Open Access
How to translate text using browser tools
30 June 2020 The Roles of Autophagy and Senescence in the Tumor Cell Response to Radiation
Nipa H. Patel, Sahib S. Sohal, Masoud H Manjili, J. Chuck Harrell, David A. Gewirtz
Author Affiliations +
Abstract

Radiation is a critical pillar in cancer therapeutics, exerting its anti-tumor DNA-damaging effects through various direct and indirect mechanisms. Radiation has served as an effective mode of treatment for a number of cancer types, providing both curative and palliative treatment; however, resistance to therapy persists as a fundamental limitation. While cancer cell death is the ideal outcome of any anti-tumor treatment, radiation induces several responses, including apoptotic cell death, mitotic catastrophe, autophagy and senescence, where autophagy and senescence may promote cell survival. In most cases, autophagy, a conventionally cytoprotective mechanism, is a “first” responder to damage incurred from chemotherapy and radiation treatment. The paradigm developed on the premise that autophagy is cytoprotective in nature has provided the rationale for current clinical trials designed with the goal of radiosensitizing cancer cells through the use of autophagy inhibitors; however, these have failed to produce consistent results. Delving further into pre-clinical studies, autophagy has actually been shown to take diverse, sometimes opposing, forms, such as acting in a cytotoxic or nonprotective fashion, which may be partially responsible for the inconsistency of clinical outcomes. Furthermore, autophagy can have both pro- and anti-tumorigenic effects, while also having an important immune modulatory function. Senescence often occurs in tandem with autophagy, which is also the case with radiation. Radiation-induced senescence is frequently followed by a phase of proliferative recovery in a subset of cells and has been proposed as a tumor dormancy model, which can contribute to resistance to therapy and possibly also disease recurrence. Senescence induction is often accompanied by a unique secretory phenotype that can either promote or suppress immune functions, depending on the expression profile of cytokines and chemokines. Novel therapeutics selectively cytotoxic to senescent cells (senolytics) may prove to prolong remission by delaying disease recurrence in patients. Accurate assessment of primary responses to radiation may provide potential targets that can be manipulated for therapeutic benefit to sensitize cancer cells to radiotherapy, while sparing normal tissue.

INTRODUCTION

The effects of radiation are largely mediated through DNA damage that is both direct and indirect, the latter via free radical generation (1). Thus, the efficacy of radiation is partly dependent on the oxygenation of tumors, which is required to generate reactive oxygen species (ROS) and incur damage (2). While cells respond with compensatory mechanisms by antioxidants, such as glutathione and superoxide dismutase (SOD), localization of radiation-induced damage increases ROS levels, tipping redox equilibrium, and ultimately resulting in cell death (3). The effect of radiation-induced damage tends to be delayed, occurring over several cell cycles, resulting in aberrant chromosomes and compromised DNA integrity. Long-term damage to normal cells/tissue at the tumor periphery remains a key issue when injury accumulates in critical organs. The nature of the tumor cell response to radiation can vary. Radiation-induced DNA strand breaks activate a multitude of DNA damage response pathways to prevent propagation of cells carrying the mutated/damaged DNA. The general consensus appears to be that radiation induces a delayed cell death, possibly through mitotic catastrophe, and other direct cell death responses including apoptosis and possibly necrosis. Several cell survival mechanisms are also activated, as alternative cell fates, as the cell attempts to repair damaged DNA and remove injured organelles. Tumor cells exposed to ionizing radiation invariably also undergo autophagy and senescence as possible means to escape cell death. This review attempts to address whether autophagy and senescence contribute to radiation sensitivity and/or refractoriness or resistance to this essential form of cancer therapy. (Fig. 1)

FIG. 1

In response to radiation treatment, tumor cells can upregulate both cell death and cell survival pathways. Whereas apoptotic cell death is the ideal outcome for clinical therapeutic treatment, tumor cells often enter into senescence and autophagy, largely in efforts to evade cell death. However, radiation-induced autophagy can assume different functional roles. Induction of the cytoprotective form of autophagy allows cells to evade apoptotic cell death and prolong survival; however, cytotoxic autophagy can facilitate either apoptotic and/or autophagic cell death. Finally, an alternative form of autophagy that does not appear to influence cell sensitivity to radiotherapy can occur, termed nonprotective autophagy. Senescence often occurs in parallel with autophagy, sharing a number of mechanistic regulators. Radiation-induced senescence allows cells to transiently arrest in efforts to repair damage. Subsequently, tumor cells may undergo apoptotic cell death if the extent of damage is excessive or may overcome the insult, allowing for continued survival. Senescence may also contribute to tumor dormancy, as a subset of senescent cells endure a prolonged growth arrest and regain proliferative capacity. Senescent cells produce a unique secretory phenotype (SASP), allowing for manipulation of the ECM and influencing surrounding cells in the tumor microenvironment (TME). Through the release of specific cytokines and chemokines, autophagy and senescence can play immune-modulatory effects to create either immune-promoting or immune-suppressive microenvironments, thereby contributing to overall tumor survival or clearance. Both autophagy and senescence have cell-autonomous, as well as cell-non-autonomous effects, adding to the complexity of responses and outcomes of clinical radiotherapeutics.

img-z2-1_103.jpg

OCCURRENCE OF APOPTOSIS IN RESPONSE TO RADIATION

Apoptosis is a process of programmed cell death characterized by chromatin condensation, DNA fragmentation, cell shrinkage, membrane blebbing and formation of apoptotic bodies (4). While irradiated tumor cells clearly do undergo apoptotic cell death, the extent of apoptosis tends to be relatively low (5–7). Clinically relevant or even significantly higher doses of radiation induced only ∼20–30% apoptosis in several experimental tumor cell lines, including breast cancer, non-small cell lung cancer (NSCLC) and colorectal cancer (8). For instance, Rodel et al. demonstrated relative levels of apoptosis between 12% and 27% induced in response to 8 Gy in colorectal cell lines with varying radiosensitivity (7). Similarly, Qu et al. reported ∼20–25% apoptosis induction in MCF-7 breast cancer cells and A549 lung cancer cells with 8 Gy (9). In agreement with these data, previously published work performed in our laboratory demonstrated low levels of apoptosis (∼20%) induced in breast, lung, colorectal, and head and neck cell lines when exposed to fractionated radiation exposure (8). It is only at higher levels of irradiation (above 10 Gy) that apoptosis becomes a more pronounced response to radiotherapy (10). This is, of course, relevant to stereotactic radiation treatment, wherein multiple, precisely focused beams of radiation are delivered to patients to achieve higher effective doses to the tumor while minimizing damage to surrounding tissue (1113). With regard to cancer treatment modalities, apoptosis or other forms of cell death are, of necessity, the desired outcomes; however, there are a number of survival mechanisms that cancer cells have employed to evade (apoptotic) cell death. Both autophagy and senescence can allow cancer cells to mitigate or perhaps delay the damage incurred by clinical therapeutic modalities, escape cell death and prolong survival.

OCCURRENCE OF AUTOPHAGY AND SENESCENCE IN RESPONSE TO RADIATION TREATMENT

Although the desired outcome of radiation therapy is tumor cell death by a pathway such as apoptosis, apoptosis is not obligatorily the sole or primary response to radiation treatment. While the effectiveness of clinical radiation therapy in promoting tumor shrinkage may, of necessity, ultimately involve apoptosis, possibly subsequent to mitotic catastrophe, studies in tumor cells in culture clearly indicate that a consistent and uniform initial response to radiation treatment is autophagy (1417). Autophagy is a cellular survival mechanism to maintain homeostasis in response to stress, such as nutrient deprivation or hypoxia (18). In response to radiotherapy, autophagic machinery is upregulated to remove damage incurred by therapy. Studies by Ren et al. (19) utilizing 30 NSCLC patient tissue samples subjected to 2 Gy (a clinically relevant dose), assessed LC3 and SQSTM1/p62, markers of autophagy, by immunohistochemical staining. Of these 30 samples, 26 demonstrated significant upregulation of LC3 and downregulation of SQSTM1/p62, indictive of autophagy induction (15, 1922).

Autophagy is generally considered to be a cytoprotective response to various forms of stress such as chemotherapy and radiotherapy (2325), although there is accumulating evidence that autophagy can also be functionally “nonprotective” (26, 27). Nonprotective autophagy is characterized by a lack of sensitization or protection against a cellular stress or therapeutic agent when autophagy is inhibited; in such cases, autophagy is neither protective nor cytotoxic in response to the therapy (28). The current clinical utilization of autophagy inhibitors to sensitize tumor cells to radiotherapy and chemotherapy relies on the paradigm that autophagy is cytoprotective in nature; however, autophagic function can vary depending on tumor cell type, cytotoxic therapy used and treatment regimen. It is, therefore, necessary to thoroughly understand the nature of the autophagy induced to improve patient outcomes to autophagy-inhibition therapy.

Autophagy is often accompanied by senescence, at least in the case of clinically relevant doses of drugs and radiation (26, 29). Senescence has long been considered to be an irreversible form of growth arrest, although recently published studies by our group, as well as others, have shown that, while senescence is a durable form of growth arrest, tumor cells can ultimately escape from the senescent state and recover self-renewal capacity (6), possibly suggesting that senescence could be a form of tumor dormancy (30). While both autophagy and senescence often occur in parallel in response to therapeutic treatment and share a number of key regulators, such as p53 and mammalian target of rapamycin (mTOR), whether they are independent or interdependent is still not fully understood; it is feasible that a direct relationship will be evident in some systems, and not others, and will occur only with certain forms or inducers of senescence (31, 32).

