Skip to main content

Cancer-associated fibroblasts in radiotherapy: Bystanders or protagonists?

Abstract

Background

The primary goal of radiotherapy (RT) is to induce cellular damage on malignant cells; however, it is becoming increasingly recognized the important role played by the tumor microenvironment (TME) in therapy outcomes. Therapeutic irradiation of tumor lesions provokes profound cellular and biological reconfigurations within the TME that ultimately may influence the fate of the therapy.

Main content

Cancer-associated fibroblasts (CAFs) are known to participate in all stages of cancer progression and are increasingly acknowledged to contribute to therapy resistance. Accumulated evidence suggests that, upon radiation, fibroblasts/CAFs avoid cell death but instead enter a permanent senescent state, which in turn may influence the behavior of tumor cells and other components of the TME. Despite the proposed participation of senescent fibroblasts on tumor radioprotection, it is still incompletely understood the impact that RT has on CAFs and the ultimate role that irradiated CAFs have on therapy outcomes. Some of the current controversies may emerge from generalizing observations obtained using normal fibroblasts and CAFs, which are different cell entities that may respond differently to radiation exposure.

Conclusion

In this review we present current knowledge on the field of CAFs role in radiotherapy; we discuss the potential tumorigenic functions of radiation-induced senescent fibroblasts and CAFs and we make an effort to integrate the knowledge emerging from preclinical experimentation with observations from the clinics.

Video Abstract

Plain English summary

Radiotherapy (RT) is currently used to treat more than 50% of all diagnosed cancer cases and counts for around 40% of all cure rates. The primary goal of RT has always been to induce damage on tumor cells; thus, it has been traditionally believed that the therapeutic efficacy of RT is mediated exclusively by its capacity to directly kill malignant cells. However, tumors are complex tissues composed of multiple cellular and acellular elements often referred as the tumor microenvironment (TME) or tumor stroma. Both, the malignant and the non-malignant cells in tumors receive the same prescribed radiation dose during treatment. In recent years, it has become more and more evident the fundamental role played by elements of the TME on therapy outcomes. One of the most abundant and influential elements of the reactive stroma in tumors are cancer-associated fibroblasts (CAFs). CAFs are also exposed to the full prescribed radiation dose during the course of radiotherapy. Accumulated evidence suggests that, upon radiation, fibroblasts/CAFs avoid cell death but instead enter a permanent senescent (growth arrest) state, which in turn may influence the behavior of tumor cells and other components of the TME. Despite the proposed participation of senescent fibroblasts on tumor radioprotection, it is still incompletely understood the impact that RT has on CAFs and the ultimate role that irradiated CAFs have on therapy outcomes. Further research using appropriate experimental models to study CAFs, and more information from clinical research is needed to unveil the ultimate role played by CAFs on radiotherapy.

Background

Radiotherapy is currently used to treat more than 50% of all diagnosed cancer cases and counts for around 40% of all cure rates [1]. Traditionally, it has been thought that the therapeutic efficacy of RT is mediated predominantly by its capacity to directly kill malignant cells. Consequently, research aiming for improved therapeutic outcomes have focused almost entirely on the cancer cell itself, encouraging dose escalation strategies or the use of radiosensitizers for enhancing the antitumor effects [1, 2]. However, in recent years, it has become increasingly evident the fundamental role played by elements of the tumor stroma on therapy outcomes [3, 4]. Hence, today, several components of the stroma have been identified to interact with the response to RT. The extracellular matrix (ECM) can dictate radiation responsiveness by influencing oxygen availability, inducing pro-survival signals via cell-matrix interactions or by controlling the bioavailability of growth factors and cytokines [5,6,7,8]. Radiation can also induce endothelial cell dysfunctions, with altered expression of cell adhesion molecules and changes in the vascular network in tumors, which altogether may translate into impaired tumor perfusion, altered immune infiltration and/or increased hypoxia [9,10,11,12]. Moreover, the inexorable tissue damage provoked by RT is normally associated with an inflammatory response [13, 14], characterized by recruitment and activation of innate and adaptive immune cells and release of potent pro-inflammatory mediators [15,16,17].

As one of the most abundant elements of the reactive stroma in solid tumors, cancer-associated fibroblasts (CAFs) [18,19,20] are exposed to the full prescribed radiation dose during the course of clinical radiotherapy [21, 22]. However, the impact that radiation has on CAFs and the potential downstream effects of radiation-induced changes on therapy outcomes remain unsettled [11, 23,24,25]. Studies investigating cytotoxic effects of ionizing radiation (IR) on stromal cells have revealed the intrinsic radio-resistant nature of fibroblasts [26, 27]. Several in vitro observations have confirmed that following exposure to high radiation doses, CAFs [26, 28, 29] and normal tissue fibroblasts evade cell death [30, 31], but acquire a senescent phenotype [26, 30] accompanied by impaired proliferative and migratory functions [26]. A role of senescent cells in cancer development has been proposed, however such effects can be ascribed as beneficial or detrimental depending on the context [32,33,34,35,36,37,38]. Based on current knowledge on the topic of radiation effects in fibroblasts, it exists a generalized view postulating the acquisition of enhanced pro-malignant functions in irradiated (senescent) fibroblasts/CAFs, irrespective of their origin and functional status [39, 40]. However, most of the existing knowledge in this field emerge from studies conducted with normal tissue fibroblasts or fibroblast cell lines. In contrast to quiescent “normal” tissue fibroblasts (NFs), CAFs (in non-irradiated conditions) actively produce numerous tumor-promoting molecules such as matrix metalloproteinases (MMPs), inflammatory cytokines, pro-angiogenic factors and miscellaneous tumor-promoting growth factors [18, 20, 41]. Thus, to understand the contribution of CAFs to therapeutic outcomes post-RT, the activated status of the cells before treatment should be taken into consideration. Additionally, in contemporary high-precision image-guided radiotherapy settings with steep dose gradients, only cells residing within the delineated tumor volume (i.e. the planning target volume), or its periphery, are exposed to the full prescribed radiation dose (i.e. CAFs) [42], whereas healthy tissues located outside the target field (NFs) normal Fbs typically receive only residual radiation doses [21, 22, 43]. In this review, we discuss the controversies and misconceptions that have emerged in this field, due to the unrecognized fact that normal tissue fibroblasts and CAFs represent two different cell entities, and that radiation effects and responses may differ substantially between them.

Normal fibroblasts versus CAFs

Fibroblasts are spindle-shaped cells of mesenchymal origin which reside in connective tissues and are responsible for the production of ECM components and connective tissue homeostasis [44]. The embryonic origin of tissue resident fibroblasts is shared by other mesenchymal lineages including adipocytes, pericytes, chondrocytes and osteoblasts [45]. The difficulty in defining fibroblasts results largely from the lack of unique markers that are not expressed in other cell types [19, 45, 46]. In practical terms, fibroblasts are often defined by their specific cellular functions (ECM synthesis and remodeling), their lack of mutations and their lack of lineage markers for epithelial cells, endothelial cells and leukocytes. When quiescent, fibroblasts and stellate cells from specific organs do not proliferate, they form few cell-to-cell connections, they synthesize little collagen and secrete low amounts of cytokines and other signal mediators [18, 47].

