Acute graft-versus-host disease (GVHD), the major complication of allogeneic bone marrow transplantation (BMT), limits the application of this curative but toxic therapy. Studies of inflammatory pathways involved in GVHD in animals have shown that the gastrointestinal (GI) tract plays a major role in the amplification of systemic disease. Damage to the GI tract increases the translocation of inflammatory stimuli such as endotoxin, which promotes further inflammation and additional GI tract damage. The GI tract is therefore critical to the propagation of the “cytokine storm” characteristic of acute GVHD. Experimental approaches to the prevention of GVHD include reducing the damage to the GI tract by fortification of the GI mucosal barrier through novel “cytokine shields” such as IL-11 or keratinocyte growth factor. Such strategies have reduced GVHD while preserving a graft-versus-leukemia effect in animal models, and they now deserve formal testing in carefully designed clinical trials.

Allogeneic bone marrow transplantation (BMT) remains the treatment of choice for a number of malignant conditions. Graft-versus-host disease (GVHD), the primary complication of allogeneic BMT, remains the major limitation to this therapeutic approach. Donor T cells are critical in the induction of acute GVHD because depletion of T cells from the bone marrow graft effectively prevents GVHD but also results in an increase in leukemic relapse.1 Although there is a fundamental requirement for donor T cells in GVHD, the process still occurs in the absence of the T-cell cytotoxicity pathways (perforin, FasL, granzyme).2,3This anomaly has forced investigators to reconsider the traditional T-cell–restricted paradigms of GVHD pathophysiology.

Increasing evidence in experimental and clinical BMT settings suggests that damage to the gastrointestinal (GI) tract during acute GVHD plays a major pathophysiologic role in the amplification of systemic disease. Endotoxin or lipopolysaccharide (LPS) is a constituent of normal bowel flora known to play an important role in GVHD pathogenesis. Early animal studies showed that death after BMT was prevented if mice were given antibiotics to decontaminate the gut; normalization of the gut flora at or before day 20 abrogated this effect.4 In the clinical setting, gram-negative gut decontamination has also been shown to reduce GVHD.5,6Furthermore, the intensity of this decontamination has recently been demonstrated to be an important predictor of GVHD severity.7,8 

Lipopolysaccharide is a potent stimulator of inflammatory cytokine production, such as tumor necrosis factor (TNF)-α, IL-1, and IL-12, which are important mediators of clinical9-11 and experimental GVHD.12-14 The production of TNF-α by monocytes and macrophages is transduced by 2 signals.15 The first is a priming signal that may be provided by interferon (IFN)-γ15 and radiation.13 The second is a triggering signal provided by bacterial products such as LPS.15 Other bacterial products, such as CpG DNA repeats, in the GI tract lumen have also been shown to have potent immuno-stimulatory properties and to induce strong Th1 responses.16 Furthermore, microbial superantigens may activate B cells by direct stimulation of major histocompatibility complex (MHC) class II molecules.17 During GVHD, the IFN-γ produced by donor T cells renders monocytes and macrophages extremely sensitive to endogenous LPS.18 Further support for the role of LPS in GVHD comes from a recent study comparing donor monocytes and macrophages that were genetically sensitive (ie, normal) or resistant to the effects of LPS for their ability to induce GVHD.19 The donor mouse strains were otherwise identical, and, in particular, their T-cell responses to host alloantigens were equivalent. Allogeneic BMT recipients of LPS-resistant bone marrow demonstrated significantly less TNF-α production and reduced GVHD than recipients of LPS-sensitive bone marrow. Significantly, the reduced production of TNF-α in recipients of LPS-resistant marrow was associated with reduced GVHD of the GI tract. The causality of the association was demonstrated by the systemic neutralization of TNF-α, which reduced GVHD of the GI tract and lessened the severity of overall disease.

Additional evidence for the importance of GI tract integrity during GVHD comes from studies of the effect of BMT conditioning on GVHD severity after allogeneic BMT. Clinical studies first suggested a correlation between GVHD severity and radiation dose (less than 1200 versus more than 1200 cGy)20,21 and more severe GVHD after conditioning regimens that included radiation therapy than those that included only chemotherapy.22 The increased incidence of conditioning-related toxicity has also been associated with GVHD incidence.23 However, these studies demonstrated an inverse correlation between conditioning intensity and compliance with immunosuppression prophylaxis, which might partially account for the increase in GVHD. Indeed, only 1 of these studies identified increased radiation intensity as an independent risk factor for GVHD,20 and there was no demonstrable association with severe disease (grades 2 to 4). By contrast, the association of increased GVHD with dose reductions in cyclosporine was maintained throughout all GVHD grades. Using clinical data alone, it has thus been difficult to separate the influence of conditioning intensity and suppression of T-cell function on subsequent GVHD severity.

We have recently examined this issue in murine BMT models in which these variables could be tightly controlled. These studies demonstrated an increase in GVHD severity in several donor–recipient strain combinations, after intensification of the conditioning regimen, by increasing the total body irradiation (TBI) dose from 900 cGy to 1300 cGy. Synergistic damage to the GI tract was caused by increased TBI and allogeneic donor cells, permitting increased translocation of LPS to the systemic circulation. In vitro, LPS triggered the most TNF-α secretion in macrophages taken from animals with the worst GI tract damage. Neutralization of TNF-α eliminated deaths from GVHD. Thus, the higher TBI dose increased gut damage after allogeneic BMT, causing higher systemic levels of inflammatory cytokines and more severe GVHD.13 High levels of inflammatory cytokines may perpetuate GVHD because TNF-α has been shown to be directly toxic to the GI tract during GVHD in other experimental systems.19,24,25 

