Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Review

The role of senescence in cancer development



Eleni Mavrogonatou, Harris Pratsinis, Dimitris Kletsas
Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos”, Athens, Greece

A R T I C LE I N FO A B S T R A C T

Keywords: While research on cancer development is traditionally focusing mainly on the neoplastic cell per se, nowadays
Senescence the role of tumor stroma in this process is indisputable. The stroma - mainly composed of extracellular matrix
Stroma (ECM) - is a source of mediators and signals originating from heterotypic cell-cell and cell-matrix interactions
Extracellular matrix that steer the progression of the disease in a context- and a cancer type-dependent manner. With advancing age
Senescence-associated secretory phenotype
the stroma exhibits alterations, important being the accumulation of senescent cells. Senescence is often trig-
Senolytic
gered by exogenous stresses, including genotoxic anticancer treatment modalities (such as chemotherapy or
radiotherapy) and is manifested as an inhibition of cell proliferation, ascribing to cellular senescence the role of a
potent antitumor barrier. On the other hand, senescent cells, through their specific senescence-associated se-
cretory phenotype (SASP) - comprising cytokines, growth factors, ECM components and ECM-degrading enzymes
- can establish an immunosuppressive, inflammatory and catabolic microenvironment that may stimulate tumor
growth and metastasis. Given that the persistent presence of senescent cells could prove detrimental for tissue
homeostasis, inclusion of a senotherapeutic arm in novel anticancer approaches seems compulsory.

1. Introduction has moved ahead to more sophisticated strategies that take into account
several parameters of the multistep process of tumor growth, the bio-
In oversimplified terms, cancer is the outcome of the unlimited and chemical alterations of the stroma (including changes in stromal cells,
uncontrolled cell proliferation and its manifestation is often associated such as the induction of senescence), the interconnection of neoplastic
with increased chronological age. The hallmarks of cancer consist of cells with the stroma, the activation of the immune system, as well as
distinct and at the same time complementary traits that are required for the putative side-effects of the treatment itself on the future progress of
the survival, proliferation and dissemination of malignant cells, as well the disease. Even though our knowledge is growing and advance has
as for the succeeding progression of the tumor, i.e. incessant pro- been achieved, new therapeutic approaches are needed targeting the
liferative signaling, escape from growth suppressors, resistance to cell induction, abolishment or disruption of molecular pathways in malig-
death, induction of angiogenesis, activation of invasion and metastasis, nant cells and metabolic components of the stroma. Still, innovative
replicative immortality, genomic instability generating random muta- drugs and therapy schemes should be cautiously designed, since dis-
tions, tumor-promoting inflammation, metabolism reprogramming to- turbance of the subtle equilibrium between tumor promotion, preven-
wards anaerobic glycolysis to support sustained cell proliferation and tion and cure could prove deleterious for cancer development.
active evasion from the elimination by immune cells [1].
Given that most of human cancers are of epithelial origin [2,3], for 2. The role of the stroma in cancer development
many years carcinogenesis and further tumor growth was believed to be
a one-dimensional process, depending solely on the accumulation of The stroma mainly consists of extracellular matrix (ECM) and covers
sequential mutations in the genes carried by epithelial cells [2]. This the region between the tissue’s basement membrane and the cancer
reductionist approach is incomplete as it is now well established that parenchyma, thus forming a structure that surrounds malignant cells
the stroma is an additional key player in cancer development [4–6]. and can often represent more than 90% of the solid tumor [2,6,7].
The primary goal of traditional anticancer therapies was merely to Embedded in the ECM, non-cancerous stromal cells include fibroblasts,
abrogate tumor cells’ unrestrained proliferation by driving them to cell endothelial cells, specialized mesenchymal cells and immune cells
cycle arrest or apoptosis. Nevertheless, coping with cancer nowadays [4,8], with fibroblasts constituting the most important cell type, since


Corresponding author at: Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research
“Demokritos”, 153 10 Athens, Greece.
E-mail address: dkletsas@bio.demokritos.gr (D. Kletsas).

https://doi.org/10.1016/j.semcancer.2019.06.018
Received 20 May 2019; Received in revised form 24 June 2019; Accepted 27 June 2019
1044-579X/ © 2019 Published by Elsevier Ltd.

Please cite this article as: Eleni Mavrogonatou, Harris Pratsinis and Dimitris Kletsas, Seminars in Cancer Biology,
https://doi.org/10.1016/j.semcancer.2019.06.018
E. Mavrogonatou, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

they are responsible for the production of the structural components of


the stroma and the basement membrane, as well as for the secretion of
cytokines, growth factors, ECM components and ECM-degrading en-
zymes that can result in local tissue remodeling and alterations in the
architecture of the tumor microenvironment [2,4,9–11]. In the course
of tumor development fibroblasts become activated, lapsing into cancer
associated fibroblasts (CAFs) that promote malignant growth, invasion,
and metastasis [12–15]. Furthermore, the decisive role of the stroma in
tumorigenesis has been evidenced in numerous studies demonstrating
on one hand cancer growth suppression by a normal stroma and on the
other hand promotion of tumor development by an activated stroma
[16–20]. Paradigms of mediators produced by the stroma that de-
termine the fate of cancer progression through heterotypic cell and cell-
matrix interactions are transforming growth factor-β (TGF-β) and in-
terleukin (IL)-6 secreted by CAFs - that can affect proliferative, mi-
gratory, survival and invasive capacities of malignant cells - [21–23] or
matrix metalloproteases (MMPs) and members of the A Disintegrin And
Metalloproteases (ADAM) protein family - that can catabolize ECM
constituents and affect tissue homeostasis, creating a permissive en-
vironment for tumor growth [5,24–31]. Evolving ECM plays a dynamic Fig. 1. Major signaling pathways leading to cellular senescence. Telomere
role in tumor behavior, as it seems to adapt in every stage of tumor- shortening or a broad spectrum of other genotoxic stresses is triggering a DNA
igenesis, influencing the progression of the disease. It may abolish damage response, which through the indicated pathways leads to a persistent
proliferative restriction and increase replicative capacity via telomerase growth arrest, the major feature of cellular senescence.
(the enzyme that secures telomere size) overexpression, inhibit tumor
suppressors, reduce chemotherapeutic responsiveness, favor angiogen- damage response (DDR) pathways, such as the activation of the ATM-
esis and enhance invasion through actin assembly remodeling or in- Chk2-p53-p21WAF1-pRb axis leading to an irreversible growth arrest
tegrin-dependent and -independent cell adhesion [1,32]. In addition, [45–48] (Fig. 1).
cancer-associated ECM alterations may negatively affect immune re- Besides replicative exhaustion, there are additional internal and
sponses, subserve the metabolic shift to pathways that feed the in- external stressors that lead to another type of senescence (usually in-
creased energy requirements of the highly proliferating malignant cells, dependent of telomere erosion), the so-called stress-induced premature
hamper DNA repair machineries and influence the activation state of senescence (SIPS) [49,50]. Among the stresses eliciting SIPS lay oxi-
innate immune cells [32]. However, predisposition to malignancy in dative stress, exposure to ionizing radiation, treatment with che-
different contexts and/or types of cancer may stem from opposing ECM motherapeutic drugs and other genotoxic insults, while oncogene ac-
alterations, rendering it difficult to make general assumptions and to tivation fuels a special type of SIPS, namely the oncogene-induced
correlate specific ECM modifications to anticipated outcomes in every senescence (OIS) [51–55]. OIS was initially described in primary
cancer type; for instance, fibrosis and collagen cross-linking have been mammalian cells after the overexpression of ras [51] and was further
associated with a greater risk for breast cancer development, but MMPs’ shown to be induced by a long list of oncogenes [54,55] and to occur in
overexpression is a poor prognostic factor in breast cancer patients, as vivo, as well [56], strongly supporting the notion that cellular senes-
well [32]. cence is a potent antitumor barrier. The shared trigger in replicative
Age-related cancer has been initially ascribed to an age-dependent senescence and SIPS seems to be the provocation of a persistent DNA
raise in genomic instability and to the exponential increase and accu- damage and the subsequent launching of a DDR [46,55,57–62] (Fig. 1).
mulation of somatic mutations and epigenetic alterations over time Irrespective of the stimulus evoking senescence, senescent cells
[33–36]. However, given the pronounced implication of the stroma in display some common features: 1. an irreversible cell cycle arrest at the
cancer progression already described above, the role of the aged stroma G1 and occasionally at the G2 phase, even in the presence of mitogens
- which undoubtedly differentiates from that of a young individual - [59,63–66], which also corroborates the original hypothesis that the
should be added to these factors [4,37]. A major alteration of the aged evolutionary role of senescence is the avoidance of cellular transfor-
stroma concerns its cellular components and stems from the accumu- mation and cancer development [42]. This growth arrest has been as-
lation of senescent cells and especially that of senescent fibroblasts [4]. sociated with the up-regulation of several tumor suppressor genes, i.e.
p16INK4a, p14ARF, pRb, p53, p21WAF1 or p27KIP1, which could be used as
3. Cellular senescence molecular markers of cellular senescence [67–73]; 2. an enlarged
morphology and high granularity due to the presence of an expanded
Cellular senescence was first described by Hayflick and Moorhead in lysosomal compartment and vacuoles, which is conspicuously distinct
normal human embryonic lung fibroblasts, back in 1961, as the per- from that of young (early-passage) cells [4,74,75]; 3. an increased
manent cessation of cell divisions in vitro [38] and was subsequently metabolic rate, e.g. higher glucose consumption and lactate production
demonstrated in several other cell types [39]. This type of cellular se- [76,77]; 4. resistance to apoptosis [78,79]; 5. nuclear and chromatin
nescence reached after serial subculturing in vitro is called replicative alterations (i.e. lamin B1 deficiency [80,81], reduction in hetero-
senescence and has been attributed to the gradual loss of telomere chromatin, site-specific changes in DNA methylation patterns and his-
length after each cell division due to the “end replication problem”, i.e. tone modification [82], appearance of DNA damage response structures
the incomplete replication of the end of chromosomes during lagging DNA-SCARS [83] and DNA damage markers, such as γH2A.X and
strand DNA synthesis [40–42]. Telomere shortening has been proven to 53BP1 foci, senescence-associated distension of satellites (SADS) and
serve as the “mitotic clock” leading normal cells to senescence, since senescence-associated heterochromatin foci (SAHF) at E2F-regulated
germ and cancer cells expressing telomerase display an infinite re- genes [84–86]); 6. a unique bioactive, pro-inflammatory and proteo-
plicative potential, while introduction of the enzyme’s catalytic subunit lytic secretome, known as the senescence-associated secretory pheno-
in telomerase-deficient cells may extend their lifespan and even render type (SASP) [87]. Given that for a long period of time it was unclear if
them immortalized [43,44]. Loss of telomeric length beyond a certain senescence does indeed occur in vivo or is an artifact of the culture
threshold is perceived by the cells as a DNA damage, which drives DNA

