1. Introduction
Idiopathic pulmonary fibrosis (IPF) is a progressive, irreversible, and fatal lung disease that is characterized by thick collagen and ‘fibroblast foci’ in the lung interstitium, resulting in severe restriction of lung capacity and function [
1]. IPF is a rare disease that affects between 200,000 and 300,000 people in the Western world [
2]. Current treatments are limited by their lack of efficacy, side effects, or inability to effectively block fibrosis, making IPF a significant unmet medical need.
Research over the past two decades has elucidated several pathological processes that are integral to IPF initiation, including alveolar epithelial cell attrition and activation of transforming growth factor β1 (TGFβ1). However, the molecular drivers of disease progression remain less well defined. Many factors, including association with known environmental agents, connective tissue diseases, and exposure to drugs or radiation therapy have been proposed [
3,
4], while aging remains the strongest risk factor [
5,
6].
Transglutaminase 2 (TG2) is a calcium-activated multifunctional enzyme well known for its ability to form protein crosslinks through ε-(γ-glutamyl) lysine isopeptide bonds, which leads to resistance to proteolytic cleavage and, ultimately, stabilization and increased stiffness of the extracellular matrix (ECM) [
7]. The role of TG2 in human disease has been widely reported, such as in fibrosis [
8,
9,
10] and cancer [
11]. During fibrosis, TG2 was shown to be involved in the accumulation of the large latent TGFβ1 complex into the ECM which facilitates release of the latent TGFβ1 activation protein LAP from the large latent TGFβ1 complex. Active TGFβ1 from LAP promotes fibroblast-to-myofibroblast transition [
8,
9], Epithelial–Mesenchymal Transition (EMT) [
12], and Endothelial–Mesenchymal Transition (EndMT) [
13] and increased expression of TG2 in M2 macrophages. This induces ECM protein expression and deposition while inhibiting matrix degradation pathways, leading to matrix accumulation and progressive fibrosis. Importantly, TGFβ1 increases the expression of TG2, forming a self-propagating cycle [
11].
The importance of TG2 in IPF is well established, with high levels of TG2 detected in IPF patient lung tissues [
9] while TG2 knockout mice develop significantly less pulmonary fibrosis compared to wild type mice when treated with bleomycin [
14].
In our previous paper, we demonstrated the presence of increased TG2 in IPF lung fibroblasts and demonstrated a relationship between TG2 and TGFβ1 in IPF fibroblasts dependent on TG2’s crosslinking activity [
9]. Importantly, we showed the potential of using site-directed TG2-selective inhibitors as therapeutic agents for IPF by demonstrating that our TG2 selective inhibitor 1-155 can reduce matrix deposition and reverse the myofibroblast phenotype in IPF cells via preventing the activation of TGFβ1. This reduced myofibroblast biomarkers, such as α smooth muscle actin (αSMA), and inhibited excessive matrix protein deposition [
9]. Importantly, 1-155 treatment led to the reduction in TG2 expression and ECM deposition in both primary human IPF and TGFβ1-treated human pulmonary fibroblasts, indicating the potential of TG2 as a disease target for IPF [
9].
In other studies, we demonstrated that treatment with the TG2-selective and irreversible inhibitor 1-155 resulted in up to a 40% reduction in collagen deposition in a mouse Angiotensin II model of nephrosclerosis and a 60% reduction in infarct size in an acute myocardial infarction mouse model [
8]. Earlier studies with our peptidic inhibitor R281 reduced collagen deposition in a rat diabetic nephropathy model [
15]. These studies, together with the data from TG2 knockout mice, support the potential use of site-directed TG2-selective inhibitors in the treatment of lung fibrosis.
Here, for the first time, we demonstrate that our TG2 inhibitor 1-155 reduces fibrosis in vivo using a bleomycin mouse model of lung fibrosis with 1-155 administered via the intranasal route. Importantly, we show that 1-155 improves mouse lung function, correlating with its effect on inhibiting fibrosis.
2. Materials and Methods
Reagents and antibodies. The general reagents were purchased from Sigma-Aldrich (Dorset, UK), unless stated below.