RADIATION AND AUTOPHAGY

Autophagy can be considered as a “first responder”, a mechanism whereby the cell attempts to reduce the impact of cellular damage and salvage components/nutrients to avoid cell death. Our group, as well as others, have shown that autophagy is induced in response to radiation treatment and chemotherapy (8, 15, 16, 26, 33); however, the function of autophagy induced in response to these various treatment modalities is not predictable. In most of the current literature, autophagy is considered to have a cytoprotective function; consequently, inhibition of cytoprotective autophagy would be anticipated to result in radiosensitization. Early work performed by Chaachouay et al. showed that autophagy inhibition with 3-methyladenine (3-MA) and chloroquine (CQ) radiosensitized MDA-MB-231 (MDA-231) and HBL-100 breast cancer cells (16). Similarly, CQ was also shown to sensitize bladder cancer cells to radiation both in vitro and in vivo, and to promote apoptosis when autophagy inhibition was combined with radiation (34). In studies by Liang et al. examining the role of autophagy in multidrug-resistant ovarian carcinoma, radiation induced relatively low levels of apoptosis; additionally, inhibition of apoptosis with ZVAD did not significantly alter survival or cell death, further confirming that apoptosis is not the primary therapeutic response of radiation, at least in this experimental model (35). These studies also demonstrated higher basal autophagy in the multidrug-resistant phenotype SKVCR cells compared to human SKOV3 ovarian carcinoma cells, suggestive of a cytoprotective function. Moreover, inhibition of autophagy with 3-MA sensitized the multidrug-resistant cells to radiation while having only modest effects on the parental SKOV3 cells. These studies support the concept of autophagy functioning as a cytoprotective mechanism employed by the tumor cells to avert cell death, and that manipulation of these processes could hold therapeutic potential. Ko et al. also demonstrated that genetic inhibition of autophagy radiosensitizes H460 and A549 cells in vitro; however, when moved to an in vivo model of immune-competent mice, autophagy inhibition reduced responses to radiation treatment (36). This observation adds another layer of intricacy to the overall role and contributions of autophagy to tumor cell growth and host immune cell modulation.

Kuwahara et al. utilized radioresistant liver cancer cell lines, which they had previously generated, to better understand the contributions of autophagy towards radioresistance (37). These investigators demonstrated autophagy induction in response to radiation treatment in both the parental HepG2 cells and in the resistant cells (HepG2-8960-R). Furthermore, exposure to rapamycin, an mTOR inhibitor and autophagy inducer, sensitized HepG2-8960-R cells to radiation (10 Gy) but not the parental cell line. Pharmacological and genetic inhibition of autophagy reduced rather than increased sensitivity to acute radiation exposure (2 Gy), suggesting that suppression of cytotoxic autophagy could contribute to radiation resistance. However, it is generally challenging to select for radiation resistance in vitro and consequently there is relatively limited literature relating to autophagy in acquired radiation resistance.

It should be recognized that even if radiation-induced autophagy was solely cytoprotective in function, this cytoprotective form of autophagy could not uniformly represent a mechanism of resistance, since essentially all tumors undergo autophagy in response to radiation treatment and not all tumors present with radiation resistance (a degree of cell death is still observed). One possibility is that some unique (but as yet unidentified) characteristics can determine when radiation-induced cytoprotective autophagy actually confers radiation resistance, rather than simply shifting the dose-response curve for radiation sensitivity. Since the protective function of autophagy is generally considered to be a consequence of interference with apoptosis, radiation resistance could be related to this intrinsic function. A related possibility is that radiation resistance only occurs when the autophagy is durable while a transient cytoprotective outcome would not be capable of conferring actual radioresistance due to the inevitable outcome of apoptotic tumor cell death.

In our previously published studies, we identified the “nonprotective” form of autophagy. As is also the case for cytoprotective autophagy, this is a functional definition, where autophagy is induced in response to radiation treatment (or chemotherapy), but where subsequent inhibition of autophagy fails to alter radiation sensitivity (26). Eng et al. demonstrated that both pharmacological inhibition of autophagy with CQ and Lys01, and genetic inhibition by genome editing of ATG7, did not alter sensitivity to radiation (or 30 different chemotherapies) of KRAS mutant tumors in vitro and in vivo. Furthermore, they were able to show that CQ-mediated sensitization was independent of autophagy, suggesting the antiproliferative effects may be due to modulation of off-target effects (27, 38). Similarly, studies performed by Schaaf et al. demonstrated that radiosensitization effects of CQ, 3-MA and ATG7 deficiency were independent of canonical autophagy pathways and may involve effects on lysosomal degradation (39). Further analyzing these studies by Schaaf et al. (39), pharmacological inhibition with CQ and 3MA did not alter radiosensitivity in MDA-MB-231 breast tumor cells when exposed to 5.6 Gy; however, work done by Chaachouay et al. (16) showed that autophagy inhibition with similar concentrations of CQ and 3MA was sufficient to radiosensitize the same MDA-MB231 cell line exposed up to 5 Gy. These contradictory observations present a conundrum within the field when the same cell line, exposed to similar radiation doses and concentrations of the autophagy inhibitor, can produce two divergent responses, leading to opposing conclusions relating to the role of autophagy. In a seminal article by Michaud et al. (138), autophagy that was induced in colorectal cancer cells by oxaliplatin or mitoxantrone proved to be nonprotective in function, in that silencing of the autophagy gene, ATG7, failed to influence drug sensitivity in the tumor cells in vitro. However, these studies did not include radiation.

Intriguingly, work by Cechakova et al. suggested that Lys05, an autophagy inhibitor, could radiosensitize H1299 (p53-null) cells, suggesting a cytoprotective autophagic function (40). However, in fact, a close examination of data reveals that sensitization, if any, is at best modest, and unlikely to be therapeutically relevant, indicating that the autophagy is likely acting in a nonprotective fashion. This observation actually serves to confirm findings from our own laboratory where we reported radiation-induced nonprotective autophagy in the same H1299 (p53-null) cells, i.e., wherein autophagy inhibition likewise failed to alter radiosensitivity (8).

Massive tumor growth can lead to low oxygen environments, resulting in hypoxia and in nutrient deprivation, common hallmarks of solid tumors, ultimately contributing to cellular damage (41). Cells can upregulate autophagy under such stressful conditions to prevent accumulation of damaged organelles by removing and recycling cellular content (42, 43) Hypoxia often interferes with the effectiveness of anti-cancer therapies. The degree of oxygenation of a tumor can determine the cytotoxicity of radiation; thus, hypoxic environments can contribute to radioresistance (44). Under hypoxic conditions, a number of intracellular pathways are upregulated to allow tumor adaptation, such as that involving hypoxia-inducible factor 1α (HIF-1α). HIF-1α is a major sensor of hypoxic conditions and is activated to allow cells to adapt and survive under low-oxygen environments; moreover, in tumor cells, activation of this pathway acts as a survival mechanism to evade apoptotic cell death and contribute to resistance (44). Hypoxia and HIF-1α can also contribute to autophagy induction, allowing for cells to remove damaged dysfunctional organelles, such as the mitochondria (44, 45). Work by Zhong et al. demonstrated that deletion of HIF-1α in MCF-7 breast cancer cells significantly increased radiosensitivity while decreasing autophagy when irradiated (45). Similarly, Sun et al. showed that under hypoxic conditions, HIF-1α induces autophagy and reduces radiosensitivity in human colorectal cancer cell lines (46). Noman et al. showed that lung carcinoma cells under hypoxic conditions upregulated autophagy to evade CTL-mediated lysis (47). Furthermore, autophagy inhibition was sufficient to reduce tumor growth and increase apoptosis in mice transplanted with B16 melanoma cells. These studies provide evidence to support the conclusion that at least in the case of hypoxia, autophagy induction may, in fact, mediate cell autonomous effects (i.e., directly in the tumor), as well as cell non-autonomous effects, through modulation of the immune system.

Studies in our laboratory examining the functional role of autophagy in radiation sensitivity showed that inhibition of radiation-induced autophagy generally did not alter radiation sensitivity or radiation-induced ROS levels. In fact, it appears that p53 status influenced radiation sensitivity through the promotion of senescence (26). Taken together, the frequent lack of alteration in radiosensitivity exhibited when autophagy inhibition is combined with radiation suggests that autophagy frequently plays a nonprotective function and may not be the primary protective mechanism contributing to loss of radiosensitivity.

In those cases where autophagy takes on functional forms other than being protective, impairment of autophagic functions could shift the cellular fate towards unfavorable outcomes. In the case of cytoprotective autophagy, inhibition of autophagy results in increased cell death (34). In contrast, in cells where autophagy is cytotoxic or cytostatic, autophagy inhibition would be counterproductive, and permissive for tumor cell growth. In the case of cytotoxic autophagy, administration of an inhibitor would result in tumor promotion, by interfering with the original antitumorigenic actions of the autophagy in the cell. For instance, studies performed by Kim et al. demonstrated reduced radiation sensitivity in NSCLC HCC827 cells when Beclin 1, a protein required for the initiation of autophagy, was silenced, when compared to wild-type cells, suggesting that autophagy was cytotoxic in nature. Furthermore, inhibition of mTOR, ultimately resulting in autophagy induction, was sufficient to sensitize these cells to radiation (48).

