This study investigated the role of inflammatory cytokines in acute graft-versus-host disease (aGVHD) incidence and severity in 113 patients who underwent reduced-intensity conditioning (RIC) allogeneic stem cell transplantation (allo-SCT). Among all tested cytokines in the first 3 months after allo-SCT, only interleukin-12 p70 (IL-12p70) levels in the first month were significantly associated with grades II to IV aGVHD development (P < .001). IL-12p70 levels were directly correlated with aGVHD severity grade (P < .001). Before aGVHD onset, blood monocytes, the main precursor pool of IL12p70-secreting dendritic cells, recovered more rapidly in patients with grades II to IV aGVHD (P = .005). Similarly, at the effector level, there was a more robust reconstitution of naive CD3+CD4+CD45RA+CD27+ T cells in patients developing grades II to IV aGVHD (P = .006). In multivariate analysis, IL-12p70 level measured in the first month was the strongest predictive factor for aGVHD development (P < .001). These findings, reconstituting a TH1 loop, support a model in which aGVHD reflects a type 1 alloreaction after RIC allo-SCT.

In standard myeloablative allogeneic stem cell transplantation (allo-SCT), several lines of evidence have suggested that inflammatory cytokines act as mediators of acute graft-versus-host disease (aGVHD). Perturbation of the cytokine network may function as a final common pathway of target organ damage, and the rapid onset of severe aGVHD can be considered a “cytokine storm.”1-4  Nonmyeloablative, or reduced-intensity conditioning (RIC), regimens before allo-SCT have emerged as an attractive modality to decrease transplant-related toxicity while preserving the graft-versus-tumor effect.5-7  The use of such less cytotoxic regimens has modified the natural history of transplant-related complications, especially of aGVHD.8,9  Our knowledge of the pathophysiology of aGVHD is based primarily on results of analyses performed in patients who underwent myeloablative allo-SCT. The aim of this study was to investigate the role of inflammatory cytokines on aGVHD incidence and severity in patients who underwent RIC allo-SCT with an HLA-identical sibling.

Patients and methods

One hundred thirteen consecutive patients treated with RIC allo-SCT in different trials at the Institut Paoli-Calmettes (Marseille, France) were included in this study. Written informed consent was obtained from each patient and donor. The studies were approved by the local ethics committee and were performed according to institutional guidelines. Eligibility criteria for RIC allo-SCT are detailed elsewhere.9  Patient and donor characteristics are summarized in Table 1.

Table 1.

Patient and donor characteristics


Characteristic

Data
Median recipient age, y (range)   49 (18-63)  
Sex  
   Recipient, no. male/no. female (% male/% female)   58/55 (51/49)  
   Female donor, no. (%)   52 (46)  
Negative CMV donor-recipient pair, no. (%)   18 (16)  
ABO incompatibility, no. (%)   42 (37)  
Diagnosis, no. (%)  
   Acute myeloblastic leukemia   32 (28)  
   Myelodysplastic syndrome   4 (4)  
   Chronic myeloid leukemia   6 (5)  
   Non-Hodgkin lymphoma   19 (17)  
   Hodgkin lymphoma   3 (3)  
   Multiple myeloma   20 (18)  
   Chronic lymphocytic leukemia   3 (3)  
   Metastatic solid tumor   26 (23)  
Disease status, no. (%)  
   Standard risk*  23 (20)  
   Advanced disease   90 (80)  
Conditioning regimen, no. (%)  
   ATG-based regimen6,9   80 (71)  
   Low-dose irradiation-based regimen   33 (29)  
GVHD prophylaxis, no. (%)  
   CSA alone   70 (62)  
   CSA + mycophenolate mofetil   43 (38)  
Graft source, no. (%)  
   Peripheral blood stem cells   108 (96)  
   Bone marrow   5 (4)  
Graft composition, median (range)  
   CD34+, × 106/kg recipient body weight   6.0 (1.4-37.0)  
   CD3+, × 106/kg recipient body weight
 
327 (14-888)
 

