Skip to main content

PERSPECTIVE article

Front. Immunol., 27 April 2018
Sec. Alloimmunity and Transplantation

“Phage Transplantation in Allotransplantation”: Possible Treatment in Graft-Versus-Host Disease?

  • 1Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland
  • 2Phage Therapy Unit, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland
  • 3Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Warsaw, Poland

Graft-versus-host disease, both acute and chronic (aGvHD, cGvHD) remains a major complication in patients undergoing hematopoietic cell transplantation (HCT) and a significant therapeutic challenge, as many patients do not respond adequately to presently available therapy. Increasing antimicrobial resistance has greatly revived interest in using bacterial viruses (phages) to combat antibiotic-resistant bacteria. In recent years, evidence has accumulated indicating that phages also have anti-inflammatory and immunomodulatory activities. This article suggests how these anti-bacterial and immunomodulatory activities of phages may be translated into a novel treatment of acute GvHD.

Introduction

aGvHD is recognized as the most common serious complication after hematopoietic cell transplantation (HCT). It results from immune attack of T cells contained in the graft which recognize foreign tissues of the recipient, activating the lymphocytes, which subsequently mount an immune response against the grafted cells. New data also point to the role of microbial imbalance, the degree of which correlates with outcome and mortality. Typical aGvHD affects the skin (maculopapular rash), gastrointestinal tract (damage to epithelial cells), and liver (damage to small bile ducts). Recently, immunobiology and therapy of aGvHD have been discussed in detail (1, 2). While corticosteroids remain the mainstay of treatment, a substantial percentage of patients fail to respond (31% of standard risk patients and 57% of high-risk patients). Although several second-line therapies are available, their value still remains uncertain, so the outcome of such steroid-resistant patients is poor (2, 3). Evidently, novel treatment options are needed.

New Insights into the Immunopathology of aGvHD—The Role of Neutrophils

Experiments in Mice

Although T cells play a major role in the initiation of aGvHD recent data highlight the significance of neutrophils and their pro-inflammatory role in the pathogenesis of GvHD, especially its later stages. Schwab et al. (4) analyzed neutrophil infiltration of the mouse ileum following allogeneic HCT and found that physical or genetic depletion of neutrophils reduced the intensity and mortality related to GvHD. The number of granulocytes in cellular infiltrates of the intestinal tract is highly predictive of severe aGvHD and transplant-related mortality (5). Importantly, neutrophil-induced tissue damage in this setting required the production of reactive oxygen species (ROS): blocking ROS markedly reduced those effects. Neutrophil infiltration was dependent on commensal bacterial flora and toll-like receptor (TLR) activation in neutrophils (6).

Clinical Data

In addition, the authors analyzed the number of neutrophils in patients after HCT and found that the severity of intestinal aGvHD strongly correlated with the number of neutrophils in aGvHD lesions and ROS-mediated oxidative injury. Furthermore, low rates of aGvHD were noted in patients with chronic granulomatous disease, whose neutrophils are deficient in ROS production (6). Granulocytes—generally considered as the first line of defense against pathogens—have recently been increasingly recognized also as antigen-presenting cells (APC) able to activate T lymphocytes. Activated T cells produce cytokines that upregulate major histocompatibility complex class II (MHC-II) and co-stimulatory molecule expression on neutrophils which differentiate into APC. Blocking of MHC-II on neutrophils abolishes their antigen-presenting function. In addition, neutrophils can activate themselves by interferon (IFN) gamma, which they produce. Upregulation of neutrophil MHC-II was shown in patients with active Wegener’s disease—neutrophils may present autoantigens and contribute to the development of autoimmune disorders; therefore, targeting neutrophils may be a new strategy to treat immune-mediated diseases (7).

Microbiota and aGvHD

The importance of bacteria in the development of aGvHD was recognized decades ago. Initial studies showed the benefit of intestinal decontamination, but later studies failed to confirm those results—probably selective decontamination would be efficient (8). Loss of intestinal bacterial diversity is associated with the development of aGvHD; on the other hand, GvHD can lead to marked alterations of intestinal flora and expansion of Enterobacteriales (Escherichia coli, Klebsiella, Enterobacter, and Enterococcus). In another study, an increase in Staphylococcaceae was noted with an early onset of aGvHD, while a relative amount of >5% of Enterobacteriaceae was associated with increased mortality due to sepsis. Recent data indicate that the presence of oral Actinobacteria and Firmicutes in the stool was positively correlated with subsequent aGvHD (911). Narrow spectrum antibiotics and metronidazole reduce the risk of GvHD (9). These data suggest that microbiota manipulation can offer a new strategy of prevention and possibly treatment of aGvHD. Intervention in the gut microbiota using prebiotics and postbiotics may be a promising option (12).

