2020 Autophagy in Cancer Recent Advances and Future Directions
2020 Autophagy in Cancer Recent Advances and Future Directions
A R T I C LE I N FO A B S T R A C T
Keywords: Autophagy is being explored as a potential therapeutic target for enhancing the cytotoxic effects of che-
Autophagy motherapeutic regimens in various malignancies. Autophagy plays a very important role in cancer pathogenesis.
Cancer Here, we discuss the updates on the modulation of autophagy via dynamic interactions with different organelles
Therapy and the exploitation of selective autophagy for exploring therapeutic strategies. We further discuss the role of
Tumor immunity
autophagy inhibitors in cancer preclinical and clinical trials, novel autophagy inhibitors, and challenges likely to
Resistance
be faced by clinicians while inducting autophagy modulators in clinical practice.
⁎
Corresponding author at: Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
E-mail addresses: [email protected] (M. Ishaq), [email protected] (R. Ojha), [email protected] (A.P. Sharma),
[email protected] (S.K. Singh).
https://doi.org/10.1016/j.semcancer.2020.03.010
Received 15 February 2019; Received in revised form 10 February 2020; Accepted 16 March 2020
Available online 19 March 2020
1044-579X/ © 2020 Elsevier Ltd. All rights reserved.
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Ishaq, et al. Seminars in Cancer Biology 66 (2020) 171–181
autophagosomes towards the lysosome enriched regions of the cell machineries, and all of them can be degraded within the individual
(peri-nuclear regions) where SNARES (Soluble NSF Attachment Pro- autophagosomes [37,38]. Since mitochondria are the initiators of var-
tein) and other proteins complete the fusion process. Autophagy is ious cell death programs and main sources of ROS; the effective and
completed once the degraded products are released back into the cy- immediate clearance of defective mitochondria is crucial for cellular
toplasm and the membranous compartment restores its pH and is re- health and homeostasis. Once marked by the specific receptors such as
cycled back to the lysosome pool [18]. NIX, BNIP3 (BCL2/adenovirus E1B 19 kDa protein-interacting protein
Evading cell death has been characterized as an important hallmark 3), P62, NBR1 (Neighbor of BRCA1 gene 1), Optineurin, and NDP52
of cancer cells. Cells follow different types of death programs when (Nuclear dot protein 52 kDa), etc. on their membranes, autophagy is
under stress, like apoptosis, necrosis, necroptosis, ferroptosis and so on very potent in clearing the defective mitochondria. In order to conserve
[25]. While autophagy is well known to play a cell survival role under and protect the physiologically active parts of the damaged mitochon-
stress or in response to various chemotherapeutic drugs but, there are dria, the damaged regions of mitochondria are specifically excised via
reports which suggest that autophagy can itself act as a form of cell the fission process and then degraded by mitophagy [39]. This also
death [26]. This type of cell death is called autophagic cell death and gives us a clear idea of how tightly mitophagy is regulated and syn-
may play an important role in developing novel cancer strategies. chronized with the fission process, that only a damaged portion of
During autophagic cell death, autophagic vesicles are accumulated in- mitochondria is engulfed by autophagosomes. Targeting the mitophagy
side the cytoplasm and many of the Atg proteins play an active role in is a promising anti-cancer therapeutic strategy [38–40]. Although mi-
the induction of autophagic cell death. This type of cell death is highly tophagy has been generally considered as a cell protective mechanism,
context-dependent and has been specifically found upregulated in the both mitophagy inhibition and induction have been implicated in reg-
apoptosis-resistant cells [27–30]. Apoptosis resistant, Bax/Bak deficient ulating tumor growth. For example, inhibition of mitophagy in KRAS-
cells undergo autophagic cell death upon treatment with therapeutic driven cancers and melanoma can be a potential target to sensitize
drugs [31]. Also, inhibition of caspase-8 one of the key proteins in- these tumors to therapy [41]. Salinomycin and Liensinine are known to
volved in the intrinsic apoptotic pathway results in the induction of inhibit mitophagy [42] and have been reported to enhance the sensi-
autophagic cell death which is dependent on Beclin-1 and Atg7 [32]. In tivity of breast cancer cells to various DNA damaging chemotherapeutic
contrast to caspase-8, activation of caspase-10 in myeloma cells inhibits drugs [43]. In colorectal cancer stem cells inhibition of mitophagy by
the induction of autophagic cell death by cleaving the BCLAF1 (BCL2- BNIP3L knockdown enhances doxorubicin (DNA damaging agent)
associated transcription factor 1) protein [33]. Autophagic cell death sensitivity. In contrast, the negative regulator of mitophagy, USP30
offers a unique window of therapeutic potential, as cancer cells are (Ubiquitin Specific Peptidase 30), represents a potential target whose
highly resistant to apoptosis. Therefore, drugs inducing autophagic cell inhibition can increase mitophagy, and suppress tumor growth [44].
death can be useful in managing the apoptosis-resistant cancers. Also, Also, the adverse tumor-promoting effect of chronic mitophagy in-
most of the cancer cells become resistant to common anticancer drugs, hibition has been observed by deletion or inactivation of genes PARKIN
which act via the initiation of apoptosis or necroptosis. But autophagic and BNIP3 [45]. Mitophagy may be the common mechanism to increase
cell death can be induced in these resistant cancer cells as well. drug resistance and promote tumor survival, and accordingly, mito-
Therefore, both inhibition and induction of autophagy should be con- phagy inhibition may be a general strategy to decrease drug resistance
sidered as therapeutic interventions. In addition, non-canonical au- [46]. However, there are reports, which suggest that mitophagy in-
tophagy has also been shown to contribute to cell death through a duction may be a therapeutically relevant strategy as well. For example,
process that is independent of the entire machinery of Atg proteins excessive mitophagy induced by ceramide by directly interacting with
[34]. Currently, autophagy inhibitor hydroxychloroquine (HCQ) is in LC3 has been shown to be a tumor suppressor mechanism in xenograft
phase I and II clinical trials in combination with several standard che- models. Ceramide induced mitophagy depends on mitochondrial fission
motherapies while autophagy induction remains to be tested clinically [47]. Similarly, FLT3-ITD (Fms-like tyrosine kinase 3–internal tandem
[28]. Therefore, an in-depth understanding of the roles of autophagy duplication) signaling axis acts as a resistance mechanism in AML
during different stages of carcinogenesis has a unique potential to guide (acute myeloid leukemia). The FLT3 provides resistance by limiting the
the development of therapeutic strategies to eliminate cancer cells, ceramide generation and accumulation on the mitochondrial outer
prevent drug resistance and also stop recurrence. membrane. Targeting FLT3 signaling activated ceramide-induced lethal
mitophagy and effectively evaded the drug resistance mechanism [48].
2. Selective autophagy and cancer: a window to novel target Similarly, a small molecule activator of SITR1 (Sirtuin-1) has been
shown to induce the mitophagy related cell death in glioblastoma [49].
Selective or cargo-specific autophagy has been shown to play a pi-
votal role in various pathological conditions including cancer. Recently, 2.2. Reticulophagy
various selective forms of autophagy have been reported such as nu-
cleophagy, RNAphagy, ferrtinophagy, etc. (Fig. 1). However, further ER (Endoplasmic Reticulum) comprised of interconnected mem-
research is required to prove a more direct association of these newly branous sheets that facilitate protein and lipid biosynthesis, proteins
discovered selective forms of autophagy and disease progression. Re- quality control, regulates vesicles trafficking, and ion homoeostasis
cently, various studies have highlighted the importance of selective [50,51]. During stress or unfavorable conditions, some proteins become
forms of autophagy like mitophagy, reticulophagy, and ribophagy as misfolded/aggregated and get accumulated in the lumen of the ER,
druggable targets. In this section, we will discuss different forms of leading to ER stress [52,53]. During ER stress two interrelated quality
selective autophagy, their importance and role in cancer cells and how control pathways- the UPR (unfolded protein response) and the ERAD
these processes can be manipulated for better therapeutic interventions. (ER-associated protein degradation) [54] pathways are activated. UPR
activation increases the folding capacity of the ER [55,56], whereas the
2.1. Mitophagy ERAD system recognizes terminally misfolded proteins and facilitates
their translocation back into the cytoplasm from the ER. In the cyto-
Mitochondria are the central hubs of cell death, metabolism, im- plasm, these misfolded proteins are then degraded by the ubiquitin-
mune surveillance, reactive oxygen species (ROS) and adenosine tri- proteasome system [57]. The UPR induces various regulatory molecules
phosphate (ATP) production [35,36]. Autophagy has an advantage over that sense elevated levels of unfolded and aggregated proteins in the
ubiquitin-proteasome system that can degrade an organelle as a whole lumen and activates downstream PERK-eIF2α signaling pathway to
without disassembling it to its constituent macromolecules. For in- attenuate the global translation. The same pathway also leads to the up-
stance, mitochondria have their own genomes and translation regulation of transcription of ER stress response genes (such as ATF4,
172
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Ishaq, et al. Seminars in Cancer Biology 66 (2020) 171–181
Fig. 1. Different types of selective autophagy in mammalian cells. Selective autophagy represents the recognition and degradation of specific cargo. Selective
autophagy receptors for the respective processes are listed. The question mark indicates yet to unidentified receptor proteins.
