Abstract
Ovarian cancer is a highly aggressive disease with poor survival rates in part due to diagnosis after dissemination throughout the peritoneal cavity. It is well-known that inflammatory signals affect ovarian cancer dissemination. Inflammation is a hallmark of cellular senescence, a stable cell cycle arrest induced by a variety of stimuli including many of the therapies used to treat patients with ovarian cancer. Indeed, recent work has illustrated that ovarian cancer cells in vitro, mouse models, and patient tumors undergo senescence in response to platinum-based or poly(ADP-ribose) polymerase (PARP) inhibitor therapies, standard-of-care therapies for ovarian cancer. This inflammatory response, termed the senescence-associated secretory phenotype (SASP), is highly dynamic and has pleiotropic roles that can be both beneficial and detrimental in cell-intrinsic and cell-extrinsic ways. Recent data on other cancer types suggest that the SASP promotes metastasis. Here, we outline what is known about the SASP in ovarian cancer and discuss both how the SASP may promote ovarian cancer dissemination and strategies to mitigate the effects of the SASP.
Keywords: disease progression, metastasis, microenvironment, secretome, therapy
INTRODUCTION
Ovarian cancer is the deadliest gynecological cancer for women worldwide (1). Approximately 19,880 new ovarian cancer diagnoses and 12,810 deaths are estimated to occur in 2022 in the United States alone (2). Ovarian cancer incidence and mortality have decreased by ∼30% since the 1970s due to improved understanding of the disease and new therapies (1). Due to a lack of screening methods and nonspecific symptoms, over 70% of ovarian cancers are diagnosed at stage III/IV when the tumors have already disseminated throughout the peritoneal cavity, and these patients have a 5-year survival rate of less than 30%. The low survival rates of metastatic ovarian cancer highlight the importance of understanding underlying mechanisms of dissemination.
Ovarian cancer therapies such as platinum-based agents and poly(ADP) ribose polymerase (PARP) inhibitors induce cellular senescence (3, 4), a stable cell cycle arrest (5). Although initially thought to be tumor suppressive, recent work has demonstrated that senescence is more context-dependent and may be protumorigenic in part through the paracrine effects of the senescence-associated secretory phenotype (SASP) (6). The SASP is composed of proinflammatory cytokines and chemokines, proteases, growth factors, bioactive lipids, and extracellular vesicles that contain a variety of cargo. Acutely, the SASP may be beneficial by promoting immunosurveillance, clearance of senescent cells, and immunotherapy sensitization (4, 7, 8). However, the long-term chronic effects of senescence and the SASP may be detrimental. For instance, the SASP can promote metastasis and invasion, chemoresistance and relapse, epithelial-to-mesenchymal transition, angiogenesis, and stemness, which are all directly linked to cancer initiation, progress, and aggressiveness (6). In addition, age correlates with ovarian cancer dissemination (9), and senescent cells accumulate in aged tissues (10). However, in the context of ovarian cancer, little is known about the mechanisms underlying how the SASP promotes dissemination. Here, we will discuss SASP regulators and effectors that have been implicated in ovarian cancer dissemination and further speculate how modulation of the SASP and SASP factors may have implications in ovarian cancer therapy.
OVARIAN CANCER DISSEMINATION
Ovarian cancer is distinct from other cancers in that its primary mechanism of metastasis is through transcoelomic metastasis, where cells are shed from the primary tumor site into the peritoneal cavity, rather than by the hematogenous route (11, 12). In transcoelomic metastasis of ovarian cancer, which we will simply call dissemination, cells overcome anoikis and undergo an epithelial-to-mesenchymal transition (EMT) to release from the primary ovarian tumor. Metastatic ascites, a fluid that accumulates in the peritoneal cavity, aids in the dissemination of ovarian cancer cells either as single cells or spheroids, which have cancer stem cell-like properties. These cells are highly chemoresistant and able to evade immune detection. Ultimately, cells seed on organs within the peritoneum, with the omentum being the most common site of dissemination. Dissemination is a complex process that involves dynamic changes in cell-intrinsic and cell-extrinsic signaling, suggesting that the microenvironment is critical toward this process. One underappreciated aspect of ovarian cancer dissemination is the contribution of the senescence-associated secretory phenotype (SASP). Herein, we will discuss the SASP, what is known about its regulation, how it may promote ovarian cancer dissemination, and potential therapeutic strategies to target this process.
SENESCENCE-ASSOCIATED SECRETORY PHENOTYPE
The SASP is a complex secretome of senescent cells that presumably evolved to trigger tissue remodeling during development or in response to DNA damage to promote clearance of these cells (13). It is a highly dynamic process whose composition and function vary in a context-dependent manner based on factors such as senescence inducer, cell type, stage of senescence, and tissue context. The SASP is typically composed of proinflammatory cytokines and chemokines, proteases, growth factors, bioactive lipids, and extracellular vesicles that can contain a variety of factors (6). Studies in other cancers have found that the SASP can promote metastasis (6). While there is currently little knowledge on how the SASP promotes dissemination in ovarian cancer, individual SASP-like components have been shown to directly influence tumor dissemination by multiple mechanisms such as resistance to anoikis, promotion of cancer stem cell phenotypes, chemotaxis and invasion, and extracellular matrix (ECM) remodeling (11). We will discuss each in more detail in the following section.
The SASP is regulated at multiple levels. The most well-described upstream regulator of the SASP is cytoplasmic double-stranded DNA [dsDNA; also termed cytoplasmic chromatin fragments (CCFs)] (14). Cytoplasmic dsDNA is sensed by cyclic GMP-AMP synthase (cGAS), which activates the stimulator of interferon genes (STING) pathway. Downstream of cytoplasmic dsDNA, regulation of the SASP occurs at the level of transcription, translation, and secretion. cGAS-STING activates NF-κB and C/EBPβ, two of the main transcription factors of SASP genes (6). In addition, high mobility group box (HMGB) proteins such as HMGB1 and HMGB2 can regulate the expression of SASP genes by modifying chromatin architecture (15). Other important transcriptional regulators of the SASP are p53, p16, and RB (16–18). Furthermore, epigenetic regulators such as enhancers and multiple active histone marks have been shown to transcriptionally upregulate SASP factors (15). At the translational level, mTOR translationally controls the SASP in multiple ways, and inhibition of mTOR blunts the SASP (19). Here we will highlight the relevance of these regulators in ovarian cancer dissemination and discuss targeting these pathways.
