Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2020

Open Access 01.12.2020 | Review

Emerging agents that target signaling pathways in cancer stem cells

verfasst von: Yue Yang, Xiaoman Li, Ting Wang, Qianqian Guo, Tao Xi, Lufeng Zheng

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2020

Abstract

Cancer stem cells (CSCs) contribute to the initiation, recurrence, and metastasis of cancer; however, there are still no drugs targeting CSCs in clinical application. There are several signaling pathways playing critical roles in CSC progression, such as the Wnt, Hedgehog, Notch, Hippo, and autophagy signaling pathways. Additionally, targeting the ferroptosis signaling pathway was recently shown to specifically kill CSCs. Therefore, targeting these pathways may suppress CSC progression. The structure of small-molecule drugs shows a good spatial dispersion, and its chemical properties determine its good druggability and pharmacokinetic properties. These characteristics make small-molecule drugs show a great advantage in drug development, which is increasingly popular in the market. Thus, in this review, we will summarize the current researches on the small-molecule compounds suppressing CSC progression, including inhibitors of Wnt, Notch, Hedgehog, and autophagy pathways, and activators of Hippo and ferroptosis pathways. These small-molecule compounds emphasize CSC importance in tumor progression and propose a new strategy to treat cancer in clinic via targeting CSCs.
Hinweise
Yue Yang and Xiaoman Li contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CSCs
Cancer stem cells
Hh
Hedgehog
LRP
Lipoprotein receptor-related protein
TNBC
Triple negative breast cancer
TBD
Terminal anchor polymerase binding domain
HNSCC
Neck squamous cell carcinoma
CRC
Colorectal cancer
TFP
Trifluoperazine
NSCLC
Non-small cell lung carcinoma
NPC
Nasopharyngeal carcinoma
HCC
Hepatocellular carcinoma
DTX
Docetaxel
SFN
Sulforaphane
PP
Pyrvinium pamoate
EMT
Epithelial-mesenchymal transition
AD
Actinomycin D
TS
Telmisartan
DLL
Delta-like ligand
NICD
Notch intracellular domain
DLT
Dose-limiting toxicity
INS
Insulinomas
LSCs
Leukemia stem cells
SMO
Smoothened
ER
Estrogen receptor
BCSC
Breast cancer stem cell
TEAD
TEA domain family members
Evo
Evodiamine
CPZ
Chlorpromazine
CQ
Chloroquine
HCQ
Hydroxychloroquine
IM
Imatinib
mCRPC
Metastatic castration-resistant prostate cancer
PPI
Proton pump inhibitor
3-MA
3-Methyladenine
Rott
Rottlerin
ROS
Reactive oxygen species
TfR1
Transferrin receptor 1
DMT1
Divalent metal transporter 1
LIP
Labile iron pool
PCBP1/2
Poly (rC)-binding protein 1/2
IRP2
Iron regulatory proteins 2
IREs
Iron-responsive elements
UTRs
Untranslated regions
Gpx4
Glutathione peroxidase 4
PLs
Phospholipids
PUFAs
Polyunsaturated fatty acids
LOXs
Lipoxygenases
AA
Arachidonic acid
ACSL4
Acyl-CoA synthetase long-chain family member
LPCAT3
Lysophosphatidylcholine acyltransferase 3
MDR
Multi-drug resistance
TMZ
Temozolomide
GSCs
Glioblastoma stem cells
DHA
Dihydroartemisinin
iCSCL
Induced cancer stem-like cells
GBM
Glioblastoma multiforme
HER
Pan-human epidermal growth factor receptor
EOC
Epithelial ovarian cancer

Background

Although traditional therapeutic methods can reduce tumor volume extensively, cancer recurrence and metastasis always occur [1]. CSCs constitute a small portion of cancer cells, but they show resistance to chemotherapy and radiotherapy. Most conventional treatment methods kill cells with a proliferative potential to shrink the tumor but have no effect on stationary CSCs. This action explains why the tumor volume is reduced but the patient survival rate is not improved [2]. Due to their self-renewal ability and therapeutic resistance, CSCs are considered to be the root of tumor growth, recurrence, metastasis, and drug resistance [3]. Notably, because of their plasticity, stationary CSCs may produce cycling CSCs, which contributes to the relapse of cancer [4]. Therefore, more specific therapies targeting CSCs may lead to better results, and combining them with the traditional therapeutic methods may even achieve healing [5]. Cancer is a disease caused by disordered cell growth due to genetic mutations. CSCs have the same genetic driver mutations as most cancer cells, but CSCs have developmental characteristics that differ from those of non-stem cells, including differences in epigenetic modifications and gene expression profiles [6]. Many changes in the signaling pathways in CSCs provide a preliminary basis for developing compounds targeting CSCs. Signaling pathways that regulate the self-renewal and differentiation of CSCs include Wnt, Hedgehog (Hh), Notch, and Hippo, and these signaling pathways have been extensively studied [7, 8]. In addition, the signaling pathways inhibiting the proliferation of CSCs, in newly discovered mechanisms, have also gradually gained attention, and many promising results have been obtained (Fig. 1) [9]. Targeting CSCs with traditional therapy (chemotherapy and radiotherapy) may yield better results in controlling tumor growth, preventing cancer recurrence and metastasis, and decreasing drug resistance. Small-molecule compounds targeting CSCs play important role in this attenuation process. This review summarizes the research status of small-molecule compounds that suppress CSC progression, including inhibitors of the Wnt, Notch, Hh, and autophagy signaling pathways and activators of Hippo and ferroptosis signaling pathways, which may provide a basis for a current treatment for cancer.

Signaling pathway regulators

CSCs display many characteristics of embryonic or tissue stem cells and often show continuous activation of one or more highly conserved signaling pathways related to development and tissue homeostasis. In these signaling pathways, the Wnt, Hh, Notch, and Hippo signaling pathways are associated with CSC self-renewal [10] and have been used to explore new drugs targeting CSCs.

Wnt signaling pathway inhibitors

The Wnt signaling pathway is highly conserved among species and is divided into the β-catenin-dependent pathway and noncanonical Wnt pathway. When Wnt ligands bind to the Frizzled protein and low-density lipoprotein receptor-related protein (LRP) coreceptors, they initiate the transduction of Wnt/β-catenin signaling, eventually leading to β-catenin stabilization, nuclear translocation, and activation of target genes [31]. Since the β-catenin-dependent pathway has been extensively characterized in mammals, regulates the pluripotency of stem cells, and plays a critical role in self-renewal and differentiation ability, it is thought that abnormal activation of the Wnt pathway promotes CSC progression and thus leads to the deterioration and metastasis of cancer. Thus, the inhibition of CSCs can be mediated through this pathway [32, 33] (Table 1).
Table 1
Small-molecule compounds inhibiting CSC progression through suppressing Wnt signaling pathway
Name
Target
Mechanism
Type of cancer
Phase
NCT number (starting time)/publication date
Assessment
Wnt974
Wnt
Inhibits the proliferation of breast CSCs
Breast cancer
Phase I
NCT01351103(May 10, 2011)
Dysgeusia [11]
Niclosamide
Wnt/β-catenin
Selectively targets ovarian CSCs
Ovarian cancer
Preclinical
July, 2014
Without significant toxicity [1214]
LRP6, β-catenin
Decreases ALDH+ population cells
Basal-like breast cancer
Preclinical
April, 2014
Wnt/β-catenin
Suppresses CSC populations and self-renewal ability
Colorectal cancer
Phase II
NCT02519582 (August 11, 2015)
ONC201
Wnt/β-catenin
Inhibits CSC self-renewal and deregulates CSC markers and CSC-related gene expression
Glioblastoma cancer
Phase I/II
NCT02038699 (January 16, 2014)
Well tolerated, Grade III neutropenia, Grade II allergic [15, 16]
Prostate cancer
Preclinical
August 2, 2017
XAV939
β-catenin
Attenuates CSC-mediated chemoresistance
Colon cancer
Preclinical
April, 2016
Induces cardiotoxicity and limited therapeutic window [17]
HNSCC
Preclinical
October, 2019
TFP
Wnt/β-catenin
Inhibits lung CSC spheroid formation and suppresses lung CSC marker expression (such as CD44/CD133)
Lung cancer
Preclinical
December 1, 2012
Induces little systemic toxicity, but grade 0–2 neurologic toxicity [18, 19]
Chelerythrine
β-catenin
Inhibits CSCs invasion, spheroid-forming ability, and the stem marker such as SOX2
NSCLC
Preclinical
January 6, 2020
Without systemic toxicity [20, 21]
FH535
Wnt/β-catenin
Deregulates pancreatic CSC marker CD24 and CD44 expression
Pancreatic cancer
Preclinical
July 26, 2016
Without side effects according to the current studies, still need experiments to prove [22]
Wnt-C59
Wnt
Decreases sphere formation of CSCs
NPC
Preclinical
June 10, 2015
Exhibits no apparent toxicity in mice; needs experiments to prove [23]
IWR-1
β-catenin
Impairs CSCs self-renewal and hampers the expression of key stem markers, and increases doxorubicin sensitivity
Osteosarcoma
Preclinical
February 1, 2018
Well tolerated in mice, but still needs to be thoroughly studied [24]
IC-2
Wnt
Reduces the population of CD44+ (liver CSCs) and the ability of sphere-forming ability
HCC
Preclinical
July, 2017
Not reported, still needs experiments to prove
Wnt
Reduces the expression of CSC marker and sphere formation ability
CRC
Preclinical
August, 2017
JIB-04
β-catenin
Inhibits the metastasis of colorectal CSCs
Colorectal cancer
Preclinical
April 26, 2018
Without general toxicity in JIB-04-treated mice, but still needs experiments to prove [25]
DTX and SFN
β-catenin
Inhibits the self-renewal ability of breast CSCs
Breast cancer
Preclinical
July, 2016
DTX has the side effect of neurological toxicity, nausea, diarrhea, and alopecia, but SFN is without significant toxicity [26, 27]
PP
β-catenin
Inhibits the self-renewal ability of breast CSCs
Breast cancer
Preclinical
March, 2016
PP shows no obvious toxicity in mice. But the poor targeting of it made the dosage large. It would be better to improve dosage form and develop new derivatives [28]
OXT-328
October, 2012
Safety according to the recent researches [28]
AD and Ts
Wnt/β-catenin
Decreases CSC number and activity, and reduces CSC marker expression (such as SOX2, ALDH1, and NOS2)
Lung cancer
Preclinical
December 3, 2019
AD has hepatoxicity, and TS is without toxic side effects in nude mice [29, 30]
In clinical trials, several small-molecule compounds have been used to target CSCs through the Wnt/β-catenin signaling pathway. For example, LGK-974 (Wnt974) can target porcupine to inhibit the post-translational acetylation of Wnt, thereby inhibiting Wnt secretion, and it has been reported that Wnt974 can inhibit the proliferation of breast CSCs (BCSCs) [34, 35]. Notably, in a phase I study, Wnt974 was found to be safe and effective in treating triple negative breast cancer (TNBC) (NCT01351103). Niclosamide has been approved by the FDA as an antihelminthic, and as a Wnt/β-catenin pathway inhibitor, it has anticancer ability that has been established by various studies. Niclosamide has been shown to selectively target ovarian CSCs [36]. In addition, niclosamide can decrease the population of ALDH+ cells by reducing the expression of LRP6 (a Wnt ligand receptor that plays a key role in the Wnt/β-catenin pathway) and β-catenin in basal-like breast cancer [37]. Notably, in a phase II trail, niclosamide was proved to safely and effectively treat colorectal cancer (CRC) [12]. Mechanistically, it can reduce the expression of many signaling components in the Wnt/β-catenin signaling pathway, the CSC population, and the self-renewal ability of CRC cells [38]. Additionally, ONC201, which is in a phase I/II study for patients with advanced cancer (NCT02038699), can inhibit CSC self-renewal and the expression of CSC-related genes in prostate and glioblastoma tumors through suppressing the Wnt signaling pathway [39].
Furthermore, there are many potential small-molecule compounds targeting CSCs through Wnt/β-catenin signaling pathway in preclinical experiments. For example, XAV939 can inhibit β-catenin signaling and thus attenuate CSC progression by interacting with the terminal anchor polymerase-binding domain (TBD) in Axin [40], which can abrogate CSC-mediated chemoresistance in head and neck squamous cell carcinoma (HNSCC) and colon cancer cells [41, 42]. Trifluoperazine (TFP) is used as an antipsychotic and an antiemetic, and it has been found to inhibit lung CSC spheroid formation ability and suppress lung CSC marker expression (such as CD44/CD133) by inhibiting Wnt/β-catenin signaling [43]. Chelerythrine chloride (Chelerythrine) can downregulate β-catenin and inhibit CSC invasion, spheroid-forming ability, and the expression of the stem marker SOX2 in non-small cell lung carcinoma (NSCLC) [44]. FH535 can suppress the expression of the pancreatic CSC marker CD24 and CD44 by inhibiting the Wnt/β-catenin signaling pathway [45]. Wnt-C59 (C59), an inhibitor of Wnt, can decrease the sphere formation ability of CSCs in a dose-dependent manner in nasopharyngeal carcinoma (NPC) [46]. IWR-1, a tankyrase inhibitor, can impair osteosarcoma CSC self-renewal, hamper the expression of key stem markers in osteosarcoma, and increase doxorubicin sensitivity in vivo by inhibiting β-catenin translocation [24]. IC-2, a novel small-molecule Wnt inhibitor, can reduce the population of CD44+ (liver CSCs) and the sphere-forming ability of hepatocellular carcinoma (HCC) cells [47]. It can also reduce the expression of CSC markers and the sphere formation ability in CRC. In addition, it can increase the sensitivity of 5-FU in the DLD-1 CRC cell line [48]. JIB-04, a selective inhibitor of histone demethylase, can inhibit the metastasis of colorectal CSCs by regulating the recruitment of β-catenin [49]. The combination of docetaxel (DTX) and sulforaphane (SFN), pyrvinium pamoate (PP), and phosphor-sulindac (OXT-328) can inhibit CSC self-renewal ability, the EMT (epithelial-mesenchymal transition), and drug resistance by decreasing β-catenin expression in BCSCs [5052]. Additionally, actinomycin D (AD) and telmisartan (TS) can also decrease the CSC number and activity and reduce CSC marker expression (such as SOX2, ALDH1, and NOS2) in lung cancer by inhibiting the Wnt-β/catenin signaling pathway [53].

Notch signaling pathway inhibitors

The Notch signaling pathway is an evolutionarily conserved pathway that is closely related to all aspects of cancer biology including CSC progression, angiogenesis, and tumor immunity [54]. The Notch pathway mainly consists of Notch receptors (Notch 1–4) and Notch ligands (Jagged 1, Jagged 2, delta-like ligand (DLL) -1, DLL-3, and DLL-4). When the receptors bind to the ligands, the Notch intracellular domain (NICD) is released into the nucleus through three cleavage processes mediated by γ-secretase, thereby activating the transcription of Notch target genes (Hes-1 and Hey-1) [55]. The activation of the Notch signaling pathway promotes tumor proliferation and metastasis; in contrast, inhibition of this pathway can eliminate CSCs and increase drug sensitivity. Therefore, genes in the Notch signaling pathway may represent potential cancer therapeutic targets [56]. Notch inhibitors can be used alone or in combination with chemotherapy agents to treat cancer and prevent recurrence [57].
Currently, inhibitors targeting the Notch signaling pathway mostly target γ-secretase or Notch ligands (Table 2). For example, MK-0752, a γ-secretase inhibitor, can decrease the population of CD44+/CD24 and ALDH+, reduce mammosphere-forming efficiency, and inhibit tumor regeneration in BCSCs [63]. In a phase I study, the combined use of MK-0752 with docetaxel increased MK-0752-induced anti-BCSC ability and improved the efficiency of docetaxel in treating breast cancer (NCT00645333). PF-03084014, another γ-secretase inhibitor, can inhibit CSC self-renewal and proliferation and induce CSC differentiation by targeting Notch signaling pathways in HCC [64]. PF-03084014 can also decrease the CD44+/CD24 and ALDH+ populations by suppressing N1ICD cleavage and the expression of Hes-1 and Hey-1 in pancreatic cancer [65]. In a phase II trail, PF-03084014 combined with gemcitabine and nab-paclitaxel increased the overall survival of patients with metastatic pancreatic adenocarcinoma (NCT02109445) compared to that of patients treated with PF-03084014. When combined with docetaxel, PF-03084014 can increase docetaxel efficiency against breast cancer; this combination is in a phase I study (NCT01876251). Mechanistically, PF-03084014 can diminish CD133+/CD44+ and ALDH+ subpopulations and eliminate BCSCs by targeting the Notch signaling pathway, thereby decreasing drug resistance [66].
Table 2
Small-molecule compounds inhibiting CSC progression through suppressing Notch signaling pathway
Name
Target
Mechanism
Type of cancer
Phase
NCT number (starting time)/publication date
Assessment
MK-0752
γ-secretase
Decreases the population of CD44+/CD24 and ALDH+, reduces mammosphere-forming efficiency, and inhibits tumor regeneration in BCSCs
Breast cancer
Phase I
NCT00645333 (March 27, 2008)
Well tolerated, but exists dose-limiting toxicity (DLT) [58]
PF-03084014
γ-secretase
Inhibits CSC self-renewal and proliferation, and induces CSCs differentiation
HCC
Preclinical
August, 2017
Induces gastrointestinal toxicity and exists DLT [59]
N1ICD, Hes-1, and Hey-1
Decreases CD44+/CD24− and ALDH+ population
Pancreatic cancer
Phase II
NCT02109445(April 9, 2014)
Notch
Diminishes CD133+/CD44+ and ALDH+ subpopulations and eliminates CSCs
Breast cancer
Phase I
NCT01876251(June 12, 2013)
RO4929097
γ-secretase
Combined with 5-FU can decrease the proportion of CSC subgroup
INS
Preclinical
February, 2018
Fatigue is the most common toxicities, but it has DLT [60]
DAPT
Notch1
Inhibits the proliferation of LSCs and regulates LSC self-renewal
Leukemia
Preclinical
December, 2006
Induces low toxicity in cell and mice [61]
Inhibits the self-renewal ability of ovarian CSCs and the expression of stem markers
Ovarian cancer
Preclinical
June, 2011
Quinomycin A
Notch ligands
Inhibits pancreatic cancer microsphere formation, the stem marker and the number of CSCs
Pancreatic cancer
Preclinical
January 19, 2016
Induces gastrointestinal toxicity [62]
Additionally, the γ-secretase inhibitor RO4929097 can significantly inhibit Notch target genes Hes1 and Hey1, and it is in a phase II clinical setting to treat breast cancer, ovarian cancer, and renal cell carcinoma [67]. The combination of RO4929097 and 5-FU can decrease the proportion of the CSC subgroup with insulinomas (INS) [68]. DAPT (GSI-IX) is a new type of Notch1 inhibitor that was initially used to treat Alzheimer’s disease, and increasing number of studies have shown that it can inhibit CSCs [69]. Studies have demonstrated that DAPT can inhibit the proliferation and self-renewal ability of leukemia stem cells (LSCs) and ovarian CSCs [70, 71]. Additionally, quinomycin A can inhibit pancreatic cancer microsphere formation, stem marker expression, and CSC number by decreasing the expression of Notch ligands [72].