ATF3, CHOP, CCNG2, GADD34, etc.) and restores the ER health and the RTN3 oligomerization. RTN3 oligomerization leads to the ER-
homeostasis [56]. fragmentation and subsequent lysosomal degradation via the autop-
Various groups have shown the interconnection between ER stress hagy-lysosomal pathway.
and cancer. Moreover, the UPR plays an important role in resistance SEC62 is a subunit of the SEC translocation machinery involved in
mechanisms against cancer therapy [58]. UPR induction has been the transport of newly synthesized proteins into the ER. Recently,
known to increase the autophagy via upregulation of ATG genes SEC62 is reported to function as an autophagy receptor during the re-
[59–61]. Interestingly, it has been shown that the accumulation of covery phase after ER stress [76]. SEC62 is located at the rough ER and
aggregated/misfolded proteins in the ER lumen leads to removal of the selectively delivers those portions of ER, which are enriched in the
ER fragment via autophagy, which also known as reticulophagy/ER- molecular chaperones and other such proteins after UPR. The function
phagy [62–64]. Just like the mitophagy, six specific ER-phagy receptors of SEC62 mediated reticulophagy is to restore the ER to the basal
have been reported- Sec62 [65], FAM134B [66], CCPG1 (cell-cycle condition after the ER stress response is over [76]. SEC62 is frequently
progression gene 1) [67], RTN3 (reticulon3) [68], ATL3 (Atlastins) [69] amplified in non-small cell lung, prostate, thyroid cancers, and head
and TEX264 [70]. The cytoplasmic sides of these receptors contain and neck cancers [77]. In addition, a recent study by Ojha et al. have
LC3/GABARAP interacting region, which helps them to bind with au- shown that SEC61 allows translocation of BRAF-MEK-ERK signaling
tophagosomes. These receptors have a distinct spatial distribution on axis into the ER lumen during BRAF and MEK inhibition treatment.
the ER and are therefore involved in ER-phagy of the different ER Interestingly, inhibition of SEC61 translocon blocks autophagy and
compartments. FAM134B is enriched at the edges of ER-sheets and induces cancer cell death in melanoma cells [78]. CCPG1, an ER-re-
helps in the autophagic degradation of fragmented portions of ER. sident transmembrane protein, contains a LIR at the N-terminal cyto-
FAM134B itself is involved in the fragmentation of ER-sheets via its RH solic domain which binds with LC3/GABARAP [79,80]. CCPG1 is
domains and in selectively targeting these ER-fragments to autopha- mainly localized on tubular-ER and is therefore involved in the au-
gosomes by its LC3/GABARAP interacting regions. Inhibition of tophagic degradation of tubular portions of the ER. CCPG1 is unique
FAM134B increased swelling and expansion of the ER, while over- among the known ER-phagy receptors as it also contains two FIRs
expression of FAM134B led to ER fragmentation and degradation [71] (FIP200 interacting regions) apart from LC3 and GABARAP binding
by reticulophagy. FAM134B is involved in various cellular signaling sites. These FIRs can directly recruit the FIP200-ULK complex and in-
and plays key roles in the pathogenesis of different human diseases itiate the autophagy directly. Although these studies do suggest the role
including cancer [72]. Interestingly, FAM134B acts as an oncogene and of reticulophagy in cancer, but further studies in preclinical models are
stimulates esophageal squamous cell carcinoma cell growth and pro- required to make reticulophagy as a druggable target. ATL3 is also the
liferation, on the other hand, it acts as a tumor suppressor and reduces autophagic adaptor for tubular-ER and belongs to the dynamin-like
colorectal adenocarcinomas cancer cells growth and proliferation [72]. GTPase family. ATL3 only contains GIMs (GABARAP interacting motif)
FAM134B mutations are common in esophageal squamous cell carci- but not the LIRs. TEX264 is the latest addition of ER-phagy receptor
noma and colorectal adenocarcinomas [73,74]. RTN3 as a new re- family. TEX264 has been shown to be the most robust of all the ER-
ticulophagy receptor, involved in the selective degradation of ER tu- phagy receptors and is accordingly is found uniformly distributed on
bules [75]. RTN3 is selectively enriched at the curved ends of ER- the ER. This leads to the speculation that drugs blocking autophagy
tubules and any increase in the local concentration of RTN3 results in could be an advantageous component of chemotherapeutic regimens
173
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Ishaq, et al. Seminars in Cancer Biology 66 (2020) 171–181
aiming at counteracting the protective effects of SEC62 overexpression ribosomes in malignant transformation is becoming more obvious, the
in tumors. drugs targeting ribosome biogenesis may become very useful in tar-
geting cancer cells with hyperactive ribosomes.
2.3. Ribophagy
3. Selective autophagy, and clinical translation
Ribosomes were found inside the autophagosomes by electron mi-
croscopy in 1960 and were believed to be passively engulfed by au- 3.1. Autophagy and the web of inter-organelle membrane contact sites
tophagosomes along with the other cytoplasmic contents [81–83]. De-
gradation of ribosomes via autophagy (ribophagy) was well established In the past few decades cell biologists have now well established
by Kraft et al. as a distinct process than the bulk nonselective autophagy that cellular organelle like mitochondria, endoplasmic reticulum, en-
during nutrient starvation [84]. Ribosomes are very stable molecules dosomes etc. are interconnected with each other via the structures
with a half-life of several days and comprise almost about 50% protein called as membrane contact sites (MCS). MCS are well-defined struc-
in normally growing cells. During nutrient starvation, cells can easily tures where the membranes of two organelles are in close proximity
degrade the highly stable ribosomes and respond to cellular stress in an (10-50 nm) and are held together by specific proteins called as tethering
effective way. Lessons from the mitophagy do provide the insights that proteins. These MCS sites act as signaling hubs and are also involved in
ribophagy may be a selective process and almost a decade later, the first diverse functions like trafficking of lipids and ions [100]. The ER forms
ribophagy receptor was hunted down by the Sabatini group in 2018 MCS with mitochondria, endosomes, and autophagosomes, therefore a
[85]. Most of the autophagy receptors interact with LC3 via their LIR specific set of proteins are enriched at ER-mitochondria, ER-endosomes,
domains and ribophagy receptor, NUFIP1 (nuclear fragile X mental and ER-autophagosomes contact sites [101]. Similarly, the mitochon-
retardation–interacting protein 1) proved no exception to that. NUFIP1 dria itself is also connected with lysosomes, endosomes, lipid droplets
via interacting with LC3 helps in the degradation of ribosomes and and peroxisomes. The ER-MCS have very specific functions and re-
leads to cell survival during starvation. cently, ER-mitochondria and ER-plasma membrane contact sites have
Both 60s and 40s ribosomal subunits are synthesized in the nu- been shown to act as a source of autophagosomes biogenesis [102,103].
cleolus, exported into the cytoplasm where they are finally assembled VMP-1 (Vacuole membrane protein 1), an ER-resident protein regulates
into the mature 80s subunit by the help of accessory proteins [86,87]. the ER-MCS and acts as a positive regulator of autophagosome bio-
The cancer biologists already were aware of the alterations in nucleolus genesis [104–106]. The ER-MCS and autophagosomes biogenesis are
size, number, and morphology, but the importance of the ribosome it- intricately linked as VMP1 inhibition increases the number of ER-MCS
self was not recognized in cancer cells [88]. Cancerous cells, in general, but blocks autophagosome formation. During the loss of ER-MCS sig-
have the high ribosome biogenesis and are believed to be addicted to naling, cells dependent on other stress response systems, to maintain
the ribosomes as well [89]. This seems logically simple, as cancer cells organelle homeostasis and integrity [107,108]. ER-mitochondria con-
are rapidly dividing and therefore in need of a constant supply of the tact sites are at the center of metabolic regulation and cell stress re-
proteins necessary for growth and proliferation. Also, most of the active sponse. Mitochondria are in contact with both smooth ER and rough
signaling cascades in cancerous cells positively regulate ribosome bio- ER, but the functional significance of rough ER-mitochondria is not
genesis. However, there is evidence that suggests that increased ribo- clear yet. Smooth ER-mitochondria contact sites are involved in Ca2+
some numbers can itself lead to cancer [90]. Various studies have lipid transport across the two organelles. There are several other ways
shown the increased ribosomal number is upstream of the oncogenic by which ER-Mitochondria may be involved in cancer progression and
signaling cascades [91]. Ribosomes were thought to be very static nano- drug resistance and have been excellently reviewed by Doghman and
machines that statically translated the genome. Ribosomes, in general, Lalli [109]. Here we will be mainly focusing on the relationship of
were thought to be highly regulated molecular machines without autophagy with ER-mitochondria contact sites and its role in cancer
having any regulatory function of their own. However, there is now progression and drug resistance. ER-mitochondria contact sites have
evidence that cancer cells during the malignant transformation may been reported to overcome the cisplatin resistance by increasing cal-
acquire the property of assembling the specialized ribosomes [92,93]. cium transport from ER to mitochondria in colorectal cancer cells
But, how do cancer cells maintain these specialized ribosomes will be [110]. Bcl2 overexpression decreases the cisplatin sensitivity of ovarian
very critical for proper cancer cell proliferation and is one of the most cancer cells by decreasing the ER-mitochondria contact sites SKVO3
critical questions among the cancer biologists. cells [111].
Rack1 (Receptor for Activated C Kinase 1) interacts with the 40s ER is also in contact with the endosomes and these contact sites are
ribosomal subunit and acts as an adaptor for various signaling mole- quite distinct from the ER-mitochondria contact sites. At the site of
cules. Rack1 deficient ribosomes have been shown to increase the ex- endosome-ER contact, a distance of 10 nM only separates the two
pression of genes involved in non-canonical autophagy [94,95]. The membranes [112]. ER-endosome contact sites regulate the position,
function of Rack1 depends on its interacting proteins so Rack1 may play dynamics, and fission of the endosomes. VAPA, VAPB, MOSPD2,
a central role in linking autophagy and ribosome function. Rack1 has STARD3, and STARD3NL are the key proteins involved in the main-
been also shown to directly interact with the Atg5 [73] and also pro- tenance of the ER-endosome contact sites [113–115]. VAPA and VAPB
mote autophagy by VPS34 complex formation [96]. Recently, Sundar- are integral ER membrane proteins and have been recently reported to
amoorthy et al elucidated the role of Rack1 in ribosomal quality control directly involved in autophagy by helping in the formation of Isolation
(RQC) by regulating the ubiquitination of 40s ribosomal subunit. Ri- membrane [116]. VAPA and VAPB directly interact with the ULK-
bosomal ubiquitination is now emerging at the center of ribosome FIP200 via their FFAT motifs. ER has been also shown to form contacts
quality control [97]. In fact, the autophagy-mediated degradation of with lysosomes for the maintenance of calcium levels in the cell. The
ribosomes is also regulated by ubiquitin protease. It is very important to interaction is specifically mediated by the IP3Rs and these receptors are
note that the de-ubiquitination of ribosomal subunits serves the signal also involved in translocation of calcium from ER into the lysosomes
for ribophagy. Listerin1 is one of the crucial RQC factors that is a [117].