Promising initial results were obtained using fecal microbiota transplantation (FMT) for the treatment of aGvHD (13). Fecal extracts lacking bacteria were effective in mediating beneficial effects of FMT, which may suggest that phages are responsible for those effects (14). In fact, successful FMT was associated with increased abundance of Caudovirales (15); thus, phages may be a key component of the microbiota responsible for the efficiency of FMT (16). Reduced phage richness was found in GvHD which is another argument for the role of phages in alleviating this pathology (17).

Evolution of Phage Therapy (PT)

The dramatic increase of antimicrobial resistance and paucity of new antibiotics resulted in a growing interest in using bacterial viruses (phages) in combating antibiotic resistance (18, 19). Moreover, as pointed out, PT is evolving from treating complications to targeting diseases (20). While the safety of PT has been confirmed and clinical trials intended to provide full evidence of PT efficacy are underway, data have also accumulated indicating that PT mediates immunomodulating effects that could be useful in the clinic, especially in treating inflammatory and autoimmune disorders. As initially postulated, phages present in the human body (especially in the intestinal tract) could dampen exacerbated immune reactions and inflammatory responses and—by translocation from the gut to other tissues—contribute to maintenance of immune homeostasis at the level of both the intestines and other tissues (“natural phage therapy”) (21, 22). This hypothesis was confirmed by Barr (23), who demonstrated that phages adhering to mucus protect the underlying epithelium from bacterial infection. Recently, we suggested that in fact phages could not only protect gut epithelium from bacterial invasion but also interact with the epithelial cells to directly inhibit inflammation (e.g., through dampening ROS production by those cells and/or infiltrating neutrophils) (24). Previously, we have shown that homologous (but not heterologous) phages inhibit ROS production in human neutrophils and monocytes induced by E. coli; phages themselves induce only minimal ROS production in phagocytes (25). This phenomenon was later confirmed and extended, revealing that phages inhibit ROS induced in neutrophils not only by bacteria but also by endotoxin (26), while the inability of T4 phage and its capsid proteins to induce ROS has been confirmed by Miernikiewicz et al. (27). Importantly, phages do not induce neutrophil degranulation (28). Phages and some of their capsid proteins have also been demonstrated to inhibit inflammatory skin and organ infiltrates in experimental mice, skin allograft rejection in mice, and autoimmune reaction using a mouse model of rheumatoid arthritis (2931). Recently, Van Belleghem et al. demonstrated a predominantly anti-inflammatory effect of phages as evaluated by their ability to influence expression and production of cytokines and specific receptors of human mononuclear cells (32). This correlates with laboratory data derived from patients on PT indicating that the therapy significantly dampens C-reactive protein (CRP) and the sedimentation rate in patients (33); in fact, in some patients, a reduction in CRP may occur shortly after the onset of therapy even though eradication of infection has not been achieved (20). Moreover, reduction of proteinuria may take place in those patients (34); as proteinuria may be considered another marker of inflammation, this finding provides another argument for anti-inflammatory action of phages (35, 36).

We postulated that intestinal phages can translocate from the gut into the blood and other organs and tissues, mediating immunomodulatory activities which may be relevant for maintenance of immune homeostasis (“natural phage therapy”) (21, 22). These assumptions have recently been confirmed by other groups. It has been demonstrated that phages are transcytosed across epithelial cell layers (>30 billion phages daily). In line with those observations, Lehti et al. (37) have shown that phages can bind and penetrate into neuroblastoma cells in vitro and persist inside human cells up to 1 day without phage entry into the nucleus and without affecting cell viability. Those data strongly suggest that “natural phage therapy” (encompassing not only anti-bacterial activities of phages but also their anti-inflammatory and immunomodulating functions) may be translated into novel forms of clinical PT reaching well beyond current antimicrobial applications.

Potential of Phages to Mitigate the Severity of an Experimentally-Induced and Clinical aGvHD

