You are on page 1of 16

Biomedicine & Pharmacotherapy 167 (2023) 115467

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

1,8-cineole (eucalyptol): A versatile phytochemical with therapeutic


applications across multiple diseases
Cosima C. Hoch a, Julie Petry a, Lena Griesbaum a, Tobias Weiser a, Kathrin Werner a,
Michael Ploch b, Admar Verschoor a, Gabriele Multhoff c, Ali Bashiri Dezfouli a, c,
Barbara Wollenberg a, *
a
Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
b
Cassella-med GmbH & Co. KG, 50670 Cologne, Germany
c
Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, 81675
Munich, Germany

A R T I C L E I N F O A B S T R A C T

Keywords: 1,8-cineole (Eucalyptol), a naturally occurring compound derived from botanical sources such as eucalyptus,
1,8-cineole rosemary, and camphor laurel, has a long history of use in traditional medicine and exhibits an array of biological
Eucalyptol properties, including anti-inflammatory, antioxidant, antimicrobial, bronchodilatory, analgesic, and pro-
Respiratory disorders
apoptotic effects. Recent evidence has also indicated its potential role in managing conditions such as Alz­
Anti-inflammatory activity
Antimicrobial
heimer’s disease, neuropathic pain, and cancer. This review spotlights the health advantages of 1,8-cineole, as
Antioxidant demonstrated in clinical trials involving patients with respiratory disorders, including chronic obstructive pul­
monary disease, asthma, bronchitis, and rhinosinusitis. In addition, we shed light on potential therapeutic ap­
plications of 1,8-cineole in various conditions, such as depression, epilepsy, peptic ulcer disease, diarrhea,
cardiac-related heart diseases, and diabetes mellitus. A comprehensive understanding of 1,8-cineole’s pharma­
codynamics and safety aspects as well as developing effective formulations, might help to leverage its therapeutic
value. This thorough review sets the stage for future research on diverse health benefits and potential uses of 1,8-
cineole in tackling complex medical conditions.

1. Introduction has been historically used in traditional medicine across different cul­
tures, such as Chinese, Indian, and Australian Aboriginal medicines, for
Plants have long been recognized as a significant reservoir of active treating a variety of conditions, including respiratory and digestive is­
pharmaceutical ingredients for drug discovery, and their naturally sues, fever, and pain [6].
occurring small molecules have emerged as a prominent class of thera­ In Germany, Soledum™ enteric-coated capsules, which contain 100
peutic compounds [1]. Among these, 1,8-cineole (1,3,3-trimethy­ mg or 200 mg (forte) of 1,8-cineole (CNL-1976) per capsule, have been
l-2-oxabicyclo[2.2.2]octane), also referred to as Eucalyptol, is a registered as authorized medicinal products for several years and are
naturally derived compound found in the essential oil fraction of various utilized in treating inflammatory respiratory disorders such as the
botanical sources, including eucalyptus, rosemary, and camphor laurel common cold, bronchitis, sinusitis, bronchial asthma, and chronic
[2]. obstructive pulmonary disease (COPD) [7].
Eucalyptus nicholii essential oil is renowned for its high 1,8-cineole After oral administration, 1,8-cineole is absorbed in the small in­
concentration, which can reach levels of up to 90% [3]. The oil com­ testine and subsequently metabolized in the liver by human cytochrome
prises mainly volatile organic compounds, including monoterpenes and P450 enzymes (CYP3A4/5). Its primary metabolites, 2alpha-hydroxy-
sesquiterpenes [4]. Due to its pleasant flavor and aroma, 1,8-cineole is and 3alpha-hydroxy-1,8-cineole, are excreted through urine [8]. In
frequently used in food, fragrances, and cosmetics [5]. Additionally, it addition to its systemic availability in the bloodstream, 1,8-cineole can

* Correspondence to: Department of Otolaryngology, Head and Neck Surgery. School of Medicine, Technical University of Munich (TUM), Ismaningerstrasse 22,
81675 Munich, Germany.
E-mail address: barbara.wollenberg@tum.de (B. Wollenberg).

https://doi.org/10.1016/j.biopha.2023.115467
Received 23 July 2023; Received in revised form 28 August 2023; Accepted 7 September 2023
Available online 9 September 2023
0753-3322/© 2023 The Author(s). Published by Elsevier Masson SAS. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/).
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

reach the bronchial system and be exhaled through the peripheral air­ 3. Mechanisms of action
ways. This allows its beneficial effects to extend throughout the entire
respiratory system, including the sinuses, which are a key source of 3.1. Anti-inflammatory mechanisms of 1,8-cineole
infection-related exacerbations [9,10].
1,8-cineole has gained widespread attention for its array of biolog­ 3.1.1. Regulation of inflammatory mediators and cytokines
ical properties, such as anti-inflammatory, antioxidant, mucolytic/ Pro-inflammatory cytokines are small proteins secreted by immune
secretolytic, bronchodilatory, and antimicrobial effects [11,12]. More­ cells, which play a crucial role in mediating and regulating inflammation
over, recent studies have highlighted the neuroprotective, analgesic, and in the body. Dysregulated production of these cytokines contributes to
pro-apoptotic properties of 1,8-cineole, underscoring its potential the development and progression of various inflammatory diseases [25].
beneficial role in a broad spectrum of conditions such as Alzheimer’s Venkataraman et al. identified the anti-inflammatory effects of 1,8-
disease, neuropathic pain, and cancer [13–15]. cineole on colonic inflammation using a dextran sodium sulfate (DSS)-
Given its extensive range of beneficial effects across multiple dis­ induced mouse model of colitis [26]. 1,8-cineole effectively prevented
eases, there has been a growing interest in exploring the mechanisms of colonic shortening, reduced myeloperoxidase (MPO) enzyme activity,
action and potential therapeutic applications of 1,8-cineole in various and decreased pro-inflammatory cytokine levels, such as interleukin
conditions, including respiratory, neurological, gastrointestinal, and (IL)− 6, IL-1β, tumor necrosis factor (TNF)-α, and IL-17A. In addition, 1,
oncological disorders. Therefore, this review aims to critically evaluate 8-cineole reduced the DSS-induced expression of pro-inflammatory
the available evidence on the effects of 1,8-cineole, mechanisms of ac­ mediators, including cyclooxygenase-2 (COX2) and inducible nitric
tion, and potential clinical applications, while identifying promising oxide synthase (iNOS), at both the protein and mRNA levels [26].
areas for future research. Moreover, in an acute pancreatitis mouse model, it has been shown that
treatment with 1,8-cineole resulted in decreased cytokine levels of
2. Pharmacological properties TNF-α, IL-1β, and IL-6, mirroring the effects seen with thalidomide, a
TNF-α inhibitor. Of note, the anti-inflammatory cytokine IL-10 increased
The pharmacological properties exhibited by 1,8-cineole are of upon 1,8-cineole treatment [27]. A study investigating trinitrobenzene
profound significance, underlining its versatile attributes. At room sulfonic acid-induced colitis in rats also demonstrated similar protective
temperature, this compound takes the form of a clear liquid, with a effects of 1,8-cineole, particularly in relation to reduced cytokine levels
melting point recorded at 1.5 ◦ C and a flash point at 49 ◦ C [16]. Marked [28].
by a logP value of 2.74, 1,8-cineole strikes an optimal equilibrium be­ In a guinea pig model of ovalbumin (OVA)-induced asthma, Bastos
tween solubility and permeability, hinting at its favorable potential for et al. found that inhalation of 1,8-cineole mitigated tracheal contrac­
oral bioavailability [17]. Emerging from isoprene units (C5H8), mono­ tions, reduced TNF-α and IL-1β levels, and inhibited eosinophil and
terpenes like 1,8-cineole are synthesized via the activation of iso­ neutrophil aggregation in bronchoalveolar lavage fluid (BALF) samples
pentenyl pyrophosphate and dimethylallyl pyrophosphate [18]. These [29]. Juergens et al. documented that 1,8-cineole decreased the release
foundational units undergo iterative linkage, culminating in the creation of prostaglandin E2 and leukotriene B4 (LTB4) from peripheral blood
of diverse terpenes distinguished by their carbon backbones, including mononuclear cells in asthmatic patients, and reduced TNF-α, IL-1β,
monoterpenes (C10) and sesquiterpenes (C15). LTB4, and thromboxane B2 in lipopolysaccharide (LPS)-stimulated pe­
In terms of pharmacokinetics, investigations have shed light on the ripheral blood monocytes of healthy individuals [30].
behavior of 1,8-cineole. In rabbits, oral administration of 200 mg/kg has Furthermore, treatment with 1,8-cineole resulted in a reduction in
led to rapid attainment of peak plasma concentration within 1 h, indi­ vascular endothelial inflammation mediated by pro-inflammatory cy­
cating efficient absorption [19]. Biotransformation investigations con­ tokines in LPS-induced systemic inflammation in mice [31] and
cerning 1,8-cineole have been conducted on brushtail possums and decreased the expression of TNF-α and IL-1β mRNA expression in human
rabbits [20,21]. These studies have successfully identified a range of umbilical vein endothelial cells (HUVECs) after LPS stimulation in vitro
hydroxylated derivatives, among them 7-hydroxy-1,8-cineole and 9-hy­ [32].
droxy-1,8-cineole. Rodent models revealed oxidative modifications 1,8-cineole has also been shown to effectively reduce Dermatopha­
yielding metabolites like 2-hydroxy-1,8-cineole and 3-hydroxy-1, goides pteronyssinus (Der p)-triggered cytokine production and intra­
8-cineole, present within 2 h post-oral dosing. Additionally, the corre­ cellular toll-like receptor (TLR)− 4 expression in human bronchial
sponding diols, cineolic acids, and hydroxylcineolic acids have been epithelial cells [33]. Moreover, in a Der p-induced mouse model, 1,
recognized as phase I metabolites, present in both urine and blood 8-cineole treatment significantly decreased airway hyper­
plasma. Subsequent conjugation formed glucuronide products, a pattern responsiveness and the number of eosinophils in BALF, as well as the
similarly observed in rat and human liver microsome studies [22]. production of IL-4, IL-13, and IL-17A [33].
Addressing the compound’s toxicity profile, the oral acute LD50 value The effectiveness of 1,8-cineole in suppressing pro-inflammatory
in rats is documented at 2480 mg/kg body weight [23]. Subacute cytokines in LPS-induced lung inflammation, human mononuclear
toxicity has been observed in rats at dose levels of 600 mg/kg body cells, and lung macrophages has been further confirmed in other studies
weight and beyond. Manifestations of this subacute toxicity included [34–36]. For instance, 1,8-cineole has demonstrated inhibitory proper­
reduced body weight and the presence of lesions in liver and kidney. No ties in both monocytes and lymphocytes, specifically targeting chemo­
indications of chronic or genotoxic effects attributed to 1,8-cineole have tactic cytokines like IL-8 and IL-5 [37]. As a matter of note, 1,8-cineole
been identified [24]. has shown promising potential as an anti-allergic agent by inhibiting the
Limited research exists regarding the metabolism of 1,8-cineole in production of IL-4. In the context of respiratory disorders, the observed
humans, and as of the present time, the identification of urinary me­ decrease in cytokine production indicates that 1,8-cineole operates via
tabolites following a single 100 mg dose of 1,8-cineole has revealed 2- an anti-inflammatory mechanism, effectively inhibiting
hydroxy-1,8-cineole and 3-hydroxy-1,8-cineole [8]. Nevertheless, cytokine-induced airway mucus hypersecretion rather than solely
given the considerably lower doses acquired through the consumption of functioning as a secretolytic agent. These findings are consistent with
1,8-cineole via herbal teas or spices, the significance of dose-dependent previous studies and highlight the potential of 1,8-cineole as an alter­
metabolism becomes pivotal in a comprehensive assessment encom­ native medication with anti-inflammatory properties which are com­
passing both pharmacological and toxicological dimensions. parable to those of steroids [7].
Finally, 1,8-cineole significantly influenced the phosphorylation and
activation of endothelial nitric oxide synthase 3 in tissue samples from
nasal polyps of chronic rhinosinusitis patients [38]. This effect is

2
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

particularly noteworthy since it suggests that 1,8-cineole may reduce macrophages of COPD patients [42].
inflammation and edema by regulating the vascular permeability. As There is also evidence that 1,8-cineole is capable of influencing
nasal nitric oxide (nNO) is a known mediator of ongoing sinus inflam­ neutrophils. In an in vivo experiment on Bleomycin-induced pulmonary
mation, the use of 1,8-cineole to reduce nNO levels presents a promising inflammation it was shown that 1,8-cineole can inhibit neutrophils
therapeutic approach for airway diseases, particularly chronic rhinosi­ pulmonary infiltration, suggesting a potential role for 1,8-cineole in
nusitis with nasal polyps (CRSwNP). regulating neutrophil migration and activation [40]. It has been
demonstrated that 1,8-cineole inhibits basophil activation mediated by
3.1.2. Modulation of immune cells IgE, which is involved in allergic reactions [43]. In a study involving an
Macrophages are phagocytic cells primarily responsible for engulfing IgE-mediated local allergic model, Nakamura et al. confirmed that the
and digesting foreign particles, debris, and pathogens, in helping to anti-inflammatory effect of 1,8-cineole in wild-type mice was attribut­
maintain tissue homeostasis and promoting tissue repair [39]. Rui et al. able to the suppression of mast cell activation when compared to mast
have shown that treatment with 1,8-cineole suppressed IL-13-induced cell-deficient mice [44]. The mechanism of action was further examined
polarization of M2 macrophage and mitigated pathological alterations in an in vitro cell system using bone-marrow-derived mast cells, which
associated with lung fibrosis in mice [40]. 1,8-cineole appeared to alter revealed that 1,8-cineole not only repressed mast cell degranulation but
M2 macrophage polarization by inhibiting the phosphorylation of also mitigated allergic dermatitis. According to the findings, the authors
signaling molecules such as STAT6 and p38 MAPK, and the expression of suggested that 1,8-cineole may have a direct or indirect impact on the
transcription factors such as KLF4 and PPAR-γ [40]. Moreover, 1, phosphorylation of PLCγ and p38 MAPK, contributing to its ability to
8-cineole hindered LPS-induced nitric oxide (NO) production in mouse suppress degranulation in mast cells.
macrophage cell lines [34], facilitated acute lung injury in a mouse
model [41], and mitigated active oxygen release by cultured alveolar

Fig. 1. The anti-inflammatory mechanisms of 1,8-cineole involve the inhibition of nuclear translocation of NF-κB p65 and PPARγ, leading to the suppression of
immune response genes. This regulatory process effectively modulates the expression of key inflammatory mediators, including TNF-alpha, IL-6, IL-8, VCAM-1, and
E-selectin. By targeting these crucial pathways, 1,8-cineole demonstrates its potential to mitigate inflammation and associated effects (Figure created using Bio­
Render, Toronto, ON, Canada).

