Introduction
Asthma is a chronic inflammatory disease of the lungs and airways that is marked by exaggerated airway-constrictive responses against diverse provocative challenges (airway hyperresponsiveness, AHR
3The abbreviations used are:
AHR
airway hyperresponsiveness
T
HT helper
IL
interleukin
SH
Src homology
EGF
epidermal growth factor
STAT
signal transducer and activator of transcription
R
RSrespiratory system resistance
i.p.
intraperitoneal(ly)
i.n.
intranasal(ly)
IFN
interferon
Ova
ovalbumin
ppm
parts per million
TSLP
thymic stromal lymphopoietin
POM
pivaloyloxymethyl
FAK
focal adhesion kinase
BALF
bronchoalveolar lavage fluid
CBC
complete blood count
FBS
fetal bovine serum
MTT
methylthiazol tetrazolium.
); accumulation of mucus and fibrin plugs in the airway that further compromise airflow (plastic bronchitis); airway and systemic eosinophilia; T helper type 2 (T
H2) cells and innate lymphoid cells type 2 (ILC2) that secrete the cytokines interleukin-4 (IL-4), IL-5, and IL-13; and vigorous IgE antibody responses (
1Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma.
,
2- Iwasaki A.
- Foxman E.F.
- Molony R.D.
Early local immune defences in the respiratory tract.
,
3Type 2 inflammation in asthma: present in most, absent in many.
). Affecting up to 30 million Americans, with 3 million experiencing severe therapy-resistant disease, asthma is one of the most common and burdensome of all chronic afflictions (
4- McDonald V.M.
- Maltby S.
- Reddel H.K.
- King G.G.
- Wark P.A.
- Smith L.
- Upham J.W.
- James A.L.
- Marks G.B.
- Gibson P.G.
Severe asthma: current management, targeted therapies and future directions: a roundtable report.
,
5- Peters M.C.
- Nguyen M.L.
- Dunican E.M.
Biomarkers of airway type-2 inflammation and integrating complex phenotypes to endotypes in asthma.
).
Current asthma therapy provides symptomatic relief for many patients, but the inability of these agents to cure disease ensures their long-term use and attendant risk of inducing serious, even life-threatening side effects. We reasoned that a safer and more effective means of treating asthma could be developed through an improved understanding of the critical immune effector pathways that drive disease expression. The development of asthma is complex and is initiated in part by airway epithelial cells that recognize exogenous antigen or pathogens through pattern recognition receptors. Airway epithelial cells then secrete the inflammatory cytokines IL-25, IL-33, and TSLP (
1Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma.
,
2- Iwasaki A.
- Foxman E.F.
- Molony R.D.
Early local immune defences in the respiratory tract.
) to promote ILC2 recruitment and enhance dendritic cell luminal sampling, migration to lymph nodes, and antigen presentation to initiate B and T cell activation (
1Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma.
,
6- Hoffmann F.
- Ender F.
- Schmudde I.
- Lewkowich I.P.
- Köhl J.
- König P.
- Laumonnier Y.
Origin, localization, and immunoregulatory properties of pulmonary phagocytes in allergic asthma.
,
7Type 2 cytokines: mechanisms and therapeutic strategies.
,
8Regulation of group 2 innate lymphoid cells.
).
Adaptive immunity is critical for maintaining the chronic inflammation associated with asthma that is mediated by T
H2 and T
H17 cells that migrate to the lungs (
3Type 2 inflammation in asthma: present in most, absent in many.
). When in the lung, T
H2 cells work cooperatively with ILC2s to coordinate production of the cytokines IL-4 and IL-13 (
8Regulation of group 2 innate lymphoid cells.
). These cytokines signal through distinct multimeric receptor complexes that share the common IL-4Rα subunit (
9- Bhattacharjee A.
- Shukla M.
- Yakubenko V.P.
- Mulya A.
- Kundu S.
- Cathcart M.K.
IL-4 and IL-13 employ discrete signaling pathways for target gene expression in alternatively activated monocytes/macrophages.
). Cytokine binding to IL-4Rα induces recruitment of the tyrosine kinases Jak1 and Jak3 (Jak1 and Tyk2 for IL-13) that then phosphorylate Tyr
631 within the Tyr-Lys-Pro-Phe docking site for the latent cytoplasmic transcription factor signal transducer and activator of transcription 6 (STAT6) (
9- Bhattacharjee A.
- Shukla M.
- Yakubenko V.P.
- Mulya A.
- Kundu S.
- Cathcart M.K.
IL-4 and IL-13 employ discrete signaling pathways for target gene expression in alternatively activated monocytes/macrophages.
). Upon binding to this motif via its Src homology 2 (SH2) domain, STAT6 is phosphorylated by Jak kinases, becoming competent to homodimerize, translocate to the nucleus, and promote STAT6-dependent gene expression (
). The absence of IL-4/IL-13/STAT6 responses in STAT6
−/− mice results in complete disease resistance in murine models of asthma, highlighting the central role of STAT6 in the development of asthma (
11- Knight J.M.
- Mak G.
- Shaw J.
- Porter P.
- McDermott C.
- Roberts L.
- You R.
- Yuan X.
- Millien V.O.
- Qian Y.
- Song L.Z.
- Frazier V.
- Kim C.
- Kim J.J.
- Bond R.A.
- et al.
Long-acting β agonists enhance allergic airway disease.
,
12- Miyata S.
- Matsuyama T.
- Kodama T.
- Nishioka Y.
- Kuribayashi K.
- Takeda K.
- Akira S.
- Sugita M.
STAT6 deficiency in a mouse model of allergen-induced airways inflammation abolishes eosinophilia but induces infiltration of CD8+ T cells.
).
In addition to STAT6, STAT5 also contributes to the development of allergic airway disease. STAT5 together with STAT6 is required for optimal T
H2 development (
13Transcriptional regulation of Th2 cell differentiation.
