ArticlePDF Available

Anti-tumor effects and mechanisms of Astragalus membranaceus (AM) and its specific immunopotentiation: Status and prospect

Authors:

Figures

Content may be subject to copyright.
Contents lists available at ScienceDirect
Journal of Ethnopharmacology
journal homepage: www.elsevier.com/locate/jethpharm
Anti-tumor eects and mechanisms of Astragalus membranaceus (AM) and its
specic immunopotentiation: Status and prospect
Shanshan Li
a,1
, Yi Sun
b,1
, Jin Huang
a
, Bin Wang
c
, Yinan Gong
a
, Yuxin Fang
a,d
, Yangyang Liu
a,d
,
Shenjun Wang
a,d
, Yi Guo
a,e
, Hong Wang
d
, Zhifang Xu
a,d,
, Yongming Guo
a,d,∗∗
a
Acupuncture Research Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
b
Nephropathy and Rheumatology Department, Second Aliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300250, China
c
Tianjin Medical University Cancer Institute of Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
d
Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
e
College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
ARTICLE INFO
Keywords:
Astragalus membranaceus
Tumor immune microenvironment
Host immunity
Organic immunity
Chemotherapy
ABSTRACT
With cancer deaths increasing, the initiation, pathophysiology and curative management of cancer is receiving
increasing attention. Traditional therapies such as surgery and chemoradiotherapy are often accompanied by
suppression of host immunity, which increase the risk of metastasis. Astragalus membranceus (AM) is commonly
utilized as one herbal medicine of traditional Chinese medicines (TCMs) with a variety of biological activities.
Studies have shown that the active ingredients of AM and AM-based TCMs, combined with chemotherapy, can
enhance anti-tumor ecacy in cancer patients, in addition to reduce complications and avoid side eects in-
duced by chemotherapy. By using various cancer models and cell lines, AM has been found to be capable of
shrinking or stabilizing tumors by direct anti-proliferation or pro-apoptosis eect on tumor cells. Further, AM
ameliorates immunosuppression by activating M1 macrophages and T cells tumor-kill function in tumor mi-
croenvironment (TME). AM is also found to improve systemic immunity which may help promoting ecacy of
chemotherapy and preventing metastasis. Thereby this review contributes to an understanding of AM as an
adjunctive therapy in the whole course of cancer treatment, at the same time providing useful information for
development of more eective anti-tumor medication. The combination of AM and immune checkpoint therapies
has a promising therapeutic prospect, and the observation of direct ecacy and mechanisms on tumor growth
and metastasis of AM combined with chemotherapies or other therapies require more in vivo validations and
further clinical investigation as well.
1. Introduction
With the rapid growth and aging of the global population, cancer
has become increasingly prominent as a leading cause of death (Bray
et al., 2018). It is predicted that the incidence of all cancer cases will
increase from 12.7 million new cases in 2008 to 22.2 million in 2030
(Bray et al., 2012). Therefore, cancer occurrence, pathophysiology and
therapeutic option development are receiving increased attention
worldwide, especially in low- and middle-income countries such as
China.
Traditional therapies such as surgery and chemoradiotherapy can
directly act on cancer cells while have several serious drawbacks.
Firstly, most cancer patients are diagnosed too late to perform surgery.
Even if there are surgical indications, a series of complications such as
bleeding, infection, lymphedema may occur after surgery. Secondly,
although chemoradiotherapy is still the main adjuvant therapy to sur-
gery or the preferred treatment for patients with advanced malignant
tumors, there are also many side eects and complications, such as
bone marrow suppression, impaired liver and kidney function, nausea
and vomiting or local radiotherapy damage. More importantly, several
regularly used chemotherapeutic drugs convert cancer cells into cancer
stem cells, thereby resulting in therapeutic resistance and accelerating
https://doi.org/10.1016/j.jep.2020.112797
Received 18 December 2019; Received in revised form 10 March 2020; Accepted 23 March 2020
Corresponding author. Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese medicine, No.10 Poyang Lake Road, Tuanbo new town,
Jinghai district, Tianjin, 301617, China.
∗∗
Corresponding author. Acu-moxibustion and Tuina Department, Tianjin University of Traditional Chinese medicine, No.10 Poyang Lake Road, Tuanbo new town,
Jinghai district, Tianjin, 301617, China.
E-mail addresses: xuzhifangmsn@hotmail.com (Z. Xu), guoymxr@163.com (Y. Guo).
1
Equal contribution to this work.
Journal of Ethnopharmacology 258 (2020) 112797
Available online 01 April 2020
0378-8741/ © 2020 Elsevier B.V. All rights reserved.
T
cancer cell metastasis by worsening host immunity (Martins-Neves
et al., 2016;Safa et al., 2015).
Accumulating evidence has conrmed that cancer cells reside in a
specialized microenvironment, or niche, namely the TME. Tumor cells
must recruit and reprogram the surrounding normal cells to serve as
contributors so that ensure their rapid proliferation, survival, local in-
vasion and remote metastasis (Casey et al., 2015). With tumor devel-
opment, there is a dynamic alteration on molecular and cellular pro-
cesses in TME involving the interactions between cancer cells and
immune cells. Given the eectiveness of T cells in mediating anti-tumor
immune responses, T cell-based immunotherapy is considered as an
important and promising therapeutic approach against cancer (Chen
and Mellman, 2017). However, the majority of patients treated with
immune monotherapies fail to achieve the desired therapeutic response.
The main reasons could be the severe immune-suppressive micro-
environment including impaired antigen presentation capability in
tumor sites that inhibit the proliferation, migration and survival of in-
ltrating T cells. How to improve the host immune status is therefore
very important in T cell-based immunotherapy (Nicolas-Boluda and
Donnadieu, 2019).
Astragalus membranaceus (AM, Huang qi in Chinese) is a plant belong
to the leguminous family. AM particularly its dry root Astragali Radix,is
a popular tonic in traditional Chinese medicine (TCM). It has been
found to have multiple biofunctions, such as immunomodulatory, anti-
hyperglycemic, anti-inammatory, anti-oxidant and anti-viral activities
(Shao et al., 2004). Modern pharmacological evidence has shown that
AM and its active ingredients have strong anti-tumor activity and en-
hance host immune function. Thus, this review will summarize the
capability of AM to reduce the side eects and complications and in-
crease the anti-tumor ecacy caused by chemotherapy in cancer pa-
tients. Meanwhile, the mechanism evidences that AM directly shrinks
focus or stabilizes cancer state, enhancing the organic immunity to
improve the ecacy of chemotherapy and prevent metastasis in a
variety of pathways will be accumulated, providing a systematic review
and evaluation of the anti-tumor eects and mechanisms of AM. More
importantly, the evidence of AM being able to regulate the tumor im-
mune microenvironment (TIME) will be provided, highlighting the
multiple anti-tumor targets and signaling pathways of AM as an im-
munity enhancer, may act as a booster for checkpoint immunotherapy
and chemotherapy.
2. Method
We searched the PubMed database for studies published unlimited,
beginning from January 2000 to January 2020. The keywords included
[Astragalus membranaceusor Huangqi]or[Astragalus] and
[tumoror canceror immunityor immune]. Language was
limited to English and Chinese. The lter process was rstly done by
search engine of the website which screened out 422 articles. We ex-
cluded 221 articles due to absence of abstract or being not t of the
theme in abstract, by the authors and screened out 201 articles. We
then excluded 123 articles due to unavailability of the full text and
being not t of the theme by the authors which screened out 78 articles.
Therefore, the number of basic, clinical and review article/meta-ana-
lysis were 40, 9 and 29 respectively. Flowchart of the search processes
is shown in Fig. 1.
3. General introduction of Astragalus membranaceus
AM was rst recorded in the Han Dynasty's "Shen Nong's Herbs"
2000 years ago [Li et al., 2019a,b]. Chinese doctor also calls it huang
qi, but this refers mainly to the roots of AM, named Astragali Radix.
AM is commonly recognized as a tonic to treat patients with a de-
ciency in vitality, which present as a lack of strength, anorexia, spon-
taneous sweating, edema, and abscesses. It can also induce urination
and promote the discharge of pus, plus the growth of new tissue
formulated as an ingredient of herbal mixtures in decoction. It has also
been used as a health food supplement as well as in Chinese ethnic
tonifying soups, gruel and tea in some Asian populations (Lin et al.,
2019;Psihogios et al., 2019).
AM is a leguminous plant, mainly distributed in the northeast, north
and northwest regions of China, Mongolia and North Korea. There are
over 2000 species of AM, like Astragalus membranaceus (Fisch.) Bge.
(Fam Leguminosae) and Astragalus membranaceus (Fisch.) Bge. Var.
mongholicus (Bge.), among which Hsiao are the most commonly ap-
plied. More than 200 compounds were identied from AM including
polysaccharides, saponins, avonoids and some others, several of which
have been found with biological activities (Li et al., 2014;Qi et al.,
2008;Zhang et al., 2015). Because polysaccharides are large molecules
with complex chemical structures, their precise chemical analysis is
very limited (Jin et al., 2014;Xie et al., 2016), but it is widely used in
the clinic. Saponin components have been explicitly included in 161
types, and astragalosides I, II, and IV, and isoastragaloside I and II
making up more than 80% of the total. There are 63 avonoids, among
them, isoavones are the most important and have been studied a lot
(Fu et al., 2014;Song et al., 2007). In recent years, AM and its com-
monly used components, including astragalus polysaccharide (APS),
astragaloside IV and formoterol abstracted from AM owers, have been
extensively studied for their remarkable anti-tumor activities (Burrell
and Swanton, 2014;Hanahan and Weinberg, 2011;Hong et al., 2017;
Schmetterer and Pickl, 2017;Skaggs et al., 2008). The commonly used
dosage forms are granula, capsule, oral liquid and injection (Dong et al.,
2010;Guo et al., 2012;Li et al., 2015).
4. Astragalus membranaceus reduces the side eects and improves
the therapeutic actions of chemotherapy in cancer patients
APS is more commonly used in a variety of tumors, including gastric
cancer and colon cancer. A systematic review based on 15 TCMs com-
bining with a commonly used drug for gastric cancer, showed that APS
injection could relieve clinical symptoms (odds ratio and 95% con-
dence intervals, 3.06 (1.01, 8.99), achieving a higher performance
status, and was superior in reducing leucopenia and gastrointestinal
reaction to chemotherapy than using FOLFOX regimen single (Zhang
et al., 2017). Aiming at solving the problem of poor systemic treatment
and large toxic side eects of non-small cell lung cancer (NSCLC),
McCulloch et al. evaluated the evidence from 34 randomized control
trials (RCT) and found that AM components and AM-based TCM in-
cluding its combination with platinum-based chemotherapy could re-
duce the risk of death in 12 months ((risk ratio [RR] = 0.67; 95% CI,
0.52 to 0.87)), with improved tumor response data (RR = 1.34; 95% CI,
1.24 to 1.46) and reduced toxic reaction of chemotherapy (McCulloch
et al., 2006). Besides, Guo et al. reported after 3 cycles of treatment,
APS injection combined with vinorelbine and cisplatin (VC) could
Fig. 1. Flow chart of the search processes.
