Open Access

Cancer‑associated fibroblasts under therapy‑induced senescence in the tumor microenvironment (Review)

  • Authors:
    • Qiuhua Zhang
    • Yijie Lou
    • Hao Fang
    • Shaopeng Sun
    • Rijuan Jin
    • Yunxi Ji
    • Zhe Chen
  • View Affiliations

  • Published online on: February 20, 2024     https://doi.org/10.3892/etm.2024.12438
  • Article Number: 150
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Current cancer treatments target tumor cells; however, the tumor microenvironment (TME) induces therapeutic resistance, tumor development and metastasis, thus rendering these treatments ineffective. Research on the TME has therefore concentrated on nonmalignant cells. Cancer‑associated fibroblasts (CAFs) are a major TME component, which contribute to cancer progression due to their diverse origins, phenotypes and functions, including cancer cell invasion and migration, extracellular matrix remodeling, tumor metabolism modulation and therapeutic resistance. Standard cancer treatment typically exacerbates the senescence‑associated secretory phenotype (SASP) of senescent cancer cells and nonmalignant cells that actively leak proinflammatory signals in the TME. Therapy‑induced senescence may impair cancer cell activity and compromise treatment responsiveness. CAFs and SASP are well‑studied in the formation and progression of cancer. The present review discusses the current data on CAF senescence caused by anticancer treatment and assesses how senescence‑like CAFs affect tumor formation. The development of senolytic medication for aging stromal cells is also highlighted. Combining cancer therapies with senolytics may boost therapeutic effects and provide novel possibilities for research.

1. Introduction

The tumor microenvironment (TME), which consists primarily of the extracellular matrix (ECM), is crucial to various aspects of tumor progression, such as tumorigenesis, metastasis, relapse and treatment resistance, making it a potential target for cancer therapies (1,2). The TME is a complex system comprising a range of cellular and noncellular elements, and cancer-associated fibroblasts (CAFs), which are highly abundant in the tumor stroma and exhibit potent regulatory effects on tumor growth (3,4). The induction of cell senescence through traditional cancer treatments is one of the most notable mechanisms of tumor suppression. However, in cancer, the process of therapy-induced senescence (TIS) is a ‘double-edged sword’ (5). Despite its indispensable role in combating tumor growth by halting cancer cell division, the chronic accumulation of senescent cells can conceivably promote tumor development (6). Specifically, secretion of the senescence-associated secretory phenotype (SASP) (7), which is induced by TIS, can significantly affect the TME through autocrine and paracrine effects (8).

The aim of the present review was to provide an overview of the most recent research regarding the biological tumor-promoting functions of CAFs. Additionally, this review aimed to emphasize the antitumorigenic properties of senescence-like CAFs within the TME, which are induced by anticancer therapies. Furthermore, a comprehensive overview of the existing senolytic therapies, and their potential advantages in the context of cancer treatment, is provided.

2. Origins, activation and markers of CAFs

Origin of CAFs

Previous studies have highlighted the complexity and diversity of CAFs, one of the diverse components of the tumor stroma, which could be due to their multiple cellular origins and the emergence of various CAF subpopulations (9,10). While the exact origin of CAFs remains unclear, they can be produced through various means. During tumorigenesis, remnant local fibroblasts in the surrounding tissues can undergo gene expression and phenotype changes in response to tumor driver stimuli. For example, ‘quiescent’ stellate cells in the liver and pancreas, which are the resident fibroblasts in these tissues, can be activated by inflammatory stimuli released by tumors to acquire a myofibroblast-like CAF phenotype (11-13). In addition, CAFs can be recruited from remotely circulating cell populations. One such precursor of CAFs is bone marrow mesenchymal stem cells, which is the most well-studied cell population among the known sources of CAFs (14,15). Moreover, nonfibroblastic lineages that are in close proximity to tumor cells, such as endothelial or epithelial cells, can undergo transdifferentiation into CAFs through endothelial-mesenchymal or epithelial-mesenchymal transition (EMT), respectively. These cells exhibit CAF-like gene expression and represent another source of heterogeneity for the population of CAFs (16,17). Furthermore, transdifferentiation from other uncommon CAF precursor cells, such as adipocytes and pericytes, has been observed under limited conditions (18,19) (Fig. 1).

Activation of CAFs

Cancer progression relies heavily on tumor stromal support to maintain continuous tumor growth and metastasis (20,21). Despite the presence of normal fibroblasts in the matrix surrounding the tumor, they do not hinder cancer cell invasion and metastasis (22). Hence, it becomes essential for cancer cells to transform normal fibroblasts into tumor-promoting CAFs, which are recruited and activated by various tumor-derived signals and specific stimuli in the TME, such as hypoxia and oxidative stress damage (23,24). Among these, transforming growth factor β (TGF-β), platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) play crucial roles as biochemical mediators in CAFs (25). It has been indicated that TGF-β has a strong interaction with ECM components in tissues and is an essential cytokine that activates the cancer stroma (26). TGF-β is typically activated locally, and as a result, CAFs produce TGF-β locally in response to its secretion by cancer cells in the surrounding area. This triggers a response in CAFs, enhancing their potential to produce tumor-promoting factors (27,28). PDGF is another growth factor secreted by tumor cells that is closely linked to cancer progression (29). Cellular processes, such as chemotaxis, cell proliferation, division and angiogenesis, are some of the numerous biological processes affected by PDGF, and overexpression of PDGF and its receptors in tumors is a common occurrence. However, most tumor cells do not express PDGF receptors. Rather, PDGF levels are upregulated, implying that PDGF mainly promotes tumor progression through paracrine signaling from other cells, such as fibroblasts and endothelial cells (30,31). Unlike the TGF-β-induced ECM over-deposition phenotype, PDGF mainly enhances fibroblast recruitment and proliferation (32). FGF2 was initially known as a ‘basic fibroblast growth factor’ that promotes cell differentiation and proliferation between epithelial and mesenchymal cells through autocrine and paracrine processes. Further research has revealed that FGF2 stimulates the activation of normal fibroblasts in vivo, conferring their ability to promote metastasis (33). Furthermore, FGF2 can act synergistically with PDGF to promote tumor angiogenesis and metastasis (34).

Markers of CAFs

CAF populations are highly heterogeneous, with differences across various tumor types and cell sources. However, unlike other cell lineages, CAFs do not express specific biomarkers, posing a challenge to their comprehensive characterization. Representative CAF markers have been established, including fibroblast activation protein, PDGF receptor-α/β, α-smooth muscle actin, fibroblast-specific protein 1/S100A4, vimentin, podoplanin, periostin and type-I collagen (35,36). Nevertheless, while these markers are commonly used to identify CAF phenotypic features, none of them offer specificity to CAFs, and the expression of these markers can also be observed in healthy tissues and other cell types. Table I (37-54) lists some typical CAF markers used in cancer studies.

