Exploration of a potent PI3 kinase/mTOR inhibitor as a novel anti-fibrotic agent in IPF

Paul F Mercer, Hannah V Woodcock, Jessica D Eley, Manuela Platé, Michal G Sulikowski, Pascal F Durrenberger, Linda Franklin, Carmel B Nanthakumar, Yim Man, Federica Genovese, Robin J McAnulty, Shuying Yang, Toby M Maher, Andrew G Nicholson, Andy D Blanchard, Richard P Marshall, Pauline T Lukey, Rachel C Chambers, Paul F Mercer, Hannah V Woodcock, Jessica D Eley, Manuela Platé, Michal G Sulikowski, Pascal F Durrenberger, Linda Franklin, Carmel B Nanthakumar, Yim Man, Federica Genovese, Robin J McAnulty, Shuying Yang, Toby M Maher, Andrew G Nicholson, Andy D Blanchard, Richard P Marshall, Pauline T Lukey, Rachel C Chambers

Abstract

Rationale: Idiopathic pulmonary fibrosis (IPF) is the most rapidly progressive and fatal of all fibrotic conditions with no curative therapies. Common pathomechanisms between IPF and cancer are increasingly recognised, including dysfunctional pan-PI3 kinase (PI3K) signalling as a driver of aberrant proliferative responses. GSK2126458 is a novel, potent, PI3K/mammalian target of rapamycin (mTOR) inhibitor which has recently completed phase I trials in the oncology setting. Our aim was to establish a scientific and dosing framework for PI3K inhibition with this agent in IPF at a clinically developable dose.

Methods: We explored evidence for pathway signalling in IPF lung tissue and examined the potency of GSK2126458 in fibroblast functional assays and precision-cut IPF lung tissue. We further explored the potential of IPF patient-derived bronchoalveolar lavage (BAL) cells to serve as pharmacodynamic biosensors to monitor GSK2126458 target engagement within the lung.

Results: We provide evidence for PI3K pathway activation in fibrotic foci, the cardinal lesions in IPF. GSK2126458 inhibited PI3K signalling and functional responses in IPF-derived lung fibroblasts, inhibiting Akt phosphorylation in IPF lung tissue and BAL derived cells with comparable potency. Integration of these data with GSK2126458 pharmacokinetic data from clinical trials in cancer enabled modelling of an optimal dosing regimen for patients with IPF.

Conclusions: Our data define PI3K as a promising therapeutic target in IPF and provide a scientific and dosing framework for progressing GSK2126458 to clinical testing in this disease setting. A proof-of-mechanism trial of this agent is currently underway.

Trial registration number: NCT01725139, pre-clinical.

Keywords: Idiopathic pulmonary fibrosis.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/

Figures

Figure 1
Figure 1
Akt phosphorylation co-localised within fibrotic foci, macrophages and bronchial epithelium in the idiopathic pulmonary fibrosis (IPF) lung. Immunofluorescence for α smooth muscle actin (αSMA, green, A and D), Akt phosphorylated at serine 473 (pAktS473, red, B) and threonine 308 (pAktT308, red, E) is shown co-localised (C and F) within fibrotic foci (A–F). Immunofluorescence for CD68 (green, G and J), pAktS473 (red, H), pAktT308 (red, K) is shown co-localised (I and L) within macrophages in airspaces (panels G–L). In panels M–R immunofluorescence for pAktS473 (red, N), pAktT308 (red, Q) is shown localised to unstained cells morphologically characteristic of bronchial epithelium (N, Q, O and R). Cells morphologically characteristic of airway smooth muscle are evident (green, M and P). DAPI-stained nuclei are highlighted in blue. 25 μm scale bars are shown.
Figure 2
Figure 2
Pan-PI3 kinase (PI3K) dual mammalian target of rapamycin (mTOR) inhibition reduces Akt phosphorylation in human idiopathic pulmonary fibrosis (IPF) lung slices. Precision cut lung slices were treated with vehicle or increasing concentrations of PI3K inhibitor (Compound 1) for 2 h (0.1% DMSO vehicle was constant for all experimental conditions). Following harvest and homogenisation, pAkt levels were assessed by Milliplex Map transforming growth factor (TGF)-β signalling beads and normalised to glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Data are shown for mean fluorescence intensity (MFI) normalised to GAPDH as ±SEM of n=4 technical replicate wells per condition; *p

