Khemraj Bairwa
,
Jagdeep Grover
,
Mihir Kania
and
Sanjay M. Jachak
*
Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, SAS Nagar (Mohali)-160062, Punjab, India. E-mail: sanjayjachak@niper.ac.in; sjachak11@gmail.com; Fax: +91 172 2214692; Tel: +91 172 2214683
First published on 28th February 2014
Curcumin, a prominent constituent of the rhizome of Curcuma longa L., possesses versatile biological properties, which is evidenced from the extensive research during the last half century. Curcumin has been shown to exhibit antioxidant, anti-inflammatory, antiviral, antibacterial, antifungal, and anticancer activities and thus has potential against various malignant diseases, such as allergies, arthritis, Alzheimer's disease, and other chronic illnesses. In the last decade it has been much explored and various synthetic analogues have been prepared and evaluated for various pharmacological activities that render it as a lead molecule against several biological targets. To accelerate this lead molecule from kitchen to clinic, around 65 clinical trials are underway worldwide to assess its therapeutic potential. Thus, there is continued interest in the synthesis of new curcumin analogues with a similar safety profile, but increased activity and improved oral bioavailability. The present review article describes recent developments in curcumin chemistry with emphasis on the semi-synthesis, synthesis pharmacological properties and SAR of various curcumin analogues reported from 1994 to mid 2013.
During the last two decades, numerous studies have shown that curcumin possess a variety of biological activities (Table 1) such as anti-carcinogen,7,8 immunomodulation,9 antioxidant,10 anti-inflammatory,11,12 anti-angiogenesis,13 antitumor,14 chemo-prevention,15 Alzheimer's disease,16 antimicrobial,17 antimalarial,18 antithrombotic,19 myocardial infarction protective,20 rheumatoid arthritis,21 inhibition of human immunodeficiency virus (HIV) replication,22 enhancement of wound healing,23 antihepatotoxic,24 psoriasis,25 hypoglycemic,26 and antihyperlipidaemic.27,28 Curcumin interacts with numerous molecular and biochemical targets which includes inflammatory cytokines, transcriptional factors, kinases, enzymes, receptors, growth factors, anti-apoptotic proteins, adhesion molecules and ligands.28–30 These diverse biological properties attracted clinicians to explore the therapeutic potential of curcumin by conducting clinical trials. Around 65 clinical trials are underway world wide to verify the therapeutic potential of curcumin for various diseases.28,31,32 In spite of the broad spectrum activities and safety profile exhibited by curcumin, it is not yet approved as a therapeutic agent due to its poor oral bioavailability.33 Curcumin is non-toxic even at high dosages, to date no toxicity has been found in any animal and human studies and has been classified as ‘generally recognized as safe’ (GRAS) by the National Cancer Institute.33,34 The pharmacokinetic and pharmacodynamics studies on curcumin has proved that one of the major limitations of using curcumin as a drug is its poor water and plasma solubility; even doses as high as 8 g of curcumin per day administered to human subject's results in an average peak serum concentration of ∼1.77 μM of curcumin.33,34 Also, because curcumin strikes the cell at several sites, it is difficult to determine which target is causing the desired effect in a certain disease. Thus, there is a continued interest in the synthesis of new curcumin analogues with a similar safety profile, but increased activity and improved oral bioavailability.
Compound | Activity | Assay/target | IC50 (μM) | References |
---|---|---|---|---|
a * indicates % inhibition in mg ml−1; ** indicates % inhibition at 6 μg ml−1; # indicates GI50 value in μM; ## indicates MIC. b Against vincristine induced cytotoxicity. c Against paclitaxel induced cytotoxicity. | ||||
Curcumin (1) | Antioxidant | Lipid peroxidation | 1.30 | 40 |
ABTS+ assay | 3.37 | 40 | ||
DPPH assay | 20.02 | 40 and 41 | ||
NBT assay | 41 | 41 | ||
Anti-inflammatory | COX-1 | 50 | 43 | |
COX-2 | >100 | 43 | ||
Antitumor | COX-2 | 15.9 | 47 | |
Fos–Jun DNA complex | 0.48* | 45 | ||
NF-κB | 8.2 | 44 | ||
Leukaemia | 3.7# | 55 | ||
NSCL | 9.2# | 55 | ||
Colon | 4.7# | 55 | ||
CNS | 5.8# | 55 | ||
Melanoma | 7.1# | 55 | ||
Ovarian | 8.9# | 55 | ||
Renal | 8.9# | 55 | ||
Prostate | 11.2# | 55 | ||
Breast | 5.9# | 55 | ||
MDR reversal activity | 0.82b | 61 | ||
MTS assay | 0.42c | |||
Cytotoxic | PC-3 | 7.7 | 48 | |
19.8 | 50 | |||
LNCaP | 3.8 | 48 | ||
19.6 | 50 | |||
DU-145 (mutant & wild AR) | Inactive | 49 | ||
HepG2 | 4.6 | 46 | ||
SMMC-7721 | 5.4 | 46 | ||
CT26 | 16.6 | 46 | ||
Hela | 7.5, 17.5 | 46 and 75 | ||
Skov3 | 7.8 | 46 | ||
A549 | 10.3 | 46 | ||
MCF-7 | 21.5 | 50 | ||
MDA-MB-231 | 25.6 | 50 | ||
BGC 823 | 16.7 | 72 | ||
CNE | 32.8 | 72 | ||
HL-60 | 11.6 | 72 | ||
KB | 35.9 | 72 | ||
LS 174T | 6.4 | 72 | ||
A16-F10 | 18.6 | 69 | ||
EGFR | 8.6 | 69 | ||
Anti-angiogenesis | HUVEC | 10.84 | 70 | |
SVR cell line | 56.2**, 9.8# | 53,55 and 76 | ||
Anti-HIV | Strand transfer | 140 | 51 | |
Antimalarial | P. falciparum (strain FCK2) | 3.25 | 50 | |
P. falciparum (strain MP-14) | 4.21 | 50 | ||
Antimicrobial | Escherichia coli | 20## | 83 and 84 | |
Staphylococcus aureus | 20## | 83 and 84 | ||
Staphylococcus epidermidis | 10## | 83 and 84 | ||
Enterococcus | 20## | 83 and 84 |
The broad-spectrum activities and the least bioavailability led the path for the semi-synthesis and synthesis of various curcumin analogues. The curcumin analogues are those which encompass some ostensible or claimed structural analogy or likeness to curcumin. The structure of curcumin includes two feruloyl moieties, non-polar heptadiene bridge and 1,3-keto–enol or diketo moiety (Fig. 2). A preliminary structure–activity relationship (SAR) study of symmetrical curcuminoids explained that the feruloyl moiety plays a critical role in various biological functions. However, curcumin is stable at a pH below 6.5. The instability of curcumin at a pH above 6.5 is caused by the active methylene group.33 It has been proved that the biological efficacy of curcumin arises from the electrophilic nature of its central β-diketone component. It is suggested that the presence of the methylene group and β-diketone moiety contributes to the instability of curcumin under physiological conditions.37 Therefore; the deletion of the β-diketone moiety may contribute to the enhancement of stability of curcuminoids. On the basis of the former information and research on curcumin, there have been considerable efforts to synthesize various bioactive analogues of curcumin and the process is still on the way of progress with a large number of successes. In the present article, we have focused on different curcumin analogues prepared by both synthetic or semi-synthetic route and being described as per following schemes;
(1) Changes in aryl substitution pattern of curcumin,
(2) Changes in the β-diketone structure which includes,
(2.1) Monocarbonyl analogues (cyclic and acyclic, symmetrical and asymmetrical),
(2.2) Dicarbonyl analogues,
(2.3) Heterocyclic analogues,
(2.4) Change in the 1,3-keto–enol moiety,
(2.5) Change in the conjugation,
(2.6) Substitution on the 1,6-heptadiene moiety (β-carbon of dicarbonyl chain),
(3) Curcumin bioconjugates,
(3.1) Amino-acid conjugates,
(3.2) Curcuminoid glycosides,
(3.3) Curcumin–taxoid conjugate,
(4) Miscellaneous.