As pointed out, ROS production by neutrophils is an important hallmark of GvHD. In this context, the ability of phages to dampen such ROS production provides a strong argument for their potential use in this setting, especially as PT does not impair human granulocyte and monocyte ability to kill standard strains and pathogens isolated from patients—thus, the therapy should not contribute to immunodeficiency in patients undergoing HCT. In fact, PT does not induce immunodeficiency and may be used in immunodeficient patients (38, 39). It should be noted that the reduction of oxidative stress may be beneficial in mice with aGvHD (40, 41). Furthermore, neutrophils can also exert beneficial effects in aGvHD when primed with G-CSF. Such neutrophils have diminished expression of MHC-II and co-stimulatory molecules, decreased IFN gamma production but high IL-10 production, consistent with “suppressor neutrophils” inducing donor Tregs responsible for aGvHD suppression in mice (42). The significant beneficial role of IL-10 in aGvHD is supported by data derived from cord blood (CD) transplantation and clinical observations indicating that CD recipients have lower incidence of aGvHD. CD contains an abundance of IL-10 producing B cells which suppress T cell proliferation and effector functions with the aid of that cytokine. In addition, there was a marked recovery of IL-10 producing B cells to levels found in healthy donors (43). These findings are supported by the data derived from patients following HCT in whom polymorphism in the IL-10 promoter region was found to have a significant effect on the outcome of transplantation. The authors believe that a high level of IL-10 production by the recipient’s cells during the early post-transplant period mitigates the alloantigen-induced immune response and GvHD-induced inflammation (44). In line with those observations, Chan et al. demonstrated that NK cells rapidly reconstitute following clinical HCT and produce IL-10, which may suppress alloimmune T cell-mediated responses (45). In a mouse model of aGvHD, IL-10 gene-modified dendritic cells induced transplant tolerance and suppressed aGvHD (46). IL-10 has also been shown to downregulate TLR-mediated human dendritic cell activation (47). In this context, the recent data of Van Belleghem et al. indicating that phage induce IL-10 production in human mononuclear cells appear to be of paramount importance (32). Previously, it has been reported that indeed phage preparations may induce IL-10 in those cells (38); in addition, phage films may induce IL-10, reducing inflammatory responses (48). However, it should also be kept in mind that IL-10 has been shown to support the growth of cytotoxic T cells and clinical studies in patients with Crohn’s disease failed to show its efficacy (49).

As already mentioned, TLR activation enhances neutrophil infiltration of GvHD lesions. Most studies confirm the role of lipopolysaccharide (LPS) and TLR4 in the development of aGvHD. In experimental bone marrow transplantation in mice, LPS antagonism reduces aGvHD while preserving graft-versus-leukemia activity (50). TLR knockout mice have attenuated aGvHD (51). Heparan sulfate, a TLR4 agonist, promotes aGvHD following bone marrow transplantation in mice (52). Although there are also data suggesting that in some experimental models TLR4 signaling may not be not absolutely required for the development of GvHD, the existing data suggest that targeting LPS and TLR4 signaling is a promising area to search for new GvHD treatment modalities (53). Phages may interfere with LPS and reduce its pro-inflammatory effects (including reduction of organ and tissue infiltration) (54) and dampen TLR expression (32).

Blockade of IL-1 reduces inflammation, and therefore this area of research is of obvious interest in studies on novel GvHD treatment modalities. IL-1 blockade indeed reduces aGvHD development, while IL-1 receptor deficiency alleviates murine aGvHD (55, 56). The donor genotype for the IL-1 receptor antagonist (IL-1Ra) polymorphism has a clear protective role against aGvHD in the clinic (57). While a clinical trial involving IL-1Ra did not show its effectiveness in preventing aGvHD, the authors believe that the agent was discontinued too early or different schedules or dosing would give different results (58). Moreover, the level of IL-1Ra was found to be markedly reduced in saliva from patients with cGvHD (59), which highlights IL-1Ra as a promising agent for future studies on its potential beneficial effect not only in aGvHD but also cGvHD. In this context, it should be noted that phages increase the production of this cytokine in human mononuclear cells (32).

Azithromycin (AZM) is active against a variety of bacteria and is also an anti-inflammatory agent modulating the functions of dendritic cells, monocytes, and granulocytes, inhibiting the NF-kappaB pathway following LPS stimulation. It also stimulates production of IL-10 in vitro. In a mouse model of aGvHD, a short course of AZM suppressed aGvHD significantly, whereas lymphocyte functions were unchanged. The authors suggest that AZM may be a novel prophylactic agent for lethal GvHD (60). Interestingly, phages exhibit similar activities to AZM: they may eliminate bacteria (in contrast to AZM phages they act very selectively, so specific bacterial targeting allowing for elimination of a given species of bacterium is possible); in addition—as pointed out earlier—phages have immunomodulating and anti-inflammatory effects (including inhibition of NF-kappaB) (54). Therefore, phages may reduce skin, gut, and liver infiltration typical for aGvHD, inhibit ROS production important for immunopathology of aGvHD, and induce IL-10, alleviating this condition. Gastrointestinal manifestations of aGvHD are often severe and pose a significant therapeutic challenge, while immune-mediated damage to intestinal epithelial cells (IEC) is a hallmark of this condition (2). We postulate that phages may interact with IEC, protecting those cells from such damage, which could offer a new form of immunotherapy of inflammatory bowel diseases (IBD) (24). There are many similarities between IBD and aGvHD (61), so this concept might also be applicable in treating aGvHD.

Could PT be Potentially Beneficial in cGvHD as Well?

The immunopathologic mechanisms responsible for cGvHD are activated already at the time of HCT and include inflammation, cell-mediated and humoral immunity, aberrant immunoregulation, and fibrosis (62). Clinical manifestations of cGvHD are similar to autoimmune collagen diseases (63).