As indicated above, tumor cells almost uniformly undergo autophagy in response to exogenous forms of stress such as chemotherapy and radiation treatment. Although the majority of the scientific literature tends to consider autophagy as a cytoprotective response to stress and as a mechanism of resistance, this premise is subject to a number of reservations. One is that autophagy is not uniformly cytoprotective; in fact, autophagy can exist in one of four functional forms, only one of which is protective; the other forms are cytotoxic, cytostatic and nonprotective autophagy (49). Consequently, efforts to exploit autophagy inhibition as a therapeutic strategy for radiosensitization (or chemo-sensitization) are unlikely to be successful unless all autophagic responses to radiation treatment, regardless of the tumor type, actually prove to be cytoprotective, which is highly unlikely based on our preclinical studies.

AUTOPHAGY AND TUMOR DORMANCY

Dormancy is traditionally considered to be a state of arrest wherein tumor cells cease to divide but still remain viable until appropriate environmental conditions are introduced to begin proliferation, leading to disease recurrence (50). Recently published work by Vera-Ramirez et al. demonstrated the role of autophagy in sustaining/promoting survival in dormant breast cancer cells. Autophagy inhibition via hydroxychloroquine (HCQ) exposure was sufficient to reduce cellular survival of dormant breast cancer cells and to reduce lung metastases in vivo (51). However, once these cells regained proliferative growth, the effect of HCQ was minimal, suggesting that autophagy could be important for cells in prolonging dormancy. However, it should be acknowledged that HCQ may have multiple off-target effects, raising reservations as to whether the observed effects are actually directly related to autophagy.

The potential involvement of autophagy in tumor dormancy was recently supported by published studies from our group showing that autophagy-deficient tumor cells entered the state of dormancy in the presence of chemotherapy, in vitro, and recovered earlier than autophagy-competent dormant cells (52, 53). Since autophagy is involved in the maintenance of DNA integrity such that autophagy-deficient tumor cells accumulate γ-H2AX foci and genomic damage leading to tumor progression (54), lack of autophagy could result in DNA fragmentation in surviving cells. We showed that a genetic knockdown of ATG5 resulted in the formation of multinuclear dormant tumor cells with higher DNA content in response to chemotherapy, in vitro and in vivo, leading to tumor recovery more rapidly than autophagy-competent dormant cells (52). A recent published review highlighted the paradoxical roles of autophagy in tumor dormancy and tumor progression (55), suggesting that the outcome of autophagy could depend on cellular pathways/proteins that are randomly impacted by autophagy.

RADIATION AND SENESCENCE

Senescence is a prolonged growth arrest generally associated with the induction of DNA damage and consequent signaling, involving induction of p21waf1 (and sometime p16), inhibition of cyclin-dependent kinases, dephosphorylation of Rb and the presumed formation of Rb-E2F complexes (56, 57). Senescence can furthermore be considered as an alternative cell fate which can be induced to allow cells to evade apoptotic cell death (58). Senescent cells present with a myriad of phenotypes, such as morphological changes (enlargement and flattening), expression of a pH6-dependent beta-galactosidase activity, secretion of chemokines and cytokines that encompass the senescence-associated secretory phenotype (SASP), and heterochromatic foci appearance (5961). Previously published studies have quite conclusively demonstrated growth arrest characteristic of senescence in response to radiation treatment (6266). Cui et al. showed that a dose of 4 Gy to cervical cancer cells induced only 16% apoptosis but did induce a long-lasting G2/M-phase arrest (67). Recently reported work from our research group demonstrated that H460 NSCLC cells also exhibit senescence induction in response to radiation treatment and arrest at the G2/M phase (26). In agreement, Luo et al. showed that a 6 Gy dose did not induce significant apoptosis in A549 and H460 cells, but rather induced a premature senescence indicated by increased SA-β-gal staining. Furthermore, knockdown of p53 inhibited radiation-induced senescence, while restoration of p53 expression sensitized cells to radiation and induced senescence (68). Studies performed by Roberson et al. demonstrated induction of accelerated senescence in response to camptothecin, a DNA-damaging agent, in p53-null H2199 human lung cancer cells. Of interest was that these investigators demonstrated that a subset of cells were able to escape the therapy-induced senescence and these cells resembled parental cells while still exhibiting SA-β-gal activity (69). In recently reported work, our group was able to demonstrate that radiation induced a transient growth arrest characterized as senescence, followed by proliferative recovery in NSCLC cells. Moreover, radiation-induced senescence was greater in p53wt H460 NSCLC cells, which were more sensitive to radiation treatment than the p53-null H460 cells (26). These data suggest that p53, an essential tumor suppressor protein commonly mutated in many cancer types, plays a role in mediating senescence induction in response to radiation-related damage, and furthermore, that differential induction of senescence may be a primary contributor to altered radiation sensitivity in various cancers (26, 70).

Aside from its tumor-suppressive functions, senescence may also present as a model of tumor dormancy (30). Damage incurred by the cells from radiation and chemotherapy can induce senescent growth arrest as the cell attempts to repair the damaged DNA. Radiation has been shown to induce a temporary period of growth arrest, followed by a phase of proliferative recovery (26, 69, 71) that could theoretically contribute to disease recurrence.

One limitation to efforts to fully understand the role and contributions of therapy-induced senescence in radiosensitivity is the absence of specific pharmacologic or genetic approaches to silence the senescence response. Nevertheless, our recent evidence of outgrowth/escape from senescence argues for the likelihood that senescence, like autophagy, may be a cytoprotective response that allows the tumor cells to escape elimination by radiation. The prolonged and sustained growth inhibition may be permissive for the ultimate regrowth of the tumor cells and the consequent disease recurrence. Given this possibility, coupled with evidence that the senescence-associated secretory phenotype (SASP) may also promote tumor growth, the recent identification of agents with “senolytic” properties, which promote apoptosis selectively in senescent cells, opens up the possibility of developing a therapeutic strategy for elimination of the residual surviving tumor cells (72, 73).

Work by Yosef et al. (74) demonstrated that human fibroblasts induced into senescence by radiation upregulated anti-apoptotic proteins, BCL-W and BCL-XL, in vivo. Targeting these proteins using a small-molecule inhibitor, ABT-737, was sufficient to eliminate these senescent cells (74). Similarly, Samaraweera et al. (75) showed senescence induction and SASP secretion in both NSCLC cells and head and neck squamous cell carcinoma (HNSCC) cells when exposed to cisplatin or taxanes. Furthermore, administration of panobinostat, an FDC-approved histone deacetylase inhibitor (HDACi), after chemotherapy, suppressed proliferation and induced cell death in both cancer cell types (75). While there are many agents that have been proposed to have properties of senolytics (72, 76), not all of these compounds have been shown to be universally effective; consequently, further work will be required to generate clearer insights as to exactly how these agents act as “senolytics” and why particular agents are effective under certain experimental conditions but not others. Taken together, selectively targeting senescent cells while they are dormant and before they begin to regain proliferative recovery may serve as a therapeutic benefit and prolong patient survival, as well as increasing the delay before disease recurrence.

AUTOPHAGY AND SENESCENCE

Autophagy and senescence are both induced in response to radiation treatment, which may in part be due to the overlapping pathways and shared regulators between these two processes.

Reactive Oxygen Species

A key mechanism by which radiation treatment exerts its indirect cytotoxic effects is through ROS generation. Increased ROS levels can be detrimental to the intracellular environment, resulting in mitochondrial dysfunction, genomic instability and misfolded proteins (77). To mitigate the ROS-mediated stress, autophagy is induced to remove and turn over damaged organelles and proteins (78). Chen et al. demonstrated ROS elevation by low-dose ionizing irradiation after high-dose irradiation promoted autophagy and induction of radioresistance in A549 NSCLC cells (79). Furthermore, treatment with N-acetyl-l-cysteine (NAC), an ROS scavenger, was sufficient to block autophagy and the associated radioresistance. Oxidative stress as well as the direct radiation effects can cause substantial DNA damage within a cell, which may induce a senescent cell cycle growth arrest as the cell attempts to repair the damaged DNA (80). Luo et al. showed that resveratrol enhanced radiosensitivity of H460 and A549 NSCLC cells by ROS-mediated DNA damage which induced a premature senescence (64, 81). ROS-induced DNA damage upregulates and activates a number of important cellular regulators, including p53, which in turn increases p21, a key cell cycle regulator in mediating senescence (82).

TP53

TP53 is an essential tumor suppressor gene, which is commonly aberrant or dysfunctional in multiple tumor cell types. p53 regulates a vast number of cellular processes, including but not limited to apoptosis, autophagy and senescence. p53 mediates the transcription of a number of cell cycle inhibitors, including p21waf1 and p16, which interfere with the interaction between cyclins and cyclin-dependent kinases that induce cell cycle arrest (83). Luo et al. demonstrated that p53 activation using Nutlin-3a radiosensitized H1299 (p53-null) cells by activating p53-p21waf1 pathways and inducing cellular senescence (68). Furthermore, depending on p53 localization, this protein can also mediate autophagy. p53 can activate autophagy by upstream inhibition of mammalian target of rapamycin (mTOR), through direct means such as generation of damage-related autophagy modulators (DRAM), as well as regulating the transcription of key autophagy-related genes (8486).