Characteristic

Data
Median recipient age, y (range)   49 (18-63)  
Sex  
   Recipient, no. male/no. female (% male/% female)   58/55 (51/49)  
   Female donor, no. (%)   52 (46)  
Negative CMV donor-recipient pair, no. (%)   18 (16)  
ABO incompatibility, no. (%)   42 (37)  
Diagnosis, no. (%)  
   Acute myeloblastic leukemia   32 (28)  
   Myelodysplastic syndrome   4 (4)  
   Chronic myeloid leukemia   6 (5)  
   Non-Hodgkin lymphoma   19 (17)  
   Hodgkin lymphoma   3 (3)  
   Multiple myeloma   20 (18)  
   Chronic lymphocytic leukemia   3 (3)  
   Metastatic solid tumor   26 (23)  
Disease status, no. (%)  
   Standard risk*  23 (20)  
   Advanced disease   90 (80)  
Conditioning regimen, no. (%)  
   ATG-based regimen6,9   80 (71)  
   Low-dose irradiation-based regimen   33 (29)  
GVHD prophylaxis, no. (%)  
   CSA alone   70 (62)  
   CSA + mycophenolate mofetil   43 (38)  
Graft source, no. (%)  
   Peripheral blood stem cells   108 (96)  
   Bone marrow   5 (4)  
Graft composition, median (range)  
   CD34+, × 106/kg recipient body weight   6.0 (1.4-37.0)  
   CD3+, × 106/kg recipient body weight
 
327 (14-888)
 

N = 113 patients and donors.

CMV indicates cytomegalovirus.

*

Standard risk disease: chronic myeloid leukemia in chronic phase, acute leukemia in first complete remission

Transplantation procedures

The RIC regimen included either fludarabine, oral busulfan, and antithymocyte globulin (ATG) (Thymoglobuline; Genzyme, Lyon, France)6,9  or fludarabine and low-dose total body irradiation.7  Supportive care has been previously reported and was similar during the entire study period.10  GVHD prophylaxis was performed with cyclosporin A (CSA) alone9  or with CSA and mycophenolate mofetil.7  Most (96%) patients received a peripheral blood stem cell allograft mobilized with granulocyte-colony-stimulating factor (G-CSF) (10 μg/kg per day).

Cytokine measurement and phenotypic analysis

Plasma samples were collected and stored at -80°C according to a prespecified schedule applied to all patients during the entire study period. Times for cytokine measurements were as follows: (1) before allo-SCT: median, day -7 (range, -23 to -3); (2) day 0: all procedures performed on day 0; (3) 1 month after allo-SCT: median, day 28 (range, days 20-41); (4) 2 months after allo-SCT: median, day 59 (range, days 49-75); and (5) 3 months after allo-SCT: median, day 91 (range, days 76-114). The levels of 10 different cytokines (interleukin-1β [IL-1β], IL-6, IL-8, IL-10, IL-12p70, IL-18, tumor necrosis factor alpha [TNF-α], interferon-alpha [IFN-α], IFN-γ, and Fas-ligand) were measured on these collected samples using commercially available enzyme-linked immunosorbent assay (ELISA) kits according to the manufacturer's instructions. Naive CD4+ T lymphocytes were identified by 4-color flow cytometry using the CD3, CD4, CD45RA, and CD27 mAbs (Beckman-Coulter, Marseille, France) as previously described.11  Monocytes were counted by an automated counter.

Clinical outcomes and statistical methods

Acute GVHD was evaluated according to standard criteria. On diagnosis of grades II to IV aGVHD, all patients were primarily treated with CSA and methylprednisolone (2 mg/kg per day). Data were computed using SEM software (Silex, Mirefleurs, France) and SPSS 9.0 for Windows (SPSS, Chicago, IL). The Mann-Whitney U test was used for comparison of continuous variables. Categorical variables were compared using the χ2 test. The association of time to aGVHD with cytokine levels and other relevant variables was evaluated in a multivariate analysis with the use of a Cox proportional hazards regression model.12 

