Preventive Azithromycin Treatment Reduces Noninfectious Lung ...

4 downloads 0 Views 2MB Size Report
1 Department of Internal Medicine, Louisiana State University Health Sciences Center .... the Jackson Laboratory (Bar Harbor, ME) or Charles River Laboratories.
Biol Blood Marrow Transplant 21 (2015) 30e38

Biology of Blood and Marrow Transplantation journal homepage: www.bbmt.org

Biology

Preventive Azithromycin Treatment Reduces Noninfectious Lung Injury and Acute Graft-versus-Host Disease in a Murine Model of Allogeneic Hematopoietic Cell Transplantation Sabarinath Venniyil Radhakrishnan 1, 2, Senthilnathan Palaniyandi 2, 3, Gunnar Mueller 4, Sandra Miklos 4, Max Hager 4, Elena Spacenko 4, Fridrik J. Karlsson 3, Elisabeth Huber 5, Nicolai A. Kittan 3, Gerhard C. Hildebrandt 2, 3, * 1

Department of Internal Medicine, Louisiana State University Health Sciences Center, Shreveport, Shreveport, Louisiana Division of Hematology and Hematologic Malignancies, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah 3 Division of Hematology and Oncology, Louisiana State University Health Sciences Center, Shreveport, Feist-Weiller Cancer Center, Shreveport, Louisiana 4 Division of Hematology and Oncology, University of Regensburg, Regensburg, Germany 5 Department of Pathology, University of Regensburg, Regensburg, Germany 2

Article history: Received 30 July 2014 Accepted 26 September 2014 Key Words: Acute graft-versus-host disease Azithromycin Noninfectious lung injury Regulatory T cell

a b s t r a c t Noninfectious lung injury and acute graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (allo-HCT) are associated with significant morbidity and mortality. Azithromycin is widely used in allogeneic HCT recipients for pulmonary chronic GVHD, although current data appear controversial. We induced GVHD and noninfectious lung injury in lethally irradiated B6D2F1 mice by transplanting bone marrow and splenic T cells from allogeneic C57BL/6 mice. Experimental groups were treated with oral azithromycin starting on day 14 until the end of week 6 or week 14 after transplantation. Azithromycin treatment resulted in improved survival and decreased lung injury; the latter characterized by improved pulmonary function, reduced peribronchial and perivascular inflammatory cell infiltrates along with diminished collagen deposition, and a decrease in lung cytokine and chemokine expression. Azithromycin also improved intestinal GVHD but did not affect liver GVHD at week 6 early after transplantation. At week 14, azithromycin decreased liver GVHD but had no effect on intestinal GVHD. In vitro, allogeneic antigenpresenting cell (APC)e dependent T cell proliferation and cytokine production were suppressed by azithromycin and inversely correlated with relative regulatory T cell (Treg) expansion, whereas no effect was seen when T cell proliferation occurred APC independently through CD3/CD28-stimulation. Further, azithromycin reduced alloreactive T cell expansion but increased Treg expansion in vivo with corresponding downregulation of MHC II on CD11cþ dendritic cells. These results demonstrate that preventive administration of azithromycin can reduce the severity of acute GVHD and noninfectious lung injury after allo-HCT, supporting further investigation in clinical trials. Ó 2015 American Society for Blood and Marrow Transplantation.

INTRODUCTION Noninfectious lung injury and acute graft-versus-host disease (GVHD) are major causes of morbidity and mortality after allogeneic hematopoietic cell transplantation (allo-HCT). In 2011, the American Thoracic Society issued a consensus statement on noninfectious lung injury

Financial disclosure: See Acknowledgments on page 37. * Correspondence and reprint requests: Gerhard C. Hildebrandt, Division of Hematology and Hematologic Malignancies, University of Utah School of Medicine, Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, UT 84112-5550. E-mail address: [email protected] (G.C. Hildebrandt).

http://dx.doi.org/10.1016/j.bbmt.2014.09.025 1083-8791/Ó 2015 American Society for Blood and Marrow Transplantation.

summarized under the term Idiopathic Pneumonia Syndrome, encompassing various clinical forms, which differ based on the time of onset, course of disease, and treatment response [1]. Various models of rodents have been established to study noninfectious lung injury after allo-HCT, and 2 major histopathologic features similar to that seen in patients receiving allo-HCT are commonly observed across models: peribronchial and perivascular lymphocytic infiltration and acute pneumonitis of the alveolar spaces and the interstitium [1,2]. Although the exact pathophysiology is not completely understood, donor CD4þ T cells activated by antigen presenting cells (APCs) [3-5] and shifting towards Th1 or Th17 phenotype [6,7], cytotoxic CD8þ T cells [8], and macrophages [5]