ubiquitin ligase and effectively inhibits the ribophagy [98]. Listerin1 is In conclusion, the MCS are at the center of cellular signaling, me-
involved in the degradation of polypeptide chains associated with tabolism, growth and cell death. Autophagosome biogenesis depending
stalled ribosomes [99]. Since post-translational modifications act as on the stimulus is initiated at ER-mitochondria contact sites, plasma
molecular signatures for selective autophagy degradation therefore the membrane, ER, recycling endosomes. Specific autophagy receptors are
association of Rack1, Listerin1 and other adaptors may be also crucial localized on the ER and on the mitochondria and may fine-tune the
for the regulation of ribosomal function. As the importance of drug resistance mechanisms in cancer cells. Targeting the MCS enriched
174
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Ishaq, et al. Seminars in Cancer Biology 66 (2020) 171–181
proteins along with the conventional anticancer agents may be the next- regulators of cellular metabolism and autophagy is one of the key
generation strategy in treating drug-resistant cancers. Further studies processes that maintain the healthy pool of the mitochondria. By reg-
are needed to define the MCS, autophagy and their dynamics under ulating the mitochondrial number autophagy indirectly contributes
different stress conditions. The regulatory loops and specificity of pro- towards maintaining the key metabolic intermediates. The final stages
teins enriched at mitochondria-ER-autophagosome biogenesis sites and of autophagy involve the recycling of digested intracellular components
other contact sites will provide a better understanding of the role with the help of lysosomes. So, autophagy is also directly involved in
played by autophagy in tumor progression and particularly in drug metabolic regulation by degrading various complex macromolecules
resistance mechanisms. into their smaller subunits. Accordingly, cancer cells are mostly ad-
dicted to functional autophagy and deletion of autophagy genes impair
3.2. Autophagy in cancer stem cells the tumor growth in melanoma [135], urothelial carcinoma, non-small-
cell lung cancer cells and a mouse model for allografts of melanoma
Cancer Stem Cells (CSCs) are reported to have higher autophagic [136]. Autophagy can recycle back amino acids, carbohydrates, and
flux compared to bulk tumor cells [118,119]. Various ATG genes are nucleotides that can essentially maintain all the central metabolic
highly expressed in CSCs for the maintenance of CSCs dormancy. The pathways like the tricarboxylic acid cycle (TCA) cycle and glycolysis.
increased autophagic flux has been reported to play a major role in CSC Since the cancer cells are limited by the availability of nutrients, au-
mediated development of tumorigenesis in leukemia [120] and breast tophagy can provide essential metabolites and act as a buffering system
cancer [121]. Additionally, autophagy induction provides resistance to during nutritional scarcity. Autophagy has been shown to play an im-
photodynamic therapy in CSCs [122], leading to the recurrence and portant role in maintaining glycolysis in breast cancer cells, chronic
progression of colorectal cancer [123]. Recently, autophagy has shown myeloid leukemia, and Ras-driven cancers [137,138]. The levels of
to play an adverse role in osimertinib induced cytotoxicity by inducing various metabolic intermediates have also been reported to inhibit or
stem cell-like properties in non-small cell lung carcinoma. Combined induce autophagy. High glucose levels in pancreatic cancer patients
treatment of epidermal growth factor receptor-tyrosin kinase inhibitor were recently reported to inhibit autophagy via SREBP1 and lead to the
and autophagy inhibitor inhibits the stemness and restored the toxicity poor disease prognosis [139].
of osimertinib [124]. On the contrary, the increased autophagy induced Acetyl-CoA acts as an autophagy inhibitor via the acetyltransferase
by nigericin has been shown to suppress CSCs characteristics in glioma EP300 [140]. Acetyl-CoA is a key metabolic intermediate involved in
cells [125]. Both autophagy and CSCs are the two arsenals deployed by the regulation of autophagy. Acetyl-CoA depletion induces autophagy
various tumors for the development of drug resistance [126], therefore while increasing its concentration even under starvation can effectively
the underlying regulatory mechanism between these two factors may be inhibit autophagy [140]. Acetyl-CoA pool in cells is generated via
critical for the development of new drug combinations. In this aspect, glycolysis or by the degradation of some amino acids, both of which are
our lab has previously reported that the treatment of CQ (chloroquine), among the key regulators of autophagy. Recently, Lee et al have shown
a well know autophagy inhibitor, increases the sensitivity towards that in glioblastoma cells acetyl-CoA can up-regulate the genes involved
gemcitabine and mitomycin [127]. Another study also demonstrated in cell migration- and adhesion via Ca2+–NFAT (nuclear factor of ac-
that CQ reduces CSCs populations in triple-negative breast CSCs [128]. tivated T cells 1) signaling axis. The crosstalk between a metabolic
Similarly, salinomycin by inhibiting autophagy has also been reported intermediate Acetyl-CoA and Autophagy may be key in determining
to reduce the proportion of breast CSC population [129,130]. Also, tumor development and metastasis. Accordingly, the autophagy-Acetyl-
salinomycin has shown to inhibit autophagy under conditions of tran- CoA-NFAT axis may be, therefore, exploited as a potential anticancer
sient and chronic acidosis. This study suggested that the development strategy [141]. Apart from glycolysis, inhibition of autophagy by CQ
and use of clinically suitable derivates of salinomycin may improve the has been shown to decreases the oxidative phosphorylation in pan-
efficacy of autophagy inhibition in the acidic tumor microenvironment creatic cancer cells [142]. In Atg7-null mice, studies have shown the
[129]. increasing circulating metabolites differently regulated compared to
Recently, drug-loaded albumin coated nanoparticle has shown to Atg7+/+ mice. Arginine was highly down-regulated in Atg7Δ/Δ mice
facilitate efficient targeted therapy in glioma CSCs during the combi- [143], providing a direct link between amino acid metabolism and
nation of chemotherapy and autophagy [131]. In addition, the en- autophagy. Further studies in mice models may provide details on how
dosomal pathway has shown to be involved in the maintenance and autophagy regulates and supplies growth-limiting factors to the cancer
survival of cancer stem cells in colorectal cancer. A combination of cells under hypoxia, ischemia and other stressful condition.
mefloquine hydrochloride, a novel RAB5/7 inhibitor with chemother-
apeutic agent-induced colorectal cancer cell death [132]. In addition, 3.4. Autophagy in tumor immunity
autophagy has shown to play an important role in the maintenance of
CSCs by the regulation of immune cells and cytokine production. Au- Autophagy has been well known to play a role in antibacterial and
tophagy inhibition modified CD44high/CD25low population in breast viral immunity [144]. Also, in Atg7-null mice, an increased antitumor
CSCs by regulating CD24 and IL-6 in autophagy-dependent breast immune response was observed, indicating as a potential reason for
cancer [133]. Autophagy gene ATG7 induced OCT4 transcription via β- reduced tumor growth [145]. Functional autophagy is necessary for the
catenin to facilitates characteristics of CSCs in prostate cancer. CSCs survival and development of NK (Natural Killer) cells, and neutrophils
maintain their characteristics by modifying monocytes/macrophages in antitumor immune response and in tumor microenvironment
toward tumor-associated macrophages to provide resistance to prostate [146,147], and is also required for the effective onset of DC (dendritic
cancer via IL6/STAT3 [134]. Altogether, the role of autophagy in CSCs cells) mediated adaptive anticancer immunity. However, the deletion of
may not be as straight forward as thought earlier, however targeting ATG5 in DCs does not affect the presentation of tumor-associated an-
autophagy and CSCs may be a potent way to block tumor drug re- tigens on MHC- I molecules [148]. In addition, Treg cells by inhibiting
sistance and recurrence. CSCs can be a cause of therapeutic escape autophagy are reported to suppress autoimmune response in DCs. Au-
leading to tumor recurrence and relapse. Controlling the recruitment tophagy has shown to be upregulated in patients and mouse models of
and sustenance of CSCs has been achieved with autophagy inhibitors. melanoma. Inhibition of autophagy in myeloid cells decreased tumor
growth and induce an anti-tumor immune response in vivo. Autophagy
3.3. Autophagy in cancer metabolism deficient MDSCs (Myeloid-derived suppressor cells) showed decreased
expression of the membrane-associated RING-CH1 (MARCH1) E3 ubi-
Autophagy contributes both directly and indirectly in maintaining quitin ligase that mediates the lysosomal degradation, which led to
the metabolic pool of cancer cells. Mitochondria are the central enhance the surface expression of MHC-II and thereby induce tumor-
175
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Ishaq, et al. Seminars in Cancer Biology 66 (2020) 171–181
specific CD4-T cells. This study illustrates that autophagy may be a promote apoptosis by inhibition of autophagy [161]. The initial phase
potential druggable target of MDSCs-mediated suppression of antitumor I/II trials of HCQ have been on adult solid tumors [162]. These included
immunity, however, this study did not correlate with in vivo findings studies on patients with pancreatic adenocarcinoma, melanoma, col-
[149]. Recently, genetic inhibition of BECN1 has shown to inhibit orectal carcinoma, myeloma, lymphoma, and renal cell carcinoma. The
tumor growth by overexpression and release of CCL5 cytokine via chemotherapeutic drugs used were temsirolimus, bortezomib, temozo-
MAPK8/JNK-JUN/c-Jun signaling pathway, leading to massive NK cells lomide, vorinostat, and doxorubicin [163–169]. The cancer patients
infiltrations into the tumor microenvironment. More importantly, the received HCQ in the range of 400 mg–600 mg BD. The study found the
authors demonstrated a strong clinical positive correlation between the dosages to be well tolerated with partial response and stable disease in a
expression of NK cell marker and CCL5 in melanoma patients and found subset of population [170]. 1200 mg /day is the highest possible dose
a significant increased survival in melanoma patients expressing a high approved by food and drug administration and these trials established
level of CCL5. Overall, these findings emphasize the importance of the safety profile of HCQ in combination with cytotoxic drugs in solid
autophagy inhibition in the exploitation of NK cell-mediated anti-tumor tumors [170]. HCQ was used in combination with 150 mg/m2 of te-
immunity [150]. mozolomide in advanced solid malignancies and melanoma [171].
