Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 8128 word(s) 8128 2021-08-05 04:39:14 |
2 format correct Meta information modification 8128 2021-08-18 04:02:39 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Tang, Y.Q.; Yap, W.H. Honokiol. Encyclopedia. Available online: https://encyclopedia.pub/entry/13267 (accessed on 28 March 2024).
Tang YQ, Yap WH. Honokiol. Encyclopedia. Available at: https://encyclopedia.pub/entry/13267. Accessed March 28, 2024.
Tang, Yin Quan, Wei Hsum Yap. "Honokiol" Encyclopedia, https://encyclopedia.pub/entry/13267 (accessed March 28, 2024).
Tang, Y.Q., & Yap, W.H. (2021, August 17). Honokiol. In Encyclopedia. https://encyclopedia.pub/entry/13267
Tang, Yin Quan and Wei Hsum Yap. "Honokiol." Encyclopedia. Web. 17 August, 2021.
Honokiol
Edit

Cancer is characterised by uncontrolled cell division and abnormal cell growth, which is largely caused by a variety of gene mutations. There are continuous efforts being made to develop effective cancer treatments as resistance to current anticancer drugs has been on the rise. Natural products represent a promising source in the search for anticancer treatments as they possess unique chemical structures and combinations of compounds that may be effective against cancer with a minimal toxicity profile or few side effects compared to standard anticancer therapy. Extensive research on natural products has shown that bioactive natural compounds target multiple cellular processes and pathways involved in cancer progression.

honokiol anticancer mechanism signalling pathway

1. Introduction

Cancer is the outcome of rampant cell division which is associated with cell cycle disorganisation [1], leading to uncontrolled cell proliferation. In addition, it also involves the dysregulation of apoptosis, immune evasion, inflammatory responses, and ultimately, metastatic spread [2]. Over the last few decades, our progressive understanding of the aetiology of cancer together with advancement of cancer treatment, detection, and prevention, have contributed towards receding cancer mortality around the world [3]. However, more than half of cancer cases were diagnosed at a later stage of cancer progression [4]. According to a study by Bray et al. [5], the worldwide estimated number of new cancer cases for the year 2018 was 18.1 million in both sexes and across all ages. Amongst all the cancer types, lung, breast, and colorectum have topped the charts with approximately 2.1 million, 2.1 million, and 1.8 million cases, respectively. On the other hand, the estimated number of deaths was approximately 9.6 million. Asia accounted for more than half of the cancer deaths (57.3%), followed by Europe (20.3%), and America (14.4%). Lung cancer has caused the highest number of deaths due to substandard prognoses. Attempts to develop the effective prevention of cancer may diminish the incidence rate for some cancers, for instance lung cancer in North America and Northern Europe. These western countries have implemented tobacco control in order to avert involuntary exposure to tobacco and minimise active smoking within the community. Unfortunately, a majority of the population are still facing an upsurge of cancer diagnosis, demanding treatment and care [5].
The common treatment regimens for cancer patients include surgery, chemotherapy, and radiotherapy [6]. Although some of these regimens represent the first-in-line options for cancer treatment, the lack of selectivity towards neoplastic cells and the development of drug toxicity has caused these therapeutic effects to recede slowly, rendering it ineffective over the years [7]. Additionally, multidrug resistance tumours pose a severe threat and have been responsible for numerous cancer-related deaths [8]. A modern approach to target multiple cell regulating pathways is mandatory in order to provide highly efficient and targeted cancer therapy. For instance, combination therapy that targets different pathways exhibit significantly lower toxicity towards normal cells compared to mono-therapy [9]. Currently, the development of anticancer drugs possessing the capability to overcome common mechanisms of chemoresistance with minimal toxicity effects would be considered a breakthrough in cancer research [2].
Approximately 70–95% of the world population continues to use traditional medicinal herbs, plants, and fruits which contain valuable bioactive compounds with therapeutic effects to maintain health, as well as to prevent or treat physical and mental illnesses [10]. These biologically active compounds provide extensive opportunities in uncovering competent anticancer agents [2][11]. A majority of the anticancer drugs that are currently in use originate from plants, marine organisms, and microorganisms, such as the well-known plant-derived anti-cancer drugs Paclitaxel (Taxol®) and Camptothecin (CPT) [12].
The Magnolia genus is widely distributed throughout the world, especially in East and South-East Asia [13]. Among the Magnolia species, Magnolia officinalis and Magnolia obovata are commonly used in traditional Chinese (known as “Houpu”) and Japanese herbal medicine [13][14]. The traditional prescriptions named Hange-koboku-to and Sai-boku-to, which contain the Magnolia bark, are still used in modern clinical practice in Japan [15]. There are several potent bioactive compounds in the Magnolia species have been identified including honokiol, magnolol, obovatol, 4-O-methylhonokiol, and several other neolignan compounds [13][15][16]. This paper highlights the potential anticancer effect of a simple biphenyl neolignan found in this Magnolia family, namely honokiol.

2. Anticancer Properties of Honokiol

2.1. In Vitro Studies

Honokiol has been shown to exhibit antiproliferation effects against numerous cancer cells, including bone, bladder, brain, breast, blood, and colon, as shown in Table 1. Generally, the concentrations used for the in vitro studies are between 0–150 μM, which majority of these concentration ranges have been shown to significantly inhibit cell proliferation or cell viability of various cancer cell lines. The trend for the IC50 values of numerous cancer cell lines were time-dependent, whereby the IC50 values decreases as duration of the experiment increases. As seen in Table 1, human blood cancer Raji cells were highly susceptible to honokiol treatment (IC50 = 0.092) compared to highly resistant human nasopharyngeal cancer HNE-1 cells (IC50 = 144.71 μM). Interestingly, honokiol has been shown to exhibit minimal cytotoxicity against on normal cell lines, including human fibroblast FB-1, FB-2, Hs68, and NIH-3T3 cells [17][18][19][20]. The low cytotoxicity of honokiol treatment against normal cell lines should be emphasised as current chemotherapeutic regimens have a considerable amount of side effects that harm cancer patients.
Many chemotherapeutic agents have been shown to induce severe systemic toxicity and several side effects due to their deficient pharmacokinetic profiles and non-specific distribution in the body [21]. In Yang et al.’s study [22], they have encapsulated honokiol into nanopolymers to enhance its permeability and specificity against cancer cells. They utilised the active targeting nanoparticles-loaded honokiol (ANTH) in their in vitro studies against human nasopharyngeal cancer HNE-1 cells, and this incorporation exhibited significantly lower IC50 values compared to free honokiol treatment. As a result, the incorporation or encapsulation of honokiol in transporting vehicles can improve the anticancer effects and concurrently overcome the water solubility issue of honokiol itself. This has shown to be a promising regimen for anticancer treatment in the future.
Furthermore, it is worthy to note that honokiol can enhance the antineoplastic effects of several chemotherapeutic agents when cells are treated in combination treatment of both honokiol and the chemotherapeutic agent. In Wang et al.’s study [23], they have shown that honokiol has enhanced the in vitro cytotoxicity of paclitaxel against human cervix cancer cell lines. The combination treatment has resulted in approximately 10–60% increase of apoptotic cells and inhibition of cell viability when compared to honokiol treatment alone [23]. In another study, honokiol potentiated the apoptotic effect of both doxorubicin and paclitaxel against human liver cancer HepG2 cells. Honokiol enhanced the apoptotic effects of paclitaxel and doxorubicin by 22% and 24% respectively [24].
Table 1. The anticancer effects of honokiol against cancer cells in in vitro experiments.
Cell Lines Mechanism of Action Concentration Used Efficacy/IC50 (Exposure Time) References
Colorectal cancer RKO Inhibit cell proliferation
Induce G1 phase cell cycle arrest
Induce apoptosis ↓ Bcl-xL; ↑ Caspase-3 & caspase-9
0–150 μM 46.76 μM
(68 h)
[25]
HCT116, HCT116-CH2, HCT116-CH3 Inhibit cell proliferation
Induce G0/G1 & G2/M phase cell cycle
arrest: ↓ cyclin D1 & A1; ↑ p53 phosphorylation
Induce apoptosis: ↓ Caspase-3; ↓ Bcl-2; ↑ Bax protein
25 μM Honokiol with
2.5 or 5.0 Gy IR
N/A [26]
HT-29 Inhibit cell growth & proliferation
Induce G1 phase cell cycle arrest: ↓ Cdk1 & cyclin B1
0–50 μM followed by
0–5 Gy IR
23.05 μM (24 h)
13.24 μM (72 h)
[27]
HCT116 & SW480 Inhibit cell proliferation via
Inhibition of Notch signalling: ↓ Notch1 & Jagged-1; ↓ Hey-1 & Hes1; ↓ γ-secretase complex; ↓ Skip1
Induce apoptosis: ↑ caspase-3/-7 activity; ↓ Bcl-2 & Bcl-xL; ↑ Bax protein; ↓ cyclin D1 & c-Myc; ↑ p21WAF1 protein
Inhibit primary and secondary colonosphere formation
0–50 μM N/A [28]
RKO & HCT116 Inhibit cell viability
Induce apoptosis: ↑ caspase-3, caspase-8 & caspase-9 activation; ↑ DR5 & cleaved PARP proteins; ↑ survivin protein; ↑ phosphorylated p53 & p53 proteins; ↓ PUMA protein
0–60 μM RKO:
38.25 μM (24 h)
HCT116:
39.64 μM (24 h)
[29]
Blood cancer B-CLL Inhibit cell viability
Induce apoptosis: ↑ caspase-3 activity; ↑ caspase-8 & caspase-9 activation; ↓ caspase-9; ↑ Bax protein; ↓ Mcl-1 protein
0–100 μM 49 μM (6 h)
38 μM (24 h)
[30]
Raji, Molt-4 Inhibit cell growth: ↓ p65; ↓ NF-κB
Induce apoptosis: ↑ JNK activation
Increase ROS activity: ↑ Nrf2 & c-Jun protein activation
0–2.5 μM Raji:
3.500 μM (24 h)
0.092 μM (72 h)
Molt-4:
0.521 μM (24 h)
[31]
Breast cancer MCF-7, MDA-MB-231, SKBR-3, ZR-75-1, BT-474 Inhibit cell viability and growth: ↓ EFGR; ↓ MAPK/PI3K pathway activity
Induce apoptosis: ↑ PARP protein degradation; ↓ caspase-8; ↑ Bax proteins
Induce G1 phase cell cycle arrest: ↓ cyclin D1; ↑ p21 & p27
0–100 μM MCF-7:
40 μM (24 h)
MDA-MB-231: 33 μM (24 h)
SKBR-3:
29 μM (24 h)
ZR-75-1:
39 μM (24 h)
BT-474:
50 μM (24 h)
[32]
MCF-7, MDA-MB-231 Inhibit cell clonogenicity
Inhibit cell anchorage-dependent colony formation
Inhibit cell growth, migration & invasion: ↓ pS6K & 4EBP1 phosphorylation; ↑ AMPK activation; ↓ mTORC1 function; ↑ LKB1 & cytosolic localisation
1–25 μM N/A [33]
MCF-7, MDA-MB-231, SUM149, SUM159 Inhibit cell migration & invasion: ↑ AMPK phosphorylation; ↑ LKB1
Inhibit stem-like characteristics: ↓ Oct4, Nanog & Sox4 protein; ↓ STAT3; ↓ iPSC inducer mRNA
5 μM N/A [34]
MCF-7, MDA-MB-231, T47D, SKBR-3, Zr-75, BT-474 Inhibit cell growth: ↓ PI3K/Akt/mTOR signalling
Inhibit cell invasion
Induce G0/G1 phase cell cycle arrest: ↓ cyclin D1 & cyclin E; ↓ Cdk2 & c-myc; ↑ PTEN
Induce apoptosis: ↑ caspase-3, caspase-6 & caspase-9 activation
0–40 μM MCF7:
34.9 μM (24 h)
13.7 μM (48 h)
13.5 μM (72 h)
10.5 μM (96 h)

MDA-MB-231:
56.9 μM (24 h)
44.4 μM (48 h)
16.0 μM (72 h)
12.0 μM (96 h)

T47D:
47.7 μM (24 h)
41.6 μM (48 h)
17.6 μM (72 h)
7.1 μM (96 h)

SKBR-3:
76.1 μM (24 h)
68.1 μM (48 h)
62.7 μM (72 h)
15.7 μM (96 h)

ZR-75:
71.1 μM (24 h)
58.1 μM (48 h)
28.7 μM (72 h)
14.5 μM (96 h)

BT-474:
80.2 μM (24 h)
65.6 μM (48 h)
39.5 μM (72 h)
15.1 μM (96 h)
[35]
MDA-MB-231 Inhibit cell proliferation: ↓ c-Src/EGFR-mediated signalling pathway; ↓ c-Myc protein
Induce G0/G1 phase cell cycle arrest: ↓ cyclin A, cyclin D1 & cyclin E; ↓ Cdk2, Cdk4 & p-pRbSer780; ↑ p27Kip−1
Induce apoptosis: ↑ caspase-3, caspase-8 & caspase-9 cascade; ↓ Bcl-2 & Bid protein; ↑ PARP cleavage
0–100 μM 59.5 μM (72 h) [36]
Lung cancer A549 Inhibit cell growth & proliferation
Induce G0/G1 phase cell cycle arrest: ↓ Cdk1 & cyclin B1
0–50 μM 12.51 μM (24 h)
7.75 μM (72 h)
[27]
A549, H460, H226, H1299 Reduce invasive potential
Inhibit PGE2-induced cell migration: ↓ PGE2 production ↓ COX-2 ↑ β-catenin degradation ↓ NF-κB/p65 activity ↓ IKKα
0–20 μM N/A [37]
A549, H1299 Inhibit cell viability and growth: ↓ class I HDAC proteins; ↓ HDAC activity; ↑ histone acetyltransferase (HAT) activity; ↑ histone H3 & H4
Induce G1 phase cell cycle arrest: ↓ cyclin D1 & cyclin D2; ↓ Cdk2, Cdk4 & Cdk6
0–60 μM N/A [38]
H460 & A549 Inhibit cell proliferation
Induce apoptosis: ↑ cathepsin D; ↑ cleaved PARP; ↑ caspase-3
Inhibit autophagy: ↑ p62; ↑ LC3-II
0–60 μM H460:
~30 μM (48 h)

A549:
~40 μM (48 h)
[39]
Pc9-BrM3 & H2030-BrM3 (brain metastatic) Inhibit cell proliferation and cell invasion: ↓ STAT3 protein phosphorylation; ↓ STAT-3 mediated mitochondrial respiratory function 0–50 μM PC9-BrM3:
28.4 μM (48 h)