Activation of tissue resident fibroblasts can occur in physiological conditions, such as wound healing or acute organ repair, or in pathologic conditions such as chronic infections, organ fibrosis, autoimmune diseases, or cancer [44, 45]. Activated fibroblasts can originate from multiple cell types including tissue-resident fibroblasts, mesenchymal progenitor cells, pericytes, organ-specific stellate cells or from trans-differentiation from different stromal cell types [18,19,20, 48]. Activation is triggered by multiple stimuli including hypoxia, injury-induced platelet activation and the secretion of potent factors by epithelial cells such as transforming growth factor- (TGF-β), platelet-derived growth factor (PDGF) and basic fibroblast growth factor (bFGF), resulting in increased proliferation, increased contractility, and expression of activation markers such as αSMA, PDGFRα/β, FAP, podoplanin (PDPN) and desmin [18, 20]. In the context of cancer, chronically activated fibroblasts/CAFs contribute to aberrant ECM deposition and desmoplasia, promote angiogenesis and regulate the infiltration and polarization of myeloid cells attracted to tumors [19]. A complex and intricate network of signaling pathways and crosstalk with epithelial cells and other stromal cells perpetuate the activation status of CAFs in tumors [18, 49, 50]. Most of the well-described tumor-promoting functions of CAFs happen in the context of paracrine and juxtracrine signaling via the secretion of multiple growth factors including TGF-β, connective tissue growth factor (CTGF), stromal-derived growth factor (SDF-1) vascular endothelial growth factor (VEGF), osteopontin (OPN) or hepatocyte growth factor (HGF) just to mention some, as well as a myriad of cytokines and chemokines such as IL-6, IL-8, IL-1, CXCL2, CXCL5, CXCL12/SDF-1a, CCL20 and others [18, 20, 50].

With the advent of single-cell sequencing, the complexity of CAF biology during tumor development and across different tumor entities has gradually become more evident [44, 51, 52]. CAFs, which were originally perceived as a homogenous population, are now understood to be a mixture of different fibroblast phenotypes with distinct behavior, comprising both tumor-promoting and tumor-restraining subtypes [46, 53,54,55]. Despite the proven existence of CAF diversity in both preclinical and clinical settings [56], the context-dependent roles of different CAF populations and their interchangeable plasticity remain largely unknown [56]. In this review, we have not put emphasis on the CAF diversity aspect since CAF heterogeneity has not been studied in the context of radiation or radiotherapy yet.

Role of senescent fibroblasts in cancer development

Cellular senescence is an established tumor suppressive mechanism that halts the proliferation of premalignant cells [37]. Replicative senescence in a physiological context is frequently associated with aging [38, 57], and it is attributed to the gradual loss of telomere length after repeated cell divisions, whereas premature cell senescence or accelerated senescence is normally triggered by exogenous stresses, including genotoxic anticancer treatments such as chemotherapy or radiotherapy [58, 59]. Despite its tumor suppressive nature, mounting evidence indicate that senescent cells can also promote tumor progression via the senescence-associated secretory phenotype or SASP [60].

In the context of cancer, senescent cells can act as a double-edged sword. On one hand, SASP promotes tissue repair through the induction of plasticity and stemness and participates in the clearance of cancer and damaged cells by attracting phagocytes, NK cells and other immune cells [61, 62]. Induction of cell senescence in fibroblasts also limit the development of desmoplastic reactions and fibrosis [63]. On the other hand, the presence of senescent cells in the tumor stroma or its surroundings can aid in the establishment of an immunosuppressive [64, 65], pro-angiogenic [66], pro-inflammatory and catabolic microenvironment [33, 67, 68] that stimulate tumor growth and cancer cell dissemination.

Senescence-associated alterations in the secretion of matricellular proteins and ECM constituents by fibroblasts have been shown to create a favorable milieu for tumor development [33]. The observed upregulation of MMPs, cathepsins, ADAMTs (a disintegrin and metalloproteinase with thrombospondin motifs) and other proteases, together with the downregulation of tissue inhibitors of matrix metalloproteinases (TIMPs) contribute to a catabolic environment that supports tumor cell invasion and metastasis. In contrast to ECM-degrading constituents, which are mainly overexpressed in senescent fibroblasts, ECM molecules have been reported to be downregulated in general [69, 70]. Senescent fibroblasts and mesenchymal cells can also promote cancer development by favoring epithelial-to-mesenchymal transition in premalignant and malignant cells [71, 72]. This phenomenon is mediated in a paracrine fashion by the release of molecules such as MMPs, IL-1β, IL-6 and reactive oxygen species (ROS). Additionally, senescent macrophages can directly or indirectly promote tumor vascularization by overexpressed secretion of pro-angiogenic factors and by promoting the recruitment and polarization of M2-macrophages [73]. Last, the SASP from both stromal and tumor cells promotes tumor growth by establishing a microenvironment that is immunosuppressive. Such induction is mediated by the secretion of immunosuppressive (Th2) cytokines that favor the recruitment of myeloid-derived suppressor cells (MDSCs) and consequently inhibition of CD8 + T-lymphocyte-mediated killing of tumor cells [64].

Pro-tumorigenic functions of irradiated (senescent) normal fibroblasts

Ionizing radiation has been shown to drive both stromal fibroblasts and cancer cells to premature senescence [30, 74, 75]. Accordingly, therapy-induced cell senescence has become a recognized side effect of anticancer treatments with potential to mediate substantial impact on therapy outcomes [59]. Soluble SASP elements such as MMPs have been associated with pro-tumorigenic effects from radiation-induced senescent fibroblasts in vitro and in vivo [30, 76, 77]. Moreover, insoluble (cell-associated) molecules such as syndecan-1 have also been shown to be overexpressed in radiation-induced senescent breast stromal fibroblasts, which reportedly have been demonstrated to mediate enhanced tumor progression [78].

In the context of radiotherapy, accumulated evidence reveals that on one hand, radiation-induced premature senescence instead of apoptosis is a major mode of cell fate in irradiated fibroblasts, and on the other hand, radiation-induced senescence is a dose-dependent factor, with higher doses being more effective in inducing premature senescence [26, 28]. Experiments related to the involvement of radiation-induced senescent fibroblasts in cancer promotion have usually been performed using normal human fibroblasts. In fact, the pro-malignant phenotype acquired by senescent normal fibroblasts post-RT have been thoroughly documented. Numerous in vitro studies have demonstrated increased invasiveness, proliferation rates and radio-resistance of tumor cells exposed to irradiated fibroblast cell lines (or their conditioned medium) when compared to non-irradiated cells (Table 1). Different soluble signal molecules have been proposed to be responsible for the radiation-induced enhanced effects including HGF [79], MMPs [80], TGF-β [81] as well as interleukin (IL) 6 and IL-8 [82]. There are also reports of pro-tumorigenic effects exerted by irradiated fibroblasts upon co-transplantation in in vivo models [30, 83]. Additionally, increased tumor incidence and growth have been observed in animal models when both pre-malignant and malignant cells are injected in pre-irradiated tissues [80, 84].