Table I

Commonly used markers of CAFs.

Table I

Commonly used markers of CAFs.

MarkerDescriptionExpression in other cells(Refs.)
FAPA 170-kDa melanoma membrane-bound gelatinase, serine integral membrane proteaseMacrophages(37,38)
PDGFR-α/βCell-surface receptor for homodimeric PDGFB and PDGFD, and for heterodimers formed by PDGFA and PDGFB, tyrosine-protein kinaseVascular smooth muscle cells, pericytes(39-42)
α-SMA Intermediate-filament-associated protein, most highly conserved globular proteinSmooth muscle cells, pericytes, myoepithelial cells(43-44)
FSP1/S100A4A member of the calcium-binding protein family, a marker of quiescent fibroblastsCancer cells(45)
VimentinClass-III intermediate filament protein, widely expressed in fibroblastsEndothelial cells, myoepithelial cells and neurons(46-48)
PDPNMembrane glycoprotein, extensively O-glycosylatedEndothelial cells(49)
Tenascin CExtracellular matrix protein, a marker of myofibroblastsCancer cells, endothelial cells(50,51)
POSTNExtracellular matrix protein, associated with cancer stem cell maintenance and metastasisCancer cells, mesenchymal stem cells(52,53)
COL1A member of group I collagen (fibrillar forming collagen), not exclusive to fibroblastsCancer cells, endothelial cells(54)

[i] α-SMA, α-smooth muscle actin; COL, type-I collagen; FAP, fibroblast activation protein; PDGFR, platelet-derived growth factor receptor; FSP1, fibroblast-specific protein 1; PDPN, podoplanin; POSTN, periostin.

3. The function of CAFs in metastasis

Tumor metastasis is frequently associated with an unfavorable prognosis for patients (55). CAFs are known to be important in tumor development and resistance to cancer therapy; therefore, the functions of CAFs should be closely examined in relation to tumor metastasis. The modulatory effects of CAFs on tumor progression have been well documented in the TME, where they promote tumor progression and metastasis through various mechanisms, including ECM modification, EMT regulation in cancer cells and the release of cytokines that support tumor growth.

CAF-mediated ECM remodeling

ECM remodeling is a vital physiological process for maintaining tissue homeostasis that occurs throughout body development, regeneration and wound healing (56,57). Fibroblasts, found in both healthy and tumor tissues, make a critical difference in producing ECM components and remodeling enzymes in the interstitial space. Tumor-derived factors activate fibroblasts, transforming them into CAFs, which facilitate communication between the tumor and its surrounding stroma. CAFs are responsible for ECM remodeling in distant organs, promoting tumor invasion and metastasis, and can create pathways within the ECM by degrading matrix proteins, allowing cancer cells to penetrate the basement membrane and enter the surrounding tissues, ultimately metastasizing to distant locations (58). This ECM remodeling process, facilitated by ECM-degrading enzymes such as rho-associated kinase and matrix metalloproteinases, serves an important role in driving malignant cell invasion and metastasis (59-61).

Role of CAF in EMT and migration

In both embryogenesis and tumor metastasis, the involvement of fibroblasts in promoting EMT is crucial. Generally, cells with a mesenchymal phenotype are more prone to invasion (62). However, carcinomas typically retain the epithelial characteristics that limit their invasive potential. Therefore, cancer cells rely on nonmalignant stromal cell types, with CAFs being the most ideal matrix partners, to facilitate invasion and metastasis. Hybrid EMT, in which malignant cells exhibit a combination of mesenchymal and epithelial characteristics, strongly promotes metastasis (17,63-65). The interaction between malignant cells and CAFs is mediated by cytokines, which is vital for EMT. In recent years, extensive research has been conducted on the CAF-secreted factors involved in EMT in various types of cancer. Among these factors, TGF-β, released by CAFs, has been extensively studied. In advanced cancer, TGF-β enhances the migratory and invasive capabilities of cancer cells by inducing a mesenchymal phenotype. For example, CAFs can activate homeobox transcript antisense intergenic RNA (HOTAIR) transcription through the secretion of TGF-β1, and SMAD2/3/4 can directly bind to the HOTAIR promoter site to increase the ability of breast cancer cells to metastasize (66). Other signaling pathways that drive cancer cells to acquire mesenchymal phenotypes, such as Janus kinase/signal transducer and activator of transcription proteins (JAK/STAT), Wnt/β-catenin, MAPK and PI3K/Akt, can be triggered by CAF-released cytokines, growth factors and chemokines, such as interleukin-6(67), hepatocyte growth factor (68) and stromal cell-derived factor-1(69).

Role of CAFs-derived exosomes in cancer progression

As previously mentioned, activated CAFs secrete various messengers that can induce remodeling of the ECM or EMT in malignant cells to facilitate tumor development and progression. In the present section, the ways in which CAF-derived exosomes promote metastasis was focused on. Exosomes, a type of extracellular vesicle, have emerged as novel messengers in intercellular communication; notably, they deliver proteins, DNA and RNA (70). Several studies have identified exosomes secreted by CAFs as key players in the crosstalk between tumors and CAFs, as well as in cancer cell invasion. For example, in gemcitabine-treated human pancreatic ductal adenocarcinoma CAFs, the expression levels of Snail and microRNA (miR)-146a were revealed to be increased in exosomes, and this enhanced cell proliferation and chemoresistance (71). In gastric carcinoma, exosomal miR-522 derived from CAFs was shown to inhibit ferroptosis by suppressing arachidonate lipoxygenase 15 and reducing lipid reactive oxygen species accumulation, resulting in chemoresistance and tumor progression (72) (Fig. 2).

4. Senescence-like CAFs in tumor progression

Cellular senescence

Cellular aging is a response to cellular stress caused by molecular damage. Common triggers of senescence include replicative exhaustion (replicative senescence), hyperactivation of oncogenes (oncogene-induced senescence), and persistent damage to DNA and chromatin structures (73). Cellular senescence is regarded as a vital intrinsic tumor-suppressing mechanism. Various anticancer therapies induce senescence in malignant cells by inducing genotoxic stress, overactivating mitotic signals or inducing oxidative stress; this blocks the growth of tumor cells and promotes immune cell infiltration (74). However, some patients experience relapse, metastasis and/or therapeutic resistance. The TME is a complex biological system consisting of tumor cells and numerous nontumor components. These nontumor components, such as the ECM, fibroblasts and immune cells, can have profound effects on tumor progression during aging (75,76). This review focuses on the impact of aging of CAFs on tumor progression.