3
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

3.1.3. PPARγ and NF-κB signaling pathways 8-cineole has been found to enhance the activities of antioxidant en­
Peroxisome proliferator-activated receptor-gamma (PPARγ) and zymes while reducing ROS levels, effectively alleviating the oxidative
nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) stress induced by monosodium urate. 1,8-cineole co-administered with
are two important cellular signaling molecules involved in inflammation bisphenol A (BPA) significantly increased the activities of antioxidant
which can be regulated by 1,8-cineole (Fig. 1). enzymes like SOD, glutathione peroxidase (GPx), and CAT, while
PPARγ is a member of the nuclear receptor superfamily and a ligand- reducing high levels of oxidative parameters, including MDA, iNOS, and
inducible transcription factor that plays a significant role in adipocyte NO in chickens [58].
function. Recent research has demonstrated that PPARγ is also crucial in The antioxidant capacity of 1,8-cineole extends beyond its ability to
regulating immune cells, cell proliferation, and in stimulating the anti- scavenge ROS directly. It has also been shown to chelate metal ions,
inflammatory activity of PPARγ [45]. Treatment with 1,8-cineole has contributing to ROS production [59]. By sequestering these ions, 1,
been shown to restore PPARγ protein activity in DSS-induced colitis 8-cineole can limit ROS formation and protect cells from oxidative
[26]. In addition, an increase in PPARγ expression has been observed in damage.
1,8-cineole-treated vascular endothelium dysfunction induced by LPS
[46]. Both 1,8-cineole and PPAR-γ agonists had comparable effects on 3.2.2. Inhibition of lipid peroxidation
blocking the protein and mRNA levels of VCAM-1, E-selectin, IL-6, and Lipid peroxidation (LP) is a detrimental process that can cause sig­
IL-8 triggered by LPS. The authors confirmed that the anti-inflammatory nificant cellular damage and has been implicated in the pathogenesis of
activity of 1,8-cineole can be reversed by PPAR-γ inhibitors [46]. various diseases, including rheumatoid arthritis, atherosclerosis,
Therefore, it is assumed that 1,8-cineole mediates its anti-inflammatory ischemia, carcinogenesis, and aging [60].
effects, at least in part by PPARγ activation. 1,8-cineole has demonstrated the ability to inhibit LP [61]. In a study
NF-κB is a crucial transcription factor in inflammation and immune examining the protective effect of 1,8-cineole against LP in rat liver
response regulation. Inflammatory stimuli such as LPS or TNF-α can microsomes, 1,8-cineole successfully inhibited the formation of thio­
activate the NF-κB pathway that increases the expression of pro- barbituric acid reactive substances, commonly used markers of LP [62].
inflammatory genes [47]. 1,8-cineole inhibited NF-κB-mediated This finding indicates that 1,8-cineole can protect cellular components,
signaling events by preventing the dissociation of the inhibitory subunit such as membranes and lipids, from oxidative damage.
IκBa and the translocation of the NF-κB subunit P65 into the nucleus [26, Furthermore, 1,8-cineole exhibited a dose-dependent protective ef­
48]. These findings are consistent with a study demonstrating fect on linoleic acid oxidation [63], an essential fatty acid that plays a
anti-inflammatory properties of 1,8-cineole in mice with acute lung critical role in cellular function and is a primary target of LP. By pre­
inflammation induced by cigarette smoke [49]. Specifically, 1,8-cineole venting the oxidation of linoleic acid, 1,8-cineole may help maintain
reduced the activation of the NF-κB p65 subunit, decreasing both in­ cellular homeostasis and protect against the development and progres­
flammatory cells and the cytokine secretion, including TNF-α. Moreover, sion of diseases associated with LP.
1,8-cineole has been shown to improve the inflammatory phenotype in
HUVECs by modulating the NF-κB expression in vitro [50]. 1,8-cineole 3.2.3. Nrf2/Keap1 signaling pathway
has been found to impede UVB-induced COX-2 protein and mRNA The antioxidant activity of 1,8-cineole is mediated, in part, by acti­
production in HaCaT cells [51]. The suppression of the aryl hydrocarbon vating the Nrf2/Keap1 system, a critical regulator of cellular protective
receptor (AhR) led to decreased COX-2 expression and diminished responses to oxidative stress and inflammation [64]. This pathway is
ERK1/2 phosphorylation in these cells. Additionally, the topical appli­ essential for maintaining cellular redox balance and promoting cell
cation of 1,8-cineole to the mouse skin effectively postponed tumor survival under stress conditions (Fig. 2).
formation, reduced the tumor growth, concurrently with obstructing the Treatment with 1,8-cineole promoted the nuclear translocation of
COX-2 expression in vivo. The bioinformatics application, TFactsS, Nrf2, a transcription factor that binds to antioxidant response elements
postulated the possibility of 1,8-cineole being able to regulate NF-κB in in the promoter regions of target genes, leading to increased expression
HaCaT cells [52]. However, deeper exploration is required to elucidate of phase II detoxifying enzymes and antioxidant proteins, such as heme
the transcription factors involved in the UVB-induced COX-2 expression oxygenase-1 and NAD(P)H: quinone oxidoreductase 1 (NOQ1) [26].
through the AhR/c-Src/EGFR/ERK pathway. 1,8-cineole’s inhibition of Additional studies have demonstrated the protective effects of 1,8-
COX2 may contribute to reducing inflammation by reducing the NF-κB cineole on liver cells and pheochromocytoma cells through activating
expression. the Nrf2/Keap1 system and increasing SOD expression [65–67]. In
various cellular and tissue models, 1,8-cineole treatment has been
3.2. Antioxidant properties of 1,8-cineole: mechanisms and cellular shown to enhance the activities of antioxidant enzymes, such as SOD,
effects GPx, and CAT, while reducing the levels of oxidative parameters
generated by different ROS inducers, such as LPS, hydrogen peroxide,
3.2.1. Scavenging of free radicals and cigarette smoke [68–70].
1,8-cineole’s antioxidant properties play a crucial role in its thera­
peutic potential, as it is effective in neutralizing reactive oxygen species 3.3. Broad-spectrum antimicrobial properties of 1,8-cineole
(ROS) and enhancing cellular defense mechanisms. Oxidative stress,
caused by ROS accumulation, is involved in the developing and pro­ 3.3.1. Antibacterial properties: activity, synergy with antibiotics, and
gression of various pathological conditions, including inflammation, impact on biofilm formation and cell morphology
aging, and neurodegenerative diseases [53]. Antimicrobial resistance is a pressing global health concern, and the
1,8-cineole has demonstrated potent free radical scavenging activity formation of biofilms by bacteria is a potential cause of resistance.
in a variety of in vitro assays, highlighting its ability to neutralize ROS Biofilms are structured communities of microorganisms that produce a
and protect cells from oxidative damage [54,55]. In grass carp hepato­ protective extracellular matrix that reduces the efficacy of antibiotics
cytes, 1,8-cineole treatment enhanced antioxidant enzymes activities, [71,72]. To address this issue, natural compounds derived from plants,
such as superoxide dismutase (SOD) and catalase (CAT), increased total such as the monoterpene 1,8-cineole, have gained significant attention
antioxidant capacity, and decreased ROS and malondialdehyde (MDA) for their potential to decrease antibiotic resistance (Table 1).
content, effectively attenuating oxidative stress caused by di 1,8-cineole has demonstrated antibacterial activity against various
(2-ethylhexyl)phthalate [56]. Gram-positive pathogenic strains, including Staphylococcus aureus, Ba­
Various models have demonstrated similar antioxidant effects, cillus subtilis, Streptococcus pyogenes, Bacillus cereus, Micrococcus flavus,
including RAW264.7 cells and ankle tissues [57]. In these studies, 1, and Staphylococcus epidermidis, as well as Gram-negative strains, such as

4
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

Table 1
A comprehensive summary of 1,8-cineole’s diverse antimicrobial properties,
including antibacterial, antifungal, and antiviral effects. HSV: herpes simplex
virus; IBV: infectious bronchitis virus; SARS-CoV-2: severe acute respiratory
syndrome coronavirus 2; IRF3: interferon regulatory factor 3.
Properties Details Potential References
Applications

Antibacterial • Effective against Gram- Development of [71,73–75,


positive strains (e.g., new antibacterial 78,80,84,
S. aureus, B. subtilis) and agents to eliminate 86–88,90,92,
Gram-negative strains antibiotic 95,96]
(e.g., E. coli, resistance and
P. aeruginosa), biofilm-associated
• Synergistic with infections
antibiotics like
gentamicin,
amoxicillin/clavulanic
acid, and penicillin G,
• Inhibits quorum sensing
and biofilm formation,
affecting bacterial
growth and motility,
• Modifies shape and size
of bacterial cells,
particularly in E. coli
and S. aureus
Antifungal • Inhibits growth of Development of [97–101]
various fungi, including new antifungal
F. oxysporum, agents to address
F. sporotrichioides, and limited efficacy and
A. tubingensis, toxicity risks of
• Disrupts fungal biofilm current antifungal
formation in F. solani medications
species complex,
Fig. 2. Molecular mechanisms of 1,8-cineole’s antioxidant activity via Nrf2/ C. albicans, and
Keap1 pathway activation. ARE: antioxidant response element; HO-1: heme C. glabrata,
oxygenase-1; NQO1: NAD(P)H: quinone oxidoreductase 1 (Figure created using • Inhibitory effect on
BioRender, Toronto, ON, Canada). dermatophytes using
1,8-cineole nano­
emulsion gel,
Escherichia coli, Pseudomonas aeruginosa, Salmonella enteritidis, Entero­ • Affects cell wall
bacter cloacae, Proteus mirabilis, and Salmonella typhimurium [73–79]. biogenesis,
mitochondrial activity,
The antimicrobial properties of 1,8-cineole can be mainly attributed to
and adhesion
its ability to cause cell membrane permeabilization [80,81]. Sun and Antiviral • Antiviral efficacy Development of [102–107]
colleagues demonstrated that treating Salmonella with 1,8-cineole against HSV-1, HSV-2, new antiviral
resulted in the downregulation of genes related to carbohydrate meta­ IBV, and influenza A agents targeting
bolism and membrane proteins at the mRNA level [82]. Similar results virus, viral replication
• Potential for SARS-CoV- and enhancing
have been reported for carbapenemase-producing Klebsiella pneumoniae 2 treatment by inhibit­ antiviral responses
isolates [83]. The antimicrobial activity of 1,8-cineole against bacteria ing Mpro enzyme crucial
possessing an outer LPS membrane is attributed to its hydrophobic na­ for viral replication,
ture [78]. • Stimulates antiviral
response by increasing
Although 1,8-cineole exhibited weaker bactericidal activity than
activity of IRF3 and
commonly used antibiotics such as gentamicin and amoxicillin (AMX)/ reducing
clavulanic acid, it significantly reduced the minimum inhibitory con­ proinflammatory
centration of antibiotics when used in combination with other drugs [84, activity of NF-kB,
85]. These findings align with a study that reported synergistic effects • Synergistic with
oseltamivir in treating
between 1,8-cineole and nisin against S. aureus [73]. Moreover, a
influenza A, enhancing
combination of AMX and 1,8-cineole has been noted to reduce the af­ immune response and
finity of the β-lactam antibiotic, AMX, for the β-lactamase enzyme as vaccine co-
well as to improve the bioavailability of AMX partly by enhancing in­ administration effects
testinal absorption and mainly by prolonging its half-time [86,87].
Accordingly, in vitro assays indicated that combining 1,8-cineole and
et al., who noted the inhibition of biofilm formation by S. pyogenes and
beta-lactam antibiotics, such as penicillin G and AMX, displayed a syn­
its mediated virulence factors [77]. Importantly, 1,8-cineole inhibited
ergistic effect against MRSA strains [88].
virulence production and biofilm formation in P. aeruginosa mediated by
At non-inhibitory doses, 1,8-cineole has been found to substantially
QS without detrimental effects on their growth [91]. In addition, a
inhibit the expression of Quorum sensing (QS), which plays a crucial role
decrease in biomass and cell viability of biofilms formed by MDR
in the formation and structure of bacterial biofilms [89], and virulence
ESBL-producing E. coli and K. pneumoniae isolates following treatment
genes in E. coli O101, particularly the luxS gene [90]. Furthermore,
with 1,8-cineole was documented [92,93]. Interestingly, S. aureus, the
non-inhibitory concentrations of 1,8-cineole and the absence of the luxS
most prevalent germ in chronic rhinosinusitis, demonstrated a reduction
gene led to a significant reduction in bacterial biofilm formation and
in biofilm formation [94]. Real-time quantitative polymerase chain re­
motility, indicating that 1,8-cineole can target both bacterial biofilm
action analysis verified that this phenomenon resulted from the
and motility. This finding agrees with a study conducted by Vijayakumar

5
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

downregulation of essential components in biofilm production (AgrA, provided cross-protection against the virus when co-administered with a
SarA, and σB) which is supported by 1,8-cineole. vaccination [114]. The combination increased the production of specific
Research findings by Li et al. indicated that 1,8-cineole can modify antibodies (IgG2a), the secretory response of IgA in nasal cavity mucosa,
the shape and size of bacterial cells, including Gram-negative and Gram- the expression of intraepithelial lymphocytes, the maturation of den­
positive bacteria [95]. The antimicrobial activity of 1,8-cineole might be dritic cells, and the expression of costimulatory proteins (CD40, CD80,
more pronounced against E. coli due to its susceptibility, compared to and CD86) in the peripheral blood [114].
S. aureus, which possesses a more robust cell wall and membrane. These Based on the behavior of similar monoterpenes, it can be inferred
observations are highly relevant in the clinical setting, given the ease that 1,8-cineole may act directly on viruses, whereas commonly used
with which virulence factors spread among Gram-negative bacteria and antivirals drugs like acyclovir target viral enzymes such as DNA poly­
the limited efficacy of conventional antibiotics in such cases. merase or RNA polymerase, as in the case of remdesivir.

