Open access peer-reviewed chapter

The Orange Peel: An Outstanding Source of Chemical Resources

Written By

Gianfranco Fontana

Submitted: 22 September 2020 Reviewed: 29 January 2021 Published: 30 September 2021

DOI: 10.5772/intechopen.96298

From the Edited Volume

Citrus - Research, Development and Biotechnology

Edited by Muhammad Sarwar Khan and Iqrar Ahmad Khan

Chapter metrics overview

989 Chapter Downloads

View Full Metrics

Abstract

Citrus sinensis (L.) Osbeck is a very common cultivar belonging to the Rutaceae family. It is largely diffused in several areas of the world characterized by mild to warm climate conditions. Its abundant worldwide production (up to 107 Tons. per year) and consumption both as the edible part of the fruit and as several types of derivative products imply the production of a huge amount of waste, such as the fruit pomace. Several ways of recycling this material have been developed in recent years: employment as fertilizer, fodder ingredient, and even cloth material. However, the chemical added value of Citrus sinensis peel has been underestimated despite the diversified and significant content of useful chemicals, such as polyphenols, polymethoxylated phenols, glycosylated flavonoids, volatile and non-volatile terpenoids, pectins, enzymes, etc. This work aims to highlight the outstanding chemical potential of Citrus sinensis peel.

Keywords

  • biological activity
  • Citrus sinensis
  • essential oil
  • flavonoids
  • orange peels
  • polymethoxyphenols

1. Introduction

Citrus sinensis (CS) (L.) Osbeck is a perennial species growing in warm climate areas of the world and largely employed as food in form of fresh fruit, with a global production of ca. 6.7X107 tons. per year (TPY) in 2016 [1], or as a processed derivative (ca. 1.85x107 TPY) such as juice, marmalade, flavor, fragrance and coloring additive, pectin.

CS is an evergreen tree, 3 to 9 mts. high with sparingly barbed branches, alternate leaves with toothed blades differently shaped, oval or elliptical, connected to the stem by winged-petioles. Axillary flowers are present singly or in whorls of 6 and possess 5 white petals and up to about 25 yellow colored stamens. The pericarp of CS has a spherical or oval shape of 6–10 cm diameter with the color changing from green to yellow-orange during the ripening; the endocarp containing juice sac glands is enclosed within a wrinkled epicarp or exocarp or flavedo containing a great number of essential oil glands protected by a waxy epidermis. Below the flavedo is the albedo, also called mesocarp, a white filamentary tissue composed of tubular-like cells.

The principal industrial application of CS is the production of frozen concentrated juice. The procedure of juice extraction eventually accompanied by the extraction of the essential oil, implies the generation of a major “by-product” constituted by a pomace, mainly containing peels, accounting for up to around 60% w/w of the original fruit mass processed [2]. This huge amount of biomass does pose serious environmental concerns because of its high level of total organic carbon (TOC) and biological oxygen demand (BOD) that make disposal procedures rather complex and demanding from both the legal and industrial points of view. This is because there is an increasing trend to modify the way of approaching this problem by reconsidering the post-production orange pomace more like a by-product rather than a waste. In the last years, many producers have subjected this material to processings involving partial acidic fermentation, drying, and packaging to biologically and chemically stabilize the biomass before its application as animal feed in zootechnics, soil conditioners in agriculture, or the manufacturing of compost and biogas [2].

Beyond the standard workup of the Citrus sinensis peel (CSP) waste, new perspectives have been being opened in the context of the high chemical added value of the CSP [3, 4, 5] also by the complete knowledge of the rich metabolomics profile of this species. The use of CS peel has been proposed for a variety of purposes that include the production of antioxidant-enriched dietary supplements in veterinary [6], the preparation of human dietary supplements, and nutraceuticals such as citric acid [7] and flavonoids [8, 9]. The extract of CS peel is the source of a huge variety of phytochemicals and has been investigated on several applications including its chemotherapeutic and chemopreventive potential for several relevant human pathologies, such as cancer [10, 11] and obesity [12]. The extraction procedures vary in function of the main components that have to be obtained: from the simple cold pressing of pomace and the extraction with water to obtain highly hydroxylated compounds to the employment of mixtures organic protic solvent/water and finally low polar organic solvents such as Chloroform and Ethyl acetate to obtain polymethoxylated phenols (PMF, see below). New extraction technologies such as ultrasounds and microwaves may help to obtain better extraction yields.

In the following sections, the chemical structures and the biological effects of these compounds will be discussed.

Advertisement

2. The chemistry of Citrus sinensis peel

2.1 Essential oils

The essential oil (EO) is mainly obtained from the CS peel as a major by-product of the juice production process by a cold-pressing method that can provide the intact blend of compounds without losing the lighter, more volatile, components of the complex mixture that can be lost in the standard EO extraction procedure that is the hydrodistillation. The last one is mainly used in small scale applications, for example in research laboratories.

The chemical composition of CSP EO [13, 14, 15] is reported in Table 1. As it can be seen, the major component is D-Limonene, accompanied by several minor components belonging to the classes of monoterpene alkenes, oxygenated monoterpenes including alcohol aldehydes and esters, sesquiterpenes as well as linear alkanes and aldehydes. This rather complex blend accounts for the numerous deal of biological activities reported for the CSP EO [14, 15, 16], which include anthelmintic, anti-aflatoxigenic [17], antibacterial [18, 19, 20], anticarcinogenic, antifungal [21], antioxidant [17], anti-tumor [22], anxiolytic [23], food preservative [24], hepatocarcinogenesis suppressant, insecticidal and larvicidal [25], pain relief and relaxant [26]. It can be argued that the main effects are due to the presence of the major component Limonene that showed several bioactivities when tested as pure compound [27]. However, it is possible that synergistic effects due to the combination of Limonene with other minor components may be speculated and should have to be demonstrated.

Comp.Comp. name%Compound.Comp. name%
1Aromadendrene0.0121β-Linalool0.4–5.6
2δ-Amorphene0.0522β-Myrcene1.3–3.3
3D-Cadinene0.01–0.0323Neral0.1–1.3
4δ-3-Carene0.1824Neryl acetate0.02
5β-Citral0.12–0.1525Nonanal0–0.1
6L-(+)-Citronellal0.01–0.126Nootkatone0.01
7Citronellyl acetate0.0127cis-β-Ocimene0.03–0.26
8α-Copaene0.0428Octanal0.02–0.8
9α-Cubebene0.02–0.2629Perillaldehyde0.03
10β-Cubebene0.0330α-Phellandrene0.02–0.07
11Decanal0.04–0.431α-Pinene0.49–0.59
12n-Dodecanal0.0632(+)-Sabinene0.2–1.0
13β-Elemene0.01–0.0233γ-Terpinene0–1.21
14Geranial0–1.834γ-Terpineol0.04–008
15Germacrene-D0.02–0.0835α-Terpineol0.07–0.42
16β-Gurjurene0.0136Terpinolene0–0.08
17Hexadecanol0.0437α-Thujene0.04
18D-LimoneneCa. 95
19L-Limonene0.02
20trans-Limonene oxide0.01

Table 1.