Table 2 shows the results and kinetics of the different cytokines measured in the entire study population. Except for IL-12p70, all other cytokines showed little variation in the blood in the first 3 months after allo-SCT. When looking at the kinetics of IL-12p70 secretion before and after allo-SCT, there was a dramatically different pattern between patients with grades II to IV aGVHD and patients with grades 0 to I aGVHD (Figure 1A). With a median follow-up of 470 (range, 125-2175) days, the incidence of grades II to IV aGVHD was 45% (95% confidence interval [CI], 36%-54%; median onset, 32 days after allo-SCT). Forty-seven (42%) patients did not experience aGVHD, and 15 (13%) patients had grade I aGVHD. In univariate analysis performed in the subgroup of 75 patients for whom all tested cytokines were measured closely and rigorously before aGVHD clinical onset, advanced disease stage (P = .04) and high IL-12p70 level (P < .001) measured around the first month after allo-SCT were significantly associated with the development of clinically significant grades II to IV aGVHD. Levels of all other cytokines did not show any significant correlation with the risk for aGVHD (Table 3). IL-12p70 levels were significantly correlated with the severity of aGVHD: grades 0 to I, median 468 pg/mL; grade II, median 2538 pg/mL; and grades III to IV, median 4615 pg/mL (P < .001; Figure 1B). In patients experiencing grades II to IV aGVHD, IL-12p70 levels decreased after aGVHD therapy (Figure 1A).

Table 2.

Cytokine levels measured before and after RIC allo-SCT



Median level (range)
Cytokine
Before allo-SCT
Day 0
1 mo after allo-SCT
2 mo after allo-SCT
3 mo after allo-SCT
IL-1β, pg/mL   5 (0-29)   0 (0-1062)   0 (0-496)   0 (0-675)   0 (0-23)  
IL-6, pg/mL   0 (0-265)   0 (0-63)   0 (0-667)   0 (0-13)   0 (0-411)  
IL-8, pg/mL   0 (0-911)   0   0 (0-630)   0 (0-149)   0 (0-139)  
IL-10, pg/mL   0 (0-51)   0 (0-142)   0 (0-71)   0 (0-124)   0 (0-664)  
IL-12p70, pg/mL   1246 (41-2643)   768 (0-2831)   1669 (0-9000)   885 (0-6790)   1973 (0-4112)  
IL-18, pg/mL   0 (0-452)   0 (0-1145)   0 (0-10453)   0 (0-3579)   0 (0-20587)  
IFN-α, pg/mL   0   0 (0-768)   0 (0-817)   0   0  
IFN-γ, pg/mL   0 (0-39)   8 (0-225)   0 (0-478)   0 (0-167)   3 (0-240)  
Fas-ligand, ng/mL   0 (0-1)   0   0 (0-1)   0 (0-15)   0 (0-1)  
TNF-α, pg/mL
 
0 (0-64)
 
0 (0-134)
 
0 (0-200)
 
0
 
0
 


Median level (range)
Cytokine
Before allo-SCT
Day 0
1 mo after allo-SCT
2 mo after allo-SCT
3 mo after allo-SCT
IL-1β, pg/mL   5 (0-29)   0 (0-1062)   0 (0-496)   0 (0-675)   0 (0-23)  
IL-6, pg/mL   0 (0-265)   0 (0-63)   0 (0-667)   0 (0-13)   0 (0-411)  
IL-8, pg/mL   0 (0-911)   0   0 (0-630)   0 (0-149)   0 (0-139)  
IL-10, pg/mL   0 (0-51)   0 (0-142)   0 (0-71)   0 (0-124)   0 (0-664)  
IL-12p70, pg/mL   1246 (41-2643)   768 (0-2831)   1669 (0-9000)   885 (0-6790)   1973 (0-4112)  
IL-18, pg/mL   0 (0-452)   0 (0-1145)   0 (0-10453)   0 (0-3579)   0 (0-20587)  
IFN-α, pg/mL   0   0 (0-768)   0 (0-817)   0   0  
IFN-γ, pg/mL   0 (0-39)   8 (0-225)   0 (0-478)   0 (0-167)   3 (0-240)  
Fas-ligand, ng/mL   0 (0-1)   0   0 (0-1)   0 (0-15)   0 (0-1)  
TNF-α, pg/mL
 