General structure for Class-1 compounds (40–62) | ||||
---|---|---|---|---|
Compounds | R1 | R2 | R3 | R4 |
40 | H | H | OCH3 | H |
41 | OH | H | OCH3 | OH |
42 | H | H | OH | H |
43 | OCH3 | CH3 | OCH3 | H |
44 | OCH3 | C3H7 | OCH3 | H |
45 | OCH3 | C3H7 | OCH3 | C3H7 |
46 | OCH3 | CH2CH2OH | OCH3 | H |
47 | OCH3 | CH2–CHCH2 | OCH3 | H |
48 | OCH3 | CH2–CHCH2 | OCH3 | CH2–CHCH2 |
49 | OCH3 | CH2(CH2)3CH3 | OCH3 | H |
50 | OCH3 | CH2(CH2)3CH3 | OCH3 | CH2(CH2)3CH3 |
51 | OCH3 | Ac | OCH3 | H |
52 | H | Ac | OCH3 | H |
53 | H | H | OCH3 | Ac |
54 | H | Ac | OCH3 | Ac |
55 | H | Ac | H | H |
56 | H | CH3 | OCH3 | H |
57 | H | CH3 | OCH3 | CH3 |
58 | H | CH3 | H | H |
59 | OCH3 | CH2CHCH(CH3)2 | OCH3 | H |
60 | H | CH2CHCH(CH3)2 | OCH3 | CH2CHCH(CH3)2 |
61 | H | CH2CH2OH | OCH3 | H |
62 | H | H | OCH3 | CH2CH2OH |
Activity | Assay/target | Compound(s) | IC50 (μM) | Ref. |
---|---|---|---|---|
a * indicates % inhibition in mg ml−1; ** indicates % inhibition at 1 μM; # indicates GI50; NA, not active (MIC > 200 mg ml−1); ND, not determined; INS = insoluble; ## indicate MIC (μg ml−1). | ||||
Antioxidant | Lipid peroxidation | 18, 19 | 1.1, 0.6 | 40 |
ABTS+ assay | 12, 18, 20, 21, 16 | 2.6, 3.3, 1.0, 2.3, 2.1 | 40 | |
DPPH assay | 38, 11, 17, 18, 19, 20, 39 | 6, 4.6, 26, 30.3, 21.7, 23.7, >300 | 40–42 | |
NBT assay | 38, 11, 20, 22 | 11.8, 4.8, 37.6, 27.8 | 41 | |
Anti-inflammatory | COX-1 | 14, 26, 23 | 0.06, 0.3, 2.6 | 44 |
COX-2 | 26 | 12.5 | 44 | |
Antitumor | COX-2 | 27 | 23.7 | 48 |
Fos–Jun DNA complex | 25 | 0.38* | 46 | |
Cytotoxic | PC-3 | 12, 14, 15, 17, 33, 34, 35, 65, 66, 67, 68, 69, 70, 71 | 1.1, 8.1, 2.4, 14.0, 7.5, 13.1, 7.4, 39, 100.3, 48.1, 52.1, >100, >100, >100 | 49–51 and 59 |
LNCaP | 12, 14, 15, 17, 33, 34, 35, 39, 65, 66, 67, 68, 69, 70, 71 | 1.3, 4.8, 2.9, 2.6, 5.9, 10.4, 7.7, 19.7, 41.8, 73.0, 55.0, 54.8, >100, >100, >100 | 49,51 and 59 | |
MCF-7 | 12, 33, 34, 35, 36, 37 | 5.4, 5.5, 4.7, 5.5, >100, 49.2 | 51 and 52 | |
MDA-MB-231 | 12, 33, 34, 35 | 4.9, 3.1, 5.5, 6.5 | 51 | |
DU-145 (mutant AR) | 12 | 45** | 50 | |
DU-145 (wild AR) | 12 | 49** | 50 | |
HepG2 | 28, 29, 30, 31, 36, 60 | 10.1, 18.2, 7.2, 15.7, 71.6, 43.6 | 47 and 52 | |
SMMC-7721 | 28, 29, 30 | 53.1, 38.6, 24.2 | 47 | |
CT26 | 28, 29, 30, 31 | 6.1, 38.6, 25.1, 10.0 | 47 | |
Hela | 28, 29, 30 | 12.1, 31.2, 15.5 | 47 | |
31, 36, 60 | 11.7, 37.1, 23.8 | 52 | ||
Skov3 | 28, 30, 31, 36, 37 | 41, 76, 62.6, 55.5, 38.3 | 47 and 52 | |
A549 | 28, 29, 31, 58, 37 | 21, 42.4, 53.7, >100, >100 | 47 and 52 | |
A431 | 36, 37 | 7.2, 9.4 | 52 | |
U-251 | 36, 37 | 7.1, 7.7 | 52 | |
HEp-2 | 36, 37 | >100, >100 | 52 | |
HEK | 39, 41, 42, 12, 56, 57, 58, 45, 47, 48, 59, 60, 46, 30, 61, 21, 54 | 200, 36, 30, 200, 79, 690, 400, >500, 270, >500, >500, >500, 25, 20, 46, 80, 49 | 54 | |
A549 cells | 16, 12, 13, 63, 64 | >25, 5.80, 19.26, >25, >25 | 56 | |
Anti-HIV | Strand transfer | 38, 39 | 3.1, 80 | 53 |
Anti-malarial | P. falciparum (strain FCK2) | 12, 21 | 7.86, 2.34 | 57 |
P. falciparum (strain MP-14) | 12, 21 | 8.4, 2.51 | 57 | |
M. tuberculosis | H37Ra strain | 40, 39 | 50, 25 | 55 |
41, 42 | 200, 200 | |||
43, 12, 44 | 50, 100, 25 | |||
45, 46, 30 | 200, 200, 200 | |||
47, 48, 49 | 100, NA, 12.5 | |||
50, 51, 21 | 100, 100, 200 | |||
52, 53, 54 | 50, 50, 100 | |||
55, 24, 32 | 25, 7.81##, 125## | 58 | ||
Antitrypanosomal | T. brucei strain 427 | 40, 39, 41, 42, 12, 56, 57, 58, 45, 47, 48, 59, 60, 46, 30, 61, 21, 54 | 4.6, 7.7, 0.87, 0.65, 0.45, 1.7, 1.6, 8.9, 1.3, 2.4, 1.6, 1.4, 1.9, 2.9, 1.8, 0.7, 1.6, 1.5 | 54 |
T. brucei ΔTbat1 | 40, 39, 41, 42, 12, 56, 57, 58, 45, 47, 48, 59, 60, 46, 30, 61, 21, 54 | 5.9, 9.5, 1.1, 1.8, 0.62, 2.0, 1.9, 8.8, 1.8, 2.2, 1.9, 1.7, 2.9, 2.9, 2.3, 0.85, 2.2, 2.0 | 54 | |
T. brucei clone B48 | 40, 39, 41, 42, 12, 56, 57, 58, 45, 47, 48, 59, 60, 46, 30, 61, 21, 54 | 2.2, 4.5, 2.7, 1, 0.77, 2.1, 1.9, 2.9, 1.6, 12, 5.8, 2, 3.1, 1.8, 1.6, 1.1, 1.2, 0.92 | 54 | |
Antileishmanial | L. major promastigotes | 40, 39, 41, 42, 12, 56, 57, 58, 45, 47, 48, 59, 60, 46, 30, 61, 21, 54 | 37, 72, 4.3, 22, 2.8, 23, 23, 87, 28, 5.7, 25, 7.6, 48, 22, 33, 9.7, 73, >100 | 54 |
L. mexicana amastigotes | 40, 39, 41, 42, 12, 56, 57, 58, 45, 47, 48, 59, 60, 46, 30, 61, 21, 54 | 37, 63, 3.2, 21, 10, 38, 61, 32, 34, 12, >100, 21, 27, 30, 37, 32, 34, 43 | 54 |
In contrast to curcumin analogues that retained the 7-carbon spacer (39), the compounds with a 5-carbon linker had lower activity. In case of the later, the introduction of a ring further decreased DPPH-scavenging activity. However, the introduction of a ring did increase anti-haemolysis activity, suggesting that the lipophilicity of these compounds might play an important role in the antioxidant activity.42
Substitution with thiomethyl group (23), which can be found in some COX-2-selective compounds, exhibited a distinct affinity towards COX-1, while substitution with sulphonylmethyl group resulted in no activity. The para-hydroxy group on the phenyl ring of curcuminoids was proved to be essential for an anti-inflammatory activity. Introduction of an additional methoxy group ortho to hydroxy group, gives no change in cytotoxic activity and COX-2 inhibitory activity. Introduction of methylenedioxy group or nitro group (24, 25) or bulkier tert-butyl groups (20) diminished the ability to inhibit COX-2 enzyme, while addition of a fluorine atom (26, 27) in aromatic ring at meta or para-positions enhanced COX-2 selectivity.43,44
Since curcumin is maximally active, DMC is intermediate and BDMC is least active, this suggests that the methoxy groups do contribute to the suppression of NF-κB activation. It has been postulated that hydrogen bonding between ortho-methoxy oxygen and phenolic hydrogen influences the planarity, conformation and ability to undergo oxidation. This may probably influence the interaction with the proteins such as NF-κB. There is no correlation between anti-oxidant activity and ability to inhibit the TNF-α-induced activation of NF-κB.45 The para-position and polar group substituted curcuminoids have a tendency to inhibit the Fos–Jun DNA complex formation more potently. Introduction of a nitro group on phenyl ring increases the ability to inhibit the Fos–Jun DNA complex formation more potently.46 Replacement of para-hydroxy group with amino methyl group (24) resulted in no activity to all tested tumor cell lines. Introduction of polar groups (28–31) at para-position of phenyl group resulted in enhancement of cytotoxic activity. These results indicated that substitution at para-position of phenyl group had significant influence on the cytotoxicity of curcumin analogues. Substitution at para-position of phenyl group is responsible for the cytotoxicity of the compounds.47 Therefore; introduction of appropriate substitution at para-position of phenyl group might be a potential option for structural modification of curcumin. Demethylation or methylation of curcumin to form the dihydroxy and trimethoxy (13, 14) derivatives significantly increased cytotoxic activity against 1A9, KB and HCT-8 cell lines. Increase in the number of hydroxyl substitutions (11) on the aryl ring with or without methoxy substitutions (12–16) or tert-butyl substitutions (20) at ortho position showed promising tumor reducing activity. Thus, the presence of catechol or 3,4-dimethoxyphenyl substituents enhanced the cytotoxic properties.41,48–50 Introduction of methoxycarbonylmethyl group (32) at the phenolic hydroxyls contributed to increased antitumor activity.50 Conversion of para-hydroxy group into methoxy, acetate and sulfamate (12, 33–35), resulted in increase in cytotoxic activity.51