Mammalian Target of Rapamycin (mTOR)

Mammalian target of rapamycin (mTOR) is an important regulator of both senescence and autophagy. mTOR prevents activation of autophagy initialization; thus, mTOR inhibition has been shown to upregulate autophagy (87). Cao et al. showed that addition of RAD001, a pharmacological mTOR inhibitor, enhanced radiosensitivity of prostate cancer cell lines, which was associated with increased autophagy (88). Studies by Nam et al. demonstrated autophagy activation in response to mTOR inhibition in glioma, lung, colorectal and breast cancer cell lines when exposed to radiation; furthermore, mTOR blockade (which promotes autophagy) resulted in premature senescence and restoration of radiosensitivity (89). Seminal work by Narita et al. showed that mTOR and autophagic machinery may be important in SASP processing during senescence (90, 91). The authors observed a specialized compartment, which they termed the TOR-autophagy spatial coupling compartment (TSACC), where products of cellular catabolic processes, such as autophagic degradation, could feed into cellular anabolic processes, to promote protein synthesis. Disruption of mTOR localization to TSACC was shown to inhibit interleukin-6/8 synthesis in Ras-induced senescence, suggesting that autophagy may play a role in SASP generation, which can reinforce the senescent phenotype.

Recently published studies from our laboratory utilizing multiple cytotoxic therapies and multiple cell lines demonstrated senescence induction and proliferative recovery independent of autophagy when the autophagy is “nonprotective” in function (92). Furthermore, we observed that autophagy inhibition did not alter the extent of senescence induction or recovery in HCT116 colorectal carcinoma cells exposed to 4 Gy. Given that both autophagy and senescence are induced in parallel in response to radiation treatment, and often occur in conjunction, it has been asserted that senescence is dependent on prior autophagy. However, a careful analysis of the literature suggests that while autophagy inhibition can delay the occurrence of senescence, it is unlikely to alter the extent of senescence induction; that is, the promotion of autophagy may merely accelerate the senescence response, but senescence is not autophagy-dependent (71, 93, 94).

BYSTANDER AND ABSCOPAL EFFECT/INVOLVEMENT OF THE IMMUNE SYSTEM IN THE RESPONSE TO RADIATION TREATMENT/DAMPS/SENESCENCE

Autophagy and senescence both can exhibit immune-modulatory effects, both endogenously and exogenously. Endogenously, autophagy is essential for T-cell activation and differentiation in response to environmental insults. Dysregulation of autophagy and an increase of senescence during aging of T cells results in a compromised immune response to pathogens in the elderly (95). Exogenously, through SASP secretion, senescent cells can regulate immune infiltration and clearance of tumor cells, while autophagy contributes to immune cell function and cytokine production (72, 96).

Senescence-Associated Secretory Phenotype

A key characteristic of senescent cells is the senescence-associated secretory phenotype (SASP), which encompasses a unique secretion profile that includes cytokines, matrix metalloproteases (MMPs), growth factors and several other soluble regulators (97). A number of these factors can alter the tumor microenvironment (TME) or promote tumor growth. Additionally, several of these cytokines and soluble factors participate in wound-healing processes that modulate extracellular matrix (ECM) remodeling, tissue repair, surrounding cells in the TME, as well as regulate immune infiltration (98). MMPs and other proteases help to remodel the ECM around tumor cells and can promote tumor migration (99, 100). Moreover, senescent cells secrete a number of pro-inflammatory cytokines and chemokines, such as IL-6 and IL-8, which can play a role in promoting tumor growth and migration. Ortiz-Montero et al. demonstrated that when MCF-7 cells were treated with conditioned media from senescent cells, IL-6 or IL-8, markers of endothelial-to-mesenchymal transition (EMT) were upregulated, and promoted tumor cell migration and invasion (101). Both of these inflammatory factors can modulate the activation of the NF-κB pathway, resulting in an increase in transcription of anti-apoptotic proteins and promoting tumor growth (102). In contrast, secretion of pro-inflammatory cytokines can promote immune infiltration, and depending on the type of immune infiltrates, can promote or inhibit tumor growth. Published studies by Meng et al. utilized radiation and the PARP inhibitor, veliparib, to promote premature senescence in B16SIY melanoma cells (103). When these senescent cells were isolated and injected into C57BL/6 mice, a number of cytokines were upregulated and an increased CD8+ T-cell proliferation was observed in coculture studies. Furthermore, in tumor-bearing mice, vaccination with senescent cells followed by irradiation was sufficient to elicit immune responses and eliminate established tumors. As exemplified, cellular senescence can play a dual role in tumor development itself, and it remains to be determined whether these soluble factors produced by senescent cells promote or antagonize tumorigenesis (104).

Bystander and Abscopal Effect

Given the functional interplay between SASP secretion and immune modulation, it is important to acknowledge the potential contributions of senescence to the abscopal effect. Depending on the soluble factors released by senescent cells, recruitment and promotion of the clonal expansion of a myriad of immune cells, ranging both dichotomies of pro-to anti-tumorigenic immune cells, can occur (72, 105, 106). Work by Xue et al. demonstrated that restoration of p53-rescued senescence induction, activation of the innate immune system and subsequent clearance of tumor cells (107). Furthermore, it has been demonstrated that the SASP released by senescent cells promote a bystander effect both in vitro and in vivo (108, 109), in which secretion can promote premature senescence in surrounding cells, possibly enhancing responses. Ruhland et al., showed that SASP secretion by senescent stromal cells can increase localization and activation of myeloid suppressor cells, creating an immunosuppressive, tumor-permissive environment (110). Reiterating the duality in function (104), SASP secretion can promote either an immune-suppressive effect to promote tumor growth (111) or immune-activating TME, promoting anti-tumor effects, immediately surrounding the tumor; however, whether this may contribute to distal tumor clearance has yet to be determined.

With increasing utilization of immunotherapies as an additive to the current treatment regimen of various cancer types, better understanding and manipulation of the abscopal effect could be of benefit, aiding to activate immune responses and increasing immune clearance in otherwise “cold” tumors.

Tumor immune surveillance could keep nascent transformed cells in check and inhibit tumor formation (112); however, “cold” tumors or weakly immunogenic tumors could escape tumor immune surveillance. Tumors that do not contain infiltrating lymphocytes are termed non-inflamed or immunogenically “cold” tumors, while those with predominant immune cell infiltrates are termed inflamed or immunologically “hot” tumors. Both “cold” tumors and weakly immunogenic tumors could be turned into “hot” or highly immunogenic tumors due to radiation exposure, which could result in inflammation and immunogenic cell death (ICD). Subsequently, tumors undergoing ICD and expressing damage-associated molecular pattern (DAMP) can be detected by antigen-presenting cells (APCs) to induce antitumor immune responses (113). This phenomenon has been characterized as abscopal effects, such that direct irradiation of tumor cells on one side of an experimental animal can result in shrinkage of the tumor on the contralateral site (114) or produce a durable anti-tumor immune response and inhibit tumor progression (115). This abscopal or bystander effect has also occasionally been observed in patients where tumors distant from the original site of irradiation have also undergone reduction (116). It is understood that this is likely to be the consequence of the establishment of anti-tumor immune responses against shared tumor antigens expressed on irradiated tumor and nonirradiated tumor cells. Paradoxically, tumor-derived cytokines/chemokines could increase myeloid-derived suppressor cells (MDSCs) or Tregs, resulting in the inhibition of the anti-tumor immune responses. To this end, the contribution(s) of tumor cell autophagy or senescence in determining the outcome of immunogenic radiation therapy remains elusive.

What has not been extensively explored is whether autophagy or senescence is involved in the abscopal or bystander effects. It is well established that autophagy plays a key role in the phagocytic process, regulating the processing and secretion of inflammatory cytokines, the processing and loading of antigenic peptides, and regulating activation of adaptive immune cells (117121). Moreover, autophagy contributes to the modulation of ICD. In their published studies, Ko et al. were able to show reduced radiosensitivity in autophagy-deficient cells in mouse tumor xenografts in wild-type immune-competent mice (36). This alteration in radiosensitivity could be rescued by recapitulating extracellular ATP in mice transplanted with ATG knockdown cells when compared to shControl transplanted mice treated with extracellular ATP and radiation, suggesting that autophagy contributes to the generation/release of ATP, a well-established DAMP recognized by the immune system. These effects were not evident in parallel experiments performed with mouse tumor xenografts in immunodeficient animals. Collectively, these experiments establish that autophagy can contribute to radiation sensitivity through its cell non-autonomous effects by modulating immune responses.

ARE PATIENT-DERIVED TUMOR XENOGRAFTS AN APPROPRIATE MODEL FOR STUDIES OF RADIATION SENSITIVITY?

As evident in the previous section, cell non-autonomous effects of autophagy and senescence, tumor microenvironment and immune cell regulation all appear to play a role in modulating sensitivity to cancer therapy; thus, the question arises whether current pre-clinical models will reflect clinical outcomes and the translational potential of novel pre-clinical therapeutics. Patient-derived xenografts (PDX) are currently considered the “gold standard” in modeling tumors and their microenvironment, in part because they can reflect the heterogeneity of tumor populations, and capture various host cell types (such as endothelial cells and fibroblasts) that compose the tumor microenvironment (122124). These have become the preferred model to study drug sensitivity in tumors as well as chemotherapy and radiation resistance (125127). PDX models are generally implanted in immunodeficient animals to prevent their rejection. When assessing effects of radiation, PDX models have been preferred to syngeneic ones, as rodents and humans differ significantly in processes governing response to radiation treatment (126). With the goal of simulating human disease response, the primary use of PDXs is justified. In one pioneering study, PDX models of glioblastoma were effectively used to determine whether amplification of EGFR in tumors was related to their radiation sensitivity (128). Further, mouse-specific radiation delivery techniques have been developed that closely mirror systems used clinically. In addition, tools like micro-CT have helped to evaluate the response of murine tumors to radiation (129). It should be noted, however, that for radiation therapy studies, athymic nude mice are the most reliable hosts for PDXs. Other immunodeficient mice are more radiation-sensitive due to altered DNA repair mechanisms resulting in significant changes to the tumor microenvironment (130). As an alternative to using murine hosts, PDXs can also be cultured using 3D model systems to study effects of radiation. This significantly reduces the costs associated with in vivo experiments but has still been shown to produce different data from that acquired in mono-layer culture experiments (130).