0 (0-64)
 
0 (0-134)
 
0 (0-200)
 
0
 
0
 

Absence of range indication means that the cytokine was not detected in any patient at the corresponding time point. Cytokine levels were measured by ELISA at regular intervals in the first 3 months after RIC allo-SCT. Median times for cytokine measurements were as follows: before allo-SCT: median, day –7 (range, –23 to –3); day 0, all procedures performed on day 0; 1 month after allo-SCT: median, day 28 (range, days 20-41); 2 months after allo-SCT: median, day 59 (range, days 49-75); and 3 months after allo-SCT: median, day 91 (range, days 76-114).

Table 3.

Univariate analysis of risk factors for occurrence of aGVHD


Characteristic

Grades 0 to I aGVHD

Grades II to IV aGVHD

P
No. patients   41   34   
Median recipient age, y (range)   48 (33-63)   51 (27-63)   .55  
Female donor, no. (%)   19 (46)   12 (35)   .33  
Negative CMV donor-recipient pair, no. (%)   8 (19)   3 (9)   .32  
Diagnosis, no. (%)    .12  
   Myeloid malignancy   20 (49)   9 (26)   
   Lymphoid malignancy   11 (27)   15 (44)   
   Metastatic solid tumor   10 (24)   10 (29)   
Disease status, no. (%)    .04  
   Standard risk*  13 (32)   4 (12)   
   Advanced disease   28 (68)   30 (88)   
Conditioning regimen, no. (%)    .66  
   ATG-based regimen   27 (66)   24 (71)   
   Low-dose irradiation-based regimen   14 (34)   10 (29)   
GVHD prophylaxis, no. (%)    .82  
   CSA alone   24 (59)   19 (56)   
   CSA + mycophenolate mofetil   17 (41)   15 (44)   
Stem cell source, no. (%)    .56  
   Bone marrow   2 (5)   0   
   Peripheral blood stem cells   39 (95)   34 (100)   
Median graft composition, (range, %)    
   CD34+, × 106/kg recipient body weight   6.2 (2.0-12.9)   5.5 (1.4-22.2)   .68  
   CD3+, × 106/kg recipient body weight   309 (14-689)   328 (131-617)   .61  
Cytokine level, median (range)    
   IL-1β, pg/mL   0 (0-17)   0 (0-496)   .72  
   IL-6, pg/mL   0 (0-667)   0 (0-33)   .66  
   IL-8, pg/mL   0 (0-336)   0 (0-630)   .75  
   IFN-α, pg/mL   0   0 (0-817)   .65  
   IFN-γ, pg/mL   0 (0-478)   0 (0-214)   .54  
   IL-10, pg/mL   0 (0-71)   0 (0-36)   .21  
   IL-12p70, pg/mL   468 (0-4638)   3606 (0-9000)   < .001  
   IL-18, pg/mL   0 (0-1495)   0 (0-10453)   .21  
   TNF-α, pg/mL   0 (0-1)   0 (0-200)   .84  
   Fas-ligand, ng/mL
 
0 (0-0.22)
 
0 (0-0.64)
 
