Open Access

Autophagy is a double‑edged sword in the therapy of colorectal cancer (Review)

  • Authors:
    • Bo Zhang
    • Lantao Liu
  • View Affiliations

  • Published online on: March 15, 2021     https://doi.org/10.3892/ol.2021.12639
  • Article Number: 378
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer is one of the leading causes of cancer‑associated mortality worldwide. The limitations of colorectal cancer treatment include various types of multidrug resistance and the contingent damage to neighboring normal cells caused by chemotherapy. Macroautophagy/autophagy and apoptosis are essential mechanisms involved in cancer cell regulation of chemotherapy. Autophagy can either cause cancer cell death or promote tumor survival during colorectal cancer. Given that autophagy is involved in chemotherapy of colorectal cancer, an improved insight into the potential interactions between apoptosis and autophagy is crucial. The present review aimed to summarize the involvement of autophagy in the regulation of colorectal cancer and its association with chemotherapy. Furthermore, the role of natural product extraction, novel chemicals and small molecules, as well as radiation, which induce autophagy in colorectal cancer cells, were reviewed. Finally, the present review aimed to provide an outlook for the regulation of autophagy as a novel approach to the treatment of cancer, particularly chemotherapy‑resistant colorectal cancer.

Introduction

Colorectal cancer is one of the most widespread malignancies in humans, which is the second most common cause of cancer-associated mortality worldwide (1). In 2020, ~147,950 people will be diagnosed with colorectal cancer and ~53,200 will succumb to the disease, including 17,930 cases and 3,640 mortalities among patients younger than 50 years (2). The major treatments for colorectal cancer include surgery, radiotherapy and chemotherapy. However, these treatments are associated with severe side effects and high relapse rates. In addition, chemoresistance is a major obstacle to overcome in patients with colon cancer (3). A recent study demonstrated that reductive stress may be a potential weapon against cancer, acting by priming tumor cells to apoptosis (4).

Autophagy is an evolutionarily conserved mechanism that maintains cellular homeostasis by degrading unnecessary or dysfunctional organelles and proteins (5). Several events, such as shortage of nutrients or energy availability, reactive oxygen species and hypoxia promote autophagy, which is accompanied by the recycling of cellular components under normal conditions in eukaryotic cells (6). In addition, autophagy is considered to serve as a cell survival mechanism implicated in drug resistance, and may be triggered by radiotherapy, chemotherapy and targeted therapy, or other factors, such as oxidative stress, causing cell injury (7). Highly autophagy-dependent cancer cells circumvent the loss of autophagy via upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) expression (8). Activation of endogenous antioxidants by Nrf2 can decrease oxidative stress, which promotes early lung tumor progression (9). In addition, Nrf2 activation promotes lung cancer metastasis by controlling oxidative homeostasis and targeted therapy, which mediates damage (10). A previous study reported that inhibition of autophagy can alleviate alcoholic fatty liver injury in alcoholic liver fibrosis model mice and Nrf2 signaling is involved in autophagy (11). Thus, novel therapeutic approaches targeting autophagy are being investigated to improve treatment options for patients with colorectal cancer.

Autophagy is a catabolic process that segregates specific intracellular cargo through engulfment into double-membrane vesicles in the cytosol, referred to as autophagosomes. Recognition of cargo chemical or extracellular stimuli induces autophagosome formation (12). The formation of autophagosomes can be activated by class III phosphoinositide 3-kinase and Beclin 1 (13). During autophagy, LC3 is transformed from its cytosolic form (LC3-I) to the LC3-II form, which is the autophagic membrane-bound form (14). The relative expression of LC3-I and II are considered to be an autophagy index (15).

Autophagy serves a controversial role in cancer, both in protecting against tumor development through isolation of injured organelles, and potentially by contributing to cancer progression (16). Autophagy may serve a pro-survival or pro-death role, depending on the type of cell and particular stimuli, the phase of progression and the intensity of Beclin 1 stimulation (17). In addition, autophagy serves an important role in colon cancer stem cell-related cancer progression (18). Use of the autophagy inhibitors, 3-methyladenine or chloroquine, has been demonstrated to increase sensitivity to chemotherapy in hepatocellular carcinoma (19). Thus, autophagy is a vital cellular mechanism underlying chemoresistance (20).

Apoptosis and autophagy are highly dynamic processes that regulate the final fate of cells (21). A previous study reported the association between autophagy and apoptosis, and related factors in colorectal cancer (22). The equilibrium between autophagy and apoptosis is shifted towards apoptosis via downregulation of JNK1 expression in HT29 colorectal cancer cells (23). Furthermore, there are several signaling pathways that are implicated in the regulation of autophagy, including the ERK (24), AMP-activated protein kinase (AMPK) (25), mTOR, silent information regulator-1, JNK and p38 signaling pathways (26).

Recently, autophagy has become one of the most extensively investigated fields in colorectal cancer research. This may be partially attributed to the development of autophagy that contributes to chemotherapy resistance. Overall, unique investigational therapies that apply natural and modified biological agents stimulated by derivatives from plants, small molecules and newly developed chemicals are an important source of potential anticancer treatments (Fig. 1). The present study aimed to discuss autophagy as a novel target for colorectal cancer therapy.

Autophagy genes

Beclin 1 was the first recognized mammalian autophagy gene, and it stimulates the nucleation of the autophagic vesicle (27). Beclin 1 is expressed at high levels in colorectal cancer tissues compared with normal tissues (28). Beclin 1 expression in vivo markedly suppresses the proliferation of colon cancer cells in xenograft models by inducing apoptosis, and overexpression may reverse aggressive phenotypes and suppress colon cancer tumor growth (29).

Autophagy-related gene (ATG)5 is a central autophagy protein that is involved in autophagosome formation. Furthermore, it is required for the ubiquitin-like conjugation systems implicated in LC3 lipidation (30). A previous study reported that ATG5 is permanently conjugated to ATG12 by E1-like ATG7 and E2-like ATG10 (31).

The phosphoinositide 3-kinase regulatory subunit 4 (PIK3R4; also named VPS15) is the regulatory subunit that regulates the production of phosphatidylinositol-3-phosphate (32). VPS15, PIK3R4 and Beclin 1 form three distinctive PI3K complexes (33). These central components, along with ATG14L, form the PI3KC3 complex 1, which is crucial for the stimulation of canonical autophagy (34).

Sequestosome 1 (p62) is a multifunctional receptor that is involved in autophagy-related signaling pathways (35,36). p62 is another important protein that targets other proteins for proteasome degradation and autophagic digestion (37). In particular, LC3-II binds to p62 to control protein packaging and to deliver it to the autophagosome (30).