H2030-BrM3:
25.7 μM (48 h)
[40]
H23, A549 & HCC827 Inhibit cell growth
Induce G1 phase cell cycle arrest: ↓EGFR; ↓ class I HDAC; ↓ class IIb HDAC6 activity; ↑ Hsp90 acetylation & EGFR degradation
0–40 μM A549:
23.55 μM (24h)
[41]
H460, A549, H358 Inhibit cell growth: ↓ c-RAF, ERK & AKT phosphorylation
Inhibit colony formation capacity
Induce apoptosis: ↑ Bax protein; ↓ Bcl-2 protein; ↑ PARP cleavage
Induce G1 phase cell cycle arrest: ↓ cyclin D1; ↑ p21 & p27; ↓ P70S6k kinase activity
Induce autophagy: ↑ LC3-I conversion to LC3-II; ↑ Sirt3 mRNA & protein; ↓ Hif-1α protein
0–80 μM H460:
30.42 μM (72 h)
A549:
50.58 μM (72 h)
H358:
59.38 μM (72 h)
[42]
A549 & 95-D Inhibit cell viability
Induce apoptosis: ↑ ER stress signalling pathway activation; ↑ GRP78, phosphorylation PERK & phosphorylated IRE1α; ↑ cleaved caspase-9 & CHOP; ↓ Bcl-2 protein; ↑ Bax, caspase-3 & caspase-9
Inhibit cell migration
0–60 μM N/A [43]
CH27, H460 & H1299 Inhibit cell growth
Induce apoptosis: ↓ Bcl-XL; ↑ mitochondrial cytochrome c release; ↑ BAD protein; ↑ caspase-1, caspase-2, caspase-3, caspase-6, caspase-8 & caspase-9 activity; ↑ PARP cleavage
0–100 μM CH27:
40.9 μM (24 h)

H460:
41.4 μM (24 h)

H1299:
34.7 μM (24 h)
[17]
MSTO-211H Inhibit cell viability
Induce apoptosis: ↑ PARP cleavage; ↑ caspase-3 activation; ↓ Bid & Bcl-xL protein; ↑ Bax protein; ↓ Mcl-1 & survivin protein; ↓ Sp1
Induce G1 phase cell cycle arrest: ↓ cyclin D1
0–22.5 μM N/A [44]
Skin cancer SK-MEL2 & MeWo Inhibit cell growth & cell proliferation
Induce apoptosis via DNA degradation
Induce cell death via mitochondrial depolarization
0–100 μM N/A [45]
A431 Inhibit cell viability & proliferation
Induce G0/G1 phase cell cycle arrest: ↓ cyclin A, cyclin D1, cyclin D2 & cyclin E; ↓ Cdk2, Cdk4 & Cdk6; ↑ p21 & p27
Induce cell apoptosis: ↑ PARP
0–75 μM N/A [46]
B16-F10 Inhibit cell proliferation
Induce cell death: ↑ Autophagosome (vacuoles) formation; ↓ cyclin D1; ↓ AKT/mTOR & Notch signalling
0–50 μM N/A [47]
B16/F-10 & SKMEL-28 Inhibit cell proliferation & viability: ↓ Notch signalling; ↓ TACE & γ-secretase complex proteins
Inhibit clonogenicity
Induce G0/G1 phase cell cycle arrest
Induce autophagy: ↑ autophagosome formation; ↑ LC3B cleavage
Inhibit cell stemness: ↓ CD271, CD166, Jarid1B & ABCB5
0–60 μM N/A [48]
UACC903 Inhibit cell growth & proliferation 0–50 μM 7.45 μM (24 h)
5.10 μM (72 h)
[27]
SKMEL-2 Inhibit cell proliferation & viability
Induce apoptotic death: ↑ caspase-3, caspase-6, caspase-8 & caspase-9; ↑ PARP cleavage; ↓ procaspase-3, procaspase-8 & procaspase-9
Induce G2/M phase cell cycle arrest: ↓ cyclin B1, cyclin D1, cyclin D2 & PCNA; ↓ Cdk2 & Cdk4; ↑ p21 & p53
0–100 μM N/A [49]
UACC-62 Inhibit cell proliferation & viability
Induce apoptotic death: ↑ caspase-3, caspase-6, caspase-8 & caspase-9; ↑ cleaved PARP; ↓ procaspase-3, procaspase-8 & procaspase-9
Induce G0/G1 phase cell cycle arrest: ↓ cyclin B1, cyclin D1 & cyclin D2; ↓ Cdk2, Cdk4 & Cdc2p34; ↓ p21 & p27
0–100 μM N/A [49]
Renal cancer A498 Inhibit cell proliferation
Inhibit colony formation capability
Inhibit cell migration and invasion: ↓ Epithelial-mesenchymal transition (EMT); ↓ cancer stem cells (CSC) properties; ↑ miR-141; ↓ ZEB2
Inhibit tumoursphere formation
0–80 μM ~12 μM (72 h) [50]
Cervix cancer KB-3-1, KB-8-5, KB-C1, KB-V1 Inhibit cell viability: ↓ EGFR-STAT3 signalling
Induce mitochondria-dependent & death receptor-dependent apoptosis: ↓ Bcl-2, Mcl-1 & survivin; ↑ PARP & caspase-3 cleavage; ↑ mitochondrial release of cytochrome c; ↑ DR5
Enhances in vitro cytotoxicity of Paclitaxel
0–75 μM KB-3-1:
12.56 μM (72 h)
KB-8-5:
12.08 μM (72 h)
KB-C1:
11.40 μM (72 h)
KB-V1:
10.39 μM (72 h)
[23]
Pancreatic cancer MiaPaCa & Colo-357 Suppress plating efficiency of cells
Reduce anchorage-independent clonogenicity growth
Suppress migration and invasion ability
0–5 μM N/A [51]
MiaPaCa & Panc1 Inhibit cell growth
Induce G1 phase cell cycle arrest: ↓ cyclin D1 & cyclin E; ↓ Cdk2 & Cdk4; ↑ p21 & p27
Induce apoptosis: ↓ Bcl-2 & Bcl-xL proteins; ↑ Bax protein; ↓ IKB-α phosphorylation; ↓ NF-κB constitutive activation
0–60 μM MiaPaCa:
43.25 μM (24 h)
31.08 μM (48 h)
18.54 μM (72 h)

Panc1:
47.44 μM (24 h)
34.17 μM (48 h)
21.86 μM (72 h)
[52]
Thyroid cancer ARO, WRO Inhibit cell growth & proliferation: ↓ ERK, JNK & p37 activation and expression; ↓ mTOR & p70S6K
Inhibit colony formation
Induce apoptosis: ↑ PARP cleavage; ↑ caspase-3, caspase-8 & PARP activation; ↓ PI3K/AKT & MAPK pathways
Induce G0/G1 cell cycle arrest: ↓ cyclin D1; ↓ Cdk2 & Cdk4; ↑ p21 & p27
Induce autophagy & autophagy flux: ↑ LC3-II
ARO & WRO:
0–60 μM

SW579:
0–40 μM
ARO:
36.3 μM (24 h)
40.1 μM (48 h)
44.8 μM (72 h)

WRO:
37.7 μM (24 h)
31.8 μM (48 h)
30.7 μM (72 h)

SW579:
19.9 μM (24 h)
10.5 μM (48 h)
8.8 μM (72 h)
[53]
Nasopharyngeal cancer HNE-1 Inhibit cell growth
Induce apoptosis
Induce G1 phase cell cycle arrest
0–150 μM (Honokiol & ATNH—Active targeting nanoparticles-loaded honokiol) Honokiol:
144.71 μM (24 h)

ATNH:
69.04 μM (24 h)
[22]
Brain cancer U251 Inhibit cell growth
Inhibit cell proliferation
Induce apoptosis
0–120 μM 61.43 μM (24 h) [54]
T98G Inhibit cell viability
Inhibit cell invasion
Induce cell apoptosis: ↑ Bax protein; ↓ Bcl-2; ↑ Bax/Bcl-2 ratio
0–50 μM N/A [55]
GBM8401 (Parental) &
GBM8401 SP
Inhibit cell proliferation & viability
Induce sub-G1 phase cell cycle arrest
Induce apoptosis: ↓ Notch3/Hes1 pathway
0–20 μM GBM8401 (Parental):
5.30 μM (48 h)

GBM8401 SP:
11.20 μM (48 h)
[29]
U251 & U-87 MG Inhibit cell viability & proliferation: ↓ PI3K/Akt & MAPK/Erk signalling pathways
Inhibit cell invasion & migration: ↓ MMP2 & MMP9; ↓ NF-κB-mediated E-cadherin pathway
Inhibit colony formation
Induce apoptosis: ↓ Bcl-2, p-AKT & p-ERK; ↑ Bax protein; ↑ caspase-3 cleavage; ↓ EGFR-STAT3 signalling
Reduce spheroid formation: ↓ CD133 & Nestin protein
0–60 μM U251:
54.00 μM (24 h)

U-87 MG:
62.50 μM (24 h)
[56]
DBTRG-05MG Inhibit cell growth
Induce apoptosis: ↓ Rb protein; ↑ PARP & Bcl-x(S/L) cleavage
Induce autophagy: ↑ Beclin-1 & LC3-II
0–50 μM ~30 μM [57]
U87 MG (Human)

BMEC (Mouse)
Inhibit cell viability
Inhibit epithelial-mesenchymal transition (EMT): ↓ Snail, β-catenin & N-cadherin; ↑ E-cadherin
Inhibit cell adhesion & invasion: ↓ VCAM-1; ↓ phosphor-VE-cadherin-mediated BMEC permeability
0–20 μM U87MG:
22.66 μM (24 h)

BMEC:
13.09 μM (24 h)
[58]
U87 MG Inhibit cell viability
Induce G1 phase cell cycle arrest: ↑ p21 & p53; ↓ cyclin D1; ↓ Cdk4 & Cdk6; ↓ p-Rb protein; ↓ E2F1
Induce apoptosis: ↓ procaspase-3; ↑ caspase-8 & caspase-9 activity
0–100 μM 52.70 μM [59]
Bone cancer HOS & U20S Inhibit cell proliferation
Inhibit colony formation
Induce G0/G1 phase cell cycle arrest: ↓ cyclin D1 & cyclin E; ↓ Cdk4
Induce mitochondria-mediated apoptosis: ↑ caspase-3 & caspase-9 activation; ↑ PARP cleavage; ↓ Bcl-2, Bcl-xL & survivin; ↑ ERK activation; ↓ proteasome activity; ↑ ER stress and subsequent ROS overgeneration; ↑ GRP78
Induce autophagy: ↑ Atg7 protein activation; ↑ Atg5; ↑ LC3B-II
0–30 μM HOS:
17.70 μM (24 h)

U20S:
21.50 μM (24 h)
[60]
SAOS-2, HOS, 143B, MG-63 M8, HU09, HU09 M132

Dunn, LM5, LM8 & LM8-LacZ (Mouse)
Inhibit cell metabolic activity
Inhibit cell proliferation
Inhibit cell migration
Induce rapid cell death via Honokiol-provoked vacuolation
0–150 μM (72 h)
SAOS-2:
48.38 μM
HOS:
51.38 μM
143B:
41.63 μM
MG-63M8:
34.88 μM
HU09:
59.25 μM
HU09M132:
31.88 μM

(72 h)
Dunn:
36.00 μM
LM5:
30.00 μM
LM8:
31.13 μM
[61]
Saos-2 & MG-63 Inhibit cell viability
Induce apoptosis: ↑ caspase-3 & PARP cleavage; ↑ Bax protein; ↓ Bcl-2; ↓ PI3K/AKT signalling pathway; ↓ miR-21
0–100 μM Saos-2:
37.85 μM (24 h)

MG-63:
38.24 μM (24h)
[62]
Oral cancer OC2 & OCSL Inhibit cell growth
Induce G0/G1 phase cell cycle arrest: ↑ cyclin E accumulation; ↑ p21 & p27; ↓ cyclin D1, ↓ Cdk2 & Cdk4
Induce apoptosis: ↓ caspase-8 & caspase-9; ↑ caspase-3 cleavage; ↓ Bid protein
Induce autophagy and autophagic flux: ↑ LC3-II; ↓ Akt/mTORC1 pathway; ↑ AMPK signalling pathway; ↑ p62
0–60 μM OC2:
35.00 μM (24 h)
22.00 μM (48 h)

OCSL:
33 μM (24 h)
13 μM (48 h)
[18]
HN-22 & HSC-4 Inhibit cell viability
Induce apoptosis: ↓ Sp1 protein; ↑ p21 & p27; ↑ PARP & caspase-3 activation; ↓ Mcl-1 & survivin protein
Induce G1 phase cell cycle arrest: ↓ cyclin D1
0–37.5 μM HN-22:
26.63 μM (48 h)

HSC-4:
30.00 μM (48 h)
[63]
Liver cancer HepG2 Inhibit cell growth & proliferation: ↓ β-catenin protein
Induce apoptosis: ↑ BAD protein; ↓ Bcl-2 protein
Upregulation of BAD protein expression
Downregulation of Bcl-2 protein level
0–2 μM N/A [64]
SMMC-7721 Inhibit cell growth
Induce G0/G1 phase cell cycle arrest
Induce apoptosis: ↓ mitochondrial potential; ↑ ROS production; ↓ Bcl-2 protein; ↑ Bax protein
0–37.5 μM N/A [65]
HepG2, HUH7, PLC/PRF5, Hep3B Inhibit cell proliferation: ↓ STAT3 activation; ↓ IL-induced Akt phosphorylation; ↓ c-Src activation; ↓ JAK1 & JAK2; ↑ SHP-1 protein
Induce sub-G1 phase cell cycle arrest: ↓ cyclin D1
Downregulation of cyclin D1 level
Induce apoptosis: ↓ Bcl-2 & Bcl-xL; ↓ survivin & Mcl-1 protein; ↑ caspase-3 activation; ↑ PARP cleavage
Enhance apoptotic effect of doxorubicin & paclitaxel
0–100 μM N/A [24]
Ovarian cancer A2780s & A2780cp Inhibit cell growth
Induce apoptosis
0–100 μM A2780s:
36.00 μM (48 h)

A2780cp:
34.70 μM (48 h)
[66]
SKOV3 & Caov-3 Inhibit cell proliferation and growth
Inhibit colony formation
Induce apoptosis: ↑ AMPK pathway activation; ↑ caspase-3, caspase-7 & caspase-9 activation; ↑ PARP cleavage
Induce G0/G1 phase cell cycle arrest
Inhibit cell migration and invasion
0–100 μM SKOV:
48.71 μM (24 h)