.53
 

Characteristic

Grades 0 to I aGVHD

Grades II to IV aGVHD

P
No. patients   41   34   
Median recipient age, y (range)   48 (33-63)   51 (27-63)   .55  
Female donor, no. (%)   19 (46)   12 (35)   .33  
Negative CMV donor-recipient pair, no. (%)   8 (19)   3 (9)   .32  
Diagnosis, no. (%)    .12  
   Myeloid malignancy   20 (49)   9 (26)   
   Lymphoid malignancy   11 (27)   15 (44)   
   Metastatic solid tumor   10 (24)   10 (29)   
Disease status, no. (%)    .04  
   Standard risk*  13 (32)   4 (12)   
   Advanced disease   28 (68)   30 (88)   
Conditioning regimen, no. (%)    .66  
   ATG-based regimen   27 (66)   24 (71)   
   Low-dose irradiation-based regimen   14 (34)   10 (29)   
GVHD prophylaxis, no. (%)    .82  
   CSA alone   24 (59)   19 (56)   
   CSA + mycophenolate mofetil   17 (41)   15 (44)   
Stem cell source, no. (%)    .56  
   Bone marrow   2 (5)   0   
   Peripheral blood stem cells   39 (95)   34 (100)   
Median graft composition, (range, %)    
   CD34+, × 106/kg recipient body weight   6.2 (2.0-12.9)   5.5 (1.4-22.2)   .68  
   CD3+, × 106/kg recipient body weight   309 (14-689)   328 (131-617)   .61  
Cytokine level, median (range)    
   IL-1β, pg/mL   0 (0-17)   0 (0-496)   .72  
   IL-6, pg/mL   0 (0-667)   0 (0-33)   .66  
   IL-8, pg/mL   0 (0-336)   0 (0-630)   .75  
   IFN-α, pg/mL   0   0 (0-817)   .65  
   IFN-γ, pg/mL   0 (0-478)   0 (0-214)   .54  
   IL-10, pg/mL   0 (0-71)   0 (0-36)   .21  
   IL-12p70, pg/mL   468 (0-4638)   3606 (0-9000)   < .001  
   IL-18, pg/mL   0 (0-1495)   0 (0-10453)   .21  
   TNF-α, pg/mL   0 (0-1)   0 (0-200)   .84  
   Fas-ligand, ng/mL
 
0 (0-0.22)
 
0 (0-0.64)
 
.53
 

Variables with P < .10 in univariate analysis were included in the multivariate analysis.

CMV indicates cytomegalovirus.

*

Standard risk disease: chronic myeloid leukemia in chronic phase, acute leukemia in first complete remission

Given that IL-12p70 is mainly produced by myeloid dendritic cells (DCs) and that peripheral blood monocytes are considered the main pool of DC precursors,13  we investigated the recovery of monocytes in the groups of patients with and without clinically significant aGVHD. Interestingly, we found more rapid recovery of monocytes before aGVHD clinical onset in patients with grades II to IV aGVHD than in patients with grades 0 to I aGVHD (median, 829/μL vs 552/μL; P = .005; Figure 1C).

At the effector level, naive CD4+ lymphocytes are the principal target for IL-12p70, which induces their differentiation into TH1 effectors.13,14  Here, we observed a significantly more robust recovery of genuine naive CD3+CD4+CD45RA+CD27+ T cells before aGVHD clinical onset in patients with grades II to IV aGVHD compared with patients with grades 0 to I aGVHD (median, 50/μL vs 16/μL; P = .006; Figure 1D). Finally, in multivariate analysis, IL-12p70 level measured before aGVHD clinical onset was the strongest predictive factor for aGVHD development and severity (P < .001; relative risk [RR], 10.7; 95% CI, 3.8-30.6).

Figure 1.

IL-12p70 plasma levels correlate with aGVHD incidence and severity. (A) Kinetics of IL-12p70 secretion before RIC allo-SCT at the time of graft infusion (day 0) and in the first 3 months after RIC allo-SCT. Median levels and SEMs are shown for patients with (▴) grades 0 to I aGVHD and patients with (▪) grades II to IV aGVHD. (B) Correlation of IL-12p70 levels with aGVHD grade in the group of 75 patients in whom IL-12p70 was measured closely and rigorously before aGVHD clinical onset in the first month after RIC allo-SCT. (C) Peripheral blood monocyte recovery. (D) Naive CD3+CD4+CD45RA+CD27+ T lymphocyte recovery as measured in the group of 75 patients closely and rigorously before aGVHD clinical onset in the first month after RIC allo-SCT. As detailed in “Patients and methods,” plasma samples were collected according to a prespecified schedule applied to all patients during the entire study period. Data represent the same single time point analysis for all patients, not the maximum level for each patient within the first month. IL-12p70 levels were measured by standard ELISA. Monocytes were counted by an automated counter. Naive CD4+ T lymphocytes were identified by 4-color flow cytometry. Short horizontal lines indicate the median values.