Hh signaling pathway inhibitors

The Classical Hh signaling pathway is critical for embryonic development. The Hh signaling pathway regulates the self-renewal of CSCs and tissue homeostasis in cancer [80]. When extracellular Hh ligands (SHh, IH, and DHh) bind to PTCH, the inhibition of PTCH on Smoothened (SMO) is decreased, thereby GlI is translocated to the nucleus and induces the transcription of target genes [81]. Abnormal activation of the Hh signaling pathway is a crucial driver of breast cancer, prostate cancer, NSCLC, gastric cancer, and hematopoietic malignancies [82]. Hh signaling pathway inhibitors have been proven to be effective in early clinical trials. In addition, the development of Hh inhibitors has drawn significant interest for anticancer drug development. Recently, it has been reported that inhibition of the Hh signaling pathway can inhibit the self-renewal and drug resistance of pancreatic and breast CSCs [83, 84] (Table 3).
Table 3
Small-molecule compounds inhibiting CSC progression through suppressing Hh signalling pathway
Name
Target
Mechanism
Type of cancer
Phase
NCT number (starting time)/publication date
Assessment
Glasdegib
Hh
Attenuates the potential of leukemia-initiation and increases the sensitivity of LSCs to chemotherapy
Leukemia
Approved
November 21, 2018
Induces common side effect of chemotherapy drugs such as fatigue, nausea, and febrile neutropenia, but also has embryo-fetal toxicity [73]
Sonidegib
SMO
Downregulates the expression of CSC markers and increases the sensitivity to paclitaxel
Breast cancer
Phase I
NCT02027376 (January 6, 2014)
Induces myalgia, fatigue, and abnormal hepatic function, and gastrointestinal toxicity and alopecia are related to the dose of Sonidegib [74, 75]
Vismodegib
SMO
Inhibits BCSC self-renewal and mammosphere formation
Breast cancer
Phase II
NCT02694224 (February 29, 2016)
DLT, hyperbilirubinemia [76]
Suppresses pancreatic CSC proliferation and survival
Pancreatic cancer
Phase II
NCT01064622(February 8, 2010)
Decreases the stem markers (such as CD44 and ALDH) of colon CSCs
Colorectal cancer
Phase II
NCT00636610(March 14, 2008)
Ciclesonide
Hh
Inhibits the growth of lung CSCs
Lung cancer
Preclinical
February 4, 2020
Well tolerated, but as corticosteroid, it may inhibit bone growth [77]
Cyclopamine
SMO
Inhibits bladder CSC self-renewal
Bladder cancer
Preclinical
March 1, 2016
Induces holoprosencephaly, dystonia, and lethargy in rodents [78]
GANT61
GLI1 and GLI2
Decreases the CSC population
Breast cancer
Preclinical
May, 2017
No side effects in the mice according to the current studies [79]
For example, ciclesonide was approved by the FDA to treat asthma, and it was found that ciclesonide can inhibit the growth of lung CSCs through Hh signaling-mediated SOX2 regulation [85]. Sonidegib, an SMO antagonist, was approved by the FDA for the treatment of advanced basal cell carcinoma [86]. Recently, it was shown that sonidegib can downregulate the expression of CSC markers and increase the sensitivity of TNBC to paclitaxel, thus improving patient survival and reducing metastasis [87]. In addition, in a phase I study, sonidegib obtained a better result in advanced TNBC when it was combined with docetaxel (NCT02027376). Vismodegib (GDC-0449), another SMO inhibitor, was approved by the FDA to treat basal cell carcinoma [88]. Recently, many studies have found that vismodegib can inhibit BCSC self-renewal and mammosphere formation [89]. In a phase II trial, vismodegib was added to neoadjuvant chemotherapy for TNBC patients (NCT02694224). It can also suppress pancreatic CSC proliferation and survival by inhibiting Hh signaling pathways [90]. In a phase II trial, vismodegib combined with gemcitabine and nab-paclitaxel was used against untreated metastatic pancreatic cancer [91]. In another phase Ib/II trial, vismodegib plus gemcitabine was used to treat metastatic pancreatic cancer [92]. Additionally, vismodegib decreased the stem markers (such as CD44 and ALDH) of colon CSCs [93], and it was used to treat untreated metastatic CRC in a phase II trial [94]. These results suggest that vismodegib can target CSCs through the Hh signaling pathway. Furthermore, glasdegib (PF-04449913), an Hh signaling pathway inhibitor, was approved by the FDA to treat acute myeloid leukemia [73]. Glasdegib can attenuate the potential of leukemia-initiation and increase the sensitivity of LSCs to chemotherapy by inhibiting the Hh signaling pathway [95]. Cyclopamine is a natural compound that can specifically target SMO and inhibit the Hh signaling pathway [96], and it was reported that cyclopamine can inhibit bladder CSC self-renewal [97]. GANT61, another Hh inhibitor, can decrease the CSC population by downregulating the expression of GLI1 and GLI2 in ER (estrogen receptor)-positive breast cancer [98].

Hippo pathway activators

The Hippo signaling pathway plays an essential role in CSC self-renewal, the EMT, and drug resistance. Upon activation of the Hippo signaling pathway, MST1/2 phosphorylate and activate LATS1/2. Then, LATS1/2 inactivate YAP/TAZ, which was subsequently translocated into the cytoplasm, and thus inhibits the expression of TEAD (TEA domain family member)-mediated genes, thereby suppressing CSC progression [106]. In contrast, inhibition of the Hippo signaling pathway activates YAP/TAZ, conferring CSC-like characteristics to the cell and leading to tumorigenesis [107]. YAP/TAZ-TEAD acts as a tumor promoter in the Hippo signaling pathway, while the other members of the Hippo signaling pathway are mostly tumor suppressor genes. Thus, targeting YAP/TAZ may serve as a strategy to inhibit CSCs (Table 4).
Table 4
Small-molecule compounds inhibiting CSC progression through activating Hippo pathway
Name
Target
Mechanism
Type of cancer
Phase
NCT number (starting time)/publication date
Assessment
Verteporfin
YAP/TAZ
Reduces the expression of CSC markers and suppresses CSC proliferation
Gastric and esophageal cancer
Preclinical
August 1, 2014
Without visible toxicity in the mice [99]
gastric cancer
Preclinical
Apr 15, 2020
Evodiamine
LATS1/2
Inhibits the proliferation of colon CSCs
Colon cancer
Preclinical
December 10, 2019
Induces low toxicity and still needs much experiments to prove [100]
Fluvastatin
YAP
Reduces the expression of CD44 and the characteristics of malignant mesothelioma stem cells
Malignant mesothelioma
Preclinical
January 28, 2017
Without any genotoxic, and relatively safe in patients [101, 102]
Atorvastatin
TAZ
Decreases MDA-MB 231 cells stemness-related features (such as the decrease of CD44+/CD24- subpopulation of cells)
Breast cancer
Phase II
NCT02416427(April 15, 2015)
Muscle loss [103]
CA3
YAP/TEAD
Suppresses tumor microsphere, formation and reduces the proportion of ALDH1+ cells
Esophageal adenocarcinoma
Preclinical
February, 2018
Without apparent toxicity in mice according to the current studies [104]
CPZ
YAP
Inhibits tumor microsphere-formation and stem marker expression
Breast cancer
Preclinical
April 1, 2019
Induces fatal hepatic failure [105]
For instance, verteporfin is a photosensitizer approved by the FDA, and it has garnered increasing interest for its anticancer role in gastric and esophageal cancer. Verteporfin can inhibit the transcriptional activity of YAP/TAZ-TEAD, reduce the expression of CSC markers, and suppress CSC proliferation [108, 109]. Evodiamine (Evo), which is isolated from the Chinese herb Evodia rutaecarpa Benham, can activate MST1/2-mediated phosphorylation of LATS1/2, which leads to YAP/TAZ phosphorylation and prevents YAP/TAZ translocation from the cytoplasm into the nucleus, and it has been shown that Evo can inhibit the proliferation of colon CSCs [110, 111]. Additionally, tanshinone IIA and limonin, which are extracted from Chinese herbs, have been shown to attenuate the stemness of cervical carcinoma stem cells by inhibiting the cytoplasmic-nuclear translocation of YAP [112, 113]. Moreover, statins such as fluvastatin can reduce the expression of CD44 by accelerating YAP phosphorylation, thereby reducing the characteristics of malignant mesothelioma stem cells and drug resistance [114]. Atorvastatin, another stain, can target TAZ in breast cancer, and is in a phase II trial (NCT02416427). Notably, atorvastatin can decrease the stemness of MDA-MB 231 cells, as evident by the decrease in the CD44+/CD24 subpopulation of cells by inducing LATS1 expression and downregulating the expression of YAP/TAZ [115].
Recently, a new type of YAP inhibitor, CA3, was screened from a chemical library and found to attenuate the transcriptional activity of YAP/TEAD, and CA3 shows an excellent ability to target CSCs and inhibit tumor growth, as evident by its role in suppressing tumor sphere formation and reducing the proportion of ALDH1+ cells [104]. In addition, the antipsychotic drug chlorpromazine (CPZ) can kill breast cancer and BCSCs, which were characterized by inhibited tumor microsphere-formation and stem marker expression through promoting YAP degradation [116].

Selective inducers of signaling pathways that contribute to cell death

According to measurable biochemical characteristics and molecular mechanisms, signaling pathways contributing to cell death mainly include apoptosis, autophagy, necroptosis, and ferroptosis. Cancer cells also undergo multiple forms of cell death during tumor development, including apoptosis, autophagy, and necrosis. Recently, ferroptosis has been shown to play a critical role in the development of cancer and may be a beneficial anticancer treatment strategy, which has gradually gained attention. Because classic apoptosis-inducing drugs have a poor effect on CSCs, current research is more intent on inhibiting CSCs by inducing autophagy and ferroptosis, which can effectively inhibit cancer recurrence and metastasis

Autophagy and CSCs

Autophagy is a self-digesting mechanism in which proteins, lipids, and damaged organelles (such as mitochondria) are sequestered into vesicles called autophagosomes for degradation and recycling. Under physiological conditions, autophagy is critical for maintaining cell homeostasis and controlling protein and organelle quality. High levels of autophagy often occur in CSCs. Autophagy can help CSCs maintain their diversity and overcome low nutrients and hypoxia in the tumor microenvironment; therefore, it promotes CSCs to metastasis, drug resistance, and immune surveillance evasion [122]. Recently, autophagy was associated with CSC progression in breast cancer, NSCLC, prostate cancer, leukemia, gastric cancer, and myeloma, and its dysfunction affects the self-renewal ability of CSCs. Therefore, autophagy can be used as a CSC target. Although promotion of autophagy in CSCs is specific, different types, periods, and microenvironments have been shown to inhibit CSC progression through autophagy. For example, in some acute myeloid leukemias, many autophagy-related genes are mutated or downregulated in patients [123]. Therefore, currently, the compounds targeting autophagy mostly inhibit the autophagy of CSCs to suppress CSC progression (Table 5).
Table 5
Small-molecule compounds targeting autophagy to inhibit CSCs
Name
Target
Mechanism
Type of cancer
Phase
NCT number (starting time)/publication date
Assessment
CQ
Autophagy
Targets CSCs by inhibiting autophagy
Breast cancer
Phase II
NCT02333890 (January 7, 2015)
Induces cardiotoxicity [117]
Autophagy
Inhibits the stemness marker of CD133+ and decreases the CSC proportions
NSCLC
Preclinical
December, 2019
HCQ
Autophagy
Eliminates LSCs
Leukemia
Phase II
NCT00771056 (October 10, 2008)
Induces retinal toxicity [118]
Pantoprazole
Autophagy
Inhibits autophagy
Prostate cancer
Phase II
NCT01748500(December 12, 2012)
DLT, grade 3 to 4 [119]
EMT/β-catenin
Inhibits the chemoresistance of gastric cancer stem cells
Gastric cancer
Preclinical
December, 2016
3-MA
Autophagy
Reduces the resistance of mesenchymal stem cells
Myeloma
Preclinical
September, 2017
No significant side effect [120]
Rott
Autophagy
Induces autophagy leading to breast CSC death
Breast cancer
Preclinical
December 23, 2013
Without toxicity in the mice [121]
For example, chloroquine (CQ) is widely used in clinical antimalarial drugs and can be used in combination with anticancer drugs to treat cancer. CQ can effectively target CSCs by inhibiting autophagy, which causes the destruction of mitochondrial structures and double-stranded DNA, and the combination of CQ and carboplatin may be a useful adjuvant drug for treating TNBC [124]. CQ can inhibit the expression of stemness markers such as CD133 and decrease the proportions of CSCs, subsequently enhancing the efficacy of cisplatin against NSCLC by inhibiting autophagy [125]. In a phase II study, CQ inhibited breast cancer (NCT02333890). In addition, hydroxychloroquine (HCQ), a derivative of CQ, shows a stronger anticancer ability by targeting autophagy [126]. Additionally, the combination of HCQ and imatinib (IM) can effectively eliminate LSCs in chronic myeloid leukemia, and it is in a phase II trial [127]. A high dose of pantoprazole, a proton pump inhibitor (PPI), can affect docetaxel resistance in metastatic castration-resistant prostate cancer (mCRPC) by inhibiting autophagy, and it is in a phase II trial [128]. Moreover, pantoprazole can inhibit the chemoresistance of gastric CSCs [129]. Therefore, pantoprazole may target CSCs by inhibiting autophagy.
Moreover, concanamycin A is a selective inhibitor of V-ATPase that can inhibit the degradation of autolysosomes but has no effect on the formation of lysosomes; therefore, lysosomal degradation inhibitors may not reduce the rate of autophagosome-mediating chelation in destroying mitochondria. In this situation, some mitochondrial-dependent drugs may reduce the efficiency of inhibitors on CSCs; therefore, targeting early autophagy such as with VPS34 or ULK1 inhibitors may lead to better results. In addition, 3-methyladenine (3-MA) can prevent the formation of autolysosomes and reduce the resistance of mesenchymal stem cells; therefore, it can provide new treatment strategies for patients with multiple myeloma resistance [130]. Rottlerin (Rott), an active molecule that is isolated from Mallotus philippensis, is used in the treatment of allergies and helminthiasis and can induce autophagy, leading to BCSC death [131]. However, it is noteworthy that, although autophagy-targeting therapy has fewer side effects on normal cells than do conventional therapies, it also has some unknown effects that remain to be explored.

Ferroptosis and CSCs

Ferroptosis is a new type of programmed cell death that is different from apoptosis, necrosis, and autophagy at the morphological and biochemical levels. It is induced by the accumulation of iron and lipid peroxidation caused by reactive oxygen species (ROS). The destruction of iron homeostasis and uncontrolled lipid peroxidation are two key features of ferroptosis [132]. The regulation of intracellular iron homeostasis is mainly regulated by IRP2 (iron regulatory protein 2). IRP2 can bind to iron-responsive elements (IREs) in mRNA 5'- and 3'-untranslated regions (UTRs) to inhibit transcription or stabilize mRNA. In the absence of iron, IRP2 binds to 5'-end IREs in FPN1 and ferritin to decrease their mRNA stability, and it binds to 3'-end IREs in TFR1 and DMT1 to stabilize mRNA. When there is excess iron in the cells, the synthesis of FPN1 and ferritin mRNA is inhibited and the degradation of TFR1 and DMT1 mRNA is enhanced (Fig. 2) [133].
Additionally, the selenoenzyme glutathione peroxidase 4 (GPX4), which can inhibit the peroxidation of phospholipids (PLs), is considered to be a regulator of ferroptosis, and inactivation of GPX4 will lead to an increase in lipid peroxides. In addition, the accumulation of iron produces lipid peroxides in a nonenzymatic or enzyme-dependent manner [134]. The nonenzymatic reaction of iron to produce lipid peroxides is the Fenton reaction, Fe2+ + H2O2 → Fe3+ + (OH) + OH ·[135, 136] (Fig. 3). Another pathway for ferroptosis is mediated by the Xc system, formed by SLC3A2 and SLC7A11, which internalizes cystine cells and expels glutamic acid, and then cystine is reduced to cysteine. Cysteine participates in the synthesis of GSH. GSH acts as an electron donor to maintain the activity of GPX4, thereby preventing ferroptosis.

Relationship between ferroptosis and CSCs

The changes in iron homeostasis in CSCs are usually manifested by high intracellular iron content. In addition, abnormal iron metabolism is associated with accelerated tumor growth and a poor prognosis for cancer patients. Therefore, because of the dependence of cancer on iron, iron-dependent mechanisms such as ferroptosis can be used as targets for drug development [137]. A higher level of iron in a CSC may affect its redox state, which is manifested by the increase in peroxidation and OH ·[138, 139].
Recently, it was reported that the levels of TfR1 and its ligand transferrin expressed by glioblastoma stem cells are higher than those expressed by non-CSCs, and iron-tracking experiments using these CSCs have shown that the level of iron intake by CSCs is greater than that internalized by non-CSCs [140], indicating that increased iron intake may also be a characteristic of CSCs. Additionally, ferritin is overexpressed in a variety of cancers, including breast cancer, pancreatic cancer, liver cancer, Hodgkin’s lymphoma, and glioblastoma. Ferritin may protect CSCs, but the degradation of ferritin produces a source of iron leading to ferroptosis. Targeting the H and L subunits of ferritin with siRNA causes a significant reduction in the growth of CSCs in vivo and in vitro [140].