There is no specific prophylactic therapy of cGvHD. Corticosteroids remain the mainstay of therapy for established syndrome with immunosuppressants as second-line therapy. However, current regimens have limited efficacy and significant side effects; about one-third of patients treated at the best centers relapse or die (64). In this regard, it is interesting that our group has recently reported that PT may be beneficial in a mouse model of collagen-induced autoimmune syndrome (31). A review of other available data on anti-inflammatory and immunomodulating effects of phages presented in this article (for example, low levels of IL-1Ra in cGvHD and upregulation of that cytokine by phages) suggests that PT could also bring benefits to patients with cGvHD (perhaps as an adjunct therapy allowing for steroid sparing).

Conclusion

The perspective presented herein is speculative, but may stimulate new directions of research that could result in a significant progress in transplantation. To obtain some preliminary data, a clinical trial of PT in IBD would be prudent before initiating such trials in immunosuppressed patients with aGvHD. Selection of most appropriate phage(s) should be based on available information on its immunomodulating activities that could be instrumental in alleviating the immunopathology of GvHD. As stated earlier, data supporting the potential applicability of T4 phages has been supplied by our group, while the data of Van Belleghem et al. (32) also suggest the potential of Staphylococcus aureus and Pseudomonas phages. It may well be that the optimal immunomodulatory effects could be mediated by a cocktail of phages targeting different immune cell receptors and cytokines. Such monovalent or polyvalent phage preparations could be produced using standard technology currently used for production of anti-bacterial phage preparations (65, 66).

aGvHD usually occurs within 100 days after HCT; therefore, a clinical trial should cover that initial period following transplantation as aGvHD prophylaxis. Such a trial should involve patients who received HCT for correction of hemopoietic deficiency (e.g., aplastic anemia) rather than patients with leukemia (to avoid serious complications like recurrence of malignancy).

aGvHD prophylaxis with cyclosporine is used in the majority of patients. Despite this prophylaxis, aGvHD may still evolve (1). Therefore, in addition to cyclosporine, phages could be added. Oral phage administration would be preferred as the intestinal tract is the common site of GvHD-related immunopathology; furthermore, such administration usually does not induce significant anti-phage antibody production while phages can penetrate the gut barrier and act also at other tissue sites (22, 23). Detailed description of PT protocol has been provided earlier (34).

Viruses provide balance to the holobiont, keeping the host and associated prokaryotes and eukaryotes functioning together as a unit (67). If indeed our endogenous phages—referred to as “bacteriophage guests”—protect human health (68), then PT using exogenous phages (which may be considered as a form of transplantation) should produce similar beneficial effects. PT in HCT may help combat antibiotic-resistant infections in those patients, enable selective decontamination and therapy-oriented manipulation of the gut microbiome, and attenuate inflammation and aberrant immunity. Therefore, PT offers new tools for personalized, preemptive, and therapeutic strategies to improve HCT results, which should be explored further.

Author Contributions

AG drafted the main part of the manuscript; EJ-M, RM, BW-D, and JB contributed parts of the manuscript; all authors approved the manuscript.

Conflict of Interest Statement

AG, RM, BW-D, and JB are co-inventors of patents owned by the Institute of Immunology and Experimental Therapy, Wrocław, covering phage preparations. The remaining author declares that they have no conflicts of interest.

Funding

The publication was supported by Wroclaw Centre of Biotechnology, programme The Leading National Research Centre (KNOW) for years 2014–2018 and statutory funds from Warsaw Medical University.

References

1. Zeiser R, Blazar BR. Acute graft-versus-host disease – biologic process, prevention, and therapy. N Engl J Med (2017) 377:2167–79. doi:10.1056/NEJMra1609337

CrossRef Full Text | Google Scholar

2. Naymagon S, Naymagon L, Wong SY, Ko HM, Renteria A, Levine J, et al. Acute graft-versus-host disease of the gut: considerations for the gastroenterologist. Nat Rev Gastroenterol Hepatol (2017) 14:711–26. doi:10.1038/nrgastro.2017.126

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Maldonado MS, RamírezVillanueva P, BertínCortes-Monroy P, Jara Arias V, SotoDonoso K, Uribe Gonzalez P, et al. Compassionate use of ruxolitinib in acute and chronic graft versus host disease refractory both to corticosteroids and extracorporeal photopheresis. Exp Hematol Oncol (2017) 6:32. doi:10.1186/s40164-017-0092-3

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Schwab L, Goroncy L, Palaniyandi S, Gautam S, Triantafyllopoulou A, Mocsai A, et al. Neutrophil granulocytes recruited upon translocation of intestinal bacteria enhance graft-versus-host disease via tissue damage. Nat Med (2014) 20:648–54. doi:10.1038/nm.3517

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Socie G, Mary JY, Lemann M, Daneshpouy M, Guardiola P, Meignin V, et al. Prognostic value of apoptotic cells and infiltrating neutrophils in graft-versus-host disease of the gastrointestinal tract in humans: TNF and Fas expression. Blood (2004) 103:50–7. doi:10.1182/blood-2003-03-0909

