oral delivery of curcumin using the Caco-2 cell model. Eur J Pharm Biopharm 77, 275282.CrossRefGoogle ScholarPubMed
Heger, M, et al., (2014) The molecular basis for the pharmacokinetics and pharmacodynamics of curcumin and its metabolites in relation to cancer. Pharmacol Rev 66, 222307.CrossRefGoogle ScholarPubMed
Nelson, KM, et al. (2017) The essential medicinal chemistry of curcumin. J Med Chem 60, 16201637.CrossRefGoogle ScholarPubMed
Jankun, J, et al. (2016) Determining whether curcumin degradation/condensation is actually bioactivation (review). Int J Mol Med 37, 11511158.CrossRefGoogle ScholarPubMed
Tsuda, T (2018) Curcumin as a functional food-derived factor: degradation products, metabolites, bioactivity, and future perspectives. Food Funct 9, 705714.CrossRefGoogle ScholarPubMed
Kurita, T and Makino, Y (2013) Novel curcumin oral dfelivery systems. Anticancer Res 33, 28072821.Google ScholarPubMed
Krishnakumar, IM, Dinesh Kumar, AR, Kuttan, R & Maliakel, B (2012) An enhanced bioavailable formulation of curcumin using fenugreek-derived soluble dietary fibre. J Funct Foods 4, 348357.Google Scholar
Song, Y, et al. (2019) Development of a liquid chromatography-tandem mass spectrometric (LC-MS/MS) method for simultaneous determination of epigallocatechin-3-gallate, silibinin, and curcumin in plasma and different tissues after oral dosing of Protandim in rats and its application in pharmacokinetic and tissue distribution studies. J Pharm Biomed Anal 170, 5462.CrossRefGoogle ScholarPubMed
Abhilash, MB, et al. (2021) Enhanced absorption of curcuminoids and 3-acetyl-11-keto-β-boswellic acid from fenugreek galactomannan hydrogel beadlets: a natural approach to the co-delivery of lipophilic phytonutrients. J Funct Foods 79, 104405.CrossRefGoogle Scholar
Joseph, A, et al. (2020) Safety assessment of a fenugreek dietary fiber-based formulation of capsaicinoids-rich red chili (Capsicum annum) extract (Capsifen®): acute and sub-chronic studies. Toxicol Rep 7, 602609.CrossRefGoogle ScholarPubMed
Vijayasteltar, L, et al. (2017) Beyond the flavor: a green formulation of Ferula asafoetida oleo-gum-resin with fenugreek dietary fibre and its gut health potential. Toxicol Rep 4, 382390.CrossRefGoogle Scholar
Vismaya, KU, et al. (2022) Natural self-emulsifying reversible hybrid-hydrogel delivery (N’SERH) of tocopherol enhances bioavailability and modulates alcohol-induced reproductive toxicity in rats. Andrologia 54(2), e14305.Google Scholar
Begum, AN, et al. (2008) Curcumin structure-function, bioavailability, and efficacy in models of neuroinflammation and Alzheimer’s disease. J Pharmacol Exp Ther 326, 196208.CrossRefGoogle ScholarPubMed
Sandur, SK, et al. (2007) Curcumin, demethoxycurcumin, bisdemethoxycurcumin, tetrahydrocurcumin and turmerones differentially regulate anti-inflammatory and anti-proliferative responses through a ROS-independent mechanism. Carcinogenesis 28, 17651773.CrossRefGoogle ScholarPubMed
Pal, A, et al. (2014) Curcumin glucuronides: assessing the proliferative activity against human cell lines. Bioorg Med Chem 22, 435439.CrossRefGoogle ScholarPubMed
Ireson, C, et al. (2001) Characterization of metabolites of the chemopreventive agent curcumin in human and rat hepatocytes and in the rat in vivo, and evaluation of their ability to inhibit phorbol ester-induced prostaglandin E2 production. Cancer Res 61, 10581064.Google ScholarPubMed
Kumar, D, et al. (2015) Enhanced bioavailability and relative distribution of free (unconjugated) curcuminoids following the oral administration of a food-grade formulation with fenugreek dietary fibre: a randomised double-blind crossover study. J Funct Foods 22, 578587.CrossRefGoogle Scholar
Garcea, G, et al. (2004) Detection of curcumin and its metabolites in hepatic tissue and portal blood of patients following oral administration. Br J Cancer 90, 10111015.CrossRefGoogle Scholar