Figure 3

GSK2126458 inhibits Akt phosphorylation and…

Figure 3

GSK2126458 inhibits Akt phosphorylation and proliferation in normal and idiopathic pulmonary fibrosis (IPF)…

Figure 3
GSK2126458 inhibits Akt phosphorylation and proliferation in normal and idiopathic pulmonary fibrosis (IPF) primary human lung fibroblasts. Control (C-LF, n=2, A) or IPF lung fibroblasts (IPF-LF, n=5, C) were pre-incubated with increasing concentrations of GSK2126458. Following 30 min stimulation with 20% FCS, pAktS473 signal was normalised to total Akt and expressed as % maximum response (maximum asymptote) and is shown as mean±SEM of n=3 replicate wells per condition. A representative Western blot is shown visualising the effect of GSK2126458 on FCS-induced phosphorylation of AktS473 in a representative C-LF line (E). Subconfluent control (C-LF, n=2, B) or IPF lung fibroblasts (IPF-LF, n=5, D) were pre-incubated with increasing concentrations of GSK2126458. Following 72 h stimulation with 10% FCS, fibroblast proliferation was measured using an MTS assay at 490 nm. Data were expressed as % maximum response (maximum asymptote) and are shown as mean±n=6 replicate wells per condition. Incorporation of EdU (5-ethynyl-2′deoxyuridine) into the DNA of sub-confluent fibroblasts was also used as a measured of proliferation following 72 h of FCS stimulation (C-LF, n=5, dashed lines; IPF-LF, n=2, solid lines (F)). IC50 values (panels A–D and F) were calculated using four-parameter non-linear regression, and representative curves are shown for each fibroblast line. FCS stimulated and unstimulated DMSO vehicle control fibroblast data points are indicated as (+) and (−), respectively (panels A–F). DMSO was maintained at 0.1% for all conditions. Activation of fibroblast caspase 3 and 7 was assessed following incubation with increasing concentrations of GSK2126458 in the presence of 10% FCS by Caspase Glo (G). FCS-stimulated (+) vehicle control (0.1% DMSO) fibroblasts are indicated. Data are expressed as fold relative to baseline cells (no FCS, 0.1% DMSO) and shown as mean±SEM of n=3 replicate wells per condition. **p<0.0001 for differences in raw data values from FCS-stimulated controls (two-way ANOVA, Tukey's multiple comparisons test).

Figure 4

GSK2126458 inhibits Akt phosphorylation in…

Figure 4

GSK2126458 inhibits Akt phosphorylation in bronchoalveolar lavage (BALF) cells from patients with idiopathic…

Figure 4
GSK2126458 inhibits Akt phosphorylation in bronchoalveolar lavage (BALF) cells from patients with idiopathic pulmonary fibrosis (IPF). IPF BALF cells (n=6) were incubated with 0.1% DMSO (vehicle control) or increasing concentrations of GSK2126458 for 30 min at 37°C (0.1% DMSO vehicle was constant for all experimental conditions). Cell lysates were assayed for total Akt and pAktS473 using Meso Scale Discovery (MSD) electro-chemiluminescence technology. pAktS473 signal was normalised to total Akt signal and expressed as % of a reference vehicle control well for each experiment (Veh). Data are shown as mean±SEM of triplicate wells per condition. IC50 values were calculated using four-parameter non-linear regression.

Figure 5

Construction of an integrated pharmacokinetic…

Figure 5

Construction of an integrated pharmacokinetic (PK)/pharmacodynamics (PD) model for GSK2126458 in patients with…

Figure 5
Construction of an integrated pharmacokinetic (PK)/pharmacodynamics (PD) model for GSK2126458 in patients with idiopathic pulmonary fibrosis (IPF). Schematic illustrating the strategy for integration of in vitro PK/PD data with human PK for clinical dose response estimation in IPF. Equation (1): Inh represents the % inhibition of pAkt (IPF myofibroblasts or bronchoalveolar lavage fluid (BALF) cell isolates) of fibroblast proliferation (IPF myofibroblasts), EMAX=maximum estimated inhibition, EC50=drug potency and shape=gradient of the response curve. ε refers to residual or unexplained variability, the subscript i indicates replicate cell line, j is individual drug concentration from each cell line.