Compound 36 and 37, exhibited selective and potent cytotoxic activity against human epidermoid carcinoma cell line A-431 and human glioblastoma cell line U-251, implying their specific potential in the chemoprevention and chemotherapy of skin cancer and glioma. The preliminary SAR information extracted from the results suggested that introduction of appropriate substituents to the 4th-positions could be a promising approach for the development of new cytotoxic curcumin analogues with special selectivity for A-431 and U-251 cell lines.52
Curcumin derivatives without hydroxyl group showed no activity against HIV-1 integrase, while compounds retaining hydroxyl groups (38, 39) showed comparable activity as that of curcumin. In addition, the compounds with catechol group exhibited better HIV-1 integrase inhibitory activity.53
Decrease in oxygenated (hydroxyl and alkyl) substituents in the aromatic rings of curcuminoids (40–50) resulted in decreased antitrypanosomal activity of the curminoids and analogs; demethylation of various analogues of curcumin tends to increase the antitrypanosomal activity and decrease the antimycobacterial activity. Whereas higher alkyl ether analogues (compound 43–62) showed better activity than the parent compound.54,55
Qiu et al. synthesized new arylidene curcumin analogues (63, 64) and established SAR which concluded that the triarylvinyl skeleton of 4-arylidene curcumin analogues acts as a phamacophore, and the 4-arylmethylene modification is an effective strategy to improve the NF-κB inhibitory activity of curcumin analogues.56
Methylation of curcumin to form dimethoxycurcumin (12) resulted in loss of antimalarial activity while diacetylcurcumin (21) retained almost similar potency as curcumin. This suggests that hydroxy group is important for antimalarial activity of curcumin analogues with substitution on the aryl ring.57
Agrawal et al. synthesized semi-synthetic analogues (24, 32) of demethoxy curcumin and screened the compounds for antitubercular activity. The objective of the synthesis of 24 and 32 was to increase the lipophilicity of demethoxycurcumin by attaching fatty acid ester chains at the phenolic hydroxy groups due to the lipophilic nature of the Mycobacterium tuberculosis cell wall. These compounds showed better antitubercular activity than the parent compound.58
Shi et al. synthesized the cytotoxic curcumin analogues conjugated with anti-androgens. The synthesized compounds were evaluated against two human prostate cancer cell lines, androgen-dependent LNCaP and androgen-independent PC-3. Compound 65, exhibited the most potent cytotoxicity against both cell lines with IC50 values of 41.8 μM against LNCaP and 39.1 μM (Table 3) against PC-3 proliferation. Compound 65 was almost two-fold more potent than flutamide and bicalutamide. SAR revealed that replacement of the nitro group on the aniline ring of the hydroxyflutamide-related substructure of 65 with a cyano group in 66 resulted in two-fold decrease of activity. Substitution of cyano and fluoro groups (67, 68) on the aniline ring in the flutamide substructure with nitro (69), chloro (70), and methyl (71) groups diminished the activity against both the cell lines. So, it could be suggested that the substituents on the flutamide substructure may be important for interaction of the conjugate molecule with the target protein.59
Constructing the asymmetric monocarbonyl curcumin analogues having phenyl ring with alkyl amide (72–78), chloro-substituted benzamide (79–83) (Table 4) and heteroaromatic amide moieties (84–85), increased the anti-angiogenic potential as compared to curcumin. The aromatic amides exhibited higher activity than alkyl amides, but benzamide, furan amide (84), thiophene amide (85) exhibited highest potential to inhibit angiogenesis process (asymmetric monocarbonyl analogues). The analogues with aromatic dienone moiety showed good inhibition of in vitro (HUVEC assay) (Table 5), especially compound with pyridyl substitution showed excellent activity. In short, curcumin analogues having a weak electron-donating substituent at 4′ position or a strong electron-withdrawing substituent at 2′ position increased the cytotoxic activity.60
General structure for Class 2.1 compounds: asymmetrical (138–148) | ||||||
---|---|---|---|---|---|---|
Com. | R1 | R2 | R3 | R4 | R5 | R6 |
138 | OCH3 | OH | H | OCH3 | OCH3 | H |
139 | OCH3 | OCH3 | H | OCH3 | OCH3 | OCH3 |
140 | OCH3 | OCH3 | H | OCH3 | OCH3 | OCH3 |
141 | H | OCH3 | OCH3 | OCH3 | OCH3 | OCH3 |
142 | OH | H | H | OCH3 | OCH3 | OCH3 |
143 | OCH3 | OH | OCH3 | OCH3 | OCH3 | OCH3 |
144 | OCH3 | COCH2CH2OH | OCH3 | OCH3 | OCH3 | OCH3 |
145 | OCH3 | COCH2OH | OCH3 | OCH3 | OCH3 | OCH3 |
146 | OCH3 | COCH2O2Me | OCH3 | OCH3 | OCH3 | OCH3 |
147 | OCH3 | OCH(CH3)OCH2CH3 | OCH3 | OCH3 | OCH3 | OCH3 |
148 | OCH3 | OCH3 | OCH3 | OCH3 | ORc | OCH3 |
Activity | Assay/target | Compound(s) | IC50 (μM) | Ref. |
---|---|---|---|---|
a Indicates IC50 for compound of series-A.b Indicates IC50 for compound of series-B.c Indicates IC50 for compound of series-C.d Indicates GI50 value in μM.e Indicates % inhibition at 10 μM.f Indicates % inhibition at 6 μg ml−1.g Indicates % inhibition at 3 μg ml−1.h Indicates zone of inhibition in mm.i Indicates % increased life span; NA, not active (MIC > 200 mg ml−1); ND, not determined. | ||||
Antioxidant | Lipid peroxidation | 170a, 170b, 170c, 171a, 171b, 172a, 172b, 172c | 2.8, 2.5, 1.3, 2.0, 2.2, 11.6, 0.9, 2.9 | 71 |
TRAP assay | 91, 92, 93, 167a, 168a, 169c | 36e, 41e, 69e, 64e, 76e, 55e | 70 | |
FRAP assay | 91, 93, 169c | 53e, 60e, 69e | 70 | |
Anti-inflammatory | COX-2 | 192c | 5.5 | 48 |
LPS-induced TNF-α | 169a, 169b, 169c, 177a, 177c, 189c, 190b, 191a, 191b | 64, 38, 34.5, 34, 60, 34, 46, 36, 38 | 75 | |
LPS-induced IL-6 | 169a, 169b, 169c, 178b, 190a | 68, 36, 70, 80, 90 | 75 | |
Antitumor | NF-κb | 91, 168a, 169a, 169c, 183a, 184a | 6.2, 4.2, 9.6, 4.4, 7.0, 5.0 | 45 |
Leukaemia | 166c | 1.6d | 63 | |
NSCL | 166c | 3.1d | 63 | |
Colon | 166c, 84a, 85a, 138, 179a, 182c, 182a, 181a, 180a, 139, 140, 141, 142, 174a, 143, 144, 145, 146, 147, 201a, 202a, 148 | 2.1d, 15d, 0.8d, 0.7d, 1.5d, >50, 2.0, 2.3, 0.3, 0.4, 1.9, 3.3, 0.9, 0.8, 0.8, 1.1, 38, 7.0, 1.5, 0.3, 7.6, 0.3 | 63 and 67 | |
CNS | 166c | 2.5d | 63 | |
Melanoma | 166c | 2.2d | 63 | |
Ovarian | 166c | 3.3d | 63 | |
Renal | 166c | 2.0d | 63 | |
Prostate | 166c | 1.7d | 63 | |
Breast | 166c | 2.5d | 63 | |
MDR reversal activity (in presence of vincristine) | 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85 | 1.26, 1.26, 1.89, 1.92, 3.31, 3.81, 3.82, 0.48, 0.72, 1.23, 0.85, 0.41, 0.44, 1.41 | 61 | |
MDR reversal activity (in presence of paclitaxel) | 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85 | 0.83, 0.72, 0.21, 0.24, 1.09, 0.32, 0.42, 0.063, 0.045, 0.1, 0.026, 0.022, 0.21, 0.18 | 61 | |
IGROVI | 202c | 1.6 | 77 | |
SK-OV-3 | 202c | 2.4 | 77 | |
Ehrlich ascites tumor | 185, 186, 187, 188 | 34.4i, 48.8i, 77.7i, 28.8i | 73 | |
MCF-7 | 149, 150, 151, 152, 153, 154 | 3.7, 4.5, 3.5, 3.7, 2.9, 1.9 | 68 | |
SH-SY5Y | 149, 150, 151, 152, 153, 154 | 4.7, 4.7, 1.0, 4.6, 4.8, 4.6 | 68 | |
Hep-G2 | 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165 | 2.9, 4.2, 1.9, 3.7, 3.6, 3.7, 6.1, 5.6, 16.6, 29.2, 12.0, 25.4, 17.2, 1.0, >100, 10.4, 11.0 | 68 and 69 | |
A16-F10 | 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165 | 5.1, 3.2, 10.7, 12.2, 7.6, 18.4, 9.7, 0.7, >100, 3.2, 7.0 | 69 | |
EGFR | 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165 | 2.2, 1.5, 6.8, 7.1, 5.6, 15.3, 6.1, 0.4, 20.8, 4.6, 5.3 | 69 | |
H460 | 149, 150, 151, 152, 153, 154 | 7.0, 7.7, 3.0, 6.7, 7.6, 7.2 | 68 | |
P388 cells IC50 (mM) | 212, 123, 124, 125, 126, 127, 128, 129, 130 | 0.77, 0.623, 0.755, 0.312, 0.371, 0.919, 0.477, 0.399, 0.221 | 66 | |