OVARIAN CANCER DISSEMINATION IS AFFECTED BY SASP-LIKE FACTORS
Senescence in Ovarian Cancer
Platinum-based therapies have been the standard-of-care for patients with ovarian cancer for decades (20); the emerging therapies for patients with ovarian cancer are poly(ADP-ribose) polymerase (PARP) inhibitors (21). Recent reports in both ovarian cancer cell lines and patient tumors demonstrate that senescence and upregulation of the SASP occur in response to these therapies (3, 4, 22, 23). In addition, senescent cells accumulate during aging (10), and aging is an independent prognostic factor for ovarian cancer (24). Together, these data suggest that senescence induction in ovarian tumors is likely due to two sources: therapies used to treat ovarian cancer and aging.
Interestingly, senescence has been shown to be both beneficial and detrimental in ovarian cancer. Induction of senescence and presence of the SASP increases ovarian cancer survival (23) and correlates with immune cell infiltration and response to immunotherapy (4, 7). On the other hand, higher senescent cell burden corresponds to increased malignant ascites of patients with ovarian cancer (25), and age accelerates tumorigenesis and dissemination in mouse models of ovarian cancer (26), although this study did not directly assess senescence. Together, these data suggest that senescence may play an important role in ovarian cancer initiation, progression, and response therapy. However, experimental evidence is just beginning to emerge about how the SASP promotes dissemination and other phenotypes in ovarian cancer. Many papers on other cancer types have demonstrated that aging, cellular senescence, and systemic SASP may contribute to metastasis and other protumorigenic phenotypes (6), which are relevant to ovarian cancer. Here, we will discuss SASP regulators and effectors that have been implicated in ovarian cancer dissemination (Fig. 1 and Table 1). One caveat to note is that while it is possible that SASP regulators and effectors are important for ovarian cancer, they are not necessarily related to the SASP or senescence. This will need to be empirically explored. We will also further speculate how modulation of the SASP and SASP factors may have implications in ovarian cancer therapy.
Table 1.
SASP Regulator | Role in Promoting SASP | Potential Role in Ovarian Cancer Dissemination | Therapeutic Target in Ovarian Cancer? | Therapeutic Agents (*in Clinical Trials; #in Ovarian Cancer) |
---|---|---|---|---|
cGAS-STING | Sensor of cytoplasmic dsDNA and promotion of downstream transcription via NF-κB (14) | Decreased ascites (27) | Upregulation increases response to immunotherapy (7, 28) | Antagonists: |
C-170 | ||||
C-171 | ||||
C-176 | ||||
H-151 | ||||
SN-011 | ||||
Agonists: | ||||
GSK3745417* | ||||
E7766* | ||||
MIW815* | ||||
MSA-2 | ||||
SNX281# | ||||
TAK-500* | ||||
TAK-676* | ||||
p53 | Transcriptional repressor (17) | Mutant p53 increases cytokine expression (29) and has more dissemination potential (30) | ||
p16/RB1 | Loss leads to transcriptional repression (16, 18) | Unknown, but patients with RB low ovarian cancer are extremely long-lived (31) | ||
NF-κB | Transcriptional activator (6) | Multiple roles in EMT, stemness, angiogenesis (32) | Inhibition affects multiple pathways (32) | Many agents [reviewed in (33)] |
C/EBPβ | Transcriptional activator (6) | Linked to a transcriptional hub including inflammatory genes that are a part of the SASP (34) and promotes migration through DAXX (35) | Helenalin acetate | |
DOT1L | Transcriptional upregulation through H3K79 methylation (36) | Downstream of C/EBP-β (40) | EPZ-5676* | |
SCG 0946 | ||||
SYC-522 | ||||
Brd4 | Transcriptional upregulation through enhancers (8) | Promotes EMT (37) | Inhibition decreases dissemination (38) and affect multiple other pathways (37) | ABBV-075 |
AZD5153# | ||||
BMS-986158* | ||||
BMS-986378* | ||||
Bromosporine | ||||
CPI-203 | ||||
GSK525762* | ||||
I-BET 151 | ||||
I-BET 762 | ||||
JQ1 | ||||
LY 303511 | ||||
MS 436 | ||||
OTX 015 | ||||
OXF BD 02 | ||||
PFI 1 | ||||
PLX51107* | ||||
SYHA1801* | ||||
XMD 8-92 | ||||
ZEN-3694# | ||||
HMGB2 | Transcriptional upregulation through insulation from heterochromatin marks (39) | Promotes dissemination downstream of CENPU (40) | Inflachromene | |
METLL3 | Enhancer-promoter loop formation (15) | Promotes invasion and migration through inhibition of CCNG2 (41) | Cpd-564 | |
STM2457 | ||||
UZH1a | ||||
mTORC1 | Translational regulation (19) | Decreased mTORC1 activity decreases migration, invasion, dissemination (42–45) | Inhibition decreases dissemination (42–45) | ABI-009# |
ATG-008# | ||||
AZD2014# | ||||
CC-115* | ||||
CC-223* | ||||
DS-3078a* | ||||
Everolimus* | ||||
HEC68498* | ||||
MLN0128# | ||||
PF-05212384# | ||||
PQR309* | ||||
Rapamycin | ||||
SAR245409# | ||||
SF1126* | ||||
TAK-228# | ||||
Temsirolimus | ||||
Torin 1 | ||||
WXFL10030390* |
Clinical trial information was obtained from clinicaltrials.gov. SASP, senescence-associated secretory phenotype.
SASP Regulators in Ovarian Cancer
cGAS-STING.