S.V. Radhakrishnan et al. / Biol Blood Marrow Transplant 21 (2015) 30e38

contribute to the tissue injury. Cytokines and chemokines such as TNF-a, IL-1b, IFN-g, and CCL2, CCL5, CXCL9-11, and CXCL1 [9-11], respectively may also be contributive to the disease process. Acute GVHD itself, sharing similar pathophysiological characteristics, may be a predisposing and driving factor of disease development. Azithromycin is a macrolide antibiotic with established anti-inflammatory properties and favorable toxicity profile, which attains very high and durable lung concentrations [12]. Azithromycin treatment has been found to decrease pulmonary exacerbations and improve lung function in patients with cystic fibrosis (CF) [13,14], chronic obstructive pulmonary disease, and non-CF bronchiectasis [15,16]. Azithromycin has also been shown to be beneficial in lung transplantation for the prevention and treatment of chronic allograft rejection [17,18]. Accordingly, azithromycin has been studied in the setting of allo-HCT to treat bronchiolitis obliterans syndrome (BOS), but studies were limited by small patient numbers and demonstrated conflicting evidence of therapeutic efficacy [19,20]. In none of these studies, azithromycin was given prophylactically with the intention to prevent lung injury. In this study, we use a well-established mouse model of noninfectious lung injury after allo-HCT to determine whether azithromycin started early and given over a prolonged time period is beneficial in preventing pulmonary damage, thereby improving pulmonary function and overall outcome. MATERIALS AND METHODS Animal experiments were done at 3 different institutions: early GVHD and pulmonary function testing experiments were done at the University of Regensburg Medical Center (UKR), Regensburg, Germany, and the splenic T cell expansion study was done at the University of Utah (UofU), Salt Lake City, Utah. All other data presented are from experiments performed at Louisiana State University Health Sciences Center Shreveport (LSUHSC-S), Louisiana. All experiments were done with the approval of the local institutional animal committees. Induction of GVHD and lung Injury Female B6D2F1 (H-2bxd) and C57BL/6 (H-2b) mice were purchased from the Jackson Laboratory (Bar Harbor, ME) or Charles River Laboratories (Sulzbach, Germany) and acclimatized in the respective animal facility for at least 1 week before starting the experiments. Animals were between 10 and 20 weeks old at the time of HCT or when used for in vitro studies. Mice underwent bone marrow transplantation according to standard protocol as previously described [9,21]. B6D2F1 mice were conditioned using total body irradiation. Irradiation was done using either a linear accelerator, 150 cGy/minute (UKR) or using a Cesium source irradiator (LSUHSC-S and UofU), at a total dose of 12 Gy. Radiation was delivered in 2 fractions, 3 hours apart to reduce gastrointestinal toxicity. Animals then received a cell mixture of 4 to 5  106 bone marrow cells and 6  106 splenocytes from either syngeneic B6D2F1 or allogeneic C57BL/6 donors via tail vein injection. Mice were housed in sterilized microisolator cages and received autoclaved (UKR) or normal (LSUHSC-S and UofU) chow and autoclaved water for the first 2 weeks. At day 14 after transplantation, syngeneic and allogeneic recipients were equally stratified based on weight loss into 2 subgroups: 1 group received autoclaved water only whereas the other group received autoclaved water supplemented with azithromycin dihydrate (Pfizer, Germany) at a concentration of 1 mg/mL from day þ14 until day þ17 and .5 mg/mL from day þ18 until end of week þ14 (UKR). At LSUHSC-S and UofU, azithromycin (Sagent Pharmaceuticals, Schaumburg, IL) dosing was increased to 3 mg/mL to attain a dose of 450 to 600 mg/kg/day to optimize study set up [22]. Survival was monitored daily and GVHD clinical scores were assessed weekly using 5 clinical parameters: weight loss, posture, mobility, fur texture, and skin integrity, as previously described [2]. Animals were sacrificed for analysis at end of week þ6 and þ14. Histopathology Lung injury was assessed by examination of lung histopathology. Hematoxylin and eosinestained lung sections were independently evaluated

31

by the pathologist for the severity of periluminal infiltrates (around airways and vessels) or parenchymal pneumonitis (involving the alveoli or interstitium), using a previously described semiquantitative scoring system [2]. Final scores for periluminal and parenchymal infiltrates were obtained by multiplication of the respective severity and extent. Liver and gut pathology were also assessed by hematoxylin and eosin staining of the tissues and independently evaluated by the pathologist, using previously described scoring systems [23,24]. Trichrome staining for collagen (NovaUltra Masson Trichrome Stain kit, IW-3006, IHC World, Woodstock, MD) was performed as per manufacturer’s protocol. For immunostaining, 4 mm of organ slices were dewaxed and rehydrated, followed by antigen unmasking using citrate buffer pH 7.2 and microwave at 300W for 30 minutes. Next, peroxidase blocking solution (S2023, Dako) was added for 5 minutes to quench endogenous peroxidase activity. CD3þ cells were then detected by rabbit IgG anti-mouse CD3 antibody (NeoMarkers, RM-9107-S [Thermo Fisher Scientific GMBH, Dreieich, Germany]) in 1:50 dilution in antibody diluent (S2022, Dako). Simple Stain Mouse MAX PO anti rabbit (414141F, Histofine, NICHIREI BIOSCIENCES INC, Tokyo, Japan) was used as the secondary antibody, visualization using DAB (K3467, Dako) and counterstained using hematoxylin. CD3þ staining was scored as cells per high power field. Bronchoalveolar Lavage Bronchoalveolar lavage (BAL) and cell surface phenotyping were performed as previously described and cells were analyzed by flow cytometry [2]. Pulmonary Function Testing Assessment of pulmonary function was performed using a Buxco lung function analysis system (Buxco Electronics, Troy, NY) consisting of a pulmonary function test/forced maneuvers analyzer (SFT3840, Buxco) plus pressure panel for mouse maneuvers (AUT6100, Buxco), and an anesthetized mouseepulmonary function test (PFT) plethysmograph (PLY3112, Buxco) as previously described [25]. Data acquisition and analysis were done using BioSystem XA software (SFT3850). Tissue Cytokine and Chemokine Expression At time of analysis, tissues were harvested and snap frozen. Supernatant of homogenized tissue was obtained as described before [26]. Total protein concentration in the supernatant was determined using Coomassie Bradford protein assay (Thermo Scientific, Rockford, IL) to allow for cytokine and chemokine concentration normalization to picograms per milligram of total protein. Cytokines and chemokines, such as TNF-a, IL-1b, CXCL1, CXCL9, CXCL10, IL-15, CCL5, CCL3, and others were assayed by Millipore’s MILLIPLEX MAG Mouse Cytokine/Chemokine kit (Billerica, MA) using Luminex xMAP technology according to manufacturer’s protocol. Cytokine/chemokine analysis was also performed on tissues using ELISA for CXCL9, IFN-g,TNF-a, CCL2, CXCL1, MIP-2, IL-6, and CXCL10 (Duoset, R&D Systems GmbH, Germany). T cell Activation and Proliferation Assays Mixed lymphocytic reaction (MLR) was done using irradiated (40 Gy) BALB/c or B6D2F1splenocytes (8  105 cells per well) as stimulators. C57BL/ 6 effector T cells were purified from splenocytes using MACS CD90$2 beads and QuadroMACS system using LS columns (Miltenyi Biotec, Auburn, CA) according to manufacturer’s protocol and cocultured in complete RPMI medium in 2:1 stimulator to effector ratio at 4  105 cells per well in a 96well flat bottom plate. CD3/CD28 T cell activation assay was performed using C57BL/6 splenocytes at a concentration of 2  105 cells per well in complete RPMI using antiCD3ε (Clone 145-2C11) and anti-CD28 (Clone 37$51, eBioscience, San Jose, CA) antibody stimulation as per manufacturer’s protocol. Cultures were incubated at 37 C and 5% CO2, pulsed at 72 hours with 3H thymidine (1 mCi) for MLR and at 48 hours for CD3/CD28 T cell activation assay and harvested 24 hours later using PHD cell harvester (Brandel, MD). Proliferation was quantified by using Wallac 1409 liquid scintillation counter. Azithromycin was added to respective groups at the concentrations 5 mg/mL, 10 mg/mL, and 20 mg/mL versus controls for the duration of the assay. Average counts per minute were obtained from triplicates, and experiments were done in duplicates. Supernatants were obtained before the addition of 3H thymidine and stored at 80 C for cytokine analysis. MLR and CD3/CD28 T cell activation assay supernatant were analyzed for IFN-g, TNF-a, IL-10, IL-17, IL-2, IL-4, and IL-6 cytokine levels using Cytometric Bead Array Mouse Th1/Th2/Th17 Cytokine Kit (BD Biosciences Pharmingen, San Diego, CA) per manufacturer’s protocol. T cell Migration Assay Migration assay was performed using murine recombinant CXCL9 (DevaTal Inc., Trenton, NJ) as chemotactic stimulant at 100 nM/well using a 24-well transwell migration plate. Purified T cells at 5  105 cells per well