Caov-3:
46.42 μM (24 h)
[20]
SKOV3, COC1, Angelen & A2780 Inhibit cell proliferation
Induce cell apoptosis: ↓ Bcl-xL; ↑ BAD protein; ↑ caspase-3 activation
Induce G1 phase cell cycle arrest
0–93.75 μM SKOV3:
62.63 μM (24 h)

COC1:
73.50 μM (24 h)

Angelen:
61.50 μM (24 h)

A2780:
55.85 μM (24 h)
[67]
Prostate cancer PC-3 & LNCaP Inhibit cell viability
Induce G0/G1 phase cell cycle arrest: ↓ cyclin D1 & cyclin E; ↓ Cdk2, Cdk4 & Cdk6; ↑ p21 & p53; ↓ Rb & E2F1 proteins; ↓ Rb phosphorylation at Ser807/811; ↑ ROS generation
0–60 μM N/A [68]
PC-3, LNCaP & C4-2 Inhibit cell growth
Induce apoptosis: ↑ caspase-3, caspase-8 & caspase-9 activation; ↑ PARP cleavage
Induce apoptosis via DNA fragmentation: ↑ Bax & Bak proteins; ↓ Mcl-1 protein
0–75 μM 18.75–37.50 μM (24 h) [69]
PC-3, LNCaP Inhibit cell viability
Induce autophagy: ↑ LC3-BII protein; ↓ mTOR pathway
Induce apoptosis via DNA fragmentation: ↑ ROS generation
0–40 μM N/A [70]
Head & neck squamous cancer Cal-33 & MD-1483 Inhibit cell growth
Induce cell apoptosis and cell cycle arrest: ↓ EGFR signalling pathway; ↓ STAT3 signalling pathway; ↓ Bcl-xL & cyclin D1; ↓ phosphorylation p42/p44 MAPK & phosphorylated Akt
0–100 μM Cal-33:
3.80 μM (72 h)

1483:
7.44 μM (72 h)
[71]
Neuroblastoma Neuro-2a Induce apoptosis via DNA fragmentation: ↑ caspase-3, caspase-6 & caspase-9 activation; ↑ Bax protein; ↓ mitochondrial membrane potential; ↑ cytochrome c releaseInduce sub-G1 phase cell cycle arrest 0–100 μM 63.3 μM (72 h) [72]
Neuro-2a & NB41A3 Inhibit cell viability
Induce autophagy: ↑ LC3-II; ↑ PI3K/Akt/mTOR signalling pathway; ↑ Grp78; ↑ ROS generation; ↑ ERK1/2; ↑ p-ERK1Induce apoptosis via DNA fragmentation
Inhibit cell migration
0–100 μM Neuro-2a:
~50 μM (72 h)
[73]
Bladder cancer T24 & 5637 Inhibit cell viability and induce apoptosis: ↑ Bax protein; ↑ PARP cleavage; ↓ Bcl-2 protein
Inhibit clonogenicity
Induce G1 phase cell cycle arrest: ↓ cyclin D1; ↑ p21 & p27
Inhibit sphere formation capacity
Inhibit cell migration & invasion: ↓ EZH2 gene expression; ↓ MMP9
Inhibit cell stemness: ↓ EZH2 gene expression; ↓ CD44 & Sox2; ↑ miR-143 overexpression
0–72 μM N/A [74]

2.2. In Vivo Studies

Based on the in vivo studies, honokiol possessed the capability to inhibit tumour growth, metastasis, and angiogenesis using different animal models, as shown in Table 2. The degree of tumour inhibition was shown to be significantly effective against each distinct cancer cell line, ranging from 0–150 mg/kg via various delivery methods of honokiol between oral gavage or injection (intraperitoneal, caudal vein, or intravenous). Honokiol was shown to downregulate the expression of Oct4, Nanog, and Sox2 which were known to be expressed in osteosarcoma, breast carcinoma and germ cell tumours [34]. According to Wang et al.’s study, they have found that the average tumour size was significantly lower than the control group without affecting their body weight, suggesting inconsequential toxicity under tested conditions when treated with a combination of honokiol and paclitaxel [23]. Indisputably, honokiol was once again proven to exhibit minor to no toxicity against normal cells.
Over the years, the development of chemo-resistance in ovarian cancer cells has hindered the outcome of treatment regimen towards ovarian cancer [75]. Despite the effectiveness of honokiol to inhibit cancer cell proliferation, delivering effective concentration towards the tumour site was deemed challenging due to its water insolubility [66]. The encapsulation of honokiol in liposome, namely Lipo-HNK by Luo and his team has displayed substantial efficacy against cisplatin-resistance ovarian cancer cell line A2780cp. The tumour volume for Lipo-HNK treated mice was 408 ± 165 mm3 compared to liposome-treated mice and control mice were 2575 ± 701 mm3 and 2828 ± 796 mm3 respectively after 21 days [66]. In addition, Lipo-HNK was also shown to prolong survival and induce intra-tumoral apoptosis in vivo. The promising in vivo properties of honokiol should consolidate its importance as a potential anticancer agent for future researches.
Zebrafish (Danio rerio) model has emerged as a newly important cancer model that complements against traditional cell culture assays and mice model due to its small size, heavy brood, and rapid maturation time. Importantly, its transparent body wall enables visibility of tumour progression and the ease of experimentation [76][77]. It was known that juvenile zebrafish (Danio rerio) or zebrafish embryos have the competency to study cancer cell invasion, metastasis, tumour-induced angiogenesis. Honokiol reduced U-87 MG human glioma/glioblastoma cell proliferation and migration in zebrafish yolk sac and in vivo xenograft nude mouse model [56]. These observations are associated with a reduction in EGFR, phosphorylated STAT3, CD133 and Nestin levels, thus highlighting the regulation of honokiol in EGFR-mediated STAT3/JAK signalling pathway to induce anti-tumour and anti-metastasis.
The subsections below will further discuss the mechanism of anticancer actions of honokiol including the induction of cancer cell death, inhibition of cell cycle progression, induction of autophagy, prevention of epithelial–mesenchymal transition (EMT), as well as the suppression of migration, invasion, and angiogenesis of cancer cells.
Table 2. The antitumour effect of honokiol in in vivo tumour bearing animal models.
Cancer Cell Line Animal Model & Site of Tumour Xenograft Dose, Duration & Route of Administration Observation & Mechanism of Action Efficacy on Tumour Inhibition References
Breast cancer
MDA-MB-231 cells Both flanks of athymic nude mice 100 mg/kg/day
28 days
IP
Induce tumour growth arrest Complete arrest of tumour growth from week 2 onwards [32]
MDA-MB-231 cells Right gluteal region of athymic nude mice 3 mg/mouse/day
Three times a week
28 days
IP
Inhibit tumour progression:
↓ Ki-67; ↑ LKB1 & pAMPK; ↑ ACC phosphorylation, ↓ pS6K & 4EBP1 phosphorylation
Tumour weight of honokiol-treated group was 0.22 g compared to control group which was 1.58 g [33]
MDA-MB-231-pLKO.1 & MDA-MB-231-LKB1shRNA cells Right gluteal region of athymic nude mice 3 mg/mouse/day
Three times a week
42 days
Oral gavage
Inhibit cell stemness: ↓ Oct4, Nanog & Sox2; ↓ pSTAT3 & Ki-67
Inhibit mammosphere formation
Decreased expression of Oct4, Nanog, Sox2

Reduce number of tumour cells showing Ki-67 & pStat3 expression
[34]
Colorectal cancer
RKO cells Axilla of BALB/c nude mice 80 mg/kg/day
Treatment on days 8–11, 14–17, 21–24, 28–31
51 days
IP
Inhibit tumour growth
Prolong survival of mice
709.9% increase in tumour growth rate in honokiol-treated group compared to 1627.6% and 1408.2% in control and vehicle groups respectively [25]
HCT116 cells Flank of athymic nude mice 200 μg/kg/day + 5 Gy irradiation
Once a week
21 days
IP
Inhibit tumour growth: ↓ CSC proteins → ↓ DCLK1, Sox-9, CD133 & CD44 Significantly lower tumour weight (<800 mg) in honokiol-IR combination, (~1500 mg) in honokiol treatment group compared to (~3300 mg) in control group [28]
Cervical cancer
KB-8-5 cells Athymic nu/nu nude mice (site of xenograft not stated) 50 mg/kg Honokiol
Three times a week
+
20 mg/kg Paclitaxel
Once a week
28 days
IP (honokiol)
Tail vein injection (paclitaxel)
Suppress tumour growth: ↓ Ki-67 tissue level
Induce apoptosis
Significantly lower average tumour volume for honokiol-paclitaxel combination treatment (573.9 mm3) compared to control (2585.4 mm3) [23]
Lung cancer
H2030-BrM3 cells Left ventricle of NOD/SCID mice 2 or 10 mg/kg/day
28 days
Oral gavage
Prevent metastasis of lung cancer cells to brain 10 mg/kg: Decrease brain metastasis for >70% [40]
H2030-BrM3 cells Left lung via left ribcage of athymic nude mice 2 or 10 mg/kg/day
Five days a week
28 days
Oral gavage
Decrease lung tumour growth
Inhibit metastasis to lymph node
10 mg/kg: Significantly reduce incidence of mediastinal adenopathy, decrement of weight of mediastinal lymph node for >80%, only 2/6 mice have lymphatic metastasis [40]
Blood cancer
Raji cells Back of BALB/c nude mice 5 mg/20 g &
10 mg/20 g
Treatment on days 8–12 & 15–19
20 days
(Route of administration not specified)
Inhibit cell proliferation
Inhibit tumour growth
Tumour growth of honokiol-treated mice was significantly lower (~90 cm3) compared to control mice (~270 cm3) [31]
HL60 cells Inoculated intraperitoneally into SCID mice 100 mg/kg/day
Treatment on Day 1–6
47 days
IP
Prolong survival of mice Median survival time of honokiol-treated mice are longer (37.5 days) compared to vehicle-treated mice (24.5 days) [78]
Pancreatic cancer
MiaPaCa cells Pancreas of immunocompromised mice 150 mg/kg/day
28 days
IP
Suppress tumour growth
Inhibit metastasis: ↓ CXCR & SHH; ↓ NF-κB & downstream pathway
Inhibit desmoplastic reaction:
↓ ECM protein; ↓ collagen I
Significant decrease in tumour growth for honokiol-treated mice (99.6 mm3) compared to vehicle-treated mice (1361.0 mm3) [51]
Skin cancer
SKMEL-2 or UACC-62 cells Right flank of athymic nude mice 50 mg/kg
Three times a week
14–54 days
IP
Decrease tumour growth SKMEL-2:
40% reduction in tumour volume

UACC-62:
50% reduction in tumour volume
[49]
Thyroid cancer
ARO cells BALB/cAnN.Cg-Foxn1nu/CrlNarl mice (site of xenograft not stated) 5 or 15 mg/kg/mouse
Every three days
21 days
Oral gavage
Decrease tumour volume & tumour weight
Induce apoptosis & autophagy
Control: ~1000 mm3; 700 mg
5 mg/kg Honokiol:
~600 mm3; 400 mg
15 mg/kg Honokiol:
~400 mm3; 200 mg
[53]
Nasopharyngeal cancer
HNE-1 cells Right dorsal aspect of right foot of BALB/c athymic nude mice Active-targeting nanoparticles-loaded HK (ATNH), Non-active-targeting nanoparticles-loaded HK (NATNH), Free Honokiol (HK)

3 mg/mouse/day
Every three days
Euthanise 50% mice after 12 days, rest are left to observe tumour growth & survival time up to 60 days;
IV
Inhibit tumour progression, Induce apoptosis
Potential inhibitor of angiogenesis & proliferation
Efficiency in tumour delay:
ATNH > NATNH > Free HK

Median survival time:
Control: 28.5 days
Free HK: 34 days
NATNH: 42.5 days
ATNH: 57.5 days
[22]
Brain cancer
U21 cells Right flank of athymic nude mice 20 mg/kg
Twice a week
27 days
Caudal vein injection
Inhibit tumour growth
Inhibit angiogenesis
Honokiol-treated mice have significant inhibition of tumour volume by 50.21% compared to vehicle

Significantly lower microvessel present in honokiol-treated cells
[54]
U-87 MG cell suspension pre-treated with honokiol or vehicle for 48h Yolk sac of Zebrafish larvae (Concentration N/A)
3 days
Injection of cells into zebrafish
Inhibit cell proliferation
Inhibit cell migration
Reduced number of cell mass compared to vehicle-treated cells [56]
U-87 MG cells Right flank near upper extremity of nude mice 100 mg/kg/day
Treatment at days 1–7
21 days
IP
Reduce tumour growth:
↓ EGFR, pSTAT3, CD133 & Nestin
Increased number of apoptotic cells in honokiol-treated tissue, Significantly lower tumour volume & tumour weight in honokiol-treated mice [56]
Bone cancer
HOS cells Dorsal area of BALB/c-nu mice 40 mg/kg/day
7 days
IP
Reduce tumour growth
Induce apoptosis & autophagy: ↑ cleaved caspase-3; ↑ LC3B-II & phosphor-ERK (ROS/ERK1/2 signalling pathway)
Significant decrease in tumour volume & weight of honokiol-treated mice (200 mm3; 0.2 g) compared to control group (~500 mm3; 0.5 g)
Increased number of TUNEL-positive cells
[18]
LM8-LacZ cells Left flank of C3H/HeNCrl mice 150 mg/kg/day
25 days;
IP
Inhibit metastasis Mean number of micrometastases decreased significantly by 41.4% in honokiol-treated mice compared to control mice [61]
Oral cancer
SAS cells Right flank of BALB/cAnN.Cg-Foxn1nu.CrlNarl nude mice 5 mg/kg or 15 mg/kg
Treatment on day 1, 4, 7, 10, 13, 16, 19, 22
35 days
Oral
Reduce tumour growth & volume Significantly reduction in tumour growth in honokiol-treated mice

29% reduction (5 mg/kg; 21 days), 40% reduction (15 mg/kg; 21 days)

