Skip to main content
Erschienen in: Journal of Translational Medicine 1/2024

Open Access 01.12.2024 | Review

Inhibitor of PD-1/PD-L1: a new approach may be beneficial for the treatment of idiopathic pulmonary fibrosis

verfasst von: Jie Tan, Qianfei Xue, Xiao Hu, Junling Yang

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2024

Abstract

Idiopathic pulmonary fibrosis (IPF) is a globally prevalent, progressive disease with limited treatment options and poor prognosis. Because of its irreversible disease progression, IPF affects the quality and length of life of patients and imposes a significant burden on their families and social healthcare services. The use of the antifibrotic drugs pirfenidone and nintedanib can slow the progression of the disease to some extent, but it does not have a reverse effect on the prognosis. The option of lung transplantion is also limited owing to contraindications to transplantation, possible complications after transplantation, and the risk of death. Therefore, the discovery of new, effective treatment methods is an urgent need. Over recent years, various studies have been undertaken to investigate the relationship between interstitial pneumonia and lung cancer, suggesting that some immune checkpoints in IPF are similar to those in tumors. Immune checkpoints are a class of immunosuppressive molecules that are essential for maintaining autoimmune tolerance and regulating the duration and magnitude of immune responses in peripheral tissues. They can prevent normal tissues from being damaged and destroyed by the immune response. While current studies have focused on PD-1/PD-L1 and CTLA-4, PD-1/PD-L1 may be the only effective immune checkpoint IPF treatment. This review discusses the application of PD-1/PD-L1 checkpoint in IPF, with the aim of finding a new direction for IPF treatment.
Hinweise

Publisher's Note

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

Introduction

Idiopathic pulmonary fibrosis (IPF) is a specific manifestation of progressive chronic pulmonary interstitial pneumonia of unknown origin and mainly occurs in the elderly [1]. Studies have shown that the incidence of IPF is increasing globally [2], and increases with age [3]. The primary clinical manifestations involve progressive and deteriorating dyspnea [4], and the diagnosis of IPF typically relies on histology and high-resolution computed tomography of the chest [5]. The characteristic pathological features of IPF include characteristic fibroblast foci, marked fibrosis, or honeycomb-shaped lungs [6]. Previous research suggests that the pathogenesis of IPF is associated with abnormally activated alveolar epithelial cells (AECs), which produce a variety of cytokines and growth factors, stimulate epithelial cell transformation into mesenchymal cells, and induce the formation of fibroblasts and myofibroblast foci. These fibroblasts and myofibroblast foci secrete excessive extracellular matrix (ECM), leading to scarring and lung remodeling [7]. The molecular mechanism of IPF is still poorly understood. However, studies have shown that transforming growth factor (TGF)-β signaling plays a central role in fibrosis progression [8]. However, since IPF has no specific clinical manifestations and most patients do not undergo relevant examinations in the early stage of the disease, the diagnosis and treatment of IPF pose a great challenge. Further, the course of the disease is irreversible, resulting in severe physical, psychological, and socio-economic burdens [9].
The current first-line protocol for diagnosing progressive IPF is the use of the antifibrotic drugs pirfenidone and nintedanib [10, 11]. Although the use of antifibrotic drugs can slow the deterioration of forced vital capacity in some patients, it does not extend survival time after diagnosis [12] and has a relatively limited effect on improving lung function and quality of life in patients with end-stage IPF [10, 11]. Lung transplantation, currently the only treatment for IPF that can improve symptoms and prolong survival, may only be used in a small number of patients with IPF [13]. Whether patients with IPF can undergo lung transplantation must be evaluated considering age, illness, comorbidities, contraindications, risk of death, and complications after transplantation [14]. Therefore, the need is urgent for treatments that can extend the lives of patients with IPF and improve their quality of life. Unfortunately, current treatments do not meet these requirements.
IPF has also been reported to share common risk factors with cancer [15]. Some articles have summarized the mechanisms underlying the interaction between IPF and lung cancer (LC), indicating that there are indeed some epidemiological, mechanistic, and genetic associations [16, 17]. One study summarized the reported incidence of LC in patients with IPF in recent years, which ranged from 2.7 to 48% [17], much higher than the incidence of LC in the general population [18]. Considering the association between these two diseases, studies have begun to test known anticancer drugs against IPF [19], with a focus on immune checkpoints.
PD-1/PD-L1 is involved in immune regulation and in maintaining the immune tolerance to autoantigens via a mechanism that is likely to be associated with the inhibition of T-cell hyperactivation and cytokine secretion [20]. Inhibiting the PD-1/PD-L1 axis can reverse the immunosuppressive targeting of the tumor microenvironment and restore the antitumor effects of T cells. Previous studies have found that PD-L1 is highly expressed in invasive lung fibroblasts and drives the progression of lung fibrosis in humanized mice [21]. Knocking down PD-L1 expression can also reduce TGF-β-induced extracellular matrix production in human and mouse lung fibroblast cell lines [22]. Other studies have suggested that PD-L1 can promote pulmonary fibrosis through the Smad3/β‑catenin pathway [23]. The role of the PD-1/PD-L1 immune checkpoint in IPF and the possible therapeutic effect of inhibiting this immune checkpoint on IPF still need to be further explored. In this review, we explored the potential of PD-1/PD-L1 inhibitors in the treatment of IPF based on current studies on PD-1/PD-L1 immune checkpoint.

Co-pathogenic mechanism and signaling pathways of IPF and LC

Potential risk factors for IPF include genetic mutations, viral infections, lifestyle choices, environmental exposures, and occupational hazards [24]. Smoking, air pollution, and occupational exposure are common risk factors for IPF and LC [25, 26]. The shared pathogenesis of IPF and LC [17, 27] includes uncontrolled cell proliferation [28] and interference with intercellular transmission [29, 30]. Studies have shown that certain cells (e.g. myofibroblasts, cancer-associated fibroblasts [CAFs]) and associated growth factors are equally involved in the disease development of IPF and LC [31]. Of these, myofibroblasts are particularly important in IPF because of their proliferative capacity and their ability to be activated under high fibrotic conditions. These abilities enable myofibroblasts to secrete and deposit ECM proteins, leading to lung fibrosis [32]. TGF-β1 induces the conversion of epithelial cells from epithelial-to-mesenchymal transition (EMT) to fibroblasts/myofibroblasts, thereby promoting fibrous proliferation. Furthermore, alveolar type II epithelial cells have been observed to differentiate into fibroblasts/myofibroblasts and produce the ECM under TGF-β1 stimulation [33].
CAFs are key cellular actors in LC, especially in non-small cell LC (NSCLS) [34]. Several hypotheses have been proposed regarding the cellular origin of CAF in tumors [16], as follows: resident fibroblasts differentiate into CAFs during tumor development under the coordination by certain signaling pathways and cancer-associated adipocytes, which exist in tumor stroma, possibly originating from circulating progenitor cells [35]; bone marrow MSCs and hematopoietic stem cells [36]; epithelial cells via EMT [37]; and vascular endothelial cells are critical to tumor angiogenesis via endothelial-to-mesenchymal transformation [38]. Therefore, TGF-β not only regulates the major molecules that promote fibrosis signaling but also promotes LC progression and cancer cell mitosis. In addition to the mesenchymal phenotype, cell transformation [27, 39] represents a common association between pulmonary fibrosis and carcinogenesis.
Abnormal activation of the following common signaling pathways (Fig. 1) has been reported to occur in both pulmonary fibrosis and lung cancer:

TGF-β/Smad pathway

This key pathway for fibrogenesis involves TGF-β1 being responsible for the activation of fibroblasts into myofibroblasts [40] and being classically signaled by the SMAD protein, which is a key pathway for fibrogenesis [41, 42]. TGF-β promotes ECM deposition, or selectively inhibits collagenase synthesis, induces the production of collagenase inhibitors or inhibits the expression and activation of protease inhibitors, and ultimately reduces collagen degradation, thus effectively promoting the formation of ECM-mediated pulmonary fibrosis [43]. In early-phase LC, TGF-β induces the arrest and apoptosis of normal and malignant cancer cells, thereby inhibiting tumor growth. Conversely, TGF-β also stimulates tumor EMT to promote tumorigenesis when cancer cells lose their oncogenic function or undergo mutations [44].

Wnt/β-catenin pathway

This pathway induces EMT by regulating certain cytokines associated with tissue infiltration. In lung fibrosis, the classical pathway of tracheal injury-induced alveolar type II epithelial cells is the activation of this signaling pathway to promote the activation of mesenchymal fibroblasts; sustained stimulation eventually leads to pulmonary fibrosis. Activation of this pathway also promotes the conversion of perivascular fibroblasts into myofibroblasts, leading to ECM accumulation and pulmonary fibrosis development [45]. The expression of the pathway-related protein TCF/LEF considerably increases following the activation of the Wnt signaling pathway in LC, thereby activating the transcription of Sox-2, c-Myc, cyclin D1, and other downstream target genes [46].

PI3K/AKT/mTOR pathway

Dysregulation of this pathway is involved in cell proliferation and apoptosis. TGF-β can activate the PI3K/Akt pathway by activating extracellular signals in addition to the Smad pathway [47]. In pulmonary fibrosis, PI3K was observed to be associated with fibroblast proliferation and differentiation [48]. Similarly, dysregulation of this signaling pathway has been observed in NSCLC and is primarily associated with cancer metastasis and invasion [49].

Notch pathway

Reactivation of the Notch pathway is essential for EMT in fibroblasts and it also promotes α-smooth muscle actin expression in fibroblasts [50]. This pathway is also involved in the NSCLC process, and in addition to stimulating NSCLC cell propagation and inhibiting NSCLC cell apoptosis, members of the Notch signaling pathway may also be able to predict disease progression and prognosis in patients with NSCLC [51]. Furthermore, activation of the Notch pathway in subpopulations of tumor cells in small cell lung cancer (SCLC) contributes to the establishment of the tumor microenvironment [52].

Sonic hedgehog (Shh) pathway

Over-expression of the Shh pathway promotes apoptosis in epithelial cells while enhancing fibroblast resistance to apoptosis [53]. In the development of early lung cancer, tumor stem cells can activate the Shh pathway and affect other tumor cells through paracrine effects, leading to tumor proliferation, spread, and EMT [54].
Gene set enrichment analysis of the IPF and NSCLC datasets revealed a common pattern of gene dysregulation [55]. However, IPF and NSCLC differ considerably at the global transcriptome level, with only a limited number of key genes exhibiting similar regulation, so successful treatment options for patients with cancer may provide unexpected benefits for patients with IPF [56]. The emergence of immune checkpoint inhibitors (ICIs) has provided a reliable and bright prospect for the immunotherapy of cancer.