The cGAS-STING pathway is activated by cytoplasmic dsDNA that is enriched in senescent cells due to induction of DNA damage (14). While this is still an emerging field in ovarian cancer, studies have found that the cGAS-STING pathway is activated by therapies that induce senescence. A recent publication in a mouse model of HGSOC found that cGAS-STING signaling is activated by cisplatin-induced senescence, which correlated with increased NK and T cell infiltration and response to immune checkpoint blockade specifically in Brca1 null tumors (4). Excitingly, knockdown of cGAS in this model suppressed the SASP and combinatory therapy response, although disseminated tumor burden was not assessed. In addition, cGAS-STING signaling is upstream of the transcription factor nuclear factor-κB (NF-κB)-mediated SASP regulation (46), which is well-known to affect ovarian cancer dissemination and is discussed in more detail below. In other cancer models, cGAS-STING activation has been shown to drive metastasis (47). However, this may not be the case in ovarian cancer. For instance, a recent study found that inhibition of the STING deubiquitinase USP35 decreases ascites volume in an ovarian cancer mouse model (27). More work is needed to fully understand the role of the cGAS-STING axis in ovarian cancer dissemination.
p53.
p53 is mutated in >90% of high-grade serous ovarian cancers (20), and p53 is a critical regulator of cellular senescence and SASP expression (17). Mounting evidence indicates that p53 is a negative regulator of the SASP, suggesting that cells with mutant p53 likely have higher SASP-like effectors. Indeed, p53-null ovarian cancers secrete higher levels of tumor-promoting chemokines, and this response is attenuated by restoring p53 function (29). Therefore, p53 loss or mutation may promote SASP in a way that is detrimental to patients with ovarian cancer outcomes. Indeed, although it has not been linked to the SASP, compared with the wild-type form, mutant p53 was found to have more metastatic potential in a mouse model of ovarian cancer (30). In addition, p53 affects other phenotypes known to be critical for metastasis, such as anoikis, EMT, angiogenesis, and stemness (48). Future work will need to be done to delineate the roles of mutant p53 in cell cycle control versus SASP in the context of ovarian cancer dissemination.
p16INK4A and RB.
The p16INK4A (p16)-retinoblastoma (RB) axis is a critical tumor suppressor pathway that is dysregulated in ∼40% of ovarian cancers (49). When altered, the p16-RB axis allows for E2F-mediated transcription of cell proliferation genes. Recent work from our laboratory and others has shown that a decrease in either p16 or RB suppresses the SASP (4, 16), although neither study was performed in ovarian cancer. Our work found that the suppression of SASP upon loss of p16 was not coupled to the cell cycle (16), suggesting that other unknown factors are at play. Interestingly, patients with RB low ovarian cancer are extremely long-lived (31), and it is intriguing to consider that this may be due in part to decreased SASP, thereby limiting dissemination.
NF-κB and C/EBP-β.
Nuclear factor-κB (NF-κB) activity is required for full transcriptional activation of the SASP (50) and is known to be downstream of cGAS-STING (46). NF-κB is well-known to have pleiotropic roles in ovarian cancer in part through promoting SASP-like actions such as stemness, angiogenesis, and inflammation (32) that affect dissemination (11). A recent report found that patients with high NF-κB pathway activity and low PI3K pathway activity have a more favorable prognosis (51), suggesting elevated NF-κB and its promoting of SASP-like factors may provide better immunosurveillance to limit dissemination, although this remains to be determined. Another important regulator of SASP activation is CCAAT/enhancer-binding protein β (C/EBP-β) (6). Interestingly, C/EBP-β has recently been linked to a transcriptional hub including inflammatory genes that are a part of the SASP (34). C/EBP-β also promotes migration of ovarian cancer cells through cooperation with death-domain-associated protein (DAXX) (35). Increased expression of both NF-κB and C/EBP-β have been linked to poor ovarian cancer survival (32, 52), but whether this is due to their contribution to SASP gene expression is currently unclear.
Epigenetic and chromatin regulators.
Multiple epigenetic and chromatin regulators have been shown to affect SASP transcription, including histone methyltransferases MLL1 and DOT1L, RNA methyltransferases METTL3/14, the epigenetic reader Brd4, and chromatin architectural proteins HMGB1/2 (15). Although not all of these have been linked to ovarian cancer dissemination, it is possible that they contribute to the disease phenotype. DOT1L and its methyl mark H3K79 are positively associated with ovarian cancer stage (53), and it may affect dissemination downstream of C/EBP-β activity or SASP expression as we have recently published (36, 52). Brd4 is also linked to ovarian cancer pathogenesis and dissemination, potentially by affecting EMT (37). HMGB2 is associated with ovarian cancer prognosis (54), and it indeed promotes dissemination downstream of the centromere protein CENPU (40). Finally, METTL3 promotes ovarian cancer invasion and migration through indirect inhibition of CCNG2 (41). Again, whether these effects are through SASP-dependent mechanisms needs to be empirically tested.
mTORC1.
mTORC1 has been shown to promote the SASP through its critical role in translation (19). Like many cancers, ovarian cancers have high mTORC1 activity, and a recent publication found that mTORC1 activity through increased p-4EBP1 correlated with ovarian cancer stage (55). Multiple reports have found that suppression of mTOR activity, either through genetic or pharmacological approaches, decreased ovarian cancer cell migration, invasion, and dissemination (42–45). What is not yet clear is whether this observation is due to reduced translation of SASP and SASP-like factors or the other signaling pathways downstream of mTORC1. As mTORC1 activity is important for a multitude of cellular processes, it is likely that it, at least in part, contributes to dissemination through regulation of SASP mRNAs.
SASP Effectors in Ovarian Cancer
The SASP is composed of multiple factors, including cytokines, chemokines, proteases, and metabolites. Previous work in other models suggests that>100 different factors are secreted as a part of the SASP (56). This has not been extensively studied in ovarian cancer. Since aging promotes ovarian cancer dissemination (9), and cancer therapy induces senescence and the SASP (3, 4, 22), it is likely that SASP factors are a key component of ovarian cancer biology. Here, we will briefly describe SASP factors that are highly relevant in ovarian cancer dissemination, namely, IL6/IL8 and MMPs. Clearly, more work is needed to shed light on other SASP factors and their roles in ovarian cancer.
IL6 and IL8.