CAFs and senescence

Tumor cells can induce the aging of stromal cells through cytokine secretion. It has been reported that the specific expression of matrix metalloproteinase 1 in large-cell carcinoma can induce fibroblast senescence and promote the progression of lung cancer (76). In addition, the gut microbiota can induce aging of hepatic stellate cells through the enterohepatic circulation of metabolites, leading to the development of liver cancer (77-79). Cancer cells cease to proliferate after TIS; however, senescent cells still exhibit metabolic activity and undergo changes in their secretory proteomes. They secrete proinflammatory factors, growth factors and proteases, collectively referred to as SASP (80). The accumulation of therapy-induced senescent cells leads to chronic inflammation and immunosuppression, which can have long-term adverse effects (74). Since most anticancer treatments are administered systemically, they become problematic when nonmalignant cells in nontumor areas undergo senescence in response to these therapies (81). The present review aims to concentrate on recent studies that refer to the induction of senescence in CAFs by antitumor therapies, such as chemotherapy, radiotherapy (RT) and targeted therapies.

Cancer therapies and the induction of senescence. CAFs and chemotherapy

Chemotherapy and RT are conventional and widely utilized treatments for cancer. High doses of chemotherapy or RT can effectively induce the apoptosis of cancer cells; however, they also pose a risk of damaging the surrounding tissues, causing serious side effects in patients (74). Consequently, an alternative therapeutic strategy is to induce senescence in malignant cells to permanently halt their ability to proliferate without triggering apoptosis. An extensively studied mechanism of cellular aging is activation of the DNA damage response (DDR) (7). The DDR is closely associated with the relapse and development of tumors, and it also provides therapeutic opportunities for tumor treatment (82). When cells are exposed to various endogenous and exogenous stressors (such as replication stress, ultraviolet, drugs and ionization radiation), DDR-associated proteases (such as ATM, ATR, CHK1 and PARP) are activated, which eventually results in senescence (83). Camptothecin, doxorubicin and etoposide, the most commonly used topoisomerase inhibitors in chemotherapy for various types of cancer, can effectively block DNA replication by causing misalignment of DNA strands after supercoil unwinding (84). Similarly, bioalkylating agents, another widely used type of chemotherapeutic drug, trigger DNA damage-mediated aging reactions by causing DNA strand crosslinking, abnormal base pairing or DNA strand breaks. During cell division, these crosslinked DNA strands break, triggering DDR-mediated senescence. Cisplatin is an alkylating agent widely used in anticancer treatments (85). Microtubule inhibitors, such as paclitaxel can arrest tumor cells in the mitotic phase by disrupting the regular dynamics of microtubule spindles (86). These cytotoxic compounds, commonly used as standard cancer therapies for various tumor types, have dual effects. They induce a senescent phenotype in cancer cells and exert an anticancer effect. However, they also induce senescence in cellular components of the TME. CAFs play an essential role in promoting tumorigenic signals inside the tumor stroma. Senescent CAFs can enhance the differentiation and proliferation of neighboring tumor cells via the secretion of SASP factors. In a xenotransplant tumor model of breast cancer, senescent fibroblasts have been reported to promote the growth of breast cancer in mice through the secretion of matrix metalloproteinases (86). It has also been demonstrated that CAFs are prone to p53-mediated senescence during chemotherapy, leading to drug resistance in mouse lung cancer models (87). Similarly, a recent study revealed that docetaxel and cisplatin treatment can strongly induce a senescence phenotype in prostate- and ovary-associated fibroblasts in vitro. These senescent fibroblasts exhibit enhanced malignant behavior through alterations in metabolism and activation of SASP (88).

CAFs and RT. RT is a commonly used treatment for various types of cancers as it causes irreparable damage to cancer cell DNA. Similar to chemotherapy, the outcome of RT, whether senescence or apoptosis is induced, is mostly dependent on the radiation dose. High doses of radiation (>10 Gy) trigger apoptosis, whereas medium doses (>0.5 Gy) primarily lead to senescence (87). Unlike chemotherapy, RT can be administered directly to the cancer site, resulting in less damage to normal tissues. However, the tissues surrounding the tumor may still be affected, leading to a greater burden on aging cells and other local side effects (89). It has been shown that in murine rectal cancer models, tumor organoids and primary stromal cells derived from patients, CAFs undergo oxidative DNA damage following RT. This damage leads to p53-mediated senescence of CAFs, ultimately resulting in resistance to chemoradiotherapy and disease progression (67). Similarly, another study demonstrated that CAFs acquire senescence-like characteristics after RT, promoting lung cancer cell proliferation and radioresistance via activation of the JAK/STAT pathway (90).

CAFs and cell cycle inhibition. Cyclin-dependent kinase (CDK) inhibitor proteins are upregulated in senescent cells. CDK4/6 enzymes play a crucial role in facilitating the transition of the cell cycle from the G1 phase to the S phase (91). In a number of cancer cells, particularly in breast cancer, the CDK4/6 axis is overactivated; therefore, targeting CDK4/6 is a promising anticancer strategy (92). CDK4/6 inhibitors (such as palbociclib, abemaciclib and ribociclib) have been approved for cancer treatment. By mimicking the function of p16 (INK4a), CDK4/6 inhibitors induce cell senescence and arrest the cell cycle (93). Data from in vitro models have demonstrated that palbociclib (PD-0332991) can cause senescence in normal fibroblasts. These aging fibroblasts significantly promote breast cancer development (94). Another study revealed that prolonged use of the CDK4/6 inhibitor palbociclib (PD-6) causes aging and induction of the SASP phenotype in normal fibroblasts, and that aging fibroblasts can promote melanoma progression in both in vivo and in vitro melanoma models. The study also revealed that this senescence is associated with the downregulation of Mdm2 rather than DNA damage (95).

5. One-two punch model for cancer treatment

The clinical dose of cancer therapy, while effectively inducing tumor cell apoptosis or senescence (first punch), can also lead to senescence of other elements in the TME, which can contribute to tumor relapse, metastasis and the development of drug resistance to cancer treatment. Eliminating these senescent cells is crucial for reducing the risk of tumor progression (second punch) (5). Senolytics, which are a type of drug that selectively eliminates aging cells, have emerged as potential therapeutics for addressing this issue. In the context of tumors, senolytic strategies typically involve a combination of a senescence-inducing drug and another drug targeting senescent cells to induce the synthesis of lethal substances. By specifically targeting and eliminating these senescent cells, senolytics hold promise in preventing tumor progression and improving the efficacy of cancer treatment. A key characteristic of aging cells is the alteration of chromatin structure, which affects gene expression. The present review aims to provide a brief introduction to the application of senolytic drugs in targeting senescent fibroblasts or stromal cells in tumors. However, there are detailed studies that focus on the introductions and applications of these drugs in other diseases (96,97).