S. Li, et al. Journal of Ethnopharmacology 258 (2020) 112797
2
improve the quality of life (QOL), physiological function, fatigue,
nausea and vomiting, pain, loss of appetite and other symptoms in
patients with advanced NSCLC compared with the patients on VC alone
(Guo et al., 2012). Very recently, a phase II double-blind randomized
placebo-controlled trial showed that APS injection displayed less con-
current chemoradiotherapy (CCRT)-associated adverse events, QOL
uctuation from the baseline, while a signicant improvement in the
pain, appetite loss, and social eating behavior during CCRT in the
CCRT/placebo groups of patients with advanced head and neck squa-
mous cell carcinoma (Hsieh et al., 2020).
Besides AM and its extracts, the prescriptions by using AM as the
main component have also been proved benecial for cancer patients.
For instance, Shenqi Fuzheng Injection (SFI) was approved as an in-
jectable Chinese medicine formula by the China Food and Drug
Administration (FDA) in the 1990s, which consists of two TCM's, AM
and Radix Codonopsis in a ratio of 1:1. In the past 30 years, SFI has been
reported to be eective in a variety of advanced cancer types. A sys-
tematic meta-analysis evaluated the ecacy of SFI in the treatment of
advanced gastric cancer involving 13 randomized controlled trials and
860 patients (Li et al., 2015). Results showed that chemotherapy
combined with SFI improved QOL, complete remission rate and partial
remission rate, and reduced adverse reactions such as nausea, vomiting,
oral mucositis and leukopenia (Li et al., 2015). In addition, SFI com-
bined with platinum-based chemotherapy improved the ecacy and
toxicity of advanced NSCLC and colorectal cancer (CRC) (Dong et al.,
2010).
In fact, many clinical trials of AM and AM-based TCM prescriptions
ecacy have been reported in Chinese journals, with capacity such as
improving QOL of cancer patients, alleviating the side eects of
radiotherapy and chemotherapy as well as improving the organic im-
munity. However, these articles are not collected by PUBMED data
base. To summarize, main components of AM and AM-based TCMs can
reduce the side eects and enhance the eect of chemotherapies, al-
though the direct curable actions on shrunk tumors has not been re-
ported yet. There are some limitations mentioned in the cited meta-
analysis, including methodological deciencies, small sample sizes,
limited to East Asian patients, which would lead to potential risks of
bias (Cao et al., 2019). It is expected that more high-quality and large-
sample RCTs are carried out and reported in the high-impact journal, to
further verify the above conclusion.
5. The mechanisms of Astragalus membranaceus anti-tumor actions
Using tumor animal models or cell lines, AM has shown direct anti-
tumor activity in various tumor models such as NSCLC, liver cancer,
gastric cancer, breast cancer, and ovarian cancer. Based on the existing
literatures, it is speculated that AM mainly exerts its anti-tumor eects
by directly inhibiting the proliferation and promoting apoptosis of
tumor cells; increasing the ecacy of chemotherapies, potentially
preventing tumor cell metastasis and improving TME by enhancing
organic or local immunity.
5.1. The anti-proliferative and pro-apoptotic actions on cancer by
Astragalus membranaceus and related pathways
Studies haves shown that AM and its main components have the
capacity to inhibit the growth of many types of tumor tissues and cells
via several signaling pathways in vitro and in vivo as shown in
Table 1(Auyeung et al., 2016). Various components of AM can inhibit
the proliferation of tumor cells and promote their apoptosis. Among
them, Park et al. found that the ethyl acetate fraction of AM (EAM)
reduced the proliferation of NSCLC cells in a dose and time-dependent
manner with increased numbers of cells in a non-proliferative state. In
addition, EAM treated cells displayed chromatin condensation, an in-
creased annexin V-positive cell population accompanied by the up-
regulated expression of cleaved caspase-8 and -9 and the accumulation
of lysed poly ADP-ribose polymerase (PARP) via extracellular regulated
protein kinases (ERK) signaling pathway (Park and Park., 2018). It has
been recently found that APS was able to inhibit the proliferation of
human gastric cancer MGC-803 cells and promote mitochondria-de-
pendent apoptosis in a concentration and time-dependent manner. The
underlying mechanisms involved APS induction of intracellular reactive
oxygen species (ROS) accumulation and further promotion of the
apoptosis (Yu et al., 2019). Zhou et al. reported that four isoavones of
AM extracts campanulin, ononin, calycosin, and formononetin could
inhibit proliferation and induce apoptosis of several breast cell lines
(MCF-7, SK-BR-3, MDA-MB-231) mediated through the PI3K/AKT/
mTOR pathways (Zhou et al., 2018). However, studies using antago-
nists of these signaling pathways need to be conducted to provide a rm
conclusion in this regard.
Formononetin is one of the major isoavonoid constituents isolated
from AM and demonstrates diverse pharmacological benets. Yang
et al. demonstrated that three cell lines of NSCLC and A549 treated with
formononetin showed inhibited cell growth in a time- and dose-de-
pendent manner with G1-phase cell cycle arrest and enhancement of
apoptosis in NSCLC mediated by the p53 signaling pathway (Yang et al.,
2014). Baicalein, another AM active ingredient, has shown its anti-
tumor ability in various cancers. It could inhibit the growth of human
nasopharyngeal carcinoma cells by inhibiting their cell proliferation
and inducing apoptosis. Baicalein could also eectively limit both
CNE1-and CNE2-transplanted tumors in nude mice with down-regula-
tion of Bcl-xl and Mcl-1 proteins and up-regulation of Bax and Bad (Guo
et al., 2019). All the evidences above indicates diverse components
abstracted from AM can inhibit the growth of tumor by targeting both
proliferation and apoptosis in several cancer types, but this direct anti-
tumor actions need clinical conformation.
5.2. The actions of Astragalus membranaceus in tumor-associated
environment
Tumor cells reside in TME which contains a variety of mesenchymal
cells, particularly broblasts, myobroblasts, endothelial cells, peri-
cytes, mesenchymal stem cells, innervating nerves and immune cells, in
addition to secreted factor from these cells (Burrell and Swanton,
2014). Many "characteristics of cancer" are associated with TME, which
has the capacity to promote proliferation and inhibit apoptosis, en-
hance angiogenesis and create hypoxia and an immune-suppressive
environment to avoid immune detection and support cancer growth,
even invasion and metastasis (Hanahan and Weinberg, 2011). Hence,
the manipulation of the immune niche of TME has been used as an
approach to treat cancer and prevent its progression.
5.2.1. Alleviation of immunosuppressive status in tumor immune
microenvironments by Astragalus membranaceus
An increasing number of immune cell subpopulations and related
molecules have been observed in TME, which play an important role in
promoting and inhibiting the occurrence and progression of cancers
(Wu et al., 2019). There are two basic proles in TME: a hotimmune
inamed prole which displays an increased distribution of CD8
+
tumor inltrating lymphocytes (TILS), accompanied by cells derived
from bone marrow such as myeloid lineages with higher levels of in-
terferon and chemokines including CXCL9, CLCL10, CXCL11 and other
pro-inammatory eector cytokines. In contrast, a coldnon-inamed
prole is characterized by immunosuppression, with high inltratio of
myeloid derived suppressor cells, M2-phynotype macrophages and
regulatory T cells (Treg, CD4
+
CD25
+
Foxp3
+
dominant), with en-
hanced expression of IL-10, IL-4 and TGF-β(Hong et al., 2017;
Schmetterer and Pickl, 2017;Skaggs et al., 2008;Namdar et al., 2018;
Safarzadeh et al., 2018;Chanmee et al., 2014;Zhou et al., 2014).
As has been shown, macrophages can be divided into M1 and M2
phenotypes. As the forerunner in the defense against tumor cells, M1-
macrophages rapidly colonize and secrete cytokines that kill tumor
S. Li, et al. Journal of Ethnopharmacology 258 (2020) 112797
3
cells, as well as support pro-inammatory cells by activating dendritic
cells and natural killer cells (Hong et al., 2017). However, advanced
tumors display severe immune-suppression, dominated by M2-macro-
phages which promote the progression of numerous cancers and predict
a poor prognosis for tumor-bearing animals and human patients (Jiang
et al., 2019). M2-macrophages polarization is induced by several im-
mune-suppressive cytokines including IL-4, IL-10, IL-13 and gluco-
corticoids, which can accelerate the metastasis and angiogenesis of
tumors (Xu et al., 2018). CD8
+
T cells are cytotoxic T lymphocytes
(CTLs) that kill tumor cells in TME, but are inhibited by Tregs and
immunosuppressive cytokines, such as IL-4, IL-10 and TGF-β. CTLs
however highly express PD-1, which can induce apoptosis by tumor
secreted PD-1 ligand (Huang et al., 2012). Tregs are key components of
tumor-acquired tolerance and inhibit most types of immune responses
(Chatila, 2005;Zhang et al., 2014).
According to TCM theory, Chinese herbal medicine with qi-toni-
fying eect, has the function of enhancing the defense ability of the
immune system. Until now, various pharmacological studies have
shown that AM has immunomodulatory properties, manifested by ac-
tivation of lymphocytes, natural killer cells and macrophages and up-
regulation of related cytokine production such as interferon and TNF-
α(Li et al., 2019a,b).
As shown in Table 2, lines of evidence have shown that AM com-
ponents could relieve the severe immunosuppressive condition in can-
cers or cancer-bearing organisms. For instance, it has demonstrated that
Arg-1 (a M2 macrophages marker) decient mice displayed shrunken
tumor size compared to those of wild type mice (Xu et al., 2018), and an
ethanol extract of AM, Astragaloside IV (AS-IV) could inhibit more than
50% expression of Arg-1 and CD206, possibly by targeting the im-
munosuppressive activity of M2-macrophages (Jiang et al., 2019).
Further, AS-IV was found to inhibit M2 conditioned medium-induced
A549 and H1299 cell invasion, migration and angiogenesis. In vivo in-
vestigation conrmed that the tumor region contained a decreased
proportion of M2-macrophages in Lewis lung cancer-bearing animals in
the AS-IV treatment group, with inhibited AMPKαactivation in M2
macrophages, while silencing AMPKαpartially abolishes the eect of
AS-IV, suggesting that AS-IV reduces the growth, migration and an-
giogenesis of lung cancer by blocking the polarization of M2 macro-
phages via the AMPK signaling pathway (Xu et al., 2018).
It is encouraging that the anti-tumor activity of APS-activated
macrophages with up-regulating the concentration of nitric oxide (NO)
and TNF-α, which may be the upstream of tumor cell proliferation in-
hibition such as G1 cell cycle arrest, and modulation of apoptosis-re-
lated genes, directly preventing the growth of cancer cells (Li et al.,
2019a,b). Recent studies reported that APS increased the M1/M2
macrophage polarization ratio in NSCLC H441 and H1299 cells, with
concomitant marked inhibition of cell proliferation, clonogenicity and
tumor sphere formation (Bamodu et al., 2019). A hydrosoluble poly-
saccharide named RAP extracted from AM(Wei et al., 2016) has been
shown directly to induce the macrophage cell line RAW264.7 to pro-
duce the inammatory cytokines TNF-α, IL-6 and iNOS, with were
blocked by TLR4 inhibitors. Further evidence supported that RAP could
activate TLR-4-related MAPKs, including phosphorylation of ERK and
p38 and induction of translocation NF-κB signaling, indicating RAP
activates macrophages via the TLR-4-MAPK-NF-κb pathway (Zhou
et al., 2017).