,
14- Knosp C.A.
- Carroll H.P.
- Elliott J.
- Saunders S.P.
- Nel H.J.
- Amu S.
- Pratt J.C.
- Spence S.
- Doran E.
- Cooke N.
- Jackson R.
- Swift J.
- Fitzgerald D.C.
- Heaney L.G.
- Fallon P.G.
- et al.
SOCS2 regulates T helper type 2 differentiation and the generation of type 2 allergic responses.
,
15- Yang X.O.
- Zhang H.
- Kim B.S.
- Niu X.
- Peng J.
- Chen Y.
- Kerketta R.
- Lee Y.H.
- Chang S.H.
- Corry D.B.
- Wang D.
- Watowich S.S.
- Dong C.
The signaling suppressor CIS controls proallergic T cell development and allergic airway inflammation.
). STAT5 is also essential for ILC2 development and is the major transcription factor activated by IL-5, which promotes eosinophil development, recruitment, and survival (
5- Peters M.C.
- Nguyen M.L.
- Dunican E.M.
Biomarkers of airway type-2 inflammation and integrating complex phenotypes to endotypes in asthma.
,
16- Kabata H.
- Moro K.
- Koyasu S.
- Asano K.
Group 2 innate lymphoid cells and asthma.
). STAT5 and STAT6 are both activated by TSLP (
17- Arima K.
- Watanabe N.
- Hanabuchi S.
- Chang M.
- Sun S.-C.
- Liu Y.-J.
Distinct signal codes generate dendritic cell functional plasticity.
,
18- Wu J.
- Dong F.
- Wang R.A.
- Wang J.
- Zhao J.
- Yang M.
- Gong W.
- Cui R.
- Dong L.
Central role of cellular senescence in TSLP-induced airway remodeling in asthma.
), underscoring the fact that multiple diverse ligands drive the activation of a limited number of STAT factors that are crucial to the expression of asthma. Thus, although targeting select cytokines is likely to be beneficial in asthma (
19- Corren J.
- Lemanske R.F.
- Hanania N.A.
- Korenblat P.E.
- Parsey M.V.
- Arron J.R.
- Harris J.M.
- Scheerens H.
- Wu L.C.
- Su Z.
- Mosesova S.
- Eisner M.D.
- Bohen S.P.
- Matthews J.G.
Lebrikizumab treatment in adults with asthma.
,
20- Wenzel S.
- Ford L.
- Pearlman D.
- Spector S.
- Sher L.
- Skobieranda F.
- Wang L.
- Kirkesseli S.
- Rocklin R.
- Bock B.
- Hamilton J.
- Ming J.E.
- Radin A.
- Stahl N.
- Yancopoulos G.D.
- et al.
Dupilumab in persistent asthma with elevated eosinophil levels.
), a more effective approach is likely to be targeting the key STAT factors that represent the final common allergic disease mediators (
21- Tan H.T.
- Sugita K.
- Akdis C.A.
Novel biologicals for the treatment of allergic diseases and asthma.
).
Our laboratories have focused on the design of small-molecule phosphopeptidomimetic inhibitors of the SH2 domain of STAT6 that prevent recruitment to the critical docking site on IL-4Rα and phosphorylation of Tyr
641. Early-generation compounds were shown to be potent ligands of STAT6
in vitro, blocking STAT6-dependent gene expression in a variety of human airway epithelial cells, breast cancer cells, and primary splenocytes (
22- Morlacchi P.
- Mandal P.K.
- McMurray J.S.
Synthesis and in vitro evaluation of a peptidomimetic inhibitor targeting the Src homology 2 (SH2) domain of STAT6.
,
23- Mandal P.K.
- Morlacchi P.
- Knight J.M.
- Link T.M.
- Lee 4th, G.R.
- Nurieva R.
- Singh D.
- Dhanik A.
- Kavraki L.
- Corry D.B.
- Ladbury J.E.
- McMurray J.S.
Targeting the Src homology 2 (SH2) domain of signal transducer and activator of transcription 6 (STAT6) with cell-permeable, phosphatase-stable phosphopeptide mimics potently inhibits Tyr641 phosphorylation and transcriptional activity.
). Furthermore, the cell-permeable, phosphatase-stable prodrug PM-242H was effective at preventing and/or reversing allergic lung disease in a murine model of asthma (
11- Knight J.M.
- Mak G.
- Shaw J.
- Porter P.
- McDermott C.
- Roberts L.
- You R.
- Yuan X.
- Millien V.O.
- Qian Y.
- Song L.Z.
- Frazier V.
- Kim C.
- Kim J.J.
- Bond R.A.
- et al.
Long-acting β agonists enhance allergic airway disease.
). Here we expand the scope of our initial
in vitro structure–activity relationship studies to include novel central and C-terminal analogs.
Discussion
Based on seminal work conducted in the mouse (
33- Grünig G.
- Warnock M.
- Wakil A.E.
- Venkayya R.
- Brombacher F.
- Rennick D.M.
- Sheppard D.
- Mohrs M.
- Donaldson D.D.
- Locksley R.M.
- Corry D.B.
Requirement for IL-13 independently of IL-4 in experimental asthma.
,
34- Wills-Karp M.
- Luyimbazi J.
- Xu X.
- Schofield B.
- Neben T.Y.
- Karp C.L.
- Donaldson D.D.
Interleukin-13: central mediator of allergic asthma.
) and verified through human clinical trials (
19- Corren J.
- Lemanske R.F.
- Hanania N.A.
- Korenblat P.E.
- Parsey M.V.
- Arron J.R.
- Harris J.M.
- Scheerens H.
- Wu L.C.
- Su Z.
- Mosesova S.
- Eisner M.D.
- Bohen S.P.
- Matthews J.G.
Lebrikizumab treatment in adults with asthma.
,
20- Wenzel S.
- Ford L.
- Pearlman D.
- Spector S.
- Sher L.