Figure 6

Effect of GSK2126458 on epithelial…

Figure 6

Effect of GSK2126458 on epithelial cell apoptosis. Activation of epithelial caspase 3 and…

Figure 6
Effect of GSK2126458 on epithelial cell apoptosis. Activation of epithelial caspase 3 and 7 was assessed in primary bronchial epithelial cultures following incubation with increasing concentrations of GSK2126458 for 72 h (0.1% DMSO vehicle was constant for all experimental conditions). Human bronchial epithelial cell (HBEC) cultures were derived from n=2 control and n=1 idiopathic pulmonary fibrosis (IPF) donor lungs. Data are expressed fold relative to baseline (0.1% DMSO) controls (Veh) and shown as mean±SEM of n=6 replicate wells per condition; **p

Figure 7

Time course of the primary…

Figure 7

Time course of the primary human fibroblast response to transforming growth factor (TGF)-β…

Figure 7
Time course of the primary human fibroblast response to transforming growth factor (TGF)-β1. Confluent lung fibroblasts (LFs) were stimulated with TGFβ1 (1 ng/mL) and lysates collected over the indicated timecourse (A, B and D). Phosphorylation of SMAD2 or AktS473 is shown by Western blot (A) and densitometry (B and D). Collagen gene expression in TGFβ1 (1 ng/mL) stimulated (C, dark triangles and dashed line) or unstimulated (C, light triangles and dashed line) was assessed in LFs over the indicated time course. Pro-collagen recovered from cell supernatants over the same time course in TGFβ1 stimulated (C, dark bar) or unstimulated (C, light bar) is also shown. Serum-free confluent primary human LFs (HLFs) were incubated with increasing concentrations of GSK2126458 or 0.1% DMSO and stimulated with TGFβ1 (1 ng/mL) for 12 h (0.1% DMSO vehicle was constant for all experimental conditions). Phosphorylation of SMAD2 and AktS473 is shown by representative Western blot (E). Densitometry illustrating the effects of GSK2126458 on pAktS473 in HLFs from n=3 donors is shown (F).

Figure 8

Pan-PI3 kinase (PI3K)/mammalian target of…

Figure 8

Pan-PI3 kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibition attenuates transforming growth factor (TGF)-β…