2
E. Mavrogonatou, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

conditions in vitro, the necessity for a reliable marker for the identifi- enzymes that cleave ECM, respectively. Up-regulation of MMPs has
cation of senescent cells was indispensable. In this direction, the se- been firmly connected with senescence in several cell types like fibro-
nescence-associated beta galactosidase (SA-β-Gal) histochemical pro- blasts [86,103,105,126–131], vascular smooth muscle cells (VSMCs)
cedure was developed, which detects senescent cells by their prominent [132,133], intervertebral disc (IVD) cells [134–136] and hepatic stel-
beta galactosidase activity at pH 6.0, resulting in a characteristic blue late cells (HSCs) [137] regardless of the stimulus inducing senescence
perinuclear staining [88,89]. Even though revolutionary, we are now and of the culture conditions [105,130,136,138]. Additionally, tissue
aware that SA-β-Gal staining should be cautiously interpreted, as the inhibitors of MMPs (TIMPs) have been mainly shown to be down-
methodology is cell density- and stimulus-dependent and may thus lead regulated during senescence [126,129,134,139]. The aforementioned
to pseudo-positive results [90–92]. The recently discovered staining for increased senescence-associated proteolytic activity is reinforced by the
Sudan Black B of lipofuscin (that accumulates in the cytoplasm of se- up-regulation of enzymes belonging to the A Disintegrin And Metallo-
nescent cells) overrides most of the limitations of SA-β-Gal staining and proteinase with Thrombospondin motifs (ADAMTS) protein family
can be performed even in archival samples [93–95]. Based on the fact [134,135,140], cathepsins [127,141], urokinase-type plasminogen ac-
that senescence is complicated and multiparametric, it seems that a tivator (uPA) and tissue plasminogen activator (tPA) [103,127,142]
combination of morphologic, molecular and functional traits is rather and plasminogen activator inhibitors (PAI) -1 and -2 [142–144]. Ex-
necessary for the characterization of the senescent phenotype [96]. tracellular PAI-1 has been also shown to be downstream of p53 in the
Since senescent cells have been thus far evidenced to accumulate induction of replicative senescence [145]. Moreover, it was found that
with age [97] and to create a permissive environment for cancerous prolonged activation of nuclear factor-erythroid 2-related factor 2
outgrowth [86,95,98–101], it remains to fully elucidate the mechan- (Nrf2) leads fibroblasts to senescence and that ECM deposited by these
isms by which senescent cells can synergize with malignant cells and senescent cells further promotes senescence through PAI-1 [125].
promote cancer development. In contrast to ECM-degrading molecules that are mainly over-
expressed in senescent cells, ECM constituents have been reported to be
4. The senescence-associated secretory phenotype (SASP) down-regulated in general, thus establishing the catabolic aspect of the
senescent phenotype [95]. In detail, collagen and collagenous proteins
As already mentioned, cellular senescence is a complex phenotype show lower expression in senescent cells, in parallel to the down-reg-
elicited by diverse inputs and manifested through multiple effector ulation of prolyl 4-hydroxylase that catalyzes collagen biosynthesis
programs. Beyond the inability to proliferate, senescent cells are char- [103,127,128,130,133,137,141,146,147]. Elastin has been demon-
acterized by a specific secretory phenotype, namely the SASP, that strated to be down-regulated in senescent cells [103,127], as well, in
consists of numerous inflammatory mediators, proteolytic enzymes, contrast to fibronectin, tenascin and laminin that have been found to be
cytokines, growth factors and even nucleic acids and proteins enclosed induced [103,146,148–152]. Regarding proteoglycans, they have been
in exosome-like small extracellular vesicles [87,102–104]. The SASP is mainly shown to be underexpressed and to display modifications con-
positively regulated by p38 MAPK [105] and nuclear factor κB (NF-κB) cerning their molecular size, binding properties, etc. in senescent fi-
[106], while it is dependent on the DDR proteins ATM, Nbs1 and Chk2, broblasts, chondrocytes, endothelial cells, VSMCs and IVD cells or in
but not on the cell cycle regulators p53 and pRb [107]. SASP has been vivo in the skin [132,134,153–158]. On the contrary, the cell surface
shown to be a conserved senescence-associated feature in several cell heparan sulfate proteoglycan syndecan-1 (SDC1) has been shown to be
types including cancer cells [103,108,109]. On one hand SASP pro- up-regulated in prematurely senescent human breast fibroblasts in vitro
motes tissue regeneration through the induction of plasticity and and in vivo [99]. Furthermore, hyaluronic acid biosynthesis - whose
stemness [110,111] and participates in the clearance of cancer cells by overexpression has been linked to the extended lifespan and resistance
attracting immune cells [112,113]; on the other hand it can establish an to cancer development of the naked mole-rat [159,160] - is reduced in
immunosuppressive, inflammatory and catabolic microenvironment senescent mesenchymal stem cells due to the down-regulation of hya-
and stimulate tumor growth and metastasis [95,114–120] (Fig. 2). In luronic acid synthases [161,162].
addition, SASP factors have been shown to reinforce senescence via Beyond quantitative changes in ECM components and degrading
autocrine and paracrine mechanisms [121], but also to affect adjacent enzymes, ECM produced by senescent cells also differentiates qualita-
cells and tissue homeostasis in a paracrine manner, even at great dis- tively and in terms of organization from that produced by young cells
tance causing a bystander effect [122–125]. [95,163]. More explicitly, the inflammatory phenotype of senescent
cells accompanied by changes in the levels of ECM components and
5. Senescence-associated changes in the extracellular matrix ECM-degrading molecules disturb elastin and collagen fiber networks
(ECM) and basal membrane integrity in the aged tissue. Additionally, it has
been shown that inappropriate collagen cross-linking observed during
It has been stated above that SASP comprises of numerous insoluble ageing renders ECM more rigid, less elastic and mechanically weaker
and soluble factors, including ECM components and proteolytic [95,163].

Fig. 2. Role of senescent cells in tumor progression. Various


factors trigger senescence of normal and cancer cells. Senescence
characterized by an irreversible growth arrest acts as an antitumor
barrier. On the other hand, the soluble and insoluble factors of the
senescence-associated secretory phenotype (SASP) stimulate
tumor growth and metastasis, but also trigger immune recognition
and tumor clearance.

3
E. Mavrogonatou, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

The great importance of the ECM interplay with cells has been va- morphogenesis [189]. Finally, co-injection of Nrf2-induced senescent
lidated by the reversal of the senescent phenotype of fibroblasts when fibroblasts with weakly malignant human squamous cells into the ear or
they were cultured onto ECM isolated from young cells. These cells the back skin of immunocompromised mice led to the development of
displayed signs of a youthful phenotype, i.e. a spindle-like morphology larger and more invasive tumors [125]. These senescent fibroblasts
and an actin structure pattern similar to that of young cells, lower SA-β- were found to deposit a senescence-promoting ECM, thus leading to a
Gal activity, reduced intracellular reactive oxygen species (ROS) levels, perpetuating series of events reinforcing senescence and promoting
increased novel DNA synthesis, improved mitochondrial function, a tumor growth [125].
better response to mitogenic signals like epidermal growth factor (EGF), Harking back to the tight relationship of the ECM with the mani-
decreased levels of p53, p21WAF1, p16INK4a and caveolin-1 and even festation of the major features of cancer discussed above, we should
longer telomeres [164]. When, in their turn, young cells were plated point out that an interrelation of the senescence-associated ECM al-
onto ECM isolated from senescent cells, their proliferation was only terations with the onset and progression of cancer could be also pro-
decelerated [164]. In contrast to this finding, young fibroblasts grown posed. As mentioned earlier, ECM in aged tissues and several age-re-
onto ECM produced by Nrf2-induced senescent cells not only showed a lated pathologies (where the accumulation of senescent cells has been
lower proliferation rate, but they also displayed significantly more SA- identified) is characterized by the loss of tensile strength and by in-
β-Gal activity compared to cells grown onto control ECM [125]. Ad- creased stiffness [95,163], which change the mechanical properties of
ditionally, it has been demonstrated that autologous decellularized the matrisome (i.e. all ECM proteins and associated factors) that serves
ECM protects adipose-derived stem cells against H2O2-induced senes- for epithelial growth and expansion and could thus rigorously interfere
cence and stimulates their proliferation in vitro [165]. with tumor progression.

6. The role of senescent cells in cancer progression 7. Therapy-induced senescence (TIS)

Since cancer has been considered as “a wound that never heals”, We have referred so far to replicative senescence and SIPS provoked
similarities in the implication of senescent cells in wound healing and in stromal fibroblasts due to the normal stressors cells routinely face.
cancer development are expected [166]. It has yet been well established What we should also take under consideration when discussing the
that transient presence of senescent cells is beneficial during normal implication of senescent cells and SASP in tumor development is the
tissue repair due to their anti-fibrotic phenotype [137,167–170], but therapy-induced senescence (TIS), a type of senescence occurring not
accumulation of senescent cells can be detrimental to local tissue only in normal stomal cells, but also in cancer cells during treatment of
homeostasis due to their pro-inflammatory properties [171–174]. In the disease. The TIS program is launched as a response to genotoxic
this vein, even though senescence is a potent tumorigenic barrier when chemotherapy and radiation and has indeed been demonstrated to be
initially induced [53,55,72,175–177], the persistent presence of se- adopted not only by the neighboring to the tumor healthy cells
nescent cells often allies with malignant cells and supports tumor ex- [86,99,190–192], but also by cancer cells themselves, depending on
pansion [178]. Indeed, accumulating senescent cells through their non- their molecular profile [177,193–202]. In detail, regarding che-
cell-autonomous activities become accomplice with cancer cells and are motherapeutic compounds, it has been shown that doxorubicin, eto-
main drivers of cancer initiation and progression [87,121,179], in- poside and cisplatin (which introduce double- or single-strand breaks
dicating that cellular senescence is a “double-edged sword” in cancer into the DNA) can lead normal human fibroblasts to premature senes-
development [180]. We have mentioned above that among all senes- cence [190,191,203] and that busulfan (which can cause DNA-DNA and
cence-associated traits, SASP is mainly culpable for this two-pronged DNA-protein cross-links) also results in the senescence of human fi-
role of cellular senescence. Although the specific outcome of the SASP is broblasts in a ROS-dependent manner [192,204]. On the other hand,
context-dependent, the net effect of its paracrine action in advanced chemotherapeutic drugs like doxorubicin, cisplatin, camphotecin (a
cancer is to directly enhance the proliferative and metastatic potential topoisomerase I inhibitor) and dicodermolide (which stabilizes micro-
of neoplastic cells or to indirectly allow them to expand by forming a tubules) have been shown to trigger accelerated senescence in several
permissive milieu, as it may locally result in tissue remodeling types of cancer cells, including colon carcinoma, fibrosarcoma, breast
[95,120]. In favor of this, it has been reported that stem cell senescence cancer, nasopharyngeal cancer and non-small cell lung cancer cells,
and a stem cell-associated SASP drive cell transformation and induction directly or through a bystander effect [195,197–199,201,205,206], a
of pituitary tumors in mice [181,182], while in the case of pediatric phenomenon that has been also verified in vivo [193,200]. Besides
pilocytic astrocytoma (PA), SASP has been shown to be a strong reg- chemotherapy, ionizing radiation used in radiotherapy has been shown
ulator of PA tumor growth by inducing OIS in proliferating PA cells to drive stromal fibroblasts [86,207,208] and cancer cells [202,209] to
through IL-1β [183]. IL-6 secreted from senescent mesenchymal stem premature senescence, as well. It is noteworthy that p53 and p16INK4a
cells promoted proliferation and migration of breast cancer cells [184] (known to be often mutated in tumors) are not always mandatory for
and IL-6 secreted by senescent osteoblasts increased breast cancer co- the manifestation of TIS in cancer cells [193–196,210].
lonization of the bone [116]. Senescence-associated alterations in the The SASP of stromal and cancer cells becoming senescent in the
production and secretion of matricellular proteins and ECM con- course of anticancer therapies has also been demonstrated to affect
stituents (hyaluronic acid, collagens, laminins and integrins) of prostate tumor progression and the therapeutic outcome. Hence, TIS has been
cells have been shown to create a favorable milieu for tumor develop- deservedly now considered a significant side-effect of anticancer
ment [185]. Furthermore, it has been demonstrated that senescent fi- treatments that should be taken into account when designing a parti-
broblasts promote the development of cancer from ovarian surface cular therapeutic scheme. Bleomycin-induced senescent human fibro-
epithelial cells at an early stage of neoplastic transformation [186], blasts enhanced the growth of co-transplanted breast cancer cells in
stimulate premalignant and malignant epithelial cells to proliferate in immunodeficient mice, which was diminished when MMP activity was
culture and to form tumors in vivo [98] and enhance the malignant inhibited [101]. Additionally, irradiation-induced senescent lung
progression of keratinocytes via MMP-1 and MMP-2 or ROS secretion stromal fibroblasts stimulated cancer cells’ growth both in vitro and in
[187,188]. This phenomenon has been in some cases attributed to vivo, partly through MMP up-regulation [86]. In contrast to the soluble
epithelial-to-mesenchymal transition (EMT) of cancer cells [98,187]. In SASP elements (and predominantly MMPs) that have been thoroughly
addition, premalignant mammary epithelial cells exposed to senescent investigated during carcinogenesis [87,120], to the best of our knowl-
human fibroblasts in vivo underwent full malignant transformation and edge the implication of the insoluble segment of the SASP in cancer
non-malignant breast epithelial cells co-cultured with senescent fibro- progression has been barely studied so far. Focusing on the insoluble
blasts in vitro showed an MMP-3-mediated disruption in their branching ECM components encompassed in SASP, we have recently shown that