Pancholi, V, et al. (2021) Safety assessment of a highly bioavailable curcumin-galactomannoside complex (CurQfen) in healthy volunteers, with a special reference to the recent hepatotoxic reports of curcumin supplements: a 90-days prospective study. Toxicol Rep 8, 12551264.CrossRefGoogle Scholar
Fança-Berthon, P, et al. (2021) Pharmacokinetics of a single dose of turmeric curcuminoids depends on formulation: results of a human crossover study. J Nutr 151, 18021816.CrossRefGoogle ScholarPubMed
Prasad, S, et al. (2014) Curcumin, a component of golden spice: from bedside to bench and back. Biotechnol Adv 32, 10531064.CrossRefGoogle ScholarPubMed
Prasad, S, Tyagi, AK & Aggarwal, BB (2014) Recent developments in delivery, bioavailability, absorption and metabolism of curcumin: the golden pigment from golden spice. Cancer Res Treat 46, 218.CrossRefGoogle ScholarPubMed
Pandey, A, et al. (2020) Reductive metabolites of curcumin and their therapeutic effects. Heliyon 6, e05469.CrossRefGoogle Scholar
Shen, L and Ji, H-F (2009) Contribution of degradation products to the anticancer activity of curcumin. Clin Cancer Res 15, 7108.CrossRefGoogle Scholar
Shoji, M, et al. (2014) Comparison of the effects of curcumin and curcumin glucuronide in human hepatocellular carcinoma HepG2 cells. Food Chem 151, 126132.CrossRefGoogle ScholarPubMed
Choudhury, AK, et al. (2015) Synthesis and evaluation of the anti-oxidant capacity of curcumin glucuronides, the major curcumin metabolites. Antioxidants (Basel) 4, 750767.CrossRefGoogle ScholarPubMed
Pal, A, et al. (2014) Curcumin glucuronides: assessing the proliferative activity against human cell lines. Bioorg Med Chem 22, 435439.CrossRefGoogle ScholarPubMed
Volak, LP, et al. (2008) Curcuminoids inhibit multiple human cytochromes P450, UDP-glucuronosyltransferase, and sulfotransferase enzymes, whereas piperine is a relatively selective CYP3A4 inhibitor. Drug Metab Dispos 36, 15941605.CrossRefGoogle ScholarPubMed
Srinivasan, K (2007) Black pepper and its pungent principle-piperine: a review of diverse physiological effects. Crit Rev Food Sci Nutr 47, 735748.CrossRefGoogle ScholarPubMed
Ziegenhagen, R, et al. (2021) Safety aspects of the use of isolated piperine ingested as a bolus. Foods (Basel, Switzerland). 10, 2121.Google ScholarPubMed
Stohs, SJ, et al. (2019) The fallacy of enzymatic hydrolysis for the determination of bioactive curcumin in plasma samples as an indication of bioavailability: a comparative study. BMC Complement Altern Med 19, 293.CrossRefGoogle ScholarPubMed
Liju, VB, et al., (2015) Enhanced bioavailability and safety of curcumagalactomannosides as a dietary ingredient. Food Funct 6, 275285.CrossRefGoogle ScholarPubMed
Szymusiak, M, et al., (2016) Bioavailability of curcumin and curcumin glucuronide in the central nervous system of mice after oral delivery of nano-curcumin. Int J Pharm 511, 415423.CrossRefGoogle ScholarPubMed
Krishnakumar, IM, Maliakel, A, Gopakumar, G, Kumar, D, Maliakel, B & Kuttan, R (2015) Improved blood–brain-barrier permeability and tissue distribution following the oral administration of a food-grade formulation of curcumin with fenugreek fibre. J Funct Foods 14, 215225.Google Scholar
Douglass, BJ & Clouatre, DL (2015) Beyond yellow curry: assessing commercial curcumin absorption technologies. J Am Coll Nutr 34, 347358.CrossRefGoogle ScholarPubMed
Jager, R, et al. (2014) Comparative absorption of curcumin formulations. Nutr J 13, 11.CrossRefGoogle ScholarPubMed
Cuomo, J, et al. (2011) Comparative absorption of a standardized curcuminoid mixture and its lecithin formulation. J Nat Prod 74, 664669.CrossRefGoogle ScholarPubMed
Schiborr, C, et al. (2014) The oral bioavailability of curcumin from micronized powder and liquid micelles is significantly increased in healthy humans and differs between sexes. Mol Nutr Food Res 58, 516527.CrossRefGoogle ScholarPubMed
