Exploration of α1-antitrypsin treatment protocol for islet transplantation: dosing plan and route of administration

Boris M Baranovski, Eyal Ozeri, Galit Shahaf, David E Ochayon, Ronen Schuster, Nofar Bahar, Noa Kalay, Pablo Cal, Mark I Mizrahi, Omer Nisim, Pnina Strauss, Eran Schenker, Eli C Lewis, Boris M Baranovski, Eyal Ozeri, Galit Shahaf, David E Ochayon, Ronen Schuster, Nofar Bahar, Noa Kalay, Pablo Cal, Mark I Mizrahi, Omer Nisim, Pnina Strauss, Eran Schenker, Eli C Lewis

Abstract

Life-long weekly infusions of human α1-antitrypsin (hAAT) are currently administered as augmentation therapy for patients with genetic AAT deficiency (AATD). Several recent clinical trials attempt to extend hAAT therapy to conditions outside AATD, including type 1 diabetes. Since the endpoint for AATD is primarily the reduction of risk for pulmonary emphysema, the present study explores hAAT dose protocols and routes of administration in attempt to optimize hAAT therapy for islet-related injury. Islet-grafted mice were treated with hAAT (Glassia™; i.p. or s.c.) under an array of clinically relevant dosing plans. Serum hAAT and immunocyte cell membrane association were examined, as well as parameters of islet survival. Results indicate that dividing the commonly prescribed 60 mg/kg i.p. dose to three 20 mg/kg injections is superior in affording islet graft survival; in addition, a short dynamic descending dose protocol (240→120→60→60 mg/kg i.p.) is comparable in outcomes to indefinite 60 mg/kg injections. While hAAT pharmacokinetics after i.p. administration in mice resembles exogenous hAAT treatment in humans, s.c. administration better imitated the physiological progressive rise of hAAT during acute phase responses; nonetheless, only the 60 mg/kg dose depicted an advantage using the s.c. route. Taken together, this study provides a platform for extrapolating an islet-relevant clinical protocol from animal models that use hAAT to protect islets. In addition, the study places emphasis on outcome-oriented analyses of drug efficacy, particularly important when considering that hAAT is presently at an era of drug-repurposing towards an extended list of clinical indications outside genetic AATD.

Keywords: diabetes; inflammation.

The American Society for Pharmacology and Experimental Therapeutics.