The addition of cyclophosphamide (Cy) to TBI also increased damage to the GI tract and the subsequent incidences of mortality and morbidity related to GVHD.14 After Cy/TBI conditioning, neutralization of IL-1 with a hamster antibody to the IL-1 receptor significantly reduced serum LPS levels and GVHD deaths, whereas neutralization of TNF-α did not. Although the expansion of donor T cells on day 13 after BMT was increased after Cy/TBI than with Cy or TBI alone, cytotoxic T lymphocyte (CTL) function was not different between groups. Taken together, these studies confirm that increasing the dose of TBI or combining TBI with cyclophosphamide increases the severity of GVHD. Importantly, the pathophysiologic mechanisms involved in this increase in GVHD appear to be focused on GI tract damage and subsequent amplification of the inflammatory effectors of GVHD rather than on the effects on donor T cells.

These studies suggest that the GI tract is not only a major target organ of GVHD but is a critical amplifier of systemic GVHD severity. We have previously suggested that acute GVHD pathophysiology can be conceptualized in 3 sequential phases (Figure1).26 In phase 1, the conditioning regimen (irradiation, chemotherapy, or both) leads to damage to and activation of host tissue by release of the inflammatory cytokines TNF-α and IL-1. These cytokines can increase the expression of MHC antigens and adhesion molecules on host antigen-presenting cells, enhancing the recognition of host MHC and minor histocompatibility antigens by mature donor T cells. Donor T-cell activation in phase 2 is characterized by the proliferation of Th1 T cells and the secretion of IL-2 and IFN-γ. IL-2 and IFN-γ induce further T-cell expansion, CTL- and NK-cell responses, and prime additional mononuclear phagocytes to produce IL-1 and TNF-α. Effector functions of mononuclear phagocytes (phase 3) are triggered by the secondary signal provided by bacterial products such as LPS. Damage to the intestinal mucosa in phase 1 and by cytolytic effectors activated in phase 2 allows translocation of LPS from the intestinal lumen to the circulation. Subsequently, LPS may stimulate additional cytokine production by gut-associated lymphocytes and macrophages in the GI tract and by keratinocytes, dermal fibroblasts, and macrophages within the skin. This mechanism may amplify local tissue injury and further promote an inflammatory response that, together with the CTL and NK component, leads to target tissue destruction in the BMT host. Damage to the GI tract in phase 3 increases LPS release, stimulating further cytokine production and causing additional GI tract damage. Thus the GI tract is critical to propagating the “cytokine storm” characteristic of acute GVHD.

Fig. 1.

The immunopathophysiology of GVHD.

Schematic representation of central role of GI tract damage during GVHD. In phase 1, the conditioning regimen (irradiation, chemotherapy, or both) leads to the damage and activation of host tissues, especially the intestinal mucosa. This allows the translocation of LPS from the intestinal lumen to the circulation, stimulating the secretion of the inflammatory cytokines TNF-α and IL-1 from host tissues, particularly macrophages. These cytokines increase the expression of MHC antigens and adhesion molecules on host tissues, enhancing the recognition of MHC and minor histocompatibility antigens by mature donor T cells. Donor T-cell activation in phase 2 is characterized by the proliferation of Th1 T cells in the presence of IL-12 and the secretion of IL-2 and IFN-γ. IL-2 and IFN-γ induce further T-cell expansion and CTL and NK cell responses, and they activate mononuclear phagocytes. The CTL and NK effectors damage tissue by perforin/granzyme, FasL, and TNF-α. In phase 3, effector functions of activated mononuclear phagocytes are triggered by the secondary signal provided by LPS and other immuno-stimulatory molecules that leak through the intestinal mucosa damaged during phases 1 and 2. This damage results in the amplification of local tissue injury, and it further promotes an inflammatory response. Damage to the GI tract in this phase, principally by inflammatory cytokines, amplifies LPS release and leads to the “cytokine storm” characteristic of severe acute GVHD. Double lines show the points at which KGF and IL-11 both interrupt this process, whereas single lines reflect interruption by IL-11 only. IL-11, but not KGF, promotes type 2 donor T-cell differentiation and inhibits IFN-γ secretion. Neither IL-11 nor KGF impairs CTL or NK function, thereby preserving GVL effects. The reduction in GI tract damage by these “cytokine shields” prevents systemic LPS translocation and reduces inflammatory cytokine production, culminating in reduced GI tract damage and subsequent death from GVHD.

Fig. 1.

The immunopathophysiology of GVHD.

Schematic representation of central role of GI tract damage during GVHD. In phase 1, the conditioning regimen (irradiation, chemotherapy, or both) leads to the damage and activation of host tissues, especially the intestinal mucosa. This allows the translocation of LPS from the intestinal lumen to the circulation, stimulating the secretion of the inflammatory cytokines TNF-α and IL-1 from host tissues, particularly macrophages. These cytokines increase the expression of MHC antigens and adhesion molecules on host tissues, enhancing the recognition of MHC and minor histocompatibility antigens by mature donor T cells. Donor T-cell activation in phase 2 is characterized by the proliferation of Th1 T cells in the presence of IL-12 and the secretion of IL-2 and IFN-γ. IL-2 and IFN-γ induce further T-cell expansion and CTL and NK cell responses, and they activate mononuclear phagocytes. The CTL and NK effectors damage tissue by perforin/granzyme, FasL, and TNF-α. In phase 3, effector functions of activated mononuclear phagocytes are triggered by the secondary signal provided by LPS and other immuno-stimulatory molecules that leak through the intestinal mucosa damaged during phases 1 and 2. This damage results in the amplification of local tissue injury, and it further promotes an inflammatory response. Damage to the GI tract in this phase, principally by inflammatory cytokines, amplifies LPS release and leads to the “cytokine storm” characteristic of severe acute GVHD. Double lines show the points at which KGF and IL-11 both interrupt this process, whereas single lines reflect interruption by IL-11 only. IL-11, but not KGF, promotes type 2 donor T-cell differentiation and inhibits IFN-γ secretion. Neither IL-11 nor KGF impairs CTL or NK function, thereby preserving GVL effects. The reduction in GI tract damage by these “cytokine shields” prevents systemic LPS translocation and reduces inflammatory cytokine production, culminating in reduced GI tract damage and subsequent death from GVHD.