Kannan, RG, et al. (2021) Brain regional pharmacokinetics following the oral administration of curcumagalactomannosides and its relation to cognitive function. Nutr Neurosci, 25(9), 19281939.CrossRefGoogle ScholarPubMed
Pan, M-H, Huang, T-M & Lin, J-K (1999) Biotransformation of curcumin through reduction and glucuronidation in mice. Drug Metab Dispos 27, 486494.Google ScholarPubMed
Pandaran Sudheeran, S, et al. (2016) Safety, tolerance, and enhanced efficacy of a bioavailable formulation of curcumin with fenugreek dietary fiber on occupational stress: a randomized, double-blind, placebo-controlled pilot study. J Clin Psychopharmacol 36, 236243.CrossRefGoogle Scholar
Jamwal, R (2018) Bioavailable curcumin formulations: a review of pharmacokinetic studies in healthy volunteers. J Integr Med 16, 367374.CrossRefGoogle ScholarPubMed
Dei Cas, M and Ghidoni, R (2019) Dietary curcumin: correlation between bioavailability and health potential. Nutrients 11, 2147.CrossRefGoogle ScholarPubMed
Maiti, P and Dunbar, GL (2018) Use of curcumin, a natural polyphenol for targeting molecular pathways in treating age-related neurodegenerative diseases. Int J Mol Sci 19(6), 1637.CrossRefGoogle ScholarPubMed
Rane, JS, Bhaumik, P & Panda, D (2017) Curcumin inhibits tau aggregation and disintegrates preformed tau filaments in vitro . J Alzheimers Dis 60, 9991014.CrossRefGoogle ScholarPubMed
Rao, PP, et al. (2015) Curcumin binding to beta amyloid: a computational study. Chem Biol Drug Des 86, 813820.CrossRefGoogle ScholarPubMed
Reddy, PH, et al. (2016) Protective effects of a natural product, curcumin, against amyloid beta induced mitochondrial and synaptic toxicities in Alzheimer’s disease. J Investig Med 64, 12201234.CrossRefGoogle ScholarPubMed
Reddy, PH, et al. (2018) Protective effects of Indian spice curcumin against amyloid-beta in Alzheimer’s disease. J Alzheimers Dis 61, 843866.CrossRefGoogle ScholarPubMed
Sunny, A, Ramalingam, K, Das, SS, Maliakel, B, Krishnakumar, IM & Ittiyavirah, S (2019) Bioavailable curcumin alleviates lipopolysaccharide-induced neuroinflammation and improves cognition in experimental animals. Pharmacogn Mag 15, 111117.Google Scholar
Lee, JW, et al. (2008) Neuro-inflammation induced by lipopolysaccharide causes cognitive impairment through enhancement of beta-amyloid generation. J Neuroinflamm 5, 37.CrossRefGoogle ScholarPubMed
Ittiyavirah, SP (2019) Ameliorating demyelination effect of curqfen® in cuprizone induced mice model of multiple sclerosis. Pharm Pharmacol Int J 7, 1720.Google Scholar
Torkildsen, O, et al. (2008) The cuprizone model for demyelination. Acta Neurol Scand Suppl 188, 7276.CrossRefGoogle ScholarPubMed
Sindhu, ER, Binitha, PP, Nair, SS, Maliakel, B, Ramadasan, K & Krishnakumar, IM (2020) Comparative neuroprotective effects of native curcumin and its galactomannoside formulation in carbofuran-induced neurotoxicity model. Nat Product Res 34, 14561460.Google Scholar
Fedoce, AdG, et al. (2018) The role of oxidative stress in anxiety disorder: cause or consequence? Free Radic Res 52, 737750.CrossRefGoogle ScholarPubMed
Lobo, V, et al. (2010) Free radicals, antioxidants and functional foods: impact on human health. Pharmacogn Rev 4, 118126.CrossRefGoogle ScholarPubMed
Khanna, A, et al. (2020) The effects of oral administration of curcumin-galactomannan complex on brain waves are consistent with brain penetration: a randomized, double-blinded, placebo-controlled pilot study. Nutr Neurosci, 25(6), 12401249.CrossRefGoogle ScholarPubMed
Bewernitz, M & Derendorf, H (2012) Electroencephalogram-based pharmacodynamic measures: a review. Int J Clin Pharmacol Ther 50, 162184.CrossRefGoogle ScholarPubMed