Expression of PD-1/PD-L1 in patients with IPF

PD-1 is an immunosuppressive molecule mainly expressed in activated T cells and can be enhanced through stimulation with tumor necrosis factor [57]. PD-L1 is the main ligand of PD-1 and is widely expressed in T cells, B cells, dendritic cells, macrophages, and other tissues [58]. Blocking negative regulatory signaling then reactivates T-cell activity and promotes tumor cell killing ability, which is mainly achieved by blocking PD-1 binding to PD-L1 [59]. ICIs primarily exert their antitumor effects by blocking tumor cell escape, helping in repositioning the immune cells and engaging in cytotoxic killing [60]. In particular, PD-1/PD-L1 inhibitors block PD-1 on T cells or PD-L1 on the surface of tumor cells, leading to antitumor effects [61]. At present, PD-1/PD-L1 inhibitors have been approved for the treatment of patients with advanced NSCLC, hepatocellular carcinoma, urothelial carcinoma, metastatic melanoma, and metastatic colorectal cancer [62]. The clinical benefit of PD-1/PD-L1 inhibitors is usually predicted by immunohistochemical determination of PD-L1 expression levels [63]; high expression (> 1 or > 50% in NSCLC) is associated with better response rates [64, 65]. Given the similar pathogeneses and pathway mechanisms of IPF and cancer, we wondered whether ICIs could benefit patients with IPF.
The methods frequently employed to detect PD-1/PD-L1 expression in human specimens include enzyme-linked immunosorbent assay (ELISA) [66], flow cytometry [67], and immunofluorescence [23]. The results of these studies are summarised in Table 1.
Table 1
Summary of clinical studies of PD-1/PD-L1 in human specimens
Reference setting
PD-1/PD-L1
IPF patients/normal or other disease numbers
Sample
Method
Expression change
Jovanovic [66]
mPD-L1
sPD-L1
12/–
23/–
Lung
Plasma
IHC
LISA
Upregulation
Elevation
Celada [67]
PD-1
PD-L1
10/5
25/24
10/5
Lung
Peripheral blood
IHC
FCM
IHC
Upregulation
Upregulation
Upregulation
Guo [23]
PD-L1
3/3
Lung
IF
Upregulation
Cui [76]
PD-L1
11/3
Lung
Mass cytometry
Upregulation
Bonham [68]
PD-1
41/13
Peripheral blood
FCM
No change
Wang [69]
PD-1
PD-L1
23//25
23//25
Peripheral blood
Peripheral blood
FCM
Upregulation
Upregulation
Milenkovic [70]
sPD-L1
30/30
Plasma
ELISA
Elevation
Guyard [71]
PD-L1
18/13
(all complicated lung cancer)
Lung
IHC
Upregulation
Kronborg-White [72]
mPD-L1
43/55
Lung
IHC
Upregulation
Asai [73]
PD-1
18/21
Peripheral blood
FCM
Upregulation
Li [74]
PD-L1
16/8
8/10
12/12
Lung
BALF
Serum
IHC
ELISA
ELISA
Upregulation
Elevation
Elevation
Ahmadvand [75]
CD274
PD-1
9/12
Lung
FCM/RT-PCR
Upregulation
Upregulation
IPF idiopathic pulmonary fibrosis, PD-1 programmed cell death 1, PD-L1 programmed death-ligand 1, IF immunofluorescence, FCM flow cytometry, mPD-L1 membrane programmed death-ligand 1, sPD-L1 soluble programmed death-ligand 1, IHC immunohistochemistry, WB Western blot, ELISA enzyme-linked immunosorbent assay, Th17 T helper 17, Tfh cells T follicular helper cells, AT2 Alveolar epithelial type 2 cells
One study reported significantly elevated levels of PD-L1 expression in the serum of IPF patients [66], while another study showed the opposite, with no change in PD-L1 expression in the peripheral blood of IPF patients [68]. However, although no alterations were observed in PD-L1 expression in peripheral blood, PD-1 levels in circulating CD4 + T cells and lung tissue in IPF patients were observed to be higher than in age-matched healthy controls in another previous study [67]. Another study observed increased expression of PD-1 and PD-L1 in CD4 + T cells in the peripheral blood of patients [69]. In addition to the determination of PD-L1 expression, studies have also observed changes in the expression of soluble PD-L1 (sPD-L1), with one study showing significantly higher sPD-L1 levels in the serum of IPF patients without surgical biopsy than in healthy controls [66], and another study observing a similar presentation [70]. Given the similarity between IPF and NSCLC, a previous study used lung tissue from patients with lung fibrosis-related cancers as a sample to explore changes in PD-L1 expression [71]. Another previous study showed that PD-L1 was highly expressed on the cell membranes of alveolar and bronchial epithelial cells in lung tissue samples from patients with IPF and pulmonary fibrosis [72]. Alternatively, other studies reported that the PD-L1 expression was not significantly altered in fibroblasts and myofibroblasts in the lung tissues of patients but was overexpressed in alveolar macrophages [66]. Asai et al. reported a significantly higher proportion of PD-1+ICOS+Tfh cells in patients with IPF [73]. Li et al. observed elevated levels of PD-L1 in lung tissue, alveolar lavage fluid, and serum from patients [74]. Ahmadvand et al. came to the same conclusion for lung tissue and reported that PD-1 was highly expressed in T cells [75].
We noticed that in this table, Bonham et al. [68]found that the expression level of PD-1 did not increase as it did in other studies, which may be related to the fact that the samples used were fresh peripheral blood PBMC, the control group was age-sex matched, and the average fluorescence intensity of PD-1 expression was used in data analysis. Celada et al. [67] and Wang et al. [69] reported the percentage of PD-1+CD4+ T cells, while Asai et al. [73] reported the proportion of PD-1+ICOS+Tfh cells in peripheral blood. These results suggest that PD-1/PD-L1 is involved in the process of pulmonary fibrosis. Nonetheless, all studies published to date have been retrospective in design and have small sample sizes; therefore, further research is required. Prospective cohort studies can be designed for further exploration, with as large a sample size as possible, and care should be taken to eliminate bias and confounding factors in the study. Prospective cohort studies can be designed to explore further, with as large a sample size as possible, in addition to being aware of bias and confounding factors in the studies.

Preclinical studies of PD-1/PD-L1 in pulmonary fibrosis

The potential role of the PD-1/PD-L1 immune checkpoint in IPF was observed, and a series of preclinical studies were initiated in the hope that this new anti-tumor drug could bring a promising future to the treatment of IPF. The role of PD-1/PD-L1 in the pathogenesis of IPF has been explored through in vivo and in vitro experiments. Table 2 summarizes the results of these studies in animal models of IPF.
Table 2
Summary of clinical studies of PD-1/PD-L1 in mice specimens
Reference setting
PD-1/PD-L1
Exposure
Sample
Cell type
Method
Expression change
Celada [67]
PD-1
Bleomycin
Lung
CD4+ T cells
FCM
Upregulation
Guo [23]
PD-L1
Bleomycin
Lung
Fibroblasts
IF
Upregulation
Cui [76]
PD-L1
Bleomycin
Lung
Fibroblasts
Mass cytometry
IF/FCM
Upregulation
 
Primary human fibrotic lung fibroblast engraftation
Lung
Fibroblasts
Upregulation
PD-1
Bleomycin
Lung
CD8+ T cells
Mass cytometry
Upregulation
Li [74]
PD-L1
Bleomycin
paraquat
Lung
Lung
 