In addition, the deletion of autophagy genes Atg5, Atg14, or Atg16l1 Rangwala et al. showed a 27 % stable disease and 14 % partial response
has shown to impair tumor growth in autophagy-competent syngeneic in wild type melanoma [165,166]. The combination of HCQ produced
mammary, prostate, and colorectal tumors [151]. The authors further synergistic activity with rapamycin by suppressing mTORC1 activity.
demonstrated that Atg5 deficient CD8+ T cells acquire an effector The mTOR inhibitor temsirolimus was used in combination with HCQ
memory phenotype. These phenotypic changes are mediated by en- in phase I trial by the same group in advanced solid tumors [164]. The
hanced glucose metabolism resulting in alterations in histone methy- combination was well tolerated with limited side effects. Vogl et al.
lation which increases in H3K4me3 density and leads to the upregula- treated 25 myeloma patients with bortezomib in combination with HCQ
tion of both metabolic and effector target genes. Interestingly, [164]. Autophagy inhibition with HCQ enhances the efficacy of pro-
regardless of the significant reduction in total CD8 + TILs (tumor-in- teasome inhibitor by leading to the accumulation of misfolded proteins
filtrating cells), Atg5 deficiency leads to a significant shift towards IFN- [134]. Forty-five percent of patients showed stable disease as their best
γ- and TNF-α-producing effector memory cells. However, restricting response. The most common adverse events in this trial were gastro-
glucose in Atg5−/− T cells and adoptive transfer with a sub-therapeutic intestinal and cytopenias [164]. Sorafenib is a multi tyrosine kinase
dose of atg5−/− T cells significantly decreased the tumor growth and mTOR inhibitor, which has been used in hepatocellular carcinoma
reduced the tumor volume. This finding highlights the unique role of [169]. It has been found to induce autophagy and the same is being
cell-autonomous T cell autophagy in anti-tumor immune response in investigated as the cause of possible drug resistance. Two important
cancer [151]. Recently, Chen at al shown that autophagy inhibitor CQ clinical trials (Table 1) are studying the use of HCQ in hepatocellular
(chloroquine) increases macrophage lysosomal pH which leads to ly- carcinoma. HCQ, when used at high doses and for a longer duration,
sosomal Ca2+ release, polarizing tumor-associated macrophages from showed retinal toxicity. Thus, baseline retinal screening and annual
M2 to M1 phenotype [152]. This study highlighted yet another me- screening after five years has been recommended for early detection of
chanism of CQ as a potent immunotherapeutic modulator in cancer changes [171]. Targeted immunotherapeutic agents including check-
cells. Moreover, PDI inhibition leads to the induction of autophagy point inhibitors have limited efficacy and are expensive. If the efficacy
which may be a factor towards the resistance to the anti-PDI antibody
by tumor cells [153,154]. Therefore, autophagy inhibitors like CQ may
Table 1
be an effective way to increase the efficacy of immunotherapy. How-
Summary of clinical trials on autophagy in cancer.
ever further in-depth studies are needed to establish the role of PD1 and
autophagy in modulating the anti-immune response. Cancer Autophagy Clinical Trial registration
Modulators trial phase number
176
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Ishaq, et al. Seminars in Cancer Biology 66 (2020) 171–181
Fig. 2. Autophagy process, activators, inhibitors, and druggable targets. Nutrient starvation, growth factor deprivation is known to induce autophagy flux. These
factors lead to an increase in mTORC1 inhibition and AMPK activation. This initiates the upregulation of ULK1 complex through a series of phosphorylation cascades.
The membranes of isolation membrane/phagophore have multiple sources such as the ER, endosomal compartment, mitochondria, and plasma membrane. LC3
lipidation sites for autophagosome precursors by the recruitment of the two-ubiquitin ligase complexes. This step leads to the elongation of the phagosphore. The
autophagy process ends up by degradation of engulfed substrates in the lysosome after fusion with autophagosomes. Specific autophagy inducers and inhibitors are
also shown in the figures.
of autophagy modulators such as HCQ is proven to be worth, the Selective autophagy has opened a new therapeutic window for ef-
treatment can be made more cost-effective. fective cancer treatment. Different cancers are addicted and more
The various signaling cascades upstream of the phagophore, pro- sensitive to one process than the other. Therefore, targeting mitophagy,
teins involved in different steps of autophagy and factors regulating the reticulophagy, and ribophagy via their respective receptors can be ex-
end stages (lysosomes), have all been exploited for the therapeutic in- plored for widening to therapy alternatives.
terventions (Fig. 2). Various inhibitors have been reported by re-
searchers to target autophagy at different stages in different cancers. 5. Future challenges
These inhibitors may be considered as potential therapeutic molecules
against various cancers. First, targeting upstream signaling molecules: Autophagy modulation studies on tumor cell lines and mouse
inhibitors include SBI-0206965, inhibitor of the autophagy kinase ULK1 models have already led to the discovery of novel chemotherapeutic
(Unc-51 like autophagy activating kinase), Spautin-1, a class III PI3 strategies. There are lots of autophagy modulators in different phases of
kinase complex and Beclin1 inhibitor [172], SAR405, a kinase inhibitor clinical trials but several questions from clinical aspects remain to be
of Vps18 [172], Gambogic acid a natural anticancer agent induces answered. First and foremost, how to monitor the levels of autophagy
Caspase mediated cleavage of autophagy proteins involved in initiation inhibition or induction in patients treated with autophagy modulators?
and elongation phages of autophagosomes biogenesis [8]. Second, au- Once the autophagy is inhibited, how can we evaluate the status of
tophagy initiation inhibitors; ATG4 inhibitors, NSC185058 and autophagy in tumor subpopulations? To obtain all tumor cells in a
NSC377071 [173], Verteporfin, inhibits autophagosome formation at particular phase of cell cycle is another bone of contention in tapping
an early stage [174]. Third, lysosomal inhibitors: ROC325 [175], Lys05 the advantage of autophagy modulation. Predicting the outcome of
[176]. DQ661 [177] and DC661 [178] are new potential lysosomal- already existing anticancer drugs in combination with different au-
autophagy inhibitors. In addition, Autophagy has also been studied in tophagy inhibitors can be very useful in developing better treatment
potentiating the radiotherapy and as radiosensitizer in preclinical trials strategies. Classification of tumors, which are best served by autophagy
[179–181]. Woon Kim et al. studied autophagy induction in cells and inhibition or by autophagy induction is still a dillema. Identification of
mouse model of lung cancer using Z-DEVD (caspase-3 inhibitor), some markers which can predict whether autophagy is a pro-survival or
RAD001 (mTOR inhibitor) and irradiation. They found that the greatest pro-death process in specific cancer will be of immense importance for
induction of autophagy and associated radiation toxicity was observed effective treatment strategies. Identifying which role of autophagy (pro-
in the treatment group where all the three modalities were used si- survival or pro-death) is dominant in particular cancer, and at which
multaneously. They suggested that combined inhibition of apoptosis stage of tumor development autophagy should be targeted are two
and mTOR during radiotherapy may be a potential therapeutic strategy pertinent questions.Finally autophagy is a normal homeostatic reg-
to enhance radiation therapy in patients with non-small cell lung cancer ulatory mechnism of tissues. The inhibition of this mechanism can lead
[180]. Similarly, in glioma cells Wu et al. found that cell protective to lysosomal storage diseases, neurodegenerative disorders, diabetes
autophagy could be induced by AgNPs and/or radiation, which was and liver dysfunction. Similar to chemotherapetic drugs the con-
verified by the use of 3-MA [181]. comitant autophagy modulators affect the normal tissues and are not
177
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Ishaq, et al. Seminars in Cancer Biology 66 (2020) 171–181
tumor selective. Thus there is possibility of life threatenening effects of controversies in autophagy, Cell Death Discov. 1 (2015), https://doi.org/10.1038/
autophagy modulation in long term use [182]. cddiscovery.2015.36.
[18] L. Yu, Y. Chen, S.A. Tooze, Autophagy pathway: Cellular and molecular me-
Selective autophagy inhibitors which can specifically inhibit for chanisms, Autophagy 14 (2018) 207–215, https://doi.org/10.1080/15548627.
example mitophagy, reticulophagy or ribophagy may become potential 2017.1378838.
therapeutic agents for preventing the development of chemoresistance. [19] J. Kaur, J. Debnath, Autophagy at the crossroads of catabolism and anabolism,
Nat. Rev. Mol. Cell Biol. 16 (2015) 461–472, https://doi.org/10.1038/nrm4024.
Since autophagy is regulated via multiple signaling circuits and selec- [20] N. Fujita, T. Itoh, H. Omori, M. Fukuda, T. Noda, T. Yoshimori, The Atg16L
tion of a target molecule most critical for tumor growth will be a step complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy,
forward towards the development of more effective treatment strate- Mol. Cell Biol. 19 (2018) 2092–2100, https://doi.org/10.1091/mbc.E07-12-1257.
[21] I. Dikic, Z. Elazar, Mechanism and medical implications of mammalian autophagy,
gies. A better understanding and more clarity in the controversial pair Nat. Rev. Mol. Cell Biol. 19 (2018) 349–364, https://doi.org/10.1038/s41580-
roles like “anti-apoptotic or pro-apoptotic”, “Anti-tumorigenic or pro- 018-0003-4.
tumorigenic” will be the key factors in selecting autophagy modulators [22] C. Munch, I. Dikic, Hitchhiking on selective autophagy, Nat. Cell Biol. 20 (2018)
122–124, https://doi.org/10.1038/s41556-018-0036-0.
for cancer treatment. For better therapeutic interventions, a gold
[23] Q. Sun, W. Fan, K. Chen, X. Ding, S. Chen, Q. Zhong, Identification of Barkor as a
standard biomarker ideally in-patient serum or plasma samples, will be mammalian autophagy-specific factor for Beclin 1 and class III phosphatidylino-
a milestone in autophagy-cancer research. Taken together, the ther- sitol 3-kinase, Proc. Natl. Acad. Sci. U. S. A. 105 (2008) 19211–19216, https://doi.
apeutic interventions of autophagy at its different stages, via its dif- org/10.1073/pnas.0810452105.
[24] N.T. Ktistakis, S.A. Tooze, Digesting the expanding mechanisms of autophagy,
ferent receptors or through its signaling modulators, will be among the Trends Cell Biol. 26 (2016) 624–635, https://doi.org/10.1016/j.tcb.2016.03.006.
crucial factors for the development of precision medicine for different [25] L. Galluzzi, et al., Molecular mechanisms of cell death: recommendations of the
types of cancer patients. nomenclature committee on cell death 2018, Cell Death Differ. 25 (2018)
486–541, https://doi.org/10.1038/s41418-017-0012-4.
[26] D.A. Gewirtz, The four faces of autophagy: implications for cancer therapy, Cancer
Funding sources Res. 74 (2014) 647–651, https://doi.org/10.1158/0008-5472.CAN-13-2966.
[27] S.S. Singh, et al., Dual role of autophagy in hallmarks of cancer, Oncogene 37
(2017) 1142–1158, https://doi.org/10.1038/s41388-017-0046-6.
Mohd Ishaq and Rani Ojha are the recipients of the postdoctoral [28] R.K. Amaravadi, A.C. Kimmelman, J. Debnath, Targeting autophagy in Cancer:
fellowship at Stanford University CA, USA. recent advances and future directions, Cancer Discov. 9 (2019) 1167–1181,
https://doi.org/10.1158/2159-8290.CD-19-0292.
[29] A. Notte, L. Leclere, C. Michiels, Autophagy as a mediator of chemotherapy-in-
Declaration of Competing Interest duced cell death in cancer, Biochem. Pharmacol. 82 (2011) 427–434, https://doi.
org/10.1016/j.bcp.2011.06.015.