Antibodies used in this study, including a rabbit monoclonal antibody against αSMA (EPR5386), a rabbit polyclonal antibody against TG2 (AB421), and a rabbit IgG control monoclonal antibody (EPR25A), were purchased from AbCam (Cambridge, UK). The secondary antibody was supplied in the DAB kit. TG2 inhibitor 1-155 (with >95% purity) was synthesized by Wuxi AppTec (Shanghai, China).
2.1. Cell Culture
Human pulmonary lung fibroblasts (PromoCell, Heidelberg, Germany) were cultured in fibroblast culture medium containing 2% (v/v) fatal calf serum, 1 ng/mL human fibroblast growth factor, and 5 µg/mL insulin (PromoCell, Heidelberg, Germany).
2.2. FN Staining
FN deposited by primary lung fibroblasts was detected as described previously [
16]. The cells (7 × 10
4/chamber in complete medium) were cultured for 72 h in chamber slides in the presence of TG2 inhibitor 1-155 (between 25 and 500 nM), while DMSO was used as the vehicle control. The cells were cultured with anti-FN antibody for 2 h and then fixed, permeabilized, and incubated with FITC-conjugated secondary antibody for 2 h at 37 °C. The cells were finally mounted with Vecta shield mounting medium (Vector Laboratories, Peterborough, UK) and the fluorescence signals visualized using a Leica epi-fluorescent microscope (Milton Keynes, UK). The fluorescence signal was measured using ImageJ (version 1.53e) and the cell EC
50 was analyzed using GraphPad Prism (version 9.4.1).
2.3. Compound Formulation
The physical and thermal properties of compound 1-155 as its native material (powder form) and formulated as a suspension were characterized by Polarized Light Microscopy (PLM) (LV100PL, Nikon, Nishioi, Japan). The native 1-155 powder was in its crystal form with a particle size of around 4–5.0 µm as measured by PLM. The particles were milled to achieve a smaller particle size of around 2–3 µm. A formulation of the 1-155 milled form was made using 0.1% (v/v) Tween 80 in 0.9% sodium chloride (w/v), pH 7.13, which formed a homogenous stable suspension at 1 mg/mL and 2.5 mg/mL with particle sizes in the respiratory range. The 1-155 native powder form was made in 5% DMSO + 95% (v/v) sulfobutylether -beta-cyclodextrin (SBECD), pH 7.7, which contains 20% SBECD in water (w/w). Formulations were prepared fresh daily prior to administration and kept at 5 °C.
The formulation of nintedanib was prepared in a 0.1% hydroxyethyl cellulose to a final concentration of 6 mg/mL.
2.4. General Animal Study Design
All animal-related research was conducted by CROs in accordance with 2010/63/EU and the national legislation regulating the use of laboratory animals in scientific research and for other purposes. Necessary procedures were taken to ensure randomization and blinding to avoid bias. The tissue samples were coded for histological analysis. The animals were acclimatized for at least a week prior to the start of the experiments.
2.5. Lung PK Analysis of 1-155
Lung PK study was conducted by Wuxi AppTec. Animal facilities and Animal Care and Use Program (Units 001369 & SYXKS(沪)2019-0024) are fully accredited by AAALAC (Association for Assessment and Accreditation of Laboratory Animal Care, International) and approved by the Science and Technology Commission of Shanghai Municipal Government. The AAALAC Guideline and the Chinese National Standards related to the care and use of laboratory animals were followed. In vivo protocols were approved by the institutional ethical committee IACUC (Institutional Animal Care and Use Committee), all in-life procedures were performed by trained and experienced scientists, in full compliance with the relevant international standards, EST123, Directive 2012/63/EU, OLAW PHS (Policy on Humane Care and Use of Laboratory Animals).
C57BL/6 male mice (body weight of 25 g, GemPharmatech, Nanjing, China) were used in the studies. The mice were kept in GC type IVC mouse cages with corncob bedding and maintained under standard laboratory conditions (temperature 20–26 °C, relative humidity 30–70%, 15~20 air changes per hour, artificial lighting with circadian cycle of 12 h). Pelleted food and water were provided ad libitum.