IHC/WB
IHC/WB
Upregulation
Upregulation
Geng [21]
PD-L1
CD274high and CD274low IPF lung normal fibroblasts injection
Lung
Fibroblasts
PCR
RNA-seq
Upregulation
Lu [77]
PD-L1
Bleomycin
Lung
Fibroblasts
WB/IHC
Upregulation
Wang [78]
PD-1
Bleomycin
Lung
/
IHC
Upregulation
Peripheral blood
CD4+ T cells
FCM
Upregulation
Zhao [83]
PD-1
Silica
Lung
/
WB/IHC FCM/PCR
Upregulation
PD-L1
Lung
/
Upregulation
PD-1 programmed cell death 1, PD-L1 programmed death-ligand 1, IF immunofluorescence, FCM flow cytometry, IHC immunohistochemistry, WB Western blot, PCR polymerase chain reaction, RNA-seq RNA sequencing
In three studies on mouse models of lung fibrosis, elevated levels of PD-L1 expression in lung tissue lesions were observed [23, 76, 77]. Furthermore, aberrant PD-1 expression was observed in a mouse model of pulmonary fibrosis [67, 76, 78]. Celada et al. observed elevated levels of PD-1 in CD4+ T cells from the lung tissue of fibrotic mice [67], whereas Cui et al. observed this phenomenon in CD8+ T cells [76]. In addition to the elevated expression of PD-1 observed in the lung tissue of fibrotic mice, Wang et al. found elevated expression of this indicator in CD4 + T cells of the peripheral blood of mice [78]. However, none of the above studies measured the specific expression levels of PD-1.
Based on research on human tissue transplantation, increasingly more immunodeficient mice have been used as preclinical animal models over recent years [66, 7981]. Some studies have also used humanized mouse models for PD-1/PD-L1 immune checkpoint related studies [21, 76, 82]. Geng et al. used NODSCIDIL2Rgc−/− (NSG) mice to establish pulmonary fibrosis models and found a significant regulatory effect of PD-L1 on invasive fibroblasts [21]. Additionally, an elevation in PD-L1 levels was observed in a humanized mouse model [76].
In addition to the abovementioned mouse models of bleomycin-induced lung fibrosis, transgenic techniques, and silica can also be utilized to establish mouse models of pulmonary fibrosis for the evaluation of PD-1/PD-L1 expression. Cui et al. detected elevated levels of PD-L1 expression in the lung tissue of IL-6 knockout mice, which is consistent with the results observed for PD-L1 in bleomycin-modelled lung fibrosis mice [76]. Zhao et al. found abnormal levels of expression of immune checkpoint molecules on these cells in a mouse model of silica-induced lung fibrosis, ultimately leading to systemic immune dysregulation [83]. Li et al. observed the same phenomenon as in BLM-induced pulmonary fibrosis in a mouse model of paraquat-induced pulmonary fibrosis, with upregulation of PD-L1 expression in lung tissue [74]. In summary, the aberrant expression levels of PD-1 and PD-L1 in certain lung cell types in a mouse model of pulmonary fibrosis suggest that PD-1/PD-L1 may play a role in the process of pulmonary fibrosis.
We observed differences in the animal models used in these studies. BLM-induced mouse pulmonary fibrosis model is the most commonly used animal model for the study of IPF, which can demonstrate the pathogenesis of human IPF [84], but it has certain limitations because its fibrosis resolves spontaneously after 28 days [85] The use of Silica in the lungs also causes persistent fibrosis, but is more similar to silicosis in humans than pulmonary fibrosis [85]. Systemic administration of paraquat causes fibrosis-like changes in the lungs of animals, but other organs (e.g., liver, kidney, etc.) also fail, increasing mortality rates [86]. Humanized mouse models can better elucidate the mechanism of human fibroblasts inducing pulmonary fibrosis in vivo, but the main issue is the availability of cells from human patients with IPF [87]. Although different modeling methods were used, the results showed increased expression levels of PD-1/PD-L1, suggesting that PD-1/PD-L1 was involved in the process of pulmonary fibrosis. Therefore, the researchers conducted further studies.
Current studies have shown that the fibrotic process is a combination of multiple cells, growth factors, and fibrosis-associated proteins, and their interactions determine the outcome of fibrosis [88]. The role of PD-1/PD-L1 in IPF can be explored from two perspectives.
Mechanistic experiments first recognized that PD-1+CD4+ T cells can promote pulmonary fibrosis and TGF-β production mainly through IL-17A, where T helper 17 (Th17) cells are the main CD4+ T cell subset expressing TGF-β. In an in vivo animal experiment, blocking PD-1 expression significantly reduced the expression of STAT3, IL-17A, and TGF-β on Th17 cells, thereby reducing the production of type I collagen by fibroblasts and attenuating lung fibrosis [67]. Similar results were observed in another study [78]. In addition, a study confirmed that maintaining the functional homeostatic balance between Th17/Treg cells is an important part of regulating autoimmunity and treating cancer [89]. Tregs can up-regulate the expression of TGF-β [90], and IL-6 allows Treg to regain the characteristics of Th17 cells [91]. Elevated IL-6 levels were also observed in patients with IPF. Therefore, we hypothesized that elevated IL-6 leads to the subpopulation conversion of Treg to Th17 cells, which increases the production of IL-17 and TGF-β and promotes lung fibrosis (Fig. 2A). Another study indicated that an increase in PD-1/PD-L1 inhibited CD4+ T cell differentiation to Treg cells, thereby reversing the effect of TGF-β on Treg cell differentiation. PD-1 promoted type I collagen production in myofibroblasts while decreasing the proliferation of myofibroblasts co-cultured with CD4+ T cells [69] (Fig. 2A). Upregulation of PD-L1 expression on fibroblasts has also been observed in studies on lung fibrosis, especially in invasive fibroblasts, which can promote lung fibrosis [21, 23, 76, 77]. We next explored the mechanism of the involvement of PD-L1 in IPF in fibroblasts.
Research suggests that high expression of PD-L1 on lung fibroblasts can be regulated through the p53 and FAK pathways. Knockdown of PD-L1 in invasive fibroblasts attenuates the severity of lung fibrosis in lung fibroblasts and mice [21]. Evidence also supports that p53 expression levels in myofibroblasts are significantly lower than those in normal lung fibroblasts [92, 93]. PD-L1 knockdown in IPF fibroblasts and targeting of PD-L1 by FAK inhibition or PD-L1-neutralizing antibodies blunts invasion and attenuates fibrosis [21]. In addition, the acquisition of p53 function could sensitize lung fibroblasts to apoptosis and prompt macrophages to clear apoptotic myofibroblasts, thereby attenuating pulmonary fibrosis symptoms in a mouse model of pulmonary fibrosis [93]. Therefore, PD-L1 on lung myofibroblasts may lead to IPF by inhibiting the p53 pathway, leading to myofibroblast resistance to apoptosis, and inhibiting phagocytosis by macrophages, leading to overproliferation of myofibroblasts (Fig. 2B). Recently research suggests that knockdown of PD-L1 in primary human lung fibroblasts significantly attenuated the expression of myofibroblast markers induced by TGF-β. TGF-β could induce PD-L1 to interact with Smad3, suggesting that PD-L1 may act as a cofactor for Smad3 to promote the process of lung fibrosis. PD-L1 knockdown attenuated TGF-β-induced GSK3β phosphorylation/inhibition and β-catenin upregulation, suggesting PD-L1-mediated GSK3β/β-catenin signaling in FMT [23]. Another study also showed a relation between the GSK3β/β-catenin signaling pathway and the progression of IPF [90]. Thereby, PD-L1 in lung fibroblasts was upregulated to promote the development of IPF through the Smad3 pathway and the β-catenin pathway (Fig. 2B).
Autophagy is also involved in the development of IPF. Inhibition of autophagy can induce myofibroblast proliferation and ECM deposition, leading to pulmonary fibrosis [94, 95]. The application of autophagy activators can attenuate the extent of pulmonary fibrosis [96]. It was found that anti-PD-L1 monoclonal antibody down-regulated the PI3K/AKT/mTOR pathway to induce autophagy, thereby inhibiting TGF-β1-induced lung fibroblast invasiveness and ECM deposition [77]. Inhibition of PI3K inhibits AKT phosphorylation and TGF-β-induced proliferation of fibroblasts as well as expression of markers in myofibroblasts [97]. Further, mTOR acts as a downstream molecule of the PI3K/AKT pathway, and inhibition of the PI3K/AKT pathway inhibits this target, upregulates myofibroblast autophagy and attenuates lung fibrosis [98] (Fig. 2C).
The mechanism of action of the PD-1/PD-L1 axis in IPF is not yet fully elucidated and further exploration is needed. We summarize the results of current PD-1/PD-L1 inhibitors in IPF studies below to explore the feasibility of targeting PD-1/PD-L1 for the treatment of IPF.
The majority of studies revealed a reduction in the extent of lung tissue lesions with anti-PD-L1 mAb in mouse models of pulmonary fibrosis [21, 67, 76, 77, 83]. Cui et al. observed that pulmonary fibrosis was reduced in a mouse fibrosis model after blocking PD-L1, thus providing a potential treatment strategy for patients with pulmonary fibrosis [76]. By establishing a humanized severe combined immunodeficiency IPF model, Geng et al. suggested that targeting PD-L1 during the early and late stages might significantly reduce the invasion of IPF fibroblasts and could not only eliminate the progression of pulmonary fibrosis but also reverse it [21]. Lu et al. found that anti-Pd-L1mAb ameliorated lung tissue disorganization and collagen deposition in mice with pulmonary fibrosis while demonstrating that it possesses the ability to activate autophagy in pulmonary fibrosis [77]. Zhao et al. conducted a similar study on silica-induced pulmonary fibrosis in mice and discovered that PD-1/PD-L1 signal inhibition significantly improved silicosis [83]. Current research is not limited to single-drug studies of PD-1/PD-L1 pathway inhibitors, and some researchers are interested in combination therapy. A study combined pirfenidone with a PD-L1 blocker to explore its treatment effects in mouse models of pulmonary fibrosis and LC [99]. Combined therapy with pirfenidone and PD-L1 blockers promoted immune cell invasion and inhibited tumor outgrowth while improving the prognosis of mice. This combination treatment regimen reduced lung fibrosis in mice. Some studies have also pointed out that human MSCs contribute to immune regulation, which has been used in several preclinical studies on IPF treatment [100]. The efficacy of ICIs in the aforementioned mouse models of pulmonary fibrosis demonstrates that PD-L1 inhibitors have a promising effect on alleviating pulmonary fibrosis.
The researchers also designed in vitro experiments to determine whether inhibition of PD-L1 reduces lung fibrosis. The in vitro experiments demonstrated that blocking the PD-1/PD-L1 pathway can inhibit STAT3 expression in CD4 + T cells, leading to low expression of IL-17A and TGF-β [67]. A previous report showed that CD274 knockdown in IPF fibroblasts by FAK inhibition or CD274-neutralizing antibodies, reduced invasion and alleviated fibrosis, whereas PD-L1 expression on invasive fibroblasts promoted lung fibrosis [21]. Inhibition of PD-1 appears to be less effective than inhibition of PD-L1 in preclinical IPF models, which may be related to the genetic ablation of PD-1 [67]. Given the elevated levels of immune checkpoint expression in patients with IPF, novel ICIs may provide new immunotherapeutic options.
The above preclinical studies have shown that inhibition of immune checkpoint pairs can attenuate the progression of pulmonary fibrosis, and that inhibition of PD-L1, in particular, can produce an antifibrotic effect. However, PD-1/PD-L1 inhibitors cannot be assumed to directly lead to results for the clinical treatment of IPF. While the fibrotic process is a complex process in which antifibrotic mediators interact with pro-fibrotic mediators in a microenvironment composed of different cell types [76], immune tolerance imbalances and the occurrence of immune-related adverse events (irAEs) require attention. Therefore, although preclinical studies have shown that targeting PD-1/PD-L1 immune checkpoints attenuates pulmonary fibrosis, more evidence is needed to support how these preclinical studies can be transferred to practical clinical applications.
ICIs, while enhancing the normal immune response, may enhance the anti-tumor effects of cellular immunity, leading to an immune tolerance imbalance and immune-related adverse events (irAEs). IrAEs can involve all organs throughout the body and are common, with data from one study showing that over 80% of patients using ICIs developed irAEs in some systems [101, 102]. In the targeted treatment of ICIs for LC, immunosuppressant-related pneumonia requires consideration. Studies have found that the use of ICIs in patients with LC with ILD is associated with a higher risk of developing immune checkpoint inhibitors related pneumonitis (CIP) than that in patients without ILD, but these studies suggest that CIP is not associated with increased mortality [103105]. The response rate, progression-free survival, and overall survival of NSCLC patients with concomitant ILD treated with ICIs were similar to those of patients without ILD, suggesting that patients with NSCLS treated with ILD could also benefit from ICI treatment [103]. However, since the presence of interstitial pneumonia is currently regarded as the exclusion criterion for ICI clinical trials [106, 107], direct evidence is lacking to prove the effect of ICIs on pulmonary fibrosis symptoms. Currently, no studies have directly validated the utility of PD-1/PD-L1 inhibitors on pulmonary fibrosis, and conclusive experimental evidence to support their therapeutic value is scarce. Studies have shown that ICIs in patients with IPF combined with squamous cell carcinoma, the addition of nidanib may prevent drug-induced pneumonia or acute exacerbation of IPF [108]. Another study showed that pirfenidone combined with PD-L1 inhibitors in the treatment of tumor pulmonary fibrosis in mice significantly delayed tumor growth, reduced pulmonary fibrosis, and improved mouse survival. This suggests that pirfenidone could be used as an adjunct to immunotherapy in cancer treatment [99]. Therefore, the study of the potential mechanism of irAEs not only contributes to the immunotherapy of tumors but also plays an important role in the treatment of IPF. Whether the combination of ICIs and antifibrotic drugs (such as nidanib and pirfenidone) can delay the pathogenesis of IPF can be considered as a research direction. In addition to investigating whether ICIs can be used to improve disease progression in patients with IPF, future studies should also clarify its role in prognosis.

