Cancer Response to Therapy-Induced Senescence: A Matter of Dose and Timing

Simple Summary Cellular senescence consists of a permanent block of cell proliferation in the presence of an active metabolism. It is a physiological process occurring when cells exhaust their proliferative potential, as signaled by critical telomere erosion. Additionally, cell senescence might be triggered as a response to different stresses: DNA damage, oxidative stress and oncogenic activation. Whatever the senescence-inducing stress is, a peculiarity of senescent cells is the production of an altered secretome, the Senescence-Associated Secretory Phenotype (SASP), which profoundly affects the cellular microenvironment. Cancer therapy, either ionizing radiations or chemotherapy, induces cellular senescence, the so-called therapy-induced senescence (TIS). The issue of whether TIS is a pro- or anti-tumorigenic process is a current and open question. Abstract Cellular senescence participates to fundamental processes like tissue remodeling in embryo development, wound healing and inhibition of preneoplastic cell growth. Most senescent cells display common hallmarks, among which the most characteristic is a permanent (or long lasting) arrest of cell division. However, upon senescence, different cell types acquire distinct phenotypes, which also depend on the specific inducing stimuli. Senescent cells are metabolically active and secrete a collection of growth factors, cytokines, proteases, and matrix-remodeling proteins collectively defined as senescence-associated secretory phenotype, SASP. Through SASP, senescent cells modify their microenvironment and engage in a dynamic dialog with neighbor cells. Senescence of neoplastic cells, at least temporarily, reduces tumor expansion, but SASP of senescent cancer cells as well as SASP of senescent stromal cells in the tumor microenvironment may promote the growth of more aggressive cancer subclones. Here, we will review recent data on the mechanisms and the consequences of cancer-therapy induced senescence, enlightening the potentiality and the risk of senescence inducing treatments.

[1]  Margarita V. Meer,et al.  Reprogramming to recover youthful epigenetic information and restore vision , 2020, Nature.

[2]  Yoko Ito,et al.  Transcription-dependent cohesin repositioning rewires chromatin loops in cellular senescence , 2020, Nature Communications.

[3]  I. Cheeseman,et al.  Cellular Mechanisms and Regulation of Quiescence. , 2020, Developmental cell.

[4]  Elizabeth L. Wilder,et al.  A Blueprint for Characterizing Senescence , 2020, Cell.

[5]  Frédérick A. Mallette,et al.  Non‐canonical ATM/MRN activities temporally define the senescence secretory program , 2020, EMBO reports.

[6]  Jae-Seon Lee,et al.  Endothelial cells under therapy-induced senescence secrete CXCL11, which increases aggressiveness of breast cancer cells. , 2020, Cancer letters.

[7]  M. Velarde,et al.  Cellular Senescence Promotes Skin Carcinogenesis through p38MAPK and p44/42MAPK Signaling , 2020, Cancer Research.

[8]  J. Shay,et al.  Dysfunctional telomeres trigger cellular senescence mediated by cyclic GMP-AMP synthase , 2020, The Journal of Biological Chemistry.

[9]  Verena Wagner,et al.  Senescence as a therapeutically relevant response to CDK4/6 inhibitors , 2020, Oncogene.

[10]  V. Hornung,et al.  Molecular mechanisms and cellular functions of cGAS–STING signalling , 2020, Nature Reviews Molecular Cell Biology.

[11]  Amanda R. Kulick,et al.  Senolytic CAR T cells reverse senescence-associated pathologies , 2020, Nature.

[12]  Amanda R. Kulick,et al.  Senescence-Induced Vascular Remodeling Creates Therapeutic Vulnerabilities in Pancreas Cancer , 2020, Cell.

[13]  O. Chinot,et al.  Bevacizumab (Avastin®) in cancer treatment: A review of 15 years of clinical experience and future outlook. , 2020, Cancer treatment reviews.

[14]  E. Lam,et al.  Senescent Stromal Cells Promote Cancer Resistance through SIRT1 Loss-Potentiated Overproduction of Small Extracellular Vesicles , 2020, Cancer Research.