Composition of C. sinensis essential oil obtained from peels.

2.2 Polyphenols

2.2.1 Flavanoids

Polyphenols extracted from the CS peel belongs to the general structural categories of flavanones (Figure 1a), flavones (Figure 1b), flavonols (Figure 1b), with and without sugar moieties attached to one or more of the hydroxyl groups [28]. It is worthy of particular mention the rarely occurring class of C-glycolflavones (Figure 1b, compounds 6365, 85, 86).

Figure 1.

Chemical structures of flavonoids from C. sinensis peels.

These compounds are produced in vivo from the biogenetic mixed pathway of the Acetate and Shikimate that implies the enantiospecific formation of the basic aromatic bicyclic framework of the flavanone, from which a huge number of flavonoids originate employing selective enzymatic hydroxylations, methylations, and glycosylation steps. As can be seen from the structures shown in Figure 1, most of the chemical entities found in the peel extract contain several methoxy fragments that decorate the carbon skeleton. This characteristic makes those molecules to get a rather apolar character that explains their presence in the hydrophobic environment of the waxy peel. On the contrary, compounds containing a major number of hydroxyl groups are less present in the peel and are instead more significantly concentrated in the juice of the pericarp. However, some glycosylated compounds are present in the peel. In these molecules, the aglicone bears a monosaccharide unit (mainly glucose) or a disaccharide, in most of the cases being Rutinose (91) – Rhamnosyl (α1 → 6) glucose – or Neohesperidose (92)- Rhamnosyl (α1 → 2) glucose (Figure 2).

Figure 2.

Chemical structures of the disaccharides most commonly bound to flavonoids of C. sinensis peel.

The composition of the peel extracts described in the literature may slightly vary depending on the cultivar and the region of harvesting but some general points are common, that is the presence of the high amount of bioactive polymethoxyflavonoids [29, 30](PMF) some of which are rather ubiquitous, e.g. Nobiletin 53, Sinensetin 51, 3′,4′,3,5,6,7,8-Heptamethoxyflavone 55; some other compounds containing one to six methoxy groups in place of the hydroxyl groups are present at variable amounts. The presence of one or more residual hydroxy groups in the molecule may result in a higher bioavailability and in other general differences in their mechanism of biological and therapeutic actions [30, 31].

The biological role of these secondary metabolites in the plant is still matter of debate. It has been proposed their involvement in the mechanism of defense of the fruits exposed to the attack of phytopathogens, such as Phytophthora citrophthora [32].

Further, the composition of the PMF blend can be employed for the chemiotaxonomic characterization of the Citrus genus [33].

However, it needs to be stressed that in many cases the reported compounds were recognized by mass spectrometry and electronic spectroscopy. It is not always a matter of simplicity to discern the exact structure of a given PMF and to discriminate between different regioisomers, normally quite similar in terms of mass and electronic spectra, if an isolation procedure is not conducted and followed by a complete bi-dimensional NMR characterization. Significant differences in the extract composition do arise also in consequence of the extraction method; non-polar solvents such as Methanol, Chloroform Ethyl acetate let to obtain PMFs-rich extracts while, on the other hand, hydroalcoholic and aqueous extracts do contain a low concentration of PMFs and a higher concentration of un-methylated polyphenols as well as glycosylated compounds.

The biological activities disclosed for the flavonoids extracted from CSP are variegated. They include antioxidant [9, 34, 35, 36, 37, 38, 39], anti-inflammatory [40, 41], antimicrobial [39, 42, 43, 44], antimalarial [45], antitrypanosomal [46], cardio-protective [47], anti-osteoporosis [48], anti-ulcer [49], vascular protective [50], anti-diabetes [51, 52], hepatoprotective [53, 54], neurotrophic [55], anti-adipogenesis and anti-obesity [56, 57, 58], anti-hypertensive [59], cataract prevention [60], sun protection [61], metabolic syndrome control [62]. Further, it has been demonstrated [63] that while both flavonoid set 40, 42, 43 and the PMFs 5153 were able to inhibit the anion transportin polypeptide OATP2B1 in HEK293 cells, only the PMF group displayed this inhibitory activity also for the OATP1B1 and OATP1B3 carriers.

The most abundant PMF occurring in CSP, Nobiletin 53, was proven to possess sevral bioactivities, such as antioxidant, anti-inflammatory, cancer preventive [64] and also a significant protective effect in vivo against the endotoxic shock [65] and ethanol-induced acute gastric lesions [66] in mice. Further, compound 53 demonstrated the capacity to induce autophagy in human keratinocyte HaCaT cells [67], vasodilatator effect in the rat aorta [68] and to protect the intestinal barrier from the demages induced by dextran sulfate sodium [69].

PMFs can be considered as especially promising lead compounds for cancer therapy as asignificant cytotoxic activity has been demonstrated toward a number of cancer cells [70, 71] with several mechanisms of action [72, 73]; the cytotypes investigated include MCF-7 [73, 74, 75, 76], Hs578T triple-negative breast cancer [73, 77]; colon cancer cells CaCo-2 [19], LoVo [78], HTC-116 [79, 80] and HT-29 [79, 81]; lung cancer cells A549 [80, 82], H460 [82, 83], H1299 [82, 83]; gastric cancer cell lines AGS, BGC-823, and SGC-7901 [84]; leukemia cells HL-60 [85]. However, data regarding a possible antitumor activity in vivo are still rather uncommon. An interesting example is the case of the significant reduction of the intestinal tumor mass in ApcMin/+ mice treated with a CSP extract containing various PMF [86]. Further, CSP extract and pure Naringin 47 were tested for their efficacy against a YM1 esophageal cancer in an animal model [87].

Given the development of pharmacological applications of CSP extract components, further investigations are needed to better understand the bioavailability, safety, and efficacy of these compounds in humans. Most of the data reported concern in vitro experimentations or animal model tests. For example, the toxicity of Hesperidin 40 was evaluated [88] in Sprague Dawley rats showing a 50% lethal dose (LD50) of about 5 g/Kg body weight (BW) and a lowest-observed-adverse-effect level (LOAEL) of ca. 1 g/Kg BW.

In general, it should be emphasized as the body of evidence concerning the actual efficacy of sweet orange-derived compounds in human health is still far to be exhaustive. For example, while this work is under typewriting, a severe acute respiratory syndrome pandemic due to a COVID-19 virus is in act and a big deal of research has been being directed toward antiviral remedies and therapies. Research on nutraceuticals is not an exception and in particular some authors have shown by computational and molecular docking methods how Hesperidin 40, the most abundant polyphenol obtained from C. sinensis, would be able to bind the spyke protein of this virus thus inhibiting its activity [89]. Despite their undoubted interest, these results need to be further investigated with different experimental approaches.

The pharmacological potential of pure Hesperidin 40 was also investigated for several relevant human morbidity, such as cancer, hypertension, and ulcer [90].