A class III PI3K is required for autophagosomes, and is further associated with the conversion of LC3-I to its membrane-bound LC3-II form (38). LC3-BI is converted to LC3-BII through lipidation by a ubiquitin-like system involving ATG7 and ATG3 that allows LC3 to become associated with autophagic vesicles (39).

Autophagy-related 16 like 1 (ATG16L1) is an autophagy gene that is also involved in the immune response. Autophagy gene polymorphisms are associated with the progression of human colon cancer (18). A non-synonymous single-nucleotide polymorphism in ATG16L1 (Thr300Ala) is associated with the development of overall persistence in human colon cancer (40).

Autophagy pathway and its regulation

The autophagy pathway includes the following stages: i) Nucleation of the autophagic vesicle; ii) elongation and closure of the autophagosome membrane to enclose cytoplasmic constituents; iii) cropping of the autophagosome with lysosome and iv) degradation of the cytoplasmic substance inside the autophagosome (41). Several signaling molecules, including AMPK, mTOR, unc-51-like autophagy activating kinase 1 (ULK1), Beclin 1, Bcl-2, LC3, p62 and ATG contribute to the regulation of individual stages during this process (42).

Previous studies have reported that PI3K/Akt/mTOR inhibitors activate autophagy at the early stage as a survival mechanism that may affect its apoptotic potential (43,44). A novel Akt inhibitor, BI-69A11, induces autophagy at the early stages via inhibition of the Akt/mTOR/p70S6 kinase signaling pathway in colon cancer (45). Furthermore, it has been demonstrated that an increase in phosphorylated-AMPK and stimulation of autophagy in IL-10−/− mice at all stages, as suggested by the accumulation of LC3-II, increases Beclin 1 expression and decreases Bcl-2 expression (46). FOXO has been suggested to be an activator of autophagy via direct transactivation of autophagy genes or via regulation of autophagy activity (5). Endoplasmic reticulum (ER) stress is required for autophagy stimulation during oxaliplatin treatment (47). In addition, a previous study demonstrated that the BRAF oncogene induces key autophagic markers, such as LC3 and Beclin 1, in colorectal tumor cells (44). It has also been suggested that BI-69A11 mediates crosstalk among Akt, heat shock protein-90 and Beclin 1, maintaining the fine equilibrium between autophagy and apoptosis (45). Silencing of the colon cancer-associated transcript 2 gene induces apoptosis, as well as autophagy, in BGC-823 gastric cancer cells via inhibition of the PI3K/mTOR signaling pathways (48). Nuclear receptor-binding factor 2 is required for the removal of apoptotic cells and to relieve inflammation during colitis in mice, and modulates autophagy as a regulatory subunit of the ATG14 L-Beclin 1-PIK3C3 complex (49,50). Vitamin D receptor shortage increases the number of apoptotic cells and markedly increases cell death in the small intestine and colon of mice via Beclin 1 and ATG16L1 (51). In addition, treatment with vitamin D3 increases vitamin D receptor and ATG16L1 expression, which are activated by autophagic responses in organoids and colitis IL-10−/− mice (51).

Knockdown of Beclin 1 using small interfering RNA markedly inhibits the stimulation of autophagy caused by rapamycin, resulting in suppression of epithelial-to-mesenchymal transition (EMT), and a decrease in invasiveness of colon cancer cells, which suggests an association between EMT and activation of autophagy (52). Enhanced unfolded protein response in differentiated colon epithelial cells is accompanied by the induction of autophagy (53). Phosphoinositide-specific phospholipase C (PLC) γ1 suppression induces autophagy, and the protein tyrosine kinase 2/PLCγ1 axis is a potential downstream effector of the AMPK activation-dependent autophagy signaling cascade in HCT-116 cells (54). Cationic amphipathic KT2 induces nuclear condensation and apoptotic cell death, and this inhibits autophagy via the suppression of autophagy-related proteins in HCT-116 cells (55).

Recently, the role of microRNAs (miRNAs/miRs) as modulators in autophagy has improved our understanding of the role of autophagy in cancer (56). However, overexpression of miR-409-3p inhibits chemotherapy-induced autophagy in a Beclin 1-dependent manner (57). Overexpression of miR-30d suppresses cell viability, and this may be due to the inhibition of autophagy and promotion of apoptosis (58). Furthermore, miR-30d decreases cell autophagy by directly targeting the mRNA of ATG5 Beclin 1, which promotes the apoptosis of colon cancer cells (58). A recent study revealed that stimulation of autophagy decreases miR-183 expression in colorectal cancer cells (59). Furthermore, high miR-183 expression attenuates rapamycin- or starvation-induced autophagy in human cancer cells (59).

Overall, the autophagy pathway is both positively and negatively regulated by various environmental and immunological signals, as well as miRNAs.

Natural products induce autophagy in colon cancer

In addition to traditional surgery, chemotherapy and radiotherapy, western medicine and natural products offer potential methods for the treatment of colon cancer (60,61). Currently, natural medicine has become an area of clinical anticancer drug research due to its multi-link, multi-target and multi-channel antitumor effects (62). A previous study reported that autophagy is involved in resistance to radiotherapy or chemotherapy (63).

A dibenzylbutyrolactone type lignin, which was isolated from Combretum fruticosum, continually induces autophagic cell death with cytoplasmic vacuolization and development of autophagosomes regulated by increasing LC3 activation and altering Beclin 1 expression in HCT-116 cells (64). Luteolin treatment upregulates the expression levels of Beclin 1, ATG-5 and LC3B-I/II in human colon cancer SW620 cells (65).

Extracts obtained from different parts of the Grias neuberthii plant may affect autophagy in colon RKO (normal p53) and SW613-B3 (mutated p53) cell lines (66). As botanical drugs, ginkgolic acids, which are extracted from the seed coat of Ginkgo biloba L., induce intrinsic apoptosis rather than autophagy, which is regulated by reactive oxygen species generation, and contributes to human colon cancer cell death (67). Another study demonstrated that salidroside, which is a phenylpropanoid glycoside extracted from Rhodiola rosea, induces autophagy accompanied by cell apoptosis by suppressing the PI3K/Akt/mTOR signaling pathway in colorectal cancer cells (68). Pancratistatin, which is extracted from the spider lily or amaryllidaceae family of angiosperms, is associated with increased autophagy and apoptosis through G2/M cell cycle arrest in HCT-15 cells (69). Rhus coriaria extract induces Beclin 1-independent autophagy and caspase-7-dependent apoptosis in human colorectal cancer Caco-2 and HT-29 cells via inactivation of the AKT/mTOR signaling pathway (70). Berbamine is a plant-derived alkaloid that may trigger the progress of autophagic vesicles in HT-29 cells, along with an increase in the expression levels of LC3B-I, ATG-12, ATG-5 and Beclin 1 (71). Dentatin is an important coumarin derivative, mainly isolated from Murraya koenigii, which induces autophagy by inhibiting the JAK/STAT signaling pathway in colon cancer HT-29 cells (72).

