The therapeutic GVL effect after allogeneic stem cell transplantation is limited by the development of GVHD. The ultimate aim of current research is to separate the 2 processes in a meaningful fashion. The IFNs are a pleiotropic group of cytokines that were originally recognized because of their ability to interfere with viral replication. However, it is now established that these cytokines play an important role in orchestrating both innate and adaptive immunity. Multiple studies have investigated the effects of both types I and II IFN on GVHD and GVL in preclinical transplant models. The results indicate variable effects that are dependent on the period of activity within the developing immune response, the presence and type of pretransplant conditioning and the differential mechanisms, and IFN sensitivity of immune pathology within individual target organs during GVHD. This Perspective discusses the current literature on the IFNs and their potential modulation within clinical transplantation, focusing particularly on enhancing the therapeutic GVL effects.

Stem cell transplantation (SCT) has been routinely used as curative therapy for hematologic malignancies for more than 3 decades. Although autologous SCT is now principally used as therapy for myeloma, non-Hodgkin lymphoma, and Hodgkin lymphoma, allogeneic SCT is more widely used to treat acute and chronic leukemia, myelodysplasia, and advanced lymphoid malignancies failing other modalities. Allogeneic SCT is limited by the consequences of donor immune activation in response to recipient alloantigens, leading to the development of GVHD. GVHD occurs in the majority of recipients of unmanipulated (T cell–replete) grafts1  and may develop early (in the first 100 days) after SCT (acute) or thereafter (chronic), typically with divergent presentations. The therapeutic potential of stem cell transplantation lies within the GVL effect whereby residual recipient-type malignancy is eradicated by donor NK cells and/or T cells. GVHD and GVL responses are closely related immune-mediated processes and are controlled in large part by cytokines. Our understanding of the IFNs in GVHD and GVL has greatly expanded in the last 5 years, opening the possibility of manipulation for therapeutic gain.

Acute GVHD is initiated by recipient tissue inflammation in response to the transplant conditioning or preparative regimen, used typically to eradicate malignancy and inhibit rejection of the transplanted allodisparate graft. This results in production of proinflammatory cytokines, typically IL-1, IL-6, and TNF.2  The resulting inflammatory environment leads to activation of recipient cells capable of presenting alloantigen to incoming donor T cells. These antigen-presenting cells (APCs) can include both hematopoietic and nonhematopoietic cells.3,4  Acute GVHD is absolutely dependent on this interaction between APC and donor αβ T cells.5  Although T-cell depletion protects from GVHD, it can also increase graft rejection, inhibit GVL effects, and delay immune reconstitution. Consequently, recipients of allogeneic SCT have a lower risk of relapse than those of syngeneic SCT or T cell–depleted allogeneic SCT.6  In murine models where donor and recipients are MHC mismatched, both CD4+ and CD8+ T cells contribute to GVHD, although MHC class II responses dominate. In clinical patients where donors and recipients are predominantly HLA matched, GVHD is mediated by both CD4+ and CD8+ T cells, but MHC class I responses tend to dominate. The final effector phase of GVHD involves the infiltration and damage of target tissues by alloreactive cells. Although the expression of alloantigens in GVHD is not tissue specific, acute GVHD tends to occur predictably in particular organs (ie, gastrointestinal [GI] tract, skin, liver, and lung). This effect is multifactorial but is related to the differential presence of functional APCs,3  the induction of tissue-homing specific integrins on donor T cells,7  organ-specific inflammation,8  and, importantly, the differential effects of inflammatory cytokines on target tissues of which IFN is the archetype. In conjunction with cytotoxic T cell–mediated host tissue damage, the proinflammatory cytokines IFN-γ and TNF are also major contributors to GVHD. It is the tissue damage resulting from both direct T-cell cytotoxicity and proinflammatory cytokine responses that results in the clinical manifestations of GVHD. The most commonly affected organs after acute onset of GVHD are skin (81% of patients), GI tract (54%), and liver (50%).9  Epithelial apoptosis in these organs results in a loss of function and subsequent clinical symptoms, such as diarrhea, cholestatic hyperbilirubinemia, and maculopapular skin rashes.10 

Therapeutic GVL responses are mediated by donor NK cells and T cells, and the antigenic targets are principally minor histocompatibility antigens (mHAs), although hematopoietic-specific and leukemia-specific responses also occur.11  Recipient mHAs are predominantly presented directly to donor CD8+ T cells by recipient hematopoietic-derived APCs within the first few weeks after transplantation, before their replacement by donor-derived APCs.12  These donor-derived APCs must subsequently cross-present recipient mHAs to donor CD8+ T cells. Both CD4+ and CD8+ T cells are important in GVL responses, with CD8+ T cells mediating direct cytotoxicity against leukemic cells, whereas CD4+ T cells are capable of both direct cytotoxicity (if the leukemia is MHC class II positive) and providing helper function to donor CD8+ T cells (via cytokines, such as IL-2 and the licensing of APCs). Importantly, recipient rather than donor APCs are most important for the induction of T-cell responses mediating both GVHD and GVL.3,13  After recognition of target antigen on leukemia cells, cytotoxic effector populations can mediate damage via multiple different pathways, including secretion of perforin and granzymes or through Fas-FasL, TNF, and/or TRAIL.14  Although T cells deficient in both Fas ligand and perforin have been demonstrated to exert GVL activity in vivo,15  these molecules are clearly required for meaningful GVL activity.16 

The generation of GVHD and an effective GVL response is thus a complex process requiring the interaction of an APC presenting relevant mHAs with a donor T cell and subsequent differentiation and acquisition of effector function. The IFNs are key molecules in orchestrating this immune response.

The IFNs were discovered by Issaacs and Lindenmann in 1957 when they were investigating viral interference, and they have subsequently been defined by this ability.17  It was not until 1978 that they were able to be purified, analyzed, and characterized.18  They are recognized as a key component of innate immunity and the first line of defense against viral infection. There are 3 distinct types (type I, type II, and type III), which are distinguished based on their structure, cognate receptors, and biologic activities. The type I IFNs are well characterized and are the hallmark IFNs important in defense against viral infection. In contrast, the type II IFNs are better known for mediating immune responses to a broad range of intracellular pathogens. The type III IFNs are more recently described and not well characterized; however, they have similar downstream effects as type I IFNs. Therefore, although IFNs were initially described because of their antiviral properties, they exhibit multiple distinct additional immunologic properties.