Many of the cytokines that are secreted as a part of the SASP have been previously linked to chemotaxis, invasion, and metastasis (6). In addition, malignant ascites derived from patients with late-stage ovarian cancer induces cellular senescence in normal peritoneal mesothelial cells to enhance cell adhesion, proliferation, and migration (57). Two of the most well-described SASP factors are IL6 and IL8, and both are known to affect ovarian cancer dissemination, progression, and survival (58–60). IL6 enriches ovarian CSCs (61), which are often found in the ascites fluid and promote dissemination through multiple mechanisms (62). Interestingly, suppression of the paracrine SASP decreases CSCs and promotes therapeutic response (22), suggesting the SASP is an important contributing factor to stemness in ovarian cancer. Moreover, IL6 facilitates EMT to promote invasion and dissemination of ovarian cancer (63). IL6 is also an immunosuppressive cytokine (64), which may decrease immunosurveillance to allow ovarian cancer cells to more easily spread throughout the peritoneal cavity. High levels of the chemokine IL8 and its receptors CXCR1/2 are also associated with ovarian cancer dissemination and chemotaxis (65). Indeed, IL8 expression in ascites fluid is higher in patients with ovarian cancer with stage III/IV tumors than those with stage I/II tumors (66). IL8 also promotes a mesenchymal phenotype, allowing for increased ovarian cancer migration (65). Together, SASP factors IL6 and IL8 are clearly two contributing factors to the dissemination observed in ovarian cancer, although it is less clear what cells are producing these factors in a complex in-vivo setting.
MMPs.
Interestingly, cytokines/chemokines are not the only factors that influence dissemination. Extracellular matrix (ECM) remodelers such as MMPs also play a role. Not all MMPs are characteristic of the SASP. SASP-related MMPs are MMP1, 3, 10, 12, 13, and 14 (67). In ovarian cancer, MMP3 is significantly associated with a higher stage (68), suggesting it plays a role in dissemination and/or invasion. MMP1/3 have also been shown to affect intraperitoneal dissemination of ovarian cancer (69). In addition, MMP1 mRNA was recently shown to be in extracellular vesicles within ascites fluid, which promoted ovarian cancer dissemination in mouse models (70).
Therapeutic Strategies
One of the major difficulties in ovarian cancer treatment is that it is often undetected until the cancer has disseminated to distant sites (1). As discussed earlier, many SASP regulators and effectors are well-known to affect dissemination. Therefore, it is intriguing to speculate that inhibition of the SASP may decrease dissemination and improve patient outcomes. Moreover, many SASP factors also promote chemoresistance (6), suggesting a dual-pronged approach to treating ovarian cancers. Because of these detrimental effects, there is increasing interest in using therapies that kill senescent cells (senolytics) or modulate the SASP (senomorphics). Recent work from the Campisi laboratory demonstrated that clearance of senescent cells using an elegantly engineered mouse model decreases both chemoresistance and metastasis in a mouse breast cancer model (71), showing proof-of-principle of this concept. Senolytic drugs are thought to work by targeting antiapoptotic pathways. However, emerging evidence suggests that senolytic effects are primarily due to elimination of the SASP (72). The senolytic and BCL2 inhibitor navitoclax have been shown to clear senescent ovarian cancer cells after treatment with PARP inhibitors and prevent cancer relapse in mice treated with doxorubicin (3, 71). No work has yet been done in ovarian cancer to investigate whether targeting the SASP using senolytics decreases dissemination.
Other regulators of the SASP may also be targetable in ovarian cancer. For instance, there has been significant excitement about bromo- and extra-terminal domain (BET) inhibitors in ovarian cancer (37), and the BET family protein Brd4 is known to affect the SASP through enhancers (8). Indeed, BET inhibitors suppress ovarian cancer dissemination in part through inhibition of STAT3 (38), a known regulator of the SASP (6). Other pathways known to regulate the SASP such as NF-κB and mTOR have been targeted in ovarian cancer, but with little success thus far (32, 73). It is possible that induction of senescence with platinum or PARP inhibitors before these therapies would provide a better therapeutic window to suppress the SASP and thereby limit dissemination.
On the other hand, recent work has demonstrated that induction of senescence within the tumor promotes immune cell infiltration and increases response to immune checkpoint blockade (4, 7). Indeed, boosting the SASP or increasing the cGAS-STING axis has multiple therapeutic benefits in mouse models of ovarian cancer (7, 28). Collectively, these data indicate that induction of the SASP may be beneficial at least in the context of the immune response to make these tumors “hot,” thereby sensitizing them to immunotherapy. More work needs to be done to assess whether this leads to a long-term, sustained therapeutic response in humans.
SUMMARY
There has been an explosion of new information on regulation of the SASP and its contents at a mechanistic level. As we have discussed, the SASP is extremely diverse and not only modulates inflammation but also ECM remodeling, stemness, chemoresistance, and many other tumor-suppressive and tumor-promoting phenotypes that may contribute to ovarian cancer dissemination. However, many questions remain to be answered, especially related to the causal role of senescence and the SASP in ovarian cancer dissemination. For instance, we still need to investigate the exact components of the SASP from ovarian cancer cells in response to different stimuli and whether those diverse profiles have similar effects on dissemination. In addition, we are just starting to understand the types of immune cells that are recruited and activated by the SASP from ovarian cancer cells. We will need to further delineate whether this is dependent on the site of the tumor (primary versus disseminated) and how that affects ovarian cancer biology. Finally, we need to be able to better understand how to fine-tune the SASP to harness its potential benefits while suppressing its negative effects.
GRANTS
This work was supported by grants from the National Institutes of Health (R01CA259111), the American Cancer Society (RSG-19-113-01), and the Department of Defense Ovarian Cancer Research Program (W81XWH-21-1-0840) (to K.M.A.).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
K.M.A. prepared figures; J.P.V., L.F.F.B., and K.M.A. drafted manuscript; J.P.V., L.F.F.B., and K.M.A. edited and revised manuscript; J.P.V., L.F.F.B., and K.M.A. approved final version of manuscript.
ACKNOWLEDGMENTS
The authors thank Drs. Raquel Buj and Naveen Tangudu for critical reading of the manuscript.