Ferroptosis and small-molecule compounds

Current studies on the activators of ferroptosis are relatively extensive, but there are few small-molecule compounds that can target CSCs by mediating ferroptosis (Fig. 4). Additionally, the role of GPX4 in ferroptosis is decisive. Some experiments have already confirmed that the dysfunction of GPX4 causes mesenchymal stem cell ferroptosis; therefore, small-molecule compounds targeting GPX4 may induce ferroptosis in CSCs, although experimental proof is currently lacking.
The following is a summary of some ferroptosis inducers. Erastin, sulfasalazine, and sorafenib can reduce the level of cystine by inhibiting the Xc system. Sulfasalazine can also inhibit the Xc system, thereby inhibiting the progression of CSCs overexpression of CD44 in gastrointestinal cancer [141]. (1S, 3R)-RSL3, altretamine and withaferin A can increase lipid peroxidation by inhibiting or silencing GPX4 function [142]. Notably, salinomycin can increase the production of lipid peroxidation by blocking iron transport and depleting ferritin, by which it can specifically kill CSCs [143]. Other studies indicate that a combination of salinomycin and docetaxel can effectively kill gastric CSCs [144]. Salinomycin can effectively destroy non-CSCs and CSCs, as well as cancer cells with a multi-drug resistance (MDR) phenotype; therefore, it shows strong antitumor activity against a variety of cancers [145]. Additionally, salinomycin-loaded gold nanoparticles show enhanced ability to target BCSCs [146]. Considering the effect of salinomycin in killing CSCs, many salinomycin derivatives have also been developed and exhibit higher activity and selectivity, such as ironomycin and the products of C20-amination, C1-esterification, C9-oxidation, and C28-dehydration. These derivatives of salinomycin can accumulate and isolate iron in the lysosome, and the accumulation of iron in the lysosome initiates the Fenton reaction, leading to increased permeability in the lysosomal membrane and cell death. They have been shown to be at least ten-fold more potent than salinomycin in vivo and in vitro. In addition, ironomycin can effectively reduce the number of CSCs in docetaxel-resistant xenograft models [143, 147]. Furthermore, ebselen, substituting pyrazole, and benzyl isothiourea, which are the inhibitors of DMT1, can selectively target BCSCs by blocking iron in lysosomes [148]. These results indicate that increasing iron levels in lysosomes can initiate cell death in a manner similar to ferroptosis, and thus specifically and effectively kill CSCs. However, the concrete mechanisms by which the drugs target DMT1 are still unclear.
Notably, recent studies have shown that ovarian CSCs rely on iron for self-renewal and metastasis [149]. This finding reveals an opportunity to treat cancer by regulating iron balance. For example, after treatment with erastin, CSCs are more likely to induce ferroptosis than are non-CSCs [149]. The combination of temozolomide (TMZ) and CQ can cause glioblastoma stem cells (GSCs) to die in the form of ferroptosis, and specifically, this combination can reduce the self-renewal of GSCs, weaken the invasion of glioblastoma, and improve the therapeutic efficiency of chemotherapy and radiotherapy, but the specific target has not been identified [150].
Recently, an increasing number of studies have shown that artemisinin derivatives have anticancer ability. For example, dihydroartemisinin (DHA) can increase iron levels in the cell and inhibit the synthesis of ferritin through the IRP-IRE axis, which increases the concentration of intracellular iron, making cancer cells (such as lung, colorectal, and breast cancer cells) more sensitive to ferroptosis [151]. In addition, it was found that DHA can inhibit sphere formation and stem marker (CD133, SOX2, and nestin) expression in glioma CSCs [152]. Therefore, DHA may inhibit CSCs through ferroptosis. Through a platform of induced cancer stem-like cells (iCSCLs) used for high-throughput screening, artesunate can induce mitochondrial dysfunction in CSCs; therefore, it can inhibit the stemness of CSCs [153]. Artesunate can induce ferroptosis in pancreatic cancer [154]. All these findings indicate that artemisinin derivatives may target CSCs through ferroptosis. Ferumoxytol is a superparamagnetic iron oxide nanoparticle approved by the FDA, and its anticancer ability is associated with ferroptosis [155]. Magnetic hyperthermia is a new method by which to selectively kill CSCs (A549 and MDA-MB-231), and the key aspect of this technology is superparamagnetic iron oxide nanoparticles [156]. The report indicated that ferumoxytol was a new material for use in magnetic hyperthermia [157]. In addition, DOX@FMT-MC, a novel magnetic hydrogel complex consisting of ferumoxytol, doxorubicin, and chitosan, was applied in the clinic to treat colon carcinoma [158]. Therefore, ferumoxytol may target CSCs by ferroptosis (Table 6).
Table 6
Small-molecule compounds that induce ferroptosis to inhibit CSCs
Name
Target
Mechanism
Type of cancer
Phase
NCT number (starting time)/publication date
Assessment
Salinomycin
Ferroptosis
Increases the production of lipid peroxidation by blocking iron transport and depleting ferritin; these can specifically kill CSCs
Breast cancer
Preclinical
October, 2017
Induces neural and muscular toxicity; changing dosages and making chemical modifications may reduce toxicity [159]
Ferroptosis
Combined with docetaxel can kill gastric CSCs
Gastric cancer
Preclinical
October, 2017
Ironomycin
Ferroptosis
Reduces the number of CSCs in docetaxel-resistant xenograft models
Breast cancer
Preclinical
February 21, 2020
The potency against CSCs is ten-fold that of salinomycin; may cause nephrotoxicity and hepatotoxicity [160]
Ebselen
Ferroptosis
Targets BCSCs by blocking iron in lysosomes
Breast cancer
Preclinical
February 21, 2020
Induces low toxicity and shows good blood-brain barrier permeability and oral absorption [161]
Substituted pyrazoles
February 21, 2020
Not reported; need studies to prove
Benzylisothioureas
February 21, 2020
Hemoglobinopathy including thalassemia [162]
TMZ and CQ
Ferroptosis
Causes glioblastoma stem cells (GSCs) to die through a form of ferroptosis and reduce the self-renewal ability of GSCs
Glioblastoma
Preclinical
August 6, 2018
TMZ is well tolerated, but may induce hematological toxicity and infection; CQ shows cardiotoxicity [117, 163]
DHA
Ferroptosis
Ferroptosis
Lung, colorectal, and breast cancer cells
Preclinical
January, 2020
Induces neurotoxicity, cardiotoxicity and the toxicity in embryos [164]
Apoptosis
Inhibits sphere formation and stem marker (CD133, SOX2, and nestin) expression in glioma CSCs
Gliomas
Preclinical
October, 2014
Artesunate
Mitochondrial
Inhibits the stemness of CSCs
Not mentioned
Preclinical
September 2, 2016
Excellently tolerated, and with low adverse effects [165, 166]
Ferroptosis
Induces cell death through ferroptosis
Pancreatic cancer
Preclinical
May 2, 2015
Ferumoxytol
Ferroptosis
Selectively kills CSCs (A549 and MDA-MB-231 cells)
Lung cancer and breast cancer
Preclinical
August 26, 2013
Well tolerated, but intravenous may cause hypersensitivity, hypotension, and gastrointestinal side effects [167169]
Sulfasalazine
System Xc
Inhibits the progression of CSCs overexpressing CD44
Gastrointestinal cancer
Preclinical
March 8, 2011
Induces gastrointestinal toxicity, and combed with other drugs, this side effect may be overcome [170]
According to the current research, the relationship between ferroptosis and CSCs has received increasing attention. In contrast, there are few reports of GPX4 in CSCs. Therefore, to some extent, the role of iron in CSCs is seemingly more dependent on GPX4 dysfunction, which causes normal stem cells to die in an iron-dependent manner [171, 172].

Multiple-signaling pathway inhibitors targeting CSCs

As we stated above, there are many signaling pathways regulating CSC progression. Hence, the compounds that simultaneously target multiple signaling pathways critical for CSC progression may yield better results than single-target treatments in controlling tumor occurrence, recurrence, and drug resistance (Table 7). For example, Z-ajoene, a compound extracted from garlic, was confirmed to inhibit CSC sphere-forming ability in glioblastoma multiforme (GBM), and Notch-, Wnt-, and Hh-related genes ware changed after treatment with Z-ajoene. In addition, Z-ajoene induced no cytotoxicity in normal cells [173]. Poziotinib, a pan-human epidermal growth factor receptor (HER) inhibitor, can decrease ovarian CSC sphere formation ability by disrupting the Wnt, Notch, and Hh signaling pathways in epithelial ovarian cancer (EOC) [176]. According to clinical studies, patients may experience diarrhea and rash when treated with poziotinib [174]. However, considering the multiple targets and anti-CSC activity of poziotinib, more studies are needed in the future. Additionally, 6-shogaol reduces the number of CD44+/CD24 cell subpopulation in BCSCs and inhibit their sphere-forming ability by inducing autophagy and inhibiting the Notch signaling pathway [177]. Importantly, 6-shogaol shows low toxicity induction in normal cells [175].
Table 7
Small-molecule compounds regulating multi-signaling pathways to inhibit CSCs
Name
Target
Mechanism
Type of cancer
Phase
NCT number (starting time)/publication date
Assessment
Z-ajoene
Notch, Wnt, and Hh
Inhibits CSC sphere-forming ability
Glioblastoma multiforme
Preclinical
July, 2014
Without cytotoxic in normal cells [173]
Poziotinib
Wnt, Notch, and Hh
Decreases ovarian CSC sphere formation ability
Epithelial ovarian cancer
Preclinical
May 21, 2020
Diarrhea and rash [174]
6-Shogaol
Notch and autophagy
Inhibits the number of CD44+/CD24− cell subpopulation and decreases sphere-forming ability
Breast cancer
Preclinical
September 10, 2015
Induces low toxicity in normal cells [175]

Conclusions

This review summarizes several well-characterized signaling pathways involved in CSCs, such as Wnt, Hh, Notch, Hippo, autophagy, and ferroptosis, and focuses on small-molecule compounds regulating these pathways (Fig. 5). Finally, these small-molecule compounds have the potential to kill CSCs, which provides a basis for cancer treatment. It is noteworthy that there are many other pathways, such as PI3K/Akt, MAPK, JAK/Stat, and TGF-β [178], essential for CSC survival, but these pathways are also widely engaged in other biological processes, and targeting them would not be specific to CSCs; however, the Wnt, Hh, Notch, and Hippo pathways are the mainstream pathways in CSCs, which have already been well studied. Therefore, in this current review, we summarize the small-molecule compounds targeting the Wnt, Hh, Notch, and Hippo pathways to kill CSCs. Although targeting ferroptosis to kill CSCs has recently been invoked, the effects have been strongly established by many studies [143, 147, 148]. Recently, although biologics have been rapidly developed, small-molecule compounds are still needed for clinical application.
Compared with biologics (such as monoclonal antibodies and antibody-drug conjugates), small-molecule compounds are inexpensive, and the generic drugs made from them are relatively simple, which means that small-molecule compounds may have the potential to compete with biologics. In addition, we present an overall assessment of these small-molecule compounds in Tables 1, 2, 3, 4, 5, and 6. Most chemotherapy drugs induce toxicity to different degrees. Therefore, we need to find a balance between anticancer effects and side effects of compounds used to target CSCs. The process from drug development to clinical use is long. Although approved drugs always induce specific toxicities, they have been well studied, and drug repurposing may offer a shortcut to prevent financial loss and detoured efforts. Therefore, in this review, we list many drugs approved by FDA, such as Wnt inhibitors (niclosamide, TFP, DTX and SFN, PP, AD and Ts); Notch inhibitors (DAPT); Hh inhibitor (glasdegib, sonidegib, vismodegib, ciclesonide); Hippo inhibitors (verteporfin, fluvastatin, atorvastatin, CPZ); autophagy regulators (CQ, HCQ, pantoprazole); ferroptosis inducers (TMZ and CQ, artesunate, ferumoxytol, sulfasalazine). These drugs display anti-CSC abilities, and some have been entered into clinical studies. In particular, an Hh inhibitor (glasdegib) has been approved by the FDA for its efficient anti-CSC action, which is similar to that of the biologics ELZONRIS. In addition, there are many new small-molecule compounds that are well tolerated, according to the current studies, but conclusive outcomes still need to be proven by more studies, and the toxicities of the following compounds have not been reported, which may be the result of insufficient experiments: Wnt inhibitors (ONC201, TFP, chelerythrine, FH535, Wnt-C59, IWR-1, IC-2, JIB-04, PP, OXT-328, OXT-328), Notch inhibitors (MK-0752, DAPT), Hh inhibitors (ciclesonide, GANT61), Hippo inhibitors (verteporfin, evodiamine, fluvastatin, CA3), autophagy regulators (3-MA, Rott), ferroptosis inducers (ebselen, substituted pyrazoles, TMZ, CQ, Artesunate, Ferumoxytol). Furthermore, we also listed some new CSC-targeting compounds that are in the preclinical or phase I/II stage: Wnt inhibitors (Wnt974, XAV939), Notch inhibitors (PF-03084014, RO4929097, quinomycin A), Hh inhibitor (cyclopamine), ferroptosis inducers (salinomycin, ironomycin, benzylisothioureas). These compounds also show anti-CSC activities, and the toxicities they induce may be reduced by changing the dosage, and developing new derivatives and combination therapies. To our surprise, some compounds are DLTs, especially the Notch inhibitors, and because of their side effect, additional research is needed to determine precise effective and safe dosage. In summary, according to the current studies, the compounds targeting CSCs are well tolerated in the mice or patients, and the compounds targeting multi-oncogenic signaling pathways may play important role in the clinic studies.
There are more detailed studies supporting the use of small-molecule compounds targeting Wnt, Hh, Notch, Hippo, and autophagy to inhibit CSCs, and many compounds are currently in clinical research. Although many small-molecule compounds activate ferroptosis, these studies have been conducted at the cell and mouse levels, and no clinical studies have been reported. However, based on the specificity and strong inhibitory ability of salinomycin and ironomycin and their derivatives on CSCs, these small-molecule compounds may have strong potential as target CSCs, and ferroptosis induced by iron in lysosomes may be a valuable prospect as a clinical target in CSCs to promote the development of anticancer drugs. However, it is noteworthy that there are different pathways in CSCs abnormally expressed; therefore, monotherapies that can target different pathways simultaneously or combined therapies may yield the best results in the future. Importantly, to our knowledge, targeting CSCs alone is sufficient for cancer therapy in the early stage of tumorigenesis, but it is not sufficient in the tumor development period, as many small-molecule compounds targeting CSCs cannot kill cancer cells, and the combined use of drugs targeting CSCs and chemotherapeutics may lead to better effects during tumor development period.

