To simulate clinical situations, the present study established a novel sRRLI model based on BLM-induced pulmonary fibrosis mice. It was observed that most of the pre-existing ILD mice presented pathological DAD patterns and lung function loss after partial thoracic irradiation. These mice tended to develop severe pneumonitis, progressive fibrosis, and even fatal outcomes within six months post-IR, compared to RILI and BIPF mono-treatment groups. Clinically, a recent retrospective study showed that the incidence of post-treatment AE was higher in LC-ILD patients who received CCRT (54.5%) followed by radiotherapy (16.2%) and chemotherapy (15.6%), compared to non-ILD patients [14]. Even when relatively low-dose palliative thoracic radiotherapy was delivered, the rate of grade ≥ 3 RP was reported to be 13.7%. Particularly in patients with a higher pulmonary fibrosis score (score 3–5), the incidence of serious RP increased to 37.5% with eventual death [15].
Autopsy analysis or lung biopsy is scarce after treatment-related severe complications in the clinic. Using an in vivo mouse model, significant pathological changes in lung injury were detailed here. At the early stage, when lung injury in the RILI and BIPF groups gradually settled into relatively stable, the sRRLI mice were still predominated by the exudative DAD with apparent massive hyaline membranes and intra-alveolar fibrin. As time passed, at six months post-IR, the organizing DAD phase became the main pattern, characterized by inter-alveolar fibroblastic proliferation, septal collagen deposition, and fibrotic foci. Severe damage, if irreversible, could eventually result in repeated injury contributing to modality. Autopsy of the lethal mouse revealed overlapping patterns of exudative, proliferative, and fibrotic DAD in bilateral lungs. Such morphological changes may be responsible for the clinical symptoms observed in LC-ILD patients with progressive dyspnea, dry cough, declining lung function, and respiratory failure [16, 17].
Common mechanisms underlying different treatment-induced lung injuries were elucidated using transcriptomic analysis here. Signaling pathways relevant to cellular damage and repair, including p53, PI3K-Akt, MAPK, JAK-STAT, HIF-1, and cellular senescence, were universally activated. A recent report using scRNA-seq analysis showed that the activation of PI3K-Akt and p53 pathways in AEC cells participated in the progression of blast-induced lung damage via modulating autophagic and oxidative stress [18]. Besides, AKT-, MAPK-, or JAK-STAT-relevant inflammatory pathways are potential therapeutic targets for treating acute lung injury (ALI), and novel agents are under investigation [19–21]. Growing evidence suggests that cellular senescence is positively associated with pulmonary fibrosis. Upregulation of the senescence-related proteins p16INK4a and p21CIP1 were previously demonstrated in RIPF or BIPF models [22, 23]. Senescent cells display senescence-associated secretory phenotype, which is involved in promoting the lung fibroblast proliferation, myofibroblast activation, and ECM production [24, 25].
We also found that the common genes Cdkn1a, Fosb, and Serpine1 were among the core positions during the development of lung injury. Our previous study has validated that Cdkn1a, also known as p21, its expression level was significantly higher in the sRRLI mice and positively correlated with macrophage infiltration via regulating CCL7 secretion in vitro [8]. Moreover, Fosb proto-oncogene functions in regulating cell proliferation, differentiation, and transformation. It is a subunit gene of the AP-1 transcription factor, which plays a crucial role in lung fibroblasts by promoting macrophage activation and collagen production [26]. Additionally, PAI-1 (Serpine1) was recently found to be associated with downregulating re-alveolarization in ALI by reducing the AEC II self-renewal [27], and evidence supported that it was a druggable target for controlling lung cell senescence and fibrosis via inhibiting TGF-β pathway [28].
Furthermore, our findings suggested a central role of epithelial cell development and immune cell migration in the progression of severe lung injury. The hyperplastic pneumocytes in residual alveoli and the influx of macrophages and CD4 + lymphocytes were presented persistently during disease development. Cumulative evidence showed that M1/M2 macrophages participated in both the acute and chronic phases of lung disease. M2 macrophages mainly dominate the progression of pulmonary fibrosis via TGF-β signaling pathway during the rehabilitation period [29]. In the sRRLI model, macrophage pools exceeded in both ipsilateral and contralateral lungs so that radiation-induced abscopal lung injury was observed along with the activation of immune responses. This differs from previous observation that focal regions of macrophages existed only in ipsilateral lungs at 26 weeks post-IR [30]. Consistently, in some clinical cases after thoracic radiotherapy, extensive ground glass abnormalities and focal consolidations were usually observed spreading to bilateral lungs rather than being limited to the irradiated area on CT images [31].
Proinflammatory and profibrotic factors are considered crucial mediators in causing serious suffering. However, there are no recommendations for reliable biomarkers in the guidelines now due to their relatively low specificity. Experts have advised that patients with higher ILD gender age physiology (ILD-GAP) index scores should be considered when delivering treatments or only observation, as radiological usual interstitial pneumonia (UIP) pattern was significantly associated with thoracic radiotherapy-related life-threatening pneumonitis [17]. Most noteworthy, a higher level of KL-6 was found to be linked to more severe AE-ILD or treatment-related ILD and thus was proposed as a potential biomarker for poor prognosis [32, 33]. Besides, peripheral blood markers such as pretreatment NLR (≥ 3.0), and pretreatment ANC (≥ 5755) were reported to be associated with severe RILI [7]. We categorized some potential mediators in the early phase of the sRRLI here, including chemokines and chemokine receptors, ILs, TGF-β family, MMPs, and TNF-α. Other factors of growing interest include pro-platelet basic protein (PPBP), growth and differentiation factor 15 (GDF 15), and serum amyloid A (SAA), which have recently been implicated in the development of lung inflammation and fibrosis [34–36].