2.2.2 Hydroxy-acids

Several hydroxylated carboxylic acids belonging to several structural sub-classes are present foremostly in the extract obtained with mixed hydro-organic solvents, such as MeOH/water and EtOH / water [37, 38, 51, 78]; these include the aliphatic Ascorbic, Citric, Kojic, Lactic, and L-Malic acids; the aromatic 4-Hydroxybenzoic, Protocatechulic, and Gallic acids. Further, the cinnamyl compounds (Figure 3) Cinnamic (93), p-Cumaric (94), Caffeic (95), Ferulic (96), Sinapinic (97) acids, and Artepillin (98) were identified in some CSP extracts that showed relevant biological activities, such as antioxidant [34, 37, 38] and antidiabetes [51].

Figure 3.

Chemical structures of cinnamic acids extracted from C. sinensis peels.

These organic acids are mainly found in free form but in some cases, they are esterified with a variety of alcoholic compounds, such as Ethanol in Ethyl gallate 99 [51], 2-Phenylethanol in Phenylethyl ester of Caffeic acid 100 [51] and (−)-Quinic acid in Chlorogenic acid 101 [51]. An interesting ester derivative (102) in which the anomeric hydroxyl of Glucose is esterified with a O-Caffeylsinapoyl acid unit was found in the methanolic extract of a Greek cultivar of C. sinensis [34].

It was shown [38] that the antioxidant properties of a CSP extract better correlated with the total phenols content (TPC) of the sample rather than with its total flavonoid content (TFC), as it can be expected from the known relevant antioxidant character of hydroxycynamic derivatives.

2.2.3 Coumarins

Coumarins are aromatic compounds biogenetically related to the o-hydroxysubstituted cynamic acids from which originate by the intramolecular condensation between the carboxylic and the o-hydroxy groups. These compounds are most commonly encountered in other species of Citrus taxa [91], such as C. aurantium (bitter orange), C. limon, (lemon), C. limetta (lime), C. paradisi (grapefruit) and only a few molecules of this class were Isolated from extracts of CSP endowed with activity against osteoporosis [48] and antioxidant [92]; these compounds are shown in Figure 4. As coumarins are relatively less common in C. sinensis cultivars compared to other species of the Citrus taxa, their rarity can be considered as a chemotaxonomic marker for C. sinensis.

Figure 4.

Chemical structure of coumarins extracted from C. sinensis peels.

2.2.4 Catechins

The NADPH dependent bioreduction of flavanols is the biogenetic origin of this class of compounds, present as minor constituents in CSP extract possessing significant antioxidant activity [38]; they are the two enantiomeric forms Catechin 113 and Epicatechin 114, together with Epigallocatechin 115 (Figure 5).

Figure 5.

Chemical structure of catechins from C. sinensis peels.

2.3 Pectins

Pectins [93] are chemically definable as complex mixtures of polyglyconic acids in which a linear polymeric backbone is structured by a series of α (1 → 4) linkages (Figure 6). The main sugar monomer is always Galacturonic acid with the presence of possible heterogeneous domains of other sugars such as Xylogalacturonan and Rhamnogalacturonan-I. A variable amount of the free carboxy functions may be esterified with methyl groups, while the hydroxy groups at C-2 and C-3 positions of the sugar monomers may be acetylated. Even though the primary structure of the main chain is linear, a possible degree of ramification, depending on the pectin source, may also be found. The differences in the pectins composition and structures, depending on their natural source, do confer them different physio-chemical properties, such as water solubility, sol–gel concentrations, etc. On the ground of the degree of methylation of the acid moieties, pectins are classified as “low methoxyl” (LMP, -COOMe/-COOH <50% mol.) or as “high methoxyl” (> 50% mol). A simplified representation of pectin structure is given in Figure 6.

Figure 6.

Minimal representation of a Homopolygalacturonic acid domain of the linear primary pectin structure with a 1/3 Mol. /Mol. Esterification degree.

Pectins find many applications in the food and drug industry as a thickening and gelling agents, excipients, and colloidal stabilizers [93].

As it has been already mentioned, the extraction method does affect the structure and the properties of the final product; the traditional acidic water extraction implies a certain degree of hydrolytic deterioration, so that new extraction technologies have been being investigated to improve the quality of the final pectins, that is microwave-assisted extraction (MAE) [94] and ultrasounds assisted extraction (USAE) [35, 95].

2.4 Enzymes

As it can be easily argued, the CSP cellular system, whose genomic profile has been fully characterized [96], is the site of a complex network of enzymatic activity. Some of the enzymes of CSP have been characterized and employed in many applications.

The acetylesterase (international enzymatic classification: EC 3.1.1.6) from CSP is known since 1947 [97] and was isolated and characterized [98]. The acetylesterase activity of the partially purified enzyme was used for the removal of the acetyl group at the 3 positions of β-lactamic antibiotics 116 [98] (Figure 7a). Further, the whole CSP, as well as pomace from the industrial waste of the orange juice production, was successfully employed to catalyze several relevant biotransformations [99] such as the conversion of Geranyl acetate 118 to Geraniol 119 (Figure 7b) and the di-acetoxynaphtalene derivative 120 to the vitamin k1 precursor 121 (Figure 7c).

Figure 7.

Chemical reactions biocatalysed by enzymes from C. sinensis peels.

Recently, partial purification and functional characterization of a Uronic acid oxidase from CSP was accomplished [100]; this enzyme promotes the oxidation by O2 of Galacturonic acid 122 to Galactaric acid 123 (Figure 7d).

2.5 Miscellaneous

2.5.1 Highly lipophilic compounds

The waxy environment of flavedo in CSP does contain several long-chain saturated and unsaturated compounds: alkanes, fatty acids, waxes, higher terpenoids.

Tetracosane, Tetratriacontanoic acid, and Ethyl pentacosanoate were identified in CSP of a Pineapple variety [101]. Further, some carotenoids were identified in the CSP extract obtained with a solvent mixture composed of Ethanol, Ethyl acetate, Petroleum ether 1: 1:1 [102]. This complex blend of carotenoids includes α- and β-Carotene, Phytoene, Phytofluene, (all-E)- and (9Z)-Violoxanthin, (all-E)-Neoxanthin, (13Z)-, (13Z’)- and (all-E)-Lutein, (9Z)-Zeaxanthin, (all-E)-Zeaxanthin; the mono and di-esters of violaxanthin, antheroxanthins, Xanthophyll, β-Citraurin with various fatty acids, including Lauraic, Myristic, Oleic, Palmitic, Stearic. The composition of the blend has been correlated with the maturity stage of the fruit.

2.5.2 Peptides

Three cyclic peptides have been isolated from the hot water extract of CSP and were structurally characterized by FAB-MS and 2D-NMR techniques [103]. Their amino-acidic sequences, including a mostly lipophlic heptapeptide 124, a di-hydroxylated heptapeptide 125, and a Glutamate-rich octapeptide 126, are reported in Figure 8.

Figure 8.

Primary structure of cyclic peptide isolated from the C. sinensis peels.