Acknowledgements

Not applicable
Not applicable
Not applicable

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
2.
Zurück zum Zitat Zhu Q, Shen Y, Chen X, He J, Liu J, Zu X. Self-renewal signalling pathway inhibitors: perspectives on therapeutic approaches for cancer stem cells. OncoTargets and therapy. 2020;13:525–40.PubMedPubMedCentralCrossRef Zhu Q, Shen Y, Chen X, He J, Liu J, Zu X. Self-renewal signalling pathway inhibitors: perspectives on therapeutic approaches for cancer stem cells. OncoTargets and therapy. 2020;13:525–40.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Nassar D, Blanpain C. Cancer stem cells: basic concepts and therapeutic implications. Annu Rev Pathol. 2016;11:47–76.PubMedCrossRef Nassar D, Blanpain C. Cancer stem cells: basic concepts and therapeutic implications. Annu Rev Pathol. 2016;11:47–76.PubMedCrossRef
4.
Zurück zum Zitat Katoh M. Canonical and non-canonical WNT signaling in cancer stem cells and their niches: cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review). Int J Oncol. 2017;51(5):1357–69.PubMedPubMedCentralCrossRef Katoh M. Canonical and non-canonical WNT signaling in cancer stem cells and their niches: cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review). Int J Oncol. 2017;51(5):1357–69.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105.PubMedCrossRef Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105.PubMedCrossRef
6.
Zurück zum Zitat Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell. 2014;14(3):275–91.PubMedCrossRef Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell. 2014;14(3):275–91.PubMedCrossRef
7.
Zurück zum Zitat An SM, Ding Q, Zhang J, Xie J, Li L. Targeting stem cell signaling pathways for drug discovery: advances in the Notch and Wnt pathways. Sci China Life Sci. 2014;57(6):575–80.PubMedCrossRef An SM, Ding Q, Zhang J, Xie J, Li L. Targeting stem cell signaling pathways for drug discovery: advances in the Notch and Wnt pathways. Sci China Life Sci. 2014;57(6):575–80.PubMedCrossRef
8.
Zurück zum Zitat Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, Yang SX, Ivy SP. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12(8):445–64.PubMedPubMedCentralCrossRef Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, Yang SX, Ivy SP. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015;12(8):445–64.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Bouvard C, Barefield C, Zhu S. Cancer stem cells as a target population for drug discovery. Future Med Chem. 2014;6(14):1567–85.PubMedCrossRef Bouvard C, Barefield C, Zhu S. Cancer stem cells as a target population for drug discovery. Future Med Chem. 2014;6(14):1567–85.PubMedCrossRef
10.
Zurück zum Zitat Takebe N, Harris PJ, Warren RQ, Ivy SP. Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nat Rev Clin Oncol. 2011;8(2):97–106.PubMedCrossRef Takebe N, Harris PJ, Warren RQ, Ivy SP. Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways. Nat Rev Clin Oncol. 2011;8(2):97–106.PubMedCrossRef
11.
Zurück zum Zitat Solzak JP, Atale RV, Hancock BA, Sinn AL, Pollok KE, Jones DR, Radovich M. Dual PI3K and Wnt pathway inhibition is a synergistic combination against triple negative breast cancer. NPJ breast cancer. 2017;3:17.PubMedPubMedCentralCrossRef Solzak JP, Atale RV, Hancock BA, Sinn AL, Pollok KE, Jones DR, Radovich M. Dual PI3K and Wnt pathway inhibition is a synergistic combination against triple negative breast cancer. NPJ breast cancer. 2017;3:17.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Burock S, Daum S, Keilholz U, Neumann K, Walther W, Stein U. Phase II trial to investigate the safety and efficacy of orally applied niclosamide in patients with metachronous or sychronous metastases of a colorectal cancer progressing after therapy: the NIKOLO trial. BMC Cancer. 2018;18(1):297.PubMedPubMedCentralCrossRef Burock S, Daum S, Keilholz U, Neumann K, Walther W, Stein U. Phase II trial to investigate the safety and efficacy of orally applied niclosamide in patients with metachronous or sychronous metastases of a colorectal cancer progressing after therapy: the NIKOLO trial. BMC Cancer. 2018;18(1):297.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Lin CK, Bai MY, Hu TM, Wang YC, Chao TK, Weng SJ, Huang RL, Su PH, Lai HC. Preclinical evaluation of a nanoformulated antihelminthic, niclosamide, in ovarian cancer. Oncotarget. 2016;7(8):8993–9006.PubMedPubMedCentralCrossRef Lin CK, Bai MY, Hu TM, Wang YC, Chao TK, Weng SJ, Huang RL, Su PH, Lai HC. Preclinical evaluation of a nanoformulated antihelminthic, niclosamide, in ovarian cancer. Oncotarget. 2016;7(8):8993–9006.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Ye T, Xiong Y, Yan Y, Xia Y, Song X, Liu L, Li D, Wang N, Zhang L, Zhu Y, et al. The anthelmintic drug niclosamide induces apoptosis, impairs metastasis and reduces immunosuppressive cells in breast cancer model. PLoS One. 2014;9(1):e85887.PubMedPubMedCentralCrossRef Ye T, Xiong Y, Yan Y, Xia Y, Song X, Liu L, Li D, Wang N, Zhang L, Zhu Y, et al. The anthelmintic drug niclosamide induces apoptosis, impairs metastasis and reduces immunosuppressive cells in breast cancer model. PLoS One. 2014;9(1):e85887.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Arrillaga-Romany I, Chi AS, Allen JE, Oster W, Wen PY, Batchelor TT. A phase 2 study of the first imipridone ONC201, a selective DRD2 antagonist for oncology, administered every three weeks in recurrent glioblastoma. Oncotarget. 2017;8(45):79298–304.PubMedPubMedCentralCrossRef Arrillaga-Romany I, Chi AS, Allen JE, Oster W, Wen PY, Batchelor TT. A phase 2 study of the first imipridone ONC201, a selective DRD2 antagonist for oncology, administered every three weeks in recurrent glioblastoma. Oncotarget. 2017;8(45):79298–304.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Arrillaga-Romany I, Odia Y, Prabhu VV, Tarapore RS, Merdinger K, Stogniew M, Oster W, Allen JE, Mehta M, Batchelor TT, et al. Biological activity of weekly ONC201 in adult recurrent glioblastoma patients. Neuro-oncology. 2020;22(1):94–102.PubMedCrossRef Arrillaga-Romany I, Odia Y, Prabhu VV, Tarapore RS, Merdinger K, Stogniew M, Oster W, Allen JE, Mehta M, Batchelor TT, et al. Biological activity of weekly ONC201 in adult recurrent glioblastoma patients. Neuro-oncology. 2020;22(1):94–102.PubMedCrossRef
17.
Zurück zum Zitat Liu M, Tu J, Gingold JA, Kong CSL, Lee DF. Cancer in a dish: progress using stem cells as a platform for cancer research. Am J Cancer Res. 2018;8(6):944–54.PubMedPubMedCentral Liu M, Tu J, Gingold JA, Kong CSL, Lee DF. Cancer in a dish: progress using stem cells as a platform for cancer research. Am J Cancer Res. 2018;8(6):944–54.PubMedPubMedCentral
18.
Zurück zum Zitat Budd GT, Bukowski RM, Lichtin A, Bauer L, Van Kirk P, Ganapathi R. Phase II trial of doxorubicin and trifluoperazine in metastatic breast cancer. Invest New Drugs. 1993;11(1):75–9.PubMedCrossRef Budd GT, Bukowski RM, Lichtin A, Bauer L, Van Kirk P, Ganapathi R. Phase II trial of doxorubicin and trifluoperazine in metastatic breast cancer. Invest New Drugs. 1993;11(1):75–9.PubMedCrossRef
19.
Zurück zum Zitat Li J, Yao QY, Xue JS, Wang LJ, Yuan Y, Tian XY, Su H, Wang SY, Chen WJ, Lu W, et al. Dopamine D2 receptor antagonist sulpiride enhances dexamethasone responses in the treatment of drug-resistant and metastatic breast cancer. Acta Pharmacol Sin. 2017;38(9):1282–96.PubMedPubMedCentralCrossRef Li J, Yao QY, Xue JS, Wang LJ, Yuan Y, Tian XY, Su H, Wang SY, Chen WJ, Lu W, et al. Dopamine D2 receptor antagonist sulpiride enhances dexamethasone responses in the treatment of drug-resistant and metastatic breast cancer. Acta Pharmacol Sin. 2017;38(9):1282–96.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Chmura SJ, Dolan ME, Cha A, Mauceri HJ, Kufe DW, Weichselbaum RR. In vitro and in vivo activity of protein kinase C inhibitor chelerythrine chloride induces tumor cell toxicity and growth delay in vivo. Clin Cancer Res. 2000;6(2):737–42.PubMed Chmura SJ, Dolan ME, Cha A, Mauceri HJ, Kufe DW, Weichselbaum RR. In vitro and in vivo activity of protein kinase C inhibitor chelerythrine chloride induces tumor cell toxicity and growth delay in vivo. Clin Cancer Res. 2000;6(2):737–42.PubMed
21.
Zurück zum Zitat Medvetz D, Sun Y, Li C, Khabibullin D, Balan M, Parkhitko A, Priolo C, Asara JM, Pal S, Yu J, et al. High-throughput drug screen identifies chelerythrine as a selective inducer of death in a TSC2-null setting. Mol Cancer Res. 2015;13(1):50–62.PubMedCrossRef Medvetz D, Sun Y, Li C, Khabibullin D, Balan M, Parkhitko A, Priolo C, Asara JM, Pal S, Yu J, et al. High-throughput drug screen identifies chelerythrine as a selective inducer of death in a TSC2-null setting. Mol Cancer Res. 2015;13(1):50–62.PubMedCrossRef
22.
Zurück zum Zitat Razak S, Afsar T, Almajwal A, Alam I, Jahan S. Growth inhibition and apoptosis in colorectal cancer cells induced by vitamin D-nanoemulsion (NVD): involvement of Wnt/beta-catenin and other signal transduction pathways. Cell Biosci. 2019;9:15.PubMedPubMedCentralCrossRef Razak S, Afsar T, Almajwal A, Alam I, Jahan S. Growth inhibition and apoptosis in colorectal cancer cells induced by vitamin D-nanoemulsion (NVD): involvement of Wnt/beta-catenin and other signal transduction pathways. Cell Biosci. 2019;9:15.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Proffitt KD, Madan B, Ke Z, Pendharkar V, Ding L, Lee MA, Hannoush RN, Virshup DM. Pharmacological inhibition of the Wnt acyltransferase PORCN prevents growth of WNT-driven mammary cancer. Cancer Res. 2013;73(2):502–7.PubMedCrossRef Proffitt KD, Madan B, Ke Z, Pendharkar V, Ding L, Lee MA, Hannoush RN, Virshup DM. Pharmacological inhibition of the Wnt acyltransferase PORCN prevents growth of WNT-driven mammary cancer. Cancer Res. 2013;73(2):502–7.PubMedCrossRef
24.
Zurück zum Zitat Martins-Neves SR, Paiva-Oliveira DI, Fontes-Ribeiro C, Bovee J, Cleton-Jansen AM, Gomes CMF. IWR-1, a tankyrase inhibitor, attenuates Wnt/beta-catenin signaling in cancer stem-like cells and inhibits in vivo the growth of a subcutaneous human osteosarcoma xenograft. Cancer Lett. 2018;414:1–15.PubMedCrossRef Martins-Neves SR, Paiva-Oliveira DI, Fontes-Ribeiro C, Bovee J, Cleton-Jansen AM, Gomes CMF. IWR-1, a tankyrase inhibitor, attenuates Wnt/beta-catenin signaling in cancer stem-like cells and inhibits in vivo the growth of a subcutaneous human osteosarcoma xenograft. Cancer Lett. 2018;414:1–15.PubMedCrossRef
25.
Zurück zum Zitat Wang L, Chang J, Varghese D, Dellinger M, Kumar S, Best AM, Ruiz J, Bruick R, Pena-Llopis S, Xu J, et al. A small molecule modulates Jumonji histone demethylase activity and selectively inhibits cancer growth. Nat Commun. 2013;4:2035.PubMedCrossRef Wang L, Chang J, Varghese D, Dellinger M, Kumar S, Best AM, Ruiz J, Bruick R, Pena-Llopis S, Xu J, et al. A small molecule modulates Jumonji histone demethylase activity and selectively inhibits cancer growth. Nat Commun. 2013;4:2035.PubMedCrossRef
26.
Zurück zum Zitat Kamal MM, Nazzal S. Development of a new class of sulforaphane-enabled self-emulsifying drug delivery systems (SFN-SEDDS) by high throughput screening: a case study with curcumin. Int J Pharm. 2018;539(1-2):147–56.PubMedCrossRef Kamal MM, Nazzal S. Development of a new class of sulforaphane-enabled self-emulsifying drug delivery systems (SFN-SEDDS) by high throughput screening: a case study with curcumin. Int J Pharm. 2018;539(1-2):147–56.PubMedCrossRef
27.
Zurück zum Zitat Kenmotsu H, Tanigawara Y. Pharmacokinetics, dynamics and toxicity of docetaxel: why the Japanese dose differs from the Western dose. Cancer Sci. 2015;106(5):497–504.PubMedPubMedCentralCrossRef Kenmotsu H, Tanigawara Y. Pharmacokinetics, dynamics and toxicity of docetaxel: why the Japanese dose differs from the Western dose. Cancer Sci. 2015;106(5):497–504.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Xu W, Lacerda L, Debeb BG, Atkinson RL, Solley TN, Li L, Orton D, McMurray JS, Hang BI, Lee E, et al. The antihelmintic drug pyrvinium pamoate targets aggressive breast cancer. PLoS One. 2013;8(8):e71508.PubMedPubMedCentralCrossRef Xu W, Lacerda L, Debeb BG, Atkinson RL, Solley TN, Li L, Orton D, McMurray JS, Hang BI, Lee E, et al. The antihelmintic drug pyrvinium pamoate targets aggressive breast cancer. PLoS One. 2013;8(8):e71508.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Koyama N, Nishida Y, Ishii T, Yoshida T, Furukawa Y, Narahara H. Telmisartan induces growth inhibition, DNA double-strand breaks and apoptosis in human endometrial cancer cells. PLoS One. 2014;9(3):e93050.PubMedPubMedCentralCrossRef Koyama N, Nishida Y, Ishii T, Yoshida T, Furukawa Y, Narahara H. Telmisartan induces growth inhibition, DNA double-strand breaks and apoptosis in human endometrial cancer cells. PLoS One. 2014;9(3):e93050.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Lamture G, Crooks PA, Borrelli MJ. Actinomycin-D and dimethylamino-parthenolide synergism in treating human pancreatic cancer cells. Drug Dev Res. 2018;79(6):287–94.PubMedPubMedCentralCrossRef Lamture G, Crooks PA, Borrelli MJ. Actinomycin-D and dimethylamino-parthenolide synergism in treating human pancreatic cancer cells. Drug Dev Res. 2018;79(6):287–94.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat de Sousa EMF, Vermeulen L. Wnt signaling in cancer stem cell biology. Cancers. 2016:8(7). de Sousa EMF, Vermeulen L. Wnt signaling in cancer stem cell biology. Cancers. 2016:8(7).
32.
33.
Zurück zum Zitat Takahashi-Yanaga F, Kahn M. Targeting Wnt signaling: can we safely eradicate cancer stem cells? Clin cancer res. 2010;16(12):3153–62.PubMedCrossRef Takahashi-Yanaga F, Kahn M. Targeting Wnt signaling: can we safely eradicate cancer stem cells? Clin cancer res. 2010;16(12):3153–62.PubMedCrossRef
34.
Zurück zum Zitat Takada R, Satomi Y, Kurata T, Ueno N, Norioka S, Kondoh H, Takao T, Takada S. Monounsaturated fatty acid modification of Wnt protein: its role in Wnt secretion. Dev Cell. 2006;11(6):791–801.PubMedCrossRef Takada R, Satomi Y, Kurata T, Ueno N, Norioka S, Kondoh H, Takao T, Takada S. Monounsaturated fatty acid modification of Wnt protein: its role in Wnt secretion. Dev Cell. 2006;11(6):791–801.PubMedCrossRef
35.
Zurück zum Zitat Zhao Z, Lu P, Zhang H, Xu H, Gao N. Nestin positively regulates the Wnt/β-catenin pathway and the proliferation, survival and invasiveness of breast cancer stem cells. Breast Cancer Res. 2014;16(4):408.PubMedPubMedCentralCrossRef Zhao Z, Lu P, Zhang H, Xu H, Gao N. Nestin positively regulates the Wnt/β-catenin pathway and the proliferation, survival and invasiveness of breast cancer stem cells. Breast Cancer Res. 2014;16(4):408.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Arend RC, Londono-Joshi AI, Samant RS, Li Y, Conner M, Hidalgo B, Alvarez RD, Landen CN, Straughn JM, Buchsbaum DJ. Inhibition of Wnt/beta-catenin pathway by niclosamide: a therapeutic target for ovarian cancer. Gynecol Oncol. 2014;134(1):112–20.PubMedCrossRef Arend RC, Londono-Joshi AI, Samant RS, Li Y, Conner M, Hidalgo B, Alvarez RD, Landen CN, Straughn JM, Buchsbaum DJ. Inhibition of Wnt/beta-catenin pathway by niclosamide: a therapeutic target for ovarian cancer. Gynecol Oncol. 2014;134(1):112–20.PubMedCrossRef
37.
Zurück zum Zitat Londono-Joshi AI, Arend RC, Aristizabal L, Lu W, Samant RS, Metge BJ, Hidalgo B, Grizzle WE, Conner M, Forero-Torres A, et al. Effect of niclosamide on basal-like breast cancers. Mol Cancer Ther. 2014;13(4):800–11.PubMedPubMedCentralCrossRef Londono-Joshi AI, Arend RC, Aristizabal L, Lu W, Samant RS, Metge BJ, Hidalgo B, Grizzle WE, Conner M, Forero-Torres A, et al. Effect of niclosamide on basal-like breast cancers. Mol Cancer Ther. 2014;13(4):800–11.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Park SY, Kim JY, Choi JH, Kim JH, Lee CJ, Singh P, Sarkar S, Baek JH, Nam JS. Inhibition of LEF1-mediated DCLK1 by niclosamide attenuates colorectal cancer stemness. Clin Cancer Res. 2019;25(4):1415–29.PubMedCrossRef Park SY, Kim JY, Choi JH, Kim JH, Lee CJ, Singh P, Sarkar S, Baek JH, Nam JS. Inhibition of LEF1-mediated DCLK1 by niclosamide attenuates colorectal cancer stemness. Clin Cancer Res. 2019;25(4):1415–29.PubMedCrossRef
39.