The study was divided into three groups and dosed with 1-155, 1 mg/mL with no suspension, 1 mg/mL with suspension, and 2.5 mg/mL of 1-155 with suspension. Compound 1-155 was given through intratracheal (IT) administration in a volume of 30 uL/mouse. At each time point, the animals were euthanized by CO2 and 400 μL blood was collected from three mice in each group. Then, the animal was perfused with ice cold 0.9% saline by systemic circulation. BALF samples were collected from the lung, then lung tissue was separated carefully and snap frozen into liquid nitrogen. The blood samples were centrifuged at 1560 g for 10 min at 4 °C to yield around 150 μL plasma. The plasma samples, lung tissues, and BALF samples were stored under −80 °C until LC-MS/MS analysis was performed. Plasma, BALF, and lung tissue samples were collected at 10 min, 30 min, 1 h, 2 h, 4 h, 6 h, and 24 h and analyzed for 1-155 using HPLC-MS using a UPLC (Shimadzu) chromatographic system equipped with an AB Sciex QTRAP 6500 mass spectrometer operated by Analyst 1.6 software packages (Applied Biosystems). Chromatographic separation was carried out through a Waters XBridge BEH C18 column 50 × 2.1 mm ID, 2.5 µm at a flow rate of 0.7 mL/min at room temperature. All data were acquired using Analyst 1.6 software (Applied Biosystems). Plasma concentration versus time data were analyzed by non-compartmental approaches using the WinNonlin software program (version 6.1, Pharsight, Mountain View, CA, USA). Non-compartmental analysis (Phoenix WinNonlin 8.3) was used to determine PK parameters.
2.6. In Vivo Efficacy Study Protocol
The efficacy was conducted by Selvita S.A. All animal-related research was conducted in accordance with 2010/63/EU and the national legislation regulating the use of laboratory animals in scientific research and for other purposes (Official Gazette 55/13). An Institutional Committee on Animal Research Ethics (CARE-Zg) oversees that animal-related procedures are not compromising animal welfare.
Additionally, 12-week-old C57BL/6 male mice (Charles River, Italy) were used in the studies. The mice were kept in type III polysulfonate cages with ALPHA-dri dust free bedding and cotton nestles and maintained under standard laboratory conditions (temperature 22± 2 °C, relative humidity 60 ± 5%, 15 air changes per hour, artificial lighting with circadian cycle of 12 h). Pelleted food and water were provided ad libitum.
Lung fibrosis was induced by intranasal (IN) administration of bleomycin (Santa Cruz Technologies, Dallas, TX, USA) with 30 µg/50 µL of 0.9% saline per mouse, which is approximately 1 mg/kg. Seven days after the bleomycin challenge, the animals were randomly divided into different treatment groups. Prior to IN administration, the mice were anesthetized with ketamine +xylazine combination. The control group received saline alone.
Table 1 shows the efficacy study design.
The animals were examined clinically twice daily and monitored according to the clinical signs and parameters based on a humane endpoints scale. The animals were weighed on D0, D3, and D6. D7 animals were weighed every day until D21.
On day 21, the mice were anesthetized and lung function measurements were performed, while the rest of the animals were overdosed with ketamine hydrochloride (Narkamon) and xylazine (Xylazine) administered intraperitoneally. In addition, the lungs of all animals were removed, weighed, formalin-fixed, and finally paraffin-embedded for histopathological analysis and analyzed together as one group.
Treatment design is shown in the table below.
2.7. Lung Function Measurement
Lung function measurements were performed on 8 animals/group (apart from 6 animals for the saline control and 5 animals for 1-155 IN treatment). The tests performed included Boyle’s law functional residual capacity (FRC) tests, quasistatic Pressure Volume tests (PV), and fast flow volume maneuver plus resistance and compliance tests. The mice were anesthetized with ketamine +xylazine combination. A pulmonary function test (PFT) was performed with measurements on Buxco® Pulmonary function testing system (DSI, MN, USA). The animals were intubated extra-orally and placed on a mechanical ventilator. A tracheal tube provided direct access to the lungs. Parameters measured using this technique included pulmonary resistance and dynamic lung compliance.
In the Buxco® software (version 2.9.0), a new PFT study was created, and animals were allocated to new measurement groups. The mice were anesthetized for surgery and after approx. 10 min, the mice were tracheotomized. Tracheostomy was performed by cutting skin in the neck area and releasing the trachea from the surrounding tissue. Tracheal tubes were placed into the trachea and fixed with a tied suture. Standard 18-gauge stainless steel tubes were used for mice and shortened to 25 mm length. The mice were then loaded into a plethysmograph for testing. Three semiautomatic maneuvers were performed: Boyle’s law functional residual capacity (FRC), quasistatic Pressure Volume test (PV), and fast flow volume maneuver.