Figure 8
Pan-PI3 kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibition attenuates transforming growth factor (TGF)-β1-induced collagen production in human lung fibroblasts (LFs), and collagen formation markers in idiopathic pulmonary fibrosis (IPF) lung tissue. Confluent LFs were incubated with increasing concentrations of GSK2126458 (A) or SB525334 (B) and stimulated for 48 h with TGFβ1 (1 ng/mL) and collagen biosynthesis assayed by molecular crowding assay (0.1% DMSO vehicle was constant for all experimental conditions). Data are expressed as mean fluorescent intensity (n=4 reads per well) and cell counts obtained from DAPI counterstaining (blue channel). Data are shown as mean±SEM of n=3 replicate wells per condition. IC50 values were calculated using four-parameter non-linear regression. Replicate experiments were carried out on human lung fibroblasts (HLFs) from a non-IPF donor and representative curves are shown. Images from each cell treatment are shown (C). 8 mm slices of IPF lung were cultured for 24 h in DMEM supplemented with 0.4% FCS and treated with vehicle, 10 μM SB525334 (open circles) or increasing concentrations of Compound 1 (inhibitor) for 0–3 days (filled grey circles) and further from 3 to 5 days (filled black circles) following media change. P1NP levels were measured in harvested supernatants (D). Data are shown as ±SEM of n=4 replicate wells per condition; *p<0.05, **p<0.01 (two-way ANOVA with Bonferroni post-hoc testing).
All figures (8)
Comment in
Similar articles
Cited by
References
    1. Lee AS, Mira-Avendano I, Ryu JH, et al. . The burden of idiopathic pulmonary fibrosis: an unmet public health need. Respir Med 2014;108:955–67. 10.1016/j.rmed.2014.03.015 - DOI - PubMed
    1. Richeldi L, du Bois RM, Raghu G, et al. . Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med 2014;370:2071–82. 10.1056/NEJMoa1402584 - DOI - PubMed
    1. King TE Jr, Bradford WZ, Castro-Bernardini S, et al. . A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med 2014;370:2083–92. 10.1056/NEJMoa1402582 - DOI - PubMed
    1. Datta A, Scotton CJ, Chambers RC. Novel therapeutic approaches for pulmonary fibrosis. Br J Pharmacol 2011;163:141–72. 10.1111/j.1476-5381.2011.01247.x - DOI - PMC - PubMed
    1. Akhurst RJ, Hata A. Targeting the TGFβ signalling pathway in disease. Nat Rev Drug Discov 2012;11:790–811. 10.1038/nrd3810 - DOI - PMC - PubMed
Show all 39 references
MeSH terms
Associated data
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3
Figure 3
GSK2126458 inhibits Akt phosphorylation and proliferation in normal and idiopathic pulmonary fibrosis (IPF) primary human lung fibroblasts. Control (C-LF, n=2, A) or IPF lung fibroblasts (IPF-LF, n=5, C) were pre-incubated with increasing concentrations of GSK2126458. Following 30 min stimulation with 20% FCS, pAktS473 signal was normalised to total Akt and expressed as % maximum response (maximum asymptote) and is shown as mean±SEM of n=3 replicate wells per condition. A representative Western blot is shown visualising the effect of GSK2126458 on FCS-induced phosphorylation of AktS473 in a representative C-LF line (E). Subconfluent control (C-LF, n=2, B) or IPF lung fibroblasts (IPF-LF, n=5, D) were pre-incubated with increasing concentrations of GSK2126458. Following 72 h stimulation with 10% FCS, fibroblast proliferation was measured using an MTS assay at 490 nm. Data were expressed as % maximum response (maximum asymptote) and are shown as mean±n=6 replicate wells per condition. Incorporation of EdU (5-ethynyl-2′deoxyuridine) into the DNA of sub-confluent fibroblasts was also used as a measured of proliferation following 72 h of FCS stimulation (C-LF, n=5, dashed lines; IPF-LF, n=2, solid lines (F)). IC50 values (panels A–D and F) were calculated using four-parameter non-linear regression, and representative curves are shown for each fibroblast line. FCS stimulated and unstimulated DMSO vehicle control fibroblast data points are indicated as (+) and (−), respectively (panels A–F). DMSO was maintained at 0.1% for all conditions. Activation of fibroblast caspase 3 and 7 was assessed following incubation with increasing concentrations of GSK2126458 in the presence of 10% FCS by Caspase Glo (G). FCS-stimulated (+) vehicle control (0.1% DMSO) fibroblasts are indicated. Data are expressed as fold relative to baseline cells (no FCS, 0.1% DMSO) and shown as mean±SEM of n=3 replicate wells per condition. **p<0.0001 for differences in raw data values from FCS-stimulated controls (two-way ANOVA, Tukey's multiple comparisons test).
Figure 4
Figure 4
GSK2126458 inhibits Akt phosphorylation in bronchoalveolar lavage (BALF) cells from patients with idiopathic pulmonary fibrosis (IPF). IPF BALF cells (n=6) were incubated with 0.1% DMSO (vehicle control) or increasing concentrations of GSK2126458 for 30 min at 37°C (0.1% DMSO vehicle was constant for all experimental conditions). Cell lysates were assayed for total Akt and pAktS473 using Meso Scale Discovery (MSD) electro-chemiluminescence technology. pAktS473 signal was normalised to total Akt signal and expressed as % of a reference vehicle control well for each experiment (Veh). Data are shown as mean±SEM of triplicate wells per condition. IC50 values were calculated using four-parameter non-linear regression.
Figure 5
Figure 5
Construction of an integrated pharmacokinetic (PK)/pharmacodynamics (PD) model for GSK2126458 in patients with idiopathic pulmonary fibrosis (IPF). Schematic illustrating the strategy for integration of in vitro PK/PD data with human PK for clinical dose response estimation in IPF. Equation (1): Inh represents the % inhibition of pAkt (IPF myofibroblasts or bronchoalveolar lavage fluid (BALF) cell isolates) of fibroblast proliferation (IPF myofibroblasts), EMAX=maximum estimated inhibition, EC50=drug potency and shape=gradient of the response curve. ε refers to residual or unexplained variability, the subscript i indicates replicate cell line, j is individual drug concentration from each cell line.
Figure 6
Figure 6
Effect of GSK2126458 on epithelial cell apoptosis. Activation of epithelial caspase 3 and 7 was assessed in primary bronchial epithelial cultures following incubation with increasing concentrations of GSK2126458 for 72 h (0.1% DMSO vehicle was constant for all experimental conditions). Human bronchial epithelial cell (HBEC) cultures were derived from n=2 control and n=1 idiopathic pulmonary fibrosis (IPF) donor lungs. Data are expressed fold relative to baseline (0.1% DMSO) controls (Veh) and shown as mean±SEM of n=6 replicate wells per condition; **p