Figures

Fig. 1.
Fig. 1.
The effect of Glassia on pancreatic islet responses and dendritic cell maturation during inflammatory conditions. (A and B) Primary mouse islets (35 per well in triplicate) were cultured for 48 hours in the absence (CT) or presence of IL-1β and IFNγ (5 ng/ml each), with or without overnight pretreatment with Glassia (0.5 mg/ml). (A) Islet viability and insulin release. (B) Supernatant levels of nitric oxide, IL-6, MCP-1, and IL-10. (C) BMDCs (3 × 105 cells per well in triplicates) were stimulated with IL-1β and IFNγ (5 ng/ml each) overnight in the presence or absence of Glassia (0.5 mg/ml). Cells were analyzed by flow cytometry. Representative results of three independent experiments. Mean ± S.E.M., *P < 0.05, **P < 0.01.
Fig. 2.
Fig. 2.
Protection of pancreatic islet allografts by Glassia. Mice were rendered hyperglycemic by single-dose STZ (225 mg/kg) and then subjected to islet allograft transplantation. Experimental groups included: 15 mg/kg (n = 3), 30 mg/kg (n = 13), 60 mg/kg (n = 10), 120 mg/kg (n = 5), and 240 mg/kg (n = 5). Control group animals received PBS vehicle (n = 9). (A) Graft survival curve. (B) Graft site histology, K signifies kidney tissue and G signifies graft site. Representative images of rejected untreated mouse graft (top), 60 mg/kg-treated rejected graft (bottom left), and accepted graft (bottom right).
Fig. 3.
Fig. 3.
The effect of intraperitoneal Glassia dose distribution treatment on hAAT serum concentration and islet graft survival. (A) Mice (n = 3–5) were treated with 60 mg/kg i.p Glassia as indicated (arrow). Serum levels of human AAT were determined with specific ELISA. Results of 3 independent experiments, mean ± S.E.M. (B) Mice were stimulated with ThG for 72 hours after treatment with Glassia (240 mg/kg i.p.). Peritoneal lavage was performed 16 hours later, and membrane-associated hAAT levels were measured by flow cytometry (top panel). Groups included nonstimulated, untreated mice (CT, red); ThG-stimulated untreated mice (ThG, yellow); nonstimulated Glassia-treated mice (CT/Glassia, blue), and ThG-stimulated Glassia-treated mice (ThG/Glassia, green), n = 3. Results are representative of 3 independent repeats. For cell-associated analysis (bottom panel), peritoneal macrophages (1×106, in triplicate) were pulsed with Glassia (0.5 mg/ml). hAAT binding was evaluated by Western blotting. (C) Mice were treated with 30 mg/kg (left) or 20 mg/kg (right) Glassia intraperitoneally at indicated time points (arrow; n = 3 or 4 in each group). Serum levels of human AAT were determined. Results of 3 independent experiments, mean ± S.E.M. (D) Islet graft survival curve. Groups include: ×3 of 20 mg/kg i.p (n = 3, yellow) and ×2 of 30 mg/kg i.p (n = 6, red). CT and 60 mg/kg treatment groups are duplicates of Fig. 2A.
Fig. 4.
Fig. 4.
The effect of subcutaneous Glassia treatment on hAAT serum concentration and islet graft survival. (A) Mice were treated with 60 and 20 mg/kg Glassia subcutaneously at indicated time points (arrow; n = 3 or 4 in each group). Serum hAAT levels were measured. Results are representative of three independent experiments, mean ± S.E.M. (B) Islet grafted mice were treated with Glassia either intraperitoneally (dashed) or subcutaneously (solid). Groups include untreated mice (CT; gray. n = 9); ×1 of 60 mg/kg i.p (n = 10); ×1 of 60 mg/kg s.c (n = 6); ×2 of 30 mg/kg i.p. (n = 6); ×2 of 30 mg/kg s.c. (n = 5); ×1 of 30 mg/kg i.p (n = 13); ×1 of 30 mg/kg s.c. (n = 2); ×1 of 15 mg/kg i.p (n = 3); ×1 of 15 mg/kg s.c. (n = 1). Islet graft survival curve.
Fig. 5.
Fig. 5.
The effect of Glassia dynamic dose treatment on graft survival. Islet-grafted mice were treated with Glassia using dynamic dose regimen. (A) Illustration representing the treatment protocol. (B) Islet graft survival curve under Glassia dynamic dose treatment (blue area). Comparison of intraperitoneal (red; n = 6) and subcutaneous (purple; n= 3) dynamic dose treatment.