[15]  P. Robbins,et al.  ATM is a key driver of NF-κB-dependent DNA-damage-induced senescence, stem cell dysfunction and aging , 2020, Aging.

[16]  Maria Patrizia Mongiardi,et al.  Glioblastoma Cells Do Not Affect Axitinib-Dependent Senescence of HUVECs in a Transwell Coculture Model , 2020, International journal of molecular sciences.

[17]  C. Brancolini,et al.  The Histone Code of Senescence , 2020, Cells.

[18]  G. Doherty,et al.  Targeting senescent cells in translational medicine , 2019, EMBO molecular medicine.

[19]  Maria Patrizia Mongiardi,et al.  Gene expression profiling of hypoxic response in different models of senescent endothelial cells , 2019, Aging Clinical and Experimental Research.

[20]  C. Schmitt,et al.  Cellular Senescence: Defining a Path Forward , 2019, Cell.

[21]  P. Sung,et al.  Chromatin‐bound cGAS is an inhibitor of DNA repair and hence accelerates genome destabilization and cell death , 2019, The EMBO journal.

[22]  P. Kapahi,et al.  From discoveries in ageing research to therapeutics for healthy ageing , 2019, Nature.

[23]  Kotb Abdelmohsen,et al.  Transcriptome signature of cellular senescence. , 2019, Nucleic acids research.

[24]  S. Stewart,et al.  Unmasking senescence: context-dependent effects of SASP in cancer , 2019, Nature Reviews Cancer.

[25]  N. Nishimura,et al.  Endothelial cells promote 3D invasion of GBM by IL-8-dependent induction of cancer stem cell properties , 2019, Scientific Reports.

[26]  J. V. van Deursen Senolytic therapies for healthy longevity , 2019, Science.

[27]  P. J. Kranzusch,et al.  Modular Architecture of the STING C-Terminal Tail Allows Interferon and NF-κB Signaling Adaptation , 2019, Cell reports.

[28]  D. Gewirtz,et al.  Tumor Cell Escape from Therapy-Induced Senescence as a Model of Disease Recurrence after Dormancy. , 2019, Cancer research.

[29]  J. Shay,et al.  Telomeres and telomerase: three decades of progress , 2019, Nature Reviews Genetics.

[30]  Soyoung Lee,et al.  The dynamic nature of senescence in cancer , 2019, Nature Cell Biology.

[31]  L. Ricci-Vitiani,et al.  Glioblastoma endothelium drives bevacizumab‐induced infiltrative growth via modulation of PLXDC1 , 2018, International journal of cancer.

[32]  David T. W. Jones,et al.  The Senescence-associated Secretory Phenotype Mediates Oncogene-induced Senescence in Pediatric Pilocytic Astrocytoma , 2018, Clinical Cancer Research.

[33]  M. Ohba,et al.  Receptor Tyrosine Kinase-Targeted Cancer Therapy , 2018, International journal of molecular sciences.

[34]  A. Aerts,et al.  Pathological effects of ionizing radiation: endothelial activation and dysfunction , 2018, Cellular and Molecular Life Sciences.

[35]  S. Kaufmann,et al.  Nuclear cGAS suppresses DNA repair and promotes tumorigenesis , 2018, Nature.

[36]  Stephen M. Douglass,et al.  Age-Related Changes in HAPLN1 Increase Lymphatic Permeability and Affect Routes of Melanoma Metastasis. , 2018, Cancer discovery.

[37]  G. Chen,et al.  ID1-induced p16/IL6 axis activation contributes to the resistant of hepatocellular carcinoma cells to sorafenib , 2018, Cell Death & Disease.

[38]  C. Mullighan,et al.  Drugging DNA repair to target T-ALL cells , 2018, Leukemia & lymphoma.

[39]  L. Ricci-Vitiani,et al.  Endothelial trans-differentiation in glioblastoma recurring after radiotherapy , 2018, Modern Pathology.

[40]  J. Duyster,et al.  Anti-Angiogenics: Current Situation and Future Perspectives , 2018, Oncology Research and Treatment.