Studies have revealed that the expression of anti-apoptotic proteins often increases in senescent cells (74,98). Senolytic drugs can target upregulated anti-apoptotic pathways in senescent cells and reactivate the apoptotic pathway to eliminate aging cells (99). Navitoclax, a selective BCL inhibitor, is primarily active against BCL family members (such as BCL-2, BCL-XL and BCL-W). When used in combination with anticancer treatments, navitoclax can potentially achieve a dual effect. In a cell line and mouse model of glioblastoma, extensive senescence has been shown to occur in the brain following RT. Aging stromal cells promote the aggressive phenotype of glioblastoma via SASP, which enhances its ability to invade and migrate in vitro and in vivo. However, the use of navitoclax (ABT-263) selectively eliminates the senescent cells and significantly reduces glioma cell growth (100). Quercetin, a natural flavonol compound, and dasatinib, a tyrosine kinase inhibitor, are widely used to target senescent cells by interfering with Src and PI3K signaling. In a liver cancer mouse model, dasatinib has been reported to effectively inhibit senescent stellate cells, thereby inhibiting tumor progression (101). FOXO4-DRI, an interfering peptide that can regulate the activity of p53, disrupts the interactions between p53 and FOXO4. This selective disruption leads to p53 nuclear exclusion and apoptosis of senescent cells (102). Research has demonstrated that RT can induce senescence-like characteristics of CAFs in non-small cell lung cancer. FOXO4-DRI enhances the radiosensitivity of NSCLC cells and alleviates RT-induced pulmonary fibrosis by targeting aging CAFs, both in vitro and in vivo (90) (Fig. 3).

6. Conclusions and future perspectives

The goal of tumor therapy is to achieve a complete cure by eliminating malignant tumors. Increasing attention has been paid to the function of the TME in tumor therapies, as it can significantly impact therapeutic outcomes. Systemic anticancer therapy induces senescence in cancer cells and triggers the development of aging components in the TME. Among these components, CAFs are critical in tumor progression. The one-two punch strategy aims to address the dual behavior of TIS, which can have both favorable and unfavorable effects on tumor therapies. This approach acknowledges both the positive outcomes and the possible challenges associated with senescence induction. By targeting both the beneficial and harmful effects of TIS, this strategy can provide significant therapeutic benefits and may be an effective approach for future studies on cancer treatment.

Acknowledgements

Not applicable.

Funding

Funding: This research was funded by the National Natural Science Foundation of China (grant no. 81773945) and the Basic Public Welfare Research Project of Zhejiang Province (grant no. LY23H280011).

Availability of data and materials

Not applicable.

Authors' contributions

QZ and YL were involved in study conceptualization. HF and SS contributed to the generation of the figures and table. RJ performed the literature investigation. QZ and YL wrote the original draft. YJ reviewed and edited the manuscript. ZC supervised the study. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

de Visser KE and Joyce JA: The evolving tumor microenvironment: From cancer initiation to metastatic. Cancer Cell. 41:374–403. 2023.PubMed/NCBI View Article : Google Scholar

2 

Roma-Rodrigues C, Mendes R, Baptista PV and Fernandes AR: Targeting tumor microenvironment for cancer therapy. Int J Mol Sci. 20(840)2019.PubMed/NCBI View Article : Google Scholar

3 

Chen Y, Zhang X, Yang H, Liang T and Bai X: The ‘Self-eating’ of cancer-associated fibroblast: A potential target for cancer. Biomed Pharmacother. 163(114762)2023.PubMed/NCBI View Article : Google Scholar

4 

Saw PE, Chen J and Song E: Targeting CAFs to overcome anticancer therapeutic resistance. Trends Cancer. 8:527–555. 2022.PubMed/NCBI View Article : Google Scholar

5 

Prasanna PG, Citrin DE, Hildesheim J, Ahmed MM, Venkatachalam S, Riscuta G, Xi D, Zheng G, Deursen JV, Goronzy J, et al: Therapy-Induced senescence: Opportunities to improve anticancer therapy. J Natl Cancer Inst. 113:1285–1298. 2021.PubMed/NCBI View Article : Google Scholar

6 

Ewald JA, Desotelle JA, Wilding G and Jarrard DF: Therapy-induced senescence in cancer. J Natl Cancer Inst. 102:1536–1546. 2010.PubMed/NCBI View Article : Google Scholar

7 

Kumari R and Jat P: Mechanisms of cellular senescence: Cell cycle arrest and senescence associated Secretory Phenotype. Front Cell Dev Biol. 9(645593)2021.PubMed/NCBI View Article : Google Scholar

8 

Hwang HJ, Lee YR, Kang D, Lee HC, Seo HR, Ryu JK, Kim YN, Ko YG, Park HJ and Lee JS: Endothelial cells under therapy-induced senescence secrete CXCL11, which increases aggressiveness of breast cancer cells. Cancer Lett. 490:100–110. 2020.PubMed/NCBI View Article : Google Scholar

9 

Chen X and Song E: Turning foes to friends: Targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 18:99–115. 2019.PubMed/NCBI View Article : Google Scholar

10 

Öhlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, Corbo V, Oni TE, Hearn SA, Lee EJ, et al: Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. 214:579–596. 2017.PubMed/NCBI View Article : Google Scholar

11 

Kalluri R: The biology and function of fibroblasts in cancer. Nat Rev Cancer. 16:582–598. 2016.PubMed/NCBI View Article : Google Scholar

12 

Kalluri R and Zeisberg M: Fibroblasts in cancer. Nat Rev Cancer. 6:392–401. 2006.PubMed/NCBI View Article : Google Scholar

13 

Filliol A, Saito Y, Nair A, Dapito DH, Yu LX, Ravichandra A, Bhattacharjee S, Affo S, Fujiwara N, Su H, et al: Opposing roles of hepatic stellate cell subpopulations in hepatocarcinogenesis. Nature. 610:356–365. 2022.PubMed/NCBI View Article : Google Scholar

14 

Quante M, Tu SP, Tomita H, Gonda T, Wang SS, Takashi S, Baik GH, Shibata W, Diprete B, Betz KS, et al: Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth. Cancer Cell. 19:257–272. 2011.PubMed/NCBI View Article : Google Scholar

15 

Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, Richardson AL, Polyak K, Tubo R and Weinberg RA: Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 449:557–563. 2007.PubMed/NCBI View Article : Google Scholar