4
E. Mavrogonatou, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

along with its profound catabolic character, the tumor-promoting senescent cells in various tissues [232,233], hence future therapies in-
phenotype of ionizing radiation-induced senescent human breast volving the modulation of the immune responses to favor senescent cell
stromal fibroblasts is filled in by the overexpression of SDC1 [99]. SDC1 clearance may be envisaged [234].
overexpression in the stroma is known to represent a poor prognostic
factor for the development of several types of cancer [99,211–216]. 9. Emerging senotherapeutics
Increased SDC1 levels in ionizing radiation-induced senescent breast
stromal fibroblasts have been found to be TGF-β-regulated via the Given that the persistent presence of senescent cells and their me-
SMAD pathway in collaboration with the transcription factor Sp1 [99]. tabolic products could impede remedy of cancer and thus prove detri-
Regarding therapy-induced senescent cancer cells, the supernatant of mental for rehabilitation, it becomes clear that inclusion of a se-
cisplatin-induced senescent A375 melanoma cells promoted the growth notherapeutic arm (i.e. a mode to specifically target senescent cells
of non-senescent A375 cells and co-transplantation of senescent with and/or their metabolic products) should perhaps be considered in the
non-senescent A375 cells resulted in accelerated tumor growth into design of novel anticancer treatments. As mentioned above, in certain
nude mice, with IL-1α and IL-8 being the key SASP factors regulating cases clearance of senescent cells is achieved by the immune system,
these events [217]. Finally, senescent tumor cells can contribute to thus limiting their detrimental effects [137]. Moreover, genetic ap-
cancer recurrence and increased mortality rates, since they are able to proaches have been developed to totally eliminate senescent cells from
retrieve their proliferative potential after a period of dormancy [218]. the organism of experimental animals, hence preventing tissue dys-
In favor of this, it has been reported that p53- and p16INK4a-deficient function and even extending lifespan [235,236]. Since, however, such
human lung cancer cells can bypass chemotherapy-induced accelerated approaches cannot be applied to humans, in the last five years an in-
senescence and resume cell cycle progression [210]. It has been also tensive search for senolytics has begun, i.e. for natural products or
proposed that the SASP of persisting therapy-induced senescent cells in synthetic compounds capable for selectively destructing senescent cells
the microenvironment of the tumor may drive escape from senescence without affecting non-senescent ones. The first senolytic compounds
and re-entry of malignant cells into proliferation, while it provokes reported were the natural product (flavonoid) quercetin and the tyr-
local and systemic inflammation [218–221]. These deleterious out- osine kinase inhibitor dasatinib [237]. The rationale behind their dis-
comes may often exacerbate the side-effects of anticancer treatments covery is related to the increased resistance of senescent cells to
and even allow cancer relapse, which could justify poor prognosis in apoptosis [78]; indeed quercetin among a plethora of other biological
patients expressing senescence markers after treatment with neoadju- effects inhibits the PI3K/Akt/mTOR pro-survival signaling pathway,
vant chemotherapy [222,223]. while dasatinib interferes with the Ephrin B ligands, known to prevent
apoptosis [237,238]. Similarly, other reported senolytics include the
8. Clearance of senescent cells in vivo small molecule inhibitors navitoclax (or ABT-263) and ABT-737 that
target the BCL-2/BCL-W/BCL-XL family of anti-apoptotic proteins
Having in mind the pro-inflammatory character of SASP, one could [239–241], the relatively selective against BCL-XL inhibitors A1331852
anticipate the elimination of senescent cells through immune system and A1155463, as well as, the flavone fisetin that is related to quercetin
responses. Indeed, in liver carcinomas of p53-deficient mice, the re- [242], the natural product piperlongumine and its synthetic analogues
activation of p53 was observed to activate a premature senescence that target the cellular oxidative stress sensor oxidation resistance 1
program and an up-regulation of inflammatory cytokines, leading to the (OXR1) [243–245], the synthetic FOXO4 D-Retro-Inverso peptide that
triggering of an innate immune response, elimination of senescent cells disrupts the FOXO4-p53 interaction [246], and geldanamycin and its
and eventually tumor regression [112]. Moreover, in mice with liver semi-synthetic derivative 17-dimethylaminoethylamino-17-demethox-
damage, normal hepatic cells are activated, proliferate and overexpress ygeldanamycin (17-DMAG) that inhibit Heat-Shock Protein-90 (HSP-
ECM molecules towards tissue repair. Persistence of this effect may lead 90) thereby destabilizing Akt and phospho-Akt [247]. Interestingly,
to fibrosis, however the activated cells enter senescence, thereby fa- senolytics may also target cancer cells led to senescence due to standard
cilitating the resolution of fibrosis due to their catabolic phenotype and, chemotherapy, thereby augmenting therapy, as shown for the histone
moreover, they attract natural killer (NK) cells effecting senescent cells’ deacetylase inhibitor panobinostat [248]. An important observation
clearance. In contrast, in mice lacking both p53 and p16INK4a (hence from all these reports was that the effects of all verified senolytics up to
their liver cells are unable to undergo senescence) the same treatment now appear to be cell-type dependent [238,249]. Notably, the dasa-
causes severe fibrosis [137]. Especially in the case of OIS in hepato- tinib-quercetin senolytic cocktail was found to be ineffective in an an-
cytes, it has been shown that the latter secrete soluble factors triggering imal model of hepatocellular carcinoma, alone or in synergy with se-
their immune-mediated clearance, a phenomenon termed “senescence nescence-inducing chemotherapy [250]. Moreover, the in vivo
surveillance”, while if these pre-malignant senescent hepatocytes are application of senolytics may be accompanied by toxic side-effects,
not eliminated, murine hepatocellular carcinomas are being developed especially if they target central anti-apoptotic cellular mechanisms
[224]. In this vein, senescent HSCs, beyond their inability to proliferate [251]. As a consequence, the use of senomorphics may be preferable,
per se, promote also an antitumor microenvironment through paracrine i.e. compounds modulating the catabolic and/or inflammatory pheno-
factors that - in a p53-dependent manner - facilitates macrophage po- type of senescent cells [252]. In this direction, many substances sup-
larization toward the M1-state, which enables senescent cell elimina- pressing the SASP have been identified, most notably interfering with
tion [225]. A mechanism underlying senescence surveillance includes the NF-κB pathway, like various flavonoids, such as apigenin and
the overexpression by senescent cells of ligands for an activating NK cell kaempferol [253] and metformin [254], or inhibiting JAK (ruxolitinib
receptor (NKG2D) as a result of DDR [226]. The importance of the [255]), or generally conveying anti-inflammatory and antioxidant ef-
immune system in limiting senescent cell accumulation in various tis- fects, such as glucocorticoids [256], rapamycin [257] or resveratrol and
sues has been shown in mice lacking perforin, an important mediator of its analogues [258]. Notably the latter seem to exert their senomorphic
immune cytotoxicity [227]. These mice in turn develop chronic in- effects through modulation of splicing factors, in accordance with the
flammation, a common denominator in most age-related diseases recent finding that knocking down the expression of the alternative
[228]. Regarding tumor development, it seems that the combination of splicing regulator polypyrimidine tract binding protein 1 (PTBP1) in-
cancer cell senescence with immune system recognition can be bene- hibits the SASP of senescent hepatocytes and, most importantly, its pro-
ficial, hence cytokine-induced “bystander” senescence has been sug- tumorigenic effects [259]. Of course, SASP suppression may also have
gested as a promising novel therapeutic modality [229–231]. In this “off-target” effects [260] and could theoretically even impair clearance
direction senescence of the immune system itself, i.e. “im- of preneoplastic cells by the immune system [259]. In fact, many of the
munosenescence”, may play an important role for the accumulation of senolytic and semomorphic compounds are long known and have been