3.3.2. Antifungal properties: inhibition of fungal growth and disruption of 3.4. The role of 1,8-cineole in bronchodilation, mucus reduction, and
biofilm formation airway clearance
Despite the availability of specific antifungal medications like
amphotericin B and voriconazole for treating fusariosis, their efficacy in Potential therapeutic applications of 1,8-cineole in respiratory dis­
clinical trials is limited, and they bear some serious toxicity risks eases are underpinned by its ability to act as a bronchodilator, enhance
[108–110]. 1,8-cineole has shown potential as an alternative antifungal mucociliary clearance, and reduce mucus production [115]. Based on a
agent with significant inhibitory effects on various fungi, including study using a rat model exposed to cigarette smoke, 1,8-cineole has been
Fusarium oxysporum, Fusarium sporotrichioides, and Aspergillus tubingensis shown to decrease mucus accumulation in the airways and alleviate lung
(Table 1) [97]. damage [116]. 1,8-cineole may also protect the lungs from reduced
1,8-cineole nanoemulsion gel demonstrated a significant inhibitory mucociliary clearance and bacterial infections associated with cigarette
effect on dermatophytes [101]. It is noteworthy that 1,8-cineole can smoking-induced cilia damage [117].
penetrate the cell membranes and damage organelles without altering A plethora of studies has revealed that 1,8-cineole exhibits myor­
the membrane potential [98]. elaxant effects on isolated tracheal smooth muscle from guinea pigs and
In vitro studies have demonstrated that 1,8-cineole can effectively Wistar rats, suggesting its bronchodilatory activity [118–120].
inhibit biofilm formation in the Fusarium solani species complex by Sagortchev et al. investigated the impact of 1,8-cineole on spontaneous
downregulating ergosterol biosynthetic genes, inhibiting adhesion, contractile behavior in guinea pig stomach smooth muscle tissue, sug­
interfering with mitochondrial activity, and hindering extracellular gesting that 1,8-cineole functions as a reversible competitive antagonist
matrix synthesis [99]. Furthermore, 1,8-cineole exhibited anti-biofilm at histamine receptors [121]. Therefore, 1,8-cineole is likely to possess
activity against mature and developing of Candida albicans and spasmolytic and secretolytic properties.
Candida glabrata biofilms, including their clinical isolates [100]. Treat­ An experimental rhinosinusitis model using ex vivo cultured human
ment with 1,8-cineole generated ROS and caused cell cycle arrest at the nasal slices co-incubated with LPS and 1,8-cineole showed a significant
G1/S phase in both species and altered the mitochondrial membrane reduction in mucin-filled goblet cells, supporting the ability of this
potential in C. glabrata. Transcriptional analysis revealed differential monoterpene to control mucus hypersecretion through cytokine inhi­
gene expression patterns of selected ABC transporters, secreted hydro­ bition [122]. A similar outcome was observed in OVA-challenged guinea
lytic enzymes, and cell wall biogenesis in C. albicans and C. glabrata pigs treated with 1,8-cineole [29].
when treated with 1,8-cineole [100]. Further evidence of 1,8-cineole’s potential benefits includes its
ability to decrease MUC5AC protein and mRNA expression in BALF and
3.3.3. Antiviral properties: inhibition of viral replication and enhancement lungs exposed to tobacco smoke [116]. Additionally, 1,8-cineole
of antiviral responses significantly reduced LPS-induced gene expression of MUC2 and
1,8-cineole has been reported to exhibit antiviral efficacy against a MUC19 as well as NF-κB activity [122]. This downregulation may be
range of viruses, including herpes simplex virus (HSV)− 1, HSV-2, in­ attributed to the inhibition of the NF-κB signaling pathway, which
fectious bronchitis virus (IBV), influenza A virus, and potentially, severe regulates MUC5AC, MUC2, and MUC19 expression [48,123].
acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (Table 1).
In vitro studies have shown that 1,8-cineole moderately inhibited 3.5. 1,8-cineole: a promising analgesic agent targeting TRP channels and
HSV-1 replication, with rates close to 40% [103]. An in vivo study P2X receptors
revealed that 1,8-cineole provided significant protection against genital
HSV-2 infection in a mouse model, with a 44% response rate [102]. A number of studies have provided evidence supporting the analgesic
Furthermore, 1,8-cineole demonstrated antiviral activity against IBV in properties of 1,8-cineole. Santos et al. conducted a study revealing the
vitro [111] and alleviated pathological viral pneumonia changes in mice independent analgesic effects of 1,8-cineole, distinct from opioids [124].
inoculated with influenza A virus (H1N1) by decreasing the levels of Notably, the analgesic effect of 1,8-cineole remained unaffected by
inflammatory cytokines (IL-4, IL-5, IL-10, and MCP-1) in nasal lavage naloxone, a mu-opioid receptor antagonist.
fluids and IL-1β, IL-6, TNF-α, and IFN-γ in lung tissues [104]. It also Furthermore, 1,8-cineole has been found to exhibit analgesic prop­
restored the expression of proinflammatory NF-kB p65, intercellular erties by activating cool temperature-sensing thermosensitive transient
adhesion molecule (ICAM)− 1, and vascular cell adhesion molecule receptor (TRP) cation channels (TRPM8) [125]. The authors also re­
(VCAM)− 1 in lung tissue after H1N1 virus infection. In silico molecular ported that 1,8-cineole inhibits a well-known sensor of painful cold
docking assays have suggested the potential efficacy of 1,8-cineole called the human transient receptor potential cation channel, subfamily
against SARS-CoV-2 Mpro, an enzyme crucial for viral replication [112, A, member 1 (TRPA1). Interestingly, using patch clamp techniques, it
113]. 1,8-cineole showed efficient binding to the active site of Mpro, was observed that 1,8-cineole could activate TRPA1 channels in the
forming hydrophobic interactions, and may thus represent a potential dorsal root ganglia (DRG), resulting in an increase in spontaneous
treatment for COVID-19 as an inhibitor of Mpro. excitatory postsynaptic currents in substantia gelatinosa neurons [126].
Noteworthy, 1,8-cineole potentiated an antiviral response in human Although the ability of 1,8-cineole to activate TRPM8 is lower compared
stem cells by increasing the activity of the antiviral transcription factor, to menthol, it effectively inhibited menthol-induced skin irritation
interferon regulatory factor 3, while reducing the NF-κB proin­ [125], indicating that its inhibitory effects are likely due to the inhibi­
flammatory activity [105]. In mice, 1,8-cineole exhibited synergistic tion of TRPA1 rather than the activation of TRPM8.
effects with oseltamivir in the treatment of influenza A [106] and Another study investigated the antinociceptive properties of 1,8-

6
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

cineole in acute and neuropathic orofacial pain using rodent models Another investigation revealed that 1,8-cineole influenced cell cycle
[127]. The results showed that pretreatment with 1,8-cineole signifi­ distribution differently based on concentration [15]. 1,8-cineole trig­
cantly reduced formalin-induced nociceptive behaviors in all mouse gered a robust G0/G1 arrest along with a considerable decrease of
strains. 1,8-cineole demonstrated antinociceptive effects and its effect S-phase cells at both concentrations (4.0–8.0 mM). At 8.0 mM, 1,
was sensitive to capsazepine, suggesting the involvement of transient 8-cineole increased the percentage of cells in the G2/M phase and
receptor potential vanilloid 1 (TRPV1) receptors. Molecular docking slightly decreased the number of G0/G1 cells compared to a lower
analysis supported the interaction between 1,8-cineole and TRPV1. concentration of 4.0 mM.
Wang et al. demonstrated that 1,8-cineole treatment reduced the
downregulation of P2X3 receptor expressions in the L4 and L5 DRG of 3.7. The effect of 1,8-cineole on platelet reactivity and blood parameters
rats with chronic constriction injury (CCI) [128]. The P2X2 and P2X3
receptors play a role in transmitting information related to pain and An investigation has shed light on platelet activation, thrombosis,
nociception through primary sensory neurons [129]. Additionally, and hemostasis, showcasing the potential for 1,8-cineole therapeutic
Zheng et al. showed that oral administration of 1,8-cineole inhibited the application [138]. 1,8-cineole demonstrated significant inhibitory ef­
overexpression of P2X2 receptor protein and mRNA in the spinal cord fects on platelet activation incited by glycoprotein VI stimulants such as
and dorsal horn of rats with CCI [14]. collagen and cross-linked collagen-related peptides. However, it
exhibited minimal restraint on thrombin or ADP-induced platelet ag­
3.6. Janus face of 1,8-cineole in apoptosis modulation gregation. It obstructed both inside-out and outside-in signaling related
to integrin αIIbβ3, along with granule secretion and the intracellular
1,8-cineole has demonstrated potential therapeutic applications calcium mobilization in platelets. In mice, it affected thrombus forma­
through both anti-apoptotic and pro-apoptotic effects. On the one hand, tion on collagen-coated surfaces under arterial flow conditions but had a
1,8-cineole has shown potential for inhibiting apoptosis and promoting minimal effect on hemostasis. Importantly, up to a concentration of
cell survival in various models. For instance, 1,8-cineole decreased the 50 µM, 1,8-cineole did not show toxic effect on platelets. Studies eluci­
protein expression of pro-apoptotic markers (Bax, Cyt-c, cleaved dating the molecular mechanisms underlying the inhibitory effects of 1,
caspase-3, and cleaved caspase-9) while increasing the levels of the anti- 8-cineole on platelet function have revealed its influence on signaling
apoptotic protein Bcl-2, resulting in the inhibition of retinal apoptosis in pathways mediated by key molecules such as AKT, Syk, LAT, and cAMP
diabetic eyes [130]. Additionally, 1,8-cineole reduced within platelets. This suggests that 1,8-cineole holds promise as a po­
isoprenaline-induced apoptosis, by decreasing the Bax/Bcl-2 ratio and tential therapeutic agent for modulating excessive platelet reactivity
the expression of cleaved caspase-3 in both H9c2 cardiomyocytes and observed in various pathophysiological conditions.
rat heart tissue [131]. Co-administration of 1,8-cineole and BPA led to a
significant reduction in apoptosis-positive cells, as well as a decrease in 4. Clinical applications of 1,8-cineole across different diseases
mRNA and protein levels of pro-apoptotic genes (Cyt-c, cleaved
caspase-3, Bax, and p53) stimulated by BPA, while promoting the 4.1. Respiratory disorders
expression of the anti-apoptotic gene Bcl-2 [58]. 1,8-cineole also alle­
viated BPA-induced necrotic injury in the bursa of Fabricius (BF) by Similar to other aromatic oils, 1,8-cineole is commonly used to treat
modulating RIPK1, RIPK3, FADD, MLKL, and caspase-8 expression [58]. respiratory tract infections due to its ability to increase the ciliary beat
The combined use of 1,8-cineole and BPA activated the PI3K/AKT frequency in the mucus membrane and its bronchodilating and anti-
signaling pathway, suggesting a potential mechanism for the inflammatory properties, as evidenced by preclinical studies. The effi­
anti-apoptotic and anti-necroptotic effects of 1,8-cineole in vitro. cacy of 1,8-cineole (Soledum™ capsules; CNL-1976) has been explored
On the other hand, recent studies have demonstrated the pro- in clinical trials for a range of respiratory disorders, including acute
apoptotic properties of 1,8-cineole in cancer cells. 1,8-cineole stimu­ rhinosinusitis, COPD, asthma, acute bronchitis, and the common cold. A
lated apoptosis in A2780 cancer cells, triggering a dose-dependent surge detailed overview of the relevant findings from these trials is compiled
in pre-G1 events [132]. It also inhibited A549 lung adenocarcinoma cell in Table 2.
migration and, when combined with simvastatin, increased G0/G1 cell
cycle arrest and sensitized cells to apoptosis [15]. 1,8-cineole induced 4.1.1. Acute rhinosinusitis
G0/G1 arrest and senescence in HepG2 cells through oxidative stress Acute rhinosinusitis arises from viral and bacterial infections, as well
and various signaling pathways such as MAPK, AMPK, and Akt/mTOR as the accumulation of thick, abnormal mucus and inflammatory sub­
[133]. In HCT116 and RKO human colon cancer cell lines, 1,8-cineole stances that impair the mucociliary transport system [149].
selectively promoted apoptosis rather than necrosis [134]. This pro­ In a prospective, randomized, double-blinded, placebo-controlled
cess was linked to survivin and Akt inactivation, along with p38 acti­ study with 152 patients with acute nonpurulent rhinosinusitis, re­
vation. These molecules triggered subsequent cleavage of PARP and searchers assessed 1,8-cineole efficacy and safety [139]. Participants
caspase-3, resulting in apoptosis. Similarly, 1,8-cineole induced received either 1,8-cineole capsules (3 ×200 mg/day) or placebo cap­
apoptosis and G2/M phase arrest in A431 cells by increasing p53 sules for 7 days. The findings indicated that the 1,8-cineole group
expression, as well as the expression of apoptotic proteins (Bax/Bcl-2, experienced a significant reduction in their symptoms-sum-score (SSS)
Cyt-c, caspase-9, and caspase-3) [135]. Molecular docking simulations and improvements in secondary endpoints, including headache, nasal
indicated hydrophobic interactions between 1,8-cineole, Bcl-2, and obstruction, and rhinological secretion.
PARP1 receptors, supporting its pro-apoptotic properties. Another prospective, randomized, double-blinded controlled study
The seemingly contradictory findings regarding the anti-apoptotic compared the efficacy and safety of 1,8-cineole (3 ×200 mg/day) and a
and pro-apoptotic effects of 1,8-cineole could stem from the varying combination of five components to treat acute viral rhinosinusitis [140].
dosages administered to the treated cells. The efficacious concentrations The study involved 150 patients, 75 in each treatment group, and used a
of 1,8-cineole reported for inhibiting in vitro cancer cell proliferation 7-day treatment period to assess the primary endpoint: improving a SSS
range from micromolar [135,136] to millimolar (mM) [133,134,137] including various rhinosinusitis symptoms. The results demonstrated
levels. This variable sensitivity can depend not only on the type of cancer that the 1,8-cineole group experienced a clinically relevant and statis­
cell but also on the individual cell line assessed. For instance, leukemia tically significant improvement in the SSS after 4 and 7 days compared
cells are among those most responsive to 1,8-cineole [136]. In a study to the control group. Thus, both medications are considered as safe for
where 1,8-cineole was used at millimolar concentrations in vitro, it treating acute viral rhinosinusitis. However, 1,8-cineole was found to be
exhibited potent antitumor activity in vivo at non-toxic dosages [134]. more effective than the alternative herbal preparation containing five

7
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

Table 2
Overview of clinical trials demonstrating beneficial effects of 1,8-cineole (CNL-1976) for the treatment of respiratory disorders. COPD: chronic obstructive pulmonary
disease; VC: vital capacity; Raw: airway resistance; sGaw: specific airways conductance; ITGV: intrathoracic gas volume; t.i.d.: three times a day; FEV1: forced
expiratory volume in the first second; BSS: bronchitis severity scale; N/A: not available; CC: common cold.
Study Study Design Population Intervention Control Main Findings (1,8-Cineole)