16 

Rhim AD, Mirek ET, Aiello NM, Maitra A, Bailey JM, McAllister F, Reichert M, Beatty GL, Rustgi AK, Vonderheide RH, et al: EMT and dissemination precede pancreatic tumor formation. Cell. 48:349–361. 2012.PubMed/NCBI View Article : Google Scholar

17 

Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, Choi H, El Rayes T, Ryu S, Troeger J, et al: Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 527:472–476. 2015.PubMed/NCBI View Article : Google Scholar

18 

Park D, Sahai E and Rullan A: SnapShot: Cancer-Associated fibroblasts. Cell. 181:486–486.e1. 2020.PubMed/NCBI View Article : Google Scholar

19 

Glabman RA, Choyke PL and Sato N: Cancer-Associated fibroblasts: Tumorigenicity and targeting for cancer therapy. Cancers (Basel). 14(3906)2022.PubMed/NCBI View Article : Google Scholar

20 

Fiori ME, Di Franco S, Villanova L, Bianca P, Stassi G and De Maria R: Cancer-associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol Cancer. 18(70)2019.PubMed/NCBI View Article : Google Scholar

21 

Zhao Y, Shen M, Wu L, Yang H, Yao Y, Yang Q, Du J, Liu L, Li Y and Bai Y: Stromal cells in the tumor microenvironment: Accomplices of tumor progression? Cell Death Dis. 14(587)2023.PubMed/NCBI View Article : Google Scholar

22 

Biffi G and Tuveson DA: Diversity and biology of cancer-associated fibroblasts. Physiol Rev. 101:147–176. 2021.PubMed/NCBI View Article : Google Scholar

23 

Fiaschi T, Marini A, Giannoni E, Taddei ML, Gandellini P, De Donatis A, Lanciotti M, Serni S, Cirri P and Chiarugi P: Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay. Cancer Res. 72:5130–5140. 2012.PubMed/NCBI View Article : Google Scholar

24 

Singh S, Singh AP and Mitra R: Cancer-Associated Fibroblasts: Major co-conspirators in tumor development. Cancers (Basel). 16(211)2024.PubMed/NCBI View Article : Google Scholar

25 

Kuzet SE and Gaggioli C: Fibroblast activation in cancer: When seed fertilizes soil. Cell Tissue Res. 365:607–619. 2016.PubMed/NCBI View Article : Google Scholar

26 

Caja L, Dituri F, Mancarella S, Caballero-Diaz D, Moustakas A, Giannelli G and Fabregat I: TGF-β and the tissue microenvironment: Relevance in fibrosis and cancer. Int J Mol Sci. 19(1294)2018.PubMed/NCBI View Article : Google Scholar

27 

Derynck R, Turley SJ and Akhurst RJ: TGFβ biology in cancer progression and immunotherapy. Nat Rev Clin Oncol. 18:9–34. 2021.PubMed/NCBI View Article : Google Scholar

28 

Chandra Jena B, Sarkar S, Rout L and Mandal M: The transformation of cancer-associated fibroblasts: Current perspectives on the role of TGF-β in CAF mediated tumor progression and therapeutic resistance. Cancer Lett. 520:222–232. 2021.PubMed/NCBI View Article : Google Scholar

29 

Neri S, Miyashita T, Hashimoto H, Suda Y, Ishibashi M, Kii H, Watanabe H, Kuwata T, Tsuboi M, Goto K, et al: Fibroblast-led cancer cell invasion is activated by epithelial-mesenchymal transition through platelet-derived growth factor BB secretion of lung adenocarcinoma. Cancer Lett. 395:20–30. 2017.PubMed/NCBI View Article : Google Scholar

30 

Bronzert DA, Pantazis P, Antoniades HN, Kasid A, Davidson N, Dickson RB and Lippman ME: Synthesis and secretion of platelet-derived growth factor by human breast cancer cell lines. Proc Natl Acad Sci USA. 84:5763–5767. 1987.PubMed/NCBI View Article : Google Scholar

31 

Shao ZM, Nguyen M and Barsky SH: Human breast carcinoma desmoplasia is PDGF initiated. Oncogene. 19:4337–4345. 2000.PubMed/NCBI View Article : Google Scholar

32 

Montori M, Scorzoni C, Argenziano ME, Balducci D, De Blasio F, Martini F, Buono T, Benedetti A, Marzioni M and Maroni L: Cancer-Associated fibroblasts in cholangiocarcinoma: Current knowledge and possible implications for therapy. J Clin Med. 11(6498)2022.PubMed/NCBI View Article : Google Scholar

33 

Liu C, Zhang Y, Lim S, Hosaka K, Yang Y, Pavlova T, Alkasalias T, Hartman J, Jensen L, Xing X, et al: A zebrafish model discovers a novel mechanism of stromal fibroblast-mediated. Clin Cancer Res. 23:4769–4779. 2017.PubMed/NCBI View Article : Google Scholar

34 

Nissen LJ, Cao R, Hedlund EM, Wang Z, Zhao X, Wetterskog D, Funa K, Bråkenhielm E and Cao Y: Angiogenic factors FGF2 and PDGF-BB synergistically promote murine tumor neovascularization and metastasis. J Clin Invest. 117:2766–2777. 2007.PubMed/NCBI View Article : Google Scholar

35 

Nurmik M, Ullmann P, Rodriguez F, Haan S and Letellier E: In search of definitions: Cancer-associated fibroblasts and their markers. Int J Cancer. 146:895–905. 2020.PubMed/NCBI View Article : Google Scholar

36 

Yamamoto Y, Kasashima H, Fukui Y, Tsujio G, Yashiro M and Maeda K: The heterogeneity of cancer-associated fibroblast subpopulations: Their origins, biomarkers, and roles in the tumor microenvironment. Cancer Sci. 114:16–24. 2023.PubMed/NCBI View Article : Google Scholar

37 

Arnold JN, Magiera L, Kraman M and Fearon DT: Tumoral immune suppression by macrophages expressing fibroblast activation protein-α and heme oxygenase-1. Cancer Immunol Res. 2:121–126. 2014.PubMed/NCBI View Article : Google Scholar

38 

Tchou J, Zhang PJ, Bi Y, Satija C, Marjumdar R, Stephen TL, Lo A, Chen H, Mies C, June CH, et al: Fibroblast activation protein expression by stromal cells and tumor-associated macrophages in human breast cancer. Hum Pathol. 44:2549–2557. 2013.PubMed/NCBI View Article : Google Scholar

39 

Jin J, Zhu SJ, Zhu ZM, Yang YJ and Ding G: Relationship between proliferation of vascular smooth muscle cells and PDGF-AA and PDGFR-alpha expression in SHRs. Sheng Li Xue Bao. 54:145–148. 2002.PubMed/NCBI(In Chinese).