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Güngör T, Teira P, Slatter M, Stussi G, Stepensky P, Moshous D, et al. Reduced-intensity conditioning and HLA-matched haemopoietic stem-cell transplantation in patients with chronic granulomatous disease: a prospective multicentre study. Lancet (2014) 383:436–48. doi:10.1016/S0140-6736(13)62069-3

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Lin A, Lore K. Granulocytes: new members of the antigen-presenting cell family. Front Immunol (2017) 8:1781. doi:10.3389/fimmu.2017.01781

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Staffas A, Burgos da Silva M, van den Brink MR. The intestinal microbiota in allogeneic hematopoietic cell transplant and graft-versus-host disease. Blood (2017) 129:927–33. doi:10.1182/blood-2016-09-691394

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Peled JU, Hanash AM, Jenq RR. Role of the intestinal mucosa in acute gastrointestinal GVHD. Blood (2016) 128:2395–402. doi:10.1182/blood-2016-06-716738

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Mancini N, Greco R, Pasciuta R, Barbanti MC, Pini G, Morrow OB, et al. Enteric microbiome markers as early predictors of clinical outcome in allogeneic hematopoietic stem cell transplant: results of a prospective study in adult patients. Open Forum Infect Dis (2017) 4:ofx215. doi:10.1093/ofid/ofx215

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Golob JL, Pergam SA, Srinivasan S, Fiedler TL, Liu C, Garcia K, et al. Stool microbiota at neutrophil recovery is predictive for severe acute graft vs host disease after hematopoietic cell transplantation. Clin Infect Dis (2017) 65:1984–91. doi:10.1093/cid/cix699

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Yoshioka K, Kakihana K, Doki N, Ohashi K. Gut microbiota and acute graft-versus-host disease. Pharmacol Res (2017) 122:90–5. doi:10.1016/j.phrs.2017.05.028

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Kakihana K, Fujioka Y, Suda W, Najima Y, Kuwata G, Sasajima S, et al. Fecal microbiota transplantation for patients with steroid-resistant acute graft-versus-host disease of the gut. Blood (2016) 28:2083–8. doi:10.1182/blood-2016-05-717652

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Ott SJ, Waetzig GH, Rehman A, Moltzau-Anderson J, Bharti R, Grasis JA, et al. Efficacy of sterile fecal filtrate transfer for treating patients with Clostridium difficile infection. Gastroenterology (2017) 152:799–811.e7. doi:10.1053/j.gastro.2016.11.010

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Zuo T, Wong SH, Lam K, Lui R, Cheung K, Tang W, et al. Bacteriophage transfer during faecal microbiota transplantation in Clostridium difficile infection is associated with treatment outcome. Gut (2017) 67:634–43. doi:10.1136/gutjnl-2017-313952

CrossRef Full Text | Google Scholar

16. Manrique P, Dills M, Young MJ. The human gut phage community and its implications for health and disease. Viruses (2017) 9:141. doi:10.3390/v9060141

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Legoff J, Resche-Rigon M, Bouquet J, Robin M, Naccache SN, Mercier-Delarue S, et al. The eukaryotic gut virome in hematopoietic stem cell transplantation: new clues in enteric graft-versus-host disease. Nat Med (2017) 23:1080–5. doi:10.1038/nm.4380

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Watts G. Phage therapy: revival of the bygone antimicrobial. Lancet (2017) 390:2539–40. doi:10.1016/S0140-6736(17)33249-X

CrossRef Full Text | Google Scholar

19. Lyon J. Phage therapy’s role in combating antibiotic-resistant pathogens. JAMA (2017) 318:1746–8. doi:10.1001/jama.2017.12938

CrossRef Full Text | Google Scholar

20. Górski A, Międzybrodzki R, Weber-Dabrowska B, Fortuna W, Letkiewicz S, Rogóż P, et al. Phage therapy: combating infections with potential for evolving from merely a treatment for complications to targeting diseases. Front Microbiol (2016) 7:1515. doi:10.3389/fmicb.2016.01515

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Górski A, Weber-Dabrowska B. The potential role of endogenous bacteriophages in controlling invading pathogens. Cell Mol Life Sci (2005) 62:511–9. doi:10.1007/s00018-004-4403-6

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Górski A, Ważna E, Weber-Dabrowska B, Dabrowska K, Świtała-Jeleń K, Międzybrodzki R. Bacteriophage translocation. FEMS Immunol Med Microbiol (2006) 46:313–9. doi:10.1111/j.1574-695X.2006.00044.x

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Barr JJ. A bacteriophage journey through the human body. Immunol Rev (2017) 279:106–22. doi:10.1111/imr.12565

CrossRef Full Text | Google Scholar

24. Górski A, Jończyk-Matysiak E, Łusiak-Szelachowska M, Międzybrodzki R, Weber-Dabrowska B, Borysowski J. Bacteriophages targeting intestinal epithelial cells: a potential novel form of immunotherapy. Cell Mol Life Sci (2018) 75:589–95.