Despite the many benefits PDX models provide, their lack of an intact immune system results in limitations, especially as immunotherapies emerge as an additional pillar of therapy and are now frequently being incorporated into treatment regimens. Multiple studies have revealed the effectiveness of combination immunotherapy and stereotactic radiation in treating a variety of mouse lesions, including brain, colon and lung (131133). Although both syngeneic and genetically-engineered mouse models (GEMM) retain functional immune systems, the many unique aspects of GEMMs are now being explored. These models, which allow for the induction of specific mutations that drive tumorigenesis, have shown utility in studying radiation therapy. Specifically, Cre recombinase and FLP-FRT systems allow for the manipulation of multiple genes to study how these differences affect response to radiation treatment.

In GEMM's of non-small cell lung cancer, genetic differences in phenotypically identical cells were shown to be related to a growth delay after irradiation (129). Although PDXs provide an effective platform for mimicking human disease response to radiation treatment, where combination immunotherapies or an understanding of specific tumorigenic mutations is concerned, syngeneic or GEMM models should be utilized for current studies. Once the baseline utility of immune-compromised mice that have been implanted with human immune cells, “humanized NOD scid gamma mice”, has further progressed, these models may be ideal to use for radiation studies.

AUTOPHAGY INHIBITION IN CLINICAL TRIALS OF RADIATION RESPONSE

While there are several pharmacological autophagy inhibitors, the only clinically approved inhibitors are chloroquine and hydroxychloroquine (HCQ) (134). HCQ is an FDA-approved drug for the treatment and prevention of malaria. HCQ is a lysomotropic agent which can also be utilized to block autophagy through its ability to inhibit the fusion of the autophagosome with the lysosome (134). In 2007, phase I/II clinical trials were performed in which HCQ was combined with temozolomide and radiotherapy in patients diagnosed with glioblastoma multiforme (22). Electron microscopy was utilized to show that radiation induced autophagic vesicle formation in patients; however, treatment with HCQ, even at relatively high concentrations, did not sufficiently inhibit autophagy. Higher doses of HCQ are required to adequately inhibit radiation-induced autophagy in patients; however, this may lead to unwanted toxicities (22, 134). Subsequent phase I/II clinical trials were also initiated in 2013 by Susan Short's group (Brazil et al.; University of Leeds, Leeds, UK) in which the combinatorial effects of lower doses of HCQ and short-course radiotherapy were examined in aged patients with high-grade glioma (HGG) (135). HCQ in combination with radiation did not improve patient outcome compared to radiation alone, and in fact, even at the lower doses of HCQ, grade 3–5 toxicities occurred more frequently in patients who received combination treatment compared to radiation alone. Several other clinical trials were initiated to study the effects of autophagy inhibition on chemo- and radiosensitivity; however, these have provided inconsistent results. HCQ as a monotherapy in patients with metastatic pancreatic adenocarcinoma provided inconsistent autophagy inhibition and poor therapeutic efficacy in a phase II trial (136). A clinical trial of HCQ treatment combined with docetaxel in patients with metastatic prostate cancer was terminated early due to a lack of improved efficacy (identifier: NCT00786682). However, a number of clinical trials have been launched utilizing higher doses of HCQ in combination with chemotherapeutics, such as bortezomib, which may show more promising results (137) (identifiers: NCT00568880, NCT01206530, NCT01506973).

SUMMARY

The premise that autophagy confers resistance to various treatment modalities has been the basis for ongoing clinical trials combining chemotherapy (or radiation treatment) with the only FDA-approved agents that have been established to inhibit autophagy (at least in vitro, the antimalarial drugs, chloroquine and hydroxychloroquine). However, there are a number of conceptual and experimental reservations relating to these clinical trials strategies. One is that, as we and others have shown in multiple published studies, autophagy is not uniformly cytoprotective, often exhibiting cytotoxic and nonprotective functions. The other is that autophagy is unlikely to actually function as a mechanism of radiation resistance if, in fact, autophagy routinely occurs in response to radiation treatment in every tumor studied to date. From a directly clinical perspective, it is highly uncertain whether chloroquine and hydroxychloroquine can achieve levels in the circulation and the tumor to actually inhibit autophagy to the extent that radiosensitization might occur. Finally, if and when more efficacious autophagy inhibitors are identified, it is necessary to acknowledge that autophagy also provides homeostatic regulation in normal tissues such as the central nervous system, and therefore autophagy inhibition might represent a double-edged sword that would induce unanticipated and undesirable (intolerable) toxicities.

While autophagy plays a role in initial sensitivity to radiation in tumor cells, accumulating literature has supported the induction of a prolonged growth arrest, characteristic of senescence, as a response to radiation treatment. Furthermore, a subset of these senescent cells is capable of regaining proliferative capacity, a possible contributor to tumor dormancy and disease recurrence. Consequently, senescent growth arrest may provide a significant contribution to radiation resistance and disease reemergence. Although the implications of senescence cannot be truly resolved due to a lack of effective inhibitors of senescence induction, a novel class of drugs has arisen that allow us to further probe this question. Senolytics selectively induce cell death in senescent cells, providing further insights relating to radiation-induced senescence, as well as a novel class of therapeutics to add to the arsenal of cancer chemotherapy. Moreover, given the fact that autophagy and senescence often occur in tandem, it is also important to further consider the therapeutic implications of incorporating autophagy inhibitors and senolytics in combination with radiotherapy as a means of eliminating residual tumor cells, which have evaded cell death.

ACKNOWLEDGMENTS

Work from the laboratory of DG was supported by the Office of the Assistant Secretary of Defense for Health Affairs through the Breast Cancer Research Program (grant no. W81XWH-14-1-0088), National Institutes of Health (grant no. 5T32DA007027 and RO1 CA 239706) and Massey Cancer Support Grant (grant no. P30 CA016059). Additional support was provided by Susan G. Komen® (to JCH, grant no. CCR19608826).

REFERENCES

1.

Baskar R, Dai J, Wenlong N, Yeo R, Yeoh K-W. Biological response of cancer cells to radiation treatment. Front Mol Biosci 2014; 1:24. Google Scholar

2.

Colliez F, Gallez B, Jordan BF. Assessing tumor oxygenation for predicting outcome in radiation oncology: a review of studies correlating tumor hypoxic status and outcome in the preclinical and clinical settings. Front Oncol 2017; 7:10. Google Scholar

3.

Zhang Y, Martin SG. Redox proteins and radiotherapy. Clin Oncol 2014; 26:289–300. Google Scholar

4.

Shinomiya N. New concepts in radiation-induced apoptosis: ‘P remitotic apoptosis’ and'postmitotic apoptosis'. J Cell Mol Med 2001; 5:240–53. Google Scholar

5.

Roninson IB, Broude EV, Chang BD. If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells. Drug Resist Updat 2001; 4:303–13. Google Scholar

6.

Gewirtz DA, Alotaibi M, Yakovlev VA, Povirk LF. Tumor cell recovery from senescence induced by radiation with PARP inhibition. Radiat Res 2016; 186:327–32. Google Scholar

7.

Rodel C, Haas J, Groth A, Grabenbauer GG, Sauer R, Rödel F. Spontaneous and radiation-induced apoptosis in colorectal carcinoma cells with different intrinsic radiosensitivities: Survivin as a radioresistance factor. Int J Radiat Oncol 2003; 55:1341–7. Google Scholar

8.

Chakradeo S, Sharma K, Alhaddad A, Bakhshwin D, Le N, Harada H, et al. Yet another function of p53–the switch that determines whether radiation-induced autophagy will be cytoprotective or nonprotective: implications for autophagy inhibition as a therapeutic strategy. Mol Pharmacol 2015; 87:803–14. Google Scholar

9.

Qu Y, Hu S, Xu X, Wang R, Yu H, Xu J, et al. Nimotuzumab enhances the radiosensitivity of cancer cells in vitro by inhibiting radiation-induced DNA damage repair. PLoS One 2013; 8:e70727. Google Scholar

10.

Lee J-J, Kim BC, Park M-J, Lee Y-S, Kim Y-N, Lee BL, et al. PTEN status switches cell fate between premature senescence and apoptosis in glioma exposed to ionizing radiation. Cell Death Differ 2011; 18:666–77. Google Scholar

11.

Lu X-Q. Stereotactic body radiation therapy systems. In: Gaya A, Mahadevan A, editors. Stereotactic body radiotherapy: A practical guide. London: Springer London; 2015. p. 27–43. Google Scholar

12.

Andrews DW, Scott CB, Sperduto PW, Flanders AE, Gaspar LE, Schell MC, et al. Whole brain radiation therapy with or without stereotactic radiosurgery boost for patients with one to three brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet 2004; 363:1665–72. Google Scholar

13.

Doyen J, Poudenx M, Gal J, Otto J, Guerder C, Naghavi AO, et al. Stereotactic ablative radiotherapy after concomitant chemo-radiotherapy in non-small cell lung cancer: A TITE-CRM phase 1 trial. Radiother Oncol 2018; 127:239–45. Google Scholar

14.

Eriksson D, Stigbrand T. Radiation-induced cell death mechanisms. Tumor Biol. 2010; 31:363–72. Google Scholar

15.