40 

Smyth LCD, Rustenhoven J, Scotter EL, Schweder P, Faull RLM, Park TIH and Dragunow M: Markers for human brain pericytes and smooth muscle cells. J Chem Neuroanat. 92:48–60. 2018.PubMed/NCBI View Article : Google Scholar

41 

Shamsi F, Piper M, Ho LL, Huang TL, Gupta A, Streets A, Lynes MD and Tseng YH: Vascular smooth muscle-derived Trpv1(+) progenitors are a source of cold-induced thermogenic adipocytes. Nat Metab. 3:485–495. 2021.PubMed/NCBI View Article : Google Scholar

42 

Sá da Bandeira D, Casamitjana J and Crisan M: Pericytes, integral components of adult hematopoietic stem cell niches. Pharmacol Ther. 171:104–113. 2017.PubMed/NCBI View Article : Google Scholar

43 

Koliaraki V, Pallangyo CK, Greten FR and Kollias G: Mesenchymal cells in colon cancer. Gastroenterology. 152:964–979. 2017.PubMed/NCBI View Article : Google Scholar

44 

Togarrati PP, Dinglasan N, Desai S, Ryan WR and Muench MO: CD29 is highly expressed on epithelial, myoepithelial, and mesenchymal stromal cells of human salivary glands. Oral Dis. 24:561–572. 2018.PubMed/NCBI View Article : Google Scholar

45 

Delangre E, Oppliger E, Berkcan S, Gjorgjieva M, Correia de Sousa M and Foti M: S100 proteins in fatty liver disease and hepatocellular carcinoma. Int J Mol Sci. 23(11030)2022.PubMed/NCBI View Article : Google Scholar

46 

Martin M, Zhang J, Miao Y, He M, Kang J, Huang HY, Chou CH, Huang TS, Hong HC, Su SH, et al: Role of endothelial cells in pulmonary fibrosis via SREBP2 activation. JCI Insight. 6(e125635)2021.PubMed/NCBI View Article : Google Scholar

47 

Kamphuis W, Kooijman L, Orre M, Stassen O, Pekny M and Hol EM: GFAP and vimentin deficiency alters gene expression in astrocytes and microglia in wild-type mice and changes the transcriptional response of reactive glia in mouse model for Alzheimer's disease. Glia. 63:1036–1056. 2015.PubMed/NCBI View Article : Google Scholar

48 

Suzuki-Inoue K: Platelets and cancer-associated thrombosis: Focusing on the platelet activation receptor CLEC-2 and podoplanin. Blood. 134:1912–1918. 2021.PubMed/NCBI View Article : Google Scholar

49 

Krishnan H, Rayes J, Miyashita T, Ishii G, Retzbach EP, Sheehan SA, Takemoto A, Chang YW, Yoneda K, Asai J, et al: Podoplanin: An emerging cancer biomarker and therapeutic target. Cancer Sci. 109:1292–1299. 2018.PubMed/NCBI View Article : Google Scholar

50 

Lowy CM and Oskarsson T: Tenascin C in metastasis: A view from the invasive front. Cell Adh Migr. 9:112–124. 2015.PubMed/NCBI View Article : Google Scholar

51 

Yoshida T, Akatsuka T and Imanaka-Yoshida K: Tenascin-C and integrins in cancer. Cell Adh Migr. 9:96–104. 2015.PubMed/NCBI View Article : Google Scholar

52 

Yue H, Li W, Chen R, Wang J, Lu X and Li J: Stromal POSTN induced by TGF-β1 facilitates the migration and invasion of ovarian cancer. Gynecol Oncol. 160:530–538. 2021.PubMed/NCBI View Article : Google Scholar

53 

Liu C, Feng X, Wang B, Wang X, Wang C, Yu M, Cao G and Wang H: Bone marrow mesenchymal stem cells promote head and neck cancer progression through Periostin-mediated phosphoinositide 3-kinase/Akt/mammalian target of rapamycin. Cancer Sci. 109:688–698. 2018.PubMed/NCBI View Article : Google Scholar

54 

Soikkeli J, Podlasz P, Yin M, Nummela P, Jahkola T, Virolainen S, Krogerus L, Heikkilä P, von Smitten K, Saksela O and Hölttä E: Metastatic outgrowth encompasses COL-I, FN1, and POSTN up-regulation and assembly to fibrillar networks regulating cell adhesion, migration, and growth. Am J Pathol. 177:387–403. 2010.PubMed/NCBI View Article : Google Scholar

55 

Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, Blaudszun AR, Yevsa T, Fricke S and Kossatz-Boehlert U: Cancer stem cells-origins and biomarkers: Perspectives for targeted personalized therapies. Front Immunol. 11(1280)2020.PubMed/NCBI View Article : Google Scholar

56 

Bonnans C, Chou J and Werb Z: Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol. 15:786–801. 2014.PubMed/NCBI View Article : Google Scholar

57 

Winkler J, Abisoye-Ogunniyan A, Metcalf KJ and Werb Z: Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun. 11(5120)2020.PubMed/NCBI View Article : Google Scholar

58 

Asif PJ, Longobardi C, Hahne M and Medema JP: The role of cancer-associated fibroblasts in cancer invasion and metastasis. Cancers (Basel). 13(4720)2021.PubMed/NCBI View Article : Google Scholar

59 

Eiró N, Fernandez-Garcia B, Vázquez J, Del Casar JM, González LO and Vizoso FJ: A phenotype from tumor stroma based on the expression of metalloproteases and their inhibitors, associated with prognosis in breast cancer. Oncoimmunology. 4(e992222)2015.PubMed/NCBI View Article : Google Scholar

60 

Neri S, Ishii G, Hashimoto H, Kuwata T, Nagai K, Date H and Ochiai A: Podoplanin-expressing cancer-associated fibroblasts lead and enhance the local invasion of cancer cells in lung adenocarcinoma. Int J Cancer. 137:784–796. 2015.PubMed/NCBI View Article : Google Scholar

61 

Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall JF, Harrington K and Sahai E: Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 9:1392–1400. 2007.PubMed/NCBI View Article : Google Scholar

62 

Pastushenko I, Mauri F, Song Y, de Cock F, Meeusen B, Swedlund B, Impens F, Van Haver D, Opitz M, Thery M, et al: Fat1 deletion promotes hybrid EMT state, tumour stemness and metastasis. Nature. 589:448–455. 2021.PubMed/NCBI View Article : Google Scholar

63 

Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, Wu CC, LeBleu VS and Kalluri R: Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature. 527:525–530. 2015.PubMed/NCBI View Article : Google Scholar