L1210 cells IC50 (mM) | 212, 123, 124, 125, 126, 127, 128, 129, 130 | 7.96, 5.56, 94.1, 32.7, 44.2, 57.1, 9.79, 9.67, >20d | 66 | |
Molt 4/C8 cells IC50 (mM) | 212, 123, 124, 125, 126, 127, 128, 129, 130, 214, 215, 217, 218, 219, 226 | 1.67, 1.94, 2.3, 9.71, 32.1, 29.2, 4.56, 1.67, >20d, 0.02, 0.03, 0.1, 0.5, 0.3, 0.7 | 66 and 81 | |
CEM cells IC50 (mM) | 212, 123, 124, 125, 126, 127, 128, 129, 130 | 1.70, 1.58, 32.3a, 6.35, 23.0, 30.0, 4.60, 1.92, >20d | 66 | |
Ant-angiogenesis | HUVEC | 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85 | 2.87, 7.06, 1.27, 1.44, 13.3, 1.50, 0.37, 0.76, 0.40, 0.46, 2.97, 0.75, 0.38, 0.38 | 60 |
B16 | 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85 | 11.48, 3.62, 1.43, 1.09, 7.03, 4.41, 0.82, 0.59, 12.81, 1.58, 3.88, 4.83, 5.46, 6.93 | 60 | |
Vero | 72, 73, 74, 75, 76, 77, 81, 82, 83, 84, 85 | 6.02, 14.48, 2.04, 3.47, 3.55, 3.47, 3.48, 27.22, 2.48, 3.11, 3.98 | 60 | |
U87 | 72, 73, 74 | 7.71, 5.56, 8.31 | 60 | |
SiHa | 72, 73, 74, 75, 77, 78, 79, 80, 81, 82, 83, 84, 85 | 12.33, 15.75, 3.94, 5.96, 7.71, 4.57, 2.30, 34.43, 3.56, 25.28, 17.67, 0.76, 6.57 | 60 | |
SVR cell line | 86, 87, 88, 89, 90, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117 | 98.2g, 97.5f, 94.4f, 94.7f, 96.9f, 58.5f, 59.2f, 89.6f, 89.5f, 87.3f, 73.4f, 96.9f, 93.7f, 85.2f, 88.7f, 60.8f, 63.5f, 96.9f, 85.2f 88.6f, 75.4g, 96.8f,62.9f, 69f, 1.3f, 68.2f, 80.8f, 68.9f, 78.4f | 62 and 64 | |
PC-3 | 167a, 169a, 175a, 179a, 180a, 81a, 182a, 172a, 184a, 193a, 194a, 195a, 190a, 214, 215, 217, 218, 219, 226 | 9.5, 3.9, 5.9, 2.9, 8.8, 2.1, 4.6, 3.6, 0.35, 5.1, 6.1, 2.4, 10.3, >5, >5, 6.9, 0.28, 0.5, 1.5 | 51 and 72 | |
LNCaP | 167a, 169a, 175a, 179a, 181a, 182a, 172a, 193a, 194a, 195a | 5.8, 2.7, 2.6, 2.2, 0.5, 1.7, 2.5, 5.1, 2.4, 1.9 | 51 | |
MCF-7 | 167a, 169a, 175a, 179a, 181a, 182a, 172a, 193a, 194a, 195a | 6.9, 2.4, 2.9, 2.5, 0.4, 2.4, 1.7, 3.5, 6.6, 1.5 | 51 | |
MDA-MB-231 | 167a, 169a, 175a, 179a, 181a, 182a, 172a, 193a, 194a, 195a | 3.9, 2.8, 2.1, 1.6, 0.6, 2.4, 2.7, 4.2, 1.7, 0.6 | 51 | |
203c, 204c, 205c, 206c, 207c, 208c, 209c, 210c, 211c, 180c, 169c, 203f, 204f, 207f, 210f, 180f, 182f, 203d, 204d, 180d, 204b | 1.1, 1.5, 3.3, 3.2, >30, >30, >30, 1.9, >10, >30, 2.6, 0.8, 2.1, >30, >30, 0.3, 3.8, 1.1, 12.9, >30, 8.6 | 78 | ||
NF-κB activation | 203c, 204c, 205c, 206c, 207c, 208c, 209c, 210c, 211c, 180c, 169c, 203f, 204f, 207f, 210f, 180f, 182f, 203d, 204d, 204d, 204b | 2.5, 2.5, 2.2, 3.8, >200, >200, 140, 48, >200, NT, 10, 1.0, 0.8, >200, 47, 0.9, 4.0, 1.5, 4.0, >200, 34 | 78 | |
CNE | 169a, 169c, 178b | 6.8, 17.6, 2.4 | 72 | |
HL-60 | 184a | <10 | 72 | |
KB | 169c, 178b, 184a, 189a, 214, 215, 217, 218, 219, 226 | <10.10, 3.3, <3.70, 24.5, 4.4, >5, 1.6, 0.6, 0.5, 2.5 | 72 and 81 | |
LS 174T | 169a, 169c, 283a | 3.7, 4.6, 4.0 | 72 | |
HeLa | 190a, 214, 215, 217, 218, 219, 226 | 6.2, 0.4, 0.5, 1.9, 0.7, 0.4, 3.6 | 72 and 81 | |
HEK | 131, 132 + 133, 134 + 135, 136 + 137 | 24, 63, 20, 25 | 54 | |
Alzheimer | Aβ | 196a, 197b, 198c, 199c, 200c, 201c | 21.5, 12.6, 6.0, 2.5, 17.6, 17.2 | 76 |
Osteoporosis | TRAP | 118, 119, 120, 121, 122 | 19.79, 9.79, 16.81, 6.45, 2.50 | 65 |
Anti-haemolysis | DPPH | 169a, 167a, 173a, 174a, 169b, 167b, 173b, 174b, 169c, 167c, 173c, 174c | 45.65, >300, 9.63, 38.07, 101.8, >300, 9.52, 15.8, 68.83, >300, 13.11, 53.66 | 42 |
M. tuberculosis | H37Ra strain | 131, 200 | 25, 25 | 55 |
Antitrypanosomal EC50 (μM) | T. brucei strain 427 | 131, 132 + 133, 134 + 135, 200, 202, 136 + 137 | 0.053, 0.75, 0.14, 4.1, 0.22, 0.089 | 54 |
T. brucei ΔTbat1 | 131, 132 + 133, 134 + 135, 136 + 137 | 0.082, 0.73, 0.29, 0.079 | 54 | |
T. brucei clone B48 | 131, 132 + 133, 134 + 135, 136 + 137 | 0.023, 0.30, 0.032, 0.045 | 54 | |
Antimalarial | P. falciparum (D6 clone) | 214, 215, 218, 219, 220, 222, 223 | 0.5, 0.4, 0.69, 1.7, 0.5, 0.4, 0.3 | 81 |
P. falciparum (W2 clone) | 214, 215, 218, 219, 220, 222, 223 | 0.5, 0.6, 0.6, 2.8, 0.5, 0.37, 0.4 | 81 | |
Antileishmanial | L. major promastigotes | 131, 132 + 133, 134 + 135, 136 + 137 | 8.9, 10, 13, 7.5 | 54 |
L. mexicana amastigotes | 131, 132 + 133, 134 + 135, 136 + 137 | 17, 7.4, 28, 29 | 54 | |
Gram-positive anti-bacterialh | S. aureus | 167a, 169a, 169b, 169c, 180a, 189a, 189b, 189c | 17, 15, 12, 11, 14, 18, 12, 15 | 82 |
S. aureus | 167a, 169a, 169b, 169c, 180a, 189a, 189b, 189c | 17, 15, 12, 11, 14, 18, 12, 15 | 82 | |
Gram-negative anti-bacterialh | Enterobacter cloaceae | 167a, 167b, 169a, 169b, 169c, 176a, 176b, 176c, 180a, 180b, 180c, 189a, 189b, 189c | 11, 10, 12, 11, 10, 10, 9, 11, 11, 10, 10, 10, 10, 11 | 82 |
Um et al. synthesized the unsymmetrical curcumin mimics with various amide moieties (72–85) and evaluated for MDR reversal activities in MDR cell line KBV20C. SAR revealed that chlorobenzamide curcumin mimics (79–83) showed strong MDR reversal activity indicating that one chloro group at the meta or para-position on benzamide resulted can increase the activity. Furan carboxamide (84) and a thiophene carboxamide (85) showed only mild MDR reversal activity.61
Transformation of curcumin into asymmetric monocarbonyl analogues with enone or dienone (86–90) moiety, helps to boost the inhibition of angiogenesis process even better than curcumin.62,63
The presence of ortho-hydroxyl group is significant for the activity. Moving the hydroxyl group from ortho to para-position (91–93), in hydroxy-substituted derivatives cytotoxicity decreased. In case of asymmetric monocarbonyl analogues, replacement of hydroxyl group by methyl (94–98), chloro (99, 101, 103, 109, 111), trifluromethane (100), methoxy (102, 104, 105), flouro (110) methylenedioxy (115–117) led to decrease in activity. Introduction of heteroaromatic ring (106–107) or naphthalene (108–109) or anthracene (112–114) ring replacing the aryl ring, decreased the cytotoxic activity in asymmetric monocarbonyl analogues.64
(1) Triazole analogues (118–121) of curcumin exhibited moderate to strong inhibitory activity against the osteoclastogenesis induced by RANKL. Substitution of hydrogen with fluoride in the phenyl group of (121) and (122) resulted in an improved activity. To conclude (3-hydroxy-4-methoxyphenyl)propenone was found to be a promising template to develop novel antiresorptive agents to treat osteoporosis.65
(2) A novel series of cytotoxic N-[4-(3-aryl-3-oxo-1-propenyl)phenylcarbonyl]-3,5-bis(phenylmethylene)-4-piperidones (123–130). The synthesized compounds showed marked cytotoxicity and selective toxicity towards different tumour cell lines along with lack of murine toxicity.66
(3) Chatchawan et al. synthesized curcuminoids (131–137) as antitrypanosomal agents. Compound 131 exhibited very high antitrypanosomal activity, which was 47-fold more active than the curcumin. Therefore, the compound 131 was taken as a lead molecule and further modification were done on this molecule. However, none of them showed higher activity than the lead 131.54
(4) SAR study of C5-curcuminoids (138–148) for their cytotoxicities against human colon cancer cells indicates that (i) bis(aryl-methylidene) acetone serves as the most promising skeleton for eliciting cytotoxicity. (ii) The 3-oxo-1,4-pentadiene structure is essential for eliciting cytotoxicity. (iii) Hexasubstituted compounds (143–147) exhibited strong activities. (iv) para-Positions allowed the introduction of additional functional groups for use as the molecular probes.67