Google Scholar

25. Przerwa A, Zimecki M, Świtała-Jeleń K, Dabrowska K, Krawczyk E, Łuczak M, et al. Effects of bacteriophages on free radical production and phagocytic functions. Med Microbiol Immunol (2006) 195:143–50. doi:10.1007/s00430-006-0011-4

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Międzybrodzki R, Świtala-Jeleń K, Fortuna W, Weber-Dabrowska B, Przerwa A, Łusiak-Szelachowska M, et al. Bacteriophage preparation inhibition of reactive oxygen species generation by endotoxin-stimulated polymorphonuclear leukocytes. Virus Res (2008) 131:233–42. doi:10.1016/j.virusres.2007.09.013

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Miernikiewicz P, Dabrowska K, Piotrowicz A, Owczarek B, Wojas-Turek J, Kicielińska J, et al. T4 phage and its head surface proteins do not stimulate inflammatory mediator production. PLoS One (2013) 8:e71036. doi:10.1371/journal.pone.0071036

CrossRef Full Text | Google Scholar

28. Borysowski J, Międzybrodzki R, Wierzbicki P, Kłosowska D, Korczak-Kowalska G, Weber-Dabrowska B, et al. A3R phages and Staphylococcus aureus lysate do not induce neutrophil degranulation. Viruses (2017) 9:iiE36. doi:10.3390/v9020036

CrossRef Full Text | Google Scholar

29. Górski A, Kniotek M, Perkowska-Ptasińska A, Mróz A, Przerwa A, Gorczyca W. Bacteriophages and transplantation tolerance. Transplant Proc (2006) 38:331–3. doi:10.1016/j.transproceed.2005.12.073

CrossRef Full Text | Google Scholar

30. Miernikiewicz P, Kłopot A, Soluch R, Szkuta P, Kęska W, Hodyra-Stefaniak K, et al. T4 Phage tail adhesin Gp12 counteracts LPS-induced inflammation in vivo. Front Microbiol (2016) 7:1112. doi:10.3389/fmicb.2016.01112

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Międzybrodzki R, Borysowski J, Kłak M, Jończyk-Matysiak E, Obmińska-Mrukowicz B, Suszko A, et al. In vivo studies on the influence of bacteriophage preparations on the autoimmune inflammatory process. Biomed Res Int (2017) 2017:3612015. doi:10.1155/2017/3612015

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Van Belleghem JD, Clement F, Merabishvili M, Lavigne R, Vaneechoutte M. Pro- and anti-inflammatory responses of peripheral blood mononuclear cells induced by Staphylococcus aureus and Pseudomonas aeruginosa phages. Sci Rep (2017) 7:8004. doi:10.1038/s41598-017-08336-9

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Międzybrodzki R, Fortuna W, Weber-Dabrowska B, Górski A. A retrospective analysis of changes in inflammatory markers in patients treated with bacterial viruses. Clin Exp Med (2009) 9:303–12. doi:10.1007/s10238-009-0044-2

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Międzybrodzki R, Borysowski J, Weber-Dabrowska B, Fortuna W, Letkiewicz S, Szufnarowski K, et al. Clinical aspects of phage therapy. Adv Virus Res (2012) 83:73–121. doi:10.1016/B978-0-12-394438-2.00003-7

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Paisley KE, Beaman M, Tooke JE, Mohamed-Ali V, Lowe GD, Shore AC. Endothelial dysfunction and inflammation in asymptomatic proteinuria. Kidney Int (2003) 63:624–33. doi:10.1046/j.1523-1755.2003.00768.x

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Trimarchi H. Role of aliskiren in blood pressure control and renoprotection. Int J Nephrol Renovasc Dis (2011) 4:41–8. doi:10.2147/IJNRD.S6653

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Lehti TA, Pajunen MI, Skog MS, Finne J. Internalization of a polysialic acid-binding Escherichia coli bacteriophage into eukaryotic neuroblastoma cells. Nat Commun (2017) 8:1915. doi:10.1038/s41467-017-02057-3

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Górski A, Międzybrodzki R, Borysowski J, Dabrowska K, Wierzbicki P, Ohams M, et al. Phage as a modulator of immune responses: practical implications for phage therapy. Adv Virus Res (2012) 83:41–71. doi:10.1016/B978-0-12-394438-2.00002-5

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Borysowski J, Górski A. Is phage therapy acceptable in the immune compromised host. Int J Infect Dis (2008) 12:466–71. doi:10.1016/j.ijid.2008.01.006

