Next Article in Journal
Repurposing Antifungals for Host-Directed Antiviral Therapy?
Next Article in Special Issue
Protective and Therapeutic Efficacy of Hesperidin versus Cisplatin against Ehrlich Ascites Carcinoma-Induced Renal Damage in Mice
Previous Article in Journal
A Lead-Based Fragment Library Screening of the Glycosyltransferase WaaG from Escherichia coli
Previous Article in Special Issue
Clinical Outcomes of Secondary Prophylactic Granulocyte Colony-Stimulating Factors in Breast Cancer Patients at a Risk of Neutropenia with Doxorubicin and Cyclophosphamide-Based Chemotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effect and Mechanism of Herbal Medicines on Cisplatin-Induced Anorexia

1
Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea
2
Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 022447, Korea
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2022, 15(2), 208; https://doi.org/10.3390/ph15020208
Submission received: 13 January 2022 / Revised: 4 February 2022 / Accepted: 5 February 2022 / Published: 9 February 2022
(This article belongs to the Special Issue Clinical Development of Cancer Treatment)

Abstract

:
Cisplatin is a well-known chemotherapeutic agent used to treat various types of cancers; however, it can also induce anorexia, which results in reduced food intake, loss of body weight, and lower quality of life. Although drugs such as megestrol acetate and cyproheptadine are used to decrease this severe feeding disorder, they can also induce side effects, such as diarrhea and somnolence, which limit their widespread use. Various types of herbal medicines have long been used to prevent and treat numerous gastrointestinal tract diseases; however, to date, no study has been conducted to analyze and summarize their effects on cisplatin-induced anorexia. In this paper, we analyze 12 animal studies that used either a single herbal medicine extract or mixtures thereof to decrease cisplatin-induced anorexia. Among the herbal medicines, Ginseng Radix was the most used, as it was included in seven studies, whereas both Glycyrrhizae Radix et Rhizoma and Angelicae Gigantis Radix were used in four studies. As for the mechanisms of action, the roles of serotonin and its receptors, cytokines, white blood cells, ghrelin, and leptin were investigated. Based on these results, we suggest that herbal medicines could be considered a useful treatment method for cisplatin-induced anorexia.

1. Introduction

Cisplatin (also known as cisplatinum or cis-diamminedichloroplatinum (II)), is a first-generation platinum-based chemotherapeutic agent widely used to treat solid cancers, such as testicular, lung, ovarian, and breast cancer, since its approval by the Food and Drug Administration (FDA) in 1978 [1,2,3]. However, along with its remarkable antitumor effect, cisplatin can also induce several side effects, such as nephrotoxicity, allergic reactions, decreased immunity to infections, and diverse gastrointestinal (GI) disorders [4,5,6]. In particular, GI disorders such as diarrhea, nausea, vomiting, and anorexia are clinically important, as they can lead to the discontinuation of therapy due to reduced dietary intake and abnormal metabolism [7,8,9,10,11]. Among them, anorexia is particularly important, as it has been reported that approximately half of patients with cancer suffer from anorexia after chemotherapy treatments [12,13,14,15,16].
Anorexia is a loss of appetite, including the occurrence of early satiety, which results in reduced food intake, loss of body weight, and lower quality of life [17]. Furthermore, when these symptoms become chronic, a loss of fat and muscle occurs, which often leads to cachexia [18,19,20]. Megestrol acetate (MGA) [21,22,23,24] and cyproheptadine [25,26] are used as first-line treatments to attenuate anorexia. They have been reported to enhance appetite and increase the body weight of patients with chemotherapy-induced anorexia [27,28,29]. However, both MGA and cyproheptadine are known to cause adverse effects. MGA is associated with impotence, deep-vein thrombosis, and gastrointestinal complications, such as diarrhea [30,31], and cyproheptadine has been reported to cause somnolence, hallucinations, tachycardia, and muscle twitching [32,33]. Therefore, efforts to find new treatments that could attenuate anorexia with fewer side effects are needed.
Herbal medicines have been used as a relatively safe remedy for many years to prevent and treat numerous diseases, making it possible to promote health and enhance the quality of life [34,35]. In particular, they have been shown to be effective against diverse GI tract disorders, such as dyspepsia, constipation, nausea, and vomiting [36,37,38]. In clinical settings, ban-xia-xie-xin-tang, a classical herbal mixture from one of the earliest books of traditional medicine, was shown to be effective in patients with chemotherapy-induced diarrhea through a significant improvement and reduction in the frequency of diarrhea with higher grades [39,40]. Another well-known herbal medicine, Dai-kenchu-to, increased the duodenum and jejunum motility in dogs when administered intraduodenally and intrajejunally [41]. Moreover, isolated guinea pig ileum Dai-kenchu-to was also shown to induce contractions accompanied by autonomous contractions [42].
In our lab, we have previously shown that orally administered herbal mixtures, such as Sip-Jeon-Dae-Bo-Tang (SJDBT) [43] and LA16001 [44], could effectively attenuate cisplatin-induced appetite loss in mice. These results suggest that herbal medicines could be considered a useful option to attenuate cisplatin-induced anorexia. Although dozens of studies have been published in the last 20 years to demonstrate the effect of herbal medicines on cisplatin-induced anorexia, to date, no reviews summarizing and analyzing their effects on cisplatin-induced feeding disorders have been published. Thus, by analyzing 12 studies that used either a single herbal medicine extract or mixtures thereof to treat cisplatin-induced anorexia in rodents, their therapeutic effect and the underlying mechanism of action are discussed.

2. Results

The included studies focused on various pathways as the pathogenesis mechanism of cisplatin-induced anorexia, as well as the curative action of herbal medicines (Table 1 and Table 2). The role of 5-HT and its receptors in cisplatin-induced anorexia was investigated in four studies [45,46,47,48]; inflammatory cytokines [43,44,47,49] and WBCs [44,47,49,50] were also studied in four studies each; the roles of ghrelin and leptin in cisplatin-induced anorexia were assessed in five [43,44,45,46,48] and two [43,44] studies, respectively.

2.1. 5-HT and Its Receptors

Serotonin (5-HT) and its receptors are involved in several physiological and pathological functions of the GI tract, including motility, secretion, and weight maintenance [49]. In the brain, 5-HT is synthesized in the dorsal and median raphe and innervates most of brain regions related to feeding regulation, including the hypothalamus, hippocampus, amygdala, and frontal cortex [50]. In the periphery, 5-HT is mostly secreted in the GI tract, especially in the secretory granules of enterochromaffin cells [51]. GI disorders are known to occur when large amounts of 5-HT are released from enterochromaffin cells and bind to 5-HT receptors [45]. Loss of appetite has also been shown to be closely related to 5-HT and its receptors, as food intake is significantly reduced following intraperitoneal injection of 5-HT [52,53,54]. In this review, four studies observed the role of 5-HT and its receptors in cisplatin-induced anorexia [45,46,47,48].
In a study by Takeda et al. [45], a single intraperitoneal injection of cisplatin induced a decrease in 6 h food intake in rats; however, the administration of 5-HT2B or 5-HT2C receptor antagonists (i.p.) attenuated the decrease in food intake, indicating that the activation of 5-HT receptors plays an important role in the development of cisplatin-induced anorexia. The authors further assessed that the plasma-acylated ghrelin level, which is the activated form of ghrelin, was significantly decreased in rats treated with both cisplatin and 5-HT2B or 5-HT2C receptor agonists. However, injection of 5-HT3 or 5-HT4 receptor agonists failed to alter the acylated ghrelin level. In this study, rikkunshito (RKT), which is composed of eight herbal medicines, improved cisplatin-induced anorexia. Among the crude drug components of RKT, 3,3′,4′,5,6,7,8-heptamethoxyflavone (HMF), hesperetin and isoliquiritigenin (found in Aurantii nobilis Pericarpium), Glycyrrhizae Radix et Rhizoma, and Zingiberis Rhizoma, showed significant inhibitory constant (Ki) values against the 5-HT2B receptor. Moreover, hesperetin and isoliquiritigenin also showed significant Ki values against the 5-HT2C receptor, suggesting that inhibiting the action of 5-HT2B or 5-HT2C receptors may prevent a decrease in food intake induced by cisplatin injection. In accordance with the study of Takeda et al., Yakabi et al. [46] also found that 5-HT2C receptors are involved in cisplatin-induced anorexia, as a 5-HT2C receptor antagonist (SB242084HCL) treatment reversed the decrease in appetite induced by cisplatin, whereas 5-HT3 receptor antagonists (granisetron or ondansetron) did not. Altogether, these results suggest that 5-HT2B and 5-HT2C receptors, but not 5-HT3 and 5-HT4 receptors, are involved in cisplatin-induced anorexia.
The role of 5-HT in the small intestine was observed by Kim et al. [47]. They showed that 5-HT concentration in small-intestine tissue increased by 1.8-fold compared to that in the normal group after cisplatin injection. In their study, water extract of Rhus verniciflua Stoke (RVX) attenuated anorexia and succeeded in decreasing 5-HT levels in the small intestine, indicating that modulating 5-HT in the small intestine may be an effective treatment for anorexia. Furthermore, RVX significantly upregulated serotonin transporters (SERT) and downregulated 5-HT3A receptors in the small intestine. SERT transports 5-HT to the epithelial cells of the GI tract, where 5-HT is metabolized. It has been reported that cisplatin injection decreases the number of SERT [55]. Thus, damage to epithelial cells due to cisplatin can increase the action of 5-HT to its receptors, such as 5-HT3A, whose activation is known to be related to chemotherapy-induced emesis [56].
In a study by Song et al. [48], He-Wei granules (HWKL), which are formulated from seven herbs, increased 5-hydroxyindole acetic-acid (5-HIAA) content and lowered 5-HT and substance P levels, which had been elevated following cisplatin injection, thus increasing the ratio of 5-HIAA/5-HT in the serum, medulla oblongata, and ileum part of the small intestine. The 5-HIAA/5-HT ratio is used to represent the effect of a drug on 5-HT [57,58], and an increase in the ratio can be interpreted as a decreased effect of 5-HT due to its metabolism to 5-HIAA. In addition, Song et al. confirmed that HWKL significantly reduced 5-HT3A receptor mRNA, and protein-expression levels in both the ileum and medulla oblongata increased after cisplatin treatment. Furthermore, the upregulated substance-P levels in the serum, medulla oblongata, and ileum decreased after HWKL administration. Substance P is a neurotransmitter produced from enterochromaffin cells similar to 5-HT that is involved in feeding behavior, and its intraperitoneal injection has been shown to reduce food intake [59,60]. Moreover, it is reported that substance P is upregulated via the interaction between 5-HT, which is increased by cisplatin injection, and the 5-HT3 receptor [61,62].