(5) Woo et al. synthesized novel benzimidazolyl curcumin mimics library (149–154). The synthesized compounds were screened against various cancer cell lines. The MTT assay of the cancer cells MCF-7, SH-SY5Y, HEP-G2, and H460 showed that compound 151 with IC50 of 1.0 and 1.9 μM has a strong inhibitory effect on the growth of SH-SY5Y and Hep-G2 cells, respectively, and that compound 154 with IC50 of 1.9 μM (Table 5) had a strong inhibitory effect on the growth of MCF-7 cancer cells. A comparison of the growth inhibitory effect of each group of curcumin mimics showed that the positional variation of the methoxy and hydroxyl functionality did not affect the potency of the anticancer activity. From SAR study, it was found that the introduction of the benzimidazole moiety increased the potency by 10–50 times.68
(6) Xu et al. synthesized α,β-unsaturated cyclohexanone analogous of curcumin and tested for antiproliferative activity against two human tumor cell lines (Hep G2 and B16-F10). The result showed that the compounds 156 and 162 displayed the most potent EGFR inhibitory activity (IC50 = 0.43 μM and 1.54 μM, respectively). The SAR study revealed that the compounds with hydroxyl substitution on R-phenyl ring (162 and 164) displayed more potent antiproliferative activity compared to other compounds. While compounds with methoxyl group on R-phenyl ring (161 and 163) showed the significant effects on the antiproliferative activity. Compounds with halogen substitution on R-phenyl ring, the activity order was as Cl ≥ F > Br in the series. Compounds with R1 substitution at meta-position (155, 157, 161 and 165) showed better antiproliferative activity than the compounds substituted at para-position (158, 159, 160 and 164).69
(7) Eliminating one carbonyl and one methylene from curcumin yields less active derivatives. But, the antioxidant potential of monocarbonyl analogues (symmetrical, asymmetrical and cyclic) retained only with the analogues possessing hydroxyl substituents (91–93, 166–169) and others were less active or inactive than curcumin. The analogues with ortho-alkyl groups to para-hydroxy group are more potent than curcumin but the increase in the bulkiness of the substituents group adjacent to the para-hydroxy group had a negative influence on the activity (170–172). The dimethoxy analogues are the most active of all monocarbonyl analogues.70,71
(8) The analogues with para-hydroxy substitution were more active while replacing the para-hydroxy group (173–174) to methoxy (175, 179–182) or sulphamate (178–180) (Table 6) did not cause any increase in cytotoxicity. Increasing the number of methoxy substituents on phenyl ring in case of symmetric, monocarbonyl analogues (181, 182), resulted in an incredible rise in selectivity against cancer cells.51 In case of symmetric monocarbonyl analogues, replacement of phenyl ring with any heteroaromatic ring (189–191) gave broad spectrum cytotoxic activity.72 The compound with cyclopropoxy (185–188) group at para-position and methoxy group at ortho- and meta-positions, showed potent antitumor and anti-angiogenic activities on mouse Ehrlish ascites tumor (EAT) in vivo, whereas the absence of methoxy group at meta-position decreased the tumor inhibitory effect.73
(9) The properties and position of the substituents and the space of the linking chain determine the anti-inflammatory activities. The compounds with cyclohexanone moiety were more effective than compounds with acetone and cyclopentanone moieties.74 Monocarbonyl analogues having a long chain allyl-oxyl substituent group or 3-(dimethylamino)propoxyl substituent group exhibited enhanced activity than curcumin (177, 178), while analogues possessing dimethylamino (176) or trifluromethane (183, 184) substituent group exhibited less activity. Fluoro substitution in cyclohexanone analogue of monocarbonyl curcumin enhanced the activity (192) while, nitro, tert-butyl substitution decreased anti-inflammatory activity. In case of bromo substitution, contradictory results were obtained. When substitution was at 2-position, no activity was observed but substitution at 3-position, anti-inflammatory activity comparable to curcumin was obtained. The monocarbonyl analogues possessing 3-methyoxy group showed superior activity than curcumin, suggesting that the presence of a 3-methyoxy group is critical for the activity. Analogues with trimethoxy substitution possessed higher activity than curcumin (181, 182). On the other hand, heteroaromatic ring substituted monocarbonyl analogues showed moderate anti-inflammatory activity (LPS induced TNF-α, IL-6 expression) (189–191).45,48,74,75
(10) Chen et al. synthesized multifunctional agents (196–198, 199–201) for the treatment of Alzheimer disease (AD). The in vitro studies showed that these compounds had better inhibitory properties against Aβ aggregation than curcumin. Structure–activity relationships suggested that introduction of flexible moieties at the linker is crucial to the inhibitory potencies of the compounds against Aβ aggregation compound 199 was found to be the most potent with IC50 value of 2.5 μM.76
(11) Difluorinated analogue of curcumin (202) showed cytotoxicity against IGROV1 and SK-OV-3 human ovarian cancer cells.77 Yadav et al. synthesized cyclohexanone analogues (203–212) of curcumin and developed a SAR study regarding their cytotoxic potential and observed that cyclohexanone analogues showed five-fold decreased in activity than cyclopentanone analogues. The analogues with fluorine substituted pyridine rings were found to be 2–3 times less active than the parent pyridine compounds 203 and 204. Replacement of the pyridine ring with different heterocyclic rings, thiophene 207, N-methylpyrrole 208, N-methylindole 209 and 4-substituted N-methylimidazole 211 exhibited no activity.78 Compound 213 as demethylating agent has been found several fold more potent than curcumin in pancreatic cancer cell lines and displayed better solubility and bioavailability. It inhibits HSP-90 and NF-κB leading to downregulation of DNA methyltransferase-1 (DNMT-1) expression.79 In addition, it was also evaluated for the treatment of colorectal cancer using colorectal cancer cells HCT-116 and HT-29. It significantly inhibited the VEGF synthesis and secretion in both colon cell lines in concert with the loss of Hypoxia-inducible factors (HIFs) expression, which transcriptionally regulate VEGF.80
(12) Substitution of the pyridyl groups with electron rich trimethoxyphenyl and dimethoxyphenyl give analogues 180 and 181 but they did not show any increase in cytotoxicity and NF-κB activation activity, N-methylpiperidone analogues had similar or slightly increased activity over cyclohexanone core. The more sterically hindered tropinone analogues showed reduced activity in all cases, indicating that bulky groups in this area of the molecule led to decreased activity. Additionally, the incorporation of a fluorine substituent into the aromatic rings did not increase the activity of these compounds.78
(13) Manohar et al. synthesized a series of monocarbonyl analogues of curcumin and evaluated for the antimalarial and cytotoxic activity against Molt4, HeLa, PC3, DU145, and KB cancer cell lines. It has been found that six analogues showed potent cytotoxic activity against these cell lines with IC50 values below 1 μM, which was better than the standard drug doxorubicin. The SAR revealed that the rigidity did not play important role in the activity as compound 214, 217, 219, and 226 having different ring sizes at the carbonyl position were showed very good activity with the IC50 in the low micro molar range in HeLa cells. However, some other compounds (216, 221, 224, and 225) have the same five- or six-member ring did not show cytotoxicity even at 50 μM. The length of side chain did not have a major effect on the cytotoxic activities; methoxy group on the phenyl ring favoured the inhibitory effect. These analogues were also screen for their antimalarial activity against CQ-sensitive (D6 clone) and CQ-resistant (W2 clone) strains of P. falciparum. Compounds 214, 215, 218, 220, 222, and 223 having the amine probe via ethylene/butylene linker with cyclopentanone ring showed better activity with IC50 ranging from 0.35–0.69 μM in comparison to standard drug chloroquine.81