Regarding the eect of AM in T cell regulation, in ex-vivo tumor
mice using clinical sample from the NSCLC cohort showed that APS
inhibited tumor growth, promoted functional maturation of dendrite
cells with consequent enhancement of T cell-mediated anti-tumor im-
mune responses, synergistically enhanced the anti-M2-mediated anti-
tumor eect of cisplatin which could explain the clinical ecacy of AM
and chemotherapy. Meanwhile, the combination of AM and IL-2 was
able to enhance anti-tumor activity by increasing inltrating IL-2 gen-
erated lymphokine-activated killer cells (LAK), although with lowered
side eects of recombinant IL-2 therapy, such as acute renal failure,
capillary leak syndrome, myocardial infarction and uid retention
compared to those patients treated with IL-2 alone (Zou and Liu, 2003),
indicating that AM could potentially have benecial eects in combi-
nation with several immune therapies (Guo et al., 2012). However,
more evidences of the potential to increase T cell activity in TME by AM
need further studied.
In recent years, immunotherapy has become an eective treatment
in a variety of cancers. Cytotoxic T-lymphocyte protein 4 (CTLA4) and
programmed cell death protein 1 (PD-1) inhibitors are currently the two
most important immunological checkpoints since they can induce ac-
tivation and clonal-proliferation of tumor-specic T cells in TME.
However, although antibodies against the immunoregulatory factors
CTLA4 and PD-L1/PD-1 have been clinically successful, only a small
percentage of patients have shown persistent responses. It has been
speculated that the main reasons for this were that the severe immune-
suppressive TME and impaired antigen presentation capability in tumor
sites inhibit the proliferation and survival of inltrating T cells, and that
suppressive organic immunity inhibits the migration of more T cells
into tumor sites (Chen and Mellman, 2017;Wu et al., 2019). AM and its
major components are good immunopotentiators in organic and tumor
local immunity, providing potential candidates as an immunoadjuvant
checkpoint for the treatment of various cancers. This could perhaps be
in combination with other eective agents such as chemotherapy, to
further enhance each other's anti-tumor eects, and further studies of
these combination therapies should be carried out in the future.
Angiogenesis is the key to the development, progression, and
Table 1
Anti-proliferation and pro-apoptotic mechanisms of Astragalus membranaceus.
Refs. Component, medication,
dose
Tumor model Control Eect indicators Mechanism index
(Park and Park,
2018)
EAM
200 μg/ml
NSCLC cell: H1299, H460,
A549, H1975
DMSO Cell survivalproliferation
apoptosis
Caspase-8, caspase -9, PARP
(Yu et al., 2019) APS4
200, 400 and 800 μg/ml
Gastric carcinoma cell:
MGC-803
NC-DMEM, positive
control- 5-FU;
Proliferation, apoptosis, the
number of cells in S phase
ROS, Bax/Bcl-2, caspase-9/3,
PARP
(Zhou et al., 2018)AM
25, 50 μg/ml
Brest cancer cell: MCF-7,
SK-BR-3, MDA-MB-231
NC-DMSO Proliferationpercentage of cell
death
PI3K/AKT/mTOR: P-PI3K, P-G53Kβ,
p-Akt, P-mTOR
(Yang et al., 2014) Formononetin
100, 150 and 200 μM
NSCLC cell: A549, NCI-H23 NC-PBS Proliferation, apoptosis, the
number of cells in S phase
P53: phosphorylation at Ser15, Ser20
Proliferation: G1 phase: p21, cyclin
A, cyclin 1
Apoptosis: caspase-3, bax, bcl-2
(Guo et al., 2019) Baicalein, cell: 25, 50,
100 μM,
animal: 1, 2, and 3 mg/
kg,ip.
NPC cell: CNE1, CNE2
animal: CNE1 and CNE2-
bearing mice
PC-DDP Apoptotic rate, tumor size,
weight,
tumor inhibition rate
Apoptosis: Bcl-xl, Mcl-1, caspase 3,
caspase 8, Bax, Bad
EAM: ethyl acetate fraction of AM; APS4: a novel cold-water-soluble polysaccharide was isolated from Astragalus membranaceus; AM: isoavones, campanulin,
ononin, calycosin and formononetin; Baicalein: an active ingredient separated from AM; NC: negative contorl; PC: positive control.
S. Li, et al. Journal of Ethnopharmacology 258 (2020) 112797
4
Table 2
Mechanism of Astragalus membranaceus on improving TIME and organic immunity.
Ref. Medication, dose, mode Tumor model Control Eect indicators Immune indicators & mechanism indexes
Enhance tumor immune microenvironment
(Xu et al., 2018) AS-IV cell: 40, 80 μM animal: 40 mg/kg,
intragastrically
Lung cancer cell: A549, H1299
animal: LLC-bearing mice
cell: NC-DMSO animal: NC-NS Tumor size, survival rate, tumor
vessel maturation, metastasis
IL-13, IL-4,M2, CD206, IL-10, TGF-
β, p-AMPKα
(Li et al., 2019a,b) APS cell: 50, 100, 200, 500 and 1000 μg/
mL
Breast cancer cell: MCF-7 NC- conditioned medium PC-5-FU (50 μg/
mL)
Proliferation, G1-phase, apoptosisMacrophages,NO, TNF-α, Bax/Bcl-2
(Bamodu et al.,
2019)
APS cell: 8 mg/ml animal: 3 mg/kg
biweekly for 16 weeks, ip.
NSCLC cell: H441, H1229, H1437
animal: LLC1 and H1437, THP-1 cells-
bearing mice
cell: NC-PBS
animal: NC-DMSO
Body weight, tumors size and
weight, metastasis
nodules
M1/M2, DCs, T cell, IL-6, IL-10, NF-
κB, CD11b, CD31
(Wei et al., 2016) RAP cell: 30, 100 μg/ml Carcinoma cell: carcinoma (4T1),
macrophage (RAW264.7)
NC-DMEM Cell viabilityNO, TNF-α, IL-6, iNOS
(Zhou et al., 2017) APS cell: 400, 100 μg/ml, animal:
500mg/kg/d, oral 25 days
Breast cancer cell: RAW264.7,
animal: EAC- bearing mice
cell: NC-DMEM, PC- LPS (100 ng/ml),
animal: NC-NS, PC-ADM (4mg/kg/d), LPS
(5 mg/kg)
Weight of tumor, organ indexes,
inhibition rate
IL-6, IL-Iβ, TNF-α
(Law et al., 2012) AST cell: 80 μg/ml animal:100 mg/kg,
once daily for oral 14 days
Colon cancer cell: HCT116, HT-29
animal: HCT116-bearing mice
NC-DMEM Tumor volumeHIF-1α, bFGF, VEGF, PTEN, P-AKT,
P-mTOR, COX-2
Enhance the organic immunity to improve chemotherapy ecacy
(Yang et al., 2013) APS animal: 100, 200, 400 mg/kg, once
per day for 10 days intragastrically
Hepatocellular carcinoma animal: H22-
bearing mice
NC-NS, PC-5-FU (20 mg/kg) Tumor weight, tumor inhibitorySpleen and thymus indexes, macrophages
phagocytotic, IL-2, IL-12, TNF-α, IL-
10
Enhance the organic immunity to prevent tumor invasion and metastasis
(Zhang et al., 2014) APS cell: 100 ng/ml animal: 50 mg/kg Lung cancer cell: A549, PC9, B16F10,
LL2, animal: B16F10 and LL2-bearing
mice
Animal: NC-PBS Tumor growth, the survival, body
weight
Tcell, IDO, spleen and thymus indexes,
Cx43
(Zhang et al., 2018) Formononetin
80, 160, 240 μM
Ovarian cancer cell: A2780, SKOV3,
normal ovarian epithelial cell: IOSE80
NC -DMEM Proliferation, apoptosis, migration
and invasion
MMP-2/9, P-ERK, caspase3/9, Bax/Bcl-
2
AS-IV: Astragaloside IV;PG2(APS): Astragalus polysaccharides; RAP: a hyperbranched heteroglycan with average molecular weight of 1334 kDa; AST: Radix Astragali; PG2: extracts of Astragalus membranaceus;AMP:
Astragalus membranaceus polysaccharide; NC: negative contorl; PC: positive control.
S. Li, et al. Journal of Ethnopharmacology 258 (2020) 112797
5
metastasis of solid tumors (Li et al., 2018). Recent studies have found
that the tumor's vascular system is highly abnormal and dysfunctional,
and therefore, the ability of immune eector cells to penetrate solid
tumors is impaired. Normalization of tumor vasculature can improve
the inltration of immune eector cells, thereby enhancing tumor-
killing or inhibiting capacity. At the same time, stimulating immune
cell function also contributes to the normalization of the tumor vessel
system (Huang et al., 2018). Therefore, it has been proposed that the
regulation of crosstalk between tumor vascular normalization and im-
mune reprogramming can enhance long-lasting anti-tumor immunity
(Law et al., 2012).
Vascular endothelial growth factor (VEGF) has been veried as the
most critical angiogenic factor in angiogenesis and vascular abnorm-
alities. Since VGEF is released by various tumor cells, it is overexpressed
in dierent human tumors and leads to tumor progression. Law et al.
found that total Astragalus saponins could down-regulate the levels of
VEGF protein in HCT 116 colon cancer cells in a time- and dose-de-
pendent manner, with the mechanisms of modulation of Akt/mTOR
signaling molecules, upregulate PTEN, decrease Akt phosphorylation
with subsequent activation of mTOR (Law et al., 2012). The anti-car-
cinogenic action of AS- IV was further illustrated in HCT 116 xeno-
grafted athymic nude mice, with inhibited tumor growth and serum
VEGF levels. Levels of p-Akt, p-mTOR, VEGF, VEGFR1 and VEGFR2 in
tumor tissues were decreased by AS- IV, indicating that AS-IV can exert
anti-tumor activity in colon cancer by regulating mTOR-VEGF signaling
(Ichinohe, 2001;Semenza, 2003). However, the role of AS-IV in im-
proving immune-vascular crosstalk requires further investigation.
5.2.2. Astragalus memeranaceus improves the chemotherapy ecacy and
prevent tumor invasion and metastasis via boosting organic immunity
5.2.2.1. Astragalus membranaceus improves the organic immunity. Tumor
cells can escape from immune surveillance due to the host immune
system including not only tumor local immunity but also organic
immunity failing to deal with tumor-associated antigens, which
promote tumor invasion and metastasis, or aect therapeutic
response (Zhang et al., 2014). Thus boosting the organic immunity is
important in anti-tumor therapy. Lines of evidences have demonstrated
that AM, as a tonic, can enhance the organic immunity in physiological
status and various disease. On the innate immune response, the aqueous
extract of AM induced the maturation, activation and migration of
monocytes in peripheral blood (Denzler et al., 2010). In addition, the
activity of macrophage migration into the peritoneal cavity and
macrophage phagocytic activity were found to be enhanced by AM
(Cho and Leung, 2007). In the meantime, AM could also aect the
acquired immune response. For instance, through in vitro and in vivo
investigations, Cho et al. found that AM exhibited mitogenic and co-
mitogenic potentials on mouse spleen cells and lymphocytes, as well as
increasing IL-2 receptor expression on splenic cells which may mediate
the proliferation of T cells. In terms of immune recovery activity, AM
restored the lymphocyte germination reaction of aged mice to the level
of normal young mice. Meanwhile, intraperitoneal injection of AM
signicantly enhanced the antibody response of the mice to sheep
erythrocytes (Cho and Leung, 2007).