Acute nonpurulent Prospective, 152 patients with acute 3 × 200 mg 1,8-cineole Placebo t.i.d. Significant reduction in symptoms-
rhinosinusitis treatment randomized, double- rhinosinusitis per day for 7 days sum-scores and improvements in
with 1,8-cineole[139] blinded, placebo- secondary endpoints (headache,
controlled study nasal obstruction, and rhino-
secretion)
Comparison of 1,8-cineole Prospective, 150 patients with acute viral 3 × 200 mg 1,8-cineole Alternative herbal Significant reduction in symptoms-
and a combination of five randomized, double- rhinosinusitis per day for 7 days preparation with sum-scores and improvements in
different components for blinded controlled five different secondary endpoints (headache,
acute viral rhinosinusitis study components sensitivity of pressure points of
[140] trigeminal nerve, impairment of
general condition, nasal
obstruction, and rhino-secretion)
Comparison of oral therapy Randomized, double- 29 patients with COPD (4 3 × 200 mg 1,8-cineole 3 × 30 mg Significant improvements in lung
with 1,8-cineole and blind, double- excluded due to acute upper per day for 7 days Ambroxol per day function parameters (VC, Raw, and
Ambroxol in patients with dummy, cross-over airway infections, 1 due to for 7 days sGaw) and significant reduction in
COPD[141] trial allergic exanthem under ITGV.
Ambroxol therapy)
1,8-cineole in patients with Double-blind, 242 patients with stable COPD 3 × 200 mg 1,8-cineole Placebo t.i.d. Significant decrease in the
stable COPD as placebo-controlled, per day for six months frequency, duration, and severity
concomitant therapy multi-center study of COPD exacerbations; significant
[142] improvement of lung function,
dyspnea, and quality of life
Anti-inflammatory efficacy Double-blind, 32 patients with steroid- 3 × 200 mg 1,8-cineole Placebo t.i.d. Significant decrease of 36% in daily
of 1,8-cineole in patients placebo-controlled dependent bronchial asthma per day for 12 weeks prednisolone dosage; steroid-like
with severe asthma[7] trial anti-inflammatory properties
1,8-cineole as a concomitant Double-blind, 247 patients with confirmed 3 × 200 mg 1,8-cineole Placebo t.i.d. Significant improvement in lung
therapy for asthma placebo-controlled, asthma per day for six months function and significant reduction
patients[143] multi-center study in dyspnea as evidenced by primary
endpoints (FEV1, asthma
symptoms, and quality of life)
1,8-cineole as add-on to Open-label, 132 patients diagnosed with 3 × 200 mg 1,8-cineole Antiviral Significant reduction in BSS and
antiviral treatment for randomized, acute bronchitis or acute on top of antiviral treatment alone cough frequency after 4 days
acute bronchitis or acute parallel-group phase tracheobronchitis treatment (Ingavirin® (Ingavirin 90 mg
tracheobronchitis[144] III trial 90 mg once daily) for 4–9 once daily) for 4–9
days days
Effects of 1,8-cineole Double-blind, 242 acute bronchitis patients 3 × 200 mg 1,8-cineole Placebo t.i.d. Significant reduction in BSS and
treatment on acute placebo-controlled, per day for 10 days frequency of cough fits after 4 days
bronchitis patients[145] multi-center study
Effects of 1,8-cineole Multi-center, non- 336 children (Group A: 3 × 100 mg 1,8-cineole N/A Significant reduction in BSS;
treatment in children and interventional study n = 178; 2–7 years; Group B: per day for 7–9 days compliance rated as very good and
toddlers with acute n = 158; 8–11 years) with good in both groups; tolerability
bronchitis[146] acute bronchitis rated as very good and good
Effects of 1,8-cineole Non-interventional 893 patients aged 4–90 years 3 × 100 mg 1,8-cineole N/A Reduction in BSS and improvement
treatment in patients with study old, diagnosed with acute per day for children under in primary symptom cough within
acute bronchitis[147] bronchitis 10 years; 3 × 200 mg 1,8- 3–4 days; well-tolerated
cineole for individuals
aged 10 and above
Impact of 1,8-cineole Phase IV, 522 adults (18–70 years) with 3 × 200 mg 1,8-cineole N/A Earliest treatment initiation
treatment timing on CC exploratory, open- at least 1 CC during previous per day for up to 15 days resulted in lower burden of disease,
progress[148] label, non- winter, 329 subsequently faster decline in symptoms and
randomized, multi- developed a CC significantly higher and faster
center trial recovering quality of life

different components. observed. Whether preoperatively administered 1,8-cineole can reduce


inflammation and thus ease sinus surgery must be evaluated in future
4.1.2. Chronic rhinosinusitis studies.
Chronic rhinosinusitis (CRS) refers to a prolonged inflammatory Experimental observations demonstrated that 1,8-cineole effectively
condition of the sinus and nasal mucosa, characterized by symptoms impedes biofilm formation in CRS patients, establishing a correlation
persisting for more than 12 weeks [150]. It is estimated that approxi­ between the inhibition of growth in both planktonic and sessile forms of
mately a quarter to a third of CRS patients develop nasal polyps [151]. E. coli and M. catarrhalis [94]. Interestingly, the bacterium S. aureus
Generally, medical practitioners manage these symptoms using a treat­ exhibited elevated sensitivity to 1,8-cineole, with a level of suscepti­
ment plan that combines steroids, nasal washes, and antibiotics when bility that cannot be attributed solely to straightforward inhibition of
the presence of purulence is confirmed [152,153]. bacterial growth. Further exploration into the effects of 1,8-cineole on
In CRS, 1,8-cineole has not been established as a routine treatment, major components of the S. aureus biofilm formation pathways revealed
yet recent data revealed a highly sensitive detection of 1,8-cineole in a substantial inhibitory impact on the transcription of AgrA, SarA, and
nasal polyps from CRSwNP patients after 14 days of oral 1,8-cineole σB genes. This novel approach to treating bacterial biofilm infections
administration prior to surgical treatment [10]. No significant correla­ may have utility beyond CRS, extending to a variety of biofilm-related
tion between the measured 1,8-cineole concentrations and the body­ infectious diseases [154].
weight or body mass index values of the examined patients was

8
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

4.1.3. COPD reducing cough frequency.


COPD patients frequently experience exertional breathlessness due A multi-center, non-interventional study evaluated the compliance,
to bronchoconstriction, excessive mucus production, airway wall tolerability, and clinical progress of 336 children with acute bronchitis
edema, and loss of terminal airway attachments. Established pharma­ during 7–9 days of therapy with 3 × 100 mg/day of 1,8-cineole [146].
cotherapy options include bronchodilators and glucocorticosteroids In the course of the treatment period, the BSS decreased significantly
[155]. from an initial value of 8.61–5.09 at 4 days and to 1.38 at 7–9 days.
In a randomized, double-blind, double-dummy, cross-over trial Remarkably, 79% of patients reported being symptom-free at the end of
involving 29 COPD patients, researchers compared the effects of one- therapy, and 1,8-cineole therapy was rated as well-tolerated and
week oral therapy with 1,8-cineole (3 ×200 mg/day) and Ambroxol acceptable.
(3 ×30 mg/day) on lung function [141]. Both 1,8-cineole and Ambroxol In another non-interventional study, a cohort of 893 patients aged
demonstrated significant improvements in lung function parameters 4–90 years old, all diagnosed with acute bronchitis, underwent an 8-day
such as vital capacity, airway resistance, and specific airway conduc­ treatment course with 1,8-cineole [147]. Children under 10 years
tance from day 1 to day 7. Forced expiratory volume in the first second received a dosage of 3 × 100 mg/day of 1,8-cineole, while individuals
(FEV1) also improved, but without significance. Only 1,8-cineole aged 10 and above, including adults, were prescribed 3 × 200 mg/day
significantly reduced intrathoracic gas volume compared to Ambroxol. of 1,8-cineole. Significant improvements were observed, with the initial
Peak flow and the symptom score for dyspnea at rest improved under 1, BSS decreasing from 7.7 to 1.4 after treatment completion. The primary
8-cineole therapy. However, the difference failed to reach statistical cough symptom showed a 95.6% improvement rate, and treatment was
significance compared to Ambroxol due to the small number of patients. well tolerated. Notably, 81.4% of participants experienced distinct relief
Thus, these findings suggest that 1,8-cineole may have an additional from their symptoms within 3–4 days.
bronchodilator effect. Additionally, the data are in accordance with a Acute bronchitis is a transient respiratory disorder that typically
previous controlled investigation, in which Ambroxol and 1,8-cineole resolves within 14 days [158]. Achieving accelerated symptom
were shown to improve FEV1 and forced vital capacity in COPD pa­ improvement by day 3 is crucial for any intervention during a common
tients versus placebo [156]. cold. 1,8-cineole exhibited faster symptom relief, although the natural
Worth et al. conducted a placebo-controlled, double-blind trial with course of the condition suggests no significant divergence beyond the
242 moderate/severe COPD patients who were either smokers or former 14-day timeframe [145].
smokers already receiving standard therapy [142]. The study investi­
gated the effects of 1,8-cineole treatment (3 ×200 mg/day) for 6 months 4.2. Neurological disorders
on this population and found a significant decrease in the frequency,
duration, and severity of COPD exacerbations in the 1,8-cineole group Considering its lipophilic nature, 1,8-cineole has the ability to tra­
compared to the control. Although improvements in secondary outcome verse the blood-brain barrier [159]. This unique characteristic has
measures, including lung function, breathlessness, and quality of life caused growing enthusiasm to investigate the impact of 1,8-cineole on
were noted, these changes did not reach statistical significance. different neurological disorders (Fig. 3).

4.1.4. Asthma 4.2.1. Alzheimer’s disease


Asthma is characterized by an inflammation that leads to bronchial Alzheimer’s disease (AD) is a chronic neurodegenerative disorder
hyperresponsiveness and reversible airway obstruction. Current treat­ characterized by cognitive decline and memory loss. Currently, the U.S.
ment guidelines for managing asthma and reducing airway inflamma­ Food and Drug Administration has approved five medications for man­
tion recommend inhaled corticosteroids regularly [157]. aging AD symptoms, including cholinesterase inhibitors, an NMDA re­
A double-blind, placebo-controlled study evaluated the effects of ceptor antagonist, and an anti-amyloid monoclonal antibody [160]. AD
prednisolone reduction on 32 asthmatic patients who received either research primarily focuses on studying the formation, aggregation, and
3 × 200 mg/day of 1,8-cineole or a placebo for 12 weeks [7]. The results functional aspects of amyloid-beta (Aβ).
demonstrated a significant decrease of 36% in prednisolone medication The administration of 1,8-cineole in rat PC12 cells (pheochromocy­
usage within the treatment group. Noteworthy, the dyspnea score was toma cells) demonstrated effective mitigation of the Aβ induced cyto­
significantly higher in the placebo group. This trial provides compelling toxicity and oxidative stress [161]. Additionally, 1,8-cineole exhibited
evidence that 1,8-cineole exhibits anti-inflammatory properties com­ the ability to inhibit Aβ42 oligomerization in vitro and protect against
parable to those of steroids. iron-induced cell death in human neuroblastoma cells [162].
The same dosage of 1,8-cineole for 6 months was analyzed as In recent years, a mounting body of evidence points toward the
adjunctive therapy in a more recent multicenter, placebo-controlled, impact of advanced glycation end products (AGEs) on natural aging and
double-blind study involving 247 asthmatic patients already on medi­ neurodegenerative disorders such as AD [163]. Research suggests that
cation [143]. As indicated by primary endpoints such as FEV1, asthma AGEs can contribute to oxidative stress, tau hyperphosphorylation, and
symptoms, and quality of life, the treatment group showed a significant Aβ production [13]. Importantly, 1,8-cineole has shown the ability to
improvement in lung function compared to the placebo group. modulate tau phosphorylation by suppressing GSK-3β activity and to
reduce Aβ production by inhibiting beta-site amyloid precursor protein
4.1.5. Acute bronchitis cleaving enzyme-1 (BACE-1), both in vitro and in vivo [13].
Acute bronchitis is commonly associated with common cold, with its Considering that oxidative damage, Aβ aggregation, and a decline in
primary symptom being a dry or productive cough, which is frequently cell viability (iron-induced) are indicative of the onset of AD pathology,
caused by viral infections [158]. Interestingly, an open-label, random­ these findings strongly support the hypothesis of a potential therapeutic
ized, parallel-group phase III clinical trial showed that supplementing 1, role of 1,8-cineole in AD, particularly in cases associated with diabetes
8-cineole to antiviral treatment significantly decreased the frequency of mellitus (DM).
cough and other acute bronchitis symptoms compared to antiviral
medication alone [144]. 4.2.2. Anxiety and depression
In a placebo-controlled clinical trial with 242 acute bronchitis pa­ Anxiety and depression are widespread mental disorders that affect a
tients, researchers assessed the impact of administering 3 × 200 mg/day substantial number of adults globally. These conditions present
of 1,8-cineole for ten days on the bronchitis severity scale (BSS) [145]. numerous challenges, as the existing therapeutics used to treat these
The 1,8-cineole-treated group substantially decreased BSS compared to conditions are associated with various limitations and drawbacks,
the placebo group on day 4. 1,8-cineole was particularly effective at leading to low response rates [164].

9
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

Fig. 3. Neurological disorders mediated by 1,8-cineole (Figure created using BioRender, Toronto, ON, Canada).

A recent study aimed to explore the anxiolytic properties of 1,8- increased during the inter-ictal stage. 1,8-cineole’s effects were
cineole through two established behavioral models used to screen dose-dependent, with higher concentrations increasing relative wavelet
anxiolytic drugs in mice: the light-dark box test and the marble-burying energy in the delta frequency band. However, the mean fractal dimen­
test [165]. The study also investigated the potential involvement of the sion, which indicates system complexity, exhibited only marginal
GABAergic transmission system in 1,8-cineole’s anxiolytic effects by changes.
administering flumazenil (FLU), an antagonist to the GABAergic trans­ Similarly, another study exploring the effects of 1,8-cineole on the
mission system. As compared to the tests conducted with 1,8-cineole activity of central neurons in land snails reported excitatory and
alone, the combined treatment with FLU resulted in a significant epileptogenic properties [170]. The findings indicated that 1,8-cineole
decrease in the total number of square lines crossed and the number of depolarized the membrane potential, increased spontaneous neuronal
entries in the lit box. The results imply that FLU exhibits an antagonistic activity, and suppressed after-hyperpolarization. It also reduced the
impact on the anxiolytic effects of 1,8-cineole and are consistent with action potential amplitude and slope. The study revealed that 1,
prior studies that suggest the involvement of the GABAergic trans­ 8-cineole inhibited potassium channels involved in repolarization and
mission system in the mechanisms underlying the anxiolytic properties after-hyperpolarization. Additionally, 1,8-cineole increased the fre­
of 1,8-cineole [166]. Correspondingly, a study by Kim et al. identified quency of driven action potentials while inhibiting calcium-activated
the effectiveness of inhaled 1,8-cineole in reducing preoperative anxiety potassium channels. At higher concentrations, 1,8-cineole induced
in humans [167]. burst firing and paroxysmal depolarization shifts in neuronal activity.
Substances that decrease immobility time are commonly associated These effects were independent of sodium currents or protein kinase A
with antidepressant properties in humans. In the forced swimming test and C phosphorylation, in contrast to calcium currents, which played a
(FST), 1,8-cineole administration led to a significant reduction in the crucial role.
immobility time of mice compared to the control group [165]. Addi­
tionally, 1,8-cineole exhibited greater antidepressant effects than
4.3. Gastrointestinal disorders
fluoxetine. However, when 1,8-cineole was administered intraperito­
neally, no significant antidepressant activity was observed [166].
4.3.1. Peptic ulcer disease
Consistent with the FST results, 1,8-cineole significantly reduced
Peptic ulcer refers to damage caused by acid to the lining of the
immobility times in mice compared to the control group in the tail
digestive tract, extending into the submucosa. These ulcers are typically
suspension test.
found in the stomach or the upper part of the small intestine, known as
the proximal duodenum [171].
4.2.3. Epilepsy and acute seizure
Experiments conducted with Wistar rats revealed that 1,8-cineole
An estimated portion of 20–30% of epilepsy patients suffer from
effectively prevented gastric lesions induced by ethanol, ethanol/HCl,
seizures unresponsive to available antiepileptic drugs [168].
and indomethacin. This suggests that the antiulcer activity of 1,8-cineole
A study investigating 1,8-cineole’s effects on acute seizures was
may involve the production of prostaglandins and/or mucus [172].
conducted using rats [169]. Brain activity was analyzed using wavelet
Similar results have been reported in previous studies by Santos et al.
and fractal analysis before and after 1,8-cineole administration. The
that 1,8-cineole significantly attenuates ethanol-induced gastric injury
analysis revealed that the delta frequency band dominated brain activ­
in a manner similar to a known lipoxygenase inhibitor known as nor­
ity. Other frequency bands decreased during the ictal stage and
dihydroguaiaretic acid [173]. 1,8-cineole reduced basal acid secretion