41% reduction (5 mg/kg; 35 days), 56% reduction (15 mg/kg; 35 days)
[18]
Prostate cancer
C4-2 cells Bilateral tibia of BALB/c nu/nu athymic nude mice 100 mg/kg/day
42 days
IP
Inhibit cell proliferation:
↑ Ki-67
Induce apoptosis: ↑ M-31
Inhibit angiogenesis: ↑ CD-31
Lower PSA value in honokiol-treated mice compared to control group [69]
PC-3 cells Left & right flanks above hind limb of nude mice 1 or 2 mg/mice
Monday, Wednesday & Friday two weeks before tumour implantation and duration of experiment after implantation
77 days
Oral gavage
Inhibit tumour growth
Inhibit cell proliferation
Inhibit neovascularisation
Induce apoptosis
Tumour volume of honokiol-treated mice are significantly lower (~330 mm3; 1 mg), (~50 mm3; 2 mg) compared to control (~400 mm3) [79]
Gastric cancer
MKN45 cells Dorsal side of BALB/c nude mice (nu/nu) 0.5 mg/kg/day & 1.5 mg/kg/day
10 days
Injection (route not stated)
Inhibit tumour growth: ↓ GRP94 overexpression 30% reduction in tumour volume
(0.5 mg/kg)
60% reduction in tumour volume
(1.5 mg/kg)

Decreased accumulation of GRP94
[80]
MKN45 & SCM-1 cells Peritoneal cavity of BALB/c nude mice 5 mg/kg
Twice a week
28 days
IP
Inhibit metastasis
Inhibit angiogenesis
Honokiol inhibited STAT-3 signalling and VEGF signalling induced by calpain/SHP-1 [81]
Ovarian cancer
SKOV3 cells Right axilla of BALB/c nude mice 1 mg liposome-encapsulated honokiol/day
48 days
IP
Inhibit tumour growth
Inhibit angiogenesis
Reduction in tumour growth rate in liposome-encapsulated honokiol-treated mice by 67–70% compared to control [66][82]
A2780s cells Right flank of athymic BALB/c nude mice 10 mg/kg Lipo-Honokiol
Twice a week
21 days
IV
Inhibit cancer growth
Prolong survival of mice
Increase intra-tumoural apoptosis
Inhibit intra-tumoural angiogenesis
Lipo-HNK treated mice have significantly smaller tumour volume
(222 ± 71 mm3) compared to liposome-treated mice
(1823 ± 606 mm3) and control mice (3921 ± 235 mm3)
[66]
A2780cp cells Right flank of athymic BALB/c nude mice 10 mg/kg Lipo-Honokiol
Twice a week
21 days
IV
Inhibit cancer growth
Prolong survival
Increase intra-tumoural apoptosis
Inhibit intra-tumoural angiogenesis
Lipo-HNK treated mice have significantly smaller tumour volume
(408 ± 165 mm3) compared to liposome-treated mice
(2575 ± 701 mm3) and control mice (2828 ± 796 mm3)
[66]

3. Mechanism of Action of Honokiol

3.1. Dual Induction of Apoptotic and Necrotic Cell Death

Apoptosis is a normal physiological process that maintains the homeostatic cellular balance in multicellular organisms [83]. Generally, apoptosis can be classified into two central pathways, namely the intrinsic pathway (mitochondrial-mediated pathway) and extrinsic pathway (death receptor-mediated pathway) [84]. The intrinsic pathway is associated with changes in mitochondrial membrane permeability that lead to imbalance in Bax/Bak ratio and release of cytochrome c and other mitochondrial proteins into cytosol [83][84]. The released cytochrome c interacts with apoptosis protease-activating factor 1 (Apaf1) and forms an apoptosome complex [85], which promotes the activation of caspase-9 and later caspase-3, initiating the caspase cascade, which executes cell death in a coordinated way [85]. For the extrinsic pathway, the binding of ligands such as tumour necrosis factor (TNF), Fas ligand (Fas-L), and TNF-related apoptosis-inducing ligand (TRAIL) to their respective death receptors (type 1 TNF receptor (TNFR1), Fas (also called CD95/Apo-1) and TRAIL receptors will turn procaspase-8 into active caspase-8 to induce apoptosis [85][86][87].
Honokiol has been shown tp initiate caspase-dependent apoptotic pathways in different types of cancer (Table 1). Chen et al. [14] found that JJ012 human chondrosarcoma cells lose their mitochondrial membrane potential when treated at 10 µM of honokiol, thus leading to apoptosis. Other studies have also shown that honokiol markedly disrupted the balance of Bax/Bcl-2 ratio [13][79][26][56][88][89][90][91]. The increasing ratio of proapoptotic to antiapoptotic Bcl-2 family proteins (Bax/Bcl-2) will induce the release of cytochrome c and other apoptogenic proteins through the mitochondrial membrane to the cytosol, subsequently leading to the activation of caspase cascade and apoptosis [26]. Furthermore, honokiol downregulated the expression of several other anti-apoptosis mRNA and proteins such as Bcl-xL [13][79][17][57], survivin [60][92], and MCL-1 [79], as well as upregulated other pro-apoptotic proteins such as BAD, BAX, and BAK proteins [79][17].
Moreover, honokiol has been shown to effectively induce apoptosis in p53-deficient cancer cells, such as MDA-MD-231 breast cancer cells, as well as lung and bladder cancer cell lines by inhibiting the activation of ras-phospholipase D [32][93][94]. Besides p53, other tumour suppressor genes that will be activated in honokiol treatment include p21 [46], p21/waf1 [95], p27 [46], p38 MAPK [96][97], and p62 [18][39].
Besides the intrinsic pathway, honokiol is capable of targeting death receptors TNF-related apoptosis-inducing ligand (TRAIL) receptors and tumour necrosis factor receptors (TNFR) resulting in a sequential activation of caspase-8 and -3, which cleaves target proteins and then leads to apoptosis [98][99][100]. Activation of the death receptor mediated apoptotic pathway is primarily inhibited by cellular-caspase-8/FADD-like IL-1β-converting enzyme (FLICE) inhibitory protein (c-FLIP), which inhibits caspase-8 activation by preventing the recruitment of caspase-8 to the death inducing signalling complex [100]. However, honokiol was able to downregulate c-FLIP through the ubiquitin/proteasome-mediated mechanism, resulting in the sensitisation of non-small cell lung cancer cells to TRAIL-mediated apoptosis [101][102].
Other than intrinsic and extrinsic pathways, honokiol can also induce apoptosis by the endoplasmic reticulum (ER) stress-induced mechanism. A variety of ER stresses result in unfolded protein accumulation responses [103][104]. For survival, the cells induce ER chaperone proteins to increase protein aggregation, temporarily halt translation, and activate the proteasome machinery to degrade misfolded proteins. However, under severe and prolonged ER stress, an unfolded protein response activates unique pathways that lead to cell death through apoptosis [105]. According to a study by Zhu et al. [43], honokiol can upregulate the expressions of ER stress-induced apoptotic signalling molecules such as GRP78, phosphorylated PERK, phosphorylated eIF2α, CHOP, Bcl-2, Bax, and cleaved caspase-9 in human lung cancer cells. Chiu et al. [106] found that honokiol also led to an increase in ER stress activity in melanoma cell lines B16F10 (mouse), human malignant melanoma, and human metastatic melanoma. Honokiol activated ER stress and down-regulated peroxisome proliferator-activated receptor-γ (PPARγ) activity resulting in PPARγ and CRT degradation through calpain-II activity in human gastric cancer cell lines [80][107][108] and human chondrosarcoma cells [14]. This was due to the ability of honokiol to upregulate and bind effectively to the glucose regulated protein 78 (GRP78) to activate apoptosis [14][109]. However, this was opposed by another study where treatment of various human gastric cancer cells with honokiol led to the induction of GRP94 cleavage but did not affect GRP78 [80].
Necrosis is known as unprogrammed cell death whereby cell swelling and destabilisation of the cell membrane results in the leakage of cellular cytoplasmic contents into the extracellular space, thus causing inflammation [110]. Besides apoptosis, honokiol has also been found to induce necrotic cell death in MCF-7 (40 μg/mL honokiol) [111], human oesophageal adenocarcinoma cells CP-A and CP-C [112], and primary human acute myelogenous leukemia HL60 [78] via p16ink4a pathway by targeting cyclophilin D to affect several downstream mechanisms. This phenomenon was also observed in transformed Barrett’s and oesophageal adenocarcinoma cells when treated with honokiol (<40 µM) by targeting their STAT3 signalling pathway, thus resulting in a decrease of Ras activity and phosphorylated ERK1/2 expression [113]. The phosphorylation of Ser727 STAT3 induces translocation towards the mitochondria followed by ROS production, ultimately leading to the induction of necrosis [114]. Taken together, honokiol demonstrates the dual induction of apoptotic and necrotic cell death.

3.2. Cell Cycle Arrest

Cancer is attributed to uncontrolled proliferation resulting from abnormal activity of different cell cycle proteins. Therefore, cell cycle regulators are becoming attractive targets in cancer therapy. Honokiol can induce cell cycle arrest in several types of cancer cells, such as in lung squamous cell carcinoma [115], prostate cancer cells [68][116], oral squamous cancer [63], UVB-induced skin cancer [117], and more as listed in Table 1, by generally inducing G0/G1 and G2/M arrest. This arrest is associated with the suppression of cyclin-B1, CDC2, and cdc25C in honokiol-treated human gastric carcinoma and human neuroglioma cells [91][118][119], downregulation of cyclin dependent kinase (CDK)-2 and CDK-4, and the upregulation of cell cycle suppressors p21 and p27 in human oral squamous cell carcinoma (OSCC) cells [18][91]. In addition, the downregulation of c-Myc and class I histone deacetylases was also identified as other contributors to cell cycle arrest at the G0/G1 phase in prostate cancer cells [91][116] and acute myeloid leukemia respectively [37][95][102].

3.3. Autophagy

Autophagy is an evolutionary conserved catabolic process that involves the delivery of dysfunctional cytoplasmic components for lysosomal degradation [120][121]. The activation of autophagy promotes cell survival and regulates cell growth during harsh and stressful conditions via a reduction of cellular energy requirements by breaking down unnecessary components [75][121]. In cancer cells, autophagy facilitates both tumour suppression and tumourigenesis by the induction of cell death and tumour growth promotion, respectively [122][123]. The regulation of mTORC complexes mTORC1 and mTORC2 is involved in controlling the autophagic process. The activation of mTORC1 plays an important role in phosphorylation of autophagy-related protein (ATG) and subsequently inhibiting autophagy, whereas the inhibition of mTORC1 complements the autophagic process [124][125]. The inhibition of mTORC1 complex will concurrently activate Unc-51-like autophagy-activating kinase (ULK) complex, inducing localisation to the phagophore and followed by class III PI3K activation [126][127]. Beclin-1 was known to play a role in tumour suppression by recruiting several proteins associated with autophagosome elongation and maturation [128]. ATGs regulate the autophagosome elongation. For instance, ATG5-ATG12/ATG16L complexes recruit microtubule-associated protein 1 light chain 3 (LC3), followed by conversion of pro-LC3 to active cytosolic isoform LC3 I by ATG4B [129][130]. Thereafter, the interaction with ATG3, ATG7, and phosphatidylethanolamine (PE) converts LC3 I to LC3 II. The LC3 II enables the autophagosome to bind to degraded substrates and mature autophagosomes are capable of fusing with lysosomes to selectively remove damaged organelles via autophagy [131].
Generally, there are two modes of autophagy known as conventional and alternative autophagy. Conventional autophagy (also known as Atg5/Atg7-dependent pathway) involves the activation of Atg5 and Atg7 which are core regulators of autophagy, and then leads to microtubule-associated protein 1A/1B light chain 3 (LC3) modification and translocation from cytosol to the isolation membrane. This LC3 translocation was considered as a reliable hallmark of autophagy. Contradictorily, alternative autophagy occurs independently without involving Atg5 and Atg7, as well as LC3 modification [122][123][131].
The regulation of autophagy in cancer remains controversial as it plays dual roles in tumour suppression and promotion. Autophagy is believed to contribute to the properties of cancer cells stemness, induction of recurrence, and the development of anticancer drugs. However, the actual mechanism of autophagy in cancer remains unclear. Several studies have highlighted the potential of honokiol to induce cell death via autophagy in human prostate cancer cells [70], human glioma cells [132], NSCLC cells [22], and human thyroid cancer cells [53].
The activation of Atg5/Atg7-dependent pathways through the upregulation of LC3B-II, Atg5, and Atg7 levels was observed in honokiol-treated osteosarcoma HOS and U2OS cells and leads to the accumulation of autophagic vacuoles [18]. According to a study by Chang et al. [57], the expression of two critical autophagic proteins, Beclin-1 and LC3, were found to have increased in the honokiol-treated glioblastoma multiforme cells (DBTRG-05MG cell line). Similarly, the expression of autophagosomal marker LC3-II was also increased in Kirsten rat sarcoma viral oncogene homolog (KRAS) mutated cell lines of non-small cell lung cancer (NSCLC).
Other signalling pathways are also found to be involved in honokiol-induced autophagy including the involvement of AMPK-mTOR signalling pathway which leads to autophagocytosis through the coordinated phosphorylation of Ulk1 in Kirsten rat sarcoma viral oncogene homolog (KRAS) mutant lung cancer and melanoma cells [48][53][59][91]. Besides this, the ROS/ERK1/2 signalling pathway is also believed to play a certain role in honokiol-induced autophagy though ERK activation and the generation of ROS in treated osteosarcoma cells [60][70][91]. All these recent studies have further supported the potential of honokiol in the induction of autophagy in cancer cells.