32

S.V. Radhakrishnan et al. / Biol Blood Marrow Transplant 21 (2015) 30e38

Figure 1. Survival at week þ6 and week þ14 after allo-HCT. Animals underwent transplantation as described in Materials and Methods. (A) Survival at week þ6 after HCT. Azithromycin was used at 1 mg/mL for the first 3 days starting on day þ14 and then at .5 mg/mL until day 42. (syngeneic control: n ¼ 5; allogeneic control: n ¼ 10; azithromycin-treated allogeneic: n ¼ 8) Survival by Wilcoxon test. (B) Clinical GVHD score at week þ6 after HCT as described in Materials and Methods. (C) Survival at week þ14 after HCT (syngeneic control: n ¼ 8; allogeneic control: n ¼ 17; azithromycin-treated allogeneic: n ¼ 15). Azithromycin was used at 1 mg/mL for the first 3 days starting on day þ14 and then at .5 mg/mL until day þ98. (D) Survival at week þ14 (syngeneic control: n ¼ 5; syngeneic azithromycin-treated group: n ¼ 4; allogeneic control: n ¼ 13; azithromycin-treated allogeneic group: n ¼ 14; survival by Wilcoxon test). Azithromycin was used at 3 mg/mL starting on day þ14 until day þ100.

and complete RPMI supplemented with azithromycin (dissolved in PBS) at concentrations of 20 mg/mL and 50 mg/mL versus PBS control were used as previously described [10].

Flow Cytometry Cells from BAL or MLR were phenotyped for CD3þ, CD4þ, CD8þ, and Foxp3 expression. Cells from BAL or MLR were washed with PBS containing 4% FBS and incubated with FcR-block antimouse CD16/CD32 (Clone:93, eBioscience, San Diego, CA) for 15 minutes at 4 C. Following a washing step, cells were incubated in a predetermined optimized concentration of antibodies in a total volume of 100 mL. For intracellular staining for FoxP3, fixation and permeabilization were done as per protocol using the fixation/ permeabilization concentrate and diluent (eBioscience). The following antibody clones were used: CD3-FITC (17A2), CD4-APC (RM4-5), CD8aPerCP (53-6.7) CD11c (N418) MHCII (M5/114.15.2), CD80 (16-10A1), CD86 (GL-1) (all obtained from Biolegend, San Diego, CA) and FoxP3 PE (FJK-16s) (eBioscience). CD8-FITC, Ly6c-FITC and CD4-PE (BD Pharmingen) were used for the experiments at UKR. Cells were analyzed on BD LSR II, BD FACS Canto or BD FACSCalibur (BD Biosciences). Flow cytometry data were analyzed using FlowJo software (Tree Star, Ashland, OR).

Splenic T cell Proliferation Transplantation was performed as described above. In the day þ7 experiments, the treatment group received azithromycin at 3 mg/mL for 2 weeks before and continued for 1 week after transplantation until analysis at day þ7. For the day þ56 experiment, azithromycin (3 mg/mL) was started at day þ14 after transplantation and continued until day þ56, the day of analysis. Total spleen count was calculated and CD4þ, CD8þ T cells, and FoxP3þ CD4þ T cells were quantified by using flow cytometry as described above (% positive cells  total spleen count). CD11c was used to identify dendritic cells and the expression of MHC class II; CD86 and CD80 were quantified using median fluorescence intensity.

Statistical Considerations All values are expressed as mean  standard error of mean (SEM). Statistical comparisons between groups were made using either students T test or 1-way ANOVA with Bonferroni comparison between groups, and the Wilcoxon rank test was used for analyzing survival data. Significance was defined as P < .05.

RESULTS Acute GVHD and Lung Injury are Reduced in Azithromycin-treated Animals after Allogeneic HCT Animals underwent transplantation and were treated with azithromycin (1 mg/mL) versus control starting at day þ14, as described above, and analyzed at day þ42. All syngeneic and azithromycin-treated allogeneic recipients survived, but only 50% of allogeneic controls were alive at time of analysis (P ¼ .02) (Figure 1A). This survival benefit was associated with less clinical GVHD (Figure 1B). We next assessed the effects of azithromycin treatment at a later time point (14 weeks after transplantation) to determine whether prolonged azithromycin is efficacious to prevent disease progression. All syngeneic animals, 29% of allogeneic control animals, and 53% of allogeneic azithromycin-treated animals were alive for the duration of the experiment (Figure 1C). In spite of significant changes in pulmonary function (discussed below) yet only mild improvement in survival, we next treated animals with azithromycin at a higher concentration of 3 mg/mL to optimize drug efficacy. Survival at week þ14 was significantly higher in the azithromycin allogeneic group compared with the control group (85.7% versus 53.8%; P ¼ .04) (Figure 1D) and all syngeneic recipients (control and treated group) survived. This experiment was performed at a different institution, with irradiation given from a different source, and animals kept in different facility, as described in Material and Methods, possibly explaining the difference in overall survival of allogeneic controls compared with previous experiments. At week þ6 after HCT, BAL fluid cellularity showed a significant increase in CD4þ and CD8þ T cells in allogeneic recipients compared with syngeneic recipients without a statistically significant difference between allogeneic azithromycin-treated animals and allogeneic controls.