- Skobieranda F.
- Wang L.
- Kirkesseli S.
- Rocklin R.
- Bock B.
- Hamilton J.
- Ming J.E.
- Radin A.
- Stahl N.
- Yancopoulos G.D.
- et al.
Dupilumab in persistent asthma with elevated eosinophil levels.
), the IL-4/IL-13-IL-4Rα-STAT5/6 signaling pathway is now recognized as critical for determining the expression of asthma and related allergic diseases. However, because of redundant and alternative pathways for activation of STAT6 (
e.g. through TSLP (
17- Arima K.
- Watanabe N.
- Hanabuchi S.
- Chang M.
- Sun S.-C.
- Liu Y.-J.
Distinct signal codes generate dendritic cell functional plasticity.
), long-acting beta agonists (
11- Knight J.M.
- Mak G.
- Shaw J.
- Porter P.
- McDermott C.
- Roberts L.
- You R.
- Yuan X.
- Millien V.O.
- Qian Y.
- Song L.Z.
- Frazier V.
- Kim C.
- Kim J.J.
- Bond R.A.
- et al.
Long-acting β agonists enhance allergic airway disease.
), and IL-4 (
35- Hou J.
- Schindler U.
- Henzel W.J.
- Ho T.C.
- Brasseur M.
- McKnight S.L.
An interleukin-4-induced transcription factor: IL-4 Stat.
)), therapeutic agents that target the extracellular limb of this pathway (including cytokines and receptors) will likely fail to completely suppress STAT6 activation. Additional redundant cytokine pathways support STAT5 activation, underscoring this concern (
36- Isaksen D.E.
- Baumann H.
- Trobridge P.A.
- Farr A.G.
- Levin S.D.
- Ziegler S.F.
Requirement for stat5 in thymic stromal lymphopoietin-mediated signal transduction.
). Thus, existing and future asthma therapeutic agents that target STAT5/6 ligands and receptors are destined to be incompletely effective in blocking unwanted allergic responses. Moreover, the agents that have been developed in this regard are all monoclonal antibodies, which are prohibitively expensive, especially for patients with mild to moderate disease. Additionally, monoclonal antibodies must be delivered systemically, increasing the chances of inducing systemic immune suppression and neutralizing antibody responses to these complex proteins.
Mimetic peptides represent a considerably less expensive alternative drug strategy that has proven effective at targeting extracellular and intracellular proteins (
37- Ahmed C.M.
- Larkin 3rd, J.
- Johnson
- Johnson H.M.
SOCS1 mimetics and antagonists: a complementary approach to positive and negative regulation of immune function.
,
38- Delbridge A.R.
- Strasser A.
The BCL-2 protein family, BH3-mimetics and cancer therapy.
,
39- McMurray J.S.
- Mandal P.K.
- Liao W.S.
- Klostergaard J.
- Robertson F.M.
The consequences of selective inhibition of signal transducer and activator of transcription 3 (STAT3) tyrosine705 phosphorylation by phosphopeptide mimetic prodrugs targeting the Src homology 2 (SH2) domain.
,
40- Uehara Y.
- Chiesa G.
- Saku K.
High-density lipoprotein-targeted therapy and apolipoprotein A-I mimetic peptides.
). The high specificity of monoclonal antibodies can be achieved through the intelligent design of mimetic peptides around known protein–protein interactions. Several groups have embraced this strategy and developed large peptides (10 amino acids) to modulate BH3-mediated apoptosis, suppressor of cytokine signaling modulation of inflammation, and high-density lipoprotein–driven cardiovascular disease (
37- Ahmed C.M.
- Larkin 3rd, J.
- Johnson
- Johnson H.M.
SOCS1 mimetics and antagonists: a complementary approach to positive and negative regulation of immune function.
,
38- Delbridge A.R.
- Strasser A.
The BCL-2 protein family, BH3-mimetics and cancer therapy.
,
40- Uehara Y.
- Chiesa G.
- Saku K.
High-density lipoprotein-targeted therapy and apolipoprotein A-I mimetic peptides.
). We have previously reported our work developing small peptidomimetics (4-mers) targeting the SH2-domain of STAT3 and the suppression of tumor growth (
24- Mandal P.K.
- Liao W.S.
- McMurray J.S.
Synthesis of phosphatase-stable, cell-permeable peptidomimetic prodrugs that target the SH2 domain of Stat3.
,
26- Mandal P.K.
- Gao F.
- Lu Z.
- Ren Z.
- Ramesh R.
- Birtwistle J.S.
- Kaluarachchi K.K.
- Chen X.
- Bast Jr, R.C.
- Liao W.S.
- McMurray J.S.
Potent and selective phosphopeptide mimetic prodrugs targeted to the Src homology 2 (SH2) domain of signal transducer and activator of transcription 3.
,
39- McMurray J.S.
- Mandal P.K.
- Liao W.S.
- Klostergaard J.
- Robertson F.M.
The consequences of selective inhibition of signal transducer and activator of transcription 3 (STAT3) tyrosine705 phosphorylation by phosphopeptide mimetic prodrugs targeting the Src homology 2 (SH2) domain.
). Here we report the development of a novel phosphopeptidomimetic small molecule that is based on the cytoplasmic docking site on IL-4Rα for STAT6, PM-43I.
Targeting the SH2 domain of STAT6, PM-43I is also a potent inhibitor of STAT5. As STAT5/6 require activation through their SH2 domains in all contexts and share a significant degree of sequence similarity, PM-43I is theoretically capable of completely inactivating STAT5 and STAT6 regardless of the activating stimulus. Indeed, the highly effective nature of PM-43I in our experimental asthma model suggests that we are achieving inhibition of most of the latent STAT5/6 within mouse airway cells at the observed therapeutic doses. As a small molecule, PM-43I is relatively easily synthesized in large quantities and to high purity. Because of the low dose required (0.25–25 μg/kg) and the need for only topical delivery to the airway to be highly effective, PM-43I should compare economically favorably to mAb-based asthma therapies. Moreover, PM-43I appears in our preliminary studies to be well tolerated and highly effective. Together, our findings indicate that PM-43I is suitable for entry into clinical development for asthma and related afflictions.