Figure 1.

IL-12p70 plasma levels correlate with aGVHD incidence and severity. (A) Kinetics of IL-12p70 secretion before RIC allo-SCT at the time of graft infusion (day 0) and in the first 3 months after RIC allo-SCT. Median levels and SEMs are shown for patients with (▴) grades 0 to I aGVHD and patients with (▪) grades II to IV aGVHD. (B) Correlation of IL-12p70 levels with aGVHD grade in the group of 75 patients in whom IL-12p70 was measured closely and rigorously before aGVHD clinical onset in the first month after RIC allo-SCT. (C) Peripheral blood monocyte recovery. (D) Naive CD3+CD4+CD45RA+CD27+ T lymphocyte recovery as measured in the group of 75 patients closely and rigorously before aGVHD clinical onset in the first month after RIC allo-SCT. As detailed in “Patients and methods,” plasma samples were collected according to a prespecified schedule applied to all patients during the entire study period. Data represent the same single time point analysis for all patients, not the maximum level for each patient within the first month. IL-12p70 levels were measured by standard ELISA. Monocytes were counted by an automated counter. Naive CD4+ T lymphocytes were identified by 4-color flow cytometry. Short horizontal lines indicate the median values.

Close modal

Previous studies established that aGVHD pathophysiology after myeloablative conditioning can be summarized as a 3-step process: phase 1, during which inflammatory mediators are produced and a proinflammatory environment is established; phase 2, during which donor T lymphocytes are activated and T-cell target interactions can take place; and phase 3, during which T cells specifically kill target host cells through various mechanisms. Other effectors, such as natural killer (NK) cells, monocytes, granulocytes, and DCs, may cause specific and nonspecific tissue lesions.15  Our study showed a specific kinetic profile and a marked increase in circulating IL-12p70 levels that significantly correlated with the incidence and severity of aGVHD, highlighting its major role in aGVHD pathogenesis. Indeed, direct and indirect evidence suggests that aGVHD is mediated by the action of donor-derived T lymphocytes thought to polarize into type 1 T cells after stimulation with IL-12 from DCs.16,17  Most important, in our study, peak secretion of IL-12p70 was concomitant with the expansion of naive CD4+ T cells, but not with other CD4+ T-cell subsets (data not shown). Type 1 lymphocytes composed of CD4+ TH1 and CD8+ T cytotoxic (Tc1) cells produce IFN-γ and Fas-ligand that severely injure multiple organs, leading to aGVHD.16,18  The role of IL-12p70 in aGVHD initiation is supported by the observation that IL-12 administration stimulates aGVHD development in a murine strain combination that normally develops a TH 2-type, autoimmune chronic GVHD syndrome, further emphasizing the implication of an afferent TH1-type response in the generation of aGVHD.19  The IL-12-driven TH1-type processes involve not only CD4+ and CD8+ T-cell responses but also a phase of monocyte/macrophage activation with the production of inflammatory cytokines such as TNF-α, IL-1, and IL-6, leading to the classical “cytokine storm” of aGVHD. In our study, IL-12p70 was the only cytokine that showed a significant correlation with aGVHD. This apparent discrepancy might be explained by the fact that some studies demonstrating the implication of other proinflammatory cytokines have been performed at the level of mRNA cytokine expression in GVHD target organs20-22  and by the difference in conditioning-related inflammation2  that is less broad and less nonspecific in our study, allowing detection of the true TH1 response.