2.2. Inflammatory Cytokines

Compelling studies have demonstrated that anorexia is associated with inflammation [63]. Injection of proinflammatory cytokines results in reduced food intake in rodents [64]. In patients with anorexia nervosa, an increase in pro-inflammatory cytokines, such as interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6), have also been reported [65]. IL-1β has been reported to suppress feeding by reducing meal frequency and size through several pathways, including both neural and humoral pathways [63,66,67,68]. TNF-α [69,70,71] and IL-6 [72] have also been reported to influence feeding behavior [73,74]. In this review, the relationship between changes in inflammatory cytokines in cisplatin-induced anorexia was analyzed in four studies [43,44,47,49].
Table 1. The effect of herbal medicine extracts in cisplatin-induced anorexia.
Table 1. The effect of herbal medicine extracts in cisplatin-induced anorexia.
Authors/
Years
Animal TypeCisplatin DosingHerbal Medicine/
Dose
Findings
GroupFood IntakeKaolin IntakeBody WeightMechanisms of Actions
Aung et al.
2003
[75]
Wistar ratsSingle
3 mg/kg,
i.p.
Scutellaria baicalensis roots [SbE]
(water extract)
1, 3, and 10 mg/kg, i.p.
Cisplatin (n = 7)--
Cisplatin + SbE (n = 6)--
Mehendale
et al.
2005
[76]
Wistar ratsSingle
3 mg/kg,
i.p.
American ginseng berries [AGBE]
(75% ethanol extract)
50, 100, and 150 mg/kg, i.p.
Cisplatin (n = 6)--
Cisplatin + AGBE (n = 6)--
Cisplatin + Ginsenoside Re (5 mg/kg, i.p.) (n = 6)---
Takeda et al.
2008
[45]
SD
rats
Single
2 mg/kg,
i.p.
Rikkunshito
[RKT];
Atractylodis Lanceae Rhizoma, Ginseng Radix, Pinelliae Tuber,
Hoelen, Zizyphi Fructus, Aurantii Nobilis Pericarpium, Glycyrrhizae
Radix, Zingiberis Rhizoma).
(water extract)
500, 1000 mg/kg, p.o.
Cisplatin (n = 4–5)--Plasma-acylated ghrelin↓
Plasma-desacylated ghrelin↓
5-HT (4 & 8 mg/kg, i.p.) (n = 4–5)---Plasma-acylated ghrelin↓
Plasma-desacylated ghrelin↓
Cisplatin + RKT (n = 4–5)--Plasma-acylated ghrelin↑
Cisplatin + RKT + GHS-R1a antagonist (0.4 μmol/rat, i.p.) (n = 4–5)---
Cisplatin + HMF
or Hesperetin or Isoliquiritigenin (n = 4–5)
---Plasma-acylated ghrelin↑
HMF, Hesperetin, Isoliquiritigenin (n = 4–5)---5-HT2B receptor:
Ki: 0.21 ± 0.01 & 5.3 ± 0.1 & 3.3 μmol/L
Hesperetin, Isoliquiritigenin (n = 4–5)---5-HT2C receptor:
Ki: 20.9 ± 7.1 & 3.5 ± 0.1 μmol/L
Yakabi et al.
2010
[46]
SD
rats
Single
2 mg/kg,
i.p.
[RKT]
(water extract)
500, 1000 mg/kg, p.o.
Cisplatin (n = 4–5)--Hypothalamic GHS-R1a↓
Cisplatin + Ghrelin
(2 nmol/rat, I.C.V.) (n = 4–5)
---
5-HT2C receptor agonist (9 mg/kg, i.p.) (n = 4–5)---
5-HT2C receptor agonist
+ RKT (n = 4–5)
---
Cisplatin + RKT (n = 4–5)--Hypothalamic GHS-R1a↑
Effect abolished by
GHS-R1a antagonist (1nmol/rat, I.C.V.)
Cisplatin + Hesperidin or Isoliquiritigenin (n = 4–5)--Effect abolished by
GHS-R1a antagonist (1 nmol/rat, I.C.V.)
Raghavendran
et al.
2011
[77]
SD
rats
Single
Pre—7 mg/kg
Post—6 mg/kg,
i.p.
Korean ginseng roots [KG; the root of Panax ginseng CA Meyer]
(water extract)
Pre—25, 50, 100 mg/kg
Post—12.5, 25, 50 mg/kg
p.o.
Cisplatin (n = 8)-Total WBC↑ Neutrophil↑ Lymphocyte↑
Stomach injury↑ Small intestine injury↑
Cisplatin + KG (pre-treatment) (n = 8) -Total WBC↓ Neutrophil↓ Lymphocyte↓
Stomach injury↓ Small intestine injury↓
Cisplatin + KG (post-treatment) (n = 8)--
Woo et al.
2016
[43]
Balb/c
mice
Single
8 mg/kg,
i.p.
Sip-jeon-dea-bo-tang
[SJDBT; Angelicae Gigantis Radix, Astragali Radix, Atractylodis Rhizoma Alba, Cinnamomi Cortex, Cnidii Rhizoma, Paeoniae Radix, Ginseng Radix, Poria Sclerotium, Rehmannia Radix, Glycyrrhizae Radix et Rhizoma]
(water extract)
678.4 mg/kg, p.o.
Cisplatin (n = 6)-Leptin↓ IL-6↓
Cisplatin + SJDBT (Single) (n = 6)--Leptin↑ IL-6↑ JAK1/STAT3↑
Cisplatin + SJDBT (multiple) (n = 6)-Leptin↑ IL-6↑
Kim et al.
2017
[47]
SD
rats
Single
6 mg/kg,
i.p.
Rhus verniciflua Stoke [RVX]
(water extract)
25, 50, 100 mg/kg, p.o.
Cisplatin (n = 6)5-HT↑ 5-HT3A receptor↑ SERT↓ TNF-α↑ IL-6↑ IL-1β↑ WBC↓ Lymphocyte↓
Bone-marrow tissue injury↑
Thymus weight↓
Spleen weight↓
Cisplatin + RVX (n = 6)5-HT↓ 5-HT3A receptor↓ SERT↑ TNF-α↓ IL-6↓ IL-1β↓ WBC↑ Lymphocyte↑
Bone-marrow tissue injury↓
Thymus weight↑
Spleen weights↑
Woo et al.
2017
[44]
Balb/c
mice
Single
8 mg/kg,
i.p.
LCBP-Anocure
[LA; Atractylodis Rhizoma Alba (AJ), Angelicae Gigantis Radix (AG), Astragali Radix (AM), Lonicerae Flos (LJ), Taraxaci Herba (TP), Prunellae Spica (PV)]-16001, 16002, 16003
(water extract)
1000 mg/kg, p.o.
Cisplatin (n = 4)-Active ghrelin↓ Leptin↓ IL-6↓ p-JAK1↓p-STAT3↓ WBC↓ Neutrophil↓
Cisplatin + LA16001 (n = 4)-Active ghrelin↑ Leptin↑ IL-6↑ p-JAK1↑ p-STAT3↑
WBC↑ Neutrophil↑
Cisplatin + LA16002 or LA16003 or SJDBT or AJ or AG or LJ or PV (n = 4)---
Cisplatin + MGA (100 mg/kg, p.o.) or AM or TP (n = 4)--
Song et al.
2017
[48]
Wistar
rats
Single
5 mg/kg,
i.p.
He-Wei granules
[HWKL; Pinelliae Tuber, Zingiberis Rhizoma Recens, Ginseng Radix et Rhizoma, Scutellariae Radix, Coptidis Rhizoma, Glycyrrhizae Radix et Rhizoma, Jujubae Fructus]
(water extract)
1.18 (low), 2.36 (middle), 4.725 (high) mg/kg, i.g.
Cisplatin (n = 12)5-HIAA↓ SP↑ 5-HT↑
5-HIAA/5-HT↓ SERT↓
MAO-A↓ 5-HT3AR↑ PPTA↑
TPH-1↑ TPH-2↑ NK-1R↑ TPH-1↑ OB↑ GH↓ GH/OB↓ GPR39↑ EGFR↓
pERK1/2↓ GSH-PX↓ Stomach injury↑ SOD↓ MDA↑
Ileum injury↑
Cisplatin + HWKL (n = 12)5-HIAA↑ SP↓ 5-HT↓ 5-HIAA/5-HT↑ SERT↑
MAO-A↑ 5-HT3AR↓ PPTA↓
TPH-1↓ TPH-2↓ NK-1R↓ TPH-1↓ OB↓ GH↑ GH/OB↑ GPR39↓ EGFR↑
pERK1/2↑ GSH-PX↑ Stomach injury↓ SOD↑ MDA↓
Ileum injury↓
Kim et al.
2019
[78]
C57BL/6 miceMultiple
(3 times—
D1, 6, and 11)
5 mg/kg,
i.p.
HemoHIM
[Atractylodis Rhizoma Alba, Angelica Gigas Nakai, Astragali Radix]
(water extract)
100, 250, 500 mg/kg, p.o.
Cisplatin (n = 7)--NK cell activity↓
Macrophage phagocytotic activity↓CD4+ T lymphocytes↓
Splenocytes↓ IL-2↓ IFN-γ↓
Cisplatin + HemoHIM (n = 7)--NK cell activity↑
Macrophage phagocytotic activity↑CD4+ T lymphocytes↑
Splenocytes↑ IL-2↑ IFN-γ↑
Chen et al.
2019
[79]
C57BL/6 miceMultiple
(9 times—
3 days/week)
5 mg/kg,
i.p.
Zhen-Qi Sijunzi [ZQ-SJZ; Ginseng Radix, Atractylodis Rhizoma Alba, Poria Sclerotium, Glycyrrhizae Preparata, Hedysari Radix, Fructus Ligustri Lucidi]
(water extract)
700 mg/kg, p.o.
Cisplatin (n = 18)-Intestinal mucosal damage↑
Cisplatin + ZQ-SJZ (n = 18)-Intestinal mucosal damage↓
Goswami
et al.
2019
[80]
C57BL/6 miceMultiple
(D0 and 4)
8 mg/kg,
i.p.
Ninjin-yoeito
[NYT; Angelicae Acutilobae Radix, Atractylodis Rhizoma Alba, Rehmanniae Radix, Poriae Cutis, Ginseng Radix, Citri Unshius Pericarpium Immaturus, Polygalae Radix, Paeoniae Radix, Astragali Radix, Schisandrae Fructus, Glycyrrhizae Radix et Rhizoma]
(water extract)
1 g/kg, p.o.
Cisplatin (n = 6)--
Cisplatin + NYT (n = 6)-IR-positive NPY neurons [Ca2+]i
Ghrelin-responsive and unresponsive NPY neurons [Ca2+]i
Abbreviations: 5-HIAA, 5-Hydroxyindole acetic acid; 5-HT, 5-Hydroxytryptamine (serotonin); EGFR, epidermal growth factor receptor; GH, ghrelin; GHS-R1a, growth hormone secretagogue receptor type 1a; GPR39, G-protein-coupled receptor 39; GSH-PX, glutathione peroxidase; HMF, 3,3′,4′,5,6,7,8-heptamethoxyflavone; IL, interleukin; IFN-γ, interferon-γ; IR, immunoreactive; Ki, inhibition constant; MAO-A, monoamine oxidase A; MDA, malonaldehyde; NK cell, natural killer cell; NPY, neuropeptide Y; OB, obestatin; RKT, Rikkunshito; RVX, Rhus verniciflua stoke; SbE, Scutellaria baicalensis extract; SERT, serotonin reuptake transporter; SOD, superoxide dismutase; TNF-α, tumor necrosis factor-α; TPH, tryptophan hydroxylase; WBC, white blood cells.
Table 2. Underlying Mechanisms of action of herbal medicine extracts in cisplatin-induced anorexia.
Table 2. Underlying Mechanisms of action of herbal medicine extracts in cisplatin-induced anorexia.
Pathways CisplatinHerbal MedicinesMeasured Locations
5-HT5-HT and
5-HT3A receptor
RVX [47]Small intestine
HWKL [48]Ileum, medulla oblongata, serum
TPH1HWKL [57]Ileum
TPH2Medulla oblongata
SERTRVX [47]Small intestine
HWKL [48]Medulla oblongata, ileum
5 HIAAIleum, serum
CytokineIL-6SJDBT [43], LA16001 [44] Fat, serum
RVX [47]Stomach
IL-1β
TNF-α
HemoHIM [78]Spleen
IL-2
IFN-γ
IL-4
WBCTotal NumberKG [77]Serum
RVX [47], LA16001 [44]
LymphocytesKG [77]
RVX [47]
HemoHIM [78]Spleen
NeutrophilsKG [77]Serum
LA16001 [44]
NK cell activityHemoHIM [78]
Macrophage
Phagocytotic
activity
Peritoneal cavity
Splenocyte
proliferation
Spleen
HormoneGhrelinRKT [45], HWKL [48]Serum
LA16001 [44]Stomach
HWKL [48]Antrum
GHS-R1aRKT [45]Hypothalamus
LeptinSJDBT [43]Fat, serum
LA16001 [44]Fat, hypothalamus
Abbreviations: ↑, increase; ↓, decrease.
Woo et al. [43] identified that 14 consecutive days of oral administration of SJDBT, an herbal extract mixture consisting of 10 herbal medicines, significantly increased plasma IL-6 levels, which had been decreased by cisplatin. Single SJDBT administration also markedly increased the levels of IL-6 in fat and serum. The authors reported that Janus kinase 1 (JAK1) and signal transducer and activator of transcription 3 (STAT3) mediate the signaling pathway to produce IL-6. Similarly, in another study conducted by Woo et al. [44], LA16001 significantly increased IL-6 levels both in fat and serum, although cisplatin only lowered the IL-6 level in the serum but not in the fat tissue of mice. LA16001 is an herbal-mixture extract composed of six medicinal herbs, with three herbal medicines (Atractylodis Rhizoma Alba, Angelica Gigas Nakai, and Astragali Radix) present in both SJDBT and LA16001. As SJDBT, LA16001 also affected JAK1 and STAT3 pathways, inducing phosphorylation of JAK1 and STAT3 and increasing IL-6.
However, in contrast to the results obtained by Woo et al., Kim et al. [47] reported that IL-6 increased after cisplatin injection, and attenuation of anorexia is related to a decrease in IL-6 in the small intestine of rats. In their study, oral administration of water extract of RVX significantly attenuated abnormal elevations of IL-6 in the stomach, along with other proinflammatory cytokines, such as TNF-α and IL-1β, following cisplatin administration.
In a study by Kim et al. [78], multiple cisplatin injections significantly decreased the secretion of interferon-γ (IFN-γ) and interleukin-2 (IL-2) and increased interleukin-4 (IL-4) levels in splenocytes obtained from cisplatin-treated mice. Oral administration of HemoHIM, a hot-water extract of Angelica Gigas Nakai, Cnidii Rhizoma, and Paeoniae Radix, upregulated IFN-γ and IL-2 and downregulated IL-4 levels in splenocytes in a dose-dependent manner. In their study, the change in the level of TNF-α after cisplatin injection was not significantly different from that of the control. The authors further assessed that HemoHIM could restore splenocyte proliferation to the control level. Given that IFN-γ and IL-2 are Th1-associated cytokines, whereas IL-4 is a Th2-associated cytokine, the authors assessed that HemoHIM could modulate Th1-/Th2-mediated immune responses in cisplatin-treated mice.