Table 1 Tumorigenic effects exerted by prematurely senescent (RT-induced) normal fibroblasts

Collectively, the accumulated knowledge clearly indicate that radiation of normal tissue fibroblasts may turn them pro-tumorigenic via acquisition of a senescent phenotype and the associated pro-tumorigenic SASP. Also, the accumulation of senescent fibroblasts in premalignant tissues by processes of normal aging or genotoxic stress (radiation) may create a favorable environment for tumor initiation and growth (Table 1).

Role of (non-senescent) CAFs in radioprotection

Aiming at understanding the contribution of CAFs in tumor radio-resistance, some groups have investigated the potential radioprotective effects exerted by CAFs (non-irradiated/non-senescent) on cancer cells (Table 2). Several in vitro studies have demonstrated radioprotective effects of CAF conditioned medium on the survival and colony-forming abilities of cervical cancer [89] and pancreatic cancer cells [90]. Also, radioprotective effects from CAFs in co-culture with NSCLC cell lines have been described [91]. In preclinical models of melanoma and lung cancer, elevated expression of insulin-like growth factor 1 (IGF-1) and the chemokine CXCL12/SDF-1 by non-irradiated CAFs have been shown to be responsible for radioprotective effects on cancer cells [92]. In a study by Zhang et al. [93], CAF-derived CXCL1 was suggested to be accountable for the induction of a radioresistant phenotype in esophageal squamous cancer (ESCC), by promoting a reduction in ROS scavenging enzyme superoxide dismutase 1 (SOD-1) in cancer cells. Whereas in a pancreatic cancer model, it has been proposed that increased expression of TGF-β and possible other soluble signal molecules from pancreatic stellate cells (PSCs) promoted EMT changes in tumor cells and acquisition of a radioresistant phenotype [83]. Moreover, in a recent study by Ebbing et al. [94], using organoids and in vivo PDX models of esophageal cancer, authors point to CAF-derived IL-6 as a major soluble factor responsible for EMT induction and therapy resistance.

Table 2 Studies reporting on radioprotective effects exerted by CAFs (non-irradiated)

In clinical settings, the enhanced expression of CAF-specific markers or CAF-related gene signatures in tumor specimens have shown potential to predict responses to radio(chemo)therapy, and is persistently associated with poor prognosis in different types of cancers, including prostate cancer [95], cervical cancer [96], breast cancer [97], colon cancer [98], oral squamous cell carcinoma [99] and rectal cancer [100] (Table 3). Such consistency in clinical findings suggest that CAFs may play important roles in the conferral of radiation resistance phenotypes across many different solid malignancies.

Table 3 Studies reporting on CAF-related predictive/prognostic markers in clinical radio(chemo) therapy

Effects of RT on CAF tumorigenic functions

In addition to the general radioprotective functions assigned to CAFs, some studies claim that radiation exposure is amplifying the intrinsic radioprotective and pro-malignant effects exerted by CAFs (Table 4). In a study by Li et al. [101], irradiated CAFs provoked induction of epithelial mesenchymal transition (EMT) and enhanced invasive capacity of pancreatic cancer cells in co-cultures. RT-activated CAFs were found to excrete increased levels of CXCL12/SDF-1, ultimately promoting a mesenchymal phenotype in cancer cells and aiding to the overall tumor progression. Again, in a pancreatic cancer model, Mantoni et al. [102] demonstrated that pancreatic stellate cells (PSC) promote radioprotection and stimulate proliferation of pancreatic cancer cells in direct co-cultures and after co-injections in animal models. In this study, interfering with β1-integrin signaling abolished the radioprotective effects. In a third preclinical study performed on pancreatic cancer, authors show that induction of iNOS and nitric oxide (NO) in CAFs by RT is responsible for elevation of intratumoral pH and induction of a proinflammatory phenotype in tumor cells in a NFκB-dependent manner [103]. In a colorectal cancer model, Tommelein et al. [104] found increased IGF signaling from irradiated CAFs, and both IGF1 and IGF-binding proteins (IGFBP2) levels were elevated in supernatants from irradiated versus non-irradiated CAFs. In another preclinical study of colorectal cancer (CRC), upon irradiation, tumor-derived interleukin1α (IL-1α) mediated polarization of cancer-associated fibroblasts (CAFs) towards a pro-inflammatory pro-tumorigenic phenotype [105]. Authors demonstrated that IL-1-dependent signaling elevates oxidative DNA damage in iCAFs, which upon irradiation undergo senescence. This causes tissue remodeling and therapy resistance that can be overcome by inhibiting IL-1. Finally, in a recent study by Meng et al. [106], authors demonstrated that radiation-induced senescent CAFs promote non-small cell lung cancer (NSCLC) cell proliferation and radio-resistance through activation of the JAK/STAT pathway in tumor cells. Selective targeting of senescent fibroblast with a senotoxic agent was able to radio-sensitize tumors.

Table 4 Studies reporting on pro-& anti-tumorigenic effects exerted by irradiated CAFs

Conversely to the current view proposing a radiation-enhanced activation of CAFs, some studies document a loss of CAF pro-tumorigenic functions after irradiation (Table 4). In an in vivo study by Grinde et al. [107], the tumor enhancing effects exerted by NSCLC-CAFs after co-injection with A549 cells in nude mice was lost when CAFs were irradiated prior to implantation. In an in vitro study performed with NSCLC-CAFs, authors demonstrate changes in the secretory profile CAFs upon a single-high dose (1 × 18 Gy) radiation. However, in functional assays, they report no effects of conditioned media from irradiated CAFs on the proliferative or migratory capacity of tumor cells, and reduced migration rates on endothelial cells (HUVECs) [108]. Arshad et al. [87] reported similar findings, showing that murine lung CAFs did not affect the intrinsic radio-sensitivity of cancer cells. In contrast, reduced expression of TGF-β and MMPs were observed in co-culture supernatants after exposure to (1 × 10 Gy) radiation. In another study by Steer et al., the radio-protective and long-term survival effects of CAFs over cancer cells were studied in 2D and 3D in vitro systems, using different sets of fibroblasts and tumor cell lines [88]. The outcomes were inconsistent among different fibroblast-tumor cell combinations. Similar observations were obtained after co-implantation of cells in xenografts. Some authors have suggested that cancer promotion by senescent stromal cells may be restricted to certain organs and tissue types and claim that the tumorigenic properties of senescent cells need to be validated in other tissues than subcutaneous lesions [109].