Advertisement

3. Conclusions

The chemical richness of the primary and secondary metabolome of C. sinesnis species is undoubtedly impressive. Thousands of different compounds belonging to dozens of structural classes have been isolated and described. The most deeply investigated are sure, on one hand, the mixtures of volatile compounds composing the blend of the essential oil and, on the other hand, polyphenols, especially flavonoids.

The chemical composition of the extract from the exocarp of C. sinensis does differ from the composition of juice, or leaf extracts for some aspects [104]: the presence of a higher amount of more lipophilic compounds such as polymethoxy-flavonoids, r carotenoids, higher alkanes; a lesser extent of lighter terpenoids, a lower content of glycosylated compounds, the absence of cyanidins and sterols.

It is also a matter of fact that several interesting bioactivities were disclosed in the last years for the C. sinensis extracts that have been variously associated with the well-recognized beneficial effects that regular sweet oranges consumption may have on human health. However, a great deal of research work is still needed to clarify the molecular basis and the mechanism of these chemopreventive effects and to relate them with precise chemical entities that can be recognized as valuable nutraceuticals, as it is already the case for the well-established antioxidants Ascorbic acid, Hesperidin, Hesperetin, Quercetin, etc.

Advertisement

Conflict of interest

The author declares no conflict of interest.

References

  1. 1. Food and Agriculture Organization of the United nations: Citrus fruit fresh and processed - Statistical Bulletin 2016 [Internet]. 2017. Available from: http://www. http://www.fao.org/3/a-i8092e.pdf [Accessed: 2020-12-16]
  2. 2. Tamburino V, Zama DA. I sottoprodotti dell’industria di trasformazione: il pastazzo di agrumi. In: Vacante V, editor. Citrus - Trattato di agrumicoltura. Il Sole 24 Ore-Edagricole; 2009. p. 459–470
  3. 3. Putnik P, Kovačević DB, Jambrak AR, Barba FJ, Cravotto G, Binello A, Lorenzo JM, Shpigelman A. Innovative “Green” and Novel Strategies for the Extraction of Bioactive Added Value Compounds from CitrusWastes—A Review. Molecules. 2017:22:680–704. DOI: 10.3390/molecules22050680
  4. 4. Senit JJ, Velasco D, Gomez Manrique A, Sanchez-Barba M, Toledo LM, Santos VE, Garcia-Ochoa F, Yustos P, Ladero M. Orange peel waste upstream integrated processing to terpenes, phenolics, pectin and monosaccharides: Optimization approaches. Ind. Crops & Prod. 2019:134:370–381. DOI: 10.1016/j.indcrop.2019.03.060
  5. 5. Lamine M, Gargouri M, Rahali FZ, Mliki A. Recovering and Characterizing Phenolic Compounds From Citrus By-Product: A Way Towards Agriculture of Subsistence and Sustainable Bioeconomy. Waste Biomass Valor. 2020. DOI: 10.1007/s1264 9-020-01306 -9
  6. 6. Williams CA. Specialized dietary supplements. In: Geor RJ, Harris PA, Coenen M, editors. Equine Applied and Clinical Nutrition. Saunders; 2013. p. 351–366. DOI: 10.1016/C2009-0-39370-8
  7. 7. Patel S, Shukla S. Fermentation of Food Wastes for Generation of Nutraceuticals and Supplements. In: Frias J, Martinez-Villaluenga C, Peñas E, editors. Fermented Foods in Health and Disease Prevention. Academic Press; 2016. p. 707–734. DOI: 10.1016/C2014-0-01734-0
  8. 8. Khan N, Mongas M, Urpi-sarda M, Llorach R, Andres-La Cueva C. Contribution of Bioactive Foods and Their Emerging Role in Immunomodulation, Inflammation, and Arthritis. In: Watson RR. Preedy VR, editors. Bioactive Food as Dietary Interventions for Arthritis and Related Inflammatory Diseases. Academic Press; 2013. p. 43–65. DOI: 10.1016/C2011-0-07467-7
  9. 9. Rehman Z. Citrus peel extract – A natural source of antioxidant. Food Chem. 2006;99:450–454. DOI: 10.1016/j.foodchem.2005.07.054
  10. 10. Abe S, Fan K, Ho CT, Ghai G, Yang K. Chemopreventive Effects of Orange Peel Extract (OPE) II. OPE Inhibits Atypical Hyperplastic Lesions in Rodent Mammary Gland. J. Med. Food 2007;10:18–24; DOI: 10.1089/jmf.2006.0215
  11. 11. Lai CS, Li S, Miyauchi Y, Suzawa M, Hob CT, Pan MH. Potent anti-cancer effects of citrus peel flavonoids in human prostate xenograft tumors. Food Funct. 2013;4:944–949; DOI: 10.1039/c3fo60037h
  12. 12. Tung YC, Chang WT, Li S, Wu JC, Badmeav V, Ho CT, Pan MH. Citrus peel extracts attenuated obesity and modulated gut 1 microbiota in a high-fat diet-induced obesity mice. Food Funct. 2018;9:3363–3373; 10.1039/C7FO02066J
  13. 13. Qiao Y, Jun Xie B, Zhang Y, Zhang Y, Fan G, Yao XL, Pan SY. Characterization of Aroma Active Compounds in Fruit Juice and Peel Oil of Jinchen Sweet Orange Fruit (Citrus sinensis (L.) Osbeck) by GC-MS and GC-O. Molecules. 2008;13:1333–1344; DOI: 10.3390/molecules13061333
  14. 14. Dosoky NS, Setzer WN. Biological Activities and Safety of Citrus spp. Essential Oils. Int. J. Mol. Sci. 2018;19:1966; DOI: 10.3390/ijms19071966
  15. 15. Ettoumi KY, Zouambia Y, Moulai-Mostefa N. Chemical composition, antimicrobial and antioxidant activities of Algerian Citrus sinensis essential oil extracted by hydrodistillation assisted by electromagnetic induction heating. J Food Sci Technol DOI: 10.1007/s13197-020-04808-5
  16. 16. Leherbauer I, Stappen I. Selected essential oils and their mechanisms for therapeutic use against public health disorders. An overview. Z. Naturforsch. 2020;75: 205–223