Urolithin A is a major ellagitannin metabolite that has been demonstrated to induce autophagy and inhibit the metastatic potential of SW620 cells (73). Betulinic acid analogue is capable of inducing autophagy by altering the expression levels of several autophagic proteins, such as Beclin-1 and ATG-5, in HT-29 cells (74). 6-C-(E-phenylethenyl) naringenin, which is a small molecule found in naringenin fortified fried beef, can induce autophagy and necrosis in human colon cancer cells (75). Treatment with ophiopogon polysaccharide-B, which is a saponin compound extracted from Radix O. japonicus, induces autophagy by increasing Beclin 1 expression and the conversion of LC3I to LC3II, by upregulating the JNK/c-Jun signaling pathway (76). Physalin B, which can be isolated from Physalis divericata, triggers autophagosome formation and accumulates LC3-II and p62 in colon cancer cells (77).

Artesunate is a semi-synthetic derivative of artemisinin, which dose-dependently induces DNA damage and apoptosis in embryonal rhabdomyosarcoma cell lines (78). Artesunate induces autophagy by increasing LC3 and Beclin 1 expression, and the occurrence of autophagosomes in HCT-116 colon cancer cells (79). Pharmacological inhibition of autophagy stimulation using hydroxychloroquine markedly improves artesunate induced apoptosis (80). Brevilin A promotes cell autophagy and apoptosis via the mitochondrial signaling pathway and PI3K/AKT/mTOR inactivation in CT26 cells (81). Codonopis bulleynana Forest ex Diels induces cell apoptosis and inhibits autophagy via NF-κB signaling pathway activation in HCT-116 and SW480 colon cancer cells (82). In addition, evodiamine activates autophagy accompanied by apoptosis in SW480 cells by enhancing LC3 II and Beclin 1 expression (83). Vitexin induces apoptosis through suppression of autophagy in multidrug-resistant colorectal cancer cells (84). Litchi exocarp and endocarp activate a premature autophagic response, as well as cell death, via autophagy inhibitor or Beclin 1 silencing, suggesting that autophagy may be originally activated as a pro-survival response (85).

Recently, combining antineoplastic agents with autophagy blockers has been suggested as a therapeutic method for the treatment of patients with cancer (86). Pre-treatment with curcumin followed by 5-fluorouracil (5-FU) treatment promotes autophagy turnover both in vitro and in vivo through AMPK/ULK1-dependent autophagy suppression and AKT alteration, which provides an explanation for the increased susceptibility of colon cancer cells or cancer xenografts to the cytotoxicity of 5-FU (87). 5-FU-resistant SNUC5 colon cancer cells exhibit lower levels of autophagy compared with parental SNUC5 cells, indicating that reduced autophagy is associated with 5-FU resistance in colon cancer cells (88). Chloroquine in combination treatment with low concentrations of 5-FU can block autophagy in HCT-116 colon cancer cells (89).

Chemicals induce autophagy in colon cancer

17-hydroxywortmannin was identified as a drug that re-sensitizes tumor necrosis factor-related apoptosis-inducing ligand-resistant colon cancer cells, along with increased Beclin 1 expression accompanied by a deficiency of caspase-8 protein (90). Furthermore, angustifoline treatment of COLO-205 cells has been observed to markedly upregulate the protein expression levels of Beclin 1 and LC3-II, which leads to the generation of autophagic cell vesicles (91). Autophagic vacuoles are formed in glioblastoma stem cells following treatment with endothelial-monocyte-activating polypeptide-II combined with temozolomide (92).

The PI3K-Akt signaling pathway regulates autophagy and apoptosis via different mechanisms; however, mTORC1-mediated autophagy appears to not be involved in cell death initiation by 2,3-dihydro-2-(naphthalene-1-yl) quinazolin-4(1H)-one from the quinazolinone series (93). Incubation of HT-29 colon cancer cells with inositol-6 phosphate induces autophagy via inhibition of the Akt/mTOR signaling pathway (94). BH3 mimetic induces autophagy and disruption of BCL2-Beclin 1 binding in mouse embryonic fibroblasts and in human colon cancer cells, which are apoptosis-lacking cell types with a shortage of BAX and BAK1 (95). MHY218 is a hydroxamic acid derivative that can induce apoptosis and autophagy based on observing the accumulation of acidic vesicular organelles in HCT-116 cells (96). Zoledronic acid, which is a third-generation bisphosphonate molecule, regulates autophagy and promotes apoptosis in colon cancer CT26 cells (97). The recently improved polyamine analogue, N1, N11-diethylnorspermine, induces autophagy which is blocked by 3-methyladenine and Beclin 1 suppression; however, apoptosis is increased in HCT-116, SW480 and HT29 colon cancer cells (98). Upregulation of dimethyl fumarate, a dimethyl ester of fumaric acid (99), is associated with the expression of apoptotic markers in human colon cancer HT-29 and colorectal carcinoma T84 cells (99).

Melatonin treatment decreases the progression of colitis-associated colon carcinogenesis (CACC) by downregulating the process of autophagy, as demonstrated by the expression pattern of several autophagy markers combined with increased Nrf2 expression in the colon of mice with CACC (100). Autophagosome formation and autophagy are associated with the efficacy of cetuximab treatment in colon cancer CACO-2 cells (101). Purvalanol is a novel cyclin-dependent kinase inhibitor that induces ER stress-mediated apoptosis and triggers autophagy in HCT-116 cells (102).

Radiation and autophagy

Radiotherapy is a primary method for cancer treatment. It is particularly crucial to overcome radioresistance and to enhance radiosensitivity in patients with colon cancer. Chloroquine has gained consideration among anticancer treatments due to its potential use as an anticancer agent and as a chemotherapy sensitizer (103). Chloroquine has lysosomotropic effects via the suppression of the fusion of autophagosomes and lysosomes (104). A study revealed that chloroquine can sensitize HCT-116 cells to radiation and can improve the therapeutic outcome of radiation therapy in vivo (105). Furthermore, ATG7 knockdown or chloroquine treatment increases apoptotic cell death in HCT-116 cells (106).