5
E. Mavrogonatou, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

tested in various clinical trials, and some of them were initially iden- [16] C.C. Park, M.J. Bissell, M.H. Barcellos-Hoff, The influence of the microenviron-
tified as anticancer drugs [238]. Moreover, depleting senescent cells in ment on the malignant phenotype, Mol. Med. Today 6 (2000) 324–329.
[17] M.J. Bissell, D.C. Radisky, A. Rizki, V.M. Weaver, O.W. Petersen, The organizing
pre-clinical studies has been shown to successfully treat chronic con- principle: microenvironmental influences in the normal and malignant breast,
ditions, such as frailty, cardiac dysfunction, vascular hyporeactivity and Differentiation 70 (2002) 537–546.
calcification, diabetes, liver steatosis, osteoporosis, IVD degeneration, [18] P.A. Kenny, M.J. Bissell, Tumor reversion: correction of malignant behavior by
microenvironmental cues, Int. J. Cancer 107 (2003) 688–695.
pulmonary fibrosis, and radiation-induced damage [261], while a re- [19] D.A. Beacham, E. Cukierman, Stromagenesis: the changing face of fibroblastic
cent in vivo study in mice, where senescent cells’ transplantation was microenvironments during tumor progression, Semin. Cancer Biol. 15 (2005)
shown to cause physical dysfunction and decreased survival, indicated 329–341.
[20] L.M. Coussens, Z. Werb, Inflammation and cancer, Nature 420 (2002) 860–867.
also that senolytics can alleviate physical dysfunction and increase late- [21] A. Calon, D.V. Tauriello, E. Batlle, TGF-beta in CAF-mediated tumor growth and
life survival without extending morbidity [262]. metastasis, Semin. Cancer Biol. 25 (2014) 15–22.
[22] N.A. Bhowmick, A. Chytil, D. Plieth, A.E. Gorska, N. Dumont, S. Shappell, et al.,
TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent
10. Concluding remarks
epithelia, Science 303 (2004) 848–851.
[23] E.A. Ebbing, A.P. van der Zalm, A. Steins, A. Creemers, S. Hermsen, R. Rentenaar,
Since senescent cells - occuring also as a side-effect of genotoxic et al., Stromal-derived interleukin 6 drives epithelial-to-mesenchymal transition
anticancer treatments - may create a favorable milieu for tumor ex- and therapy resistance in esophageal adenocarcinoma, Proc. Natl. Acad. Sci. U. S.
A. 116 (2019) 2237–2242.
pansion or even assist malignant cells to escape and promote cancer [24] J. D’Armiento, T. DiColandrea, S.S. Dalal, Y. Okada, M.T. Huang, A.H. Conney,
relapse, it seems crucial that future anticancer therapies will not only et al., Collagenase expression in transgenic mouse skin causes hyperkeratosis and
target neoplastic cells, but they will also aim at the elimination of ac- acanthosis and increases susceptibility to tumorigenesis, Mol. Cell. Biol. 15 (1995)
5732–5739.
cumulating senescent cells or the modification of their senescence-as- [25] C.J. Sympson, M.J. Bissell, Z. Werb, Mammary gland tumor formation in trans-
sociated secretome. Given the great heterogeneity in various cancer genic mice overexpressing stromelysin-1, Semin. Cancer Biol. 6 (1995) 159–163.
types, a general therapeutic scheme could not always be applied and [26] C.L. Wilson, K.J. Heppner, P.A. Labosky, B.L. Hogan, L.M. Matrisian, Intestinal
tumorigenesis is suppressed in mice lacking the metalloproteinase matrilysin,
personalized optimization may be necessary towards an effective cancer Proc. Natl. Acad. Sci. U. S. A. 94 (1997) 1402–1407.
remedy. [27] R. Masson, O. Lefebvre, A. Noel, M.E. Fahime, M.P. Chenard, C. Wendling, et al.,
In vivo evidence that the stromelysin-3 metalloproteinase contributes in a para-
crine manner to epithelial cell malignancy, J. Cell Biol. 140 (1998) 1535–1541.
Acknowledgements [28] M.D. Sternlicht, M.J. Bissell, Z. Werb, The matrix metalloproteinase stromelysin-1
acts as a natural mammary tumor promoter, Oncogene 19 (2000) 1102–1113.
This work was partly supported by the EU Horizon 2020 project [29] M.D. Sternlicht, A. Lochter, C.J. Sympson, B. Huey, J.P. Rougier, J.W. Gray, et al.,
The stromal proteinase MMP3/stromelysin-1 promotes mammary carcinogenesis,
MSCA-RISE-2014, action No. 645756 “GLYCANC - Matrix glycans as
Cell 98 (1999) 137–146.
multifunctional pathogenesis factors and therapeutic targets in cancer” [30] M. Egeblad, Z. Werb, New functions for the matrix metalloproteinases in cancer
and by the project “Target Identification and Development of Novel progression, Nat. Rev. Cancer 2 (2002) 161–174.
Approaches for Health and Environmental Applications” (MIS [31] V.L. Veenstra, H. Damhofer, C. Waasdorp, L.B. van Rijssen, M.J. van de Vijver,
F. Dijk, et al., ADAM12 is a circulating marker for stromal activation in pancreatic
5002514), which is implemented under the Action for the Strategic cancer and predicts response to chemotherapy, Oncogenesis 7 (2018) 87.
Development on the Research and Technological Sectors, funded by the [32] M.W. Pickup, J.K. Mouw, V.M. Weaver, The extracellular matrix modulates the
Operational Programme "Competitiveness, Entrepreneurship and hallmarks of cancer, EMBO Rep. 15 (2014) 1243–1253.
[33] T. Finkel, M. Serrano, M.A. Blasco, The common biology of cancer and ageing,
Innovation" (NSRF 2014-2020) and co-financed by Greece and the Nature 448 (2007) 767–774.
European Union (European Regional Development Fund). [34] B. Milholland, A. Auton, Y. Suh, J. Vijg, Age-related somatic mutations in the
cancer genome, Oncotarget 6 (2015) 24627–24635.
[35] S. Yamashita, T. Kishino, T. Takahashi, T. Shimazu, H. Charvat, Y. Kakugawa,
References et al., Genetic and epigenetic alterations in normal tissues have differential im-
pacts on cancer risk among tissues, Proc. Natl. Acad. Sci. U. S. A. 115 (2018)
[1] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144 1328–1333.
(2011) 646–674. [36] V. Calvanese, E. Lara, A. Kahn, M.F. Fraga, The role of epigenetics in aging and
[2] B. Elenbaas, R.A. Weinberg, Heterotypic signaling between epithelial tumor cells age-related diseases, Ageing Res. Rev. 8 (2009) 268–276.
and fibroblasts in carcinoma formation, Exp. Cell Res. 264 (2001) 169–184. [37] B. Brouwers, D. Fumagalli, S. Brohee, S. Hatse, O. Govaere, G. Floris, et al., The
[3] D.C. Radisky, M.J. Bissell, Cancer. Respect thy neighbor!, Science 303 (2004) footprint of the ageing stroma in older patients with breast cancer, Breast Cancer
775–777. Res. 19 (2017) 78.
[4] A. Elkhattouti, M. Hassan, C.R. Gomez, Stromal fibroblast in age-related cancer: [38] L. Hayflick, P.S. Moorhead, The serial cultivation of human diploid cell strains,
role in tumorigenesis and potential as novel therapeutic target, Front. Oncol. 5 Exp. Cell Res. 25 (1961) 585–621.
(2015) 158. [39] D. Kletsas, Cellular Senescence and Cancer Development: Antagonistic and
[5] T.D. Tlsty, P.W. Hein, Know thy neighbor: stromal cells can contribute oncogenic Synergistic Relations, (2012), pp. 209–232.
signals, Curr. Opin. Genet. Dev. 11 (2001) 54–59. [40] C.B. Harley, A.B. Futcher, C.W. Greider, Telomeres shorten during ageing of
[6] C.J. Baglole, D.M. Ray, S.H. Bernstein, S.E. Feldon, T.J. Smith, P.J. Sime, et al., human fibroblasts, Nature 345 (1990) 458–460.
More than structural cells, fibroblasts create and orchestrate the tumor micro- [41] A.M. Olovnikov, A theory of marginotomy. The incomplete copying of template
environment, Immunol. Invest. 35 (2006) 297–325. margin in enzymic synthesis of polynucleotides and biological significance of the
[7] A. Orimo, R.A. Weinberg, Stromal fibroblasts in cancer: a novel tumor-promoting phenomenon, J. Theor. Biol. 41 (1973) 181–190.
cell type, Cell Cycle 5 (2006) 1597–1601. [42] L. Hayflick, The limited in vitro lifetime of human diploid cell strains, Exp. Cell
[8] P.A. Kenny, G.Y. Lee, M.J. Bissell, Targeting the tumor microenvironment, Front. Res. 37 (1965) 614–636.
Biosci. 12 (2007) 3468–3474. [43] J.W. Shay, W.E. Wright, Hallmarks of telomeres in ageing research, J. Pathol. 211
[9] W. Yang, W. Han, S. Ye, D. Liu, J. Wu, H. Liu, et al., Fibroblast activation protein- (2007) 114–123.
alpha promotes ovarian cancer cell proliferation and invasion via extracellular and [44] A.G. Bodnar, M. Ouellette, M. Frolkis, S.E. Holt, C.P. Chiu, G.B. Morin, et al.,
intracellular signaling mechanisms, Exp. Mol. Pathol. 95 (2013) 105–110. Extension of life-span by introduction of telomerase into normal human cells,
[10] N.A. Bhowmick, E.G. Neilson, H.L. Moses, Stromal fibroblasts in cancer initiation Science 279 (1998) 349–352.
and progression, Nature 432 (2004) 332–337. [45] U. Herbig, W.A. Jobling, B.P. Chen, D.J. Chen, J.M. Sedivy, Telomere shortening
[11] H. Kiaris, G. Trimis, A.G. Papavassiliou, Regulation of tumor-stromal fibroblast triggers senescence of human cells through a pathway involving ATM, p53, and
interactions: implications in anticancer therapy, Curr. Med. Chem. 15 (2008) p21(CIP1), but not p16(INK4a), Mol. Cell 14 (2004) 501–513.
3062–3067. [46] J. Campisi, F. d’Adda di Fagagna, Cellular senescence: when bad things happen to
[12] R.M. Bremnes, T. Donnem, S. Al-Saad, K. Al-Shibli, S. Andersen, R. Sirera, et al., good cells, Nat. Rev. Mol. Cell Biol. 8 (2007) 729–740.
The role of tumor stroma in cancer progression and prognosis: emphasis on car- [47] F. d’Adda di Fagagna, P.M. Reaper, L. Clay-Farrace, H. Fiegler, P. Carr, T. Von
cinoma-associated fibroblasts and non-small cell lung cancer, J. Thorac. Oncol. 6 Zglinicki, et al., A DNA damage checkpoint response in telomere-initiated senes-
(2011) 209–217. cence, Nature 426 (2003) 194–198.
[13] R. Kalluri, M. Zeisberg, Fibroblasts in cancer, Nat. Rev. Cancer 6 (2006) 392–401. [48] C. Lopez-Otin, M.A. Blasco, L. Partridge, M. Serrano, G. Kroemer, The hallmarks of
[14] K. Rasanen, A. Vaheri, Activation of fibroblasts in cancer stroma, Exp. Cell Res. aging, Cell 153 (2013) 1194–1217.
316 (2010) 2713–2722. [49] O. Toussaint, V. Royer, M. Salmon, J. Remacle, Stress-induced premature senes-
[15] O. De Wever, P. Demetter, M. Mareel, M. Bracke, Stromal myofibroblasts are cence and tissue ageing, Biochem. Pharmacol. 64 (2002) 1007–1009.
drivers of invasive cancer growth, Int. J. Cancer 123 (2008) 2229–2238. [50] C.J. Sherr, R.A. DePinho, Cellular senescence: mitotic clock or culture shock? Cell

6
E. Mavrogonatou, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

102 (2000) 407–410. and aging, Front. Genet. 8 (2017) 138.