This article is part of the special collection “Tumor Host Interactions in Metastasis.” Mythreye Karthikeyan, PhD, and Nadine Hempel, PhD, served as Guest Editors of this collection.
REFERENCES
- 1.Torre LA, Trabert B, DeSantis CE, Miller KD, Samimi G, Runowicz CD, Gaudet MM, Jemal A, Siegel RL. Ovarian cancer statistics, 2018. CA Cancer J Clin 68: 284–296, 2018. doi: 10.3322/caac.21456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin 72: 7–33, 2022. doi: 10.3322/caac.21708. [DOI] [PubMed] [Google Scholar]
- 3.Fleury H, Malaquin N, Tu V, Gilbert S, Martinez A, Olivier MA, Sauriol A, Communal L, Leclerc-Desaulniers K, Carmona E, Provencher D, Mes-Masson AM, Rodier F. Exploiting interconnected synthetic lethal interactions between PARP inhibition and cancer cell reversible senescence. Nat Commun 10: 2556, 2019. doi: 10.1038/s41467-019-10460-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Paffenholz SV, Salvagno C, Ho YJ, Limjoco M, Baslan T, Tian S, Kulick A, de Stanchina E, Wilkinson JE, Barriga FM, Zamarin D, Cubillos-Ruiz JR, Leibold J, Lowe SW. Senescence induction dictates response to chemo- and immunotherapy in preclinical models of ovarian cancer. Proc Natl Acad Sci USA 119: e2117754119, 2022. doi: 10.1073/pnas.2117754119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Herranz N, Gil J. Mechanisms and functions of cellular senescence. J Clin Invest 128: 1238–1246, 2018. doi: 10.1172/JCI95148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Chambers CR, Ritchie S, Pereira BA, Timpson P. Overcoming the senescence-associated secretory phenotype (SASP): a complex mechanism of resistance in the treatment of cancer. Mol Oncol 15: 3242–3255, 2021. doi: 10.1002/1878-0261.13042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hao X, Zhao B, Zhou W, Liu H, Fukumoto T, Gabrilovich D, Zhang R. Sensitization of ovarian tumor to immune checkpoint blockade by boosting senescence-associated secretory phenotype. iScience 24: 102016, 2021. doi: 10.1016/j.isci.2020.102016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Tasdemir N, Banito A, Roe JS, Alonso-Curbelo D, Camiolo M, Tschaharganeh DF, Huang CH, Aksoy O, Bolden JE, Chen CC, Fennell M, Thapar V, Chicas A, Vakoc CR, Lowe SW. BRD4 connects enhancer remodeling to senescence immune surveillance. Cancer Discov 6: 612–629, 2016. doi: 10.1158/2159-8290.CD-16-0217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Mikuła-Pietrasik J, Uruski P, Sosińska P, Maksin K, Piotrowska-Kempisty H, Kucińska M, Murias M, Szubert S, Woźniak A, Szpurek D, Sajdak S, Piwocka K, Tykarski A, Książek K. Senescent peritoneal mesothelium creates a niche for ovarian cancer metastases. Cell Death Dis 7: e2565, 2016. doi: 10.1038/cddis.2016.417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol 75: 685–705, 2013. doi: 10.1146/annurev-physiol-030212-183653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Weidle UH, Birzele F, Kollmorgen G, Rueger R. Mechanisms and targets involved in dissemination of ovarian cancer. Cancer Genomics Proteomics 13: 407–423, 2016. doi: 10.21873/cgp.20004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Tan DS, Agarwal R, Kaye SB. Mechanisms of transcoelomic metastasis in ovarian cancer. Lancet Oncol 7: 925–934, 2006. doi: 10.1016/S1470-2045(06)70939-1. [DOI] [PubMed] [Google Scholar]
- 13.Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol 15: 482–496, 2014. doi: 10.1038/nrm3823. [DOI] [PubMed] [Google Scholar]
- 14.Miller KN, Dasgupta N, Liu T, Adams PD, Vizioli MG. Cytoplasmic chromatin fragments-from mechanisms to therapeutic potential. eLife 10: e63728, 2021. doi: 10.7554/eLife.63728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hao X, Wang C, Zhang R. Chromatin basis of the senescence-associated secretory phenotype. Trends Cell Biol 32: 513–526, 2022. doi: 10.1016/j.tcb.2021.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Buj R, Leon KE, Anguelov MA, Aird KM. Suppression of p16 alleviates the senescence-associated secretory phenotype. Aging (Albany NY) 13: 3290–3312, 2021. doi: 10.18632/aging.202640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sheekey E, Narita M. p53 in senescence – it’s a marathon not a sprint. FEBS J, 2021. doi: 10.1111/febs.16325. [DOI] [PubMed] [Google Scholar]
- 18.Sturmlechner I, Zhang C, Sine CC, van Deursen EJ, Jeganathan KB, Hamada N, Grasic J, Friedman D, Stutchman JT, Can I, Hamada M, Lim DY, Lee JH, Ordog T, Laberge RM, Shapiro V, Baker DJ, Li H, van Deursen JM. p21 produces a bioactive secretome that places stressed cells under immunosurveillance. Science 374: eabb3420, 2021. doi: 10.1126/science.abb3420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Payea MJ, Anerillas C, Tharakan R, Gorospe M. Translational control during cellular senescence. Mol Cell Biol 41: e00512-20, 2021. doi: 10.1128/MCB.00512-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Bowtell DD, Bohm S, Ahmed AA, Aspuria PJ, Bast RC Jr, Beral V, et al. Rethinking ovarian cancer II: reducing mortality from high-grade serous ovarian cancer. Nat Rev Cancer 15: 668–679, 2015. doi: 10.1038/nrc4019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Previs RA, Secord AA. Ovarian cancer: clinical trial breakthroughs and impact on management. Obstet Gynecol Clin North Am 46: 67–88, 2019. doi: 10.1016/j.ogc.2018.09.005. [DOI] [PubMed] [Google Scholar]