Light emitting-diode irradiation induces markedly higher LC-3 and Beclin 1 expression levels and autophagosome formation in irradiated HT-29 or HCT-116 human colon cancer cells via photoreceptor Opsin 3 (107). Photodynamic therapy (PDT) is a simple method for invasive cancer treatment (108). Autophagy has been detected directly after the 5-ALA-mediated PDT process, with the strongest expression of autophagy-related proteins in human colon carcinoma SW620 cells (109). Photosensitive agents, such as protoporphyrin IX, induce double membrane autophagosomes (110). The radiosensitivity of colorectal cancer cells is associated with autophagy of tumor associated macrophages (TAMs), and promoting TAM autophagy may increase the radiosensitivity of colorectal cancer cells (111).

Conclusions and future direction

Autophagy can either stimulate tumor survival or induce cancer cell death in colorectal cancer. This ‘double-edged sword’ role of autophagy in colorectal cancer is dependent on the cancer stage and conditions of the microenvironment. Furthermore, autophagy is promoted in response to high-energy requests in the earlier phase of cell transformation. Autophagy is an adaptive tumor cell response in the later phases in human colorectal cancer cells. Combinatorial therapeutic methods may be of value in colorectal tumor therapy. Thus, an improved understanding of the molecular mechanisms underlying the interaction between autophagy and apoptosis are crucial for identifying the effects of combinatorial treatments on human colorectal cancer cells. In conclusion, autophagy is considered a novel therapeutic target for the treatment of chemoresistant colorectal cancer.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

BZ and LL confirmed the authenticity of all the raw data. BZ and LL performed the literature review. BZ drafted the initial manuscript, while LL revised the manuscript for important intellectual content. Both authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

AMPK

AMP-activated protein kinase

ATG

autophagy-related gene

ULK1

unc-51-like autophagy activating kinase 1

ER

endoplasmic reticulum

ATG16L1

autophagy related 16 like 1

EMT

epithelial-to-mesenchymal transition

PLC

phosphoinositide-specific phospholipase C

5-FU

5-fluorouracil

CACC

colon carcinogenesis

Nrf2

nuclear factor erythroid 2-related factor 2

PDT

photodynamic therapy

TAM

tumor-associated macrophage

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, Cercek A, Smith RA and Jemal A: Colorectal cancer statistics, 2020. CA Cancer J Clin. 70:145–164. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Saad ED and Buyse M: Statistical considerations for trials in adjuvant treatment of colorectal cancer. Cancers. 12:34422020. View Article : Google Scholar

4 

Bellezza I, Riuzzi F, Chiappalupi S, Arcuri C, Giambanco I, Sorci G and Donato R: Reductive stress in striated muscle cells. Cell Mol Life Sci. 77:3547–3565. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Cheng Z: The FoxO-Autophagy axis in health and disease. Trends Endocrinol Metab. 30:658–671. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Tschan MP and Simon HU: The role of autophagy in anticancer therapy: Promises and uncertainties. J Intern Med. 268:410–418. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Mathew R, Karantza-Wadsworth V and White E: Role of autophagy in cancer. Nat Rev Cancer. 7:961–967. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Towers CG, Fitzwalter BE, Regan D, Goodspeed A, Morgan MJ, Liu CW, Gustafson DL and Thorburn A: Cancer cells upregulate NRF2 signaling to adapt to autophagy inhibition. Dev Cell. 50:690–703.e6. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Wiel C, Le Gal K, Ibrahim MX, Jahangir CA, Kashif M, Yao H, Ziegler DV, Xu X, Ghosh T, Mondal T, et al: BACH1 stabilization by antioxidants stimulates lung cancer metastasis. Cell. 178:330–345.e322. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Lignitto L, LeBoeuf SE, Homer H, Jiang S, Askenazi M, Karakousi TR, Pass HI, Bhutkar AJ, Tsirigos A, Ueberheide B, et al: Nrf2 activation promotes lung cancer metastasis by inhibiting the degradation of Bach1. Cell. 178:316–329.e318. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Xie ZY, Xiao ZH and Wang FF: Inhibition of autophagy reverses alcohol-induced hepatic stellate cells activation through activation of Nrf2-Keap1-ARE signaling pathway. Biochimie. 147:55–62. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Gump JM and Thorburn A: Autophagy and apoptosis: What is the connection? Trends Cell Biol. 21:387–392. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Helgason GV, Holyoake TL and Ryan KM: Role of autophagy in cancer prevention, development and therapy. Essays Biochem. 55:133–151. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Rothenberg C, Srinivasan D, Mah L, Kaushik S, Peterhoff CM, Ugolino J, Fang S, Cuervo AM, Nixon RA and Monteiro MJ: Ubiquilin functions in autophagy and is degraded by chaperone-mediated autophagy. Hum Mol Genet. 19:3219–3232. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Burman C and Ktistakis NT: Autophagosome formation in mammalian cells. Semin Immunopathol. 32:397–413. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Aredia F, Guaman Ortiz LM, Giansanti V and Scovassi AI: Autophagy and cancer. Cells. 1:520–534. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Amaravadi R, Kimmelman AC and White E: Recent insights into the function of autophagy in cancer. Genes Dev. 30:1913–1930. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Roy BC, Ahmed I, Ramalingam S, Jala V, Haribabu B, Ramamoorthy P, Ashcraft J, Valentino J, Anant S, Sampath V, et al: Co-localization of autophagy-related protein p62 with cancer stem cell marker dclk1 may hamper dclk1's elimination during colon cancer development and progression. Oncotarget. 10:2340–2354. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Song J, Guo X, Xie X, Zhao X, Li D, Deng W, Song Y, Shen F, Wu M and Wei L: Autophagy in hypoxia protects cancer cells against apoptosis induced by nutrient deprivation through a Beclin1-dependent way in hepatocellular carcinoma. J Cell Biochem. 112:3406–3420. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Ho CJ and Gorski SM: Molecular mechanisms underlying autophagy-mediated treatment resistance in cancer. Cancers (Basel). 11:17752019. View Article : Google Scholar

21 

Chen Y, Zhang W, Guo X, Ren J and Gao A: The crosstalk between autophagy and apoptosis was mediated by phosphorylation of Bcl-2 and beclin1 in benzene-induced hematotoxicity. Cell Death Dis. 10:7722019. View Article : Google Scholar : PubMed/NCBI

22 

Xie Q, Liu Y and Li X: The interaction mechanism between autophagy and apoptosis in colon cancer. Transl Oncol. 13:1008712020. View Article : Google Scholar : PubMed/NCBI