5.2.2.2. Astragalus membranaceus improves chemotherapy ecacy via
boosting organic immunity. Chemotherapy is considered to be an
important approach for treating malignant tumors, with
immunosuppression being one of the common clinical manifestations
in chemotherapy. Alleviating immunosuppression is one of the
important obstacles of chemotherapy to overcome. Lines of evidence
have conrmed that the immunomodulatory eects of AM are closely
related to its protective eects of immune organs by regulation of
lymphocytes, leukocytes and macrophages in chemotherapy-induced
immunosuppression (Huang et al., 2007;Li et al., 2008;Wang et al.,
2012;Zhang et al., 2009). In the cyclophosphamide-induced
immunosuppressive mice, the spleen index, peripheral blood
leukocyte and bone marrow cell counts showed accelerated recovery
and increased splenic natural killer cell activity, and peritoneal
macrophage phagocytosis was increased by AM. At the same time,
levels of IFN-γ, IL-12P70, IL-6 and IL-17 were also up-regulated by AM,
indicating that AM can enhance organic immunity in
immunosuppressive mice (Huang et al., 2007;Li et al., 2008;Wang
et al., 2012;Zhang et al., 2009). Astragalus oligosaccharide (AOS),
degraded from APS was found to alleviate cyclophosphamide-induced
immunosuppression mediated by stimulating the secretion of GM-CSF,
which promoted the dierentiation of progenitor cells after
proliferation in bone marrow, spleen and thymus (Zhu et al., 2017).
Multiple clinical trials mentioned in Section 4have shown that AM
can reduce chemotherapy-related adverse reactions and improve e-
cacy in cancer patients. In the mechanism studies as shown in Table 2,it
was found that APS could induce connexin 43 which is involved in
facilitating the passage of chemotherapeutic drugs to bystander tumor
cells; And decrease indoleamine 2, 3-dioxygenase, which can deplete
tryptophan, reduce the active T cell number and destroy immune sur-
veillance, providing the evidence that APS could be suitable combina-
tions with chemotherapies (Phacharapiyangkul et al., 2019). In the
meantime, SFI was designed to elucidate the in vivo immuno-enhance-
ment eects of SFI in immunosuppressed mice induced by cyclopho-
sphamide treatment. Results showed that SFI treatment accelerated
recovery dose-dependently of spleen index, peripheral white blood cell
and bone marrow cell counts, enhanced T cell and B cell proliferation
responses, as well as splenic nature killer cell activity and peritoneal
macrophage phagocytosis, and restored the level of interleukin-2 in the
serum, providing experimental evidences for supporting clinical e-
cacy of combinational SFI chemotherapy in cancer patients (Wang
et al., 2012).
5.2.2.3. Astragalus membranaceus enhances the organic immunity so that
to prevent tumor invasion and metastasis. The vast majority of cancer-
related deaths are the result of metastasis due to the scarcity of current
therapeutic options for patients with advanced metastatic tumors.
During metastasis, the crosstalk between cancer cells and the immune
system dictates the fate of tumor progression. For example, monocyte
counts in peripheral blood play a premetastatic role, and patient-
derived monocytes showed a greater ability to promote cancer cell
invasion and angiogenesis compared to monocytes from healthy donors
(Chittezhath et al., 2014). On the one hand, immune evasion by tumors
is to establish an immunosuppressive environment that inhibits the
development of anti-tumor immune responses in both the local and
systemic immune environment. These tumor-educated myelocytes,
especially tumor-associated macrophages (TAMs) and tumor-
associated neutrophils (TANs), can inhibit the anti-tumor immune
response through the production of immunosuppressive cytokines, T-
cell expression of co-inhibitory molecules, the reduction of amino acids
which are vital for the functioning of T-cells, and the production of ROS
(Fleming et al., 2018). Similarly, TAM counts are related to poor
clinical outcomes in several types of cancer (Campbell et al., 2011;
Steidl et al., 2010), and higher neutrophil-to-lymphocyte ratios in
peripheral blood are associated with poor survival (Templeton et al.,
2014). The potential mechanism involved is that immunosuppressive
cells can migrate to the pre-metastatic niche and create a suitable
environment for tumor growth (Blomberg et al., 2018;Wang et al.,
2017;Wculek and Malanchi, 2015,2019), and then drive tumor cells
migration and extravasation to the pre-metastatic region far from
primary tumor site (Hiratsuka et al., 2002;Kowanetz et al., 2010;
Qian et al., 2011). Therefore, alleviating the systemic
immunosuppressive environment has become an attractive strategy
for combating metastasis. In view of the signicant immunosuppression
and pro-metastatic role of tumor-inltrating M2-macrophages, N2-
neutrophils, and Tregs, more researchers are exploring strategies for
recruiting, polarization and eector molecules. AM may be a potential
organic immune enhancer for preventing tumor metastasis (Blomberg
S. Li, et al. Journal of Ethnopharmacology 258 (2020) 112797
6
et al., 2018).
As mentioned above, chemotherapy contributes to induce organic
immunosuppressive status, while AM could alleviate such an im-
munosuppressive condition (Table 2). Zhou et al. also found that
TLR4
+/+
and MyD88
+/+
wild-type tumor-bearing mice treated with
APS displayed reduced tumor size, up-regulated levels of immune organ
indexes and increased circulatory levels of pro-inammatory cytokines
including TNF-α, IL-1βand IL-6 (Zhou et al., 2017). Given that the
cytoplasmic portion of CD45 contains protein tyrosine phosphatase
activity and is critical for TCR-mediated T cell activation, AS-II was
found to enhance the proliferation of primary splenocytes and the de-
phosphorylation of Tyr505 in primary T cells mediated by CD45 PTPase
modulation. Furthermore, AS-II could induce Th1 polarization, which
manifested with signicantly increased IL-2 and IFN-γtranscription and
secretion from primary splenocytes upon TCR stimulation. In vivo stu-
dies showed that oral administration of AS-II restored the proliferation
of splenic T cells and the production of IFN-γand IL-2 in CTX-induced
immunosuppressed mice. Taken together, these results show that AS-II
enhances T cell activation by regulating the activity of CD45 PTPase
(Wan et al., 2013).
AM could partially restore the organic immune function of tumor-
bearing mice and cyclophosphamide-treated mice as well (Choi et al.,
2014). Yang et al. used the H22 tumor-bearing mouse model to study
the in vivo therapeutic eect of APS against cancer. The results showed
that APS can not only enhance tumor deterioration, but also increase
body weight, spleen index and thymus index and the phagocytotic
ability of macrophages in mice with tumors. In addition, APS admin-
istration increased levels of IL-2, ILI-2 and TNF-α, indicating that APS
inhibits tumor growth at least to some extent by improving organic
immunity. In the in vitro setting of H22 in hepatoma cells treated with
ASP, it was observed that ASP could signicantly inhibit tumor growth
as well as elevate serum cytokine levels (TNF-α, IL-2 and IFN-γ) and
activate immune cells (macrophages, lymphocytes and NK cells),
thereby inducing tumor cells apoptosis (Yang et al., 2013).
Few studies support that AM may prevent tumor-related metastasis
via boosting organic immunity. Wang et al. found APS combined with
cisplatin (DDP) could alleviate the pathological changes of the
recurrent tumor tissues and the metastasis-related protein expressions
of CD44, CD62P and osteopontin. following Lewis lung carcinoma
(LLC) surgery (Liu et al., 2018). Another study showed formononetin
suppressed the migration and invasion of ovarian cancer cells by using
in vitro wound healing and trans well chamber assay accompanied with
decreased expression of MMP-2/9 proteins and phosphorylation level of
ERK, indicating formononetin may have potential to prevent cancer
metastasis but further evidence should be provided by future in-
vestigation (Zhang et al., 2018).
6. Conclusion
AM is commonly used as a tonic in TCM with multiple bioactivities.
To conclude in Fig. 2, it has been demonstrated that AM and its active
constituents, combined with chemotherapies, are capable of dimin-
ishing the side eects and complications induced by chemotherapies
and enhancing the ecacy in cancer patients. Utilizing various cancer
models and cell lines, AM has been found to capable of shrinking or
stabilizing tumors by direct anti-proliferation or by pro-apoptosis of
tumor cells. Meanwhile, the crucial mechanism of AM immune reg-
ulation is to alleviate the immune-suppressive status by activating M1
macrophages via the TLR4/MyD88/NFκB signaling pathway and en-
hancing tumor-kill capacity of T cells in tumor environment. In addi-
tion, it was found that AM could also improve systemic immunity which
may help promoting ecacy of chemotherapy and preventing metas-
tasis. Thereby this review contributes to an understanding of AM as an
adjunctive therapy in the whole course of cancer treatment, with pro-
viding useful information for development of more eective anti-tumor
medication. However, due to the methodological quality of the current
clinical trials, it is prospected that rigorously designed RCTs are war-
ranted to generate a high level of clinical evidence. The combination of
AM and immune checkpoint therapies would has a promising prospect,
and observation of direct ecacy and mechanisms on tumor growth
and metastasis by AM combined with chemotherapies or immune
checkpoint therapies require more in vivo validations and further clin-
ical investigation as well.
Fig. 2. Anti-tumor eects and mechanisms of Astragalus membranaceus and its specic immunopotentiation.
S. Li, et al. Journal of Ethnopharmacology 258 (2020) 112797
7
Declaration of competing interest
The authors have no conict of interests regarding this paper.
Acknowledgment
This study was nancially supported by the National Natural
Science Foundation of China (NSFC) No. 81704146, 81273868,
81873369, and 81873368, the Tianjin Municipal Bureau of Labor and
Social Security No.2018015.
Abbreviation
AM Astragalus membranaceus
APS Astragalus polysaccharides
AS-IV Astragaloside IV
(CCRT) concurrent, chemoradiotherapy
CTLA4 cytotoxic T-lymphocyte protein 4
CTLs cytotoxic T lymphocytes
CRC colorectal cancer
EAM ethyl acetate fraction of AM
ERK extracellular regulated protein kinases
HCC hepatocellular carcinoma
NMA network meta-analysis
LAK lymphokine-activated killer
NSCLC non-small-cell lung cancer
NO Nitric oxide
QOL quality of life
PARP poly ADP-ribose polymerase
PD-1 programmed cell death protein 1
ROS reactive oxygen species
SFI Shenqi Fuzheng Injection
TAMs tumor-associated macrophages
TME tumor microenvironment
TANs tumor-associated neutrophils
TCMs traditional Chinese medicines
TIM tumor immune microenvironment
TILs tumor inltrating lymphocytes
TLR4 Toll-like receptor 4
VEGF Vascular endothelial growth factor
VC vinorelbine and cisplatin
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://
doi.org/10.1016/j.jep.2020.112797.
References
Auyeung, K.K., Han, Q.B., Ko, J.K., 2016. Astragalus membranaceus: a review of its
protection against inammation and gastrointestinal cancers. Am. J. Chin. Med. 44,
122.