Klimesch, W, Schimke, H & Pfurtscheller, G (1993) Alpha frequency, cognitive load and memory performance. Brain Topogr 5, 241251.CrossRefGoogle ScholarPubMed
Fernandez, T, et al. (1995) EEG activation patterns during the performance of tasks involving different components of mental calculation. Electroencephalogr Clin Neurophysiol 94, 175182.CrossRefGoogle ScholarPubMed
Lal, SK and Craig, A (2001) A critical review of the psychophysiology of driver fatigue. Biol Psychol 55, 173194.CrossRefGoogle ScholarPubMed
Sarlak, Z, Oryan, S & Moghaddasi, M (2015) Interaction between the antioxidant activity of curcumin and cholinergic system on memory retention in adult male Wistar rats. Iran J Basic Med Sci 18, 398403.Google ScholarPubMed
Roopun, AK, et al. (2010) Cholinergic neuromodulation controls directed temporal communication in neocortex in vitro . Front Neural Circuits 4, 8.Google ScholarPubMed
Tiago-Costa, E, Quelhas-Costa, E & Santos-Baptista, J (2016) Changes in EEG amplitude (alpha and beta waves) with thermal environment. DYNA 83, 8793.CrossRefGoogle Scholar
Yin, T, et al. (2022) The association between dietary antioxidant micronutrients and cardiovascular disease in adults in the United States: a cross-sectional study. Front Nutr 8, 799095.CrossRefGoogle ScholarPubMed
Li, H, et al. (2019) Curcumin, the golden spice in treating cardiovascular diseases. Biotechnol Adv 38, 107343.CrossRefGoogle ScholarPubMed
Panahi, Y, et al. (2017) Curcuminoids modify lipid profile in type 2 diabetes mellitus: a randomized controlled trial. Complement Ther Med 33, 15.CrossRefGoogle Scholar
Rahimi, HR, et al. (2016) The effect of nano-curcumin on HbA1c, fasting blood glucose, and lipid profile in diabetic subjects: a randomized clinical trial. Avicenna J Phytomed 6, 567577.Google ScholarPubMed
Jin, T, et al. (2018) Curcumin and other dietary polyphenols: potential mechanisms of metabolic actions and therapy for diabetes and obesity. Am J Physiol Endocrinol Metab 314, E201E205.CrossRefGoogle ScholarPubMed
Saji, S, et al. (2018) Curcumin-galactomannoside complex inhibits pathogenesis in Ox-LDL-challenged human peripheral blood mononuclear cells. Inflammopharmacology 26, 12731282.CrossRefGoogle ScholarPubMed
Baszczuk, A, et al. (2014) Hyperhomocysteinemia, lipid and lipoprotein disturbances in patients with primary hypertension. Adv Med Sci 59, 6873.CrossRefGoogle ScholarPubMed
Esterbauer, H, Wag, G & Puhl, H (1993) Lipid peroxidation and its role in atherosclerosis. Br Med Bull 49, 566576.CrossRefGoogle ScholarPubMed
Krishnareddy, NT, et al. (2018) A novel curcumin-galactomannoside complex delivery system improves hepatic function markers in chronic alcoholics: a double-blinded, randomized, placebo-controlled study. Biomed Res Int 2018, 9159281.CrossRefGoogle Scholar
Campbell, MS, et al. (2019) Influence of enhanced bioavailable curcumin on obesity-associated cardiovascular disease risk factors and arterial function: a double-blinded, randomized, controlled trial. Nutrition 62, 135139.CrossRefGoogle Scholar
Campbell, M, et al. (2017) Responsiveness to curcumin intervention is associated with reduced aortic stiffness in young, obese men with higher initial stiffness. J Funct Foods 29, 154160.CrossRefGoogle Scholar
Vlachopoulos, C, Aznaouridis, K & Stefanadis, C (2010) Prediction of cardiovascular events and all-cause mortality with arterial stiffness: a systematic review and meta-analysis. J Am Coll Cardiol 55, 13181327.CrossRefGoogle ScholarPubMed
Tang, X, et al. (2003) Interleukin-13 upregulates vasodilatory 15-lipoxygenase eicosanoids in rabbit aorta. Arterioscler Thromb Vasc Biol 23, 17681774.CrossRefGoogle ScholarPubMed
Cardilo-Reis, L, et al. (2012) Interleukin-13 protects from atherosclerosis and modulates plaque composition by skewing the macrophage phenotype. EMBO Mol Med 4, 10721086.CrossRefGoogle ScholarPubMed