Phosphopeptidomimetics represent a new class of pharmacologic agents that have been developed successfully to target the SH2 domains of a variety of signaling proteins (
33- Grünig G.
- Warnock M.
- Wakil A.E.
- Venkayya R.
- Brombacher F.
- Rennick D.M.
- Sheppard D.
- Mohrs M.
- Donaldson D.D.
- Locksley R.M.
- Corry D.B.
Requirement for IL-13 independently of IL-4 in experimental asthma.
,
26- Mandal P.K.
- Gao F.
- Lu Z.
- Ren Z.
- Ramesh R.
- Birtwistle J.S.
- Kaluarachchi K.K.
- Chen X.
- Bast Jr, R.C.
- Liao W.S.
- McMurray J.S.
Potent and selective phosphopeptide mimetic prodrugs targeted to the Src homology 2 (SH2) domain of signal transducer and activator of transcription 3.
,
39- McMurray J.S.
- Mandal P.K.
- Liao W.S.
- Klostergaard J.
- Robertson F.M.
The consequences of selective inhibition of signal transducer and activator of transcription 3 (STAT3) tyrosine705 phosphorylation by phosphopeptide mimetic prodrugs targeting the Src homology 2 (SH2) domain.
) but have not previously been evaluated for the blockade of multiple STAT factors or for the treatment of asthma. Previously, we developed the phosphopeptidomimetic prodrug PM-242H to target STAT6 selectively and demonstrated that this novel agent was effective at both preventing and reversing asthma-like allergic airway disease in mice (
11- Knight J.M.
- Mak G.
- Shaw J.
- Porter P.
- McDermott C.
- Roberts L.
- You R.
- Yuan X.
- Millien V.O.
- Qian Y.
- Song L.Z.
- Frazier V.
- Kim C.
- Kim J.J.
- Bond R.A.
- et al.
Long-acting β agonists enhance allergic airway disease.
). Although PM-242H proved the validity of this therapeutic strategy, this agent has an unfavorably high minimum effective dose of 2.5 mg/kg. The iterative redesign of PM-242H as presented here was undertaken largely to improve compound potency but also to determine the efficacy of more broadly cross-reactive STAT inhibitors.
Our
in vitro observations were critically important in informing our choice of compounds selected for subsequent
in vivo testing. Specifically, based on
in vitro testing, we selected PM-43I as one compound that inhibited STAT5 and STAT6 and PM-86I as a compound that was highly selective for inhibiting STAT6. Although PM-43I inhibited STAT6 less effectively than PM-86I
in vitro, PM-43I performed significantly better
in vivo regarding inhibition of multiple parameters of allergic airway disease (
Fig. 2). We presume that PM-43I was more effective because of its ability to inhibit STAT5 in addition to STAT6. The superior
in vivo response may be due to the inhibition of the Stat5-dependent, ILC2-driven innate immune responses that promote Stat6-depandant T
H2 adaptive immunity, both of which contribute to the expression of this disease phenotype (
12- Miyata S.
- Matsuyama T.
- Kodama T.
- Nishioka Y.
- Kuribayashi K.
- Takeda K.
- Akira S.
- Sugita M.
STAT6 deficiency in a mouse model of allergen-induced airways inflammation abolishes eosinophilia but induces infiltration of CD8+ T cells.
,
13Transcriptional regulation of Th2 cell differentiation.
,
14- Knosp C.A.
- Carroll H.P.
- Elliott J.
- Saunders S.P.
- Nel H.J.
- Amu S.
- Pratt J.C.
- Spence S.
- Doran E.
- Cooke N.
- Jackson R.
- Swift J.
- Fitzgerald D.C.
- Heaney L.G.
- Fallon P.G.
- et al.
SOCS2 regulates T helper type 2 differentiation and the generation of type 2 allergic responses.
,
15- Yang X.O.
- Zhang H.
- Kim B.S.
- Niu X.
- Peng J.
- Chen Y.
- Kerketta R.
- Lee Y.H.
- Chang S.H.
- Corry D.B.
- Wang D.
- Watowich S.S.
- Dong C.
The signaling suppressor CIS controls proallergic T cell development and allergic airway inflammation.
,
16- Kabata H.
- Moro K.
- Koyasu S.
- Asano K.
Group 2 innate lymphoid cells and asthma.
,
17- Arima K.
- Watanabe N.
- Hanabuchi S.
- Chang M.
- Sun S.-C.
- Liu Y.-J.
Distinct signal codes generate dendritic cell functional plasticity.
,
18- Wu J.
- Dong F.
- Wang R.A.
- Wang J.
- Zhao J.
- Yang M.
- Gong W.
- Cui R.
- Dong L.
Central role of cellular senescence in TSLP-induced airway remodeling in asthma.
,
41- Gavino A.C.
- Nahmod K.
- Bharadwaj U.
- Makedonas G.
- Tweardy D.J.
STAT3 inhibition prevents lung inflammation, remodeling, and accumulation of Th2 and Th17 cells in a murine asthma model.
,
42- Simeone-Penney M.C.
- Severgnini M.
- Tu P.
- Homer R.J.
- Mariani T.J.
- Cohn L.
- Simon A.R.
Airway epithelial STAT3 is required for allergic inflammation in a murine model of asthma.
,
43Interleukin-13 in asthma pathogenesis.
,
44- Hao Y.
- Kuang Z.
- Jing J.
- Miao J.
- Mei L.Y.
- Lee R.J.
- Kim S.
- Choe S.
- Krause D.C.
- Lau G.W.
Mycoplasma pneumoniae modulates STAT3-STAT6/EGFR-FOXA2 signaling to induce overexpression of airway mucins.