CAFs impact on RT and vice-versa: observations from the clinics

In clinical settings, radiotherapy continue being a safe and efficient way to treat most known solid malignancies [1]. Exacerbated tumor growth after curative, adjuvant or neoadjuvant (chemo)radiotherapy is scantly observed, even though local tumor progression under or post-treatment may occur in minor subgroups of patients. In contrast, improved outcomes are frequently observed when radiation is used pre- or post-operative as opposed to surgery alone. Such long-term benefit from RT treatment have been reported for many different types of malignancies, ranging from early-stage breast cancer [110] to locally advanced colorectal cancer [111]. Considering that nearly all solid neoplasms contain tumor stroma and CAFs at all developmental stages, the evidence from the clinics suggests that the potential activation of CAFs post-RT (if happening at all) is not a dominant force that dictate the fate of the therapy. On the other hand, there is not much evidence on the impact of radiotherapy on CAFs in clinical settings. A study by Verset et al. on rectal cancer explored the impact of (chemo)radiotherapy on CAFs by comparing α-SMA/KI-67 ratios in tumor specimens collected before and after radiotherapy. Results showed that the α-SMA/neoplastic epithelial area ratio was higher after neoadjuvant therapy, and that α-SMA/epithelial area ratio was an adverse prognostic factor regarding recurrence-free survival [98]. Studies showing prognostic and predictive potential of CAF-markers in the clinics have been presented in the previous chapter (Table 3).

Concluding remarks

Scientific efforts over the last couple of decades have uncovered the important role played by the different elements of the tumor stroma in (radio)therapy outcomes. Cancer-associated fibroblasts have been proposed to participate significantly in tumor responses to radiotherapy. However, despite their prominent participation in tumor development and therapy resistance, the impact of RT on CAFs and the role of CAFs on RT outcomes remain elusive. Most existing literature in this field argue that radiation mediate changes in the mesenchymal components that favor tumor establishment and progression, however the impact that RT has on CAFs in vivo and the downstream effects of the potential RT-mediated changes remain controversial and still insufficiently investigated.

A potential source of controversy may emerge from the unrecognized fact that CAFs and normal fibroblasts are different cell entities, behaving differently, and probably responding differently to radiation exposure even when considering that both may survive and turn senescent after exposure to substantial radiation doses. Collectively, most published studies on the effects of radiation performed with normal tissue fibroblasts or fibroblast cell lines demonstrate enhanced pro-tumorigenic functions from irradiated (senescent) cells induced primarily in a paracrine fashion, by increased secretion of soluble growth factors, inflammatory mediators, and proteolytic enzymes (Table 1). Moreover, numerous studies have documented enhanced radioprotection of tumor cells cultured in the presence of (non-irradiated) CAFs (Table 2). These observations are in accordance with other studies performed on clinical specimens, demonstrating clear associations between high expression levels of CAF markers or CAF signature genes and poor prognosis (Table 3).

Findings on the impact of RT on CAFs and on the role of irradiated CAFs on tumor cell behavior are more controversial. While some studies claim that radiation exposure may affect CAF negatively through growth arrest and impaired mobility, others argue that exposure of CAFs to radiation can promote a more aggressive phenotype capable of conferring enhanced radio-resistance on tumor cells (Table 4). Most published studies have been performed on in vitro culture conditions, using single radiation doses or regimens, and have collected data at specific time-points, normally few hours/days post IR. These approaches gloss over potential differences related to different RT-regimens, that are crucial factors in modern radiotherapy [112] and radiotherapy-immunotherapy settings [4, 113,114,115,116] and also overlook potential long-term effects of RT on CAFs. Besides, most preclinical in vivo studies use co-injections of tumor cells and fibroblasts/CAFs orthotopically or in subcutaneous pockets. In transplantation experiments with admixed cells, it is frequently observed that non-tumoral cells disappear quickly after implantation [76, 107, 117]. Tumor growth effects in such experiments are believed to be related to initial tumor engraftment rather than tumor growth, and are therefore not optimal to study effects from transplanted (irradiated) cells. Additionally, there is little or no evidence on the impact that RT has on CAFs in vivo (preclinical) or in clinical settings. Preclinical models reproducing stroma-rich tumors resembling the human scenario, where endogenous CAFs can be targeted, tracked and/or regulated, could represent attractive models to explore CAF-mediated effects from RT and vice versa. In future efforts, the CAF heterogeneity aspect, including aged-fibroblasts, should also be taken into consideration. It is still unknown which CAF subtype (if any) is mostly responsible for conferring tumor cell radio-resistance. We still don’t know if radiation affects all CAF subtypes in the same way, or if the induction of cell senescence by RT unifies all subpopulations into a single CAF cell phenotype with specific functions.

In the clinics, radiotherapy is considered to be a safe and effective way to treat non-metastatic cancers with curative intent. Moreover, the use of RT in adjuvant and neoadjuvant settings has been proved to give better survival rates than surgery alone in many different tumor types [1]. Considering that basically all solid neoplasms contain CAFs at all developmental stages, the evidence from the clinics suggests that the potential activation of CAFs post-RT is not a dominant force that dictate the fate of the therapy. At least, it remains unexplained why local tumor recurrence or exacerbated tumor growth is not more frequently observed following RT, if we assume that radiation triggers enhanced pro-tumorigenic functions in CAFs. Further research using appropriate models to study CAFs and more information from clinical research is needed to clarify the ultimate role played by CAFs on radiotherapy.

Search strategy and selection criteria

Data for this review were identified by searches of MEDLINE, PubMed and references from relevant articles using search terms such as “cancer-associated fibroblasts”, “tumor microenvironment”, “radiation”, “radiotherapy”, “cell senescence”, “stress-induced senescence”. Only articles published between 1999 and 2022 have been included.

Availability of data and materials

Data sharing not applicable to this article as no datasets have been generated or analyzed for the creation of the study.

Abbreviations

α-SMA:

Alpha-smooth muscle actin

CAFs:

Cancer-associated fibroblasts

ECM:

Extracellular matrix

EMT:

Epithelial-mesenchymal transition

IR:

Ionizing radiation

MMPs:

Matrix metalloproteinases

NFs:

Normal fibroblasts

NSCLC:

Non-small cell lung cancer

RT:

Radiotherapy

SASP:

Senescence-associated secretory phenotype

TME:

Tumor microenvironment

References

  1. Sharma RA, Plummer R, Stock JK, Greenhalgh TA, Ataman O, Kelly S, et al. Clinical development of new drug-radiotherapy combinations. Nat Rev Clin Oncol. 2016;13(10):627–42.

    Article  CAS  PubMed  Google Scholar 

  2. Begg AC, Stewart FA, Vens C. Strategies to improve radiotherapy with targeted drugs. Nat Rev Cancer. 2011;11(4):239–53.