Data were calculated using automated data acquisition software Buxco® FinePointe™. Statistical significance was determined from mean values using t-test (significance level set to 0.05).
2.8. Histopathological Evaluation
Whole lungs were embedded in paraffin and stained according to Crossman‘s Trichrome method. Pulmonary histological changes were assessed using Matsuse modification of the Ashcroft score. Slices of the whole pulmonary area were subjected to histopathological analysis and evaluated at 10× magnification and a total Ashcroft score for each animal was calculated as a mean value.
Statistical analysis and graphical presentation were performed using GraphPad Prism software (version 9.4.1). Outliers were identified using Grubbs test. For the evaluation of the Ashcroft score results, non-parametric statistics (Wilcoxon Signed Rank Test, Mann–Whitney test) was performed using group median. Differences between groups were considered statistically significant when p < 0.05.
2.9. Histological Analysis of Lung Tissues
Histochemistry for collagen analysis was performed using Picro-Sirius red staining. Immunohistochemistry was performed to detect the presence of TG2 and αSMA in the lung tissue slides. Following xylene dewaxing, the tissues were rehydrated using a gradient of alcohol (100–70%) and antigen retrieval was performed in a citrate-based antigen retrieval solution (pH 6) at 100 °C. Following rinsing the samples, the samples were blocked with 5% BSA in PBS, pH 7.4, for 1 h at room temperature. Novolink DAB kit (Leica) was used to block the endogenous hydrogen peroxidase. Suitable primary antibody in 1% BSA in PBS, pH 7.4, was incubated with the tissues at 4 °C overnight. Following rinsing, the slides were incubated with Novolink polymer for 30 min. The slides were rinsed, and signals were revealed using a DAB Substrate Buffer before drying and mounting the slides using Vector mounting media. Images were obtained using an EVOS imaging system.
Image analysis was conducted using Image J software. Data are expressed as the mean ± SEM for at least three independent replicate experiments. Statistical analysis was undertaken using Microsoft Excel, one way ANOVA with Bonferroni correction post hoc test, and a p value of <0.05 was considered to indicate statistical significance. Subsequent F-test two samples for variances and identification of significant differences between individual groups by Student’s t-test or Welch’s t-test were carried out.
2.10. Tolerability Study Setup
The tolerability study was conducted by Selvita S.A. See
Section 2.6 for further information.
The mice were assigned to 1 control group and 2 testing groups, each consisting of 6 males per group. Compound 1-155 at a dose of 3 mg/kg and 10 mg/kg was administered IN once daily for 14 days, while Group 1 received the vehicle (0.1% Tween 80 in saline (0.9% sodium chloride) alone as a control.
Subsequent observations and examinations included clinical signs (daily), body weight (daily), food consumption (daily), clinical laboratory investigation, and bioanalysis evaluation. At termination, following macroscopic examination, organ weights were determined and histopathology performed for selected tissue (lung and trachea).
4. Discussion
Several studies have identified TG2 as a promising disease target for IPF [
9,
14,
18]. It was reported that when challenged with bleomycin, TG2-deficient mice showed less fibrosis and metabolic changes in the lungs, compared to the wild type animals [
19]. However, the use of small molecule selective TG2 inhibitors, according to the available literature, has not been tested for efficacy in animal lung fibrosis models. Current IPF treatments, nintedanib and pirfenidone, can slow the rate of decline in lung function and disease progression. Nintedanib is a pan intracellular inhibitor, targeting multiple growth factors like tyrosine kinase receptors, including PDGF receptors, VEGF receptors, and FGF receptors [
20]. Pirfenidone functions via affecting various pathways, including interfering with growth factors (e.g., TGFβ, PDGF, and basic FGF), upregulating MMPs to attenuate matrix accumulation, modulating cytokines (e.g., interleukins), and regulating the activity and proliferation of T and B lymphocytes [
21]. Given the broad spectrum of physiological and pathological pathways affected by these treatments, they can cause side effects, such as diarrhea and nausea. Cases of abnormal liver function in nintedanib patients were reported, and both drugs are not recommended for patients with severe liver disease [
22,
23]. Therefore, there is an urgent unmet clinical need for novel treatments that are specific to fibrosis.