Type I IFN

The type I IFNs encompass a large family of cytokines that include a single IFN-β isotype, more than 13 IFN-α isotypes, and multiple other less-described subtypes.19  Most cell types can be induced to secrete type I IFN in response to viral infection. The major pathway through which this occurs is activation of the cytosolic receptors retinoic acid-inducible gene I and melanoma differentiation-associated gene 5 by double-stranded RNA.20  Type I IFN production is induced after activation of 5 of the 11 TLRs.21  However, the most potent producers are plasmacytoid dendritic cells (DCs),22  which preferentially express TLR7 and TLR9 that recognize single-stranded RNA and CpG-rich DNA, respectively.23  After engagement of these receptors, the MyD88-IFN regulatory protein (IRF7) pathway is crucial for type I IFN production, with extended retainment of CpG and MyD88/IRF7 complexes in endosomal vesicles responsible for the ability of plasmacytoid DCs to induce high levels of type I IFN.24  All type I IFNs signal through the same receptor, which is composed of 2 subunits, IFNAR1 and IFNAR2.25  Importantly, the mouse has a type I IFN system comparable with that in humans, with multiple cytokine subsets and both IFNAR1 and IFNAR2 components of the receptor,25  which is expressed on essentially all cells.26  After signaling through the type I IFN receptor, downstream responses are transmitted through the kinases tyrosine kinase 2 and JAK1, which recruits STAT1 to form the STAT1-STAT2 heterodimer complex that dissociates and migrates into the nucleus.19  In the nucleus, this heterodimer associates with IFN regulatory factor 9 to form the IFN-stimulated gene factor 3 that binds to the IFN-stimulated response element promoter sequence to activate transcription of the IFN-inducible genes.19 

Type I IFN signaling is important in both the innate and adaptive immune responses. These effects were initially studied in the context of viral infection, with signaling resulting in antiproliferative responses in conjunction with up-regulation of MHC class I expression.27  During the adaptive immune response, type I IFN signaling is vital for the expansion and differentiation of effector cytotoxic T lymphocytes (CTLs).28  The injection of synthetic dsRNA (polyinosinic acid/polycytidylic acid) to induce type I IFN augments the response of TCR transgenic CD8+ T cells to directly presented antigen. Increased levels of type I IFN have also been shown to promote cross-presentation of exogenous recipient mHAs to CD8+ T cells by APCs, a process that is independent of CD4+ T-cell help.29  Bystander activation of T cells can also occur in the presence of high levels of type I IFN, as proliferation is observed without up-regulation of CD69 and CD25.30  More recently, there has been renewed interest in the role of type I IFN in immunoediting and tumor clearance. Early NK antitumor responses are dependent on type I IFN signaling.31  Homeostatic numbers of NK cells in mice with a deficiency of both the AR1 and AR2 components of the IFNAR are reduced, but the cytotoxicity activity of naive NK cells is similar.31  However, IL-2–mediated activation and subsequent killing are defective in mice lacking the type I IFN receptor.31  This suggests that, although type I IFN signaling does not affect the baseline activity of NK cells, it serves to enhance antitumor responses after activation. Type I IFN signaling is also important for CD8 T-cell responses against tumor. The induction of type I IFN with CpG via the TLR9 receptor in combination with DC-targeted vaccines effectively activates tumor-specific CD8+ T cells, leading to prolonged survival in tumor models, and increased serum levels of IFN-α are observed.32  Further evidence for the importance of type I IFN in antitumor function demonstrates that they play a role in immunoediting.33  IFNAR1-deficient mice are more susceptible to induced sarcomas, and tumors arising in these mice have an “uneditited” phenotype, as they are rapidly rejected via a T cell–dependent mechanism when transplanted into wild-type mice.33  Interestingly, tumor rejection occurs via effects on hematopoietic cells, rather than via direct effects on tumor cells.33  Therefore, type I IFN contributes to both innate and adaptive arms of the antitumor immune response.

In conjunction with effects observed through NK and CD8 T cells, type I IFNs also modulate DC function. Type I IFN promotes the maturation of DCs34  and enhances cross-presentation to CD8 T cells by CD8α+ DC and subsequent rejection of tumor.35  The mechanism by which IFN-β administration induces beneficial outcomes in multiple sclerosis patients has been investigated in experimental autoimmune encephalitis.36  These studies demonstrate that type I IFN signaling down-regulates the Th17-mediated inflammatory response by inhibiting osteopontin expression in DCs, resulting in derepression of the potent inhibitory cytokine IL-27.37,38  The ability of type I IFN signaling to reduce IL-17 production has also been documented in patients with ulcerative colitis, with treatment over a period of 12 weeks resulting in a significant decrease in IL-17A mRNA expression in colonic biopsies.39  Therefore, type I IFN signaling in DCs modulates subsequent cellular and cytokine differentiation responses.

IFN-α was routinely used to treat patients with chronic myeloid leukemia in the pre-tyrosine kinase inhibitor (ie, imatinib, dasatinib, nilotinab) era, with responses thought primarily to relate to antiproliferative effects.40,41  IFN-α administration is now undergoing a renaissance as combination therapy with tyrosine kinase inhibitors in an effort to promote leukemia stem cell cycling and render these cells sensitive to the latter agents. The finding that IFN-α administration in vivo induces the transition of HSCs from a dormant stage into active cell cycle was surprising, considering the antiproliferative activity observed in in vitro culture systems.42  The increased proliferation of HSCs results in increased susceptibility to elimination by antiproliferative chemotherapeutic drugs. Clinical studies investigating the effects of IFN-α administration after allogeneic SCT demonstrated enhancement of both GVHD and GVL responses.43,44  The effects of IFN-α and mechanism of disease control were recently addressed by our group in preclinical models. This study demonstrated differential effects on donor CD4+ and CD8+ T cells45  (Figure 1). Type I IFN signaling indirectly inhibits donor CD4+ T cells, putatively via recipient APCs, resulting in alleviation of GVHD. In contrast, signaling enhanced donor CD8+ T-cell responses and increased susceptibility of host cells to killing by donor CD8+ T cells, both promoting GVHD and improving protective GVL responses. A mild reduction in hepatic GVHD histopathology in the absence of type I IFN signaling has also been demonstrated in a system of GVHD directed toward minor histocompatibility antigens only, although in these studies the overall effects of type I IFN on GVHD outcomes was limited.46  Clinical data from the 1990s demonstrated a detrimental effect of type I IFN administration before transplantation, with increases in severe GVHD.47,48  This is consistent with MHC class I–restricted effects on GVHD in clinical transplantation where donor and recipients are HLA-matched. It was also recently demonstrated that type I IFN enhances the cross-presentation of exogenous recipient antigens by donor APCs within MHC class I after SCT.49  The contribution of cross-presentation to GVL responses was not investigated in our studies because of the models used being MHC mismatched, but this effect would be expected to enhance therapeutic GVL responses in patients.

Figure 1

The differential effects of type I IFN signaling on MHC class I and MHC class II–dependent GVHD and GVL responses. Signaling on host hematopoietic tissue decreases donor CD4 T-cell proliferation and differentiation via inhibitory effects on recipient APCs, resulting in reduced T-cell differentiation and GVHD of the colon. The inhibition of Th17 differentiation has been associated with inhibition of APC-derived osteopontin, which promotes IL-27 secretion. The signaling of donor APC by type I IFN promotes cross-presentation (ie, the presentation of exogenous recipient allogeneic peptides within MHC class I) and the expansion of donor CTLs. This effect is further amplified by direct signaling to donor NK and CD8 T cells, which together promote cytotoxicity (eg, mediated by perforin/granzyme) against GVHD target tissue and residual recipient leukemia (ie, GVL). Finally, the type I IFNs enhance the susceptibility of residual leukemia to CD8 and NK cell–mediated cytotoxicity, further amplifying GVL responses.

Figure 1

The differential effects of type I IFN signaling on MHC class I and MHC class II–dependent GVHD and GVL responses. Signaling on host hematopoietic tissue decreases donor CD4 T-cell proliferation and differentiation via inhibitory effects on recipient APCs, resulting in reduced T-cell differentiation and GVHD of the colon. The inhibition of Th17 differentiation has been associated with inhibition of APC-derived osteopontin, which promotes IL-27 secretion. The signaling of donor APC by type I IFN promotes cross-presentation (ie, the presentation of exogenous recipient allogeneic peptides within MHC class I) and the expansion of donor CTLs. This effect is further amplified by direct signaling to donor NK and CD8 T cells, which together promote cytotoxicity (eg, mediated by perforin/granzyme) against GVHD target tissue and residual recipient leukemia (ie, GVL). Finally, the type I IFNs enhance the susceptibility of residual leukemia to CD8 and NK cell–mediated cytotoxicity, further amplifying GVL responses.