S.V. Radhakrishnan et al. / Biol Blood Marrow Transplant 21 (2015) 30e38

Neutrophils were higher in the allogeneic groups but did not statistically differ from syngeneic controls (Figure 2AeC); in addition, pulmonary function tests showed no significant differences between syngeneic and allogeneic groups (Figure 2DeF). As reported in prior studies using this murine allo-HCT model, histopathologic changes indicative of noninfectious lung injury [25] were evident in the lungs of allogeneic control-treated animals and significantly reduced in allogeneic recipients receiving azithromycin (Figure 2G). This was associated with less gut GVHD in the azithromycintreated animals (Figure 2H) when compared with allogeneic controls, whereas no difference in liver pathology was found (Figure 2I). At week þ14 after HCT, BAL fluid cellularity revealed a significant decrease in CD4þ T cells in allogeneic azithromycin recipients compared with allogeneic controls, as well as a not statistically significant decrease in CD8þ T cells (Figure 2J,K). Lung function was assessed in surviving animals. Forced expiratory volume 50, lung compliance, and forced vital capacity were significantly better in the azithromycin-treated allogeneic group compared with the

33

allogeneic controls and similar to the syngeneic group (Figure 2LeN). Lung injury was predominantly periluminal with no difference in the parenchymal injury between groups. The periluminal lung injury, consisting largely of CD3þ T cells and collagen deposition, was decreased in the azithromycin-treated allogeneic group compared with allogeneic controls (Figure 2OeR). Along with a reduction in lung injury, there was a significant reduction in hepatic GVHD changes with a decrease in lymphocytic infiltration of the bile ducts and diminished portal duct expansion secondary to inflammation. There was no difference in gut histopathology observed between groups (Figure 2S,T). In line with the observed effects on histopathology at week þ6 in the lung, increased levels of CXCL1, IFN-g, and CXCL9 were observed in allogeneic controls compared with the syngeneic animals, and their expression was decreased in allogeneic recipients treated with azithromycin (Figure 3AeD). A significant difference in inflammatory cytokine and chemokine levels in the gut between the syngeneic group and the allogeneic controls was observed for CCL2, IFN-g, CXCL9, and TNF-a. Azithromycin-treated

Figure 2. Histopathology and pulmonary function at week þ6 and week þ14 after HCT. Animals underwent transplantation as described in Materials and Methods (A-C) Bronchoalveolar lavage (BAL) cell counts, CD4þ T cells, CD8þ T cells, and neutrophils by flow cytometry at week þ6 after HCT. (D-F) Pulmonary function test at week þ6 after HCT: FEV50, FVC, and compliance. (G-I) Pathology of the lung, gut, and liver by H&E staining at week þ6 after HCT using scoring systems as described in Materials and Methods. (syngeneic control: n ¼ 5; allogeneic control: n ¼ 5; azithromycin-treated allogeneic: n ¼ 8). (J,K) BAL cell counts CD4þ T cells and CD8þ T cells by flow cytometry at week þ14 after HCT. (L-N) Pulmonary function test at week þ14 after HCT: FEV50, FVC, and compliance. (O) H/E, CD3þ IHC (400x) and Trichrome (100x) in allogeneic control and allogeneic azithromycin-treated at week þ14 after HCT. (P,Q) Lung pathology at week þ14 by H&E staining scored using a semiquantitative scoring system as described in Materials and Method. (R) CD3þ cell quantification in the lung as cells per high power field at week þ14. (S,T) Pathology of the gut and liver by H&E staining at week þ14 after HCT using scoring systems as described in Materials and Methods. Results shown as mean  SEM (syn control: n ¼ 5; syn azithromycin: n ¼ 4; allogeneic control: n ¼ 7; allogeneic azithromycin: n ¼ 11). For H/E and CD3þ IHC, Zeiss Axioskop 40 microscope at 400 x magnifications and for trichrome stain Nikon EclipseE400 microscope at 100 x magnification was used.

34

S.V. Radhakrishnan et al. / Biol Blood Marrow Transplant 21 (2015) 30e38

Figure 3. Tissue cytokine levels at week þ6 and week þ14 after allo-HCT. Animals underwent transplantation as described in Materials and Methods. (A-D) Cytokine concentration in the lung analyzed in the lung supernatant by ELISA at week þ6 after HCT as described in Materials and Methods: IFN-g, CXCL1, MIP2, and CXCL9 respectively in pg/mg total protein. (E-H) Cytokine concentration in the gut by ELISA at week þ6 after HCT: IFN-g, CCL-2, CXCL9, and TNF-a respectively in pg/mg total protein. (syngeneic control: n ¼ 5; allogeneic control: n ¼ 5; azithromycin-treated allogeneic: n ¼ 8). Results shown as mean  SEM. (I-P) Cytokine concentration in the lung at week þ14 analyzed in the lung supernatant using Luminex technology as described in Materials and Methods: CXCL9, CXCL10, CCL5, IL-1b, IL-15, TNF-a, CCL3, and CXCL1 respectively in pg/mg total lung protein. Results shown as mean  SEM (syngeneic control: n ¼ 4; azithromycin-treated syngeneic: n ¼ 3; allogeneic control: n ¼ 4; azithromycin-treated allogeneic n ¼ 6).

allogeneic recipients had a significant reduction in CCL2 and IFN-g, but there was no difference in the other cytokines when compared with allogeneic controls (Figure 3EeH). No differences between syngeneic and allogeneic recipients were seen for CXCL1, MIP-2, IL-6, and CXCL10 in the gut and for MIP-2, IL-6,TNF-a, and CCL2 in lung, and there was no difference in hepatic expression of any of the tested inflammatory mediators between groups at this time point (data not shown). At week þ14 after HCT, azithromycin-treated allogeneic recipients demonstrated significant suppression of CXCL9, CCL5, IL-1b, IL-15, and TNF-a in the lungs compared with the allogeneic controls and a nonsignificant decrease was seen for CXCL10 (Figure 3IeN). CCL3 and CXCL1 levels were elevated in both allogeneic groups when compared with the syngeneic recipients, but they were not altered by azithromycin treatment (Figure 3O,P). There was no difference in cytokine expression in the liver between syngeneic and allogeneic groups (data not shown).