[41]  E. Passegué,et al.  Lysosome activation clears aggregates and enhances quiescent neural stem cell activation during aging , 2018, Science.

[42]  C. Lovly,et al.  Mechanisms of receptor tyrosine kinase activation in cancer , 2018, Molecular Cancer.

[43]  Frédérick A. Mallette,et al.  Targeting the Senescence-Overriding Cooperative Activity of Structurally Unrelated H3K9 Demethylases in Melanoma. , 2018, Cancer cell.

[44]  Soyoung Lee,et al.  Senescence-associated reprogramming promotes cancer stemness , 2017, Nature.

[45]  Yoko Ito,et al.  Spatial and Temporal Control of Senescence. , 2017, Trends in cell biology.

[46]  K. Kaestner,et al.  Cytoplasmic chromatin triggers inflammation in senescence and cancer , 2017, Nature.

[47]  Sun-Young Han,et al.  The Role of Kinase Modulators in Cellular Senescence for Use in Cancer Treatment , 2017, Molecules.

[48]  L. Zender,et al.  Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence , 2017, Nature Cell Biology.

[49]  Zhijian J. Chen,et al.  cGAS is essential for cellular senescence , 2017, Proceedings of the National Academy of Sciences.

[50]  M. Lieber,et al.  Non-homologous DNA end joining and alternative pathways to double-strand break repair , 2017, Nature Reviews Molecular Cell Biology.

[51]  D. Aebersold,et al.  Senescence as biologic endpoint following pharmacological targeting of receptor tyrosine kinases in cancer , 2017, Biochemical pharmacology.

[52]  B. Kennedy,et al.  Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse. , 2017, Cancer discovery.

[53]  L. Zender,et al.  The senescence-associated secretory phenotype induces cellular plasticity and tissue regeneration , 2017, Genes & development.

[54]  G. Bernardini,et al.  Axitinib induces senescence-associated cell death and necrosis in glioma cell lines: The proteasome inhibitor, bortezomib, potentiates axitinib-induced cytotoxicity in a p21(Waf/Cip1) dependent manner , 2016, Oncotarget.

[55]  G. Semenza,et al.  Hypoxia-Inducible Factors: Master Regulators of Cancer Progression. , 2016, Trends in cancer.

[56]  Ji Min Park,et al.  Radiosensitizing effect of lapatinib in human epidermal growth factor receptor 2-positive breast cancer cells , 2016, Oncotarget.

[57]  S. Razin,et al.  Small molecule compounds that induce cellular senescence , 2016, Aging cell.

[58]  Masashi Narita,et al.  NOTCH1 mediates a switch between two distinct secretomes during senescence , 2016, Nature Cell Biology.

[59]  J. Campisi,et al.  Context-dependent effects of cellular senescence in cancer development , 2016, British Journal of Cancer.

[60]  A. Pezeshki,et al.  Naturally occurring p16Ink4a-positive cells shorten healthy lifespan , 2016, Nature.

[61]  M. Velarde,et al.  Mitochondrial Dysfunction Induces Senescence with a Distinct Secretory Phenotype. , 2016, Cell metabolism.

[62]  Jing Wang,et al.  Dasatinib induces DNA damage and activates DNA repair pathways leading to senescence in non-small cell lung cancer cell lines with kinase-inactivating BRAF mutations , 2015, Oncotarget.

[63]  G. Santoni,et al.  Axitinib induces DNA damage response leading to senescence, mitotic catastrophe, and increased NK cell recognition in human renal carcinoma cells , 2015, Oncotarget.

[64]  J. Arribas,et al.  Effect of cellular senescence on the growth of HER2-positive breast cancers. , 2015, Journal of the National Cancer Institute.

[65]  C. Abbadie,et al.  The unfolded protein response and cellular senescence. A review in the theme: cellular mechanisms of endoplasmic reticulum stress signaling in health and disease. , 2015, American journal of physiology. Cell physiology.

[66]  J. Hoeijmakers,et al.  An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. , 2014, Developmental cell.

[67]  R. Chen,et al.  EGF promotes mammalian cell growth by suppressing cellular senescence , 2014, Cell Research.