  17. 17. Singh P, Shukla R, Prakash B, Kumar A, Singh S, Mishra PK, Dubey NK. Chemical profile, antifungal, antiaflatoxigenic and antioxidant activity of Citrus maxima Burm. and Citrus sinensis (L.) Osbeck essential oils and their cyclic monoterpene, DL-limonene. Food Chem. Toxicol. 2010;48:1734–1740; DOI:10.1016/j.fct.2010.04.001
  18. 18. O’Bryan CA, Crandall PG, Chalova VI, Ricke SC. Orange Essential Oils Antimicrobial Activities against Salmonella spp. J. Food Sc. 2008;73:M264-M267; DOI: 10.1111/j.1750-3841.2008.00790.x
  19. 19. Fisher K, Phillips CA. The effect of lemon, orange and bergamot essential oils and their components on the survival of Campylobacter jejuni, Escherichia coli O157, Listeria monocytogenes, Bacillus cereus and Staphylococcus aureus in vitro and in food systems. J. Appl. Microbiol. 2006;101:1232–1240; DOI:10.1111/j.1365-2672.2006.03035.x
  20. 20. Muthaiyan A, Martin EM, Natesan S, Crandall PG, Wilkinson BJ, Ricke SC. Antimicrobial effect and mode of action of terpeneless cold-pressed Valencia orange essential oil on methicillin-resistant Staphylococcus aureus. J. Appl. Microbiol. 2012;112:1020–1033; DOI:10.1111/j.1365-2672.2012.05270.x
  21. 21. Hung PV, Chi PTL, Phi NTL. Comparison of antifungal activities of Vietnamese citrus essential oils. Nat. Prod. Res. 2013;27:506–508; DOI: 10.1080/14786419.2012.706293
  22. 22. Najar B, Shortrede JE, Pistelli L, Buhagiar J. Chemical Composition and in Vitro Cytotoxic Screening of Sixteen Commercial Essential Oils on Five Cancer Cell Lines. Chem. Biodiversity 2020;17:e1900478; DOI: 10.1002/cbdv.201900478
  23. 23. Faturi CB, Leite JR, Alves PB, Canton AC, Teixeira-Silva F. Anxiolytic-like effect of sweet orange aroma in Wistar rats. Prog. Neuro-Psychoph. 2010;34:605–609; DOI:10.1016/j.pnpbp.2010.02.020
  24. 24. Uçar Y. Antioxidant Effect of Nanoemulsions Based on Citrus Peel Essential Oils: Prevention of Lipid Oxidation in Trout. Eur. J. Lipid Sci. Technol. 2020;122:1900405–1900419; DOI: 10.1002/ejlt.201900405
  25. 25. Ciriminna R, Meneguzzo F, Pagliaro M. Orange Oil. In: Nollet LML, Rathore HS editors. Green Pesticides Handbook – Essential Oils for Pest Control. CRC Press; 2017. p. 215–265
  26. 26. Lehrner J, Eckersberger C, Walla P, Pötsch G, Deecke L. Ambient odor of orange in a dental office reduces anxiety and improves mood in female patients. Physiology & Behavior 2000;71:83–86
  27. 27. Anandakumar P, Kamaraj S, Vanitha MK. D-limonene: A multifunctional compound with potent therapeutic effects. J. Food Biochem. 2020;45:e13566; DOI: 10.1111/jfbc.13566
  28. 28. Li S, Lo CY, Ho CT. Hydroxylated Polymethoxyflavones and Methylated Flavonoids in Sweet Orange (Citrus sinensis) Peel. J. Agric. Food Chem. 2006;54:4176–4185. DOI: 0.1021/jf060234n
  29. 29. Gao Z, Gao W, Zeng SL, Li P, Liu EH. Chemical structures, bioactivities and molecular mechanisms of citrus polymethoxyflavones. J. Funct. Foods 2018;40:498–509; DOI: 10.1016/j.jff.2017.11.036
  30. 30. Owis AL. Citrus Polymethoxyflavones: Biofunctional Molecules of Therapeutic Interest. Studies in Nat. Prod. Chem. 2019;59: 509–530. DOI: 10.1016/B978-0-444-64179-3.00015-3
  31. 31. Lai CS, Wu JC, Ho CT, Pan MH. Disease chemopreventive effects and molecular mechanisms of hydroxylated polymethoxyflavones. Biofactors 2015;41:301–313; DOI 10.1002/biof.1236
  32. 32. Del Río JA, Gómez P, Báidez AG, Arcas MC, Botía JM, Ortuño A. Changes in the Levels of Polymethoxyflavones and Flavanones as Part of the Defense Mechanism of Citrus sinensis (Cv. Valencia Late) Fruits against Phytophthora citrophthora. J. Agric. Food Chem. 2004;52:1913–1917; DOI: 10.1021/jf035038k
  33. 33. Mizuno M, Iinuma M, Ohara M, Tanaka T, Iwamasa M. Chemotaxonomy of the Genus Citrus Based on Polymethoxyflavones. Chem. Pharm. Bull. 1991;39:945–949; DOI: 10.1248/cpb.39.942
  34. 34. Kanaze FI, Termentzi A, Gabrieli C, Niopas I, Georgarakis M, Kokkaloua E. The phytochemical analysis and antioxidant activity assessment of orange peel (Citrus sinensis) cultivated in Greece–Crete indicates a new commercial source of hesperidin. Biomed. Chromatogr. 2009;23:239–249. DOI: 10.1002/bmc.1090
  35. 35. Montero-Calderon A, Cortes C, Zulueta A, Frigola A, Esteve MJ. Green solvents and Ultrasound- Assisted Extraction of bioactive orange (Citrus sinensis) peel compounds. Scientific Reports. 2019;9:16120–16128. DOI: 10.1038/s41598-019-52717-1
  36. 36. Manthey JA. Fractionation of Orange Peel Phenols in Ultrafiltered Molasses and Mass Balance Studies of Their Antioxidant Levels. J. Agric. Food Chem. 2004;52:7586–7592. DOI: 0.1021/jf049083j
  37. 37. Anagnostopoulou MA, Kefalas P, Kokkalou E,. Assimopoulou AN, Papageorgiou VP. Analysis of antioxidant compounds in sweet orange peel by HPLC–diode array detection–electrospray ionization mass spectrometry. Biomed. Chromatogr. 2005;19:138–148. DOI: 10.1002/bmc.430
  38. 38. Liew SS, Ho WY, Yeap SK, Bin Sharifudin SA. Phytochemical composition and in vitro antioxidant activities of Citrus sinensis peel extracts. PeerJ. 2018;6:e5331. DOI: 10.7717/peerj.5331
  39. 39. Guo C, Shan Y, Yang Z, Zhang L, Ling W, Liang Y, Ouyang Z, Zhong B, Zhang J. Chemical composition, antioxidant, antibacterial, and tyrosinase inhibition activity of extracts from Newhall navel orange (Citrus sinensis Osbeck cv. Newhall) peel. J Sci Food Agric 2020;100:2664–2674. DOI: 10.1002/jsfa.10297