Azithromycin Decreases T cell proliferation in Response to Alloantigen Presented by APCs through the Induction of Regulatory T cells We then tested whether azithromycin alters alloreactive T cell responses when alloantigens are presented by APCs, thereby modulating the development of GVHD. Significant suppression of responder C57BL/6 T cell proliferation to B6D2F1 or BALB/c splenocytes as stimulators was observed as the concentration of azithromycin was increased (Figure 4A,B). Decreased proliferation was associated with reduced levels of cytokines (IFN-g, TNF-a, IL-6, IL-10, IL-17) in the MLR supernatant, whereas no differences were seen for IL-2; IL-4 was undetectable (Table 1). In contrast, when T cell activation occurred directly through anti-CD3/CD28 stimulation and in the absence of alloreactive APCs, no differences in T cell proliferation (Figure 4C) or cytokine expression were seen (Table 1). Azithromycin-related decrease of T cell proliferation in MLRs was associated with an increase in the percentage of

S.V. Radhakrishnan et al. / Biol Blood Marrow Transplant 21 (2015) 30e38

35

Figure 4. Azithromycin decreases T cell proliferation in response to alloantigen presented by APCs through the induction of regulatory T cells. Mixed lymphocytic reaction (MLR) using C57BL/6 T cells as responders and irradiated (A) B6D2F1 or (B) BALB/c splenocytes as stimulators at varying concentrations of azithromycin. (C) Anti-CD3/CD28 induced T cell proliferation using C57BL/6 as responders at varying concentrations of azithromycin. (D-E) Percentage of regulatory CD4þ Foxp3þ T cells (Tregs) of all CD4þ T cells was quantified by flow cytometry in (D) MLR and (E) Anti-CD3/CD28 activation. (F) In vitro T cell migration assay using CXCL9 at different concentrations of azithromycin compared to control as described in Material and Methods. Results shown as mean  SEM; all experiments done in triplicates.

CD4þFoxP3þ regulatory T cell (Tregs) of all CD4þ T cells (Figure 4D), whereas when using anti-CD3/CD28 activation, no difference in Treg percentage was found (Figure 4E), suggesting that azithromycin induces the expansion of CD4þ FOXP3þ Tregs only in the presence of alloreactive APCs.

To test whether azithromycin directly alters the migratory properties of T cells, we performed a T cell migration assay using CXCL9 as the chemo attractant. The addition of azithromycin in 2 different concentrations (20 mg/mL and 50 mg/ mL) did not affect the number of transwell migrated cells

Table 1 Inflammatory Cytokine Expression in T Cell Proliferation Cytokines in pg/mL

Azithromycin (mg/mL)

cpm IFN-G TNF IL-2 IL-10 IL-17 IL-6 IL-4 cpm IFN-G TNF IL-2 IL-10 IL-17 IL-6 IL-4

20053 590.5 139.1 27.0 115.5 24.7 169.3 . 54868 9248 381.7 735.3 30.9 37.68 82.45 .

0 MLR

CD3/CD28

5       

1041 59.9 8.0 0.6 8.8 0.9 8.3

      

1873 467 20.64 87.64 2.3 8.3 12.2

17497 476.0 114.2 23.4 99.1 19.3 139.6 . 51182 9860 382.1 630.9 29.5 38.61 79.63 .

10       

1237 42.6 5.4* 1.2 14.4 3.3 16.3

      

2071 626.5 11.71 21.86 3.6 5.4 5.1

15340 363.6 115.4 23.4 98.9 17.6 120.7 . 51351 8570 366.5 675.4 36.58 27.01 85.22 .

20       

1157* 18.6y 5.6* 0.2 4.3 1.1 11.7*

      

1447 697.7 18.27 40.2 6.9 4.2 6.0

12902 218.7 104.6 25.2 65.0 9.4 96.7 . 51659 8994 371.9 709.9 44.01 35.58 75.23 .

      

1317y 19.2z 2.5y 1.0 6.8y 1.5z 9.4y

      

862 557.6 19.73 40.1 7.6 6.0 15.3

Supernatants of T cell proliferation assays (MLR versus anti-CD3/CD28 stimulation) with varying concentrations of azithromycin were analyzed for IFN-g, TNF-a, IL-2, IL-10, IL-17, IL-6, IL-4 using cytometric bead array as described in Materials and Methods. Results shown are mean  SEM; all experiments done in triplicates. CPM indicates count per minute; ., below detection. * P < .05. y P < .01. z P < .001.

36

S.V. Radhakrishnan et al. / Biol Blood Marrow Transplant 21 (2015) 30e38

(Figure 4F), suggesting that azithromycin does not have direct effects on T cell migration in vitro. Azithromycin Suppresses Alloreactive Splenic T cell Proliferation in Vivo at Day þ7 and Day þ56 after HCT and Induces Regulatory T cells As the addition of azithromycin suppressed T cell proliferation in vitro in an APC-dependent fashion, we next tested if azithromycin is able to decrease alloreactive T cell proliferation in vivo at 2 different time points after transplantation: at day þ7 and day þ56. Splenic T cell expansion was determined on day þ7 and day þ56 after bone marrow transplantation. On day þ7, a significant decrease in both CD4þ and CD8þ splenic T cells was observed in the azithromycin-treated group when compared with controltreated animals (Figure 5A,B), and the proportion of CD4þFoxp3þTregs was slightly although not significantly increased in the azithromycin-treated group when compared to allogeneic controls (Figure 5C). The suppression of splenic T cell proliferation of CD4þ and CD8þ cells was still evident at day þ56, although it was not statistically significant (Figure 5D,E), and a significant induction of CD4þ Foxp3þ Tregs in the azithromycin-treated group compared to the allogeneic controls was observed (Figure 5F). These changes were accompanied by a corresponding suppression of MHC class II expression on the CD11cþ dendritic cells in the azithromycin-treated group compared with the allogeneic control group (Figure 5G). There was no significant difference in the expression of CD80þ or CD86þ on these dendritic cells (data not shown). DISCUSSION Noninfectious pulmonary complications after allo-HCT present a major cause for transplantation-related morbidity and mortality and can present as both restrictive and obstructive lung injury. Although azithromycin is commonly used in patients with late-occurring obstructive lung injury after allo-HCT, well-controlled data on its use in these patients are limited. Khalid et al. reported a small series of patients with BOS after allo-HCT, in which treatment with azithromycin resulted in stabilization of disease [20], whereas 2 other groups did not show clear efficacy in this

