|
Fig. 1. Molecular structure of doxorubicin conjugates.
Meng-lei et al. obtained DOX and LNA conjugates and tested them in vitro on three cancer cell lines: human breast cancer (MCF-7, MDA-MB-231) and human liver cancer (HepG2). The calculated IC50 (the concentration of active substance needed to inhibit cell growth by 50%) of DOX-LNA against MCF-7 was 4.7 μM, against MDA-MB-231 was 3.1 μM, and for HepG2 – 6.8 μM. DOX used as a reference substance was characterised with higher IC50 values for all tested cell lines (MCF-7, MDA-MB-231 HepG2) and was equal to 8.3 μM, 10.2 μM and 14,3 μM, respectively. A decrease in the IC50 indicates an increase in drug toxicity, therefore the DOX–LNA conjugate was more cytotoxic than free DOX regarding to tested cancer cells (Huan et al., 2009).
Chhikara et al. synthesized amide bond linked conjugates of DOX and saturated FA containing from 6 to 18 carbon atoms. The antitumor effect of the newly obtained compounds was evaluated in vitro in a human leukemia cell line (CCRF-CEM), breast adenocarcinoma (MDA-MB-468), ovarian adenocarcinoma (SK-OV-3), and colon adenocarcinoma (HT-29) cell lines up to 120 h at 1 μM. The results indicated higher antiproliferative activity in colon and ovarian cell lines when compared to breast and leukemia cancer cells. The overall pattern in different cell lines showed that the lauric acid containing derivative was the most effective in in vitro studies (Chhikara et al., 2011).
Liang et al., conjugated DOX with either unsaturated LNA or saturated PA by a hydrazone or an amide bond (DOX-hyd-LNA, DOX-ami-LNA, DOX-hyd-PA, and DOX-ami-PA) to increase the therapeutic activity of DOX and to decrease its totoxic effect toward normal tissues. The conclusions were that the cytotoxicity of DOX-hyd-LNA on a liver cancer cell line (HepG2) and breast cancer (MCF-7 and MDA-231) cells was higher compared to that of unmodified DOX and to that of other obtained derivatives. In addition DOX-hyd-LNA exhibited less toxicity in vitro and in vivo. These results were due to higher DOX-hyd-LNA stability in serum, leading to increased distribution of DOX within tumor tissues, so a greater amount of DOX could be released inside tumor cells (Liang et al., 2014).
Doxorubicin and DHA conjugates (DHA–DOX) were synthesized by Wang et al. using a hydrazide bond. The antitumor activity was then evaluated in vitro against leukemia cells (L1210) and in vivo in experimental animal models including leukemia (L1210) and melanoma (B16) tumors. The IC50 values of DHA–DOX and free DOX in in vitro studies were 1.4 μM and 0.15 μM, respectively. In animal tumor models, DHA–DOX was significantly more efficacious than free DOX (Wang et al., 2006).
N-acylhydrazones derived by the synthesis of doxorubicin and saturated (margaric acid), unsaturated (oleic, linoleic, α-linolenic acids) FA were obtained by Effenberger et al.. All compounds were tested for anticancer activity in cells of human leukemia (HL-60), melanoma (518A2), breast (MCF-7/Topo) and cervix (KB–V1/Vbl) carcinomas. The margaric hydrazone was more cytotoxic than unsaturated FA conjugates and even three times more than DOX. In addition, the treatment of HL-60 and 518A2 cells with heptadecanoyl, linolenoyl hydrazones had an impact on the mRNA expression, resulting in the changes of Bax to Bcl-2 ratio, which is a potential molecular marker of cancer tissues and determines the susceptibility of cells to the intrinsic apoptotic pathway triggered by mitochondrial dysfunction (Effenberger et al., 2010; Khodapasand et al., 2015).
Arcamone et al. used C1–C3 carboxylic acids as well as capryloate to synthesise DOX derivatives. In vivo antitumor activity against mouse sarcoma derived from ascites (Sarcoma 180), murine virus induced sarcoma (MSV-M), intravenously transplanted Gross leukemia, and transplanted mammary carcinoma was evaluated in mouse models. In all the experimental settings tested, DOX capryloate was the most active compound (Arcamone et al., 1974).