(14) Monocarbonyl analogues were more effective against Gram-positive organisms than against Gram-negative. The cyclopentanone and cyclohexanone analogues (167, 169, 176, 180, and 189) showed moderate activity; however, had low activity than curcumin, which illustrated that the space structure of the linking C-strain can make some influence on the anti-bacterial ability. Compounds with cyclohexanone and acetone moiety were more effective than compounds with cyclopentanone moieties. But, the most of the compounds exhibited better activity against the Gram-negative E. cloacae which is completely resistant to the clinical drug ampicillin.82
Fig. 3 Chemical structures of curcumin analogues with changes in the β-diketone structure, dicarbonyl analogues (Class 2.2). |
Activity | Assay/target | Compound(s) | ED50 (μM) | Ref. |
---|---|---|---|---|
a Indicates % inhibition at 1 μM. | ||||
Cytotoxic | KB | 230, 231, 232, 233, 234, 235, 236, 237 | 16.5, 20.0, 19.5, 19.7, 15.5, 3.8, 14.4, 4.5 | 83 |
A549 | 227, 228, 231, 233, 234, 235, 236, 237 | 20.0, 20.0, 16.5, 17.5, 18.0, 4.4, 18.8, 8.9 | 83 | |
MCF-7 | 227, 228, 230, 231, 233, 234, 235, 236, 237 | 20.0, 14.5, 15.0, 13.5, 17.5, 15.0, 7.0, 16.7, 4.0 | 83 | |
IA9 | 227, 228, 229, 230, 231, 233, 234, 235, 236, 237 | 6.0, 4.8, 3.9, 4.0, <10, 5.0, 3.9, <0.63, 13.6, 2.1 | 83 | |
HCT-8 | 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237 | 7.5, 7.5, 10.0, 12.0, 5.8, 8.0, 9.7, 6.0, 1.8, 18.3, 3.0 | 83 | |
SK-MEL-2 | 230, 234, 235, 237 | 16.0, 7.0, 2.0, 6.0 | 83 | |
U-87-MG | 230, 234, 235, 236, 237 | 16.3, 15.0, 3.8, 18.8, 8.7 | 83 | |
HOS | 227, 228, 229, 230, 232, 233, 234, 235, 236, 237 | <10, <10, 16.0, 8.0, 9.0, 10.0, 12.0, 0.97, 18.0, <2.5 | 83 | |
PC-3 | 230, 234, 235, 237 | 16.0, 14.0, 5.0, 8.0 | 83 | |
KB-VIN | 230, 232, 234, 235, 237 | 16.5, 19.5, 15.0, 4.5, 7.4 | 83 | |
DU-145 | 238 | 40a | 83 |
(1) The pyrazole analogues of curcumin demonstrated increase in antioxidant, COX-2 inhibitory activity, antitumor and antimalerial activity as compared to curcumin, whereas the isoxazole analogues also showed increase in activity as compared to curcumin but not as better as pyrazole analogues (239–244) (Table 8).86
General structure for Class 2.3 compounds (260–278) | ||||
---|---|---|---|---|
Compounds | R1 | R2 | R3 | R4 |
260 | OCH3 | OH | OCH3 | OH |
261 | OCH3 | CH2–(CH2)3–CH3 | OCH3 | H |
262 | OCH3 | H | OCH3 | CH2–(CH2)3–CH3 |
263 | OCH3 | CH2–(CH2)3–CH3 | OCH3 | CH2–(CH2)3–CH3 |
264 | OCH3 | CH2CHC(CH3)2 | OCH3 | H |
265 | OCH3 | H | OCH3 | CH2CHC(CH3)2 |
266 | OCH3 | CH2CHC(CH3)2 | OCH3 | CH2CHC(CH3)2 |
267 | OCH3 | CH2CH3 | OCH3 | H |
268 | OCH3 | H | OCH3 | CH2CH3 |
269 | OCH3 | CH2CH3 | OCH3 | CH2CH3 |
270 | OCH3 | Ac | OCH3 | H |
271 | OCH3 | H | OCH3 | Ac |
272 | OCH3 | Ac | OCH3 | Ac |
273 | OCH3 | Me | OCH3 | H |
274 | OCH3 | H | OCH3 | CH3 |
275 | OCH3 | CH3 | OCH3 | CH3 |
276 | H | CH3 | OCH3 | CH3 |
277 | OCH3 | CH3 | H | CH3 |
278 | H | CH3 | H | CH3 |
(2) Both pyrazole (239) and isoxazole (243) analogues having two methoxy group showed better antioxidant activity than curcumin while the other analogues (240 and 244) were not as much active. The pyrazole analogues proved to be better than the isoxazole analogues (239 and 242).86
(3) It has been postulated that pyrazole analogues increased COX-1 inhibitory activity slightly as compared to curcumin but there was a two-fold increase in COX-2 inhibition. Thus pyrazole analogue showed significant enhancement in the selectivity towards COX-2 enzyme (COX-2/COX-1 = 0.70) compared to curcumin (COX-2/COX-1 = 0.43). Since these compounds exhibited good COX inhibitory and antioxidant activities, they were investigated for in vivo anti-inflammatory activity using carrageenan induced rat paw edema assay at 75 mg kg−1. Among these compounds, pyrazole analogue of curcumin, exhibited the highest activity. Molecular docking studies further supported the strong inhibitory activity of pyrazole analogue compared to curcumin and isoxazole analogues (239–244).86
(4) Pyrazole analogue (239–241) of curcumin showed a broad spectrum cytotoxic activity against KB, A549, CAKI-1, MCF-7, 1A9, HCT-8, SK-MEl-2, U-87-MG, HOS, PC-3, LNCaP, MDA-MB-231, KB-VIN, HepG2, and LNCaP (clone FGC) cell lines (Table 9), where as isoxazole analogue (243) showed better antitumor activity than curcumin in the hepatocellular carcinoma HA22T/VGH cell line, which is known to innately produce remarkable amounts of drug resistance and anti-apoptotic factors.51,83,85
Activity | Assay/target | Compound(s) | IC50 (μM) | Ref. |
---|---|---|---|---|
a * indicates % inhibition at 100 μM; ** indicates IC50 values in nM; # indicates ED50 value; NA, not active (MIC > 200 mg ml−1); ND, not determined. | ||||
Antioxidant | DPPH | 239, 240, 243, 244 | 9.7, 15.45, 10.71, 18.96 | 86 |
Anti-inflammatory | COX-1 | 239, 240, 243 | 87.0*, 73.9*, 80.8* | 86 |
COX-2 | 239, 240, 241, 243, 244 | 61.0*, 43.8*, 35.8*, 58.1*, 35.0* | 86 | |
Cytotoxic | KB | 239, 240, 241 | 1.0#, 9.0#, 10.7# | 83 |
A549 | 239, 240, 241 | 1.7#, >20#, 20# | 83 | |
CAKI-1 | 239 | 2.4# | 83 | |
MCF-7 | 239, 240, 241, 242, 243 | 3.9#, 8.8#, 13.5#, 15.8, 13.1 | 51,83 and 85 | |
MCF-7R | 243 | 12.0 | 85 | |
IA-9 | 239, 240, 241 | 1.8#, 4.2#, 1.2# | 83 | |
HCT-8 | 239, 240, 241 | 1.9#, 7.0#, 10.0# | 83 | |
SK-MEL-2 | 239, 240, 241 | 2.4#, 13.6#, 14.1# | 83 | |
U-87-MG | 239, 240, 241 | 2.4#, 14.0# | 83 | |
HOS | 240, 241 | 10.2#, 10.5# | 83 | |
PC-3 | 239, 240, 241, 242 | 2.8#, 13.0#, 15.0#, 16.2 | 51 and 83 | |
KB-VIN | 240, 241 | >20#, 9.5# | 83 | |
HepG2 | 239 | 2.5# | 83 | |
LNCaP clone FCG | 239 | 2.0# | 83 | |
LNCaP | 239, 242 | 3.4, 12.1 | 51 and 83 | |
HA22T/VGH | 243 | 12.8 | 85 | |
MDA-MB-231 | 239, 242 | 6.6, 20.4 | 51 | |
BAECs** | 239, 240, 241, 255, 256, 257 | 0.52, 1.8, 5.8, 9.3, 2.4, 8.7 | 88 | |
Antimalarial | P. falciparum (strain FCK2) | 239, 243, 245, 246, 247, 248 | 0.48, 8.44, 8.48, 2.42, 0.87, 4.65 | 57 |
P. falciparum (strain MP-14) | 239, 246, 247 | 3.9, 14.3, 4.9 | 57 | |
Inflammatory bowel disease | MMP-9 activity in Caco-2 cell line | 239, 258, 259 | 19.4, 17.6, 8.0 | 84 |
M. tuberculosis | H37Ra strain | 239, 240, 241, 254, 243, 249/244 (1:1), 250, 260, 279, 280, 261, 262, 263, 264/265 (2:1), 266, 267/268 (5:2), 269, 270, 271, 272, 273, 274, 275, 276/277 (1:1), 278, 251, 253 | 200, 100, 25, 25, 1.56, 12.5, 12.5, 6.25, ND, 100, 0.39, 6.25, NA, 1.56, ND, 12.5, ND, 1.56, 1.56, 1.56, 0.09, 0.78, 0.39, 100, ND. 6.25, 200 | 55 |
(5) Pyrazole analogues demonstrated moderate increase of anti-malarial potency against CQ-S and CQ-R. Pyrazole derivative of curcumin inhibited the CQ-S and CQ-R P. falciparum cultures at nanomolar concentrations. Electron withdrawing substituents at N-phenyl curcumin pyrazole system affect the anti-malarial potency of corresponding compounds (245–248). Previous reports showed that electron-withdrawing groups enhanced the biological activity, while electron donating groups decreased the activity. In N-(substituted) phenylcurcumin pyrazole analogues, 3-nitrophenylpyrazole curcumin (247) was found to be most potent.57
(6) Pyrazole analogues of the curcumin showed enhanced inhibition of matrix metalloproteinases on Caco-2 cell line (239, 258, and 259).84