setting [19,27]. However, in both of these studies, azithromycin was initiated for established BOS, and it remains unanswered whether preemptive use of azithromycin early after allo-HCT, because of its anti-inflammatory and bacteriostatic effects, would affect acute GVHD and early lung injury, potentially altering the overall clinical course of lung injury after allogeneic HCT. Animal studies on the use of azithromycin in allo-HCT are also limited. Recently, using a murine HCT model, Iwamoto et al. showed that azithromycin, when given very early, can result in improved survival and decreased early tissue injury of the liver and intestine after allo-HCT [28]. Using a well-established murine model for lung injury after allo-HCT [29], we demonstrated that preventive early treatment with azithromycin given over a prolonged period of time decreases lung injury and improves overall survival. Histological hallmarks in murine models of lung disease after allogeneic HCT include lymphocytic peribronchiolitis, interstitial pneumonitis, fibrosis, and BOS [2,5,29-32]. In our model, lung injury after allo-HCT has been classified more as a interstitial pneumonitis form of acute lung injury, but our group and others have shown that this lung injury in mice worsens over time, with inflammatory changes progressively shifting toward peribronchial and perivascular structures, along with increasing pulmonary obstruction [25,29]. We now show that the preemptive use of azithromycin, initiated early at 14 days after transplantation, before major significant structural changes have occurred, results in a decrease in lung pathology and lung function decline when assessed at 6 weeks and 3 months later. Consistent with a similar study by Cohen et al. [29], we did not observe bronchiolitis obliterans in this model. Periluminal infiltrates at 14 weeks showed a higher percentage of CD3þ cells in control-treated versus azithromycin-treated animals. In the BAL of allogeneic controls, a predominance of CD4þ T cells compared with the CD8þ T cells was found, supporting previous reports that CD4þ T cells may be more involved in chronic lung injury, whereas CD8þ T cells are rather increased early [10,29]. Decreased lung injury in azithromycin-treated animals was associated with a significant reduction in BAL CD4þ and, to a lesser degree, CD8þ T cells, as well as in decreased CD3þ

Figure 5. Azithromycin suppresses alloreactive splenic T cell proliferation in vivo at day þ7 and day þ56 after HCT and induces regulatory T cells. Animals underwent transplantation as described in Materials and Methods. (A,B) Splenic CD4þ T cell and CD8þ T cell proliferation in allogeneic controls and allogeneic azithromycintreated animals expressed as cells/spleen. (C) Percentage of regulatory CD4þ Foxp3þ T cells of total cells in the spleen of allogeneic controls and allogeneic azithromycin recipients (n ¼ 6 in both groups). (D,E) Splenic CD4þ T cell and CD8þ T cell proliferation in the allogeneic controls and azithromycin-treated animals expressed as cells per spleen. (F) Percentage of regulatory CD4þ Foxp3þ T cells of total cells in the spleen of allogeneic controls and allogeneic azithromycin recipients (G) MHC class II expression on CD11cþ dendritic cells in the control and azithromycin-treated recipients (control n ¼ 9, azithromycin-treated n ¼ 10). Results shown as mean  SEM.

S.V. Radhakrishnan et al. / Biol Blood Marrow Transplant 21 (2015) 30e38

periluminal T cell infiltrates along with decreased lung fibrosis. In addition to its bacteriostatic effects, azithromycin has known anti-inflammatory properties. T cell recruitment to the lungs partially depends on the expression of CXCL9 and CXCL10 [11], with donor T cellederived CCL5 being an amplifying factor [10]. It is likely that azithromycin exerted direct anti-inflammatory properties, suppressing pulmonary cytokine and chemokine expression and, thereby, decreased T cell infiltration and lung pathology in this model. Early in the post-transplantation period at week 6, azithromycintreated animals demonstrated suppressed pulmonary IFN-g and CXCL9 expression, associated with less inflammation and improved survival. In addition, at this time, diminished gut injury was observed along with a reduction of intestinal CCL2, IFN-g, and CXCL9, in keeping with an IFN-gemediated Th1 response during acute GVHD. At a later time point at week 14, when IFN-g expression is known to be not elevated in the lung in this model [29], interestingly, the suppressive effects of azithromycin treatment on CXCL9 along with CXCL10 were sustained. Although these chemokines are classically considered “interferon gamma inducible,” other proinflammatory cytokines likely contribute to their expression as well [33]. This persistence of decreased expression could be a compound result of local suppression of inflammation and decreased number of infiltrating alloreactive T cells. IL-15 has been shown to contribute to GVHD severity [34], most likely through the expansion of CD8þ memory T cells and maturation of dendritic cells [35], and in our study azithromycin treatment resulted in decreased IL15 levels in the lungs after allo-HCT, suggesting that IL-15 suppression possibly contributes to reduction in pulmonary T cell infiltration and lung injury. Further supportive of the anti-inflammatory effects of azithromycin in our model, TNFa and IL-1b, cytokines associated with increased tissue damage and lung injury [36], were significantly reduced in the lungs of azithromycin-treated allo recipients and correlated with less pathology and improved survival in this group at week 14. CXCL1, which is known to be suppressed by azithromycin during pulmonary inflammation [37], was also decreased both early and late after allo-HCT in our model, yet neutrophils do not seem to play a critical role in this model of lung injury (unpublished data). Interestingly, we could demonstrate that azithromycin can suppress T cell expansion and T cell cytokine production in vitro and that this was only seen in the presence of alloantigen presenting cells, but not when T cell responses were induced specifically via CD3/CD28 activation. Furthermore, decreased T cell activation was associated with the induction of Tregs both in vitro and in vivo. Consistently, azithromycintreated bone marrowederived dendritic cells, when used in vitro as stimulators, are known to decrease T cell proliferation [38]. In our study, we were able to show in vivo suppression of MHC class II expression on dendritic cells in azithromycin-treated animals similar to the reported in vitro suppression of MHC class II expression on CD11cþ dendritic cells as reported by Iwamoto et al. [38]. Our in vivo findings further demonstrate a corresponding increase in the Treg expansion. This was not observed when T cell activation occurred through T cell receptor/costimulatory molecule antibody stimulation in the actual absence of APCs. This suggests that the observed effects of azithromycin on alloreactive T cell proliferation seem not related to direct effects on T cells, but rather are mediated through its actions on APCs. Tregs have been shown to prevent the development of