  40. 40. Gosslau A, Chen KY, Ho CT, Li S. Anti-inflammatory effects of characterized orange peel extracts enrichedwith bioactive polymethoxyflavones. Food Sci. Human Well. 2014;3:26–35; DOI:10.1016/j.fshw.2014.02.002
  41. 41. Hagenlocher Y, Feilhauer K, Schäffer M, Bischoff SC, Lorentz A. Citrus peel polymethoxyflavones nobiletin and tangeretin suppress LPS- and IgE-mediated activation of human intestinal mast cells. Eur J Nutr 2017;56:1609–1620; DOI 10.1007/s00394-016-1207-z
  42. 42. Liu L, Xu X, Cheng D, Yao X, Pan S. Structure−Activity Relationship of Citrus Polymethoxylated Flavones and Their Inhibitory Effects on Aspergillus niger. J. Agric. Food Chem. 2012;60:4336–4341. DOI: 10.1021/jf3012163
  43. 43. Shetty SB, Mahin-Syed-Ismail P, Varghese S, Thomas-George B, Kandathil-Thajuraj P, Baby O, Haleem S, Sreedhar S, Devang-Divakar D. Antimicrobial effects of Citrus sinensis peel extracts against dental caries bacteria: An in vitro study. J Clin Exp Dent. 2016;8:e70–7. DOI:10.4317/jced.52493
  44. 44. Ortuño A, Báidez A, Gómez P, Arcas MC, Porras I, García-Lidón A, Del Río JA. Citrus paradisi and Citrus sinensis flavonoids: Their influence in the defence mechanism against Penicillium digitatum. Food Chem. 2006;98:351–358; DOI:10.1016/j.foodchem.2005.06.017
  45. 45. Bagavan A, Rahuman AA, Kamaraj C, Kaushik NK, Mohanakrishnan D, Sahal D. Antiplasmodial activity of botanical extracts against Plasmodium falciparum. Parasitol Res 2011;108:1099–1109. DOI: 10.1007/s00436-010-2151-0
  46. 46. Nakanishi M, Hino M, Yoshimura M, Amakura Y, Nomoto H. Identification of sinensetin and nobiletin as major antitrypanosomal factors in a citrus cultivar. Exp. Parasitol. 2019;200:24–29; DOI: 10.1016/j.exppara.2019.03.008
  47. 47. Lara Testai L, Calderone V. Nutraceutical Value of Citrus Flavanones and Their Implications in Cardiovascular Disease. Nutrients. 2017;9:502–515. DOI:10.3390/nu9050502
  48. 48. Shalaby NMM, Abd-Alla HI, Ahmed HH, Basoudan N. Protective effect of Citrus sinensis and Citrus aurantifolia against osteoporosis and their phytochemical constituents. J. Med. Plant. Res. 2011;5:579–588
  49. 49. Selmi S, Rtibi K, Grami D, Sebai H, Marzouki L. Protective effects of orange (Citrus sinensis L.) peel aqueous extract and hesperidin on oxidative stress and peptic ulcer induced by alcohol in rat. Lipids in Health and Disease. 2017;16:152–164. DOI: 10.1186/s12944-017-0546-y
  50. 50. Chen PY, Li S, Koh YC, Wu JC, Yang MJ, Ho CT, Pan MH. Oolong Tea Extract and Citrus Peel Polymethoxyflavones Reduce Transformation of L-Carnitine to Trimethylamine-N-Oxide and Decrease Vascular Inflammation in L-Carnitine Feeding Mice. J. Agric. Food Chem. 2019;67:7869–7879. DOI: 10.1021/acs.jafc.9b03092
  51. 51. Sathiyabama RG, Gandhia GR, Denadaib M, Sridharanc G, Jothic G, Sasikumard P, Siqueira Quintanse JdS, Narain N, Cuevas LE, Melo Coutinho HD, Barbosa Ramose AG, Quintans-Júniore LJ, Queiroz Gurgela R. Evidence of insulin-dependent signalling mechanisms produced by Citrus sinensis (L.) Osbeck fruit peel in an insulin resistant diabetic animal model. Food Chem. Toxicol. 2018;116:86–99. DOI: 10.1016/j.fct.2018.03.050
  52. 52. Ahmeda OM, Hassanb MA, Abdel-Twabc SM, Abdel Azeem MN. Navel orange peel hydroethanolic extract, naringin and naringenin have anti-diabetic potentials in type 2 diabetic rats. Biomed. & Pharmacotherapy 2017;94:197–205. DOI: 10.1016/j.biopha.2017.07.094
  53. 53. Ahmed OM, Fahim H, Ahmed HY, Al-Muzafar HM, Ahmed RR , Amin KA, El-Nahass ES, Abdelazeem WH. The Preventive Effects and the Mechanisms of Action of Navel Orange Peel Hydroethanolic Extract, Naringin, and Naringenin in N-Acetyl-p-aminophenol-Induced Liver Injury in Wistar Rats. Oxidative Medicine and Cellular Longevity. 2019; DOI: 10.1155/2019/2745352
  54. 54. Kim TW, Lee DR, Choi BK, Kang HK, Jung JY, Lim SW, Hwan Yang S, Suh JW. Hepatoprotective effects of polymethoxyflavones against acute and chronic carbon tetrachloride intoxication. Food Chem. Toxicol. 2016;91: 91–99; DOI: 10.1016/j.fct.2016.03.004
  55. 55. Chiu SP, Wu MJ, Chen PY, Ho YR, Tai MH, Ho CT, Yen JH. Neurotrophic action of 5-hydroxylated polymethoxyflavones: 5-demethylnobiletin and gardenin A stimulate neuritogenesis in PC12 cells. J. Agric. Food Chem. 2013;61:9453–9463; DOI: 10.1021/jf4024678
  56. 56. Lai CS, Ho MH, Tsai ML, Li S, Badmaev V, Ho CT, Pan MH. Suppression of Adipogenesis and Obesity in High-Fat Induced Mouse Model by Hydroxylated Polymethoxyflavones. J. Agric. Food Chem. 2013;61:10320–10328; DOI: 10.1021/jf402257t
  57. 57. Sergeev IN, Li S, Ho CT, Rawson NE, Dushenkov S. Polymethoxyflavones Activate Ca2+-Dependent Apoptotic Targets in Adipocytes. J. Agric. Food Chem. 2009;57:5771–5776; DOI:10.1021/jf901071k
  58. 58. Yu Wang Y, Lee PS, Chen YF, Ho CT, Pan MH. Suppression of Adipogenesis by 5-Hydroxy-3,6,7,8,30,40-Hexamethoxyflavone from Orange Peel in 3T3-L1 Cells. J. Med. Food 2016;19:830–835; DOI: 10.1089/jmf.2016.0060
  59. 59. Li GJ, Wang J, Cheng YJ, Tan X, Zhai YL, Wang Q, Gao FJ, Liu GL, Xin Zhao X, Hua Wang H. Prophylactic Effects of Polymethoxyflavone-Rich Orange Peel Oil on Nω-Nitro-L-Arginine-Induced Hypertensive Rats. Appl. Sci. 2018;8:752–768; DOI:10.3390/app8050752
  60. 60. Miyata Y, Oshitari T, Okuyama Y, Shimada A, Takahashi H, Natsugari H, Kosano H. Polymethoxyflavones as agents that prevent formation of cataract: Nobiletin congeners show potent growth inhibitory effects in human lens epithelial cells. Bioorg. Med. Chem. Lett. 2013;23:183–187; DOI: 10.1016/j.bmcl.2012.10.133