10
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

volume without affecting stimulated acid secretion, implying a potential concentrations of atrial natriuretic peptide and brain natriuretic peptide
lack of antisecretory activity [172]. Additionally, 1,8-cineole increased in rat hearts [131].
mucus levels, protected against –SH groups depletion, reduced LP, and Of note, 1,8-cineole treatment restored gap junction protein Cx43
inhibited MPO activity, thus preventing neutrophil infiltration in the levels and distribution back to the intercalated disks [182]. This resto­
gastric mucosa [172]. In a chronic ulcer model, 1,8-cineole decreased ration may improve anisotropic conduction, potentially enhancing RV
gastric area lesions, promoted regeneration and restoration of mucus function. Additionally, 1,8-cineole restored SERCA2a and ryanodine
levels in glandular cells, and stimulated cell proliferation, as indicated receptor 2, proteins involved in calcium regulation, which have been
by reactivity to PCNA, Ki-67, and BrdU [172]. linked to RV hypertrophy in PAH [183]. 1,8-cineole was also found to
inhibit the activation of dynamin-related protein 1 and promote mito­
4.3.2. Diarrhea chondrial fusion by increasing MFN2. Furthermore, 1,8-cineole partially
Even though mortality rates have improved, diarrhea remains a restored mitochondrial transcription factor A levels, suggesting a po­
major cause of death in children under 5, accounting for approximately tential role in mitochondrial biogenesis. Although 1,8-cineole decreased
15% of all death cases [174]. This translates into nearly 1.4 million oxygen consumption rates, it increased the expression of most assessed
deaths in children annually in developing countries. Prolonged diarrhea electron transport chain subunits, except for NADH dehydrogenase
episodes can also lead to malnutrition, rendering it a crucial factor in (ubiquinone) 1 beta subcomplex subunit 8.
developing nutritional deficiencies.
In a study conducted by Jalilzadeh-Amin et al., 1,8-cineole was 4.5. Diabetes mellitus
shown to inhibit various parameters associated with diarrhea, including
the condition, such as the frequency and severity of diarrhea, the total DM encompasses a range of metabolic disorders characterized by
number of stools, and the weight of wet stools, although these effects did elevated blood glucose levels resulting from insulin resistance, inade­
not reach statistical significance [175]. Interestingly, the inhibitory ef­ quate insulin production, or excessive glucagon secretion [184].
fects of 1,8-cineole were lower than those of loperamide and atropine, Emerging research has elucidated potential advantages of 1,8-cineole in
commonly used as standard antidiarrheal drugs. Studies on enter­ the management of renal dysfunction, glycemic control, and dyslipide­
opooling, which measures intestinal contents volume, showed that 1, mia in diabetic animal models.
8-cineole pretreatment reduced intraluminal contents [175]. These ef­ 1,8-cineole has demonstrated its capacity to attenuate markers of
fects, resulting from reduced secretion into the intestine, suggest that 1, renal dysfunction in diabetic rats and ameliorate renal impairment in
8-cineole may enhance electrolyte reabsorption, demonstrating the in­ both rat models of diabetic nephropathy and db/db mice by modulating
hibition of excessive secretion or increased absorption of other intestinal transforming growth factor beta-1 (TGF-β1) [185,186]. Additionally, 1,
contents. Additionally, 1,8-cineole decreased normal intestinal propul­ 8-cineole showed encouraging potential in hindering podocyte barrier
sive movement and transit in rats. These findings are consistent with dysfunction and consequent proteinuria in db/db mice [187]. In studies
observations by Magalhães et al., who noted delayed gastric emptying conducted on diabetic rats, 1,8-cineole has shown promising potential in
and affected liquid transit after intravenous administration of 1, ameliorating hyperglycemia and improving insulin sensitivity [185].
8-cineole in rats [176]. The delay in intestinal motility caused by 1, These effects are likely due to the compound’s ability to enhance the
8-cineole may contribute to increased water absorption from feces, functionality of pancreatic β-cells. Recent reports have underscored the
reducing watery feces consistency. capability of 1,8-cineole to diminish blood glucose levels in db/db mice
Experimental data suggest that 1,8-cineole exerts its inhibitory effect by activating glyoxalase 1 and mitigating various glycated products
on gastrointestinal hypermotility primarily through its anticholinergic [185,186]. Moreover, 1,8-cineole has exhibited lipid-lowering effects,
properties [175]. This implies that 1,8-cineole acts by blocking or substantiated by investigations employing zebrafish atherosclerosis
reducing the activity of cholinergic receptors in the gastrointestinal models [188]. Collectively, these findings point to the promise to apply
tract, resulting in a reduction in smooth muscle contractions and 1,8-cineole as a therapeutic intervention for addressing the multifarious
motility. facets of diabetic complications.

4.4. Cardiac hypertrophy-related heart disorders 5. Future directions and concluding remarks

Pulmonary Arterial Hypertension (PAH) is a rare and life-threatening 1,8-cineole has been recognized for its health benefits, which have
pulmonary vascular disorder, affecting approximately 15–50 cases per been appreciated since ancient times and continue to be relevant today.
million people [177]. Cardiac hypertrophy is characterized by car­ 1,8-cineole exhibits a wide range of biological effects, including anti-
diomyocyte enlargement, increased protein synthesis, and sarcomere inflammatory, antioxidant, antibacterial, antiviral, mucolytic/secreto­
reorganization [178]. This process can become maladaptive due to lytic, bronchodilatory, analgesic, pro-apoptotic, and numerous other
metabolic dysfunction, increased stiffness, dilatation, and fibrosis, ulti­ activities crucial for health-promoting properties. Clinical trials on pa­
mately leading to heart failure [179]. tients with COPD, asthma, bronchitis, and rhinosinusitis support these
Recent studies have demonstrated the hypotensive effects of 1,8- positive effects. By modulating key signaling pathways such as NF-κB,
cineole in normotensive rats and its ability to cause vasodilation in MAPK, PPAR-γ, and Nrf2/Keap1, 1,8-cineole demonstrates potent anti-
isolated aortic rings constricted with phenylephrine [180,181]. inflammatory and antioxidative properties. Additionally, it targets
Alves-Silva et al. developed an in vitro model to mimic the afterload important effector molecules entities like the MUC gene family, caspases
experienced by the right ventricle (RV) during PAH progression [182]. (specifically caspase-3, − 8 and − 9), p53, ROS, p38, P2X receptors, and
They investigated the potential of 1,8-cineole to counteract cell hyper­ TRP channels, further enhancing its therapeutic potential.
trophy in H9c2 cells and neonatal rat ventricular cardiomyocytes The potential of 1,8-cineole as an effective anticancer agent has been
exposed to high pressure. The results showed that 1,8-cineole effectively substantiated by its effects on tumor angiogenesis, invasion, and
reversed cell hypertrophy by reducing cell surface area, sarcomere metastasis [189]. These effects can be attributed to the significant
thickness, and hypertrophic gene expression. Moreover, 1,8-cineole downregulation of crucial factors including TGF-β1, Fascin-1, vascular
reversed RV hypertrophy and fibrosis, improving cardiac function as endothelial growth factors, matrix metalloproteinase-9, and the
observed through microscopy and echocardiography analysis. These epithelial-mesenchymal transition (EMT) marker vimentin. Notably,
findings are consistent with a study by Wang et al., in which 1,8-cineole EMT regulation relies on the Wnt/β-catenin pathway, which 1,8-cineole
enhanced cell viability, inhibited cardiac hypertrophy, attenuated car­ has been shown to inhibit, underscoring its potential to impede the
diac remodeling, improved cardiac function, and decreased the acquisition of invasive and metastatic properties by tumors [65,190].

11
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

While limited research has explored the antiparasitic effects of 1, their work.
8-cineole, a recent study has demonstrated promising results, high­
lighting its potential as an antimalarial agent capable of impacting the References
parasite erythrocytic cycle and mitigating severe disease [191]. How­
ever, it is important to note that these findings exceed the scope of this [1] G.M. Cragg, D.J. Newman, Natural products: a continuing source of novel drug
leads. Biochim Biophys. Acta 1830 (6) (2013) 3670–3695, https://doi.org/
review article and warrant further investigation in future research. 10.1016/j.bbagen.2013.02.008.
Straddling the rich history of traditional medicine and the ad­ [2] G.H. Seol, K.Y. Kim, Eucalyptol and its role in chronic diseases, Adv. Exp. Med.
vancements of contemporary science, 1,8-cineole stands poised to Biol. 929 (2016) 389–398, https://doi.org/10.1007/978-3-319-41342-6_18.
[3] A.E. Sadlon, D.W. Lamson, Immune-modifying and antimicrobial effects of
address modern challenges. Its versatility is evident in its ability to Eucalyptus oil and simple inhalation devices. Alter. Med Rev. 15 (1) (2010)
combat conditions like multi-drug resistance, COVID-19, cancer, dia­ 33–47.
betes, Alzheimer’s disease, hypertension, and heart failure. In a world [4] S. Aparicio, et al., Properties of 1,8-cineole: a thermophysical and theoretical
study. J. Phys. Chem. B 111 (12) (2007) 3167–3177, https://doi.org/10.1021/
grappling with complex diseases, 1,8-cineole does not simply present as jp067405b.
an alternative, but as an innovative solution, hinting at the dawn of more [5] X. Poitou, C. Thibon, P. Darriet, 1,8-cineole in french red wines: evidence for a
tailored and effective treatments. contribution related to its various origins. J. Agric. Food Chem. 65 (2) (2017)
383–393, https://doi.org/10.1021/acs.jafc.6b03042.
Despite the promising applications of 1,8-cineole, the clinical
[6] N. Chandorkar, et al., A systematic and comprehensive review on current
translation is still in its early stages and faces several complications and understanding of the pharmacological actions, molecular mechanisms, and
limitations. While considered as safe and well-tolerated at normal clinical implications of the genus Eucalyptus. Phytomedicine 1 (4) (2021),
therapeutic doses, excessive use and overdosage of 1,8-cineole through 100089 https://doi.org/10.1016/j.phyplu.2021.100089.
[7] U.R. Juergens, et al., Anti-inflammatory activity of 1.8-cineol (eucalyptol) in
oral ingestion, inhalation, or skin application can be hazardous, lead to bronchial asthma: a double-blind placebo-controlled trial. Respir. Med 97 (3)
potential acute toxicity, including inflammatory gastrointestinal condi­ (2003) 250–256, https://doi.org/10.1053/rmed.2003.1432.
tions and rare symptoms like nausea, vomiting, and diarrhea [18,107]. [8] M. Duisken, et al., Metabolism of 1,8-cineole by human cytochrome P450
enzymes: identification of a new hydroxylated metabolite. Biochim Biophys. Acta
To fully harness the previous research knowledge, an integrated 1722 (3) (2005) 304–311, https://doi.org/10.1016/j.bbagen.2004.12.019.
approach is necessary. This approach should aim to understand the exact [9] J. Beauchamp, F. Kirsch, A. Buettner, Real-time breath gas analysis for
mechanisms underlying the pharmacological activities and safety as­ pharmacokinetics: monitoring exhaled breath by on-line proton-transfer-reaction
mass spectrometry after ingestion of eucalyptol-containing capsules, J. Breath.
pects of 1,8-cineole, develop safe and effective formulations, enhance Res 4 (2) (2010), 026006, https://doi.org/10.1088/1752-7155/4/2/026006.
clinical understanding, and consider economic and production aspects. [10] C. MacKenzie, et al., Determination of orally administered 1,8-Cineol in nasal
Additionally, exploring combination therapies to enhance the 1, polyp tissues from chronic rhinosinusitis patients using gas chromatography:
mass spectrometry. Sci. Rep. 13 (1) (2023) 3605, https://doi.org/10.1038/
8-cineole activity in conjunction with other available agents holds s41598-023-29941-x.
promise for improved clinical outcomes. [11] A.K. Dhakad, et al., Biological, medicinal and toxicological significance of
In summary, this review article has not only provided a compre­ Eucalyptus leaf essential oil: a review. J. Sci. Food Agric. 98 (3) (2018) 833–848,
https://doi.org/10.1002/jsfa.8600.
hensive understanding of the properties and therapeutic potential of 1,8-
[12] Z.M. Cai, et al., 1,8-Cineole: a review of source, biological activities, and
cineole in the realm of health sciences but has also shed light on the application. J. Asian Nat. Prod. Res 23 (10) (2021) 938–954, https://doi.org/
potential benefits it might offer to patients dealing with a variety of 10.1080/10286020.2020.1839432.
diseases. This valuable contribution paves the way for further explora­ [13] F. An, et al., 1,8-Cineole Ameliorates Advanced Glycation End Products-Induced
Alzheimer’s Disease-like Pathology In Vitro and In Vivo. Molecules 27 (12)
tion and utilization of 1,8-cineole as an approach to diverse medical (2022) https://doi.org/10.3390/molecules27123913.
conditions. [14] X.B. Zheng, et al., Effects of 1,8-cineole on neuropathic pain mediated by P2X2
receptor in the spinal cord dorsal horn. Sci. Rep. 9 (1) (2019) 7909, https://doi.
org/10.1038/s41598-019-44282-4.
Funding [15] B. Rodenak-Kladniew, et al., Anti-cancer mechanisms of linalool and 1,8-cineole
in non-small cell lung cancer A549 cells. Heliyon 6 (12) (2020), e05639 https://
This research did not receive any specific grant from funding doi.org/10.1016/j.heliyon.2020.e05639.
[16] S. Griffin, S.G. Wyllie, J. Markham, Determination of octanol–water partition
agencies in the public, commercial, or not-for-profit sectors. coefficient for terpenoids using reversed-phase high-performance liquid
chromatography. J. Chromatogr. A 864 (2) (1999) 221–228, https://doi.org/
CRediT authorship contribution statement 10.1016/S0021-9673(99)01009-2.
[17] D. Prasanthi, P.K. Lakshmi, Terpenes: Effect of lipophilicity in enhancing
transdermal delivery of alfuzosin hydrochloride. J. Adv. Pharm. Technol. Res 3
Cosima C. Hoch: Conceptualization, Investigation, Writing – orig­ (4) (2012) 216–223, https://doi.org/10.4103/2231-4040.104712.
inal draft preparation, Visualization. Julie Petry: Writing – review & [18] M. Bhowal, M. Gopal, Eucalyptol: safety and pharmacological profile, RGUHS J.
Pharm. Sci. (2015) 5.
editing, Visualization. Lena Griesbaum: Writing – review & editing,
[19] M. Miyazawa, et al., Hydroxycineole: four new metabolites of 1,8-cineole in
Visualization. Tobias Weiser: Writing – review & editing, Investigation. rabbits. J. Agric. Food Chem. 37 (1) (1989) 222–226, https://doi.org/10.1021/
Kathrin Werner: Writing – review & editing. Michael Ploch: Writing – jf00085a051.
[20] R. Boyle, S. McLean, N.W. Davies, Biotransformation of 1,8-cineole in the
review & editing. Admar Verschoor: Writing – review & editing.
brushtail possum (Trichosurus vulpecula, Xenobiotica 30 (9) (2000) 915–932,
Gabriele Multhoff: Investigation, Writing – review & editing. Ali https://doi.org/10.1080/004982500433336.
Bashiri Dezfouli: Investigation, Writing – review & editing. Barbara [21] K. Horst, M. Rychlik, Quantification of 1,8-cineole and of its metabolites in
Wollenberg: Conceptualization, Writing – review & editing, humans using stable isotope dilution assays. Mol. Nutr. Food Res. 54 (10) (2010)
1515–1529, https://doi.org/10.1002/mnfr.200900528.
Supervision. [22] M. Miyazawa, M. Shindo, T. Shimada, Oxidation of 1,8-cineole, the monoterpene
cyclic ether originated from eucalyptus polybractea, by cytochrome P450 3A
Declaration of Competing Interest enzymes in rat and human liver microsomes. Drug Metab. Dispos. 29 (2) (2001)
200–205.
[23] P.M. Jenner, et al., Food flavourings and compounds of related structure I. Acute
Michael Ploch is employed by Cassella-med GmbH & Co. KG, located oral toxicity. Food Cosmet. Toxicol. 2 (1964) 327–343, https://doi.org/10.1016/
at Gereonsmühlengasse 1, 50670 Cologne, Germany. He provided S0015-6264(64)80192-9.
[24] M. De Vincenzi, et al., Constituents of aromatic plants: eucalyptol. Fitoterapia 73
guidance during the writing process, but was not involved in the design, (3) (2002) 269–275, https://doi.org/10.1016/s0367-326x(02)00062-x.
exploration, or graphical representation of this review article. As such, [25] C.A. Dinarello, Proinflammatory cytokines. Chest 118 (2) (2000) 503–508,
any potential conflict of interest does not affect the objectivity of this https://doi.org/10.1378/chest.118.2.503.
[26] B. Venkataraman, et al., Molecular Docking Identifies 1,8-Cineole (Eucalyptol) as
review article. The other authors—Cosima C. Hoch, Julie Petry, Lena A Novel PPARγ Agonist That Alleviates Colon Inflammation. Int J. Mol. Sci. 24 (7)
Griesbaum, Tobias Weiser, Kathrin Werner, Admar Verschoor, Gabriele (2023) https://doi.org/10.3390/ijms24076160.
Multhoff, Ali Bashiri Dezfouli, and Barbara Wollenberg—assert that this [27] P.R. Lima, et al., 1,8-cineole (eucalyptol) ameliorates cerulein-induced acute
pancreatitis via modulation of cytokines, oxidative stress and NF-κB activity in
research was carried out without any conflicts of interest influencing