[30] J. Lorente, et al., The interplay between autophagy and tumorigenesis: exploiting
None.
autophagy as a means of anticancer therapy, Biol. Rev. Camb. Philos. Soc. 93
(2017) 152–165, https://doi.org/10.1111/brv.12337.
References [31] S. Shimizu, et al., Role of Bcl-2 family proteins in a non-apoptotic programmed cell
death dependent on autophagy genes, Nat. Cell Biol. 6 (2004) 1221–1228.
[32] L. Yu, et al., Regulation of an ATG7-beclin 1 program of autophagic cell death by
[1] J. Doherty, E.H. Baehrecke, Life, death and autophagy, Nat. Cell Biol. 20 (2018) caspase-8, Science 304 (2004) 1500–1502.
1110–1117, https://doi.org/10.1038/s41556-018-0201-5. [33] L. Lamy, et al., Control of autophagic cell death by caspase-10 in multiple mye-
[2] C.F. Bento, et al., Mammalian autophagy: how does it work? Annu. Rev. Biochem. loma, Cancer Cell 23 (2013) 435–449, https://doi.org/10.1016/j.ccr.2013.02.
85 (2016) 685–713, https://doi.org/10.1146/annurev-biochem-060815-014556. 017.
[3] N. Mizushima, A brief history of autophagy from cell biology to physiology and [34] F. Scarlatti, R. Maffei, I. Beau, P. Codogno, R. Ghidoni, Role of non-canonical
disease, Nat. Cell Biol. 20 (2018) 521–527, https://doi.org/10.1038/s41556-018- Beclin 1-independent autophagy in cell death induced by resveratrol in human
0092-5. breast cancer cells, Cell Death Differ. 8 (2008) 1318–1329, https://doi.org/10.
[4] B. Levine, D.J. Klionsky, Autophagy wins the 2016 Nobel Prize in Physiology or 1038/cdd.2008.51.
Medicine: breakthroughs in baker’s yeast fuel advances in biomedical research, [35] P.M. Quiros, T. Langer, C. Lopez-Otin, New roles for mitochondrial proteases in
Proc. Natl. Acad. Sci. U.S.A. 114 (2017) 201–205, https://doi.org/10.1073/pnas. health, ageing and disease, Nat. Rev. Mol. Cell Biol. 16 (2015) 345–359, https://
1619876114. doi.org/10.1038/nrm3984.
[5] G. Marino, M. Niso-Santano, E.H. Baehrecke, G. Kroemer, Self-consumption: the [36] N.M. Held, R.H. Houtkooper, Mitochondrial quality control pathways as de-
interplay of autophagy and apoptosis, Nat. Rev. Mol. Cell Biol. 15 (2014) 81–94, terminants of metabolic health, Bioessays 37 (2015) 867–876, https://doi.org/10.
https://doi.org/10.1038/nrm3735. 1002/bies.201500013.
[6] R. Ojha, M. Ishaq, S.K. Singh, Caspase-mediated crosstalk between autophagy and [37] S. Pickles, P. Vigie, R.J. Youle, Mitophagy and Quality Control Mechanisms in
apoptosis: mutual adjustment or matter of dominance, J. Cancer Res. Ther. 11 Mitochondrial Maintenance, Current Biol.: CB 28 (2018) R170–r185, https://doi.
(2015) 514–524, https://doi.org/10.4103/0973-1482.163695. org/10.1016/j.cub.2018.01.004.
[7] M. Ishaq, et al., Functional inhibition of Hsp70 by Pifithrin-mu switches Gambogic [38] S.M. Yoo, Y.K. Jung, A molecular approach to mitophagy and mitochondrial dy-
acid induced caspase dependent cell death to caspase independent cell death in namics, Mol. Cells 41 (2018) 18–26, https://doi.org/10.14348/molcells.2018.
human bladder cancer cells, Biochim. Biophys. Acta 1863 (2016) 2560–2573, 2277.
https://doi.org/10.1016/j.bbamcr.2016.07.001. [39] S.R. Yoshii, N. Mizushima, Autophagy machinery in the context of mammalian
[8] M. Ishaq, et al., Gambogic acid induced oxidative stress dependent caspase acti- mitophagy, Biochim. Biophys. Acta 1853 (2015) 2797–2801, https://doi.org/10.
vation regulates both apoptosis and autophagy by targeting various key molecules 1016/j.bbamcr.2015.01.013.
(NF-kappaB, Beclin-1, p62 and NBR1) in human bladder cancer cells, Biochim. [40] A.V. Kulikov, E.A. Luchkina, V. Gogvadze, B. Zhivotovsky, Mitophagy: Link to
Biophys. Acta 1840 (2014) 3374–3384, https://doi.org/10.1016/j.bbagen.2014. cancer development and therapy, Biochem. Biophys. Res. Commun. 482 (2017)
08.019. 432–439, https://doi.org/10.1016/j.bbrc.2016.10.088.
[9] M.F. Czyzyk-Krzeska, J. Meller, D.R. Plas, Not all autophagy is equal, Autophagy 8 [41] A.M. Strohecker, E. White, Targeting mitochondrial metabolism by inhibiting
(2012) 1155–1156, https://doi.org/10.4161/auto.20650. autophagy in BRAF-driven cancers, Cancer Discov. 4 (2014) 766–772, https://doi.
[10] D.C. Rubinsztein, J.E. Gestwicki, L.O. Murphy, D.J. Klionsky, Potential therapeutic org/10.1158/2159-8290.Cd-14-0196.
applications of autophagy, Nat. Rev. Drug Discov. 6 (2007) 304–312, https://doi. [42] J.R. Jangamreddy, et al., Salinomycin induces activation of autophagy, mitophagy
org/10.1038/nrd2272. and affects mitochondrial polarity: differences between primary and cancer cells,
[11] A.V. Onorati, M. Dyczynski, R. Ojha, R.K. Amaravadi, Targeting autophagy in Biochim. Biophys. Acta 1833 (2013) 2057–2069, https://doi.org/10.1016/j.
cancer, Cancer 124 (2018) 3307–3318, https://doi.org/10.1002/cncr.31335. bbamcr.2013.04.011.
[12] S. Mostowy, Autophagy and bacterial clearance: a not so clear picture, Cell. [43] J. Zhou, et al., A novel autophagy/mitophagy inhibitor liensinine sensitizes breast
Microbiol. 15 (2013) 395–402, https://doi.org/10.1111/cmi.12063. cancer cells to chemotherapy through DNM1L-mediated mitochondrial fission,
[13] B. Levine, N. Mizushima, H.W. Virgin, Autophagy in immunity and inflammation, Autophagy 11 (2015) 1259–1279, https://doi.org/10.1080/15548627.2015.
Nature 469 (2011) 323–335, https://doi.org/10.1038/nature09782. 1056970.
[14] D. Gatica, V. Lahiri, D.J. Klionsky, Cargo recognition and degradation by selective [44] J.R. Liang, et al., USP30 deubiquitylates mitochondrial Parkin substrates and re-
autophagy, Nat. Cell Biol. 20 (2018) 233–242, https://doi.org/10.1038/s41556- stricts apoptotic cell death, EMBO Rep. 16 (2015) 618–627, https://doi.org/10.
018-0037-z. 15252/embr.201439820.
[15] A.L. Anding, E.H. Baehrecke, Cleaning house: selective autophagy of Organelles, [45] J.A. Williams, et al., Chronic deletion and acute knockdown of parkin have dif-
Dev. Cell 41 (2017) 10–22, https://doi.org/10.1016/j.devcel.2017.02.016. ferential responses to acetaminophen-induced mitophagy and liver injury in mice,
[16] S. Kaushik, A.M. Cuervo, The coming of age of chaperone-mediated autophagy. J. Biol. Chem. 290 (2015) 10934–10946, https://doi.org/10.1074/jbc.M114.
Nature reviews, Mol. Cell Biol. 19 (2018) 365–381, https://doi.org/10.1038/ 602284.
s41580-018-0001-6. [46] Lauren E. Drake, Maya Z. Springer, Logan P. Poole, Casey J. Kim, F. Kay, Macleod
[17] L.M. Lindqvist, A.K. Simon, E.H. Baehrecke, Current questions and possible Expanding perspectives on the significance of mitophagy in cancer, Sem. Cancer
178
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Ishaq, et al. Seminars in Cancer Biology 66 (2020) 171–181
Biol. 47 (2017) 110–124, https://doi.org/10.1016/j.semcancer.2017.04.008. Sec63 in signal transduction, oncology and personalized medicine, Signal
[47] R.D. Sentelle, et al., Ceramide targets autophagosomes to mitochondria and in- Transduct. Target. Ther. 2 (2017) 17002, https://doi.org/10.1038/sigtrans.
duces lethal mitophagy, Nat. Chem. Biol. 8 (2008) 831–838. 2017.2.
[48] M. Dany, et al., Targeting FLT3-ITD signaling mediates ceramide-dependent mi- [78] R. Ojha, et al., ER translocation of the MAPK pathway drives therapy resistance in
tophagy and attenuates drug resistance in AML, Blood 128 (2016) 1944–1958. BRAF mutant melanoma, Cancer Discov. (2018), https://doi.org/10.1158/2159-
[49] Zhi-qiang Yao, et al., A novel small-molecule activator of Sirtuin-1 induces autop- 8290.Cd-18-0348.
hagic cell death/mitophagy as a potential therapeutic strategy in glioblastoma, Cell [79] M.D. Smith, S. Wilkinson, CCPG1, a cargo receptor required for reticulophagy and
Death Dis. 9 (2018) 767, https://doi.org/10.1038/s41419-018-0799-z. endoplasmic reticulum proteostasis, Autophagy 14 (2018) 1090–1091, https://
[50] L.M. Westrate, J.E. Lee, W.A. Prinz, G.K. Voeltz, Form follows function: the im- doi.org/10.1080/15548627.2018.1441473.
portance of endoplasmic reticulum shape, Annu. Rev. Biochem. 84 (2015) [80] N. Mizushima, A dual binding receptor for ER-phagy, Dev. Cell 44 (2018)
791–811, https://doi.org/10.1146/annurev-biochem-072711-163501. 133–135, https://doi.org/10.1016/j.devcel.2018.01.001.
[51] D.S. Schwarz, M.D. Blower, The endoplasmic reticulum: structure, function and [81] E.L. Eskelinen, F. Reggiori, M. Baba, A.L. Kovacs, P.O. Seglen, Seeing is believing:
response to cellular signaling, Cell. Mol. Life Sci. 73 (2016) 79–94, https://doi. the impact of electron microscopy on autophagy research, Autophagy 7 (2011)
org/10.1007/s00018-015-2052-6. 935–956.