). Another compound, PM-63I, also inhibited STAT5 and was at least 10 times more effective than PM-43I at inhibiting STAT6 (
Fig. 1,
a–c). However, we did not pursue
in vivo studies with this compound because of the enhanced cellular toxicity exhibited against two cell lines at the lower tested doses (
Fig. 1,
d and
e). Nonetheless, the
in vivo potency of PM-43I, which potentially extends to the entire family of molecules we have synthesized, suggests that such
in vitro toxicity might be irrelevant
in vivo. Therefore, further studies with PM-63I using the same mouse model to determine the therapeutic potential of this compound may be warranted.
The chronic, incurable nature of asthma mandates that approved treatments and pharmaceuticals be selected so that they minimize long-term side effects. However, standard asthma care includes the use of pharmaceutical agents with relatively poor long-term safety records. The original long-acting beta agonists approved for use in the United States, formoterol and salmeterol, lead to loss of disease control and excess asthma-related mortality in a small subset of patients that is in part, but not completely (
45Regular treatment with formoterol for chronic asthma: serious adverse events.
), alleviated by the co-formulation with inhaled corticosteroids, as now mandated by the Food and Drug Administration. However, inhaled and especially oral corticosteroids have their own notorious, long-term safety concerns that include laryngitis, diabetes, weight gain, hypertension, cataracts, etc. Serious fungal infectious complications of chronic corticosteroid use (
46Intranasal fungal (Alternaria) infection related to nasal steroid spray.
,
47- Peter E.
- Bakri F.
- Ball D.M.
- Cheney R.T.
- Segal B.H.
Invasive pulmonary filamentous fungal infection in a patient receiving inhaled corticosteroid therapy.
,
48- Leav B.A.
- Fanburg B.
- Hadley S.
Invasive pulmonary aspergillosis associated with high-dose inhaled fluticasone.
,
49- Fairfax A.J.
- David V.
- Douce G.
Laryngeal aspergillosis following high dose inhaled fluticasone therapy for asthma.
) are particularly concerning because asthma and related conditions, such as chronic rhinosinusitis, are increasingly recognized as manifestations of airway mycosis (airway fungal infection) (
30- Porter P.C.
- Lim D.J.
- Maskatia Z.K.
- Mak G.
- Tsai C.L.
- Citardi M.J.
- Fakhri S.
- Shaw J.L.
- Fothergil A.
- Kheradmand F.
- Corry D.B.
- Luong A.
Airway surface mycosis in chronic TH2-associated airway disease.
). A primary benefit of agents such as PM-43I that are highly selective in their inhibition of immune pathways is that they are less likely to compromise protective immunity compared with broad-spectrum immunosuppressants like corticosteroids. Indeed, our preliminary toxicity studies suggest that PM-43I did not impact airway antifungal or antiviral immunity even when given in excessive doses (
Fig. 5h and
Fig. S2).
An important component of the drug delivery strategy presented here is the direct delivery of prodrug to the lungs and deposition on the target cells of the airway, where it is then converted, extracellularly and intracellularly, to active drug by removal of the POM groups by reactive oxygen species, ions, and/or esterases. Delivery of the drug by aerosol bypasses the caustic gastric and first-pass metabolism to allow immediate cellular absorption and intracellular activation of PM-43I in the lungs, where the drug persists for more than 24 h (
Fig. 6). It is important to note that extracellular activation, by removal of the protective POM groups, abrogates membrane permeability and systemic drug activity (
Fig. 2,
h and
i), allowing efficient renal clearance of nonabsorbed drug within hours of airway delivery (
Fig. 6). We cannot be certain which airway cells primarily take up PM-43I but presume that airway epithelial and smooth muscle cells are main PM-43I targets, given their proximity to the airway lumen. Other airway cells are likely to be targeted also, and the more they are affected by PM-43I, the stronger the suppressive effect on allergic disease expression should be.
The efficient renal clearance of non-POM drug is a critical observation and supports the apparent lack of toxicity observed in the 8-month toxicity study (
Fig. 7). The potential toxicity of the inactive POM group byproducts, formaldehyde and pivalic acid, could be a concerning factor, but it is well known that both formaldehyde and pivalic acid are readily metabolized and excreted at the therapeutic range of PM-43I (
50Pivalate-generating prodrugs and carnitine homeostasis in man.
,
51- Jia Y.Y.
- Lu C.T.
- Feng J.
- Song Y.
- Zhao J.Y.
- Wang S.
- Sun Y.
- Wen A.D.
- Yang Z.F.
- Hang T.J.
Impact on L-carnitine homeostasis of short-term treatment with the pivalate prodrug tenofovir dipivoxil.
,
52- Mitkus R.J.
- Hess M.A.
- Schwartz S.L.
Pharmacokinetic modeling as an approach to assessing the safety of residual formaldehyde in infant vaccines.
,
53Committee to Review the Formaldehyde Assessment in the National Toxicology Program 12th Report on Carcinogens, Board on Environmental Studies and Toxicology, Division on Earth and Life Sciences, National Research Council
Review of the Formaldehyde Assessment in the National Toxicology Program 12th Report on Carcinogens: 2: Review of the Formaldehyde Profile in the National Toxicology Program 12th Report on Carcinogens.
,
54- Patnaik A.
- Rowinsky E.K.
- Villalona M.A.
- Hammond L.A.
- Britten C.D.
- Siu L.L.
- Goetz A.
- Felton S.A.
- Burton S.
- Valone F.H.
- Eckhardt S.G.
A phase I study of pivaloyloxymethyl butyrate, a prodrug of the differentiating agent butyric acid, in patients with advanced solid malignancies.
). Additionally, numerous drugs containing POM groups (adefovir, pivampicillin, cefditoren, valproate, etc.) are now Food and Drug Administration–approved and regularly used in the clinic.