CrossRef Full Text | Google Scholar

40. Amer J, Weiss L, Reich S, Shapira MY, Slavin S, Fibach E. The oxidative status of blood cells in a murine model of graft-versus-host disease. Ann Hematol (2007) 86:753–8. doi:10.1007/s00277-007-0321-7

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Im KI, Kim N, Lim JY, Nam YS, Lee ES, Kim EJ, et al. The free radical scavenger NecroX-7 attenuates acute graft-versus-host disease via reciprocal regulation of Th1/regulatory T cells and inhibition of HMGB1 release. J Immunol (2015) 194:5223–32. doi:10.4049/jimmunol.1402609

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Perobelli SM, Mercadante AC, Galvani RG, Gonçalves-Silva T, Alves AP, Pereira-Neves A, et al. G-CSF-induced suppressor IL-10+ neutrophils promote regulatory T cells that inhibit graft-versus-host disease in a long-lasting and specific way. J Immunol (2016) 197:3725–34. doi:10.4049/jimmunol.1502023

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Sarvaria A, Basar R, Mehta RS, Shaim H, Muftuoglu M, Khoder A, et al. IL-10+ regulatory B cells are enriched in cord blood and may protect against cGVHD after cord blood transplantation. Blood (2016) 128:1346–61. doi:10.1182/blood-2016-01-695122

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Lin MT, Storer B, Martin PJ, Tseng LH, Gooley T, Chen PJ, et al. Relation of an interleukin-10 promoter polymorphism to graft-versus-host disease and survival after hematopoietic-cell transplantation. N Engl J Med (2003) 34:2201–10. doi:10.1056/NEJMoa022060

CrossRef Full Text | Google Scholar

45. Chan YLT, Zuo J, Inman C, Croft W, Begum J, Croudace J, et al. NK cells produce high levels of IL-10 early after allogeneic stem cell transplantation and suppress development of acute GVHD. Eur J Immunol (2017) 48:316–29. doi:10.1002/eji.201747134

CrossRef Full Text | Google Scholar

46. Wan J, Huang F, Hao S, Hu W, Liu Ch, Zhang W, et al. Interleukin-10 gene-modified dendritic cell-induced type 1 regulatory T cells induce transplant-tolerance and impede graft versus host disease after allogeneic stem cell transplantation. Cell Physiol Biochem (2017) 43:353–66. doi:10.1159/000480415

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Brencicova E, Jagger AL, Evans HG, Georgouli M, Laios A, Attard Montalto S, et al. Interleukin-10 and prostaglandin E2 have complementary but distinct suppressive effects on toll-like receptor-mediated dendritic cell activation in ovarian carcinoma. PLoS One (2017) 12:e0175712. doi:10.1371/journal.pone.0175712

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Sun Y, Li Y, Wu B, Wang J, Lu X, Qu S, et al. Biological responses to M13 bacteriophage modified titanium surfaces in vitro. Acta Biomater (2017) 58:527–38. doi:10.1016/j.actbio.2017.06.019

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Herfarth H, Schölmerich J. IL-10 therapy in Crohn’s disease: at the crossroads. Treatment of Crohn’s disease with the anti-inflammatory cytokine interleukin 10. Gut (2002) 50:146–7. doi:10.1136/gut.50.2.146

CrossRef Full Text | Google Scholar

50. Cooke KR, Gerbitz A, Crawford JM, Teshima T, Hill GR, Tesolin A, et al. LPS antagonism reduces graft-versus-host disease and preserves graft-versus-leukemia activity after experimental bone marrow transplantation. J Clin Invest (2001) 107:1581–9. doi:10.1172/JCI12156

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Zhao Y, Liu Q, Yang L, He D, Wang L, Tian J, et al. TLR4 inactivation protects from graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Cell Mol Immunol (2013) 10:165–75. doi:10.1038/cmi.2012.58

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Brennan TV, Lin L, Huang X, Cardona DM, Li Z, Dredge K, et al. Heparan sulfate, an endogenous TLR4 agonist, promotes acute GVHD after allogeneic stem cell transplantation. Blood (2012) 120:2899–908. doi:10.1182/blood-2011-07-368720

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Heidegger S, van den Brink MRM, Haas T, Poeck H. The role of pattern-recognition receptors in graft-versus-host disease and graft-versus-leukemia after allogeneic stem cell transplantation. Front Immunol (2014) 5:337. doi:10.3389/fimmu.2014.00337

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Górski A, Dabrowska K, Międzybrodzki R, Weber-Dabrowska B, Łusiak-Szelachowska M, Jończyk-Matysiak E, et al. Phages and immunomodulation. Future Microbiol (2017) 12:905–14. doi:10.2217/fmb-2017-0049

CrossRef Full Text | Google Scholar

55. Liang Y, Ma S, Zhang Y, Wang Y, Cheng Q, Wu Y, et al. IL-1β and TLR4 signaling are involved in the aggravated murine acute graft-versus-host disease caused by delayed bortezomib administration. J Immunol (2014) 192:1277–85. doi:10.4049/jimmunol.1203428