Paglin S, Hollister T, Delohery T, Hackett N, Mcmahill M, Sphicas E, et al. A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles 1. Cancer Res 2001; 61:439–44. Google Scholar

16.

Chaachouay H, Ohneseit P, Toulany M, Kehlbach R, Multhoff G, Rodemann HP. Autophagy contributes to resistance of tumor cells to ionizing radiation. Radiother Oncol 2011; 99:287–92. Google Scholar

17.

Lomonaco SL, Finniss S, Xiang C, DeCarvalho A, Umansky F, Kalkanis SN, et al. The induction of autophagy by gamma-radiation contributes to the radioresistance of glioma stem cells. Int J Cancer 2009; 125:717–22. Google Scholar

18.

Gammoh N, Wilkinson S, Gammoh N, Wilkinson S. Autophagy in cancer biology and therapy. Front Biol 2014; 9:35–50. Google Scholar

19.

Ren J, Liu T, Han Y, Wang Q, Chen Y, Li G, et al. GSK-3beta inhibits autophagy and enhances radiosensitivity in non-small cell lung cancer. Diagn Pathol 2018; 13:33. Google Scholar

20.

Ito H, Daido S, Kanzawa T, Kondo S, Kondo Y. Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int J Oncol 2005; 26:1401–10. Google Scholar

21.

Classen F, Kranz P, Riffkin H, Pompsch M, Wolf A, Göpelt K, et al. Autophagy induced by ionizing radiation promotes cell death over survival in human colorectal cancer cells. Exp Cell Res 2019; 374:29–37. Google Scholar

22.

Rosenfeld MR, Ye X, Supko JG, Desideri S, Grossman SA, Brem S, et al. A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy 2014; 10:1359–68. Google Scholar

23.

Apel A, Herr I, Schwarz H, Rodemann HP, Mayer A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res 2008; 68:1485–94. Google Scholar

24.

Mani J, Vallo S, Rakel S, Antonietti P, Gessler F, Blaheta R, et al. Chemoresistance is associated with increased cytoprotective autophagy and diminished apoptosis in bladder cancer cells treated with the BH3 mimetic (–)-Gossypol (AT-101). BMC Cancer 2015; 15:224. Google Scholar

25.

Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, et al. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest 2007; 117:326–36. Google Scholar

26.

Xu J, Patel NH, Saleh T, Cudjoe EK, Alotaibi M, Wu Y, et al. Differential radiation sensitivity in p53 wild-type and p53-deficient tumor cells associated with senescence but not apoptosis or (nonprotective) autophagy. Radiat Res 2018; 190:538–57. Google Scholar

27.

Eng CH, Wang Z, Tkach D, Toral-Barza L, Ugwonali S, Liu S, et al. Macroautophagy is dispensable for growth of KRAS mutant tumors and chloroquine efficacy. Proc Natl Acad Sci U S A 2016; 113:182–7. Google Scholar

28.

Gewirtz DA. Cytoprotective and nonprotective autophagy in cancer therapy. Autophagy 2013; 9:1263–5. Google Scholar

29.

Huang Y-H, Yang P-M, Chuah Q-Y, Lee Y-J, Hsieh Y-F, Peng C-W, et al. Autophagy promotes radiation-induced senescence but inhibits bystander effects in human breast cancer cells. Autophagy 2014; 10:1212–28. Google Scholar

30.

Saleh T, Tyutyunyk-Massey L, Gewirtz DA. tumor cell escape from therapy-induced senescence as a model of disease recurrence after dormancy. Cancer Res 2019; 79:1044–6. Google Scholar

31.

Gewirtz DA. Autophagy and senescence in cancer therapy. J Cell Physiol 2014; 229:6–9. Google Scholar

32.

Gewirtz DA. Autophagy and senescence. Autophagy 2013; 9:808–12. Google Scholar

33.

Jo GH, Bogler O, Chwae Y-J, Yoo H, Lee SH, Park JB, et al. Radiation-induced autophagy contributes to cell death and induces apoptosis partly in malignant glioma cells. Cancer Res Treat 2015; 47:221–41. Google Scholar

34.

Wang F, Tang J, Li P, Si S, Yu H, Yang X, et al. Chloroquine enhances the radiosensitivity of bladder cancer cells by inhibiting autophagy and activating apoptosis. Cell Physiol Biochem 2018; 45:54–66. Google Scholar

35.

Liang B, Kong D, Liu Y, Liang N, He M, Ma S, et al. Autophagy inhibition plays the synergetic killing roles with radiation in the multi-drug resistant SKVCR ovarian cancer cells. Radiat Oncol 2012; 7:213. Google Scholar

36.

Ko A, Kanehisa A, Martins I, Senovilla L, Chargari C, Dugue D, et al. Autophagy inhibition radiosensitizes in vitro, yet reduces radioresponses in vivo due to deficient immunogenic signalling. Cell Death Differ 2014; 21:92–9. Google Scholar

37.

Kuwahara Y, Oikawa T, Ochiai Y, Roudkenar MH, Fukumoto M, Shimura T, et al. Enhancement of autophagy is a potential modality for tumors refractory to radiotherapy. Cell Death Dis 2011; 2:e177. Google Scholar

38.

Gallagher LE, Radhi OA, Abdullah MO, McCluskey AG, Boyd M, Chan EYW. Lysosomotropism depends on glucose: a chloroquine resistance mechanism. Cell Death Dis 2017; 8:e3014. Google Scholar

39.

Schaaf MBE, Jutten B, Keulers TG, Savelkouls KGM, Peeters HJM, van den Beucken T, et al. Canonical autophagy does not contribute to cellular radioresistance. Radiother Oncol 2015; 114:406–12. Google Scholar

40.

Cechakova L, Ondrej M, Pavlik V, Jost P, Cizkova D, Bezrouk A, et al. A Potent autophagy inhibitor (Lys05) enhances the impact of ionizing radiation on human lung cancer cells H1299. Int J Mol Sci 2019; 20:5881. Google Scholar

41.

Brahimi-Horn MC, Chiche J, Pouysségur J. Hypoxia and cancer. J Mol Med 2007; 85:1301–7. Google Scholar

42.

Song J, Guo X, Xie X, Zhao X, Li D, Deng W, et al. Autophagy in hypoxia protects cancer cells against apoptosis induced by nutrient deprivation through a beclin1-dependent way in hepatocellular carcinoma. J Cell Biochem 2011; 112:3406–20. Google Scholar

43.

Schlie K, Spowart JE, Hughson LRK, Townsend KN, Lum JJ. When cells suffocate: Autophagy in cancer and immune cells under low oxygen. Int J Cell Biol 2011; 2011:1–13. Google Scholar

44.

Zhang M, Qiu Q, Li Z, Sachdeva M, Min H, Cardona DM, et al. HIF-1 alpha regulates the response of primary sarcomas to radiation therapy through a cell autonomous mechanism. Radiat Res 2015; 183:594. Google Scholar

45.

Zhong R, Xu H, Chen G, Zhao G, Gao Y, Liu X, et al. The role of hypoxia-inducible factor-1alpha in radiation-induced autophagic cell death in breast cancer cells. Tumor Biol 2015; 36:7077–83. Google Scholar

46.

Sun Y, Xing X, Liu Q, Wang Z, Xin Y, Zhang P, et al. Hypoxia-induced autophagy reduces radiosensitivity by the HIF-1alpha/miR-210/Bcl-2 pathway in colon cancer cells. Int J Oncol 2015; 46:750–6. Google Scholar

47.

Noman MZ, Janji B, Kaminska B, Van Moer K, Pierson S, Przanowski P, et al. Blocking hypoxia-induced autophagy in tumors restores cytotoxic T-cell activity and promotes regression. Cancer Res 2011; 71:5976–86. Google Scholar

48.

Kim EJ, Jeong J-H, Bae S, Kang S, Kim CH, Lim Y-B. mTOR inhibitors radiosensitize PTEN-deficient non-small-cell lung cancer cells harboring an EGFR activating mutation by inducing autophagy. J Cell Biochem 2013; 114:1248–56. Google Scholar

49.

Gewirtz DA. The four faces of autophagy: Implications for cancer therapy. Cancer Res 2014; 74:647–51. Google Scholar

50.

Endo H, Inoue M. Dormancy in cancer. Cancer Sci 2019; 110:474–80. Google Scholar

51.

Vera-Ramirez L, Vodnala SK, Nini R, Hunter KW, Green JE. Autophagy promotes the survival of dormant breast cancer cells and metastatic tumour recurrence. Nat Commun 2018; 9:1944. Google Scholar

52.

Aqbi HF, Tyutyunyk-Massey L, Keim RC, Butler SE, Thekkudan T, Joshi S, et al. Autophagy-deficient breast cancer shows early tumor recurrence and escape from dormancy. Oncotarget 2018; 9:22113–22. Google Scholar

53.

Payne KK, Keim RC, Graham L, Idowu MO, Wan W, Wang XY, et al. Tumor-reactive immune cells protect against metastatic tumor and induce immunoediting of indolent but not quiescent tumor cells. J Leukoc Biol 2016; 100:625–35. Google Scholar

54.

Vessoni AT, Filippi-Chiela EC, Menck CF, Lenz G. Autophagy and genomic integrity. Cell Death Differ 2013; 20:1444–54. Google Scholar

55.

Flynn ALB, Schiemann WP. Autophagy in breast cancer metastatic dormancy: Tumor suppressing or tumor promoting functions? J Cancer Metastasis Treat 2019;5:43. Google Scholar

56.