Conclusions and outlook

With the gradual exploration of the pathogenesis of IPF, new progress has been made in the treatment of IPF. Nonetheless, room for progress before satisfactory efficacy can be achieved. Current experiments have confirmed that the PD-1/PD-L1 pathway can interact with various cell types and pathways and is involved in promoting fibrosis and immune regulation in IPF. Simultaneously, animal experiments have revealed that the application of PD-1/PD-L1 inhibitors reduces the symptoms of pulmonary fibrosis. In this summary review, we present the effects of PD-1/PD-L1 in IPF; ongoing research suggests that it may offer a novel direction for future IPF therapy. Nevertheless, the following problems remain and, hence, require further investigations:
First, most studies published to date have reported PD-1/PD-L1 expression levels in IPF and confirmed its effect on pulmonary fibrosis; furthermore, the application of PD-1/PD-L1 ICIs has been proven to reduce lung fibrosis. However, the results are concentrated on animals and preclinical studies are poorly supported by complete clinical evidence. We have summarized in this review the role of PD-1/PD-L1 in the pathogenesis of IPF, and we still have a long way to go to translate these mechanisms into effective anti-pulmonary fibrosis therapies. The development of prognostic animal models and ex vivo primary human lung tissue models can help us achieve the transition from experiments to clinical applications [109]. Because the presence of pulmonary fibrosis is currently an exclusion criterion for receiving immune checkpoint inhibitors for lung cancer in clinical trials, the selection of patients for clinical trials with ICIs is difficult. Therefore, a more in-depth analysis of the mechanism of immune checkpoint involvement in pulmonary fibrosis, an accurate search for key therapeutic targets, and accurate and verified predictors of IPF progression, to identify target patients for inclusion in the experiment, is a reliable choice. A full understanding of the molecular mechanism of pulmonary fibrosis and stratified evaluation of IPF patients are helpful to tailor treatment and evaluate prognosis, thus improving clinical efficacy. Referring to the research methods in chronic obstructive pulmonary disease, the reasonable stratification of IPF patients to explore the relationship between different factors and disease progression and prognosis is also conducive to the treatment of IPF [110]. In addition, the development of single-cell genomics methodologies [111], molecular imaging of fibrosis [112], and other technologies can help us better conduct precision medicine research and provide help for the future development of IPF immunotherapy.
An important contribution of certain cytokines [113], such as IL-17A [67], IL-6 [114], and IL-8 [115] in pulmonary fibrosis has been observed. Studies have shown that simultaneous blockade of IL-6, CD47, and PD-L1 attenuates pulmonary fibrosis and may be associated with enhanced phagocytosis of fibroblasts and elimination of inhibition of adaptive immunity [76].In patients with COVID-19 combined with pulmonary fibrosis, and IL-6 antibodies showed better efficacy against COVID-19-induced cytokine storm. Therefore, IL-6 antibodies may be of benefit to patients with IPF [116]. IL-8 promotes senescence of IPF fibrous mesenchymal progenitor cells (MPCs) and up-regulation of PD-L1, allowing IPF MPCs to evade clearing by immune cells and thus exerting a pro-fibrotic effect [115]. In addition to this, there have been some studies exploring the effect of the combination of CD47 and PD-L1 antibodies in IPF [117, 118]. Studying the interrelationship between cytokines and PD-1/PD-L1 in the pathogenesis of IPF, further elucidating the role of PD-1/PD-L1 in the pathogenesis of IPF, and then exploring how to apply PD-1/PD-L1 inhibitors in the clinical treatment of IPF are the directions we can proceed in the future.
Second, EMT is still regarded as an important link in the development of pulmonary fibrosis in IPF. BLM-induced upregulation of PD-L1 was found to lead to EMT progression and fibroblast-like morphological changes in AEC. Alveolar epithelial cells are the progenitor cells of fibroblasts in vivo [119], and inhibition of EMT in AEC suppresses pulmonary fibrosis [120]. PD-L1 can induce EMT in AECs to promote lung fibrosis by directly binding to vimentin and inhibiting vimentin ubiquitination. Vimentin promotes BLM- and PD-L1-induced fibrosis in Human Pulmonary Alveolar Epithelial Cells, thereby promoting lung fibrosis [74]. Vimentin, as an important stimulator of EMT, is also a key factor in lung fibrosis. Therefore, exploring the interrelationship between EMT-related proteins and PD-1/PD-L1 thereby further investigating the specific pathogenesis of IPF could also bring new insights for the treatment of IPF.
Third, as the role of macrophages in the process of lung fibrosis is revealed, the understanding of lung macrophages is becoming clearer [121, 122]. For instance, one study showed that PD-L1 produced an immunosuppressive cell phenotype by delivering negative signals to macrophages and that PD-L1 antibody application reversed the phenotype [123]. In recent years, research on PD-1/PD-L1 immune checkpoints and tumor-associated macrophages has opened up new perspectives for the treatment of tumors [121, 124128]. Activation of DDR1 in macrophages was found to play a role in IPF through inflammatory vesicle activation and macrophage responses [129]. Jovanovic et al. found PD-L1 expression on alveolar macrophages in IPF and suggested that PD-L1-positive alveolar macrophages have a regulatory/inhibitory phenotype with reduced phagocytosis, thus promoting the development of pulmonary fibrosis [66]. Therefore, we can focus more on how PD-L1 regulates the role of macrophages, which in turn are involved in lung fibrosis, to obtain useful insights.
With the development of nanomedicine, researchers have begun to explore whether nanomedicine can successfully solve real clinical problems. With this in mind, researchers have devised various approaches that have looked to enhance the effectiveness of immunotherapy, such as the application of PD-1/PD-L1 drugs in tumors [130]. The current study found that preparing iron death inhibitors as inhalable nanomedicines for the treatment of IPF may result in better efficacy [131]. Nanodesigned carbon monoxide donors may also improve BLM-induced pulmonary fibrosis [132]. The feasibility of preparing PD-1/PD-L1 inhibitors as nanomedicines or adopting a nanomedicine approach to IPF also needs to be further explored. In addition, green nanomaterials deserve our attention because of their low cost, low pollution, and safety to the environment and human health [133, 134].
Additionally, an increasing number of studies have investigated the mechanisms behind the role of traditional Chinese medicine (TCM) in pulmonary fibrosis, and the material basis and therapeutic mechanism of TCM treatment for IPF have been preliminarily explored and revealed [135, 136]. Researches are beginning to focus on the relationship between TCM and PD-1/PD-L1 immune checkpoints [137, 138]. For instance, berberine is a Chinese medicinal ingredient that blocks PD-L1 expression in tumor cells and enhances the sensitivity of tumor cells to T cells. Berberine exerts its antitumor effects by degrading PD-L1 through a ubiquitin/proteasome-dependent pathway [137]. A previous study used oral Gancao Ganjiang decoction to treat bleomycin drip-induced pulmonary fibrosis in a mouse model and observed that this decoction reduced the PD-1 levels in lung tissues and the PD-1 expression in peripheral blood CD4+ T cells, thereby diminishing the inflammatory response and fibrosis in the lungs [78].
Most TCM components applied in IPF treatment have multiple targets, pathways, and levels of action. At the experimental stage, several active TCM ingredients have displayed tremendous value in IPF treatment, providing a reference for new treatment ideas for IPF. For example, Bufei huoxue capsule, as a TCM formulation, can alleviate BLM-induced pulmonary fibrosis in mice by inhibiting the TGF-β/Smad2/3 signaling pathway [139]. Considering the critical function of the PD-1/PD-L1 immune checkpoints in IPF, is it viable to treat IPF using the aforementioned nanomedicines and TCM? This question requires researchers to continue conducting further investigations. Using network pharmacology methods to design multi-target drug molecules for specific signal nodes, combining molecular docking technology with traditional experimental methods, can better help us understand the potential mechanism of IPF and the development of new drugs in the future.
In conclusion, ICIs, particularly PD-L1 inhibitors, may be effective as a treatment for IPF and warrant further exploration and development.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declared no potential competing interests with respect to the research, authorship, and/or publication of this article.
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.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Raghu G, Remy-Jardin M, Richeldi L, Thomson CC, Inoue Y, Johkoh T, et al. Idiopathic pulmonary fibrosis (an update) and progressive pulmonary fibrosis in adults: an official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med. 2022;205(9):e18–47.PubMedPubMedCentralCrossRef Raghu G, Remy-Jardin M, Richeldi L, Thomson CC, Inoue Y, Johkoh T, et al. Idiopathic pulmonary fibrosis (an update) and progressive pulmonary fibrosis in adults: an official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med. 2022;205(9):e18–47.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Hutchinson J, Fogarty A, Hubbard R, McKeever T. Global incidence and mortality of idiopathic pulmonary fibrosis: a systematic review. Eur Respir J. 2015;46(3):795–806.PubMedCrossRef Hutchinson J, Fogarty A, Hubbard R, McKeever T. Global incidence and mortality of idiopathic pulmonary fibrosis: a systematic review. Eur Respir J. 2015;46(3):795–806.PubMedCrossRef
3.
Zurück zum Zitat Jo HE, Randhawa S, Corte TJ, Moodley Y. Idiopathic pulmonary fibrosis and the elderly: diagnosis and management considerations. Drugs Aging. 2016;33(5):321–34.PubMedCrossRef Jo HE, Randhawa S, Corte TJ, Moodley Y. Idiopathic pulmonary fibrosis and the elderly: diagnosis and management considerations. Drugs Aging. 2016;33(5):321–34.PubMedCrossRef
4.
Zurück zum Zitat Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, et al. Diagnosis of idiopathic pulmonary fibrosis an official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med. 2018;198(5):e44-68.PubMedCrossRef Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, et al. Diagnosis of idiopathic pulmonary fibrosis an official ATS/ERS/JRS/ALAT clinical practice guideline. Am J Respir Crit Care Med. 2018;198(5):e44-68.PubMedCrossRef
5.