Figure 7

Time course of the primary…

Figure 7

Time course of the primary human fibroblast response to transforming growth factor (TGF)-β…

Figure 7
Time course of the primary human fibroblast response to transforming growth factor (TGF)-β1. Confluent lung fibroblasts (LFs) were stimulated with TGFβ1 (1 ng/mL) and lysates collected over the indicated timecourse (A, B and D). Phosphorylation of SMAD2 or AktS473 is shown by Western blot (A) and densitometry (B and D). Collagen gene expression in TGFβ1 (1 ng/mL) stimulated (C, dark triangles and dashed line) or unstimulated (C, light triangles and dashed line) was assessed in LFs over the indicated time course. Pro-collagen recovered from cell supernatants over the same time course in TGFβ1 stimulated (C, dark bar) or unstimulated (C, light bar) is also shown. Serum-free confluent primary human LFs (HLFs) were incubated with increasing concentrations of GSK2126458 or 0.1% DMSO and stimulated with TGFβ1 (1 ng/mL) for 12 h (0.1% DMSO vehicle was constant for all experimental conditions). Phosphorylation of SMAD2 and AktS473 is shown by representative Western blot (E). Densitometry illustrating the effects of GSK2126458 on pAktS473 in HLFs from n=3 donors is shown (F).

Figure 8

Pan-PI3 kinase (PI3K)/mammalian target of…

Figure 8

Pan-PI3 kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibition attenuates transforming growth factor (TGF)-β…