23 

Vasilevskaya IA, Selvakumaran M, Roberts D and O'Dwyer PJ: JNK1 inhibition attenuates hypoxia-induced autophagy and sensitizes to chemotherapy. Mol Cancer Res. 14:753–763. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Martinez-Lopez N, Athonvarangkul D, Mishall P, Sahu S and Singh R: Autophagy proteins regulate ERK phosphorylation. Nat Commun. 4:27992013. View Article : Google Scholar : PubMed/NCBI

25 

Zadra G, Batista JL and Loda M: Dissecting the dual role of AMPK in cancer: From experimental to human studies. Mol Cancer Res. 13:1059–1072. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Sui X, Kong N, Ye L, Han W, Zhou J, Zhang Q, He C and Pan H: p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett. 344:174–179. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Chifenti B, Locci MT, Lazzeri G, Guagnozzi M, Dinucci D, Chiellini F, Filice ME, Salerno MG and Battini L: Autophagy-related protein LC3 and Beclin-1 in the first trimester of pregnancy. Clin Exp Reprod Med. 40:33–37. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Yang Z, Ghoorun RA, Fan X, Wu P, Bai Y, Li J, Chen H, Wang L and Wang J: High expression of Beclin-1 predicts favorable prognosis for patients with colorectal cancer. Clin Res Hepatol Gastroenterol. 39:98–106. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Zhang MY, Wang LY, Zhao S, Guo XC, Xu YQ, Zheng ZH, Lu H and Zheng HC: Effects of Beclin 1 overexpression on aggressive phenotypes of colon cancer cells. Oncol Lett. 17:2441–2450. 2019.PubMed/NCBI

30 

Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Overvatn A, Bjorkoy G and Johansen T: p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. The J Biol Chem. 282:24131–24145. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Mizushima N, Noda T, Yoshimori T, Tanaka Y, Ishii T, George MD, Klionsky DJ, Ohsumi M and Ohsumi Y: A protein conjugation system essential for autophagy. Nature. 395:395–398. 1998. View Article : Google Scholar : PubMed/NCBI

32 

Menon MB and Dhamija S: Beclin 1 phosphorylation-at the center of autophagy regulation. Front Cell Dev Biol. 6:1372018. View Article : Google Scholar : PubMed/NCBI

33 

Backer JM: The regulation and function of Class III PI3Ks: Novel roles for Vps34. Biochem J. 410:1–17. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Matsunaga K, Saitoh T, Tabata K, Omori H, Satoh T, Kurotori N, Maejima I, Shirahama-Noda K, Ichimura T, Isobe T, et al: Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally regulate autophagy at different stages. Nat Cell Biol. 11:385–396. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Wasik AM, Grabarek J, Pantovic A, Cieslar-Pobuda A, Asgari HR, Bundgaard-Nielsen C, Rafat M, Dixon IM, Ghavami S and Los MJ: Reprogramming and carcinogenesis-parallels and distinctions. Int Rev Cell Mol Biol. 308:167–203. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Ren F, Shu G, Liu G, Liu D and Zhou J, Yuan L and Zhou J: Knockdown of p62/sequestosome 1 attenuates autophagy and inhibits colorectal cancer cell growth. Mol Cell Biochem. 385:95–102. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Lilienbaum A: Relationship between the proteasomal system and autophagy. Int J Biochem Mol Biol. 4:1–26. 2013.PubMed/NCBI

38 

Pattingre S, Espert L, Biard-Piechaczyk M and Codogno P: Regulation of macroautophagy by mTOR and Beclin 1 complexes. Biochimie. 90:313–323. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Satoh J, Motohashi N, Kino Y, Ishida T, Yagishita S, Jinnai K, Arai N, Nakamagoe K, Tamaoka A, Saito Y, et al: LC3, an autophagosome marker, is expressed on oligodendrocytes in Nasu-Hakola disease brains. Orphanet J Rare Dis. 9:682014. View Article : Google Scholar : PubMed/NCBI

40 

Nicoli ER, Dumitrescu T, Uscatu CD, Popescu FD, Streaţă I, Serban Şoşoi S, Ivanov P, Dumitrescu A, Bărbălan A, Lungulescu D, et al: Determination of autophagy gene ATG16L1 polymorphism in human colorectal cancer. Rom J Morphol Embryol. 55:57–62. 2014.PubMed/NCBI

41 

D'Arcy MS: Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 43:582–592. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Wang CW and Klionsky DJ: The molecular mechanism of autophagy. Mol Med. 9:65–76. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Seitz C, Hugle M, Cristofanon S, Tchoghandjian A and Fulda S: The dual PI3K/mTOR inhibitor NVP-BEZ235 and chloroquine synergize to trigger apoptosis via mitochondrial-lysosomal cross-talk. Int J Cancer. 132:2682–2693. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Goulielmaki M, Koustas E, Moysidou E, Vlassi M, Sasazuki T, Shirasawa S, Zografos G, Oikonomou E and Pintzas A: BRAF associated autophagy exploitation: BRAF and autophagy inhibitors synergise to efficiently overcome resistance of BRAF mutant colorectal cancer cells. Oncotarget. 7:9188–9221. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Pal I, Parida S, Prashanth Kumar BN, Banik P, Kumar Dey K, Chakraborty S, Bhutia SK and Mandal M: Blockade of autophagy enhances proapoptotic potential of BI-69A11, a novel Akt inhibitor, in colon carcinoma. Eur J Pharmacol. 765:217–227. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Talero E, Alcaide A, Avila-Roman J, Garcia-Maurino S, Vendramini-Costa D and Motilva V: Expression patterns of sirtuin 1-AMPK-autophagy pathway in chronic colitis and inflammation-associated colon neoplasia in IL-10-deficient mice. Int Immunopharmacol. 35:248–256. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Li DD, Xie B, Wu XJ, Li JJ, Ding Y, Wen XZ, Zhang X, Zhu SG, Liu W, Zhang XS and Peng RQ: Late-stage inhibition of autophagy enhances calreticulin surface exposure. Oncotarget. 7:80842–80854. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Yu ZY, Wang Z, Lee KY, Yuan P and Ding J: Effect of silencing colon cancer-associated transcript 2 on the proliferation, apoptosis and autophagy of gastric cancer BGC-823 cells. Oncol Lett. 15:3127–3132. 2018.PubMed/NCBI