Close modal

Although these data suggest there would be improved tumor clearance after treatment with IFN-α before transplantation, clinical SCT data in the chronic myeloid leukemia setting combined with our own data suggest that the detrimental effects of GVHD overwhelm this protective response. Because extensive data indicate that type I IFN signaling after SCT promotes GVL responses, the most effective therapeutic use of type I IFN is probably in SCT patients at a high risk of relapse. In this setting, type I IFN would be predicted to increase the sensitivity of malignancy to killing while enhancing the donor NK and CD8+ T-cell responses that mediate GVL. Whether newer pegylated versions of type I IFN will be more efficacious in this setting will be a critical issue.

Type II IFN

IFN-γ is the only type II IFN. It is produced by CD4+ Th1 lymphocytes, CD8+ T cells, NK cells, B cells, NKT cells, and APCs, with production stimulated by the secretion of IL-12 and IL-18, principally by APCs.50  IFN-γ signals through the IFN-γR, which is expressed on most cell types and consists of 2 subunits, IFN-γRα and IFN-γRβ.51  The type II IFN receptor interacts with JAK1 and JAK2 after ligand-dependent rearrangement of the receptor subunits, resulting in phosphorylation of STAT1. Homodimers of STAT1 form and translocate into the nucleus to bind IFN-γ–activated site elements and initiate transcription.51  A regulator of the JAK-STAT signaling pathway important for both type I and type II IFN signaling is the suppressor of cytokine signaling proteins, which inhibit signaling.52 

IFN-γ has diverse functions that are involved in both innate and adaptive immunity and host defense. During the innate immune response, IFN-γ is important for the activation of macrophages, resting NK cell activity, and up-regulation of MHC class II on the macrophage cell surface.53,54  Consequently, mice deficient in the IFN-γR demonstrate increased susceptibility to multiple intracellular pathogens.53,54  After initiation of the adaptive immune response, IFN-γ regulates Th1 differentiation and function and subsequently inhibits Th2 differentiation. Signaling directly on CD8 T cells stimulates development and increases abundance during acute viral infection.55  Paradoxically, IFN-γ has also been implicated in down-regulation of the immune responses and T-cell homeostasis, with signaling inducing apoptosis in activated CD456,57  and CD858  T cells. Furthermore, production of IFN-γ by alloreactive CD4+ Treg is important for their function, and generation of CD4+ Treg is impaired in IFN-γ–deficient mice.59  During the immunosurveillance phase of tumor immunity, IFN-γ protects against the development of both induced and spontaneous tumors in a variety of models.60  Studies indicate that γδ T cells are an important source of IFN-γ during antitumor responses.61  However, although increased innate immune cell recruitment, MHC class I expression on APCs, and increased Th1 development inhibit tumor growth, the more recently identified functions of IFN-γ–like enhancement of Treg development and decreased neutrophil recruitment may enhance tumor development.62  Although type I and type II IFNs exhibit distinct immunologic properties, interaction between their respective signaling pathways was recently demonstrated in the context of Listeria monocytogenes infection. This study demonstrated that type I IFN signaling in macrophages and DCs resulted in down-regulation of the IFN-γR and subsequent responsiveness to IFN-γ.63 

In the SCT setting, rapid IFN-γ release occurs by NK and NKT cells activated early after transplantation by interaction with activating or inhibitory killer inhibitory receptors. In the case of NKT cells, this cytokine secretion can occur within hours of TCR ligation by CD1 days loaded with glycolipid (relative to days for conventional T cells responding to peptide within MHC), and this appears critical for the licensing of recipient APCs and the subsequent control of GVHD and GVL effects.64,65  γδ T cells are also implicated in activation of host APCs via a cell contact– dependent mechanism, resulting in increased IFN-γ production by stimulated T cells and amplification of GVHD.66 

Early studies investigating the effects of IFN-γ production by the donor graft on transplant outcome yielded contradictory results. When donor cells unable to produce IFN-γ were transplanted into nonirradiated recipients, there was a delay in GVHD mortality, which was attributed to enhanced Th2 differentiation67  and impairment of CTL function68  relative to recipients of WT grafts. In contrast, enhanced GVHD was observed in lethally irradiated recipients of IFN-γ−/− grafts or after IFN-γ neutralization.69,70  Consistent with this, the administration of recombinant IFN-γ provided protection from GVHD in CD4-dependent systems.71  Mechanistic studies revealed that in this setting IFN-γ signaling reduced proliferation and IL-2 production from donor T cells in response to allogeneic APCs. A subsequent study demonstrated that, although IFN-γ signaling attenuates CD8-dependent GVHD, it enhances therapeutic GVL responses in the absence of CD4 T cells.72  These results suggest that recipient conditioning regimen alters the overall effect of IFN-γ signaling on recipient versus the donor graft. In addition, the location and timing of IFN-γ production may impact effects observed.73  Subsequently, studies investigating links between IFN-γ polymorphisms and GVHD development have also yielded conflicting results.74  Because of inconsistent data obtained using donors unable to produce IFN-γ or broad neutralization/administration of cytokine, investigations were completed by our group using both IFN-γ−/− and IFN-γR−/− mice as donors and recipients. Surprisingly, IFN-γ did not protect from GVHD via direct effects on donor T cells, as had been previously concluded. Rather, IFN-γ was required to act within the recipient to protect from GVHD.75  These studies revealed that IFN-γ signaling on donor cells augmented GVHD by enhancing Th1 expansion and subsequent pathogenic responses in the GI tract. Furthermore, IFN-γ is directly cytotoxic to the GI tract. In our hands, this effect of IFN-γ as a mediator of GVHD within the GI tract (and ultimately mortality) was more penetrant than other inflammatory cytokines, such as TNF or IL-6, but there are important caveats. Critically, an overall protective (rather than pathogenic) effect of IFN-γ signaling was observed because of direct signaling on lung parenchyma, which prevents donor T-cell migration and expansion in that organ and subsequent idiopathic pneumonia syndrome (Figure 2). These effects have since been confirmed by 2 groups,76,77  and IFN-γ has been shown to up-regulate PDL-1 expression on lung parenchyma, putatively resulting in donor T-cell apoptosis at this site. In conjunction with this direct signaling being responsible for protection from GVHD, IFN-γ is also an important mediator of the protective effects of exogenous IL-12 and IL-18 when administered early after bone marrow transplantation (BMT).78,79  In this setting, IFN-γ induces Fas-dependent apoptosis of donor CD4 T cells. Further studies indicated that IFN-γ production by the donor only is required for protection from GVHD by IL-12.70  Interestingly, donor-derived IFN-γ has been shown to be responsible for the cytopenias and marrow suppression associated with GVHD.80  This effect is mediated by IFN-γ signaling in recipient tissue via an as yet unknown intermediary. It is important to note that IFN-γ plays a role in the induction of immunomodulatory molecules that are both suppressive (eg, indoleamine-2-3-dioxygenase and nitric oxide synthase) and pathogenic (eg, TNF family members).73  Therefore, the effects of IFN-γ production and signaling after SCT are highly dependent on variables, such as conditioning regimen, the balance of intermediate molecules invoked, and the GVHD target organs involved. Whereas cause-and-effect studies in clinical BMT are extremely difficult, multiple studies using both genotyping81  and mRNA analysis82,83  have correlated cytokine induction to severe acute and chronic GVHD. Unfortunately, these studies were too small or did not attempt to dissect organ-specific effects.