64 

Lüönd F, Sugiyama N, Bill R, Bornes L, Hager C, Tang F, Santacroce N, Beisel C, Ivanek R, Bürglin T, et al: Distinct contributions of partial and full EMT to breast cancer malignancy. Dev Cell. 56:3203–3221.e11. 2021.PubMed/NCBI View Article : Google Scholar

65 

Fares J, Fares MY, Khachfe HH, Salhab HA and Fares Y: Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct Target Ther. 5(28)2020.PubMed/NCBI View Article : Google Scholar

66 

Ren Y, Jia HH, Xu YQ, Zhou X, Zhao XH, Wang YF, Song X, Zhu ZY, Sun T, Dou Y, et al: Paracrine and epigenetic control of CAF-induced metastasis: The role of HOTAIR stimulated by TGF-ß1 secretion. Mol Cancer. 17(5)2018.PubMed/NCBI View Article : Google Scholar

67 

Nicolas AM, Pesic M, Engel E, Ziegler PK, Diefenhardt M, Kennel KB, Buettner F, Conche C, Petrocelli V, Elwakeel E, et al: Inflammatory fibroblasts mediate resistance to neoadjuvant therapy in rectal cancer. Cancer Cell. 40:168–184. 2022.PubMed/NCBI View Article : Google Scholar

68 

Jia C, Wang G, Wang T, Fu B, Zhang Y, Huang L, Deng Y, Chen G, Wu X, Chen J, et al: Cancer-associated Fibroblasts induce epithelial-mesenchymal transition via the transglutaminase 2-dependent IL-6/IL6R/STAT3 axis in hepatocellular carcinoma. Int J Biol Sci. 16:2542–2558. 2020.PubMed/NCBI View Article : Google Scholar

69 

Wang Y, Lan W, Xu M, Song J, Mao J, Li C, Du X, Jiang Y, Li E, Zhang R and Wang Q: Cancer-associated fibroblast-derived SDF-1 induces epithelial-mesenchymal transition of lung adenocarcinoma via CXCR4/β-catenin/PPARδ signalling. Cell Death Dis. 12(214)2021.PubMed/NCBI View Article : Google Scholar

70 

Dai J, Su Y, Zhong S, Cong L, Liu B, Yang J, Tao Y, He Z, Chen C and Jiang Y: Exosomes: Key players in cancer and potential therapeutic strategy. Signal Transduct Target Ther. 5(145)2020.PubMed/NCBI View Article : Google Scholar

71 

Richards KE, Zeleniak AE, Fishel ML, Wu J, Littlepage LE and Hill R: Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene. 36:1770–1778. 2017.PubMed/NCBI View Article : Google Scholar

72 

Zhang H, Deng T, Liu R, Ning T, Yang H, Liu D, Zhang Q, Lin D, Ge S, Bai M, et al: CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol Cancer. 19(43)2020.PubMed/NCBI View Article : Google Scholar

73 

Martínez-Zamudio RI, Robinson L, Roux PF and Bischof O: SnapShot: Cellular senescence pathways. Cell. 170:816–816.e11. 2017.PubMed/NCBI View Article : Google Scholar

74 

Wang L, Lankhorst L and Bernards R: Exploiting senescence for the treatment of cancer. Nat Rev Cancer. 22:340–355. 2022.PubMed/NCBI View Article : Google Scholar

75 

Bahcecioglu G, Yue X, Howe E, Guldner I, Stack MS, Nakshatri H, Zhang S and Zorlutuna P: Aged breast extracellular matrix drives mammary epithelial cells to an invasive. Adv Sci (Weinh). 8(e2100128)2021.PubMed/NCBI View Article : Google Scholar

76 

Bancaro N, Calì B, Troiani M, Elia AR, Arzola RA, Attanasio G, Lai P, Crespo M, Gurel B, Pereira R, et al: Apolipoprotein E induces pathogenic senescent-like myeloid cells in prostate. Cancer Cell. 41:602–619.e11. 2023.PubMed/NCBI View Article : Google Scholar

77 

Nguyen PT, Kanno K, Pham QT, Kikuchi Y, Kakimoto M, Kobayashi T, Otani Y, Kishikawa N, Miyauchi M, Arihiro K, et al: Senescent hepatic stellate cells caused by deoxycholic acid modulates malignant. J Cancer Res Clin Oncol. 146:3255–3268. 2020.PubMed/NCBI View Article : Google Scholar

78 

Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, Iwakura Y, Oshima K, Morita H, Hattori M, et al: Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 499:97–101. 2013.PubMed/NCBI View Article : Google Scholar

79 

Loo TM, Kamachi F, Watanabe Y, Yoshimoto S, Kanda H, Arai Y, Nakajima-Takagi Y, Iwama A, Koga T, Sugimoto Y, et al: Gut Microbiota Promotes Obesity-Associated Liver Cancer through PGE(2)-Mediated. Cancer Discov. 7:522–538. 2017.PubMed/NCBI View Article : Google Scholar

80 

Chambers CR, Ritchie S, Pereira BA and Timpson P: Overcoming the senescence-associated secretory phenotype (SASP): A complex mechanism of resistance in the treatment of cancer. Mol Oncol. 15:3242–3255. 2021.PubMed/NCBI View Article : Google Scholar

81 

Ewald JA, Desotelle JA, Wilding G and Jarrard DF: Therapy-induced senescence in cancer. J Natl Cancer Inst. 102:1536–1546. 2010.PubMed/NCBI View Article : Google Scholar

82 

Groelly FJ, Fawkes M, Dagg RA, Blackford AN and Tarsounas M: Targeting DNA damage response pathways in cancer. Nat Rev Cancer. 23:78–94. 2023.PubMed/NCBI View Article : Google Scholar

83 

Shiloh Y: ATM and related protein kinases: Safeguarding genome integrity. Nat Rev Cancer. 3:155–168. 2003.PubMed/NCBI View Article : Google Scholar

84 

Buzun K, Bielawska A, Bielawski K and Gornowicz A: DNA topoisomerases as molecular targets for anticancer drugs. J Enzyme Inhib Med Chem. 35:1781–1799. 2020.PubMed/NCBI View Article : Google Scholar

85 

Aasland D, Götzinger L, Hauck L, Berte N, Meyer J, Effenberger M, Schneider S, Reuber EE, Roos WP, Tomicic MT, et al: Temozolomide induces senescence and repression of DNA repair pathways in glioblastoma cells via activation of ATR-CHK1, p21, and NF-κB. Cancer Res. 79:99–113. 2019.PubMed/NCBI View Article : Google Scholar