Zurück zum Zitat Martinez FJ, Collard HR, Pardo A, Raghu G, Richeldi L, Selman M, et al. Idiopathic pulmonary fibrosis. Nat Rev Dis Primers. 2017;3:17074.PubMedCrossRef Martinez FJ, Collard HR, Pardo A, Raghu G, Richeldi L, Selman M, et al. Idiopathic pulmonary fibrosis. Nat Rev Dis Primers. 2017;3:17074.PubMedCrossRef
6.
Zurück zum Zitat Wolters PJ, Collard HR, Jones KD. Pathogenesis of idiopathic pulmonary fibrosis. Annu Rev Pathol. 2014;9:157–79.PubMedCrossRef Wolters PJ, Collard HR, Jones KD. Pathogenesis of idiopathic pulmonary fibrosis. Annu Rev Pathol. 2014;9:157–79.PubMedCrossRef
7.
Zurück zum Zitat King TE Jr, Pardo A, Selman M. Idiopathic pulmonary fibrosis. Lancet. 2011;378(9807):1949–61.PubMedCrossRef King TE Jr, Pardo A, Selman M. Idiopathic pulmonary fibrosis. Lancet. 2011;378(9807):1949–61.PubMedCrossRef
9.
Zurück zum Zitat Tian Y, Li H, Qiu T, Dai J, Zhang Y, Chen J, et al. Loss of PTEN induces lung fibrosis via alveolar epithelial cell senescence depending on NF-κB activation. Aging Cell. 2019;18(1):e12858.PubMedCrossRef Tian Y, Li H, Qiu T, Dai J, Zhang Y, Chen J, et al. Loss of PTEN induces lung fibrosis via alveolar epithelial cell senescence depending on NF-κB activation. Aging Cell. 2019;18(1):e12858.PubMedCrossRef
11.
Zurück zum Zitat Raghu G, Selman M. Nintedanib and pirfenidone. New antifibrotic treatments indicated for idiopathic pulmonary fibrosis offer hopes and raises questions. Am J Respir Crit Care Med. 2015;191(3):252–4.PubMedCrossRef Raghu G, Selman M. Nintedanib and pirfenidone. New antifibrotic treatments indicated for idiopathic pulmonary fibrosis offer hopes and raises questions. Am J Respir Crit Care Med. 2015;191(3):252–4.PubMedCrossRef
12.
Zurück zum Zitat Vancheri C, Kreuter M, Richeldi L, Ryerson CJ, Valeyre D, Grutters JC, et al. Nintedanib with add-on pirfenidone in idiopathic pulmonary fibrosis. Results of the INJOURNEY trial. Am J Respir Crit Care Med. 2018;197(3):356–63.PubMedCrossRef Vancheri C, Kreuter M, Richeldi L, Ryerson CJ, Valeyre D, Grutters JC, et al. Nintedanib with add-on pirfenidone in idiopathic pulmonary fibrosis. Results of the INJOURNEY trial. Am J Respir Crit Care Med. 2018;197(3):356–63.PubMedCrossRef
15.
Zurück zum Zitat de Groot P, Munden RF. Lung cancer epidemiology, risk factors, and prevention. Radiol Clin North Am. 2012;50(5):863–76.PubMedCrossRef de Groot P, Munden RF. Lung cancer epidemiology, risk factors, and prevention. Radiol Clin North Am. 2012;50(5):863–76.PubMedCrossRef
18.
Zurück zum Zitat Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86.PubMedCrossRef Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86.PubMedCrossRef
23.
Zurück zum Zitat Guo X, Sunil C, Adeyanju O, Parker A, Huang S, Ikebe M, et al. PD-L1 mediates lung fibroblast to myofibroblast transition through Smad3 and β-catenin signaling pathways. Sci Rep. 2022;12(1):3053.PubMedPubMedCentralCrossRef Guo X, Sunil C, Adeyanju O, Parker A, Huang S, Ikebe M, et al. PD-L1 mediates lung fibroblast to myofibroblast transition through Smad3 and β-catenin signaling pathways. Sci Rep. 2022;12(1):3053.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Phan THG, Paliogiannis P, Nasrallah GK, Giordo R, Eid AH, Fois AG, et al. Emerging cellular and molecular determinants of idiopathic pulmonary fibrosis. Cell Mol Life Sci CMLS. 2021;78(5):2031–57.PubMedCrossRef Phan THG, Paliogiannis P, Nasrallah GK, Giordo R, Eid AH, Fois AG, et al. Emerging cellular and molecular determinants of idiopathic pulmonary fibrosis. Cell Mol Life Sci CMLS. 2021;78(5):2031–57.PubMedCrossRef
26.
Zurück zum Zitat Richeldi L, Collard HR, Jones MG. Idiopathic pulmonary fibrosis. Lancet. 2017;389(10082):1941–52.PubMedCrossRef Richeldi L, Collard HR, Jones MG. Idiopathic pulmonary fibrosis. Lancet. 2017;389(10082):1941–52.PubMedCrossRef
27.
Zurück zum Zitat Tzouvelekis A, Gomatou G, Bouros E, Trigidou R, Tzilas V, Bouros D. Common pathogenic mechanisms between idiopathic pulmonary fibrosis and lung cancer. Chest. 2019;156(2):383–91.PubMedCrossRef Tzouvelekis A, Gomatou G, Bouros E, Trigidou R, Tzilas V, Bouros D. Common pathogenic mechanisms between idiopathic pulmonary fibrosis and lung cancer. Chest. 2019;156(2):383–91.PubMedCrossRef
28.
Zurück zum Zitat Gerarduzzi C, Di Battista JA. Myofibroblast repair mechanisms post-inflammatory response: a fibrotic perspective. Inflamm Res. 2017;66(6):451–65.PubMedCrossRef Gerarduzzi C, Di Battista JA. Myofibroblast repair mechanisms post-inflammatory response: a fibrotic perspective. Inflamm Res. 2017;66(6):451–65.PubMedCrossRef
29.
Zurück zum Zitat Koval M, Billaud M, Straub AC, Johnstone SR, Zarbock A, Duling BR, et al. Spontaneous lung dysfunction and fibrosis in mice lacking connexin 40 and endothelial cell connexin 43. Am J Pathol. 2011;178(6):2536–46.PubMedPubMedCentralCrossRef Koval M, Billaud M, Straub AC, Johnstone SR, Zarbock A, Duling BR, et al. Spontaneous lung dysfunction and fibrosis in mice lacking connexin 40 and endothelial cell connexin 43. Am J Pathol. 2011;178(6):2536–46.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Chen R, Chen Y, Yuan Y, Zou X, Sun Q, Lin H, et al. Cx43 and AKAP95 regulate G1/S conversion by competitively binding to cyclin E1/E2 in lung cancer cells. Thoracic Cancer. 2020;11(6):1594–602.PubMedPubMedCentralCrossRef Chen R, Chen Y, Yuan Y, Zou X, Sun Q, Lin H, et al. Cx43 and AKAP95 regulate G1/S conversion by competitively binding to cyclin E1/E2 in lung cancer cells. Thoracic Cancer. 2020;11(6):1594–602.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Sivakumar P, Ammar R, Thompson JR, Luo Y, Streltsov D, Porteous M, et al. Integrated plasma proteomics and lung transcriptomics reveal novel biomarkers in idiopathic pulmonary fibrosis. Respir Res. 2021;22(1):273.PubMedPubMedCentralCrossRef Sivakumar P, Ammar R, Thompson JR, Luo Y, Streltsov D, Porteous M, et al. Integrated plasma proteomics and lung transcriptomics reveal novel biomarkers in idiopathic pulmonary fibrosis. Respir Res. 2021;22(1):273.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Wei P, Xie Y, Abel PW, Huang Y, Ma Q, Li L, et al. Transforming growth factor (TGF)-β1-induced miR-133a inhibits myofibroblast differentiation and pulmonary fibrosis. Cell Death Dis. 2019;10(9):670.PubMedPubMedCentralCrossRef Wei P, Xie Y, Abel PW, Huang Y, Ma Q, Li L, et al. Transforming growth factor (TGF)-β1-induced miR-133a inhibits myofibroblast differentiation and pulmonary fibrosis. Cell Death Dis. 2019;10(9):670.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Wong KY, Cheung AH, Chen B, Chan WN, Yu J, Lo KW, et al. Cancer-associated fibroblasts in nonsmall cell lung cancer: from molecular mechanisms to clinical implications. Int J Cancer. 2022;151(8):1195–215.PubMedPubMedCentralCrossRef Wong KY, Cheung AH, Chen B, Chan WN, Yu J, Lo KW, et al. Cancer-associated fibroblasts in nonsmall cell lung cancer: from molecular mechanisms to clinical implications. Int J Cancer. 2022;151(8):1195–215.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Bochet L, Lehuédé C, Dauvillier S, Wang YY, Dirat B, Laurent V, et al. Adipocyte-derived fibroblasts promote tumor progression and contribute to the desmoplastic reaction in breast cancer. Can Res. 2013;73(18):5657–68.CrossRef Bochet L, Lehuédé C, Dauvillier S, Wang YY, Dirat B, Laurent V, et al. Adipocyte-derived fibroblasts promote tumor progression and contribute to the desmoplastic reaction in breast cancer. Can Res. 2013;73(18):5657–68.CrossRef
36.
Zurück zum Zitat Raz Y, Cohen N, Shani O, Bell RE, Novitskiy SV, Abramovitz L, et al. Bone marrow-derived fibroblasts are a functionally distinct stromal cell population in breast cancer. J Exp Med. 2018;215(12):3075–93.PubMedPubMedCentralCrossRef Raz Y, Cohen N, Shani O, Bell RE, Novitskiy SV, Abramovitz L, et al. Bone marrow-derived fibroblasts are a functionally distinct stromal cell population in breast cancer. J Exp Med. 2018;215(12):3075–93.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Petersen OW, Lind Nielsen H, Gudjonsson T, Villadsen R, Rønnov-Jessen L, Bissell MJ. The plasticity of human breast carcinoma cells is more than epithelial to mesenchymal conversion. Breast Cancer Res. 2001;3(4):213–7.PubMedCrossRef Petersen OW, Lind Nielsen H, Gudjonsson T, Villadsen R, Rønnov-Jessen L, Bissell MJ. The plasticity of human breast carcinoma cells is more than epithelial to mesenchymal conversion. Breast Cancer Res. 2001;3(4):213–7.PubMedCrossRef
38.
Zurück zum Zitat Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Can Res. 2007;67(21):10123–8.CrossRef Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Can Res. 2007;67(21):10123–8.CrossRef
39.
Zurück zum Zitat Tang Z, Ding Y, Shen Q, Zhang C, Li J, Nazar M, et al. KIAA1199 promotes invasion and migration in non-small-cell lung cancer (NSCLC) via PI3K-Akt mediated EMT. J Mol Med. 2019;97(1):127–40.PubMedCrossRef Tang Z, Ding Y, Shen Q, Zhang C, Li J, Nazar M, et al. KIAA1199 promotes invasion and migration in non-small-cell lung cancer (NSCLC) via PI3K-Akt mediated EMT. J Mol Med. 2019;97(1):127–40.PubMedCrossRef
40.
Zurück zum Zitat Thannickal VJ, Lee DY, White ES, Cui Z, Larios JM, Chacon R, et al. Myofibroblast differentiation by transforming growth factor-beta1 is dependent on cell adhesion and integrin signaling via focal adhesion kinase. J Biol Chem. 2003;278(14):12384–9.PubMedCrossRef Thannickal VJ, Lee DY, White ES, Cui Z, Larios JM, Chacon R, et al. Myofibroblast differentiation by transforming growth factor-beta1 is dependent on cell adhesion and integrin signaling via focal adhesion kinase. J Biol Chem. 2003;278(14):12384–9.PubMedCrossRef
42.
Zurück zum Zitat Bonniaud P, Margetts PJ, Ask K, Flanders K, Gauldie J, Kolb M. TGF-beta and Smad3 signaling link inflammation to chronic fibrogenesis. J Immunol. 2005;175(8):5390–5.PubMedCrossRef Bonniaud P, Margetts PJ, Ask K, Flanders K, Gauldie J, Kolb M. TGF-beta and Smad3 signaling link inflammation to chronic fibrogenesis. J Immunol. 2005;175(8):5390–5.PubMedCrossRef
43.
Zurück zum Zitat Chanda D, Otoupalova E, Smith SR, Volckaert T, De Langhe SP, Thannickal VJ. Developmental pathways in the pathogenesis of lung fibrosis. Mol Aspects Med. 2019;65:56–69.PubMedCrossRef Chanda D, Otoupalova E, Smith SR, Volckaert T, De Langhe SP, Thannickal VJ. Developmental pathways in the pathogenesis of lung fibrosis. Mol Aspects Med. 2019;65:56–69.PubMedCrossRef
44.
Zurück zum Zitat Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol. 2019;20(2):69–84.PubMedCrossRef Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol. 2019;20(2):69–84.PubMedCrossRef
45.
Zurück zum Zitat Katoh M. Multi-layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β-catenin signaling activation (Review). Int J Mol Med. 2018;42(2):713–25.PubMedPubMedCentral Katoh M. Multi-layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β-catenin signaling activation (Review). Int J Mol Med. 2018;42(2):713–25.PubMedPubMedCentral
46.