[52] S.A. Oakes, F.R. Papa, The role of endoplasmic reticulum stress in human pa- [82] H. Nakatogawa, Spoon-feeding ribosomes to autophagy, Mol. Cell 71 (2018)
thology, Annu. Rev. Pathol. 10 (2015) 173–194, https://doi.org/10.1146/ 197–199, https://doi.org/10.1016/j.molcel.2018.07.003.
annurev-pathol-012513-104649. [83] M. Nofal, J.D. Rabinowitz, Ribosomes on the night shift, Science (New York, N.Y.)
[53] E. Chevet, C. Hetz, A. Samali, Endoplasmic reticulum stress-activated cell repro- 360 (2018) 710–711, https://doi.org/10.1126/science.aat7121.
gramming in oncogenesis, Cancer Discov. 5 (2015) 586–597, https://doi.org/10. [84] C. Kraft, A. Deplazes, M. Sohrmann, M. Peter, Mature ribosomes are selectively
1158/2159-8290.Cd-14-1490. degraded upon starvation by an autophagy pathway requiring the Ubp3p/Bre5p
[54] J. Hwang, L. Qi, Quality Control in the Endoplasmic Reticulum: Crosstalk between ubiquitin protease, Nat. Cell Biol. 10 (2008) 602–610, https://doi.org/10.1038/
ERAD and UPR pathways, Trends Biochem. Sci. 43 (2018) 593–605, https://doi. ncb1723.
org/10.1016/j.tibs.2018.06.005. [85] G.A. Wyant, et al., NUFIP1 is a ribosome receptor for starvation-induced ribo-
[55] A. Chakrabarti, A.W. Chen, J.D. Varner, A review of the mammalian unfolded phagy, Science (New York, N.Y.) 360 (2018) 751–758, https://doi.org/10.1126/
protein response, Biotechnol. Bioeng. 108 (2011) 2777–2793, https://doi.org/10. science.aar2663.
1002/bit.23282. [86] A. Fatica, D. Tollervey, Making ribosomes, Curr. Opin. Cell Biol. 14 (2002)
[56] R. Ojha, R.K. Amaravadi, Targeting the unfolded protein response in cancer, 313–318.
Pharmacol. Res. 120 (2017) 258–266, https://doi.org/10.1016/j.phrs.2017.04. [87] G.A. Brar, J.S. Weissman, Ribosome profiling reveals the what, when, where and
003. how of protein synthesis, Nat. Rev. Mol. Cell Biol. 16 (2015) 651–664, https://doi.
[57] J.A. Olzmann, R.R. Kopito, J.C. Christianson, The mammalian endoplasmic re- org/10.1038/nrm4069.
ticulum-associated degradation system, Cold Spring Harb. Perspect. Biol. 5 (2013), [88] S.O. Sulima, I.J.F. Hofman, K. De Keersmaecker, J.D. Dinman, How ribosomes
https://doi.org/10.1101/cshperspect.a013185. translate Cancer, Cancer Discov. 7 (2017) 1069–1087, https://doi.org/10.1158/
[58] H. Kim, A. Bhattacharya, L. Qi, Endoplasmic reticulum quality control in cancer: 2159-8290.Cd-17-0550.
friend or foe, Semin. Cancer Biol. 33 (2015) 25–33, https://doi.org/10.1016/j. [89] J. Pelletier, G. Thomas, S. Volarevic, Ribosome biogenesis in cancer: new players
semcancer.2015.02.003. and therapeutic avenues, Nat. Rev. Cancer 18 (2018) 51–63, https://doi.org/10.
[59] T. Yorimitsu, D.J. Klionsky, Endoplasmic reticulum stress: a new pathway to in- 1038/nrc.2017.104.
duce autophagy, Autophagy 3 (2007) 160–162. [90] A. de Las Heras-Rubio, L. Perucho, R. Paciucci, J. Vilardell, L.L. ME, Ribosomal
[60] T. Verfaillie, M. Salazar, G. Velasco, P. Agostinis, Linking ER stress to autophagy: proteins as novel players in tumorigenesis, Cancer Metastasis Rev. 33 (2014)
potential implications for Cancer therapy, Int. J. Cell Biol. 2010 (2010) 930509, , 115–141, https://doi.org/10.1007/s10555-013-9460-6.
https://doi.org/10.1155/2010/930509. [91] M.T. Pedersen, K.B. Jensen, Cell biology: unconventional translation in cancer,
[61] M. Smith, S. Wilkinson, ER homeostasis and autophagy, Essays Biochem. 61 Nature 541 (2017) 471–472, https://doi.org/10.1038/nature21115.
(2017) 625–635, https://doi.org/10.1042/ebc20170092. [92] M. Bucci, RNA modifications: ribosomes get decorated, Nat. Chem. Biol. 14 (2017)
[62] L. Li, J. Xu, L. Chen, Z. Jiang, Receptor-mediated reticulophagy: a novel promising 1, https://doi.org/10.1038/nchembio.2543.
therapy target for diseases, Acta Biochim. Biophys. Sin. 48 (2016) 774–776, [93] E. Brighenti, D. Trere, M. Derenzini, Targeted cancer therapy with ribosome
https://doi.org/10.1093/abbs/gmw057. biogenesis inhibitors: a real possibility? Oncotarget 6 (2015) 38617–38627,
[63] P. Grumati, I. Dikic, A. Stolz, ER-phagy at a glance, J. Cell. Sci. 131 (2018), https://doi.org/10.18632/oncotarget.5775.
https://doi.org/10.1242/jcs.217364. [94] S. Erbil, et al., RACK1 is an interaction partner of ATG5 and a novel regulator of
[64] M. Loi, I. Fregno, C. Guerra, M. Molinari, Eat it right: ER-phagy and recovER- autophagy, J. Biol. Chem. 291 (2016) 16753–16765, https://doi.org/10.1074/jbc.
phagy, Biochem. Soc. Trans. 46 (2018) 699–706, https://doi.org/10.1042/ M115.708081.
bst20170354. [95] H.D. Kim, E. Kong, Y. Kim, J.S. Chang, J. Kim, RACK1 depletion in the ribosome
[65] F. Fumagalli, et al., Translocon component Sec62 acts in endoplasmic reticulum induces selective translation for non-canonical autophagy, Cell Death Dis. 8
turnover during stress recovery, Nat. Cell Biol. 18 (2016) 1173–1184, https://doi. (2017) e2800, https://doi.org/10.1038/cddis.2017.204.
org/10.1038/ncb3423. [96] Y. Zhao, et al., RACK1 promotes autophagy by enhancing the Atg14L-Beclin 1-
[66] R.M. Bhaskara, et al., Curvature induction and membrane remodeling by Vps34-Vps15 complex formation upon phosphorylation by AMPK, Cell Rep. 13
FAM134B reticulon homology domain assist selective ER-phagy, Nature Commun. (2015) 1407–1417, https://doi.org/10.1016/j.celrep.2015.10.011.
10 (2019) 2370, https://doi.org/10.1038/s41467-019-10345-3. [97] E. Sundaramoorthy, et al., ZNF598 and RACK1 regulate mammalian ribosome-
[67] M.D. Smith, et al., CCPG1 is a non-canonical autophagy cargo receptor essential associated quality control function by mediating regulatory 40S ribosomal ubi-
for ER-Phagy and pancreatic ER proteostasis, Dev. Cell 44 (2017) 217–232, quitylation, Mol. Cell 65 (2017) 751–760, https://doi.org/10.1016/j.molcel.2016.
https://doi.org/10.1016/j.devcel.2017. 12.026 e754.
[68] P. Grumati, et al., Full length RTN3 regulates turnover of tubular endoplasmic [98] B. Ossareh-Nazari, et al., Ubiquitylation by the Ltn1 E3 ligase protects 60S ribo-
reticulum via selective autophagy, Elife 6 (2017) e25555, , https://doi.org/10. somes from starvation-induced selective autophagy, J. Cell Biol. 204 (2014)
7554/eLife.25555. 909–917, https://doi.org/10.1083/jcb.201308139.
[69] Q. Chen, Y. Xiao, P. Chai, P. Zheng, J. Teng, J. Chen, ATL3 is a tubular ER-Phagy [99] Y. Matsuo, et al., Ubiquitination of stalled ribosome triggers ribosome-associated
receptor for GABARAP-Mediated selective autophagy, Curr. Biol. 29 (2019) quality control, Nat. Commun. 8 (2017) 159, https://doi.org/10.1038/s41467-
846–855, https://doi.org/10.1016/j.cub.2019.01.041 e6. 017-00188-1.
[70] H. Chino, T. Hatta, T. Natsume, N. Mizushima, Intrinsically disordered protein [100] M.J. Phillips, G.K. Voeltz, Structure and function of ER membrane contact sites
TEX264 mediates ER-phagy, Mol. Cell 74 (2019) 909–921, https://doi.org/10. with other organelles, Nat. Rev. Mol. Cell Biol. 17 (2016) 69–82, https://doi.org/
1016/j.molcel.2019.03.033 e6. 10.1038/nrm.2015.8.
[71] A. Khaminets, et al., Regulation of endoplasmic reticulum turnover by selective [101] H. Wu, P. Carvalho, G.K. Voeltz, Here, there, and everywhere: the importance of
autophagy, Nature 522 (2015) 354–358, https://doi.org/10.1038/nature14498. ER membrane contact sites, Science (New York, N.Y.) 361 (2018), https://doi.org/
[72] F. Islam, V. Gopalan, A.K. Lam, Retreg1 (FAM134B): a new player in human 10.1126/science.aan5835.
diseases: 15 years after the discovery in cancer, J. Cell. Physiol. 233 (2018) [102] D. Molino, A.C. Nascimbeni, F. Giordano, P. Codogno, E. Morel, ER-driven
4479–4489, https://doi.org/10.1002/jcp.26384. membrane contact sites: Evolutionary conserved machineries for stress response
[73] M.H. Haque, et al., Identification of novel FAM134B (JK1) mutations in oeso- and autophagy regulation? Commun. Integr. Biol. 10 (2017) e1401699, , https://
phageal squamous cell carcinoma, Sci. Rep. 6 (2016) 29173, https://doi.org/10. doi.org/10.1080/19420889.2017.1401699.
1038/srep29173. [103] A.C. Nascimbeni, et al., ER-plasma membrane contact sites contribute to autop-
[74] F. Islam, et al., Promoter hypermethylation inactivate tumor suppressor FAM134B hagosome biogenesis by regulation of local PI3P synthesis, EMBO J. 36 (2017)
and is associated with poor prognosis in colorectal cancer, Genes Chromosomes 2018–2033, https://doi.org/10.15252/embj.201797006.