37

bronchiolitis obliterans in lung transplant recipients [39,40]. Azithromycin achieves high drug concentrations in the lung [12], and it is possible that in addition to its systemic effects, sustained pulmonary induction of Tregs occurs during disease development. Azithromycin is usually a well-tolerated antibiotic with convenient dosing that attains high lung tissue concentrations [12]. Despite recent concerns related to its arrhythmogenic potential [41], with an increased cardiovascular risk specifically for patients with high cardiovascular risk scores, none of the studies on the use of azithromycin in lung transplantation have shown increased azithromycinassociated severe adverse effects [17]. Long-term use of azithromycin in CF patients has shown an increased incidence of infection with nontuberculous mycobacteria secondary to its inhibitory effects on autophagy [42], and monitoring HCT recipients, due to their immunocompromised state, for nontuberculous mycobacteria infections may be beneficial when given azithromycin over a prolonged time period. In summary, our study showed that azithromycin treatment given prophylactically before or started early after transplantation improved survival and decreased GVHD severity and noninfectious lung injury in allo HCT recipients. The beneficial effects of azithromycin seemed multipronged, involving the suppression of inflammation in the absence of microbial infection, cytokine and chemokine-related modulation of T cell migration, and inhibition of alloreactive proliferative T cell responses through the induction of Tregs. Our study supports the value of azithromycin in the prevention and treatment of noninfectious lung injury after allo-HCT under controlled experimental conditions in association with possible beneficial effects on acute GVHD. ACKNOWLEDGMENTS Financial disclosure: Funding provided by the Department of Hematology/Oncology University of Regensburg, Huntsman Cancer Institute, University of Utah and Feist-WeillerCancer Center Louisiana State University Health Sciences Center Shreveport. No involvement of the funding source in the design, data collection, analysis, interpretation, or publishing the research. Conflict of interest statement: There are no conflicts of interest to report. Authorship contributions: S.V.R., S.P., F.J.K., N.K., and G.C.H. conceived and designed the experiments. S.V.R., S.P., G.M., G.C.H., S.M., M.H., E.S., F.J.K., N.K., and E.H. performed the experiments. S.V.R., S.P., G.M., F.J.K., and E.H. analyzed the data. E.H. contributed reagents/materials/analysis tools. S.V.R., S.P., and G.C.H. wrote the paper. REFERENCES 1. Panoskaltsis-Mortari A, Griese M, Madtes DK, et al. An official American Thoracic Society research statement: noninfectious lung injury after hematopoietic stem cell transplantation: idiopathic pneumonia syndrome. Am J Respir Crit Care Med. 2011;183:1262-1279. 2. Cooke KR, Kobzik L, Martin TR, et al. An experimental model of idiopathic pneumonia syndrome after bone marrow transplantation: I. The roles of minor H antigens and endotoxin. Blood. 1996;88: 3230-3239. 3. Cooke KR, Krenger W, Hill G, et al. Host reactive donor T cells are associated with lung injury after experimental allogeneic bone marrow transplantation. Blood. 1998;92:2571-2580. 4. Korngold R, Sprent J. Features of T cells causing H-2-restricted lethal graft-vs.-host disease across minor histocompatibility barriers. J Exp Med. 1982;155:872-883. 5. Panoskaltsis-Mortari A, Taylor PA, Yaeger TM, et al. The critical early proinflammatory events associated with idiopathic pneumonia syndrome in irradiated murine allogeneic recipients are due to donor T cell

38

6.

7.

8.

9.

10.

11.

12.

13.

14. 15. 16.

17.

18.

19.

20.

21.

22.