3.2. Paclitaxel conjugates
Paclitaxel (PTX) is a complex taxane diterpene which was originally isolated from Taxus brevifolia (Wani et al., 1971). It is the active compound in Taxol®, which is one of the most effective anticancer drugs. It has been approved for the treatment of breast, ovarian, and lung cancers as well as Kaposi's sarcoma. However, PTX is not capable of crossing the blood-brain barrier to reach to a notable extent the brain tissue (Sparreboom et al., 1996). The mechanism of PTX action is based on binding to the microtubules of β-tubulin, causing the formation of parallel bundles of microtubules and inhibiting cell division at the G2-M phase of the cell cycle (Schiff and Horwitz, 1980; Yvon et al., 1999). Due to the fact, that PTX is extensively metabolized by the liver and is usually rapidly eliminated from the circulatory system, there is a search for various modifications that would extend its half-life and improve cellular uptake, thereby potentiating the drug's therapeutic effect (Xi Yu et al., 2010). PTX can be conjugated with FA in 2′-hydroxy and 7-hydroxy positions (Fig. 2).
Fig. 2. Molecular structure of paclitaxel conjugates.
Paclitaxel oleate (PTX-oleate) was synthesized in the reaction of oleoyl chloride with paclitaxel by Lundberg et al.. The esterification of the 2′-hydroxyl group of paclitaxel was preferred, because of the much higher reactivity of this position compared to the 7-hydroxyl one. The conjugate demonstrated an in vitro cytotoxic activity against a human cervical cancer cell line (HeLa) with a significant increase in activity along with the increase of incubation time. IC50 for HeLa cells were 5500, 500, 150, and 100 nM for 24, 48, 72 and 96 h of incubation, respectively. In addition, the in vitro incubation of PTX-oleate with human plasma resulted in greater proportion found in the lipoprotein pool when compared to PTX (Lundberg et al., 2003).
Bardley et al. coupled DHA to paclitaxel at the 2′-hydroxyl position. The conjugate showed increased antitumor activity in a mice lung tumor model (M109) and in human colon (HT-29) mice implanted carcinoma when compared with PTX. The modified drug appeared to be less toxic before it was metabolized by tumor cells to the cytotoxically active free PTX form, therefore 4.4-fold higher molar doses could be delivered to mice (Bradley et al., 2001).
Bedikian et al. covalently conjugated the essential fatty acid DHA to the paclitaxel molecule (DHA-PTX) at the 2′-hydroxyl position. Preclinical studies have demonstrated increased activity of DHA-PTX compared to PTX. In the presented study, the efficacy and toxicity profiles of DHA–PTX were compared with those of dacarbazine – a structural analogue of imidazole carboxamide used as a chemotherapy drug (Bedikian et al., 2011).
Lack of efficacy of PTX against drug-resistant tumors such as colon, pancreatic, melanoma, and renal cancers is reported to be due to the overexpression of P-glycoprotein (Pgp), a transporter effluxing out hydrophobic anticancer agents including PTX. Kuznetsova et al., used PUFAs such as DHA, LA, LNA to modify second-generation toxoids: SB-T-1103, SB-T-1104, SB-T-1213, SBT-1214, SB-T-1216, and SB-T-1217 in 2′-hydroxy position. This way obtained PUFA–taxoid conjugates were assessed for their efficacy against a drug-sensitive (Pgp-) human ovarian tumor xenograft (A121) and a drug-resistant (Pgp+) human colon tumor xenograft (DLD-1) in SCID mice. While PTX was ineffective, LNA–SB-T-1213 and DHA–SB-T-1213 exhibited strong antitumor activity. LA-SB-T-1213 did not show meaningful efficacy in the same assay, which confirmed the selectiveness of drug-conjugate tumor cell targeting between n-3 PUFA (LNA, DHA) and n-6 PUFA (LA) (Kuznetsova et al., 2006).