CrossRef Full Text | Google Scholar

56. Jankovic D, Ganesan J, Bscheider M, Stickel N, Weber FC, Guarda G, et al. The Nlrp3 inflammasome regulates acute graft-versus-host disease. J Exp Med (2013) 210:1899–910. doi:10.1084/jem.20130084

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Cullup H, Dickinson AM, Jackson GH, Taylor PR, Cavet J, Middleton PG. Donor interleukin 1 receptor antagonist genotype associated with acute graft-versus-host disease in human leucocyte antigen-matched sibling allogeneic transplants. Br J Haematol (2001) 113:807–13. doi:10.1046/j.1365-2141.2001.02811.x

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Antin JH, Weisdorf D, Neuberg D. Interleukin-1 blockade does not prevent acute graft-versus-host disease: results of a randomized, double-blind, placebo-controlled trial of interleukin-1 receptor antagonist in allogeneic bone marrow transplantation. Blood (2002) 100:3479–82. doi:10.1182/blood-2002-03-0985

CrossRef Full Text | Google Scholar

59. Devic I, Shi M, Schubert MM, Lloid M, Izutsu KT, Pan C, et al. Proteomic analysis of saliva from patients with oral chronic graft-versus-host disease. Biol Blood Marrow Transplant (2014) 20:1048–55. doi:10.1016/j.bbmt.2014.03.031

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Iwamoto S, Azuma E, Kumamoto T, Hirayama M, Yoshida T, Ito M, et al. Efficacy of azithromycin in preventing lethal graft-versus-host disease. Clin Exp Immunol (2013) 171:338–45. doi:10.1111/cei.12023

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Nalle SC, Turner JR. Intestinal barrier loss as a critical pathogenic link between inflammatory bowel disease and graft-versus-host disease. Mucosal Immunol (2015) 8:720–30. doi:10.1038/mi.2015.40

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Flowers ME, Martin PJ. How we treat chronic graft-versus-host disease. Blood (2015) 125:606–15. doi:10.1182/blood-2014-08-551994

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Ratanatharathorn V, Ayash L, Lazarus HM, Fu J, Uberti JP. Chronic graft-versus-host disease: clinical manifestation and therapy. Bone Marrow Transplant (2001) 28:121–9. doi:10.1038/sj.bmt.1703111

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Lee SJ, Nguyen TD, Onstad L, Bar M, Krakow EF, Salit RB, et al. Success of immunosuppressive treatments in patients with chronic graft-versus-host disease. Biol Blood Marrow Transplant (2018) 24:555–62. doi:10.1016/j.bbmt.2017.10.042

CrossRef Full Text | Google Scholar

65. Pirnay JP, Verbeken G, Ceyssens PJ, Huys I, De Vos D, Ameloot C, et al. The magistral phage. Viruses (2018) 10:64. doi:10.3390/v10020064

CrossRef Full Text | Google Scholar

66. Pirnay JP, Merabishvili M, Van Raemdonck H, De Vos D, Verbeken G. Bacteriophage production in compliance with regulatory requirements. In: Azeredo J, Sillankorva S, editors. Bacteriophage Therapy. Methods in Molecular Biology. Vol 1693. New York, NY: Humana Press (2018). p. 233–52.

Google Scholar

67. Grasis JA. The intra-dependence of viruses and the holobiont. Front Immunol (2017) 8:1501. doi:10.3389/fimmu.2017.01501

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Guglielmi G. Do bacteriophage guests protect human health? Science (2017) 358:982–3. doi:10.1126/science.358.6366.982

CrossRef Full Text | Google Scholar

Keywords: GvHD, hematopoietic cell transplantation, immunomodulation, inflammation, phage, phage therapy

Citation: Górski A, Jończyk-Matysiak E, Międzybrodzki R, Weber-Dąbrowska B and Borysowski J (2018) “Phage Transplantation in Allotransplantation”: Possible Treatment in Graft-Versus-Host Disease? Front. Immunol. 9:941. doi: 10.3389/fimmu.2018.00941

Received: 20 December 2017; Accepted: 16 April 2018;
Published: 27 April 2018

Edited by:

Hans-Jochem Kolb, Kolb Consulting UG, Germany

Reviewed by:

Thomas Luft, Universitätsklinikum Heidelberg, Germany
Benedetto Bruno, Università degli Studi di Torino, Italy

Copyright: © 2018 Górski, Jończyk-Matysiak, Międzybrodzki, Weber-Dąbrowska and Borysowski. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Andrzej Górski, agorski@ikp.pl

This article is dedicated to John A. Hansen, M.D., Professor of Medicine, Fred Hutchinson Cancer Research Center and University of Washington, Seattle, a great physician and scientist, on the occasion of his 75th birthday.

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.