[51] M. Serrano, A.W. Lin, M.E. McCurrach, D. Beach, S.W. Lowe, Oncogenic ras pro- [83] F. Rodier, D.P. Munoz, R. Teachenor, V. Chu, O. Le, D. Bhaumik, et al., DNA-
vokes premature cell senescence associated with accumulation of p53 and SCARS: distinct nuclear structures that sustain damage-induced senescence growth
p16INK4a, Cell 88 (1997) 593–602. arrest and inflammatory cytokine secretion, J. Cell. Sci. 124 (2011) 68–81.
[52] O. Toussaint, E.E. Medrano, T. von Zglinicki, Cellular and molecular mechanisms [84] M. Narita, S. Nunez, E. Heard, M. Narita, A.W. Lin, S.A. Hearn, et al., Rb-mediated
of stress-induced premature senescence (SIPS) of human diploid fibroblasts and heterochromatin formation and silencing of E2F target genes during cellular se-
melanocytes, Exp. Gerontol. 35 (2000) 927–945. nescence, Cell 113 (2003) 703–716.
[53] C. Michaloglou, L.C. Vredeveld, M.S. Soengas, C. Denoyelle, T. Kuilman, C.M. van [85] E.C. Swanson, B. Manning, H. Zhang, J.B. Lawrence, Higher-order unfolding of
der Horst, et al., BRAFE600-associated senescence-like cell cycle arrest of human satellite heterochromatin is a consistent and early event in cell senescence, J. Cell
naevi, Nature 436 (2005) 720–724. Biol. 203 (2013) 929–942.
[54] V.G. Gorgoulis, T.D. Halazonetis, Oncogene-induced senescence: the bright and [86] A. Papadopoulou, D. Kletsas, Human lung fibroblasts prematurely senescent after
dark side of the response, Curr. Opin. Cell Biol. 22 (2010) 816–827. exposure to ionizing radiation enhance the growth of malignant lung epithelial
[55] J. Bartkova, N. Rezaei, M. Liontos, P. Karakaidos, D. Kletsas, N. Issaeva, et al., cells in vitro and in vivo, Int. J. Oncol. 39 (2011) 989–999.
Oncogene-induced senescence is part of the tumorigenesis barrier imposed by [87] J.P. Coppe, C.K. Patil, F. Rodier, Y. Sun, D.P. Munoz, J. Goldstein, et al.,
DNA damage checkpoints, Nature 444 (2006) 633–637. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions
[56] C.J. Sarkisian, B.A. Keister, D.B. Stairs, R.B. Boxer, S.E. Moody, L.A. Chodosh, of oncogenic RAS and the p53 tumor suppressor, PLoS Biol. 6 (2008) 2853–2868.
Dose-dependent oncogene-induced senescence in vivo and its evasion during [88] G.P. Dimri, X. Lee, G. Basile, M. Acosta, G. Scott, C. Roskelley, et al., A biomarker
mammary tumorigenesis, Nat. Cell Biol. 9 (2007) 493–505. that identifies senescent human cells in culture and in aging skin in vivo, Proc.
[57] M. Fumagalli, F. Rossiello, M. Clerici, S. Barozzi, D. Cittaro, J.M. Kaplunov, et al., Natl. Acad. Sci. U. S. A. 92 (1995) 9363–9367.
Telomeric DNA damage is irreparable and causes persistent DNA-damage-response [89] B.Y. Lee, J.A. Han, J.S. Im, A. Morrone, K. Johung, E.C. Goodwin, et al.,
activation, Nat. Cell Biol. 14 (2012) 355–365. Senescence-associated beta-galactosidase is lysosomal beta-galactosidase, Aging
[58] Q. Chen, A. Fischer, J.D. Reagan, L.J. Yan, B.N. Ames, Oxidative DNA damage and Cell 5 (2006) 187–195.
senescence of human diploid fibroblast cells, Proc. Natl. Acad. Sci. U. S. A. 92 [90] J. Severino, R.G. Allen, S. Balin, A. Balin, V.J. Cristofalo, Is beta-galactosidase
(1995) 4337–4341. staining a marker of senescence in vitro and in vivo? Exp. Cell Res. 257 (2000)
[59] A. Di Leonardo, S.P. Linke, K. Clarkin, G.M. Wahl, DNA damage triggers a pro- 162–171.
longed p53-dependent G1 arrest and long-term induction of Cip1 in normal human [91] V.J. Cristofalo, SA beta Gal staining: biomarker or delusion, Exp. Gerontol. 40
fibroblasts, Genes Dev. 8 (1994) 2540–2551. (2005) 836–838.
[60] R. Di Micco, M. Fumagalli, A. Cicalese, S. Piccinin, P. Gasparini, C. Luise, et al., [92] N.C. Yang, M.L. Hu, The limitations and validities of senescence associated-beta-
Oncogene-induced senescence is a DNA damage response triggered by DNA hyper- galactosidase activity as an aging marker for human foreskin fibroblast Hs68 cells,
replication, Nature 444 (2006) 638–642. Exp. Gerontol. 40 (2005) 813–819.
[61] J.W. Shay, I.B. Roninson, Hallmarks of senescence in carcinogenesis and cancer [93] K. Evangelou, N. Lougiakis, S.V. Rizou, A. Kotsinas, D. Kletsas, D. Munoz-Espin,
therapy, Oncogene 23 (2004) 2919–2933. et al., Robust, universal biomarker assay to detect senescent cells in biological
[62] F.A. Mallette, M.F. Gaumont-Leclerc, G. Ferbeyre, The DNA damage signaling specimens, Aging Cell 16 (2017) 192–197.
pathway is a critical mediator of oncogene-induced senescence, Genes Dev. 21 [94] E.A. Georgakopoulou, K. Tsimaratou, K. Evangelou, P.J. Fernandez Marcos,
(2007) 43–48. V. Zoumpourlis, I.P. Trougakos, et al., Specific lipofuscin staining as a novel
[63] J.H. Chen, K. Stoeber, S. Kingsbury, S.E. Ozanne, G.H. Williams, C.N. Hales, Loss biomarker to detect replicative and stress-induced senescence. A method applic-
of proliferative capacity and induction of senescence in oxidatively stressed able in cryo-preserved and archival tissues, Aging (Albany NY) 5 (2013) 37–50.
human fibroblasts, J. Biol. Chem. 279 (2004) 49439–49446. [95] E. Mavrogonatou, H. Pratsinis, A. Papadopoulou, N.K. Karamanos, D. Kletsas,
[64] G.H. Stein, L.F. Drullinger, R.S. Robetorye, O.M. Pereira-Smith, J.R. Smith, Extracellular matrix alterations in senescent cells and their significance in tissue
Senescent cells fail to express cdc2, cycA, and cycB in response to mitogen sti- homeostasis, Matrix Biol. 75–76 (2019) 27–42.
mulation, Proc. Natl. Acad. Sci. U. S. A. 88 (1991) 11012–11016. [96] A. Hernandez-Segura, J. Nehme, M. Demaria, Hallmarks of cellular senescence,
[65] V.V. Ogryzko, T.H. Hirai, V.R. Russanova, D.A. Barbie, B.H. Howard, Human fi- Trends Cell Biol. 28 (2018) 436–453.
broblast commitment to a senescence-like state in response to histone deacetylase [97] J.M. van Deursen, The role of senescent cells in ageing, Nature 509 (2014)
inhibitors is cell cycle dependent, Mol. Cell. Biol. 16 (1996) 5210–5218. 439–446.
[66] Z. Mao, Z. Ke, V. Gorbunova, A. Seluanov, Replicatively senescent cells are ar- [98] A. Krtolica, S. Parrinello, S. Lockett, P.Y. Desprez, J. Campisi, Senescent fibroblasts
rested in G1 and G2 phases, Aging (Albany NY) 4 (2012) 431–435. promote epithelial cell growth and tumorigenesis: a link between cancer and
[67] A.J. Brenner, M.R. Stampfer, C.M. Aldaz, Increased p16 expression with first se- aging, Proc. Natl. Acad. Sci. U. S. A. 98 (2001) 12072–12077.
nescence arrest in human mammary epithelial cells and extended growth capacity [99] E. Liakou, E. Mavrogonatou, H. Pratsinis, S. Rizou, K. Evangelou, P.N. Panagiotou,
with p16 inactivation, Oncogene 17 (1998) 199–205. et al., Ionizing radiation-mediated premature senescence and paracrine interac-
[68] T. Kuilman, C. Michaloglou, W.J. Mooi, D.S. Peeper, The essence of senescence, tions with cancer cells enhance the expression of syndecan 1 in human breast
Genes Dev. 24 (2010) 2463–2479. stromal fibroblasts: the role of TGF-beta, Aging (Albany NY) 8 (2016) 1650–1669.
[69] D.A. Alcorta, Y. Xiong, D. Phelps, G. Hannon, D. Beach, J.C. Barrett, Involvement [100] A. Krtolica, J. Campisi, Cancer and aging: a model for the cancer promoting effects
of the cyclin-dependent kinase inhibitor p16 (INK4a) in replicative senescence of of the aging stroma, Int. J. Biochem. Cell Biol. 34 (2002) 1401–1414.
normal human fibroblasts, Proc. Natl. Acad. Sci. U. S. A. 93 (1996) 13742–13747. [101] D. Liu, P.J. Hornsby, Senescent human fibroblasts increase the early growth of
[70] P. Atadja, H. Wong, I. Garkavtsev, C. Veillette, K. Riabowol, Increased activity of xenograft tumors via matrix metalloproteinase secretion, Cancer Res. 67 (2007)
p53 in senescing fibroblasts, Proc. Natl. Acad. Sci. U. S. A. 92 (1995) 8348–8352. 3117–3126.
[71] C.M. Beausejour, A. Krtolica, F. Galimi, M. Narita, S.W. Lowe, P. Yaswen, et al., [102] A.R. Davalos, J.P. Coppe, J. Campisi, P.Y. Desprez, Senescent cells as a source of
Reversal of human cellular senescence: roles of the p53 and p16 pathways, EMBO inflammatory factors for tumor progression, Cancer Metastasis Rev. 29 (2010)
J. 22 (2003) 4212–4222. 273–283.
[72] Z. Chen, L.C. Trotman, D. Shaffer, H.K. Lin, Z.A. Dotan, M. Niki, et al., Crucial role [103] D.N. Shelton, E. Chang, P.S. Whittier, D. Choi, W.D. Funk, Microarray analysis of
of p53-dependent cellular senescence in suppression of Pten-deficient tumor- replicative senescence, Curr. Biol. 9 (1999) 939–945.
igenesis, Nature 436 (2005) 725–730. [104] M. Takasugi, R. Okada, A. Takahashi, D. Virya Chen, S. Watanabe, E. Hara, Small
[73] J. Krishnamurthy, C. Torrice, M.R. Ramsey, G.I. Kovalev, K. Al-Regaiey, L. Su, extracellular vesicles secreted from senescent cells promote cancer cell prolifera-
et al., Ink4a/Arf expression is a biomarker of aging, J. Clin. Invest. 114 (2004) tion through EphA2, Nat. Commun. 8 (2017) 15729.
1299–1307. [105] A. Freund, C.K. Patil, J. Campisi, p38MAPK is a novel DNA damage response-
[74] S. Lee, C.A. Schmitt, The dynamic nature of senescence in cancer, Nat. Cell Biol. 21 independent regulator of the senescence-associated secretory phenotype, EMBO J.
(2019) 94–101. 30 (2011) 1536–1548.
[75] D. Kletsas, Aging of fibroblasts, in: S.C. Kaul, R. Wadhwa (Eds.), Aging of Cells in [106] A. Salminen, A. Kauppinen, K. Kaarniranta, Emerging role of NF-kappaB signaling
and Outside the Body, Springer, Netherlands, Dordrecht, 2003, pp. 27–46. in the induction of senescence-associated secretory phenotype (SASP), Cell. Signal.
[76] J. Kaplon, L. Zheng, K. Meissl, B. Chaneton, V.A. Selivanov, G. Mackay, et al., A 24 (2012) 835–845.
key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-in- [107] F. Rodier, J.P. Coppe, C.K. Patil, W.A. Hoeijmakers, D.P. Munoz, S.R. Raza, et al.,
duced senescence, Nature 498 (2013) 109–112. Persistent DNA damage signalling triggers senescence-associated inflammatory
[77] J.R. Dorr, Y. Yu, M. Milanovic, G. Beuster, C. Zasada, J.H. Dabritz, et al., Synthetic cytokine secretion, Nat. Cell Biol. 11 (2009) 973–979.
lethal metabolic targeting of cellular senescence in cancer therapy, Nature 501 [108] B. Schnabl, C.A. Purbeck, Y.H. Choi, C.H. Hagedorn, D. Brenner, Replicative se-
(2013) 421–425. nescence of activated human hepatic stellate cells is accompanied by a pro-
[78] E. Wang, Senescent human fibroblasts resist programmed cell death, and failure to nounced inflammatory but less fibrogenic phenotype, Hepatology 37 (2003)
suppress bcl2 is involved, Cancer Res. 55 (1995) 2284–2292. 653–664.
[79] B. Hampel, F. Malisan, H. Niederegger, R. Testi, P. Jansen-Durr, Differential reg- [109] S.R. Schwarze, V.X. Fu, J.A. Desotelle, M.L. Kenowski, D.F. Jarrard, The identifi-
ulation of apoptotic cell death in senescent human cells, Exp. Gerontol. 39 (2004) cation of senescence-specific genes during the induction of senescence in prostate
1713–1721. cancer cells, Neoplasia 7 (2005) 816–823.
[80] A. Freund, R.M. Laberge, M. Demaria, J. Campisi, Lamin B1 loss is a senescence- [110] L. Mosteiro, C. Pantoja, N. Alcazar, R.M. Marion, D. Chondronasiou, M. Rovira,
associated biomarker, Mol. Biol. Cell 23 (2012) 2066–2075. et al., Tissue damage and senescence provide critical signals for cellular repro-
[81] T. Shimi, V. Butin-Israeli, S.A. Adam, R.B. Hamanaka, A.E. Goldman, C.A. Lucas, gramming in vivo, Science 354 (2016).
et al., The role of nuclear lamin B1 in cell proliferation and senescence, Genes Dev. [111] B. Ritschka, M. Storer, A. Mas, F. Heinzmann, M.C. Ortells, J.P. Morton, et al., The
25 (2011) 2579–2593. senescence-associated secretory phenotype induces cellular plasticity and tissue
[82] C. Sidler, O. Kovalchuk, I. Kovalchuk, Epigenetic regulation of cellular senescence regeneration, Genes Dev. 31 (2017) 172–183.