References

    1. Ashkenazi E, Baranovski BM, Shahaf G, Lewis EC. (2013) Pancreatic islet xenograft survival in mice is extended by a combination of alpha-1-antitrypsin and single-dose anti-CD4/CD8 therapy. PLoS One 8:e63625.
    1. Austin GE, Mullins RH, Morin LG. (1987) Non-enzymic glycation of individual plasma proteins in normoglycemic and hyperglycemic patients. Clin Chem 33:2220–2224.
    1. Balbi B, Ferrarotti I, Miravitlles M. (2016) Efficacy of augmentation therapy for emphysema associated with α1-antitrypsin deficiency: enough is enough. Eur Respir J 47:35–38.
    1. Braza F, Brouard S, Chadban S, Goldstein DR. (2016) Role of TLRs and DAMPs in allograft inflammation and transplant outcomes. Nat Rev Nephrol 12:281–290.
    1. Daemen MA, Heemskerk VH, van’t Veer C, Denecker G, Wolfs TG, Vandenabeele P, Buurman WA. (2000) Functional protection by acute phase proteins alpha(1)-acid glycoprotein and alpha(1)-antitrypsin against ischemia/reperfusion injury by preventing apoptosis and inflammation. Circulation 102:1420–1426.
    1. Dhami R, Zay K, Gilks B, Porter S, Wright JL, Churg A. (1999) Pulmonary epithelial expression of human alpha1-antitrypsin in transgenic mice results in delivery of alpha1-antitrypsin protein to the interstitium. J Mol Med (Berl) 77:377–385.
    1. Ehlers MR. (2014) Immune-modulating effects of alpha-1 antitrypsin. Biol Chem 395:1187–1193.
    1. Elshikha AS, Lu Y, Chen MJ, Akbar M, Zeumer L, Ritter A, Elghamry H, Mahdi MA, Morel L, Song S. (2016) Alpha 1 antitrypsin inhibits dendritic cell activation and attenuates nephritis in a mouse model of lupus. PLoS One 11:e0156583.
    1. Feng Y, Hu L, Xu Q, Yuan H, Ba L, He Y, Che H. (2015) Cytoprotective role of alpha-1 antitrypsin in vascular endothelial cell under hypoxia/reoxygenation condition. J Cardiovasc Pharmacol 66:96–107.
    1. Feng Y, Xu J, Zhou Q, Wang R, Liu N, Wu Y, Yuan H, Che H. (2016) Alpha-1 antitrypsin prevents the development of preeclampsia through suppression of oxidative stress. Front Physiol 7:176.
    1. Finotti P, Pagetta A. (2004) A heat shock protein70 fusion protein with alpha1-antitrypsin in plasma of type 1 diabetic subjects. Biochem Biophys Res Commun 315:297–305.
    1. Fleixo-Lima G, Ventura H, Medini M, Bar L, Strauss P, Lewis EC. (2014) Mechanistic evidence in support of alpha1-antitrypsin as a therapeutic approach for type 1 diabetes. J Diabetes Sci Technol 8:1193–1203.
    1. Flotte TR, Trapnell BC, Humphries M, Carey B, Calcedo R, Rouhani F, Campbell-Thompson M, Yachnis AT, Sandhaus RA, McElvaney NG, et al. (2011) Phase 2 clinical trial of a recombinant adeno-associated viral vector expressing α1-antitrypsin: interim results. Hum Gene Ther 22:1239–1247.
    1. Gao W, Zhao J, Kim H, Xu S, Chen M, Bai X, Toba H, Cho HR, Zhang H, Keshavjeel Set al. (2014) alpha1-Antitrypsin inhibits ischemia reperfusion-induced lung injury by reducing inflammatory response and cell death. J Heart Lung Transplant 33:309–315.
    1. Gottlieb PA, Alkanani AK, Michels AW, Lewis EC, Shapiro L, Dinarello CA, Zipris D. (2014) α1-Antitrypsin therapy downregulates toll-like receptor-induced IL-1β responses in monocytes and myeloid dendritic cells and may improve islet function in recently diagnosed patients with type 1 diabetes. J Clin Endocrinol Metab 99:E1418–E1426.
    1. Grimstein C, Choi YK, Satoh M, Lu Y, Wang X, Campbell-Thompson M, Song S. (2010) Combination of alpha-1 antitrypsin and doxycycline suppresses collagen-induced arthritis. J Gene Med 12:35–44.