(7) The isoxazole analogues (249–254, 260–280) were synthesized and evaluated for their antimycobacterial activity. The most active class of analogues, with mono-O-methylcurcumin isoxazole (273) being the most active compound (MIC 0.09 mg ml−1). It was 1131-fold more active than curcumin (1). The structural requirements for a curcuminoid analogue to exhibit antimycobacterial activity are the presence of an isoxazole ring and two unsaturated bonds on the heptyl chain. The presence of a suitable para-alkoxyl group (270–274) on the aromatic ring which is attached in close proximity to the nitrogen function of the isoxazole ring and a free para-hydroxyl group (261) on another aromatic ring enhanced the biological activity.55
(8) Cristina et al. synthesized pyrazole analogues (281–286) of curcumin. The SAR of aggregation inhibitors based on curcumin, revealed that the predominant features affecting inhibition of amyloid aggregation is the co-planarity of the two aromatic rings and the distance between them (from 8 to 16 Å). On the other hand, the different chains present on the pyrazole ring did not contribute to the interaction. However, the formation of pyrazole moiety, locks the keto–enol tautomerism in an enol type arrangement, important for its Aβ-binding capability.87
(9) A novel hydrazino curcuminoids (239–241) and hydrazinobenzoyl (255–257) curcuminoids were synthesized and evaluated for anti-angiogenic activity using bovine aortic endothelial cells (BAECs). Among the six analogues tested, the compound 239 showed the most potent growth inhibitory activity against BAECs with an IC50 of 0.52 nM. Compound 256 also showed an enhanced anti-proliferative activity against BAECs. However, the potency of 256 was relatively weaker than that of 239.88
Curcumin was converted into diarylheptylamine and the synthesized new analogues were evaluated for (i) inhibition of iNOS expression; (ii) inhibition of NO production; (iii) inhibition of biosynthesis of COX-2 downstream product PGE2; and (iv) cytotoxicity (Table 10). These analogues exhibited moderate inhibition of NO and iNOS, whereas they showed significant inhibitory activity of PGE2 (289–292) (Fig. 4).90
Activity | Assay/target | Compound(s) | IC50 (μM) | Ref. |
---|---|---|---|---|
a * indicates % inhibition at 10 μM; ** indicates oxidation potential; $ indicates % inhibition at 1 μg ml−1. | ||||
Antioxidant | Lipid peroxidation | 287 | 85* | 89 |
DPPH assay | 287 | 0.28** | 89 | |
Anti-inflammatory | LPS-induced PGE2 production | 289, 290, 291 | 1.4, 1.0, 7.4 | 90 |
292 | 16.0 | |||
LPS-induced NO production | 289, 290, 291 | 72, 100, 68 | 90 | |
292 | 62 | |||
Cytotoxic | HA22T/VGH | 288 | 5.9 | 85 |
MCF-7 | 287, 288 | 74$, 7.1 | 85 | |
MCF-7R | 288 | 9.3 | 85 |
Fig. 5 Chemical structures of curcumin analogues with changes in the β-diketone structure, change in the conjugation (Class 2.5). |
The antimycobacterial activity of compound (5–8) against M. tuberculosis H37Ra strain was less than isoxazole analogues of curcumin. This reflected that structural requirements for the curcuminoid analogues to exhibit antimycobacterial activity are the presence of an isoxazole ring and two double bonds on the heptyl chain.55
The parent curcuminoids exhibited low antitrypanosomal activity (T. brucei strain 427) (Table 11). The number and nature of the oxygen functions on the aromatic rings strongly determined the antitrypanosomal activity of the tetrahydro analogues. Compound 295 was almost 9-fold more active than compound 294. Compound 296 was even 42-fold more active than the methylated analogue 6.54
Activity | Assay/target | Compound(s) | IC50 (μM) | Ref. |
---|---|---|---|---|
a * indicates % inhibition at 10 μM; ** indicates % inhibition at 10 μM; NA, not active (MIC > 200 mg ml−1); ND, not determined. | ||||
Antioxidant | TRAP assay | 6 | 94* | 70 |
FRAP assay | 6 | 60* | 70 | |
Lipid peroxidation | 6 | 1.83 | 40 | |
DPPH assay | 6, 7, 8 | 18.22, 21.6, 23.6 | 40 | |
ABTS+ assay | 6 | 2.52 | 40 | |
Anti-inflammatory | LPS-induced iNOS expression | 6, 7, 8 | 62**, 64**, 67** | 91 |
LPS-induced iNOS Promoter activity | 6, 7, 8 | 42**, 34**, 40** | 91 | |
COX-1 | 293 | 1.14 | 44 | |
COX-2 | 293 | 5.13 | 44 | |
cPLA2 | 6 | 40# | 90 | |
5-LOX | 6 | 2.99 | 91 | |
M. tuberculosis | H37Ra strain | 5, 6, 7, 8 | 200, 50, 100, NA | 55 |
Antitrypanosomal | T. brucei strain 427 | 6, 7, 8, 294, 295, 296, 297 + 298, 299, 300 | 21, 17, 78, 22, 2.5, 0.50, 3.07, 1.9, 33 | 54 |
T. brucei ΔTbat1 | 6, 7, 8, 294, 295, 296, 297 + 298, 299, 300 | 37, 38, 61, 34, 4.0, 0.52, 2.9, 1.5, 22 | 54 | |
T. brucei clone B48 | 6, 7, 8, 294, 295, 296, 297 + 298, 299, 300 | 12, 5.9, >100, 18, 5.1, 0.43, 2.5, 1.7, 19 | 54 | |
Antileishmanial | L. major promastigotes | 6, 7, 8, 294, 295, 296, 297 + 298, 299, 300 | 90, >100, >100, >100, 80, 11, >100, >100, 41 | 54 |
L. mexicana amastigotes | 6, 7, 8, 294, 295, 296, 297 + 298, 299, 300 | 43, >100, >100, >50, 64, 16, 40, 53, >100 | 54 | |
Cytotoxic | HEK | 6, 7, 8, 294, 295, 296, 297 + 298, 299, 300 | 120, 370, ND, ND, 350, 130, 77, 150, 150 | 54 |
General structure for Class-2.6 compounds (301–325) | |||||
---|---|---|---|---|---|
Compound(s) | R1 | R2 | R3 | R4 | R5 |
301 | CH3 | H | H | H | H |
302 | CH3 | H | OCH3 | OCH3 | H |
303 | CH2C6H5 | H | H | H | H |
304 | H | H | OCH3 | OH | H |
305 | H | H | OCH3 | OCH3 | H |
306 | H | H | H | H | H |
307 | H | H | H | H | H |
308 | CH3 | H | OCH3 | OH | H |
309 | CH2CHCH2 | H | OCH3 | OH | H |
310 | SC(S)OEt | H | OCH3 | OH | H |
311 | CH2CHCH2 | H | OCH3 | OCH3 | H |
312 | OCOCH3 | H | H | OH | H |
313 | OCOCH3 | H | H | OCH3 | H |
314 | OCOCH3 | H | H | H | H |
315 | CH2CH2COOEt | H | OCH3 | H | H |
316 | CH2CH2COOEt | H | H | OCH3 | OCH3 |
317 | CH2CH2COOEt | H | OCH3 | OCH3 | OCH3 |
318 | CH2CH2COOEt | H | N(CH3)2 | OCH3 | H |
319 | CH2CH2COOEt | H | OCH3 | OH | H |
320 | CHCHCOOEt | H | OCH3 | OTHP | H |
321 | CHCHCOOEt | H | OCH3 | OCH3 | H |
322 | CHCHCOOMe | H | OCH3 | OCH3 | H |
323 | CHCHCONHEt | H | OCH3 | OCH3 | H |
324 | CHCHCN | H | OCH3 | OCH3 | H |
325 | CHCHCH2OH | H | OCH3 | OCH3 | H |
Substitution at central methylene carbon with 4-ethoxycarbonylethyl ester enhances the anti-androgen activity (315–319 and 329). The replacement of ethoxy with methoxy or N-ethylamino resulted in loss of activity, which implied that a long chain ester may be more favourable. The reduction of ester to alcohol also led to loss of activity, emphasizing the necessity for an ester in the side chain. The nitrile functional group did not enhance the anti-androgen activity or the cytotoxicity. The –CHCHCOOEt side chain is superior to –CHCHCOOMe, –CH3, –CHCONHEt, –CHCHCN, and –CHCHCH2OH (320–325). The monoalkylated analogues were more potent in inhibiting NF-κB than dialkylated analogues and curcumin.45,49,93,94
Replacement of an acidic proton from the central methylene with benzylidene derivatives proved to be as effective antimalarial as curcumin. The 4-hydroxy-3-methoxy-benzaylidene derivative of curcumin was more active than curcumin, 4-hydroxy-benzylidene derivative and benzylidene derivative (326–328). This suggested that the presence of methoxy group (electron donor group) at the meta position of 4-hydroxy-3-methoxy-benzylidene derivative of curcumin appears to play an important role for the potency of antimalarial compounds.57
Qiu et al. synthesized new 4-arylidene curcumin analogues as potential anticancer agents and were screened against A549 cell growth. Most of the 4-arylidene curcumin analogues (4-arylidene-1,7-bisarylhepta-1,6-diene-3,5-diones, 330–341) showed potent anticancer activities with a GI50 in the sub-micromolar range (0.23–0.93 μM), except 332, 333, 335, and 338 which exhibited slightly lower activities (GI50 of 1.12–2.62 μM) (Table 13). The potency of 4-arylidene analogues was improved by 10–60 folds over the parent compound curcumin in this assay. 4-Hydroxymethylene curcumin analogues (339–341), however, showed poor activity, suggesting the importance of the third arylvinyl moiety.56