3.4. Epithelial-Mesenchymal Transition (EMT)

Migratory mesenchymal-like cells are involved in embryonic development, tissue repair, and regeneration, as well as several pathological processes like tissue fibrosis, tumour invasiveness, and metastasis [133][134]. These migratory mesenchymal cells originate from the conversion of the epithelial cells, and this process is known as epithelial-mesenchymal transition (EMT). This plasticity of cellular phenotypes provides a new insight into possible therapeutic interventions in cancer [134].
EMT is characterised by the loss of epithelial markers such as cytokeratins and E-cadherin, followed by an increase in mesenchymal markers such as N-cadherin and vimentin [135]. The cellular processes of EMT are composed of several key transcription factors (such as TWIST, SNAI1, SNAI2, ZEB1/2) that act in concert with epigenetic mechanisms and post-translational protein modifications to coordinate cellular alterations [133][136]. The application of gene expression signatures combining multiple EMT-linked genes has proven useful to evaluate EMT as a contributing factor in tumour development in human cancers. However, the EMT process was shown to be incomplete in tumours, venturing in between multiple translational states and expressing a mixture of both epithelial and mesenchymal genes. This hybrid in partial EMT can be more aggressive than tumour cells with a complete EMT phenotype [135]. In addition, EMT contributes to tumour metastatic progression and resistance towards cancer treatment, resulting in poor clinical outcomes [134][135].
Honokiol has been shown to block and inhibit EMT in many cancer cells such as breast cancer, melanoma, bladder cancer, human non-small cell lung cancer, and gastric cancer (Table 1). Honokiol reduced steroid receptor coactivator-3 (SRC-3), matrix metalloproteinase (MMP)-2, and Twist1, preventing the invasion of urinary bladder cancer cells [102][137]. In addition, honokiol was also capable of inducing E-cadherin and repressing N-cadherin expression, thus inhibiting the EMT process in J82 bladder cancer cells [102][137]. In breast cancer cells, honokiol inhibits the recruitment of Stat3 on mesenchymal transcription factor Zeb1 promoter, resulting in decreased Zeb1 expression and nuclear translocation [138]. In addition, honokiol increases E-cadherin expression via the Stat3-mediated release of Zeb1 from E-cadherin promoter [138]. Collectively, many studies have reported that honokiol effectively inhibits EMT in breast cancer cells, evidence has been found to support a cross-talk between honokiol and Stat3/Zeb1/E-cadherin axis [138]. On the other hand, EMT is inhibited by modulating the miR-141/ZEB2 signalling in renal cell carcinoma (A-498) [50].
Honokiol inhibited the EMT-driven migration of human NSCLC cells in vitro by targeting c-FLIP through N-cadherin/snail signalling as N-cadherin and snail are downstream targets of c-FLIP [139]. Twist1, a basic helix-loop-helix domain-containing transcription factor, promotes tumour metastasis by inducing EMT, and can be upregulated by multiple factors, including SRC-1, STAT3, MSX2, HIF-1α, integrin-linked kinase, and NF-κB. The capability of honokiol in targeting Twist1 can be regarded as a promising therapy for metastatic cancer [102][140].
Honokiol was found to inhibit breast cancer cell metastasis and eliminate human oral squamous cell carcinoma cell by blocking EMT through the modulation of Snail/Slug protein translation [141][142]. Honokiol markedly downregulated endogenous Snail, Slug, and vimentin expression and upregulated E-cadherin expression in MDA-MB-231, MCF7, and 4T1 breast cancer cells [142]. As primary EMT inducers, Snail and Slug dictate the induction of EMT by targeting E-cadherin and vimentin [138][142]. Furthermore, when cells were treated with honokiol, Snail and Slug expression levels were decreased from 12 h to 24 h in a time-dependent manner, suggesting that honokiol can reverse the EMT process via the downregulation of Snail and Slug in breast cancer cell lines [142]. Besides that, EMT was inhibited in human oral squamous cell carcinoma cell via the disruption of Wnt/β-catenin signalling pathway [141]. It was reported that the protein levels of mesenchymal markers such as Slug and Snail were markedly suppressed, while β-catenin and its downstream Cyclin D1 were inhibited [141]. It is known that β-catenin could mediate EMT [141][143], which plays a crucial role in cancer invasion and metastasis. The EMT markers such as Snail and Slug are also the target genes of β-catenin [144]. Therefore, the suppression of Snail and Slug in honokiol treated human oral squamous cell carcinoma cells was believed to be due to the inhibition of Wnt/β-catenin signalling pathway [141]. Similarly, in U87MG human glioblastoma cell and melanoma cells, Snail, N-cadherin and β-catenin expression levels were decreased, whereas E-cadherin expression was increased after honokiol treatment [58][106].

3.5. Suppression of Migration, Invasion and Angiogenesis of Cancer Cells

Metastasis is known to be the major cause of death in cancer patients [145]. It involves the migration and invasion of tumour cells into neighbouring tissues and distant organs via intravasation into blood or lymphatic system [146][147]. The formation of invadopodium was stimulated by epidermal growth factor (EGF) and is crucial for the degradation of the extracellular matrix and remodelling membrane proteins, promoting metastasis [145]. Therefore, one of the important steps in cancer management is to control tumour cell metastasis, especially for early-stage cancer patients [147]. Various studies have reported that honokiol has the capability to suppress tumour metastasis in different types of cancer including breast cancer [33][142][148], non-small cell lung cancer [37][149] ovarian carcinoma cells [20], lung cancer [43], U251 human glioma, as well as U-87MG and T98G human glioblastoma cell [56][58][88], oral squamous cell carcinoma (OSCC) [18], bladder cancer cell [137], pancreatic cancer [51], renal cell carcinoma [150][151], and gastric cancer cells [107]. For instance, the percentage of invading urinary bladder cancer (UBC) cells was significantly reduced by 67% and 92% upon 2.4 μg/mL and 4.8 μg/mL of honokiol treatment, respectively [137]. Similarly, tumour cell migration was inhibited by 38–66% in A549 cells, by 37–62% in H1299 cells, 12% to 58% in H460 cells and 32% to 69% in H226 cells, in a concentration-dependent manner after treatment with honokiol [37].
Furthermore, honokiol also demonstrated an inhibitory effect on the expression of matrix metalloproteinases (MMPs) such as MMP-2 and MMP-9 proteins, which play an essential role in the metastatic process of tumour cells, as well as the regulation of angiogenesis in the maintenance of tumour cell survivability [37][56][137]. MMPs are a group of extracellular matrix degrading enzymes that control various normal cellular processes, such as cell growth, differentiation, apoptosis, and migration [147]. However, MMP activity was increased in many tumour cells. The overexpression of MMP-2 and MMP-9 are associated with pro-oncogenic events such as neovascularisation, tumour cell proliferation, and metastasis because it can degrade the extracellular matrix, basement membranes, and adhesion molecules (intercellular adhesion molecule, ICAM, and vascular cell adhesion molecule) and become invasive [51][147][152].
The transition from an epithelial-to-mesenchymal (EMT) phenotype facilitates the breakdown of extracellular matrix followed by the subsequent invasion of the surrounding tissues in order to enter the bloodstream and/or lymph nodes, and travel to distant organ sites. Once cells have reached the distant organ sites, they undergo mesenchymal-to-epithelial transition and begin the establishment of distal metastasis by the surviving cancer cells followed by the outgrowth of secondary tumours [51][153]. Honokiol has been shown to inhibit the invasion of HT-1080 human fibrosarcoma cells and U937 leukemic cells by inhibiting MMP-9 [154]. In addition, honokiol also reduced the protein levels of MMP2 and MMP9 in U251 human glioma and U-87 MG human glioblastoma cell lines in a dose-dependent manner [56]. The expression of MMP-2 and MMP-9 were also found to be decreased in both honokiol-treated A549 and H1299 cells (NSCLC cell lines), consistent with the decreased nuclear accumulation of β-catenin as both MMP-2 and MMP-9 are the downstream targets of β-catenin [37][155][156]. In the J82 bladder cancer cell, honokiol repressed the expression of SRC-3, MMP-2, and Twist1 genes which were involved in cancer cell invasion [137].
Another proposed mechanism for the inhibitory effects of honokiol on invasion and metastasis is through the liver kinase B1 (LKB1)/adenine monophosphate-activated protein kinase (AMPK) axis. Honokiol treatment increased the expression and cytoplasmic translocation of tumour-suppressor LKB1 in breast cancer cells, which led to the phosphorylation and functional activation of AMPK and resulted in the inhibition of cell invasion and metastasis [33][51]. The activation of AMPK suppresses mTOR signalling, decreasing the phosphorylation of p70 kDA ribosomal protein S6 kinase 1 (p70S6K1) and eukaryotic translation initiation factor 4E (eIF4E)-binding protein (4EBP1). This will ultimately inhibit the reorganisation of the actin cytoskeleton in cells, subsequently inhibiting cell migration [33].
In human renal carcinoma cell (RCC) 786-0, honokiol significantly upregulated the expression of metastasis suppressor gene (KISS-1), genes encoding TIMP metalloproteinase inhibitor 4 (TIMP4), and KISS-1 receptor (KISS-1R). In addition, honokiol markedly suppressed the expression of genes encoding chemokine (C-X-C motif) ligand 12 (CXCL12), chemokine (C-C motif) ligand 7 (CCL7), interleukin-18 (IL18) and matrix metalloproteinase 7 (MMP7). It was proven that honokiol significantly upregulated KISS1 and KISS1R in the 786-0 cells when treated with honokiol since recent studies showed that the activation of KISS1/KISS1R signalling by kisspeptin treatment decreases the motility and invasive capacity of conventional RCC, and overexpression of KISS1 inhibits the invasion of RCC cells Caki-1 [14][157]. In short, the activation of KISS1/KISS1R signalling by honokiol suppresses the multistep process of metastasis, including invasion and colony formation, in RCC cells 786-0 [157].
Angiogenesis is the formation of new blood vessels for supplying nutrients and oxygen to tissues and cells. In tumourigenesis, angiogenesis is important for the development and progression of malignant tumours [158]. The endothelial cells in growing cancer are active due to the release of cell growth and motility promoting proteins, creating a network of blood vessels to overcome its oxygen tension [159]. Vascular endothelial growth factor (VEGF) and fibroblast growth factor-2 (FGF2) are among the factors that play an important role in tumour angiogenesis [147]. In human renal cancer cell lines (786-0 and Caki-1), honokiol induced down-regulation of the expression of VEGF and heme oxygenase-1 (HO-1) via the Ras signalling pathway thus inhibit angiogenesis [160][161].
In retinal pigment epithelial (RPE) cell lines, honokiol inhibited the binding of hypoxia- inducible-factor (HIF) to hypoxia-response elements present on the VEGF promoter, thereby inhibiting the secretion of VEGF protein [162][163]. This decrement of VEGF levels resulted in reduced proliferation of human retinal microvascular endothelial cells (hRMVECs) [162]. Therefore, honokiol is said to possess both anti-HIF and anti-angiogenic properties.
In the overexpression of VEGF-D Lewis lung carcinoma cell-induced tumours in C57BL/6 mice, honokiol was shown to significantly inhibit tumour-associated lymphangiogenesis and metastasis. Furthermore, a remarkable delay in tumour growth and prolonged life span in honokiol-treated mice were also observed [164]. In another study, honokiol inhibited VEGF-D-induced survival, proliferation, and microcapillary tube formation in both human umbilical vein endothelial cells (HUVECs) and lymphatic vascular endothelial cells (HLECs). These observations are believed to be due to the inhibition in Akt and MAPK phosphorylation and downregulation of VEGFR-2 expressions in HUVECs as well as VEGFR-3 of HLECs [95][154][165]. Collectively, honokiol has been shown to exert direct and indirect effects on tumour suppression via anti-metastasis, anti-angiogenesis, and anti-lymphangiogenesis by mainly affecting HIF- and VEGF/VEGFR- dependent pathways. However, an in-depth mechanism of honokiol on the inhibition of metastatic progression and spread should be further explored in the future.