Figure 2

The differential effects of IFN-γ on GVHD target organs and GVL. After conditioning induced tissue damage, NK and NKT cells are activated through activating receptor ligands (eg, NKG2D) and inhibitory receptors (eg, killer inhibitory receptors [KIR]) and TCR ligation (in the case of NKT cells), resulting in rapid IFN-γ release and licensing of recipient APCs (ie, up-regulation of CD40 and CD80/86). Transplanted donor T cells are activated after interaction with these host APCs to undergo expansion and Th1/Tc1 differentiation. IFN-γ production by donor T cells augments gut damage via (1) promoting Th1 and Tc1 differentiation and subsequent cytolysis against target tissue (eg, via perforin/granzyme), (2) direct cytopathic effects of the cytokine itself on the GI tract, and (3) priming monocytes/macrophages such that they secrete quantities of TNF and lymphotoxin after signaling by TLR ligands that are themselves directly cytopathic to the GI tract. In contrast to these pathogenic effects, IFN-γ secretion by donor T cells is critical in the lung parenchyma to induce PDL1 expression, which subsequently inhibits donor T-cell survival and infiltration within this organ, preventing the development of idiopathic pneumonia syndrome (IPS). IFN-γ also promotes GVL effects via (1) the enhancement of CTL expansion and function (eg, perforin/granzyme-dependent killing) and (2) acting on leukemia cells directly (eg, to up-regulate MHC expression and death receptors), rendering them more susceptible to T cell–mediated cytolysis.

Figure 2

The differential effects of IFN-γ on GVHD target organs and GVL. After conditioning induced tissue damage, NK and NKT cells are activated through activating receptor ligands (eg, NKG2D) and inhibitory receptors (eg, killer inhibitory receptors [KIR]) and TCR ligation (in the case of NKT cells), resulting in rapid IFN-γ release and licensing of recipient APCs (ie, up-regulation of CD40 and CD80/86). Transplanted donor T cells are activated after interaction with these host APCs to undergo expansion and Th1/Tc1 differentiation. IFN-γ production by donor T cells augments gut damage via (1) promoting Th1 and Tc1 differentiation and subsequent cytolysis against target tissue (eg, via perforin/granzyme), (2) direct cytopathic effects of the cytokine itself on the GI tract, and (3) priming monocytes/macrophages such that they secrete quantities of TNF and lymphotoxin after signaling by TLR ligands that are themselves directly cytopathic to the GI tract. In contrast to these pathogenic effects, IFN-γ secretion by donor T cells is critical in the lung parenchyma to induce PDL1 expression, which subsequently inhibits donor T-cell survival and infiltration within this organ, preventing the development of idiopathic pneumonia syndrome (IPS). IFN-γ also promotes GVL effects via (1) the enhancement of CTL expansion and function (eg, perforin/granzyme-dependent killing) and (2) acting on leukemia cells directly (eg, to up-regulate MHC expression and death receptors), rendering them more susceptible to T cell–mediated cytolysis.

Close modal

In conjunction with effects on immune reconstitution and T-cell activation after transplantation, IFN-γ also mediates GVL effects through direct interaction with tumor cells. This may result in the induction of apoptosis, suppression of proliferation, and increased sensitivity to cytotoxic T-cell killing via the up-regulation of Fas and MHC.84  However, further studies using tumor that does not express IFN-γR in models of BMT indicated that IFN-γ still promotes GVL effects, even in the absence of direct signaling by the cytokine with leukemic cells.85  Therefore, IFN-γ improves GVL via both direct effects on leukemic cells and improving antileukemia responses because of effects on T-cell priming and differentiation (Figure 2). It is tempting to consider that IFN-γ administration may prevent GVHD while enhancing GVL (ie, IFN-γ inhibits the infiltration of alloreactive cells into some GVHD target tissues while promoting CTL expansion and activity, particularly within the lymphohematopoietic system where leukemia predominantly resides). However, the known pathogenic effects of the cytokine, particularly within the GI tract, would suggest this as an unwise therapeutic strategy in patients.

Type III IFN

Type III IFNs were discovered in 2003 with the group composing 3 subtypes in humans, IFN-λ1, IFN-λ2, and IFN-λ3 or IL-29, IL-28A, and IL-28B, respectively. Mice only express IFN-λ2 and IFN-λ3. Type III IFNs signal through a distinct receptor complex consisting of the IFN-λR1 and IL10-R2 subunits,86  with signaling activating a similar set of transcription factors to that seen after type I IFN signaling.87  There is different expression of the receptors for type I and type III IFN, with most cell types expressing the type I IFN receptor whereas the IFN-λR1 is highly restricted to cells of epithelial origin, suggesting that this cytokine system may have evolved to play an important role in the prevention of viral invasion through the skin and mucosal surfaces.88 

Similar to type I IFN, the expression of type III IFN is induced by signaling through pattern recognition receptors, such as TLRs.89  Although the cell types responsible for type III IFN require clarification, preliminary studies demonstrate limited production in the CNS compared with the liver.88  Initial studies suggested redundancy between the type I and type III IFN systems. Whereas recipients lacking receptors to both type I IFN and type III IFN are more susceptible to respiratory viral infections compared with those lacking type I IFN signaling only, mice lacking only type III IFN receptor display only slightly enhanced susceptibility to infection, unlike type I IFN receptor-deficient mice, which are extremely susceptible.90  However, the site of infection appears to be an important factor toward the redundancy of type III IFN, as mice lacking the type III IFN receptor are highly susceptible to rotavirus infection.91  Therefore, despite expression of the type III IFN receptor in both the lung and small intestine, type III IFN appears to play a nonredundant role in antiviral infections only in the GI tract.

There has been extensive interest in the potential of type III IFN as an alternative to type I IFN for the treatment for chronic hepatitis C infection. Although IFN-α is a potent antiviral agent, the broad expression of the type I IFN receptor results in responses in many organs apart from the target tissue. Thus, serious complications include the development of neutropenia and lymphopenia resulting from IFNAR1 expression in hematopoietic cells,92  depression resulting from CNS effects,93  and constitutional symptoms (fevers, chills, myalgias). The limitation of type III IFN receptor expression to epithelial cells suggests that such complications may be avoided by type III IFN administration. Furthermore, 3 independent genome-wide association studies have identified single nucleotide polymorphisms in the IL-28B gene region to be associated with response of patients with chronic hepatitis C to pegylated IFN-α treatment.94,95  Therefore, IFN-λ is currently being investigated as an alternative therapeutic agent to type I IFNs, with results indicating that IFN-λ elicits antiviral activity without significant side effects.96,97  However, the lack of IFN-λ expression on hematopoietic cells suggests it is unlikely that IFN-λ administration would result in comparable GVL effects to type 1 IFN. However, it will be interesting to investigate the role of IFN-λ in GVHD, particularly considering the surprising colon-specific GVHD observed in recipients of BMT who lack the IFNAR1.45 

In conclusion, the broad range of outcomes and complex interactions after signaling with the various IFNs has resulted in conflicting studies observed in multiple experimental models and disease states. Although there has been limited investigation into cross-regulation between the IFNs, there is evidence that this does occur.63  Therefore, the ability of blocking one type of IFN to effect signaling and production of the other groups requires consideration when interpreting studies. Furthermore, the general lack of cross-species reactivity seen with many cytokines, including the IFNs, severely limits the ability of xenogeneic models of GVHD to provide informative data if the cytokine in question is capable of mediating significant end-organ damage and should probably be avoided in this setting.