Although triggering signals for aGVHD induction, such as the role of toll-like receptors, are yet to be established, our findings are in line with previous data showing a role for IL-18, another IFN-γ-inducing cytokine, in aGVHD pathophysiology.23  Here, we could reconstitute a genuine TH1 loop, supporting a model in which aGVHD primarily reflects a type 1 alloreaction (rapid monocytes/DC recovery, IL-12p70 secretion, naive CD4+ T-cell expansion, TH1 and Tc1 cell differentiation) in the context of RIC allo-SCT, consistent with data from the myeloablative setting indicating a major role of the antigen-presenting cell (APC) network (monocyte/macrophage/DC) in the pathophysiology of aGVHD.24  However, this study did not address the role of APC origin in aGVHD initiation. Indeed, given the specific role of host-derived APCs compared with donor-derived APCs in the initiation of aGVHD,25  monitoring of the chimerism status of APCs, especially at the site of aGVHD target organs, would allow further insight into aGVHD mechanisms in the RIC setting.

Obviously, IL-12p70 is not the exclusive cytokine involved in aGVHD pathogenesis after RIC allo-SCT.26  However, the fine functions of immune effectors tend to be more evident in less toxic regimens,11,27  offering new opportunities for a better understanding of aGVHD pathophysiology. From a practical point of view, close and early monitoring of IL-12p70 can be a useful indicator of aGVHD, and anti-IL-12-based therapy can represent a potential tool for controlling alloreactivity.

Prepublished online as Blood First Edition Paper, September 1, 2005; DOI 10.1182/blood-2005-07-2919.

Supported by several grants from the French Ministry of Health as part of the Programme Hospitalier de Recherche Clinique (PHRC). Also supported by an “interface” research contract sponsored by the INSERM (M.M.).

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.

We thank N. Baratier for technical assistance, the nursing staff for providing excellent care for our patients, and the physicians of the Hematology and Medical Oncology Departments at the Institut Paoli-Calmettes for their important study contributions and dedicated patient care. We thank D. Maraninchi and C. Mawas for their continuous support. We also thank the “Association pour la Recherche sur le Cancer (ARC),” the “Ligue Nationale contre le Cancer,” the “Fondation de France,” the “Fondation contre la Leucémie,” the “Etablissement Français des Greffes (EFG),” the “Association Cent pour Sang la Vie,” the “Association Laurette Fuguain,” and the GEFLUC, for their generous and continuous support for our clinical work and basic research.