Figure 8
Pan-PI3 kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibition attenuates transforming growth factor (TGF)-β1-induced collagen production in human lung fibroblasts (LFs), and collagen formation markers in idiopathic pulmonary fibrosis (IPF) lung tissue. Confluent LFs were incubated with increasing concentrations of GSK2126458 (A) or SB525334 (B) and stimulated for 48 h with TGFβ1 (1 ng/mL) and collagen biosynthesis assayed by molecular crowding assay (0.1% DMSO vehicle was constant for all experimental conditions). Data are expressed as mean fluorescent intensity (n=4 reads per well) and cell counts obtained from DAPI counterstaining (blue channel). Data are shown as mean±SEM of n=3 replicate wells per condition. IC50 values were calculated using four-parameter non-linear regression. Replicate experiments were carried out on human lung fibroblasts (HLFs) from a non-IPF donor and representative curves are shown. Images from each cell treatment are shown (C). 8 mm slices of IPF lung were cultured for 24 h in DMEM supplemented with 0.4% FCS and treated with vehicle, 10 μM SB525334 (open circles) or increasing concentrations of Compound 1 (inhibitor) for 0–3 days (filled grey circles) and further from 3 to 5 days (filled black circles) following media change. P1NP levels were measured in harvested supernatants (D). Data are shown as ±SEM of n=4 replicate wells per condition; *p<0.05, **p<0.01 (two-way ANOVA with Bonferroni post-hoc testing).
All figures (8)
Figure 7
Figure 7
Time course of the primary human fibroblast response to transforming growth factor (TGF)-β1. Confluent lung fibroblasts (LFs) were stimulated with TGFβ1 (1 ng/mL) and lysates collected over the indicated timecourse (A, B and D). Phosphorylation of SMAD2 or AktS473 is shown by Western blot (A) and densitometry (B and D). Collagen gene expression in TGFβ1 (1 ng/mL) stimulated (C, dark triangles and dashed line) or unstimulated (C, light triangles and dashed line) was assessed in LFs over the indicated time course. Pro-collagen recovered from cell supernatants over the same time course in TGFβ1 stimulated (C, dark bar) or unstimulated (C, light bar) is also shown. Serum-free confluent primary human LFs (HLFs) were incubated with increasing concentrations of GSK2126458 or 0.1% DMSO and stimulated with TGFβ1 (1 ng/mL) for 12 h (0.1% DMSO vehicle was constant for all experimental conditions). Phosphorylation of SMAD2 and AktS473 is shown by representative Western blot (E). Densitometry illustrating the effects of GSK2126458 on pAktS473 in HLFs from n=3 donors is shown (F).
Figure 8
Figure 8
Pan-PI3 kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibition attenuates transforming growth factor (TGF)-β1-induced collagen production in human lung fibroblasts (LFs), and collagen formation markers in idiopathic pulmonary fibrosis (IPF) lung tissue. Confluent LFs were incubated with increasing concentrations of GSK2126458 (A) or SB525334 (B) and stimulated for 48 h with TGFβ1 (1 ng/mL) and collagen biosynthesis assayed by molecular crowding assay (0.1% DMSO vehicle was constant for all experimental conditions). Data are expressed as mean fluorescent intensity (n=4 reads per well) and cell counts obtained from DAPI counterstaining (blue channel). Data are shown as mean±SEM of n=3 replicate wells per condition. IC50 values were calculated using four-parameter non-linear regression. Replicate experiments were carried out on human lung fibroblasts (HLFs) from a non-IPF donor and representative curves are shown. Images from each cell treatment are shown (C). 8 mm slices of IPF lung were cultured for 24 h in DMEM supplemented with 0.4% FCS and treated with vehicle, 10 μM SB525334 (open circles) or increasing concentrations of Compound 1 (inhibitor) for 0–3 days (filled grey circles) and further from 3 to 5 days (filled black circles) following media change. P1NP levels were measured in harvested supernatants (D). Data are shown as ±SEM of n=4 replicate wells per condition; *p<0.05, **p<0.01 (two-way ANOVA with Bonferroni post-hoc testing).