Zurück zum Zitat Prabhu VV, Lulla AR, Madhukar NS, Ralff MD, El-Deiry WS. Cancer stem cell-related gene expression as a potential biomarker of response for first-in-class imipridone ONC201 in solid tumors. Plos One. 2017;12(8):e0180541.PubMedPubMedCentralCrossRef Prabhu VV, Lulla AR, Madhukar NS, Ralff MD, El-Deiry WS. Cancer stem cell-related gene expression as a potential biomarker of response for first-in-class imipridone ONC201 in solid tumors. Plos One. 2017;12(8):e0180541.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Huang SMA, Mishina YM, Liu S, Cheung A, Stegmeier F, Michaud GA, Charlat O, Wiellette E, Zhang Y, Wiessner S. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature. 2009;461(7264):614–20.PubMedCrossRef Huang SMA, Mishina YM, Liu S, Cheung A, Stegmeier F, Michaud GA, Charlat O, Wiellette E, Zhang Y, Wiessner S. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature. 2009;461(7264):614–20.PubMedCrossRef
41.
Zurück zum Zitat Roy S, Roy S, Kar M, Chakraborty A, Kumar A, Delogu F, Asthana S, Hande MP, Banerjee B. Combined treatment with cisplatin and the tankyrase inhibitor XAV-939 increases cytotoxicity, abrogates cancer-stem-like cell phenotype and increases chemosensitivity of head-and-neck squamous-cell carcinoma cells. Mutat Res. 2019;846:503084.CrossRef Roy S, Roy S, Kar M, Chakraborty A, Kumar A, Delogu F, Asthana S, Hande MP, Banerjee B. Combined treatment with cisplatin and the tankyrase inhibitor XAV-939 increases cytotoxicity, abrogates cancer-stem-like cell phenotype and increases chemosensitivity of head-and-neck squamous-cell carcinoma cells. Mutat Res. 2019;846:503084.CrossRef
42.
Zurück zum Zitat Wu X, Luo F, Li J, Zhong X, Liu K. Tankyrase 1 inhibitior XAV939 increases chemosensitivity in colon cancer cell lines via inhibition of the Wnt signaling pathway. Int J Oncol. 2016;48(4):1333–40.PubMedPubMedCentralCrossRef Wu X, Luo F, Li J, Zhong X, Liu K. Tankyrase 1 inhibitior XAV939 increases chemosensitivity in colon cancer cell lines via inhibition of the Wnt signaling pathway. Int J Oncol. 2016;48(4):1333–40.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Yeh CT, Wu AT, Chang PM, Chen KY, Yang CN, Yang SC, Ho CC, Chen CC, Kuo YL, Lee PY, et al. Trifluoperazine, an antipsychotic agent, inhibits cancer stem cell growth and overcomes drug resistance of lung cancer. Am J Respir Crit Care Med. 2012;186(11):1180–8.PubMedCrossRef Yeh CT, Wu AT, Chang PM, Chen KY, Yang CN, Yang SC, Ho CC, Chen CC, Kuo YL, Lee PY, et al. Trifluoperazine, an antipsychotic agent, inhibits cancer stem cell growth and overcomes drug resistance of lung cancer. Am J Respir Crit Care Med. 2012;186(11):1180–8.PubMedCrossRef
44.
Zurück zum Zitat Heng WS, Cheah SC. Chelerythrine chloride downregulates beta-catenin and inhibits stem cell properties of non-small cell lung carcinoma. Molecules. 2020;25(1):224.PubMedCentralCrossRef Heng WS, Cheah SC. Chelerythrine chloride downregulates beta-catenin and inhibits stem cell properties of non-small cell lung carcinoma. Molecules. 2020;25(1):224.PubMedCentralCrossRef
45.
Zurück zum Zitat Liu L, Zhi Q, Shen M, Gong FR, Zhou BP, Lian L, Shen B, Chen K, Duan W, Wu MY, et al. FH535, a beta-catenin pathway inhibitor, represses pancreatic cancer xenograft growth and angiogenesis. Oncotarget. 2016;7(30):47145–62.PubMedPubMedCentralCrossRef Liu L, Zhi Q, Shen M, Gong FR, Zhou BP, Lian L, Shen B, Chen K, Duan W, Wu MY, et al. FH535, a beta-catenin pathway inhibitor, represses pancreatic cancer xenograft growth and angiogenesis. Oncotarget. 2016;7(30):47145–62.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Cheng Y, Phoon YP, Jin X, Chong SY, Ip JC, Wong BW, Lung ML. Wnt-C59 arrests stemness and suppresses growth of nasopharyngeal carcinoma in mice by inhibiting the Wnt pathway in the tumor microenvironment. Oncotarget. 2015;6(16):14428–39.PubMedPubMedCentralCrossRef Cheng Y, Phoon YP, Jin X, Chong SY, Ip JC, Wong BW, Lung ML. Wnt-C59 arrests stemness and suppresses growth of nasopharyngeal carcinoma in mice by inhibiting the Wnt pathway in the tumor microenvironment. Oncotarget. 2015;6(16):14428–39.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Seto K, Sakabe T, Itaba N, Azumi J, Oka H, Morimoto M, Umekita Y, Shiota G. A novel small-molecule WNT inhibitor, IC-2, has the potential to suppress liver cancer stem cells. Anticancer Res. 2017;37(7):3569–79.PubMed Seto K, Sakabe T, Itaba N, Azumi J, Oka H, Morimoto M, Umekita Y, Shiota G. A novel small-molecule WNT inhibitor, IC-2, has the potential to suppress liver cancer stem cells. Anticancer Res. 2017;37(7):3569–79.PubMed
48.
Zurück zum Zitat Urushibara S, Tsubota T, Asai R, Azumi J, Ashida K, Fujiwara Y, Shiota G. WNT/beta-catenin signaling inhibitor IC-2 suppresses sphere formation and sensitizes colorectal cancer cells to 5-fluorouracil. Anticancer Res. 2017;37(8):4085–91.PubMed Urushibara S, Tsubota T, Asai R, Azumi J, Ashida K, Fujiwara Y, Shiota G. WNT/beta-catenin signaling inhibitor IC-2 suppresses sphere formation and sensitizes colorectal cancer cells to 5-fluorouracil. Anticancer Res. 2017;37(8):4085–91.PubMed
49.
Zurück zum Zitat Kim MS, Cho HI, Yoon HJ, Ahn YH, Park EJ, Jin YH, Jang YK. JIB-04, a small molecule histone demethylase inhibitor, selectively targets colorectal cancer stem cells by inhibiting the Wnt/beta-catenin signaling pathway. Sci Rep. 2018;8(1):6611.PubMedPubMedCentralCrossRef Kim MS, Cho HI, Yoon HJ, Ahn YH, Park EJ, Jin YH, Jang YK. JIB-04, a small molecule histone demethylase inhibitor, selectively targets colorectal cancer stem cells by inhibiting the Wnt/beta-catenin signaling pathway. Sci Rep. 2018;8(1):6611.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Huang J, Tao C, Yu Y, Yu F, Zhang H, Gao J, Wang D, Chen Y, Gao J, Zhang G. Simultaneous targeting of differentiated breast cancer cells and breast cancer stem cells by combination of docetaxel- and dulforaphane-loaded self-assembled poly(D, L-lactide-co-glycolide)/hyaluronic acid block copolymer-based nanoparticles. J Biomed Nanotechnol. 2016;12(7):1463.PubMedCrossRef Huang J, Tao C, Yu Y, Yu F, Zhang H, Gao J, Wang D, Chen Y, Gao J, Zhang G. Simultaneous targeting of differentiated breast cancer cells and breast cancer stem cells by combination of docetaxel- and dulforaphane-loaded self-assembled poly(D, L-lactide-co-glycolide)/hyaluronic acid block copolymer-based nanoparticles. J Biomed Nanotechnol. 2016;12(7):1463.PubMedCrossRef
51.
Zurück zum Zitat Liang XU, Zhang LE, Chun HU, Liang S, Fei X. WNT pathway inhibitor pyrvinium pamoate inhibits the self-renewal and metastasis of breast cancer stem cells. Int J Oncol. 2016;48(3):1175–86.PubMedCrossRef Liang XU, Zhang LE, Chun HU, Liang S, Fei X. WNT pathway inhibitor pyrvinium pamoate inhibits the self-renewal and metastasis of breast cancer stem cells. Int J Oncol. 2016;48(3):1175–86.PubMedCrossRef
52.
Zurück zum Zitat Zhu C, Cheng KW, Ouyang N, Huang L, Sun Y, Constantinides P, Rigas B. Phosphosulindac (OXT-328) selectively targets breast cancer stem cells in vitro and in human breast cancer xenografts. Stem Cells, 75. 2012;30:2065.PubMedPubMedCentralCrossRef Zhu C, Cheng KW, Ouyang N, Huang L, Sun Y, Constantinides P, Rigas B. Phosphosulindac (OXT-328) selectively targets breast cancer stem cells in vitro and in human breast cancer xenografts. Stem Cells, 75. 2012;30:2065.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Green R, Howell M, Khalil R, Nair R, Yan J, Foran E, Katiri S, Banerjee J, Singh M, Bharadwaj S, et al. Actinomycin D and telmisartan combination targets lung cancer stem cells through the Wnt/beta catenin pathway. Sci Rep. 2019;9(1):18177.PubMedPubMedCentralCrossRef Green R, Howell M, Khalil R, Nair R, Yan J, Foran E, Katiri S, Banerjee J, Singh M, Bharadwaj S, et al. Actinomycin D and telmisartan combination targets lung cancer stem cells through the Wnt/beta catenin pathway. Sci Rep. 2019;9(1):18177.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Schweisguth F. Regulation of notch signaling activity. Current biology : CB. 2004;14(3):R129–38.PubMedCrossRef Schweisguth F. Regulation of notch signaling activity. Current biology : CB. 2004;14(3):R129–38.PubMedCrossRef
56.
Zurück zum Zitat Nefedova Y, Sullivan DM, Bolick SC, Dalton WS, Gabrilovich DI. Inhibition of Notch signaling induces apoptosis of myeloma cells and enhances sensitivity to chemotherapy. Blood. 2008;111(4):2220–9.PubMedCrossRef Nefedova Y, Sullivan DM, Bolick SC, Dalton WS, Gabrilovich DI. Inhibition of Notch signaling induces apoptosis of myeloma cells and enhances sensitivity to chemotherapy. Blood. 2008;111(4):2220–9.PubMedCrossRef
57.
Zurück zum Zitat Vandana V, Raghu N, S. TG, Murugesan K, Ashok G, B. KS, Gobianand K, Gowdru KC, M. BK: Targeting Notch signalling pathway of cancer stem cells. Stem Cell Investigation 2018, 5:5-5.PubMedPubMedCentralCrossRef Vandana V, Raghu N, S. TG, Murugesan K, Ashok G, B. KS, Gobianand K, Gowdru KC, M. BK: Targeting Notch signalling pathway of cancer stem cells. Stem Cell Investigation 2018, 5:5-5.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Krop I, Demuth T, Guthrie T, Wen PY, Mason WP, Chinnaiyan P, Butowski N, Groves MD, Kesari S, Freedman SJ, et al. Phase I pharmacologic and pharmacodynamic study of the gamma secretase (Notch) inhibitor MK-0752 in adult patients with advanced solid tumors. J Clin Oncol. 2012;30(19):2307–13.PubMedCrossRef Krop I, Demuth T, Guthrie T, Wen PY, Mason WP, Chinnaiyan P, Butowski N, Groves MD, Kesari S, Freedman SJ, et al. Phase I pharmacologic and pharmacodynamic study of the gamma secretase (Notch) inhibitor MK-0752 in adult patients with advanced solid tumors. J Clin Oncol. 2012;30(19):2307–13.PubMedCrossRef
59.
Zurück zum Zitat Samon JB, Castillo-Martin M, Hadler M, Ambesi-Impiobato A, Paietta E, Racevskis J, Wiernik PH, Rowe JM, Jakubczak J, Randolph S, et al. Preclinical analysis of the gamma-secretase inhibitor PF-03084014 in combination with glucocorticoids in T-cell acute lymphoblastic leukemia. Mol Cancer Ther. 2012;11(7):1565–75.PubMedPubMedCentralCrossRef Samon JB, Castillo-Martin M, Hadler M, Ambesi-Impiobato A, Paietta E, Racevskis J, Wiernik PH, Rowe JM, Jakubczak J, Randolph S, et al. Preclinical analysis of the gamma-secretase inhibitor PF-03084014 in combination with glucocorticoids in T-cell acute lymphoblastic leukemia. Mol Cancer Ther. 2012;11(7):1565–75.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Tolcher AW, Messersmith WA, Mikulski SM, Papadopoulos KP, Kwak EL, Gibbon DG, Patnaik A, Falchook GS, Dasari A, Shapiro GI, et al. Phase I study of RO4929097, a gamma secretase inhibitor of Notch signaling, in patients with refractory metastatic or locally advanced solid tumors. J Clin Oncol. 2012;30(19):2348–53.PubMedPubMedCentralCrossRef Tolcher AW, Messersmith WA, Mikulski SM, Papadopoulos KP, Kwak EL, Gibbon DG, Patnaik A, Falchook GS, Dasari A, Shapiro GI, et al. Phase I study of RO4929097, a gamma secretase inhibitor of Notch signaling, in patients with refractory metastatic or locally advanced solid tumors. J Clin Oncol. 2012;30(19):2348–53.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Zhou Y, Gregor VE, Sun Z, Ayida BK, Winters GC, Murphy D, Simonsen KB, Vourloumis D, Fish S, Froelich JM, et al. Structure-guided discovery of novel aminoglycoside mimetics as antibacterial translation inhibitors. Antimicrob Agents Chemother. 2005;49(12):4942–9.PubMedPubMedCentralCrossRef Zhou Y, Gregor VE, Sun Z, Ayida BK, Winters GC, Murphy D, Simonsen KB, Vourloumis D, Fish S, Froelich JM, et al. Structure-guided discovery of novel aminoglycoside mimetics as antibacterial translation inhibitors. Antimicrob Agents Chemother. 2005;49(12):4942–9.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Harvey JH, McFadden M, Andrews WG, Byrne PJ, Ahlgren JD, Woolley PV. Phase I study of echinomycin administered on an intermittent bolus schedule. Cancer Treat Rep. 1985;69(12):1365–8.PubMed Harvey JH, McFadden M, Andrews WG, Byrne PJ, Ahlgren JD, Woolley PV. Phase I study of echinomycin administered on an intermittent bolus schedule. Cancer Treat Rep. 1985;69(12):1365–8.PubMed
63.
Zurück zum Zitat Schott AF, Landis MD, Dontu G, Griffith KA, Layman RM, Krop I, Paskett LA, Wong H, Dobrolecki LE, Lewis MT, et al. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res. 2013;19(6):1512–24.PubMedPubMedCentralCrossRef Schott AF, Landis MD, Dontu G, Griffith KA, Layman RM, Krop I, Paskett LA, Wong H, Dobrolecki LE, Lewis MT, et al. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res. 2013;19(6):1512–24.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Wu CX, Xu A, Zhang CC, Olson P, Chen L, Lee TK, Cheung TT, Lo CM, Wang XQ. Notch inhibitor PF-03084014 inhibits hepatocellular carcinoma growth and metastasis via suppression of cancer stemness due to reduced activation of Notch1-Stat3. Mol Cancer Ther. 2017;16(8):1531–43.PubMedCrossRef Wu CX, Xu A, Zhang CC, Olson P, Chen L, Lee TK, Cheung TT, Lo CM, Wang XQ. Notch inhibitor PF-03084014 inhibits hepatocellular carcinoma growth and metastasis via suppression of cancer stemness due to reduced activation of Notch1-Stat3. Mol Cancer Ther. 2017;16(8):1531–43.PubMedCrossRef
65.
Zurück zum Zitat Yabuuchi S, Pai SG, Campbell NR, de Wilde RF, De Oliveira E, Korangath P, Streppel MM, Rasheed ZA, Hidalgo M, Maitra A, et al. Notch signaling pathway targeted therapy suppresses tumor progression and metastatic spread in pancreatic cancer. Cancer Lett. 2013;335(1):41–51.PubMedPubMedCentralCrossRef Yabuuchi S, Pai SG, Campbell NR, de Wilde RF, De Oliveira E, Korangath P, Streppel MM, Rasheed ZA, Hidalgo M, Maitra A, et al. Notch signaling pathway targeted therapy suppresses tumor progression and metastatic spread in pancreatic cancer. Cancer Lett. 2013;335(1):41–51.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Zhang CC, Yan Z, Zong Q, Fang DD, Painter C, Zhang Q, Chen E, Lira ME, John-Baptiste A, Christensen JG. Synergistic effect of the gamma-secretase inhibitor PF-03084014 and docetaxel in breast cancer models. Stem Cells Transl Med. 2013;2(3):233–42.PubMedPubMedCentralCrossRef Zhang CC, Yan Z, Zong Q, Fang DD, Painter C, Zhang Q, Chen E, Lira ME, John-Baptiste A, Christensen JG. Synergistic effect of the gamma-secretase inhibitor PF-03084014 and docetaxel in breast cancer models. Stem Cells Transl Med. 2013;2(3):233–42.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Chanh H, Laura P, SM F, Ratna M, Adele H, Silvia M, Shulian S, Anna P, Yongzhao S, Farbod D. The novel gamma secretase inhibitor RO4929097 reduces the tumor initiating potential of melanoma. Plos One. 2011;6(9):e25264.CrossRef Chanh H, Laura P, SM F, Ratna M, Adele H, Silvia M, Shulian S, Anna P, Yongzhao S, Farbod D. The novel gamma secretase inhibitor RO4929097 reduces the tumor initiating potential of melanoma. Plos One. 2011;6(9):e25264.CrossRef
68.
Zurück zum Zitat Capodanno Y, Buishand FO, Pang LY, Kirpensteijn J, Mol JA, Argyle DJ. Notch pathway inhibition targets chemoresistant insulinoma cancer stem cells. Endocr Relat Cancer. 2018;25(2):131–44.PubMedCrossRef Capodanno Y, Buishand FO, Pang LY, Kirpensteijn J, Mol JA, Argyle DJ. Notch pathway inhibition targets chemoresistant insulinoma cancer stem cells. Endocr Relat Cancer. 2018;25(2):131–44.PubMedCrossRef
69.
Zurück zum Zitat Zhao ZL, Zhang L, Huang CF, Ma SR, Bu LL, Liu JF, Yu GT, Liu B, Gutkind JS, Kulkarni AB, et al. NOTCH1 inhibition enhances the efficacy of conventional chemotherapeutic agents by targeting head neck cancer stem cell. Sci Rep. 2016;6:24704.PubMedPubMedCentralCrossRef Zhao ZL, Zhang L, Huang CF, Ma SR, Bu LL, Liu JF, Yu GT, Liu B, Gutkind JS, Kulkarni AB, et al. NOTCH1 inhibition enhances the efficacy of conventional chemotherapeutic agents by targeting head neck cancer stem cell. Sci Rep. 2016;6:24704.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Gal H, Amariglio N, Trakhtenbrot L, Jacob-Hirsh J, Margalit O, Avigdor A, Nagler A, Tavor S, Ein-Dor L, Lapidot T, et al. Gene expression profiles of AML derived stem cells; similarity to hematopoietic stem cells. Leukemia. 2006;20(12):2147–54.PubMedCrossRef Gal H, Amariglio N, Trakhtenbrot L, Jacob-Hirsh J, Margalit O, Avigdor A, Nagler A, Tavor S, Ein-Dor L, Lapidot T, et al. Gene expression profiles of AML derived stem cells; similarity to hematopoietic stem cells. Leukemia. 2006;20(12):2147–54.PubMedCrossRef
71.