Close modal

Although cytokines clearly play important roles in incidences of morbidity and mortality related to systemic GVHD, their relative importance as mediators of damage in GVHD target organs is less well established. The unusual cluster of GVHD target organs (skin, gut, and liver) is inadequately explained by the systemic release of cytokines. For example, intravenous infusion of TNF-α and IL-1 does not cause the lymphomononuclear cell infiltration of liver and skin observed in GVHD. Furthermore, the absence of GVHD toxicity in other visceral organs, such as the kidney, argues against circulating cytokines as the sole cause of tissue-specific damage. The infiltrates seen in GVHD target organs are generally thought to contain T cells responding to alloantigens on host tissues. As mentioned above, LPS leakage through skin or mucosa may act as an adjuvant to the antigens expressed in these tissues, attracting and activating alloreactive donor T cells. A second possibility is that tissue-specific neoantigens are expressed at these sites as the result of ongoing inflammation. Such inflammation may alter ligands for homing receptors on T cells (eg, selectins) that enables them to traffic into specific tissues.

It is clear in murine systems that cytolytic T-lymphocyte effectors contribute to GVHD. The application of knock-out technology and the identification of pertinent mutant mice have provided important tools for dissecting effector mechanisms. Three cytolytic pathways have been identified as important to GVHD: the perforin/granzyme B pathway, the Fas/FasL pathway, and direct cytokine-mediated injury. Donor cells from perforin-deficient and granzyme B-deficient knock-out mice can mediate lethal GVHD, but the onset of clinical manifestations is significantly delayed.27,28 Similarly, when mutant mice deficient in FasL (gld) are used as donors, GVHD occurs in an attenuated fashion.3 When FasL-deficient mice are crossed with those having perforin or granzyme B knock-outs, the use of lymphocytes from these donors further diminishes but does not abrogate GVHD.27 Interestingly, the Fas pathway is important in the development of hepatic and cutaneous GVHD.24 

GVHD histopathology in the GI tract has been described in 3 phases.29 The early phases of GI tract changes have been described in animal models that do not use chemotherapy or radiation to condition the host; therefore, direct comparisons to clinical GVHD after BMT are not possible. This initial proliferative phase results in increased crypt cell mitotic activity, crypt lengthening, and intraepithelial lymphocytes. In experimental systems, this phase seems to be linked to IFN-γ production,30 which increases MHC class II expression and gut permeability by altering tight junction integrity31 and may modulate crypt stem-cell turnover.29 The histologic features of the GI tract in clinical GVHD and experimental GVHD after myeloablative conditioning are consistent with the destructive and atrophic phases, characterized by villus blunting, lamina propria inflammation, crypt destruction (with crypt stem-cell loss), and mucosal atrophy. These features can be induced in animals by the administration of exogenous cytokines, including TNF-α32 and IL-1.33 Furthermore, the inhibition of IFN-γ,34 TNF-α,25IL-1,33 or nitric oxide35 can reduce GI tract histopathology in animals with GVHD. In contrast, CTL effectors do not appear to play a dominant role in experimental GVHD of the GI tract,3,13,14,19,24,36 despite the ability of intraepithelial lymphocytes to induce Fas-mediated apoptosis of host type tumor cells.37 It is clear, when these findings are considered in aggregate, that cytokines and cellular effectors combine to produce specific target organ damage and systemic toxicity of acute GVHD.

The experimental studies referenced above used novel cytokine antagonists and transgenic/knock-out mice. As yet, it has not been possible to confirm the extent to which the principals derived from these studies apply to humans. The histologic features of end-stage intestinal GVHD are similar in animals and humans.29 The diarrhea characteristic of GVHD may occur because of a number of mechanisms, including enterocyte damage and epithelial disaccharidase deficiency with an excess of luminal sugars and osmotic water loss, an increase in the proportion of immature enterocytes with subsequent enzyme deficiency and impaired water transport, and protein and water exudation through a hyperpermeable epithelium. In the large intestine, damage to colonic enterocytes also impairs water reabsorption.29 

Increasing the intensity of the conditioning regimen in murine models of GVHD to the levels used clinically amplifies GVHD of the GI tract. This increase in gut injury and LPS leak is a common proximal pathway for the dysregulation of inflammatory cytokines after allogeneic BMT. As discussed above, recent work has confirmed that GVHD of the GI tract is principally mediated by inflammatory cytokines. From this perspective, it should be possible to interrupt the process of GI tract damage at a number of steps. First, attempts to eradicate or significantly reduce the load of gram-negative organisms from the GI tract are current practice in a number of transplant centers. Unfortunately, endotoxemia has been noted to be common after BMT even with gut decontamination; it occurs in association with biochemical parameters of gut damage.38 Although gut decontamination can reduce clinical GVHD,5,7 the effect is at best partial. It has not been shown to benefit the recipients of unrelated donor BMT, who are in the greatest need for improved GVHD prophylaxis.8 Inhibition of systemic LPS with neutralizing proteins is under investigation in experimental GVHD models. However, the success of this approach assumes that LPS is the only or primary immuno-stimulatory constituent of the GI tract lumen that amplifies GVHD. There are numerous additional putative immuno-stimulatory molecules, such as soluble peptidoglycan, lipoteichoic acid,39 bacterial DNA,40 and heat-shock proteins,41 suggesting that neutralization of LPS alone may be only partially successful in preventing GVHD.