Macip S, Igarashi M, Fang L, Chen A, Pan Z, Lee SW, et al. Inhibition of p21-mediated ROS accumulation can rescue p21-induced senescence. EMBO J 2002; 21:2180–8. Google Scholar

57.

Chang B-D, Swift ME, Shen M, Fang J, Broude EV, Roninson IB. Molecular determinants of terminal growth arrest induced in tumor cells by a chemotherapeutic agent. Proc Natl Acad Sci U S A 2002; 99:389–94. Google Scholar

58.

Cerella C, Grandjenette C, Dicato M, Diederich M. Roles of apoptosis and cellular senescence in cancer and aging. Curr Drug Targets 2016; 17:405–15. Google Scholar

59.

Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of cellular senescence. Trends Cell Biol 2018; 28:436–53. Google Scholar

60.

Narita M, Nunez S, Heard E, Narita M, Lin AW, Hearn SA, et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 2003; 113:703–16. Google Scholar

61.

Pare R, Soon PS, Shah A, Lee CS. Differential expression of senescence tumour markers and its implications on survival outcomes of breast cancer patients. PLoS One 2019; 14:e0214604. Google Scholar

62.

Liao E-C, Hsu Y-T, Chuah Q-Y, Lee Y-J, Hu J-Y, Huang T-C, et al. Radiation induces senescence and a bystander effect through metabolic alterations. Cell Death Dis 2014; 5:e1255. Google Scholar

63.

He X, Yang A, McDonald DG, Riemer EC, Vanek KN, Schulte BA, et al. MiR-34a modulates ionizing radiation-induced senescence in lung cancer cells. Oncotarget 2017; 8:69797–807. Google Scholar

64.

Luo H, Wang L, Schulte BA, Yang A, Tang S, Wang GY, et al. Resveratrol enhances ionizing radiation-induced premature senescence in lung cancer cells. Int J Oncol 2013; 43:1999–2006. Google Scholar

65.

Jones KR, Elmore LW, Jackson-Cook C, Demasters G, Povirk LF, Holt SE, et al. p53-Dependent accelerated senescence induced by ionizing radiation in breast tumour cells. Int J Radiat Biol 2005; 81:445–58. Google Scholar

66.

Mirzayans R, Scott A, Cameron M, Murray D. Induction of accelerated senescence by gamma radiation in human solid tumor-derived cell lines expressing wild-type TP53. Radiat Res 2005; 163:53–62. Google Scholar

67.

Cui F, Hou J, Huang C, Sun X, Zeng Y, Cheng H, et al. C-Myc regulates radiation-induced G2/M cell cycle arrest and cell death in human cervical cancer cells. J Obstet Gynaecol Res 2017; 43:729–35. Google Scholar

68.

Luo H, Yount C, Lang H, Yang A, Riemer EC, Lyons K, et al. Activation of p53 with Nutlin-3a radiosensitizes lung cancer cells via enhancing radiation-induced premature senescence. Lung Cancer 2013; 81:167–73. Google Scholar

69.

Roberson RS, Kussick SJ, Vallieres E, Chen S-YJ, Wu DY. Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers. Cancer Res 2005; 65:2795–803. Google Scholar

70.

Lehmann BD, McCubrey JA, Jefferson HS, Paine MS, Chappell WH, Terrian DM. A dominant role for p53-dependent cellular senescence in radiosensitization of human prostate cancer cells. Cell Cycle 2007; 6:595–605. Google Scholar

71.

Alotaibi M, Sharma K, Saleh T, Povirk LF, Hendrickson EA, Gewirtz DA. Radiosensitization by PARP inhibition in DNA repair proficient and deficient tumor cells: Proliferative recovery in senescent cells. Radiat Res 2016; 185:229–45. Google Scholar

72.

Saleh T, Tyutynuk-Massey L, Cudjoe EK, Idowu MO, Landry JW, Gewirtz DA. Non-cell autonomous effects of the senescence-associated secretory phenotype in cancer therapy. Front Oncol 2018; 8:164. Google Scholar

73.

Short S, Fielder E, Miwa S, von Zglinicki T. Senolytics and senostatics as adjuvant tumour therapy. EBioMedicine 2019; 41:683–92. Google Scholar

74.

Yosef R, Pilpel N, Tokarsky-Amiel R, Biran A, Ovadya Y, Cohen S, et al. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat Commun 2016; 7:11190. Google Scholar

75.

Samaraweera L, Adomako A, Rodriguez-Gabin A, McDaid HM. A novel indication for panobinostat as a senolytic drug in NSCLC and HNSCC. Sci Rep 2017; 7:1900. Google Scholar

76.

Malavolta M, Bracci M, Santarelli L, Sayeed MA, Pierpaoli E, Giacconi R, et al. Inducers of senescence, toxic compounds, and senolytics: The multiple faces of Nrf2-activating phytochemicals in cancer adjuvant therapy. Mediators Inflamm 2018; 2018:1–32. Google Scholar

77.

Pelicano H, Carney D, Huang P. ROS stress in cancer cells and therapeutic implications. Drug Resist Updat 2004; 7:97–110. Google Scholar

78.

Azad MB, Chen Y, Gibson SB. Regulation of autophagy by reactive oxygen species (ROS): Implications for cancer progression and treatment. Antioxid Redox Signal 2009; 11:777–90. Google Scholar

79.

Chen N, Wu L, Yuan H, Wang J. ROS/autophagy/Nrf2 pathway mediated low-dose radiation induced radio-resistance in human lung adenocarcinoma A549 cell. Int J Biol Sci 2015; 11:833–44. Google Scholar

80.

Sabin RJ, Anderson RM. Cellular senescence - its role in cancer and the response to ionizing radiation. Genome Integr 2011; 2:7. Google Scholar

81.

Luo H, Yang A, Schulte BA, Wargovich MJ, Wang GY. Resveratrol induces premature senescence in lung cancer cells via ROS-mediated DNA damage. PLoS One 2013; 8:e60065. Google Scholar

82.

Fitzgerald AL, Osman AA, Xie T-X, Patel A, Skinner H, Sandulache V, et al. Reactive oxygen species and p21Waf1/Cip1 are both essential for p53-mediated senescence of head and neck cancer cells. Cell Death Dis 2015; 6:e1678. Google Scholar

83.

Artuso M, Esteve A, Bresil H, Vuillaume M, Hall J. The role of the Ataxia telangiectasia gene in the p53, WAF1/CIP1(p21)- and GADD45-mediated response to DNA damage produced by ionising radiation. Oncogene 1995; 11:1427–35. Google Scholar

84.

White E. Autophagy and p53. Cold Spring Harb Perspect Med 2016; 6:a026120. Google Scholar

85.

Crighton D, Wilkinson S, O'Prey J, Syed N, Smith P, Harrison PR, et al. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 2006; 126:121–34. Google Scholar

86.

Cui L, Song Z, Liang B, Jia L, Ma S, Liu X. Radiation induces autophagic cell death via the p53/DRAM signaling pathway in breast cancer cells. Oncol Rep 2016; 35:3639–47. Google Scholar

87.

Paquette M, El-Houjeiri L, Pause A. mTOR pathways in cancer and autophagy. Cancers (Basel) 2018; 10:18. Google Scholar

88.

Cao C, Subhawong T, Albert JM, Kim KW, Geng L, Sekhar KR, et al. Inhibition of mammalian target of rapamycin or apoptotic pathway induces autophagy and radiosensitizes PTEN null prostate cancer cells. Cancer Res 2006; 66:10040–7. Google Scholar

89.

Nam HY, Han MW, Chang HW, Lee YS, Lee M, Lee HJ, et al. Radioresistant cancer cells can be conditioned to enter senescence by mTOR inhibition. Cancer Res 2013; 73:4267–77. Google Scholar

90.

Narita M, Young ARJ, Arakawa S, Samarajiwa SA, Nakashima T, Yoshida S, et al. Spatial coupling of mTOR and autophagy augments secretory phenotypes. Science 2011; 332:966–70. Google Scholar

91.

Young ARJ, Narita M, Narita M. Spatio-temporal association between mTOR and autophagy during cellular senescence. Autophagy 2011; 7:1387–8. Google Scholar

92.

Saleh T, Tyutyunyk-Massey L, Patel NH, Cudjoe EK Jr , Alotaibi M, Gewirtz DA. Studies of non-protective autophagy provide evidence that recovery from therapy-induced senescence is independent of early autophagy. Int J Mol Sci 2020; 21:1427. Google Scholar

93.

Narita M, Young ARJ, Narita M. Autophagy facilitates oncogene-induced senescence. Autophagy 2009; 5:1046–7. Google Scholar

94.

Young ARJ, Narita M, Ferreira M, Kirschner K, Sadaie M, Darot JFJ, et al. Autophagy mediates the mitotic senescence transition. Genes Dev 2009; 23:798–803. Google Scholar

95.

Macian F. Autophagy in T cell function and aging. Front Cell Dev Biol 2019; 7:213. Google Scholar

96.

Jiang G-M, Tan Y, Wang H, Peng L, Chen H-T, Meng X-J, et al. The relationship between autophagy and the immune system and its applications for tumor immunotherapy. Mol Cancer 2019; 18:17. Google Scholar

97.

Coppe J-P, Desprez P-Y, Krtolica A, Campisi J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu Rev Pathol 2010; 5:99–118. Google Scholar

98.

Ritschka B, Storer M, Mas A, Heinzmann F, Ortells MC, Morton JP, et al. The senescence-associated secretory phenotype induces cellular plasticity and tissue regeneration. Genes Dev 2017; 31:172–83. Google Scholar

99.