  61. 61. Li G, Tan F, Zhang Q, Tan A, Cheng Y, Zhou Q, Liu M, Tan X, Huang L, Rouseff R, Wu H, Zhao X, Liang G, Zhao X. Protective effects of polymethoxyflavone-rich cold-pressed orange peel oil against ultraviolet B-induced photoaging on mouse skin. J. Funct. Foods 2020; 67:103834–103844; DOI: 10.1016/j.jff.2020.103834
  62. 62. Zeng SL, Li SZ, Xiao PT, Cai YY, Chu C, Chen BZ, Li P, Li J, Liu EH. Citrus polymethoxyflavones attenuate metabolic syndrome by regulating gut microbiome and amino acid metabolism. Sci. Adv. 2020;6: eaax6208; DOI: 10.1126/sciadv.aax6208
  63. 63. Bajraktari-Sylejmani G, Weiss J. Potential Risk of Food-Drug Interactions: Citrus Polymethoxyflavones and Flavanones as Inhibitors of the Organic Anion Transporting Polypeptides (OATP) 1B1, 1B3, and 2B1. Eur. J. Drug Metab. Pharm. 2020;45:809–815; DOI: 10.1007/s13318-020-00634-4
  64. 64. Li S, Wang H, Guo L, Zhao H, Ho CT. Chemistry and bioactivity of nobiletin and its metabolites. J. Funct. Food 2014;6;2–10. DOI: 10.1016/j.jff.2013.12.011
  65. 65. Li W, Wang X, Niu X, Zhang H, He Z, Wang Y,1 Zhi W, Liu F. Protective Effects of Nobiletin Against Endotoxic Shock in Mice Through Inhibiting TNF-α, IL-6, and HMGB1 and Regulating NF-κB Pathway. Inflammation 2016;39:786–797. DOI: 10.1007/s10753-016-0307-5
  66. 66. Li W, Wang X, Zhi W, Zhang H, He Z, Wang Y, Liu F, Niu X, Zhang X. The gastroprotective effect of nobiletin against ethanol-induced acute gastric lesions in mice: impact on oxidative stress and inflammation. Immunopharm. Immunot. 2017;39:354–363; DOI: /10.1080/08923973.2017.1379088
  67. 67. Abe S, Hirose S, Nishitani M, Yoshida I, Tsukayama M, Tsuji A, Yuasa K. Citrus peel polymethoxyflavones, sudachitin and nobiletin, induce distinct cellular responses in human keratinocyte HaCaT cells. Biosci. Biotechnol. Biochem. 2018;82:2064–2071; DOI: 10.1080/09168451.2018.1514246
  68. 68. Kaneda H, Otomo R, Sasaki N, Omi T, Sato T, Kaneda T. Endothelium-independent vasodilator effects of nobiletin in rat aorta. J. Pharmacol. Sci. 2019;140:48–53; DOI: 10.1016/j.jphs.2019.04.004
  69. 69. Wen X, Zhao H, Wang L, Wang L, Du G, Guan W, Liu J, Cao X, Jiang X, Tian J, Wang M, Ho CT, Li S. Nobiletin Attenuates DSS-Induced Intestinal Barrier Damage through the HNF4α-Claudin-7 Signaling Pathway. J. Agric. Food Chem. 2020;68:4641–4649; DOI: 10.1021/acs.jafc.0c01217
  70. 70. Koolaji N, Shammugasamy B, Schindeler A, Dong Q, Dehghani F, Valtchev P. citrus Peel Flavonoids as Potential Cancer Prevention Agents. Current Developments In Nutrition. 2020;4:ID nzaa025;DOI: 10.1093/cdn/nzaa025
  71. 71. Tung, YC, Chou YC, Hung WL, Cheng AC, Yu RC, Ho CT, Pan MH. Polymethoxyflavones: Chemistry and Molecular Mechanisms for Cancer Prevention and Treatment. Curr. Pharmacol. Rep. 2019;5:98–113; DOI: 10.1007/s40495-019-00170-z
  72. 72. Wang L, Wang J, Fang L, Zheng Z, Zhi D, Wang S, Li S, Ho CT, Zhao H. Anticancer Activities of Citrus Peel Polymethoxyflavones Related to Angiogenesis and Others. Biomed. Res. Int. 2014; Article ID 453972; DOI: 10.1155/2014/453972
  73. 73. Chan EWC, Soo OYM, Tan YH, Wong SK, Chan HT. Nobiletin and tangeretin (citrus polymethoxyflavones): an overview on their chemistry, pharmacology and cytotoxic activities against breast cancer. J. Chin. Pharm. Sci. 2020;29:443–454; DOI: 10.5246/jcps.2020.07.042
  74. 74. Sergeev IN, Ho CT, Li S, Colby J, Dushenkov S. Apoptosis-inducing activity of hydroxylated polymethoxyflavones and polymethoxyflavones from orange peel in human breast cancer cells. Mol. Nutr. Food Res. 2007;51:1478–1484. DOI: 10.1002/mnfr.200700136
  75. 75. Rahideh ST, Keramatipour M, Nourbakhsh M, Koohdani F, Hoseini M, Talebi S, Shidfar F. Comparison of the effects of Nobiletin and Letrozole on the activity and expression of aromatase in MCF-7 breast cancer cell line. Biochem. Cell. Biol. 2017;95:468–473;DOI: 10.1139/bcb-2016-0206
  76. 76. Sergeev IN, Li S, Colby J, Ho CT, Dushenkov S. Polymethoxylated flavones induce Ca2+−mediated apoptosis in breast cancer cells. Life Sci. 2006;80:245–253; DOI: 10.1016/j.lfs.2006.09.006
  77. 77. Borah N, Gunawardana S, Torres H, McDonnell S, Van slambrouck S. 5,6,7,3′,4′,5'-Hexamethoxyflavone inhibits growth of triple-negative breast cancer cells via suppression of MAPK and Akt signaling pathways and arresting cell cycle. Int. J. Oncol. 2017;51:1685–1693; DOI: 10.3892/ijo.2017.4157
  78. 78. Ademosun AO, Oboh G, Passamonti S, Tramer F, Ziberna L, Augusti Boligon A, Athayde ML. Inhibition of metalloproteinase and proteasome activities in colon cancer cells by citrus peel extracts. J Basic Clin Physiol Pharmacol 2015;26: 471–477. DOI: 10.1515/jbcpp-2013-0127
  79. 79. Kaur J, Kaur G. An insight into the role of citrus bioactives in modulation of colon cancer. J. Funct. Foods. 2015;13:239–261;DOI: 10.1016/j.jff.2014.12.043
  80. 80. Qiu P, Cui Y, Xiao H, Han Z, Ma H, Tang Y, Xu H, Zhang L. 5-Hydroxy polymethoxyflavones inhibit glycosaminoglycan biosynthesis in lung and colon cancer cells. J. Funct. Foods. 2017;30:39–47. DOI: 10.1016/j.jff.2017.01.008
  81. 81. Silva I, Estrada MF, Pereira CV, Bento da Silva A, Bronze MR, Alves PM,. Duarte CMM, Brito C, Serra AT. Polymethoxylated Flavones from Orange Peels Inhibit Cell Proliferation in a 3D Cell Model of Human Colorectal Cancer. Nutrition and Cancer. 2018;70:257–266. DOI: 10.1080/01635581.2018.1412473