Xi-Yu et al. synthesized conjugated linoleic acid and PTX hybrid molecule (CLA-PTX) using the hydroxyl (2′-position) group. The conjugate showed higher cellular uptake efficiency in murine glioma cells (C6) in comparison to PTX and showed lower in vitro cytotoxicity. The antitumor efficacy in brain tumor-bearing rats after administering CLA-PTX was significantly higher than that after giving Taxol (Xi Yu et al., 2010).
3.3. Cytarabine conjugates
Cytarabine, also known as cytosine arabinoside (ara-C), is a pyrimidine nucleoside-based anticancer drug with arabinose sugar, widely used for the treatment of leukemia. Cytarabine is predominantly used against acute myelogenous leukemia (AML) and non-Hodgkin's lymphoma (NHL), acute lymphoblastic leukemia (ALL), and erythroleukemia (Jabbour et al., 2007; Shah and Agarwal, 2008). Many research groups have been evaluating the ara-C prodrug approach using various fatty acid modifications at 2′-hydroxyl, 5′-hydroxyl and 4-amino groups of cytarabine, making the drug more lipophilic and protecting it from cytidine deaminase (Fig. 3).
Fig. 3. Molecular structure of cytarabine conjugates.
Liu. et al. has used the simultaneous amino acid and fatty acid derivatization obtaining 4-amino acid (Val, Met, Tyr, Glu and Arg) and fatty acid conjugates with chain length of 10, 14 and 18 carbons. Subsequently, a direct coupling of amino acid fatty acyls derivatives and cytarabine at 4-NH2 position was performed forming a peptide bond. The antiproliferative activity was evaluated in a human leukemia cell line (HL-60) and cervical cancer cells (HeLa), and varied depending on the type of amino acid and FA used. Methionine derivatives showed better antiproliferative activity when compared to the other synthesized amino acid containing conjugates. Additionally, the antitumor effect decreased with the increase of carbon chain length and did not improve biological activity (Liu et al., 2009).
Fatty acid-substituted conjugates of cytarabine at 5′-hydroxyl position were characterised with significantly different properties than N4 derivatives (A. M. Bergman et al., 2004). An ester derivative of cytarabine and elaidic acid (ara-C-5′-elaidic, CP-4055) facilitated cellular accumulation and retention of ara-C in tumor cells. Unlike cytarabine, the cellular uptake of this derivative is independent on nucleoside transporters (Breistøl et al., 1999), which caused high cytotoxic effect in solid tumor and leukemia cells in vitro and in vivo (A M Bergman et al., 2004).
The cytotoxic mechanism of cytarabine involves phosphorylation of the 5′-hydroxyl group of arabinose sugar into triphosphate, therefore the effectiveness of fatty acid ester derivatives depends on the concentration of the parent drug delivered intracellularly after the ester hydrolysis and availability of the 5′-hydroxyl group for phosphorylation (Plunkett et al., 1987).
The 2′-hydroxyl position of arabinose sugar in cytarabine was also explored for functionalizing with fatty acids by Chhikara et al.. The C2′ and C5′ myristic acid substituted ara-C derivatives (ara-C-2′-myristoyl, ara-C-5′-myristoyl and ara-C-2′,5′-dimyristoyl) were synthesized and screened for their cytotoxic activity. The C5’ substituted derivative was not able to inhibit the proliferation of leukemia cells (CCRF-CEM) even after 96 h at a concentration of 1 μM. 2′-Fatty acyl and 2′,5′-disubstituted derivatives of ara-C inhibited the growth of cancer cells by approximately 36–76% at a concentration of 1 μM after 96 h incubation. Enhanced cytotoxic activity of 2′,5′-disubstituted derivative after 96 h compared to that after 24 h indicates that the conjugate slowly releases cytarabine and may behave as a prodrug for sustained parent ara-C delivery, which may be beneficial for the therapy (Chhikara et al., 2010).
3.4. Gemcitabine conjugates
Gemcitabine (GEM) is a deoxycytidine analog used as chemotherapeutic drug for treatment of various solid tumors, including pancreatic cancer (Burris et al., 1997), non-small cell lung cancer, ovarian cancer and breast cancer (Hui and Reitz, 1997). It can inhibit both DNA synthesis and ribonucleotide reductase (Galmarini et al., 2002).