7
E. Mavrogonatou, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

[112] W. Xue, L. Zender, C. Miething, R.A. Dickins, E. Hernando, V. Krizhanovsky, et al., [139] Y. Liu, I. Drozdov, R. Shroff, L.E. Beltran, C.M. Shanahan, Prelamin A accelerates
Senescence and tumour clearance is triggered by p53 restoration in murine liver vascular calcification via activation of the DNA damage response and senescence-
carcinomas, Nature 445 (2007) 656–660. associated secretory phenotype in vascular smooth muscle cells, Circ. Res. 112
[113] M. Hoare, Y. Ito, T.W. Kang, M.P. Weekes, N.J. Matheson, D.A. Patten, et al., (2013) e99–109.
NOTCH1 mediates a switch between two distinct secretomes during senescence, [140] A.D. Theocharis, S.S. Skandalis, C. Gialeli, N.K. Karamanos, Extracellular matrix
Nat. Cell Biol. 18 (2016) 979–992. structure, Adv. Drug Deliv. Rev. 97 (2016) 4–27.
[114] R.M. Laberge, Y. Sun, A.V. Orjalo, C.K. Patil, A. Freund, L. Zhou, et al., MTOR [141] H. Kamino, M. Hiratsuka, T. Toda, R. Nishigaki, M. Osaki, H. Ito, et al., Searching
regulates the pro-tumorigenic senescence-associated secretory phenotype by pro- for genes involved in arteriosclerosis: proteomic analysis of cultured human um-
moting IL1A translation, Nat. Cell Biol. 17 (2015) 1049–1061. bilical vein endothelial cells undergoing replicative senescence, Cell Struct. Funct.
[115] D. Eyman, M. Damodarasamy, S.R. Plymate, M.J. Reed, CCL5 secreted by senes- 28 (2003) 495–503.
cent aged fibroblasts induces proliferation of prostate epithelial cells and expres- [142] M.D. West, J.W. Shay, W.E. Wright, M.H. Linskens, Altered expression of plas-
sion of genes that modulate angiogenesis, J. Cell. Physiol. 220 (2009) 376–381. minogen activator and plasminogen activator inhibitor during cellular senescence,
[116] X. Luo, Y. Fu, A.J. Loza, B. Murali, K.M. Leahy, M.K. Ruhland, et al., Stromal- Exp. Gerontol. 31 (1996) 175–193.
initiated changes in the bone promote metastatic niche development, Cell Rep. 14 [143] P. Comi, R. Chiaramonte, J.A. Maier, Senescence-dependent regulation of type 1
(2016) 82–92. plasminogen activator inhibitor in human vascular endothelial cells, Exp. Cell Res.
[117] P.D. Angelini, M.F. Zacarias Fluck, K. Pedersen, J.L. Parra-Palau, M. Guiu, 219 (1995) 304–308.
C. Bernado Morales, et al., Constitutive HER2 signaling promotes breast cancer [144] G. Untergasser, H.B. Koch, A. Menssen, H. Hermeking, Characterization of epi-
metastasis through cellular senescence, Cancer Res. 73 (2013) 450–458. thelial senescence by serial analysis of gene expression: identification of genes
[118] M.K. Ruhland, A.J. Loza, A.H. Capietto, X. Luo, B.L. Knolhoff, K.C. Flanagan, et al., potentially involved in prostate cancer, Cancer Res. 62 (2002) 6255–6262.
Stromal senescence establishes an immunosuppressive microenvironment that [145] R.M. Kortlever, P.J. Higgins, R. Bernards, Plasminogen activator inhibitor-1 is a
drives tumorigenesis, Nat. Commun. 7 (2016) 11762. critical downstream target of p53 in the induction of replicative senescence, Nat.
[119] T. Eggert, K. Wolter, J. Ji, C. Ma, T. Yevsa, S. Klotz, et al., Distinct functions of Cell Biol. 8 (2006) 877–884.
senescence-associated immune responses in liver tumor surveillance and tumor [146] H. Yin, J.G. Pickering, Cellular senescence and vascular disease: novel routes to
progression, Cancer Cell 30 (2016) 533–547. better understanding and therapy, Can. J. Cardiol. 32 (2016) 612–623.
[120] J.P. Coppe, P.Y. Desprez, A. Krtolica, J. Campisi, The senescence-associated se- [147] M.R. Khorramizadeh, E.E. Tredget, C. Telasky, Q. Shen, A. Ghahary, Aging dif-
cretory phenotype: the dark side of tumor suppression, Annu. Rev. Pathol. 5 ferentially modulates the expression of collagen and collagenase in dermal fibro-
(2010) 99–118. blasts, Mol. Cell. Biochem. 194 (1999) 99–108.
[121] J.C. Acosta, A. O’Loghlen, A. Banito, S. Raguz, J. Gil, Control of senescence by [148] T. Kumazaki, R.S. Robetorye, S.C. Robetorye, J.R. Smith, Fibronectin expression
CXCR2 and its ligands, Cell Cycle 7 (2008) 2956–2959. increases during in vitro cellular senescence: correlation with increased cell area,
[122] S. Havaki, A. Kotsinas, E. Chronopoulos, D. Kletsas, A. Georgakilas, Exp. Cell Res. 195 (1991) 13–19.
V.G. Gorgoulis, The role of oxidative DNA damage in radiation induced bystander [149] A.M. Choi, D.R. Olsen, K.G. Cook, S.F. Deamond, J. Uitto, S.A. Bruce, Differential
effect, Cancer Lett. 356 (2015) 43–51. extracellular matrix gene expression by fibroblasts during their proliferative life
[123] G. Nelson, J. Wordsworth, C. Wang, D. Jurk, C. Lawless, C. Martin-Ruiz, et al., A span in vitro and at senescence, J. Cell. Physiol. 151 (1992) 147–155.
senescent cell bystander effect: senescence-induced senescence, Aging Cell 11 [150] F. Pagani, L. Zagato, J.A. Maier, G. Ragnotti, D.A. Coviello, C. Vergani, Expression
(2012) 345–349. and alternative splicing of fibronectin mRNA in human diploid endothelial cells
[124] J.C. Acosta, A. Banito, T. Wuestefeld, A. Georgilis, P. Janich, J.P. Morton, et al., A during aging in vitro, Biochim. Biophys. Acta 1173 (1993) 172–178.
complex secretory program orchestrated by the inflammasome controls paracrine [151] K. Korybalska, E. Kawka, A. Kusch, F. Aregger, D. Dragun, A. Jorres, et al.,
senescence, Nat. Cell Biol. 15 (2013) 978–990. Recovery of senescent endothelial cells from injury, J. Gerontol. A Biol. Sci. Med.
[125] P. Hiebert, M.S. Wietecha, M. Cangkrama, E. Haertel, E. Mavrogonatou, Sci. 68 (2013) 250–257.
M. Stumpe, et al., Nrf2-mediated fibroblast reprogramming drives cellular senes- [152] M.Y. Lee, Y. Wang, P.M. Vanhoutte, Senescence of cultured porcine coronary ar-
cence by targeting the matrisome, Dev. Cell 46 (2018) 145–161 e10. terial endothelial cells is associated with accelerated oxidative stress and activa-
[126] V. Bizot-Foulon, B. Bouchard, W. Hornebeck, L. Dubertret, B. Bertaux, tion of NFkB, J. Vasc. Res. 47 (2010) 287–298.
Uncoordinate expressions of type I and III collagens, collagenase and tissue in- [153] K. Takeda, A. Gosiewska, B. Peterkofsky, Similar, but not identical, modulation of
hibitor of matrix metalloproteinase 1 along in vitro proliferative life span of expression of extracellular matrix components during in vitro and in vivo aging of
human skin fibroblasts. Regulation by all-trans retinoic acid, Cell Biol. Int. 19 human skin fibroblasts, J. Cell. Physiol. 153 (1992) 450–459.
(1995) 129–135. [154] K. Yudoh, T. Nguyen v, H. Nakamura, K. Hongo-Masuko, T. Kato, K. Nishioka,
[127] W.D. Funk, C.K. Wang, D.N. Shelton, C.B. Harley, G.D. Pagon, W.K. Hoeffler, Potential involvement of oxidative stress in cartilage senescence and development
Telomerase expression restores dermal integrity to in vitro-aged fibroblasts in a of osteoarthritis: oxidative stress induces chondrocyte telomere instability and
reconstituted skin model, Exp. Cell Res. 258 (2000) 270–278. downregulation of chondrocyte function, Arthritis Res. Ther. 7 (2005) R380–91.
[128] A. Ghahary, Y.J. Shen, B. Nedelec, R. Wang, P.G. Scott, E.E. Tredget, Collagenase [155] R. Yentrapalli, O. Azimzadeh, Z. Barjaktarovic, H. Sarioglu, A. Wojcik, M. Harms-
production is lower in post-burn hypertrophic scar fibroblasts than in normal fi- Ringdahl, et al., Quantitative proteomic analysis reveals induction of premature
broblasts and is reduced by insulin-like growth factor-1, J. Invest. Dermatol. 106 senescence in human umbilical vein endothelial cells exposed to chronic low-dose
(1996) 476–481. rate gamma radiation, Proteomics 13 (2013) 1096–1107.
[129] A.J. Millis, M. Hoyle, H.M. McCue, H. Martini, Differential expression of me- [156] A. Passi, R. Albertini, A. Bardoni, S. Rindi, R. Salvini, G. Pallavicini, et al.,
talloproteinase and tissue inhibitor of metalloproteinase genes in aged human fi- Modifications of proteoglycans produced by human skin fibroblast cultures during
broblasts, Exp. Cell Res. 201 (1992) 373–379. replicative senescence, Cell Biochem. Funct. 11 (1993) 263–269.
[130] H. Pratsinis, A. Armatas, A. Dimozi, M. Lefaki, P. Vassiliu, D. Kletsas, Paracrine [157] A. Passi, R. Albertini, F. Campagnari, G. De Luca, Modifications of proteoglycans
anti-fibrotic effects of neonatal cells and living cell constructs on young and se- extracted from monolayer cultures of young and senescent human skin fibroblasts,
nescent human dermal fibroblasts, Wound Repair Regen. 21 (2013) 842–851. FEBS Lett. 420 (1997) 175–178.
[131] J.P. Coppe, C.K. Patil, F. Rodier, A. Krtolica, C.M. Beausejour, S. Parrinello, et al., [158] D.A. Carrino, J.M. Sorrell, A.I. Caplan, Age-related changes in the proteoglycans of
A human-like senescence-associated secretory phenotype is conserved in mouse human skin, Arch. Biochem. Biophys. 373 (2000) 91–101.
cells dependent on physiological oxygen, PLoS One 5 (2010) e9188. [159] R. Buffenstein, Negligible senescence in the longest living rodent, the naked mole-
[132] D.G. Burton, P.J. Giles, A.N. Sheerin, S.K. Smith, J.J. Lawton, E.L. Ostler, et al., rat: insights from a successfully aging species, J. Comp. Physiol. B 178 (2008)
Microarray analysis of senescent vascular smooth muscle cells: a link to athero- 439–445.
sclerosis and vascular calcification, Exp. Gerontol. 44 (2009) 659–665. [160] Y. Kulaberoglu, B. Bhushan, F. Hadi, S. Chakrabarti, W.T. Khaled, K.S. Rankin,
[133] S.E. Gardner, M. Humphry, M.R. Bennett, M.C. Clarke, Senescent vascular smooth et al., The material properties of naked mole-rat hyaluronan, Sci. Rep. 9 (2019)
muscle cells drive inflammation through an Interleukin-1alpha-dependent senes- 6632.
cence-associated secretory phenotype, Arterioscler. Thromb. Vasc. Biol. 35 (2015) [161] E. Karousou, S. Misra, S. Ghatak, K. Dobra, M. Gotte, D. Vigetti, et al., Roles and
1963–1974. targeting of the HAS/hyaluronan/CD44 molecular system in cancer, Matrix Biol.
[134] A. Dimozi, E. Mavrogonatou, A. Sklirou, D. Kletsas, Oxidative stress inhibits the 59 (2017) 3–22.
proliferation, induces premature senescence and promotes a catabolic phenotype [162] E.M. Jung, O. Kwon, K.S. Kwon, Y.S. Cho, S.K. Rhee, J.K. Min, et al., Evidences for
in human nucleus pulposus intervertebral disc cells, Eur. Cell. Mater. 30 (2015) correlation between the reduced VCAM-1 expression and hyaluronan synthesis
89–102 discussion 103. during cellular senescence of human mesenchymal stem cells, Biochem. Biophys.
[135] C.L. Le Maitre, A.J. Freemont, J.A. Hoyland, Accelerated cellular senescence in Res. Commun. 404 (2011) 463–469.
degenerate intervertebral discs: a possible role in the pathogenesis of inter- [163] C. Frantz, K.M. Stewart, V.M. Weaver, The extracellular matrix at a glance, J. Cell.
vertebral disc degeneration, Arthritis Res. Ther. 9 (2007) R45. Sci. 123 (2010) 4195–4200.
[136] S.S. Vamvakas, E. Mavrogonatou, D. Kletsas, Human nucleus pulposus inter- [164] H.R. Choi, K.A. Cho, H.T. Kang, J.B. Lee, M. Kaeberlein, Y. Suh, et al., Restoration
vertebral disc cells becoming senescent using different treatments exhibit a similar of senescent human diploid fibroblasts by modulation of the extracellular matrix,
transcriptional profile of catabolic and inflammatory genes, Eur. Spine J. 26 Aging Cell 10 (2011) 148–157.
(2017) 2063–2071. [165] X. Yu, Y. He, Z. Chen, Y. Qian, J. Wang, Z. Ji, et al., Autologous decellularized
[137] V. Krizhanovsky, M. Yon, R.A. Dickins, S. Hearn, J. Simon, C. Miething, et al., extracellular matrix protects against H2O2-induced senescence and aging in adi-
Senescence of activated stellate cells limits liver fibrosis, Cell 134 (2008) 657–667. pose-derived stem cells and stimulates proliferation in vitro, Biosci. Rep. (2019).
[138] P. Stephens, H. Cook, J. Hilton, C.J. Jones, M.F. Haughton, F.S. Wyllie, et al., An [166] S. Goruppi, G.P. Dotto, Mesenchymal stroma: primary determinant and ther-
analysis of replicative senescence in dermal fibroblasts derived from chronic leg apeutic target for epithelial cancer, Trends Cell Biol. 23 (2013) 593–602.
wounds predicts that telomerase therapy would fail to reverse their disease-spe- [167] D.G. Burton, V. Krizhanovsky, Physiological and pathological consequences of
cific cellular and proteolytic phenotype, Exp. Cell Res. 283 (2003) 22–35. cellular senescence, Cell. Mol. Life Sci. 71 (2014) 4373–4386.

8
E. Mavrogonatou, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

[168] Y. Li, J. Liang, T. Yang, J. Monterrosa Mena, C. Huan, T. Xie, et al., Hyaluronan [198] Z. Han, W. Wei, S. Dunaway, J.W. Darnowski, P. Calabresi, J. Sedivy, et al., Role of
synthase 2 regulates fibroblast senescence in pulmonary fibrosis, Matrix Biol. 55 p21 in apoptosis and senescence of human colon cancer cells treated with camp-
(2016) 35–48. tothecin, J. Biol. Chem. 277 (2002) 17154–17160.
[169] M. Demaria, N. Ohtani, S.A. Youssef, F. Rodier, W. Toussaint, J.R. Mitchell, et al., [199] X. Wang, S.C. Wong, J. Pan, S.W. Tsao, K.H. Fung, D.L. Kwong, et al., Evidence of
An essential role for senescent cells in optimal wound healing through secretion of cisplatin-induced senescent-like growth arrest in nasopharyngeal carcinoma cells,
PDGF-AA, Dev. Cell 31 (2014) 722–733. Cancer Res. 58 (1998) 5019–5022.
[170] M. Demaria, P.Y. Desprez, J. Campisi, M.C. Velarde, Cell autonomous and non- [200] I.B. Roninson, Tumor cell senescence in cancer treatment, Cancer Res. 63 (2003)
autonomous effects of senescent cells in the skin, J. Invest. Dermatol. 135 (2015) 2705–2715.
1722–1726. [201] K. Fang, C.C. Chiu, C.H. Li, Y.T. Chang, H.T. Hwang, Cisplatin-induced senescence
[171] M. Muller, Z. Li, P.K. Maitz, Pseudomonas pyocyanin inhibits wound repair by and growth inhibition in human non-small cell lung cancer cells with ectopic
inducing premature cellular senescence: role for p38 mitogen-activated protein transfer of p16INK4a, Oncol. Res. 16 (2007) 479–488.
kinase, Burns 35 (2009) 500–508. [202] K.R. Jones, L.W. Elmore, C. Jackson-Cook, G. Demasters, L.F. Povirk, S.E. Holt,
[172] A. Sindrilaru, T. Peters, S. Wieschalka, C. Baican, A. Baican, H. Peter, et al., An et al., p53-Dependent accelerated senescence induced by ionizing radiation in
unrestrained proinflammatory M1 macrophage population induced by iron im- breast tumour cells, Int. J. Radiat. Biol. 81 (2005) 445–458.
pairs wound healing in humans and mice, J. Clin. Invest. 121 (2011) 985–997. [203] S. Vamvakas, E.H. Vock, W.K. Lutz, On the role of DNA double-strand breaks in
[173] M.S. Bitar, S.M. Abdel-Halim, F. Al-Mulla, Caveolin-1/PTRF upregulation con- toxicity and carcinogenesis, Crit. Rev. Toxicol. 27 (1997) 155–174.
stitutes a mechanism for mediating p53-induced cellular senescence: implications [204] V. Probin, Y. Wang, D. Zhou, Busulfan-induced senescence is dependent on ROS
for evidence-based therapy of delayed wound healing in diabetes, Am. J. Physiol. production upstream of the MAPK pathway, Free Radic. Biol. Med. 42 (2007)
Endocrinol. Metab. 305 (2013) E951–63. 1858–1865.
[174] J.S. Vande Berg, R. Rudolph, C. Hollan, P.L. Haywood-Reid, Fibroblast senescence [205] L.E. Klein, B.S. Freeze, A.B. Smith 3rd, S.B. Horwitz, The microtubule stabilizing
in pressure ulcers, Wound Repair Regen. 6 (1998) 38–49. agent discodermolide is a potent inducer of accelerated cell senescence, Cell Cycle
[175] M. Braig, S. Lee, C. Loddenkemper, C. Rudolph, A.H. Peters, B. Schlegelberger, 4 (2005) 501–507.
et al., Oncogene-induced senescence as an initial barrier in lymphoma develop- [206] X. Di, A.T. Bright, R. Bellott, E. Gaskins, J. Robert, S. Holt, et al., A chemotherapy-
ment, Nature 436 (2005) 660–665. associated senescence bystander effect in breast cancer cells, Cancer Biol. Ther. 7
[176] M. Collado, J. Gil, A. Efeyan, C. Guerra, A.J. Schuhmacher, M. Barradas, et al., (2008) 864–872.
Tumour biology: senescence in premalignant tumours, Nature 436 (2005) 642. [207] M. Suzuki, K. Suzuki, S. Kodama, M. Watanabe, Phosphorylated histone H2AX foci
[177] C.A. Schmitt, J.S. Fridman, M. Yang, S. Lee, E. Baranov, R.M. Hoffman, et al., A persist on rejoined mitotic chromosomes in normal human diploid cells exposed to
senescence program controlled by p53 and p16INK4a contributes to the outcome ionizing radiation, Radiat. Res. 165 (2006) 269–276.
of cancer therapy, Cell 109 (2002) 335–346. [208] K. Naka, A. Tachibana, K. Ikeda, N. Motoyama, Stress-induced premature senes-
[178] J. Campisi, Aging, cellular senescence, and cancer, Annu. Rev. Physiol. 75 (2013) cence in hTERT-expressing ataxia telangiectasia fibroblasts, J. Biol. Chem. 279
685–705. (2004) 2030–2037.
[179] M. Hoare, M. Narita, Transmitting senescence to the cell neighbourhood, Nat. Cell [209] R. Mirzayans, A. Scott, M. Cameron, D. Murray, Induction of accelerated senes-
Biol. 15 (2013) 887–889. cence by gamma radiation in human solid tumor-derived cell lines expressing
[180] J. Campisi, Aging and cancer: the double-edged sword of replicative senescence, J. wild-type TP53, Radiat. Res. 163 (2005) 53–62.
Am. Geriatr. Soc. 45 (1997) 482–488. [210] R.S. Roberson, S.J. Kussick, E. Vallieres, S.Y. Chen, D.Y. Wu, Escape from therapy-
[181] J.M. Gonzalez-Meljem, S. Haston, G. Carreno, J.R. Apps, S. Pozzi, C. Stache, et al., induced accelerated cellular senescence in p53-null lung cancer cells and in human
Stem cell senescence drives age-attenuated induction of pituitary tumours in lung cancers, Cancer Res. 65 (2005) 2795–2803.
mouse models of paediatric craniopharyngioma, Nat. Commun. 8 (2017) 1819. [211] M. Mathe, Z. Suba, Z. Nemeth, P. Tatrai, T. Fule, G. Borgulya, et al., Stromal
[182] J.M. Gonzalez-Meljem, J.P. Martinez-Barbera, Senescence drives non-cell auton- syndecan-1 expression is an adverse prognostic factor in oral carcinomas, Oral
omous tumorigenesis in the pituitary gland, Mol. Cell. Oncol. 5 (2018) e1435180. Oncol. 42 (2006) 493–500.
[183] J.L. Buhl, F. Selt, T. Hielscher, R. Guiho, J. Ecker, F. Sahm, et al., The senescence- [212] T. Szarvas, H. Reis, G. Kramer, S.F. Shariat, F. Vom Dorp, S. Tschirdewahn, et al.,
associated secretory phenotype mediates oncogene-induced senescence in pedia- Enhanced stromal syndecan-1 expression is an independent risk factor for poor
tric pilocytic astrocytoma, Clin. Cancer Res. 25 (2019) 1851–1866. survival in bladder cancer, Hum. Pathol. 45 (2014) 674–682.
[184] G.H. Di, Y. Liu, Y. Lu, J. Liu, C. Wu, H.F. Duan, IL-6 secreted from senescent [213] F. Zong, E. Fthenou, F. Mundt, T. Szatmari, I. Kovalszky, L. Szilak, et al., Specific
mesenchymal stem cells promotes proliferation and migration of breast cancer syndecan-1 domains regulate mesenchymal tumor cell adhesion, motility and
cells, PLoS One 9 (2014) e113572. migration, PLoS One 6 (2011) e14816.
[185] C.C. Sprenger, S.R. Plymate, M.J. Reed, Aging-related alterations in the extra- [214] D. Vigetti, M. Gotte, M.S. Pavao, A.D. Theocharis, Cellular microenvironment in
cellular matrix modulate the microenvironment and influence tumor progression, human pathologies, Biomed. Res. Int. 2013 (2013) 946958.
Int. J. Cancer 127 (2010) 2739–2748. [215] V. Nikolova, C.Y. Koo, S.A. Ibrahim, Z. Wang, D. Spillmann, R. Dreier, et al.,
[186] K. Lawrenson, B. Grun, E. Benjamin, I.J. Jacobs, D. Dafou, S.A. Gayther, Senescent Differential roles for membrane-bound and soluble syndecan-1 (CD138) in breast
fibroblasts promote neoplastic transformation of partially transformed ovarian cancer progression, Carcinogenesis 30 (2009) 397–407.
epithelial cells in a three-dimensional model of early stage ovarian cancer, [216] Z. Piperigkou, B. Mohr, N. Karamanos, M. Gotte, Shed proteoglycans in tumor
Neoplasia 12 (2010) 317–325. stroma, Cell Tissue Res. 365 (2016) 643–655.
[187] N. Malaquin, C. Vercamer, F. Bouali, S. Martien, E. Deruy, N. Wernert, et al., [217] X. Sun, B. Shi, H. Zheng, L. Min, J. Yang, X. Li, et al., Senescence-associated se-
Senescent fibroblasts enhance early skin carcinogenic events via a paracrine MMP- cretory factors induced by cisplatin in melanoma cells promote non-senescent
PAR-1 axis, PLoS One 8 (2013) e63607. melanoma cell growth through activation of the ERK1/2-RSK1 pathway, Cell
[188] K. Gosselin, S. Martien, A. Pourtier, C. Vercamer, P. Ostoich, L. Morat, et al., Death Dis. 9 (2018) 260.
Senescence-associated oxidative DNA damage promotes the generation of neo- [218] T. Saleh, L. Tyutynuk-Massey, E.K. Cudjoe Jr., M.O. Idowu, J.W. Landry,
plastic cells, Cancer Res. 69 (2009) 7917–7925. D.A. Gewirtz, Non-cell autonomous effects of the senescence-associated secretory
[189] S. Parrinello, J.P. Coppe, A. Krtolica, J. Campisi, Stromal-epithelial interactions in phenotype in cancer therapy, Front. Oncol. 8 (2018) 164.
aging and cancer: senescent fibroblasts alter epithelial cell differentiation, J. Cell. [219] M. Demaria, M.N. O’Leary, J. Chang, L. Shao, S. Liu, F. Alimirah, et al., Cellular
Sci. 118 (2005) 485–496. senescence promotes adverse effects of chemotherapy and cancer relapse, Cancer
[190] S.J. Robles, P.W. Buehler, A. Negrusz, G.R. Adami, Permanent cell cycle arrest in Discov. 7 (2017) 165–176.
asynchronously proliferating normal human fibroblasts treated with doxorubicin [220] S.G. Rao, J.G. Jackson, SASP: Tumor Suppressor or Promoter? Yes!, Trends Cancer
or etoposide but not camptothecin, Biochem. Pharmacol. 58 (1999) 675–685. 2 (2016) 676–687.
[191] W. Zhao, Z.X. Lin, Z.Q. Zhang, Cisplatin-induced premature senescence with [221] M.C. Velarde, M. Demaria, J. Campisi, Senescent cells and their secretory phe-
concomitant reduction of gap junctions in human fibroblasts, Cell Res. 14 (2004) notype as targets for cancer therapy, Interdiscip. Top. Gerontol. 38 (2013) 17–27.
60–66. [222] R. Sidi, G. Pasello, I. Opitz, A. Soltermann, M. Tutic, H. Rehrauer, et al., Induction
[192] V. Probin, Y. Wang, A. Bai, D. Zhou, Busulfan selectively induces cellular senes- of senescence markers after neo-adjuvant chemotherapy of malignant pleural
cence but not apoptosis in WI38 fibroblasts via a p53-independent but extra- mesothelioma and association with clinical outcome: an exploratory analysis, Eur.
cellular signal-regulated kinase-p38 mitogen-activated protein kinase-dependent J. Cancer 47 (2011) 326–332.
mechanism, J. Pharmacol. Exp. Ther. 319 (2006) 551–560. [223] B. Robl, C. Pauli, S.M. Botter, B. Bode-Lesniewska, B. Fuchs, Prognostic value of
[193] R.H. te Poele, A.L. Okorokov, L. Jardine, J. Cummings, S.P. Joel, DNA damage is tumor suppressors in osteosarcoma before and after neoadjuvant chemotherapy,
able to induce senescence in tumor cells in vitro and in vivo, Cancer Res. 62 (2002) BMC Cancer 15 (2015) 379.
1876–1883. [224] T.W. Kang, T. Yevsa, N. Woller, L. Hoenicke, T. Wuestefeld, D. Dauch, et al.,
[194] I.B. Roninson, E.V. Broude, B.D. Chang, If not apoptosis, then what? Treatment- Senescence surveillance of pre-malignant hepatocytes limits liver cancer devel-
induced senescence and mitotic catastrophe in tumor cells, Drug Resist. Updat. 4 opment, Nature 479 (2011) 547–551.
(2001) 303–313. [225] A. Lujambio, L. Akkari, J. Simon, D. Grace, D.F. Tschaharganeh, J.E. Bolden, et al.,
[195] B.D. Chang, Y. Xuan, E.V. Broude, H. Zhu, B. Schott, J. Fang, et al., Role of p53 and Non-cell-autonomous tumor suppression by p53, Cell 153 (2013) 449–460.
p21waf1/cip1 in senescence-like terminal proliferation arrest induced in human [226] A. Sagiv, D.G. Burton, Z. Moshayev, E. Vadai, F. Wensveen, S. Ben-Dor, et al.,
tumor cells by chemotherapeutic drugs, Oncogene 18 (1999) 4808–4818. NKG2D ligands mediate immunosurveillance of senescent cells, Aging (Albany
[196] J.A. Ewald, J.A. Desotelle, G. Wilding, D.F. Jarrard, Therapy-induced senescence NY) 8 (2016) 328–344.
in cancer, J. Natl. Cancer Inst. 102 (2010) 1536–1546. [227] Y. Ovadya, T. Landsberger, H. Leins, E. Vadai, H. Gal, A. Biran, et al., Impaired
[197] L.W. Elmore, C.W. Rehder, X. Di, P.A. McChesney, C.K. Jackson-Cook, immune surveillance accelerates accumulation of senescent cells and aging, Nat.
D.A. Gewirtz, et al., Adriamycin-induced senescence in breast tumor cells involves Commun. 9 (2018) 5435.
functional p53 and telomere dysfunction, J. Biol. Chem. 277 (2002) 35509–35515. [228] C. Franceschi, J. Campisi, Chronic inflammation (inflammaging) and its potential