2.3. White Blood Cells (WBCs)

Due to malnutrition, hematological complications are often observed in patients with anorexia [81]. Changes in the total number of WBCs characterized by lymphocytopenia or neutropenia are often present [81,82]. However, the changes in hematological parameters in cisplatin-induced anorexia have not yet been clearly elucidated. Four experiments analyzed the total number of WBCs, neutrophils, lymphocytes, and monocytes after injection of cisplatin and herbal medicines [44,47,49,50].
Raghavendran et al. [77] showed that 72 h after intraperitoneal injection of cisplatin (7 mg/kg), the number of WBCs, such as neutrophils, lymphocytes, and monocytes, significantly increased compared to the control. However, oral administration of 50 mg/kg of Korean ginseng root extract (KG) 1 h before cisplatin treatment prevented the increase in hematological parameters. Nevertheless, when KG was treated 2 h after injection of 6 mg/kg of cisplatin, its modulatory effects disappeared.
In contrast to the study by Raghavendran et al., Woo et al. [44] reported that the number of WBCs and neutrophil decreased when measured 3 days after the injection of 8 mg/kg of cisplatin in mice. LA16001 was administered for three consecutive days, and the number of WBCs and neutrophils in the blood significantly increased. In accordance with the study of Woo et al., Kim et al. [47] reported that 6 mg/kg of cisplatin lowered the number of WBCs and lymphocytes. RVX (100 mg/kg) significantly increased the number of WBCs and lymphocytes. However, the number of neutrophils remained unchanged after cisplatin injection.
In a study by Kim et al. [78], changes in the ratio of CD4+/CD8+ T lymphocytes, activities of natural killer (NK) cells, and phagocytosis activity of macrophages were assessed. The ratio of CD4+/CD8+ T lymphocytes and the number of CD4+ T lymphocytes are known to determine immunological ability [83]. Although the ratio of CD4+/CD8+ T lymphocytes was not affected by injection with either cisplatin or herbal medicine, CD4+ T lymphocytes significantly increased in splenocytes after medium and high doses of HemoHIM oral treatment. These changes were not observed in the blood. Furthermore, the suppressed activity of NK cells was alleviated after HemoHIM treatment at the highest dose. Moreover, the phagocytic activity of macrophages that initiate the innate immune response decreased significantly following cisplatin injection, whereas HemoHIM treatment significantly alleviated the phagocytic activities of macrophages. Altogether, these results suggest that HemoHIM could restore and enhance immune-cell activity suppressed by chemotherapy treatment.

2.4. Ghrelin

Ghrelin is an endogenous ligand of the growth hormone secretagogue receptor 1 (GHS-R1), which consists of 28 amino acids [84]. It is mainly secreted from the stomach, and upon secretion, it increases appetite [85,86,87]. Two major forms of ghrelin are found in the stomach and plasma: acylated ghrelin and desacylated ghrelin [88]. Acylated ghrelin is involved in the regulation of food intake, gastrointestinal motility, and energy expenditure, whereas desacylated ghrelin, although controversial, has been reported to be devoid of the biological activities of acylated ghrelin [84]. When ghrelin is administered peripherally or intracerebroventricularly (ICV) in rodents, it improves gastrointestinal motility and food intake [89,90,91]. In contrast, ICV injection of [D-Lys-3]-GHRP-6, a GHS-R1 antagonist, inhibited food intake increased by ghrelin [59]. Changes in plasma ghrelin levels have been reported in functional dyspepsia, chronic gastritis, and gastric ulcers, suggesting that ghrelin levels are closely associated with gastrointestinal disorders [92,93]. In total, five studies analyzed the modulatory effects of herbal medicines on ghrelin levels [44,45,46,48,80].
First, Takeda et al. [45] showed that both plasma-acylated- and -desacylated-ghrelin levels significantly decreased after cisplatin treatment. They also demonstrated that 5-HT2B and 5-HT2C agonists could decrease plasma-acylated-ghrelin levels in rats. Moreover, by demonstrating that cisplatin could significantly decrease acylated-ghrelin levels in vagotomized rats, the authors reported that the action of cisplatin primarily occurs in the peripheral tissues. As mentioned in the 5-HT section, RKT oral treatment succeeded in preventing a cisplatin-induced decrease in plasma-acylated-ghrelin levels when measured 2 h after its administration. They also identified that HMF, hesperidin, and isoliquiritigenin, which are components of RKT, significantly inhibited the cisplatin-induced decrease in plasma-acylated-ghrelin levels.
In a study by Yakabi et al. [46], cisplatin significantly inhibited relative mRNA levels of GHS-R1a in the hypothalamus but not in the stomach 6 h after the treatment. However, when ghrelin was injected (ICV) into cisplatin-injected rats, no significant changes in food intake were observed compared to saline-treated rats, owing to decreased GHS-R1a in the hypothalamus.
Both Woo et al. [44] and Song et al. [48] showed that herbal medicines could increase ghrelin content in the stomach. Woo et al. showed that oral administration of LA16001 significantly increased acylated-ghrelin levels in the stomach and food intake in cisplatin-injected mice. Song et al. [48] also confirmed the effect of HWKL, as it significantly increased the ghrelin content and the ratio of ghrelin/obestatin in the serum and stomach.
Goswami et al. [80] demonstrated that oral treatment with 1 mg/kg/day of ninjin’yoeito (NYT) for 10 days could increase both food intake and body weight in cisplatin-treated mice. To further analyze the mechanism of action of NYT, they focused on first-order neurons of the arcuate nucleus (ARC), which are known to receive feeding-related signals from the periphery [94]. NYT (10 μg/mL) administration for 10 min increased cytosolic Ca2+ concentration ([Ca2+]i) in a single neuron isolated from ARC, showing that NYT could modulate the action of ARC neurons. Furthermore, both NYT and ghrelin (10−8 M) increased [Ca2+]i in ARC neurons immunoreactive to neuropeptide Y (NPY-IR). NPY neurons are reported to play a critical role in feeding regulation [95], as their deletion results in reduced feeding and body weight in adult mice [96,97]. In addition, NYT increased [Ca2+]i in ghrelin-responsive and -unresponsive NPY neurons. Among 81 single ARC NPY-IR neurons, eight neurons (9.9%) responded to NYT, only 30 (37.0%) responded to ghrelin only, 18 (22.2%) responded to both, and 25 (30.9%) responded to none. Among 26 NPY neurons that responded to NYT, 18 (69.2%) responded to ghrelin, indicating that NYT activates both ghrelin-responsive (69.2%) and ghrelin-unresponsive (30.8%) NPY neurons.