Modifications on GEM involve the 3′-hydroxyl, 5′-hydroxyl group and 4-amino group and thus can transform it into an ester or amide derivative (Fig. 4). Bender et al. reported that gemcitabine amides were better than the esters in terms of increased plasma half-life, since the amides can block the site of deamination for deoxycytidine deaminase and therefore lower the metabolism of gemcitabine (Bender et al., 2009).
Fig. 4. Molecular structure of gemcitabine conjugates.
Immordino et al. synthesized a series of lipophilic gemcitabine prodrugs by linking the 4-amino group with valeroyl, heptanoyl, lauroyl and stearoyl acyl derivatives. Cytotoxicity of these derivatives, free or encapsulated in liposomes, was between two- and sevenfold that of GEM. Encapsulation of prodrugs in liposomes protected them from degradation in plasma, thus ensured longer plasma half-time and better intracellular release of the free drug (Immordino et al., 2004).
Bergman et al. used elaidic fatty acid to modify GEM at the 5′position by esterification. The cytotoxic activity and mechanism of action of the new lipophilic prodrug (CP-4126) was described in rodent leukemia cell lines, human leukemia and various solid tumor cell lines and xenografts. In conclusion, the gemcitabine-elaidic acid derivative showed equal antitumor activity to GEM in various xenograft models. Furthermore, oral administration compared to intraperitoneal administration resulted in equal antitumor activity (Bergman et al., 2011).
Tao et al. covalently coupled conjugated linoleic acid (CLA) to the 4-amino group of GEM to obtain CLA–GEM amide. In vitro tests in human breast tumor cells (MCF-7) showed an increased plasma prodrug stability and antitumor activity. The CLA–GEM conjugate was found to significantly inhibit tumor cell growth in a nucleoside transporter independent manner, in contrast to unmodified GEM. An In vivo pharmacokinetic study resulted in longer CLA–GEM half-life and better bioavailability compared to free GEM (Tao et al., 2012).
To enhance the drug efficacy and reduce the adverse effects Li et al. modified the N4-position of gemcitabine with DHA. The cytotoxicity of DHA, GEM and DHA-GEM was evaluated in human breast cancer cells (MCF-7 and MB-MDA-231), liver cancer (HepG2 and Bel-7402), lung cancer (A549), gastric cancer (SCG-7901) and mouse hepatoma cancer (H22) cells. The results showed that DHA-GEM had high efficacy and lower toxicity than the free GEM (Li et al., 2014).
3.5. Ciprofloxacin conjugates
Ciprofloxacin (CP) is a commonly used broad-spectrum fluoroquinolone (FQ) antibiotic with low side effects, which does not show cytotoxic effect against normal cell lines. It has been also shown to have antiproliferative and apoptotic activities in several cancer cells (Aranha et al., 2000; El-Rayes et al., 2002; Herold et al., 2002). Hussy et al. described the inhibitory effect of several FQs against mammalian DNA topoisomerases I and II and DNA polymerase and reported that CP was the most potent FQ inhibitor of all tested enzymes (Hussy et al., 1986). FA modifications of CP can be carried out only on the NH moiety in piperazine ring (Fig. 5).
Fig. 5. Molecular structure of ciprofloxacin conjugates.
Azéma et al., (2009) modified the ciprofloxacin piperazine NH group using a 5–16 carbon chain length FA and forming oxoethylalcanoate, alkoxycarbonyl, alkanoyl conjugates. The in vitro antitumor activity of obtained compounds and CP has been determined for prostate (PC-3), glioblastoma (U373-MG), colorectal (LoVo), lung (A549), and breast (MCF-7) human cancer cell lines. Only the amide derivative of CP and capric acid showed potent in vitro antitumor activity. No correlation was reported between in vitro antitumor activity and chain length (Azéma et al., 2009).