Bamodu, O.A., Kuo, K.T., Wang, C.H., Huang, W.C., Wu, A., Tsai, J.T., Lee, K.Y., Yeh, C.T.,
Wang, L.S., 2019. Astragalus polysaccharides (PG2) enhances the M1 polarization of
macrophages, functional maturation of dendritic cells, and T cell-mediated antic-
ancer immune responses in patients with lung cancer. Nutrients 11.
Blomberg, O.S., Spagnuolo, L., de Visser, K.E., 2018. Immune regulation of metastasis:
mechanistic insights and therapeutic opportunities. Dis. Model. Mech. 11.
Bray, F., Ferlay, J., Soerjomataram, I., Siegel, R.L., Torre, L.A., Jemal, A., 2018. Global
cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide
for 36 cancers in 185 countries. Ca - Cancer J. Clin. 68, 394424.
Bray, F., Jemal, A., Grey, N., Ferlay, J., Forman, D., 2012. Global cancer transitions ac-
cording to the Human Development Index (2008-2030): a population-based study.
Lancet Oncol. 13, 790801.
Burrell, R.A., Swanton, C., 2014. The evolution of the unstable cancer genome. Curr.
Opin. Genet. Dev. 24, 6167.
Campbell, M.J., Tonlaar, N.Y., Garwood, E.R., Huo, D., Moore, D.H., Khramtsov, A.I., Au,
A., Baehner, F., Chen, Y., Malaka, D.O., Lin, A., Adeyanju, O.O., Li, S., Gong, C.,
McGrath, M., Olopade, O.I., Esserman, L.J., 2011. Proliferating macrophages asso-
ciated with high grade, hormone receptor negative breast cancer and poor clinical
outcome. Breast Canc. Res. Treat. 128, 703711.
Cao, A., He, H., Wang, Q., Li, L., An, Y., Zhou, X., 2019. Evidence of Astragalus injection
combined platinum-based chemotherapy in advanced nonsmall cell lung cancer pa-
tients: a systematic review and meta-analysis. Medicine 98, e14798.
Casey, S.C., Amedei, A., Aquilano, K., Azmi, A.S., Benencia, F., Bhakta, D., Bilsland, A.E.,
Boosani, C.S., Chen, S., Ciriolo, M.R., Crawford, S., Fujii, H., Georgakilas, A.G., Guha,
G., Halicka, D., Helferich, W.G., Heneberg, P., Honoki, K., Keith, W.N., Kerkar, S.P.,
Mohammed, S.I., Niccolai, E., Nowsheen, S., Vasantha Rupasinghe, H.P., Samadi, A.,
Singh, N., Talib, W.H., Venkateswaran, V., Whelan, R.L., Yang, X., Felsher, D.W.,
2015. Cancer prevention and therapy through the modulation of the tumor micro-
environment. Semin. Canc. Biol. 35 (Suppl. l) S199-199S223.
Chanmee, T., Ontong, P., Konno, K., Itano, N., 2014. Tumor-associated macrophages as
major players in the tumor microenvironment. Cancers 6, 16701690.
Chatila, T.A., 2005. Role of regulatory T cells in human diseases. J. Allergy Clin.
Immunol. 116, 949959 quiz 960.
Chen, D.S., Mellman, I., 2017. Elements of cancer immunity and the cancer-immune set
point. Nature 541, 321330.
Chittezhath, M., Dhillon, M.K., Lim, J.Y., Laoui, D., Shalova, I.N., Teo, Y.L., Chen, J.,
Kamaraj, R., Raman, L., Lum, J., Thamboo, T.P., Chiong, E., Zolezzi, F., Yang, H., Van
Ginderachter, J.A., Poidinger, M., Wong, A.S., Biswas, S.K., 2014. Molecular proling
reveals a tumor-promoting phenotype of monocytes and macrophages in human
cancer progression. Immunity 41, 815829.
Cho, W.C., Leung, K.N., 2007. In vitro and in vivo immunomodulating and im-
munorestorative eects of Astragalus membranaceus. J. Ethnopharmacol. 113,
132141.
Choi, Y.K., Cho, S.G., Woo, S.M., Yun, Y.J., Park, S., Shin, Y.C., Ko, S.G., 2014. Herbal
extract SH003 suppresses tumor growth and metastasis of MDA-MB-231 breast
cancer cells by inhibiting STAT3-IL-6 signaling. Mediat. Inamm. 492173.
Denzler, K.L., Waters, R., Jacobs, B.L., Rochon, Y., Langland, J.O., 2010. Regulation of
inammatory gene expression in PBMCs by immunostimulatory botanicals. PloS One
5 e12561.
Dong, J., Su, S.Y., Wang, M.Y., Zhan, Z., 2010. Shenqi fuzheng, an injection concocted
from Chinese medicinal herbs, combined with platinum-based chemotherapy for
advanced non-small cell lung cancer: a systematic review. J. Exp. Clin. Canc. Res. : CR
29, 137.
Fleming, V., Hu, X., Weber, R., Nagibin, V., Groth, C., Altevogt, P., Utikal, J., Umansky,
V., 2018. Targeting myeloid-derived suppressor cells to bypass tumor-induced im-
munosuppression. Front. Immunol. 9, 398.
Fu, J., Wang, Z., Huang, L., Zheng, S., Wang, D., Chen, S., Zhang, H., Yang, S., 2014.
Review of the botanical characteristics, phytochemistry, and pharmacology of
Astragalus membranaceus (Huangqi). Phytother Res. : PTR 28, 12751283.
Guo, J., You, H., Li, D., 2019. Baicalein exerts anticancer eect in nasopharyngeal car-
cinoma in vitro and in vivo. Oncology Res. 27, 601611.
Guo, L., Bai, S.P., Zhao, L., Wang, X.H., 2012. Astragalus polysaccharide injection in-
tegrated with vinorelbine and cisplatin for patients with advanced non-small cell lung
cancer: eects on quality of life and survival. Med. Oncol. 29, 16561662.
Hanahan, D., Weinberg, R.A., 2011. Hallmarks of cancer: the next generation. Cell 144,
646674.
Hiratsuka, S., Nakamura, K., Iwai, S., Murakami, M., Itoh, T., Kijima, H., Shipley, J.M.,
Senior, R.M., Shibuya, M., 2002. MMP9 induction by vascular endothelial growth
factor receptor-1 is involved in lung-specic metastasis. Canc. Cell 2, 289300.
Hong, J.W., Lim, J.H., Chung, C.J., Kang, T.J., Kim, T.Y., Kim, Y.S., Roh, T.S., Lew, D.H.,
2017. Immune tolerance of human dental pulp-derived mesenchymal stem cells
mediated by CD4CD25FoxP3regulatory T-cells and induced by TGF-β1 and IL-10.
Yonsei Med. J. 58, 10311039.
Hsieh, C.H., Lin, C.Y., Hsu, C.L., Fan, K.H., Huang, S.F., Liao, C.T., Lee, L.Y., Ng, S.K., Yen,
T.C., Chang, J.T., Lin, J.R., Wang, H.M., 2020. Incorporation of Astragalus poly-
saccharides injection during concurrent chemoradiotherapy in advanced pharyngeal
or laryngeal squamous cell carcinoma: preliminary experience of a phase II double-
blind, randomized trial. J. Canc. Res. Clin. Oncol. 146, 3341.
Huang, C.S., Liu, L., Liu, J., Chen, Z., Guo, J., Li, C.Z., Zhou, D.G., Wang, Z.H., 2012.
Association of chemotherapy-induced leucopenia with treatment outcomes in ad-
vanced non-small-cell lung cancer cases receiving the NP regimen. Asian Pac. J.
Cancer Prev. APJCP : Asian Pac. J. Cancer Prev. APJCP 13, 44814485.
Huang, G.C., Wu, L.S., Chen, L.G., Yang, L.L., Wang, C.C., 2007. Immuno-enhancement
eects of Huang Qi Liu Yi Tang in a murine model of cyclophosphamide-induced
leucopenia. J. Ethnopharmacol. 109, 229235.
Huang, Y., Kim, B., Chan, C.K., Hahn, S.M., Weissman, I.L., Jiang, W., 2018. Improving
immune-vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol. 18,
195203.
Ichinohe, T., 2001. Tumor angiogenesis and microcirculation. Int. J. Hematol. 74, 479.
Jiang, Y.X., Chen, Y., Yang, Y., Chen, X.X., Zhang, D.D., 2019. Screening ve qi-tonifying
herbs on M2 phenotype macrophages. Evid. base Compl. Alternative Med. : eCAM
2019, 9549315.
Jin, M., Zhao, K., Huang, Q., Shang, P., 2014. Structural features and biological activities
of the polysaccharides from Astragalus membranaceus. Int. J. Biol. Macromol. 64,
257266.
Kowanetz, M., Wu, X., Lee, J., Tan, M., Hagenbeek, T., Qu, X., Yu, L., Ross, J., Korsisaari,
N., Cao, T., Bou-Reslan, H., Kallop, D., Weimer, R., Ludlam, M.J., Kaminker, J.S.,
Modrusan, Z., van Bruggen, N., Peale, F.V., Carano, R., Meng, Y.G., Ferrara, N., 2010.
Granulocyte-colony stimulating factor promotes lung metastasis through mobiliza-
tion of Ly6G+Ly6C+ granulocytes. Proc. Natl. Acad. Sci. U.S.A. 107, 2124821255.
Law, P.C., Auyeung, K.K., Chan, L.Y., Ko, J.K., 2012. Astragalus saponins downregulate
vascular endothelial growth factor under cobalt chloride-stimulated hypoxia in colon
cancer cells. BMC Compl. Alternative Med. 12, 160.
Li, H., Jiang, T., Li, M.Q., Zheng, X.L., Zhao, G.J., 2018. Transcriptional regulation of
S. Li, et al. Journal of Ethnopharmacology 258 (2020) 112797
8
macrophages polarization by MicroRNAs. Front. Immunol. 9, 1175.
Li, J., Bao, Y., Lam, W., Li, W., Lu, F., Zhu, X., Liu, J., Wang, H., 2008. Immunoregulatory
and anti-tumor eects of polysaccharopeptide and Astragalus polysaccharides on
tumor-bearing mice. Immunopharmacol. Immunotoxicol. 30, 771782.
Li, J., Wang, J.C., Ma, B., Gao, W., Chen, P., Sun, R., Yang, K.H., 2015. Shenqi Fuzheng
Injection for advanced gastric cancer: a systematic review of randomized controlled
trials. Chin. J. Integr. Med. 21, 7179.
Li, W., Song, K., Wang, S., Zhang, C., Zhuang, M., Wang, Y., Liu, T., 2019a. Anti-tumor
potential of astragalus polysaccharides on breast cancer cell line mediated by mac-
rophage activation. Mater. Sci. Eng. C Mater. Mater Appl. 98, 685695.
Li, X., Qu, L., Dong, Y., Han, L., Liu, E., Fang, S., Zhang, Y., Wang, T., 2014. A review of
recent research progress on the astragalus genus. Molecules : J. Synth. Chem. Nat.
Prod. Chem. 19, 1885018880.
Li, Y., Guo, S., Zhu, Y., Yan, H., Qian, D.W., Wang, H.Q., Yu, J.Q., Duan, J.A., 2019b.
Flowers of Astragalus membranaceus var. mongholicus as a novel high potential by-
product: phytochemical characterization and antioxidant activity. Molecules : J.
Synth. Chem. Nat. Prod. Chem. 24.