    Article  CAS  PubMed  Google Scholar 

  3. Barker HE, Paget JT, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer. 2015;15(7):409–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hellevik T, Martinez-Zubiaurre I. Radiotherapy and the Tumor Stroma: The Importance of Dose and Fractionation. Front Oncol. 2014;4:1.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Nia HT, Munn LL, Jain RK. Physical traits of cancer. Science. 2020;370(6516):eaaz0868.

  6. Barcellos-Hoff MH. The radiobiology of TGFbeta. Semin Cancer Biol. 2022;86(Pt 3):857–67.

    Article  CAS  PubMed  Google Scholar 

  7. Lan Y, Moustafa M, Knoll M, Xu C, Furkel J, Lazorchak A, et al. Simultaneous targeting of TGF-beta/PD-L1 synergizes with radiotherapy by reprogramming the tumor microenvironment to overcome immune evasion. Cancer Cell. 2021;39(10):1388–403.

    Article  CAS  PubMed  Google Scholar 

  8. Park CC, Zhang HJ, Yao ES, Park CJ, Bissell MJ. Beta1 integrin inhibition dramatically enhances radiotherapy efficacy in human breast cancer xenografts. Cancer Res. 2008;68(11):4398–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Garcia-Barros M, Paris F, Cordon-Cardo C, Lyden D, Rafii S, Haimovitz-Friedman A, et al. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science. 2003;300(5622):1155–9.

    Article  CAS  PubMed  Google Scholar 

  10. Park HJ, Griffin RJ, Hui S, Levitt SH, Song CW. Radiation-induced vascular damage in tumors: implications of vascular damage in ablative hypofractionated radiotherapy (SBRT and SRS). Radiat Res. 2012;177(3):311–27.

    Article  CAS  PubMed  Google Scholar 

  11. Martinez-Zubiaurre I, Chalmers AJ, Hellevik T. Radiation-induced transformation of immunoregulatory networks in the tumor stroma. Front Immunol. 2018;9:1679.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Stapleton S, Jaffray D, Milosevic M. Radiation effects on the tumor microenvironment: implications for nanomedicine delivery. Adv Drug Deliv Rev. 2017;109:119–30.

  13. Surace L, Guckenberger M, van den Broek M. Radiation holidays stimulate tumor immunity. Oncotarget. 2015;6(18):15716–7.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Schaue D, Micewicz ED, Ratikan JA, Xie MW, Cheng G, McBride WH. Radiation and inflammation. Semin Radiat Oncol. 2015;25(1):4–10.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Gupta A, Probst HC, Vuong V, Landshammer A, Muth S, Yagita H, et al. Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J Immunol. 2012;189(2):558–66.

    Article  CAS  PubMed  Google Scholar 

  16. Burnette BC, Liang H, Lee Y, Chlewicki L, Khodarev NN, Weichselbaum RR, et al. The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. 2011;71(7):2488–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rodriguez-Ruiz ME, Vitale I, Harrington KJ, Melero I, Galluzzi L. Immunological impact of cell death signaling driven by radiation on the tumor microenvironment. Nat Immunol. 2020;21(2):120–34.

    Article  CAS  PubMed  Google Scholar 

  18. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16(9):582–98.

    Article  CAS  PubMed  Google Scholar 

  19. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020;20(3):174–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Biffi G, Tuveson DA. Diversity and biology of cancer-associated fibroblasts. Physiol Rev. 2021;101(1):147–76.

    Article  CAS  PubMed  Google Scholar 

  21. Stieb S, McDonald B, Gronberg M, Engeseth GM, He R, Fuller CD. Imaging for target delineation and treatment planning in radiation oncology: current and emerging techniques. Hematol Oncol Clin N Am. 2019;33(6):963–75.

    Article  Google Scholar 

  22. Giglioli FR, Clemente S, Esposito M, Fiandra C, Marino C, Russo S, et al. Frontiers in planning optimization for lung SBRT. Phys Med. 2017;44:163–70.

    Article  PubMed  Google Scholar 

  23. Wang Z, Tang Y, Tan Y, Wei Q, Yu W. Cancer-associated fibroblasts in radiotherapy: challenges and new opportunities. Cell Commun Signal. 2019;17(1):47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ji X, Zhu X, Lu X. Effect of cancer-associated fibroblasts on radiosensitivity of cancer cells. Future Oncol. 2017;13(17):1537–50.

    Article  CAS  PubMed  Google Scholar 

  25. Hellevik T, Berzaghi R, Lode K, Islam A, Martinez-Zubiaurre I. Immunobiology of cancer-associated fibroblasts in the context of radiotherapy. J Transl Med. 2021;19(1):437.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Hellevik T, Pettersen I, Berg V, Winberg JO, Moe BT, Bartnes K, et al. Cancer-associated fibroblasts from human NSCLC survive ablative doses of radiation but their invasive capacity is reduced. Radiat Oncol. 2012;7:59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Tachiiri S, Katagiri T, Tsunoda T, Oya N, Hiraoka M, Nakamura Y. Analysis of gene-expression profiles after gamma irradiation of normal human fibroblasts. Int J Radiat Oncol Biol Phys. 2006;64(1):272–9.

    Article  CAS  PubMed  Google Scholar 

  28. Berzaghi R, Islam A, Hellevik T, Martinez-Zubiaurre I. Secretion rates and protein composition of extracellular vesicles released by cancer-associated fibroblasts after radiation. J Radiat Res. 2021;62(3):401–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yang N, Lode K, Berzaghi R, Islam A, Martinez-Zubiaurre I, Hellevik T. Irradiated tumor fibroblasts avoid immune recognition and retain immunosuppressive functions over natural killer cells. Front Immunol. 2020;11: 602530.

    Article  CAS  PubMed  Google Scholar 

  30. Papadopoulou A, Kletsas D. Human lung fibroblasts prematurely senescent after exposure to ionizing radiation enhance the growth of malignant lung epithelial cells in vitro and in vivo. Int J Oncol. 2011;39(4):989–99.

    CAS  PubMed  Google Scholar 

  31. Pereira BI, Devine OP, Vukmanovic-Stejic M, Chambers ES, Subramanian P, Patel N, et al. Senescent cells evade immune clearance via HLA-E-mediated NK and CD8(+) T cell inhibition. Nat Commun. 2019;10(1):2387.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Lecot P, Alimirah F, Desprez PY, Campisi J, Wiley C. Context-dependent effects of cellular senescence in cancer development. Br J Cancer. 2016;114(11):1180–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Mavrogonatou E, Pratsinis H, Kletsas D. The role of senescence in cancer development. Semin Cancer Biol. 2020;62:182–91.

    Article  CAS  PubMed  Google Scholar 

  34. Wang L, Lankhorst L, Bernards R. Exploiting senescence for the treatment of cancer. Nat Rev Cancer. 2022;22(6):340–55.