Hong, M, et al. (2015) Current status of herbal medicines in chronic liver disease therapy: the biological effects, molecular targets and future prospects. Int J Mol Sci 16, 2870528745.CrossRefGoogle ScholarPubMed
Bielak-Zmijewska, A, et al. (2019) The role of curcumin in the modulation of ageing. Int J Mol Sci 20(5), 1239.CrossRefGoogle ScholarPubMed
Sandur, SK, et al. (2007) Role of pro-oxidants and antioxidants in the anti-inflammatory and apoptotic effects of curcumin (diferuloylmethane). Free Radic Biol Med 43, 568580.CrossRefGoogle ScholarPubMed
Sikora, E, et al. (2010) The promise of slow down ageing may come from curcumin. Curr Pharm Des 16, 884892.CrossRefGoogle ScholarPubMed
Deguchi, A (2015) Curcumin targets in inflammation and cancer. Endocr Metab Immune Disord Drug Targets 15, 8896.CrossRefGoogle Scholar
Tasneem, S, et al. (2019) Molecular pharmacology of inflammation: medicinal plants as anti-inflammatory agents. Pharmacol Res 139, 126140.CrossRefGoogle Scholar
Chin, KY (2016) The spice for joint inflammation: anti-inflammatory role of curcumin in treating osteoarthritis. Drug Des Devel Ther 10, 30293042.CrossRefGoogle ScholarPubMed
Mohan, R, et al. (2019) Curcumin-galactomannosides mitigate alcohol-induced liver damage by inhibiting oxidative stress, hepatic inflammation, and enhance bioavailability on TLR4/MMP events compared to curcumin. J Biochem Mol Toxicol 33, e22315.CrossRefGoogle ScholarPubMed
Banerjee, P, et al. (2013) Inflammation and MMPs in alcohol-induced liver diseases and protective action of antioxidants. Indian J Biochem Biophys 50, 377386.Google ScholarPubMed
Gustot, T, et al. (2006) Differential liver sensitization to Toll-like receptor pathways in mice with alcoholic fatty liver. Hepatology 43, 9891000.CrossRefGoogle ScholarPubMed
Jurenka, JS (2009) Anti-inflammatory properties of curcumin, a major constituent of curcuma longa: a review of preclinical and clinical research. Altern Med Rev 14, 141153.Google Scholar
Shimizu, K, et al. (2019) Anti-inflammatory action of curcumin and its use in the treatment of lifestyle-related diseases. Eur Cardiol 14, 117122.CrossRefGoogle ScholarPubMed
Menon, VP and Sudheer, AR (2007) Antioxidant and anti-inflammatory properties of curcumin. Adv Exp Med Biol 595, 105125.CrossRefGoogle ScholarPubMed
Sheethal, S, et al. (2020) Anti-ulcerative effect of curcumin-galactomannoside complex on acetic acid-induced experimental model by inhibiting inflammation and oxidative stress. Inflammation 43, 14111422.CrossRefGoogle Scholar
Sangeeth Saji, KI, Jose, SP, Ratheesh, M, Sandya, S & Asha, S (2021) Curcumin-galactomannoside interaction enhances the bioavailability of curcumin and inhibits inflammation and oxidative stress associated with experimental arthritis. Virol Mycol 10, 203.Google Scholar
Khanna, A, et al. (2020) Curcumagalactomannoside/glucosamine combination improved joint health among osteoarthritic subjects as compared to chondroitin sulfate/glucosamine: double-blinded, randomized controlled study. J Altern Complement Med 26, 945955.CrossRefGoogle ScholarPubMed
Thomas, JV, et al. (2021) Influence of a low-dose supplementation of curcumagalactomannoside complex (CurQfen) in knee osteoarthritis: a randomized, open-labeled, active-controlled clinical trial. Phytother Res. 35(3), 14431455.CrossRefGoogle Scholar
Zhu, X, et al. (2018) Effectiveness and safety of glucosamine and chondroitin for the treatment of osteoarthritis: a meta-analysis of randomized controlled trials. J Orthop Surg Res 13, 170.CrossRefGoogle ScholarPubMed
Zhu, X, et al. (2018) Comparative effectiveness of glucosamine, chondroitin, acetaminophen or celecoxib for the treatment of knee and/or hip osteoarthritis: a network meta-analysis. Clin Exp Rheumatol 36, 595602.Google ScholarPubMed
Clegg, DO, et al. (2006) Glucosamine, chondroitin sulfate, and the two in combination for painful knee osteoarthritis. N Engl J Med 354, 795808.CrossRefGoogle ScholarPubMed