Lin, C., Cao, S.M., Chang, E.T., Liu, Z., Cai, Y., Zhang, Z., Chen, G., Huang, Q.H., Xie, S.H.,
Zhang, Y., Yun, J., Jia, W.H., Zheng, Y., Liao, J., Chen, Y., Lin, L., Liu, Q., Ernberg, I.,
Huang, G., Zeng, Y., Zeng, Y.X., Adami, H.O., Ye, W., 2019. Chinese nonmedicinal
herbal diet and risk of nasopharyngeal carcinoma: a population-based case-control
study. Cancer 125, 44624470.
Liu, D., Li, Y., Wang, X., Wang, Y., Ma, Y., Chen, Y., Ming, H., 2018. [Astragalus poly-
saccharide combined with cisplatin inhibits growth of recurrent tumor and down-
regulats the expression of CD44, CD62P and osteopontin in tumor tissues in mice
bearing Lewis lung cancer]. Chin. J. Cell. Mol. Immunol. 34, 11051110.
Martins-Neves, S.R., Paiva-Oliveira, D.I., Wijers-Koster, P.M., Abrunhosa, A.J., Fontes-
Ribeiro, C., Bovée, J.V., Cleton-Jansen, A.M., Gomes, C.M., 2016. Chemotherapy
induces stemness in osteosarcoma cells through activation of Wnt/β-catenin sig-
naling. Canc. Lett. 370, 286295.
McCulloch, M., See, C., Shu, X.J., Broman, M., Kramer, A., Fan, W.Y., Gao, J., Lieb, W.,
Shieh, K., Colford Jr., J.M., 2006. Astragalus-based Chinese herbs and platinum-
based chemotherapy for advanced non-small-cell lung cancer: meta-analysis of ran-
domized trials. J. Clin. Oncol. : O. J. Am. Soc. Clin. Oncol. 24, 419430.
Namdar, A., Mirzaei, R., Memarnejadian, A., Boghosian, R., Samadi, M., Mirzaei, H.R.,
Farajifard, H., Zavar, M., Azadmanesh, K., Elahi, S., Noorbakhsh, F., Rezaei, A.,
Hadjati, J., 2018. Prophylactic DNA vaccine targeting Foxp3+ regulatory T cells
depletes myeloid-derived suppressor cells and improves anti-melanoma immune re-
sponses in a murine model. Canc. Immunol. Immunother. : CII 67, 367379.
Nicolas-Boluda, A., Donnadieu, E., 2019. Obstacles to T cell migration in the tumor mi-
croenvironment. Comp. Immunol. Microbiol. Infect. Dis. 63, 2230.
Park, H.J., Park, S.H., 2018. Induction of apoptosis by ethyl acetate fraction of Astragalus
membranaceus in human non-small cell lung cancer cells: - apoptosis induction by
Astragalus membranaceus. J. Pharmacopuncture 21, 268276.
Phacharapiyangkul, N., Wu, L.H., Lee, W.Y., Kuo, Y.H., Wu, Y.J., Liou, H.P., Tsai, Y.E.,
Lee, C.H., 2019. The extracts of Astragalus membranaceus enhance chemosensitivity
and reduce tumor indoleamine 2, 3-dioxygenase expression. Int. J. Med. Sci. 16,
11071115.
Psihogios, A., Ennis, J.K., Seely, D., 2019. Naturopathic oncology care for pediatric
cancers: a practice survey. Integr. Canc. Ther. 18 1534735419878504.
Qi, L.W., Yu, Q.T., Yi, L., Ren, M.T., Wen, X.D., Wang, Y.X., Li, P., 2008. Simultaneous
determination of 15 marker constituents in various radix Astragali preparations by
solid-phase extraction and high-performance liquid chromatography. J. Separ. Sci.
31, 97106.
Qian, B.Z., Li, J., Zhang, H., Kitamura, T., Zhang, J., Campion, L.R., Kaiser, E.A., Snyder,
L.A., Pollard, J.W., 2011. CCL2 recruits inammatory monocytes to facilitate breast-
tumour metastasis. Nature 475, 222225.
Safa, A.R., Saadatzadeh, M.R., Cohen-Gadol, A.A., Pollok, K.E., Bijangi-Vishehsaraei, K.,
2015. Glioblastoma stem cells (GSCs) epigenetic plasticity and interconversion be-
tween dierentiated non-GSCs and GSCs. Genes & diseases 2, 152163.
Safarzadeh, E., Orangi, M., Mohammadi, H., Babaie, F., Baradaran, B., 2018. Myeloid-
derived suppressor cells: important contributors to tumor progression and metastasis.
J. Cell. Physiol. 233, 30243036.
Schmetterer, K.G., Pickl, W.F., 2017. The IL-10/STAT3 axis: contributions to immune
tolerance by thymus and peripherally derived regulatory T-cells. Eur. J. Immunol. 47,
12561265.
Semenza, G.L., 2003. Targeting HIF-1 for cancer therapy. Nat. Rev. Canc. 3, 721732.
Shao, B.M., Xu, W., Dai, H., Tu, P., Li, Z., Gao, X.M., 2004. A study on the immune
receptors for polysaccharides from the roots of Astragalus membranaceus, a Chinese
medicinal herb. Biochem. Biophys. Res. Commun. 320, 11031111.
Skaggs, B.J., Singh, R.P., Hahn, B.H., 2008. Induction of immune tolerance by activation
of CD8+ T suppressor/regulatory cells in lupus-prone mice. Hum. Immunol. 69,
790796.
Song, J.Z., Mo, S.F., Yip, Y.K., Qiao, C.F., Han, Q.B., Xu, H.X., 2007. Development of
microwave assisted extraction for the simultaneous determination of isoavonoids
and saponins in radix astragali by high performance liquid chromatography. J. Separ.
Sci. 30, 819824.
Steidl, C., Lee, T., Shah, S.P., Farinha, P., Han, G., Nayar, T., Delaney, A., Jones, S.J.,
Iqbal, J., Weisenburger, D.D., Bast, M.A., Rosenwald, A., Muller-Hermelink, H.K.,
Rimsza, L.M., Campo, E., Delabie, J., Braziel, R.M., Cook, J.R., Tubbs, R.R., Jae,
E.S., Lenz, G., Connors, J.M., Staudt, L.M., Chan, W.C., Gascoyne, R.D., 2010. Tumor-
associated macrophages and survival in classic Hodgkin's lymphoma. N. Engl. J. Med.
362, 875885.
Templeton, A.J., McNamara, M.G., Šeruga, B., Vera-Badillo, F.E., Aneja, P., Ocaña, A.,
Leibowitz-Amit, R., Sonpavde, G., Knox, J.J., Tran, B., Tannock, I.F., Amir, E., 2014.
Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: a systematic review
and meta-analysis. J. Natl. Cancer Inst. 106, dju124.
Wan, C.P., Gao, L.X., Hou, L.F., Yang, X.Q., He, P.L., Yang, Y.F., Tang, W., Yue, J.M., Li, J.,
Zuo, J.P., 2013. Astragaloside II triggers T cell activation through regulation of CD45
protein tyrosine phosphatase activity. Acta Pharmacol. Sin. 34, 522530.
Wang, D., Sun, H., Wei, J., Cen, B., DuBois, R.N., 2017. CXCL1 is critical for premetastatic
niche formation and metastasis in colorectal cancer. Canc. Res. 77, 36553665.
Wang, J., Tong, X., Li, P., Cao, H., Su, W., 2012. Immuno-enhancement eects of Shenqi
Fuzheng Injection on cyclophosphamide-induced immunosuppression in Balb/c mice.
J. Ethnopharmacol. 139, 788795.
Wculek, S.K., Malanchi, I., 2015. Neutrophils support lung colonization of metastasis-
initiating breast cancer cells. Nature 528, 413417.
Wculek, S.K., Malanchi, I., 2019. Author Correction: neutrophils support lung coloniza-
tion of metastasis-initiating breast cancer cells. Nature 571, E2.
Wei, W., Xiao, H.T., Bao, W.R., Ma, D.L., Leung, C.H., Han, X.Q., Ko, C.H., Lau, C.B.,
Wong, C.K., Fung, K.P., Leung, P.C., Bian, Z.X., Han, Q.B., 2016. TLR-4 may mediate
signaling pathways of Astragalus polysaccharide RAP induced cytokine expression of
RAW264.7 cells. J. Ethnopharmacol. 179, 243252.
Wu, X., Gu, Z., Chen, Y., Chen, B., Chen, W., Weng, L., Liu, X., 2019. Application of PD-1
blockade in cancer immunotherapy. Comput. Struct. Biotechnol. J. 17, 661674.
Xie, J.H., Jin, M.L., Morris, G.A., Zha, X.Q., Chen, H.Q., Yi, Y., Li, J.E., Wang, Z.J., Gao, J.,
Nie, S.P., Shang, P., Xie, M.Y., 2016. Advances on bioactive polysaccharides from
medicinal plants. Crit. Rev. Food Sci. Nutr. 56 (Suppl. 1), S60S84.
Xu, F., Cui, W.Q., Wei, Y., Cui, J., Qiu, J., Hu, L.L., Gong, W.Y., Dong, J.C., Liu, B.J., 2018.
Astragaloside IV inhibits lung cancer progression and metastasis by modulating
macrophage polarization through AMPK signaling. J. Exp. Clin. Canc. Res. : CR 37,
207.
Yang, B., Xiao, B., Sun, T., 2013. Antitumor and immunomodulatory activity of Astragalus
membranaceus polysaccharides in H22 tumor-bearing mice. Int. J. Biol. Macromol.
62, 287290.
Yang, Y., Zhao, Y., Ai, X., Cheng, B., Lu, S., 2014. Formononetin suppresses the pro-
liferation of human non-small cell lung cancer through induction of cell cycle arrest
and apoptosis. Int. J. Clin. Exp. Pathol. 7, 84538461.
Yu, J., Ji, H., Dong, X., Feng, Y., Liu, A., 2019. Apoptosis of human gastric carcinoma
MGC-803 cells induced by a novel Astragalus membranaceus polysaccharide via in-
trinsic mitochondrial pathways. Int. J. Biol. Macromol. 126, 811819.
Zhang, A., Zheng, Y., Que, Z., Zhang, L., Lin, S., Le, V., Liu, J., Tian, J., 2014.
Astragaloside IV inhibits progression of lung cancer by mediating immune function of
Tregs and CTLs by interfering with IDO. J. Canc. Res. Clin. Oncol. 140, 18831890.
Zhang, D., Zheng, J., Ni, M., Wu, J., Wang, K., Duan, X., Zhang, X., Zhang, B., 2017.
Comparative ecacy and safety of Chinese herbal injections combined with the
FOLFOX regimen for treating gastric cancer in China: a network meta-analysis.
Oncotarget 8, 6887368889.
Zhang, J., Liu, L., Wang, J., Ren, B., Zhang, L., Li, W., 2018. Formononetin, an isoavone
from Astragalus membranaceus inhibits proliferation and metastasis of ovarian
cancer cells. J. Ethnopharmacol. 221, 9199.
Zhang, K., Pugliese, M., Pugliese, A., Passantino, A., 2015. Biological active ingredients of
traditional Chinese herb Astragalus membranaceus on treatment of diabetes: a sys-
tematic review. Mini Rev. Med. Chem. 15, 315329.