[68]  K. Aldape,et al.  A randomized trial of bevacizumab for newly diagnosed glioblastoma. , 2014, The New England journal of medicine.

[69]  Jimin Shin,et al.  Depletion of ERK2 but not ERK1 abrogates oncogenic Ras-induced senescence. , 2013, Cellular signalling.

[70]  A. Mercurio,et al.  VEGF targets the tumour cell , 2013, Nature Reviews Cancer.

[71]  A. Rodríguez-Baeza,et al.  Programmed Cell Senescence during Mammalian Embryonic Development , 2013, Cell.

[72]  Soyoung Lee,et al.  Synthetic lethal metabolic targeting of cellular senescence in cancer therapy , 2013, Nature.

[73]  Le Xu,et al.  Sunitinib induces cellular senescence via p53/Dec1 activation in renal cell carcinoma cells , 2013, Cancer science.

[74]  Kelly J. Morris,et al.  A complex secretory program orchestrated by the inflammasome controls paracrine senescence , 2013, Nature Cell Biology.

[75]  S. Haferkamp,et al.  Vemurafenib induces senescence features in melanoma cells. , 2013, The Journal of investigative dermatology.

[76]  T. Shlomi,et al.  A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence , 2013, Nature.

[77]  Matthew J. Brauer,et al.  A high-content cellular senescence screen identifies candidate tumor suppressors, including EPHA3 , 2013, Cell cycle.

[78]  Hiroshi Mitani,et al.  Unrepairable DNA double-strand breaks that are generated by ionising radiation determine the fate of normal human cells , 2012, Journal of Cell Science.

[79]  M. Priault,et al.  Apoptosis and autophagy have opposite roles on imatinib-induced K562 leukemia cell senescence , 2012, Cell Death and Disease.

[80]  P. Muti,et al.  SASP mediates chemoresistance and tumor-initiating-activity of mesothelioma cells , 2012, Oncogene.

[81]  F. d’Adda di Fagagna,et al.  Is cellular senescence an example of antagonistic pleiotropy? , 2012, Aging cell.

[82]  C. Woodworth,et al.  Inhibition of the epidermal growth factor receptor by erlotinib prevents immortalization of human cervical cells by Human Papillomavirus type 16. , 2011, Virology.

[83]  N. LeBrasseur,et al.  Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders , 2011, Nature.

[84]  D. Owen,et al.  Inhibition of VEGF induces cellular senescence in colorectal cancer cells , 2011, International journal of cancer.

[85]  F. DiMeco,et al.  Endothelial cells create a stem cell niche in glioblastoma by providing NOTCH ligands that nurture self-renewal of cancer stem-like cells. , 2011, Cancer research.

[86]  B. Kim,et al.  PTEN status switches cell fate between premature senescence and apoptosis in glioma exposed to ionizing radiation , 2011, Cell Death and Differentiation.

[87]  D. Peeper,et al.  The essence of senescence. , 2010, Genes & development.

[88]  David F Jarrard,et al.  Therapy-induced senescence in cancer. , 2010, Journal of the National Cancer Institute.

[89]  H. Hsu,et al.  Depletion of securin induces senescence after irradiation and enhances radiosensitivity in human cancer cells regardless of functional p53 expression. , 2010, International journal of radiation oncology, biology, physics.

[90]  M. Karin,et al.  Immunity, Inflammation, and Cancer , 2010, Cell.

[91]  S. Lowe,et al.  TAp63 induces senescence and suppresses tumorigenesis in vivo , 2009, Nature Cell Biology.

[92]  Judith Campisi,et al.  Senescence-Associated Secretory Phenotypes Reveal Cell-Nonautonomous Functions of Oncogenic RAS and the p53 Tumor Suppressor , 2008, PLoS biology.

[93]  Jonathan Melamed,et al.  Chemokine Signaling via the CXCR2 Receptor Reinforces Senescence , 2008, Cell.

[94]  D. Peeper,et al.  Oncogene-Induced Senescence Relayed by an Interleukin-Dependent Inflammatory Network , 2008, Cell.