2.5. Leptin

Leptin is a hormone secreted by fat cells that is known to reduce body fat and increase energy expenditure. Since its discovery in 1994, leptin has gained much attention in feeding regulations [98]. Although its role in chemotherapy-induced anorexia remains unclear, it is seen as an indicator of the severity of the disorder, as serum leptin levels have been reported to be low in anorexia-induced humans and rodents [16,99].
In a study by Woo et al. [43], 8 mg/kg of cisplatin significantly reduced serum and fat leptin levels in mice. However, multiple SJDBT administration for 14 days, starting from three days after cisplatin injection, significantly increased both serum and fat leptin levels. Single SJDBT administration also succeeded in ameliorating leptin levels in fat and serum. The authors further suggested that SJDBT administration could increase leptin by activating the JAK1/STAT3-mediated signaling pathway in adipocytes, as demonstrated by Western blotting in fat tissues. Furthermore, in their next study, Woo et al. [44] showed that cisplatin injection reduced leptin in fat but not in the hypothalamus 4 h after its injection in mice. Although leptin levels were not altered in the hypothalamus, LA16001 significantly increased leptin levels in adipocytes.

2.6. Most Used Herbal Medicines

Ginseng Radix was mentioned in seven studies [43,45,46,48,50,80,81], and both Glycyrrhizae Radix et Rhizoma [45,46,48,80] and Angelicae Gigantis Radix [43,44,49,81] were included in four herbal medicine extracts. Pinelliae Tuber [45,46,48], Zingiberis Rhizoma [45,46,48], Astragali Radix [43,44,81], and Paeoniae Radix [43,49,81] were used in three studies each. Scutellariae Radix [48,75], Cinnamomi Cortex [43,80], and Rehmanniae Radix [43,80] were only mentioned in two studies each.

3. Discussion

In this review, we examined the effects of herbal medicines in an animal model of cisplatin-induced anorexia. A total of 12 papers were included. Eight studies used a mixture of various herbal medicine extracts [43,44,45,46,48,78,79,80], and four used single herbal medicine extracts [47,50,78,79]. To our knowledge, this is the first review to summarize and analyze the effects of herbal medicines in an animal model of cisplatin-induced feeding disorder.
With the increase in the number of cancer patients, cisplatin has been cited as one of the most used anticancer medications due to its broad efficacy in the treatment of cancers [100]. However, anorexia is a serious dose-limiting side effect caused by cisplatin that could decrease survival [101], as well as quality of life in cancer patients [102]. Thus, it is critical to find new approaches for drug development, as the number of drug options is limited. In the included studies, cisplatin was administered to rodents to mimic feeding disorders induced in clinical settings. Most studies used a single cisplatin injection; however, in two studies, cisplatin was injected multiple times (3 and 15 times) [78,79]. In addition, the doses used were also different for each experiment, with the lowest dose being 2 mg/kg [45,46] and the highest dose being 8 mg/kg [43,44,81] (Table 1). The therapeutic range for cisplatin in humans (60 kg) is 35 mg/m2, which corresponds to approximately 5 mg/kg in rodents [103].
As for the pathogenesis mechanism of anorexia, changes in 5-HT, inflammatory cytokines, WBCs, ghrelin, and leptin were analyzed in the studies (Table 2 and Figure 1).
When the level of 5-HT is increased, it is known to induce a feeling of satiety and reduced appetite [104,105,106]. There are reports confirming that plasma 5-HT levels are elevated in cancer patients [107,108,109] and in the small intestine of animals [110,111] after cisplatin treatment. In accordance with these results, in the included studies, 5-HT was shown to be increased after cisplatin injection in the small intestine, serum, and brain, whereas the administration of herbal medicine (RVX and HWKL) succeeded in reducing 5-HT levels and ameliorating food intake in mice [47,48]. Along with 5-HT, 5-HT receptors are also closely involved in anorexia. Takeda et al. [45] and Yakabi et al. [46] suggested that the activation of 5-HT2B and 5-HT2C receptors may induce anorexia, as injection of their antagonist prevented a decrease in food intake. The activities of 5-HT2B and 5-HT2C receptors have also been related to loss of appetite and decreased food intake in other studies [112,113,114]. Although 5-HT2B receptors are mostly peripherally distributed, such as in the gastrointestinal tract, stomach fundus, and blood vessels, 5-HT2C receptors are known to exist centrally, mainly in the brain. 5-HT2C receptor stimulation by administration of selective agonists is known to induce a marked decrease in food intake, and knockout of the 5-HT2C receptor subtype in mice caused leptin-independent hyperphagia and hypoactivity, leading to obesity [115]. Contrary to 5-HT2B and 5-HT2C receptors, the role of 5-HT3 receptors in anorexia is still controversial. 5-HT3 receptors have been considered to play an important role in GI tract diseases, and antagonists such as ondansetron have been used for the prevention and treatment of vomiting [116,117]. However, in the included studies, subcutaneous administration of ondansetron, a 5-HT3 receptor antagonist, had no effect on the decrease in plasma-ghrelin concentration and food intake caused by cisplatin, suggesting that 5-HT3 receptors may not be closely involved in anorexia [45]. Furthermore, although Kim et al. [47] and Song et al. [48] showed a change in 5-HT3A receptor mRNA expression in the mouse brain and small intestine after cisplatin treatment, ondansetron failed to increase food consumption, suggesting that modulating the activity of 5-HT3A receptors may not be effective for the treatment of anorexia.
Regarding the change in inflammatory cytokines after cisplatin injection, TNF-α, IL-1β, and IL-6 increased in the stomach tissue when measured 5 days after a dose of 6 mg/kg of cisplatin [47]. In splenocytes, IL-2 and IFN-γ levels decreased, while IL-4 levels increased. TNF-α levels remained unchanged 14 days after multiple cisplatin injections [78]. In the studies by Woo et al. [43,44], IL-6 slightly decreased only in the serum when measured 4 h after 8 mg/kg of cisplatin treatment but not when measured 14 days after cisplatin injection in the serum or fat. Among the treatments, RVX significantly attenuated the increased TNF-α, IL-1β, and IL-6 levels in the stomach [47], and HemoHIM increased IL-2 and IFN-γ levels [78]. Both SJDBT [43] and LA16001 [44] significantly increased IL-6 levels in serum and fat tissue. RVX, SJDBT, and LA16001 all succeeded in decreasing anorexia in rodents, but their effects on IL-6 were different. The pathogenesis of inflammation-associated anorexia induced by cisplatin is unclear, and the role of IL-6 needs further investigation, as IL-6 was also shown to be highly elevated in both anorexia and obese patients [118].
The total WBC count was also different, according to previous studies. WBC decreased after cisplatin injection and increased after herbal medicines in two studies [44,47], whereas in the study by Raghavendran et al. [77] WBC increased after cisplatin treatment and decreased after KG administration. The difference in the WBC count (k/μL) after cisplatin injection may be due to the difference in the measurement time (48 h [44], 72 h [77], or 120 h [47]), animals used (Balb/c mice [44] or SD rats [78]), or the dose used (6 mg/kg [47], 7 mg/kg [77], or 8 mg/kg [44]).
The most frequently used herb in the 12 papers was Ginseng Radix, which was included in six herbal mixtures [43,45,46,50,80,81]. The components contributing to the therapeutic effect of ginseng radix on various diseases have been well studied, and it has been proven that Ginseng Radix has anticancer, antioxidant, and anti-inflammatory effects [119,120,121]. In particular, the anti-inflammatory effect of Ginseng Radix may have been effective in treating anorexia. For example, it has been reported that ginseng, a polysaccharide extracted from Ginseng Radix, inhibits the p38 MAP kinase pathway and NF-κB in vitro and inhibits proinflammatory cytokines in vivo [122]. Ginsenoside Rg3, another component of Ginseng Radix, was shown to inhibit phorbol-ester-induced COX-2 and NF-κB induction [123].
Angelicae Gigantis Radix was also included in four studies [43,44,49,81], making it the second most used medicinal herb among the reviewed studies. Angelicae Gigantis Radix attenuated scratching behavior, and 2,4-dinitrochlorobenzene-induced atopic dermatitis-like skin lesions by reducing serum IgE, histamine, TNF-α, IL-6, and COX-2 expression in skin tissue from mouse models [124]. Atractylodis Rhizoma Alba is a medicinal herb that was used in three of the papers reviewed [43,44,81]. Atractylon, which is one of the components of Atractylodis Rhizoma Alba, significantly inhibited NO and prostaglandin E2 production, as well as inducible NO synthase and cyclooxygenase-2 expression, in LPS-induced RAW 264.7 cells [125].
For future studies, it would be valuable to observe the effect of herbal-mixture extracts that have Ginseng Radix, Angelicae Gigantis Radix, or Atractylodis Rhizoma as their components. As a candidate, Bojungikki-tang could be considered, as it is a mixture of eight herbal extracts, including Ginseng Radix, Angelicae Gigantis Radix, and Atractylodis Rhizoma. Moreover, it has been widely used in the past to improve problems associated with digestive disease, such as decreased gastrointestinal motility and gastric injury [126,127]. In addition, ban-xia-xie-xin-tang and dai-kenchu-to could also be effective against cisplatin-induced anorexia, as they both have Ginseng Radix as a component. Ban-xia-xie-xin-tang is composed of seven herbs and has been reported to be effective for the treatment of various digestive inflammations, such as colitis, esophagitis, and gastritis [39,128]. In addition, dai-kenchu-to has been reported to increase blood flow in the intestinal tract [129], stimulate intestinal motility [130,131], and prevent bacterial translocation [132].
In conclusion, based on the results obtained from all included studies, we suggest that herbal medicines could be considered as an effective treatment method for cisplatin-induced anorexia. However, more well-designed clinical trials and experimental studies should be conducted to clarify their effect and to increase the understanding of the mechanisms of action. We believe that this review will help other researchers in the field of herbal medicines and cisplatin-induced anorexia to better understand its mechanism in the future.

4. Methods

A search was conducted of all studies on herbal medicines and cisplatin-induced anorexia in the National Library of Medicine (MEDLINE) using PubMed, and Google Scholar (Figure 2). Extensive searches were undertaken for articles written in English, as non-English studies were excluded. Studies electronically published until the end of September 2021 were included. The literature search was performed using the following keywords: “anorexia”, “cisplatin”, “feeding disorder”, and “herbal medicines”. After the initial search, duplicates, bibliographies, study protocols, clinical trials, and non-English studies were excluded. Twelve animal studies were included in this study.