Reductions in the doses of chemoradiotherapy to condition BMT recipients should also reduce GVHD, as demonstrated in experimental models.13,14 This reduction is the result of reduced priming of mononuclear cells by lower TBI doses and of subsequent reductions in TNF-α production.13 It is also likely that low TBI doses fail to sensitize the GI tract to secondary damage by inflammatory cytokines. The use of non-myeloablative conditioning should therefore also reduce GI tract damage after allogeneic BMT. The activation of donor T cells is enhanced by inflammatory cytokines,42 and a temporal separation of the inflammatory milieu induced by conditioning (which is thought to be self-limited) and donor lymphocyte infusions may interrupt the cytokine cascade that damages the GI tract and increases systemic GVHD.12,43 

Much of the therapeutic potential of allogeneic BMT relates to the graft-versus-leukemia (GVL) effect, which is mediated by the cytotoxic pathways of donor T and NK cells, including perforin/granzyme and Fas/FasL.44 In experimental GVHD models, the absence of perforin in donor T cells results in an almost complete loss of GVL,45-47 whereas the absence of FasL does not diminish GVL effects.47 Inhibition of inflammatory cytokine production after BMT might offer an approach to separate GVHD and GVL. However, TNF-α may not be an ideal target because the p55 TNF-α receptor is critical for donor CTL activity after BMT and contributes to the GVL effect.14 GVL effects are also diminished when TNF-α is neutralized in experimental models of GVHD.47 IL-1 may be a more attractive target for neutralization because the inhibition of IL-1 does not inhibit CTL generation or the GVL effect.14Neutralization of inflammatory cytokines must therefore be approached on an individual basis with respect to the potential to separate GVHD and GVL.

An alternative approach to prevent GI tract damage during allogeneic BMT may permit the exploitation of intensive conditioning as an antileukemic modality without requiring T-cell depletion. This approach involves strengthening the GI mucosal barrier before BMT conditioning to prevent the entry of immunostimulatory molecules from the GI tract lumen into the circulation. Because direct shielding of the GI tract from TBI is not feasible, this effort relies on pharmacologic agents that provide a “cytokine shield” to reduce mucosal sensitivity to radiation, chemotherapy, or both. This approach is attractive because it blocks inflammatory cytokine dysregulation before the initiation of the cascade. In addition, by acting as indirect cytokine antagonists, these shields would not impede the physiological functions of cytokines in cellular differentiation (as might be the case with complete neutralization of TNF-α and IL-1). Two growth factors, IL-11 and KGF, have recently shown particular promise as cytokine shields.

IL-11, a member of the IL-6 cytokine family, is produced by a variety of tissues, including the central nervous system, thymus, lung, bone, skin, and connective tissue, and has pleiotropic effects.48 IL-11 stimulates megakaryopoiesis and accelerates neutrophil recovery after myelosuppressive therapy.49-51 In addition, IL-11 has potent anti-inflammatory effects by virtue of its ability to inhibit nuclear translocation of nuclear factor-κB (NF-κB).52,53 Preclinical studies have demonstrated the efficacy of IL-11 in treating inflammatory disorders, among them oxygen- and radiation-induced lung damage,54,55inflammatory bowel disease,56 and sepsis.57 In addition, IL-11 down-regulates IL-12 production by macrophages,58 which suggests that IL-11 may also modulate T-cell–mediated inflammation. Significantly, IL-11 has direct protective effects on the GI tract epithelium in models of injury by chemotherapy and radiation,59-63 surgery,64,65and ischemia.66 Small bowel crypt recovery is improved through the protection of clonogenic crypt cells,60reductions in apoptosis,62 and increases in cellular mitotic index.59,64,65 These studies confirmed that IL-11 has trophic effects on mucosal epithelium and suggested that continuing therapy with IL-11 after BMT conditioning would not impair mucosal healing. Of interest was the observation that maximum crypt protection occurred if IL-11 treatment was begun before irradiation and was continued for 3 days.60 

In experimental studies examining the possible use of IL-11 as a cytokine shield to prevent GVHD, IL-11 administration was begun 2 days before conditioning and was continued for 7 to 14 days after BMT. When used in this fashion, IL-11 almost completely prevented GVHD of the small bowel, and it reduced serum endotoxin levels after BMT by 80%. Treatment with IL-11 also reduced TNF-α serum levels and suppressed TNF-α secretion by macrophages to LPS stimulation in vitro. Donor CTL responses to host antigens were not affected by IL-11. Surprisingly, IL-11 administration polarized the donor T-cell cytokine responses to host antigen after BMT with a 2-fold reduction in IFN-γ and IL-2 secretion and a 10-fold increase in IL-4. This polarization of T-cell responses was associated with reduced IFN-γ serum levels and decreased IL-12 production in mixed lymphocyte cultures. Therefore, IL-11 inhibits GVHD pathophysiology at multiple steps (Figure 1), enabling IL-11 to dramatically reduce GVHD mortality and morbidity rates after allogeneic BMT.67 The 10-day schedule of IL-11 treatment, beginning just before BMT, also provides long-term protection of the GI tract and improved immune reconstitution.67 Further studies showed that IL-11 spared donor CTL/NK function and preserved a GVL effect that was mediated by perforin, improving long-term leukemia-free survival.45These data confirm the ability of IL-11 to separate GVHD and GVL in experimental animal studies.