Daily, JW, Yang, M & Park, S (2016) Efficacy of turmeric extracts and curcumin for alleviating the symptoms of joint arthritis: a systematic review and meta-analysis of randomized clinical trials. J Med Food 19, 717729.CrossRefGoogle ScholarPubMed
Goh, J, et al. (2020) Examination of curcumin and fenugreek soluble fiber supplementation on submaximal and maximal aerobic performance indices. J Funct Morphol Kinesiol 5, 34.CrossRefGoogle ScholarPubMed
Camic, CLCL, et al. (2010) The effects of 4 weeks of an arginine-based supplement on the gas exchange threshold and peak oxygen uptake. Appl Physiol Nutr Metab 35, 286293.CrossRefGoogle ScholarPubMed
Hassan, F-u, et al. (2019) Curcumin as an alternative epigenetic modulator: mechanism of action and potential effects. Front Genet 10, 514.CrossRefGoogle ScholarPubMed
Sharifi-Rad, J, et al. (2020) Turmeric and its major compound curcumin on health: bioactive effects and safety profiles for food, pharmaceutical, biotechnological and medicinal applications. Front Pharmacol 11, 01021.CrossRefGoogle ScholarPubMed
Banik, K, et al. (2021) A novel bioavailable curcumin-galactomannan complex modulates the genes responsible for the development of chronic diseases in mice: a RNA sequence analysis. Life Sci, 287, 120074.CrossRefGoogle Scholar
Prasad, KN, et al. (2002) Pros and cons of antioxidant use during radiation therapy. Cancer Treat Rev 28, 7991.CrossRefGoogle ScholarPubMed
Liju, VB, et al. (2020) Amelioration of radiation-induced damages in mice by curcuminoids: the role of bioavailability. Nutr Cancer, 73(4), 617629.CrossRefGoogle ScholarPubMed
Ratheesh, M, et al. (2021) Curcumin-galactomannoside complex inhibits the proliferation of human cervical cancer cells: possible role in cell cycle arrest and apoptosis. Asian Pac J Cancer Prev: APJCP 22, 17131720.Google Scholar
Jiang, S, et al. (2017) Curcumin as a potential protective compound against cardiac diseases. Pharmacol Res 119, 373383.CrossRefGoogle Scholar
Tomeh, MA, Hadianamrei, R & Zhao, X (2019) A review of curcumin and its derivatives as anticancer agents. Int J Mol Sci 20(5), 1033.CrossRefGoogle ScholarPubMed
Borek, C (2004) Antioxidants and radiation therapy. J Nutr 134, 3207S3209S.CrossRefGoogle ScholarPubMed
Borek, C (2004) Dietary antioxidants and human cancer. Integr Cancer Ther 3, 333341.CrossRefGoogle ScholarPubMed
Lopresti, AL (2018) The problem of curcumin and its bioavailability: could its gastrointestinal influence contribute to its overall health-enhancing effects? Adv Nutr 9, 4150.CrossRefGoogle ScholarPubMed
Jabczyk, M, et al. (2021) Curcumin and its potential impact on microbiota. Nutrients 13, 2004.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Oral administration of curcumin results mainly in conjugated curcumin detected in plasma, and intravenous or intraperitoneal administration results mainly in reduced curcumin metabolites, while there are minor levels of free curcumin detected in plasma following any administration method.

Figure 1

Fig. 2. CGM results in higher bioavailability and tissue distribution of free curcuminoids with improved effects on inflammatory, oxidative stress and endogenous antioxidant pathways in multiple organ systems. AChE, acetylcholine esterase; CAT, catalase; CRP, C-reactive protein; Dopa, dopamine; Glu, glutamate; GPx, glutathione peroxidase; GSH, Reduced Glutathione; HDL, High density lipoprotein; IL-1β, interleukin-1β; IL-6, interleukin-6; i-NOS, inducible nitric oxide synthase; MMP-2 & 9, matrix metalloproteinase-2 & 9; NF-κB, nuclear factor kappa B; Ser, serotonin; sVCAM, solublevascular cell adhesion molecule; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor α.

Figure 2

Table 1. Pre-clinical studies

Figure 3

Table 2. Clinical studies