49 

Wu MY, Liu L, Wang EJ, Xiao HT, Cai CZ, Wang J, Su H, Wang Y, Tan J, Zhang Z, et al: PI3KC3 complex subunit NRBF2 is required for apoptotic cell clearance to restrict intestinal inflammation. Autophagy. 1–16. 2020. View Article : Google Scholar

50 

Young LN, Cho K, Lawrence R, Zoncu R and Hurley JH: Dynamics and architecture of the NRBF2-containing phosphatidylinositol 3-kinase complex I of autophagy. Proc Natl Acad Sci USA. 113:8224–8229. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Lu R, Zhang YG, Xia Y and Sun J: Imbalance of autophagy and apoptosis in intestinal epithelium lacking the vitamin D receptor. FASEB J. 33:11845–11856. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Shen H, Yin L, Deng G, Guo C, Han Y, Li Y, Cai C, Fu Y, Liu S and Zeng S: Knockdown of Beclin-1 impairs epithelial-mesenchymal transition of colon cancer cells. J Cell Biochem. 119:7022–7031. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Tuncer S, Sade-Memisoglu A, Keskus AG, Sheraj I, Guner G, Akyol A and Banerjee S: Enhanced expression of HNF4alpha during intestinal epithelial differentiation is involved in the activation of ER stress. FEBS J. 287:2504–2523. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Dai L, Chen X, Lu X, Wang F, Zhan Y, Song G, Hu T, Xia C and Zhang B: Phosphoinositide-specific phospholipase Cgamma1 inhibition induces autophagy in human colon cancer and hepatocellular carcinoma cells. Sci Rep. 7:139122017. View Article : Google Scholar : PubMed/NCBI

55 

Maraming P, Klaynongsruang S, Boonsiri P, Peng SF, Daduang S, Leelayuwat C, Pientong C, Chung JG and Daduang J: The cationic cell-penetrating KT2 peptide promotes cell membrane defects and apoptosis with autophagy inhibition in human HCT 116 colon cancer cells. J Cell Physiol. 234:22116–22129. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Frankel LB and Lund AH: MicroRNA regulation of autophagy. Carcinogenesis. 33:2018–2025. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Tan S, Shi H, Ba M, Lin S, Tang H, Zeng X and Zhang X: miR-409-3p sensitizes colon cancer cells to oxaliplatin by inhibiting Beclin-1-mediated autophagy. Int J Mol Med. 37:1030–1038. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Zhang R, Xu J, Zhao J and Bai J: Mir-30d suppresses cell proliferation of colon cancer cells by inhibiting cell autophagy and promoting cell apoptosis. Tumour Biol. 39:10104283177039842017. View Article : Google Scholar : PubMed/NCBI

59 

Huangfu L, Liang H, Wang G, Su X, Li L, Du Z, Hu M, Dong Y, Bai X, Liu T, et al: miR-183 regulates autophagy and apoptosis in colorectal cancer through targeting of UVRAG. Oncotarget. 7:4735–4745. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Sommerer C and Zeier M: Clinical manifestation and management of ADPKD in Western countries. Kidney Dis (Basel). 2:120–127. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Liu D and Liang XC: New developments in the pharmacodynamics and pharmacokinetics of combination of Chinese medicine and Western medicine. Chin J Integr Med. 23:312–319. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, et al: Naturally occurring anti-cancer compounds: Shining from Chinese herbal medicine. Chin Med. 14:482019. View Article : Google Scholar : PubMed/NCBI

63 

Xiong L, Liu Z, Ouyang G, Lin L, Huang H, Kang H, Chen W, Miao X and Wen Y: Autophagy inhibition enhances photocytotoxicity of Photosan-II in human colorectal cancer cells. Oncotarget. 8:6419–6432. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Moura AF, Lima KSB, Sousa TS, Marinho-Filho JDB, Pessoa C, Silveira ER, Pessoa ODL, Costa-Lotufo LV, Moraes MO and Araujo AJ: In vitro antitumor effect of a lignan isolated from Combretum fruticosum, trachelogenin, in HCT-116 human colon cancer cells. Toxicol In vitro. 47:129–136. 2018. View Article : Google Scholar : PubMed/NCBI

65 

Potocnjak I, Simic L, Gobin I, Vukelic I and Domitrovic R: Antitumor activity of luteolin in human colon cancer SW620 cells is mediated by the ERK/FOXO3a signaling pathway. Toxicol In Vitro. 66:1048522020. View Article : Google Scholar : PubMed/NCBI

66 

Guaman-Ortiz LM, Romero-Benavides JC, Suarez AI, Torres-Aguilar S, Castillo-Veintimilla P, Samaniego-Romero J, Ortiz-Diaz K and Bailon-Moscoso N: Cytotoxic property of grias neuberthii extract on human colon cancer cells: A crucial role of autophagy. Evid Based Complement Alternat Med. 2020:15653062020. View Article : Google Scholar : PubMed/NCBI

67 

Liu Y, Yang B, Zhang L, Cong X, Liu Z, Hu Y, Zhang J and Hu H: Ginkgolic acid induces interplay between apoptosis and autophagy regulated by ROS generation in colon cancer. Biochem Biophys Res Commun. 498:246–253. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Fan XJ, Wang Y, Wang L and Zhu M: Salidroside induces apoptosis and autophagy in human colorectal cancer cells through inhibition of PI3K/Akt/mTOR pathway. Oncol Rep. 36:3559–3567. 2016. View Article : Google Scholar : PubMed/NCBI

69 

Xiong Y, Xiong YJ, Liu DY and Shen RR: Pancratistatin inhibits the growth of colorectal cancer cells by inducing apoptosis, autophagy, and G2/M cell cycle arrest. Med Sci Monit. 25:6015–6022. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Athamneh K, Hasasna HE, Samri HA, Attoub S, Arafat K, Benhalilou N, Rashedi AA, Dhaheri YA, AbuQamar S, Eid A and Iratni R: Rhus coriaria increases protein ubiquitination, proteasomal degradation and triggers non-canonical Beclin-1-independent autophagy and apoptotic cell death in colon cancer cells. Sci Rep. 7:116332017. View Article : Google Scholar : PubMed/NCBI

71 

Mou L, Liang B, Liu G, Jiang J, Liu J, Zhou B, Huang J, Zang N, Liao Y, Ye L and Liang H: Berbamine exerts anticancer effects on human colon cancer cells via induction of autophagy and apoptosis, inhibition of cell migration and MEK/ERK signalling pathway. J BUON. 24:1870–1875. 2019.PubMed/NCBI

72 

Xiong W, Dong J and Kong S: Dentatin exerts anticancer effects on human colon cancer cell lines via cell cycle arrest, autophagy, inhibition of cell migration and JAK/STAT signalling pathway. J BUON. 24:1488–1493. 2019.PubMed/NCBI