Within SCT, the effects of IFN-γ are complex, highly temporally and conditioning dependent, with both beneficial and detrimental effects seen that are organ specific. Therefore, it will be important for future preclinical studies to use more sophisticated reagents that remove the ability of individual cell populations to produce or respond to IFN-γ. Ideally, this will involve inducible systems so that these issues can be more clearly delineated without the potential influence of developmental abnormalities that may be seen in knockout animals. Given current information, the exogenous administration of IFN-γ or its direct inhibition after clinical SCT is likely to result in highly unpredictable and potentially undesirable effects. However, promising preclinical results have been demonstrated by the inhibition of downstream signaling pathways of IFN-γ, such as Stat-1,98  or signaling pathways potentially upstream of IFN-γ, such as the Jak2 pathway,99  and this may be a viable alternative clinical strategy.

The role of type III IFNs in transplant outcome remains to be explored, but it is probable that, like type I IFN, target organ-specific effects will again be seen. In contrast, the type I IFNs have clear potential for immunomodulation to control relapse based on their ability to promote GVL effects. However, the administration of type 1 IFNs will be best tolerated in a setting temporally distant from conditioning where suppressive MHC class II–dependent effects dominate. Thus, the most appropriate timing would appear to be late after BMT, perhaps in conjunction with donor lymphocyte infusions, such that effects on antigen cross-presentation within donor DCs and direct effects on T cells may be invoked in the absence of the inflammatory milieu seen after conditioning and/or GVHD. Even then, the side effects, including the exacerbation of GVHD, will probably limit the use of this cytokine to patients at very high risk of relapse. Thus, future prospective clinical studies will need to initially focus on the ability of exogenous type I IFN administration to improve GVL and limit relapse in very high risk settings.

R.J.R. is a Leukemia Foundation Queensland PhD Scholarship Recipient. G.R.H. is a National Health and Medical Research Council Australia Fellow and a Queensland Health Senior Clinical Research Fellow.

Contribution: R.J.R. and G.R.H. reviewed the literature and wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Geoffrey R. Hill, Bone Marrow Transplantation Laboratory, Queensland Institute of Medical Research, 300 Herston Road, Herston, QLD, 4006, Australia; e-mail: geoff.hill@qimr.edu.au.

1
Beatty
 
PG
Clift
 
RA
Mickelson
 
EM
, et al. 
Marrow transplantation from related donors other than HLA-identical siblings.
N Engl J Med
1985
, vol. 
313
 
13
(pg. 
765
-
771
)
2
Ferrara
 
JL
Cooke
 
KR
Teshima
 
T
The pathophysiology of acute graft-versus-host disease.
Int J Hematol
2003
, vol. 
78
 
3
(pg. 
181
-
187
)
3
Koyama
 
M
Kuns
 
RD
Olver
 
SD
, et al. 
Recipient nonhematopoietic antigen-presenting cells are sufficient to induce lethal acute graft-versus-host disease.
Nat Med
2011
, vol. 
18
 
1
(pg. 
135
-
142
)
4
Toubai
 
T
Tawara
 
I
Sun
 
Y
, et al. 
Induction of acute GVHD by sex-mismatched H-Y antigens in the absence of functional radio-sensitive host hematopoietic-derived antigen presenting cells.
Blood
2012
, vol. 
119
 
16
(pg. 
3844
-
3853
)
5
Shlomchik
 
WD
Graft-versus-host disease.
Nat Rev Immunol
2007
, vol. 
7
 
5
(pg. 
340
-
352
)
6
Horowitz
 
MM
Gale
 
RP
Sondel
 
PM
, et al. 
Graft-versus-leukemia reactions after bone marrow transplantation.
Blood
1990
, vol. 
75
 
3
(pg. 
555
-
562
)
7
Wysocki
 
CA
Panoskaltsis-Mortari
 
A
Blazar
 
BR
Serody
 
JS
Leukocyte migration and graft-versus-host disease.
Blood
2005
, vol. 
105
 
11
(pg. 
4191
-
4199
)
8
Hill
 
GR
Crawford
 
JM
Cooke
 
KJ
Brinson
 
YS
Pan
 
L
Ferrara
 
JLM
Total body irradiation and acute graft versus host disease: the role of gastrointestinal damage and inflammatory cytokines.
Blood
1997
, vol. 
90
 
8
(pg. 
3204
-
3213
)
9
Martin
 
PJ
Schoch
 
G
Fisher
 
L
, et al. 
A retrospective analysis of therapy for acute graft-versus-host disease: initial treatment.
Blood
1990
, vol. 
76
 
8
(pg. 
1464
-
1472
)
10
Ferrara
 
JL
Levine
 
JE
Reddy
 
P
Holler
 
E
Graft-versus-host disease.
Lancet
2009
, vol. 
373
 
9674
(pg. 
1550
-
1561
)
11
Rezvani
 
K
Barrett
 
AJ
Characterizing and optimizing immune responses to leukaemia antigens after allogeneic stem cell transplantation.
Best Pract Res Clin Haematol
2008
, vol. 
21
 
3
(pg. 
437
-
453
)
12
Shlomchik
 
WD
Couzens
 
MS
Tang
 
CB
, et al. 
Prevention of graft versus host disease by inactivation of host antigen-presenting cells.
Science
1999
, vol. 
285
 
5426
(pg. 
412
-
415
)
13
Reddy
 
P
Maeda
 
Y
Liu
 
C
Krijanovski
 
OI
Korngold
 
R
Ferrara
 
JL
A crucial role for antigen-presenting cells and alloantigen expression in graft-versus-leukemia responses.
Nat Med
2005
, vol. 
11
 
11
(pg. 
1244
-
1249
)
14
Kägi
 
D
Vignaux
 
F
Ledermann
 
B
, et al. 
Fas and perforin pathways as major mechanisms of T cell-mediated cytotoxicity.
Science
1994
, vol. 
265
 (pg. 
528
-
530
)
15
Winter
 
H
Hu
 
H-M
Urba
 
WJ
Fox
 
BA
Tumor regression after adoptive transfer of effector T cells is independent of perforin or Fas ligand (APO-1L/CD95L).
J Immunol
1999
, vol. 
163
 
8
(pg. 
4462
-
4472
)
16
Schmaltz
 
C
Alpdogan
 
O
Horndasch
 
KJ
, et al. 
Differential use of Fas ligand and perforin cytotoxic pathways by donor T cells in graft-versus-host disease and graft-versus-leukemia effect.
Blood
2001
, vol. 
97
 
9
(pg. 
2886
-
2895
)
17
Isaacs
 
A
Lindenmann
 
J
Virus interference: I. The interferon.
Proc R Soc Lond B Biol Sci
1957
, vol. 
147
 
927
(pg. 
258
-
267
)
18
Rubinstein
 
M
Rubinstein
 
S
Familletti
 
PC
, et al. 
Human leukocyte interferon purified to homogeneity.
Science
1978
, vol. 
202
 
4374
(pg. 
1289
-
1290
)
19
Decker
 
T
Muller
 
M
Stockinger
 
S
The yin and yang of type I interferon activity in bacterial infection.
Nat Rev Immunol
2005
, vol. 
5
 
9
(pg. 
675
-
687
)
20
Kawai
 
T
Akira
 
S
The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors.
Nat Immunol
2010
, vol. 
11
 