Numerous drugs targeting immune molecules have been evaluated in asthma, many of which have proven successful (e.g. drugs targeting IL-5 and IgE), but not all. PM-43I offers several distinct advantages over all of these agents. First, as a small, easily synthesized molecule, it avoids the complexities and expense of biological agents such as monoclonal antibodies. Second, although biologic agents that individually target STAT6-activating cytokines have been developed, none individually blocks STAT6 completely and none block both STAT5 and STAT6 as does PM-43I. PM-43I is therefore predicted to be both more effective and less expensive than existing agents.
The direct delivery by aerosolization and activation of PM-43I in the lungs is an additional novel aspect of this drug that minimizes possible systemic toxic effects that are unavoidable with parenterally administered biologics. Assuming immediate and complete POM group breakdown and prodrug molar equivalence for the observed PM-43I therapeutic range (0.25–25 μg/kg), a 15-min aerosol treatment would produce a formaldehyde concentration (0.006–0.6 ppm) below the Occupational Safety and Health Administration (OSHA)–defined 15-min permissible exposure limit of 2 ppm and the 8-h permissible exposure limit of 0.75 ppm (
53Committee to Review the Formaldehyde Assessment in the National Toxicology Program 12th Report on Carcinogens, Board on Environmental Studies and Toxicology, Division on Earth and Life Sciences, National Research Council
Review of the Formaldehyde Assessment in the National Toxicology Program 12th Report on Carcinogens: 2: Review of the Formaldehyde Profile in the National Toxicology Program 12th Report on Carcinogens.
). Furthermore, the Environmental Protection Agency estimated average indoor formaldehyde concentration of 0.03 ppm is well above the optimal PM-43I therapeutic dose (2.5 μg/kg, 0.006 ppm). Together, these data suggest that concerns regarding PM-43I–related toxicities related to the POM groups are negligible.
Although PM-43I demonstrated dose-dependent efficacy
in vivo, the inverse
in vivo relationship between dose and efficacy is unusual; the drug begins to lose efficacy at doses higher than the maximally effective dose of 0.25 μg/kg. This issue highlights the complexities of targeting the intracellular compartment where many distinct potential targets exist, including more than 140 SH2 domain–containing proteins that are potentially cross-reactive targets. Although PM-43I did not cross-react at highly effective pharmacologic doses with other STAT proteins of considerable sequence homology (
Fig. S1a) and other critical immune signaling proteins, such as AKT and FAK (
Fig. 1C), potential cross-reactivity with the SH2 domains of suppressors of cytokine signaling 1 and 3 (SOCS1 and SOCS3) could block their ability to inhibit STAT6 activity, in effect extending the activity of STAT6 that was not inactivated by drug (
55- Endo T.A.
- Masuhara M.
- Yokouchi M.
- Suzuki R.
- Sakamoto H.
- Mitsui K.
- Matsumoto A.
- Tanimura S.
- Ohtsubo M.
- Misawa H.
- Miyazaki T.
- Leonor N.
- Taniguchi T.
- Fujita T.
- Kanakura Y.
- et al.
A new protein containing an SH2 domain that inhibits JAK kinases.
,
56- Helman D.
- Sandowski Y.
- Cohen Y.
- Matsumoto A.
- Yoshimura A.
- Merchav S.
- Gertler A.
Cytokine-inducible SH2 protein (CIS3) and JAK2 binding protein (JAB) abolish prolactin receptor-mediated STAT5 signaling.
,
57- Losman J.A.
- Chen X.P.
- Hilton D.
- Rothman P.
Cutting edge: SOCS-1 is a potent inhibitor of IL-4 signal transduction.
). Similarly, protein tyrosine phosphatase nonreceptor type 6 (PTPN6, SHP-1) is an SH2 domain–containing phosphatase that dephosphorylates active STAT factors, especially STAT5 and STAT6 (
58- Haque S.J.
- Harbor P.
- Tabrizi M.
- Yi T.
- Williams B.R.
Protein-tyrosine phosphatase Shp-1 is a negative regulator of IL-4- and IL-13-dependent signal transduction.
,
59Interaction of growth hormone-activated STATs with SH2-containing phosphotyrosine phosphatase SHP-1 and nuclear JAK2 tyrosine kinase.
). Inhibition of this protein via its SH2 domain with PM-43I could potentially produce paradoxically enhanced STAT5/6 activity at supratherapeutic doses of PM-43I. If additional testing suggests that PM-43I possesses an excessively narrow therapeutic window related to such potential cross-reactivity, then PM-63I could be explored further as a therapeutic alternative, or PM-43I could be further chemically modified to reduce such cross-reactivity while preserving specificity for STAT5/6.
Concluding remarks
We have developed a potent inhibitor of STAT5 and STAT6 that can be stably delivered to the lungs and inhibit the induction of or reverse pre-existing allergic lung disease in a murine model of allergic lung disease. The lead compound does not appear to impair immunity or present any apparent toxicity but can skew circulating humoral immunity toward a less dominant TH2 profile. Our studies confirm the feasibility to target the SH2 domain therapeutically and indicate that PM-43I is suitable for entry into clinical development.
Experimental procedures
Drug/inhibitor
1,2-dilauroyl-sn-glycero-3-phosphocholine (DLPC, 770335) was acquired from Avanti Polar Lipids (Alabaster, AL) and used as a vehicle at a 1:5 (drug:DLPC) ratio. In brief, drug and DLPC were solubilized in t-butyl alcohol at the identified mass ratio, frozen, and lyophilized. Individual treatments were suspended in PBS and sonicated for 30 s before use.
Fluorescence polarization assays
Serial dilutions of peptides (200 μm) in fluorescence polarization buffer (50 mm NaCl, 10 mm HEPES, 1 mm Na4EDTA, 2 mm DTT, and 1% NP-40) were prepared. Aliquots of STAT6 (480 nm) and FAM-Ala-pTyr-Lys-Pro-Phe-Gln-Asp-Leu-Ile-NH2 (20 nm) were plated in a 96-well plate, and peptide dilutions were added. Fluorescence polarization was then read on a Tecan Infinite F200Pro plate reader, and IC50 values were obtained from the nonlinear regression analysis.