[95]  S. Signoretti,et al.  VHL loss actuates a HIF-independent senescence programme mediated by Rb and p400 , 2008, Nature Cell Biology.

[96]  Sandy Chang,et al.  Dysfunctional telomeres activate an ATM‐ATR‐dependent DNA damage response to suppress tumorigenesis , 2007, The EMBO journal.

[97]  T. Lange,et al.  Protection of telomeres through independent control of ATM and ATR by TRF2 and POT1 , 2007, Nature.

[98]  B. A. Ballif,et al.  ATM and ATR Substrate Analysis Reveals Extensive Protein Networks Responsive to DNA Damage , 2007, Science.

[99]  Carlos Cordon-Cardo,et al.  Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas , 2007, Nature.

[100]  K. Kiura,et al.  Gefitinib induces premature senescence in non-small cell lung cancer cells with or without EGFR gene mutation. , 2007, Oncology reports.

[101]  Aaron Bensimon,et al.  Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication , 2006, Nature.

[102]  D. Guernsey,et al.  Neosis—a paradigm of self‐renewal in cancer , 2005, Cell biology international.

[103]  J. Shay,et al.  BRAFE600-associated senescence-like cell cycle arrest of human naevi , 2005, Nature.

[104]  M. Barbacid,et al.  Tumour biology: Senescence in premalignant tumours , 2005, Nature.

[105]  L. Elmore,et al.  p53-Dependent accelerated senescence induced by ionizing radiation in breast tumour cells , 2005, International journal of radiation biology.

[106]  Szu-Yu Chen,et al.  Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers. , 2005, Cancer research.

[107]  L. Elmore,et al.  Evasion of a Single-Step, Chemotherapy-Induced Senescence in Breast Cancer Cells: Implications for Treatment Response , 2005, Clinical Cancer Research.

[108]  Kenneth J. Hillan,et al.  Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer , 2004, Nature Reviews Drug Discovery.

[109]  T. Lange,et al.  DNA Damage Foci at Dysfunctional Telomeres , 2003, Current Biology.

[110]  Masashi Narita,et al.  Reversal of human cellular senescence: roles of the p53 and p16 pathways , 2003, The EMBO journal.

[111]  I. Roninson,et al.  Tumor cell senescence in cancer treatment. , 2003, Cancer research.

[112]  Soyoung Lee,et al.  A Senescence Program Controlled by p53 and p16INK4a Contributes to the Outcome of Cancer Therapy , 2002, Cell.

[113]  S. Joel,et al.  DNA damage is able to induce senescence in tumor cells in vitro and in vivo. , 2002, Cancer research.

[114]  J. Campisi,et al.  Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[115]  Michael R. Speicher,et al.  Securin Is Required for Chromosomal Stability in Human Cells , 2001, Cell.

[116]  E. Kandel,et al.  A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents. , 1999, Cancer research.

[117]  J. Sham,et al.  Evidence of cisplatin-induced senescent-like growth arrest in nasopharyngeal carcinoma cells. , 1998, Cancer research.

[118]  S. Lowe,et al.  Oncogenic ras Provokes Premature Cell Senescence Associated with Accumulation of p53 and p16INK4a , 1997, Cell.

[119]  D. Hanahan,et al.  Patterns and Emerging Mechanisms of the Angiogenic Switch during Tumorigenesis , 1996, Cell.

[120]  C. Harley,et al.  Telomeres shorten during ageing of human fibroblasts , 1990, Nature.

[121]  J. Folkman Tumor angiogenesis: therapeutic implications. , 1971, The New England journal of medicine.

[122]  R. Pallini,et al.  Axitinib exposure triggers endothelial cells senescence through ROS accumulation and ATM activation , 2019, Oncogene.

[123]  Y. Shiloh,et al.  The ATM protein kinase: regulating the cellular response to genotoxic stress, and more. , 2013, Nature reviews. Molecular cell biology.

[124]  I. Bayazitov,et al.  A perivascular niche for brain tumor stem cells. , 2007, Cancer cell.

[125]  A. Bikfalvi,et al.  Tumor angiogenesis , 2020, Advances in cancer research.