5
(pg. 
373
-
384
)
21
Noppert
 
SJ
Fitzgerald
 
KA
Hertzog
 
PJ
The role of type I interferons in TLR responses.
Immunol Cell Biol
2007
, vol. 
85
 
6
(pg. 
446
-
457
)
22
Asselin-Paturel
 
C
Boonstra
 
A
Dalod
 
M
, et al. 
Mouse type I IFN-producing cells are immature APCs with plasmacytoid morphology.
Nat Immunol
2001
, vol. 
2
 
12
(pg. 
1144
-
1150
)
23
Ito
 
T
Wang
 
YH
Liu
 
YJ
Plasmacytoid dendritic cell precursors/type I interferon-producing cells sense viral infection by Toll-like receptor (TLR) 7 and TLR9.
Springer Semin Immunopathol
2005
, vol. 
26
 
3
(pg. 
221
-
229
)
24
Honda
 
K
Ohba
 
Y
Yanai
 
H
, et al. 
Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction.
Nature
2005
, vol. 
434
 
7036
(pg. 
1035
-
1040
)
25
de Weerd
 
NA
Samarajiwa
 
SA
Hertzog
 
PJ
Type I interferon receptors: biochemistry and biological functions.
J Biol Chem
2007
, vol. 
282
 
28
(pg. 
20053
-
20057
)
26
Biron
 
CA
Interferons alpha and beta as immune regulators: a new look.
Immunity
2001
, vol. 
14
 
6
(pg. 
661
-
664
)
27
Hwang
 
SY
Hertzog
 
PJ
Holland
 
KA
, et al. 
A null mutation in the gene encoding a type I interferon receptor component eliminates antiproliferative and antiviral responses to interferons alpha and beta and alters macrophage responses.
Proc Natl Acad Sci U S A
1995
, vol. 
92
 
24
(pg. 
11284
-
11288
)
28
Kolumam
 
GA
Thomas
 
S
Thompson
 
LJ
Sprent
 
J
Murali-Krishna
 
K
Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection.
J Exp Med
2005
, vol. 
202
 
5
(pg. 
637
-
650
)
29
Le Bon
 
A
Etchart
 
N
Rossmann
 
C
, et al. 
Cross-priming of CD8+ T cells stimulated by virus-induced type I interferon.
Nat Immunol
2003
, vol. 
4
 
10
(pg. 
1009
-
1015
)
30
Tough
 
DF
Borrow
 
P
Sprent
 
J
Induction of bystander T cell proliferation by viruses and type I interferon in vivo.
Science
1996
, vol. 
272
 
5270
(pg. 
1947
-
1950
)
31
Swann
 
JB
Hayakawa
 
Y
Zerafa
 
N
, et al. 
Type I IFN contributes to NK cell homeostasis, activation, and antitumor function.
J Immunol
2007
, vol. 
178
 
12
(pg. 
7540
-
7549
)
32
Horkheimer
 
I
Quigley
 
M
Zhu
 
J
Huang
 
X
Chao
 
NJ
Yang
 
Y
Induction of type I IFN is required for overcoming tumor-specific T cell tolerance following stem cell transplantation.
Blood
2009
, vol. 
113
 
21
(pg. 
5330
-
5339
)
33
Dunn
 
GP
Bruce
 
AT
Sheehan
 
KC
, et al. 
A critical function for type I interferons in cancer immunoediting.
Nat Immunol
2005
, vol. 
6
 
7
(pg. 
722
-
729
)
34
Longhi
 
MP
Trumpfheller
 
C
Idoyaga
 
J
, et al. 
Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Th1 immunity with poly IC as adjuvant.
J Exp Med
2009
, vol. 
206
 
7
(pg. 
1589
-
1602
)
35
Fuertes
 
MB
Kacha
 
AK
Kline
 
J
, et al. 
Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8alpha+ dendritic cells.
J Exp Med
2011
, vol. 
208
 
10
(pg. 
2005
-
2016
)
36
Tourbah
 
A
Lyon-Caen
 
O
Interferons in multiple sclerosis: ten years' experience.
Biochimie
2007
, vol. 
89
 
6
(pg. 
899
-
902
)
37
Guo
 
B
Chang
 
EY
Cheng
 
G
The type I IFN induction pathway constrains Th17-mediated autoimmune inflammation in mice.
J Clin Invest
2008
, vol. 
118
 
5
(pg. 
1680
-
1690
)
38
Shinohara
 
ML
Kim
 
JH
Garcia
 
VA
Cantor
 
H
Engagement of the type I interferon receptor on dendritic cells inhibits T helper 17 cell development: role of intracellular osteopontin.
Immunity
2008
, vol. 
29
 
1
(pg. 
68
-
78
)
39
Moschen
 
AR
Geiger
 
S
Krehan
 
I
Kaser
 
A
Tilg
 
H
Interferon-alpha controls IL-17 expression in vitro and in vivo.
Immunobiology
2008
, vol. 
213
 
9
(pg. 
779
-
787
)
40
Allan
 
NC
Richards
 
SM
Shepherd
 
PC
UK Medical Research Council randomised, multicentre trial of interferon-alpha n1 for chronic myeloid leukaemia: improved survival irrespective of cytogenetic response. The UK Medical Research Council's Working Parties for Therapeutic Trials in Adult Leukaemia.
Lancet
1995
, vol. 
345
 
8962
(pg. 
1392
-
1397
)
41
Italian Cooperative Study Group on Chronic Myeloid Leukemia
Interferon alfa-2a as compared with conventional chemotherapy for the treatment of chronic myeloid leukemia.
N Engl J Med
1994
, vol. 
330
 
12
(pg. 
820
-
825
)
42
Essers
 
MA
Offner
 
S
Blanco-Bose
 
WE
, et al. 
IFN-alpha activates dormant haematopoietic stem cells in vivo.
Nature
2009
, vol. 
458
 
7240
(pg. 
904
-
908
)
43
Porter
 
D
Roth
 
M
McGarigle
 
C
, et al. 
Induction of graft versus host disease as immunotherapy for relapsed chronic myeloid leukemia.
N Engl J Med
1994
, vol. 
330
 
2
(pg. 
100
-
106
)
44
Kolb
 
H
Mittermuller
 
J
Clemm
 
C
, et al. 
Donor leukocyte transfusions for treatment of recurrent chronic myelogenousleukemia in marrow transplant patients.
Blood
1990
, vol. 
76
 
12
(pg. 
2462
-
2465
)
45
Robb
 
RJ
Kreijveld
 
E
Kuns
 
RD
, et al. 
Type I-IFNs control GVHD and GVL responses after transplantation.
Blood
2011
, vol. 
118
 
12
(pg. 
3399
-
3409
)
46
Li
 
H
Matte-Martone
 
C
Tan
 
HS
, et al. 
Graft-versus-host disease is independent of innate signaling pathways triggered by pathogens in host hematopoietic cells.
J Immunol
2011
, vol. 
186
 
1
(pg. 
230
-
241
)
47
Hehlmann
 
R
Hochhaus
 
A
Kolb
 
HJ
, et al. 
Interferon-alpha before allogeneic bone marrow transplantation in chronic myelogenous leukemia does not affect outcome adversely, provided it is discontinued at least 90 days before the procedure.
Blood
1999
, vol. 
94
 
11
(pg. 
3668
-
3677
)
48
Morton
 
AJ
Gooley
 
T
Hansen
 
JA
, et al. 
Association between pretransplant interferon-alpha and outcome after unrelated donor marrow transplantation for chronic myelogenous leukemia in chronic phase.
Blood
1998
, vol. 
92
 