S.V. Radhakrishnan et al. / Biol Blood Marrow Transplant 21 (2015) 30e38

infusion and potentiated by cyclophosphamide. J Clin Invest. 1997;100: 1015-1027. Clark JG, Madtes DK, Hackman RC, et al. Lung injury induced by alloreactive Th1 cells is characterized by host-derived mononuclear cell inflammation and activation of alveolar macrophages. J Immunol. 1998; 161:1913-1920. Carlson MJ, West ML, Coghill JM, et al. In vitro-differentiated TH17 cells mediate lethal acute graft-versus-host disease with severe cutaneous and pulmonary pathologic manifestations. Blood. 2009;113:1365-1374. Liu Q, Ning J, Zhang Y, et al. Idiopathic pneumonia syndrome in mice after allogeneic bone marrow transplantation: association between idiopathic pneumonia syndrome and acute graft-versus-host disease. Transpl Immunol. 2010;23:12-17. Hildebrandt GC, Duffner UA, Olkiewicz KM, et al. A critical role for CCR2/MCP-1 interactions in the development of idiopathic pneumonia syndrome after allogeneic bone marrow transplantation. Blood. 2004; 103:2417-2426. Hildebrandt GC, Olkiewicz KM, Choi S, et al. Donor T-cell production of RANTES significantly contributes to the development of idiopathic pneumonia syndrome after allogeneic stem cell transplantation. Blood. 2005;105:2249-2257. Hildebrandt GC, Corrion LA, Olkiewicz KM, et al. Blockade of CXCR3 receptor:ligand interactions reduces leukocyte recruitment to the lung and the severity of experimental idiopathic pneumonia syndrome. J Immunol. 2004;173:2050-2059. Togami K, Chono S, Morimoto K. Distribution characteristics of clarithromycin and azithromycin, macrolide antimicrobial agents used for treatment of respiratory infections, in lung epithelial lining fluid and alveolar macrophages. Biopharm Drug Dispos. 2011;32:389-397. Southern KW, Barker PM, Solis-Moya A, Patel L. Macrolide antibiotics for cystic fibrosis. Cochrane Database Syst Rev. 2012;. http://dx.doi.org/ 10.1002/14651858.CD002203.pub4. Spagnolo P, Fabbri LM, Bush A. Long-term macrolide treatment for chronic respiratory disease. Eur Respir J. 2013;42:239-251. Albert RK, Connett J, Bailey WC, et al. Azithromycin for prevention of exacerbations of COPD. N Engl J Med. 2011;365:689-698. Wong C, Jayaram L, Karalus N, et al. Azithromycin for prevention of exacerbations in non-cystic fibrosis bronchiectasis (EMBRACE): a randomised, double-blind, placebo-controlled trial. Lancet. 2012;380: 660-667. Vos R, Vanaudenaerde BM, Verleden SE, et al. A randomised controlled trial of azithromycin to prevent chronic rejection after lung transplantation. Eur Respir J. 2011;37:164-172. Jain R, Hachem RR, Morrell MR, et al. Azithromycin is associated with increased survival in lung transplant recipients with bronchiolitis obliterans syndrome. J Heart Lung Transplant. 2010;29:531-537. Lam DC, Lam B, Wong MK, et al. Effects of azithromycin in bronchiolitis obliterans syndrome after hematopoietic SCTea randomized doubleblinded placebo-controlled study. Bone Marrow Transplant. 2011;46: 1551-1556. Khalid M, Al Saghir A, Saleemi S, et al. Azithromycin in bronchiolitis obliterans complicating bone marrow transplantation: a preliminary study. Eur Respir J. 2005;25:490-493. Krenger W, Snyder KM, Byon JC, et al. Polarized type 2 alloreactive CD4þ and CD8þ donor T cells fail to induce experimental acute graftversus-host disease. J Immunol. 1995;155:585-593. Bosnar M, Bosnjak B, Cuzic S, et al. Azithromycin and clarithromycin inhibit lipopolysaccharide-induced murine pulmonary neutrophilia mainly through effects on macrophage-derived granulocyte-macrophage colony-stimulating factor and interleukin-1beta. J Pharmacol Exp Ther. 2009;331:104-113.

23. Kaplan DH, Anderson BE, McNiff JM, et al. Target antigens determine graft-versus-host disease phenotype. J Immunol. 2004;173:5467-5475. 24. Hill GR, Cooke KR, Teshima T, et al. Interleukin-11 promotes T cell polarization and prevents acute graft-versus-host disease after allogeneic bone marrow transplantation. J Clin Invest. 1998;102:115-123. 25. Miklos S, Mueller G, Chang Y, et al. Pulmonary function changes in experimental graft-versus-host disease of the lung. Biol Blood Marrow Transplant. 2008;14:1004-1016. 26. Bouazzaoui A, Spacenko E, Mueller G, et al. Chemokine and chemokine receptor expression analysis in target organs of acute graft-versus-host disease. Genes Immun. 2009;10:687-701. 27. Lemonnier F, Rivaud E, Neveu H, et al. Azithromycin in bronchiolitis obliterans syndrome after hematopoietic SCT. Bone Marrow Transplant. 2012;47:1374. 28. Iwamoto S, Azuma E, Kumamoto T, et al. Efficacy of azithromycin in preventing lethal graft-versus-host disease. Clin Exp Immunol. 2013; 171:338-345. 29. Shankar G, Bryson JS, Jennings CD, et al. Idiopathic pneumonia syndrome in mice after allogeneic bone marrow transplantation. Am J Respir Cell Mol Biol. 1998;18:235-242. 30. Flowers ME, Lee S, Vogelsang G. An update on how to treat chronic GVHD. Blood. 2003;102:2312. 31. Panoskaltsis-Mortari A, Tram KV, Price AP, et al. A new murine model for bronchiolitis obliterans post-bone marrow transplant. Am J Respir Crit Care Med. 2007;176:713-723. 32. Panoskaltsis-Mortari A, Strieter RM, Hermanson JR, et al. Induction of monocyte- and T-cell-attracting chemokines in the lung during the generation of idiopathic pneumonia syndrome following allogeneic murine bone marrow transplantation. Blood. 2000;96: 834-839. 33. Loos T, Dekeyzer L, Struyf S, et al. TLR ligands and cytokines induce CXCR3 ligands in endothelial cells: enhanced CXCL9 in autoimmune arthritis. Lab Invest. 2006;86:902-916. 34. Blaser BW, Roychowdhury S, Kim DJ, et al. Donor-derived IL-15 is critical for acute allogeneic graft-versus-host disease. Blood. 2005;105: 894-901. 35. Roychowdhury S, Blaser BW, Freud AG, et al. IL-15 but not IL-2 rapidly induces lethal xenogeneic graft-versus-host disease. Blood. 2005;106: 2433-2435. 36. Gerbitz A, Nickoloff BJ, Olkiewicz K, et al. A role for tumor necrosis factor-alpha-mediated endothelial apoptosis in the development of experimental idiopathic pneumonia syndrome. Transplantation. 2004; 78:494-502. 37. Tsai WC, Rodriguez ML, Young KS, et al. Azithromycin blocks neutrophil recruitment in Pseudomonas endobronchial infection. Am J Respir Crit Care Med. 2004;170:1331-1339. 38. Iwamoto S, Kumamoto T, Azuma E, et al. The effect of azithromycin on the maturation and function of murine bone marrow-derived dendritic cells. Clin Exp Immunol. 2011;166:385-392. 39. Bhorade SM, Chen H, Molinero L, et al. Decreased percentage of CD4þFoxP3þ cells in bronchoalveolar lavage from lung transplant recipients correlates with development of bronchiolitis obliterans syndrome. Transplantation. 2010;90:540-546. 40. Gregson AL, Hoji A, Palchevskiy V, et al. Protection against bronchiolitis obliterans syndrome is associated with allograft CCR7þ CD45RA- T regulatory cells. PLoS One. 2010;5:e11354. 41. Ray WA, Murray KT, Hall K, et al. Azithromycin and the risk of cardiovascular death. N Engl J Med. 2012;366:1881-1890. 42. Renna M, Schaffner C, Brown K, et al. Azithromycin blocks autophagy and may predispose cystic fibrosis patients to mycobacterial infection. J Clin Invest. 2011;121:3554-3563.