Aerosol characterization
Drug (PM-43I) or DLPC was aerosolized using an Aerotech II nebulizer with a flow rate of 10 liters/min. Aerosols were captured using an all-glass impinger under a vacuum. Aerosol particle size was determined with an Andersen cascade impactor. Captured drug was quantified by HPLC-MS and used to calculate the drug dose for animal studies as reported previously (
60Antiviral therapy with small particle aerosols.
,
61Ribavirin aerosol treatment of influenza.
,
62Inhalation Studies: Foundations and Techniques.
,
63Comparative deposition of inhaled aerosols in experimental animals and humans: a review.
).
Cell culture
A549, BEAS-2B, and MD468 cells were acquired from the American Type Culture Collection (Manassas, VA). Cells were maintained in 50% Dulbecco's modified Eagle's medium and 50% F-12 complete medium until sufficient confluency and switched to 2% FBS 24 h prior to stimulation. Cells were pretreated up to 2 h with identified drugs at various concentrations, stimulated with IL-4 (2 ng/ml, R&D Systems), EGF (100 ng/ml, R&D Systems), or IFN-γ (25 ng/ml, R&D Systems) for identified times and harvested for protein analysis.
Western blotting
Total protein was harvested from cells with radioimmune precipitation assay buffer (9806S, Cell Signaling Technology, Danvers, MA), quantified with BCA protein assay reagent (23227, Thermo Fisher Scientific, Rockford, IL), and denatured with Laemmli sample buffer (161-0737, Bio-Rad) according to the manufacturers' protocols. Proteins were analyzed by separation in hand-poured SDS-PAGE gels with standard electrophoretic units (Bio-Rad) and transferred to polyvinylidene difluoride membranes with an iBlot gel transfer device (IB1001, Invitrogen). After blocking with 2% FBS PBS + 0.05% Tween 20, membranes were probed for pSTAT6, STAT6, pSTAT5, STAT5, pSTAT3, STAT3, pAKT, AKT, pFAK, FAK, pSTAT1, STAT1, and β-actin. Protein signals were detected using the ChemiDoc XRS+ system (Bio-Rad).
Mice
4- to 8-week-old female BALB/c, C57BL/6, and CR1 mice were purchased from The Jackson Laboratory (Bar Harbor, ME) and housed in the American Association for Accreditation of Laboratory Animal Care–accredited Transgenic Mouse Facility at Baylor College of Medicine under specific pathogen–free conditions. All experimental protocols were approved by the Institutional Animal Care and Use Committee of Baylor College of Medicine and followed federal guidelines.
Infectious allergic lung disease/reversal models
Mice were challenged intranasally every other day with 4 × 10
5 A. niger conidia for a minimum of six challenges and treated with the identified STAT6 inhibitors at identified doses and/or vehicle (DLPC) as described (infectious allergic lung disease model,
Fig. 3a; reversal model,
Fig. 7a) for each experiment. Following the final challenge, mice rested for a day, and allergic airway disease was assessed.
Immunosuppression model
Mice were concurrently sensitized to Ova-alum i.p. and immunosuppressed with 5 μg of PM-43I, PM-86I, or vehicle control (DLPC), as detailed in
Fig. 4. Briefly, mice were vaccinated with alum-bound ovalbumin once a week for 2 weeks and PM-43I (5 μg), PM-86I (5 μg), or DLPC (25 μg) i.p. (systemic immunosuppression) or i.n. (local immunosuppression). Splenocytes were harvested and assessed for Ova-specific responses.
Allergic airway disease analysis
Allergic airway disease was assessed in accordance with previous reports (
28- Porter P.
- Susarla S.C.
- Polikepahad S.
- Qian Y.
- Hampton J.
- Kiss A.
- Vaidya S.
- Sur S.
- Ongeri V.
- Yang T.
- Delclos G.L.
- Abramson S.
- Kheradmand F.
- Corry D.B.
Link between allergic asthma and airway mucosal infection suggested by proteinase-secreting household fungi.
). Increases in R
RS because of intravenous acetylcholine, bronchoalveolar lavage (BAL) fluid differential counts, and lung IL-4–, IL-17A–, and IFN–γ secreting cells were quantified as described previously (
25- Lee S.H.
- Prince J.E.
- Rais M.
- Kheradmand F.
- Ballantyne C.M.
- Weitz-Schmidt G.
- Smith C.W.
- Corry D.B.
Developmental control of integrin expression regulates Th2 effector homing.
,
28- Porter P.
- Susarla S.C.
- Polikepahad S.
- Qian Y.
- Hampton J.
- Kiss A.
- Vaidya S.
- Sur S.
- Ongeri V.
- Yang T.
- Delclos G.L.
- Abramson S.
- Kheradmand F.
- Corry D.B.
Link between allergic asthma and airway mucosal infection suggested by proteinase-secreting household fungi.
).
Ovalbumin restimulation
Splenocytes from Ova-sensitized mice were assessed for antigen-specific recall by ELISpot for relevant cytokines. In brief, cells were cultured in the presence of medium or whole ovalbumin (1 mg/ml) overnight, and plates were developed for IL-4, INF-γ, and IL-17 as described previously.
Histology
Lungs were perfused with 4% paraformaldehyde and embedded in paraffin. Lung sections were cut and stained for mucus with the periodic acid-Schiff kit (395B) from Sigma-Aldrich (St. Louis, MO) or cells with hematoxylin and eosin. Lung sections were immunohistologically stained for p STAT6 and pSTAT5.