    Article  CAS  PubMed  Google Scholar 

  35. Wyld L, Bellantuono I, Tchkonia T, Morgan J, Turner O, Foss F, et al. Senescence and cancer: a review of clinical implications of senescence and senotherapies. Cancers. 2020;12(8):2134-53.

  36. Rodier F, Campisi J. Four faces of cellular senescence. J Cell Biol. 2011;192(4):547–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, et al. Cellular senescence: defining a path forward. Cell. 2019;179(4):813–27.

    Article  CAS  PubMed  Google Scholar 

  38. Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol. 2013;75:685–705.

    Article  CAS  PubMed  Google Scholar 

  39. Bao CH, Wang XT, Ma W, Wang NN, Un Nesa E, Wang JB, et al. Irradiated fibroblasts promote epithelial-mesenchymal transition and HDGF expression of esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 2015;458(2):441–7.

    Article  CAS  PubMed  Google Scholar 

  40. Hawsawi NM, Ghebeh H, Hendrayani SF, Tulbah A, Al-Eid M, Al-Tweigeri T, et al. Breast carcinoma-associated fibroblasts and their counterparts display neoplastic-specific changes. Cancer Res. 2008;68(8):2717–25.

    Article  CAS  PubMed  Google Scholar 

  41. Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 2010;17(2):135–47.

    Article  CAS  PubMed  Google Scholar 

  42. Hoffman D, Dragojevic I, Hoisak J, Hoopes D, Manger R. Lung stereotactic body radiation therapy (SBRT) dose gradient and PTV volume: a retrospective multi-center analysis. Radiat Oncol. 2019;14(1):162.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Filice A, Casali M, Ciammella P, Galaverni M, Fioroni F, Iotti C, et al. Radiotherapy planning and molecular imaging in lung cancer. Curr Radiopharm. 2020;13(3):204–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Buechler MB, Pradhan RN, Krishnamurty AT, Cox C, Calviello AK, Wang AW, et al. Cross-tissue organization of the fibroblast lineage. Nature. 2021;593(7860):575–9.

    Article  CAS  PubMed  Google Scholar 

  45. Lendahl U, Muhl L, Betsholtz C. Identification, discrimination and heterogeneity of fibroblasts. Nat Commun. 2022;13(1):3409.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ohlund D, Elyada E, Tuveson D. Fibroblast heterogeneity in the cancer wound. J Exp Med. 2014;211(8):1503–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lynch MD, Watt FM. Fibroblast heterogeneity: implications for human disease. J Clin Invest. 2018;128(1):26–35.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Arina A, Idel C, Hyjek EM, Alegre ML, Wang Y, Bindokas VP, et al. Tumor-associated fibroblasts predominantly come from local and not circulating precursors. Proc Natl Acad Sci U S A. 2016;113(27):7551–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Mishra P, NBanerjee D, Ben-Baruch A. Chemokines at the crossroads of tumor-fibroblast interactions that promote malignancy. J Leukoc Biol. 2011;89:31–9.

  50. Jung JG, Le A. Targeting Metabolic cross talk between cancer cells and cancer-associated fibroblasts. Adv Exp Med Biol. 2018;1063:167–78.

    Article  CAS  PubMed  Google Scholar 

  51. Dominguez CX, Muller S, Keerthivasan S, Koeppen H, Hung J, Gierke S, et al. Single-Cell RNA sequencing reveals stromal evolution into LRRC15(+) myofibroblasts as a determinant of patient response to cancer immunotherapy. Cancer Discov. 2020;10(2):232–53.

    Article  CAS  PubMed  Google Scholar 

  52. Joseph DB, Henry GH, Malewska A, Reese JC, Mauck RJ, Gahan JC, et al. Single-cell analysis of mouse and human prostate reveals novel fibroblasts with specialized distribution and microenvironment interactions. J Pathol. 2021;255(2):141–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ohlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. 2017;214(3):579–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Menezes S, Okail MH, Jalil SMA, Kocher HM, Cameron AJM. Cancer-associated fibroblasts in pancreatic cancer: new subtypes, new markers, new targets. J Pathol. 2022;257(4):526–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Jenkins BH, Buckingham JF, Hanley CJ, Thomas GJ. Targeting cancer-associated fibroblasts: challenges, opportunities and future directions. Pharmacol Ther. 2022;240: 108231.

    Article  CAS  PubMed  Google Scholar 

  56. Chen Y, McAndrews KM, Kalluri R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat Rev Clin Oncol. 2021;18(12):792–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Harley CB, Futcher AB, Greider CW. Telomeres shorten during ageing of human fibroblasts. Nature. 1990;345(6274):458–60.

    Article  CAS  PubMed  Google Scholar 

  58. Toussaint O, Dumont P, Remacle J, Dierick JF, Pascal T, Frippiat C, et al. Stress-induced premature senescence or stress-induced senescence-like phenotype: One in vivo reality, two possible definitions? ScientificWorldJournal. 2002;2:230–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Gewirtz DA, Holt SE, Elmore LW. Accelerated senescence: an emerging role in tumor cell response to chemotherapy and radiation. Biochem Pharmacol. 2008;76(8):947–57.

    Article  CAS  PubMed  Google Scholar 

  60. Kuilman T, Peeper DS. Senescence-messaging secretome: SMS-ing cellular stress. Nat Rev Cancer. 2009;9(2):81–94.

    Article  CAS  PubMed  Google Scholar 

  61. Giannoula Y, Kroemer G, Pietrocola F. Cellular senescence and the host immune system in aging and age-related disorders. Biomed J. 2023.

  62. Kale A, Sharma A, Stolzing A, Desprez PY, Campisi J. Role of immune cells in the removal of deleterious senescent cells. Immun Ageing. 2020;17:16.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C, et al. Senescence of activated stellate cells limits liver fibrosis. Cell. 2008;134(4):657–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Toso A, Revandkar A, Di Mitri D, Guccini I, Proietti M, Sarti M, et al. Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Rep. 2014;9(1):75–89.

    Article  CAS  PubMed  Google Scholar 

  65. Buechler MB, Turley SJ. A short field guide to fibroblast function in immunity. Semin Immunol. 2018;35:48–58.

    Article  CAS  PubMed  Google Scholar 

  66. Davalos AR, Coppe JP, Campisi J, Desprez PY. Senescent cells as a source of inflammatory factors for tumor progression. Cancer Metastasis Rev. 2010;29(2):273–83.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 2008;6(12):2853–68.

    Article  CAS  PubMed  Google Scholar 

  68. Rodier F, Munoz DP, Teachenor R, Chu V, Le O, Bhaumik D, et al. DNA-SCARS: distinct nuclear structures that sustain damage-induced senescence growth arrest and inflammatory cytokine secretion. J Cell Sci. 2011;124(Pt 1):68–81.