Author Contributions

Conceptualization, W.K.; methodology, W.K.; formal analysis, D.M.; investigation, D.M.; data curation, D.M.; writing—original draft preparation, D.M. and W.K.; writing—review and editing, B.K., S.-G.K. and W.K.; project administration, W.K.; funding acquisition, S.-G.K. and W.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) No. 2020R1A5A2019413 and No. 2020R1F1A1070512.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dasari, S.; Bernard Tchounwou, P. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Ghosh, S. Cisplatin: The first metal based anticancer drug. Bioorganic Chem. 2019, 88, 102925. [Google Scholar] [CrossRef] [PubMed]
  3. Ho, G.Y.; Woodward, N.; Coward, J.I.G. Cisplatin versus carboplatin: Comparative review of therapeutic management in solid malignancies. Crit. Rev. Oncol. /Hematol. 2016, 102, 37–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Florea, A.-M.; Büsselberg, D. Cisplatin as an Anti-Tumor Drug: Cellular Mechanisms of Activity, Drug Resistance and Induced Side Effects. Cancers 2011, 3, 1351–1371. [Google Scholar] [CrossRef] [PubMed]
  5. Astolfi, L.; Ghiselli, S.; Guaran, V.; Chicca, M.; Simoni, E.; Olivetto, E.; Lelli, G.; Martini, A. Correlation of adverse effects of cisplatin administration in patients affected by solid tumours: A retrospective evaluation. Oncol. Rep. 2013, 29, 1285–1292. [Google Scholar] [CrossRef] [Green Version]
  6. Oun, R.; Moussa, Y.E.; Wheate, N.J. The side effects of platinum-based chemotherapy drugs: A review for chemists. Dalton Trans. 2018, 47, 6645–6653. [Google Scholar] [CrossRef]
  7. Shahid, F.; Farooqui, Z.; Khan, F. Cisplatin-induced gastrointestinal toxicity: An update on possible mechanisms and on available gastroprotective strategies. Eur. J. Pharmacol. 2018, 827, 49–57. [Google Scholar] [CrossRef]
  8. Le-Rademacher, J.G.; Crawford, J.; Evans, W.J.; Jatoi, A. Overcoming obstacles in the design of cancer anorexia/weight loss trials. Crit. Rev. Oncol. /Hematol. 2017, 117, 30–37. [Google Scholar] [CrossRef]
  9. Bearcroft, C.P.; Domizio, P.; Mourad, F.H.; André, E.A.; Farthing, M.J.G. Cisplatin impairs fluid and electrolyte absorption in rat small intestine: A role for 5-hydroxytryptamine. Gut 1999, 44, 174. [Google Scholar] [CrossRef] [Green Version]
  10. Song, M.Y.; Ku, S.K.; Kim, H.J.; Han, J.S. Low molecular weight fucoidan ameliorating the chronic cisplatin-induced delayed gastrointestinal motility in rats. Food Chem. Toxicol. 2012, 50, 4468–4478. [Google Scholar] [CrossRef]
  11. Vera, G.; Chiarlone, A.; Martín, M.I.; Abalo, R. Altered feeding behaviour induced by long-term cisplatin in rats. Auton. Neurosci. 2006, 126-127, 81–92. [Google Scholar] [CrossRef] [PubMed]
  12. Churm, D.; Andrew, I.M.; Holden, K.; Hildreth, A.J.; Hawkins, C. A questionnaire study of the approach to the anorexia-cachexia syndrome in patients with cancer by staff in a district general hospital. Supportive Care Cancer Off. J. Multinatl. Assoc. Supportive Care Cancer 2009, 17, 503–507. [Google Scholar] [CrossRef] [PubMed]
  13. Mitchell, E.P. Gastrointestinal Toxicity of Chemotherapeutic Agents. Semin. Oncol. 2006, 33, 106–120. [Google Scholar] [CrossRef] [PubMed]
  14. Ohnuma, T.; Holland, J.F. Nutritional Consequences of Cancer Chemotherapy and Immunotherapy. Cancer Res. 1977, 37, 2395. [Google Scholar]
  15. Kokal, W.A. The impact of antitumor therapy on nutrition. Cancer 1985, 55, 273–278. [Google Scholar] [CrossRef]
  16. Hattori, T.; Yakabi, K.; Takeda, H. Cisplatin-induced anorexia and ghrelin. Vitam. Horm. 2013, 92, 301–317. [Google Scholar]
  17. Van Cutsem, E.; Arends, J. The causes and consequences of cancer-associated malnutrition. Eur. J. Oncol. Nurs. 2005, 9, S51–S63. [Google Scholar] [CrossRef]
  18. Sinno, M.H.; Coquerel, Q.; Boukhettala, N.; Coëffier, M.; Gallas, S.; Terashi, M.; Ibrahim, A.; Breuillé, D.; Déchelotte, P.; Fetissov, S.O. Chemotherapy-induced anorexia is accompanied by activation of brain pathways signaling dehydration. Physiol. Behav. 2010, 101, 639–648. [Google Scholar] [CrossRef]
  19. Jahn, P.; Renz, P.; Stukenkemper, J.; Book, K.; Kuss, O.; Jordan, K.; Horn, I.; Thoke-Colberg, A.; Schmoll, H.-J.; Landenberger, M. Reduction of chemotherapy-induced anorexia, nausea, and emesis through a structured nursing intervention: A cluster-randomized multicenter trial. Supportive Care Cancer 2009. [CrossRef]
  20. Mantovani, G.; Macciò, A.; Massa, E.; Madeddu, C. Managing cancer-related anorexia/cachexia. Drugs 2001, 61, 499–514. [Google Scholar] [CrossRef]
  21. Fernández-Lucas, M.; Díaz-Domínguez, M.E.; Ruiz-Roso, G.; Raoch, V.; Teruel-Briones, J.L.; Quereda-Rodríguez-Navarro, C. Anorexia and megestrol acetate: Treatment versus placebo controlled study. Nefrol. (Engl. Ed.) 2014, 34, 416–417. [Google Scholar]
  22. Aoyagi, T.; Terracina, K.P.; Raza, A.; Matsubara, H.; Takabe, K. Cancer cachexia, mechanism and treatment. World J. Gastrointest Oncol. 2015, 7, 17–29. [Google Scholar] [CrossRef]
  23. Cuvelier, G.D.; Baker, T.J.; Peddie, E.F.; Casey, L.M.; Lambert, P.J.; Distefano, D.S.; Wardle, M.G.; Mychajlunow, B.A.; Romanick, M.A.; Dix, D.B.; et al. A randomized, double-blind, placebo-controlled clinical trial of megestrol acetate as an appetite stimulant in children with weight loss due to cancer and/or cancer therapy. Pediatric Blood Cancer 2014, 61, 672–679. [Google Scholar] [CrossRef]
  24. Schacter, L.; Rozencweig, M.; Canetta, R.; Kelley, S.; Nicaise, C.; Smaldone, L. Megestrol acetate: Clinical experience. Cancer Treat. Rev. 1989, 16, 49–63. [Google Scholar] [CrossRef]
  25. Harrison, M.E.; Norris, M.L.; Robinson, A.; Spettigue, W.; Morrissey, M.; Isserlin, L. Use of cyproheptadine to stimulate appetite and body weight gain: A systematic review. Appetite 2019, 137, 62–72. [Google Scholar] [CrossRef]
  26. Najib, K.; Moghtaderi, M.; Karamizadeh, Z.; Fallahzadeh, E. Beneficial effect of cyproheptadine on body mass index in undernourished children: A randomized controlled trial. Iran J. Pediatr. 2014, 24, 753–758. [Google Scholar] [PubMed]
  27. Ruiz Garcia, V.; López-Briz, E.; Carbonell Sanchis, R.; Gonzalvez Perales, J.L.; Bort-Marti, S. Megestrol acetate for treatment of anorexia-cachexia syndrome. Cochrane Database Syst. Rev. 2013, 2013. [Google Scholar] [CrossRef]
  28. Pascual López, A.; Roqué i Figuls, M.; Urrútia Cuchi, G.; Graciela Berenstein, E.; Almenar Pasies, B.; Balcells Alegre, M.; Herdman, M. Systematic review of megestrol acetate in the treatment of anorexia-cachexia syndrome. J. Pain Symptom Manag. 2004, 27, 360–369. [Google Scholar] [CrossRef]
  29. Krasaelap, A.; Madani, S. Cyproheptadine: A Potentially Effective Treatment for Functional Gastrointestinal Disorders in Children. Pediatric Ann. 2017, 46, e120–e125. [Google Scholar] [CrossRef] [Green Version]
  30. DiSaia, P.J.; Morrow, C.P. Unusual side effect of megestrol acetate. Am. J. Obstet. Gynecol. 1977, 129, 460–461. [Google Scholar] [CrossRef]
  31. Nelson, K.A.; Walsh, D.; Hussein, M. A phase II study of low-dose megestrol acetate using twice-daily dosing for anorexia in nonhormonally dependent cancer. Am. J. Hosp. Palliat. Care 2002, 19, 206–210. [Google Scholar] [CrossRef]
  32. von Mühlendahl, K.E.; Krienke, E.G. Toxicity of cyproheptadine. Side effects and accidental overdosage (author's transl). Mon. Fur Kinderheilkd. 1978, 126, 123–126. [Google Scholar]
  33. Wortsman, J.; Soler, N.G.; Hirschowitz, J. Side effects of cyproheptadine. Br. Med. J. 1978, 1, 1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hoa, T.P.; Duong, T.B.T.; Thi, H.D.N.; Anna, M.P.; Ali, H.E.; Gianfranco, P. Herbal Medicine for Slowing Aging and Aging-associated Conditions: Efficacy, Mechanisms and Safety. Curr. Vasc. Pharmacol. 2020, 18, 369–393. [Google Scholar]
  35. Ardalan, M.-R.; Rafieian-Kopaei, M. Is the safety of herbal medicines for kidneys under question? J. Nephropharmacol. 2013, 2, 11–12. [Google Scholar] [PubMed]
  36. Langmead, L.; Rampton, D.S. Review article: Herbal treatment in gastrointestinal and liver disease--benefits and dangers. Aliment. Pharmacol. Ther. 2001, 15, 1239–1252. [Google Scholar] [CrossRef]
  37. Bahmani, M.; Zargaran, A.; Rafieian-Kopaei, M. Identification of medicinal plants of Urmia for treatment of gastrointestinal disorders. Rev. Bras. De Farmacogn. 2014, 24, 468–480. [Google Scholar] [CrossRef] [Green Version]
  38. Anheyer, D.; Frawley, J.; Koch, A.K.; Lauche, R.; Langhorst, J.; Dobos, G.; Cramer, H. Herbal Medicines for Gastrointestinal Disorders in Children and Adolescents: A Systematic Review. Pediatrics 2017, 139, e20170062. [Google Scholar] [CrossRef] [Green Version]