12
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

mice. Life Sci. 92 (24–26) (2013) 1195–1201, https://doi.org/10.1016/j. [53] S. Reuter, et al., Oxidative stress, inflammation, and cancer: how are they linked?
lfs.2013.05.009. Free Radic. Biol. Med 49 (11) (2010) 1603–1616, https://doi.org/10.1016/j.
[28] F.A. Santos, et al., 1,8-cineole (eucalyptol), a monoterpene oxide attenuates the freeradbiomed.2010.09.006.
colonic damage in rats on acute TNBS-colitis. Food Chem. Toxicol. 42 (4) (2004) [54] S. Ryu, et al., 1,8-Cineole ameliorates oxygen-glucose deprivation/reoxygenation-
579–584, https://doi.org/10.1016/j.fct.2003.11.001. induced ischaemic injury by reducing oxidative stress in rat cortical neuron/glia.
[29] V.P. Bastos, et al., Inhaled 1,8-cineole reduces inflammatory parameters in J. Pharm. Pharm. 66 (12) (2014) 1818–1826, https://doi.org/10.1111/
airways of ovalbumin-challenged Guinea pigs. Basic Clin. Pharm. Toxicol. 108 (1) jphp.12295.
(2011) 34–39, https://doi.org/10.1111/j.1742-7843.2010.00622.x. [55] A. Rašković, et al., Antioxidant activity of rosemary (Rosmarinus officinalis L.)
[30] U.R. Juergens, et al., Antiinflammatory effects of euclyptol (1.8-cineole) in essential oil and its hepatoprotective potential. BMC Complement Alter. Med 14
bronchial asthma: inhibition of arachidonic acid metabolism in human blood (2014) 225, https://doi.org/10.1186/1472-6882-14-225.
monocytes ex vivo. Eur. J. Med Res 3 (9) (1998) 407–412. [56] Y. Cui, et al., DEHP-induce damage in grass carp hepatocytes and the remedy of
[31] F. Jiang, et al., Preparation and protective effects of 1,8-cineole-loaded self- Eucalyptol. Ecotoxicol. Environ. Saf. 206 (2020), 111151 https://doi.org/
microemulsifying drug delivery system on lipopolysaccharide-induced 10.1016/j.ecoenv.2020.111151.
endothelial injury in mice. Eur. J. Pharm. Sci. 127 (2019) 14–23, https://doi.org/ [57] C. Yin, et al., Eucalyptol alleviates inflammation and pain responses in a mouse
10.1016/j.ejps.2018.10.012. model of gout arthritis. Br. J. Pharm. 177 (9) (2020) 2042–2057, https://doi.org/
[32] Z. Kutlu, et al., Biochemical research of the effects of essential oil obtained from 10.1111/bph.14967.
the fruit of Myrtus communis l. on cell damage associated with [58] L. Liu, et al., 1,8-cineole alleviates bisphenol A-induced apoptosis and necroptosis
lipopolysaccharide-induced endotoxemia in a human umbilical cord vein in bursa of Fabricius in chicken through regulating oxidative stress and PI3K/AKT
endothelial cells. Biochem Genet 59 (1) (2021) 315–334, https://doi.org/ pathway. Ecotoxicol. Environ. Saf. 226 (2021), 112877 https://doi.org/10.1016/
10.1007/s10528-020-10005-y. j.ecoenv.2021.112877.
[33] H.S. Lee, et al., Effect of 1.8-cineole in dermatophagoides pteronyssinus- [59] D.S. Antonella, et al., Antimutagenic and antioxidant activities of some
stimulated bronchial epithelial cells and mouse model of asthma., in: Biol Pharm bioflavours from wine. Food Chem. Toxicol. 60 (2013) 141–146, https://doi.org/
Bull, 39, 2016, pp. 946–952, https://doi.org/10.1248/bpb.b15-00876. 10.1016/j.fct.2013.07.042.
[34] C. Zhao, et al., 1,8-cineol attenuates LPS-induced acute pulmonary inflammation [60] M. Rodrigues, et al., Thymus mastichina: composition and biological properties
in mice. Inflammation 37 (2) (2014) 566–572, https://doi.org/10.1007/s10753- with a focus on antimicrobial activity. Pharm. (Basel) 13 (12) (2020) https://doi.
013-9770-4. org/10.3390/ph13120479.
[35] U.R. Juergens, M. Stöber, H. Vetter, Inhibition of cytokine production and [61] H.K. Moon, et al., Effects of 1,8-cineole on hypertension induced by chronic
arachidonic acid metabolism by eucalyptol (1.8-cineole) in human blood exposure to nicotine in rats. J. Pharm. Pharm. 66 (5) (2014) 688–693, https://
monocytes in vitro. Eur. J. Med Res 3 (11) (1998) 508–510. doi.org/10.1111/jphp.12195.
[36] N. Yadav, H. Chandra, Suppression of inflammatory and infection responses in [62] A. Ben Hsouna, et al., Chemical composition and hepatoprotective effect of
lung macrophages by eucalyptus oil and its constituent 1,8-cineole: Role of essential oil from Myrtus communis L. flowers against CCL(4)-induced acute
pattern recognition receptors TREM-1 and NLRP3, the MAP kinase regulator hepatotoxicity in rats. RSC Adv. 9 (7) (2019) 3777–3787, https://doi.org/
MKP-1, and NFκB. PLoS One 12 (11) (2017), e0188232 https://doi.org/10.1371/ 10.1039/c8ra08204a.
journal.pone.0188232. [63] E. Hodaj-Çeliku, et al., Antioxidant Activity and Chemical Composition of
[37] U.R. Juergens, et al., Inhibitory activity of 1,8-cineol (eucalyptol) on cytokine Essential Oils of some Aromatic and Medicinal Plants from Albania. Nat. Prod.
production in cultured human lymphocytes and monocytes. Pulm. Pharm. Ther. Commun. 12 (5) (2017) 785–790.
17 (5) (2004) 281–287, https://doi.org/10.1016/j.pupt.2004.06.002. [64] F. He, X. Ru, T. Wen, NRF2, a transcription factor for stress response and beyond.
[38] M. Koennecke, et al., Increased phosphorylation of eNOS in nasal polyps of Int J. Mol. Sci. 21 (13) (2020) https://doi.org/10.3390/ijms21134777.
chronic rhinosinusitis patients can be diminished by 1,8-cineol. Nitric Oxide 78 [65] L. Chen, et al., Cineole regulates Wnt/β-catenin pathway through Nrf2/keap1/
(2018) 89–94, https://doi.org/10.1016/j.niox.2018.06.002. ROS to inhibit bisphenol A-induced apoptosis, autophagy inhibition and
[39] T.A. Wynn, K.M. Vannella, Macrophages in Tissue Repair, Regeneration, and immunosuppression of grass carp hepatocytes. Fish. Shellfish Immunol. 131
Fibrosis. Immunity 44 (3) (2016) 450–462, https://doi.org/10.1016/j. (2022) 30–41, https://doi.org/10.1016/j.fsi.2022.09.067.
immuni.2016.02.015. [66] Z. Jiang, et al., The Essential Oils and Eucalyptol From Artemisia vulgaris L.
[40] Y. Rui, et al., Eucalyptol prevents bleomycin-induced pulmonary fibrosis and M2 Prevent Acetaminophen-Induced Liver Injury by Activating Nrf2-Keap1 and
macrophage polarization. Eur. J. Pharm. 931 (2022), 175184 https://doi.org/ Enhancing APAP Clearance Through Non-Toxic Metabolic Pathway. Front Pharm.
10.1016/j.ejphar.2022.175184. 10 (2019) 782, https://doi.org/10.3389/fphar.2019.00782.
[41] K.Y. Kim, H.S. Lee, G.H. Seol, Eucalyptol suppresses matrix metalloproteinase-9 [67] M. Porres-Martínez, et al., In vitro neuroprotective potential of the monoterpenes
expression through an extracellular signal-regulated kinase-dependent nuclear α-pinene and 1,8-cineole against H2O2-induced oxidative stress in PC12 cells.
factor-kappa B pathway to exert anti-inflammatory effects in an acute lung Z. Nat. C. J. Biosci. 71 (7–8) (2016) 191–199, https://doi.org/10.1515/znc-2014-
inflammation model. J. Pharm. Pharm. 67 (8) (2015) 1066–1074, https://doi. 4135.
org/10.1111/jphp.12407. [68] Y. Di, et al., Effects of dietary 1,8-cineole supplementation on growth
[42] U. Rantzsch, et al., Anti-inflammatory effects of Myrtol standardized and other performance, antioxidant capacity, immunity, and intestine health of broilers.
essential oils on alveolar macrophages from patients with chronic obstructive Anim. (Basel) (18) (2022) 12, https://doi.org/10.3390/ani12182415.
pulmonary disease., Suppl 4, Eur. J. Med Res 14 (Suppl 4) (2009) 205–209, [69] A. Taheri Mirghaed, S.M. Hoseini, M. Ghelichpour, Effects of dietary 1,8-cineole
https://doi.org/10.1186/2047-783x-14-s4-205. supplementation on physiological, immunological and antioxidant responses to
[43] Z. Ikawati, S. Wahyuono, K. Maeyama, Screening of several Indonesian medicinal crowding stress in rainbow trout (Oncorhynchus mykiss, Fish. Shellfish Immunol.
plants for their inhibitory effect on histamine release from RBL-2H3 cells. 81 (2018) 182–188, https://doi.org/10.1016/j.fsi.2018.07.027.
J. Ethnopharmacol. 75 (2–3) (2001) 249–256, https://doi.org/10.1016/s0378- [70] O. Ciftci, et al., Antioxidative effects of curcumin, β-myrcene and 1,8-cineole
8741(01)00201-x. against 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced oxidative stress in rats liver.
[44] T. Nakamura, et al., Eucalyptus oil reduces allergic reactions and suppresses mast Toxicol. Ind. Health 27 (5) (2011) 447–453, https://doi.org/10.1177/
cell degranulation by downregulating IgE-FcεRI signalling. Sci. Rep. 10 (1) 0748233710388452.
(2020) 20940, https://doi.org/10.1038/s41598-020-77039-5. [71] F. Guzzo, et al., Plant derived natural products against pseudomonas aeruginosa
[45] M. Hernandez-Quiles, M.F. Broekema, E. Kalkhoven, PPARgamma in Metabolism, and staphylococcus aureus: antibiofilm activity and molecular mechanisms.
Immunity, and Cancer: Unified and Diverse Mechanisms of Action. Front Molecules 25 (21) (2020) https://doi.org/10.3390/molecules25215024.
Endocrinol. (Lausanne) 12 (2021), 624112 https://doi.org/10.3389/ [72] H. Koo, et al., Targeting microbial biofilms: current and prospective therapeutic
fendo.2021.624112. strategies. Nat. Rev. Microbiol 15 (12) (2017) 740–755, https://doi.org/10.1038/
[46] K.G. Linghu, et al., 1,8-Cineole Ameliorates LPS-Induced Vascular Endothelium nrmicro.2017.99.
Dysfunction in Mice via PPAR-γ Dependent Regulation of NF-κB. Front Pharm. 10 [73] A. Farhanghi, et al., Antibacterial interactions of pulegone and 1,8-cineole with
(2019) 178, https://doi.org/10.3389/fphar.2019.00178. monolaurin ornisin against Staphylococcus aureus. Food Sci. Nutr. 10 (8) (2022)
[47] P.P. Tak, G.S. Firestein, NF-kappaB: a key role in inflammatory diseases. J. Clin. 2659–2666, https://doi.org/10.1002/fsn3.2870.
Invest 107 (1) (2001) 7–11, https://doi.org/10.1172/jci11830. [74] D. Kifer, V. Mužinić, M. Klarić, Antimicrobial potency of single and combined
[48] J.F. Greiner, et al., 1,8-Cineol inhibits nuclear translocation of NF-κB p65 and NF- mupirocin and monoterpenes, thymol, menthol and 1,8-cineole against
κB-dependent transcriptional activity. Biochim Biophys. Acta 1833 (12) (2013) Staphylococcus aureus planktonic and biofilm growth. J. Antibiot. (Tokyo) 69 (9)
2866–2878, https://doi.org/10.1016/j.bbamcr.2013.07.001. (2016) 689–696, https://doi.org/10.1038/ja.2016.10.
[49] E. Kennedy-Feitosa, et al., Eucalyptol attenuates cigarette smoke-induced acute [75] M. Soković, et al., Antibacterial effects of the essential oils of commonly
lung inflammation and oxidative stress in the mouse. Pulm. Pharm. Ther. 41 consumed medicinal herbs using an in vitro model. Molecules 15 (11) (2010)
(2016) 11–18, https://doi.org/10.1016/j.pupt.2016.09.004. 7532–7546, https://doi.org/10.3390/molecules15117532.
[50] K. Linghu, et al., Ameliorating effects of 1,8-cineole on LPS-induced human [76] Z. Jiang, et al., Protective Effects of 1,8-Cineole Microcapsules Against
umbilical vein endothelial cell injury by suppressing NF-κB signaling in vitro. Eur. Inflammation and Gut Microbiota Imbalance Associated Weight Loss Induced by
J. Pharm. 789 (2016) 195–201, https://doi.org/10.1016/j.ejphar.2016.07.039. Heat Stress in Broiler Chicken. Front Pharm. 11 (2020), 585945 https://doi.org/
[51] J. Lee, et al., 1,8-cineole prevents UVB-induced skin carcinogenesis by targeting 10.3389/fphar.2020.585945.
the aryl hydrocarbon receptor. Oncotarget 8 (62) (2017) 105995–106008, [77] K. Vijayakumar, et al., Eucalyptol inhibits biofilm formation of Streptococcus
https://doi.org/10.18632/oncotarget.22519. pyogenes and its mediated virulence factors. J. Med Microbiol 69 (11) (2020)
[52] A. Essaghir, et al., Transcription factor regulation can be accurately predicted 1308–1318, https://doi.org/10.1099/jmm.0.001253.
from the presence of target gene signatures in microarray gene expression data.,
e120, Nucleic Acids Res 38 (11) (2010), https://doi.org/10.1093/nar/gkq149.