    Article  CAS  PubMed  Google Scholar 

  69. Mavrogonatou E, Pratsinis H, Papadopoulou A, Karamanos NK, Kletsas D. Extracellular matrix alterations in senescent cells and their significance in tissue homeostasis. Matrix Biol. 2019;75–76:27–42.

    Article  PubMed  Google Scholar 

  70. Bizot-Foulon V, Bouchard B, Hornebeck W, Dubertret L, Bertaux B. Uncoordinate expressions of type I and III collagens, collagenase and tissue inhibitor of matrix metalloproteinase 1 along in vitro proliferative life span of human skin fibroblasts. Regulation by all-trans retinoic acid. Cell Biol Int. 1995;19(2):129–35.

    Article  CAS  PubMed  Google Scholar 

  71. Parrinello S, Coppe JP, Krtolica A, Campisi J. Stromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiation. J Cell Sci. 2005;118(Pt 3):485–96.

    Article  CAS  PubMed  Google Scholar 

  72. Di GH, Liu Y, Lu Y, Liu J, Wu C, Duan HF. IL-6 secreted from senescent mesenchymal stem cells promotes proliferation and migration of breast cancer cells. PLoS ONE. 2014;9(11): e113572.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Kelly J, Ali Khan A, Yin J, Ferguson TA, Apte RS. Senescence regulates macrophage activation and angiogenic fate at sites of tissue injury in mice. J Clin Invest. 2007;117(11):3421–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Jones KR, Elmore LW, Jackson-Cook C, Demasters G, Povirk LF, Holt SE, et al. p53-Dependent accelerated senescence induced by ionizing radiation in breast tumour cells. Int J Radiat Biol. 2005;81(6):445–58.

    Article  CAS  PubMed  Google Scholar 

  75. Mirzayans R, Scott A, Cameron M, Murray D. Induction of accelerated senescence by gamma radiation in human solid tumor-derived cell lines expressing wild-type TP53. Radiat Res. 2005;163(1):53–62.

    Article  CAS  PubMed  Google Scholar 

  76. Liu D, Hornsby PJ. Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Res. 2007;67(7):3117–26.

    Article  CAS  PubMed  Google Scholar 

  77. Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Liakou E, Mavrogonatou E, Pratsinis H, Rizou S, Evangelou K, Panagiotou PN, et al. Ionizing radiation-mediated premature senescence and paracrine interactions with cancer cells enhance the expression of syndecan 1 in human breast stromal fibroblasts: the role of TGF-beta. Aging. 2016;8(8):1650–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Patel ZS, Grugan KD, Rustgi AK, Cucinotta FA, Huff JL. Ionizing radiation enhances esophageal epithelial cell migration and invasion through a paracrine mechanism involving stromal-derived hepatocyte growth factor. Radiat Res. 2012;177(2):200–8.

    Article  CAS  PubMed  Google Scholar 

  80. Tsai KK, Chuang EY, Little JB, Yuan ZM. Cellular mechanisms for low-dose ionizing radiation-induced perturbation of the breast tissue microenvironment. Cancer Res. 2005;65(15):6734–44.

    Article  CAS  PubMed  Google Scholar 

  81. Kamochi N, Nakashima M, Aoki S, Uchihashi K, Sugihara H, Toda S, et al. Irradiated fibroblast-induced bystander effects on invasive growth of squamous cell carcinoma under cancer-stromal cell interaction. Cancer Sci. 2008;99(12):2417–27.

    Article  CAS  PubMed  Google Scholar 

  82. Rodier F, Coppe JP, Patil CK, Hoeijmakers WA, Munoz DP, Raza SR, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009;11(8):973–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Al-Assar O, Demiciorglu F, Lunardi S, Gaspar-Carvalho MM, McKenna WG, Muschel RM, et al. Contextual regulation of pancreatic cancer stem cell phenotype and radioresistance by pancreatic stellate cells. Radiother Oncol. 2014;111(2):243–51.

    Article  PubMed  Google Scholar 

  84. Barcellos-Hoff MH, Ravani SA. Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res. 2000;60(5):1254–60.

    CAS  PubMed  Google Scholar 

  85. Ohuchida K, Mizumoto K, Murakami M, Qian LW, Sato N, Nagai E, et al. Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res. 2004;64(9):3215–22.

    Article  CAS  PubMed  Google Scholar 

  86. Tsai KK, Stuart J, Chuang YY, Little JB, Yuan ZM. Low-dose radiation-induced senescent stromal fibroblasts render nearby breast cancer cells radioresistant. Radiat Res. 2009;172(3):306–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Arshad A, Deutsch E, Vozenin MC. Simultaneous irradiation of fibroblasts and carcinoma cells repress the secretion of soluble factors able to stimulate carcinoma cell migration. PLoS ONE. 2015;10(1): e0115447.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Steer A, Cordes N, Jendrossek V, Klein D. Impact of cancer-associated fibroblast on the radiation-response of solid xenograft tumors. Front Mol Biosci. 2019;6:70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Chu TY, Yang JT, Huang TH, Liu HW. Crosstalk with cancer-associated fibroblasts increases the growth and radiation survival of cervical cancer cells. Radiat Res. 2014;181(5):540–7.

    Article  CAS  PubMed  Google Scholar 

  90. Hwang RF, Moore T, Arumugam T, Ramachandran V, Amos KD, Rivera A, et al. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 2008;68(3):918–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Ji X, Ji J, Shan F, Zhang Y, Chen Y, Lu X. Cancer-associated fibroblasts from NSCLC promote the radioresistance in lung cancer cell lines. Int J Clin Exp Med. 2015;8(5):7002–8.

    PubMed  PubMed Central  Google Scholar 

  92. Wang Y, Gan G, Wang B, Wu J, Cao Y, Zhu D, et al. Cancer-associated fibroblasts promote irradiated cancer cell recovery through autophagy. EBioMedicine. 2017;17:45–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zhang H, Yue J, Jiang Z, Zhou R, Xie R, Xu Y, et al. CAF-secreted CXCL1 conferred radioresistance by regulating DNA damage response in a ROS-dependent manner in esophageal squamous cell carcinoma. Cell Death Dis. 2017;8(5): e2790.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Ebbing EA, van der Zalm AP, Steins A, Creemers A, Hermsen S, Rentenaar R, et al. Stromal-derived interleukin 6 drives epithelial-to-mesenchymal transition and therapy resistance in esophageal adenocarcinoma. Proc Natl Acad Sci U S A. 2019;116(6):2237–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Zhang R, Liu F. Cancer-associated fibroblast-derived gene signatures predict radiotherapeutic survival in prostate cancer patients. J Transl Med. 2022;20(1):453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Kim KH, Chang JS, Byun HK, Kim YB. A novel gene signature associated with poor response to chemoradiotherapy in patients with locally advanced cervical cancer. J Gynecol Oncol. 2022;33(1): e7.