Cancer 57 (2018) 240–251, https://doi.org/10.1002/gcc.22525. [104] M.I. Molejon, A. Ropolo, A.L. Re, V. Boggio, M.I. Vaccaro, The VMP1-Beclin 1
[75] P. Grumati, et al., Full length RTN3 regulates turnover of tubular endoplasmic interaction regulates autophagy induction, Sci. Rep. 3 (2013) 1055, https://doi.
reticulum via selective autophagy, eLife 6 (2017), https://doi.org/10.7554/eLife. org/10.1038/srep01055.
25555. [105] L.C. Tabara, R. Escalante, VMP1 establishes ER-Microdomains that regulate
[76] F. Fumagalli, et al., Translocon component Sec62 acts in endoplasmic reticulum membrane contact sites and autophagy, PLoS One 11 (2016) e0166499, , https://
turnover during stress recovery, Nat. Cell Biol. 18 (2016) 1173–1184, https://doi. doi.org/10.1371/journal.pone.0166499.
org/10.1038/ncb3423. [106] M.I. Molejon, A. Ropolo, M.I. Vaccaro, VMP1 is a new player in the regulation of
[77] M. Linxweiler, B. Schick, R. Zimmermann, Let’s talk about Secs: Sec61, Sec62 and the autophagy-specific phosphatidylinositol 3-kinase complex activation,
179
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Ishaq, et al. Seminars in Cancer Biology 66 (2020) 171–181
Autophagy 9 (2013) 933–935, https://doi.org/10.4161/auto.24390. [135] X. Xie, J.Y. Koh, S. Price, E. White, J.M. Mehnert, Atg7 overcomes senescence and
[107] Y. Saheki, P. De Camilli, Endoplasmic reticulum-plasma membrane contact sites, promotes growth of BrafV600E-Driven melanoma, Cancer Discov. 5 (2015)
Annu. Rev. Biochem. 86 (2017) 659–684, https://doi.org/10.1146/annurev- 410–423, https://doi.org/10.1158/2159-8290.Cd-14-1473.
biochem-061516-044932. [136] R. Amaravadi, A.C. Kimmelman, E. White, Recent insights into the function of
[108] R. Salvador-Gallego, M.J. Hoyer, G.K. Voeltz, SnapShot: Functions of Endoplasmic autophagy in cancer, Genes Dev. 30 (2016) 1913–1930, https://doi.org/10.1101/
Reticulum Membrane Contact Sites, Cell 171 (2017) 1224, https://doi.org/10. gad.287524.116.
1016/j.cell.2017.11.005 e1221. [137] R. Lock, et al., Autophagy facilitates glycolysis during Ras-mediated oncogenic
[109] M. Doghman-Bouguerra, E. Lalli, ER-mitochondria interactions: both strength and transformation, Mol. Biol. Cell 22 (2011) 165–178, https://doi.org/10.1091/mbc.
weakness within cancer cells, Biochim. Biophys. Acta (BBA) Mol. Cell Res. 1866 E10-06-0500.
(2019) 650–662, https://doi.org/10.1016/j.bbamcr.2019.01.009. [138] A.C. Kimmelman, E. White, Autophagy and tumor metabolism, Cell Metab. 25
[110] Q. Xie, et al., ABT737 reverses cisplatin resistance by regulating ER-mitochondria (2017) 1037–1043, https://doi.org/10.1016/j.cmet.2017.04.004.
Ca2+ signal transduction in human ovarian cancer cells, Int. J. Oncol. 49 (2016) [139] C. Zhou, et al., High glucose microenvironment accelerates tumor growth via
2507–2519, https://doi.org/10.3892/ijo.2016.3733. SREBP1-autophagy axis in pancreatic cancer, J. Exp. Clin. Cancer Res. 38 (2019)
[111] L. Xu, et al., Bcl-2 overexpression reduces cisplatin cytotoxicity by decreasing ER- 302, https://doi.org/10.1186/s13046-019-1288-7.
mitochondrial Ca2+ signaling in SKOV3 cells, Oncology Report 39 (2017) [140] G. Mariño, et al., Regulation of autophagy by cytosolic acetyl-coenzyme A, Mol.
985–992, https://doi.org/10.3892/or.2017.6164. Cell 53 (2014) 710–725, https://doi.org/10.1016/j.molcel.2014.01.016.
[112] T. Di Mattia, C. Tomasetto, F. Alpy, Faraway, so close! functions of endoplasmic [141] J.V. Lee, et al., Acetyl-CoA promotes glioblastoma cell adhesion and migration
reticulum-endosome contacts, Biochim. Biophys. Acta (BBA) Mol. Cell Res. Lipids through Ca2+-NFAT signaling, Genes Dev. 32 (2018) 497–511, https://doi.org/
1865 (2019) 158490, , https://doi.org/10.1016/j.bbalip.2019.06.016. 10.1101/gad.311027.117.
[113] F. Alpy, et al., STARD3 or STARD3NL and VAP form a novel molecular tether [142] A. Yang, et al., Autophagy is critical for pancreatic tumor growth and progression
between late endosomes and the ER, J. Cell Sci. 126 (2013) 5500–5512, https:// in tumors with p53 alterations, Cancer Discov. 4 (2014) 905–913, https://doi.org/
doi.org/10.1242/jcs.139295. 10.1158/2159-8290.CD-14-0362.
[114] T. Di Mattia, et al., Identification of MOSPD2, a novel scaffold for endoplasmic [143] U. Santanam, et al., Atg7 cooperates with Pten loss to drive prostate cancer tumor
reticulum membrane contact sites, EMBO Report. 19 (2018), https://doi.org/10. growth, Genes Dev. 30 (2016) 399–407, https://doi.org/10.1101/gad.274134.
15252/embr.201745453 pii: e45453. 115.
[115] C. Raiborg, E.M. Wenzel, H. Stenmark, ER-endosome contact sites: molecular [144] M. Desai, R. Fang, J. Sun, The role of autophagy in microbial infection and im-
compositions and functions, EMBO J. 34 (2015) 1848–1858, https://doi.org/10. munity, Immunotargets Ther. 4 (2015) 13–26, https://doi.org/10.2147/ITT.
15252/embj.201591481. S76720.
[116] Y.G. Zhao, N. Liu, G. Miao, Y. Chen, H. Zhao, H. Zhang, The ER contact proteins [145] G. Karsli-Uzunbas, et al., Autophagy is required for glucose homeostasis and lung
VAPA/B interact with multiple autophagy proteins to modulate autophagosome tumor maintenance, Cancer Discov. 4 (2014) 914–927, https://doi.org/10.1158/
biogenesis, Curr. Biol. 28 (2018) 1234–1245, https://doi.org/10.1016/j.cub.2018. 2159-8290.Cd-14-0363.
03.002 e4. [146] G. Ghislat, T. Lawrence, Autophagy in dendritic cells, Cell. Mol. Immunol. 15
[117] P. Atakpa, N.B. Thillaiappan, S. Mataragka, D.L. Prole, C.W. Taylor, 3IP receptors (2018) 944–952, https://doi.org/10.1038/cmi.2018.2.
preferentially associate with ER-Lysosome contact sites and selectively deliver [147] M. Das, S.V. Kaveri, J. Bayry, Cross-presentation of antigens by dendritic cells: role
Ca2+ to lysosomes, Cell Report 25 (2018) 3180–3193, https://doi.org/10.1016/j. of autophagy, Oncotarget 6 (2015) 28527–28528, https://doi.org/10.18632/
celrep.2018.11.064 e7. oncotarget.5268.
[118] M. Cojoc, K. Mabert, M.H. Muders, A. Dubrovska, A role for cancer stem cells in [148] H.K. Lee, et al., In vivo requirement for Atg5 in antigen presentation by dendritic
therapy resistance: cellular and molecular mechanisms, Semin. Cancer Biol. 31 cells, Immunity. 32 (2010) 227–239, https://doi.org/10.1016/j.immuni.2009.12.
(2015) 16–27, https://doi.org/10.1016/j.semcancer.2014.06.004. 006.
[119] R. Ojha, S. Bhattacharyya, S.K. Singh, Autophagy in Cancer Stem Cells: A Potential [149] T. Alissafi, et al., Autophagy orchestrates the regulatory program of tumor-asso-
Link Between Chemoresistance, Recurrence, and Metastasis, Biores. Open Access 4 ciated myeloid-derived suppressor cells, J. Clin. Invest. 128 (2018) 3840–3852,
(2015) 97–108, https://doi.org/10.1089/biores.2014.0035. https://doi.org/10.1172/JCI120888.
[120] M. Mortensen, A.S. Watson, A.K. Simon, Lack of autophagy in the hematopoietic [150] M.Z. Noman, G. Berchem, B. Janji, Targeting autophagy blocks melanoma growth
system leads to loss of hematopoietic stem cell function and dysregulated myeloid by bringing natural killer cells to the tumor battlefield, Autophagy 4 (2018)
proliferation, Autophagy 7 (2011) 1069–1070. 730–732, https://doi.org/10.1080/15548627.2018.1427398.
[121] C. Gong, et al., Beclin 1 and autophagy are required for the tumorigenicity of [151] L. DeVorkin, et al., Autophagy regulation of metabolism is required for CD8+ t cell
breast cancer stem-like/progenitor cells, Oncogene 32 (2013) 2261–2272, https:// anti-tumor immunity, Cell Report 9 (2019) 502–513, https://doi.org/10.1016/j.
doi.org/10.1038/onc.2012.252 2272e.2261-2211. celrep.2019.03.037 e5.
[122] M.F. Wei, et al., Autophagy promotes resistance to photodynamic therapy-induced [152] D. Chen, et al., Chloroquine modulates antitumor immune response by resetting
apoptosis selectively in colorectal cancer stem-like cells, Autophagy 10 (2014) tumor-associated macrophages toward M1 phenotype, Nat. Commun. 9 (2018)
1179–1192, https://doi.org/10.4161/auto.28679. 873, https://doi.org/10.1038/s41467-018-03225-9.
[123] N.K. Lytle, A.G. Barber, T. Reya, Stem cell fate in cancer growth, progression and [153] C.A. Clark, H.B. Gupta, T.J. Curiel, Tumor cell-intrinsic CD274/PD-L1: a novel
therapy resistance, Nat. Rev. Cancer 18 (2018) 669–680, https://doi.org/10. metabolic balancing act with clinical potential, Autophagy 13 (2017) 987–988,
1038/s41568-018-0056-x. https://doi.org/10.1080/15548627.2017.1280223.