Zurück zum Zitat Yang S, Liu Y, Li MY, Ng CSH, Yang SL, Wang S, et al. FOXP3 promotes tumor growth and metastasis by activating Wnt/β-catenin signaling pathway and EMT in non-small cell lung cancer. Mol Cancer. 2017;16(1):124.PubMedPubMedCentralCrossRef Yang S, Liu Y, Li MY, Ng CSH, Yang SL, Wang S, et al. FOXP3 promotes tumor growth and metastasis by activating Wnt/β-catenin signaling pathway and EMT in non-small cell lung cancer. Mol Cancer. 2017;16(1):124.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Hu X, Xu Q, Wan H, Hu Y, Xing S, Yang H, et al. PI3K-Akt-mTOR/PFKFB3 pathway mediated lung fibroblast aerobic glycolysis and collagen synthesis in lipopolysaccharide-induced pulmonary fibrosis. Lab Invest. 2020;100(6):801–11.PubMedCrossRef Hu X, Xu Q, Wan H, Hu Y, Xing S, Yang H, et al. PI3K-Akt-mTOR/PFKFB3 pathway mediated lung fibroblast aerobic glycolysis and collagen synthesis in lipopolysaccharide-induced pulmonary fibrosis. Lab Invest. 2020;100(6):801–11.PubMedCrossRef
48.
Zurück zum Zitat Wang J, Hu K, Cai X, Yang B, He Q, Wang J, et al. Targeting PI3K/AKT signaling for treatment of idiopathic pulmonary fibrosis. Acta Pharma Sinica B. 2022;12(1):18–32.CrossRef Wang J, Hu K, Cai X, Yang B, He Q, Wang J, et al. Targeting PI3K/AKT signaling for treatment of idiopathic pulmonary fibrosis. Acta Pharma Sinica B. 2022;12(1):18–32.CrossRef
50.
Zurück zum Zitat Hussain M, Xu C, Ahmad M, Yang Y, Lu M, Wu X, et al. Notch signaling: linking embryonic lung development and asthmatic airway remodeling. Mol Pharmacol. 2017;92(6):676–93.PubMedCrossRef Hussain M, Xu C, Ahmad M, Yang Y, Lu M, Wu X, et al. Notch signaling: linking embryonic lung development and asthmatic airway remodeling. Mol Pharmacol. 2017;92(6):676–93.PubMedCrossRef
52.
Zurück zum Zitat Lim JS, Ibaseta A, Fischer MM, Cancilla B, O’Young G, Cristea S, et al. Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature. 2017;545(7654):360–4.PubMedPubMedCentralCrossRef Lim JS, Ibaseta A, Fischer MM, Cancilla B, O’Young G, Cristea S, et al. Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature. 2017;545(7654):360–4.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Mailleux AA, Moshai EF, Crestani B. Sonic Hedgehog signaling in pulmonary fibrosis: a spiky issue? Am J Physiol Lung Cell Mol Physiol. 2013;304(6):L391–3.PubMedCrossRef Mailleux AA, Moshai EF, Crestani B. Sonic Hedgehog signaling in pulmonary fibrosis: a spiky issue? Am J Physiol Lung Cell Mol Physiol. 2013;304(6):L391–3.PubMedCrossRef
55.
Zurück zum Zitat Ulke HM, Mutze K, Lehmann M, Wagner DE, Heinzelmann K, Günther A, et al. The oncogene ECT2 contributes to a hyperplastic, proliferative lung epithelial cell phenotype in idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol. 2019;61(6):713–26.PubMedCrossRef Ulke HM, Mutze K, Lehmann M, Wagner DE, Heinzelmann K, Günther A, et al. The oncogene ECT2 contributes to a hyperplastic, proliferative lung epithelial cell phenotype in idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol. 2019;61(6):713–26.PubMedCrossRef
57.
Zurück zum Zitat Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8(3):239–45.PubMedCrossRef Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol. 2007;8(3):239–45.PubMedCrossRef
59.
Zurück zum Zitat Efremova M, Rieder D, Klepsch V, Charoentong P, Finotello F, Hackl H, et al. Targeting immune checkpoints potentiates immunoediting and changes the dynamics of tumor evolution. Nat Commun. 2018;9(1):32.PubMedPubMedCentralCrossRef Efremova M, Rieder D, Klepsch V, Charoentong P, Finotello F, Hackl H, et al. Targeting immune checkpoints potentiates immunoediting and changes the dynamics of tumor evolution. Nat Commun. 2018;9(1):32.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8(9):1069–86.PubMedCrossRef Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8(9):1069–86.PubMedCrossRef
62.
Zurück zum Zitat Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16:223–49.PubMedCrossRef Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16:223–49.PubMedCrossRef
63.
Zurück zum Zitat Shi H, Zhang W, Zhang L, Zheng Y, Dong T. Comparison of different predictive biomarker testing assays for PD-1/PD-L1 checkpoint inhibitors response: a systematic review and network meta-analysis. Front Immunol. 2023;14:1265202.PubMedPubMedCentralCrossRef Shi H, Zhang W, Zhang L, Zheng Y, Dong T. Comparison of different predictive biomarker testing assays for PD-1/PD-L1 checkpoint inhibitors response: a systematic review and network meta-analysis. Front Immunol. 2023;14:1265202.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Ready N, Hellmann MD, Awad MM, Otterson GA, Gutierrez M, Gainor JF, et al. First-line nivolumab plus ipilimumab in advanced non-small-cell lung cancer (CheckMate 568): outcomes by programmed death ligand 1 and tumor mutational burden as biomarkers. J Clin Oncol. 2019;37(12):992–1000.PubMedPubMedCentralCrossRef Ready N, Hellmann MD, Awad MM, Otterson GA, Gutierrez M, Gainor JF, et al. First-line nivolumab plus ipilimumab in advanced non-small-cell lung cancer (CheckMate 568): outcomes by programmed death ligand 1 and tumor mutational burden as biomarkers. J Clin Oncol. 2019;37(12):992–1000.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Deng H, Zhao Y, Cai X, Chen H, Cheng B, Zhong R, et al. PD-L1 expression and tumor mutation burden as pathological response biomarkers of neoadjuvant immunotherapy for early-stage non-small cell lung cancer: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2022;170:103582.PubMedCrossRef Deng H, Zhao Y, Cai X, Chen H, Cheng B, Zhong R, et al. PD-L1 expression and tumor mutation burden as pathological response biomarkers of neoadjuvant immunotherapy for early-stage non-small cell lung cancer: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2022;170:103582.PubMedCrossRef
66.
Zurück zum Zitat Jovanovic D, Roksandic Milenkovic M, Kotur Stevuljevic J, Markovic J, Ceriman V, Kontic M, et al. Membrane PD-L1 expression and soluble PD-L1 plasma levels in idiopathic pulmonary fibrosis-a pilot study. J Thorac Dis. 2018;10(12):6660–9.PubMedPubMedCentralCrossRef Jovanovic D, Roksandic Milenkovic M, Kotur Stevuljevic J, Markovic J, Ceriman V, Kontic M, et al. Membrane PD-L1 expression and soluble PD-L1 plasma levels in idiopathic pulmonary fibrosis-a pilot study. J Thorac Dis. 2018;10(12):6660–9.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Wang B, Bai W, Ma H, Li F. Regulatory effect of PD1/PD-ligand 1 (PD-L1) on treg cells in patients with idiopathic pulmonary fibrosis. Med Sci Monit. 2021;27:e927577.PubMedPubMedCentral Wang B, Bai W, Ma H, Li F. Regulatory effect of PD1/PD-ligand 1 (PD-L1) on treg cells in patients with idiopathic pulmonary fibrosis. Med Sci Monit. 2021;27:e927577.PubMedPubMedCentral
70.
Zurück zum Zitat Roksandic Milenkovic M, Klisic A, Ceriman V, Kotur Stevuljevic J, Savic Vujovic K, Mirkov D, et al. Oxidative stress and inflammation parameters-novel biomarkers for idiopathic pulmonary fibrosis. Eur Rev Med Pharmacol Sci. 2022;26(3):927–34.PubMed Roksandic Milenkovic M, Klisic A, Ceriman V, Kotur Stevuljevic J, Savic Vujovic K, Mirkov D, et al. Oxidative stress and inflammation parameters-novel biomarkers for idiopathic pulmonary fibrosis. Eur Rev Med Pharmacol Sci. 2022;26(3):927–34.PubMed
71.
Zurück zum Zitat Guyard A, Danel C, Théou-Anton N, Debray MP, Gibault L, Mordant P, et al. Morphologic and molecular study of lung cancers associated with idiopathic pulmonary fibrosis and other pulmonary fibroses. Respir Res. 2017;18(1):120.PubMedPubMedCentralCrossRef Guyard A, Danel C, Théou-Anton N, Debray MP, Gibault L, Mordant P, et al. Morphologic and molecular study of lung cancers associated with idiopathic pulmonary fibrosis and other pulmonary fibroses. Respir Res. 2017;18(1):120.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Asai Y, Chiba H, Nishikiori H, Kamekura R, Yabe H, Kondo S, et al. Aberrant populations of circulating T follicular helper cells and regulatory B cells underlying idiopathic pulmonary fibrosis. Respir Res. 2019;20(1):244.PubMedPubMedCentralCrossRef Asai Y, Chiba H, Nishikiori H, Kamekura R, Yabe H, Kondo S, et al. Aberrant populations of circulating T follicular helper cells and regulatory B cells underlying idiopathic pulmonary fibrosis. Respir Res. 2019;20(1):244.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Li Q, Deng MS, Wang RT, Luo H, Luo YY, Zhang DD, et al. PD-L1 upregulation promotes drug-induced pulmonary fibrosis by inhibiting vimentin degradation. Pharmacol Res. 2023;187:106636.PubMedCrossRef Li Q, Deng MS, Wang RT, Luo H, Luo YY, Zhang DD, et al. PD-L1 upregulation promotes drug-induced pulmonary fibrosis by inhibiting vimentin degradation. Pharmacol Res. 2023;187:106636.PubMedCrossRef
76.
Zurück zum Zitat Cui L, Chen SY, Lerbs T, Lee JW, Domizi P, Gordon S, et al. Activation of JUN in fibroblasts promotes pro-fibrotic programme and modulates protective immunity. Nat Commun. 2020;11(1):2795.PubMedPubMedCentralCrossRef Cui L, Chen SY, Lerbs T, Lee JW, Domizi P, Gordon S, et al. Activation of JUN in fibroblasts promotes pro-fibrotic programme and modulates protective immunity. Nat Commun. 2020;11(1):2795.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Lu Y, Zhong W, Liu Y, Chen W, Zhang J, Zeng Z, et al. Anti-PD-L1 antibody alleviates pulmonary fibrosis by inducing autophagy via inhibition of the PI3K/Akt/mTOR pathway. Int Immunopharmacol. 2022;104:108504.PubMedCrossRef Lu Y, Zhong W, Liu Y, Chen W, Zhang J, Zeng Z, et al. Anti-PD-L1 antibody alleviates pulmonary fibrosis by inducing autophagy via inhibition of the PI3K/Akt/mTOR pathway. Int Immunopharmacol. 2022;104:108504.PubMedCrossRef
78.
Zurück zum Zitat Wang D, Gong L, Li Z, Chen H, Xu M, Rong R, et al. Antifibrotic effect of Gancao Ganjiang decoction is mediated by PD-1 / TGF-β1 / IL-17A pathway in bleomycin-induced idiopathic pulmonary fibrosis. J Ethnopharmacol. 2021;281:114522.PubMedCrossRef Wang D, Gong L, Li Z, Chen H, Xu M, Rong R, et al. Antifibrotic effect of Gancao Ganjiang decoction is mediated by PD-1 / TGF-β1 / IL-17A pathway in bleomycin-induced idiopathic pulmonary fibrosis. J Ethnopharmacol. 2021;281:114522.PubMedCrossRef
79.
Zurück zum Zitat Walsh NC, Kenney LL, Jangalwe S, Aryee KE, Greiner DL, Brehm MA, et al. Humanized mouse models of clinical disease. Annu Rev Pathol. 2017;12:187–215.PubMedCrossRef Walsh NC, Kenney LL, Jangalwe S, Aryee KE, Greiner DL, Brehm MA, et al. Humanized mouse models of clinical disease. Annu Rev Pathol. 2017;12:187–215.PubMedCrossRef
80.
Zurück zum Zitat Yan J, Zheng X, You W, He W, Xu GK. A Bionic-homodimerization strategy for optimizing modulators of protein-protein interactions: from statistical mechanics theory to potential clinical translation. Adv Sci. 2022;9(11):e2105179.CrossRef Yan J, Zheng X, You W, He W, Xu GK. A Bionic-homodimerization strategy for optimizing modulators of protein-protein interactions: from statistical mechanics theory to potential clinical translation. Adv Sci. 2022;9(11):e2105179.CrossRef
81.