Chrzanowska et al., (2020) coupled ciprofloxacin (CP) with saturated and unsaturated fatty acids, and evaluated cytotoxicity, apoptosis-inducing effects and inhibition of IL-6 release in human primary (SW480) and metastatic (SW620) colon cancer, metastatic prostate cancer (PC3) and normal (HaCaT) cell lines. The oleic acid conjugate showed 13 times lower IC50 value (7.7 μM) for CP alone (101.4 μM) in the PC3 cell line, which was found to be the most sensitive to the presence of the obtained conjugates. The normal cell line (HaCaT) was much less sensitive to oleic acid-CP derivative and CP. The IC50 values were 132.5 μM and 222.1 μM, respectively.
Modification of fatty acid with heterocyclic compounds, which express none or weak cytotoxic activity, results in strong cytotoxic effect against tumor cells and weak effect against normal cell lines.
Ahmad et al. synthesized oleic acid and caproleic acid derivatives bearing 1,3,4-oxadiazol-2-thione (a), 1,2,4-triazol-3-thione (b) and 1,2,4-triazolo[3,4-b]-1,3,4-thiadiazine (c) moieties (Fig. 6). All compounds were tested in vitro for antitumor activity against three human cancer cell lines: human hepatocellular carcinoma (Hep3B), human breast adenocarcinoma (MCF-7), human cervical carcinoma (HeLa) and normal peripheral blood mononuclear cells (PBMC) by MTT assay. The results showed moderate cytotoxicity against different human cancer cell lines in range: 7.4–19.9 μM. Caproleic acid derivatives expressed higher cytotoxicity than the oleic acid ones. Cytotoxicity of starting compounds and of heterocyclic moieties was not tested in the studies (A. Ahmad et al., 2017).
Fig. 6. Molecular structure of 1,3,4-oxadiazol-2-thione (a), 1,2,4-triazol-3-thione (b) and 1,2,4-triazolo[3,4-b]-1,3,4-thiadiazine (c) conjugates.
Yang et al. reported the synthesis of different amide derivatives of saturated FA with different carbon chain lengths (C10–C24) and quaternary salts of piperazine (Fig. 7). Compounds were assayed towards growth inhibitory activity against six human cancer cell types: prostate cancer (PC-3), gastric cancer (NUGC-3), breast cancer (MDA-MB-231), renal cancer (ACHN), colon cancer (HCT-15), and non-small cell lung cancer (NCI–H23). The palmitic acid derivatives exhibited the most potent inhibitory profile. For compounds with a relatively shorter or longer carbon chain (C10 or C24), the cell growth inhibition was weak, or was not observed at all, irrespective of cell type. Perifosine and NSC126188 (a piperazine alkyl derivative) were used as positive references for comparison of in vitro activities (Yang et al., 2011).
Fig. 7. Molecular structure of piperazine quaternary salts.
Jubie et al. synthesized some novel 1,3,4-oxadiazoles (a) and 1,2,4-triazoles (b) derivatives of stearic and gamma-linolenic acids (Fig. 8). Cytotoxicity of these compounds was evaluated by the growth inhibition assay in human lung carcinoma cell lines (A-549) cells in vitro. Conjugates of gamma-linolenic acid showed more potent cytotoxicity than stearic acid. No reference compound was used in the study. (Jubie et al., 2012).
Fig. 8. Molecular structure of 1,3,4-oxadiazol-2-thiol (a), 1,2,4-triazol-3-thiol (b).
Amides of FA (palmitic, stearic, oleic, elaidic and linoleic acids) and benzylamine derivatives: benzylamine, (R)-methylbenzylamine, (S)-methylbenzylamine as well as pyrrolidine, piperidine, morpholine were obtained by Santos et al. (Fig. 9). Antiproliferative in vitro study resulted in a clear structure-activity relationship in seven human cancer cell lines: glioma (U251), breast epithelial carcinoma (MCF-7), ovarian (OVCAR-3), ovarian with a phenotype of multiple drug resistance (NCI-ADR/RES), kidney (786–0), non-small cell lung cancer (NCI–H460), prostate (PC-3) as well as two normal cell lines: human keratinocyte (HaCaT) and kidney epithelial cells from the African green monkey (VERO). The presence of a five membered heterocyclic compound with unsaturated chain resulted in significantly greater antiproliferative activity when compared to saturated chain derivatives. Pyrrolidine conjugates of oleic and linoleic acids were the most effective in inhibiting cell proliferation in most of the cell lines. No information about the cytotoxic activity of pyrrolidine was provided in the paper (Santos et al., 2015).