  39. Yang, M.; Chen, J.; Xu, L.; Shi, X.; Zhou, X.; An, R. A Network Pharmacology Approach to Uncover the Molecular Mechanisms of Herbal Formula Ban-Xia-Xie-Xin-Tang. Evid.-Based Complementary Altern. Med. 2018, 2018, 4050714. [Google Scholar] [CrossRef]
  40. Jong, M.-S.; Hwang, S.-J.; Chen, Y.-C.; Chen, T.-J.; Chen, F.-J.; Chen, F.-P. Prescriptions of Chinese Herbal Medicine for Constipation Under the National Health Insurance in Taiwan. J. Chin. Med. Assoc. 2010, 73. [Google Scholar] [CrossRef] [Green Version]
  41. Jin, X.-L.; Shibata, C.; Naito, H.; Ueno, T.; Funayama, Y.; Fukushima, K.; Matsuno, S.; Sasakimd, I. Intraduodenal and Intrajejunal Administration of the Herbal Medicine, Dai-Kenchu-Tou, Stimulates Small Intestinal Motility via Cholinergic Receptors in Conscious Dogs. Dig. Dis. Sci. 2001, 46, 1171–1176. [Google Scholar] [CrossRef] [PubMed]
  42. Satoh, K.; Hayakawa, T.; Kase, Y.; Ishige, A.; Sasaki, H.; Nishikawa, S.; Kurosawa, S.; Yakabi, K.; Nakamura, T. Mechanisms for Contractile Effect of Dai-kenchu-to in Isolated Guinea Pig Ileum. Dig. Dis. Sci. 2001, 46, 250–256. [Google Scholar] [CrossRef]
  43. Woo, S.M.; Choi, Y.K.; Kim, A.J.; Yun, Y.J.; Shin, Y.C.; Cho, S.G.; Ko, S.G. Sip-jeon-dea-bo-tang, a traditional herbal medicine, ameliorates cisplatin-induced anorexia via the activation of JAK1/STAT3-mediated leptin and IL-6 production in the fat tissue of mice. Mol. Med. Rep. 2016, 13, 2967–2972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Woo, S.M.; Lee, K.M.; Lee, G.R.; Park, J.Y.; Lee, H.J.; Bahn, H.J.; Yoon, H.S.; Kim, J.Y.; Shin, Y.C.; Cho, S.G.; et al. Novel herbal medicine LA16001 ameliorates cisplatin-induced anorexia. Mol. Med. Rep. 2018, 17, 2665–2672. [Google Scholar] [CrossRef] [PubMed]
  45. Takeda, H.; Sadakane, C.; Hattori, T.; Katsurada, T.; Ohkawara, T.; Nagai, K.; Asaka, M. Rikkunshito, an herbal medicine, suppresses cisplatin-induced anorexia in rats via 5-HT2 receptor antagonism. Gastroenterology 2008, 134, 2004–2013. [Google Scholar] [CrossRef]
  46. Yakabi, K.; Kurosawa, S.; Tamai, M.; Yuzurihara, M.; Nahata, M.; Ohno, S.; Ro, S.; Kato, S.; Aoyama, T.; Sakurada, T.; et al. Rikkunshito and 5-HT2C receptor antagonist improve cisplatin-induced anorexia via hypothalamic ghrelin interaction. Regul. Pept. 2010, 161, 97–105. [Google Scholar] [CrossRef] [PubMed]
  47. Kim, H.S.; Kim, H.G.; Im, H.J.; Lee, J.S.; Lee, S.B.; Kim, W.Y.; Lee, H.W.; Lee, S.K.; Byun, C.K.; Son, C.G. Antiemetic and Myeloprotective Effects of Rhus verniciflua Stoke in a Cisplatin-Induced Rat Model. Evid.-Based Complementary Altern. Med. Ecam 2017, 2017, 9830342. [Google Scholar]
  48. Song, Z.H.; Chang, H.; Han, N.; Liu, Z.H.; Wang, Z.L.; Gao, H.; Yin, J. He-Wei granules inhibit chemotherapy-induced vomiting (CINV) in rats by reducing oxidative stress and regulating 5-HT, substance P, ghrelin and obestatin. Rsc. Adv. 2017, 7, 43866–43878. [Google Scholar] [CrossRef] [Green Version]
  49. Hasler, W.L. Serotonin and the GI tract. Curr. Gastroenterol. Rep. 2009, 11, 383–391. [Google Scholar] [CrossRef] [PubMed]
  50. Haleem, D.J. Serotonin neurotransmission in anorexia nervosa. Behav. Pharmacol. 2012, 23, 478–495. [Google Scholar] [CrossRef]
  51. Martin, A.M.; Young, R.L.; Leong, L.; Rogers, G.B.; Spencer, N.J.; Jessup, C.F.; Keating, D.J. The diverse metabolic roles of peripheral serotonin. Endocrinology 2017, 158, 1049–1063. [Google Scholar] [CrossRef]
  52. Simansky, K.J. Serotonergic control of the organization of feeding and satiety. Behav. Brain Res. 1995, 73, 37–42. [Google Scholar] [CrossRef]
  53. Jason, C.G.H.; Joanne, A.H.; Clare, L.L.; John, E.B. Serotonin (5-HT) Drugs: Effects on Appetite Expression and Use for the Treatment of Obesity. Curr. Drug Targets 2005, 6, 201–213. [Google Scholar]
  54. Samanin, R.; Mennini, T.; Ferraris, A.; Bendotti, C.; Borsini, F.; Garattini, S. m-Chlorophenylpiperazine: A central serotonin agonist causing powerful anorexia in rats. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1979, 308, 159–163. [Google Scholar] [CrossRef]
  55. Gershon, M. Serotonin receptors and transporters—roles in normal and abnormal gastrointestinal motility. Aliment. Pharmacol. Ther. 2004, 20, 3–14. [Google Scholar] [CrossRef] [PubMed]
  56. Lee, G.; Yang, E.; Kim, S.; Park, C.-S.; Park, Y.S.; Jin, Y.-H. Parapheromones suppress chemotherapy side effects. J. Pharmacol. Exp. Ther. 2018, 367, 215–221. [Google Scholar] [CrossRef] [PubMed]
  57. Jayamohananan, H.; Manoj Kumar, M.K.; T P, A. 5-HIAA as a Potential Biological Marker for Neurological and Psychiatric Disorders. Adv. Pharm. Bull. 2019, 9, 374–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Stenfors, C.; Ross, S.B. Changes in extracellular 5-HIAA concentrations as measured by in vivo microdialysis technique in relation to changes in 5-HT release. Psychopharmacology 2004, 172, 119–128. [Google Scholar] [CrossRef]
  59. Asakawa, A.; Inui, A.; Kaga, T.; Katsuura, G.; Fujimiya, M.; Fujino, M.A.; Kasuga, M. Antagonism of ghrelin receptor reduces food intake and body weight gain in mice. Gut 2003, 52, 947. [Google Scholar] [CrossRef] [Green Version]
  60. Karagiannides, I.; Torres, D.; Tseng, Y.-H.; Bowe, C.; Carvalho, E.; Espinoza, D.; Pothoulakis, C.; Kokkotou, E. Substance P as a novel anti-obesity target. Gastroenterology 2008, 134, 747–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Yamamoto, K.; Asano, K.; Tasaka, A.; Ogura, Y.; Kim, S.; Ito, Y.; Yamatodani, A. Involvement of substance P in the development of cisplatin-induced acute and delayed pica in rats. Br. J. Pharmacol. 2014, 171, 2888–2899. [Google Scholar] [CrossRef] [Green Version]
  62. Rojas, C.; Li, Y.; Zhang, J.; Stathis, M.; Alt, J.; Thomas, A.G.; Cantoreggi, S.; Sebastiani, S.; Pietra, C.; Slusher, B.S. The antiemetic 5-HT3 receptor antagonist Palonosetron inhibits substance P-mediated responses in vitro and in vivo. J. Pharmacol. Exp. Ther. 2010, 335, 362–368. [Google Scholar] [CrossRef] [Green Version]
  63. Gautron, L.; Layé, S. Neurobiology of inflammation-associated anorexia. Front Neurosci 2010, 3, 3. [Google Scholar] [CrossRef] [Green Version]
  64. Goehler, L.E.; Busch, C.R.; Tartaglia, N.; Relton, J.; Sisk, D.; Maier, S.F.; Watkins, L.R. Blockade of cytokine induced conditioned taste aversion by subdiaphragmatic vagotomy: Further evidence for vagal mediation of immune-brain communication. Neurosci. Lett. 1995, 185, 163–166. [Google Scholar] [CrossRef]
  65. Solmi, M.; Veronese, N.; Favaro, A.; Santonastaso, P.; Manzato, E.; Sergi, G.; Correll, C.U. Inflammatory cytokines and anorexia nervosa: A meta-analysis of cross-sectional and longitudinal studies. Psychoneuroendocrinology 2015, 51, 237–252. [Google Scholar] [CrossRef] [PubMed]
  66. Langhans, W.; Savoldelli, D.; Weingarten, S. Comparison of the feeding responses to bacterial lipopolysaccharide and interleukin-1β. Physiol. Behav. 1993, 53, 643–649. [Google Scholar] [CrossRef]
  67. Bretdibat, J.L.; Bluthe, R.M.; Kent, S.; Kelley, K.W.; Dantzer, R. Lipopolysaccharide and Interleukin-1 Depress Food-Motivated Behavior in Mice by a Vagal-Mediated Mechanism. Brain Behav. Immun. 1995, 9, 242–246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Layé, S.; Gheusi, G.; Cremona, S.; Combe, C.; Kelley, K.; Dantzer, R.; Parnet, P. Endogenous brain IL-1 mediates LPS-induced anorexia and hypothalamic cytokine expression. Am. J. Physiology. Regul. Integr. Comp. Physiol. 2000, 279, R93–R98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Porter, M.H.; Arnold, M.; Langhans, W. TNF-α tolerance blocks LPS-induced hypophagia but LPS tolerance fails to prevent TNF-α-induced hypophagia. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 1998, 274, R741–R745. [Google Scholar] [CrossRef]
  70. Porter, M.H.; Hrupka, B.J.; Altreuther, G.; Arnold, M.; Langhans, W. Inhibition of TNF-α production contributes to the attenuation of LPS-induced hypophagia by pentoxifylline. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 2000, 279, R2113–R2120. [Google Scholar] [CrossRef]
  71. Bodnar, R.J.; Pasternak, G.W.; Mann, P.E.; Paul, D.; Warren, R.; Donner, D.B. Mediation of Anorexia by Human Recombinant Tumor Necrosis Factor through a Peripheral Action in the Rat. Cancer Res. 1989, 49, 6280. [Google Scholar] [PubMed]
  72. Fattori, E.; Cappelletti, M.; Costa, P.; Sellitto, C.; Cantoni, L.; Carelli, M.; Faggioni, R.; Fantuzzi, G.; Ghezzi, P.; Poli, V. Defective inflammatory response in interleukin 6-deficient mice. J. Exp. Med. 1994, 180, 1243–1250. [Google Scholar] [CrossRef] [PubMed]