References

  1. Foster, I. Cancer: A cell cycle defect. Radiography 2008, 14, 144–149.
  2. Cabral, C.; Efferth, T.; Pires, I.M.; Severino, P.; Lemos, M.F.L. Natural Products as a Source for New Leads in Cancer Research and Treatment. Evid.-Based Complement. Altern. Med. 2018, 2018, 8243680.
  3. Wu, S.; Zhu, W.; Thompson, P.; Hannun, Y.A. Evaluating intrinsic and non-intrinsic cancer risk factors. Nat. Commun. 2018, 9, 3490.
  4. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34.
  5. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424.
  6. DeVita, V.T.; Canellos, G.P. New therapies and standard of care in oncology. Nat. Rev. Clin. Oncol. 2011, 8, 67–68.
  7. Marqus, S.; Pirogova, E.; Piva, T.J. Evaluation of the use of therapeutic peptides for cancer treatment. J. Biomed. Sci. 2017, 24, 21.
  8. Mitra, S.; Dash, R. Natural Products for the Management and Prevention of Breast Cancer. Evid.-Based Complement. Altern. Med. 2018, 2018, 8324696.
  9. Bayat Mokhtari, R.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination therapy in combating cancer. Oncotarget 2017, 8, 38022–38043.
  10. Robinson, M.M.Z.; Zhang, X. The World Medicines Situation 2011. Traditional Medicines: Global Situation, Issues and Challenges; World Health Organization: Geneva, Switzerland, 2011.
  11. Seelinger, M.; Popescu, R.; Giessrigl, B.; Jarukamjorn, K.; Unger, C.; Wallnöfer, B.; Fritzer-Szekeres, M.; Szekeres, T.; Diaz, R.; Jäger, W.; et al. Methanol extract of the ethnopharmaceutical remedy Smilax spinosa exhibits anti-neoplastic activity. Int. J. Oncol. 2012, 41, 1164–1172.
  12. Amaral, R.G.; dos Santos, S.A.; Andrade, L.N.; Severino, P.; Carvalho, A.A. Natural Products as Treatment against Cancer: A Historical and Current Vision. Clin. Oncol. 2019, 4, 1562.
  13. Arora, S.; Singh, S.; Piazza, G.A.; Contreras, C.M.; Panyam, J.; Singh, A.P. Honokiol: A novel natural agent for cancer prevention and therapy. Curr. Mol. Med. 2012, 12, 1244–1252.
  14. Chen, Y.J.; Wu, C.L.; Liu, J.F.; Fong, Y.C.; Hsu, S.F.; Li, T.M.; Su, Y.C.; Liu, S.H.; Tang, C.H. Honokiol induces cell apoptosis in human chondrosarcoma cells through mitochondrial dysfunction and endoplasmic reticulum stress. Cancer Lett. 2010, 291, 20–30.
  15. Lee, J.D.; Lee, J.Y.; Baek, B.J.; Lee, B.D.; Koh, Y.W.; Lee, W.-S.; Lee, Y.-J.; Kwon, B.-M. The inhibitory effect of honokiol, a natural plant product, on vestibular schwannoma cells. Laryngoscope 2012, 122, 162–166.
  16. Amblard, F.; Delinsky, D.; Arbiser, J.L.; Schinazi, R.F. Facile purification of honokiol and its antiviral and cytotoxic properties. J. Med. Chem. 2006, 49, 3426–3427.
  17. Yang, S.E.; Hsieh, M.T.; Tsai, T.H.; Hsu, S.L. Down-modulation of Bcl-XL, release of cytochrome c and sequential activation of caspases during honokiol-induced apoptosis in human squamous lung cancer CH27 cells. Biochem. Pharmacol. 2002, 63, 1641–1651.
  18. Huang, K.J.; Kuo, C.H.; Chen, S.H.; Lin, C.Y.; Lee, Y.R. Honokiol inhibits in vitro and in vivo growth of oral squamous cell carcinoma through induction of apoptosis, cell cycle arrest and autophagy. J. Cell. Mol. Med. 2018, 22, 1894–1908.
  19. Huang, L.; Zhang, K.; Guo, Y.; Huang, F.; Yang, K.; Chen, L.; Huang, K.; Zhang, F.; Long, Q. Honokiol protects against doxorubicin cardiotoxicity via improving mitochondrial function in mouse hearts. Sci. Rep. 2017, 7, 11989.
  20. Lee, J.S.; Sul, J.Y.; Park, J.B.; Lee, M.S.; Cha, E.Y.; Ko, Y.B. Honokiol induces apoptosis and suppresses migration and invasion of ovarian carcinoma cells via AMPK/mTOR signaling pathway. Int. J. Mol. Med. 2019, 43, 1969–1978.
  21. Olusanya, T.O.B.; Haj Ahmad, R.R.; Ibegbu, D.M.; Smith, J.R.; Elkordy, A.A. Liposomal Drug Delivery Systems and Anticancer Drugs. Molecules 2018, 23, 907.
  22. Yang, B.; Ni, X.; Chen, L.; Zhang, H.; Ren, P.; Feng, Y.; Chen, Y.; Fu, S.; Wu, J. Honokiol-loaded polymeric nanoparticles: An active targeting drug delivery system for the treatment of nasopharyngeal carcinoma. Drug Deliv. 2017, 24, 660–669.
  23. Wang, X.; Beitler, J.J.; Wang, H.; Lee, M.J.; Huang, W.; Koenig, L.; Nannapaneni, S.; Amin, A.R.M.R.; Bonner, M.; Shin, H.J.C.; et al. Honokiol Enhances Paclitaxel Efficacy in Multi-Drug Resistant Human Cancer Model through the Induction of Apoptosis. PLoS ONE 2014, 9, e86369.
  24. Rajendran, P.; Li, F.; Shanmugam, M.K.; Vali, S.; Abbasi, T.; Kapoor, S.; Ahn, K.S.; Kumar, A.P.; Sethi, G. Honokiol inhibits signal transducer and activator of transcription-3 signaling, proliferation, and survival of hepatocellular carcinoma cells via the protein tyrosine phosphatase SHP-1. J. Cell. Physiol. 2012, 227, 2184–2195.
  25. Chen, F.; Wang, T.; Wu, Y.-F.; Gu, Y.; Xu, X.-L.; Zheng, S.; Hu, X. Honokiol: A potent chemotherapy candidate for human colorectal carcinoma. World J. Gastroenterol. 2004, 10, 3459–3463.
  26. He, Z.; Subramaniam, D.; Ramalingam, S.; Dhar, A.; Postier, R.G.; Umar, S.; Zhang, Y.; Anant, S. Honokiol radiosensitizes colorectal cancer cells: Enhanced activity in cells with mismatch repair defects. Am. J. Physiol.-Gastrointest. Liver Physiol. 2011, 301, G929–G937.
  27. Lin, J.M.; Prakasha Gowda, A.S.; Sharma, A.K.; Amin, S. In vitro growth inhibition of human cancer cells by novel honokiol analogs. Bioorg. Med. Chem. 2012, 20, 3202–3211.
  28. Ponnurangam, S.; Mammen, J.M.; Ramalingam, S.; He, Z.; Zhang, Y.; Umar, S.; Subramaniam, D.; Anant, S. Honokiol in combination with radiation targets notch signaling to inhibit colon cancer stem cells. Mol. Cancer Ther. 2012, 11, 963–972.
  29. Lai, I.C.; Shih, P.-H.; Yao, C.-J.; Yeh, C.-T.; Wang-Peng, J.; Lui, T.-N.; Chuang, S.-E.; Hu, T.-S.; Lai, T.-Y.; Lai, G.-M. Elimination of Cancer Stem-Like Cells and Potentiation of Temozolomide Sensitivity by Honokiol in Glioblastoma Multiforme Cells. PLoS ONE 2015, 10, e0114830.
  30. Battle, T.E.; Arbiser, J.; Frank, D.A. The natural product honokiol induces caspase-dependent apoptosis in B-cell chronic lymphocytic leukemia (B-CLL) cells. Blood 2005, 106, 690–697.
  31. Gao, D.Q.; Qian, S.; Ju, T. Anticancer activity of Honokiol against lymphoid malignant cells via activation of ROS-JNK and attenuation of Nrf2 and NF-kappaB. J. BUON 2016, 21, 673–679.
  32. Wolf, I.; O’Kelly, J.; Wakimoto, N.; Nguyen, A.; Amblard, F.; Karlan, B.Y.; Arbiser, J.L.; Koeffler, H.P. Honokiol, a natural biphenyl, inhibits in vitro and in vivo growth of breast cancer through induction of apoptosis and cell cycle arrest. Int. J. Oncol. 2007, 30, 1529–1537.
  33. Nagalingam, A.; Arbiser, J.L.; Bonner, M.Y.; Saxena, N.K.; Sharma, D. Honokiol activates AMP-activated protein kinase in breast cancer cells via an LKB1-dependent pathway and inhibits breast carcinogenesis. Breast Cancer Res. 2012, 14, R35.
  34. Sengupta, S.; Nagalingam, A.; Muniraj, N.; Bonner, M.Y.; Mistriotis, P.; Afthinos, A.; Kuppusamy, P.; Lanoue, D.; Cho, S.; Korangath, P.; et al. Activation of tumor suppressor LKB1 by honokiol abrogates cancer stem-like phenotype in breast cancer via inhibition of oncogenic Stat3. Oncogene 2017, 36, 5709–5721.
  35. Liu, H.; Zang, C.; Emde, A.; Planas-Silva, M.D.; Rosche, M.; Kuhnl, A.; Schulz, C.O.; Elstner, E.; Possinger, K.; Eucker, J. Anti-tumor effect of honokiol alone and in combination with other anti-cancer agents in breast cancer. Eur. J. Pharmacol. 2008, 591, 43–51.
  36. Park, E.J.; Min, H.Y.; Chung, H.J.; Hong, J.Y.; Kang, Y.J.; Hung, T.M.; Youn, U.J.; Kim, Y.S.; Bae, K.; Kang, S.S.; et al. Down-regulation of c-Src/EGFR-mediated signaling activation is involved in the honokiol-induced cell cycle arrest and apoptosis in MDA-MB-231 human breast cancer cells. Cancer Lett. 2009, 277, 133–140.
  37. Singh, T.; Katiyar, S.K. Honokiol Inhibits Non-Small Cell Lung Cancer Cell Migration by Targeting PGE2-Mediated Activation of β-Catenin Signaling. PLoS ONE 2013, 8, e60749.
  38. Singh, T.; Prasad, R.; Katiyar, S.K. Inhibition of class I histone deacetylases in non-small cell lung cancer by honokiol leads to suppression of cancer cell growth and induction of cell death in vitro and in vivo. Epigenetics 2013, 8, 54–65.
  39. Lv, X.; Liu, F.; Shang, Y.; Chen, S.Z. Honokiol exhibits enhanced antitumor effects with chloroquine by inducing cell death and inhibiting autophagy in human non-small cell lung cancer cells. Oncol. Rep. 2015, 34, 1289–1300.
  40. Pan, J.; Lee, Y.; Zhang, Q.; Xiong, D.; Wan, T.C.; Wang, Y.; You, M. Honokiol Decreases Lung Cancer Metastasis through Inhibition of the STAT3 Signaling Pathway. Cancer Prev. Res. 2017, 10, 133–141.
  41. Liou, S.-F.; Hua, K.-T.; Hsu, C.-Y.; Weng, M.-S. Honokiol from Magnolia spp. induces G1 arrest via disruption of EGFR stability through repressing HDAC6 deacetylated Hsp90 function in lung cancer cells. J. Funct. Foods 2015, 15, 84–96.
  42. Luo, L.-X.; Li, Y.; Liu, Z.-Q.; Fan, X.-X.; Duan, F.-G.; Li, R.-Z.; Yao, X.-J.; Leung, E.L.-H.; Liu, L. Honokiol Induces Apoptosis, G1 Arrest, and Autophagy in KRAS Mutant Lung Cancer Cells. Front. Pharmacol. 2017, 8, 199.
  43. Zhu, J.; Xu, S.; Gao, W.; Feng, J.; Zhao, G. Honokiol induces endoplasmic reticulum stress-mediated apoptosis in human lung cancer cells. Life Sci. 2019, 221, 204–211.
  44. Chae, J.I.; Jeon, Y.J.; Shim, J.H. Downregulation of Sp1 is involved in honokiol-induced cell cycle arrest and apoptosis in human malignant pleural mesothelioma cells. Oncol. Rep. 2013, 29, 2318–2324.
  45. Mannal, P.W.; Schneider, J.; Tangada, A.; McDonald, D.; McFadden, D.W. Honokiol produces anti-neoplastic effects on melanoma cells in vitro. J. Surg. Oncol. 2011, 104, 260–264.
  46. Chilampalli, C.; Guillermo, R.; Kaushik, R.S.; Young, A.; Chandrasekher, G.; Fahmy, H.; Dwivedi, C. Honokiol, a chemopreventive agent against skin cancer, induces cell cycle arrest and apoptosis in human epidermoid A431 cells. Exp. Biol. Med. 2011, 236, 1351–1359.
  47. Kaushik, G.; Ramalingam, S.; Subramaniam, D.; Rangarajan, P.; Protti, P.; Rammamoorthy, P.; Anant, S.; Mammen, J.M. Honokiol induces cytotoxic and cytostatic effects in malignant melanoma cancer cells. Am. J. Surg. 2012, 204, 868–873.
  48. Kaushik, G.; Kwatra, D.; Subramaniam, D.; Jensen, R.A.; Anant, S.; Mammen, J.M.V. Honokiol affects melanoma cell growth by targeting the AMP-activated protein kinase signaling pathway. Am. J. Surg. 2014, 208, 995–1002.
  49. Guillermo-Lagae, R.; Santha, S.; Thomas, M.; Zoelle, E.; Stevens, J.; Kaushik, R.S. Antineoplastic Effects of Honokiol on Melanoma. Biomed Res. Int. 2017, 2017, 5496398.
  50. Li, W.; Wang, Q.; Su, Q.; Ma, D.; An, C.; Ma, L.; Liang, H. Honokiol suppresses renal cancer cells’ metastasis via dual-blocking epithelial-mesenchymal transition and cancer stem cell properties through modulating miR-141/ZEB2 signaling. Mol. Cells 2014, 37, 383–388.
  51. Averett, C.; Bhardwaj, A.; Arora, S.; Srivastava, S.K.; Khan, M.A.; Ahmad, A.; Singh, S.; Carter, J.E.; Khushman, M.D.; Singh, A.P. Honokiol suppresses pancreatic tumor growth, metastasis and desmoplasia by interfering with tumor–stromal cross-talk. Carcinogenesis 2016, 37, 1052–1061.
  52. Arora, S.; Bhardwaj, A.; Srivastava, S.K.; Singh, S.; McClellan, S.; Wang, B.; Singh, A.P. Honokiol arrests cell cycle, induces apoptosis, and potentiates the cytotoxic effect of gemcitabine in human pancreatic cancer cells. PLoS ONE 2011, 6, e21573.
  53. Lu, C.H.; Chen, S.H.; Chang, Y.S.; Liu, Y.W.; Wu, J.Y.; Lim, Y.P.; Yu, H.I.; Lee, Y.R. Honokiol, a potential therapeutic agent, induces cell cycle arrest and program cell death in vitro and in vivo in human thyroid cancer cells. Pharmacol. Res. 2017, 115, 288–298.
  54. Wang, X.; Duan, X.; Yang, G.; Zhang, X.; Deng, L.; Zheng, H.; Deng, C.; Wen, J.; Wang, N.; Peng, C.; et al. Honokiol crosses BBB and BCSFB, and inhibits brain tumor growth in rat 9L intracerebral gliosarcoma model and human U251 xenograft glioma model. PLoS ONE 2011, 6, e18490.