73 

Zhao W, Shi F, Guo Z, Zhao J, Song X and Yang H: Metabolite of ellagitannins, urolithin A induces autophagy and inhibits metastasis in human sw620 colorectal cancer cells. Mol Carcinog. 57:193–200. 2018. View Article : Google Scholar : PubMed/NCBI

74 

Dutta D, Chakraborty B, Sarkar A, Chowdhury C and Das P: A potent betulinic acid analogue ascertains an antagonistic mechanism between autophagy and proteasomal degradation pathway in HT-29 cells. BMC Cancer. 16:232016. View Article : Google Scholar : PubMed/NCBI

75 

Zhao Y, Fan D, Ru B, Cheng KW, Hu S, Zhang J, Li ET and Wang M: 6-C-(E-phenylethenyl)naringenin induces cell growth inhibition and cytoprotective autophagy in colon cancer cells. Eur J Cancer. 68:38–50. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Chen M, Guo Y, Zhao R, Wang X, Jiang M, Fu H and Zhang X: Ophiopogonin B induces apoptosis, mitotic catastrophe and autophagy in A549 cells. Int J Oncol. 49:316–324. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Ma YM, Han W, Li J, Hu LH and Zhou YB: Physalin B not only inhibits the ubiquitin-proteasome pathway but also induces incomplete autophagic response in human colon cancer cells in vitro. Acta Pharmacol Sin. 36:517–527. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Beccafico S, Morozzi G, Marchetti MC, Riccardi C, Sidoni A, Donato R and Sorci G: Artesunate induces ROS- and p38 MAPK-mediated apoptosis and counteracts tumor growth in vivo in embryonal rhabdomyosarcoma cells. Carcinogenesis. 36:1071–1083. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Chen K, Shou LM, Lin F, Duan WM, Wu MY, Xie X, Xie YF, Li W and Tao M: Artesunate induces G2/M cell cycle arrest through autophagy induction in breast cancer cells. Anticancer Drugs. 25:652–662. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Jiang F, Zhou JY, Zhang D, Liu MH and Chen YG: Artesunate induces apoptosis and autophagy in HCT116 colon cancer cells, and autophagy inhibition enhances the artesunateinduced apoptosis. Int J Mol Med. 42:1295–1304. 2018.PubMed/NCBI

81 

You P, Wu H, Deng M, Peng J, Li F and Yang Y: Brevilin A induces apoptosis and autophagy of colon adenocarcinoma cell CT26 via mitochondrial pathway and PI3K/AKT/mTOR inactivation. Biomed Pharmacother. 98:619–625. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Luan Y, Li Y, Zhu L, Zheng S, Mao D, Chen Z and Cao Y: Codonopis bulleynana Forest ex Diels inhibits autophagy and induces apoptosis of colon cancer cells by activating the NF-κB signaling pathway. Int J Mol Med. 41:1305–1314. 2018.PubMed/NCBI

83 

Wang D, Ge S, Chen Z and Song Y: Evodiamine exerts anticancer effects via induction of apoptosis and autophagy and suppresses the migration and invasion of human colon cancer cells. J BUON. 24:1824–1829. 2019.PubMed/NCBI

84 

Bhardwaj M, Cho HJ, Paul S, Jakhar R, Khan I, Lee SJ, Kim BY, Krishnan M, Khaket TP, Lee HG and Kang SC: Vitexin induces apoptosis by suppressing autophagy in multi-drug resistant colorectal cancer cells. Oncotarget. 9:3278–3291. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Emanuele S, Notaro A, Palumbo Piccionello A, Maggio A, Lauricella M, D'Anneo A, Cernigliaro C, Calvaruso G and Giuliano M: Sicilian Litchi fruit extracts induce autophagy versus apoptosis switch in human colon cancer cells. Nutrients. 10:14902018. View Article : Google Scholar

86 

Levy JMM, Towers CG and Thorburn A: Targeting autophagy in cancer. Nature reviews Cancer. 17:528–542. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Zhang P, Lai ZL, Chen HF, Zhang M, Wang A, Jia T, Sun WQ, Zhu XM, Chen XF, Zhao Z and Zhang J: Curcumin synergizes with 5-fluorouracil by impairing AMPK/ULK1-dependent autophagy, AKT activity and enhancing apoptosis in colon cancer cells with tumor growth inhibition in xenograft mice. J Exp Clin Cancer Res. 36:1902017. View Article : Google Scholar : PubMed/NCBI

88 

Yao CW, Kang KA, Piao MJ, Ryu YS, Fernando PMDJ, Oh MC, Park JE, Shilnikova K, Na SY, Jeong SU, et al: Reduced autophagy in 5-fluorouracil resistant colon cancer cells. Biomol Ther (Seoul). 25:315–320. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Zamame Ramirez JA, Romagnoli GG, Falasco BF, Gorgulho CM, Sanzochi Fogolin C, Dos Santos DC, Junior JPA, Lotze MT, Ureshino RP and Kaneno R: Blocking drug-induced autophagy with chloroquine in HCT-116 colon cancer cells enhances DC maturation and T cell responses induced by tumor cell lysate. Int Immunopharmacol. 84:1064952020. View Article : Google Scholar : PubMed/NCBI

90 

Dai S, Yang S, Hu X, Sun W, Tawa G, Zhu W, Schimmer AD, He C, Fang B, Zhu H and Zheng W: 17-Hydroxy wortmannin restores TRAIL's response by ameliorating increased Beclin 1 level and autophagy function in TRAIL-resistant colon cancer cells. Mol Cancer Ther. 18:1265–1277. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Ding Z, Chen Q, Xiong B, Cun Y, Wang H and Xu M: Angustifoline inhibits human colon cancer cell growth by inducing autophagy along with mitochondrial-mediated apoptosis, suppression of cell invasion and migration and stimulating G2/M cell cycle arrest. J BUON. 24:130–135. 2019.PubMed/NCBI

92 

Zhou W, Liu L, Xue Y, Zheng J, Liu X, Ma J, Li Z and Liu Y: Combination of endothelial-monocyte-activating polypeptide-II with temozolomide suppress malignant biological behaviors of human glioblastoma stem cells via miR-590-3p/MACC1 inhibiting PI3K/AKT/mTOR signal pathway. Front Mol Neurosci. 10:682017. View Article : Google Scholar : PubMed/NCBI