- 22.Nacarelli T, Fukumoto T, Zundell JA, Fatkhutdinov N, Jean S, Cadungog MG, Borowsky ME, Zhang R. NAMPT Inhibition suppresses cancer stem-like cells associated with therapy-induced senescence in ovarian cancer. Cancer Res 80: 890–900, 2020. doi: 10.1158/0008-5472.CAN-19-2830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Calvo L, Cheng S, Skulimowski M, Clement I, Portelance L, Zhan Y, Carmona E, Lafontaine J, de Ladurantaye M, Rahimi K, Provencher D, Mes-Masson A-M, Rodier F. Cellular senescence is a central response to cytotoxic chemotherapy in high-grade serous ovarian cancer (Preprint). bioRxiv, 2018. doi: 10.1101/425199. [DOI]
- 24.Chan JK, Urban R, Cheung MK, Osann K, Shin JY, Husain A, Teng NN, Kapp DS, Berek JS, Leiserowitz GS. Ovarian cancer in younger vs older women: a population-based analysis. Br J Cancer 95: 1314–1320, 2006. [Erratum in Br J Cancer 96: 1492, 2007]. doi: 10.1038/sj.bjc.6603457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Uruski P, Mikuła-Pietrasik J, Naumowicz E, Kaźmierczak K, Gaiday AN, Królak J, Nowakowski B, Moszyński R, Tykarski A, Książek K. Patient-specific variables determine the extent of cellular senescence biomarkers in ovarian tumors in vivo. Biomedicines 9: 330, 2021. doi: 10.3390/biomedicines9040330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hou X, Zhai Y, Hu K, Liu CJ, Udager A, Pearce CL, Fearon ER, Cho KR. Aging accelerates while multiparity delays tumorigenesis in mouse models of high-grade serous carcinoma. Gynecologic oncology 165: 552–559, 2022. doi: 10.1016/j.ygyno.2022.03.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhang J, Chen Y, Chen X, Zhang W, Zhao L, Weng L, Tian H, Wu Z, Tan X, Ge X, Wang P, Fang L. Deubiquitinase USP35 restrains STING-mediated interferon signaling in ovarian cancer. Cell Death Differ 28: 139–155, 2021. doi: 10.1038/s41418-020-0588-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Grabosch S, Bulatovic M, Zeng F, Ma T, Zhang L, Ross M, Brozick J, Fang Y, Tseng G, Kim E, Gambotto A, Elishaev E, Edwards RP, Vlad AM. Cisplatin-induced immune modulation in ovarian cancer mouse models with distinct inflammation profiles. Oncogene 38: 2380–2393, 2019. doi: 10.1038/s41388-018-0581-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Son DS, Kabir SM, Dong YL, Lee E, Adunyah SE. Inhibitory effect of tumor suppressor p53 on proinflammatory chemokine expression in ovarian cancer cells by reducing proteasomal degradation of IκB. PLoS One 7: e51116, 2012. doi: 10.1371/journal.pone.0051116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ren YA, Mullany LK, Liu Z, Herron AJ, Wong KK, Richards JS. Mutant p53 promotes epithelial ovarian cancer by regulating tumor differentiation, metastasis, and responsiveness to steroid hormones. Cancer Res 76: 2206–2218, 2016. doi: 10.1158/0008-5472.CAN-15-1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Garsed DW, Alsop K, Fereday S, Emmanuel C, Kennedy CJ, Etemadmoghadam D, et al. Homologous recombination DNA repair pathway disruption and retinoblastoma protein loss are associated with exceptional survival in high-grade serous ovarian cancer. Clin Cancer Res 24: 569–580, 2018. doi: 10.1158/1078-0432.CCR-17-1621. [DOI] [PubMed] [Google Scholar]
- 32.Harrington BS, Annunziata CM. NF-κB signaling in ovarian cancer. Cancers (Basel) 11: 1182, 2019. doi: 10.3390/cancers11081182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ramadass V, Vaiyapuri T, Tergaonkar V. Small molecule NF-κB pathway inhibitors in clinic. Int J Mol Sci 21: 5164, 2020. doi: 10.3390/ijms21145164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Jordan KR, Sikora MJ, Slansky JE, Minic A, Richer JK, Moroney MR, Hu J, Wolsky RJ, Watson ZL, Yamamoto TM, Costello JC, Clauset A, Behbakht K, Kumar TR, Bitler BG. The Capacity of the Ovarian Cancer Tumor Microenvironment to Integrate Inflammation Signaling Conveys a Shorter Disease-free Interval. Clin Cancer Res 26: 6362–6373, 2020. doi: 10.1158/1078-0432.CCR-20-1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Liu SB, Lin XP, Xu Y, Shen ZF, Pan WW. DAXX promotes ovarian cancer ascites cell proliferation and migration by activating the ERK signaling pathway. J Ovarian Res 11: 90, 2018. doi: 10.1186/s13048-018-0462-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Leon KE, Buj R, Lesko E, Dahl ES, Chen CW, Tangudu NK, Imamura-Kawasawa Y, Kossenkov AV, Hobbs RP, Aird KM. DOT1L modulates the senescence-associated secretory phenotype through epigenetic regulation of IL1A. J Cell Biol 220: e202008101, 2021. doi: 10.1083/jcb.202008101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Drumond-Bock AL, Bieniasz M. The role of distinct BRD4 isoforms and their contribution to high-grade serous ovarian carcinoma pathogenesis. Mol Cancer 20: 145, 2021. doi: 10.1186/s12943-021-01424-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Andrikopoulou A, Liontos M, Koutsoukos K, Dimopoulos MA, Zagouri F. Clinical perspectives of BET inhibition in ovarian cancer. Cell Oncol (Dordr) 44: 237–249, 2021. doi: 10.1007/s13402-020-00578-6. [DOI] [PubMed] [Google Scholar]