Our previous work has demonstrated that there is a self-propagating cycle between TG2 and TGFβ1 in driving fibroblasts into myofibroblast transition during IPF, which can be inhibited by TG2 small molecule inhibitor 1-155 [
9]. 1-155 is a TG2 inhibitor with high potency and selectivity, and has been effective in inhibiting fibrosis in several in vivo fibrosis models, including acute myocardial infarction [
8] angiotensin II-induced cardiac and renal fibrosis [
14]. No toxic effects were observed in these animal models for over 21 days when 1-155 was delivered s.c. using a mini pump [
8,
10].
Our current challenge in developing 1-155 as a treatment for fibrotic disease is its relatively high clearance, which may potentially limit systemic side effects but would limit the concentration distribution into the lungs when administrated IV or orally [
10]. Therefore, we have overcome these characteristics of 1-155 by administering it to the lung directly via the IN route. The advantage of topical administration via inhalation of the drug as used in humans should improve its potency and once cleared rapidly in the plasma, limit potential side effects.
By delivering 1-155 in a stable suspension IN, we demonstrate that 1-155, which has good cell permeability [
10], has a better chance to be retained by permeating into the lung sub-compartments and providing longer duration in the lung. At 3 mg/kg in suspension, 1-155 treatment resulted in a significant reduction in fibrosis when the presence of collagen in the lung tissue was analyzed in both lung tissue sections and the whole lung slices. Compared to its vehicle control, 1-155 treatment significantly reduced the Ashcroft scores, an indicator of pulmonary fibrosis, with a reduction in lung weight. Importantly, 1-155 showed a lower average Ashcroft score (2.7) than the positive control treatment nintedanib (3.0). Our findings agree with the effect of 1-155 on inhibiting fibrosis in the heart and kidneys, as we reported previously [
8,
10].
Further analysis revealed that the inhibition of bleomycin-induced pulmonary fibrosis was in part due to a significant reduction in lung myofibroblasts, as confirmed by the myofibroblast marker αSMA. Furthermore, 1-155 treatment led to the reduction in the high level of TG2 in bleomycin-treated lungs, validating that TG2 could be a reliable disease marker for progressive pulmonary fibrosis [
9,
14], such as IPF. In the future, it would also be interesting to explore the effect of 1-155 on pulmonary fibrosis in a longer-term chronic fibrosis model and investigate the possibility of co-staining intracellular TG2 with αSMA positive cells [
9].
The overall aim of our study was to demonstrate the potential of 1-155 as a treatment for IPF. Therefore, it was important for us to correlate pathology data with clinical readouts, i.e., the lung function analysis. For the first time, we are able to demonstrate a very clear correlation between the inhibition of pulmonary fibrosis by TG2 inhibitor 1-155 both by histopathology and its effects on improving lung function using the bleomycin-induced pulmonary fibrosis mouse model. Significant improvements in key lung function parameters by 1-155 include cord and dynamic compliance. This suggests that 1-155 treatment can help alleviate the restrictive problems associated with a diseased lung, as seen in the bleomycin-treated lungs of mice and in patients with IPF where myofibroblast proliferation and increased deposition of collagen restricts access of air to the lung tissue. In line with the improvements shown in compliance, an improvement was also shown for lung vital capacity and inspiratory capacity (
p = 0.06), suggesting these parameters were improved with 1-155 treatment. Although not significant, forced vital capacity and peaked respiratory flow were also improved by 1-155 treatment, compared to its vehicle control. Forced vital capacity decline is predictive of mortality in patients with IPF and is presently used as a clinical trial endpoint to define disease progression [
24]. It is important to note that a much higher dosage of nintedanib (60 mg/kg, twice daily) was used compared to 1-155 (3 mg/kg, twice daily).
In a tolerability study, 1-155 delivered IN at 3 mg/kg and 10 mg/kg in healthy animals caused no serious toxic effects over 14 days and in the efficacy study, 1-155-treated animals did show a small improvement in body weight between day 9 to day 21. However, despite the above observations, a lower survival rate was observed in the animals treated IN with 1-155 when bleomycin treatment was introduced.