  55. Jeong, J.J.; Lee, J.H.; Chang, K.C.; Kim, H.J. Honokiol exerts an anticancer effect in T98G human glioblastoma cells through the induction of apoptosis and the regulation of adhesion molecules. Int. J. Oncol. 2012, 41, 1358–1364.
  56. Fan, Y.; Xue, W.; Schachner, M.; Zhao, W. Honokiol Eliminates Glioma/Glioblastoma Stem Cell-Like Cells Via JAK-STAT3 Signaling and Inhibits Tumor Progression by Targeting Epidermal Growth Factor Receptor. Cancers 2018, 11, 22.
  57. Chang, K.H.; Yan, M.D.; Yao, C.J.; Lin, P.C.; Lai, G.M. Honokiol-induced apoptosis and autophagy in glioblastoma multiforme cells. Oncol. Lett. 2013, 6, 1435–1438.
  58. Joo, Y.N.; Eun, S.Y.; Park, S.W.; Lee, J.H.; Chang, K.C.; Kim, H.J. Honokiol inhibits U87MG human glioblastoma cell invasion through endothelial cells by regulating membrane permeability and the epithelial-mesenchymal transition. Int. J. Oncol. 2014, 44, 187–194.
  59. Lin, C.J.; Chen, T.L.; Tseng, Y.Y.; Wu, G.J.; Hsieh, M.H.; Lin, Y.W.; Chen, R.M. Honokiol induces autophagic cell death in malignant glioma through reactive oxygen species-mediated regulation of the p53/PI3K/Akt/mTOR signaling pathway. Toxicol. Appl. Pharmacol. 2016, 304, 59–69.
  60. Huang, K.; Chen, Y.; Zhang, R.; Wu, Y.; Ma, Y.; Fang, X.; Shen, S. Honokiol induces apoptosis and autophagy via the ROS/ERK1/2 signaling pathway in human osteosarcoma cells in vitro and in vivo. Cell Death Dis. 2018, 9, 157.
  61. Steinmann, P.; Walters, D.K.; Arlt, M.J.; Banke, I.J.; Ziegler, U.; Langsam, B.; Arbiser, J.; Muff, R.; Born, W.; Fuchs, B. Antimetastatic activity of honokiol in osteosarcoma. Cancer 2012, 118, 2117–2127.
  62. Yang, J.; Zou, Y.; Jiang, D. Honokiol suppresses proliferation and induces apoptosis via regulation of the miR21/PTEN/PI3K/AKT signaling pathway in human osteosarcoma cells. Int. J. Mol. Med. 2018, 41, 1845–1854.
  63. Kim, D.W.; Ko, S.M.; Jeon, Y.J.; Noh, Y.W.; Choi, N.J.; Cho, S.D.; Moon, H.S.; Cho, Y.S.; Shin, J.C.; Park, S.M.; et al. Anti-proliferative effect of honokiol in oral squamous cancer through the regulation of specificity protein 1. Int. J. Oncol. 2013, 43, 1103–1110.
  64. Xu, Q.; Tong, F.; He, C.; Song, P.; Xu, Q.; Chen, Z. The inhibition effect of Honokiol in liver cancer. Int. J. Clin. Exp. Med. 2018, 11, 10673–10678.
  65. Han, L.L.; Xie, L.P.; Li, L.H.; Zhang, X.W.; Zhang, R.Q.; Wang, H.Z. Reactive oxygen species production and Bax/Bcl-2 regulation in honokiol-induced apoptosis in human hepatocellular carcinoma SMMC-7721 cells. Environ. Toxicol. Pharmacol. 2009, 28, 97–103.
  66. Luo, H.; Zhong, Q.; Chen, L.J.; Qi, X.R.; Fu, A.F.; Yang, H.S.; Yang, F.; Lin, H.G.; Wei, Y.Q.; Zhao, X. Liposomal honokiol, a promising agent for treatment of cisplatin-resistant human ovarian cancer. J. Cancer Res. Clin. Oncol. 2008, 134, 937–945.
  67. Li, Z.; Liu, Y.; Zhao, X.; Pan, X.; Yin, R.; Huang, C.; Chen, L.; Wei, Y. Honokiol, a natural therapeutic candidate, induces apoptosis and inhibits angiogenesis of ovarian tumor cells. Eur. J. Obstet. Gynecol. Reprod. Biol. 2008, 140, 95–102.
  68. Hahm, E.R.; Singh, S.V. Honokiol causes G0-G1 phase cell cycle arrest in human prostate cancer cells in association with suppression of retinoblastoma protein level/phosphorylation and inhibition of E2F1 transcriptional activity. Mol. Cancer Ther. 2007, 6, 2686–2695.
  69. Shigemura, K.; Arbiser, J.L.; Sun, S.Y.; Zayzafoon, M.; Johnstone, P.A.; Fujisawa, M.; Gotoh, A.; Weksler, B.; Zhau, H.E.; Chung, L.W. Honokiol, a natural plant product, inhibits the bone metastatic growth of human prostate cancer cells. Cancer 2007, 109, 1279–1289.
  70. Hahm, E.R.; Sakao, K.; Singh, S.V. Honokiol activates reactive oxygen species-mediated cytoprotective autophagy in human prostate cancer cells. Prostate 2014, 74, 1209–1221.
  71. Leeman-Neill, R.J.; Cai, Q.; Joyce, S.C.; Thomas, S.M.; Bhola, N.E.; Neill, D.B.; Arbiser, J.L.; Grandis, J.R. Honokiol inhibits epidermal growth factor receptor signaling and enhances the antitumor effects of epidermal growth factor receptor inhibitors. Clin. Cancer Res. 2010, 16, 2571–2579.
  72. Lin, J.W.; Chen, J.T.; Hong, C.Y.; Lin, Y.L.; Wang, K.T.; Yao, C.J.; Lai, G.M.; Chen, R.M. Honokiol traverses the blood-brain barrier and induces apoptosis of neuroblastoma cells via an intrinsic bax-mitochondrion-cytochrome c-caspase protease pathway. Neuro-Oncology 2012, 14, 302–314.
  73. Yeh, P.S.; Wang, W.; Chang, Y.A.; Lin, C.J.; Wang, J.J.; Chen, R.M. Honokiol induces autophagy of neuroblastoma cells through activating the PI3K/Akt/mTOR and endoplasmic reticular stress/ERK1/2 signaling pathways and suppressing cell migration. Cancer Lett. 2016, 370, 66–77.
  74. Zhang, Q.; Zhao, W.; Ye, C.; Zhuang, J.; Chang, C.; Li, Y.; Huang, X.; Shen, L.; Li, Y.; Cui, Y.; et al. Honokiol inhibits bladder tumor growth by suppressing EZH2/miR-143 axis. Oncotarget 2015, 6, 37335–37348.
  75. Bao, L.; Jaramillo, M.C.; Zhang, Z.; Zheng, Y.; Yao, M.; Zhang, D.D.; Yi, X. Induction of autophagy contributes to cisplatin resistance in human ovarian cancer cells. Mol. Med. Rep. 2015, 11, 91–98.
  76. Zhang, Q.; Cheng, J.; Xin, Q. Effects of tetracycline on developmental toxicity and molecular responses in zebrafish (Danio rerio) embryos. Ecotoxicology 2015, 24, 707–719.
  77. Hill, D.; Chen, L.; Snaar-Jagalska, E.; Chaudhry, B. Embryonic zebrafish xenograft assay of human cancer metastasis. F1000Research 2018, 7, 1682.
  78. Li, L.; Han, W.; Gu, Y.; Qiu, S.; Lu, Q.; Jin, J.; Luo, J.; Hu, X. Honokiol Induces a Necrotic Cell Death through the Mitochondrial Permeability Transition Pore. Cancer Res. 2007, 67, 4894.
  79. Hahm, E.R.; Arlotti, J.A.; Marynowski, S.W.; Singh, S.V. Honokiol, a constituent of oriental medicinal herb magnolia officinalis, inhibits growth of PC-3 xenografts in vivo in association with apoptosis induction. Clin. Cancer Res. 2008, 14, 1248–1257.
  80. Sheu, M.L.; Liu, S.H.; Lan, K.H. Honokiol induces calpain-mediated glucose-regulated protein-94 cleavage and apoptosis in human gastric cancer cells and reduces tumor growth. PLoS ONE 2007, 2, e1096.
  81. Liu, S.H.; Wang, K.B.; Lan, K.H.; Lee, W.J.; Pan, H.C.; Wu, S.M.; Peng, Y.C.; Chen, Y.C.; Shen, C.C.; Cheng, H.C.; et al. Calpain/SHP-1 Interaction by Honokiol Dampening Peritoneal Dissemination of Gastric Cancer in nu/nu Mice. PLoS ONE 2012, 7, e43711.
  82. Liu, Y.; Chen, L.; He, X.; Fan, L.; Yang, G.; Chen, X.; Lin, X.; Du, L.; Li, Z.; Ye, H.; et al. Enhancement of therapeutic effectiveness by combining liposomal honokiol with cisplatin in ovarian carcinoma. Int. J. Gynecol. Cancer 2008, 18, 652–659.
  83. Goldar, S.; Khaniani, M.S.; Derakhshan, S.M.; Baradaran, B. Molecular mechanisms of apoptosis and roles in cancer development and treatment. Asian Pac. J. Cancer Prev. 2015, 16, 2129–2144.
  84. Hassan, M.; Watari, H.; AbuAlmaaty, A.; Ohba, Y.; Sakuragi, N. Apoptosis and molecular targeting therapy in cancer. Biomed Res. Int. 2014, 2014, 150845.
  85. Wong, R.S.Y. Apoptosis in cancer: From pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011, 30, 87.
  86. Gerl, R.; Vaux, D.L. Apoptosis in the development and treatment of cancer. Carcinogenesis 2005, 26, 263–270.
  87. Letai, A. Apoptosis and Cancer. Annu. Rev. Cancer Biol. 2017, 1, 275–294.
  88. Jeong, Y.-H.; Hur, J.H.; Jeon, E.-J.; Park, S.-J.; Hwang, T.J.; Lee, S.A.; Lee, W.K.; Sung, J.M. Honokiol Improves Liver Steatosis in Ovariectomized Mice. Molecules 2018, 23, 194.
  89. Huang, J.-S.; Yao, C.-J.; Chuang, S.-E.; Yeh, C.-T.; Lee, L.-M.; Chen, R.-M.; Chao, W.-J.; Whang-Peng, J.; Lai, G.-M. Honokiol inhibits sphere formation and xenograft growth of oral cancer side population cells accompanied with JAK/STAT signaling pathway suppression and apoptosis induction. BMC Cancer 2016, 16, 245.
  90. Prasad, R.; Katiyar, S.K. Honokiol, an Active Compound of Magnolia Plant, Inhibits Growth, and Progression of Cancers of Different Organs. Adv. Exp. Med. Biol. 2016, 928, 245–265.
  91. Banik, K.; Ranaware, A.M.; Deshpande, V.; Nalawade, S.P.; Padmavathi, G.; Bordoloi, D.; Sailo, B.L.; Shanmugam, M.K.; Fan, L.; Arfuso, F.; et al. Honokiol for cancer therapeutics: A traditional medicine that can modulate multiple oncogenic targets. Pharmacol. Res. 2019, 144, 192–209.
  92. Wang, X.; Beitler, J.J.; Huang, W.; Chen, G.; Qian, G.; Magliocca, K.; Patel, M.R.; Chen, A.Y.; Zhang, J.; Nannapaneni, S.; et al. Honokiol Radiosensitizes Squamous Cell Carcinoma of the Head and Neck by Downregulation of Survivin. Clin. Cancer Res. 2018, 24, 858–869.
  93. Garcia, A.; Zheng, Y.; Zhao, C.; Toschi, A.; Fan, J.; Shraibman, N.; Brown, H.A.; Bar-Sagi, D.; Foster, D.A.; Arbiser, J.L. Honokiol Suppresses Survival Signals Mediated by Ras-Dependent Phospholipase D Activity in Human Cancer Cells. Clin. Cancer Res. 2008, 14, 4267.
  94. Fried, L.E.; Arbiser, J.L. Honokiol, a multifunctional antiangiogenic and antitumor agent. Antioxid. Redox Signal. 2009, 11, 1139–1148.
  95. Li, H.Y.; Ye, H.G.; Chen, C.Q.; Yin, L.H.; Wu, J.B.; He, L.C.; Gao, S.M. Honokiol induces cell cycle arrest and apoptosis via inhibiting class I histone deacetylases in acute myeloid leukemia. J. Cell. Biochem. 2015, 116, 287–298.
  96. Deng, J.; Qian, Y.; Geng, L.; Chen, J.; Wang, X.; Xie, H.; Yan, S.; Jiang, G.; Zhou, L.; Zheng, S. Involvement of p38 mitogen-activated protein kinase pathway in honokiol-induced apoptosis in a human hepatoma cell line (hepG2). Liver Int. 2008, 28, 1458–1464.
  97. Hasegawa, S.; Yonezawa, T.; Ahn, J.Y.; Cha, B.Y.; Teruya, T.; Takami, M.; Yagasaki, K.; Nagai, K.; Woo, J.T. Honokiol inhibits osteoclast differentiation and function in vitro. Biol. Pharm. Bull. 2010, 33, 487–492.
  98. Tse, A.K.; Wan, C.K.; Shen, X.L.; Yang, M.; Fong, W.F. Honokiol inhibits TNF-alpha-stimulated NF-kappaB activation and NF-kappaB-regulated gene expression through suppression of IKK activation. Biochem. Pharmacol. 2005, 70, 1443–1457.
  99. Li, J.; Shao, X.; Wu, L.; Feng, T.; Jin, C.; Fang, M.; Wu, N.; Yao, H. Honokiol: An effective inhibitor of tumor necrosis factor-alpha-induced up-regulation of inflammatory cytokine and chemokine production in human synovial fibroblasts. Acta Biochim. Biophys. Sin. 2011, 43, 380–386.
  100. Xu, H.L.; Tang, W.; Du, G.H.; Kokudo, N. Targeting apoptosis pathways in cancer with magnolol and honokiol, bioactive constituents of the bark of Magnolia officinalis. Drug Discov. Ther. 2011, 5, 202–210.
  101. Raja, S.M.; Chen, S.; Yue, P.; Acker, T.M.; Lefkove, B.; Arbiser, J.L.; Khuri, F.R.; Sun, S.Y. The natural product honokiol preferentially inhibits cellular FLICE-inhibitory protein and augments death receptor-induced apoptosis. Mol. Cancer Ther. 2008, 7, 2212–2223.
  102. Rauf, A.; Patel, S.; Imran, M.; Maalik, A.; Arshad, M.U.; Saeed, F.; Mabkhot, Y.N.; Al-Showiman, S.S.; Ahmad, N.; Elsharkawy, E. Honokiol: An anticancer lignan. Biomed. Pharmacother. 2018, 107, 555–562.
  103. Schroder, M.; Kaufman, R.J. ER stress and the unfolded protein response. Mutat. Res. 2005, 569, 29–63.
  104. Cao, S.S.; Kaufman, R.J. Endoplasmic reticulum stress and oxidative stress in cell fate decision and human disease. Antioxid. Redox Signal. 2014, 21, 396–413.
  105. Ferri, K.F.; Kroemer, G. Organelle-specific initiation of cell death pathways. Nat. Cell Biol. 2001, 3, E255–E263.
  106. Chiu, C.-S.; Tsai, C.-H.; Hsieh, M.-S.; Tsai, S.-C.; Jan, Y.-J.; Lin, W.-Y.; Lai, D.-W.; Wu, S.-M.; Hsing, H.-Y.; Arbiser, J.L.; et al. Exploiting Honokiol-induced ER stress CHOP activation inhibits the growth and metastasis of melanoma by suppressing the MITF and β-catenin pathways. Cancer Lett. 2019, 442, 113–125.