Zhang, R.P., Zhang, X.P., Ruan, Y.F., Ye, S.Y., Zhao, H.C., Cheng, Q.H., Wu, D.J., 2009.
Protective eect of Radix Astragali injection on immune organs of rats with ob-
structive jaundice and its mechanism. World J. Gastroenterol. 15, 28622869.
Zhou, D., Huang, C., Kong, L., Li, J., 2014. Novel therapeutic target of hepatocellular
carcinoma by manipulation of macrophage colony-stimulating factor/tumor-asso-
ciated macrophages axis in tumor microenvironment. Hepatol. Res. : O. J. Jpn. Soc.
Hepatol. 44, E318E319.
Zhou, L., Liu, Z., Wang, Z., Yu, S., Long, T., Zhou, X., Bao, Y., 2017. Astragalus poly-
saccharides exerts immunomodulatory eects via TLR4-mediated MyD88-dependent
signaling pathway in vitro and in vivo. Sci. Rep. 7, 44822.
Zhou, R., Chen, H., Chen, J., Chen, X., Wen, Y., Xu, L., 2018. Extract from Astragalus
membranaceus inhibit breast cancer cells proliferation via PI3K/AKT/mTOR sig-
naling pathway. BMC Compl. Alternative Med. 18, 83.
Zhu, Z.Y., Zhang, J.Y., Liu, F., Chen, L., Chen, L.J., Tang, Y., 2017. Characterization and
lymphocyte proliferation activity of an oligosaccharide degraded from Astragalus
polysaccharide. MedChemComm 8, 15211530.
Zou, Y.H., Liu, X.M., 2003. [Eect of astragalus injection combined with chemotherapy
on quality of life in patients with advanced non-small cell lung cancer]. Chin. J.
Integrated Tradit. West Med. 23, 733735.
S. Li, et al. Journal of Ethnopharmacology 258 (2020) 112797
9
... It has also the ability that can help to boost humoral and cellular immunity, thereby enhancing the effect of inhibition on anti-tumour cells and thereby stimulating apoptotic cell death, through using different cancer models and cell lines. It can also lessen the damage to the immune system through radiation therapy; on the other hand, it involves the balance of tumour adjunctive chemotherapy (32). Research findings have shown that to minimize complications and prevent chemotherapy-induced side effects, the compounds of AM coupled with chemotherapy can improve anti-tumour effects in people with cancer. ...
Article
Full-text available
Astragalus used mainly in Chinese traditional medicine, and in some parts of other Asian countries like Japan. It is one of the fundamental herbs found among medicinal and food plants. The plant contains various beneficial compounds, which provide enormous benefits to human health, such as the improvisation of the immune system, and it prevents and helps in the treatments of many diseases. These compounds act against various diseases like cancer, heart disease, a neurodegenerative disorder, and kidney and urinary-related problems. These elements also perform some crucial activities like anti-microbial activity, anti-ox-idative activity, anti-hypertensive effect, etc., which is the precursor for the cause of various diseases. The dried root or the root powder of the plant is widely used as a nutrient supplement and used in foodstuffs includes ice cream, jelly, mayonnaise, syrups, sauce, candies, etc. The bark, root, flower, and stem extracts used widely in cosmetics, such as skin protection and skin whitening. Astragalus membranaceus (AM) is a natural substitute for food, medicine, and cosmetic ingredients as it is always good to replace synthetic ones with natural compounds. Therefore, this review describes the importance of its biologically active molecules and their applications in different health benefits in various aspects.
... It has also the ability that can help to boost humoral and cellular immunity, thereby enhancing the effect of inhibition on anti-tumour cells and thereby stimulating apoptotic cell death, through using different cancer models and cell lines. It can also lessen the damage to the immune system through radiation therapy; on the other hand, it involves the balance of tumour adjunctive chemotherapy (32). Research findings have shown that to minimize complications and prevent chemotherapy-induced side effects, the compounds of AM coupled with chemotherapy can improve anti-tumour effects in people with cancer. ...
Article
Astragalus used mainly in Chinese traditional medicine, and in some parts of other Asian countries like Japan. It is one of the fundamental herbs found among medicinal and food plants. The plant contains various beneficial compounds, which provide enormous benefits to human health, such as the improvisation of the immune system, and it prevents and helps in the treatments of many diseases. These compounds act against various diseases like cancer, heart disease, a neurodegenerative disorder, and kidney and urinary-related problems. These elements also perform some crucial activities like anti-microbial activity, anti-ox-idative activity, anti-hypertensive effect, etc., which is the precursor for the cause of various diseases. The dried root or the root powder of the plant is widely used as a nutrient supplement and used in foodstuffs includes ice cream, jelly, mayonnaise, syrups, sauce, candies, etc. The bark, root, flower, and stem extracts used widely in cosmetics, such as skin protection and skin whitening. Astragalus membranaceus (AM) is a natural substitute for food, medicine, and cosmetic ingredients as it is always good to replace synthetic ones with natural compounds. Therefore, this review describes the importance of its biologically active molecules and their applications in different health benefits in various aspects.
Article
Full-text available
Background Calycosin may be a potential candidate regarding chemotherapeutic agent, because already some studies against multivarious cancer have been made with this natural compound. Aim This review elucidated a brief overview of previous studies on calycosin potential effects on various cancers and its potential mechanism of action. Methodology Data retrieved by systematic searches of Google Scholar, PubMed, Science Direct, Web of Science, and Scopus by using keywords including calycosin, cancer types, anti‐cancer mechanism, synergistic, and pharmacokinetic and commonly used tools are BioRender, ChemDraw Professional 16.0, and ADMETlab 2.0. Results Based on our review, calycosin is available in nature and effective against around 15 different types of cancer. Generally, the anti‐cancer mechanism of this compound is mediated through a variety of processes, including regulation of apoptotic pathways, cell cycle, angiogenesis and metastasis, oncogenes, enzymatic pathways, and signal transduction process. These study conducted in various study models, including in silico, in vitro, preclinical and clinical models. The molecular framework behind the anti‐cancer effect is targeting some oncogenic and therapeutic proteins and multiple signaling cascades. Therapies based on nano‐formulated calycosin may make excellent nanocarriers for the delivery of this compound to targeted tissue as well as particular organ. This natural compound becomes very effective when combined with other natural compounds and some standard drugs. Moreover, proper use of this compound can reverse resistance to existing anti‐cancer drugs through a variety of strategies. Calycosin showed better pharmacokinetic properties with less toxicity in human bodies. Conclusion Calycosin exhibits excellent potential as a therapeutic drug against several cancer types and should be consumed until standard chemotherapeutics are available in pharma markets.
Article
The Astragalus propinquus is used in traditional Chinese medicines (TCM) for centuries. However, its mechanism of action and functional targets remain elusive. The purpose of the study is to conduct a pharmacological network analysis to identify the interaction of A. propinquus derived components with the target proteins involved in rheumatoid arthritis (RA) and diabetes. The chemical components of A. propinquus were obtained from Traditional Chinese Medicine Systems Pharmacology Database (TCMSP), and the target-protein related genes were collected from DisGeNET and GeneCards databases. A total of 16 A. propinquus phytochemical compounds and 147 potential targets were screened for RA and diabetes-related target proteins STRING and Cytoscape software were used to establish the protein interaction networks. Finally, the pharmacological network was integrated to investigate the mechanism underlying the potential target protein network of A. propinquus. The results showed that the A. propinquus derived components such as quercetin, hederagenin, isorhamnetin, calycosin, 7-O-methylisomucronulatol, formononetin, and kaempferol exhibited potential binding efficacy with screened 89 target proteins, suggesting it as one of the natural medicines for the treatment of RA and diabetes. Gene Ontology and KEGG analysis revealed that certain target pathways were involved in RA and diabetes, which are also elaborated in this study. The molecular docking analysis demonstrates that the compound hederagenin acts as a potent inhibitor for RA and diabetics.
Article
Full-text available
Purpose Concurrent chemoradiotherapy (CCRT) is one of the standard treatments for patients with advanced head and neck squamous cell carcinoma (HNSCC). However, CCRT may lead to decreased quality of life (QoL) and treatment compliance. This study aimed to determine the effects of PG2 (Astragalus polysaccharides) injection on CCRT-associated adverse events (AEs) and patients’ compliance with the CCRT course. Methods In this phase II double-blind randomized placebo-controlled trial, PG2 injection (sterile powder form) or placebo was administrated three times per week in parallel with CCRT to patients with HNSCC. The chemotherapy regimen included 50 mg/m² cisplatin every 2 weeks with daily tegafur–uracil (300 mg/m²) and leucovorin (60 mg/day). Results The study was terminated prematurely due to the successful launch of a newly formulated PG2 injection (lyophilized form). A total of 17 patients were enrolled. The baseline demographics and therapeutic compliance were comparable between the CCRT/PG2 and CCRT/placebo groups. During CCRT, severe treatment-associated AEs were less frequent in the CCRT/PG2 group than in the CCRT/placebo group. Furthermore, less QoL fluctuations from the baseline during CCRT were noted in the CCRT/PG2 group than in the CCRT/placebo group, with a significant difference in the pain, appetite loss, and social eating behavior. The tumor response, disease-specific survival and overall survival did not differ between the two groups. Conclusion This preliminary study demonstrated PG2 injection exhibited an excellent safety profile, and has potential in ameliorating the deterioration in QoL and the AEs associated with active anticancer treatment among patients with advanced pharyngeal or laryngeal HNSCC under CCRT. Further research in patients with other cancer types or treatment modalities may widen PG2’s application in clinical settings.
Article
Full-text available
Background: The majority of pediatric oncology patients report use of complementary and alternative medicine. Some naturopathic doctors (NDs) provide supportive pediatric oncology care; however, little information exists to formally describe this clinical practice. A survey was conducted with members of the Oncology Association of Naturopathic Physicians (OncANP.org) to describe recommendations across four therapeutic domains: natural health products (NHPs), nutrition, physical medicine, and mental/emotional support. Results: We had 99 respondents with a wide variance of clinical experience and aptitude to treat children with cancer. Of the majority (52.5%) of respondents who choose not to treat these children, the three primary reasons for this are lack of public demand (45.1%), institutional or clinic restrictions (21.6%), and personal reasons/comfort (19.6%). The 10 most frequently considered NHPs by all NDs are fish-derived omega-3 fatty acid (83.3%), vitamin D (83.3%), probiotics (82.1%), melatonin (73.8%), vitamin C (72.6%), homeopathic Arnica (69.0%), turmeric/curcumin (67.9%), glutamine (66.7%), Astragalus membranaceus (64.3%), and Coriolus versicolor/PSK (polysaccharide K) extracts (61.9%). The top 5 nutritional recommendations are anti-inflammatory diets (77.9%), dairy restriction (66.2%), Mediterranean diet (66.2%), gluten restriction (61.8%), and ketogenic diet (57.4%). The top 5 physical modality interventions are exercise (94.1%), acupuncture (77.9%), acupressure (72.1%), craniosacral therapy (69.1%), and yoga (69.1%). The top 5 mental/emotional interventions are meditation (79.4%), art therapy (77.9%), mindfulness-based stress reduction (70.6%), music therapy (70.6%), and visualization therapy (67.6%). Conclusion: The results of our clinical practice survey highlight naturopathic interventions across four domains with a strong rationale for further inquiry in the care of children with cancer.