A member of the fibroblast growth factor family FGF-7, KGF shows specificity for epithelial tissues that express its receptors, including gut epithelial cells, hepatocytes,68 skin keratinocytes,69 alveolar type 2 cells,70mammary epithelium,71 and urothelium.72 In early studies, KGF administration before autologous BMT dramatically protected the gut epithelium from injury by lethal chemoradiotherapy.73 This protection appears to result from a potent trophic effect on intestinal epithelium68 and from improved survival of crypt stem cells,74 perhaps through reduced oxidative damage (nonselenium glutathione peroxidase)75 and enhanced DNA repair (DNA polymerase-α, -δ, and -ε).76 

In 2 recent studies, investigators have examined the effect of human recombinant KGF on experimental GVHD.77,78 When KGF was given before conditioning only, it reduced the mortality rate and GVHD target organ histopathology.78 When KGF was administered from 3 days before to 7 days after BMT, it abolished GVHD of the GI tract and further improved survival.77 As expected, this improvement was associated with a reduction in serum LPS and TNF-α levels. Therapy with KGF also preserved donor T-cell responses (CTL activity, proliferation, and IL-2 production) to host antigens and significantly improved leukemia-free survival and when a lethal dose of P815 leukemia was given at the time of BMT (42% vs 4%; P < .001).77 KGF has a favorable clinical toxicity profile,79 and its administration, like that of IL-11, thus offers a novel approach to the separation of GVL effects from GVHD (Figure 1).

Insights into the pathophysiology of GVHD have confirmed a central role of GI tract damage in this process and have predicted for the success of novel approaches to preventing GVHD in experimental models. It is important to note that the majority of data regarding our understanding of GVHD pathophysiology derive from murine models that use inbred strains of donors and recipients with highly defined and often limited genetic disparity compared with our own human outbred species. The experimental conditions of these BMT models were highly controlled, and the mice spent their entire lives, from conception to death, in specific pathogen-free conditions. Both circumstances help to maximize the clarity of experimental results, but they may also lead to certain distortions with respect to the more variable clinical reality of human transplantation. It is unlikely that the mechanisms of murine and human GVHD differ completely, but the relative contribution of a given effector pathway to specific organ damage may well differ between humans and mice. Certainly, the use of intensive conditioning schedules and pharmacologic immunosuppression in clinical BMT is in contrast to most published murine systems in which GVHD target organ damage has been mediated primarily by T cells. Strong evidence supports the concept that inflammatory cytokines, GI tract damage, and LPS translocation are important mediators of clinical GVHD.9-11 Reduced GVHD severity seen in patients conditioned with nonmyeloablative conditioning regimens provides further support for this assertion.80 Nevertheless, some initial clinical studies of specific cytokine antagonists have yielded less compelling results than the murine models predict, and the effect of any single cytokine inhibitor may be small because of complex interactions of multiple, potentially redundant cytokines. Clinical testing of novel biologic response modifiers often occurs under high-risk conditions in patients with advanced disease, and considerable time may be required for these novel therapeutic regimens to be introduced under more standard risk situations, in which their true value in the prevention and treatment of GVHD can be discerned.

The preclinical data offer a compelling rationale for the testing of agents that can protect the GI tract and that therefore may modulate the inflammatory amplification of acute GVHD. Success in animal models does not always predict clinical efficacy, however. Given the potential toxicities of these agents and the severity of illness experienced by many patients under allogeneic BMT, the use of such agents should be tested in carefully monitored clinical protocols designed to test the feasibility and safety of this approach and its potential efficacy. If successful, the concept of “cytokine shields” may allow the potent antileukemic effect of high-dose chemoradiotherapy to be used without the induction of life-threatening GVHD and may permit traditional allogeneic BMT to be undertaken in a patient population that would otherwise be ineligible for this therapy.