Hassona Y, Cirillo N, Heesom K, Parkinson EK, Prime SS. Senescent cancer-associated fibroblasts secrete active MMP-2 that promotes keratinocyte dis-cohesion and invasion. Br J Cancer 2014; 111:1230–7. Google Scholar

100.

Webb AH, Gao BT, Goldsmith ZK, Irvine AS, Saleh N, Lee RP, et al. Inhibition of MMP-2 and MMP-9 decreases cellular migration, and angiogenesis in in vitro models of retinoblastoma. BMC Cancer 2017; 17:434. Google Scholar

101.

Ortiz-Montero P, Londoño-Vallejo A, Vernot J-P. Senescence-associated IL-6 and IL-8 cytokines induce a self- and cross-reinforced senescence/inflammatory milieu strengthening tumorigenic capabilities in the MCF-7 breast cancer cell line. Cell Commun Signal 2017; 15:17. Google Scholar

102.

Korkaya H, Liu S, Wicha MS. Regulation of cancer stem cells by cytokine networks: Attacking Cancer's inflammatory roots. Clin Cancer Res 2011; 17:6125–9. Google Scholar

103.

Meng Y, Efimova E V, Hamzeh KW, Darga TE, Mauceri HJ, Fu Y-X, et al. Radiation-inducible immunotherapy for cancer: senescent tumor cells as a cancer vaccine. Mol Ther 2012; 20:1046–55. Google Scholar

104.

Schosserer M, Grillari J, Breitenbach M. The dual role of cellular senescence in developing tumors and their response to cancer therapy. Front Oncol 2017; 7:278. Google Scholar

105.

Sagiv A, Krizhanovsky V. Immunosurveillance of senescent cells: the bright side of the senescence program. Biogerontology 2013; 14:617–28. Google Scholar

106.

Burton DGA, Stolzing A. Cellular senescence: Immunosurveillance and future immunotherapy. Ageing Res Rev 2018; 43:17–25. Google Scholar

107.

Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizhanovsky V, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 2007; 445:656–60. Google Scholar

108.

Da Silva PFL, Ogrodnik M, Kucheryavenko O, Glibert J, Miwa | Satomi, Cameron K, et al. The bystander effect contributes to the accumulation of senescent cells in vivo. Aging Cell 2019; 18:e12848. Google Scholar

109.

Di X, Bright AT, Bellott R, Gaskins E, Robert J, Holt S, et al. A chemotherapy-associated senescence bystander effect in breast cancer cells. Cancer Biol Ther 2008; 7:864–72. Google Scholar

110.

Ruhland MK, Loza AJ, Capietto A-H, Luo X, Knolhoff BL, Flanagan KC, et al. Stromal senescence establishes an immunosuppressive microenvironment that drives tumorigenesis. Nat Commun 2016; 7:11762. Google Scholar

111.

Demaria M, O'Leary MN, Chang J, Shao L, Liu S, Alimirah F, et al. cellular senescence promotes adverse effects of chemotherapy and cancer relapse. Cancer Discov 2017; 7:165–76. Google Scholar

112.

Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases–elimination, equilibrium and escape. Curr Opin Immunol 2014; 27:16–25. Google Scholar

113.

Deng L, Liang H, Xu M, Yang X, Burnette B, Arina A, et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type i interferon-dependent antitumor immunity in immunogenic tumors. Immunity 2014; 41:843–52. Google Scholar

114.

Rodriguez-Ruiz ME, Rodriguez I, Garasa S, Barbes B, Solorzano JL, Perez-Gracia JL, et al. Abscopal effects of radiotherapy are enhanced by combined immunostimulatory mAbs and are dependent on CD8 T cells and crosspriming. Cancer Res 2016; 76:5994–6005. Google Scholar

115.

Filatenkov A, Baker J, Mueller AMS, Kenkel J, Ahn G-O, Dutt S, et al. Ablative tumor radiation can change the tumor immune cell microenvironment to induce durable complete remissions. Clin Cancer Res 2015; 21:3727–39. Google Scholar

116.

Azami A, Suzuki N, Azami Y, Seto I, Sato A, Takano Y, et al. Abscopal effect following radiation monotherapy in breast cancer: A case report. Mol Clin Oncol 2018; 9:283–6. Google Scholar

117.

Levine B, Deretic V. Unveiling the roles of autophagy in innate and adaptive immunity. Nat Rev Immunol 2007; 7:767–77. Google Scholar

118.

Schmid D, Münz C. Innate and adaptive immunity through autophagy. Immunity 2007; 27:11–21. Google Scholar

119.

Chemali M, Radtke K, Desjardins M, English L. Alternative pathways for MHC class I presentation: a new function for autophagy. Cell Mol Life Sci 2011; 68:1533–41. Google Scholar

120.

Xu Y, Jagannath C, Liu X-D, Sharafkhaneh A, Kolodziejska KE, Eissa NT. Toll-like receptor 4 is a sensor for autophagy associated with innate immunity. Immunity 2007; 27:135–44. Google Scholar

121.

Li Y, Wang L-X, Yang G, Hao F, Urba WJ, Hu H-M. Efficient cross-presentation depends on autophagy in tumor cells. Cancer Res 2008; 68:6889–95. Google Scholar

122.

Hidalgo M, Amant F, Biankin AV., Budinska E, Byrne AT, Caldas C, et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov 2014; 4:998–1013. Google Scholar

123.

Jung J, Lee CH, Seol HS, Choi YS, Kim E, Lee EJ, et al. Generation and molecular characterization of pancreatic cancer patient-derived xenografts reveals their heterologous nature. Oncotarget 2016; 7:62533–46. Google Scholar

124.

Cassidy JW, Caldas C, Bruna A. Maintaining tumor heterogeneity in patient-derived tumor xenografts. Cancer Res 2015; 75:2963–8. Google Scholar

125.

Koga Y, Ochiai A. Systematic review of patient-derived xenograft models for preclinical studies of anti-cancer drugs in solid tumors. Cells 2019; 8:418. Google Scholar

126.

Banuelos CA, Banath JP, MacPhail SH, Zhao J, Eaves CA, O'Connor MD, et al. Mouse but not human embryonic stem cells are deficient in rejoining of ionizing radiation-induced DNA double-strand breaks. DNA Repair (Amst) 2008; 7:1471–83. Google Scholar

127.

Turner TH, Alzubi MA, Sohal SS, Olex AL, Dozmorov MG, Harrell JC. Characterizing the efficacy of cancer therapeutics in patient-derived xenograft models of metastatic breast cancer. Breast Cancer Res Treat 2018; 170:221–34. Google Scholar

128.

Sarkaria JN, Carlson BL, Schroeder MA, Grogan P, Brown PD, Giannini C, et al. Use of an orthotopic xenograft model for assessing the effect of epidermal growth factor receptor amplification on glioblastoma radiation response. Clin Cancer Res 2006; 12:2264–71. Google Scholar

129.

Castle KD, Chen M, Wisdom AJ, Kirsch DG. Genetically engineered mouse models for studying radiation biology. Transl Cancer Res 2017; 6:S900–13. Google Scholar

130.

Willey CD, Gilbert AN, Anderson JC, Gillespie GY. Patient-derived xenografts as a model system for radiation research. Semin Radiat Oncol 2015; 25:273–80. Google Scholar

131.

Wang Y, Deng W, Li N, Neri S, Sharma A, Jiang W, et al. Combining immunotherapy and radiotherapy for cancer treatment: Current challenges and future directions. Front Pharmacol 2018; 9:185. Google Scholar

132.

Demaria S, Coleman CN, Formenti SC. Radiotherapy: Changing the game in immunotherapy. Trends Cancer 2016; 2:286–94. Google Scholar

133.

Son CH, Fleming GF, Moroney JW. Potential role of radiation therapy in augmenting the activity of immunotherapy for gynecologic cancers. Cancer Manag Res 2017; 9:553–63. Google Scholar

134.

Chude CI, Amaravadi RK, Chude CI, Amaravadi RK. Targeting autophagy in cancer: Update on clinical trials and novel inhibitors. Int J Mol Sci 2017; 18:1279. Google Scholar

135.

Brazil L, Swampillai A, Mak K, Hackshaw A, Edwards D, Mesiri P, et al. P01.072 hydroxychloroquine and short course radiotherapy for elderly patients with glioma: a randomised study. Neuro Oncol 2018; 20:iii246. Google Scholar

136.

Wolpin BM, Rubinson DA, Wang X, Chan JA, Cleary JM, Enzinger PC, et al. Phase II and pharmacodynamic study of autophagy inhibition using hydroxychloroquine in patients with metastatic pancreatic adenocarcinoma. Oncologist 2014; 19:637–8. Google Scholar

137.

Vogl DT, Stadtmauer EA, Tan K-S, Heitjan DF, Davis LE, Pontiggia L, et al. Combined autophagy and proteasome inhibition: A phase 1 trial of hydroxychloroquine and bortezomib in patients with relapsed/refractory myeloma. Autophagy 2014; 10:1380–90. Google Scholar

138.

Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 2011; 334:1573–7. Google Scholar
©2020 by Radiation Research Society. All rights of reproduction in any form reserved.
Nipa H. Patel, Sahib S. Sohal, Masoud H Manjili, J. Chuck Harrell, and David A. Gewirtz "The Roles of Autophagy and Senescence in the Tumor Cell Response to Radiation," Radiation Research 194(2), 103-115, (30 June 2020). https://doi.org/10.1667/RADE-20-00009
Received: 20 December 2019; Accepted: 15 May 2020; Published: 30 June 2020
Back to Top