References

    1. Lee AS, Mira-Avendano I, Ryu JH, et al. . The burden of idiopathic pulmonary fibrosis: an unmet public health need. Respir Med 2014;108:955–67. 10.1016/j.rmed.2014.03.015
    1. Richeldi L, du Bois RM, Raghu G, et al. . Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med 2014;370:2071–82. 10.1056/NEJMoa1402584
    1. King TE Jr, Bradford WZ, Castro-Bernardini S, et al. . A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med 2014;370:2083–92. 10.1056/NEJMoa1402582
    1. Datta A, Scotton CJ, Chambers RC. Novel therapeutic approaches for pulmonary fibrosis. Br J Pharmacol 2011;163:141–72. 10.1111/j.1476-5381.2011.01247.x
    1. Akhurst RJ, Hata A. Targeting the TGFβ signalling pathway in disease. Nat Rev Drug Discov 2012;11:790–811. 10.1038/nrd3810
    1. Horan GS, Wood S, Ona V. , et al. Partial inhibition of integrin alpha(v)beta6 prevents pulmonary fibrosis without exacerbating inflammation. Am J Respir Crit Care Med 2008;177:56–65. 10.1164/rccm.200706-805OC
    1. Parker MW, Rossi D, Peterson M, et al. . Fibrotic extracellular matrix activates a profibrotic positive feedback loop. J Clin Invest 2014;124:1622–35. 10.1172/JCI71386
    1. Daniels CE, Jett JR. Does interstitial lung disease predispose to lung cancer? Curr Opin Pulm Med 2005;11:431–7. 10.1097/
    1. Rabinovich EI, Kapetanaki MG, Steinfeld I, et al. . Global methylation patterns in idiopathic pulmonary fibrosis. PLoS ONE 2012;7:e33770 10.1371/journal.pone.0033770
    1. Selman M, Pardo A. Revealing the pathogenic and aging-related mechanisms of the enigmatic idiopathic pulmonary fibrosis. an integral model. Am J Respir Crit Care Med 2014;189:1161–72. 10.1164/rccm.201312-2221PP
    1. Vancheri C. Common pathways in idiopathic pulmonary fibrosis and cancer. Eur Respir Rev 2013;22:265–72. 10.1183/09059180.00003613
    1. Groves AM, Win T, Screaton NJ. , et al. Idiopathic pulmonary fibrosis and diffuse parenchymal lung disease: implications from initial experience with 18F-FDG PET/CT. J Nucl Med 2009;50:538–45. 10.2967/jnumed.108.057901
    1. Vanhaesebroeck B, Guillermet-Guibert J, Graupera M. , et al. The emerging mechanisms of isoform-specific PI3K signalling. Nat Rev Mol Cell Biol 2010;11:329–41. 10.1038/nrm2882
    1. Conte E, Gili E, Fruciano M. , et al. PI3K p110γ overexpression in idiopathic pulmonary fibrosis lung tissue and fibroblast cells: in vitro effects of its inhibition. Lab Invest 2013;93:566–76. 10.1038/labinvest.2013.6
    1. Horowitz JC, Lee DY, Waghray M. , et al. Activation of the pro-survival phosphatidylinositol 3-kinase/AKT pathway by transforming growth factor-beta1 in mesenchymal cells is mediated by p38 MAPK-dependent induction of an autocrine growth factor. J Biol Chem 2004;279:1359–67. 10.1074/jbc.M306248200
    1. Kavanaugh WM, Klippel A, Escobedo JA. , et al. Modification of the 85-kilodalton subunit of phosphatidylinositol-3 kinase in platelet-derived growth factor-stimulated cells. Mol Cell Biol 1992;12:3415–24. 10.1128/MCB.12.8.3415
    1. White ES, Atrasz RG, Hu B. , et al. Negative regulation of myofibroblast differentiation by PTEN (Phosphatase and Tensin Homolog Deleted on chromosome 10). Am J Respir Crit Care Med 2006;173:112–21. 10.1164/rccm.200507-1058OC
    1. Xia H, Diebold D, Nho R. , et al. Pathological integrin signaling enhances proliferation of primary lung fibroblasts from patients with idiopathic pulmonary fibrosis. J Exp Med 2008;205:1659–72. 10.1084/jem.20080001
    1. Conte E, Fruciano M, Fagone E, 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:e24663 10.1371/journal.pone.0024663
    1. Le Cras TD, Korfhagen TR, Davidson C. , et al. Inhibition of PI3K by PX-866 prevents transforming growth factor-alpha-induced pulmonary fibrosis. Am J Pathol 2010;176:679–86. 10.2353/ajpath.2010.090123