  82. 82. Charoensinphon N, Qiu P, Dong P, Zheng J, Ngauv P, Cao Y, Li S, Ho CT, Xiao H. 5-Demethyltangeretin inhibits human nonsmall cell lung cancer cell growth by inducing G2/M cell cycle arrest and apoptosis. Mol. Nutr. Food Res. 2013;57:2103–2111; DOI: 10.1002/mnfr.201300136
  83. 83. Xiao H, Yang CS, Li S, Jin H, Ho CT, Patel T. Monodemethylated polymethoxyflavones from sweet orange (Citrus sinensis) peel inhibit growth of human lung cancer cells by apoptosis. Mol. Nutr. Food Res. 2009;53:398–406; DOI 10.1002/mnfr.200800057
  84. 84. Wang Y, Chen Y, Zhang H, Chen J, Cao J, Chen Q, Li X, Sun C. Polymethoxyflavones from citrus inhibited gastric cancer cell proliferation through inducing apoptosis by upregulating RARβ, both in vitro and in vivo. Food Chem. Toxicol. 2020;146:111811–111821; DOI: 10.1016/j.fct.2020.111811
  85. 85. Li S, Pan MH, Lai CS, Lo CY, Dushenkovc S, Ho CT. Isolation and syntheses of polymethoxyflavones and hydroxylated polymethoxyflavones as inhibitors of HL-60 cell lines. Bioorg. Med. Chem. 2007;15:3381–338. DOI: 10.1016/j.bmc.2007.03.021
  86. 86. Fan K, Kurihara N, Abe S, Ho CT, Ghai G, Yang K. Chemopreventive Effects of Orange Peel Extract (OPE) I. OPE Inhibits Intestinal Tumor Growth in ApcMin+ Mice. Med. Food 2007;10:11–17. DOI: 10.1089/jmf.2006.0214
  87. 87. Tajaldinia M, Samadia F, Khosravib A, Ghasemnejadd A, Asadie J. Polymethoxylated Protective and anticancer effects of orange peel extract and naringin in doxorubicin treated esophageal cancer stem cell xenograft tumor mouse model. Biomed. Pharm. 2020;121:e109594. DOI: 10.1016/j.biopha.2019.109594
  88. 88. Lia Y, Kandhare AD, Mukherjee AA, Bodhankar SL. Acute and sub-chronic oral toxicity studies of hesperidin isolated from orange peel extract in Sprague Dawley rats. Regul. Toxicol Pharm. 2019;105:77–85. DOI: 10.1016/j.yrtph.2019.04.001
  89. 89. Bellavite P, Donzelli A. Hesperidin and SARS-CoV-2: New Light on the Healthy Function of Citrus Fruits. Antioxidants. 2020;9:742–760. DOI: 10.3390/antiox9080742
  90. 90. Ganeshpurkar A, Saluja A. The pharmacological potential of hesperidin. Indian J. Biochem. Biophys. 2019;56:287–300
  91. 91. Murray RDH. Naturally Occurring Plant Cumarins. In: Herz W, Grisebach H, Kirby GW, editors. Progress in the Chemistry of Organic Natural Products. Springer-Verlag. New York; 1978. p. 199–400
  92. 92. Mohamed TK. Chemical constituents and antioxidant activity of Citrus paradisi (star-ruby red grapefruit) and Citrus sinensis (blood sweet orange) Egyptian cultivars. Asian J. Chem. 2004:887815
  93. 93. Cunha AG, Gandini A. Turning polysaccharides into hydrophobic materials: a critical review. Part 2. Hemicelluloses, chitin/chitosan, starch, pectin and alginates. Cellulose. 2010;17:1045–1065. DOI: 10.1007/s10570-010-9435-5
  94. 94. Sua DL, Li PJ, Quekc SY, Huang ZQ, Yuan YJ, Li GY, Shan Y. Efficient extraction and characterization of pectin from orange peel by a combined surfactant and microwave assisted process. Food Chem. 2019;286:1–7. DOI: 10.1016/j.foodchem.2019.01.200
  95. 95. Hosseini SS, Khodaiyan F, Kazemi M, Najari Z. Optimization and characterization of pectin extracted from sour orange peel by ultrasound assisted method. Int. J. Biol. Macromol. 2019;125:621–629. DOI: 10.1016/j.ijbiomac.2018.12.096
  96. 96. Jiao WB, Huang D, Xing F, Hu Y, Deng XX, Xu Q, Chen LL. Genome-wide characterization and expression analysis of genetic variants in sweet oranged. The Plant Journal 2013;75:954–964. DOI: 10.1111/tpj.12254
  97. 97. Jansen EF, Jang R, Mac Donnel LR. Citrus acetylesterase. Arch Biochem. 1947;15:415–431
  98. 98. Pasta P, Verga R, Zambianchi F, Daminati M. Acetyl Esterase from Mediterranean Oranges: Partial Purification, Immobilisation and Biotransformations. Biocatal. & Biotrasf. 2004;22:221–224. DOI: 10.1080/10242420410001697089
  99. 99. Fontana G, Bruno M, Maggio A, Rosselli S. Functional investigation and applications of the acetylesterase activity of the Citrus sinensis (L.) Osbeck peel. Nat. Prod. Res. 2020; DOI: 10.1080/14786419.2020.1737055
  100. 100. Wei Y, Tan YL, Ang FL, Zhao H. Identification and Characterization of Citrus Peel Uronic Acid Oxidase. ChemBioChem 2020;21:797–800. DOI: 10.1002/cbic.201900546
  101. 101. Rani G, Yadav L, Kalidhar SB. Chemical Examination of Citrus sinensis Flavedo Variety Pineapple. Indian J. Pharm. Sci. 2009;71:677–679
  102. 102. Lux PE, Carle R, Zacarías L, Rodrigo MJ, Schweiggert RM, Steingass CB. Genuine Carotenoid Profiles in Sweet Orange [Citrus sinensis (L.) Osbeck cv. Navel] Peel and Pulp at Different Maturity Stages. J. Agric. Food Chem. 2019; 67:13164–13175. DOI: 10.1021/acs.jafc.9b06098
  103. 103. Matsubara Y, Yusa T, Sawabe A, Itzuka Y, Takekuma S, Yoshida Y. Structures of New Cyclic Peptides in Young Unshiu (Citrus unshiu Marcov.), Orange (Citrus sinensis Osbeck.) and Amanatzu (Citrus natzudaidai) Peelings. Agric. Biol. Chem. 1991;55:2923–2929
  104. 104. Favela-Hernández JMJ, González-Santiago O, Ramírez-Cabrera MA. Esquivel-Ferriño PC, Camacho-Corona MdR. Chemistry and Pharmacology of Citrus sinensis. Molecules. 2016;21, 247–271. DOI: 10.3390/molecules21020247

Written By

Gianfranco Fontana

Submitted: 22 September 2020 Reviewed: 29 January 2021 Published: 30 September 2021