13
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

[78] V. Cepas, et al., Relationship between biofilm formation and antimicrobial [104] Y. Li, et al., 1, 8-Cineol Protect Against Influenza-Virus-Induced Pneumonia in
resistance in gram-negative bacteria. Micro Drug Resist 25 (1) (2019) 72–79, Mice. Inflammation 39 (4) (2016) 1582–1593, https://doi.org/10.1007/s10753-
https://doi.org/10.1089/mdr.2018.0027. 016-0394-3.
[79] M. Şimşek, R. Duman, Investigation of effect of 1,8-cineole on antimicrobial [105] J. Müller, et al., 1,8-Cineole potentiates IRF3-mediated antiviral response in
activity of chlorhexidine gluconate. Pharmacogn. Res 9 (3) (2017) 234–237, human stem cells and in an ex vivo model of rhinosinusitis, Clin. Sci. (Lond. ) 130
https://doi.org/10.4103/0974-8490.210329. (15) (2016) 1339–1352, https://doi.org/10.1042/cs20160218.
[80] A.M. Ojeda-Sana, et al., New insights into antibacterial and antioxidant activities [106] Y.N. Lai, et al., Combinations of 1,8-cineol and oseltamivir for the treatment of
of rosemary essential oils and their main components. Food Control 31 (1) (2013) influenza virus A (H3N2) infection in mice. J. Med Virol. 89 (7) (2017)
189–195, https://doi.org/10.1016/j.foodcont.2012.09.022. 1158–1167, https://doi.org/10.1002/jmv.24755.
[81] K. Hąc-Wydro, K. Szydło, The influence of environmentally friendly pesticide - [107] C. Colalto, Volatile molecules for COVID-19: A possible pharmacological strategy?
Eucalyptol - alone and in combination with terpinen-4-ol - on model bacterial Drug Dev. Res 81 (8) (2020) 950–968, https://doi.org/10.1002/ddr.21716.
membranes. Colloids Surf. B Biointerfaces 146 (2016) 918–923, https://doi.org/ [108] J. Guarro, Fusariosis, a complex infection caused by a high diversity of fungal
10.1016/j.colsurfb.2016.07.044. species refractory to treatment. Eur. J. Clin. Microbiol Infect. Dis. 32 (12) (2013)
[82] Y. Sun, et al., Effects of 1,8-cineole on Carbohydrate Metabolism Related Cell 1491–1500, https://doi.org/10.1007/s10096-013-1924-7.
Structure Changes of Salmonella. Front Microbiol 9 (2018) 1078, https://doi.org/ [109] R.J. Hamill, Amphotericin B formulations: a comparative review of efficacy and
10.3389/fmicb.2018.01078. toxicity. Drugs 73 (9) (2013) 919–934, https://doi.org/10.1007/s40265-013-
[83] C.L. Moo, et al., Antimicrobial activity and mode of action of 1,8-cineol against 0069-4.
carbapenemase-producing Klebsiella pneumoniae. Sci. Rep. 11 (1) (2021) 20824, [110] M.T. Levine, P.H. Chandrasekar, Adverse effects of voriconazole: Over a decade of
https://doi.org/10.1038/s41598-021-00249-y. use. Clin. Transpl. 30 (11) (2016) 1377–1386, https://doi.org/10.1111/
[84] S. Hriouech, et al., The antistaphylococcal activity of amoxicillin/clavulanic acid, ctr.12834.
gentamicin, and 1,8-cineole alone or in combination and their efficacy through a [111] Z. Yang, et al., Anti-infectious bronchitis virus (IBV) activity of 1,8-cineole: effect
rabbit model of methicillin-resistant Staphylococcus aureus Osteomyelitis. Evid. on nucleocapsid (N) protein. J. Biomol. Struct. Dyn. 28 (3) (2010) 323–330,
Based Complement Altern. Med 2020 (2020) 4271017, https://doi.org/10.1155/ https://doi.org/10.1080/07391102.2010.10507362.
2020/4271017. [112] J. Silva, et al., Essential oils as antiviral agents. potential of essential oils to treat
[85] W. Mączka, et al., Can eucalyptol replace antibiotics? Molecules 26 (16) (2021) SARS-CoV-2 infection: an in-silico investigation. Int J. Mol. Sci. 21 (10) (2020)
https://doi.org/10.3390/molecules26164933. https://doi.org/10.3390/ijms21103426.
[86] A.A. Akhmouch, et al., Synergistic Action of AMX Associated with 1,8-Cineole [113] S. Panikar, et al., Essential oils as an effective alternative for the treatment of
and Its Effect on the ESBL Enzymatic Resistance Mechanism. Antibiot. (Basel) (8) COVID-19: Molecular interaction analysis of protease (M(pro)) with
(2022) 11, https://doi.org/10.3390/antibiotics11081002. pharmacokinetics and toxicological properties, J. Infect. Public Health 14 (5)
[87] A.A. Akhmouch, et al., The Combination of Amoxicillin and 1,8-Cineole Improves (2021) 601–610, https://doi.org/10.1016/j.jiph.2020.12.037.
the Bioavailability and the Therapeutic Effect of Amoxicillin in a Rabbit Model. [114] Y. Li, et al., Intranasal co-administration of 1,8-cineole with influenza vaccine
Antibiot. (Basel) 11 (10) (2022) https://doi.org/10.3390/antibiotics11101294. provide cross-protection against influenza virus infection. Phytomedicine 34
[88] P. Kwiatkowski, et al., Antibacterial Activity of Selected Essential Oil Compounds (2017) 127–135, https://doi.org/10.1016/j.phymed.2017.08.014.
Alone and in Combination with β-Lactam Antibiotics Against MRSA Strains. Int J. [115] D.M. Galan, et al., Eucalyptol (1,8-cineole): an underutilized ally in respiratory
Mol. Sci. 21 (19) (2020) https://doi.org/10.3390/ijms21197106. disorders? J. Essent. Oil Res. 32 (2) (2020) 103–110, https://doi.org/10.1080/
[89] J.S. Kumar, et al., Detection of quorum sensing molecules and biofilm formation 10412905.2020.1716867.
in Ralstonia solanacearum. Curr. Microbiol 72 (3) (2016) 297–305, https://doi. [116] N. Yu, et al., Eucalyptol protects lungs against bacterial invasion through
org/10.1007/s00284-015-0953-0. attenuating ciliated cell damage and suppressing MUC5AC expression. J. Cell
[90] Y. Wang, et al., 1,8-Cineole inhibits biofilm formation and bacterial pathogenicity Physiol. 234 (5) (2019) 5842–5850, https://doi.org/10.1002/jcp.26359.
by suppressing luxS gene expression in Escherichia coli., in: Front Pharmacol, 13, [117] P.L. Leopold, et al., Smoking is associated with shortened airway cilia. PLoS One 4
2022, 988245, https://doi.org/10.3389/fphar.2022.988245. (12) (2009), e8157 https://doi.org/10.1371/journal.pone.0008157.
[91] V. Karuppiah, R. Thirunanasambandham, G. Thangaraj, Anti-quorum sensing and [118] L.N. Coelho-de-Souza, et al., Relaxant effects of the essential oil of Eucalyptus
antibiofilm potential of 1,8-cineole derived from Musa paradisiaca against tereticornis and its main constituent 1,8-cineole on guinea-pig tracheal smooth
Pseudomonas aeruginosa strain PAO1. World J. Microbiol Biotechnol. 37 (4) muscle. Planta Med 71 (12) (2005) 1173–1175, https://doi.org/10.1055/s-2005-
(2021) 66, https://doi.org/10.1007/s11274-021-03029-y. 873173.
[92] N.M. Vazquez, et al., Cell death and biomass reduction in biofilms of multidrug [119] N.R. Nascimento, et al., 1,8-Cineole induces relaxation in rat and guinea-pig
resistant extended spectrum β-lactamase-producing uropathogenic Escherichia airway smooth muscle. J. Pharm. Pharm. 61 (3) (2009) 361–366, https://doi.org/
coli isolates by 1,8-cineole. PLoS One 15 (11) (2020), e0241978 https://doi.org/ 10.1211/jpp/61.03.0011.
10.1371/journal.pone.0241978. [120] Á. Pereira-Gonçalves, et al., 1,8-Cineole blocks voltage-gated L-type calcium
[93] N.M. Vazquez, S. Moreno, E.M. Galván, Exposure of multidrug-resistant Klebsiella channels in tracheal smooth muscle. Pflug. Arch. 470 (12) (2018) 1803–1813,
pneumoniae biofilms to 1,8-cineole leads to bacterial cell death and biomass https://doi.org/10.1007/s00424-018-2201-5.
disruption. Biofilm 4 (2022), 100085 https://doi.org/10.1016/j. [121] P. Sagortchev, J. Lukanov, A.M. Beer, Untersuchungen zu den Wirkungen von
bioflm.2022.100085. 1,8-Cineol auf die Aktivität von Histaminrezeptoren. Z. für Phytother. 33 (S 01)
[94] M. Schürmann, et al., The therapeutic effect of 1,8-cineol on pathogenic bacteria (2012) P05, https://doi.org/10.1055/s-0032-1313245.
species present in chronic rhinosinusitis. Front. Microbiol. (2019) 10, https://doi. [122] H. Sudhoff, et al., 1,8-Cineol Reduces Mucus-Production in a Novel Human Ex
org/10.3389/fmicb.2019.02325. Vivo Model of Late Rhinosinusitis. PLoS One 10 (7) (2015), e0133040 https://doi.
[95] L. Li, et al., Antibacterial activity of leaf essential oil and its constituents from org/10.1371/journal.pone.0133040.
Cinnamomum longepaniculatum. Int J. Clin. Exp. Med 7 (7) (2014) 1721–1727. [123] H. Jono, et al., Transforming growth factor-beta -Smad signaling pathway
[96] A. Merghni, et al., Assessment of the antibiofilm and antiquorum sensing cooperates with NF-kappa B to mediate nontypeable Haemophilus influenzae-
activities of Eucalyptus globulus essential oil and its main component 1,8-cineole induced MUC2 mucin transcription, J. Biol. Chem. 277 (47) (2002)
against methicillin-resistant Staphylococcus aureus strains. Micro Pathog. 118 45547–45557, https://doi.org/10.1074/jbc.M206883200.
(2018) 74–80, https://doi.org/10.1016/j.micpath.2018.03.006. [124] F.A. Santos, V.S. Rao, Antiinflammatory and antinociceptive effects of 1,8-cineole
[97] C. Morcia, M. Malnati, V. Terzi, In vitro antifungal activity of terpinen-4-ol, a terpenoid oxide present in many plant essential oils, Phytother. Res 14 (4)
eugenol, carvone, 1,8-cineole (eucalyptol) and thymol against mycotoxigenic (2000) 240–244, https://doi.org/10.1002/1099-1573(200006)14:4<240::aid-
plant pathogens. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk ptr573>3.0.co;2-x.
Assess. 29 (3) (2012) 415–422, https://doi.org/10.1080/ [125] M. Takaishi, et al., 1,8-cineole, a TRPM8 agonist, is a novel natural antagonist of
19440049.2011.643458. human TRPA1. Mol. Pain. 8 (2012) 86, https://doi.org/10.1186/1744-8069-8-
[98] D. Yu, et al., Antifungal modes of action of tea tree oil and its two characteristic 86.
components against Botrytis cinerea. J. Appl. Microbiol 119 (5) (2015) [126] C.Y. Jiang, et al., 1,8- and 1,4-cineole enhance spontaneous excitatory
1253–1262, https://doi.org/10.1111/jam.12939. transmission by activating different types of transient receptor potential channels
[99] Y. Zhang, et al., Study on the anti-biofilm mechanism of 1,8-cineole against in the rat spinal substantia gelatinosa. J. Neurochem 136 (4) (2016) 764–777,
Fusarium solani species complex. Front Pharm. 13 (2022) 1010593, https://doi. https://doi.org/10.1111/jnc.13433.
org/10.3389/fphar.2022.1010593. [127] J.M. Melo Júnior, et al., Acute and neuropathic orofacial antinociceptive effect of
[100] Z. Shahina, et al., Rosemary essential oil and its components 1,8-cineole and eucalyptol. Inflammopharmacology 25 (2) (2017) 247–254, https://doi.org/
α-pinene induce ROS-dependent lethality and ROS-independent virulence 10.1007/s10787-017-0324-5.
inhibition in Candida albicans. PLoS One 17 (11) (2022), e0277097 https://doi. [128] L.X. Wang, Z.J. Wang, Animal and cellular models of chronic pain. Adv. Drug
org/10.1371/journal.pone.0277097. Deliv. Rev. 55 (8) (2003) 949–965, https://doi.org/10.1016/s0169-409x(03)
[101] L. Li, et al., Preparation, characterization, ex vivo transdermal properties and skin 00098-x.
irritation evaluation of 1,8-cineole nanoemulsion gel. Int J. Pharm. 624 (2022), [129] Y.L. Zhang, et al., Microencapsulated Schwann cell transplantation inhibits P2X3
121982 https://doi.org/10.1016/j.ijpharm.2022.121982. receptor expression in dorsal root ganglia and neuropathic pain. Neural Regen.
[102] K.Z. Bourne, et al., Plant products as topical microbicide candidates: assessment Res 13 (11) (2018) 1961–1967, https://doi.org/10.4103/1673-5374.238715.
of in vitro and in vivo activity against herpes simplex virus type 2. Antivir. Res 42 [130] D.Y. Kim, et al., Eucalyptol Inhibits Amyloid-β-Induced Barrier Dysfunction in
(3) (1999) 219–226, https://doi.org/10.1016/s0166-3542(99)00020-0. Glucose-Exposed Retinal Pigment Epithelial Cells and Diabetic Eyes. Antioxid.
[103] A. Astani, J. Reichling, P. Schnitzler, Comparative study on the antiviral activity (Basel) 9 (2020) 10, https://doi.org/10.3390/antiox9101000.
of selected monoterpenes derived from essential oils, Phytother. Res 24 (5) (2010)
673–679, https://doi.org/10.1002/ptr.2955.