Zurück zum Zitat Jiang LY, Zhang XL, Du P, Zheng JH. gamma-Secretase inhibitor, DAPT inhibits self-renewal and stemness maintenance of ovarian cancer stem-like cells in vitro. Chinese journal of cancer research = Chung-kuo yen cheng yen chiu. 2011;23(2):140–6.PubMedPubMedCentralCrossRef Jiang LY, Zhang XL, Du P, Zheng JH. gamma-Secretase inhibitor, DAPT inhibits self-renewal and stemness maintenance of ovarian cancer stem-like cells in vitro. Chinese journal of cancer research = Chung-kuo yen cheng yen chiu. 2011;23(2):140–6.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Ponnurangam S, Dandawate PR, Dhar A, Tawfik OW, Parab RR, Mishra PD, Ranadive P, Sharma R, Mahajan G, Umar S, et al. Quinomycin A targets Notch signaling pathway in pancreatic cancer stem cells. Oncotarget. 2016;7(3):3217–32.PubMedCrossRef Ponnurangam S, Dandawate PR, Dhar A, Tawfik OW, Parab RR, Mishra PD, Ranadive P, Sharma R, Mahajan G, Umar S, et al. Quinomycin A targets Notch signaling pathway in pancreatic cancer stem cells. Oncotarget. 2016;7(3):3217–32.PubMedCrossRef
73.
Zurück zum Zitat Norsworthy KJ, By K, Subramaniam S, Zhuang L, Del Valle PL, Przepiorka D, Shen YL, Sheth CM, Liu C, Leong R, et al. FDA approval summary: glasdegib for newly diagnosed acute myeloid leukemia. Clin Cancer Res. 2019;25(20):6021–5.PubMedCrossRef Norsworthy KJ, By K, Subramaniam S, Zhuang L, Del Valle PL, Przepiorka D, Shen YL, Sheth CM, Liu C, Leong R, et al. FDA approval summary: glasdegib for newly diagnosed acute myeloid leukemia. Clin Cancer Res. 2019;25(20):6021–5.PubMedCrossRef
74.
Zurück zum Zitat Chen L, Silapunt S, Migden MR. Sonidegib for the treatment of advanced basal cell carcinoma: a comprehensive review of sonidegib and the BOLT trial with 12-month update. Future Oncol. 2016;12(18):2095–105.PubMedCrossRef Chen L, Silapunt S, Migden MR. Sonidegib for the treatment of advanced basal cell carcinoma: a comprehensive review of sonidegib and the BOLT trial with 12-month update. Future Oncol. 2016;12(18):2095–105.PubMedCrossRef
75.
Zurück zum Zitat Minami H, Ando Y, Ma BB, Hsiang Lee J, Momota H, Fujiwara Y, Li L, Fukino K, Ito K, Tajima T, et al. Phase I, multicenter, open-label, dose-escalation study of sonidegib in Asian patients with advanced solid tumors. Cancer Sci. 2016;107(10):1477–83.PubMedPubMedCentralCrossRef Minami H, Ando Y, Ma BB, Hsiang Lee J, Momota H, Fujiwara Y, Li L, Fukino K, Ito K, Tajima T, et al. Phase I, multicenter, open-label, dose-escalation study of sonidegib in Asian patients with advanced solid tumors. Cancer Sci. 2016;107(10):1477–83.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Abou-Alfa GK, Lewis LD, LoRusso P, Maitland M, Chandra P, Cheeti S, Colburn D, Williams S, Simmons B, Graham RA. Pharmacokinetics and safety of vismodegib in patients with advanced solid malignancies and hepatic impairment. Cancer Chemother Pharmacol. 2017;80(1):29–36.PubMedPubMedCentralCrossRef Abou-Alfa GK, Lewis LD, LoRusso P, Maitland M, Chandra P, Cheeti S, Colburn D, Williams S, Simmons B, Graham RA. Pharmacokinetics and safety of vismodegib in patients with advanced solid malignancies and hepatic impairment. Cancer Chemother Pharmacol. 2017;80(1):29–36.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Belvisi MG, Bundschuh DS, Stoeck M, Wicks S, Underwood S, Battram CH, Haddad el B, Webber SE, Foster ML: Preclinical profile of ciclesonide, a novel corticosteroid for the treatment of asthma. J Pharmacol Exp Ther 2005, 314(2):568-574.PubMedCrossRef Belvisi MG, Bundschuh DS, Stoeck M, Wicks S, Underwood S, Battram CH, Haddad el B, Webber SE, Foster ML: Preclinical profile of ciclesonide, a novel corticosteroid for the treatment of asthma. J Pharmacol Exp Ther 2005, 314(2):568-574.PubMedCrossRef
78.
Zurück zum Zitat Everson JL, Sun MR, Fink DM, Heyne GW, Melberg CG, Nelson KF, Doroodchi P, Colopy LJ, Ulschmid CM, Martin AA, et al. Developmental toxicity assessment of piperonyl butoxide exposure targeting sonic hedgehog signaling and forebrain and face morphogenesis in the mouse: an in vitro and in vivo study. Environ Health Perspect. 2019;127(10):107006.PubMedPubMedCentralCrossRef Everson JL, Sun MR, Fink DM, Heyne GW, Melberg CG, Nelson KF, Doroodchi P, Colopy LJ, Ulschmid CM, Martin AA, et al. Developmental toxicity assessment of piperonyl butoxide exposure targeting sonic hedgehog signaling and forebrain and face morphogenesis in the mouse: an in vitro and in vivo study. Environ Health Perspect. 2019;127(10):107006.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Lauth M, Bergstrom A, Shimokawa T, Toftgard R. Inhibition of GLI-mediated transcription and tumor cell growth by small-molecule antagonists. Proc Natl Acad Sci U S A. 2007;104(20):8455–60.PubMedPubMedCentralCrossRef Lauth M, Bergstrom A, Shimokawa T, Toftgard R. Inhibition of GLI-mediated transcription and tumor cell growth by small-molecule antagonists. Proc Natl Acad Sci U S A. 2007;104(20):8455–60.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Amakye D, Jagani Z, Dorsch M. Unraveling the therapeutic potential of the Hedgehog pathway in cancer. Nat Med. 2013;19(11):1410–22.PubMedCrossRef Amakye D, Jagani Z, Dorsch M. Unraveling the therapeutic potential of the Hedgehog pathway in cancer. Nat Med. 2013;19(11):1410–22.PubMedCrossRef
83.
Zurück zum Zitat Huang FT, Zhuan-Sun YX, Zhuang YY, Wei SL, Tang J, Chen WB, Zhang SN. Inhibition of hedgehog signaling depresses self-renewal of pancreatic cancer stem cells and reverses chemoresistance. Int J Oncol. 2012;41(5):1707–14.PubMedCrossRef Huang FT, Zhuan-Sun YX, Zhuang YY, Wei SL, Tang J, Chen WB, Zhang SN. Inhibition of hedgehog signaling depresses self-renewal of pancreatic cancer stem cells and reverses chemoresistance. Int J Oncol. 2012;41(5):1707–14.PubMedCrossRef
84.
Zurück zum Zitat Liu S, Dontu G, Mantle ID, Patel S, Ahn NS, Jackson KW, Suri P, Wicha MS. Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res. 2006;66(12):6063–71.PubMedPubMedCentralCrossRef Liu S, Dontu G, Mantle ID, Patel S, Ahn NS, Jackson KW, Suri P, Wicha MS. Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res. 2006;66(12):6063–71.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Choi HS, Kim SL, Kim JH, Lee DS. The FDA-approved anti-asthma medicine ciclesonide inhibits lung cancer stem cells through Hedgehog signaling-mediated SOX2 regulation. Int J Mol Sci. 2020;21(3):1014.PubMedCentralCrossRef Choi HS, Kim SL, Kim JH, Lee DS. The FDA-approved anti-asthma medicine ciclesonide inhibits lung cancer stem cells through Hedgehog signaling-mediated SOX2 regulation. Int J Mol Sci. 2020;21(3):1014.PubMedCentralCrossRef
86.
Zurück zum Zitat Casey D, Demko S, Shord S, Zhao H, Chen H, He K, Putman A, Helms W, Keegan P, Pazdur R. FDA approval summary: sonidegib for locally advanced basal cell carcinoma. Clin Cancer Res. 2017;23(10):2377–81.PubMedCrossRef Casey D, Demko S, Shord S, Zhao H, Chen H, He K, Putman A, Helms W, Keegan P, Pazdur R. FDA approval summary: sonidegib for locally advanced basal cell carcinoma. Clin Cancer Res. 2017;23(10):2377–81.PubMedCrossRef
87.
Zurück zum Zitat Cazet AS, Hui MN, Elsworth BL, Wu SZ, Roden D, Chan CL, Skhinas JN, Collot R, Yang J, Harvey K, et al. Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer. Nat Commun. 2018;9(1):2897.PubMedCrossRef Cazet AS, Hui MN, Elsworth BL, Wu SZ, Roden D, Chan CL, Skhinas JN, Collot R, Yang J, Harvey K, et al. Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer. Nat Commun. 2018;9(1):2897.PubMedCrossRef
88.
Zurück zum Zitat Ingram I. Vismodegib granted FDA approval for treatment of basal cell carcinoma; 2012. Ingram I. Vismodegib granted FDA approval for treatment of basal cell carcinoma; 2012.
89.
Zurück zum Zitat Li W, Yang H, Li X, Han L, Xu N, Shi A. Signaling pathway inhibitors target breast cancer stem cells in triple-negative breast cancer. Oncol Rep. 2019;41(1):437–46.PubMed Li W, Yang H, Li X, Han L, Xu N, Shi A. Signaling pathway inhibitors target breast cancer stem cells in triple-negative breast cancer. Oncol Rep. 2019;41(1):437–46.PubMed
90.
Zurück zum Zitat Singh BN, Fu J, Srivastava RK, Shankar S. Hedgehog signaling antagonist GDC-0449 (Vismodegib) inhibits pancreatic cancer stem cell characteristics: molecular mechanisms. PLoS One. 2011;6(11):e27306.PubMedPubMedCentralCrossRef Singh BN, Fu J, Srivastava RK, Shankar S. Hedgehog signaling antagonist GDC-0449 (Vismodegib) inhibits pancreatic cancer stem cell characteristics: molecular mechanisms. PLoS One. 2011;6(11):e27306.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat De Jesus-Acosta A, Sugar EA, O'Dwyer PJ, Ramanathan RK, Von Hoff DD, Rasheed Z, Zheng L, Begum A, Anders R, Maitra A, et al. Phase 2 study of vismodegib, a hedgehog inhibitor, combined with gemcitabine and nab-paclitaxel in patients with untreated metastatic pancreatic adenocarcinoma. Br J Cancer. 2020;122(4):498–505.PubMedCrossRef De Jesus-Acosta A, Sugar EA, O'Dwyer PJ, Ramanathan RK, Von Hoff DD, Rasheed Z, Zheng L, Begum A, Anders R, Maitra A, et al. Phase 2 study of vismodegib, a hedgehog inhibitor, combined with gemcitabine and nab-paclitaxel in patients with untreated metastatic pancreatic adenocarcinoma. Br J Cancer. 2020;122(4):498–505.PubMedCrossRef
92.
Zurück zum Zitat Catenacci DV, Junttila MR, Karrison T, Bahary N, Horiba MN, Nattam SR, Marsh R, Wallace J, Kozloff M, Rajdev L, et al. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a Hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J Clin Oncol. 2015;33(36):4284–92.PubMedPubMedCentralCrossRef Catenacci DV, Junttila MR, Karrison T, Bahary N, Horiba MN, Nattam SR, Marsh R, Wallace J, Kozloff M, Rajdev L, et al. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a Hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J Clin Oncol. 2015;33(36):4284–92.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Wu C, Hu S, Cheng J, Wang G, Tao K. Smoothened antagonist GDC-0449 (Vismodegib) inhibits proliferation and triggers apoptosis in colon cancer cell lines. Exp Ther Med. 2017;13(5):2529–36.PubMedPubMedCentralCrossRef Wu C, Hu S, Cheng J, Wang G, Tao K. Smoothened antagonist GDC-0449 (Vismodegib) inhibits proliferation and triggers apoptosis in colon cancer cell lines. Exp Ther Med. 2017;13(5):2529–36.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Berlin J, Bendell JC, Hart LL, Firdaus I, Gore I, Hermann RC, Mulcahy MF, Zalupski MM, Mackey HM, Yauch RL, et al. A randomized phase II trial of vismodegib versus placebo with FOLFOX or FOLFIRI and bevacizumab in patients with previously untreated metastatic colorectal cancer. Clin Cancer Res. 2013;19(1):258–67.PubMedCrossRef Berlin J, Bendell JC, Hart LL, Firdaus I, Gore I, Hermann RC, Mulcahy MF, Zalupski MM, Mackey HM, Yauch RL, et al. A randomized phase II trial of vismodegib versus placebo with FOLFOX or FOLFIRI and bevacizumab in patients with previously untreated metastatic colorectal cancer. Clin Cancer Res. 2013;19(1):258–67.PubMedCrossRef
95.
Zurück zum Zitat Fukushima N, Minami Y, Kakiuchi S, Kuwatsuka Y, Hayakawa F, Jamieson C, Kiyoi H, Naoe T. Small-molecule Hedgehog inhibitor attenuates the leukemia-initiation potential of acute myeloid leukemia cells. Cancer Sci. 2016;107(10):1422–9.PubMedPubMedCentralCrossRef Fukushima N, Minami Y, Kakiuchi S, Kuwatsuka Y, Hayakawa F, Jamieson C, Kiyoi H, Naoe T. Small-molecule Hedgehog inhibitor attenuates the leukemia-initiation potential of acute myeloid leukemia cells. Cancer Sci. 2016;107(10):1422–9.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Chen JK, Taipale J, Young KE, Maiti T, Beachy PA. Small molecule modulation of Smoothened activity. Proc Natl Acad Sci U S A. 2002;99(22):14071–6.PubMedPubMedCentralCrossRef Chen JK, Taipale J, Young KE, Maiti T, Beachy PA. Small molecule modulation of Smoothened activity. Proc Natl Acad Sci U S A. 2002;99(22):14071–6.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Li C, Du Y, Yang Z, He L, Wang Y, Hao L, Ding M, Yan R, Wang J, Fan Z. GALNT1-mediated glycosylation and activation of sonic Hedgehog signaling maintains the self-renewal and tumor-initiating capacity of bladder cancer stem cells. Cancer Res. 2016;76(5):1273–83.PubMedCrossRef Li C, Du Y, Yang Z, He L, Wang Y, Hao L, Ding M, Yan R, Wang J, Fan Z. GALNT1-mediated glycosylation and activation of sonic Hedgehog signaling maintains the self-renewal and tumor-initiating capacity of bladder cancer stem cells. Cancer Res. 2016;76(5):1273–83.PubMedCrossRef
98.
Zurück zum Zitat Kurebayashi J, Koike Y, Ohta Y, Saitoh W, Yamashita T, Kanomata N, Moriya T. Anti-cancer stem cell activity of a hedgehog inhibitor GANT61 in estrogen receptor-positive breast cancer cells. Cancer Sci. 2017;108(5):918–30.PubMedPubMedCentralCrossRef Kurebayashi J, Koike Y, Ohta Y, Saitoh W, Yamashita T, Kanomata N, Moriya T. Anti-cancer stem cell activity of a hedgehog inhibitor GANT61 in estrogen receptor-positive breast cancer cells. Cancer Sci. 2017;108(5):918–30.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Michy T, Massias T, Bernard C, Vanwonterghem L, Henry M, Guidetti M, Royal G, Coll JL, Texier I, Josserand V, et al. Verteporfin-loaded lipid nanoparticles improve ovarian cancer photodynamic therapy in vitro and in vivo. Cancers. 2019;11(11):1760.PubMedCentralCrossRef Michy T, Massias T, Bernard C, Vanwonterghem L, Henry M, Guidetti M, Royal G, Coll JL, Texier I, Josserand V, et al. Verteporfin-loaded lipid nanoparticles improve ovarian cancer photodynamic therapy in vitro and in vivo. Cancers. 2019;11(11):1760.PubMedCentralCrossRef
100.
Zurück zum Zitat Su T, Yang X, Deng JH, Huang QJ, Huang SC, Zhang YM, Zheng HM, Wang Y, Lu LL, Liu ZQ. Evodiamine, a novel NOTCH3 methylation stimulator, significantly suppresses lung carcinogenesis in vitro and in vivo. Front Pharmacol. 2018;9:434.PubMedPubMedCentralCrossRef Su T, Yang X, Deng JH, Huang QJ, Huang SC, Zhang YM, Zheng HM, Wang Y, Lu LL, Liu ZQ. Evodiamine, a novel NOTCH3 methylation stimulator, significantly suppresses lung carcinogenesis in vitro and in vivo. Front Pharmacol. 2018;9:434.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Lopez-Aguilar E, Sepulveda-Vildosola AC, Rivera-Marquez H, Cerecedo-Diaz F, Valdez-Sanchez M, Villasis-Keever MA. Security and maximal tolerated doses of fluvastatin in pediatric cancer patients. Arch Med Res. 1999;30(2):128–31.PubMedCrossRef Lopez-Aguilar E, Sepulveda-Vildosola AC, Rivera-Marquez H, Cerecedo-Diaz F, Valdez-Sanchez M, Villasis-Keever MA. Security and maximal tolerated doses of fluvastatin in pediatric cancer patients. Arch Med Res. 1999;30(2):128–31.PubMedCrossRef
102.
Zurück zum Zitat Robison RL, Suter W, Cox RH. Carcinogenicity and mutagenicity studies with fluvastatin, a new, entirely synthetic HMG-CoA reductase inhibitor. Fundam Appl Toxicol. 1994;23(1):9–20.PubMedCrossRef Robison RL, Suter W, Cox RH. Carcinogenicity and mutagenicity studies with fluvastatin, a new, entirely synthetic HMG-CoA reductase inhibitor. Fundam Appl Toxicol. 1994;23(1):9–20.PubMedCrossRef
103.
Zurück zum Zitat Prado CM, Antoun S, Sawyer MB, Baracos VE. Two faces of drug therapy in cancer: drug-related lean tissue loss and its adverse consequences to survival and toxicity. Curr Opin Clin Nutr Metab Care. 2011;14(3):250–4.PubMedCrossRef Prado CM, Antoun S, Sawyer MB, Baracos VE. Two faces of drug therapy in cancer: drug-related lean tissue loss and its adverse consequences to survival and toxicity. Curr Opin Clin Nutr Metab Care. 2011;14(3):250–4.PubMedCrossRef
104.
Zurück zum Zitat Song S, Xie M, Scott AW, Jin J, Ma L, Dong X, Skinner HD, Johnson RL, Ding S, Ajani JA. A novel YAP1 inhibitor targets CSC-enriched radiation-resistant cells and exerts strong antitumor activity in esophageal adenocarcinoma. Mol Cancer Ther. 2018;17(2):443–54.PubMedCrossRef Song S, Xie M, Scott AW, Jin J, Ma L, Dong X, Skinner HD, Johnson RL, Ding S, Ajani JA. A novel YAP1 inhibitor targets CSC-enriched radiation-resistant cells and exerts strong antitumor activity in esophageal adenocarcinoma. Mol Cancer Ther. 2018;17(2):443–54.PubMedCrossRef
105.