86 

Mikuła-Pietrasik J, Witucka A, Pakuła M, Uruski P, Begier-Krasińska B, Niklas A, Tykarski A and Książek K: Comprehensive review on how platinum- and taxane-based chemotherapy of ovarian cancer affects biology of normal cells. Cell Mol Life Sci. 76:681–697. 2019.PubMed/NCBI View Article : Google Scholar

87 

Burdelya LG, Komarova EA, Hill JE, Browder T, Tararova ND, Mavrakis L, DiCorleto PE, Folkman J and Gudkov AV: Inhibition of p53 response in tumor stroma improves efficacy of anticancer treatment by increasing antiangiogenic effects of chemotherapy and radiotherapy in mice. Cancer Res. 66:9356–9361. 2006.PubMed/NCBI View Article : Google Scholar

88 

Pardella E, Pranzini E, Nesi I, Parri M, Spatafora P, Torre E, Muccilli A, Castiglione F, Fambrini M, Sorbi F, et al: Therapy-Induced stromal senescence promoting aggressiveness of prostate and ovarian cancer. Cells. 11(4026)2022.PubMed/NCBI View Article : Google Scholar

89 

Li M, You L, Xue J and Lu Y: Ionizing radiation-induced cellular senescence in normal, non-transformed cells and the involved DNA damage response: A mini review. Front Pharmacol. 9(522)2018.PubMed/NCBI View Article : Google Scholar

90 

Meng J, Li Y, Wan C, Sun Y, Dai X, Huang J, Hu Y, Gao Y, Wu B, Zhang Z, et al: Targeting senescence-like fibroblasts radiosensitizes non-small cell lung cancer and reduces radiation-induced pulmonary fibrosis. JCI Insight. 6(e146334)2021.PubMed/NCBI View Article : Google Scholar

91 

Malumbres M and Barbacid M: Cell cycle, CDKs and cancer: A changing paradigm. Nat Rev Cancer. 9:153–166. 2009.PubMed/NCBI View Article : Google Scholar

92 

Wagner V and Gil J: Senescence as a therapeutically relevant response to CDK4/6 inhibitors. Oncogene. 39:5165–5176. 2020.PubMed/NCBI View Article : Google Scholar

93 

Coppé JP, Rodier F, Patil CK, Freund A, Desprez PY and Campisi J: Tumor suppressor and aging biomarker p16(INK4a) induces cellular senescence without the associated inflammatory secretory phenotype. J Biol Chem. 286:36396–36403. 2011.PubMed/NCBI View Article : Google Scholar

94 

Capparelli C, Chiavarina B, Whitaker-Menezes D, Pestell TG, Pestell RG, Hulit J, Andò S, Howell A, Martinez-Outschoorn UE, Sotgia F and Lisanti MP: CDK inhibitors (p16/p19/p21) induce senescence and autophagy in cancer-associated fibroblasts, ‘fueling’ tumor growth via paracrine interactions, without an increase in neo-angiogenesis. Cell Cycle. 11:3599–3610. 2012.PubMed/NCBI View Article : Google Scholar

95 

Guan X, LaPak KM, Hennessey RC, Yu CY, Shakya R, Zhang J and Burd CE: Stromal senescence by prolonged CDK4/6 inhibition potentiates tumor growth. Mol Cancer Res. 15:237–249. 2017.PubMed/NCBI View Article : Google Scholar

96 

Campisi J, Kapahi P, Lithgow GJ, Melov S, Newman JC and Verdin E: From discoveries in ageing research to therapeutics for healthy ageing. Nature. 571:183–192. 2019.PubMed/NCBI View Article : Google Scholar

97 

Chaib S, Tchkonia T and Kirkland JL: Cellular senescence and senolytics: The path to the clinic. Nat Med. 28:1556–1568. 2022.PubMed/NCBI View Article : Google Scholar

98 

Yosef R, Pilpel N, Tokarsky-Amiel R, Biran A, Ovadya Y, Cohen S, Vadai E, Dassa L, Shahar E, Condiotti R, et al: Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat Commun. 7(11190)2016.PubMed/NCBI View Article : Google Scholar

99 

Jochems F, Thijssen B, De Conti G, Jansen R, Pogacar Z, Groot K, Wang L, Schepers A, Wang C, Jin H, et al: The Cancer SENESCopedia: A delineation of cancer cell senescence. Cell Rep. 36(109441)2021.PubMed/NCBI View Article : Google Scholar

100 

Fletcher-Sananikone E, Kanji S, Tomimatsu N, Di Cristofaro LFM, Kollipara RK, Saha D, Floyd JR, Sung P, Hromas R, Burns TC, et al: Elimination of radiation-induced senescence in the brain tumor microenvironment attenuates glioblastoma recurrence. Cancer Res. 81:5935–5947. 2021.PubMed/NCBI View Article : Google Scholar

101 

Li F, Huangyang P, Burrows M, Guo K, Riscal R, Godfrey J, Lee KE, Lin N, Lee P, Blair IA, et al: FBP1 loss disrupts liver metabolism and promotes tumorigenesis through a hepatic stellate cell senescence secretome. Nat Cell Biol. 22:728–739. 2020.PubMed/NCBI View Article : Google Scholar

102 

Baar MP, Brandt RMC, Putavet DA, Klein JDD, Derks KWJ, Bourgeois BRM, Stryeck S, Rijksen Y, van Willigenburg H, Feijtel DA, et al: Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell. 169:132–147.e16. 2017.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2024
Volume 27 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Q, Lou Y, Fang H, Sun S, Jin R, Ji Y and Chen Z: Cancer‑associated fibroblasts under therapy‑induced senescence in the tumor microenvironment (Review). Exp Ther Med 27: 150, 2024
APA
Zhang, Q., Lou, Y., Fang, H., Sun, S., Jin, R., Ji, Y., & Chen, Z. (2024). Cancer‑associated fibroblasts under therapy‑induced senescence in the tumor microenvironment (Review). Experimental and Therapeutic Medicine, 27, 150. https://doi.org/10.3892/etm.2024.12438
MLA
Zhang, Q., Lou, Y., Fang, H., Sun, S., Jin, R., Ji, Y., Chen, Z."Cancer‑associated fibroblasts under therapy‑induced senescence in the tumor microenvironment (Review)". Experimental and Therapeutic Medicine 27.4 (2024): 150.
Chicago
Zhang, Q., Lou, Y., Fang, H., Sun, S., Jin, R., Ji, Y., Chen, Z."Cancer‑associated fibroblasts under therapy‑induced senescence in the tumor microenvironment (Review)". Experimental and Therapeutic Medicine 27, no. 4 (2024): 150. https://doi.org/10.3892/etm.2024.12438