  107. Liu, S.H.; Shen, C.C.; Yi, Y.C.; Tsai, J.J.; Wang, C.C.; Chueh, J.T.; Lin, K.L.; Lee, T.C.; Pan, H.C.; Sheu, M.L. Honokiol inhibits gastric tumourigenesis by activation of 15-lipoxygenase-1 and consequent inhibition of peroxisome proliferator-activated receptor-γ and COX-2-dependent signals. Br. J. Pharmacol. 2010, 160, 1963–1972.
  108. Liu, S.H.; Lee, W.J.; Lai, D.W.; Wu, S.M.; Liu, C.Y.; Tien, H.R.; Chiu, C.S.; Peng, Y.C.; Jan, Y.J.; Chao, T.H.; et al. Honokiol confers immunogenicity by dictating calreticulin exposure, activating ER stress and inhibiting epithelial-to-mesenchymal transition. Mol. Oncol. 2015, 9, 834–849.
  109. Martin, S.; Lamb, H.K.; Brady, C.; Lefkove, B.; Bonner, M.Y.; Thompson, P.; Lovat, P.E.; Arbiser, J.L.; Hawkins, A.R.; Redfern, C.P. Inducing apoptosis of cancer cells using small-molecule plant compounds that bind to GRP78. Br. J. Cancer 2013, 109, 433–443.
  110. Lee, S.Y.; Ju, M.K.; Jeon, H.M.; Jeong, E.K.; Lee, Y.J.; Kim, C.H.; Park, H.G.; Han, S.I.; Kang, H.S. Regulation of Tumor Progression by Programmed Necrosis. Oxidative Med. Cell. Longev. 2018, 2018, 3537471.
  111. Tian, W.; Xu, D.; Deng, Y.-C. Honokiol, a multifunctional tumor cell death inducer. Die Pharm. Int. J. Pharm. Sci. 2012, 67, 811–816.
  112. Chen, G.; Izzo, J.; Demizu, Y.; Wang, F.; Guha, S.; Wu, X.; Hung, M.-C.; Ajani, J.A.; Huang, P. Different redox states in malignant and nonmalignant esophageal epithelial cells and differential cytotoxic responses to bile acid and honokiol. Antioxid. Redox Signal. 2009, 11, 1083–1095.
  113. Yu, C.; Zhang, Q.; Zhang, H.Y.; Zhang, X.; Huo, X.; Cheng, E.; Wang, D.H.; Arbiser, J.L.; Spechler, S.J.; Souza, R.F. Targeting the intrinsic inflammatory pathway: Honokiol exerts proapoptotic effects through STAT3 inhibition in transformed Barrett’s cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G561–G569.
  114. Meier, J.A.; Hyun, M. Stress-induced dynamic regulation of mitochondrial STAT3 and its association with cyclophilin D reduce mitochondrial ROS production. Sci. Signal. 2017, 10.
  115. Cen, M.; Yao, Y.; Cui, L.; Yang, G.; Lu, G.; Fang, L.; Bao, Z.; Zhou, J. Honokiol induces apoptosis of lung squamous cell carcinoma by targeting FGF2-FGFR1 autocrine loop. Cancer Med. 2018, 7, 6205–6218.
  116. Hahm, E.R.; Singh, K.B.; Singh, S.V. c-Myc is a novel target of cell cycle arrest by honokiol in prostate cancer cells. Cell Cycle 2016, 15, 2309–2320.
  117. Vaid, M.; Sharma, S.D.; Katiyar, S.K. Honokiol, a phytochemical from the Magnolia plant, inhibits photocarcinogenesis by targeting UVB-induced inflammatory mediators and cell cycle regulators: Development of topical formulation. Carcinogenesis 2010, 31, 2004–2011.
  118. Guo, C.; Ma, L.; Zhao, Y.; Peng, A.; Cheng, B.; Zhou, Q.; Zheng, L.; Huang, K. Inhibitory effects of magnolol and honokiol on human calcitonin aggregation. Sci. Rep. 2015, 5, 13556.
  119. Yan, B.; Peng, Z.Y. Honokiol induces cell cycle arrest and apoptosis in human gastric carcinoma MGC-803 cell line. Int. J. Clin. Exp. Med. 2015, 8, 5454–5461.
  120. Grimmel, M.; Backhaus, C.; Proikas-Cezanne, T. WIPI-Mediated Autophagy and Longevity. Cells 2015, 4, 202–217.
  121. Yun, C.W.; Lee, S.H. The Roles of Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 3466.
  122. Galluzzi, L.; Pietrocola, F.; Bravo-San Pedro, J.M.; Amaravadi, R.K.; Baehrecke, E.H.; Cecconi, F.; Codogno, P.; Debnath, J.; Gewirtz, D.A.; Karantza, V.; et al. Autophagy in malignant transformation and cancer progression. EMBO J. 2015, 34, 856–880.
  123. Lin, L.; Baehrecke, E.H. Autophagy, cell death, and cancer. Mol. Cell Oncol. 2015, 2, e985913.
  124. Paquette, M.; El-Houjeiri, L.; Pause, A. mTOR Pathways in Cancer and Autophagy. Cancers 2018, 10, 18.
  125. Murray, J.T.; Tee, A.R. Mechanistic Target of Rapamycin (mTOR) in the Cancer Setting. Cancers 2018, 10, 168.
  126. Itakura, E.; Mizushima, N. Atg14 and UVRAG: Mutually exclusive subunits of mammalian Beclin 1-PI3K complexes. Autophagy 2009, 5, 534–536.
  127. Torii, S.; Yoshida, T.; Arakawa, S.; Honda, S.; Nakanishi, A.; Shimizu, S. Identification of PPM1D as an essential Ulk1 phosphatase for genotoxic stress-induced autophagy. EMBO Rep. 2016, 17, 1552–1564.
  128. Maiuri, M.C.; Criollo, A.; Kroemer, G. Crosstalk between apoptosis and autophagy within the Beclin 1 interactome. EMBO J. 2010, 29, 515–516.
  129. Carlsson, S.R.; Simonsen, A. Membrane dynamics in autophagosome biogenesis. J. Cell Sci. 2015, 128, 193.
  130. Mochida, K.; Oikawa, Y.; Kimura, Y.; Kirisako, H.; Hirano, H.; Ohsumi, Y.; Nakatogawa, H. Receptor-mediated selective autophagy degrades the endoplasmic reticulum and the nucleus. Nature 2015, 522, 359–362.
  131. Wrighton, K.H. Selecting ER for eating. Nat. Rev. Mol. Cell Biol. 2015, 16, 389.
  132. Chio, C.C.; Chen, K.Y.; Chang, C.K.; Chuang, J.Y.; Liu, C.C.; Liu, S.H.; Chen, R.M. Improved effects of honokiol on temozolomide-induced autophagy and apoptosis of drug-sensitive and -tolerant glioma cells. BMC Cancer 2018, 18, 379.
  133. Nieto, M.A.; Huang, R.Y.; Jackson, R.A.; Thiery, J.P. EMT: 2016. Cell 2016, 166, 21–45.
  134. Brabletz, T.; Kalluri, R.; Nieto, M.A.; Weinberg, R.A. EMT in cancer. Nat. Rev. Cancer 2018, 18, 128–134.
  135. Roche, J. The Epithelial-to-Mesenchymal Transition in Cancer. Cancers 2018, 10, 52.
  136. Røsland, G.V.; Dyrstad, S.E.; Tusubira, D.; Helwa, R.; Tan, T.Z.; Lotsberg, M.L.; Pettersen, I.K.N.; Berg, A.; Kindt, C.; Hoel, F.; et al. Epithelial to mesenchymal transition (EMT) is associated with attenuation of succinate dehydrogenase (SDH) in breast cancer through reduced expression of SDHC. Cancer Metab. 2019, 7, 6.
  137. Shen, L.; Zhang, F.; Huang, R.; Yan, J.; Shen, B. Honokiol inhibits bladder cancer cell invasion through repressing SRC-3 expression and epithelial-mesenchymal transition. Oncol. Lett. 2017, 14, 4294–4300.
  138. Avtanski, D.B.; Nagalingam, A.; Bonner, M.Y.; Arbiser, J.L.; Saxena, N.K.; Sharma, D. Honokiol inhibits epithelial-mesenchymal transition in breast cancer cells by targeting signal transducer and activator of transcription 3/Zeb1/E-cadherin axis. Mol. Oncol. 2014, 8, 565–580.
  139. Lv, X.-Q.; Qiao, X.-R.; Su, L.; Chen, S.-Z. Honokiol inhibits EMT-mediated motility and migration of human non-small cell lung cancer cells in vitro by targeting c-FLIP. Acta Pharmacol. Sin. 2016, 37, 1574–1586.
  140. Qin, L.; Liu, Z.; Chen, H.; Xu, J. The steroid receptor coactivator-1 regulates twist expression and promotes breast cancer metastasis. Cancer Res. 2009, 69, 3819–3827.
  141. Yao, C.-J.; Lai, G.-M.; Yeh, C.-T.; Lai, M.-T.; Shih, P.-H.; Chao, W.-J.; Whang-Peng, J.; Chuang, S.-E.; Lai, T.-Y. Honokiol Eliminates Human Oral Cancer Stem-Like Cells Accompanied with Suppression of Wnt/β-Catenin Signaling and Apoptosis Induction. Evid.-Based Complement. Altern. Med. 2013, 2013, 146136.
  142. Wang, W.D.; Shang, Y.; Li, Y.; Chen, S.Z. Honokiol inhibits breast cancer cell metastasis by blocking EMT through modulation of Snail/Slug protein translation. Acta Pharm. Sin. 2019, 40, 1219–1227.
  143. Galichon, P.; Hertig, A. Epithelial to mesenchymal transition as a biomarker in renal fibrosis: Are we ready for the bedside? Fibrogenesis Tissue Repair 2011, 4, 11.
  144. Conacci-Sorrell, M.; Ngouenet, C.; Anderson, S.; Brabletz, T.; Eisenman, R.N. Stress-induced cleavage of Myc promotes cancer cell survival. Genes Dev. 2014, 28, 689–707.
  145. Yamaguchi, H.; Wyckoff, J.; Condeelis, J. Cell migration in tumors. Curr. Opin. Cell Biol. 2005, 17, 559–564.
  146. Seyfried, T.N.; Huysentruyt, L.C. On the origin of cancer metastasis. Crit. Rev. Oncog. 2013, 18, 43–73.
  147. Tay, R.Y.; Fernández-Gutiérrez, F.; Foy, V.; Burns, K.; Pierce, J.; Morris, K.; Priest, L.; Tugwood, J.; Ashcroft, L.; Lindsay, C.R.; et al. Prognostic value of circulating tumour cells in limited-stage small-cell lung cancer: Analysis of the concurrent once-daily versus twice-daily radiotherapy (CONVERT) randomised controlled trial. Ann. Oncol. 2019, 30, 1114–1120.
  148. Singh, T.; Katiyar, S.K. Honokiol, a phytochemical from Magnolia spp., inhibits breast cancer cell migration by targeting nitric oxide and cyclooxygenase-2. Int. J. Oncol. 2011, 38, 769–776.
  149. Zhang, J.; Zhang, Y.; Shen, W.; Fu, R.; Ding, Z.; Zhen, Y.; Wan, Y. Cytological effects of honokiol treatment and its potential mechanism of action in non-small cell lung cancer. Biomed. Pharmacother. 2019, 117, 109058.
  150. Cheng, S.; Castillo, V.; Welty, M.; Eliaz, I.; Sliva, D. Honokiol inhibits migration of renal cell carcinoma through activation of RhoA/ROCK/MLC signaling pathway. Int. J. Oncol. 2016, 49, 1525–1530.
  151. Balan, M.; Chakraborty, S.; Flynn, E.; Zurakowski, D.; Pal, S. Honokiol inhibits c-Met-HO-1 tumor-promoting pathway and its cross-talk with calcineurin inhibitor-mediated renal cancer growth. Sci. Rep. 2017, 7, 5900.
  152. Alizadeh, A.M.; Shiri, S.; Farsinejad, S. Metastasis review: From bench to bedside. Tumor Biol. 2014, 35, 8483–8523.
  153. Klein, C.A. Cancer. The metastasis cascade. Science 2008, 321, 1785–1787.
  154. Bai, X.; Cerimele, F.; Ushio-Fukai, M.; Waqas, M.; Campbell, P.M.; Govindarajan, B.; Der, C.J.; Battle, T.; Frank, D.A.; Ye, K.; et al. Honokiol, a small molecular weight natural product, inhibits angiogenesis in vitro and tumor growth in vivo. J. Biol. Chem. 2003, 278, 35501–35507.
  155. Kolligs, F.T.; Bommer, G.; Goke, B. Wnt/beta-catenin/tcf signaling: A critical pathway in gastrointestinal tumorigenesis. Digestion 2002, 66, 131–144.
  156. Hlubek, F.; Spaderna, S.; Jung, A.; Kirchner, T.; Brabletz, T. Beta-catenin activates a coordinated expression of the proinvasive factors laminin-5 gamma2 chain and MT1-MMP in colorectal carcinomas. Int. J. Cancer 2004, 108, 321–326.
  157. Cheng, S.; Castillo, V.; Eliaz, I.; Sliva, D. Honokiol suppresses metastasis of renal cell carcinoma by targeting KISS1/KISS1R signaling. Int. J. Oncol. 2015, 46, 2293–2298.
  158. Nishida, N.; Yano, H.; Nishida, T.; Kamura, T.; Kojiro, M. Angiogenesis in cancer. Vasc. Health Risk Manag. 2006, 2, 213–219.
  159. Rajabi, M.; Mousa, S.A. The Role of Angiogenesis in Cancer Treatment. Biomedicines 2017, 5, 34.
  160. Tonini, T.; Rossi, F.; Claudio, P.P. Molecular basis of angiogenesis and cancer. Oncogene 2003, 22, 6549–6556.
  161. Banerjee, P.; Basu, A.; Arbiser, J.L.; Pal, S. The natural product honokiol inhibits calcineurin inhibitor-induced and Ras-mediated tumor promoting pathways. Cancer Lett. 2013, 338, 292–299.
  162. Vavilala, D.T.; Ponnaluri, V.K.C.; Kanjilal, D.; Mukherji, M. Evaluation of anti-HIF and anti-angiogenic properties of honokiol for the treatment of ocular neovascular diseases. PLoS ONE 2014, 9, e113717.
  163. Vavilala, D.T.; O’Bryhim, B.E.; Ponnaluri, V.K.; White, R.S.; Radel, J.; Symons, R.C.; Mukherji, M. Honokiol inhibits pathological retinal neovascularization in oxygen-induced retinopathy mouse model. Biochem. Biophys. Res. Commun. 2013, 438, 697–702.
  164. Wen, J.; Wang, X.; Pei, H.; Xie, C.; Qiu, N.; Li, S.; Wang, W.; Cheng, X.; Chen, L. Anti-psoriatic effects of Honokiol through the inhibition of NF-kappaB and VEGFR-2 in animal model of K14-VEGF transgenic mouse. J. Pharmacol. Sci. 2015, 128, 116–124.
  165. Hu, J.; Chen, L.J.; Liu, L.; Chen, X.; Chen, P.L.; Yang, G.; Hou, W.L.; Tang, M.H.; Zhang, F.; Wang, X.H.; et al. Liposomal honokiol, a potent anti-angiogenesis agent, in combination with radiotherapy produces a synergistic antitumor efficacy without increasing toxicity. Exp. Mol. Med. 2008, 40, 617–628.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 634
Revisions: 2 times (View History)
Update Date: 18 Aug 2021
1000/1000