Zurück zum Zitat Solmi M, Murru A, Pacchiarotti I, Undurraga J, Veronese N, Fornaro M, Stubbs B, Monaco F, Vieta E, Seeman MV, et al. Safety, tolerability, and risks associated with first- and second-generation antipsychotics: a state-of-the-art clinical review. Ther Clin Risk Manag. 2017;13:757–77.PubMedPubMedCentralCrossRef Solmi M, Murru A, Pacchiarotti I, Undurraga J, Veronese N, Fornaro M, Stubbs B, Monaco F, Vieta E, Seeman MV, et al. Safety, tolerability, and risks associated with first- and second-generation antipsychotics: a state-of-the-art clinical review. Ther Clin Risk Manag. 2017;13:757–77.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Zheng L, Xiang C, Li X, Guo Q, Gao L, Ni H, Xia Y, Xi T. STARD13-correlated ceRNA network-directed inhibition on YAP/TAZ activity suppresses stemness of breast cancer via co-regulating Hippo and Rho-GTPase/F-actin signaling. J Hematol Oncol. 2018;11(1):72.PubMedPubMedCentralCrossRef Zheng L, Xiang C, Li X, Guo Q, Gao L, Ni H, Xia Y, Xi T. STARD13-correlated ceRNA network-directed inhibition on YAP/TAZ activity suppresses stemness of breast cancer via co-regulating Hippo and Rho-GTPase/F-actin signaling. J Hematol Oncol. 2018;11(1):72.PubMedPubMedCentralCrossRef
107.
108.
Zurück zum Zitat Giraud J, Molina-Castro S, Seeneevassen L, Sifré E, Izotte J, Tiffon C, Staedel C, Boeuf H, Fernandez S, Barthelemy P, et al. Verteporfin targeting YAP1/TAZ-TEAD transcriptional activity inhibits the tumorigenic properties of gastric cancer stem cells. Int J Cancer. 2019;146(8):2255–67.PubMedCrossRef Giraud J, Molina-Castro S, Seeneevassen L, Sifré E, Izotte J, Tiffon C, Staedel C, Boeuf H, Fernandez S, Barthelemy P, et al. Verteporfin targeting YAP1/TAZ-TEAD transcriptional activity inhibits the tumorigenic properties of gastric cancer stem cells. Int J Cancer. 2019;146(8):2255–67.PubMedCrossRef
109.
Zurück zum Zitat Song S, Ajani JA, Honjo S, Maru DM, Chen Q, Scott AW, Heallen TR, Xiao L, Hofstetter WL, Weston B, et al. Hippo coactivator YAP1 upregulates SOX9 and endows esophageal cancer cells with stem-like properties. Cancer Res. 2014;74(15):4170–82.PubMedPubMedCentralCrossRef Song S, Ajani JA, Honjo S, Maru DM, Chen Q, Scott AW, Heallen TR, Xiao L, Hofstetter WL, Weston B, et al. Hippo coactivator YAP1 upregulates SOX9 and endows esophageal cancer cells with stem-like properties. Cancer Res. 2014;74(15):4170–82.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Zhao S, Xu K, Jiang R, Li DY, Guo XX, Zhou P, Tang JF, Li LS, Zeng D, Hu L, et al. Evodiamine inhibits proliferation and promotes apoptosis of hepatocellular carcinoma cells via the Hippo-yes-associated protein signaling pathway. Life Sci. 2020;117424. Zhao S, Xu K, Jiang R, Li DY, Guo XX, Zhou P, Tang JF, Li LS, Zeng D, Hu L, et al. Evodiamine inhibits proliferation and promotes apoptosis of hepatocellular carcinoma cells via the Hippo-yes-associated protein signaling pathway. Life Sci. 2020;117424.
111.
Zurück zum Zitat Kim H, Yu Y, Choi S, Lee H, Yu J, Lee JH, Kim WY. Evodiamine eliminates colon cancer stem cells via suppressing Notch and Wnt signaling. Molecules. 2019;24(24):4520.PubMedCentralCrossRef Kim H, Yu Y, Choi S, Lee H, Yu J, Lee JH, Kim WY. Evodiamine eliminates colon cancer stem cells via suppressing Notch and Wnt signaling. Molecules. 2019;24(24):4520.PubMedCentralCrossRef
112.
Zurück zum Zitat Qin J, Shi H, Xu Y, Zhao F, Wang Q. Tanshinone IIA inhibits cervix carcinoma stem cells migration and invasion via inhibiting YAP transcriptional activity. Biomed Pharmacother. 2018;105:758–65.PubMedCrossRef Qin J, Shi H, Xu Y, Zhao F, Wang Q. Tanshinone IIA inhibits cervix carcinoma stem cells migration and invasion via inhibiting YAP transcriptional activity. Biomed Pharmacother. 2018;105:758–65.PubMedCrossRef
113.
Zurück zum Zitat Zhao W, Wu M, Cui L, Du W. Limonin attenuates the stemness of cervical carcinoma cells by promoting YAP nuclear-cytoplasmic translocation. Food Chem Toxicol. 2019;125:621–8.PubMedCrossRef Zhao W, Wu M, Cui L, Du W. Limonin attenuates the stemness of cervical carcinoma cells by promoting YAP nuclear-cytoplasmic translocation. Food Chem Toxicol. 2019;125:621–8.PubMedCrossRef
114.
Zurück zum Zitat Tanaka K, Osada H, Murakami-Tonami Y, Horio Y, Hida T, Sekido Y. Statin suppresses Hippo pathway-inactivated malignant mesothelioma cells and blocks the YAP/CD44 growth stimulatory axis. Cancer Lett. 2017;385:215–24.PubMedCrossRef Tanaka K, Osada H, Murakami-Tonami Y, Horio Y, Hida T, Sekido Y. Statin suppresses Hippo pathway-inactivated malignant mesothelioma cells and blocks the YAP/CD44 growth stimulatory axis. Cancer Lett. 2017;385:215–24.PubMedCrossRef
115.
Zurück zum Zitat Koohestanimobarhan S, Salami S, Imeni V, Mohammadi Z, Bayat O. Lipophilic statins antagonistically alter the major epithelial-to-mesenchymal transition signaling pathways in breast cancer stem-like cells via inhibition of the mevalonate pathway. J Cell Biochem. 2018. https://doi.org/10.1002/jcb.27544.CrossRef Koohestanimobarhan S, Salami S, Imeni V, Mohammadi Z, Bayat O. Lipophilic statins antagonistically alter the major epithelial-to-mesenchymal transition signaling pathways in breast cancer stem-like cells via inhibition of the mevalonate pathway. J Cell Biochem. 2018. https://​doi.​org/​10.​1002/​jcb.​27544.CrossRef
116.
Zurück zum Zitat Yang CE, Lee WY, Cheng HW, Chung CH, Mi FL, Lin CW. The antipsychotic chlorpromazine suppresses YAP signaling, stemness properties, and drug resistance in breast cancer cells. Chem Biol Interact. 2019;302:28–35.PubMedCrossRef Yang CE, Lee WY, Cheng HW, Chung CH, Mi FL, Lin CW. The antipsychotic chlorpromazine suppresses YAP signaling, stemness properties, and drug resistance in breast cancer cells. Chem Biol Interact. 2019;302:28–35.PubMedCrossRef
117.
Zurück zum Zitat Al-Bari MA. Chloroquine analogues in drug discovery: new directions of uses, mechanisms of actions and toxic manifestations from malaria to multifarious diseases. J Antimicrob Chemother. 2015;70(6):1608–21.PubMedCrossRef Al-Bari MA. Chloroquine analogues in drug discovery: new directions of uses, mechanisms of actions and toxic manifestations from malaria to multifarious diseases. J Antimicrob Chemother. 2015;70(6):1608–21.PubMedCrossRef
118.
Zurück zum Zitat Shi TT, Yu XX, Yan LJ, Xiao HT. Research progress of hydroxychloroquine and autophagy inhibitors on cancer. Cancer Chemother Pharmacol. 2017;79(2):287–94.PubMedCrossRef Shi TT, Yu XX, Yan LJ, Xiao HT. Research progress of hydroxychloroquine and autophagy inhibitors on cancer. Cancer Chemother Pharmacol. 2017;79(2):287–94.PubMedCrossRef
119.
Zurück zum Zitat Brana I, Ocana A, Chen EX, Razak AR, Haines C, Lee C, Douglas S, Wang L, Siu LL, Tannock IF, et al. A phase I trial of pantoprazole in combination with doxorubicin in patients with advanced solid tumors: evaluation of pharmacokinetics of both drugs and tissue penetration of doxorubicin. Invest New Drugs. 2014;32(6):1269–77.PubMedCrossRef Brana I, Ocana A, Chen EX, Razak AR, Haines C, Lee C, Douglas S, Wang L, Siu LL, Tannock IF, et al. A phase I trial of pantoprazole in combination with doxorubicin in patients with advanced solid tumors: evaluation of pharmacokinetics of both drugs and tissue penetration of doxorubicin. Invest New Drugs. 2014;32(6):1269–77.PubMedCrossRef
120.
Zurück zum Zitat Dai S, Wang B, Li W, Wang L, Song X, Guo C, Li Y, Liu F, Zhu F, Wang Q, et al. Systemic application of 3-methyladenine markedly inhibited atherosclerotic lesion in ApoE(-/-) mice by modulating autophagy, foam cell formation and immune-negative molecules. Cell Death Dis. 2016;7(12):e2498.PubMedPubMedCentralCrossRef Dai S, Wang B, Li W, Wang L, Song X, Guo C, Li Y, Liu F, Zhu F, Wang Q, et al. Systemic application of 3-methyladenine markedly inhibited atherosclerotic lesion in ApoE(-/-) mice by modulating autophagy, foam cell formation and immune-negative molecules. Cell Death Dis. 2016;7(12):e2498.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Huang M, Tang SN, Upadhyay G, Marsh JL, Jackman CP, Srivastava RK, Shankar S. Rottlerin suppresses growth of human pancreatic tumors in nude mice, and pancreatic cancer cells isolated from Kras(G12D) mice. Cancer Lett. 2014;353(1):32–40.PubMedCrossRef Huang M, Tang SN, Upadhyay G, Marsh JL, Jackman CP, Srivastava RK, Shankar S. Rottlerin suppresses growth of human pancreatic tumors in nude mice, and pancreatic cancer cells isolated from Kras(G12D) mice. Cancer Lett. 2014;353(1):32–40.PubMedCrossRef
122.
Zurück zum Zitat Nazio F, Bordi M, Cianfanelli V, Locatelli F, Cecconi F. Autophagy and cancer stem cells: molecular mechanisms and therapeutic applications. Cell Death Differ. 2019;26(4):690–702.PubMedPubMedCentralCrossRef Nazio F, Bordi M, Cianfanelli V, Locatelli F, Cecconi F. Autophagy and cancer stem cells: molecular mechanisms and therapeutic applications. Cell Death Differ. 2019;26(4):690–702.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Auberger P, Puissant A. Autophagy, a key mechanism of oncogenesis and resistance in leukemia. Blood. 2017;129(5):547–52.PubMedCrossRef Auberger P, Puissant A. Autophagy, a key mechanism of oncogenesis and resistance in leukemia. Blood. 2017;129(5):547–52.PubMedCrossRef
124.
Zurück zum Zitat Liang DH, Choi DS, Ensor JE, Kaipparettu BA, Bass BL, Chang JC. The autophagy inhibitor chloroquine targets cancer stem cells in triple negative breast cancer by inducing mitochondrial damage and impairing DNA break repair. Cancer Lett. 2016;376(2):249–58.PubMedPubMedCentralCrossRef Liang DH, Choi DS, Ensor JE, Kaipparettu BA, Bass BL, Chang JC. The autophagy inhibitor chloroquine targets cancer stem cells in triple negative breast cancer by inducing mitochondrial damage and impairing DNA break repair. Cancer Lett. 2016;376(2):249–58.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Hao C, Liu G, Tian G. Autophagy inhibition of cancer stem cells promotes the efficacy of cisplatin against non-small cell lung carcinoma. Ther Adv Respir Dis. 2019;13:1753466619866097.PubMedPubMedCentralCrossRef Hao C, Liu G, Tian G. Autophagy inhibition of cancer stem cells promotes the efficacy of cisplatin against non-small cell lung carcinoma. Ther Adv Respir Dis. 2019;13:1753466619866097.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Rothe K, Porter V, Jiang X. Current outlook on autophagy in human leukemia: foe in cancer stem cells and drug resistance, friend in new therapeutic interventions. Int J Mol Sci. 2019;20(3):461.PubMedCentralCrossRef Rothe K, Porter V, Jiang X. Current outlook on autophagy in human leukemia: foe in cancer stem cells and drug resistance, friend in new therapeutic interventions. Int J Mol Sci. 2019;20(3):461.PubMedCentralCrossRef
127.
Zurück zum Zitat Horne GA, Stobo J, Kelly C, Mukhopadhyay A, Latif AL, Dixon-Hughes J, McMahon L, Cony-Makhoul P, Byrne J, Smith G, et al. A randomised phase II trial of hydroxychloroquine and imatinib versus imatinib alone for patients with chronic myeloid leukaemia in major cytogenetic response with residual disease. Leukemia. 2020. Horne GA, Stobo J, Kelly C, Mukhopadhyay A, Latif AL, Dixon-Hughes J, McMahon L, Cony-Makhoul P, Byrne J, Smith G, et al. A randomised phase II trial of hydroxychloroquine and imatinib versus imatinib alone for patients with chronic myeloid leukaemia in major cytogenetic response with residual disease. Leukemia. 2020.
128.
Zurück zum Zitat Hansen AR, Tannock IF, Templeton A, Chen E, Evans A, Knox J, Prawira A, Sridhar SS, Tan S, Vera-Badillo F, et al. Pantoprazole affecting docetaxel resistance pathways via autophagy (PANDORA): phase II trial of high dose pantoprazole (autophagy inhibitor) with docetaxel in metastatic castration-resistant prostate cancer (mCRPC). Oncologist. 2019;24(9):1188–94.PubMedPubMedCentralCrossRef Hansen AR, Tannock IF, Templeton A, Chen E, Evans A, Knox J, Prawira A, Sridhar SS, Tan S, Vera-Badillo F, et al. Pantoprazole affecting docetaxel resistance pathways via autophagy (PANDORA): phase II trial of high dose pantoprazole (autophagy inhibitor) with docetaxel in metastatic castration-resistant prostate cancer (mCRPC). Oncologist. 2019;24(9):1188–94.PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Feng S, Zheng Z, Feng L, Yang L, Chen Z, Lin Y, Gao Y, Chen Y. Proton pump inhibitor pantoprazole inhibits the proliferation, selfrenewal and chemoresistance of gastric cancer stem cells via the EMT/betacatenin pathways. Oncol Rep. 2016;36(6):3207–14.PubMedCrossRef Feng S, Zheng Z, Feng L, Yang L, Chen Z, Lin Y, Gao Y, Chen Y. Proton pump inhibitor pantoprazole inhibits the proliferation, selfrenewal and chemoresistance of gastric cancer stem cells via the EMT/betacatenin pathways. Oncol Rep. 2016;36(6):3207–14.PubMedCrossRef
130.
Zurück zum Zitat Yang H, Zheng Y, Zhang Y, Cao Z, Jiang Y. Mesenchymal stem cells derived from multiple myeloma patients protect against chemotherapy through autophagy-dependent activation of NF-kappaB signaling. Leuk Res. 2017;60:82–8.PubMedCrossRef Yang H, Zheng Y, Zhang Y, Cao Z, Jiang Y. Mesenchymal stem cells derived from multiple myeloma patients protect against chemotherapy through autophagy-dependent activation of NF-kappaB signaling. Leuk Res. 2017;60:82–8.PubMedCrossRef
131.
Zurück zum Zitat Kumar D, Shankar S, Srivastava RK. Rottlerin-induced autophagy leads to the apoptosis in breast cancer stem cells: molecular mechanisms. Mol Cancer. 2013;12(1):171.PubMedPubMedCentralCrossRef Kumar D, Shankar S, Srivastava RK. Rottlerin-induced autophagy leads to the apoptosis in breast cancer stem cells: molecular mechanisms. Mol Cancer. 2013;12(1):171.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat El Hout M, Dos Santos L, Hamai A, Mehrpour M. A promising new approach to cancer therapy: targeting iron metabolism in cancer stem cells. Semin Cancer Biol. 2018;53:125–38.PubMedCrossRef El Hout M, Dos Santos L, Hamai A, Mehrpour M. A promising new approach to cancer therapy: targeting iron metabolism in cancer stem cells. Semin Cancer Biol. 2018;53:125–38.PubMedCrossRef
134.
Zurück zum Zitat Shah R, Shchepinov MS, Pratt DA. Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS central science. 2018;4(3):387–96.PubMedPubMedCentralCrossRef Shah R, Shchepinov MS, Pratt DA. Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS central science. 2018;4(3):387–96.PubMedPubMedCentralCrossRef
135.
Zurück zum Zitat Conrad M, Kagan VE, Bayir H, Pagnussat GC, Head B, Traber MG, Stockwell BR. Regulation of lipid peroxidation and ferroptosis in diverse species. Genes Dev. 2018;32(9-10):602–19.PubMedPubMedCentralCrossRef Conrad M, Kagan VE, Bayir H, Pagnussat GC, Head B, Traber MG, Stockwell BR. Regulation of lipid peroxidation and ferroptosis in diverse species. Genes Dev. 2018;32(9-10):602–19.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Stoyanovsky DA, Tyurina YY, Shrivastava I, Bahar I, Tyurin VA, Protchenko O, Jadhav S, Bolevich SB, Kozlov AV, Vladimirov YA, et al. Iron catalysis of lipid peroxidation in ferroptosis: regulated enzymatic or random free radical reaction? Free Radic Biol Med. 2019;133:153–61.PubMedCrossRef Stoyanovsky DA, Tyurina YY, Shrivastava I, Bahar I, Tyurin VA, Protchenko O, Jadhav S, Bolevich SB, Kozlov AV, Vladimirov YA, et al. Iron catalysis of lipid peroxidation in ferroptosis: regulated enzymatic or random free radical reaction? Free Radic Biol Med. 2019;133:153–61.PubMedCrossRef
137.
Zurück zum Zitat Recalcati S, Gammella E, Cairo G. Dysregulation of iron metabolism in cancer stem cells. Free Radic Biol Med. 2019;133:216–20.PubMedCrossRef Recalcati S, Gammella E, Cairo G. Dysregulation of iron metabolism in cancer stem cells. Free Radic Biol Med. 2019;133:216–20.PubMedCrossRef
138.
Zurück zum Zitat Chanvorachote P, Luanpitpong S. Iron induces cancer stem cells and aggressive phenotypes in human lung cancer cells. Am J Physiol Cell Physiol. 2016;310(9):C728–39.PubMedCrossRef Chanvorachote P, Luanpitpong S. Iron induces cancer stem cells and aggressive phenotypes in human lung cancer cells. Am J Physiol Cell Physiol. 2016;310(9):C728–39.PubMedCrossRef
139.
Zurück zum Zitat Raggi C, Gammella E, Correnti M, Buratti P, Forti E, Andersen JB, Alpini G, Glaser S, Alvaro D, Invernizzi P, et al. Dysregulation of iron metabolism in cholangiocarcinoma stem-like cells. Sci Rep. 2017;7(1):17667.PubMedPubMedCentralCrossRef Raggi C, Gammella E, Correnti M, Buratti P, Forti E, Andersen JB, Alpini G, Glaser S, Alvaro D, Invernizzi P, et al. Dysregulation of iron metabolism in cholangiocarcinoma stem-like cells. Sci Rep. 2017;7(1):17667.PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat Schonberg DL, Miller TE, Wu Q, Flavahan WA, Das NK, Hale JS, Hubert CG, Mack SC, Jarrar AM. Karl RT et al: preferential iron trafficking characterizes glioblastoma stem-like cells. Cancer Cell. 2015;28(4):441–55.PubMedPubMedCentralCrossRef Schonberg DL, Miller TE, Wu Q, Flavahan WA, Das NK, Hale JS, Hubert CG, Mack SC, Jarrar AM. Karl RT et al: preferential iron trafficking characterizes glioblastoma stem-like cells. Cancer Cell. 2015;28(4):441–55.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Ishimoto T, Nagano O, Yae T, Tamada M, Motohara T, Oshima H, Oshima M, Ikeda T, Asaba R, Yagi H, et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth. Cancer Cell. 2011;19(3):387–400.PubMedCrossRef Ishimoto T, Nagano O, Yae T, Tamada M, Motohara T, Oshima H, Oshima M, Ikeda T, Asaba R, Yagi H, et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth. Cancer Cell. 2011;19(3):387–400.PubMedCrossRef