Fig. 9. Molecular structure of the most active pyrrolidine derivatives.
Venepally et al. synthesized a series of novel ethyl derivatives [1-ethyl-6-fluoro-7-(fatty amido)-1,4-dihydro-4-oxoquinoline-3-carboxylate] containing an amide linkage at the C-7 position (Fig. 10). The amine group was covalently coupled with saturated FA: C6, C8, C12, C15, C16, C18 and unsaturated FA: undecylenic (C11), oleic (C18). All synthesized compounds were screened against four cancer cell lines: human prostate (DU145), lung cancer (A549), ovarian cancer (SKOV3), breast cancer (MCF-7) and normal lung cell line (IMR-90). DOX was used as a standard positive control. The FQs derivatives with hexanoic, octanoic, lauric and myristic fatty acids exhibited promising cytotoxicity against all tested tumor cell lines with hexanoic acid derivative showing the highest one. None of intermediate compounds obtained in multistep synthesis of the final quinolone derivatives were tested for cytotoxic activity (Venepally et al., 2016).
Fig. 10. Molecular structure of 1-ethyl-6-fluoro-7-(fatty amido)-1,4-dihydro-4-oxoquinoline-3-carboxylate.
In another work Venepally et al. obtained N-fatty acyl derivatives of saturated FA: C4, C6, C8, C12, C14, C16, C18 and unsaturated FA: undecylenic (C11:1), oleic (C18:1) and dimethoxy tryptamine (Fig. 11). Compounds were tested for antitumor activity against different human cancer cell lines: lung cancer (A549), prostate cancer (PC-3), breast cancer (MDA-MB-231), liver cancer (HepG2) and normal cell line (HUVEC). Most of derivatives showed a significant cytotoxic effect. Conjugates of butyric acid (C4) and oleic acid (C18:1) were the most promising ones (IC50 < 16 μM). Only final derivatives were screened for antitumor activity (Venepally et al., 2017).
Fig. 11. Molecular structure of dimethoxy tryptamine derivatives.
Jóźwiak et al. synthesized eleven esters of alloxanthoxyletin and fatty acids (saturated: C3, C6, C8, C12, C14, C16, C18 and unsaturated: oleic, α-linolenic, conjugated linolenic and docosahexaenoic) (Fig. 12) and screened for their anticancer toxicity using tumor cell lines such as human melanoma cells (HTB-140), human epithelial lung carcinoma cells (A549) and a normal cell line – human keratinocyte line (HaCaT). Derivatives of unsaturated FA showed high cytotoxic potential against cancer cells with IC50 of 14.4–39.4 μM, with the compound bearing DHA acid being the most effective. The increasing level of unsaturation of the hydrocarbon chain resulted in an increased compound's activity. In addition, alloxanthoxyletin derivatives showed greater cytotoxic effects against tumor cell lines compared to normal cells (HaCaT). (Jóźwiak et al., 2019). 7-Hydroxyalloxanthyletin used as a starting compound expressed significantly lower cytotoxic activity against the majority of tested tumor cell lines. It showed a cytotoxic effect against Vero cells of African green monkey (IC50 = 41 μg/ml), breast carcinoma MCF-7 cells (IC50 > 50 μg/ml), epidermoid carcinoma KB cells (IC50 = 48 μg/ml) and small cell lung cancer NCI–H187 (IC50 = 11 μg/ml) (Promsuwan and Yenjai, 2013).
Fig. 12. Molecular structure of alloxanthoxyletin derivatives.