    1. Guttman O, Baranovski BM, Schuster R, Kaner Z, Freixo-Lima GS, Bahar N, Kalay N, Mizrahi MI, Brami I, Ochayon DE, et al. (2015) Acute-phase protein α1-anti-trypsin: diverting injurious innate and adaptive immune responses from non-authentic threats. Clin Exp Immunol 179:161–172.
    1. Iskender I, Sakamoto J, Nakajima D, Lin H, Chen M, Kim H, Guan Z, Del Sorbo L, Hwang D, Waddell TK, et al. (2016) Human α1-antitrypsin improves early post-transplant lung function: Pre-clinical studies in a pig lung transplant model. J Heart Lung Transplant 35:913–921.
    1. Janciauskiene SM, Nita IM, Stevens T. (2007) Alpha1-antitrypsin, old dog, new tricks. Alpha1-antitrypsin exerts in vitro anti-inflammatory activity in human monocytes by elevating cAMP. J Biol Chem 282:8573–8582.
    1. Jin JF, Zhu LL, Chen M, Xu HM, Wang HF, Feng XQ, Zhu XP, Zhou Q. (2015) The optimal choice of medication administration route regarding intravenous, intramuscular, and subcutaneous injection. Patient Prefer Adherence 9:923–942.
    1. Jonigk D, Al-Omari M, Maegel L, Müller M, Izykowski N, Hong J, Hong K, Kim SH, Dorsch M, Mahadeva R, et al. (2013) Anti-inflammatory and immunomodulatory properties of α1-antitrypsin without inhibition of elastase. Proc Natl Acad Sci USA 110:15007–15012.
    1. Kalis M, Kumar R, Janciauskiene S, Salehi A, Cilio CM. (2010) α 1-antitrypsin enhances insulin secretion and prevents cytokine-mediated apoptosis in pancreatic β-cells. Islets 2:185–189.
    1. Koulmanda M, Bhasin M, Fan Z, Hanidziar D, Goel N, Putheti P, Movahedi B, Libermann TA, Strom TB. (2012) Alpha 1-antitrypsin reduces inflammation and enhances mouse pancreatic islet transplant survival. Proc Natl Acad Sci USA 109:15443–15448.
    1. Koulmanda M, Sampathkumar RS, Bhasin M, Qipo A, Fan Z, Singh G, Movahedi B, Duggan M, Chipashvili V, Strom TB. (2014) Prevention of nonimmunologic loss of transplanted islets in monkeys. Am J Transplant 14:1543–1551.
    1. Lewis EC. (2012) Expanding the clinical indications for α(1)-antitrypsin therapy. Mol Med 18:957–970.
    1. Lewis EC, Mizrahi M, Toledano M, Defelice N, Wright JL, Churg A, Shapiro L, Dinarello CA. (2008a) alpha1-Antitrypsin monotherapy induces immune tolerance during islet allograft transplantation in mice. Proc Natl Acad Sci USA 105:16236–16241.
    1. Lewis EC, Mizrahi M, Toledano M, Defelice N, Wright JL, Churg A, Shapiro L, Dinarello CA. (2008b) alpha1-Antitrypsin monotherapy induces immune tolerance during islet allograft transplantation in mice. Proc Natl Acad Sci USA 105:16236–16241.
    1. Lewis EC, Shapiro L, Bowers OJ, Dinarello CA. (2005) Alpha1-antitrypsin monotherapy prolongs islet allograft survival in mice. Proc Natl Acad Sci USA 102:12153–12158.
    1. Lior Y, Geyra A, Lewis EC. (2016) Therapeutic compositions and uses of alpha1-antitrypsin: a patent review (2012 - 2015). Expert Opin Ther Pat 26:581–589.
    1. Lu Y, Tang M, Wasserfall C, Kou Z, Campbell-Thompson M, Gardemann T, Crawford J, Atkinson M, Song S. (2006) Alpha1-antitrypsin gene therapy modulates cellular immunity and efficiently prevents type 1 diabetes in nonobese diabetic mice. Hum Gene Ther 17:625–634.
    1. Ma H, Lu Y, Li H, Campbell-Thompson M, Parker M, Wasserfall C, Haller M, Brantly M, Schatz D, Atkinson M, et al. (2010) Intradermal alpha1-antitrypsin therapy avoids fatal anaphylaxis, prevents type 1 diabetes and reverses hyperglycaemia in the NOD mouse model of the disease. Diabetologia 53:2198–2204.