142.
Zurück zum Zitat Liang C, Zhang X, Yang M, Dong X. Recent progress in ferroptosis inducers for cancer therapy. Adv Mater. 2019;31(51):e1904197.PubMedCrossRef Liang C, Zhang X, Yang M, Dong X. Recent progress in ferroptosis inducers for cancer therapy. Adv Mater. 2019;31(51):e1904197.PubMedCrossRef
143.
Zurück zum Zitat Mai TT, Hamai A, Hienzsch A, Caneque T, Muller S, Wicinski J, Cabaud O, Leroy C, David A, Acevedo V, et al. Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat Chem. 2017;9(10):1025–33.PubMedPubMedCentralCrossRef Mai TT, Hamai A, Hienzsch A, Caneque T, Muller S, Wicinski J, Cabaud O, Leroy C, David A, Acevedo V, et al. Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat Chem. 2017;9(10):1025–33.PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Li L, Cui D, Ye L, Li Y, Zhu L, Yang L, Bai B, Nie Z, Gao J, Cao Y. Codelivery of salinomycin and docetaxel using poly(D,L-lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles to target both gastric cancer cells and cancer stem cells. Anticancer Drugs. 2017;28(9):989–1001.PubMedCrossRef Li L, Cui D, Ye L, Li Y, Zhu L, Yang L, Bai B, Nie Z, Gao J, Cao Y. Codelivery of salinomycin and docetaxel using poly(D,L-lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles to target both gastric cancer cells and cancer stem cells. Anticancer Drugs. 2017;28(9):989–1001.PubMedCrossRef
145.
Zurück zum Zitat Antoszczak M. A medicinal chemistry perspective on salinomycin as a potent anticancer and anti-CSCs agent. Eur J Med Chem. 2019;164:366–77.PubMedCrossRef Antoszczak M. A medicinal chemistry perspective on salinomycin as a potent anticancer and anti-CSCs agent. Eur J Med Chem. 2019;164:366–77.PubMedCrossRef
146.
Zurück zum Zitat Zhao Y, Zhao W, Lim YC, Liu T. Salinomycin-loaded gold nanoparticles for treating cancer stem cells by ferroptosis-induced cell death. Mol Pharm. 2019;16(6):2532–9.PubMedCrossRef Zhao Y, Zhao W, Lim YC, Liu T. Salinomycin-loaded gold nanoparticles for treating cancer stem cells by ferroptosis-induced cell death. Mol Pharm. 2019;16(6):2532–9.PubMedCrossRef
149.
Zurück zum Zitat Basuli D, Tesfay L, Deng Z, Paul B, Yamamoto Y, Ning G, Xian W, McKeon F, Lynch M, Crum CP, et al. Iron addiction: a novel therapeutic target in ovarian cancer. Oncogene. 2017;36(29):4089–99.PubMedPubMedCentralCrossRef Basuli D, Tesfay L, Deng Z, Paul B, Yamamoto Y, Ning G, Xian W, McKeon F, Lynch M, Crum CP, et al. Iron addiction: a novel therapeutic target in ovarian cancer. Oncogene. 2017;36(29):4089–99.PubMedPubMedCentralCrossRef
150.
Zurück zum Zitat Buccarelli M, Marconi M, Pacioni S, De Pascalis I, D'Alessandris QG, Martini M, Ascione B, Malorni W, Larocca LM, Pallini R, et al. Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis. Cell Death Dis. 2018;9(8):841.PubMedPubMedCentralCrossRef Buccarelli M, Marconi M, Pacioni S, De Pascalis I, D'Alessandris QG, Martini M, Ascione B, Malorni W, Larocca LM, Pallini R, et al. Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis. Cell Death Dis. 2018;9(8):841.PubMedPubMedCentralCrossRef
151.
Zurück zum Zitat Chen GQ, Benthani FA, Wu J, Liang D, Bian ZX, Jiang X. Artemisinin compounds sensitize cancer cells to ferroptosis by regulating iron homeostasis. Cell Death Differ. 2020;27(1):242–54.PubMedCrossRef Chen GQ, Benthani FA, Wu J, Liang D, Bian ZX, Jiang X. Artemisinin compounds sensitize cancer cells to ferroptosis by regulating iron homeostasis. Cell Death Differ. 2020;27(1):242–54.PubMedCrossRef
152.
Zurück zum Zitat Cao L, Duanmu W, Yin Y, Zhou Z, Ge H, Chen T, Tan L, Yu A, Hu R, Fei L, et al. Dihydroartemisinin exhibits anti-glioma stem cell activity through inhibiting p-AKT and activating caspase-3. Pharmazie. 2014;69(10):752–8.PubMed Cao L, Duanmu W, Yin Y, Zhou Z, Ge H, Chen T, Tan L, Yu A, Hu R, Fei L, et al. Dihydroartemisinin exhibits anti-glioma stem cell activity through inhibiting p-AKT and activating caspase-3. Pharmazie. 2014;69(10):752–8.PubMed
153.
Zurück zum Zitat Subedi A, Futamura Y, Nishi M, Ryo A, Watanabe N, Osada H. High-throughput screening identifies artesunate as selective inhibitor of cancer stemness: involvement of mitochondrial metabolism. Biochem Biophys Res Commun. 2016;477(4):737–42.PubMedCrossRef Subedi A, Futamura Y, Nishi M, Ryo A, Watanabe N, Osada H. High-throughput screening identifies artesunate as selective inhibitor of cancer stemness: involvement of mitochondrial metabolism. Biochem Biophys Res Commun. 2016;477(4):737–42.PubMedCrossRef
154.
Zurück zum Zitat Eling N, Reuter L, Hazin J, Hamacher-Brady A, Brady NR. Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience. 2015;2(5):517–32.PubMedPubMedCentralCrossRef Eling N, Reuter L, Hazin J, Hamacher-Brady A, Brady NR. Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience. 2015;2(5):517–32.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Su Y, Zhao B, Zhou L, Zhang Z, Shen Y, Lv H, AlQudsy LHH, Shang P. Ferroptosis, a novel pharmacological mechanism of anti-cancer drugs. Cancer Lett. 2020;483:127–36.PubMedCrossRef Su Y, Zhao B, Zhou L, Zhang Z, Shen Y, Lv H, AlQudsy LHH, Shang P. Ferroptosis, a novel pharmacological mechanism of anti-cancer drugs. Cancer Lett. 2020;483:127–36.PubMedCrossRef
156.
Zurück zum Zitat Sadhukha T, Niu L, Wiedmann TS, Panyam J. Effective elimination of cancer stem cells by magnetic hyperthermia. Mol Pharm. 2013;10(4):1432–41.PubMedCrossRef Sadhukha T, Niu L, Wiedmann TS, Panyam J. Effective elimination of cancer stem cells by magnetic hyperthermia. Mol Pharm. 2013;10(4):1432–41.PubMedCrossRef
157.
Zurück zum Zitat Bullivant JP, Zhao S, Willenberg BJ, Kozissnik B, Batich CD, Dobson J. Materials characterization of feraheme/ferumoxytol and preliminary evaluation of its potential for magnetic fluid hyperthermia. Int J Mol Sci. 2013;14(9):17501–10.PubMedPubMedCentralCrossRef Bullivant JP, Zhao S, Willenberg BJ, Kozissnik B, Batich CD, Dobson J. Materials characterization of feraheme/ferumoxytol and preliminary evaluation of its potential for magnetic fluid hyperthermia. Int J Mol Sci. 2013;14(9):17501–10.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Chen B, Xing J, Li M, Liu Y, Ji M. DOX@ferumoxytol-medical chitosan as magnetic hydrogel therapeutic system for effective magnetic hyperthermia and chemotherapy in vitro. Colloids Surf B Biointerfaces. 2020;190:110896.PubMedCrossRef Chen B, Xing J, Li M, Liu Y, Ji M. DOX@ferumoxytol-medical chitosan as magnetic hydrogel therapeutic system for effective magnetic hyperthermia and chemotherapy in vitro. Colloids Surf B Biointerfaces. 2020;190:110896.PubMedCrossRef
159.
Zurück zum Zitat Antoszczak M. A comprehensive review of salinomycin derivatives as potent anticancer and anti-CSCs agents. Eur J Med Chem. 2019;166:48–64.PubMedCrossRef Antoszczak M. A comprehensive review of salinomycin derivatives as potent anticancer and anti-CSCs agents. Eur J Med Chem. 2019;166:48–64.PubMedCrossRef
160.
161.
Zurück zum Zitat Azad GK, Tomar RS. Ebselen, a promising antioxidant drug: mechanisms of action and targets of biological pathways. Mol Biol Rep. 2014;41(8):4865–79.PubMedCrossRef Azad GK, Tomar RS. Ebselen, a promising antioxidant drug: mechanisms of action and targets of biological pathways. Mol Biol Rep. 2014;41(8):4865–79.PubMedCrossRef
162.
Zurück zum Zitat Zhang Z, Kodumuru V, Sviridov S, Liu S, Chafeev M, Chowdhury S, Chakka N, Sun J, Gauthier SJ, Mattice M, et al. Discovery of benzylisothioureas as potent divalent metal transporter 1 (DMT1) inhibitors. Bioorg Med Chem Lett. 2012;22(15):5108–13.PubMedCrossRef Zhang Z, Kodumuru V, Sviridov S, Liu S, Chafeev M, Chowdhury S, Chakka N, Sun J, Gauthier SJ, Mattice M, et al. Discovery of benzylisothioureas as potent divalent metal transporter 1 (DMT1) inhibitors. Bioorg Med Chem Lett. 2012;22(15):5108–13.PubMedCrossRef
163.
Zurück zum Zitat Stepanovic A, Nikitovic M. Severe hematologic temozolomide-related toxicity and lifethreatening infections. J BUON. 2018;23(1):7–13.PubMed Stepanovic A, Nikitovic M. Severe hematologic temozolomide-related toxicity and lifethreatening infections. J BUON. 2018;23(1):7–13.PubMed
164.
Zurück zum Zitat Luo Y, Che MJ, Liu C, Liu HG, Fu XW, Hou YP. Toxicity and related mechanisms of dihydroartemisinin on porcine oocyte maturation in vitro. Toxicol Appl Pharmacol. 2018;341:8–15.PubMedCrossRef Luo Y, Che MJ, Liu C, Liu HG, Fu XW, Hou YP. Toxicity and related mechanisms of dihydroartemisinin on porcine oocyte maturation in vitro. Toxicol Appl Pharmacol. 2018;341:8–15.PubMedCrossRef
165.
Zurück zum Zitat Li H, Xu K, Pian G, Sun S. Artesunate and sorafenib: combinatorial inhibition of liver cancer cell growth. Oncol Lett. 2019;18(5):4735–43.PubMedPubMedCentral Li H, Xu K, Pian G, Sun S. Artesunate and sorafenib: combinatorial inhibition of liver cancer cell growth. Oncol Lett. 2019;18(5):4735–43.PubMedPubMedCentral
166.
Zurück zum Zitat Slezakova S, Ruda J: Anticancer activity of artemisinin and its derivatives. Anticancer Res 2017, 37(11). Slezakova S, Ruda J: Anticancer activity of artemisinin and its derivatives. Anticancer Res 2017, 37(11).
167.
Zurück zum Zitat Kaittanis C, Shaffer TM, Ogirala A, Santra S, Perez JM, Chiosis G, Li Y, Josephson L, Grimm J. Environment-responsive nanophores for therapy and treatment monitoring via molecular MRI quenching. Nat Commun. 2014;5:3384.PubMedCrossRef Kaittanis C, Shaffer TM, Ogirala A, Santra S, Perez JM, Chiosis G, Li Y, Josephson L, Grimm J. Environment-responsive nanophores for therapy and treatment monitoring via molecular MRI quenching. Nat Commun. 2014;5:3384.PubMedCrossRef
168.
Zurück zum Zitat Nguyen KL, Yoshida T, Kathuria-Prakash N, Zaki IH, Varallyay CG, Semple SI, Saouaf R, Rigsby CK, Stoumpos S, Whitehead KK, et al. Multicenter safety and practice for off-label diagnostic use of ferumoxytol in MRI. Radiology. 2019;293(3):554–64.PubMedCrossRef Nguyen KL, Yoshida T, Kathuria-Prakash N, Zaki IH, Varallyay CG, Semple SI, Saouaf R, Rigsby CK, Stoumpos S, Whitehead KK, et al. Multicenter safety and practice for off-label diagnostic use of ferumoxytol in MRI. Radiology. 2019;293(3):554–64.PubMedCrossRef
169.
Zurück zum Zitat Shepshelovich D, Rozen-Zvi B, Avni T, Gafter U, Gafter-Gvili A. Intravenous versus oral iron supplementation for the treatment of anemia in CKD: an updated systematic review and meta-analysis. American journal of kidney diseases : the official journal of the National Kidney Foundation. 2016;68(5):677–90.CrossRef Shepshelovich D, Rozen-Zvi B, Avni T, Gafter U, Gafter-Gvili A. Intravenous versus oral iron supplementation for the treatment of anemia in CKD: an updated systematic review and meta-analysis. American journal of kidney diseases : the official journal of the National Kidney Foundation. 2016;68(5):677–90.CrossRef
170.
Zurück zum Zitat Miller RC, Petereit DG, Sloan JA, Liu H, Martenson JA, Bearden JD 3rd, Sapiente R, Seeger GR, Mowat RB, Liem B, et al. N08C9 (Alliance): a phase 3 randomized study of sulfasalazine versus placebo in the prevention of acute diarrhea in patients receiving pelvic radiation therapy. Int J Radiat Oncol Biol Phys. 2016;95(4):1168–74.PubMedPubMedCentralCrossRef Miller RC, Petereit DG, Sloan JA, Liu H, Martenson JA, Bearden JD 3rd, Sapiente R, Seeger GR, Mowat RB, Liem B, et al. N08C9 (Alliance): a phase 3 randomized study of sulfasalazine versus placebo in the prevention of acute diarrhea in patients receiving pelvic radiation therapy. Int J Radiat Oncol Biol Phys. 2016;95(4):1168–74.PubMedPubMedCentralCrossRef
171.
Zurück zum Zitat Hangauer MJ, Viswanathan VS, Ryan MJ, Bole D, Eaton JK, Matov A, Galeas J, Dhruv HD, Berens ME, Schreiber SL, et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature. 2017;551(7679):247–50.PubMedPubMedCentralCrossRef Hangauer MJ, Viswanathan VS, Ryan MJ, Bole D, Eaton JK, Matov A, Galeas J, Dhruv HD, Berens ME, Schreiber SL, et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature. 2017;551(7679):247–50.PubMedPubMedCentralCrossRef
172.
Zurück zum Zitat Viswanathan VS, Ryan MJ, Dhruv HD, Gill S, Eichhoff OM, Seashore-Ludlow B, Kaffenberger SD, Eaton JK, Shimada K, Aguirre AJ, et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature. 2017;547(7664):453–7.PubMedPubMedCentralCrossRef Viswanathan VS, Ryan MJ, Dhruv HD, Gill S, Eichhoff OM, Seashore-Ludlow B, Kaffenberger SD, Eaton JK, Shimada K, Aguirre AJ, et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature. 2017;547(7664):453–7.PubMedPubMedCentralCrossRef
173.
Zurück zum Zitat Jung Y, Park H, Zhao HY, Jeon R, Ryu JH, Kim WY. Systemic approaches identify a garlic-derived chemical, Z-ajoene, as a glioblastoma multiforme cancer stem cell-specific targeting agent. Mol Cells. 2014;37(7):547–53.PubMedPubMedCentralCrossRef Jung Y, Park H, Zhao HY, Jeon R, Ryu JH, Kim WY. Systemic approaches identify a garlic-derived chemical, Z-ajoene, as a glioblastoma multiforme cancer stem cell-specific targeting agent. Mol Cells. 2014;37(7):547–53.PubMedPubMedCentralCrossRef
174.
Zurück zum Zitat Kim TY, Han HS, Lee KW, Zang DY, Rha SY, Park YI, Kim JS, Lee KH, Park SH, Song EK, et al. A phase I/II study of poziotinib combined with paclitaxel and trastuzumab in patients with HER2-positive advanced gastric cancer. Gastric Cancer. 2019;22(6):1206–14.PubMedCrossRef Kim TY, Han HS, Lee KW, Zang DY, Rha SY, Park YI, Kim JS, Lee KH, Park SH, Song EK, et al. A phase I/II study of poziotinib combined with paclitaxel and trastuzumab in patients with HER2-positive advanced gastric cancer. Gastric Cancer. 2019;22(6):1206–14.PubMedCrossRef
175.
Zurück zum Zitat Zhu Y, Warin RF, Soroka DN, Chen H, Sang S. Metabolites of ginger component [6]-shogaol remain bioactive in cancer cells and have low toxicity in normal cells: chemical synthesis and biological evaluation. PLoS One. 2013;8(1):e54677.PubMedPubMedCentralCrossRef Zhu Y, Warin RF, Soroka DN, Chen H, Sang S. Metabolites of ginger component [6]-shogaol remain bioactive in cancer cells and have low toxicity in normal cells: chemical synthesis and biological evaluation. PLoS One. 2013;8(1):e54677.PubMedPubMedCentralCrossRef
176.
Zurück zum Zitat Lee H, Kim JW, Choi DK, Yu JH, Kim JH, Lee DS, Min SH. Poziotinib suppresses ovarian cancer stem cell growth via inhibition of HER4-mediated STAT5 pathway. Biochem Biophys Res Commun. 2020;526(1):158–64.PubMedCrossRef Lee H, Kim JW, Choi DK, Yu JH, Kim JH, Lee DS, Min SH. Poziotinib suppresses ovarian cancer stem cell growth via inhibition of HER4-mediated STAT5 pathway. Biochem Biophys Res Commun. 2020;526(1):158–64.PubMedCrossRef
177.
Zurück zum Zitat Ray A, Vasudevan S, Sengupta S. 6-Shogaol inhibits breast cancer cells and stem cell-like spheroids by modulation of Notch signaling pathway and induction of autophagic cell death. PLoS One. 2015;10(9):e0137614.PubMedPubMedCentralCrossRef Ray A, Vasudevan S, Sengupta S. 6-Shogaol inhibits breast cancer cells and stem cell-like spheroids by modulation of Notch signaling pathway and induction of autophagic cell death. PLoS One. 2015;10(9):e0137614.PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat Dreesen O, Brivanlou AH. Signaling pathways in cancer and embryonic stem cells. Stem Cell Rev. 2007;3(1):7–17.PubMedCrossRef Dreesen O, Brivanlou AH. Signaling pathways in cancer and embryonic stem cells. Stem Cell Rev. 2007;3(1):7–17.PubMedCrossRef
Metadaten
Titel
Emerging agents that target signaling pathways in cancer stem cells
verfasst von
Yue Yang
Xiaoman Li
Ting Wang
Qianqian Guo
Tao Xi
Lufeng Zheng
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2020
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-00901-6

Weitere Artikel der Ausgabe 1/2020

Journal of Hematology & Oncology 1/2020 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.