  73. Johnson, P.M.; Vogt, S.K.; Burney, M.W.; Muglia, L.J. COX-2 inhibition attenuates anorexia during systemic inflammation without impairing cytokine production. Am. J. Physiol.-Endocrinol. Metab. 2002, 282, E650–E656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Braun, T.P.; Marks, D.L. Pathophysiology and treatment of inflammatory anorexia in chronic disease. J. Cachexia Sarcopenia Muscle 2010, 1, 135–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Aung, H.H.; Dey, L.; Mehendale, S.; Xie, J.T.; Wu, J.A.; Yuan, C.S. Scutellaria baicalensis extract decreases cisplatin-induced pica in rats. Cancer Chemother. Pharmacol. 2003, 52, 453–458. [Google Scholar] [CrossRef]
  76. Mehendale, S.; Aung, H.; Wang, A.; Yin, J.J.; Wang, C.Z.; Xie, J.T.; Yuan, C.S. American ginseng berry extract and ginsenoside Re attenuate cisplatin-induced kaolin intake in rats. Cancer Chemother. Pharmacol. 2005, 56, 63–69. [Google Scholar] [CrossRef]
  77. Raghavendran, H.R.; Rekha, S.; Shin, J.W.; Kim, H.G.; Wang, J.H.; Park, H.J.; Choi, M.K.; Cho, J.H.; Son, C.G. Effects of Korean ginseng root extract on cisplatin-induced emesis in a rat-pica model. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2011, 49, 215–221. [Google Scholar] [CrossRef]
  78. Kim, S.K.; Kwon, D.A.; Lee, H.S.; Kim, H.K.; Kim, W.K. Preventive Effect of the Herbal Preparation, HemoHIM, on Cisplatin-Induced Immune Suppression. Evid.-Based Complementary Altern. Med. Ecam 2019, 2019, 3494806. [Google Scholar] [CrossRef] [Green Version]
  79. Chen, J.M.; Yang, T.T.; Cheng, T.S.; Hsiao, T.F.; Chang, P.M.H.; Leu, J.Y.; Wang, F.S.; Hsu, S.L.; Huang, C.Y.F.; Lai, J.M. Modified Sijunzi decoction can alleviate cisplatin-induced toxicity and prolong the survival time of cachectic mice by recovering muscle atrophy. J. Ethnopharmacol. 2019, 233, 47–55. [Google Scholar] [CrossRef]
  80. Goswami, C.; Dezaki, K.; Wang, L.; Inui, A.; Seino, Y.; Yada, T. Ninjin-yoeito activates ghrelin-responsive and unresponsive NPY neurons in the arcuate nucleus and counteracts cisplatin-induced anorexia. Neuropeptides 2019, 75, 58–64. [Google Scholar] [CrossRef]
  81. De Filippo, E.; Marra, M.; Alfinito, F.; Di Guglielmo, M.L.; Majorano, P.; Cerciello, G.; De Caprio, C.; Contaldo, F.; Pasanisi, F. Hematological complications in anorexia nervosa. Eur. J. Clin. Nutr. 2016, 70, 1305–1308. [Google Scholar] [CrossRef] [Green Version]
  82. Hütter, G.; Ganepola, S.; Hofmann, W.-K. The hematology of anorexia nervosa. Int. J. Eat. Disord. 2009, 42, 293–300. [Google Scholar] [CrossRef]
  83. Janssen, E.M.; Lemmens, E.E.; Wolfe, T.; Christen, U.; von Herrath, M.G.; Schoenberger, S.P. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature 2003, 421, 852–856. [Google Scholar] [CrossRef]
  84. Müller, T.D.; Nogueiras, R.; Andermann, M.L.; Andrews, Z.B.; Anker, S.D.; Argente, J.; Batterham, R.L.; Benoit, S.; Bowers, C.Y.; Broglio, F. Ghrelin. Mol. Metab. 2015, 4, 437–460. [Google Scholar] [CrossRef] [PubMed]
  85. Kojima, M.; Hosoda, H.; Date, Y.; Nakazato, M.; Matsuo, H.; Kangawa, K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 1999, 402, 656–660. [Google Scholar] [CrossRef]
  86. Cummings, D.E.; Shannon, M.H. Roles for Ghrelin in the Regulation of Appetite and Body Weight. Arch. Surg. 2003, 138, 389–396. [Google Scholar] [CrossRef] [Green Version]
  87. Wren, A.M.; Small, C.J.; Ward, H.L.; Murphy, K.G.; Dakin, C.L.; Taheri, S.; Kennedy, A.R.; Roberts, G.H.; Morgan, D.G.A.; Ghatei, M.A.; et al. The Novel Hypothalamic Peptide Ghrelin Stimulates Food Intake and Growth Hormone Secretion. Endocrinology 2000, 141, 4325–4328. [Google Scholar] [CrossRef] [PubMed]
  88. Hosoda, H.; Kojima, M.; Matsuo, H.; Kangawa, K. Ghrelin and des-acyl ghrelin: Two major forms of rat ghrelin peptide in gastrointestinal tissue. Biochem. Biophys. Res. Commun. 2000, 279, 909–913. [Google Scholar] [CrossRef]
  89. Tschöp, M.; Smiley, D.L.; Heiman, M.L. Ghrelin induces adiposity in rodents. Nature 2000, 407, 908–913. [Google Scholar] [CrossRef]
  90. Asakawa, A.; Inui, A.; Kaga, T.; Yuzuriha, H.; Nagata, T.; Ueno, N.; Makino, S.; Fujimiya, M.; Niijima, A.; Fujino, M.A.; et al. Ghrelin is an appetite-stimulatory signal from stomach with structural resemblance to motilin. Gastroenterology 2001, 120, 337–345. [Google Scholar] [CrossRef] [PubMed]
  91. Nakazato, M.; Murakami, N.; Date, Y.; Kojima, M.; Matsuo, H.; Kangawa, K.; Matsukura, S. A role for ghrelin in the central regulation of feeding. Nature 2001, 409, 194–198. [Google Scholar] [CrossRef] [PubMed]
  92. Shinomiya, T.; Fukunaga, M.; Akamizu, T.; Irako, T.; Yokode, M.; Kangawa, K.; Nakai, Y.; Nakai, Y. Plasma acylated ghrelin levels correlate with subjective symptoms of functional dyspepsia in female patients. Scand. J. Gastroenterol. 2005, 40, 648–653. [Google Scholar] [CrossRef]
  93. Isomoto, H.; Ueno, H.; Nishi, Y.; Yasutake, T.; Tanaka, K.; Kawano, N.; Ohnita, K.; Mizuta, Y.; Inoue, K.; Nakazato, M.; et al. Circulating Ghrelin Levels in Patients with Various Upper Gastrointestinal Diseases. Dig. Dis. Sci. 2005, 50, 833–838. [Google Scholar] [CrossRef]
  94. Schwartz, M.W.; Woods, S.C.; Porte, D.; Seeley, R.J.; Baskin, D.G. Central nervous system control of food intake. Nature 2000, 404, 661–671. [Google Scholar] [CrossRef]
  95. Kohno, D.; Nakata, M.; Maekawa, F.; Fujiwara, K.; Maejima, Y.; Kuramochi, M.; Shimazaki, T.; Okano, H.; Onaka, T.; Yada, T. Leptin suppresses ghrelin-induced activation of neuropeptide Y neurons in the arcuate nucleus via phosphatidylinositol 3-kinase-and phosphodiesterase 3-mediated pathway. Endocrinology 2007, 148, 2251–2263. [Google Scholar] [CrossRef]
  96. Gropp, E.; Shanabrough, M.; Borok, E.; Xu, A.W.; Janoschek, R.; Buch, T.; Plum, L.; Balthasar, N.; Hampel, B.; Waisman, A. Agouti-related peptide–expressing neurons are mandatory for feeding. Nat. Neurosci. 2005, 8, 1289–1291. [Google Scholar] [CrossRef]
  97. Luquet, S.; Perez, F.A.; Hnasko, T.S.; Palmiter, R.D. NPY/AgRP neurons are essential for feeding in adult mice but can be ablated in neonates. Science 2005, 310, 683–685. [Google Scholar] [CrossRef] [Green Version]
  98. Zhang, Y.; Proenca, R.; Maffei, M.; Barone, M.; Leopold, L.; Friedman, J.M. Positional cloning of the mouse obese gene and its human homologue. Nature 1994, 372, 425–432. [Google Scholar] [CrossRef]
  99. Hebebrand, J.; Muller, T.; Holtkamp, K.; Herpertz-Dahlmann, B. The role of leptin in anorexia nervosa: Clinical implications. Mol. Psychiatry 2007, 12, 23–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Torres, L.S. Cancer Drug Resistance Research Perspectives; Nova Publishers: New York, NY, USA, 2007. [Google Scholar]
  101. Garcia, J.M.; Scherer, T.; Chen, J.-a.; Guillory, B.; Nassif, A.; Papusha, V.; Smiechowska, J.; Asnicar, M.; Buettner, C.; Smith, R.G. Inhibition of cisplatin-induced lipid catabolism and weight loss by ghrelin in male mice. Endocrinology 2013, 154, 3118–3129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Yakabi, K.; Sadakane, C.; Noguchi, M.; Ohno, S.; Ro, S.; Chinen, K.; Aoyama, T.; Sakurada, T.; Takabayashi, H.; Hattori, T. Reduced Ghrelin Secretion in the Hypothalamus of Rats due to Cisplatin-Induced Anorexia. Endocrinology 2010, 151, 3773–3782. [Google Scholar] [CrossRef] [Green Version]
  103. Lin, M.-T.; Ko, J.-L.; Liu, T.-C.; Chao, P.-T.; Ou, C.-C. Protective effect of D-methionine on body weight loss, anorexia, and nephrotoxicity in cisplatin-induced chronic toxicity in rats. Integr. Cancer Ther. 2018, 17, 813–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yabut, J.M.; Crane, J.D.; Green, A.E.; Keating, D.J.; Khan, W.I.; Steinberg, G.R. Emerging Roles for Serotonin in Regulating Metabolism: New Implications for an Ancient Molecule. Endocr. Rev. 2019, 40, 1092–1107. [Google Scholar] [CrossRef] [PubMed]
  105. de Matos Feijó, F.; Bertoluci, M.C.; Reis, C. Serotonin and hypothalamic control of hunger: A review. Rev. Da Assoc. Médica Bras. (Engl. Ed.) 2011, 57, 74–77. [Google Scholar] [CrossRef]
  106. Halford, J.C.; Blundell, J.E. Separate systems for serotonin and leptin in appetite control. Ann. Med. 2000, 32, 222–232. [Google Scholar] [CrossRef] [PubMed]
  107. Barnes, N.M.; Ge, J.; Jones, W.G.; Naylor, R.J.; Rudd, J.A. Cisplatin induced emesis: Preliminary results indicative of changes in plasma levels of 5-hydroxytryptamine. Br. J. Cancer 1990, 62, 862–864. [Google Scholar] [CrossRef] [Green Version]
  108. Cubeddu, L.X.; Hoffmann, I.S.; Fuenmayor, N.T.; Finn, A.L. Efficacy of Ondansetron (Gr 38032F) and the Role of Serotonin in Cisplatin-Induced Nausea and Vomiting. N. Engl. J. Med. 1990, 322, 810–816. [Google Scholar] [CrossRef]
  109. Wilder-Smith, O.H.G.; Borgeat, A.; Chappuis, P.; Fathi, M.; Forni, M. Urinary serotonin metabolite excretion during cisplatin chemotherapy. Cancer 1993, 72, 2239–2241. [Google Scholar] [CrossRef]