    Article  CAS  PubMed  Google Scholar 

  97. Strell C, Stenmark Tullberg A, Jetne Edelmann R, Akslen LA, Malmstrom P, Ferno M, et al. Prognostic and predictive impact of stroma cells defined by PDGFRb expression in early breast cancer: results from the randomized SweBCG91RT trial. Breast Cancer Res Treat. 2021;187(1):45–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Verset L, Tommelein J, Moles Lopez X, Decaestecker C, Boterberg T, De Vlieghere E, et al. Impact of neoadjuvant therapy on cancer-associated fibroblasts in rectal cancer. Radiother Oncol. 2015;116(3):449–54.

    Article  PubMed  Google Scholar 

  99. Matsuoka Y, Yoshida R, Nakayama H, Nagata M, Hirosue A, Tanaka T, et al. The tumour stromal features are associated with resistance to 5-FU-based chemoradiotherapy and a poor prognosis in patients with oral squamous cell carcinoma. APMIS. 2015;123(3):205–14.

    Article  CAS  PubMed  Google Scholar 

  100. Saigusa S, Toiyama Y, Tanaka K, Yokoe T, Okugawa Y, Fujikawa H, et al. Cancer-associated fibroblasts correlate with poor prognosis in rectal cancer after chemoradiotherapy. Int J Oncol. 2011;38(3):655–63.

    Article  CAS  PubMed  Google Scholar 

  101. Li D, Qu C, Ning Z, Wang H, Zang K, Zhuang L, et al. Radiation promotes epithelial-to-mesenchymal transition and invasion of pancreatic cancer cell by activating carcinoma-associated fibroblasts. Am J Cancer Res. 2016;6(10):2192–206.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Mantoni TS, Lunardi S, Al-Assar O, Masamune A, Brunner TB. Pancreatic stellate cells radioprotect pancreatic cancer cells through beta1-integrin signaling. Cancer Res. 2011;71(10):3453–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Pereira PMR, Edwards KJ, Mandleywala K, Carter LM, Escorcia FE, Campesato LF, et al. iNOS regulates the therapeutic response of pancreatic cancer cells to radiotherapy. Cancer Res. 2020;80(8):1681–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Tommelein J, De Vlieghere E, Verset L, Melsens E, Leenders J, Descamps B, et al. Radiotherapy-activated cancer-associated fibroblasts promote tumor progression through paracrine IGF1R activation. Cancer Res. 2018;78(3):659–70.

    Article  CAS  PubMed  Google Scholar 

  105. Nicolas AM, Pesic M, Engel E, Ziegler PK, Diefenhardt M, Kennel KB, et al. Inflammatory fibroblasts mediate resistance to neoadjuvant therapy in rectal cancer. Cancer Cell. 2022;40(2):168–84.

    Article  CAS  PubMed  Google Scholar 

  106. Meng J, Li Y, Wan C, Sun Y, Dai X, Huang J, et al. Targeting senescence-like fibroblasts radiosensitizes non-small cell lung cancer and reduces radiation-induced pulmonary fibrosis. JCI Insight. 2021;6(23).

  107. Grinde MT, Vik J, Camilio KA, Martinez-Zubiaurre I, Hellevik T. Ionizing radiation abrogates the pro-tumorigenic capacity of cancer-associated fibroblasts co-implanted in xenografts. Sci Rep. 2017;7:46714.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Hellevik T, Pettersen I, Berg V, Bruun J, Bartnes K, Busund LT, et al. Changes in the secretory profile of NSCLC-associated fibroblasts after ablative radiotherapy: potential impact on angiogenesis and tumor growth. Transl Oncol. 2013;6(1):66–74.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Liu D, Hornsby PJ. Fibroblast stimulation of blood vessel development and cancer cell invasion in a subrenal capsule xenograft model: stress-induced premature senescence does not increase effect. Neoplasia. 2007;9(5):418–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. van Maaren MC, de Munck L, de Bock GH, Jobsen JJ, van Dalen T, Linn SC, et al. 10 year survival after breast-conserving surgery plus radiotherapy compared with mastectomy in early breast cancer in the Netherlands: a population-based study. Lancet Oncol. 2016;17(8):1158–70.

    Article  PubMed  Google Scholar 

  111. van Gijn W, Marijnen CA, Nagtegaal ID, Kranenbarg EM, Putter H, Wiggers T, et al. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer: 12-year follow-up of the multicentre, randomised controlled TME trial. Lancet Oncol. 2011;12(6):575–82.

    Article  PubMed  Google Scholar 

  112. Ramroth J, Cutter DJ, Darby SC, Higgins GS, McGale P, Partridge M, et al. Dose and fractionation in radiation therapy of curative intent for non-small cell lung cancer: meta-analysis of randomized trials. Int J Radiat Oncol Biol Phys. 2016;96(4):736–47.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Demaria S, Guha C, Schoenfeld J, Morris Z, Monjazeb A, Sikora A, et al. Radiation dose and fraction in immunotherapy: One-size regimen does not fit all settings, so how does one choose? Journal for immunotherapy of cancer. 2021;9(4).

  114. Dovedi SJ, Cheadle EJ, Popple AL, Poon E, Morrow M, Stewart R, et al. Fractionated radiation therapy stimulates antitumor immunity mediated by both resident and infiltrating polyclonal T-cell populations when combined with PD-1 blockade. Clin Cancer Res. 2017;23(18):5514–26.

    Article  CAS  PubMed  Google Scholar 

  115. Frey B, Hehlgans S, Rodel F, Gaipl US. Modulation of inflammation by low and high doses of ionizing radiation: Implications for benign and malign diseases. Cancer Lett. 2015;368(2):230–7.

    Article  CAS  PubMed  Google Scholar 

  116. Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, et al. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T Cell immunotherapy. Cancer Cell. 2013;24(5):589–602.

    Article  CAS  PubMed  Google Scholar 

  117. Fabris VT, Sahores A, Vanzulli SI, Colombo L, Molinolo AA, Lanari C, et al. Inoculated mammary carcinoma-associated fibroblasts: contribution to hormone independent tumor growth. BMC Cancer. 2010;10:293.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Open access funding provided by UiT The Arctic University of Norway (incl University Hospital of North Norway) This work was supported by grants from the Norwegian Regional Health Authorities (Grant #: HNF1423-18); The Norwegian Cancer Society (project ID#: 198164) and the Aakre Foundation at UiT.

Author information

Authors and Affiliations

Authors

Contributions

IM was the main contributor to the conception of the study. IMZ and TH drafted the manuscript and revised it critically for important intellectual content. The two authors read and approved the final version of the manuscript.

Corresponding author

Correspondence to Inigo Martinez-Zubiaurre.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Martinez-Zubiaurre, I., Hellevik, T. Cancer-associated fibroblasts in radiotherapy: Bystanders or protagonists?. Cell Commun Signal 21, 108 (2023). https://doi.org/10.1186/s12964-023-01093-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-023-01093-5