2
(pg. 
394
-
401
)
49
Wang
 
X
Li
 
H
Matte-Martone
 
C
, et al. 
Mechanisms of antigen presentation to T cells in murine graft-vs-host disease: cross-presentation and the appearance of cross-presentation.
Blood
2011
, vol. 
118
 
24
(pg. 
6426
-
6437
)
50
Schroder
 
K
Hertzog
 
PJ
Ravasi
 
T
Hume
 
DA
Interferon-gamma: an overview of signals, mechanisms and functions.
J Leukoc Biol
2004
, vol. 
75
 
2
(pg. 
163
-
189
)
51
Platanias
 
LC
Mechanisms of type-I- and type-II-interferon-mediated signalling.
Nat Rev Immunol
2005
, vol. 
5
 
5
(pg. 
375
-
386
)
52
Krebs
 
DL
Hilton
 
DJ
SOCS proteins: negative regulators of cytokine signaling.
Stem Cells
2001
, vol. 
19
 
5
(pg. 
378
-
387
)
53
Huang
 
S
Hendriks
 
W
Althage
 
A
, et al. 
Immune response in mice that lack the interferon-gamma receptor.
Science
1993
, vol. 
259
 
5102
(pg. 
1742
-
1745
)
54
Dalton
 
DK
Pitts-Meek
 
S
Keshav
 
S
Figari
 
IS
Bradley
 
A
Stewart
 
TA
Multiple defects of immune cell function in mice with disrupted interferon-gamma genes.
Science
1993
, vol. 
259
 
5102
(pg. 
1739
-
1742
)
55
Whitmire
 
JK
Tan
 
JT
Whitton
 
JL
Interferon-gamma acts directly on CD8+ T cells to increase their abundance during virus infection.
J Exp Med
2005
, vol. 
201
 
7
(pg. 
1053
-
1059
)
56
Refaeli
 
Y
Van Parijs
 
L
Alexander
 
SI
Abbas
 
AK
Interferon gamma is required for activation-induced death of T lymphocytes.
J Exp Med
2002
, vol. 
196
 
7
(pg. 
999
-
1005
)
57
Chu
 
CQ
Wittmer
 
S
Dalton
 
DK
Failure to suppress the expansion of the activated CD4 T cell population in interferon gamma-deficient mice leads to exacerbation of experimental autoimmune encephalomyelitis.
J Exp Med
2000
, vol. 
192
 
1
(pg. 
123
-
128
)
58
Badovinac
 
VP
Tvinnereim
 
AR
Harty
 
JT
Regulation of antigen-specific CD8+ T cell homeostasis by perforin and interferon-gamma.
Science
2000
, vol. 
290
 
5495
(pg. 
1354
-
1358
)
59
Sawitzki
 
B
Kingsley
 
CI
Oliveira
 
V
Karim
 
M
Herber
 
M
Wood
 
KJ
IFN-gamma production by alloantigen-reactive regulatory T cells is important for their regulatory function in vivo.
J Exp Med
2005
, vol. 
201
 
12
(pg. 
1925
-
1935
)
60
Dunn
 
GP
Koebel
 
CM
Schreiber
 
RD
Interferons, immunity and cancer immunoediting.
Nat Rev Immunol
2006
, vol. 
6
 
11
(pg. 
836
-
848
)
61
Gao
 
Y
Yang
 
W
Pan
 
M
, et al. 
Gamma delta T cells provide an early source of interferon gamma in tumor immunity.
J Exp Med
2003
, vol. 
198
 
3
(pg. 
433
-
442
)
62
Zaidi
 
MR
Merlino
 
G
The two faces of interferon-gamma in cancer.
Clin Cancer Res
2011
, vol. 
17
 
19
(pg. 
6118
-
6124
)
63
Rayamajhi
 
M
Humann
 
J
Penheiter
 
K
Andreasen
 
K
Lenz
 
LL
Induction of IFN-alpha beta enables Listeria monocytogenes to suppress macrophage activation by IFN-gamma.
J Exp Med
2010
, vol. 
207
 
2
(pg. 
327
-
337
)
64
Morris
 
ES
Macdonald
 
KP
Rowe
 
V
, et al. 
NKT cell-dependent leukemia eradication following stem cell mobilization with potent G-CSF analogs.
J Clin Invest
2005
, vol. 
115
 
11
(pg. 
3093
-
3103
)
65
Morris
 
ES
MacDonald
 
KP
Kuns
 
RD
, et al. 
Induction of natural killer T cell-dependent alloreactivity by administration of granulocyte colony-stimulating factor after bone marrow transplantation.
Nat Med
2009
, vol. 
15
 
4
(pg. 
436
-
441
)
66
Maeda
 
Y
Reddy
 
P
Lowler
 
KP
Liu
 
C
Bishop
 
DK
Ferrara
 
JL
Critical role of host gamma delta T cells in experimental acute graft-versus-host disease.
Blood
2005
, vol. 
106
 
2
(pg. 
749
-
755
)
67
Ellison
 
CA
Fischer
 
JM
Hayglass
 
KT
Gartner
 
JG
Murine graft-versus-host disease in an F1-hybrid model using IFN-gamma gene knockout donors.
J Immunol
1998
, vol. 
161
 (pg. 
631
-
640
)
68
Puliaev
 
R
Nguyen
 
P
Finkelman
 
FD
Via
 
CS
Differential requirement for IFN-gamma in CTL maturation in acute murine graft-versus-host disease.
J Immunol
2004
, vol. 
173
 
2
(pg. 
910
-
919
)
69
Murphy
 
WJ
Welniak
 
LA
Taub
 
DD
, et al. 
Differential effects of the absence of interferon-gamma and IL-4 in acute graft-versus-host disease after allogeneic bone marrow transplantation in mice.
J Clin Invest
1998
, vol. 
102
 
9
(pg. 
1742
-
1748
)
70
Yang
 
YG
Dey
 
BR
Sergio
 
JJ
Pearson
 
DA
Sykes
 
M
Donor-derived interferon gamma is required for inhibition of acute graft-versus-host disease by interleukin 12.
J Clin Invest
1998
, vol. 
102
 
12
(pg. 
2126
-
2135
)
71
Brok
 
HP
Heidt
 
PJ
van der Meide
 
PH
Zurcher
 
C
Vossen
 
JM
Interferon-gamma prevents graft-versus-host disease after allogeneic bone marrow transplantation in mice.
J Immunol
1993
, vol. 
151
 
11
(pg. 
6451
-
6459
)
72
Yang
 
YG
Qi
 
J
Wang
 
MG
Sykes
 
M
Donor-derived interferon gamma separates graft-versus-leukemia effects and graft-versus-host disease induced by donor CD8 T cells.
Blood
2002
, vol. 
99
 
11
(pg. 
4207
-
4215
)
73
Lu
 
Y
Waller
 
EK
Dichotomous role of interferon-gamma in allogeneic bone marrow transplant.
Biol Blood Marrow Transplant
2009
, vol. 
15
 
11
(pg. 
1347
-
1353
)
74
Dickinson
 
AM
Harrold
 
JL
Cullup
 
H
Haematopoietic stem cell transplantation: can our genes predict clinical outcome?
Expert Rev Mol Med
2007
, vol. 
9
 
29
(pg. 
1
-
19
)
75
Burman
 
AC
Banovic
 
T
Kuns
 
RD
, et al. 
IFN-gamma differentially controls the development of idiopathic pneumonia syndrome and GVHD of the GI tract.
Blood
2007
, vol. 
110
 