    1. Knight SD, Adams ND, Burgess JL. , et al. Discovery of GSK2126458, a Highly Potent Inhibitor of PI3K and the Mammalian Target of Rapamycin. ACS Med Chem Lett 2010;1:39–43. 10.1021/ml900028r
    1. Raghu G, Collard HR, Egan JJ. , et al. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med 2011;183:788–824. 10.1164/rccm.2009-040GL
    1. Keerthisingam CB, Jenkins RG, Harrison NK. , et al. Cyclooxygenase-2 deficiency results in a loss of the anti-proliferative response to transforming growth factor-beta in human fibrotic lung fibroblasts and promotes bleomycin-induced pulmonary fibrosis in mice. Am J Pathol 2001;158:1411–22. 10.1016/S0002-9440(10)64092-8
    1. Crowley C, Klanrit P, Butler CR. , et al. Surface modification of a POSS-nanocomposite material to enhance cellular integration of a synthetic bioscaffold. Biomaterials 2016;83:283–93. 10.1016/j.biomaterials.2016.01.005
    1. Leeming DJ, Larsen DV, Zhang C. , et al. Enzyme-linked immunosorbent serum assays (ELISAs) for rat and human N-terminal pro-peptide of collagen type I (PINP)—assessment of corresponding epitopes. Clin Biochem 2010;43:1249–56. 10.1016/j.clinbiochem.2010.07.025
    1. Chambers RC, Laurent GJ, Westergren-Thorsson G. Cadmium inhibits proteoglycan and procollagen production by cultured human lung fibroblasts. Am J Respir Cell Mol Biol 1998;19:498–506. 10.1165/ajrcmb.19.3.3242
    1. Chen CZ, Peng YX, Wang ZB, et al. . The Scar-in-a-Jar: studying potential antifibrotic compounds from the epigenetic to extracellular level in a single well. Br J Pharmacol 2009;158:1196–209. 10.1111/j.1476-5381.2009.00387.x
    1. Hong M, Wilkes MC, Penheiter SG. , et al. Non-Smad transforming growth factor-β signaling regulated by focal adhesion kinase binding the p85 subunit of phosphatidylinositol 3-kinase. J Biol Chem 2011;286:17841–50. 10.1074/jbc.M111.233676
    1. Bozulic L, Hemmings BA. PIKKing on PKB: regulation of PKB activity by phosphorylation. Curr Opin Cell Biol 2009;21:256–61. 10.1016/j.ceb.2009.02.002
    1. Islam MR, Jones SJ, Macluskey M, et al. . Is there a pAkt between VEGF and oral cancer cell migration? Cell Signal 2014;26:1294–302. 10.1016/j.cellsig.2014.02.004
    1. Vincent EE, Elder DJ, Thomas EC. , et al. Akt phosphorylation on Thr308 but not on Ser473 correlates with Akt protein kinase activity in human non-small cell lung cancer. Br J Cancer 2011;104:1755–61. 10.1038/bjc.2011.132
    1. Foukas LC, Berenjeno IM, Gray A. , et al. Activity of any class IA PI3K isoform can sustain cell proliferation and survival. Proc Natl Acad Sci USA 2010;107:11381–6. 10.1073/pnas.0906461107
    1. Fruman DA, Rommel C. PI3K and cancer: lessons, challenges and opportunities. Nat Rev Drug Discov 2014;13:140–56. 10.1038/nrd4204
    1. Gibbons MA, MacKinnon AC, Ramachandran P. , et al. Ly6Chi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. Am J Respir Crit Care Med 2011;184:569–81. 10.1164/rccm.201010-1719OC
    1. Murray LA, Rosada R, Moreira AP, et al. . Serum amyloid P therapeutically attenuates murine bleomycin-induced pulmonary fibrosis via its effects on macrophages. PLoS ONE 2010;5:e9683 10.1371/journal.pone.0009683
    1. Prasse A, Pechkovsky DV, Toews GB. , et al. A vicious circle of alveolar macrophages and fibroblasts perpetuates pulmonary fibrosis via CCL18. Am J Respir Crit Care Med 2006;173:781–92. 10.1164/rccm.200509-1518OC
    1. Stahl M, Schupp J, Jäger B, et al. . Lung collagens perpetuate pulmonary fibrosis via CD204 and M2 macrophage activation. PLoS ONE 2013;8:e81382 10.1371/journal.pone.0081382
    1. Marone R, Cmiljanovic V, Giese B. , et al. Targeting phosphoinositide 3-kinase: moving towards therapy. Biochim Biophys Acta 2008;1784:159–85. 10.1016/j.bbapap.2007.10.003
    1. Anderton MJ, Mellor HR, Bell A. , et al. Induction of heart valve lesions by small-molecule ALK5 inhibitors. Toxicol Pathol 2011;39:916–24. 10.1177/0192623311416259

Source: PubMed

3
订阅