Reprints: James L. M. Ferrara, Departments of Internal Medicine and Pediatrics, University of Michigan Cancer Center, 1500 E Medical Center Drive, Ann Arbor, MI 48109-0560; e-mail:ferrara@umich.edu.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Ferrara
JLM
Deeg
HJ
Graft versus host disease.
N Engl J Med.
324
1991
667
2
Baker
MB
Podack
ER
Levy
RB
Perforin- and Fas-mediated cytotoxic pathways are not required for allogeneic resistance to bone marrow grafts in mice.
Biol Blood Marrow Transplant.
1
1995
69
3
Baker
MB
Altman
NH
Podack
ER
Levy
RB
The role of cell-mediated cytotoxicity in acute GVHD after MHC-matched allogeneic bone marrow transplantation in mice.
J Exp Med.
183
1996
2645
4
van Bekkum
DW
Roodenburg
J
Heidt
PJ
van der Waaij
D
Mitigation of secondary disease of allogeneic mouse radiation chimeras by modification of the intestinal microflora.
J Natl Cancer Inst.
52
1974
401
5
Storb
R
Prentice
RL
Buckner
CD
et al
Graft–versus–host disease and survival in patients with aplastic anemia treated by marrow grafts from HLA-identical siblings: beneficial effect of a protective environment.
N Engl J Med.
308
1983
302
6
Moller
J
Skirhoj
P
Hoiby
N
Peterson
FB
Protection against graft versus host disease by gut sterilization?
Exp Haematol.
10
1982
101
7
Beelen
DW
Haralambie
E
Brandt
H
et al
Evidence that sustained growth suppression of intestinal anaerobic bacteria reduces the risk of acute graft-versus-host disease after sibling marrow transplantation.
Blood.
80
1992
2668
8
Beelen
DW
Elmaagacli
A
Muller
KD
Hirche
H
Schaefer
UW
Influence of intestinal bacterial decontamination using metronidazole and ciprofloxacin or ciprofloxacin alone on the development of acute graft-versus-host disease after marrow transplantation in patients with hematologic malignancies: final results and long term follow-up of an open-label prospective randomized trial.
Blood.
93
1999
3267
9
Antin
JH
Weinstein
HJ
Guinan
EC
et al
Recombinant human interleukin-1 receptor antagonist in the treatment of steroid-resistant graft–versus–host disease.
Blood.
84
1994
1342
10
Holler
E
Kolb
HJ
Moller
A
et al
Increased serum levels of tumor necrosis factor alpha precede major complications of bone marrow transplantation.
Blood.
75
1990
1011
11
Holler
E
Kolb
HJ
Mittermueller
J
et al
Modulation of acute graft-versus-host disease after allogeneic bone marrow transplantation by tumor necrosis factor α (TNF-α) release in the course of pretransplant conditioning: role of conditioning regimens and prophylactic application of a monoclonal antibody neutralizing human TNF-α (MAK 195F).
Blood.
86
1995
890
12
Xun
CQ
Thompson
JS
Jennings
CD
Brown
SA
Widmer
MB
Effect of total body irradiation, busulfan-cyclophosphamide, or cyclophosphamide conditioning on inflammatory cytokine release and development of acute and chronic graft-versus-host disease in H-2-incompatible transplanted SCID mice.
Blood.
83
1994
2360
13
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.
90
1997
3204
14
Hill
GR
Teshima
T
Gerbita
A
et al
Differential roles of IL-1 and TNF-α on graft-versus-host disease and graft-versus leukemia.
J Clin Invest.
104
1999
459
15
Nestel
FP
Price
KS
Seemayer
TA
Lapp
WS
Macrophage priming and lipopolysaccharide-triggered release of tumor necrosis factor alpha during graft-versus-host disease.
J Exp Med.
175
1992
405
16
Hartmann
G
Weiner
GJ
Kreig
AM
CpG: a potent signal for growth, activation, and maturation of human dendritic cells.
Proc Natl Acad Sci U S A.
6
1999
9305
17
Scholl
PR
Geha
RS
MHC class II signaling in B-cell activation.
Immunol Today.
9
1994
418
18
Nestel
F
Kichian
K
You-Ten
K
Desbarats
J
Price
K
Lapp
WS
The role of endotoxin in the pathogenesis of acute graft-versus-host disease.
Graft-vs-Host Disease.
2nd ed.
Ferrara
JLM
Deeg
HJ
Burakoff
SJ
1997
501
Marcel Dekker
New York
19
Cooke
KR
Hill
GR
Crawford
JM
et al
TNF-α production to LPS stimulation by donor cells predicts the severity of experimental acute graft–versus–host disease.
J Clin Invest.
102
1998
1882
20
Nash
A
Pepe
MS
Storb
R
et al
Acute graft-versus-host disease: analysis of risk factors after allogeneic marrow transplantation and prophylaxis with cyclosporine and methotrexate.
Blood.
80
1992
1838
21
Clift
RA
Buckner
CD
Appelbaum
FR
et al
Allogeneic marrow transplantation in patients with acute myeloid leukemia in first remission: a randomized trial of two irradiation regimens.
Blood.
76
1990
1867
22
Clift
RA
Buckner
CD
Thomas
WI
et al
Marrow transplantation for chronic myeloid leukemia: a randomized study comparing cyclophosphamide and total body irradiation with busulfan and cyclophosphamide.
Blood.
84
1994
2036
23
Deeg
HJ
Spitzer
TR
Cottler-Fox
M
Cahill
R
Pickle
LW
Conditioning-related toxicity and acute graft-versus-host disease in patients given methotrexate/cyclosporine prophylaxis.
Bone Marrow Transplant.
7
1991
193
24
Hattori
K
Hirano
T
Miyajima
H
et al
Differential effects of anti-Fas ligand and anti-tumor necrosis factor alpha antibodies on acute graft-versus-host disease pathologies.
Blood.
91
1998
4051
25
Piguet
PF
Grau
GE
Allet
B
Vassalli
PJ
Tumor necrosis factor/cachectin is an effector of skin and gut lesions of the acute phase of graft-versus-host disease.
J Exp Med.
166
1987
1280
26
Antin
JH
Ferrara
JLM
Cytokine dysregulation and acute graft-versus-host disease.
Blood.
80
1992
2964
27
Braun
YM
Lowin
B
French
L
Acha-Orbea
H
Tschopp
J
Cytotoxic T cells deficient in both functional Fas ligand and perforin show residual cytolytic activity yet lose their capacity to induce lethal acute graft-versus-host disease.
J Exp Med.
183
1996
657
28
Graubert
TA
Russell
JH
Ley
T
The role of granzyme B in murine models of acute graft-versus-host disease and graft rejection.
Blood.
87
1996
1232
29
Mowat
A
Intestinal Graft versus Host Disease.
Graft-vs-Host Disease.
2nd ed.
Ferrara
JLM
Deeg
HJ
Burakoff
SJ
1997
337
Marcel Dekker
New York
30
Garside
P
Reid
S
Steel
M
Mowat
AM
Differential cytokine production associated with distinct phases of murine graft-versus-host reaction.
Immunology.
82
1994
211
31
Madara
JL
Stafford
J
Interferon-γ directly affects barrier function of cultured intestinal epithelial monolayers.
J Clin Invest.
83
1989
724
32
Garside
P
Bunce
C
Tomlinson
RC
Nichois
BL
Mowat
AM
Analysis of the enteropathic effects of tumour necrosis factor α.
Cytokine.
5
1994
24
33
Mowat
AM
Hutton
AK
Garside
P
Steel
MA
A role for IL-1α in immunologically mediated enteropathy.
Immunology.
80
1993
110
34
Mowat
A
Antibodies to IFN-gamma prevent immunological mediated intestinal damage in murine graft-versus-host reactions.
Immunology.
68
1989
18
35
Garside
P
Hutton
AK
Severn
A
Liew
FY
Mowat
AM
Nitric oxide mediates intestinal pathology in graft-vs-host disease.
Eur J Immunol.
22
1992
2141
36
Thiele
DL
Eigenbrodt
ML
Bryde
SE
Eigenbrodt
EH
Lipsky
PE
Intestinal graft-versus-host disease is initiated by donor T cells distinct from classic cytotoxic T lymphocytes.
J Clin Invest.
84
1989
1947
37
Lin
T
Brunner
T
Tietz
B
et al
Fas-ligand mediated killing by intestinal intraepithelial lymphocytes: participation in intestinal graft-versus-host disease.
J Clin Invest.
101
1998
570
38
Jackson
SK
Parton
J
Barnes
RA
Poynton
CH
Fegan
C
Effect of IgM-enriched intravenous immunoglobulin (pentaglobulin) on endotoxaemia and anti-endotoxin antibodies in bone marrow transplantation.
Eur J Clin Invest.
23
1993
540
39
Schwandner
R
Dziarski
R
Wesche
H
Rothe
M
Kirschning
CJ
Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2.
J Biol Chem.
274
1999
17,406
40
Cowdery
JS
Chace
JH
Yi
AK
Krieg
AM
Bacterial DNA induces NK cells to produce IFN-gamma in vivo and increases the toxicity of lipopolysaccharides.
J Immunol.
156
1996
4570
41
Retzlaff
C
Yamamato
Y
Hoffman
PS
Freidman
H
Klein
TW
Bacterial heat shock proteins directly induce cytokine mRNA and interleukin-1 secretion in macrophage cultures.
Infect Immunol.
62
1994
5689
42
Lichtman
A
Krenger
W
Ferrara
JLM
Cytokine Networks: In: Ferrara JLM, Deeg HJ, Burakoff SJ, eds. Graft–Versus–Host Disease.
2nd ed.
1997
179
Marcel Dekker
New York
43
Truitt
RL
Atasoylu
AA
Impact of pretransplant conditioning and donor T cells on chimerism, graft-versus-host disease, graft-versus-leukemia reactivity, and tolerance after bone marrow transplantation.
Blood.
77
1991
2515
44
Truitt
RL
Johnson
BD
McCabe
C
Weiler
MB
Graft versus leukemia.
Graft-vs-Host Disease.
2nd ed.
Ferrara
JLM
Deeg
HJ
Burakoff
SJ
1997
385
Marcel Dekker
New York
45
Teshima
T
Hill
GR
Pan
L
et al
Interleukin-11 improves separate graft-versus-leukemia effects from graft-versus-host disease after bone marrow transplantation.
J Clin Invest.
104
1999
317
46
Pan
L
Teshima
T
Hill
GR
et al
Granulocyte colony-stimulating factor-mobilized allogeneic stem cell transplantation maintains graft-versus-leukemia effects through a perforin-dependent pathway while preventing graft-versus-host disease.
Blood.
93
1999
4071
47
Tsukada
N
Kobata
T
Aivawa
Y
Yagita
H
Okumura
K
Graft-versus-leukemia effect and graft–versus–host disease can be differentiated by cytotoxic mechanisms in a murine model of bone marrow transplantation.
Blood.
93
1999
2738
48
Du
X
Williams
DA
Interleukin IL-11: review of molecular, cell biology, and clinical use.
Blood.
11
1997
3897
49
Du
XX
Neben
T
Goldman
S
Williams
DA
Effects of recombinant human interleukin-11 on hematopoietic reconstitution in transplant mice: acceleration of recovery of peripheral blood neutrophils and platelets.
Blood.
81
1993
27
50
Teicher
BA
Chen
YN
Ara
G
et al
Interaction of interleukin-11 with cytotoxic therapies in vitro against CEM cells and in vivo against EMT-6 murine mammary carcinoma.
Int J Cancer.
67
1996
864
51
Maze
R
Moritz
T
Williams
DA
Increased survival and multilineage hematopoietic protection from delayed and severe myelosuppressive effects of a nitrosourea with recombinant interleukin-11.
Cancer Res.
54
1994
4947
52
Trepicchio
WL
Bozza
M
Pedneault
G
Dorner
AJ
Recombinant human IL-11 attenuates the inflammatory response through down regulation of proinflammatory cytokine release and nitric oxide production.
J Immunol.
157
1996
3627
53
Trepicchio
WL
Wang
L
Bozza
M
Dorner
AJ
Interleukin-11 regulates macrophage effector function through the inhibition of NF-kB.
J Immunol.
159
1997
5661
54
Waxman
AB
Einarsson
O
Seres
T
et al
Targeted lung expression of interleukin-11 enhances murine tolerance of 100% oxygen and diminishes hyperoxia-induced DNA fragmentation.
J Clin Invest.
101
1999
1970
55
Redlich
CA
Gao
X
Rockwell
S
Kelley
M
Elias
JA
IL-11 enhances survival and decreases TNF production after radiation-induced thoracic injury.
J Immunol.
157
1996
1705
56
Qui
BS
Pfieffer
CJ
Keith
JC
Protection by recombinant interleukin-11 against experimental TNB-induced colitis.
Dig Dis Sci.
41
1996
1625
57
Opal
SM
Jhung
JW
Keith
JCJ
Goldman
SJ
Palardy
JE
Parejo
NA
Additive effects of human recombinant interleukin-11 and granulocyte colony-stimulating factor in experimental gram-negative sepsis.
Blood.
93
1999
3467
58
Leng
SX
Elias
JA
Interleukin-11 inhibits macrophage interleukin-12 production.
J Immunol.
159
1997
2161
59
Du
XX
Doerschuk
CM
Orazi
A
Williams
DA
A bone marrow stromal-derived growth factor, interleukin-11, stimulates recovery of small intestinal mucosal cells after cytoablative therapy.
Blood.
83
1994
33
60
Potten
CS
Protection of the small intestinal clonogenic stem cells from radiation-induced damage by pretreatment with interleukin 11 also increases murine survival time.
Int J Cancer.
14
1996
452
61
Keith
JC
Albert
L
Sonis
ST
Pfeiffer
CJ
Schaub
RG
IL-11, apleotropic cytokine: exciting new effects of IL-11 on gastrointestinal mucosal biology.
Stem Cells.
12
1994
79
62
Orazi
A
Du
X
Yang
Z
Kashai
M
Williams
DA
Interleukin-11 prevents apoptosis and accelerates recovery of small intestinal mucosa in mice treated with combined chemotherapy and radiation.
Lab Invest.
75
1996
33
63
Sonis
S
Muska
A
O'Brien
J
Van Vugt
A
Langer-Safer
P
Keith
J
Alteration in the frequency, severity and duration of chemotherapy-induced mucositis in hamsters by IL-11.
Eur J Cancer.
31B
1995
261
64
Liu
Q
Du
XX
Schindel
DT
et al
Trophic effects of IL-11 in rats with experimental short bowel syndrome.
J Pediatr Surg.
31
1996
1047
65
Fiore
NF
Ledniczky
G
Liu
Q
et al
Comparison of interleukin-11 and epidermal growth factor on residual small intestine after massive small bowel resection.
J Pediatr Surg.
33
1999
24
66
Du
X
Liu
Q
Yang
Z
et al
Protective effects of interleukin-11 in a murine model of ischemic bowel necrosis.
Am J Physiol.
272
1997
G545
67
Hill
GR
Cooke
KR
Teshima
T
et al
Interleukin-11 promotes T cell polarization and prevents acute graft-versus-host disease after allogeneic bone marrow transplantation.
J Clin Invest.
102
1998
115
68
Housley
R
Morris
C
Boyle
W
et al
Keratinocyte growth factor induces proliferation of hepatocytes and epithelial cells throughout the rat gastrointestinal tract.
J Clin Invest.
94
1994
1764
69
Pierce
G
Yanagihara
D
Klopchin
K
et al
Stimulation of all epithelial elements during skin regeneration by keratinocyte growth factor.
J Exp Med.
179
1994
831
70
Panos
R
Rubin
J
Aaronson
S
Mason
R
Keratinocyte growth factor/scatter factor are heparin-binding growth factors for alveolar type II cells in fibroblast-conditioned medium.
J Clin Invest.
92
1993
969
71
Ulich
T
Yi
E
Cardiff
R
et al
Keratinocyte growth factor is a growth factor for mammary epithelium in vivo.
Am J Pathol.
144
1994
862
72
Yi
E
Shabaik
A
Lacey
D
et al
Keratinocyte growth factor causes proliferation of urothelium in vivo.
J Urol.
154
1995
1566
73
Farrell
C
Bready
J
Rex
K
et al
Keratinocyte growth factor protects mice from chemotherapy and radiation-induced gastrointestinal injury and mortality.
Cancer Res.
58
1998
933
74
Khan
WB
Shui
C
Ning
S
Knox
SJ
Enhancement of murine intestinal stem cell survival after irradiation by keratinocyte growth factor.
Radiat Res.
148
1997
248
75
Frank
S
Muna
B
Werner
S
The human homologue of a bovine-none-selenium glutathione peroxidase is a novel keratinocyte growth factor-regulated gene.
Oncogene.
14
1997
915
76
Takeoka
M
Ward
W
Pollack
H
Kamp
D
Panos
R
KGF facilitates repair of radiation-induced DNA damage in alveolar epithelial cells.
Am J Physiol.
27
1997
L1174
77
Krijanovski
OI
Hill
GR
Cooke
KR
Teshima
T
Brinson
YS
Ferrara
JLM
Keratinocyte growth factor (KGF) separates graft-versus-leukemia effects from graft-versus-host disease.
Blood.
94
1999
825
78
Panoskaltsis-Mortari
A
Lacey
DL
Vallera
DA
Blazer
BR
Keratinocyte growth factor administered before conditioning ameliorates graft-versus-host disease after allogeneic bone marrow transplantation in mice.
Blood.
92
1998
3960
79
Serdar
C
Heard
R
Prathikanti
D
et al
Safety, pharmacokinetics and biologic activity of rHuKGF in normal volunteers: results of a placebo-controlled randomized double-blind phase 1 study [abstract].
Blood.
90(suppl 1)
1997
172a
80
Adkins
D
Brown
R
Khoury
H
et al
Low-intensity conditioning regimen for related donor allogeneic peripheral blood stem cell (PBSC) transplant using low-dose (550 cGy) high dose rate (30 cGy/min) single-exposure total body irradiation (TBI) [abstract].
Blood.
92
1998
138a
Sign in via your Institution