  110. Schwörer, H.; Racké, K.; Kilbinger, H. Cisplatin increases the release of 5-hydroxytryptamine (5-HT) from the isolated vascularly perfused small intestine of the guinea-pig: Involvement of 5-HT3 receptors. Naunyn-Schmiedeberg’s Arch. Pharmacol. 1991, 344, 143–149. [Google Scholar] [CrossRef]
  111. Kamato, T.; Ito, H.; Nagakura, Y.; Nishida, A.; Yuki, H.; Yamano, M.; Miyata, K. Mechanisms of cisplatin- and m-chlorophenylbiguinide-induced emesis in ferrets. Eur. J. Pharmacol. 1993, 238, 369–376. [Google Scholar] [CrossRef]
  112. Murotani, T.; Ishizuka, T.; Isogawa, Y.; Karashima, M.; Yamatodani, A. Possible involvement of serotonin 5-HT2 receptor in the regulation of feeding behavior through the histaminergic system. Neuropharmacology 2011, 61, 228–233. [Google Scholar] [CrossRef] [PubMed]
  113. Banas, S.M.; Doly, S.; Boutourlinsky, K.; Diaz, S.L.; Belmer, A.; Callebert, J.; Collet, C.; Launay, J.-M.; Maroteaux, L. Deconstructing Antiobesity Compound Action: Requirement of Serotonin 5-HT2B Receptors for Dexfenfluramine Anorectic Effects. Neuropsychopharmacology 2011, 36, 423–433. [Google Scholar] [CrossRef] [PubMed]
  114. Nonogaki, K.; Abdallah, L.; Goulding, E.H.; Bonasera, S.J.; Tecott, L.H. Hyperactivity and Reduced Energy Cost of Physical Activity in Serotonin 5-HT2C Receptor Mutant Mice. Diabetes 2003, 52, 315. [Google Scholar]
  115. Nonogaki, K.; Strack, A.M.; Dallman, M.F.; Tecott, L.H. Leptin-independent hyperphagia and type 2 diabetes in mice with a mutated serotonin 5-HT 2C receptor gene. Nat. Med. 1998, 4, 1152–1156. [Google Scholar] [CrossRef] [PubMed]
  116. Freeman, A.; Cunningham, K.; Tyers, M. Selectivity of 5-HT3 receptor antagonists and anti-emetic mechanisms of action. Anti-Cancer Drugs 1992, 3, 79–85. [Google Scholar] [CrossRef]
  117. Hesketh, P.J. Clinical science review: Comparative review of 5-HT3 receptor antagonists in the treatment of acute chemotherapy-induced nausea and vomiting. Cancer Investig. 2000, 18, 163–173. [Google Scholar] [CrossRef]
  118. Raymond, N.C.; Dysken, M.; Bettin, K.; Eckert, E.D.; Crow, S.J.; Markus, K.; Pomeroy, C. Cytokine production in patients with anorexia nervosa, bulimia nervosa, and obesity. Int. J. Eat. Disord. 2000, 28, 293–302. [Google Scholar] [CrossRef]
  119. Kim, J.H.; Yi, Y.-S.; Kim, M.-Y.; Cho, J.Y. Role of ginsenosides, the main active components of Panax ginseng, in inflammatory responses and diseases. J. Ginseng Res. 2017, 41, 435–443. [Google Scholar] [CrossRef] [Green Version]
  120. Kim, H.G.; Yoo, S.R.; Park, H.J.; Lee, N.H.; Shin, J.W.; Sathyanath, R.; Cho, J.H.; Son, C.G. Antioxidant effects of Panax ginseng C.A. Meyer in healthy subjects: A randomized, placebo-controlled clinical trial. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2011, 49, 2229–2235. [Google Scholar] [CrossRef]
  121. Kiefer, D.; Pantuso, T. Panax ginseng. Am. Fam. Physician 2003, 68, 1539–1542. [Google Scholar]
  122. Ahn, J.-Y.; Choi, I.-S.; Shim, J.-Y.; Yun, E.-K.; Yun, Y.-S.; Jeong, G.; Song, J.-Y. The immunomodulator ginsan induces resistance to experimental sepsis by inhibiting Toll-like receptor-mediated inflammatory signals. Eur. J. Immunol. 2006, 36, 37–45. [Google Scholar] [CrossRef] [PubMed]
  123. Keum, Y.S.; Han, S.S.; Chun, K.S.; Park, K.K.; Park, J.H.; Lee, S.K.; Surh, Y.J. Inhibitory effects of the ginsenoside Rg3 on phorbol ester-induced cyclooxygenase-2 expression, NF-kappaB activation and tumor promotion. Mutat. Res. 2003, 523-524, 75–85. [Google Scholar] [CrossRef]
  124. Ok, S.; Oh, S.R.; Jung, T.S.; Jeon, S.O.; Jung, J.W.; Ryu, D.S. Effects of Angelica gigas Nakai as an Anti-Inflammatory Agent in In Vitro and In Vivo Atopic Dermatitis Models. Evid.-Based Complementary Altern. Med. Ecam 2018, 2018, 2450712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Chen, L.-G.; Jan, Y.-S.; Tsai, P.-W.; Norimoto, H.; Michihara, S.; Murayama, C.; Wang, C.-C. Anti-inflammatory and Antinociceptive Constituents of Atractylodes japonica Koidzumi. J. Agric. Food Chem. 2016, 64, 2254–2262. [Google Scholar] [CrossRef] [PubMed]
  126. Kwon, H.E.; Kim, J.N.; Kwon, M.J.; Lee, J.R.; Kim, S.C.; Nam, J.H.; Kim, B.J. The traditional medicine bojungikki-tang increases intestinal motility. Pharmacogn. Mag. 2021, 17, 1. [Google Scholar]
  127. Lee, M.-Y.; Shin, I.-S.; Jeon, W.-Y.; Seo, C.-S.; Ha, H.; Huh, J.-I.; Shin, H.-K. Protective effect of Bojungikki-tang, a traditional herbal formula, against alcohol-induced gastric injury in rats. J. Ethnopharmacol. 2012, 142, 346–353. [Google Scholar] [CrossRef] [PubMed]
  128. Chen, F.-p.; Chen, F.-j.; Jong, M.-s.; Tsai, H.-l.; Wang, J.-r.; Hwang, S.-j. Modern use of Chinese herbal formulae fromShang-Han Lun. Chin. Med. J. 2009, 122, 1889–1894. [Google Scholar]
  129. Murata, P.; Kase, Y.; Ishige, A.; Sasaki, H.; Kurosawa, S.; Nakamura, T. The herbal medicine Dai-kenchu-to and one of its active components [6]-shogaol increase intestinal blood flow in rats. Life Sci. 2002, 70, 2061–2070. [Google Scholar] [CrossRef] [Green Version]
  130. Shibata, C.; Sasaki, I.; Naito, H.; Ueno, T.; Matsuno, S. The herbal medicine Dai-Kenchu-Tou stimulates upper gut motility through cholinergic and 5-hydroxytryptamine 3 receptors in conscious dogs. Surgery 1999, 126, 918–924. [Google Scholar] [CrossRef]
  131. Kawasaki, N.; Nakada, K.; Suzuki, Y.; Furukawa, Y.; Hanyu, N.; Kashiwagi, H. Effect of Dai-kenchu-to on gastrointestinal motility and gastric emptying. Int. J. Surg. 2009, 7, 218–222. [Google Scholar] [CrossRef] [Green Version]
  132. Yoshikawa, K.; Kurita, N.; Higashijima, J.; Miyatani, T.; Miyamoto, H.; Nishioka, M.; Shimada, M. Kampo medicine “Dai-kenchu-to” prevents bacterial translocation in rats. Dig. Dis. Sci. 2008, 53, 1824–1831. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The pathogenesis mechanism of cisplatin-induced anorexia and the mechanisms of action of herbal extracts. Intraperitoneal administration of cisplatin induces anorexia (red), whereas administration herbal extract increases appetite (blue). The brain, serum, small intestine, stomach, and spleen were affected by both cisplatin and herbal-extract treatments. Abbreviation: GHS-R1a, growth hormone secretagogue receptor type 1a; GSH-PX, glutathione peroxidase; 5-HIAA, 5-Hydroxyindole acetic acid; HMF, 3,3′,4′,5,6,7,8-heptamethoxyflavone; 5-HT, 5-Hydroxytryptamine (serotonin); 5-HTR, 5-HT receptor; IFN-γ, interferon-γ; MAO-A, monoamine oxidase A; MDA, malonaldehyde; SERT, serotonin reuptake transporter; SOD, superoxide dismutase; TNF-α, tumor necrosis factor-α; TPH, tryptophan hydroxylase.
Figure 1. The pathogenesis mechanism of cisplatin-induced anorexia and the mechanisms of action of herbal extracts. Intraperitoneal administration of cisplatin induces anorexia (red), whereas administration herbal extract increases appetite (blue). The brain, serum, small intestine, stomach, and spleen were affected by both cisplatin and herbal-extract treatments. Abbreviation: GHS-R1a, growth hormone secretagogue receptor type 1a; GSH-PX, glutathione peroxidase; 5-HIAA, 5-Hydroxyindole acetic acid; HMF, 3,3′,4′,5,6,7,8-heptamethoxyflavone; 5-HT, 5-Hydroxytryptamine (serotonin); 5-HTR, 5-HT receptor; IFN-γ, interferon-γ; MAO-A, monoamine oxidase A; MDA, malonaldehyde; SERT, serotonin reuptake transporter; SOD, superoxide dismutase; TNF-α, tumor necrosis factor-α; TPH, tryptophan hydroxylase.
Pharmaceuticals 15 00208 g001
Figure 2. Flow chart of article inclusion protocol. Identification through searches of MEDLINE (PubMed) and Google Scholar yielded 1165 articles, which were screened by abstract and full-text examinations. Finally, a total of 12 articles assessing the effect of herbal extracts in cisplatin-induced anorexia in rodents were included in our review.
Figure 2. Flow chart of article inclusion protocol. Identification through searches of MEDLINE (PubMed) and Google Scholar yielded 1165 articles, which were screened by abstract and full-text examinations. Finally, a total of 12 articles assessing the effect of herbal extracts in cisplatin-induced anorexia in rodents were included in our review.
Pharmaceuticals 15 00208 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Min, D.; Kim, B.; Ko, S.-G.; Kim, W. Effect and Mechanism of Herbal Medicines on Cisplatin-Induced Anorexia. Pharmaceuticals 2022, 15, 208. https://doi.org/10.3390/ph15020208

AMA Style

Min D, Kim B, Ko S-G, Kim W. Effect and Mechanism of Herbal Medicines on Cisplatin-Induced Anorexia. Pharmaceuticals. 2022; 15(2):208. https://doi.org/10.3390/ph15020208

Chicago/Turabian Style

Min, Daeun, Bonglee Kim, Seong-Gyu Ko, and Woojin Kim. 2022. "Effect and Mechanism of Herbal Medicines on Cisplatin-Induced Anorexia" Pharmaceuticals 15, no. 2: 208. https://doi.org/10.3390/ph15020208

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop