Abstract
Cancer therapies, including conventional chemotherapy, radiation, and molecularly targeted agents, can lead to tumor eradication through a variety of mechanisms. In addition to their effects on tumor cell growth and survival, these regimens can also influence the surrounding tumor-immune microenvironment in ways that ultimately impact therapy responses. A unique biological outcome of cancer therapy is induction of cellular senescence. Senescence is a damage-induced stress program that leads to both the durable arrest of tumor cells and remodeling the tumor-immune microenvironment through activation of a collection pleiotropic cytokines, chemokines, growth factors, and proteinases known as the senescence-associated secretory phenotype (SASP). Depending on the cancer context and the mechanism of action of the therapy, the SASP produced following therapy-induced senescence (TIS) can promote anti-tumor immunity that enhances therapeutic efficacy, or alternatively chronic inflammation that leads to therapy failure and tumor relapse. Thus, a deeper understanding of the mechanisms regulating the SASP and components necessary for robust anti-immune surveillance in different cancer and therapy contexts are key to harnessing senescence for tumor control. Here we draw a roadmap to modulate TIS and its immune-stimulating features for cancer immunotherapy.
Keywords: Cellular senescence, senescence-associated secretory phenotype, tumor microenvironment, senotherapeutics, immunotherapy
1. Introduction
Cytotoxic regimens that directly kill tumor cells while sparing normal cells have historically been the primary pursuit of cancer medicine and believed necessary to achieve cure. Through a better understanding of the mechanisms by which cytotoxic and other cancer drugs work, we have come to understand treatment does not need to directly kill tumor cells to achieve clinically-relevant tumor responses [1–3]. Indeed, there are numerous biological outcomes of cancer therapy. In addition to inducing non-immunogenic (apoptosis) and immunogenic (pyroptosis, necroptosis) cell death, cancer therapies can also block tumor cell growth through a transient (quiescence) or more durable (senescence) cell cycle arrest that impacts not only the tumor itself but also its surrounding tumor microenvironment (TME) in ways that dictate therapy responses [2, 4]. The TME includes a diverse array of cellular and non-cellular components, including stromal fibroblasts, vascular and lymphatic vessels, immune cells, and the extracellular matrix (ECM) that can exert both tumor suppressive and promoting effects [5]. Over the past decade the golden age of cancer immunotherapy has emerged, based on the principle that reactivating the immune system, and in particular cytotoxic T cells, can be harnessed as a means to indirectly kill tumor cells systemically throughout the body [6, 7]. Thus, understanding the biological outcomes of cancer therapy and the mechanisms by which they impact both cancer cells and the surrounding tumor-immune microenvironment is critical to engineering the next generation of cancer therapeutics and treatment paradigms.
Senescence is a damage-induced cellular response to cancer therapy. Cellular senescence was first described by Leonard Hayflick and colleagues, who observed that human diploid fibroblasts undergo a finite number of doublings before irreversibly arresting [8, 9]. As such, this process was named senescence (derived from the Latin root senex meaning “old”) in reference to its association with cellular aging. However, we now know senescence to be a conserved response to many different types of external and internal cellular stress, including not just telomere shortening associated with cell division (i.e. replicative senescence) but also oncogenic, genotoxic, metabolic, and oxidative damage, all of which can accumulate following cancer therapy [10–12]. Though initially thought a cell culture artefact, over the past two decades a plethora of tools and models to remove senescent cells in normal and disease contexts in in vivo settings, as well as observation of senescence-related markers in patients with cancer and other age-related chronic diseases, has revealed senescence to be a bona fide biological process. In fact, senescence has been shown to be a rather broad physiological response to tissue damage that plays a pleiotropic role in aging, embryonic development, wound healing, tissue regeneration, and, importantly, response to oncogenesis and cancer therapy [13–18].
Hallmarks of senescence include: (a) stable cell cycle arrest mediated by two tumor suppressor pathways, p53/p21CIP1 and p16INK4a/RB, that lead to transcriptional and epigenetic repression (visible as senescence-associated heterochromatin foci (SAHF)) of key cell cycle genes even after the senescence inducer is removed; (b) changes in cellular morphology (cell flattening, enlargement, multinucleation); (c) dissolution of the nuclear envelope (Lamin B1 loss); (d) enhanced lysosomal activity as marked by senescence-associated beta-galactosidase (SA-β-gal); (e) upregulation of anti-apoptotic and pro-survival pathways (e.g. BCL-2 family members) leading to resistance to cell death; (f) macromolecular damage (e.g. DNA, protein damage) and a persistent DNA damage response (DDR); and (g) altered metabolism (e.g. mitochondrial dysfunction, increased glycolysis, lipid accumulation) [19]. Enhanced transcription and protein synthesis in senescent cells converges to promote a unique aspect of senescence, the senescence-associated secretory phenotype (SASP), which allows senescent cells to communicate damage signals with neighboring cells, including immune cells, fibroblasts, endothelial cells, and adjacent non-tumor epithelial cells in their surrounding microenvironment in a non-cell autonomous manner [20, 21]. Enhancer regions of hundreds of SASP factors are made accessible, transcribed by NF-κB and other factors, and translated in an mTOR-dependent manner, resulting in the robust secretion of inflammatory cytokines and chemokines and angiogenic, growth, and ECM degrading signals [22]. The SASP is also associated with upregulation of cell surface molecules that modulate the interaction between senescent cells and the immune system [23, 24]. It is important to note that these hallmarks listed above are neither specific nor universal to all forms of cellular senescence, and that multiple markers are currently necessary to distinguish senescence from other biological outcomes of cancer therapy [10].
The dynamic nature of the SASP and its ability to modulate surrounding tissue microenvironments and immune responses in different ways is thought to contribute to the many contrasting physiological phenomenon associated with senescence. Indeed, senescence can be anti- or pro-tumorigenic depending on the senescence inducer, the duration of senescence, the SASP factors produced, and tissue and disease context [25, 26]. Markers of cellular senescence have been identified in pre-cancerous lesions in various solid organs in humans, including the lungs, prostate, pancreas, and skin, and are subsequently lost during neoplastic progression [18, 27]. As such it has been postulated that senescence may be tumor suppressive and act as a roadblock to tumor development by preventing the proliferation of potentially malignant cells. Supporting this hypothesis, oncogene-induced senescence following aberrant RAS activation leads to arrest of pre-malignant cells and secretion of pro-inflammatory SASP factors that promotes innate and adaptive immune clearance of incipient cancer cells and blocks tumor formation [28]. Similarly, following treatment of established KRAS mutant cancers with RAS pathway targeted therapies, we have shown that therapy-induced senescence (TIS) inhibits tumor growth and leads to influx of cytotoxic CD8+ T cells and Natural Killer (NK) cells that promote tumor regression [29, 30]. Conversely, there is a large body of evidence demonstrating that unresolved senescence, and in particular the SASP following chemotherapy, can be tumor promoting through secretion of immune suppressive factors and attraction of immune suppressive cell types, as well as production of angiogenic and other growth factors that lead to increased invasion and metastasis of adjacent non-senescent tumor cells [31–33]. Moreover, it is now appreciated that senescence is likely not a permanent or irreversible state, and that tumor cells that bypass senescence through acquisition of genomic instability (e.g. polyploidy) can gain enhanced stemness and tumorigenic potential that contributes to treatment relapse [34–37].
In order to effectively leverage senescence and its immune stimulating properties for cancer treatment, we need a better understanding of: (a) the biological markers of senescence in cancer and following therapy, (b) the context dependent regulation of the SASP and its impact on immunity in different cancer settings, and c) those senescence and SASP factors that are necessary for immune activation and tumor suppression. It is possible that senescence is a more common outcome of cancer therapy than initially appreciated, but may have different markers and immune and tumor outcomes in various settings. This review will focus on the known impact of senescence on the tumor-immune microenvironment, the mechanisms by which this is regulated and can be manipulated, and in the end propose a strategy for how to best harness senescence for cancer immunotherapy.
2. SASP remodeling of the tumor microenvironment: Lessons from genetic models of senescence.
In vivo mouse models offer the ability to tightly control senescence in specific tumor and non-tumor cell types within the context of an intact tissue microenvironment, and have been instrumental in uncovering the impact of the SASP on remodeling the TME in cancer [15]. Genetic induction of senescence in pre-malignant or tumor contexts in vivo has been achieved through activation of oncogenes (RAS, BRAF) or modulation of tumor suppressor genes (p53, p21, p27KIP1, PTEN), leading to both cell cycle arrest and SASP production through accumulation of DNA damage and/or activation of p53 and RB pathways [28, 38–42]. Below we highlight current evidence from these models demonstrating how the SASP affects immune activity and stromal remodeling in a manner that ultimately impacts tumor onset and progression.
2.1. Immune surveillance
Incipient tumor cells, including those driven into senescence by oncogenic insults, undergo immune surveillance as a failsafe mechanism to block tumor development [43, 44]. This initially involves the innate immune system, including phagocytic macrophages that engulf pre-malignant cells, and NK cells that can secrete cytolytic granules to eliminate damaged target cells that express stress ligands [45–47]. The adaptive immune system is also activated following presentation of antigens on MHC Class I (MHC-I) molecules on tumor cells and MHC Class II (MHC-II) molecules on dendritic cells (DCs) to cytotoxic (CD8+) and helper (CD4+) T cells, respectively, which upon binding to matched T cell receptors (TCRs) can lead to T cell clonal expansion and killing of cells harboring tumor-specific antigens through similar mechanisms as NK cells [48]. Tumorigenic cells also secrete soluble factors in the form of cytokines (e.g. interleukins (ILs)) that can control immune cell activity, and chemokines (e.g. C-C motif chemokine ligands (CCLs) and C-X-C chemokine ligands (CXCLs)) that attract immune cells into tissues, leading to the initiation and maintenance of immune surveillance at the tumor site [49, 50].
Many of these immunomodulatory cell surface and secreted factors involved in cancer immune surveillance are expressed and produced by senescent cells as part of the SASP and contribute to its tumor suppressive functions. In mouse models of NrasG12V-driven senescence in the liver, senescence not only inhibits the proliferation of incipient cancer cells, but also leads to their clearance by the innate (phagocytic macrophages) and adaptive (CD4+ helper T cells) immune system that is necessary to block liver tumor development [28]. Recent evidence using similar models of KrasG12V-induced senescence suggests that immune-mediated clearance is dependent on p21 and its ability to induce the SASP chemokine CXCL14 that attracts macrophages to surveil senescent cells and activate cytotoxic T cell responses in the liver [40]. Upregulation of MHC-I and MHC-II on human melanocytes and melanoma cells through SASP factors such as IL-1β following mutant NRAS and BRAF overexpression also leads to increased CD8+ and CD4+ T cell proliferation and cytotoxicity in vitro and in vivo [51, 52].
Reactivation of senescence in established tumors can also have powerful anti-tumor effects through the SASP. Genetically restoring p53 tumor suppressor expression in HrasG12V mutant hepatocellular carcinoma (HCC) produced senescence and SASP factors such as CCL2, IL-15, and CXCL1 that led to innate immune-mediated tumor regressions through macrophage, neutrophil, and NK cell mobilization [39]. A follow-up study demonstrated that anti-tumor NK cell immunity was mediated in part by (a) expression of stimulatory NK cell receptor ligands (e.g. RAE-1, ULBP1, H60a) on senescent tumor cells that bind to the activating NKG2D receptor on NK cells to trigger cytotoxicity, and (b) SASP-associated secretion of CCL2 that attracted NK cells into the liver TME [53]. Moreover, p53-induced senescence in hepatic stellate cells leads to induction of SASP factors such as IL-6 and interferon-γ (IFN-γ) that promote polarization of macrophages from an immune suppressive M2 to an immune stimulatory M1 state where they are capable of phagocytosing and clearing senescent cells [54]. Thus, the SASP can contribute to anti-tumor immunity through secreted and cell surface proteins that can activate both the innate and adaptive arms of the immune system.
2. Immune escape
Tumors undergo immune editing as they evolve over time, leading to downregulation of immune-stimulating receptors and ligands and upregulation of inhibitory checkpoints that block the ability of the innate and adaptive immune system to recognize and eliminate pre-malignant and malignant tumor cells [43, 44]. In addition, anti-inflammatory cytokines and other chemokines secreted by tumor cells can lead to an influx of suppressive immune cells, most notably regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), that inhibit NK and CD8+ T cell cytotoxicity and sedate anti-tumor immunity [55].
Prolonged senescence and chronic SASP induction can also lead to similar mechanisms of immune escape. For example, BRAFV600E mutant melanocytic nevi undergo senescence but are able to escape immune surveillance, and as such accumulate with age in humans [38]. Recent work suggests that senescent nevi do not express activating NKG2D receptor ligands, and that their secretion of SASP factors such as IL-6 even leads to upregulation of the inhibitory MHC molecule HLA-E that suppresses NK and T cell clearance of these pre-malignant lesions [56, 57]. Moreover, if pre-malignant hepatocytes are not effectively cleared by the immune system following NrasG12V-induced senescence, the SASP through chemokine CCL2 can recruit immature suppressive myeloid cells (e.g. MDSCs) that inhibit NK cell function and promote progression to HCC [58]. In agreement, increased senescence markers in peritumoral tissue correlated with worsened prognosis in HCC patients [58]. These findings underscore the differential effects of acute and timely SASP versus a chronic and unresolved SASP on immune responses in cancer.
Certain SASP components can even regulate different immune responses depending on the cancer type. Whereas activation of COX-2 and production of the lipid SASP factor species prostaglandin E2 (PGE2) is necessary for macrophage maturation and surveillance of pre-malignant hepatocytes following NrasG12V-induced senescence in the liver [59], HrasG12V-mediated induction of PGE2 in senescent thyrocytes conversely promotes immune suppressive M2 macrophage polarization [60]. The cell type undergoing senescence, as well as the senescence-inducing stimuli, also impacts immune responses. Stromal senescence following p27 induction in fibroblasts can promote tumor progression through an IL-6-mediated influx of MDSCs and Tregs that inhibit anti-tumor cytotoxic T cell responses [41]. Similarly, PTEN tumor suppressor loss, which induces senescence in pre-malignant prostate lesions through activation of the oncogenic PI3K pathway and subsequent p53 stabilization [42], produces an immune suppressive SASP including factors such as CSF-1, IL-1β, IL-6, IL-10, and CXCL1 that attract MDSCs that inhibit cytotoxic NK and T cell responses and promote the transition to prostate adenocarcinoma [61, 62]. These findings suggest that the SASP can lead to differential anti-tumor (e.g. NK cells, helper and cytotoxic T cells, M1 macrophages) and pro-tumor (e.g. M2 macrophages, MDSCs, Tregs) immunity in different contexts.
2.3. Vascular remodeling
Besides direct immune modulation via release of cytokines and chemokines, the SASP may indirectly affect immune activity by impacting other stromal cells in the TME, in particular endothelial cells. Senescent cells produce high levels of pro-angiogenic SASP factors of the vascular endothelial factor (VEGF), platelet-derived growth factor (PDGF), and fibroblast growth factor (FGF) families that can mediate new blood vessel formation and vascular remodeling [31]. Early efforts to understand the impact of the SASP on the TME and how that ultimately influences tumor progression demonstrated that senescent fibroblasts secrete paracrine factors such as VEGF that promote new blood vessel formation (i.e. angiogenesis) and breast cancer tumorigenesis and progression in immunocompromised mice [63, 64]. In addition, lymphangiogenesis provoked by secretion of CXCL12 from BRAFV600E mutant senescent thyroid cancer cells at the invasive front can also promote metastatic dissemination [65].
Though SASP-mediated induction of blood or lymphatic vessel formation can have tumor-propagating properties, it can also have immune stimulating properties by facilitating immune cell trafficking into tumors. We have recently shown that therapy-induced senescence in pancreatic cancer models leads to induction of the pro-inflammatory SASP factors CCL5, CXCL1, and IL-6 that mediate a phenomenon known as endothelial activation [66]. This led to the upregulation of cell adhesion molecules such as VCAM-1 on blood vessels that facilitated the extravasation of CD8+ T cells that express its ligand, VLA-4, into tumors [30]. Thus, depending on the tumor context, both the pro-angiogenic and pro-inflammatory SASP can lead to vascular remodeling that can promote tumor cell intravasation through blood vessels and dissemination to distant organs or conversely immune cell extravasation and infiltration into tumors. More research is needed to understand the role of the pro-angiogenic SASP and senescence-mediated vascular remodeling in immune responses in other cancer settings. Nonetheless, evidence from preclinical models that senescence can directly or indirectly modulate anti-tumor immune responses through the SASP, and in some instances effectively block tumor formation or even shrink existing established tumors, has paved the way toward exploring whether senescence-inducing cancer therapies can achieve similar outcomes.
3. Therapy-induced senescence and immune regulation in cancer
Many FDA-approved chemo-, radio-, and targeted therapies have now been shown to induce senescence in cancer cells in vitro and tumor models in vivo depending on the dose, cancer type, and genetics of the tumor (Fig. 1), as summarized extensively in other reviews [17, 67]. Studies in preclinical mouse models that allow functional dissection of tumor and immune responses in a physiologically-intact setting have informed us of the mechanisms by which TIS regulates of the immune system, highlighting roles for the SASP in mediating anti-tumor immune surveillance, or, opposingly, the generation of a tumor-permissive immune suppressive microenvironment. The off-target effects of systemic therapies on senescence induction in non-tumor cells, as well as prevalent loss of tumor suppressor pathways (p53/p21, p16/RB) associated with cellular senescence in late-stage tumors, likely contribute to these diverse tumor and immune responses to TIS. Here we detail the known effects of different classes of senescence-inducing therapies on immune regulation, stromal remodeling, and immunotherapy outcomes in preclinical cancer models and clinical cancer patient samples (summarized in Table 1).
Fig. 1. Overview of senescence regulation of tumor-immune interactions following cancer therapy and potential senotherapeutic interventions.
(1) Certain cancer therapies, including radiation, chemotherapy, and targeted therapies such as CDK4/6 and AURKA inhibitors, can cause cellular senescence through induction of DNA damage, a prolonged DDR orchestrated by ATM/ATR, and/or downstream activation of p53 and RB-regulated pathways, leading to cell cycle arrest. (2) RB and epigenetic modifiers lead to chromatin changes in senescent cells that repress accessibility at cell cycle genes but increase open chromatin and binding of BRD4 to enhancer regions at SASP gene loci. (3) RB, ATM/ATR, p38MAPK, GATA4, and NOTCH1, as well as the cGAS-STING pathway that is activated by binding cytosolic double-stranded DNA, also interact with and activate TFs such as NF-κB, C/EBPβ, and STATs that bind to SASP gene promoters and promote their transcription. (4) Depending on the mechanism of action of the therapy, the cancer and cell type it acts on, and the upstream epigenetic and transcriptional regulators activated, the composition of the SASP can be incredibly heterogeneous, comprising pro-inflammatory, anti-inflammatory, and angiogenic factors, as well as lipids, MMPs, and other growth factors. (5) Many of these SASP factors highlighted have been shown to modulate the immune system and the vasculature in both tumor suppressive and tumor-promoting ways. In addition, the upregulation of cell surface proteins on senescent cells as part of the SASP can allow them to be recognized by (MHC-I, NK ligands) or alternatively evade (HLA-E, PD-L1) the immune system. (6) The secretome and surfaceome of senescent tumor cells can be leveraged for immunotherapy, including ICB therapies to block PD-L1 and HLA-E, CAR-T cells and antibodies engineered to bind senescence-related surface proteins, and neutralizing antibodies to target immune suppressive SASP factors, such as IL-6, IL-8, and TGF-β, and their receptors. (7) Similarly, senomorphic agents targeting SASP transcriptional regulation, including inhibitors of BRD4 (JQ1), STAT3 (Ruxolitinib), NF-κB (BAY 11–7082, Metformin), and p38MAPK (SB203580) can be used to modulate specific SASP programs. (8) Alternatively, senolytic agents targeting pro-survival and anti-apoptotic pathways in senescent cells such as BCL-2 (Navitoclax), MDM2 (UBX101), and PI3K signaling (Dasatinib + Quercetin) can be used to kill deleterious senescent tumor cells. AURKA, Aurora Kinase A; CAR-T, chimeric antigen receptor (CAR) T cell; DDR, DNA damage response; ICB, immune checkpoint blockade; mAb, monoclonal antibodies; MMP, matrix metalloproteinases; NCID, NOTCH intracellular domain; Pol II, RNA polymerase II; SASP, senescence-associated secretory phenotype; TF, transcription factor.
Table 1.
Senescence-inducing therapies and their known effects on immune and immunotherapy responses in cancer.
Therapy Class | Therapy/Combination | Cancer Type (model) | Senescence Biomarkers | Immune Response | Tumor Response | Refs |
---|---|---|---|---|---|---|
Ionizing Radiation (IR) | IR | IR-induced osteosarcoma mouse model | SA-β-gal, p16, p21, SASP (IL-6, CCL2/3/4/5) | NKT ↑ | ↓ | 88 |
IR | NSCLC human cell line xenografts | SA-β-gal, STING, NF-κB, L1, p21, SASP (IFN-β, IL1α, IL-6) | Mac ↑ | ↓ (distal tumor) | 98 | |
IR + PARPi (ex vivo in tumor cells) | Melanoma and PDAC syngeneic transplant mouse models | SA-β-gal, p16, p21, SASP (CCL5, IFNβ, CXCL9/10/11) | DC ↑ CD8+T ↑ NK ↑ | ↓ | 103 | |
Chemotherapy | Cyclophosphamide | B cell lymphoma syngeneic transplant mouse model | SA-β-gal, NF-κB, p15, SASP (IL-6, IL-8, ICAM-1, CXCL1) | NK ↑ | ↓ | 74 |
Doxorubicin or melphalan | MM syngeneic transplant mouse model | SA-β-gal, p16, p53, NK ligands (RAE-1, MICA, MULT-1, PVR) | NK ↑ | ↓ | 75–77 | |
Cisplatin + irinotecan (ex vivo in tumor cells) | Ovarian cancer syngeneic transplant mouse models | SA-β-gal, STING, p16, yH2AX, SASP (IL-6, VEGFA, GM-CSF) | DC ↑ CD8+T ↑ | ↓ | 78 | |
Docetaxel | PCa GEMM | SA-β-gal, p16, p21, SASP (GM-CSF, CSF-1, IL-10, CCL2, CXCL1/2) | MDSC ↑ NK ↓ CD8+T ↓ | ↑ | 61 | |
Mitoxantrone, other agents | PCa human xenografts; PCa patient samples | SA-β-gal, p16, SASP (IL-6, IL-8, MMPs, AREG), PD-L1 | CD8+T ↓ | ↑ | 72 | |
Aurora kinase inhibitors | MLN8054/MLN8237 (AURKAi) | Melanoma human xenografts, PDXs, syngeneic transplant mouse models | SA-β-gal, NF-κB, SASP (IL-6, IL-8, CCL5, CXCL1/2) | Mac ↑ CD8+T ↑ | ↓ | 111, 113 |
MLN8237 (AURKAi) | Melanoma patient samples | SASP (CCL5) | CD8+T ↑ | N/A | 113 | |
AZD1152 (AURKBi) | Melanoma and CRC syngeneic transplant mouse models | SA-β-gal, p21 | CD8+T ↑ | - | 114 | |
Cell Cycle inhibitors | Abemaciclib (CDK4/6i) | ER+ breast cancer GEMM and PDXs | SA-β-gal, MHC-I | CD8+T ↑ Treg ↓ | ↓ | 124 |
Abemaciclib (CDK4/6i) | Melanoma syngeneic transplant mouse models | SA-β-gal, SASP (CCL20, CX3CL1) | ↓ T cell suppression signature | - | 126 | |
Palbociclib (CDK4/6i) + Trametinib (MEKi) | LUAD GEMM | SA-β-gal, NF-κB, p15, SASP (TNFα, ICAM-1, IL-15, NKG2D ligands) | NK ↑ | ↓ | 29 | |
Palbociclib (CDK4/6i) + Trametinib (MEKi) | PDAC GEMM | SA-β-gal, SASP (VEGFs, PDGFs, MMPs, IL-6, CXCL1, CCL5), MHC-I, PD-L1 | CD8+T ↑ | - | 30 | |
Palbociclib (CDK4/6i) (ex vivo in fibroblasts) | Melanoma syngeneic transplant mouse models | SA-β-gal, NF-κB, p16, SASP (IL-6, MMP3, CCL6, CCL8, CCL11) | MDSC ↑ | ↑ | 127 | |
XL413 (CDC7i) | HCC GEMM and human xenografts | SA-β-gal, p16 | Mac ↑ CD8+T ↑ CD4+T ↑ | ↓ | 128 | |
Senescenceinducing + Immunotherapy Combinations | Cisplatin + irinotecan (chemotherapy) + α-PD-1 ICB | Ovarian cancer syngeneic transplant mouse models | SA-β-gal, STING, p16, yH2AX, SASP (IL-6, VEGFA, GM-CSF) | CD8+ T infiltration + activation ↑, DC ↑ | ↓↓ | 78 |
Mitoxantrone (chemotherapy) + α-PD-1 ICB | PCa human xenografts | SA-β-gal, p16, SASP (IL-6, IL-8, MMPS, AREG), PD-L1 | CD8+T infiltration + activation ↑ | ↓ | 72 | |
MLN8237 (AURKAi) + α-CD137 (T cell agonist) | Melanoma syngeneic transplant mouse models | SA-β-gal, NF-κB, SASP (IL-6, IL-8, CCL5, CXCL1/2) | CD8+T infiltration + activation ↑ | ↓↓ | 113 | |
AZD1152 (AURKBi) + α-CTLA-4 ICB | Melanoma and CRC syngeneic transplant models | SA-β-gal, p21 | CD8+T ↑ | ↓ | 114 | |
Abemaciclib (CDK4/6i) + α-PD-1 ICB | ER+ breast cancer GEMM | SA-β-gal, MHC-I | CD8+T ↑ Treg ↓ | ↓↓ | 124 | |
Abemaciclib (CDK4/6i) + α-PD-1/CTLA-4 ICB | Melanoma syngeneic transplant mouse models | SA-β-gal, SASP (CCL20, CX3CL1) | ↓ T cell suppression signature | ↓ | 126 | |
Palbociclib (CDK4/6i) + Trametinib (MEKi) + α-PD-1 ICB | PDAC GEMM | SA-β-gal, NF-κB, SASP (VEGFs, MMPs, PDGFs, IL-6, CXCL1, CCL5), MHC-I, PD-L1 | CD8+T numbers + activation ↑ | ↓ | 30 |
AURKA, Aurora Kinase A; AURKB, Aurora Kinase B; CRC, colorectal cancer; DC, dendritic cell; ER, estrogen receptor; GEMM, genetically engineered mouse model; HCC, hepatocellular carcinoma; i, inhibitor; ICB, immune checkpoint blockade; IR, ionizing radiation; L1, LINE-1; LUAD, lung adenocarcinoma; Mac, macrophage; MDSC, myeloid-derived suppressor cell; MM, Multiple Myeloma; NK, Natural Killer cell; NKT, Natural Killer T cell; NSCLC, non-small cell lung cancer; Pca, prostate cancer; PDAC, pancreatic ductal adenocarcinoma; PDX, patient-derived xenograft; SA-β-gal, senescence-associated beta-galactosidase; SASP, senescence-associated secretory phenotype. For tumor response: (↑) = increased tumor growth compared to control; (-) = no change in tumor growth compared to control; (↓) = decreased tumor growth compared to control; (↓↓) = further decrease in tumor growth compared to single agents alone. Ex vivo indicates that cells were treated with drugs in culture prior to their implantation into mouse models with existing tumors.
3.1. Chemotherapy
Chemotherapies can trigger cellular apoptosis or senescence depending on dose and mechanism of action of the chemotherapeutic agent [67, 68]. Low dose chemotherapy induces senescence over time through induction of DNA damage and a prolonged DDR mediated by ATM/ATR that converges to activate p53/21 and RB pathways and block cell division (Fig. 1). Classes of chemotherapeutic agents that promote senescence include: (a) DNA synthesis inhibitors (e.g. gemcitabine, 5-fluorouracil (5-FU), methotrexate) that lead to replication fork stalling and DNA damage; (b) topoisomerase inhibitors (e.g doxorubicin, etoposide, mitoxantrone) that prevent DNA re-ligation after supercoil unwinding, leading to single and double-strand DNA breaks (DSBs); (c) platinum-based and alkylating agents (e.g. cisplatin, cyclophosphamide, melphalan) that induce extensive genotoxic damage through DNA-crosslinking; and (d) microtubule inhibitors (e.g. docetaxel, paclitaxel) that interfere with microtubule spindles during the metaphase-to-anaphase transition to activate the mitotic checkpoint [17]. Markers of senescence, including SA-β-gal, p16, p21, and p53, as well as SASP factors IL-6, IL-8 (CXCL8), amphiregulin (AREG), VEGF, and CCL2 have been identified locally and in circulation in breast, lung, and prostate cancer patient samples following treatment with different classes of chemotherapeutic agents [20, 69–72]. In mice, senescence has been shown to be critical for effective chemotherapy responses in B cell lymphomas that have disabled apoptotic pathways through BCL-2 overexpression [73], suggesting that chemotherapy-induced senescence may contribute to tumor suppression in certain contexts.
Some of these tumor suppressive aspects of chemotherapy may depend on senescence-mediated anti-tumor immunity. Cyclophosphamide treatment drives an NF-κB-mediated SASP that activates NK cell immune surveillance in B cell lymphoma models [74]. In multiple myeloma, low doses of doxorubicin and melphalan induce expression of ligands on senescent tumor cells for the activating NK cell receptors NKG2D (RAE-1, MICA, MULT-1) and DNAM-1 (PVR) that lead to NK cell recognition and cytotoxicity toward tumor cells [75–77]. A recent study showed that intraperitoneal injection of chemotherapy-induced senescent cells resulted in their infiltration into established ovarian tumors in mice and an increased influx of DCs and CD8+ T cells that sensitized resistant tumors to immunotherapy targeting the PD-1 checkpoint on T cells [78]. This demonstrates the proof-of-concept that senescent tumors cells following chemotherapy treatment could serve as an immune adjuvant to promote anti-tumor immunity and immunotherapy efficacy.
Conversely, chemotherapy-induced senescence can also lead to immune suppression in tumors and inflammation and fibrosis in normal tissues. Chemotherapeutic induction of matrix metalloproteinases (MMPs) as part of the SASP leads to cleavage of NKG2D ligands on the surface of senescent tumor cells that allows them to evade NK cell surveillance and persist following cancer therapy [57]. Moreover, in a prostate cancer model, docetaxel treatment drove an influx of MDSCs through induction of anti-inflammatory SASP factors that led to NK and T cell suppression and chemoresistance [61]. Due to their systemic administration, chemotherapies can promote senescence in stromal cells and normal tissues that drives local and systemic inflammation leading to chemotoxicity, accelerated aging, and tumor relapse and metastasis [32, 79–82]. Bleomycin, which is used to treat Hodgkin’s lymphoma and testicular germ cell tumors, induces senescence in fibroblasts in the lungs of normal mice that leads to development of idiopathic pulmonary fibrosis (IPF) through secretion of fibrogenic SASP factors such as TGFβ, PAI1, IL-6, and MMPs, as well as a class of lipids known as leukotrienes [83, 84]. In addition, chemotherapy treatment in prostate cancer patients and xenograft models led to secretion of the SASP factor AREG from senescent stromal cells that promoted expression of the immune inhibitory checkpoint PD-L1 on tumor cells that contributed to T cell suppression and chemoresistance [72]. Thus, induction of senescence in bystander cells and tissues can limit the anti-tumor activity of systemic cytotoxic therapies. In the future it will be important to understand how the mechanisms of action of different classes of chemotherapy may dictate the degree and type of senescence induction and SASP-mediated immune and tumor responses in preclinical and clinical settings.
3.2. Radiation
Unlike the systemic effects of senescence-inducing chemotherapy, radiotherapy is administered locally to the tumor site, limiting damage to noncancerous tissues. Ionizing radiation (IR) therapy can trigger senescence through induction of DSBs and an ATM/ATR-dependent DDR, leading to activation of p53 and its target p21 (Fig. 1). IR induces senescence markers in vitro and in vivo, including SA-β-gal, p53, p21, and various SASP factors, and preclinical evidence suggests that senescence plays a major role in radiosensitivity in cancer [85–89].
Radiotherapy is known to promote anti-tumor immunity in cancer patients [90, 91]. Indeed, CD8+ T cells are required for IR-mediated tumor ablation in mice [92, 93]. Besides triggering the presentation of immunogenic tumor antigens [94], IR-induced DNA damage and accumulation of cytoplasmic double-stranded DNA fragments also activates nucleic acid sensors, such as cGAS-STING and the downstream NF-κB pathway, to stimulate production of SASP-associated cytokines and chemokines and expression of NK cell activating ligands on irradiated cells that promote innate and adaptive immune surveillance [95–102]. For instance, release of extracellular vesicles (EVs) containing cGAS-STING-activating DNA:RNA hybrids and retrotransposable elements (RTEs) and SASP factors IL-6, IL-1α, and IFN-β following tumor irradiation can mediate paracrine senescence, macrophage activation, and immune-mediated tumor regression in distal non-irradiated tumors through a rare phenomenon known as the abscopal effect [98]. IR has also been shown to induce RB-dependent senescence and IL-6 secretion that promotes immune surveillance by Natural Killer T (NKT) cells in osteosarcomas [88]. Combining IR with PARP inhibition to reinforce DNA repair suppression and exacerbate DNA damage in melanoma and pancreatic tumor models led to production of inflammatory SASP factors such as CCL5, IFN-β, and CXCL9/10/11 that promoted DC proliferation and enhanced both CD8+ T cell and NK cell anti-tumor immunity [103]. Interestingly, in an immune-deficient glioblastoma model, radiation-induced senescence accelerated tumor growth [87], underscoring the importance of immune responses in determining the therapeutic outcome of senescence-inducing radiotherapy.
As with chemotherapy, some of the immune stimulating aspects of radiation therapy are counterbalanced by off-target effects in normal cells. Radiation-induced immunosenescence can inhibit effective immune responses [104, 105], and clearance of senescent immune cells restores T cell proliferation and macrophage phagocytic activity [106]. Furthermore, persistent senescence following radiation in normal tissues contributes to chronic inflammation, fibrosis, and tissue dysfunction reminiscent of accelerated aging [107, 108]. Therefore, to improve therapy outcomes it will be important to develop therapies which target senescence more specifically to tumor cells.
3.3. Aurora kinase inhibitors
Aurora kinases are mitotic regulatory proteins responsible for proper segregation of chromosomes during mitosis and are commonly overexpressed in human cancers [109, 110]. Inhibition of Aurora kinases is known to induce senescence through blocking mitosis (Fig. 1), leading to genomic instability, polyploidy, and an ATM/CHK2-mediated DDR [111, 112]. In melanoma models, Aurora kinase A (AURKA) inhibition with MLN8054 or MLN8237 induced DNA damage, SA-β-gal expression, and an NF-κB-driven SASP that promoted macrophage recruitment, immune surveillance, and long-term immune control of tumors [111]. Follow-up studies revealed that AURKA inhibition could also induce SASP production of chemokine CCL5 to recruit T cells into melanoma lesions, and that combining AURKA inhibitors with a T cell agonist antibody targeting CD137 further enhanced anti-tumor responses [113]. This provided one of the first examples that senescence-inducing and immunotherapies could synergize through the ability of the SASP to recruit cytotoxic immune cells into the TME. Supporting these findings, melanoma patients that gained clinical benefit from treatment with AURKA inhibitors displayed increased CCL5 expression associated with greater infiltration of activated cytotoxic T cells [113]. Similarly, treatment with an Aurora kinase B (AURKB) inhibitor in melanoma models was shown to promote T cell-driven anti-tumor immunity and synergize with an antibody targeting the immune checkpoint CTLA-4 on T cells in a manner that was partially dependent on p21-mediated senescence in tumor cells [114]. As Aurora kinase inhibitors have also been shown to act directly on T cells and conversely inhibit their activation and proliferation [115–117], moving forward it will be important to consider the correct dose and schedule for combining senescence-inducing and immunotherapies to maximize immune activity and therapeutic benefit.
3.4. CDK inhibitors
Cyclin-dependent kinases (CDKs) regulate cell cycle checkpoints that control growth and arrest in both normal and malignant cells [118, 119]. As cyclins and CDKs are aberrantly hyperactivated, and their inhibitors, including p16, p21, and p27, are commonly suppressed in cancer cells, the use of pharmacological CDK inhibitors is a promising approach to induce cell cycle arrest in tumors that have lost cell cycle control [120]. The most clinically developed and well-studied CDK inhibitors are dual CDK4/6 inhibitors, which include the FDA-approved compounds palbociclib, ribociclib, and abemaciclib [121]. CDK4/6 inhibitors act in a way as a p16 mimetic to inhibit the CDK4 and CDK6 kinases that regulate the G1/S phase checkpoint by blocking their ability to phosphorylate RB, which can then sequester E2Fs to arrest cells in G1 (Fig. 1). As such, a functional RB protein is required for CDK4/6 inhibitors to have anti-neoplastic activity [122]. Strong RB activation following CDK4/6 inhibitor treatment alone can in certain cell types and contexts lead to senescence, for example in CDK4-amplified liposarcomas, estrogen receptor (ER)-positive breast cancers, and melanomas, as well as in normal fibroblasts [123]. While studies have begun to elucidate certain mechanisms necessary for CDK4/6 inhibitors to mediate senescence, including MDM2 downregulation, ATRX redistribution, HRAS suppression, and mTOR activation, the determinants governing whether CDK4/6 inhibitors induce transient quiescent or durable senescent states in different cancer settings are not completely understood [122].
It is now appreciated that CDK4/6 inhibitors can also directly (through their effects on immune cells) or indirectly (through induction of senescence) promote anti-tumor immunity in various cancer settings. Abemaciclib treatment in ER+ breast cancer models induced SA-β-gal activity, increased MHC-I levels, and triggered expression of endogenous retroviral elements and a type III interferon response that led to increased cytotoxic CD8+ T cell infiltration and synergy with anti-PD-1 immune checkpoint blockade (ICB) [124]. CDK4/6 inhibition also prevented the expansion of immune suppressive Tregs, though this response is likely a direct effect of CDK4/6 inhibitors on Treg function [124, 125]. Similarly, abemaciclib could reverse tumor intrinsic immune suppression associated with ICB resistance in human melanoma patients, induce SASP components such as CCL20 and CX3CL1, and improve combined anti-PD-1/CTLA-4 ICB efficacy in melanoma models [126]. It is important to note that CDK4/6 inhibitors can also have off-target effects on other stromal cells in the TME. Normal fibroblasts treated with palbociclib underwent senescence and produced an NF-κB-driven SASP that promoted MDSC infiltration and enhanced melanoma growth in vivo [127]. Targeting alternative cell cycle regulators may offer even more specificity towards activating senescence in cancer cells. For instance, inhibition of DNA replication kinase CDC7 with XL413 was recently shown to induce senescence specifically in tumors with p53 mutations and lead to SASP-mediated macrophage and CD4+ and CD8+ T cell immune surveillance in liver tumors [128].
Combining CDK4/6 inhibitors with other molecularly targeted therapies directed at cancer-specific oncogenic driver pathways also provides a means to increase selectivity and reinforce cellular senescence in tumor cells. We and others have shown that palbociclib in combination with the MEK inhibitor trametinib (T/P) can induce senescence and a robust SASP in KRAS-driven lung and pancreatic cancer models [29, 30, 129]. In KrasG12D mutant; p53-null lung adenocarcinoma models, SASP-mediated TNFα secretion and ICAM-1 and NKG2D ligand expression on senescent tumor cells following T/P treatment led to an influx of NK cells, NK cell-mediated tumor clearance, and long-lasting tumor control [29]. In KrasG12D mutant; p53-null pancreatic cancer models, a different SASP response and outcome on the surrounding TME was observed. T/P-induced senescence produced a SASP rich in both angiogenic factors, including VEGFs, FGFs, PDGFs, and MMPs that led to increased vascularization, as well as inflammatory factors such as IL-6, CXCL1, and CCL5 that simulated endothelial cell activation, culminating in increased vascular trafficking of cytotoxic CD8+ T cells into pancreatic tumors [30]. T cell influx, combined with increased MHC-I and PD-L1 expression on senescent tumors, made these immunologically “cold” pancreatic tumors “hot” and sensitized them to anti-PD-1 ICB [30].
Collectively, evidence from these different classes of cancer therapies suggests that senescence could be a means to enhance immunotherapy efficacy through SASP-mediated remodeling of the TME. Indeed, genomic correlates and transcriptional signatures related to senescence and the SASP are also enriched in cancer patients that respond well to PD-1/PD-L1 ICB [126, 130, 131], supporting the potential clinical relevance of senescence as an immunotherapy booster. Though more evidence connecting TIS and the mechanisms by which it achieves immune modulation is needed in vivo and especially in clinical samples overall, these results highlight the dynamic changes that senescence-inducing targeted therapies can have on the surrounding TME. A deeper knowledge of how the SASP is regulated in different therapy scenarios and which SASP factors mediate these divergent immune responses will allow us to strategize how to fully harness the SASP for cancer immunotherapy going forward.
4. Dynamic SASP regulation in cancer
The SASP is regulated on multiple levels, including epigenetically and transcriptionally, at the protein translation and secretion stage, and at the cellular and tissue level. Here we examine dynamic SASP regulation following senescence induction to gain insight into how the SASP output can be optimized for therapy in different cancer settings.
4.1. Epigenetic regulation
Senescent cells undergo a vast array of epigenetic changes, including altered chromatin occupancy, composition, and modifications [132], which in addition to repressing cell cycle genes can prime SASP gene expression (Fig. 1). RB forms a complex with histone deacetylases (HDACs) and the methyltransferase SUV39H1 to mediate H3K9 trimethylation (H3K9me3), often referred to as SAHF, at E2F target and cell cycle regulatory genes to repress their expression and maintain senescent cells in a growth arrested state [133–135]. Despite increased facultative chromatin at SAHF loci, in general senescent cells have reduced heterochromatin globally, correlating with loss of gene silencing and increased accessibility and expression of SASP genes and other senescence-related transcriptional targets [136]. Recent work demonstrated that the chromatin modifier HMGB2 is critical for limiting repressive heterochromatin on SASP gene loci during SAHF formation at cell cycle genes [137]. HDACs such as SIRT1 that deacetylate H3K27 and components of polycomb repressor complex 2 (PRC2) such as methyltransferase EZH2 that trimethylates H3K27 (H3K27me3) to condense chromatin and block enhancer occupancy at the p16 gene locus as well as SASP genes are silenced during senescence [138, 139]. Chromatin readers such as the bromo and extra terminal (BET) domain protein 4 (BRD4) can then be recruited and are free to bind open enhancer regions [140]. BRD4 activity is required for SASP gene expression during oncogene-induced senescence and is crucial for senescence-mediated NK cell immune surveillance and tumor suppression in the liver [140].
In addition, degradation of the methyltransferases G9a and GLP leads to further loss of H3K9 methylation and activates promoters at SASP genes including IL6 and IL8 [141]. Some chromatin modifiers, such as the disruptor of telomeric silencing 1-like (DOT1L), can lead to increased H3K79me2/3 occupancy at specific SASP genes, such as IL1A [142]. Finally, reorganization of histone variants including removal of macroH2A1 [143] and accumulation of H2A.J [144] at SASP loci reinforces SASP gene expression during senescence. This demonstrates that remodeling of chromatin landscapes, particularly in enhancer regions, is critical for SASP gene program regulation and may be necessary for immune clearance of senescent cells. A more comprehensive view of the epigenetic changes that influence expression of particular SASP gene loci in oncogene-induced senescence and other contexts, including TIS, will provide a deeper understanding of dynamic SASP transcriptional regulation. Further application of 3D and 4D DNA topological analyses (i.e. Hi-C, 3C) may help to provide additional granularity into how DNA structure regulates SASP expression [145–147].
4.2. Transcriptional regulation
Cell cycle and DDR pathways unite to activate the expression of SASP genes through downstream transcriptional regulators (Fig. 1). Apart from their canonical roles in mediating senescence-associated cell cycle arrest, p21 and p16 have very recently been shown to activate and maintain the SASP, possibly through RB-mediated transcriptional mechanisms [40, 148]. In addition to activating RB, it has been well-established that many senescence-inducing stimuli produce DNA damage that directly or indirectly, through mobilization of DDR proteins such as ATM/ATR and CHK1/CHK2, converges to activate a set of common transcription factors (TFs) that regulate SASP gene expression, including NF-κB, C/EBP-β, and GATA4 [21, 74, 149–151] (Fig. 1).
NF-κB and C/EBPβ can directly bind to DNA and synergistically activate the expression of mainly pro-inflammatory SASP genes such as IL1A, IL6, and IL8 [21, 74, 149]. Following DNA damage, ATM and ATR activate GATA4 upstream of NF-κB to regulate SASP expression [151]. The p38MAPK pathway is activated by oxidative stress and can also reinforce NF-κB transcriptional activity and stabilize SASP mRNA transcripts independent of the DDR [152–154]. Additionally, senescent cells harbor high levels of cytoplasmic DNA fragments due in part to loss of Lamin B1 and nuclear envelope dissolution [155, 156]. Cytosolic DNA is sensed by cGAS, which activates the downstream STING pathway resulting in the activation of interferon regulatory factor 3 (IRF3) and NF-κB and induction of interferon genes (e.g. IFN-β) and other pro-inflammatory SASP factors in senescent cells [100–102, 157]. Interestingly, p38MAPK has been shown to inhibit STING-mediated induction of interferon signaling while leaving NF-κB-driven SASP intact [102]. These inflammatory pathways downstream of the DDR also induce the expression of ligands for activating receptors NKG2D and DNAM-1 on NK cells that are important for their recognition and targeting of senescent cells [158, 159]. Underscoring their importance as SASP transcriptional regulators, NF-κB or cGAS-STING suppression alone is sufficient to ablate the expression of many pro-inflammatory SASP factors and subsequent immune surveillance of senescent cells. NF-κB signaling blockade inhibits key SASP factors (TNFα, CCL2) and cell adhesion molecules (ICAM-1) necessary for NK cell infiltration and targeting of senescent tumors [29, 74], and leads to recruitment of macrophages and suppressive MDSCs into tumors [160]. cGAS or STING genetic knockout in mice leads to reduced expression of SASP factors IL-1α, IL-1β, IL-6, IL-8, and CXCL10 in pre-malignant hepatocytes and inhibits their clearance by the innate and adaptive immune system [100, 102].
Other transcriptional regulators have been shown to generate a different SASP milieu beyond canonical IL-1α, IL-6, IL-8, and other pro-inflammatory SASP factors (Fig. 1). The JAK2/STAT3 pathway, which can be induced by autocrine IL-6 signaling, regulates the expression of immunosuppressive SASP factors (e.g., GM-CSF, M-CSF (CSF-1), IL-10, CXCL1, CXCL2) following Pten loss-induced senescence in prostate cancer that hinders immune surveillance [61]. JAK2 inhibitor treatment reprogrammed the immunosuppressive SASP, reversed MDSC-driven immune suppression, and promoted T cell-mediated anti-tumor immunity following docetaxel-induced senescence [61]. In oncogene-induced senescence, activation of NOTCH1 suppresses C/EBPβ activity to block the pro-inflammatory SASP and produces an alternative TGFβ-rich SASP, highlighting a pivotal role of NOTCH1 in governing functional composition of SASP [161]. Inhibition of activated NOTCH signaling promoted increased infiltration of T cells and clearance of pre-malignant NrasG12V senescent hepatocytes [161], suggesting that targeting NOTCH in senescent tumors cells could be a potential strategy to enhance immune surveillance in tumors. Interestingly, recent work suggests that p21 can mediate a unique SASP (termed PASP) through facilitating RB transcriptional complexes with STAT and SMAD TFs, including STAT1/3 and SMAD2/3, which promote the secretion of pro-inflammatory SASP factors such as CXCL14 that potentiate immune surveillance in the liver [40]. P53 itself may also play dichotomous roles in SASP regulation. Whereas p53 restoration in established liver tumors can promote a pro-inflammatory SASP [39], other reports have demonstrated that p53 can actually suppress p38MAPK and NF-KB activity, and that p53 loss further enhances pro-inflammatory SASP production [20, 153, 162–164]. Thus, different TFs and TF complexes can regulate different arms of the SASP and contribute to its dynamic regulation of the immune system following TIS (Fig. 1).
4.3. Post-transcriptional regulation
mTOR regulates the translation of SASP proteins, including IL-1α, which in turn reinforces NF-κB activation to promote the expression of other pro-inflammatory SASP genes [165]. Indeed, IL-1α knockout was sufficient to suppress NF-κB-driven SASP production and macrophage recruitment into preinvasive pancreatic lesions following KrasG12D induction [166]. Other SASP factors, including IL-6, IL-8, VEGF, CXCL1/2, CCL2, CCL20, TGFβ, and lipid species such as prostaglandins have been shown to reinforce autocrine senescence and SASP as well as promote paracrine senescence in neighboring cells through activation of downstream TFs such as NF-κB [21, 59, 149, 167–169]. mTOR also regulates the translation of MK2, which phosphorylates the RNA binding protein ZFP36L1 and inhibits its ability to degrade SASP transcripts [170]. As such, mTOR inhibition using rapamycin abolishes SASP induction [165, 170, 171]. In vivo, rapamycin treatment blocked recruitment of T cells, B cells, NK cells, and macrophages into the liver and subsequent immune clearance of senescent NrasG12V mutant hepatocytes [170]. Enzymatic cleavage of SASP-associated proteins can also impact their maturation and regulation. IL-1 signaling in senescent cells is mediated in part by inflammasome components NLRP3 and TLR2, whose activation is required for caspase-1 cleavage of pro-IL-1α, -IL1β, and -IL-18 to their mature forms [168, 172]. Moreover, genetic loss of SASP factor TIMP1 in prostate cancer has recently been shown to lead to activation of MMPs, which in turn can regulate soluble factors present in the SASP through their proteinase activity [31, 173].
In addition, transcriptional derepression of RTEs such as LINE-1 (L1) that occurs during late senescence (following earlier SASP induction) leads to induction of type I interferon signaling that maintains the SASP [174]. PTBP1 has been shown to control SASP secretion by regulating the splicing of genes encoding proteins involved in intracellular trafficking. Paradoxically, SASP suppression following PTBP1 blockade limited liver tumorigenesis despite impairing immune surveillance [175]. Non-coding RNAs such as microRNAs (miRNAs) can also regulate SASP transcription, mRNA stability, and translation [176]. For example, miR-146a/b is induced by NF-κB but subsequently forms a negative feedback loop to suppress NF-κB activity and reduce IL-1α, IL-6, and IL-8 expression [177]. It is now come to be appreciated that SASP proteins, as well as DNA:RNA hybrids, miRNAs, and L1, can also be released from senescent cells in the form of EVs such as exosomes [98, 178–180] and signal in a paracrine manner. Still, whether soluble vs. vesicle-bound DNA, RNA, and protein species associated with the SASP may have differential effects on neighboring cells and immune responses is still unclear.
4.4. Tissue and cell type dependent SASP regulation
The SASP produced following senescence induction is highly variable and dependent on the senescence-inducing stimuli and cell and tissue type it acts on. For example, the same cells treated with different genotoxic therapies can have dramatically divergent SASPs with few overlapping genes [181]. The SASP composition following treatment with the same senescence-inducing agent can also be heterogenous across cancer cells with diverse genetic backgrounds, different cell types within the same tumors, and even among cells of the same type derived from different donors [12, 20, 182]. This may be partially dependent on the unique epigenetic footprints of cells from distinct origins [182]. As such, epithelial and stromal cells treated with the same senescence-inducing drugs have quantitatively and qualitatively different SASP profiles [20, 179, 181–184]. In addition, within a population of senescent cells, there can also be marked heterogeneity in SASP gene expression at the single cell level suggesting that different cells within a population may contribute to different SASP profiles [185]. A more comprehensive and high-throughput analysis of the SASP in distinct cell populations at the single cell level in different tumor and therapy contexts will be necessary to fully grasp the cellular mechanisms leading to differential SASP profiles and subsequent immune and tumor responses to senescence.
The resident tissue microenvironment that tumors reside in may also impact SASP expression and its outcomes on immune responses. RAS overexpression in pre-malignant hepatocytes or restoration of p53 in liver tumor cells or hepatic stellate cells leads to SASP-dependent innate and adaptive immune activation that effectively controls HCC inception and progression [28, 39, 54], suggesting the SASP promotes anti-tumor immunity in the liver. Similarly, in KrasG12D mutant; p53-null lung adenocarcinoma, TIS following T/P treatment activated an NF-κB-driven pro-inflammatory SASP that triggered NK cell immunity and immune-mediated tumor regressions [29]. In contrast, T/P treatment in genetically similar KrasG12D mutant; p53-null pancreatic cancer models triggered a SASP enriched in mainly pro-angiogenic factors that failed to activate NK cell-mediated tumor responses but rather promoted vascular remodeling that indirectly facilitated T cell infiltration [30]. This suggests that the same senescence-inducing stimuli can lead to different SASP outputs and immune and stromal outcomes based on the tissue. Our own preliminary experiments suggest that transplanting the same tumor cell line into different tissues in mice is sufficient to alter the SASP profile and subsequent immune and stromal responses in the TME following treatment with the same senescence-inducing therapies (unpublished). Therefore, it will be critical to characterize the types of SASP activated and their impact on the TME in each therapy and tumor context to (a) understand the specific SASP factors and regulators necessary for anti-tumor immunity and immunotherapy efficacy and (b) guide the development of strategies to harness or target the SASP to promote tumor responses following cancer therapy.
5. Modulating senescence with senotherapeutics for cancer immunotherapy
Combining senescence-inducing therapies with other agents to remove deleterious senescent cells or modulate SASP regulators or specific SASP factors in different cancer contexts may be necessary to optimize TIS for cancer therapy. The use of senotherapeutics to eliminate senescent cells (i.e. senolytics) or modulate the SASP (i.e. senomorphs) are promising strategies to block senescence-mediated immune suppression and/or potentiate SASP-mediated anti-tumor immunity (Fig. 1). Alternatively, overlaying senescence-inducing therapies with novel immunotherapy strategies directed toward the vulnerabilities that arise in senescent tumor contexts may allow us to further leverage the immune modulatory functions of senescence for tumor eradication (Fig. 1).
5.1. Senolytics
Senescence and the resulting SASP can in certain contexts suppress anti-tumor immune responses and promote cancer progression and relapse following cancer therapy [22, 31, 186]. Inducible ablation of p16-positive senescent cells in transgenic mice has been shown to mitigate age-related morbidities and inflammation, increase lifespan and healthspan, and reduce tumor initiation and relapse following therapy [32, 187–191]. This exciting proof-of-concept has led to the development and characterization of numerous senolytic agents to selectively eliminate senescent cells by targeting their vulnerabilities, most prominently anti-apoptotic pathways that allow them to survive and persist [192]. The most well-characterized senolytic agents include the multi-kinase inhibitor dasatinib, the BCL-2 family inhibitor ABT-263 (navitoclax), UBX0101 (MDM2 inhibitor) and FOXO4-DRI (FOXO4 blocker) that activate p53-mediated apoptosis, and natural products fisetin and quercetin that can inhibit pro-survival PI3K signaling [193] (Fig. 1). These agents have been shown to clear senescent cells in various tissues in old mice, reduce systemic SASP and inflammation, and alleviate age-related pathologies to extend health span and lifespan, thus blunting many of the deleterious effects of senescence during aging [190, 191, 194–198]. While ongoing clinical trials evaluating dasatinib and quercetin (D + Q) treatment have demonstrated select removal of senescent cells in humans and promising mitigation of physical dysfunction in IPF patients [199, 200], other trials testing UBX0101 in human osteoarthritis patients failed to show efficacy [201], highlighting that there is still much work to be done to effectively translate these senolytic approaches to chronic human diseases.
Senolytic strategies could also be used to potentiate or overcome resistance to cancer therapy and mitigate associated chemotoxicities. A promising tactic to leverage senolytics for cancer therapy is to combine a “one-two punch” of a senescence-inducing drug (1) followed by a senolytic agent (2) to induce synthetic lethality, as first described by Rene Bernards and colleagues [202]. Multiple studies have now shown that induction of tumor cell senescence with chemotherapy, radiation, and targeted Aurora kinase, PARP, or CDK4/6 inhibitors leads to induced susceptibility to senolytic BCL-2 family inhibitors and senescent cell apoptosis [203–209]. Another recent example is that CDC7 inhibitors induce senescence in HCC cells with p53 mutations that leads to synthetic lethality with sertraline, an antidepressant, or the mTOR inhibitor AZD8055 [128]. In addition, MDM2 inhibitor treatment in combination with senescence-inducing AURKA inhibition was shown to not only result in the death of senescent melanoma cells, but also lead to enhanced SASP expression of factors including CCL1, CCL5, and CXCL9 that further promoted the recruitment of both innate (macrophages, NK cells) and adaptive (T cells) immune cells and anti-tumor responses [210]. This suggests that senolytics could be used in certain settings to further enhance the immune stimulatory features of acute senescence while simultaneously removing detrimental senescent cells and chronic SASP expression. Navitoclax, FOXO4-DRI, and D + Q treatment has also been shown to eliminate senescent bystander cells following chemotherapy and radiation-induced senescence, leading to reduced treatment-related toxicities and morbidities and cancer recurrence [32, 84, 196, 211–213]. Thus, senolytics could be used as adjuvant therapy to maximize the effects of standard radiation and chemotherapy drugs that induce senescence. Indeed, numerous clinical trials combining navitoclax with chemotherapy or targeted therapy agents known to induce senescence have been carried out [214]; however, this approach has so far shown limited efficacy in various solid tumor settings.
Several roadblocks must be overcome to effectively apply senolytic agents to cancer therapy. First, the mechanisms by which many senolytic agents eliminate senescent cells need to be firmly defined. For instance, it is unclear how D + Q, or other natural products such as fisetin or sertraline selectively target senescent cells, as each of these drugs has multiple targets. Second, the activity of senolytic agents is not universal and is highly cell and tumor type specific. Response to navitoclax following TIS varies amongst cancer cell lines, which may be the result of genetic alterations in BCL-2 family or other pro-apoptotic genes [182]. Similarly, while D + Q treatment is ineffective in combination with senescence-inducing chemotherapy in liver cancer, this senolytic cocktail enhances the anti-tumor efficacy of senescence-inducing radiation in melanoma models [215, 216]. Biomarkers predictive of senolytic responses will thus need be identified. Finally, senolytic agents such as BCL-2 family inhibitors and activators of p53-mediated apoptosis (i.e. MDM2 inhibitors) are not specific for tumor or senescent cells and target cell death pathways common to all cells, leading to toxicities in patients [217, 218]. The recent development of galactose-modified nanoparticles and prodrugs that allow selective delivery of senolytic payloads to senescent cells through their high β-galactosidase activity may be an innovative means to overcome off-target toxicities [206, 219–221]. Still, additional on-target toxicities may arise from elimination of physiologically-relevant senescent cells, such as macrophages, megakaryocytes, and osteoblast progenitors [222–224], that could disrupt tissue or organismal homeostasis. It will be especially important to consider the impact of excessive senescent cell ablation in elderly individuals, where senescent cells may represent a large percentage of cells in a tissue and their removal could potentially threaten tissue integrity and function [225, 226]. Given the off-target effects of current senolytic drugs and potentially determinantal consequences of removing senescent cells throughout the body, a safer and more elegant approach may be to remodel SASP phenotypes and interactions between senescent cells and the tumor-immune microenvironment through senomorphic strategies.
5.2. Senomorphs
The use of senomorphic agents to abolish or remodel the SASP to limit tumor-propagating inflammation or promote anti-tumor immunity following TIS also holds therapeutic potential in cancer. The majority of senomorphs target key SASP regulators that can block all or parts of the SASP while leaving senescent cells and other tumor suppressive aspects of senescence in place [227]. Compounds targeting SASP epigenetic, transcriptional and post-transcriptional regulators, including JAK2/STAT3 (Ruxolitinib, NVP-BSK805) [61, 228, 229], NF-κB (metformin, BAY 11–7082) [74, 230–232], p38MAPK (SB203580, UR-13756) [153, 156, 233], mTOR (Rapamycin, RAD001) [165, 170, 232], L1 (lamivudine) [174], STING (H-151) [234, 235], and BRD4 (JQ1, iBET762) [140, 236, 237] have been shown to decrease SASP production (Fig. 1). However, the impact of SASP suppression on immune responses and pro- and anti-tumorigenic outcomes depends on the context. JAK2/STAT3 inhibition has been shown to block immune suppressive SASP programs to potentiate anti-tumor immune responses in Pten-null prostate cancer [61]. Likewise, the p38MAPK inhibitor SB203580 can reduce expression of the immune checkpoint HLA-E on senescent cells that blocks NK and T cell immune surveillance [56]. In contrast, mTOR, cGAS-STING, or BRD4 signaling blockade has been shown to inhibit anti-tumor immune surveillance that could contribute to tumor escape [102, 140, 170]. Thus, the impact of senomorphic agents on the TME is context-dependent and requires further evaluation in preclinical and clinical settings. Of note, these approaches are exquisitely dose dependent, with some drugs (e.g. BET, BRD4 inhibitors) mediating SASP suppression at low doses but senolysis at high doses [140, 236, 238].
An alternative senomorphic approach is to directly target specific SASP molecules that lead to immune suppression and tumor promotion. Monoclonal antibodies and other agents targeting cytokine signaling have been widely used in other cancer and inflammatory disease settings and are far along in clinical development [239]. These include drugs targeting IL-6/IL-6R (Tocilizumab, Siltuximab) [240, 241], IL-1α/IL-1β/IL1R (Anakinra, Canakinumab, Rilonacept) [242–244], and TNFα (Etanercept, Infliximab) [245, 246]. Though IL-6/IL-6R targeting alone or in combination with chemotherapy has largely failed to demonstrate therapeutic benefit in clinical trials for solid tumor malignancies [247–249], the IL-6R monoclonal antibody tocilizumab administered in combination with chemotherapeutic agents carboplatin and doxorubicin did lead to immune stimulation, including M1 macrophage polarization and activation of T cell effector functions, in patients with recurrent epithelial ovarian cancer [250]. Additionally, inhibition of IL-8 expression using liposome-encapsulated siRNAs reduced the growth of ovarian cancer xenografts [251]. Clinical efforts to target IL-8 or its receptors CXCR1/2 are underway, including the use of Reparixin in combination with paclitaxel in breast cancer patients that has already shown early promising results [252]. These studies push forward the paradigm that neutralizing immune suppressive SASP factors could be a strategy to enhance immune surveillance and anti-tumor responses following chemotherapy-induced senescence. As IL-8 has also been implicated in blunting the effects of ICB [253, 254], use of IL-8 signaling antagonists could be yet another strategy to improve immunotherapy outcomes following treatment with senescence-inducing therapies.
In the future it will be of interest to test inhibitors of other regulators of SASP-mediated immune suppression, including NOTCH and TGFβ antagonists that are under clinical development [255, 256], as well as activators of the pro-inflammatory SASP, such as STING agonists that are now being widely tested in the setting of cancer immunotherapy [257], for their ability to boost anti-tumor immunity following TIS. Though implementation of senomorphs may be safer than senolytics due to senescent and other normal cells remaining intact, senomorphic approaches still suffer from a lack of specificity and universality, and more work is needed to understand how their use in different cancer contexts may impact the SASP milieu in ways that ultimately drives tumor suppression or promotion.
5.3. Senescence-directed immunotherapy strategies
Alterations in both the secretome and surfaceome of senescent cells provide promising targets for existing and new immunotherapy strategies (Fig. 1). Senescent cells express altered levels of immunomodulatory cell surface molecules that prime them for recognition by cytotoxic lymphocytes, such as NKG2D receptor ligands that have been shown to be required for NK cell killing of senescent cells [258], and MHC-I and B2M [30, 51, 259] required for antigen presentation to CD8+ T cells. However, senescent cells also upregulate immune checkpoint proteins, such as PD-L1 and HLA-E, that block effective NK and T cell surveillance necessary for tumor eradication. Use of FDA-approved ICB immunotherapies targeting the PD-1/PD-L1 axis has been one strategy to boost the anti-tumor activity of senescence-inducing agents [260]. For example, we and others have shown that treatment with CDK4/6 inhibitors that induce senescence alone or in combination with other targeted therapies activate the inhibitory checkpoint PD-L1, and as such synergize with anti-PD-1/PD-L1 blockade to promote CD8+ T cell-mediated tumor clearance in melanoma, breast, lung, pancreatic, and colon cancers [30, 124–126, 261]. In addition, senescent tumor cells that evade NK cell immunity through shedding of NKG2D ligands [57] or upregulation of HLA-E molecules that inhibit NK cell activation through the receptor NKG2A [56] can be targeted with antibodies that are currently in clinical development to block the NKG2A checkpoint [262] or the shedding of the NKG2D ligands MICA/B [263].
Senescent cells also express unique cell surface proteins that can be used to design and engineer antibody and chimeric antigen receptor (CAR) approaches to immunologically target senescent cells following cancer therapy (Fig. 1). Elucidation of the surfaceome in senescent cells has identified potential targets for these approaches, such as urokinase-type plasminogen activator receptor (uPAR) [264], NOTCH1 [161], DPP4 [265], and membrane-bound Vimentin [266]. One strategy has been to design antibodies targeting these molecules (e.g. DPP4) to promote antibody-dependent cell-mediated cytotoxicity (ADCC) through innate immune cells such as macrophages, neutrophils, and NK cells that express Fc receptors that can bind to the constant region of these antibodies and kill antibody-positive senescent cells [265]. More recently, Amor and colleagues engineered CAR-T cells to target uPAR expressed on senescent cells, which they demonstrated could effectively eliminate senescent liver cells following fibrosis and lung tumor cells following TIS [264]. These methods highlight innovative ways to more specifically and effectively engineer the immune system to target senescent cells following cancer therapy.
As the SASP can also promote the infiltration and immune suppressive characteristics of MDSCs and other myeloid cells known to inhibit NK and T cell functionality, targeting these populations and the signals that attract them is another approach to further augment anti-tumor immunity following TIS. Antibodies can be utilized to neutralize immune suppressive SASP factors such as CSF-1, IL-6, PGE2, IL-1β, and TGFβ that are known to attract and activate MDSCs [267]. Orthogonally, clinically developed CSF-1R, CCR2, and CXCR2 inhibitors, the latter of which have been shown to inhibit MDSC infiltration following chemotherapy-induced senescence in prostate cancer models [268], can be utilized to block recruitment and/or reeducate suppressive myeloid cell populations that infiltrate following senescence induction [269, 270]. In all, these examples underscore that by understanding how the SASP impacts immune surveillance in cancer, it is possible to take advantage of immunomodulatory effects of the SASP to create rational combinations of senescence-inducing and immunotherapies to further enhance anti-tumor responses.
6. Future Directions and concluding remarks
6.1. Defining better markers of cellular senescence
Recent advances in genetic engineering and in vivo model systems allowing manipulation of senescence in tumor contexts with intact TMEs has shed light on the broad role of senescence in regulating tumor-immune microenvironments in preclinical settings. Still, there is a great degree of heterogeneity and context dependency that dictates the impact of senescence on immune and tumor responses in cancer, including but not limited to: (a) the senescence inducer (and its dose), (b) duration and time after insult, (c) the cell type it acts on, (d) genetic and epigenetic alterations found within a tumor, and (e) the composition of the surrounding TME. A clear limitation to developing a consensus framework on the role of senescence in cancer biology and tumor immunology has been the lack of specific and reproducible markers of cellular senescence, especially in vivo, to differentiation senescent cells from other cell states associated with cell cycle withdrawal, including quiescence, post-mitotic terminal differentiation, and dormancy [10, 16, 271]. While SA-β-gal, p16, and p21 have been used widely as senescence markers, these factors are not universal [179, 182, 183, 185, 272, 273] and can be present in other cell types, particularly macrophages [224, 274]. Certain types of senescence do not even produce a SASP [275, 276]. Comparison of transcriptional and proteomic signatures across different cell types following senescence induction has begun to pinpoint common genes and pathways associated with senescence [179, 182, 183, 272]. It will be critical to link these senescence genes with their transcriptional regulators and chromatin changes across the genome to comprehensively map senescence transcriptional regulatory networks. Multi-parametric approaches and machine learning tools currently being implemented should allow systematic discovery of not only core senescence genes but also markers and regulators of different senescence subtypes or SASP programs [182, 277]. Ongoing single cell and spatial transcriptomic efforts [278] will supply further granularity within the context of intratumoral heterogeneity to better separate SASP-secreting senescent cells from other tumor and stromal cell populations within tissues. Importantly, such analysis will help us to differentiate “good” senescence populations that contribute to immune stimulation and tumor constraint from “bad” senescent cells that promote immune suppression and disease relapse following therapy.
6.2. Characterizing immune-stimulatory senescence-inducing therapies and SASP regulators
It will be critical to define the SASP factors and regulators necessary for immune-mediated tumor control systematically across cancer settings. This will require detailed analysis of both the SASP output and immune responses in human cancer patients and physiologically intact murine tumor models following TIS, and manipulation of specific SASP components to understand how they functionally modulate tumor-immune microenvironments (Fig. 2). As a first step, we must identify cancer therapies that can robustly induce senescence and anti-tumor immunity. Several groups have developed screening approaches using single senescence markers or gene reporters to identify pro-senescence therapies [128, 203, 279]; however, such systems do not readout the SASP and cannot predict the impact of candidate target inhibition on immune responses and remodeling of the TME. Creative genetic and pharmacological screening platforms to (a) uncover new targets and drugs that can induce multiple senescence and SASP markers specifically in genetically-defined tumor cells in vitro and (b) rapidly evaluate their impact on tumor-immune interactions in immunocompetent mouse models with genetically engineered tumors [280] or in 3D co-culture systems that incorporate TME stromal components [281, 282] will be necessary to characterize those senescence-inducing therapies that produce robust anti-tumor immunity and mediate tumor control (Fig. 2). Use of patient-derived xenografts (PDX) in mice with a humanized immune system or patient-derived explants (PDEs) will be also be important to confirm findings made in murine systems in the human setting [283, 284]. Functional experiments to block, modulate, or induce specific SASP regulators following therapy-induced senescence are then required to identify candidate SASP factors that are essential for anti-tumor immunity across cancer types and could serve as potential biomarkers of immune and tumor responses following cancer therapy in the clinic (Fig. 2). Importantly, as the majority of work on senescence-mediated TME remodeling has focused on a handful of immune cells, most prominently NK cells, macrophages, MDSCs, and T cells, utilization of more high-throughput and unbiased technologies will help us understand how other immune (B cells, γδ T cells, granulocytes, innate lymphoid cells (ILCs)) and stromal (endothelial cells, pericytes, fibroblasts, nerves, microbiota) populations are impacted by and respond to SASP cues and contribute to tumor control.
Fig. 2. Pipeline to identify immune-stimulatory senescence-inducing cancer therapies and SASP biomarkers for the clinic.
(1) In vitro genetic and pharmacological screens can be performed on genetically-defined cancer cell lines engineered with fluorescent senescence and SASP reporters to identify new senescence-inducing therapies for defined cancer contexts. These candidate senescence-inducing therapies can then be tested in immunocompetent mouse models harboring transplanted or autochthonous tumors to evaluate their anti-tumor efficacy. (2) The impact of therapy-induced senescence on tumor-immune interactions can be assessed using in vitro co-culture systems and in vivo tumor mouse models with intact TMEs. (3) SASP factors and regulators that functionally impact tumor-immune interactions can then be identified by inducing or suppressing their expression genetically or pharmacologically through the pipeline in (1). (4) The immunomodulatory effects of senescence-inducing therapies and SASP biomarkers of immune and tumor responses can be validated in tissue and liquid biopsies from cancer patients treated in the clinic. mAb, monoclonal antibodies; TME, tumor microenvironment.
This pipeline will unveil new senescence-inducing therapies for different tumor and tumor genetic contexts, as well as elucidate the mechanisms by which the SASP can remodel tumor-immune microenvironments for tumor control. Still, a caveat to studying the immune modulatory mechanisms of TIS or pursuing it as a therapeutic strategy for cancer treatment is that non-tumor cells, in particular stromal fibroblasts, can also undergo senescence following treatment and may impact immune responses and tumor outcomes in negative ways [186]. Thus, innovative approaches are still needed to design better therapies that specifically induce senescence only in tumor cells, or to engineer methods to deliver payloads of pro-senescence molecules specifically to the tumor. As senescence-inducing therapies alone are unlikely to mediate complete tumor remission, understanding the new dependencies that arise through senescence-mediating remodeling of the tumor-immune landscape will allow for development of rational combinatorial therapies with senolytic and senomorphic agents and immunotherapies to fully harness the capabilities of senescence for cancer therapy.
6.3. Translating senescence to cancer therapy in the clinic
Finally, how do we apply what we have learned from preclinical models to inform the clinical application of TIS (Fig. 2)? Given the lack of generalizable senescence markers and difficulty clearly discerning senescence phenotypes in patient samples, our knowledge on the frequency and impact of senescence responses following chemo-, radio-, and targeted therapies in the clinic is limited. Still, some studies have begun to assess senescence biomarkers is tissue biopsies and even circulating T cells and correlate expression with chemotherapy outcomes and toxicities [69, 285]. As bulk and even single cell transcriptomics are becoming more commonplace with longitudinal tracking of tumor responses to therapy [286], transcriptional profiles from patient tumors may be the most straightforward path to defining TIS [58, 287]. In addition, other studies have begun to look at blood biomarkers (i.e. liquid biopsies) as a readout of senescence in animal models and human patients [72, 179, 264, 288, 289]. A number of senoprobes are now being tested for in vivo detection of senescent cells [219, 290], including a β-galactosidase PET probe for radiographical imaging that is in human trials in liver cancer patients [291]. Though promising, these approaches rely on use of one phenotype associated with senescence and cannot indicate which cell types are senescent and secreting SASP.
Correlating SASP factor expression with (a) tumor responses and progression-free and overall survival and (b) immune responses locally (by biopsy) and peripherally in the blood has yet to be achieved clinically. Such analysis will be paramount to understanding which aspects of senescence and SASP are necessary for different immune effects, and whether senescence is associated with a positive or negative impact on therapeutic efficacy (Fig. 2). Indeed, each senescence-inducing therapy (and its dose) in a given cancer context may have a different effect on the SASP, immune surveillance, and tumor responses, and broad cross-comparison between treatments and cancer types will elucidate key SASP components that contribute to “good” senescence mediating tumor control and “bad” senescence promoting tumor relapse. While informative and hypothesis-driving, it is important to note that correlative analysis of independent markers of senescent, tumor, and immune cells does not prove cause and effect but rather an association that can further be followed up and substantiated at the bench. As reproducible biomarkers of senescence and SASP and the processes that govern their effects on tumor-immune microenvironments are unveiled, we envision that cancer therapies that induce senescence will be more readily applied in different settings to promote tumor control, create new therapeutic vulnerabilities and rational combinatorial therapy regimens, and be used to potentiate immunotherapy to fully harness the immune system to fight cancer.
Acknowledgements
We thank members of the Ruscetti laboratory (Katherine Murphy) and our colleagues Eric Baehrecke, Dohoon Kim, and Wen Xue for their helpful advice and feedback on the manuscript. This work was supported by a R00 CA241110-02 grant from the National Cancer Institute (NCI). Figures were created with BioRender.com.
Funding Source
All sources of funding should also be acknowledged and you should declare any involvement of study sponsors in the study design; collection, analysis and interpretation of data; the writing of the manuscript; the decision to submit the manuscript for publication. If the study sponsors had no such involvement, this should be stated.
ABBREVIATIONS
- 3C
chromosome conformation capture
- 5-FU
5- fluorouracil
- ADCC
antibody-dependent cellular cytotoxicity
- AREG
amphiregulin
- ATM
ataxia telangiectasia mutated
- ATR
ataxia telangiectasia and rad3-related
- ATRX
alpha thalassemia/mental retardation syndrome X-linked
- AURKA
Aurora kinase A
- AURKB
Aurora kinase B
- B2M
Beta-2 microglobulin
- BCL-2
B-cell lymphoma 2
- BET
Bromodomain and extra terminal
- BRD4
bromo and extra terminal domain protein 4
- C/EBP-β
CCAAT/enhancer binding protein beta
- CAR
chimeric antigen receptor
- CCL
C-C motif chemokine ligand
- CCR
C-C motif chemokine receptor
- CDC7
cell division cycle 7
- CDK
cyclin-dependent kinase
- cGAS
cyclic GMP-AMP synthase
- CHK1
checkpoint kinase 1
- CHK2
checkpoint kinase 2
- CIP
CDK interacting protein
- COX-2
cyclooxygenase-2
- CSF-1
colony-stimulating factor 1
- CTLA-4
cytotoxic T-lymphocyte-associated protein 4
- CXCL
C-X-C motif chemokine ligand
- CXCR
C-X-C motif chemokine receptor
- D + Q
Dasatinib and Quercetin
- DC
dendritic cell
- DDR
DNA damage response
- DNAM-1
DNAX accessory molecule-1
- DOT1L
disruptor of telomeric silencing 1-like
- DPP4
dipeptidyl peptidase 4
- DSB
double-strand break
- ECM
extracellular matrix
- ER
estrogen receptor
- EV
extracellular vesicle
- EZH2
enhancer of zeste homolog 2
- Fc
fragment crystallizable
- FDA
U.S. Food and Drug Administration
- FGF
fibroblast growth factor
- FOXO4
forkhead box O4
- GATA4
GATA binding protein 4
- GEMM
genetically engineered mouse model
- GLP
G9a-like protein
- GM-CSF
granulocyte-macrophage colony-stimulating factor
- HCC
hepatocellular carcinoma
- HDAC
histone deacetylase
- Hi-C
high-throughput chromosome conformation capture
- HLA-E
major histocompatibility antigen, Class I, E
- HMGB2
high mobility group box 2
- ICAM-1
intercellular adhesion molecule-1
- ICB
immune checkpoint blockade
- IFN
interferon
- IL
interleukin
- ILC
innate lymphoid cells
- IPF
idiopathic pulmonary fibrosis
- IR
ionizing radiation
- IRF
interferon regulatory factor
- JAK2
janus kinase 2
- KIP
kinase inhibitory protein
- L1
LINE-1
- MDM2
murine double minute 2
- MDSC
Myeloid-derived suppressor cell
- MEK
mitogen-activated protein kinase (MAPK) kinase
- MHC
major histocompatibility complex
- MICA
MHC class I polypeptide–related sequence A
- MICB
MHC class I polypeptide–related sequence B
- miRNA
microRNA
- MK2
mitogen-activated protein kinase (MAPK)-activated protein kinase
- MMP
matrix metalloproteinase
- mTOR
mammalian target of rapamycin
- MULT-1
murine UL16 binding protein-like transcript-1
- NF-κB
nuclear factor kappa B
- NK cells
Natural Killer cells
- NKG2A
NK group 2 member A
- NKG2D
natural killer group 2 member D
- NKT cell
Natural Killer T cell
- NLRP3
NLR family pyrin domain containing 3
- p38MAPK
p38 mitogen-activated protein kinase
- PAI-1
plasminogen activator inhibitor-1
- PARP
Poly (ADP-ribose) polymerase
- PASP
p21-activated secretory phenotype
- PET
positron emission tomography
- PD-1
programmed cell death protein-1
- PD-L1
programmed death ligand-1
- PDE
patient-derived explant
- PDX
patient-derived xenograft
- PGE2
prostaglandin E2
- PI3K
phosphoinositide 3-kinase
- PRC2
polycomb repressive complex 2
- PTBP1
polypyrimidine tract binding protein 1
- PTEN
phosphatase and tensin homolog
- RAE-1
retinoic acid early inducible gene 1
- RB
retinoblastoma
- RTE
retrotransposable element
- SA-β-gal
senescence-associated beta-galactosidase
- SAHF
senescence-associated heterochromatin foci
- SASP
senescence-associated secretory phenotype
- SIRT1
Sirtuin 1
- STAT
signal transducer and activator of transcription
- STING
stimulator of interferon genes
- SUV39h
suppressor of variegation 3–9 homolog
- TCR
T cell receptor
- TF
transcription factor
- TGFβ
transforming growth factor beta
- TIMP1
TIMP metallopeptidase inhibitor 1
- TIS
therapy-induced senescence
- TLR2
Toll-like receptor 2
- TME
tumor microenvironment
- TNFα
tumor necrosis factor alpha
- Treg
regulatory T cell
- ULBP1
UL16 binding protein 1
- uPAR
urokinase-type plasminogen activator receptor
- VCAM-1
vascular cell adhesion molecule-1
- VEGF
vascular endothelial growth factor
- VLA-4
very late antigen-4
- γδ T
gamma-delta T cell
Footnotes
Conflict of Interest Statement
The authors declare that there are no conflicts of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Galluzzi L, Zitvogel L, Kroemer G, Immunological Mechanisms Underneath the Efficacy of Cancer Therapy, Cancer Immunol Res 4(11) (2016) 895–902. [DOI] [PubMed] [Google Scholar]
- [2].Sherr CJ, Bartek J, Cell Cycle–Targeted Cancer Therapies, Annual Review of Cancer Biology 1(1) (2017) 41–57. [Google Scholar]
- [3].Rixe O, Fojo T, Is Cell Death a Critical End Point for Anticancer Therapies or Is Cytostasis Sufficient?, Clinical Cancer Research 13(24) (2007) 7280–7287. [DOI] [PubMed] [Google Scholar]
- [4].Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Annicchiarico-Petruzzelli M, Antonov AV, Arama E, Baehrecke EH, Barlev NA, Bazan NG, Bernassola F, Bertrand MJM, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Boya P, Brenner C, Campanella M, Candi E, Carmona-Gutierrez D, Cecconi F, Chan FK, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Cohen GM, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D’Angiolella V, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, DeBerardinis RJ, Deshmukh M, Di Daniele N, Di Virgilio F, Dixit VM, Dixon SJ, Duckett CS, Dynlacht BD, El-Deiry WS, Elrod JW, Fimia GM, Fulda S, Garcia-Saez AJ, Garg AD, Garrido C, Gavathiotis E, Golstein P, Gottlieb E, Green DR, Greene LA, Gronemeyer H, Gross A, Hajnoczky G, Hardwick JM, Harris IS, Hengartner MO, Hetz C, Ichijo H, Jaattela M, Joseph B, Jost PJ, Juin PP, Kaiser WJ, Karin M, Kaufmann T, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Knight RA, Kumar S, Lee SW, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, Lopez-Otin C, Lowe SW, Luedde T, Lugli E, MacFarlane M, Madeo F, Malewicz M, Malorni W, Manic G, Marine JC, Martin SJ, Martinou JC, Medema JP, Mehlen P, Meier P, Melino S, Miao EA, Molkentin JD, Moll UM, Munoz-Pinedo C, Nagata S, Nunez G, Oberst A, Oren M, Overholtzer M, Pagano M, Panaretakis T, Pasparakis M, Penninger JM, Pereira DM, Pervaiz S, Peter ME, Piacentini M, Pinton P, Prehn JHM, Puthalakath H, Rabinovich GA, Rehm M, Rizzuto R, Rodrigues CMP, Rubinsztein DC, Rudel T, Ryan KM, Sayan E, Scorrano L, Shao F, Shi Y, Silke J, Simon HU, Sistigu A, Stockwell BR, Strasser A, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Thorburn A, Tsujimoto Y, Turk B, Vanden Berghe T, Vandenabeele P, Vander Heiden MG, Villunger A, Virgin HW, Vousden KH, Vucic D, Wagner EF, Walczak H, Wallach D, Wang Y, Wells JA, Wood W, Yuan J, Zakeri Z, Zhivotovsky B, Zitvogel L, Melino G, Kroemer G, Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018, Cell Death Differ 25(3) (2018) 486–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Anderson NM, Simon MC, The tumor microenvironment, Curr Biol 30(16) (2020) R921–R925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Kaiser J, Couzin-Frankel J, Cancer immunotherapy sweeps Nobel for medicine, Science 362(6410) (2018) 13. [DOI] [PubMed] [Google Scholar]
- [7].Sharma P, Siddiqui BA, Anandhan S, Yadav SS, Subudhi SK, Gao J, Goswami S, Allison JP, The Next Decade of Immune Checkpoint Therapy, Cancer Discov 11(4) (2021) 838–857. [DOI] [PubMed] [Google Scholar]
- [8].Hayflick L, The Limited in Vitro Lifetime of Human Diploid Cell Strains, Exp Cell Res 37 (1965) 614–36. [DOI] [PubMed] [Google Scholar]
- [9].Hayflick L, Moorhead PS, The serial cultivation of human diploid cell strains, Exp Cell Res 25 (1961) 585–621. [DOI] [PubMed] [Google Scholar]
- [10].Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, Campisi J, Collado M, Evangelou K, Ferbeyre G, Gil J, Hara E, Krizhanovsky V, Jurk D, Maier AB, Narita M, Niedernhofer L, Passos JF, Robbins PD, Schmitt CA, Sedivy J, Vougas K, von Zglinicki T, Zhou D, Serrano M, Demaria M, Cellular Senescence: Defining a Path Forward, Cell 179(4) (2019) 813–827. [DOI] [PubMed] [Google Scholar]
- [11].Kuilman T, Michaloglou C, Mooi WJ, Peeper DS, The essence of senescence, Genes Dev 24(22) (2010) 2463–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Ewald JA, Desotelle JA, Wilding G, Jarrard DF, Therapy-induced senescence in cancer, J Natl Cancer Inst 102(20) (2010) 1536–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Munoz-Espin D, Serrano M, Cellular senescence: from physiology to pathology, Nat Rev Mol Cell Biol 15(7) (2014) 482–96. [DOI] [PubMed] [Google Scholar]
- [14].Rhinn M, Ritschka B, Keyes WM, Cellular senescence in development, regeneration and disease, Development 146(20) (2019). [DOI] [PubMed] [Google Scholar]
- [15].Prieto LI, Graves SI, Baker DJ, Insights from In Vivo Studies of Cellular Senescence, Cells 9(4) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Sharpless NE, Sherr CJ, Forging a signature of in vivo senescence, Nat Rev Cancer 15(7) (2015) 397–408. [DOI] [PubMed] [Google Scholar]
- [17].Wang B, Kohli J, Demaria M, Senescent Cells in Cancer Therapy: Friends or Foes?, Trends Cancer 6(10) (2020) 838–857. [DOI] [PubMed] [Google Scholar]
- [18].Collado M, Serrano M, Senescence in tumours: evidence from mice and humans, Nat Rev Cancer 10(1) (2010) 51–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Hernandez-Segura A, Nehme J, Demaria M, Hallmarks of Cellular Senescence, Trends Cell Biol 28(6) (2018) 436–453. [DOI] [PubMed] [Google Scholar]
- [20].Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, Nelson PS, Desprez PY, Campisi J, Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor, PLoS Biol 6(12) (2008) 2853–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, Aarden LA, Mooi WJ, Peeper DS, Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network, Cell 133(6) (2008) 1019–31. [DOI] [PubMed] [Google Scholar]
- [22].Faget DV, Ren Q, Stewart SA, Unmasking senescence: context-dependent effects of SASP in cancer, Nat Rev Cancer 19(8) (2019) 439–453. [DOI] [PubMed] [Google Scholar]
- [23].Prata L, Ovsyannikova IG, Tchkonia T, Kirkland JL, Senescent cell clearance by the immune system: Emerging therapeutic opportunities, Semin Immunol 40 (2018) 101275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Kale A, Sharma A, Stolzing A, Desprez PY, Campisi J, Role of immune cells in the removal of deleterious senescent cells, Immun Ageing 17 (2020) 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Rao SG, Jackson JG, SASP: Tumor Suppressor or Promoter? Yes!, Trends Cancer 2(11) (2016) 676–687. [DOI] [PubMed] [Google Scholar]
- [26].Schosserer M, Grillari J, Breitenbach M, The Dual Role of Cellular Senescence in Developing Tumors and Their Response to Cancer Therapy, Front Oncol 7 (2017) 278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Braig M, Schmitt CA, Oncogene-induced senescence: putting the brakes on tumor development, Cancer Res 66(6) (2006) 2881–4. [DOI] [PubMed] [Google Scholar]
- [28].Kang TW, Yevsa T, Woller N, Hoenicke L, Wuestefeld T, Dauch D, Hohmeyer A, Gereke M, Rudalska R, Potapova A, Iken M, Vucur M, Weiss S, Heikenwalder M, Khan S, Gil J, Bruder D, Manns M, Schirmacher P, Tacke F, Ott M, Luedde T, Longerich T, Kubicka S, Zender L, Senescence surveillance of pre-malignant hepatocytes limits liver cancer development, Nature 479(7374) (2011) 547–51. [DOI] [PubMed] [Google Scholar]
- [29].Ruscetti M, Leibold J, Bott MJ, Fennell M, Kulick A, Salgado NR, Chen CC, Ho YJ, Sanchez-Rivera FJ, Feucht J, Baslan T, Tian S, Chen HA, Romesser PB, Poirier JT, Rudin CM, de Stanchina E, Manchado E, Sherr CJ, Lowe SW, NK cell-mediated cytotoxicity contributes to tumor control by a cytostatic drug combination, Science 362(6421) (2018) 1416–1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Ruscetti M, Morris J.P.t., Mezzadra R, Russell J, Leibold J, Romesser PB, Simon J, Kulick A, Ho YJ, Fennell M, Li J, Norgard RJ, Wilkinson JE, Alonso-Curbelo D, Sridharan R, Heller DA, de Stanchina E, Stanger BZ, Sherr CJ, Lowe SW, Senescence-Induced Vascular Remodeling Creates Therapeutic Vulnerabilities in Pancreas Cancer, Cell 181(2) (2020) 424–441 e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Coppe JP, Desprez PY, Krtolica A, Campisi J, The senescence-associated secretory phenotype: the dark side of tumor suppression, Annu Rev Pathol 5 (2010) 99–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].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(2) (2017) 165–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Gonzalez-Meljem JM, Apps JR, Fraser HC, Martinez-Barbera JP, Paracrine roles of cellular senescence in promoting tumourigenesis, Br J Cancer 118(10) (2018) 1283–1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Milanovic M, Fan DNY, Belenki D, Dabritz JHM, Zhao Z, Yu Y, Dorr JR, Dimitrova L, Lenze D, Monteiro Barbosa IA, Mendoza-Parra MA, Kanashova T, Metzner M, Pardon K, Reimann M, Trumpp A, Dorken B, Zuber J, Gronemeyer H, Hummel M, Dittmar G, Lee S, Schmitt CA, Senescence-associated reprogramming promotes cancer stemness, Nature 553(7686) (2018) 96–100. [DOI] [PubMed] [Google Scholar]
- [35].Triana-Martinez F, Loza MI, Dominguez E, Beyond Tumor Suppression: Senescence in Cancer Stemness and Tumor Dormancy, Cells 9(2) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Ritschka B, Storer M, Mas A, Heinzmann F, Ortells MC, Morton JP, Sansom OJ, Zender L, Keyes WM, The senescence-associated secretory phenotype induces cellular plasticity and tissue regeneration, Genes Dev 31(2) (2017) 172–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Sikora E, Czarnecka-Herok J, Bojko A, Sunderland P, Therapy-induced polyploidization and senescence: Coincidence or interconnection?, Semin Cancer Biol (2020). [DOI] [PubMed] [Google Scholar]
- [38].Michaloglou C, Vredeveld LC, Soengas MS, Denoyelle C, Kuilman T, van der Horst CM, Majoor DM, Shay JW, Mooi WJ, Peeper DS, BRAFE600-associated senescence-like cell cycle arrest of human naevi, Nature 436(7051) (2005) 720–4. [DOI] [PubMed] [Google Scholar]
- [39].Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizhanovsky V, Cordon-Cardo C, Lowe SW, Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas, Nature 445(7128) (2007) 656–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Sturmlechner I, Zhang C, Sine CC, Deursen E.-J.v., Jeganathan KB, Hamada N, Grasic J, Friedman D, Stutchman JT, Can I, Hamada M, Lim DY, Lee J-H, Ordog T, Laberge R-M, Shapiro V, Baker DJ, Li H, Deursen J.M.v., p21 produces a bioactive secretome that places stressed cells under immunosurveillance, Science 374(6567) (2021) eabb3420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Ruhland MK, Loza AJ, Capietto AH, Luo X, Knolhoff BL, Flanagan KC, Belt BA, Alspach E, Leahy K, Luo J, Schaffer A, Edwards JR, Longmore G, Faccio R, DeNardo DG, Stewart SA, Stromal senescence establishes an immunosuppressive microenvironment that drives tumorigenesis, Nat Commun 7 (2016) 11762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Chen Z, Trotman LC, Shaffer D, Lin HK, Dotan ZA, Niki M, Koutcher JA, Scher HI, Ludwig T, Gerald W, Cordon-Cardo C, Pandolfi PP, Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis, Nature 436(7051) (2005) 725–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Schreiber RD, Old LJ, Smyth MJ, Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion, Science 331(6024) (2011) 1565–70. [DOI] [PubMed] [Google Scholar]
- [44].Gonzalez H, Hagerling C, Werb Z, Roles of the immune system in cancer: from tumor initiation to metastatic progression, Genes Dev 32(19–20) (2018) 1267–1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Demaria O, Cornen S, Daeron M, Morel Y, Medzhitov R, Vivier E, Harnessing innate immunity in cancer therapy, Nature 574(7776) (2019) 45–56. [DOI] [PubMed] [Google Scholar]
- [46].Chen S, Lai SWT, Brown CE, Feng M, Harnessing and Enhancing Macrophage Phagocytosis for Cancer Therapy, Front Immunol 12 (2021) 635173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Shimasaki N, Jain A, Campana D, NK cells for cancer immunotherapy, Nat Rev Drug Discov 19(3) (2020) 200–218. [DOI] [PubMed] [Google Scholar]
- [48].Chen DS, Mellman I, Oncology meets immunology: the cancer-immunity cycle, Immunity 39(1) (2013) 1–10. [DOI] [PubMed] [Google Scholar]
- [49].Ozga AJ, Chow MT, Luster AD, Chemokines and the immune response to cancer, Immunity 54(5) (2021) 859–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Briukhovetska D, Dorr J, Endres S, Libby P, Dinarello CA, Kobold S, Interleukins in cancer: from biology to therapy, Nat Rev Cancer 21(8) (2021) 481–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Petti C, Molla A, Vegetti C, Ferrone S, Anichini A, Sensi M, Coexpression of NRASQ61R and BRAFV600E in human melanoma cells activates senescence and increases susceptibility to cell-mediated cytotoxicity, Cancer Res 66(13) (2006) 6503–11. [DOI] [PubMed] [Google Scholar]
- [52].van Tuyn J, Jaber-Hijazi F, MacKenzie D, Cole JJ, Mann E, Pawlikowski JS, Rai TS, Nelson DM, McBryan T, Ivanov A, Blyth K, Wu H, Milling S, Adams PD, Oncogene-Expressing Senescent Melanocytes Up-Regulate MHC Class II, a Candidate Melanoma Suppressor Function, J Invest Dermatol 137(10) (2017) 2197–2207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Iannello A, Thompson TW, Ardolino M, Lowe SW, Raulet DH, p53-dependent chemokine production by senescent tumor cells supports NKG2D-dependent tumor elimination by natural killer cells, J Exp Med 210(10) (2013) 2057–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Lujambio A, Akkari L, Simon J, Grace D, Tschaharganeh DF, Bolden JE, Zhao Z, Thapar V, Joyce JA, Krizhanovsky V, Lowe SW, Non-cell-autonomous tumor suppression by p53, Cell 153(2) (2013) 449–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Shalapour S, Karin M, Immunity, inflammation, and cancer: an eternal fight between good and evil, J Clin Invest 125(9) (2015) 3347–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Pereira BI, Devine OP, Vukmanovic-Stejic M, Chambers ES, Subramanian P, Patel N, Virasami A, Sebire NJ, Kinsler V, Valdovinos A, LeSaux CJ, Passos JF, Antoniou A, Rustin MHA, Campisi J, Akbar AN, Senescent cells evade immune clearance via HLA-E-mediated NK and CD8(+) T cell inhibition, Nat Commun 10(1) (2019) 2387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Munoz DP, Yannone SM, Daemen A, Sun Y, Vakar-Lopez F, Kawahara M, Freund AM, Rodier F, Wu JD, Desprez PY, Raulet DH, Nelson PS, van ‘t Veer LJ, Campisi J, Coppe JP, Targetable mechanisms driving immunoevasion of persistent senescent cells link chemotherapy-resistant cancer to aging, JCI Insight 5 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Eggert T, Wolter K, Ji J, Ma C, Yevsa T, Klotz S, Medina-Echeverz J, Longerich T, Forgues M, Reisinger F, Heikenwalder M, Wang XW, Zender L, Greten TF, Distinct Functions of Senescence-Associated Immune Responses in Liver Tumor Surveillance and Tumor Progression, Cancer Cell 30(4) (2016) 533–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Goncalves S, Yin K, Ito Y, Chan A, Olan I, Gough S, Cassidy L, Serrao E, Smith S, Young A, Narita M, Hoare M, COX2 regulates senescence secretome composition and senescence surveillance through PGE2, Cell Rep 34(11) (2021) 108860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Mazzoni M, Mauro G, Erreni M, Romeo P, Minna E, Vizioli MG, Belgiovine C, Rizzetti MG, Pagliardini S, Avigni R, Anania MC, Allavena P, Borrello MG, Greco A, Senescent thyrocytes and thyroid tumor cells induce M2-like macrophage polarization of human monocytes via a PGE2-dependent mechanism, J Exp Clin Cancer Res 38(1) (2019) 208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Toso A, Revandkar A, Di Mitri D, Guccini I, Proietti M, Sarti M, Pinton S, Zhang J, Kalathur M, Civenni G, Jarrossay D, Montani E, Marini C, Garcia-Escudero R, Scanziani E, Grassi F, Pandolfi PP, Catapano CV, Alimonti A, Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity, Cell Rep 9(1) (2014) 75–89. [DOI] [PubMed] [Google Scholar]
- [62].Garcia AJ, Ruscetti M, Arenzana TL, Tran LM, Bianci-Frias D, Sybert E, Priceman SJ, Wu L, Nelson PS, Smale ST, Wu H, Pten null prostate epithelium promotes localized myeloid-derived suppressor cell expansion and immune suppression during tumor initiation and progression, Mol Cell Biol 34(11) (2014) 2017–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Krtolica A, Parrinello S, Lockett S, Desprez PY, Campisi J, Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging, Proc Natl Acad Sci U S A 98(21) (2001) 12072–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Coppe JP, Kauser K, Campisi J, Beausejour CM, Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence, J Biol Chem 281(40) (2006) 29568–74. [DOI] [PubMed] [Google Scholar]
- [65].Kim YH, Choi YW, Lee J, Soh EY, Kim JH, Park TJ, Senescent tumor cells lead the collective invasion in thyroid cancer, Nat Commun 8 (2017) 15208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Hunt BJ, Jurd KM, Endothelial cell activation. A central pathophysiological process, BMJ 316(7141) (1998) 1328–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Saleh T, Bloukh S, Carpenter VJ, Alwohoush E, Bakeer J, Darwish S, Azab B, Gewirtz DA, Therapy-Induced Senescence: An “Old” Friend Becomes the Enemy, Cancers (Basel) 12(4) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Childs BG, Baker DJ, Kirkland JL, Campisi J, van Deursen JM, Senescence and apoptosis: dueling or complementary cell fates?, EMBO Rep 15(11) (2014) 1139–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Sanoff HK, Deal AM, Krishnamurthy J, Torrice C, Dillon P, Sorrentino J, Ibrahim JG, Jolly TA, Williams G, Carey LA, Drobish A, Gordon BB, Alston S, Hurria A, Kleinhans K, Rudolph KL, Sharpless NE, Muss HB, Effect of cytotoxic chemotherapy on markers of molecular age in patients with breast cancer, J Natl Cancer Inst 106(4) (2014) dju057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].te Poele RH, Okorokov AL, Jardine L, Cummings J, Joel SP, DNA damage is able to induce senescence in tumor cells in vitro and in vivo, Cancer research 62(6) (2002) 1876–83. [PubMed] [Google Scholar]
- [71].Roberson RS, Kussick SJ, Vallieres E, Chen SY, Wu DY, Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers, Cancer research 65(7) (2005) 2795–803. [DOI] [PubMed] [Google Scholar]
- [72].Xu Q, Long Q, Zhu D, Fu D, Zhang B, Han L, Qian M, Guo J, Xu J, Cao L, Chin YE, Coppe JP, Lam EW, Campisi J, Sun Y, Targeting amphiregulin (AREG) derived from senescent stromal cells diminishes cancer resistance and averts programmed cell death 1 ligand (PD-L1)-mediated immunosuppression, Aging Cell 18(6) (2019) e13027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Schmitt CA, Fridman JS, Yang M, Lee S, Baranov E, Hoffman RM, Lowe SW, A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy, Cell 109(3) (2002) 335–46. [DOI] [PubMed] [Google Scholar]
- [74].Chien Y, Scuoppo C, Wang X, Fang X, Balgley B, Bolden JE, Premsrirut P, Luo W, Chicas A, Lee CS, Kogan SC, Lowe SW, Control of the senescence-associated secretory phenotype by NF-kappaB promotes senescence and enhances chemosensitivity, Genes Dev 25(20) (2011) 2125–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Antonangeli F, Soriani A, Ricci B, Ponzetta A, Benigni G, Morrone S, Bernardini G, Santoni A, Natural killer cell recognition of in vivo drug-induced senescent multiple myeloma cells, Oncoimmunology 5(10) (2016) e1218105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Soriani A, Iannitto ML, Ricci B, Fionda C, Malgarini G, Morrone S, Peruzzi G, Ricciardi MR, Petrucci MT, Cippitelli M, Santoni A, Reactive oxygen species- and DNA damage response-dependent NK cell activating ligand upregulation occurs at transcriptional levels and requires the transcriptional factor E2F1, J Immunol 193(2) (2014) 950–60. [DOI] [PubMed] [Google Scholar]
- [77].Soriani A, Zingoni A, Cerboni C, Iannitto ML, Ricciardi MR, Di Gialleonardo V, Cippitelli M, Fionda C, Petrucci MT, Guarini A, Foa R, Santoni A, ATM-ATR-dependent upregulation of DNAM-1 and NKG2D ligands on multiple myeloma cells by therapeutic agents results in enhanced NK-cell susceptibility and is associated with a senescent phenotype, Blood 113(15) (2009) 3503–11. [DOI] [PubMed] [Google Scholar]
- [78].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(1) (2021) 102016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Bent EH, Gilbert LA, Hemann MT, A senescence secretory switch mediated by PI3K/AKT/mTOR activation controls chemoprotective endothelial secretory responses, Genes Dev 30(16) (2016) 1811–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Murali B, Ren Q, Luo X, Faget DV, Wang C, Johnson RM, Gruosso T, Flanagan KC, Fu Y, Leahy K, Alspach E, Su X, Ross MH, Burnette B, Weilbaecher KN, Park M, Mbalaviele G, Monahan JB, Stewart SA, Inhibition of the Stromal p38MAPK/MK2 Pathway Limits Breast Cancer Metastases and Chemotherapy-Induced Bone Loss, Cancer Res 78(19) (2018) 5618–5630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Yao Z, Murali B, Ren Q, Luo X, Faget DV, Cole T, Ricci B, Thotala D, Monahan J, van Deursen JM, Baker D, Faccio R, Schwarz JK, Stewart SA, Therapy-Induced Senescence Drives Bone Loss, Cancer Res 80(5) (2020) 1171–1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Sun Y, Campisi J, Higano C, Beer TM, Porter P, Coleman I, True L, Nelson PS, Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B, Nat Med 18(9) (2012) 1359–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Wiley CD, Brumwell AN, Davis SS, Jackson JR, Valdovinos A, Calhoun C, Alimirah F, Castellanos CA, Ruan R, Wei Y, Chapman HA, Ramanathan A, Campisi J, Jourdan Le Saux C, Secretion of leukotrienes by senescent lung fibroblasts promotes pulmonary fibrosis, JCI Insight 4(24) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Schafer MJ, White TA, Iijima K, Haak AJ, Ligresti G, Atkinson EJ, Oberg AL, Birch J, Salmonowicz H, Zhu Y, Mazula DL, Brooks RW, Fuhrmann-Stroissnigg H, Pirtskhalava T, Prakash YS, Tchkonia T, Robbins PD, Aubry MC, Passos JF, Kirkland JL, Tschumperlin DJ, Kita H, LeBrasseur NK, Cellular senescence mediates fibrotic pulmonary disease, Nat Commun 8 (2017) 14532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Chang BD, Broude EV, Dokmanovic M, Zhu H, Ruth A, Xuan Y, Kandel ES, Lausch E, Christov K, Roninson IB, A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents, Cancer Res 59(15) (1999) 3761–7. [PubMed] [Google Scholar]
- [86].Jones KR, Elmore LW, Jackson-Cook C, Demasters G, Povirk LF, Holt SE, Gewirtz DA, p53-Dependent accelerated senescence induced by ionizing radiation in breast tumour cells, Int J Radiat Biol 81(6) (2005) 445–58. [DOI] [PubMed] [Google Scholar]
- [87].Jeon HY, Kim JK, Ham SW, Oh SY, Kim J, Park JB, Lee JY, Kim SC, Kim H, Irradiation induces glioblastoma cell senescence and senescence-associated secretory phenotype, Tumour Biol 37(5) (2016) 5857–67. [DOI] [PubMed] [Google Scholar]
- [88].Kansara M, Leong HS, Lin DM, Popkiss S, Pang P, Garsed DW, Walkley CR, Cullinane C, Ellul J, Haynes NM, Hicks R, Kuijjer ML, Cleton-Jansen AM, Hinds PW, Smyth MJ, Thomas DM, Immune response to RB1-regulated senescence limits radiation-induced osteosarcoma formation, J Clin Invest 123(12) (2013) 5351–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Xu J, Patel NH, Saleh T, Cudjoe EK Jr., Alotaibi M, Wu Y, Lima S, Hawkridge AM, Gewirtz DA, Differential Radiation Sensitivity in p53 Wild-Type and p53-Deficient Tumor Cells Associated with Senescence but not Apoptosis or (Nonprotective) Autophagy, Radiat Res 190(5) (2018) 538–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Nesslinger NJ, Sahota RA, Stone B, Johnson K, Chima N, King C, Rasmussen D, Bishop D, Rennie PS, Gleave M, Blood P, Pai H, Ludgate C, Nelson BH, Standard treatments induce antigen-specific immune responses in prostate cancer, Clin Cancer Res 13(5) (2007) 1493–502. [DOI] [PubMed] [Google Scholar]
- [91].Lhuillier C, Rudqvist NP, Elemento O, Formenti SC, Demaria S, Radiation therapy and anti-tumor immunity: exposing immunogenic mutations to the immune system, Genome Med 11(1) (2019) 40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Lee Y, Auh SL, Wang Y, Burnette B, Wang Y, Meng Y, Beckett M, Sharma R, Chin R, Tu T, Weichselbaum RR, Fu YX, Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment, Blood 114(3) (2009) 589–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Crittenden MR, Zebertavage L, Kramer G, Bambina S, Friedman D, Troesch V, Blair T, Baird JR, Alice A, Gough MJ, Tumor cure by radiation therapy and checkpoint inhibitors depends on pre-existing immunity, Sci Rep 8(1) (2018) 7012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Gameiro SR, Jammeh ML, Wattenberg MM, Tsang KY, Ferrone S, Hodge JW, Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing, Oncotarget 5(2) (2014) 403–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Fang C, Mo F, Liu L, Du J, Luo M, Men K, Na F, Wang W, Yang H, Wei X, Oxidized mitochondrial DNA sensing by STING signaling promotes the antitumor effect of an irradiated immunogenic cancer cell vaccine, Cell Mol Immunol 18(9) (2021) 2211–2223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Gasser S, Orsulic S, Brown EJ, Raulet DH, The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor, Nature 436(7054) (2005) 1186–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Lam AR, Bert NL, Ho SS, Shen YJ, Tang LF, Xiong GM, Croxford JL, Koo CX, Ishii KJ, Akira S, Raulet DH, Gasser S, RAE1 ligands for the NKG2D receptor are regulated by STING-dependent DNA sensor pathways in lymphoma, Cancer research 74(8) (2014) 2193–2203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Tesei A, Arienti C, Bossi G, Santi S, De Santis I, Bevilacqua A, Zanoni M, Pignatta S, Cortesi M, Zamagni A, Storci G, Bonafe M, Sarnelli A, Romeo A, Cavallo C, Bartolazzi A, Rossi S, Soriani A, Strigari L, TP53 drives abscopal effect by secretion of senescence-associated molecular signals in non-small cell lung cancer, J Exp Clin Cancer Res 40(1) (2021) 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Deng L, Liang H, Xu M, Yang X, Burnette B, Arina A, Li XD, Mauceri H, Beckett M, Darga T, Huang X, Gajewski TF, Chen ZJ, Fu YX, Weichselbaum RR, STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors, Immunity 41(5) (2014) 843–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Glück S, Guey B, Gulen MF, Wolter K, Kang TW, Schmacke NA, Bridgeman A, Rehwinkel J, Zender L, Ablasser A, Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence, Nature cell biology 19(9) (2017) 1061–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Yang H, Wang H, Ren J, Chen Q, Chen ZJ, cGAS is essential for cellular senescence, Proceedings of the National Academy of Sciences of the United States of America 114(23) (2017) E4612–e4620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].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(7676) (2017) 402–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Meng Y, Efimova EV, Hamzeh KW, Darga TE, Mauceri HJ, Fu YX, Kron SJ, Weichselbaum RR, Radiation-inducible immunotherapy for cancer: senescent tumor cells as a cancer vaccine, Mol Ther 20(5) (2012) 1046–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Sadhu S, Decker C, Sansbury BE, Marinello M, Seyfried A, Howard J, Mori M, Hosseini Z, Arunachalam T, Finn AV, Lamar JM, Jourd’heuil D, Guo L, MacNamara KC, Spite M, Fredman G, Radiation-Induced Macrophage Senescence Impairs Resolution Programs and Drives Cardiovascular Inflammation, J Immunol 207(7) (2021) 1812–1823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Rosen EM, Fan S, Rockwell S, Goldberg ID, The molecular and cellular basis of radiosensitivity: implications for understanding how normal tissues and tumors respond to therapeutic radiation, Cancer Invest 17(1) (1999) 56–72. [PubMed] [Google Scholar]
- [106].Palacio L, Goyer ML, Maggiorani D, Espinosa A, Villeneuve N, Bourbonnais S, Moquin-Beaudry G, Le O, Demaria M, Davalos AR, Decaluwe H, Beauséjour C, Restored immune cell functions upon clearance of senescence in the irradiated splenic environment, Aging Cell 18(4) (2019) e12971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Le ON, Rodier F, Fontaine F, Coppe JP, Campisi J, DeGregori J, Laverdière C, Kokta V, Haddad E, Beauséjour CM, Ionizing radiation-induced long-term expression of senescence markers in mice is independent of p53 and immune status, Aging Cell 9(3) (2010) 398–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Hansel C, Barr S, Schemann AV, Lauber K, Hess J, Unger K, Zitzelsberger H, Jendrossek V, Klein D, Metformin Protects against Radiation-Induced Acute Effects by Limiting Senescence of Bronchial-Epithelial Cells, Int J Mol Sci 22(13) (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Zhang J, Li B, Yang Q, Zhang P, Wang H, Prognostic value of Aurora kinase A (AURKA) expression among solid tumor patients: a systematic review and meta-analysis, Jpn J Clin Oncol 45(7) (2015) 629–36. [DOI] [PubMed] [Google Scholar]
- [110].Carvajal RD, Tse A, Schwartz GK, Aurora kinases: new targets for cancer therapy, Clin Cancer Res 12(23) (2006) 6869–75. [DOI] [PubMed] [Google Scholar]
- [111].Liu Y, Hawkins OE, Su Y, Vilgelm AE, Sobolik T, Thu YM, Kantrow S, Splittgerber RC, Short S, Amiri KI, Ecsedy JA, Sosman JA, Kelley MC, Richmond A, Targeting aurora kinases limits tumour growth through DNA damage-mediated senescence and blockade of NF-kappaB impairs this drug-induced senescence, EMBO Mol Med 5(1) (2013) 149–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Huck JJ, Zhang M, McDonald A, Bowman D, Hoar KM, Stringer B, Ecsedy J, Manfredi MG, Hyer ML, MLN8054, an inhibitor of Aurora A kinase, induces senescence in human tumor cells both in vitro and in vivo, Mol Cancer Res 8(3) (2010) 373–84. [DOI] [PubMed] [Google Scholar]
- [113].Vilgelm AE, Johnson CA, Prasad N, Yang J, Chen SC, Ayers GD, Pawlikowski JS, Raman D, Sosman JA, Kelley M, Ecsedy JA, Shyr Y, Levy SE, Richmond A, Connecting the Dots: Therapy-Induced Senescence and a Tumor-Suppressive Immune Microenvironment, J Natl Cancer Inst 108(6) (2016) djv406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Punt S, Malu S, McKenzie JA, Manrique SZ, Doorduijn EM, Mbofung RM, Williams L, Silverman DA, Ashkin EL, Dominguez AL, Wang Z, Chen JQ, Maiti SN, Tieu TN, Liu C, Xu C, Forget MA, Haymaker C, Khalili JS, Satani N, Muller F, Cooper LJN, Overwijk WW, Amaria RN, Bernatchez C, Heffernan TP, Peng W, Roszik J, Hwu P, Aurora kinase inhibition sensitizes melanoma cells to T-cell-mediated cytotoxicity, Cancer Immunol Immunother 70(4) (2021) 1101–1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Blas-Rus N, Bustos-Morán E, Pérez de Castro I, de Cárcer G, Borroto A, Camafeita E, Jorge I, Vázquez J, Alarcón B, Malumbres M, Martín-Cófreces NB, Sánchez-Madrid F, Aurora A drives early signalling and vesicle dynamics during T-cell activation, Nat Commun 7 (2016) 11389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Bustos-Morán E, Blas-Rus N, Alcaraz-Serna A, Iborra S, González-Martínez J, Malumbres M, Sánchez-Madrid F, Aurora A controls CD8(+) T cell cytotoxic activity and antiviral response, Sci Rep 9(1) (2019) 2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Betts BC, Veerapathran A, Pidala J, Yang H, Horna P, Walton K, Cubitt CL, Gunawan S, Lawrence HR, Lawrence NJ, Sebti SM, Anasetti C, Targeting Aurora kinase A and JAK2 prevents GVHD while maintaining Treg and antitumor CTL function, Sci Transl Med 9(372) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Sherr CJ, Roberts JM, CDK inhibitors: positive and negative regulators of G1-phase progression, Genes Dev 13(12) (1999) 1501–12. [DOI] [PubMed] [Google Scholar]
- [119].Rubin SM, Sage J, Skotheim JM, Integrating Old and New Paradigms of G1/S Control, Mol Cell 80(2) (2020) 183–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Shapiro GI, Harper JW, Anticancer drug targets: cell cycle and checkpoint control, J Clin Invest 104(12) (1999) 1645–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Sherr CJ, Beach D, Shapiro GI, Targeting CDK4 and CDK6: From Discovery to Therapy, Cancer Discov 6(4) (2016) 353–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Klein ME, Kovatcheva M, Davis LE, Tap WD, Koff A, CDK4/6 Inhibitors: The Mechanism of Action May Not Be as Simple as Once Thought, Cancer Cell 34(1) (2018) 9–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Wagner V, Gil J, Senescence as a therapeutically relevant response to CDK4/6 inhibitors, Oncogene 39(29) (2020) 5165–5176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Goel S, DeCristo MJ, Watt AC, BrinJones H, Sceneay J, Li BB, Khan N, Ubellacker JM, Xie S, Metzger-Filho O, Hoog J, Ellis MJ, Ma CX, Ramm S, Krop IE, Winer EP, Roberts TM, Kim HJ, McAllister SS, Zhao JJ, CDK4/6 inhibition triggers anti-tumour immunity, Nature 548(7668) (2017) 471–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, Chhabra S, Huang W, Liu H, Aref AR, Ivanova E, Paweletz CP, Bowden M, Zhou CW, Herter-Sprie GS, Sorrentino JA, Bisi JE, Lizotte PH, Merlino AA, Quinn MM, Bufe LE, Yang A, Zhang Y, Zhang H, Gao P, Chen T, Cavanaugh ME, Rode AJ, Haines E, Roberts PJ, Strum JC, Richards WG, Lorch JH, Parangi S, Gunda V, Boland GM, Bueno R, Palakurthi S, Freeman GJ, Ritz J, Haining WN, Sharpless NE, Arthanari H, Shapiro GI, Barbie DA, Gray NS, Wong KK, CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation, Cancer Discov 8(2) (2018) 216–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Jerby-Arnon L, Shah P, Cuoco MS, Rodman C, Su MJ, Melms JC, Leeson R, Kanodia A, Mei S, Lin JR, Wang S, Rabasha B, Liu D, Zhang G, Margolais C, Ashenberg O, Ott PA, Buchbinder EI, Haq R, Hodi FS, Boland GM, Sullivan RJ, Frederick DT, Miao B, Moll T, Flaherty KT, Herlyn M, Jenkins RW, Thummalapalli R, Kowalczyk MS, Canadas I, Schilling B, Cartwright ANR, Luoma AM, Malu S, Hwu P, Bernatchez C, Forget MA, Barbie DA, Shalek AK, Tirosh I, Sorger PK, Wucherpfennig K, Van Allen EM, Schadendorf D, Johnson BE, Rotem A, Rozenblatt-Rosen O, Garraway LA, Yoon CH, Izar B, Regev A, A Cancer Cell Program Promotes T Cell Exclusion and Resistance to Checkpoint Blockade, Cell 175(4) (2018) 984–997 e24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Guan X, LaPak KM, Hennessey RC, Yu CY, Shakya R, Zhang J, Burd CE, Stromal Senescence By Prolonged CDK4/6 Inhibition Potentiates Tumor Growth, Mol Cancer Res 15(3) (2017) 237–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Wang C, Vegna S, Jin H, Benedict B, Lieftink C, Ramirez C, de Oliveira RL, Morris B, Gadiot J, Wang W, du Chatinier A, Wang L, Gao D, Evers B, Jin G, Xue Z, Schepers A, Jochems F, Sanchez AM, Mainardi S, Te Riele H, Beijersbergen RL, Qin W, Akkari L, Bernards R, Inducing and exploiting vulnerabilities for the treatment of liver cancer, Nature 574(7777) (2019) 268–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Franco J, Balaji U, Freinkman E, Witkiewicz AK, Knudsen ES, Metabolic Reprogramming of Pancreatic Cancer Mediated by CDK4/6 Inhibition Elicits Unique Vulnerabilities, Cell Rep 14(5) (2016) 979–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Kamal Y, Cheng C, Frost HR, Amos CI, Predictors of disease aggressiveness influence outcome from immunotherapy treatment in renal clear cell carcinoma, Oncoimmunology 8(1) (2019) e1500106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Brenner E, Schörg BF, Ahmetlić F, Wieder T, Hilke FJ, Simon N, Schroeder C, Demidov G, Riedel T, Fehrenbacher B, Schaller M, Forschner A, Eigentler T, Niessner H, Sinnberg T, Böhm KS, Hömberg N, Braumüller H, Dauch D, Zwirner S, Zender L, Sonanini D, Geishauser A, Bauer J, Eichner M, Jarick KJ, Beilhack A, Biskup S, Döcker D, Schadendorf D, Quintanilla-Martinez L, Pichler BJ, Kneilling M, Mocikat R, Röcken M, Cancer immune control needs senescence induction by interferon-dependent cell cycle regulator pathways in tumours, Nat Commun 11(1) (2020) 1335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Adams PD, Remodeling of chromatin structure in senescent cells and its potential impact on tumor suppression and aging, Gene 397(1–2) (2007) 84–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Narita M, Nunez S, Heard E, Narita M, Lin AW, Hearn SA, Spector DL, Hannon GJ, Lowe SW, Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence, Cell 113(6) (2003) 703–16. [DOI] [PubMed] [Google Scholar]
- [134].Chicas A, Wang X, Zhang C, McCurrach M, Zhao Z, Mert O, Dickins RA, Narita M, Zhang M, Lowe SW, Dissecting the unique role of the retinoblastoma tumor suppressor during cellular senescence, Cancer Cell 17(4) (2010) 376–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Braig M, Lee S, Loddenkemper C, Rudolph C, Peters AH, Schlegelberger B, Stein H, Dorken B, Jenuwein T, Schmitt CA, Oncogene-induced senescence as an initial barrier in lymphoma development, Nature 436(7051) (2005) 660–5. [DOI] [PubMed] [Google Scholar]
- [136].Lee JH, Kim EW, Croteau DL, Bohr VA, Heterochromatin: an epigenetic point of view in aging, Exp Mol Med 52(9) (2020) 1466–1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].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(3) (2016) 325–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Ito T, Teo YV, Evans SA, Neretti N, Sedivy JM, Regulation of Cellular Senescence by Polycomb Chromatin Modifiers through Distinct DNA Damage- and Histone Methylation-Dependent Pathways, Cell reports 22(13) (2018) 3480–3492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Hayakawa T, Iwai M, Aoki S, Takimoto K, Maruyama M, Maruyama W, Motoyama N, SIRT1 suppresses the senescence-associated secretory phenotype through epigenetic gene regulation, PLoS One 10(1) (2015) e0116480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].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(6) (2016) 612–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Takahashi A, Imai Y, Yamakoshi K, Kuninaka S, Ohtani N, Yoshimoto S, Hori S, Tachibana M, Anderton E, Takeuchi T, Shinkai Y, Peters G, Saya H, Hara E, DNA damage signaling triggers degradation of histone methyltransferases through APC/C(Cdh1) in senescent cells, Mol Cell 45(1) (2012) 123–31. [DOI] [PubMed] [Google Scholar]
- [142].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(8) (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Chen H, Ruiz PD, McKimpson WM, Novikov L, Kitsis RN, Gamble MJ, MacroH2A1 and ATM Play Opposing Roles in Paracrine Senescence and the Senescence-Associated Secretory Phenotype, Mol Cell 59(5) (2015) 719–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Contrepois K, Coudereau C, Benayoun BA, Schuler N, Roux PF, Bischof O, Courbeyrette R, Carvalho C, Thuret JY, Ma Z, Derbois C, Nevers MC, Volland H, Redon CE, Bonner WM, Deleuze JF, Wiel C, Bernard D, Snyder MP, Rübe CE, Olaso R, Fenaille F, Mann C, Histone variant H2A.J accumulates in senescent cells and promotes inflammatory gene expression, Nat Commun 8 (2017) 14995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Criscione SW, Teo YV, Neretti N, The Chromatin Landscape of Cellular Senescence, Trends Genet 32(11) (2016) 751–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Chandra T, Ewels PA, Schoenfelder S, Furlan-Magaril M, Wingett SW, Kirschner K, Thuret JY, Andrews S, Fraser P, Reik W, Global reorganization of the nuclear landscape in senescent cells, Cell Rep 10(4) (2015) 471–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Zirkel A, Nikolic M, Sofiadis K, Mallm JP, Brackley CA, Gothe H, Drechsel O, Becker C, Altmuller J, Josipovic N, Georgomanolis T, Brant L, Franzen J, Koker M, Gusmao EG, Costa IG, Ullrich RT, Wagner W, Roukos V, Nurnberg P, Marenduzzo D, Rippe K, Papantonis A, HMGB2 Loss upon Senescence Entry Disrupts Genomic Organization and Induces CTCF Clustering across Cell Types, Mol Cell 70(4) (2018) 730–744 e6. [DOI] [PubMed] [Google Scholar]
- [148].Buj R, Leon KE, Anguelov MA, Aird KM, Suppression of p16 alleviates the senescence-associated secretory phenotype, Aging (Albany NY) 13(3) (2021) 3290–3312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Acosta JC, O’Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, Fumagalli M, Da Costa M, Brown C, Popov N, Takatsu Y, Melamed J, d’Adda di Fagagna F, Bernard D, Hernando E, Gil J, Chemokine signaling via the CXCR2 receptor reinforces senescence, Cell 133(6) (2008) 1006–18. [DOI] [PubMed] [Google Scholar]
- [150].Nerlov C, The C/EBP family of transcription factors: a paradigm for interaction between gene expression and proliferation control, Trends Cell Biol 17(7) (2007) 318–24. [DOI] [PubMed] [Google Scholar]
- [151].Kang C, Xu Q, Martin TD, Li MZ, Demaria M, Aron L, Lu T, Yankner BA, Campisi J, Elledge SJ, The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4, Science (New York, N.Y.) 349(6255) (2015) aaa5612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Alspach E, Flanagan KC, Luo X, Ruhland MK, Huang H, Pazolli E, Donlin MJ, Marsh T, Piwnica-Worms D, Monahan J, Novack DV, McAllister SS, Stewart SA, p38MAPK plays a crucial role in stromal-mediated tumorigenesis, Cancer Discov 4(6) (2014) 716–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Freund A, Patil CK, Campisi J, p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype, EMBO J 30(8) (2011) 1536–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Barascu A, Le Chalony C, Pennarun G, Genet D, Imam N, Lopez B, Bertrand P, Oxidative stress induces an ATM-independent senescence pathway through p38 MAPK-mediated lamin B1 accumulation, EMBO J 31(5) (2012) 1080–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Ivanov A, Pawlikowski J, Manoharan I, van Tuyn J, Nelson DM, Rai TS, Shah PP, Hewitt G, Korolchuk VI, Passos JF, Wu H, Berger SL, Adams PD, Lysosome-mediated processing of chromatin in senescence, J Cell Biol 202(1) (2013) 129–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Freund A, Laberge RM, Demaria M, Campisi J, Lamin B1 loss is a senescence-associated biomarker, Mol Biol Cell 23(11) (2012) 2066–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Omer A, Barrera MC, Moran JL, Lian XJ, Di Marco S, Beausejour C, Gallouzi IE, G3BP1 controls the senescence-associated secretome and its impact on cancer progression, Nat Commun 11(1) (2020) 4979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Chan CJ, Smyth MJ, Martinet L, Molecular mechanisms of natural killer cell activation in response to cellular stress, Cell Death Differ 21(1) (2014) 5–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Raulet DH, Guerra N, Oncogenic stress sensed by the immune system: role of natural killer cell receptors, Nat Rev Immunol 9(8) (2009) 568–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [160].Lesina M, Wormann SM, Morton J, Diakopoulos KN, Korneeva O, Wimmer M, Einwachter H, Sperveslage J, Demir IE, Kehl T, Saur D, Sipos B, Heikenwalder M, Steiner JM, Wang TC, Sansom OJ, Schmid RM, Algul H, RelA regulates CXCL1/CXCR2-dependent oncogene-induced senescence in murine Kras-driven pancreatic carcinogenesis, J Clin Invest 126(8) (2016) 2919–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [161].Hoare M, Ito Y, Kang TW, Weekes MP, Matheson NJ, Patten DA, Shetty S, Parry AJ, Menon S, Salama R, Antrobus R, Tomimatsu K, Howat W, Lehner PJ, Zender L, Narita M, NOTCH1 mediates a switch between two distinct secretomes during senescence, Nature cell biology 18(9) (2016) 979–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [162].Wiley CD, Velarde MC, Lecot P, Liu S, Sarnoski EA, Freund A, Shirakawa K, Lim HW, Davis SS, Ramanathan A, Gerencser AA, Verdin E, Campisi J, Mitochondrial Dysfunction Induces Senescence with a Distinct Secretory Phenotype, Cell Metab 23(2) (2016) 303–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Pribluda A, Elyada E, Wiener Z, Hamza H, Goldstein RE, Biton M, Burstain I, Morgenstern Y, Brachya G, Billauer H, Biton S, Snir-Alkalay I, Vucic D, Schlereth K, Mernberger M, Stiewe T, Oren M, Alitalo K, Pikarsky E, Ben-Neriah Y, A senescence-inflammatory switch from cancer-inhibitory to cancer-promoting mechanism, Cancer Cell 24(2) (2013) 242–56. [DOI] [PubMed] [Google Scholar]
- [164].Nacarelli T, Lau L, Fukumoto T, Zundell J, Fatkhutdinov N, Wu S, Aird KM, Iwasaki O, Kossenkov AV, Schultz D, Noma KI, Baur JA, Schug Z, Tang HY, Speicher DW, David G, Zhang R, NAD(+) metabolism governs the proinflammatory senescence-associated secretome, Nat Cell Biol 21(3) (2019) 397–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [165].Laberge RM, Sun Y, Orjalo AV, Patil CK, Freund A, Zhou L, Curran SC, Davalos AR, Wilson-Edell KA, Liu S, Limbad C, Demaria M, Li P, Hubbard GB, Ikeno Y, Javors M, Desprez PY, Benz CC, Kapahi P, Nelson PS, Campisi J, MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation, Nat Cell Biol 17(8) (2015) 1049–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [166].Lau L, Porciuncula A, Yu A, Iwakura Y, David G, Uncoupling the Senescence-Associated Secretory Phenotype from Cell Cycle Exit via Interleukin-1 Inactivation Unveils Its Protumorigenic Role, Mol Cell Biol 39(12) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [167].Hoare M, Narita M, Transmitting senescence to the cell neighbourhood, Nat Cell Biol 15(8) (2013) 887–9. [DOI] [PubMed] [Google Scholar]
- [168].Acosta JC, Banito A, Wuestefeld T, Georgilis A, Janich P, Morton JP, Athineos D, Kang TW, Lasitschka F, Andrulis M, Pascual G, Morris KJ, Khan S, Jin H, Dharmalingam G, Snijders AP, Carroll T, Capper D, Pritchard C, Inman GJ, Longerich T, Sansom OJ, Benitah SA, Zender L, Gil J, A complex secretory program orchestrated by the inflammasome controls paracrine senescence, Nat Cell Biol 15(8) (2013) 978–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Wiley CD, Sharma R, Davis SS, Lopez-Dominguez JA, Mitchell KP, Wiley S, Alimirah F, Kim DE, Payne T, Rosko A, Aimontche E, Deshpande SM, Neri F, Kuehnemann C, Demaria M, Ramanathan A, Campisi J, Oxylipin biosynthesis reinforces cellular senescence and allows detection of senolysis, Cell Metab 33(6) (2021) 1124–1136 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Herranz N, Gallage S, Mellone M, Wuestefeld T, Klotz S, Hanley CJ, Raguz S, Acosta JC, Innes AJ, Banito A, Georgilis A, Montoya A, Wolter K, Dharmalingam G, Faull P, Carroll T, Martínez-Barbera JP, Cutillas P, Reisinger F, Heikenwalder M, Miller RA, Withers D, Zender L, Thomas GJ, Gil J, mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype, Nature cell biology 17(9) (2015) 1205–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [171].Wang R, Yu Z, Sunchu B, Shoaf J, Dang I, Zhao S, Caples K, Bradley L, Beaver LM, Ho E, Löhr CV, Perez VI, Rapamycin inhibits the secretory phenotype of senescent cells by a Nrf2-independent mechanism, Aging Cell 16(3) (2017) 564–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [172].Hari P, Millar FR, Tarrats N, Birch J, Quintanilla A, Rink CJ, Fernandez-Duran I, Muir M, Finch AJ, Brunton VG, Passos JF, Morton JP, Boulter L, Acosta JC, The innate immune sensor Toll-like receptor 2 controls the senescence-associated secretory phenotype, Sci Adv 5(6) (2019) eaaw0254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [173].Guccini I, Revandkar A, D’Ambrosio M, Colucci M, Pasquini E, Mosole S, Troiani M, Brina D, Sheibani-Tezerji R, Elia AR, Rinaldi A, Pernigoni N, Ruschoff JH, Dettwiler S, De Marzo AM, Antonarakis ES, Borrelli C, Moor AE, Garcia-Escudero R, Alajati A, Attanasio G, Losa M, Moch H, Wild P, Egger G, Alimonti A, Senescence Reprogramming by TIMP1 Deficiency Promotes Prostate Cancer Metastasis, Cancer Cell 39(1) (2021) 68–82 e9. [DOI] [PubMed] [Google Scholar]
- [174].De Cecco M, Ito T, Petrashen AP, Elias AE, Skvir NJ, Criscione SW, Caligiana A, Brocculi G, Adney EM, Boeke JD, Le O, Beauséjour C, Ambati J, Ambati K, Simon M, Seluanov A, Gorbunova V, Slagboom PE, Helfand SL, Neretti N, Sedivy JM, L1 drives IFN in senescent cells and promotes age-associated inflammation, Nature 566(7742) (2019) 73–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Georgilis A, Klotz S, Hanley CJ, Herranz N, Weirich B, Morancho B, Leote AC, D’Artista L, Gallage S, Seehawer M, Carroll T, Dharmalingam G, Wee KB, Mellone M, Pombo J, Heide D, Guccione E, Arribas J, Barbosa-Morais NL, Heikenwalder M, Thomas GJ, Zender L, Gil J, PTBP1-Mediated Alternative Splicing Regulates the Inflammatory Secretome and the Pro-tumorigenic Effects of Senescent Cells, Cancer cell 34(1) (2018) 85–102.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Bischof O, Martinez-Zamudio RI, MicroRNAs and lncRNAs in senescence: A re-view, IUBMB Life 67(4) (2015) 255–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Bhaumik D, Scott GK, Schokrpur S, Patil CK, Orjalo AV, Rodier F, Lithgow GJ, Campisi J, MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8, Aging (Albany NY) 1(4) (2009) 402–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Terlecki-Zaniewicz L, Lämmermann I, Latreille J, Bobbili MR, Pils V, Schosserer M, Weinmüllner R, Dellago H, Skalicky S, Pum D, Almaraz JCH, Scheideler M, Morizot F, Hackl M, Gruber F, Grillari J, Small extracellular vesicles and their miRNA cargo are anti-apoptotic members of the senescence-associated secretory phenotype, Aging (Albany NY) 10(5) (2018) 1103–1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [179].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(1) (2020) e3000599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Takasugi M, Emerging roles of extracellular vesicles in cellular senescence and aging, Aging Cell 17(2) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [181].Ozcan S, Alessio N, Acar MB, Mert E, Omerli F, Peluso G, Galderisi U, Unbiased analysis of senescence associated secretory phenotype (SASP) to identify common components following different genotoxic stresses, Aging (Albany NY) 8(7) (2016) 1316–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Jochems F, Thijssen B, De Conti G, Jansen R, Pogacar Z, Groot K, Wang L, Schepers A, Wang C, Jin H, Beijersbergen RL, Leite de Oliveira R, Wessels LFA, Bernards R, The Cancer SENESCopedia: A delineation of cancer cell senescence, Cell Rep 36(4) (2021) 109441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [183].Hernandez-Segura A, de Jong TV, Melov S, Guryev V, Campisi J, Demaria M, Unmasking Transcriptional Heterogeneity in Senescent Cells, Curr Biol 27(17) (2017) 2652–2660 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Zhang H, Pan KH, Cohen SN, Senescence-specific gene expression fingerprints reveal cell-type-dependent physical clustering of up-regulated chromosomal loci, Proc Natl Acad Sci U S A 100(6) (2003) 3251–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [185].Wiley CD, Flynn JM, Morrissey C, Lebofsky R, Shuga J, Dong X, Unger MA, Vijg J, Melov S, Campisi J, Analysis of individual cells identifies cell-to-cell variability following induction of cellular senescence, Aging Cell 16(5) (2017) 1043–1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Ruhland MK, Alspach E, Senescence and Immunoregulation in the Tumor Microenvironment, Front Cell Dev Biol 9 (2021) 754069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [187].Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, Kirkland JL, van Deursen JM, Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders, Nature 479(7372) (2011) 232–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, Saltness RA, Jeganathan KB, Verzosa GC, Pezeshki A, Khazaie K, Miller JD, van Deursen JM, Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan, Nature 530(7589) (2016) 184–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Alimirah F, Pulido T, Valdovinos A, Alptekin S, Chang E, Jones E, Diaz DA, Flores J, Velarde MC, Demaria M, Davalos AR, Wiley CD, Limbad C, Desprez P-Y, Campisi J, Cellular Senescence Promotes Skin Carcinogenesis through p38MAPK and p44/42MAPK Signaling, Cancer Research 80(17) (2020) 3606–3619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Jeon OH, Kim C, Laberge RM, Demaria M, Rathod S, Vasserot AP, Chung JW, Kim DH, Poon Y, David N, Baker DJ, van Deursen JM, Campisi J, Elisseeff JH, Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment, Nat Med 23(6) (2017) 775–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [191].Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ, Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline, Nature 562(7728) (2018) 578–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [192].Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, Palmer AK, Ikeno Y, Hubbard GB, Lenburg M, O’Hara SP, LaRusso NF, Miller JD, Roos CM, Verzosa GC, LeBrasseur NK, Wren JD, Farr JN, Khosla S, Stout MB, McGowan SJ, Fuhrmann-Stroissnigg H, Gurkar AU, Zhao J, Colangelo D, Dorronsoro A, Ling YY, Barghouthy AS, Navarro DC, Sano T, Robbins PD, Niedernhofer LJ, Kirkland JL, The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs, Aging Cell 14(4) (2015) 644–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Kirkland JL, Tchkonia T, Senolytic drugs: from discovery to translation, J Intern Med 288(5) (2020) 518–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [194].Yousefzadeh MJ, Zhu Y, McGowan SJ, Angelini L, Fuhrmann-Stroissnigg H, Xu M, Ling YY, Melos KI, Pirtskhalava T, Inman CL, McGuckian C, Wade EA, Kato JI, Grassi D, Wentworth M, Burd CE, Arriaga EA, Ladiges WL, Tchkonia T, Kirkland JL, Robbins PD, Niedernhofer LJ, Fisetin is a senotherapeutic that extends health and lifespan, EBioMedicine 36 (2018) 18–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [195].Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, Inman CL, Ogrodnik MB, Hachfeld CM, Fraser DG, Onken JL, Johnson KO, Verzosa GC, Langhi LGP, Weigl M, Giorgadze N, LeBrasseur NK, Miller JD, Jurk D, Singh RJ, Allison DB, Ejima K, Hubbard GB, Ikeno Y, Cubro H, Garovic VD, Hou X, Weroha SJ, Robbins PD, Niedernhofer LJ, Khosla S, Tchkonia T, Kirkland JL, Senolytics improve physical function and increase lifespan in old age, Nat Med 24(8) (2018) 1246–1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [196].Baar MP, Brandt RMC, Putavet DA, Klein JDD, Derks KWJ, Bourgeois BRM, Stryeck S, Rijksen Y, van Willigenburg H, Feijtel DA, van der Pluijm I, Essers J, van Cappellen WA, van IWF, Houtsmuller AB, Pothof J, de Bruin RWF, Madl T, Hoeijmakers JHJ, Campisi J, de Keizer PLJ, Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging, Cell 169(1) (2017) 132–147 e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [197].Iske J, Seyda M, Heinbokel T, Maenosono R, Minami K, Nian Y, Quante M, Falk CS, Azuma H, Martin F, Passos JF, Niemann CU, Tchkonia T, Kirkland JL, Elkhal A, Tullius SG, Senolytics prevent mt-DNA-induced inflammation and promote the survival of aged organs following transplantation, Nat Commun 11(1) (2020) 4289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [198].Robbins PD, Jurk D, Khosla S, Kirkland JL, LeBrasseur NK, Miller JD, Passos JF, Pignolo RJ, Tchkonia T, Niedernhofer LJ, Senolytic Drugs: Reducing Senescent Cell Viability to Extend Health Span, Annu Rev Pharmacol Toxicol 61 (2021) 779–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [199].Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, Prata L, Masternak MM, Kritchevsky SB, Musi N, Kirkland JL, Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study, EBioMedicine 40 (2019) 554–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [200].Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, Herrmann SM, Jensen MD, Jia Q, Jordan KL, Kellogg TA, Khosla S, Koerber DM, Lagnado AB, Lawson DK, LeBrasseur NK, Lerman LO, McDonald KM, McKenzie TJ, Passos JF, Pignolo RJ, Pirtskhalava T, Saadiq IM, Schaefer KK, Textor SC, Victorelli SG, Volkman TL, Xue A, Wentworth MA, Wissler Gerdes EO, Zhu Y, Tchkonia T, Kirkland JL, Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease, EBioMedicine 47 (2019) 446–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [201].Dolgin E, Send in the senolytics, Nat Biotechnol 38(12) (2020) 1371–1377. [DOI] [PubMed] [Google Scholar]
- [202].Wang L, Bernards R, Taking advantage of drug resistance, a new approach in the war on cancer, Front Med 12(4) (2018) 490–495. [DOI] [PubMed] [Google Scholar]
- [203].Wang L, Leite de Oliveira R, Wang C, Fernandes Neto JM, Mainardi S, Evers B, Lieftink C, Morris B, Jochems F, Willemsen L, Beijersbergen RL, Bernards R, High-Throughput Functional Genetic and Compound Screens Identify Targets for Senescence Induction in Cancer, Cell Rep 21(3) (2017) 773–783. [DOI] [PubMed] [Google Scholar]
- [204].Saleh T, Carpenter VJ, Tyutyunyk-Massey L, Murray G, Leverson JD, Souers AJ, Alotaibi MR, Faber AC, Reed J, Harada H, Gewirtz DA, Clearance of therapy-induced senescent tumor cells by the senolytic ABT-263 via interference with BCL-XL -BAX interaction, Mol Oncol 14(10) (2020) 2504–2519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [205].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(1) (2019) 2556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [206].Galiana I, Lozano-Torres B, Sancho M, Alfonso M, Bernardos A, Bisbal V, Serrano M, Martinez-Manez R, Orzaez M, Preclinical antitumor efficacy of senescence-inducing chemotherapy combined with a nanoSenolytic, J Control Release 323 (2020) 624–634. [DOI] [PubMed] [Google Scholar]
- [207].Shahbandi A, Rao SG, Anderson AY, Frey WD, Olayiwola JO, Ungerleider NA, Jackson JG, BH3 mimetics selectively eliminate chemotherapy-induced senescent cells and improve response in TP53 wild-type breast cancer, Cell Death Differ 27(11) (2020) 3097–3116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [208].Whittle JR, Vaillant F, Surgenor E, Policheni AN, Giner G, Capaldo BD, Chen HR, Liu HK, Dekkers JF, Sachs N, Clevers H, Fellowes A, Green T, Xu H, Fox SB, Herold MJ, Smyth GK, Gray DHD, Visvader JE, Lindeman GJ, Dual Targeting of CDK4/6 and BCL2 Pathways Augments Tumor Response in Estrogen Receptor-Positive Breast Cancer, Clin Cancer Res 26(15) (2020) 4120–4134. [DOI] [PubMed] [Google Scholar]
- [209].Wu H, Schiff DS, Lin Y, Neboori HJ, Goyal S, Feng Z, Haffty BG, Ionizing radiation sensitizes breast cancer cells to Bcl-2 inhibitor, ABT-737, through regulating Mcl-1, Radiat Res 182(6) (2014) 618–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [210].Vilgelm AE, Pawlikowski JS, Liu Y, Hawkins OE, Davis TA, Smith J, Weller KP, Horton LW, McClain CM, Ayers GD, Turner DC, Essaka DC, Stewart CF, Sosman JA, Kelley MC, Ecsedy JA, Johnston JN, Richmond A, Mdm2 and aurora kinase a inhibitors synergize to block melanoma growth by driving apoptosis and immune clearance of tumor cells, Cancer Res 75(1) (2015) 181–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [211].Acklin S, Zhang M, Du W, Zhao X, Plotkin M, Chang J, Campisi J, Zhou D, Xia F, Depletion of senescent-like neuronal cells alleviates cisplatin-induced peripheral neuropathy in mice, Sci Rep 10(1) (2020) 14170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [212].Chang J, Wang Y, Shao L, Laberge RM, Demaria M, Campisi J, Janakiraman K, Sharpless NE, Ding S, Feng W, Luo Y, Wang X, Aykin-Burns N, Krager K, Ponnappan U, Hauer-Jensen M, Meng A, Zhou D, Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice, Nat Med 22(1) (2016) 78–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [213].Pan J, Li D, Xu Y, Zhang J, Wang Y, Chen M, Lin S, Huang L, Chung EJ, Citrin DE, Wang Y, Hauer-Jensen M, Zhou D, Meng A, Inhibition of Bcl-2/xl With ABT-263 Selectively Kills Senescent Type II Pneumocytes and Reverses Persistent Pulmonary Fibrosis Induced by Ionizing Radiation in Mice, Int J Radiat Oncol Biol Phys 99(2) (2017) 353–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [214].Paez-Ribes M, Gonzalez-Gualda E, Doherty GJ, Munoz-Espin D, Targeting senescent cells in translational medicine, EMBO Mol Med 11(12) (2019) e10234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [215].He K, Barsoumian HB, Yang L, Sezen D, Wasley MD, Masrorpour F, Cortez MA, Welsh JW, Senolytic Cocktail Dasatinib Plus Quercetin Enhances the Antitumor Effect of Senescence-Inducing Radiotherapy in a Preclinical Model of Melanoma, Int J Radiat Oncol Biol Phys 111(3S) (2021) S57. [Google Scholar]
- [216].Kovacovicova K, Skolnaja M, Heinmaa M, Mistrik M, Pata P, Pata I, Bartek J, Vinciguerra M, Senolytic Cocktail Dasatinib+Quercetin (D+Q) Does Not Enhance the Efficacy of Senescence-Inducing Chemotherapy in Liver Cancer, Front Oncol 8 (2018) 459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [217].Kaefer A, Yang J, Noertersheuser P, Mensing S, Humerickhouse R, Awni W, Xiong H, Mechanism-based pharmacokinetic/pharmacodynamic meta-analysis of navitoclax (ABT-263) induced thrombocytopenia, Cancer Chemother Pharmacol 74(3) (2014) 593–602. [DOI] [PubMed] [Google Scholar]
- [218].Pi L, Rooprai J, Allan DS, Atkins H, Bredeson C, Fulcher AJ, Ito C, Ramsay T, Shorr, Stanford WL, Sabloff M, Christou G, Evaluating dose-limiting toxicities of MDM2 inhibitors in patients with solid organ and hematologic malignancies: A systematic review of the literature, Leuk Res 86 (2019) 106222. [DOI] [PubMed] [Google Scholar]
- [219].Munoz-Espin D, Rovira M, Galiana I, Gimenez C, Lozano-Torres B, Paez-Ribes M, Llanos S, Chaib S, Munoz-Martin M, Ucero AC, Garaulet G, Mulero F, Dann SG, VanArsdale T, Shields DJ, Bernardos A, Murguia JR, Martinez-Manez R, Serrano M, A versatile drug delivery system targeting senescent cells, EMBO Mol Med 10(9) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [220].Guerrero A, Guiho R, Herranz N, Uren A, Withers DJ, Martínez-Barbera JP, Tietze LF, Gil J, Galactose-modified duocarmycin prodrugs as senolytics, Aging Cell 19(4) (2020) e13133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [221].Gonzalez-Gualda E, Paez-Ribes M, Lozano-Torres B, Macias D, Wilson JR 3rd, Gonzalez-Lopez C, Ou HL, Miron-Barroso S, Zhang Z, Lerida-Viso A, Blandez JF, Bernardos A, Sancenon F, Rovira M, Fruk L, Martins CP, Serrano M, Doherty GJ, Martinez-Manez R, Munoz-Espin D, Galacto-conjugation of Navitoclax as an efficient strategy to increase senolytic specificity and reduce platelet toxicity, Aging Cell 19(4) (2020) e13142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [222].Besancenot R, Chaligne R, Tonetti C, Pasquier F, Marty C, Lecluse Y, Vainchenker W, Constantinescu SN, Giraudier S, A senescence-like cell-cycle arrest occurs during megakaryocytic maturation: implications for physiological and pathological megakaryocytic proliferation, PLoS Biol 8(9) (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [223].Li C, Chai Y, Wang L, Gao B, Chen H, Gao P, Zhou FQ, Luo X, Crane JL, Yu B, Cao X, Wan M, Programmed cell senescence in skeleton during late puberty, Nat Commun 8(1) (2017) 1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [224].Hall BM, Balan V, Gleiberman AS, Strom E, Krasnov P, Virtuoso LP, Rydkina E, Vujcic S, Balan K, Gitlin II, Leonova KI, Consiglio CR, Gollnick SO, Chernova OB, Gudkov AV, p16(Ink4a) and senescence-associated beta-galactosidase can be induced in macrophages as part of a reversible response to physiological stimuli, Aging (Albany NY) 9(8) (2017) 1867–1884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [225].Grosse L, Wagner N, Emelyanov A, Molina C, Lacas-Gervais S, Wagner KD, Bulavin DV, Defined p16(High) Senescent Cell Types Are Indispensable for Mouse Healthspan, Cell Metab 32(1) (2020) 87–99 e6. [DOI] [PubMed] [Google Scholar]
- [226].Omori S, Wang TW, Johmura Y, Kanai T, Nakano Y, Kido T, Susaki EA, Nakajima T, Shichino S, Ueha S, Ozawa M, Yokote K, Kumamoto S, Nishiyama A, Sakamoto T, Yamaguchi K, Hatakeyama S, Shimizu E, Katayama K, Yamada Y, Yamazaki S, Iwasaki K, Miyoshi C, Funato H, Yanagisawa M, Ueno H, Imoto S, Furukawa Y, Yoshida N, Matsushima K, Ueda HR, Miyajima A, Nakanishi M, Generation of a p16 Reporter Mouse and Its Use to Characterize and Target p16(high) Cells In Vivo, Cell Metab 32(5) (2020) 814–828 e6. [DOI] [PubMed] [Google Scholar]
- [227].Di Micco R, Krizhanovsky V, Baker D, d’Adda di Fagagna F, Cellular senescence in ageing: from mechanisms to therapeutic opportunities, Nat Rev Mol Cell Biol 22(2) (2021) 75–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [228].Xu M, Tchkonia T, Ding H, Ogrodnik M, Lubbers ER, Pirtskhalava T, White TA, Johnson KO, Stout MB, Mezera V, Giorgadze N, Jensen MD, LeBrasseur NK, Kirkland JL, JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age, Proc Natl Acad Sci U S A 112(46) (2015) E6301–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [229].Farr JN, Xu M, Weivoda MM, Monroe DG, Fraser DG, Onken JL, Negley BA, Sfeir JG, Ogrodnik MB, Hachfeld CM, LeBrasseur NK, Drake MT, Pignolo RJ, Pirtskhalava T, Tchkonia T, Oursler MJ, Kirkland JL, Khosla S, Targeting cellular senescence prevents age-related bone loss in mice, Nat Med 23(9) (2017) 1072–1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [230].Moiseeva O, Deschênes-Simard X, St-Germain E, Igelmann S, Huot G, Cadar AE, Bourdeau V, Pollak MN, Ferbeyre G, Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation, Aging Cell 12(3) (2013) 489–98. [DOI] [PubMed] [Google Scholar]
- [231].Mosteiro L, Pantoja C, Alcazar N, Marion RM, Chondronasiou D, Rovira M, Fernandez-Marcos PJ, Munoz-Martin M, Blanco-Aparicio C, Pastor J, Gomez-Lopez G, De Martino A, Blasco MA, Abad M, Serrano M, Tissue damage and senescence provide critical signals for cellular reprogramming in vivo, Science 354(6315) (2016). [DOI] [PubMed] [Google Scholar]
- [232].Zhang B, Fu D, Xu Q, Cong X, Wu C, Zhong X, Ma Y, Lv Z, Chen F, Han L, Qian M, Chin YE, Lam EW, Chiao P, Sun Y, The senescence-associated secretory phenotype is potentiated by feedforward regulatory mechanisms involving Zscan4 and TAK1, Nat Commun 9(1) (2018) 1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [233].Alimbetov D, Davis T, Brook AJ, Cox LS, Faragher RG, Nurgozhin T, Zhumadilov Z, Kipling D, Suppression of the senescence-associated secretory phenotype (SASP) in human fibroblasts using small molecule inhibitors of p38 MAP kinase and MK2, Biogerontology 17(2) (2016) 305–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [234].Aguado J, Chaggar HK, Gomez-Inclan C, Shaker MR, Leeson HC, Mackay-Sim A, Wolvetang EJ, Inhibition of the cGAS-STING pathway ameliorates the premature senescence hallmarks of Ataxia-Telangiectasia brain organoids, Aging Cell 20(9) (2021) e13468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [235].Haag SM, Gulen MF, Reymond L, Gibelin A, Abrami L, Decout A, Heymann M, van der Goot FG, Turcatti G, Behrendt R, Ablasser A, Targeting STING with covalent small-molecule inhibitors, Nature 559(7713) (2018) 269–273. [DOI] [PubMed] [Google Scholar]
- [236].Go S, Kang M, Kwon SP, Jung M, Jeon OH, Kim BS, The Senolytic Drug JQ1 Removes Senescent Cells via Ferroptosis, Tissue Eng Regen Med 18(5) (2021) 841–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [237].Thompson PJ, Shah A, Apostolopolou H, Bhushan A, BET Proteins Are Required for Transcriptional Activation of the Senescent Islet Cell Secretome in Type 1 Diabetes, Int J Mol Sci 20(19) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [238].Wakita M, Takahashi A, Sano O, Loo TM, Imai Y, Narukawa M, Iwata H, Matsudaira T, Kawamoto S, Ohtani N, Yoshimori T, Hara E, A BET family protein degrader provokes senolysis by targeting NHEJ and autophagy in senescent cells, Nat Commun 11(1) (2020) 1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [239].Rider P, Carmi Y, Cohen I, Biologics for Targeting Inflammatory Cytokines, Clinical Uses, and Limitations, Int J Cell Biol 2016 (2016) 9259646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [240].Emery P, Keystone E, Tony HP, Cantagrel A, van Vollenhoven R, Sanchez A, Alecock E, Lee J, Kremer J, IL-6 receptor inhibition with tocilizumab improves treatment outcomes in patients with rheumatoid arthritis refractory to anti-tumour necrosis factor biologicals: results from a 24-week multicentre randomised placebo-controlled trial, Ann Rheum Dis 67(11) (2008) 1516–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [241].van Rhee F, Casper C, Voorhees PM, Fayad LE, Gibson D, Kanhai K, Kurzrock R, Long-term safety of siltuximab in patients with idiopathic multicentric Castleman disease: a prespecified, open-label, extension analysis of two trials, Lancet Haematol 7(3) (2020) e209–e217. [DOI] [PubMed] [Google Scholar]
- [242].Cohen S, Hurd E, Cush J, Schiff M, Weinblatt ME, Moreland LW, Kremer J, Bear MB, Rich WJ, McCabe D, Treatment of rheumatoid arthritis with anakinra, a recombinant human interleukin-1 receptor antagonist, in combination with methotrexate: results of a twenty-four-week, multicenter, randomized, double-blind, placebo-controlled trial, Arthritis Rheum 46(3) (2002) 614–24. [DOI] [PubMed] [Google Scholar]
- [243].Kuemmerle-Deschner JB, Ramos E, Blank N, Roesler J, Felix SD, Jung T, Stricker K, Chakraborty A, Tannenbaum S, Wright AM, Rordorf C, Canakinumab (ACZ885, a fully human IgG1 anti-IL-1beta mAb) induces sustained remission in pediatric patients with cryopyrin-associated periodic syndrome (CAPS), Arthritis Res Ther 13(1) (2011) R34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [244].Hoffman HM, Throne ML, Amar NJ, Sebai M, Kivitz AJ, Kavanaugh A, Weinstein SP, Belomestnov P, Yancopoulos GD, Stahl N, Mellis SJ, Efficacy and safety of rilonacept (interleukin-1 Trap) in patients with cryopyrin-associated periodic syndromes: results from two sequential placebo-controlled studies, Arthritis Rheum 58(8) (2008) 2443–52. [DOI] [PubMed] [Google Scholar]
- [245].Klareskog L, van der Heijde D, de Jager JP, Gough A, Kalden J, Malaise M, Martin Mola E, Pavelka K, Sany J, Settas L, Wajdula J, Pedersen R, Fatenejad S, Sanda M, T.s. investigators, Therapeutic effect of the combination of etanercept and methotrexate compared with each treatment alone in patients with rheumatoid arthritis: double-blind randomised controlled trial, Lancet 363(9410) (2004) 675–81. [DOI] [PubMed] [Google Scholar]
- [246].Jobanputra P, Barton P, Bryan S, Burls A, The effectiveness of infliximab and etanercept for the treatment of rheumatoid arthritis: a systematic review and economic evaluation, Health Technol Assess 6(21) (2002) 1–110. [DOI] [PubMed] [Google Scholar]
- [247].Fizazi K, De Bono JS, Flechon A, Heidenreich A, Voog E, Davis NB, Qi M, Bandekar R, Vermeulen JT, Cornfeld M, Hudes GR, Randomised phase II study of siltuximab (CNTO 328), an anti-IL-6 monoclonal antibody, in combination with mitoxantrone/prednisone versus mitoxantrone/prednisone alone in metastatic castration-resistant prostate cancer, Eur J Cancer 48(1) (2012) 85–93. [DOI] [PubMed] [Google Scholar]
- [248].Dorff TB, Goldman B, Pinski JK, Mack PC, Lara PN Jr., Van Veldhuizen PJ Jr., Quinn DI, Vogelzang NJ, Thompson IM Jr., Hussain MH, Clinical and correlative results of SWOG S0354: a phase II trial of CNTO328 (siltuximab), a monoclonal antibody against interleukin-6, in chemotherapy-pretreated patients with castration-resistant prostate cancer, Clin Cancer Res 16(11) (2010) 3028–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [249].Angevin E, Tabernero J, Elez E, Cohen SJ, Bahleda R, van Laethem JL, Ottensmeier C, Lopez-Martin JA, Clive S, Joly F, Ray-Coquard I, Dirix L, Machiels JP, Steven N, Reddy M, Hall B, Puchalski TA, Bandekar R, van de Velde H, Tromp B, Vermeulen J, Kurzrock R, A phase I/II, multiple-dose, dose-escalation study of siltuximab, an anti-interleukin-6 monoclonal antibody, in patients with advanced solid tumors, Clin Cancer Res 20(8) (2014) 2192–204. [DOI] [PubMed] [Google Scholar]
- [250].Dijkgraaf EM, Santegoets SJ, Reyners AK, Goedemans R, Wouters MC, Kenter GG, van Erkel AR, van Poelgeest MI, Nijman HW, van der Hoeven JJ, Welters MJ, van der Burg SH, Kroep JR, A phase I trial combining carboplatin/doxorubicin with tocilizumab, an anti-IL-6R monoclonal antibody, and interferon-α2b in patients with recurrent epithelial ovarian cancer, Ann Oncol 26(10) (2015) 2141–9. [DOI] [PubMed] [Google Scholar]
- [251].Merritt WM, Lin YG, Spannuth WA, Fletcher MS, Kamat AA, Han LY, Landen CN, Jennings N, De Geest K, Langley RR, Villares G, Sanguino A, Lutgendorf SK, Lopez-Berestein G, Bar-Eli MM, Sood AK, Effect of interleukin-8 gene silencing with liposome-encapsulated small interfering RNA on ovarian cancer cell growth, J Natl Cancer Inst 100(5) (2008) 359–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [252].Schott AF, Goldstein LJ, Cristofanilli M, Ruffini PA, McCanna S, Reuben JM, Perez RP, Kato G, Wicha M, Phase Ib Pilot Study to Evaluate Reparixin in Combination with Weekly Paclitaxel in Patients with HER-2-Negative Metastatic Breast Cancer, Clin Cancer Res 23(18) (2017) 5358–5365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [253].Schalper KA, Carleton M, Zhou M, Chen T, Feng Y, Huang SP, Walsh AM, Baxi V, Pandya D, Baradet T, Locke D, Wu Q, Reilly TP, Phillips P, Nagineni V, Gianino N, Gu J, Zhao H, Perez-Gracia JL, Sanmamed MF, Melero I, Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors, Nat Med 26(5) (2020) 688–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [254].Yuen KC, Liu LF, Gupta V, Madireddi S, Keerthivasan S, Li C, Rishipathak D, Williams P, Kadel EE 3rd, Koeppen H, Chen YJ, Modrusan Z, Grogan JL, Banchereau R, Leng N, Thastrom A, Shen X, Hashimoto K, Tayama D, van der Heijden MS, Rosenberg JE, McDermott DF, Powles T, Hegde PS, Huseni MA, Mariathasan S, High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade, Nat Med 26(5) (2020) 693–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [255].Moore G, Annett S, McClements L, Robson T, Top Notch Targeting Strategies in Cancer: A Detailed Overview of Recent Insights and Current Perspectives, Cells 9(6) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [256].Kim BG, Malek E, Choi SH, Ignatz-Hoover JJ, Driscoll JJ, Novel therapies emerging in oncology to target the TGF-β pathway, J Hematol Oncol 14(1) (2021) 55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [257].Le Naour J, Zitvogel L, Galluzzi L, Vacchelli E, Kroemer G, Trial watch: STING agonists in cancer therapy, Oncoimmunology 9(1) (2020) 1777624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [258].Sagiv A, Burton DG, Moshayev Z, Vadai E, Wensveen F, Ben-Dor S, Golani O, Polic B, Krizhanovsky V, NKG2D ligands mediate immunosurveillance of senescent cells, Aging (Albany NY) 8(2) (2016) 328–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [259].Althubiti M, Lezina L, Carrera S, Jukes-Jones R, Giblett SM, Antonov A, Barlev N, Saldanha GS, Pritchard CA, Cain K, Macip S, Characterization of novel markers of senescence and their prognostic potential in cancer, Cell Death Dis 5 (2014) e1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [260].Chen L, Han X, Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future, J Clin Invest 125(9) (2015) 3384–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [261].Knudsen ES, Kumarasamy V, Chung S, Ruiz A, Vail P, Tzetzo S, Wu J, Nambiar R, Sivinski J, Chauhan SS, Seshadri M, Abrams SI, Wang J, Witkiewicz AK, Targeting dual signalling pathways in concert with immune checkpoints for the treatment of pancreatic cancer, Gut 70(1) (2021) 127–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [262].Andre P, Denis C, Soulas C, Bourbon-Caillet C, Lopez J, Arnoux T, Blery M, Bonnafous C, Gauthier L, Morel A, Rossi B, Remark R, Breso V, Bonnet E, Habif G, Guia S, Lalanne AI, Hoffmann C, Lantz O, Fayette J, Boyer-Chammard A, Zerbib R, Dodion P, Ghadially H, Jure-Kunkel M, Morel Y, Herbst R, Narni-Mancinelli E, Cohen RB, Vivier E, Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells, Cell 175(7) (2018) 1731–1743 e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [263].Ferrari de Andrade L, Tay RE, Pan D, Luoma AM, Ito Y, Badrinath S, Tsoucas D, Franz B, May KF Jr., Harvey CJ, Kobold S, Pyrdol JW, Yoon C, Yuan GC, Hodi FS, Dranoff G, Wucherpfennig KW, Antibody-mediated inhibition of MICA and MICB shedding promotes NK cell-driven tumor immunity, Science (New York, N.Y.) 359(6383) (2018) 1537–1542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [264].Amor C, Feucht J, Leibold J, Ho YJ, Zhu C, Alonso-Curbelo D, Mansilla-Soto J, Boyer JA, Li X, Giavridis T, Kulick A, Houlihan S, Peerschke E, Friedman SL, Ponomarev V, Piersigilli A, Sadelain M, Lowe SW, Senolytic CAR T cells reverse senescence-associated pathologies, Nature 583(7814) (2020) 127–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [265].Kim KM, Noh JH, Bodogai M, Martindale JL, Yang X, Indig FE, Basu SK, Ohnuma K, Morimoto C, Johnson PF, Biragyn A, Abdelmohsen K, Gorospe M, Identification of senescent cell surface targetable protein DPP4, Genes & development 31(15) (2017) 1529–1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [266].Frescas D, Roux CM, Aygun-Sunar S, Gleiberman AS, Krasnov P, Kurnasov OV, Strom E, Virtuoso LP, Wrobel M, Osterman AL, Antoch MP, Mett V, Chernova OB, Gudkov AV, Senescent cells expose and secrete an oxidized form of membrane-bound vimentin as revealed by a natural polyreactive antibody, Proc Natl Acad Sci U S A 114(9) (2017) E1668–E1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [267].Law AMK, Valdes-Mora F, Gallego-Ortega D, Myeloid-Derived Suppressor Cells as a Therapeutic Target for Cancer, Cells 9(3) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [268].Di Mitri D, Toso A, Chen JJ, Sarti M, Pinton S, Jost TR, D’Antuono R, Montani E, Garcia-Escudero R, Guccini I, Da Silva-Alvarez S, Collado M, Eisenberger M, Zhang Z, Catapano C, Grassi F, Alimonti A, Tumour-infiltrating Gr-1+ myeloid cells antagonize senescence in cancer, Nature 515(7525) (2014) 134–7. [DOI] [PubMed] [Google Scholar]
- [269].Bejarano L, Jordao MJC, Joyce JA, Therapeutic Targeting of the Tumor Microenvironment, Cancer Discov 11(4) (2021) 933–959. [DOI] [PubMed] [Google Scholar]
- [270].Di Mitri D, Toso A, Alimonti A, Molecular Pathways: Targeting Tumor-Infiltrating Myeloid-Derived Suppressor Cells for Cancer Therapy, Clin Cancer Res 21(14) (2015) 3108–12. [DOI] [PubMed] [Google Scholar]
- [271].Roy AL, Sierra F, Howcroft K, Singer DS, Sharpless N, Hodes RJ, Wilder EL, Anderson JM, A Blueprint for Characterizing Senescence, Cell 183(5) (2020) 1143–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [272].Marthandan S, Baumgart M, Priebe S, Groth M, Schaer J, Kaether C, Guthke R, Cellerino A, Platzer M, Diekmann S, Hemmerich P, Conserved Senescence Associated Genes and Pathways in Primary Human Fibroblasts Detected by RNA-Seq, PLoS One 11(5) (2016) e0154531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [273].Idda ML, McClusky WG, Lodde V, Munk R, Abdelmohsen K, Rossi M, Gorospe M, Survey of senescent cell markers with age in human tissues, Aging (Albany NY) 12(5) (2020) 4052–4066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [274].Liu JY, Souroullas GP, Diekman BO, Krishnamurthy J, Hall BM, Sorrentino JA, Parker JS, Sessions GA, Gudkov AV, Sharpless NE, Cells exhibiting strong p16 (INK4a) promoter activation in vivo display features of senescence, Proc Natl Acad Sci U S A 116(7) (2019) 2603–2611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [275].Coppe JP, Rodier F, Patil CK, Freund A, Desprez PY, Campisi J, Tumor suppressor and aging biomarker p16(INK4a) induces cellular senescence without the associated inflammatory secretory phenotype, J Biol Chem 286(42) (2011) 36396–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [276].Wang B, Brandenburg S, Hernandez-Segura A, van Vliet T, Jongbloed EM, Wilting SM, Ohtani N, Jager A, Demaria M, CDK4/6 inhibition promotes senescent states with restricted secretory phenotype and reduced toxicity, bioRxiv (2020) 2020.06.05.135715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [277].Kohli J, Wang B, Brandenburg SM, Basisty N, Evangelou K, Varela-Eirin M, Campisi J, Schilling B, Gorgoulis V, Demaria M, Algorithmic assessment of cellular senescence in experimental and clinical specimens, Nat Protoc 16(5) (2021) 2471–2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [278].Andersson A, Bergenstråhle J, Asp M, Bergenstråhle L, Jurek A, Fernández Navarro J, Lundeberg J, Single-cell and spatial transcriptomics enables probabilistic inference of cell type topography, Commun Biol 3(1) (2020) 565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [279].Kalathur M, Toso A, Chen J, Revandkar A, Danzer-Baltzer C, Guccini I, Alajati A, Sarti M, Pinton S, Brambilla L, Di Mitri D, Carbone G, Garcia-Escudero R, Padova A, Magnoni L, Tarditi A, Maccari L, Malusa F, Kalathur RK, A.P. L, Cozza G, Ruzzene M, Delaleu N, Catapano CV, Frew IJ, Alimonti A, A chemogenomic screening identifies CK2 as a target for pro-senescence therapy in PTEN-deficient tumours, Nat Commun 6 (2015) 7227. [DOI] [PubMed] [Google Scholar]
- [280].Gould SE, Junttila MR, de Sauvage FJ, Translational value of mouse models in oncology drug development, Nat Med 21(5) (2015) 431–9. [DOI] [PubMed] [Google Scholar]
- [281].Jenkins RW, Aref AR, Lizotte PH, Ivanova E, Stinson S, Zhou CW, Bowden M, Deng J, Liu H, Miao D, He MX, Walker W, Zhang G, Tian T, Cheng C, Wei Z, Palakurthi S, Bittinger M, Vitzthum H, Kim JW, Merlino A, Quinn M, Venkataramani C, Kaplan JA, Portell A, Gokhale PC, Phillips B, Smart A, Rotem A, Jones RE, Keogh L, Anguiano M, Stapleton L, Jia Z, Barzily-Rokni M, Canadas I, Thai TC, Hammond MR, Vlahos R, Wang ES, Zhang H, Li S, Hanna GJ, Huang W, Hoang MP, Piris A, Eliane JP, Stemmer-Rachamimov AO, Cameron L, Su MJ, Shah P, Izar B, Thakuria M, LeBoeuf NR, Rabinowits G, Gunda V, Parangi S, Cleary JM, Miller BC, Kitajima S, Thummalapalli R, Miao B, Barbie TU, Sivathanu V, Wong J, Richards WG, Bueno R, Yoon CH, Miret J, Herlyn M, Garraway LA, Van Allen EM, Freeman GJ, Kirschmeier PT, Lorch JH, Ott PA, Hodi FS, Flaherty KT, Kamm RD, Boland GM, Wong KK, Dornan D, Paweletz CP, Barbie DA, Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids, Cancer Discov 8(2) (2018) 196–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [282].Yuki K, Cheng N, Nakano M, Kuo CJ, Organoid Models of Tumor Immunology, Trends Immunol 41(8) (2020) 652–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [283].Olson B, Li Y, Lin Y, Liu ET, Patnaik A, Mouse Models for Cancer Immunotherapy Research, Cancer Discov 8(11) (2018) 1358–1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [284].Powley IR, Patel M, Miles G, Pringle H, Howells L, Thomas A, Kettleborough C, Bryans J, Hammonds T, MacFarlane M, Pritchard C, Patient-derived explants (PDEs) as a powerful preclinical platform for anti-cancer drug and biomarker discovery, Br J Cancer 122(6) (2020) 735–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [285].Jennings CJ, Murer B, O’Grady A, Hearn LM, Harvey BJ, Kay EW, Thomas W, Differential p16/INK4A cyclin-dependent kinase inhibitor expression correlates with chemotherapy efficacy in a cohort of 88 malignant pleural mesothelioma patients, Br J Cancer 113(1) (2015) 69–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [286].Bailey C, Black JRM, Reading JL, Litchfield K, Turajlic S, McGranahan N, Jamal-Hanjani M, Swanton C, Tracking Cancer Evolution through the Disease Course, Cancer Discov 11(4) (2021) 916–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [287].Schleich K, Kase J, Dorr JR, Trescher S, Bhattacharya A, Yu Y, Wailes EM, Fan DNY, Lohneis P, Milanovic M, Lau A, Lenze D, Hummel M, Chapuy B, Leser U, Reimann M, Lee S, Schmitt CA, H3K9me3-mediated epigenetic regulation of senescence in mice predicts outcome of lymphoma patients, Nat Commun 11(1) (2020) 3651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [288].Englund DA, Sakamoto AE, Fritsche CM, Heeren AA, Zhang X, Kotajarvi BR, Lecy DR, Yousefzadeh MJ, Schafer MJ, White TA, Atkinson EJ, LeBrasseur NK, Exercise reduces circulating biomarkers of cellular senescence in humans, Aging Cell 20(7) (2021) e13415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [289].Schafer MJ, Zhang X, Kumar A, Atkinson EJ, Zhu Y, Jachim S, Mazula DL, Brown AK, Berning M, Aversa Z, Kotajarvi B, Bruce CJ, Greason KL, Suri RM, Tracy RP, Cummings SR, White TA, LeBrasseur NK, The senescence-associated secretome as an indicator of age and medical risk, JCI Insight 5(12) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [290].Lozano-Torres B, Galiana I, Rovira M, Garrido E, Chaib S, Bernardos A, Munoz-Espin D, Serrano M, Martinez-Manez R, Sancenon F, An OFF-ON Two-Photon Fluorescent Probe for Tracking Cell Senescence in Vivo, J Am Chem Soc 139(26) (2017) 8808–8811. [DOI] [PubMed] [Google Scholar]
- [291].Krueger MA, Cotton JM, Zhou B, Wolter K, Schwenck J, Kuehn A, Fuchs K, Maurer A, Fougere CL, Zender L, Pichler BJ, Abstract 1146: [18F]FPyGal: A novel ß-galactosidase specific PET tracer for in vivo imaging of tumor senescence, Cancer Research 79(13 Supplement) (2019) 1146–1146. [Google Scholar]