Zurück zum Zitat He W, Zhang Z, Yang W, Zheng X, You W, Yao Y, et al. Turing milk into pro-apoptotic oral nanotherapeutic: De novo bionic chiral-peptide supramolecule for cancer targeted and immunological therapy. Theranostics. 2022;12(5):2322–34.PubMedPubMedCentralCrossRef He W, Zhang Z, Yang W, Zheng X, You W, Yao Y, et al. Turing milk into pro-apoptotic oral nanotherapeutic: De novo bionic chiral-peptide supramolecule for cancer targeted and immunological therapy. Theranostics. 2022;12(5):2322–34.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Habiel DM, Espindola MS, Kitson C, Azzara AV, Coelho AL, Stripp B, et al. Characterization of CD28(null) T cells in idiopathic pulmonary fibrosis. Mucosal Immunol. 2019;12(1):212–22.PubMedCrossRef Habiel DM, Espindola MS, Kitson C, Azzara AV, Coelho AL, Stripp B, et al. Characterization of CD28(null) T cells in idiopathic pulmonary fibrosis. Mucosal Immunol. 2019;12(1):212–22.PubMedCrossRef
83.
Zurück zum Zitat Zhao Y, Hao C, Li M, Qu Y, Guo Y, Deng X, et al. PD-1/PD-L1 inhibitor ameliorates silica-induced pulmonary fibrosis by maintaining systemic immune homeostasis. Biomed Pharmacother. 2022;148:112768.PubMedCrossRef Zhao Y, Hao C, Li M, Qu Y, Guo Y, Deng X, et al. PD-1/PD-L1 inhibitor ameliorates silica-induced pulmonary fibrosis by maintaining systemic immune homeostasis. Biomed Pharmacother. 2022;148:112768.PubMedCrossRef
84.
Zurück zum Zitat Li S, Shi J, Tang H. Animal models of drug-induced pulmonary fibrosis: an overview of molecular mechanisms and characteristics. Cell Biol Toxicol. 2022;38(5):699–723.PubMedCrossRef Li S, Shi J, Tang H. Animal models of drug-induced pulmonary fibrosis: an overview of molecular mechanisms and characteristics. Cell Biol Toxicol. 2022;38(5):699–723.PubMedCrossRef
85.
Zurück zum Zitat Carrington R, Jordan S, Pitchford SC, Page CP. Use of animal models in IPF research. Pulm Pharmacol Ther. 2018;51:73–8.PubMedCrossRef Carrington R, Jordan S, Pitchford SC, Page CP. Use of animal models in IPF research. Pulm Pharmacol Ther. 2018;51:73–8.PubMedCrossRef
86.
Zurück zum Zitat Murray RE, Gibson JE. A comparative study of paraquat intoxication in rats, guinea pigs and monkeys. Exp Mol Pathol. 1972;17(3):317–25.PubMedCrossRef Murray RE, Gibson JE. A comparative study of paraquat intoxication in rats, guinea pigs and monkeys. Exp Mol Pathol. 1972;17(3):317–25.PubMedCrossRef
87.
Zurück zum Zitat Habiel DM, Espindola MS, Coelho AL, Hogaboam CM. Modeling idiopathic pulmonary fibrosis in humanized severe combined immunodeficient mice. Am J Pathol. 2018;188(4):891–903.PubMedPubMedCentralCrossRef Habiel DM, Espindola MS, Coelho AL, Hogaboam CM. Modeling idiopathic pulmonary fibrosis in humanized severe combined immunodeficient mice. Am J Pathol. 2018;188(4):891–903.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Knochelmann HM, Dwyer CJ, Bailey SR, Amaya SM, Elston DM, Mazza-McCrann JM, et al. When worlds collide: Th17 and treg cells in cancer and autoimmunity. Cell Mol Immunol. 2018;15(5):458–69.PubMedPubMedCentralCrossRef Knochelmann HM, Dwyer CJ, Bailey SR, Amaya SM, Elston DM, Mazza-McCrann JM, et al. When worlds collide: Th17 and treg cells in cancer and autoimmunity. Cell Mol Immunol. 2018;15(5):458–69.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Koch MA, Tucker-Heard G, Perdue NR, Killebrew JR, Urdahl KB, Campbell DJ. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat Immunol. 2009;10(6):595–602.PubMedPubMedCentralCrossRef Koch MA, Tucker-Heard G, Perdue NR, Killebrew JR, Urdahl KB, Campbell DJ. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat Immunol. 2009;10(6):595–602.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, Pappu BP, et al. Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity. 2008;29(1):44–56.PubMedPubMedCentralCrossRef Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, Pappu BP, et al. Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity. 2008;29(1):44–56.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Cisneros J, Hagood J, Checa M, Ortiz-Quintero B, Negreros M, Herrera I, et al. Hypermethylation-mediated silencing of p14(ARF) in fibroblasts from idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2012;303(4):L295-303.PubMedPubMedCentralCrossRef Cisneros J, Hagood J, Checa M, Ortiz-Quintero B, Negreros M, Herrera I, et al. Hypermethylation-mediated silencing of p14(ARF) in fibroblasts from idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2012;303(4):L295-303.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Wang K, Zhang T, Lei Y, Li X, Jiang J, Lan J, et al. Identification of ANXA2 (annexin A2) as a specific bleomycin target to induce pulmonary fibrosis by impeding TFEB-mediated autophagic flux. Autophagy. 2018;14(2):269–82.PubMedPubMedCentralCrossRef Wang K, Zhang T, Lei Y, Li X, Jiang J, Lan J, et al. Identification of ANXA2 (annexin A2) as a specific bleomycin target to induce pulmonary fibrosis by impeding TFEB-mediated autophagic flux. Autophagy. 2018;14(2):269–82.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Araya J, Kojima J, Takasaka N, Ito S, Fujii S, Hara H, et al. Insufficient autophagy in idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2013;304(1):L56-69.PubMedCrossRef Araya J, Kojima J, Takasaka N, Ito S, Fujii S, Hara H, et al. Insufficient autophagy in idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2013;304(1):L56-69.PubMedCrossRef
97.
Zurück zum Zitat Conte E, Fruciano M, Fagone E, Gili E, Caraci F, Iemmolo M, et al. Inhibition of PI3K prevents the proliferation and differentiation of human lung fibroblasts into myofibroblasts: the role of class I P110 isoforms. PLoS ONE. 2011;6(10):e24663.PubMedPubMedCentralCrossRef Conte E, Fruciano M, Fagone E, Gili E, Caraci F, Iemmolo M, et al. Inhibition of PI3K prevents the proliferation and differentiation of human lung fibroblasts into myofibroblasts: the role of class I P110 isoforms. PLoS ONE. 2011;6(10):e24663.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Yu JZ, Ying Y, Liu Y, Sun CB, Dai C, Zhao S, et al. Antifibrotic action of Yifei Sanjie formula enhanced autophagy via PI3K-AKT-mTOR signaling pathway in mouse model of pulmonary fibrosis. Biomed Pharmacother. 2019;118:109293.PubMedCrossRef Yu JZ, Ying Y, Liu Y, Sun CB, Dai C, Zhao S, et al. Antifibrotic action of Yifei Sanjie formula enhanced autophagy via PI3K-AKT-mTOR signaling pathway in mouse model of pulmonary fibrosis. Biomed Pharmacother. 2019;118:109293.PubMedCrossRef
99.
Zurück zum Zitat Qin W, Zou J, Huang Y, Liu C, Kang Y, Han H, et al. Pirfenidone facilitates immune infiltration and enhances the antitumor efficacy of PD-L1 blockade in mice. Oncoimmunology. 2020;9(1):1824631.PubMedPubMedCentralCrossRef Qin W, Zou J, Huang Y, Liu C, Kang Y, Han H, et al. Pirfenidone facilitates immune infiltration and enhances the antitumor efficacy of PD-L1 blockade in mice. Oncoimmunology. 2020;9(1):1824631.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Yang S, Liu P, Jiang Y, Wang Z, Dai H, Wang C. Therapeutic applications of mesenchymal stem cells in idiopathic pulmonary fibrosis. Front Cell Dev Biol. 2021;9:639657.PubMedPubMedCentralCrossRef Yang S, Liu P, Jiang Y, Wang Z, Dai H, Wang C. Therapeutic applications of mesenchymal stem cells in idiopathic pulmonary fibrosis. Front Cell Dev Biol. 2021;9:639657.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Cramer P, Bresalier RS. Gastrointestinal and hepatic complications of immune checkpoint inhibitors. Curr Gastroenterol Rep. 2017;19(1):3.PubMedCrossRef Cramer P, Bresalier RS. Gastrointestinal and hepatic complications of immune checkpoint inhibitors. Curr Gastroenterol Rep. 2017;19(1):3.PubMedCrossRef
102.
Zurück zum Zitat Hofmann L, Forschner A, Loquai C, Goldinger SM, Zimmer L, Ugurel S, et al. Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy. Eur J Cancer. 2016;60:190–209.PubMedCrossRef Hofmann L, Forschner A, Loquai C, Goldinger SM, Zimmer L, Ugurel S, et al. Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of anti-PD-1 therapy. Eur J Cancer. 2016;60:190–209.PubMedCrossRef
103.
Zurück zum Zitat Tasaka Y, Honda T, Nishiyama N, Tsutsui T, Saito H, Watabe H, et al. Non-inferior clinical outcomes of immune checkpoint inhibitors in non-small cell lung cancer patients with interstitial lung disease. Lung Cancer. 2021;155:120–6.PubMedCrossRef Tasaka Y, Honda T, Nishiyama N, Tsutsui T, Saito H, Watabe H, et al. Non-inferior clinical outcomes of immune checkpoint inhibitors in non-small cell lung cancer patients with interstitial lung disease. Lung Cancer. 2021;155:120–6.PubMedCrossRef
104.
Zurück zum Zitat Kanai O, Kim YH, Demura Y, Kanai M, Ito T, Fujita K, et al. Efficacy and safety of nivolumab in non-small cell lung cancer with preexisting interstitial lung disease. Thorac Cancer. 2018;9(7):847–55.PubMedPubMedCentralCrossRef Kanai O, Kim YH, Demura Y, Kanai M, Ito T, Fujita K, et al. Efficacy and safety of nivolumab in non-small cell lung cancer with preexisting interstitial lung disease. Thorac Cancer. 2018;9(7):847–55.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Dobre IA, Frank AJ, D’Silva KM, Christiani DC, Okin D, Sharma A, et al. Outcomes of patients with interstitial lung disease receiving programmed cell death 1 inhibitors: a retrospective case series. Clin Lung Cancer. 2021;22(5):e738–44.PubMedPubMedCentralCrossRef Dobre IA, Frank AJ, D’Silva KM, Christiani DC, Okin D, Sharma A, et al. Outcomes of patients with interstitial lung disease receiving programmed cell death 1 inhibitors: a retrospective case series. Clin Lung Cancer. 2021;22(5):e738–44.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–39.PubMedPubMedCentralCrossRef Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–39.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Horn L, Spigel DR, Vokes EE, Holgado E, Ready N, Steins M, et al. Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung cancer: two-year outcomes from two randomized, open-label, phase III trials (CheckMate 017 and CheckMate 057). J Clin Oncol. 2017;35(35):3924–33.PubMedPubMedCentralCrossRef Horn L, Spigel DR, Vokes EE, Holgado E, Ready N, Steins M, et al. Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung cancer: two-year outcomes from two randomized, open-label, phase III trials (CheckMate 017 and CheckMate 057). J Clin Oncol. 2017;35(35):3924–33.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Yamakawa H, Oba T, Ohta H, Tsukahara Y, Kida G, Tsumiyama E, et al. Nintedanib allows retreatment with atezolizumab of combined non-small cell lung cancer/idiopathic pulmonary fibrosis after atezolizumab-induced pneumonitis: a case report. BMC Pulm Med. 2019;19(1):156.PubMedPubMedCentralCrossRef Yamakawa H, Oba T, Ohta H, Tsukahara Y, Kida G, Tsumiyama E, et al. Nintedanib allows retreatment with atezolizumab of combined non-small cell lung cancer/idiopathic pulmonary fibrosis after atezolizumab-induced pneumonitis: a case report. BMC Pulm Med. 2019;19(1):156.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Albayrak L, Türksoy VA, Khalilov R, Eftekhari A. Investigation of heavy metal exposure and trace element levels in acute exacerbation of COPD. J King Saud Univ Sci. 2023;35(1):6.CrossRef Albayrak L, Türksoy VA, Khalilov R, Eftekhari A. Investigation of heavy metal exposure and trace element levels in acute exacerbation of COPD. J King Saud Univ Sci. 2023;35(1):6.CrossRef