- 39.Aird KM, Iwasaki O, Kossenkov AV, Tanizawa H, Fatkhutdinov N, Bitler BG, Le L, Alicea G, Yang TL, Johnson FB, Noma KI, Zhang R. HMGB2 orchestrates the chromatin landscape of senescence-associated secretory phenotype gene loci. J Cell Biol 215: 325–334, 2016. doi: 10.1083/jcb.201608026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li H, Zhang H, Wang Y. Centromere protein U facilitates metastasis of ovarian cancer cells by targeting high mobility group box 2 expression. Am J Cancer Res 8: 835–851, 2018. [PMC free article] [PubMed] [Google Scholar]
- 41.Bi X, Lv X, Liu D, Guo H, Yao G, Wang L, Liang X, Yang Y. METTL3 promotes the initiation and metastasis of ovarian cancer by inhibiting CCNG2 expression via promoting the maturation of pri-microRNA-1246. Cell Death Discov 7: 237, 2021. doi: 10.1038/s41420-021-00600-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lucà R, Assenza MR, Maiullari F, Pieroni L, Maiullari S, Federici G, Marini F, Rizzi R, Urbani A, Soddu S, Moretti F. Inhibition of the mTOR pathway and reprogramming of protein synthesis by MDM4 reduce ovarian cancer metastatic properties. Cell Death Dis 12: 558, 2021. doi: 10.1038/s41419-021-03828-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Xiao Y, Yu Y, Jiang P, Li Y, Wang C, Zhang R. The PI3K/mTOR dual inhibitor GSK458 potently impedes ovarian cancer tumorigenesis and metastasis. Cell Oncol (Dordr) 43: 669–680, 2020. doi: 10.1007/s13402-020-00514-8. [DOI] [PubMed] [Google Scholar]
- 44.Sun C, Li X, Guo E, Li N, Zhou B, Lu H, Huang J, Xia M, Shan W, Wang B, Li K, Weng D, Xu X, Gao Q, Wang S, Hu J, Lu Y, Mills GB, Chen G. MCP-1/CCR-2 axis in adipocytes and cancer cell respectively facilitates ovarian cancer peritoneal metastasis. Oncogene 39: 1681–1695, 2020. doi: 10.1038/s41388-019-1090-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Spaeth-Cook D, Burch M, Belton R, Demoret B, Grosenbacher N, David J, Stets C, Cohen D, Shakya R, Hays JL, Chen JL. Loss of TXNIP enhances peritoneal metastasis and can be abrogated by dual TORC1/2 inhibition. Oncotarget 9: 35676–35686, 2018. doi: 10.18632/oncotarget.26281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dou Z, Ghosh K, Vizioli MG, Zhu J, Sen P, Wangensteen KJ, Simithy J, Lan Y, Lin Y, Zhou Z, Capell BC, Xu C, Xu M, Kieckhaefer JE, Jiang T, Shoshkes-Carmel M, Tanim K, Barber GN, Seykora JT, Millar SE, Kaestner KH, Garcia BA, Adams PD, Berger SL. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature 550: 402–406, 2017. doi: 10.1038/nature24050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Bakhoum SF, Ngo B, Laughney AM, Cavallo JA, Murphy CJ, Ly P, Shah P, Sriram RK, Watkins TBK, Taunk NK, Duran M, Pauli C, Shaw C, Chadalavada K, Rajasekhar VK, Genovese G, Venkatesan S, Birkbak NJ, McGranahan N, Lundquist M, LaPlant Q, Healey JH, Elemento O, Chung CH, Lee NY, Imielenski M, Nanjangud G, Pe'er D, Cleveland DW, Powell SN, Lammerding J, Swanton C, Cantley LC. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 553: 467–472, 2018. doi: 10.1038/nature25432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Babaei G, Aliarab A, Asghari Vostakolaei M, Hotelchi M, Neisari R, Gholizadeh-Ghaleh Aziz S, Bazl MR. Crosslink between p53 and metastasis: focus on epithelial-mesenchymal transition, cancer stem cell, angiogenesis, autophagy, and anoikis. Mol Biol Rep 48: 7545–7557, 2021. doi: 10.1007/s11033-021-06706-1. [DOI] [PubMed] [Google Scholar]
- 49.Corney DC, Flesken-Nikitin A, Choi J, Nikitin AY. Role of p53 and Rb in ovarian cancer. Adv Exp Med Biol 622: 99–117, 2008. doi: 10.1007/978-0-387-68969-2_9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lopes-Paciencia S, Saint-Germain E, Rowell MC, Ruiz AF, Kalegari P, Ferbeyre G. The senescence-associated secretory phenotype and its regulation. Cytokine 117: 15–22, 2019. doi: 10.1016/j.cyto.2019.01.013. [DOI] [PubMed] [Google Scholar]
- 51.van Lieshout L, van de Stolpe A, van der Ploeg P, Bowtell D, de Hullu J, Piek J. Signal transduction pathway activity in high-grade, serous ovarian carcinoma reveals a more favorable prognosis in tumors with low PI3K and high NF-κB pathway activity: a novel approach to a long-standing enigma. Cancers (Basel) 12: 2660, 2020. doi: 10.3390/cancers12092660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Liu D, Zhang XX, Li MC, Cao CH, Wan DY, Xi BX, Tan JH, Wang J, Yang ZY, Feng XX, Ye F, Chen G, Wu P, Xi L, Wang H, Zhou JF, Feng ZH, Ma D, Gao QL. C/EBPβ enhances platinum resistance of ovarian cancer cells by reprogramming H3K79 methylation. Nat Commun 9: 1739, 2018. doi: 10.1038/s41467-018-03590-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhang X, Liu D, Li M, Cao C, Wan D, Xi B, Li W, Tan J, Wang J, Wu Z, Ma D, Gao Q. Prognostic and therapeutic value of disruptor of telomeric silencing-1-like (DOT1L) expression in patients with ovarian cancer. J Hematol Oncol 10: 29, 2017. doi: 10.1186/s13045-017-0400-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ouellet V, Le Page C, Guyot MC, Lussier C, Tonin PN, Provencher DM, Mes-Masson AM. SET complex in serous epithelial ovarian cancer. Int J Cancer 119: 2119–2126, 2006. doi: 10.1002/ijc.22054. [DOI] [PubMed] [Google Scholar]
- 55.Alabdullah ML, Ahmad DA, Moseley P, Madhusudan S, Chan S, Rakha E. The mTOR downstream regulator (p-4EBP1) is a novel independent prognostic marker in ovarian cancer. J Obstet Gynaecol 39: 522–528, 2019. doi: 10.1080/01443615.2018.1534091. [DOI] [PubMed] [Google Scholar]