3
(pg. 
1064
-
1072
)
76
Yi
 
T
Chen
 
Y
Wang
 
L
, et al. 
Reciprocal differentiation and tissue-specific pathogenesis of Th1, Th2, and Th17 cells in graft-versus-host disease.
Blood
2009
, vol. 
114
 
14
(pg. 
3101
-
3112
)
77
Wang
 
H
Asavaroengchai
 
W
Yeap
 
BY
, et al. 
Paradoxical effects of IFN-gamma in graft-versus-host disease reflect promotion of lymphohematopoietic graft-versus-host reactions and inhibition of epithelial tissue injury.
Blood
2009
, vol. 
113
 
15
(pg. 
3612
-
3619
)
78
Yang
 
Y
Sergio
 
JJ
Pearson
 
DA
Szot
 
GL
Shimizu
 
A
Sykes
 
M
Interleukin-12 preserves the graft-versus-leukemia effect of allogeneic CD8 T cells while inhibiting CD4-dependent graft-versus-host disease in mice.
Blood
1997
, vol. 
90
 
11
(pg. 
4651
-
4660
)
79
Reddy
 
P
Teshima
 
T
Kukuruga
 
M
, et al. 
Interleukin-18 regulates acute graft-versus-host disease by enhancing Fas-mediated donor T cell apoptosis.
J Exp Med
2001
, vol. 
194
 
10
(pg. 
1433
-
1440
)
80
Delisle
 
JS
Gaboury
 
L
Belanger
 
MP
Tasse
 
E
Yagita
 
H
Perreault
 
C
Graft-versus-host disease causes failure of donor hematopoiesis and lymphopoiesis in interferon-gamma receptor-deficient hosts.
Blood
2008
, vol. 
112
 
5
(pg. 
2111
-
2119
)
81
Cavet
 
J
Dickinson
 
AM
Norden
 
J
Taylor
 
PR
Jackson
 
GH
Middleton
 
PG
Interferon-gamma and interleukin-6 gene polymorphisms associate with graft-versus-host disease in HLA-matched sibling bone marrow transplantation.
Blood
2001
, vol. 
98
 
5
(pg. 
1594
-
1600
)
82
Das
 
H
Imoto
 
S
Murayama
 
T
, et al. 
Kinetic analysis of cytokine gene expression in patients with GVHD after donor lymphocyte infusion.
Bone Marrow Transplant
2001
, vol. 
27
 
4
(pg. 
373
-
380
)
83
Ritchie
 
D
Seconi
 
J
Wood
 
C
Walton
 
J
Watt
 
V
Prospective monitoring of tumor necrosis factorα and interferon gamma to predict the onset of acute and chronic graft-versus-host disease after allogeneic stem cell transplantation.
Biol Blood Marrow Transplant
2005
, vol. 
11
 
9
(pg. 
706
-
712
)
84
Ikeda
 
H
Old
 
LJ
Schreiber
 
RD
The roles of IFN gamma in protection against tumor development and cancer immunoediting.
Cytokine Growth Factor Rev
2002
, vol. 
13
 
2
(pg. 
95
-
109
)
85
Yang
 
Y
Wang
 
H
Yu
 
H
, et al. 
IFN-gamma promotes graft-versus-leukemia effects without directly interacting with leukemia cells in mice after allogeneic hematopoietic cell transplantation.
Blood
2011
, vol. 
118
 
13
(pg. 
3721
-
3724
)
86
Kotenko
 
SV
Gallagher
 
G
Baurin
 
VV
, et al. 
IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex.
Nat Immunol
2003
, vol. 
4
 
1
(pg. 
69
-
77
)
87
Sheppard
 
P
Kindsvogel
 
W
Xu
 
W
, et al. 
IL-28, IL-29 and their class II cytokine receptor IL-28R.
Nat Immunol
2003
, vol. 
4
 
1
(pg. 
63
-
68
)
88
Sommereyns
 
C
Paul
 
S
Staeheli
 
P
Michiels
 
T
IFN-lambda (IFN-lambda) is expressed in a tissue-dependent fashion and primarily acts on epithelial cells in vivo.
PLoS Pathog
2008
, vol. 
4
 
3
pg. 
e1000017
 
89
Ank
 
N
West
 
H
Bartholdy
 
C
Eriksson
 
K
Thomsen
 
AR
Paludan
 
SR
Lambda interferon (IFN-lambda), a type III IFN, is induced by viruses and IFNs and displays potent antiviral activity against select virus infections in vivo.
J Virol
2006
, vol. 
80
 
9
(pg. 
4501
-
4509
)
90
Mordstein
 
M
Neugebauer
 
E
Ditt
 
V
, et al. 
Lambda interferon renders epithelial cells of the respiratory and GI tracts resistant to viral infections.
J Virol
2010
, vol. 
84
 
11
(pg. 
5670
-
5677
)
91
Pott
 
J
Mahlakoiv
 
T
Mordstein
 
M
, et al. 
IFN-lambda determines the intestinal epithelial antiviral host defense.
Proc Natl Acad Sci U S A
2011
, vol. 
108
 
19
(pg. 
7944
-
7949
)
92
Aghemo
 
A
Rumi
 
MG
Colombo
 
M
Pegylated interferons alpha2a and alpha2b in the treatment of chronic hepatitis C.
Nat Rev Gastroenterol Hepatol
2010
, vol. 
7
 
9
(pg. 
485
-
494
)
93
Valentine
 
AD
Meyers
 
CA
Kling
 
MA
Richelson
 
E
Hauser
 
P
Mood and cognitive side effects of interferon-alpha therapy.
Semin Oncol
1998
, vol. 
25
 
1 Suppl 1
(pg. 
39
-
47
)
94
Ge
 
D
Fellay
 
J
Thompson
 
AJ
, et al. 
Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance.
Nature
2009
, vol. 
461
 
7262
(pg. 
399
-
401
)
95
Tanaka
 
Y
Nishida
 
N
Sugiyama
 
M
, et al. 
Genome-wide association of IL28B with response to pegylated interferon-alpha and ribavirin therapy for chronic hepatitis C.
Nat Genet
2009
, vol. 
41
 
10
(pg. 
1105
-
1109
)
96
Muir
 
AJ
Shiffman
 
ML
Zaman
 
A
, et al. 
Phase 1b study of pegylated interferon lambda 1 with or without ribavirin in patients with chronic genotype 1 hepatitis C virus infection.
Hepatology
2010
, vol. 
52
 
3
(pg. 
822
-
832
)
97
Ramos
 
EL
Preclinical and clinical development of pegylated interferon-lambda 1 in chronic hepatitis C.
J Interferon Cytokine Res
2010
, vol. 
30
 
8
(pg. 
591
-
595
)
98
Ma
 
H
Lu
 
C
Ziegler
 
J
, et al. 
Absence of Stat1 in donor CD4 T cells promotes the expansion of Tregs and reduces graft-versus-host disease in mice.
J Clin Invest
2011
, vol. 
121
 
7
(pg. 
2554
-
2569
)
99
Betts
 
BC
Abdel-Wahab
 
O
Curran
 
SA
, et al. 
Janus kinase-2 inhibition induces durable tolerance to alloantigen by human dendritic cell-stimulated T cells yet preserves immunity to recall antigen.
Blood
2011
, vol. 
118
 
19
(pg. 
5330
-
5339
)
Sign in via your Institution