111.
Zurück zum Zitat Cao J, Cusanovich DA, Ramani V, Aghamirzaie D, Pliner HA, Hill AJ, et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science. 2018;361(6409):1380–5.PubMedPubMedCentralCrossRef Cao J, Cusanovich DA, Ramani V, Aghamirzaie D, Pliner HA, Hill AJ, et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science. 2018;361(6409):1380–5.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Montesi SB, Désogère P, Fuchs BC, Caravan P. Molecular imaging of fibrosis: recent advances and future directions. J Clin Investig. 2019;129(1):24–33.PubMedPubMedCentralCrossRef Montesi SB, Désogère P, Fuchs BC, Caravan P. Molecular imaging of fibrosis: recent advances and future directions. J Clin Investig. 2019;129(1):24–33.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Hunter CA, Jones SA. IL-6 as a keystone cytokine in health and disease. Nat Immunol. 2015;16(5):448–57.PubMedCrossRef Hunter CA, Jones SA. IL-6 as a keystone cytokine in health and disease. Nat Immunol. 2015;16(5):448–57.PubMedCrossRef
114.
Zurück zum Zitat Epstein Shochet G, Brook E, Bardenstein-Wald B, Shitrit D. TGF-β pathway activation by idiopathic pulmonary fibrosis (IPF) fibroblast derived soluble factors is mediated by IL-6 trans-signaling. Respir Res. 2020;21(1):56.PubMedPubMedCentralCrossRef Epstein Shochet G, Brook E, Bardenstein-Wald B, Shitrit D. TGF-β pathway activation by idiopathic pulmonary fibrosis (IPF) fibroblast derived soluble factors is mediated by IL-6 trans-signaling. Respir Res. 2020;21(1):56.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Yang L, Xia H, Gilbertsen A, Smith K, Racila E, Bitterman PB, et al. IL-8 concurrently promotes idiopathic pulmonary fibrosis mesenchymal progenitor cell senescence and PD-L1 expression enabling escape from immune cell surveillance. Am J Physiol Lung Cell Mol Physiol. 2023;324(6):L849–62.PubMedCrossRef Yang L, Xia H, Gilbertsen A, Smith K, Racila E, Bitterman PB, et al. IL-8 concurrently promotes idiopathic pulmonary fibrosis mesenchymal progenitor cell senescence and PD-L1 expression enabling escape from immune cell surveillance. Am J Physiol Lung Cell Mol Physiol. 2023;324(6):L849–62.PubMedCrossRef
116.
Zurück zum Zitat Mohammadi A, Balan I, Yadav S, Matos WF, Kharawala A, Gaddam M, et al. Post-COVID-19 pulmonary fibrosis. Cureus. 2022;14(3):e22770.PubMedPubMedCentral Mohammadi A, Balan I, Yadav S, Matos WF, Kharawala A, Gaddam M, et al. Post-COVID-19 pulmonary fibrosis. Cureus. 2022;14(3):e22770.PubMedPubMedCentral
117.
Zurück zum Zitat Liu J, Wang L, Zhao F, Tseng S, Narayanan C, Shura L, et al. Pre-clinical development of a humanized anti-CD47 antibody with anti-cancer therapeutic potential. PLoS ONE. 2015;10(9):e0137345.PubMedPubMedCentralCrossRef Liu J, Wang L, Zhao F, Tseng S, Narayanan C, Shura L, et al. Pre-clinical development of a humanized anti-CD47 antibody with anti-cancer therapeutic potential. PLoS ONE. 2015;10(9):e0137345.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Wang Y, Ni H, Zhou S, He K, Gao Y, Wu W, et al. Tumor-selective blockade of CD47 signaling with a CD47/PD-L1 bispecific antibody for enhanced anti-tumor activity and limited toxicity. Cancer Immunol Immunother CII. 2021;70(2):365–76.PubMedCrossRef Wang Y, Ni H, Zhou S, He K, Gao Y, Wu W, et al. Tumor-selective blockade of CD47 signaling with a CD47/PD-L1 bispecific antibody for enhanced anti-tumor activity and limited toxicity. Cancer Immunol Immunother CII. 2021;70(2):365–76.PubMedCrossRef
119.
Zurück zum Zitat Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, et al. Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci USA. 2006;103(35):13180–5.PubMedPubMedCentralCrossRef Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, et al. Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci USA. 2006;103(35):13180–5.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Zhang X, Zou Y, Liu Y, Cao Y, Zhu J, Zhang J, et al. Inhibition of PIM1 kinase attenuates bleomycin-induced pulmonary fibrosis in mice by modulating the ZEB1/E-cadherin pathway in alveolar epithelial cells. Mol Immunol. 2020;125:15–22.PubMedCrossRef Zhang X, Zou Y, Liu Y, Cao Y, Zhu J, Zhang J, et al. Inhibition of PIM1 kinase attenuates bleomycin-induced pulmonary fibrosis in mice by modulating the ZEB1/E-cadherin pathway in alveolar epithelial cells. Mol Immunol. 2020;125:15–22.PubMedCrossRef
121.
Zurück zum Zitat Singh A, Chakraborty S, Wong SW, Hefner NA, Stuart A, Qadir AS, et al. Nanoparticle targeting of de novo profibrotic macrophages mitigates lung fibrosis. Proc Natl Acad Sci USA. 2022;119(15):e2121098119.PubMedPubMedCentralCrossRef Singh A, Chakraborty S, Wong SW, Hefner NA, Stuart A, Qadir AS, et al. Nanoparticle targeting of de novo profibrotic macrophages mitigates lung fibrosis. Proc Natl Acad Sci USA. 2022;119(15):e2121098119.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Aran D, Looney AP, Liu L, Wu E, Fong V, Hsu A, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol. 2019;20(2):163–72.PubMedPubMedCentralCrossRef Aran D, Looney AP, Liu L, Wu E, Fong V, Hsu A, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol. 2019;20(2):163–72.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Hartley GP, Chow L, Ammons DT, Wheat WH, Dow SW. Programmed cell death ligand 1 (PD-L1) signaling regulates macrophage proliferation and activation. Cancer Immunol Res. 2018;6(10):1260–73.PubMedCrossRef Hartley GP, Chow L, Ammons DT, Wheat WH, Dow SW. Programmed cell death ligand 1 (PD-L1) signaling regulates macrophage proliferation and activation. Cancer Immunol Res. 2018;6(10):1260–73.PubMedCrossRef
125.
Zurück zum Zitat Fang W, Zhou T, Shi H, Yao M, Zhang D, Qian H, et al. Progranulin induces immune escape in breast cancer via up-regulating PD-L1 expression on tumor-associated macrophages (TAMs) and promoting CD8(+) T cell exclusion. J Exp Clin Cancer Res CR. 2021;40(1):4.PubMedCrossRef Fang W, Zhou T, Shi H, Yao M, Zhang D, Qian H, et al. Progranulin induces immune escape in breast cancer via up-regulating PD-L1 expression on tumor-associated macrophages (TAMs) and promoting CD8(+) T cell exclusion. J Exp Clin Cancer Res CR. 2021;40(1):4.PubMedCrossRef
126.
Zurück zum Zitat Batista A, Rodvold JJ, Xian S, Searles SC, Lew A, Iwawaki T, et al. IRE1α regulates macrophage polarization, PD-L1 expression, and tumor survival. PLoS Biol. 2020;18(6):e3000687.PubMedPubMedCentralCrossRef Batista A, Rodvold JJ, Xian S, Searles SC, Lew A, Iwawaki T, et al. IRE1α regulates macrophage polarization, PD-L1 expression, and tumor survival. PLoS Biol. 2020;18(6):e3000687.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Miyasato Y, Takashima Y, Takeya H, Yano H, Hayano A, Nakagawa T, et al. The expression of PD-1 ligands and IDO1 by macrophage/microglia in primary central nervous system lymphoma. J Clin Exp Hematopathol. 2018;58(2):95–101.CrossRef Miyasato Y, Takashima Y, Takeya H, Yano H, Hayano A, Nakagawa T, et al. The expression of PD-1 ligands and IDO1 by macrophage/microglia in primary central nervous system lymphoma. J Clin Exp Hematopathol. 2018;58(2):95–101.CrossRef
129.
Zurück zum Zitat Wang H, Wen Y, Wang L, Wang J, Chen H, Chen J, et al. DDR1 activation in macrophage promotes IPF by regulating NLRP3 inflammasome and macrophage reaction. Int Immunopharmacol. 2022;113(Pt A):109294.PubMedCrossRef Wang H, Wen Y, Wang L, Wang J, Chen H, Chen J, et al. DDR1 activation in macrophage promotes IPF by regulating NLRP3 inflammasome and macrophage reaction. Int Immunopharmacol. 2022;113(Pt A):109294.PubMedCrossRef
130.
Zurück zum Zitat Tran TH, Phuong Tran TT. Targeting the PD-1/PD-L1 axis for cancer treatment: a review on nanotechnology. Royal Soc Open Sci. 2022;9(4):211991.CrossRef Tran TH, Phuong Tran TT. Targeting the PD-1/PD-L1 axis for cancer treatment: a review on nanotechnology. Royal Soc Open Sci. 2022;9(4):211991.CrossRef
132.
Zurück zum Zitat Guo C, Zhang C, Xia Z, Song B, Hu W, Cui Y, et al. Nano-designed CO donor ameliorates bleomycin-induced pulmonary fibrosis via macrophage manipulation. J Controlled Release. 2022;341:566–77.CrossRef Guo C, Zhang C, Xia Z, Song B, Hu W, Cui Y, et al. Nano-designed CO donor ameliorates bleomycin-induced pulmonary fibrosis via macrophage manipulation. J Controlled Release. 2022;341:566–77.CrossRef
133.
Zurück zum Zitat Gupta D, Boora A, Thakur A, Gupta TK. Green and sustainable synthesis of nanomaterials: recent advancements and limitations. Environ Res. 2023;231(Pt 3):116316.PubMedCrossRef Gupta D, Boora A, Thakur A, Gupta TK. Green and sustainable synthesis of nanomaterials: recent advancements and limitations. Environ Res. 2023;231(Pt 3):116316.PubMedCrossRef
134.
Zurück zum Zitat Ahmadov IS, Bandaliyeva AA, Nasibova AN, Hasanova FV, Khalilov RI. The synthesis of the silver nanodrugs in the medicinal plant baikal skullcap (scutellaria baicalensis georgi) and their antioxidant, antibacterial activity. Adv Biol Earth Sci. 2020;5(2):103–18. Ahmadov IS, Bandaliyeva AA, Nasibova AN, Hasanova FV, Khalilov RI. The synthesis of the silver nanodrugs in the medicinal plant baikal skullcap (scutellaria baicalensis georgi) and their antioxidant, antibacterial activity. Adv Biol Earth Sci. 2020;5(2):103–18.
137.
Zurück zum Zitat Liu Y, Liu XJ, Zhang N, Yin MX, Dong JW, Zeng QX, et al. Berberine diminishes cancer cell PD-L1 expression and facilitates antitumor immunity via inhibiting the deubiquitination activity of CSN5. Acta Pharma Sinica B. 2020;10(12):2299–312.CrossRef Liu Y, Liu XJ, Zhang N, Yin MX, Dong JW, Zeng QX, et al. Berberine diminishes cancer cell PD-L1 expression and facilitates antitumor immunity via inhibiting the deubiquitination activity of CSN5. Acta Pharma Sinica B. 2020;10(12):2299–312.CrossRef
139.
Zurück zum Zitat Li Y, Qin W, Liang Q, Zeng J, Yang Q, Chen Y, et al. Bufei huoxue capsule alleviates bleomycin-induced pulmonary fibrosis in mice via TGF-β1/Smad2/3 signaling. J Ethnopharmacol. 2023;316:116733.PubMedCrossRef Li Y, Qin W, Liang Q, Zeng J, Yang Q, Chen Y, et al. Bufei huoxue capsule alleviates bleomycin-induced pulmonary fibrosis in mice via TGF-β1/Smad2/3 signaling. J Ethnopharmacol. 2023;316:116733.PubMedCrossRef
Metadaten
Titel
Inhibitor of PD-1/PD-L1: a new approach may be beneficial for the treatment of idiopathic pulmonary fibrosis
verfasst von
Jie Tan
Qianfei Xue
Xiao Hu
Junling Yang
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2024
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-024-04884-7

Weitere Artikel der Ausgabe 1/2024

Journal of Translational Medicine 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.