- 56.Basisty N, Kale A, Jeon OH, Kuehnemann C, Payne T, Rao C, Holtz A, Shah S, Sharma V, Ferrucci L, Campisi J, Schilling B. A proteomic atlas of senescence-associated secretomes for aging biomarker development. PLoS Biol 18: e3000599, 2020. doi: 10.1371/journal.pbio.3000599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mikuła-Pietrasik J, Uruski P, Matuszkiewicz K, Szubert S, Moszyński R, Szpurek D, Sajdak S, Tykarski A, Książek K. Ovarian cancer-derived ascitic fluids induce a senescence-dependent pro-cancerogenic phenotype in normal peritoneal mesothelial cells. Cell Oncol (Dordr) 39: 473–481, 2016. doi: 10.1007/s13402-016-0289-1. [DOI] [PubMed] [Google Scholar]
- 58.Lane D, Matte I, Rancourt C, Piché A. Prognostic significance of IL-6 and IL-8 ascites levels in ovarian cancer patients. BMC Cancer 11: 210, 2011. doi: 10.1186/1471-2407-11-210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wen W, Liang W, Wu J, Kowolik CM, Buettner R, Scuto A, Hsieh MY, Hong H, Brown CE, Forman SJ, Horne D, Morgan R, Wakabayashi M, Dellinger TH, Han ES, Yim JH, Jove R. Targeting JAK1/STAT3 signaling suppresses tumor progression and metastasis in a peritoneal model of human ovarian cancer. Mol Cancer Ther 13: 3037–3048, 2014. doi: 10.1158/1535-7163.MCT-14-0077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Mehner C, Miller E, Hockla A, Coban M, Weroha SJ, Radisky DC, Radisky ES. Targeting an autocrine IL-6-SPINK1 signaling axis to suppress metastatic spread in ovarian clear cell carcinoma. Oncogene 39: 6606–6618, 2020. doi: 10.1038/s41388-020-01451-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wang Y, Zong X, Mitra S, Mitra AK, Matei D, Nephew KP. IL-6 mediates platinum-induced enrichment of ovarian cancer stem cells. JCI Insight 3: e122360, 2018. doi: 10.1172/jci.insight.122360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zong X, Nephew KP. Ovarian cancer stem cells: role in metastasis and opportunity for therapeutic targeting. Cancers (Basel) 11: 934, 2019. doi: 10.3390/cancers11070934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.So KA, Min KJ, Hong JH, Lee JK. Interleukin-6 expression by interactions between gynecologic cancer cells and human mesenchymal stem cells promotes epithelial-mesenchymal transition. Int J Oncol 47: 1451–1459, 2015. doi: 10.3892/ijo.2015.3122. [DOI] [PubMed] [Google Scholar]
- 64.Macciò A, Madeddu C. Inflammation and ovarian cancer. Cytokine 58: 133–147, 2012. doi: 10.1016/j.cyto.2012.01.015. [DOI] [PubMed] [Google Scholar]
- 65.Wen J, Zhao Z, Huang L, Wang L, Miao Y, Wu J. IL-8 promotes cell migration through regulating EMT by activating the Wnt/β-catenin pathway in ovarian cancer. J Cell Mol Med 24: 1588–1598, 2020. doi: 10.1111/jcmm.14848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zhang L, Liu W, Wang X, Wang X, Sun H. Prognostic value of serum IL-8 and IL-10 in patients with ovarian cancer undergoing chemotherapy. Oncol Lett 17: 2365–2369, 2018. doi: 10.3892/ol.2018.9842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Coppé JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol 5: 99–118, 2010. doi: 10.1146/annurev-pathol-121808-102144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wang S, Jia J, Liu D, Wang M, Wang Z, Li X, Wang H, Rui Y, Liu Z, Guo W, Nie J, Dai H. Matrix metalloproteinase expressions play important role in prediction of ovarian cancer outcome. Sci Rep 9: 11677, 2019. doi: 10.1038/s41598-019-47871-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Agarwal A, Covic L, Sevigny LM, Kaneider NC, Lazarides K, Azabdaftari G, Sharifi S, Kuliopulos A. Targeting a metalloprotease-PAR1 signaling system with cell-penetrating pepducins inhibits angiogenesis, ascites, and progression of ovarian cancer. Mol Cancer Ther 7: 2746–2757, 2008. doi: 10.1158/1535-7163.MCT-08-0177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yokoi A, Yoshioka Y, Yamamoto Y, Ishikawa M, Ikeda SI, Kato T, Kiyono T, Takeshita F, Kajiyama H, Kikkawa F, Ochiya T. Malignant extracellular vesicles carrying MMP1 mRNA facilitate peritoneal dissemination in ovarian cancer. Nat Commun 8: 14470, 2017. doi: 10.1038/ncomms14470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Demaria M, O'Leary MN, Chang J, Shao L, Liu S, Alimirah F, Koenig K, Le C, Mitin N, Deal AM, Alston S, Academia EC, Kilmarx S, Valdovinos A, Wang B, de Bruin A, Kennedy BK, Melov S, Zhou D, Sharpless NE, Muss H, Campisi J. Cellular senescence promotes adverse effects of chemotherapy and cancer relapse. Cancer Discov 7: 165–176, 2017. doi: 10.1158/2159-8290.CD-16-0241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Birch J, Gil J. Senescence and the SASP: many therapeutic avenues. Genes Dev 34: 1565–1576, 2020. doi: 10.1101/gad.343129.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Rinne N, Christie EL, Ardasheva A, Kwok CH, Demchenko N, Low C, Tralau-Stewart C, Fotopoulou C, Cunnea P. Targeting the PI3K/AKT/mTOR pathway in epithelial ovarian cancer, therapeutic treatment options for platinum-resistant ovarian cancer. Cancer Drug Resist 4: 573–595, 2021. doi: 10.20517/cdr.2021.05. [DOI] [PMC free article] [PubMed] [Google Scholar]