9
E. Mavrogonatou, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

contribution to age-associated diseases, J. Gerontol. A Biol. Sci. Med. Sci. 69 B.R.M. Bourgeois, et al., Targeted apoptosis of senescent cells restores tissue
(Suppl 1) (2014) S4–9. homeostasis in response to chemotoxicity and aging, Cell 169 (2017) 132–147
[229] H. Braumuller, T. Wieder, E. Brenner, S. Assmann, M. Hahn, M. Alkhaled, et al., T- e16.
helper-1-cell cytokines drive cancer into senescence, Nature 494 (2013) 361–365. [247] H. Fuhrmann-Stroissnigg, Y.Y. Ling, J. Zhao, S.J. McGowan, Y. Zhu, R.W. Brooks,
[230] S. Hubackova, K. Krejcikova, J. Bartek, Z. Hodny, IL1- and TGFbeta-Nox4 sig- et al., Identification of HSP90 inhibitors as a novel class of senolytics, Nat.
naling, oxidative stress and DNA damage response are shared features of re- Commun. 8 (2017) 422.
plicative, oncogene-induced, and drug-induced paracrine’ bystander senescence’, [248] L. Samaraweera, A. Adomako, A. Rodriguez-Gabin, H.M. McDaid, A novel in-
Aging (Albany NY) 4 (2012) 932–951. dication for Panobinostat as a senolytic drug in NSCLC and HNSCC, Sci. Rep. 7
[231] T. Wieder, E. Brenner, H. Braumuller, O. Bischof, M. Rocken, Cytokine-induced (2017) 1900.
senescence for cancer surveillance, Cancer Metastasis Rev. 36 (2017) 357–365. [249] V. Myrianthopoulos, The emerging field of senotherapeutic drugs, Future Med.
[232] D.G.A. Burton, A. Stolzing, Cellular senescence: immunosurveillance and future Chem. 10 (2018) 2369–2372.
immunotherapy, Ageing Res. Rev. 43 (2018) 17–25. [250] K. Kovacovicova, M. Skolnaja, M. Heinmaa, M. Mistrik, P. Pata, I. Pata, et al.,
[233] T. Fulop, A. Larbi, G. Dupuis, A. Le Page, E.H. Frost, A.A. Cohen, et al., Senolytic cocktail dasatinib+Quercetin (D+Q) does not enhance the efficacy of
Immunosenescence and inflamm-aging as two sides of the same coin: friends or senescence-inducing chemotherapy in liver cancer, Front. Oncol. 8 (2018) 459.
foes? Front. Immunol. 8 (2017) 1960. [251] C.M. Rudin, C.L. Hann, E.B. Garon, M. Ribeiro de Oliveira, P.D. Bonomi,
[234] L. Prata, I.G. Ovsyannikova, T. Tchkonia, J.L. Kirkland, Senescent cell clearance by D.R. Camidge, et al., Phase II study of single-agent navitoclax (ABT-263) and
the immune system: emerging therapeutic opportunities, Semin. Immunol. (2019). biomarker correlates in patients with relapsed small cell lung cancer, Clin. Cancer
[235] D.J. Baker, B.G. Childs, M. Durik, M.E. Wijers, C.J. Sieben, J. Zhong, et al., Res. 18 (2012) 3163–3169.
Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan, Nature 530 [252] E.C. Kim, J.R. Kim, Senotherapeutics: emerging strategy for healthy aging and age-
(2016) 184–189. related disease, BMB Rep. 52 (2019) 47–55.
[236] D.J. Baker, T. Wijshake, T. Tchkonia, N.K. LeBrasseur, B.G. Childs, B. van de Sluis, [253] H. Lim, H. Park, H.P. Kim, Effects of flavonoids on senescence-associated secretory
et al., Clearance of p16Ink4a-positive senescent cells delays ageing-associated phenotype formation from bleomycin-induced senescence in BJ fibroblasts,
disorders, Nature 479 (2011) 232–236. Biochem. Pharmacol. 96 (2015) 337–348.
[237] Y. Zhu, T. Tchkonia, T. Pirtskhalava, A.C. Gower, H. Ding, N. Giorgadze, et al., The [254] O. Moiseeva, X. Deschenes-Simard, E. St-Germain, S. Igelmann, G. Huot,
Achilles’ heel of senescent cells: from transcriptome to senolytic drugs, Aging Cell A.E. Cadar, et al., Metformin inhibits the senescence-associated secretory pheno-
14 (2015) 644–658. type by interfering with IKK/NF-kappaB activation, Aging Cell 12 (2013)
[238] A. Soto-Gamez, M. Demaria, Therapeutic interventions for aging: the case of cel- 489–498.
lular senescence, Drug Discov. Today 22 (2017) 786–795. [255] M. Xu, T. Tchkonia, H. Ding, M. Ogrodnik, E.R. Lubbers, T. Pirtskhalava, et al.,
[239] J. Chang, Y. Wang, L. Shao, R.M. Laberge, M. Demaria, J. Campisi, et al., JAK inhibition alleviates the cellular senescence-associated secretory phenotype
Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells and frailty in old age, Proc. Natl. Acad. Sci. U. S. A. 112 (2015) E6301–10.
in mice, Nat. Med. 22 (2016) 78–83. [256] R.M. Laberge, L. Zhou, M.R. Sarantos, F. Rodier, A. Freund, P.L. de Keizer, et al.,
[240] R. Yosef, N. Pilpel, R. Tokarsky-Amiel, A. Biran, Y. Ovadya, S. Cohen, et al., Glucocorticoids suppress selected components of the senescence-associated se-
Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL, Nat. cretory phenotype, Aging Cell 11 (2012) 569–578.
Commun. 7 (2016) 11190. [257] R. Wang, Z. Yu, B. Sunchu, J. Shoaf, I. Dang, S. Zhao, et al., Rapamycin inhibits the
[241] Y. Zhu, T. Tchkonia, H. Fuhrmann-Stroissnigg, H.M. Dai, Y.Y. Ling, M.B. Stout, secretory phenotype of senescent cells by a Nrf2-independent mechanism, Aging
et al., Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 Cell 16 (2017) 564–574.
family of anti-apoptotic factors, Aging Cell 15 (2016) 428–435. [258] E. Latorre, V.C. Birar, A.N. Sheerin, J.C.C. Jeynes, A. Hooper, H.R. Dawe, et al.,
[242] Y. Zhu, E.J. Doornebal, T. Pirtskhalava, N. Giorgadze, M. Wentworth, Small molecule modulation of splicing factor expression is associated with rescue
H. Fuhrmann-Stroissnigg, et al., New agents that target senescent cells: the fla- from cellular senescence, BMC Cell Biol. 18 (2017) 31.
vone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463, Aging (Albany [259] A. Georgilis, S. Klotz, C.J. Hanley, N. Herranz, B. Weirich, B. Morancho, et al.,
NY) 9 (2017) 955–963. PTBP1-mediated alternative splicing regulates the inflammatory secretome and
[243] Y. Wang, J. Chang, X. Liu, X. Zhang, S. Zhang, X. Zhang, et al., Discovery of pi- the pro-tumorigenic effects of senescent cells, Cancer Cell 34 (2018) 85–102 e9.
perlongumine as a potential novel lead for the development of senolytic agents, [260] J.L. Kirkland, T. Tchkonia, Cellular senescence: a translational perspective,
Aging (Albany NY) 8 (2016) 2915–2926. EBioMedicine 21 (2017) 21–28.
[244] X. Zhang, S. Zhang, X. Liu, Y. Wang, J. Chang, X. Zhang, et al., Oxidation re- [261] J.L. Kirkland, T. Tchkonia, Y. Zhu, L.J. Niedernhofer, P.D. Robbins, The clinical
sistance 1 is a novel senolytic target, Aging Cell (2018) e12780. potential of senolytic drugs, J. Am. Geriatr. Soc. 65 (2017) 2297–2301.
[245] X. Liu, Y. Wang, X. Zhang, Z. Gao, S. Zhang, P. Shi, et al., Senolytic activity of [262] M. Xu, T. Pirtskhalava, J.N. Farr, B.M. Weigand, A.K. Palmer, M.M. Weivoda,
piperlongumine analogues: synthesis and biological evaluation, Bioorg. Med. et al., Senolytics improve physical function and increase lifespan in old age, Nat.
Chem. 26 (2018) 3925–3938. Med. 24 (2018) 1246–1256.
[246] M.P. Baar, R.M.C. Brandt, D.A. Putavet, J.D.D. Klein, K.W.J. Derks,

10

You might also like