Fungal burden
Fungal burden in fungus-challenged mice was determined with serial dilutions on Sabouraud agar plates (84088, Sigma-Aldrich) of lung homogenates in the presence of 100 mg/ml chloramphenicol (C0378, Sigma-Aldrich). After overnight incubation at 37 °C, plates were assessed for fungal colony-forming units per lung from dilution.
Influenza infection
Mice were placed on PM-43I (1.5 μg) or DLPC aerosol therapy prior to challenge with an LD
30 of aerosolized influenza (H3N2), as detailed in
Fig. 6a. Changes in weight and survival were monitored throughout the experiment.
Toxicity
Mice were sensitized to Ova-alum i.p. (once per week twice) and treated with PM-43I (5 μg) or DLPC for 8 months, as described in
Fig. 1a. In brief, sensitized mice were treated intranasally every other day with ovalbumin plus PM-43I or DLPC and monitored for changes in weight. Airway hyperreactivity and Ova-specific antibodies were assessed as described. Blood and spleens were harvested at the end of the experiment for further analysis. Blood samples were submitted to the pathology core at the University of Texas MD Anderson Cancer Center for complete blood counts and blood chemistry. Detailed cellular analysis of splenocytes was performed by the flow cytometry core at Baylor College of Medicine.
Flow cytometric analysis
Spleens were disassociated using an Octo dissociator (Miltenyi Biotech) in RPMI 1640 containing (Gibco) 10% heat-inactivated FBS (HyClone), 2 mm EDTA (Gibco), 0.2 mg/ml DNase I (Sigma), and 1 mg/ml collagenase II (Gibco). The suspension was incubated at room temperature for 20 min. Digestion was stopped by addition of EDTA to a 12 mm concentration. Cells were filtered through a 40-μm mesh. Cells were collected, and red blood cells were lysed in RBC lysis buffer (eBioscience) for 5 min at room temperature. Cells were washed and resuspended in flow buffer (DPBS without Ca2+/Mg2+ (Gibco) supplemented with 2% fetal calf serum, 25 mm HEPES, and 2 mm EDTA). A portion of the cells was used for enumeration and trypan blue viability assessment using a ViCell (Beckman Coulter). Fc receptors were blocked on ice for 10 min using flow buffer containing a 1:250 dilution of anti-mouse CD16/32 (BD Biosciences). Cells were incubated on ice for 30 min with one of two panels of antibodies, panel 1 or panel 2. Panel 1 included anti-mouse CD5, CD4, CD44, CD8, CD25, CD161, and CD62L (BD Biosciences). Panel 2 included anti-mouse CD5, Ly6G CD19, Ly6C, CD21/35, CD11b, CD11c, CD161, CD23 (BD Biosciences), and anti-mouse major histocompatibility complex II (Biolegend). Both groups were washed and resuspended in flow buffer with 1 drop/ml Nuc Blue Fixed DAPI Stain (Life Technologies). Single-stained DAPI control and fluorescence minus one controls (FMOs) were prepared from the remaining pooled spleen cells. Cells were run on a LSRII flow cytometer (BD Biosciences) and analyzed using FlowJo (Tree Star) according to immunophenotyping gating strategies outlined by the International Mouse Phenotyping Consortium.
Ova-specific ELISA
Serum samples were serially diluted and incubated in 96-well plates coated with ovalbumin. Captured antibodies were detected with biotinylated anti-IgE and anti-IgG2a and developed with streptavidin–alkaline phosphatase. Substrate was added, and the signal was detected by optical density at 405 nm.
Pharmacokinetic analysis
Mice were treated intranasally with 5 μg of PM-43I, and tissue samples were harvested at identified time points. Drug was quantified in tissue samples by HPLC-MS.
Analytical method
Tissue samples were homogenized in sample buffer (50% acetonitrile, 50% 0.1% formic acid). Serum and captured aerosol samples were mixed with acetonitrile at a 1:1 ratio. All samples and standards were separated on a C18-column (Phenomenex) and detected by MS-MS (Quattro Premier XE).
Author contributions
J. M. K., P. Mandal, D. B. C., and J. S. M. conceptualization; J. M. K. and J. S. M. data curation; J. M. K., P. Morlacchi, G. M., E. L., M. M., C. L., B. S., E. F., B. G., J. S., D. B. C., and J. S. M. formal analysis; J. M. K., P. Mandal, D. B. C., and J. S. M. supervision; J. M. K., P. Mandal, P. Morlacchi, G. M., E. L., M. M., C. L., D. B. C., and J. S. M. investigation; J. M. K., P. Morlacchi, G. M., M. M., C. L., J. S., D. B. C., and J. S. M. methodology; J. M. K., D. B. C., and J. S. M. writing-original draft; J. M. K., P. Mandal, P. Morlacchi, G. M., E. L., M. M., C. L., B. S., E. F., B. G., J. S., A. V., M. M. S., D. C., D. B. C., and J. S. M. writing-review and editing; P. Mandal, A. V., M. M. S., D. C., D. B. C., and J. S. M. resources; P. Mandal, D. B. C., and J. S. M. validation; A. V., M. M. S., D. C., D. B. C., and J. S. M. funding acquisition; A. V., M. M. S., and D. C. project administration; D. B. C. and J. S. M. visualization.
Article info
Publication history
Published online: May 08, 2018
Received in revised form:
April 2,
2018
Received:
October 22,
2017
Edited by Peter Cresswell
Footnotes
This article contains Figs. S1–S4.
This study was funded by National Institutes of Health Grants R41AI25007 and R01 HL117181, Veterans Affairs Office of Research and Development Grants I01BX002221 and I01 CX001673, the American Asthma Foundation, and the Biology of Inflammation Center, Baylor College of Medicine. J. M. K., P. Mandal, P. Morlacchi, D. B. C., and Atrapos Therapeutics, LLC hold intellectual property rights in compound PM-43I. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
We dedicate this manuscript to the memory of John S. McMurray, Ph.D. who died on March 28, 2017.