[124] L. Li, et al., Protective autophagy decreases osimertinib cytotoxicity through [154] M. Robainas, R. Otano, S. Bueno, S. Ait-Oudhia, Understanding the role of PD-L1/
regulation of stem cell-like properties in lung cancer, Cancer Letter 452 (2019) PD1 pathway blockade and autophagy in cancer therapy, Onco. Ther. 10 (2017)
191–202, https://doi.org/10.1016/j.canlet.2019.03.027. 1803–1807, https://doi.org/10.2147/ott.S132508.
[125] Y. Lei, et al., Targeting autophagy in cancer stem cells as an anticancer therapy, [155] Y. Wang, et al., Chloroquine enhances the cytotoxicity of topotecan by inhibiting
Cancer Lett. 393 (2017) 33–39, https://doi.org/10.1016/j.canlet.2017.02.012. autophagy in lung cancer cells, Chin. J. Cancer 30 (2011) 690–700, https://doi.
[126] E. Batlle, H. Clevers, Cancer stem cells revisited, Nat. Med. 23 (2017) 1124–1134, org/10.5732/cjc.011.10056.
https://doi.org/10.1038/nm.4409. [156] P. Pellegrini, et al., Acidic extracellular pH neutralizes the autophagy-inhibiting
[127] R. Ojha, V. Jha, S.K. Singh, Gemcitabine and mitomycin induced autophagy reg- activity of chloroquine: implications for cancer therapies, Autophagy 10 (2014)
ulates cancer stem cell pool in urothelial carcinoma cells, Biochim. Biophys. Acta 562–571, https://doi.org/10.4161/auto.27901.
1863 (2016) 347–359, https://doi.org/10.1016/j.bbamcr.2015.12.002. [157] D. Fu, et al., Imaging the intracellular distribution of tyrosine kinase inhibitors in
[128] D.S. Choi, et al., Chloroquine eliminates cancer stem cells through deregulation of living cells with quantitative hyperspectral stimulated Raman scattering, Nat.
Jak2 and DNMT1, Stem Cells (Dayton, Ohio) 32 (2014) 2309–2323, https://doi. Chem. 6 (2014) 614–622, https://doi.org/10.1038/nchem.1961.
org/10.1002/stem.1746. [158] X. Sui, et al., Autophagy and chemotherapy resistance: a promising therapeutic
[129] P. Pellegrini, et al., Tumor acidosis enhances cytotoxic effects and autophagy in- target for cancer treatment, Cell Death Dis. 4 (2013) e838, https://doi.org/10.
hibition by salinomycin on cancer cell lines and cancer stem cells, Oncotarget 7 1038/cddis.2013.350.
(2016) 35703–35723, https://doi.org/10.18632/oncotarget.9601. [159] R.A. Barnard, et al., Phase I clinical trial and pharmacodynamic evaluation of
[130] W. Yue, et al., Inhibition of the autophagic flux by salinomycin in breast cancer combination hydroxychloroquine and doxorubicin treatment in pet dogs treated
stem-like/progenitor cells interferes with their maintenance, Autophagy 9 (2013) for spontaneously occurring lymphoma, Autophagy 10 (2014) 1415–1425,
714–729, https://doi.org/10.4161/auto.23997. https://doi.org/10.4161/auto.29165.
[131] Lu Lu, Xinkun Shen, Bailong Tao, Chuanchuan Lin, Ke Li, Zhong Luo, Kaiyong Cai, [160] K.L. Cook, et al., Hydroxychloroquine inhibits autophagy to potentiate anti-
The nanoparticle-facilitated autophagy inhibition of cancer stem cells for im- estrogen responsiveness in ER+ breast cancer, Clin. Cancer Res. 20 (2014)
proved chemotherapeutic effects on glioblastomas, J. Mater. Chem. B 12 (2019). 3222–3232, https://doi.org/10.1158/1078-0432.Ccr-13-3227.
[132] M. Takeda, et al., Disruption of endolysosomal RAB5/7 efficiently eliminates [161] H.O. Lee, A. Mustafa, G.R. Hudes, W.D. Kruger, Hydroxychloroquine destabilizes
colorectal Cancer stem cells, Cancer Res. 79 (2019) 1426–1437, https://doi.org/ Phospho-S6 in human renal carcinoma cells, PLoS One 10 (2015) e0131464, ,
10.1158/0008-5472.CAN-18-2192. https://doi.org/10.1371/journal.pone.0131464.
[133] P. Maycotte, K.L. Jones, M.L. Goodall, J. Thorburn, A. Thorburn, Supports breast [162] C.I. Chude, R.K. Amaravadi, Targeting autophagy in Cancer: update on clinical
Cancer stem cell maintenance by regulating IL6 secretion, Mol. Cancer Res. 13 trials and novel inhibitors, Int. J. Mol. Sci. 18 (2017), https://doi.org/10.3390/
(2015) 651–658, https://doi.org/10.1158/1541-7786.MCR-14-0487. ijms18061279.
[134] H. Huang, et al., Reciprocal network between Cancer stem-Like cells and macro- [163] B.M. Wolpin, et al., Phase II and pharmacodynamic study of autophagy inhibition
phages facilitates the progression and androgen deprivation therapy resistance of using hydroxychloroquine in patients with metastatic pancreatic adenocarcinoma,
prostate Cancer, Clin. Cancer Res. 24 (2018) 4612–4626, https://doi.org/10. Oncologist 19 (2014) 637–638, https://doi.org/10.1634/theoncologist.2014-
1158/1078-0432.CCR-18-0461. 0086.
180
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.
M. Ishaq, et al. Seminars in Cancer Biology 66 (2020) 171–181
[164] D.T. Vogl, et al., Combined autophagy and proteasome inhibition: a phase 1 trial https://doi.org/10.1080/15548627.2015.1033601.
of hydroxychloroquine and bortezomib in patients with relapsed/refractory [173] D. Akin, et al., A novel ATG4B antagonist inhibits autophagy and has a negative
myeloma, Autophagy 10 (2014) 1380–1390, https://doi.org/10.4161/auto. impact on osteosarcoma tumors, Autophagy 10 (2014) 2021–2035, https://doi.
29264. org/10.4161/auto.32229.
[165] R. Rangwala, et al., Phase I trial of hydroxychloroquine with dose-intense temo- [174] E. Donohue, et al., Inhibition of autophagosome formation by the benzoporphyrin
zolomide in patients with advanced solid tumors and melanoma, Autophagy 10 derivative verteporfin, J. Biol. Chem. 286 (2011) 7290–7300, https://doi.org/10.
(2014) 1369–1379, https://doi.org/10.4161/auto.29118. 1074/jbc.M110.139915.
[166] R. Rangwala, et al., Combined MTOR and autophagy inhibition: phase I trial of [175] S.T. Nawrocki, et al., The novel autophagy inhibitor ROC-325 augments the an-
hydroxychloroquine and temsirolimus in patients with advanced solid tumors and tileukemic activity of azacitidine, Leukemia (July (29)) (2019), https://doi.org/
melanoma, Autophagy 10 (2014) 1391–1402, https://doi.org/10.4161/auto. 10.1038/s41375-019-0529-2.
29119. [176] R.K. Amaravadi, J.D. Winkler, Lys05: a new lysosomal autophagy inhibitor,
[167] M.R. Rosenfeld, et al., A phase I/II trial of hydroxychloroquine in conjunction with Autophagy 8 (2012) 1383–1384, https://doi.org/10.4161/auto.20958.
radiation therapy and concurrent and adjuvant temozolomide in patients with [177] V.W. Rebecca, et al., A unified approach to targeting the lysosome’s degradative
newly diagnosed glioblastoma multiforme, Autophagy 10 (2014) 1359–1368, and growth signaling roles, Cancer Discov. 7 (2017) 1266–1283, https://doi.org/
https://doi.org/10.4161/auto.28984. 10.1158/2159-8290.Cd-17-0741.
[168] D. Mahalingam, et al., Combined autophagy and HDAC inhibition: a phase I safety, [178] V.W. Rebecca, et al., PPT1 promotes tumor growth and is the molecular target of
tolerability, pharmacokinetic, and pharmacodynamic analysis of hydroxy- chloroquine derivatives in Cancer, Cancer Discov. (2018), https://doi.org/10.
chloroquine in combination with the HDAC inhibitor vorinostat in patients with 1158/2159-8290.Cd-18-0706.
advanced solid tumors, Autophagy 10 (2014) 1403–1414, https://doi.org/10. [179] Y. Kuwahara, et al., Enhancement of autophagy is a potential modality for tumors
4161/auto.29231. refractory to radiotherapy, Cell Death Dis. 2 (2008), https://doi.org/10.1038/
[169] J.M. Llovet, et al., Sorafenib in advanced hepatocellular carcinoma, N. Engl. J. cddis.2011.56 e177.
Med. 359 (2008) 378–390, https://doi.org/10.1056/NEJMoa0708857. [180] K.W. Kim, M. Hwang, L. Moretti, J.J. Jaboin, Y.I. Cha, B. Lu, Autophagy upre-
[170] J.S. Carew, S.T. Nawrocki, Drain the lysosome: Development of the novel orally gulation by inhibitors of caspase-3 and mTOR enhances radiotherapy in a mouse
available autophagy inhibitor ROC-325, Autophagy 13 (2017) 765–766, https:// model of lung cancer, Autophagy 4 (2008) 659–668.
doi.org/10.1080/15548627.2017.1280222. [181] H. Wu, et al., Is the autophagy a friend or foe in the silver nanoparticles associated
[171] A.L. Cheng, et al., Efficacy and safety of sorafenib in patients in the Asia-Pacific radiotherapy for glioma? Biomaterials 62 (2015) 47–57, https://doi.org/10.1016/
region with advanced hepatocellular carcinoma: a phase III randomised, double- j.biomaterials.2015.05.033.
blind, placebo-controlled trial, Lancet Oncol. 10 (2009) 25–34, https://doi.org/ [182] A. David, D.A. Gewirtz, The challenge of developing autophagy inhibition as a
10.1016/s1470-2045(08)70285-7. therapeutic strategy, Cancer Res. 19 (2019) 5610–5614, https://doi.org/10.1158/
[172] B. Pasquier, SAR405, a PIK3C3/Vps34 inhibitor that prevents autophagy and sy- 0008-5472.
nergizes with MTOR inhibition in tumor cells, Autophagy 11 (2015) 725–726,
181
Descargado para Heenry Luis Dávila Gómez ([email protected]) en HS EMEA/LA Infomed de ClinicalKey.es por Elsevier en diciembre 04, 2020.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2020. Elsevier Inc. Todos los derechos reservados.