Article
Full-text available
Background An association between a nonmedicinal herbal diet and nasopharyngeal carcinoma (NPC) has often been hypothesized but never thoroughly investigated. Methods This study enrolled a total of 2469 patients with incident NPC and 2559 population controls from parts of Guangdong and Guangxi Provinces in southern China between 2010 and 2014. Questionnaire information was collected on the intake of traditional herbal tea and herbal soup as well as the specific herbal plants used in soups and other potentially confounding lifestyle factors. Multivariate logistic regression models were used to estimate odds ratios (ORs) with 95% confidence intervals (CIs) for the NPC risk in association with herbal tea and soup intake. Results Ever consumption of herbal tea was not associated with NPC risk (OR, 1.03; 95% CI, 0.91‐1.17). An inverse association was observed for NPC among ever drinkers of herbal soup (OR, 0.78; 95% CI, 0.67‐0.90) but without any monotonic trend with an increasing frequency or duration of herbal soup consumption. Inverse associations with NPC risk were detected with 9 herbal plants used in herbal soup, including Ziziphus jujuba, Fructus lycii, Codonopsis pilosula, Astragalus membranaceus, Semen coicis, Smilax glabra, Phaseolus calcaratus, Morinda officinalis, and Atractylodes macrocephala (OR range, 0.31‐0.79). Conclusions Consuming herbal soups including specific plants, but not herbal tea, was inversely associated with NPC. If replicated, these results might provide potential for NPC prevention in endemic areas.
Article
Full-text available
Background: Recently, we demonstrated that Astragalus polysaccharide (PG2), the active ingredient in dried roots of astragalus membranaceus, ameliorates cancer symptom clusters and improves quality of life (QoL) in patients with metastatic disease by modulating inflammatory cascade against the background roles of inflammatory cells, including macrophages, dendritic cells (DCs), and cytotoxic T lymphocytes (CTLs) in tumor initiation, metastasis, and progression. Nevertheless, the role of PG2 in the modulation of anticancer immunogenicity and therapeutic response remains relatively underexplored and unclear. Purpose: The present study investigates how and to what extent PG2 modulates cellular and biochemical components of the inflammatory cascade and enhances anticancer immunity, as well as the therapeutic implication of these bio-events in patients with lung cancer. Methods and results: Herein, we demonstrated that PG2 significantly increased the M1/M2 macrophage polarization ratio in non-small cell carcinoma (NSCLC) H441 and H1299 cells. This PG2-induced preferential pharmacologic up-regulation of tumoral M1 population in vitro positively correlated with the downregulation of tumor-promoting IL-6 and IL-10 expression in NSCLC cell-conditioned medium, with concomitant marked inhibition of cell proliferation, clonogenicity, and tumorsphere formation. Our ex vivo results, using clinical sample from our NSCLC cohort, demonstrated that PG2 also promoted the functional maturation of DCs with consequent enhancement of T cell-mediated anticancer immune responses. Consistent with the in vitro and ex vivo results, our in vivo studies showed that treatment with PG2 elicited significant time-dependent depletion of the tumor-associated M2 population, synergistically enhanced the anti-M2-based anticancer effect of cisplatin, and inhibited xenograft tumor growth in the NSCLC mice models. Moreover, in the presence of PG2, cisplatin-associated dyscrasia and weight-loss was markedly suppressed. Conclusion: These results do indicate a therapeutically-relevant role for PG2 in modulating the M1/M2 macrophage pool, facilitating DC maturation and synergistically enhancing the anticancer effect of conventional chemotherapeutic agent, cisplatin, thus laying the foundation for further exploration of the curative relevance of PG2 as surrogate immunotherapy and/or clinical feasibility of its use for maintenance therapy in patients with lung cancer.
Article
Full-text available
Astragalus membranaceus has been shown to possess anti-inflammation and antitumor properties. Several studies have indicated that extracts of Astragalus membranaceus (PG2) have growth inhibitory effects on tumor. However, the effect of PG2 on enhancing the chemotherapy, modulating tumor immune escape and their mechanism of action is unknown and need further investigation. Connexin (Cx) 43 is ubiquitous in cells and involved in facilitating the passage of chemotherapeutic drugs to bystander tumor cells. The indoleamine 2, 3-dioxygenase (IDO) depletes tryptophan, reduces the active T cell number and destroys immune surveillance. Herein, we provide evidence that the treatment of PG2 induced Cx43 expression, decreases IDO expression and enhances the distribution of chemotherapeutic drug. However, the effects of combination therapy (PG2 plus cisplatin) in animal models significantly retarded tumor growth and prolonged the survival. We believe that the information provided in this study may aid in the design of future therapy of PG2, suggest suitable combinations with chemotherapies.
Article
Full-text available
Change history: In this Letter, the final two pages of the Supplementary Information, which included two tables listing primer and antibodies, respectively, were missing; see accompanying Amendment for the missing Supplementary Tables. This Letter has not been corrected online.
Article
Full-text available
The programmed cell death protein 1 (PD-1) pathway has received considerable attention due to its role in eliciting the immune checkpoint response of T cells, resulting in tumor cells capable of evading immune surveillance and being highly refractory to conventional chemotherapy. Application of anti-PD-1/PD-L1 antibodies as checkpoint inhibitors is rapidly becoming a promising therapeutic approach in treating tumors, and some of them have successfully been commercialized in the past few years. However, not all patients show complete responses and adverse events have been noted, suggesting a better understanding of PD-1 pathway mediated immunosuppression is needed to predict patient response and improve treatment efficacy. Here, we review the progresses on the studies of the mechanistic role of PD-1 pathway in the tumor immune evasion, recent clinical development and commercialization of PD-1 pathway inhibitors, the toxicities associated with PD-1 blockade observed in clinical trials as well as how to improve therapeutic efficacy and safety of cancer immunotherapy.
Article
Full-text available
Baicalein, an active ingredient separated from Astragalus membranaceus, has shown its anticancer ability in various cancers. However, its effect on nasopharyngeal carcinoma has not been explored yet. The present study aimed to investigate the effect of baicalein on the growth, proliferation, apoptosis, and cell cycle of human nasopharyngeal carcinoma cells, as well as transplanted nude mouse xenograft. The results showed that baicalein inhibited the growth and proliferation of CNE1 and CNE2 cells in a time- and concentration-dependent manner. It also caused a significant increase in the number of cells in the G0/G1 phase and a decrease in the G₂/M phase, thereby reducing the number of cells entering mitosis and inhibiting the proliferation of tumor cells. Baicalein also significantly induced apoptosis of CNE1 and CNE2 cells. Western blots showed that baicalein decreased the expression of Bcl-xl and Mcl-1 and increased the expression of Bax, Bad, and caspase 3, 8, and 9. In CNE1- and CNE2-transplanted tumors of mice, baicalein significantly inhibited tumor growth. In conclusion, baicalein could inhibit the growth and proliferation of human nasopharyngeal carcinoma cells, change their cell cycle, and induce apoptosis. Baicalein also effectively limits both CNE1- and CNE2-transplanted tumors in nude mice. Downregulation of Bcl-xl and Mcl-1 proteins and upregulation of Bax and Bad may be involved in the mechanism.
Article
Full-text available
Background: Platinum-based chemotherapy is one of the standard treatments for advanced nonsmall cell lung cancer (NSCLC). Despite on an effective treatment for advanced NSCLC patients, its high toxicity and limited clinical effects have raised big concerns. Astragalus injection (AGI) has been commonly employed as an adjutant chemotherapy drug for NSCLC in China. This review was conducted to evaluate the beneficial of AGI in combination with platinum-based chemotherapy in advanced NSCLC. Methods: We collected all studies about AGI plus platinum-based chemotherapy for advanced NSCLC in the PubMed, EMBASE, China National Knowledge Infrastructure Database, the Cochrane Library, Wanfang Database, China Biological Medicine Database, and Chinese Scientific Journal Database established on July 2018 without language restriction. Cochrane handbook was applied to assess the quality of included trials. Stata (version 12.0) and RevMan (version 5.3) were employed for data analysis. The quality of the evidence was assessed with the GRADE approach. Results: Nineteen randomized controlled trials (RCTs) including 1635 patients were included to determine the effectiveness and safety of AGI combined with platinum-based chemotherapy in the treatment of NSCLC. The result of meta-analysis indicated that comparing with chemotherapy alone, AGI combined chemotherapy could significantly improve the objective response rate (relative risk [RR] = 1.19, 95% confidence interval [CI] [1.06, 1.33], P = .002), the Karnofsky performance status (RR = 2.28, 95% CI [1.63, 3.18], P < .00001), and 1-year survival rate (RR = 1.40, 95% CI [1.16, 1.70], P = .0005), meanwhile increase the percentages of CD3 (weighted mean differences [WMD] = 11.98, 95% CI [8.0, 15.96], P < .00001), CD4 (WMD = 2.98, 95% CI [0.45, 5.52], P = .02), CD4/CD8 (WMD = 0.33, 95% CI [0.20, 0.46], P < .00001), and NK cells (WMD = 9.5, 95% CI [7.25, 11.76], P < .00001), decrease the incidence of leukopenia (RR = 0.52, 95% CI [0.44, 0.61], P < .00001), platelet toxicity (RR = 0.62, 95% CI [0.50, 0.76], P < .00001), and vomiting (RR = 0.72, 95% CI [0.60, 0.87], P = .0006). Based on the system evaluation results, the GRADE system recommendation grading method was adopted to evaluate the evidence quality. The results showed that the level of evidence was low. Conclusions: The AGI apparently has attenuation and synergistic efficacy to platinum-based chemotherapy patients. However, considering the limits of articles included in the present researches, the recommendation is likely to be weak. High-quality RCTs are urgently used to generate conclusive results.
Article
Objective To investigate the effects of Astragalus polysaccharide (APS) combined with cisplatin (DDP) on the pathological morphology of recurrent tumor and the expression of metastasis-related proteins CD44, CD62P and osteopontin (OPN) following Lewis lung carcinoma (LLC) surgery. Methods LLC cell suspension was injected subcutaneously into palmula of left hind limb of C57BL/6J mice as a tumor-supply group. The other 80 mice were randomized into 8 groups: model group, APS-treated groups at different concentrations of 50, 100 and 200 μg/mL, 6 mg/kg DDP-treated group, and 3 mg/kg DDP combined with 50, 100, 200 μg/mL APS-treated groups. The palmula tumor cells were collected from the tumor-supply group 10 days after LLC injection and then injected subcutaneously into all of the 80 mice to establish the recurrent and metastatic mouse models of lung cancer. Subsequently, corresponding different substances were administrated intraperitoneally in the different treated groups since the next day. After 15 days' administration, all the mice were sacrificed by cervical spine dislocation. Morphological characteristics were observed by H&E staining, and the protein expression of CD44, CD62P and OPN were measured by immunohistochemistry. Results Compared with the model group, the pathological changes of the recurrent tissues in each treatment group were alleviated to some extent, especially in the DDP combined with 200 μg/mL APS group; the expression of CD44, CD62P and OPN in each treatment group decreased, especially in the DDP group and DDP combined with 100 and 200 μg/mL APS-treated groups. Conclusion APS combined with DDP could significantly inhibit the growth and metastasis of Lewis lung cancer cells, which might be associated with the reduced expression of CD44, CD62P and OPN.