14
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

[131] Y. Wang, et al., 1, 8-cineole attenuates cardiac hypertrophy in heart failure by [159] L. Dao, et al., Study on the opening effect of eucalyptol on the blood-brain barrier
inhibiting the miR-206-3p/SERP1 pathway. Phytomedicine 91 (2021), 153672 and its brain pharmacokinetics. Biomed. Chromatogr. 37 (6) (2023), e5631
https://doi.org/10.1016/j.phymed.2021.153672. https://doi.org/10.1002/bmc.5631.
[132] A.N. Abdalla, et al., Proapoptotic Activity of Achillea membranacea Essential Oil [160] J. Karlawish, J.D. Grill, The approval of Aduhelm risks eroding public trust in
and Its Major Constituent 1,8-Cineole against A2780 Ovarian Cancer Cells. Alzheimer research and the FDA. Nat. Rev. Neurol. 17 (9) (2021) 523–524,
Molecules 25 (7) (2020) https://doi.org/10.3390/molecules25071582. https://doi.org/10.1038/s41582-021-00540-6.
[133] B. Rodenak-Kladniew, et al., 1,8-Cineole promotes G0/G1 cell cycle arrest and [161] A. Khan, et al., 1,8-cineole (eucalyptol) mitigates inflammation in amyloid Beta
oxidative stress-induced senescence in HepG2 cells and sensitizes cells to anti- toxicated PC12 cells: relevance to Alzheimer’s disease. Neurochem Res 39 (2)
senescence drugs. Life Sci. 243 (2020), 117271 https://doi.org/10.1016/j. (2014) 344–352, https://doi.org/10.1007/s11064-013-1231-9.
lfs.2020.117271. [162] K. Paul, et al., Is 1,8-Cineole-Rich Extract of Small Cardamom Seeds More
[134] S. Murata, et al., Antitumor effect of 1, 8-cineole against colon cancer. Oncol. Rep. Effective in Preventing Alzheimer’s Disease than 1,8-Cineole Alone? Neuromol.
30 (6) (2013) 2647–2652, https://doi.org/10.3892/or.2013.2763. Med. 22 (1) (2020) 150–158, https://doi.org/10.1007/s12017-019-08574-2.
[135] S. Sampath, et al., Bioactive compound 1,8-Cineole selectively induces G2/M [163] E. Rönnemaa, et al., Impaired insulin secretion increases the risk of Alzheimer
arrest in A431 cells through the upregulation of the p53 signaling pathway and disease. Neurology 71 (14) (2008) 1065–1071, https://doi.org/10.1212/01.
molecular docking studies. Phytomedicine 46 (2018) 57–68, https://doi.org/ wnl.0000310646.32212.3a.
10.1016/j.phymed.2018.04.007. [164] R. Buller, V. Legrand, Novel treatments for anxiety and depression: hurdles in
[136] H. Moteki, et al., Specific induction of apoptosis by 1,8-cineole in two human bringing them to the market. Drug Discov. Today 6 (23) (2001) 1220–1230,
leukemia cell lines, but not a in human stomach cancer cell line. Oncol. Rep. 9 (4) https://doi.org/10.1016/s1359-6446(01)02043-8.
(2002) 757–760. [165] G. Dougnon, M. Ito, Inhalation Administration of the Bicyclic Ethers 1,8- and 1,4-
[137] J.-D. Cha, Y.-H. Kim, J.-Y. Kim, Essential oil and 1, 8-cineole from Artemisia cineole Prevent Anxiety and Depressive-Like Behaviours in Mice. Molecules 25
lavandulaefolia induces apoptosis in KB cells via mitochondrial stress and caspase (8) (2020) https://doi.org/10.3390/molecules25081884.
activation. Food Sci. Biotechnol. 19 (2010) 185–191. [166] T.E. Ceremuga, et al., Investigation of the Anxiolytic and Antidepressant Effects of
[138] K.A. Alatawi, et al., 1,8-Cineole affects agonists-induced platelet activation, Eucalyptol (1,8-Cineole), a Compound From Eucalyptus, in the Adult Male
thrombus formation and haemostasis. Cells 10 (10) (2021) https://doi.org/ Sprague-Dawley Rat. Aana J. 85 (4) (2017) 277–284.
10.3390/cells10102616. [167] K.Y. Kim, et al., The effect of 1,8-cineole inhalation on preoperative anxiety: a
[139] W. Kehrl, U. Sonnemann, U. Dethlefsen, Therapy for acute nonpurulent randomized clinical trial, Evid. Based Complement Altern. Med 2014. (2014),
rhinosinusitis with cineole: results of a double-blind, randomized, placebo- 820126, https://doi.org/10.1155/2014/820126.
controlled trial. Laryngoscope 114 (4) (2004) 738–742, https://doi.org/10.1097/ [168] D. Schmidt, Drug treatment of epilepsy: options and limitations. Epilepsy Behav.
00005537-200404000-00027. 15 (1) (2009) 56–65, https://doi.org/10.1016/j.yebeh.2009.02.030.
[140] S. Tesche, et al., The value of herbal medicines in the treatment of acute non- [169] M. Culić, et al., Wavelet and fractal analysis of rat brain activity in seizures
purulent rhinosinusitis, Results a Double-Blind, Random, Control. Trial Eur. Arch. evoked by camphor essential oil and 1,8-cineole, Gen. Physiol. Biophys. (2009)
Otorhinolaryngol. 265 (11) (2008) 1355–1359, https://doi.org/10.1007/s00405- 33–40, 28 Spec No.
008-0683-z. [170] Z. Zeraatpisheh, J. Vatanparast, Eucalyptol induces hyperexcitability and
[141] M. Wittmann, et al., Therapy with expectorants in COPD patients: A double-blind epileptiform activity in snail neurons by inhibiting potassium channels. Eur. J.
randomized study comparing Ambroxol and Cineol. Atemwegs- und Pharm. 764 (2015) 70–78, https://doi.org/10.1016/j.ejphar.2015.06.050.
Lungenkrankh. 24 (2) (1998) 67–74. [171] A. Lanas, F.K.L. Chan, Peptic ulcer disease., in: Lancet, 390, 2017, pp. 613–624,
[142] H. Worth, C. Schacher, U. Dethlefsen, Concomitant therapy with Cineole https://doi.org/10.1016/s0140-6736(16)32404-7.
(Eucalyptole) reduces exacerbations in COPD: a placebo-controlled double-blind [172] G.F. Rocha Caldas, et al., Gastroprotective Mechanisms of the Monoterpene 1,8-
trial. Respir. Res 10 (1) (2009) 69, https://doi.org/10.1186/1465-9921-10-69. Cineole (Eucalyptol, PLoS One 10 (8) (2015), e0134558, https://doi.org/
[143] H. Worth, U. Dethlefsen, Patients with asthma benefit from concomitant therapy 10.1371/journal.pone.0134558.
with cineole: a placebo-controlled, double-blind trial. J. Asthma 49 (8) (2012) [173] F.A. Santos, V.S. Rao, 1,8-cineol, a food flavoring agent, prevents ethanol-induced
849–853, https://doi.org/10.3109/02770903.2012.717657. gastric injury in rats, Dig. Dis. Sci. 46 (2) (2001) 331–337, https://doi.org/
[144] P. Kardos, O. Khaletskaya, O. Kropova, Efficacy and safety of Cineole (Soledum®) 10.1023/a:1005604932760.
in the treatment of patients with acute bronchitis: results of an open-label [174] R.E. Black, et al., Global, regional, and national causes of child mortality in 2008:
randomized clinical phase III study. Clin. Phytoscience 7 (1) (2021) 83, https:// a systematic analysis. lancet 375 (9730) (2010) 1969–1987.
doi.org/10.1186/s40816-021-00319-8. [175] G. Jalilzadeh-Amin, M. Maham, The application of 1,8-cineole, a terpenoid oxide
[145] J. Fischer, U. Dethlefsen, Efficacy of cineole in patients suffering from acute present in medicinal plants, inhibits castor oil-induced diarrhea in rats. Pharm.
bronchitis: a placebo-controlled double-blind trial., in: Cough, 9, 2013, p. 25, Biol. 53 (4) (2015) 594–599, https://doi.org/10.3109/13880209.2014.935862.
https://doi.org/10.1186/1745-9974-9-25. [176] P.J. Magalhães, et al., Intestinal myorelaxant and antispasmodic effects of the
[146] C. Nauert, W. Oestreich, Nichtinterventionelle Studie an 336 Kindern (2 – 11J) essential oil of Croton nepetaefolius and its constituents cineole, methyl-eugenol
mit akuter Bronchitis: Compliance, Verträglichkeit und klinischer Verlauf unter and terpineol, Phytother. Res.: Int. J. Devoted Pharmacol. Toxicol. Eval. Nat.
einer Therapie mit Cineol. Z. für Phytother. 36 (S 01) (2015) V05, https://doi. Prod. Deriv. 12 (3) (1998) 172–177.
org/10.1055/s-0035-1565933. [177] E.M. Lau, et al., Epidemiology and treatment of pulmonary arterial hypertension.
[147] Schmidt, M., Cineol bei akuter Bronchitis: Eine nicht interventionelle Studie bei Nat. Rev. Cardiol. 14 (10) (2017) 603–614.
Kindern und Erwachsenen. Journal Pharmakol. u. Ther., 2013. [178] Q. Wu, et al., Sulforaphane protects H9c2 cardiomyocytes from angiotensin II-
[148] P. Kardos, et al., Benefits of early cineole administration during common cold: an induced hypertrophy. Herz 39 (3) (2014) 390.
exploratory clinical trial. Eur. Respir. J. 60 (suppl 66) (2022) 2486, https://doi. [179] P. Mendes-Ferreira, et al., Distinct right ventricle remodeling in response to
org/10.1183/13993003.congress-2022.2486. pressure overload in the rat. Am. J. Physiol. -Heart Circ. Physiol. 311 (1) (2016)
[149] R.S. Leung, R. Katial, The diagnosis and management of acute and chronic H85–H95.
sinusitis. Prim. care: Clin. Off. Pract. 35 (1) (2008) 11–24. [180] N.V. Pinto, et al., Endothelium-dependent vasorelaxant effects of the essential oil
[150] D. Hastan, et al., Chronic rhinosinusitis in Europe–an underestimated disease. A from aerial parts of Alpinia zerumbet and its main constituent 1, 8-cineole in rats.
GA2LEN study, Allergy 66 (9) (2011) 1216–1223, https://doi.org/10.1111/ Phytomedicine 16 (12) (2009) 1151–1155.
j.1398-9995.2011.02646.x. [181] S. Lahlou, et al., Cardiovascular effects of 1, 8-cineole, a terpenoid oxide present
[151] W.W. Stevens, R.P. Schleimer, R.C. Kern, Chronic rhinosinusitis with nasal polyps, in many plant essential oils, in normotensive rats. Can. J. Physiol. Pharmacol. 80
J. Allergy Clin. Immunol. Pr. 4 (4) (2016) 565–572, https://doi.org/10.1016/j. (12) (2002) 1125–1131.
jaip.2016.04.012. [182] J.M. Alves-Silva, et al., 1,8-Cineole ameliorates right ventricle dysfunction
[152] S.A. Joe, R. Thambi, J. Huang, A systematic review of the use of intranasal associated with pulmonary arterial hypertension by restoring connexin43 and
steroids in the treatment of chronic rhinosinusitis. Otolaryngol. Head. Neck Surg. mitochondrial homeostasis. Pharm. Res 180 (2022), 106151 https://doi.org/
139 (3) (2008) 340–347, https://doi.org/10.1016/j.otohns.2008.05.628. 10.1016/j.phrs.2022.106151.
[153] G. Bachmann, G. Hommel, O. Michel, Effect of irrigation of the nose with isotonic [183] N.F. Ruopp, B.A. Cockrill, Diagnosis and treatment of pulmonary arterial
salt solution on adult patients with chronic paranasal sinus disease. Eur. Arch. hypertension: a review. Jama 327 (14) (2022) 1379–1391, https://doi.org/
Otorhinolaryngol. 257 (10) (2000) 537–541, https://doi.org/10.1007/ 10.1001/jama.2022.4402.
s004050000271. [184] N.A. ElSayed, et al., 2. Classification and diagnosis of diabetes: standards of care
[154] J.P. O’Gara, ica and beyond: biofilm mechanisms and regulation in in diabetes-2023. Diabetes Care 46 (Suppl 1) (2023) S19–s40, https://doi.org/
Staphylococcus epidermidis and Staphylococcus aureus, FEMS Microbiol Lett. 10.2337/dc23-S002.
270 (2) (2007) 179–188, https://doi.org/10.1111/j.1574-6968.2007.00688.x. [185] S. Mahdavifard, M. Nakhjavani, 1,8 cineole protects type 2 diabetic rats against
[155] P.M. Calverley, et al., Salmeterol and fluticasone propionate and survival in diabetic nephropathy via inducing the activity of glyoxalase-I and lowering the
chronic obstructive pulmonary disease. N. Engl. J. Med 356 (8) (2007) 775–789, level of transforming growth factor-1β. J. Diabetes Metab. Disord. 21 (1) (2022)
https://doi.org/10.1056/NEJMoa063070. 567–572, https://doi.org/10.1007/s40200-022-01014-2.
[156] P. Kaspar, et al., Sekretolytika im Vergleich, Änderung der Ziliarfrequenz und [186] D.Y. Kim, et al., Eucalyptol ameliorates Snail1/β-catenin-dependent diabetic
Lungenfunktion nach Therapie mit Cineol und Ambroxol, Atemw-Lungenkrkh 20 disjunction of renal tubular epithelial cells and tubulointerstitial fibrosis.
(1994) 605–614. Oncotarget 8 (63) (2017) 106190–106205, https://doi.org/10.18632/
[157] R. Buhl, et al., Guidelines for diagnosis and treatment of asthma patients, oncotarget.22311.
Pneumologie 60 (3) (2006) 139–177, https://doi.org/10.1055/s-2005-919153. [187] D.Y. Kim, et al., Eucalyptol Inhibits Advanced Glycation End Products-Induced
[158] R.P. Wenzel, A.A. Fowler 3rd, Clinical practice. Acute bronchitis. N. Engl. J. Med Disruption of Podocyte Slit Junctions by Suppressing Rage-Erk-C-Myc Signaling
355 (20) (2006) 2125–2130, https://doi.org/10.1056/NEJMcp061493.

15
C.C. Hoch et al. Biomedicine & Pharmacotherapy 167 (2023) 115467

Pathway. Mol. Nutr. Food Res 62 (19) (2018), e1800302 https://doi.org/ and MMP-9. PLoS One 17 (1) (2022), e0258998 https://doi.org/10.1371/journal.
10.1002/mnfr.201800302. pone.0258998.
[188] K.H. Cho, 1,8-cineole protected human lipoproteins from modification by [190] K.L. Bruchhage, et al., 1,8-cineol inhibits the Wnt/β-catenin signaling pathway
oxidation and glycation and exhibited serum lipid-lowering and anti- through GSK-3 dephosphorylation in nasal polyps of chronic rhinosinusitis
inflammatory activity in zebrafish, BMB Rep. 45 (10) (2012) 565–570, https:// patients. Eur. J. Pharm. 835 (2018) 140–146, https://doi.org/10.1016/j.
doi.org/10.5483/bmbrep.2012.45.10.044. ejphar.2018.07.060.
[189] H.M.I. Abdallah, et al., 1,8 Cineole and Ellagic acid inhibit hepatocarcinogenesis [191] E.C.D. Santos, et al., The monoterpene 1,8-cineole prevents cerebral edema in a
via upregulation of MiR-122 and suppression of TGF-β1, FSCN1, Vimentin, VEGF, murine model of severe malaria., in: PLoS One, 17, 2022, https://doi.org/
10.1371/journal.pone.0268347.

16

You might also like