Activity | Assay/target | Compound(s) | IC50 (μM) | Ref. |
---|---|---|---|---|
a * indicates % inhibition at 10 μM; ** indicates ED50 value in μg ml−1; *** indicates EC50 (μM); # indicates % inhibition at 1 μM; ## indicates % inhibition at 5 μM; $ indicates GI50 in μM. | ||||
Antioxidant | TRAP assay | 301, 303 | 52*, 55* | 70 |
FRAP assay | 303 | 39* | 70 | |
DPPH assay*** | 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314 | 22.6, >100, >100, >100, 23.2, 23.2, 29.7, >100, 26.3, 32.8, 47.8 | 92 | |
Antitumor | NF-κB | 338 | 6.3 | 45 |
DU145/wtAR | 315, 316, 317, 318 | 30–40#, 30–40#, 30–40#, 30–40# | 92 | |
PC-3/wtAR | 315, 302 | 30–40#, 5.2 | 92 | |
KB | 302 | 7.5** | 83 | |
A549$ | 302 | 7.9** | 83 | |
330, 331, 332, 333, 334, 335, 336, 337, 338 | 1.64, 0.93, 1.12, 2.62, 0.65, 2.00, 0.43, 0.69, 1.31 | 56 | ||
MCF-7 | 302 | 6.5** | 83 | |
IA9 | 302 | 1.1** | 83 | |
HCT-8 | 302 | 2.2** | 83 | |
SK-MEL-2 | 302 | 4.6** | 83 | |
U87-MG | 302 | 3.0** | 83 | |
HOS | 302 | 4.0** | 83 | |
KB-VIN | 302 | 6.7** | 83 | |
HepG2 | 302 | 5.4** | 83 | |
LNCaP clone FGC | 302 | 13.0** | 83 | |
Anti-androgenic | LNCaP | 319, 320, 321, 322, 323, 324, 325, 319, 329 | 96## & 1.5, 2.6, 94## & 0.2, 0.4, 0.6, 60## & 0.2, 0.2, 30, 160 | 49,93 and 94 |
PC-3 | 302, 319, 320, 321, 322, 323, 324, 325, 319, 329 | 3.5, 90## & 5.1, 3.1, 60## & 1.0, 0.2, 1.2, 30## & 0.7, 1.3, 6.4, 6.3 | 49,93 and 94 | |
Antimalarial | P. falciparum | 326, 327, 328 | 3.89, 5.85, 0.92 | 57 |
Activity | Target | Compound(s) | MIC | Ref. |
---|---|---|---|---|
a * indicates zone of inhibition in mM; ** indicates IC50 values in μM. | ||||
Antimicrobial | Micrococci | 343, 344, 345, 346, 347, 348, 349, 350 | 2.5, 5, 10, 10, 10, 10, 10, 5 | 95 and 96 |
Enterococcus | 349 | 5 | 96 | |
Klebsiella aeruginosa | 342, 343, 350 | 5, 5, 10 | 95 and 96 | |
Staphylococcus saprophyticus | 345, 349, 350 | 10, 5, 10 | 95 | |
Enterobacter cloaceae | 343, 345, 349, 350 | 2.5, 5, 10, 10 | 95 and 96 | |
Escherichia coli | 343, 349, 350 | 10, 10, 10 | 95 and 96 | |
Pseudomonas aeruginosa | 349, 350 | 2.5, 2.5 | 96 | |
Pseudomonas pyocynin | 349, 350 | 2.5, 5 | 96 | |
Staphylococcus epidermidis | 349, 350 | 10, 10 | 96 | |
Staphylococcus aureus | 349, 350 | 10, 5 | 96 | |
Enterococcus aerogen | 350 | 10 | 96 | |
Antifungal | Candida albincans | 349 | 24* | 96 |
DPPH-scavenging and anti-haemolysis** | DPPH | 351, 352, 353, 354, 345, 355, 356, 357, 346, 358 | 0.068, 0.082, 0.055, 0.058, 0.049, 0.048, 0.036, 0.092, 0.076, 0.098 | 97 |
β-carotene bleaching method | 351, 352, 353, 354, 345, 355, 356, 357, 346, 358 | 0.064, 0.077, 0.047, 0.06, 0.037, 0.043, 0.03, 0.086, 0.066, 0.091 | 97 |
The enhancement in the activity of these curcumin bioconjugates may be due to:
(i) Enhanced metabolic stability due to masking of phenolic hydroxyl groups and delay in their glucuronide formation during metabolism, (ii) better cellular uptake due to the transportation of the conjugate via the amino acid carrier protein, i.e., drug smuggling, and (iii) more solubility of the conjugates because of enhancement in polar character (hydrophilicity). The curcumin analogues with alkyl-substituted amino acids (351–358), such as alanine, valine, serine and cysteine, exhibited lower IC50 values than did curcumin in antioxidant assays. The enhancement in the antioxidant activities of curcumin amino acid conjugates could be principally attributed to the scavenging activity of the keto–enol group in curcumin. The phenolic hydroxyl of curcumin was substituted with amino acids blocking the route of the HAT (hydrogen atom transfer) mechanism of antioxidant activity. Here, the mechanism involved may be sequential proton loss electron transfer (SPLET), because the keto–enol moiety of curcumin has a more easily dissociable proton, and this is supposed to be faster than other mechanisms as the enolic proton is more acidic than the other two phenolic groups on curcumin.97
Activity | Assay/target | Compound(s) | EC50 (μM) | Ref. |
---|---|---|---|---|
a # indicates GI50, _aindicates no EC50 value could be obtained, _bindicates drug potency was too low to obtain an EC50 value, _cindicates drug increased cell number. | ||||
Anticancer | MDA-MB-231 cells | 362, 363, 367, 364, 361, 368, 365, 366, 369 | 2.55, _a, 8.64, _a, _a, 20.05, 1.54, 1.10, _c | 99 |
SKBr3 cells | 362, 363, 367, 364, 361, 368, 365, 366, 369 | 1.16, _a, 9.78, _b, _c, 6.21, 0.51, 0.23, 0.27 | 99 | |
HUVEC | 376, 377, 378, 379, 380, 381, 382, 383, 384, 385, 386, 387, 388, 389 | 2.2, 12.6, 2.0, 9.3, 7.2, 10.8, >50, >50, >50, >50, 9.2, 1.5, 22.8, 9.9 | 100 | |
Alzheimer | Aβ | 370, 371, 372, 373 | >50, >50, 37.8, >50 | 76 |
Anticancer | HCT-116 | 374, 375 | >50#, 22# | 67 |
Woo et al. synthesized symmetrical bis-alkynyl pyridine and thiophene derivatives (376–389) by Sonogashira reaction and tested for antiangiogenic activity using human umbilical vein endothelial cells (HUVEC). Compounds 376, 378, and 380 (Fig. 8), with rigid mimetic structure of curcumin, showed the potent growth inhibitory activity.100
Various metal complexes of curcumin and curcumin analogues101–105 sugar conjugates of curcumin,106,107 dipeptide conjugate, fatty acid and folic acid conjugates of curcumin,108 dihydropyridone derivatives of curcumin,109 cyclopalladated complexes of curcumin,110 unsymmetrical curcumin analogues111 and monofunctional curcumin derivative, clicked curcumin dimer112 have been synthesized. The various metal complexes include copper and manganese complexes of curcumin and their analogues while various mono and diglycosylcurcumin derivatives were synthesized. The monofunctional curcumin derivatives includes monocarboxylic acid derivative monoazide derivative, monoalkyne derivative, monotriazole–PEG derivative has been synthesized.
Curcumin in general showed better biological activities as compared to DMC and BDMC. Enhanced antioxidant activity was observed as the number of phenolic groups on the aryl rings increased. The picture is more complex in case of anticancer and cytotoxic activities. In general curcumin analogues with cyclohexanone moiety showed better anti-inflammatory, cytotoxic activities than compounds with acetone and cyclopentanone moieties. Some heterocyclic analogues also showed promising COX inhibitory activities. Our group has synthesized and screened the pyrazole and isoxazole analogues of curcumin against COX-1/COX-2. The pyrazole analogues of curcumin showed better antioxidant, COX-2 inhibitory, antitumor and antimalarial activities as compared to curcumin and isoxazole analogues. Hydrogenated analogues showed better antioxidant, similar anti-inflammatory and cytotoxic activities as that of curcumin. In-spite of better activity profile of curcumin analogues, their pharmacokinetic and pharmacodynamic studies have not been performed so far. Therefore, further studies are required to reveal detailed mechanism of action of these analogues and to make them suitable candidate for clinical trials.
ABTS | 2,2′-Azino-bis(3-ethylbenzothiazoline-6-sulphonic acid) |
BDMC | Bisdemethoxycurcumin |
COX | Cyclooxygenase |
cPLA2 | Cytosolic phospholipase A2 |
DHC | Dihydrocurcumin |
DMC | Demethoxycurcumin |
DPPH | 2,2-Diphenyl-1-picrylhydrazyl |
FRAP | Ferric reducing ability of plasma |
HHC | Hexahydrocurcumin |
IL | Interleukin |
LOX | Lipoxygenase |
LPS | Lipopolysaccharide |
LT | Leukotriene |
NBT | Nitroblue tetrazolium |
NF-κB | Nuclear factor-κB |
NO | Nitric oxide |
OHC | Octahydrocurcumin |
PGE2 | Prostaglandin E2 |
SAR | Structure–activity relationship |
THC | Tetrahydrocurcumin |
TNF-α | Tumor necrosis factor-α |
TRAP | Telomeric repeat amplification protocol |
This journal is © The Royal Society of Chemistry 2014 |