    1. Ochayon DE, Mizrahi M, Shahaf G, Baranovski BM, Lewis EC. (2013) Human α1-Antitrypsin Binds to Heat-Shock Protein gp96 and Protects from Endogenous gp96-Mediated Injury In vivo. Front Immunol 4:320.
    1. Ozeri E, Mizrahi M, Shahaf G, Lewis EC. (2012) α-1 antitrypsin promotes semimature, IL-10-producing and readily migrating tolerogenic dendritic cells. J Immunol 189:146–153.
    1. Pamarthi MF, Taylor GM, Wilson J, Scuderi P, Arora V. (2008) Pharmacokinetics of Subcutaneously Administered Alpha-1 Antitrypsin. J Allergy Clin Immunol 123:S163.
    1. Pileggi A, Molano RD, Song S, Zahr E, SanJose S, Villate S, Wasserfall C, Ricordi C, Atkinson MA, Inverardi L. (2008) Alpha-1 antitrypsin treatment of spontaneously diabetic nonobese diabetic mice receiving islet allografts. Transplant Proc 40:457–458.
    1. Pott GB, Chan ED, Dinarello CA, Shapiro L. (2009) Alpha-1-antitrypsin is an endogenous inhibitor of proinflammatory cytokine production in whole blood. J Leukoc Biol 85:886–895.
    1. Rachmiel M, Strauss P, Dror N, Benzaquen H, Horesh O, Tov N, Weintrob N, Landau Z, Ben-Ami M, Haim A, et al. (2016) Alpha-1 antitrypsin therapy is safe and well tolerated in children and adolescents with recent onset type 1 diabetes mellitus. Pediatr Diabetes 17:351–359.
    1. Shahaf G, Moser H, Ozeri E, Mizrahi M, Abecassis A, Lewis EC. (2011) α-1-antitrypsin gene delivery reduces inflammation, increases T-regulatory cell population size and prevents islet allograft rejection. Mol Med 17:1000–1011.
    1. Sorrells S, Camprubi S, Griffin R, Chen J, Ayguasanosa J. (2015) SPARTA clinical trial design: exploring the efficacy and safety of two dose regimens of alpha1-proteinase inhibitor augmentation therapy in alpha1-antitrypsin deficiency. Respir Med 109:490–499.
    1. Stolk J, Nieuwenhuizen W, Stoller JK, Aboussouan L. (2005) High dose intravenous AAT and plasma neutrophil derived fibrinogen fragments. Thorax 60:84.
    1. Subramanian S, Shahaf G, Ozeri E, Miller LM, Vandenbark AA, Lewis EC, Offner H. (2011) Sustained expression of circulating human alpha-1 antitrypsin reduces inflammation, increases CD4+FoxP3+ Treg cell population and prevents signs of experimental autoimmune encephalomyelitis in mice. Metab Brain Dis 26:107–113.
    1. Subramaniyam D, Zhou H, Liang M, Welte T, Mahadeva R, Janciauskiene S. (2010) Cholesterol rich lipid raft microdomains are gateway for acute phase protein, SERPINA1. Int J Biochem Cell Biol 42:1562–1570.
    1. Tawara I, Sun Y, Lewis EC, Toubai T, Evers R, Nieves E, Azam T, Dinarello CA, Reddy P. (2012) Alpha-1-antitrypsin monotherapy reduces graft-versus-host disease after experimental allogeneic bone marrow transplantation. Proc Natl Acad Sci USA 109:564–569.
    1. Tilg H, Vannier E, Vachino G, Dinarello CA, Mier JW. (1993) Antiinflammatory properties of hepatic acute phase proteins: preferential induction of interleukin 1 (IL-1) receptor antagonist over IL-1 beta synthesis by human peripheral blood mononuclear cells. J Exp Med 178:1629–1636.
    1. Wewers MD, Casolaro MA, Sellers SE, Swayze SC, McPhaul KM, Wittes JT, Crystal RG. (1987) Replacement therapy for alpha 1-antitrypsin deficiency associated with emphysema. N Engl J Med 316:1055–1062.
    1. Zhou X, Liu Z, Shapiro L, Yang J, Burton GF. (2015) Low-density lipoprotein receptor-related protein 1 mediates α1-antitrypsin internalization in CD4+ T lymphocytes. J Leukoc Biol 98:1027–1035.

Source: PubMed

3
Předplatit