4. Conclusion and perspectives
Considering saturated fatty acids, only short-carbon chain molecules show antitumor activity. There was no anticancer activity reported for saturated FA with carbon chains longer than C10. The activity for unsaturated FA increases with the elongation of the carbon chain in the molecule as well as with the level of unsaturation. Besides the number of double bonds in the molecule, the location of the double bond (n-3, n-6, n-9) has also an important impact on the activity of FA. Among unsaturated FA, long-chain polyunsaturated (PUFAs) molecules show the greatest antitumor activity. PUFAs n-3 are a more promising group compared to PUFAs n-6, as they play a role in the prevention of the development of different cancer types in vivo and in vitro. PUFAs n-6 (arachidonic acid, linoleic acid) can promote or inhibit tumor development depending on the mechanism of metabolic action induced within the cell, therefore they possible use in cancer treatment is so far controversial. Modifications of existing anticancer drugs by conjugating them with FA may increase their tissue selectivity and can potentially make chemotherapy more effective and less toxic. Improved drug stability, solubility and therefore bioavailability limits undesirable physiological interactions and prolongs circulation half-time. All this increases the amount of active substance that can reach the intended target, minimizing side effects and improving the quality of chemotherapy. Conjugates of FA with commonly used anticancer drugs express higher cytotoxicity against tumor cell lines compared to normal cell lines.
The literature review revealed that heterocyclic structures exhibit a cytotoxic activity which can be improved by the modifications with FA. The starting heterocyclic compounds, if, did not show cytotoxic effects against treated cells or exhibited significantly lower efficacy compared to the FA-heterocycle conjugate, as shown in the reviewed studies. Among the short-chain acyl derivatives, both saturated and unsaturated conjugates expressed anticancer activity. Condensation with long-chain FA resulted in a high cytotoxic effect only in case of unsaturated chains used.
Syntheses of different conjugates of existing drugs with fatty acids are well known and described in the literature. Mechanisms of action of these hybrid molecules have been tested, proving an increase of the drugs’ selectivity against tumor cells. As a consequence, a lesser amount of a drug is needed to achieve the same effect in comparison to the non-modified drug.
To perform a complete evaluation of connections of fatty acids with heterocyclic compounds, further research is required, since the cytotoxicity tests for normal cell lines were not included in many studies. In most cases, the presented results were in vivo and in vitro preliminary studies conducted only on tumor cell lines. The mechanisms of cytotoxic activity of new fatty acid combinations with heterocyclic compounds are mostly unknown. Furthermore, conjugates of fatty acids with heterocyclic moieties require additional research determining the pharmacokinetics of new combinations. This may result in finding new potentially active molecules, which mechanisms of action haven't been described yet.
Although a number of fatty acid and cytostatic drugs connections have been approved and tested in phase I, II or III of the clinical trial process, e.g. taxoprexin, which is a prodrug of paclitaxel bound to the docosahexaenoic acid. Phase II studies have demonstrated favourable results with this new paclitaxel formulation in human patients with lung melanoma and other cancers (Homsi et al., 2010). However, compounds have not been introduced into treatment because their efficacy as a first-line therapy for metastatic melanoma does not exceed that seen with other single-agent chemotherapies such as dacarbazine (Bedikian et al., 2011). Further exploration of new connections of DHA with other chemotherapy or targeted agents could be considered. Shougang et al. connects docotaxel, a currently used drug in first- or second-line lung cancer treatment, with DHA as a tumor-targeting ligand. The researchers conclude that DHA-docotaxel may exhibit a potential therapeutic effect against lung cancer metastasis to bone (Jiang et al., 2018).
In order to develop tumor specific chemotherapeutic drugs, new formulations of drug delivery systems are tested. Gulzar et al. formulated an oil-in-water nanoemulsion of a next generation taxoid DHA-SBT-1214 and evaluated its biodistribution and pharmacokinetics. The results from this study demonstrated effective encapsulation of the drug in a nanoemulsion and this nanoemulsion showed sustained plasma levels and enhanced tumor delivery relative to the solution form (G. Ahmad et al., 2017).
This FA modifications, which increase the activity and the uptake selectivity of the compounds, and alter drug delivery methods may be the key to unlocking the true medical potential of FA, in particular PUFAs.
Acknowledgements
This work was supported by the Polish National Science Centre under the grant PRELUDIUM number 2017/25/N/NZ7/01583. This work was supported by the Polish National Science Centre under the grant PRELUDIUM number 2017/25/N/NZ7/01583.