93 

Hussain A, Qazi AK, Mupparapu N, Kumar A, Mintoo MJ, Mahajan G, Sharma PR, Singh SK, Bharate SB, Zargar MA, et al: A novel PI3K axis selective molecule exhibits potent tumor inhibition in colorectal carcinogenesis. Mol Carcinog. 55:2135–2155. 2016. View Article : Google Scholar : PubMed/NCBI

94 

Pandurangan AK, Ismail S, Esa NM and Munusamy MA: Inositol-6 phosphate inhibits the mTOR pathway and induces autophagy-mediated death in HT-29 colon cancer cells. Arch Med Sci. 14:1281–1288. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Pedro JM, Wei Y, Sica V, Maiuri MC, Zou Z, Kroemer G and Levine B: BAX and BAK1 are dispensable for ABT-737-induced dissociation of the BCL2-BECN1 complex and autophagy. Autophagy. 11:452–459. 2015. View Article : Google Scholar : PubMed/NCBI

96 

Choi PR, Kang YJ, Sung B, Kim JH, Moon HR, Chung HY, Kim SE, Park MI, Park SJ and Kim ND: MHY218-induced apoptotic cell death is enhanced by the inhibition of autophagy in AGS human gastric cancer cells. Int J Oncol. 47:563–572. 2015. View Article : Google Scholar : PubMed/NCBI

97 

Zhu J, Liu M, Liu Y, Zhang Y, Yang B and Zhang W: Zoledronic acid regulates autophagy and induces apoptosis in colon cancer cell line CT26. Biomed Res Int. 2017:72035842017. View Article : Google Scholar : PubMed/NCBI

98 

Gurkan AC, Arisan ED, Yerlikaya PO, Ilhan H and Unsal NP: Inhibition of autophagy enhances DENSpm-induced apoptosis in human colon cancer cells in a p53 independent manner. Cell Oncol (Dordr). 41:297–317. 2018. View Article : Google Scholar : PubMed/NCBI

99 

Kaluzki I, Hailemariam-Jahn T, Doll M, Kaufmann R, Balermpas P, Zoller N, Kippenberger S and Meissner M: Dimethylfumarate inhibits colorectal carcinoma cell proliferation: Evidence for cell cycle arrest, apoptosis and autophagy. Cells. 8:13292019. View Article : Google Scholar

100 

Trivedi PP, Jena GB, Tikoo KB and Kumar V: Melatonin modulated autophagy and Nrf2 signaling pathways in mice with colitis-associated colon carcinogenesis. Mol Carcinog. 55:255–267. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Guo GF, Wang YX, Zhang YJ, Chen XX, Lu JB, Wang HH, Jiang C, Qiu HQ and Xia LP: Predictive and prognostic implications of 4E-BP1, Beclin-1, and LC3 for cetuximab treatment combined with chemotherapy in advanced colorectal cancer with wild-type KRAS: Analysis from real-world data. World J Gastroenterol. 25:1840–1853. 2019. View Article : Google Scholar : PubMed/NCBI

102 

Coker-Gürkan A, Arisan ED, Obakan P, Akalin K, Özbey U and Palavan-Unsal N: Purvalanol induces endoplasmic reticulum stress-mediated apoptosis and autophagy in a time-dependent manner in HCT116 colon cancer cells. Oncol Rep. 33:2761–2770. 2015. View Article : Google Scholar : PubMed/NCBI

103 

Shiratori H, Kawai K, Hata K, Tanaka T, Nishikawa T, Otani K, Sasaki K, Kaneko M, Murono K, Emoto S, et al: The combination of temsirolimus and chloroquine increases radiosensitivity in colorectal cancer cells. Oncol Rep. 42:377–385. 2019.PubMed/NCBI

104 

Solomon VR and Lee H: Chloroquine and its analogs: A new promise of an old drug for effective and safe cancer therapies. Eur J Pharmacol. 625:220–233. 2009. View Article : Google Scholar : PubMed/NCBI

105 

Rouschop KM, van den Beucken T, Dubois L, Niessen H, Bussink J, Savelkouls K, Keulers T, Mujcic H, Landuyt W, Voncken JW, et al: The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5. J Clin Invest. 120:127–141. 2010. View Article : Google Scholar : PubMed/NCBI

106 

Ouyang G, Xiong L, Liu Z, Lam B, Bui B, Ma L, Chen X, Zhou P, Wang K, Zhang Z, et al: Inhibition of autophagy potentiates the apoptosis-inducing effects of photodynamic therapy on human colon cancer cells. Photodiagnosis Photodyn Ther. 21:396–403. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Yoshimoto T, Morine Y, Takasu C, Feng R, Ikemoto T, Yoshikawa K, Iwahashi S, Saito Y, Kashihara H, Akutagawa M, et al: Blue light-emitting diodes induce autophagy in colon cancer cells by Opsin 3. Ann Gastroenterol Surg. 2:154–161. 2018. View Article : Google Scholar : PubMed/NCBI

108 

Wu H, Minamide T and Yano T: Role of photodynamic therapy in the treatment of esophageal cancer. Dig Endosc. 31:508–516. 2019. View Article : Google Scholar : PubMed/NCBI

109 

Ziółkowska B, Woźniak M and Ziółkowski P: Co-expression of autophagic markers following photodynamic therapy in SW620 human colon adenocarcinoma cells. Mol Med Rep. 14:2548–2554. 2016. View Article : Google Scholar : PubMed/NCBI

110 

Kou J, Dou D and Yang L: Porphyrin photosensitizers in photodynamic therapy and its applications. Oncotarget. 8:81591–81603. 2017. View Article : Google Scholar : PubMed/NCBI

111 

Shao LN, Zhu BS, Xing CG, Yang XD, Young W and Cao JP: Effects of autophagy regulation of tumor-associated macrophages on radiosensitivity of colorectal cancer cells. Mol Med Rep. 13:2661–2670. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 21 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang B and Zhang B: Autophagy is a double‑edged sword in the therapy of colorectal cancer (Review). Oncol Lett 21: 378, 2021
APA
Zhang, B., & Zhang, B. (2021). Autophagy is a double‑edged sword in the therapy of colorectal cancer (Review). Oncology Letters, 21, 378. https://doi.org/10.3892/ol.2021.12639
MLA
Zhang, B., Liu, L."Autophagy is a double‑edged sword in the therapy of colorectal cancer (Review)". Oncology Letters 21.5 (2021): 378.
Chicago
Zhang, B., Liu, L."Autophagy is a double‑edged sword in the therapy of colorectal cancer (Review)". Oncology Letters 21, no. 5 (2021): 378. https://doi.org/10.3892/ol.2021.12639