1
Holler E, Kolb HJ, Moller A, et al. Increased serum levels of tumor necrosis factor alpha precede major complications of bone marrow transplantation.
Blood
.
1990
;
75
:
1011
-1016.
2
Holler E, Kolb HJ, Mittermuller J, et al. Modulation of acute graft-versus-host-disease after allogeneic bone marrow transplantation by tumor necrosis factor alpha (TNF alpha) release in the course of pretransplant conditioning: role of conditioning regimens and prophylactic application of a monoclonal antibody neutralizing human TNF alpha (MAK 195F).
Blood
.
1995
;
86
:
890
-899.
3
Krenger W, Ferrara JL. Dysregulation of cytokines during graft-versus-host disease.
J Hematother
.
1996
;
5
:
3
-14.
4
Krenger W, Hill GR, Ferrara JL. Cytokine cascades in acute graft-versus-host disease.
Transplantation
.
1997
;
64
:
553
-558.
5
Giralt S, Estey E, Albitar M, et al. Engraftment of allogeneic hematopoietic progenitor cells with purine analog-containing chemotherapy: harnessing graft-versus-leukemia without myeloablative therapy.
Blood
.
1997
;
89
:
4531
-4536.
6
Slavin S, Nagler A, Naparstek E, et al. Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases.
Blood
.
1998
;
91
:
756
-763.
7
Niederwieser D, Maris M, Shizuru JA, et al. Lowdose total body irradiation (TBI) and fludarabine followed by hematopoietic cell transplantation (HCT) from HLA-matched or mismatched unrelated donors and postgrafting immunosuppression with cyclosporine and mycophenolate mofetil (MMF) can induce durable complete chimerism and sustained remissions in patients with hematological diseases.
Blood
.
2003
;
101
:
1620
-1629.
8
Mielcarek M, Martin PJ, Leisenring W, et al. Graft-versus-host disease after nonmyeloablative versus conventional hematopoietic stem cell transplantation.
Blood
.
2003
;
102
:
756
-762.
9
Mohty M, Bay JO, Faucher C, et al. Graft-versus-host disease following allogeneic transplantation from HLA-identical sibling with antithymocyte globulin-based reduced-intensity preparative regimen.
Blood
.
2003
;
102
:
470
-476.
10
Mohty M, Jacot W, Faucher C, et al. Infectious complications following allogeneic HLA-identical sibling transplantation with antithymocyte globulin-based reduced intensity preparative regimen.
Leukemia
.
2003
;
17
:
2168
-2177.
11
Mohty M, Blaise D, Faucher C, et al. Impact of plasmacytoid dendritic cells on outcome after reduced-intensity conditioning allogeneic stem cell transplantation.
Leukemia
.
2005
;
19
:
1
-6.
12
Cox DR. Regression models and life-tables (with discussions), Series B.
J R Stat Soc
.
1972
;
34
:
187
-220.
13
Rissoan MC, Soumelis V, Kadowaki N, et al. Reciprocal control of T helper cell and dendritic cell differentiation.
Science
.
1999
;
283
:
1183
-1186.
14
Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity.
Nat Rev Immunol
.
2003
;
3
:
133
-146.
15
Ferrara JL, Levy R, Chao NJ. Pathophysiologic mechanisms of acute graft-vs-host disease.
Biol Blood Marrow Transplant
.
1999
;
5
:
347
-356.
16
Krenger W, Ferrara JL. Graft-versus-host disease and the Th1/Th2 paradigm.
Immunol Res
.
1996
;
15
:
50
-73.
17
Williamson E, Garside P, Bradley JA, Mowat AM. IL-12 is a central mediator of acute graft-versus-host disease in mice.
J Immunol
.
1996
;
157
:
689
-699.
18
Krenger W, Cooke KR, Crawford JM, et al. Transplantation of polarized type 2 donor T cells reduces mortality caused by experimental graft-versus-host disease.
Transplantation
.
1996
;
62
:
1278
-1285.
19
Via CS, Rus V, Gately MK, Finkelman FD. IL-12 stimulates the development of acute graft-versus-host disease in mice that normally would develop chronic, autoimmune graft-versus-host disease.
J Immunol
.
1994
;
153
:
4040
-4047.
20
Guy-Grand D, Vassalli P. Gut injury in mouse graft-versus-host reaction: study of its occurrence and mechanisms.
J Clin Invest
.
1986
;
77
:
1584
-1595.
21
Piguet PF, Grau GE, Allet B, Vassalli P. Tumor necrosis factor/cachectin is an effector of skin and gut lesions of the acute phase of graft-vs-host disease.
J Exp Med
.
1987
;
166
:
1280
-1289.
22
Ferrara JL, Abhyankar S, Gilliland DG. Cytokine storm of graft-versus-host disease: a critical effector role for interleukin-1.
Transplant Proc
.
1993
;
25
:
1216
-1217.
23
Reddy P, Ferrara JL. Role of interleukin-18 in acute graft-vs-host disease.
J Lab Clin Med
.
2003
;
141
:
365
-371.
24
Holler E, Rogler G, Herfarth H, et al. Both donor and recipient NOD2/CARD15 mutations associate with transplant-related mortality and GvHD following allogeneic stem cell transplantation.
Blood
.
2004
;
104
:
889
-894.
25
Shlomchik WD, Couzens MS, Tang CB, et al. Prevention of graft versus host disease by inactivation of host antigen-presenting cells.
Science
.
1999
;
285
:
412
-415.
26
Kaiser T, Kamal H, Rank A, et al. Proteomics applied to the clinical follow-up of patients after allogeneic hematopoietic stem cell transplantation.
Blood
.
2004
;
104
:
340
-349.
27
Mohty M, Bagattini S, Chabannon C, et al. CD8+ T cell dose affects development of acute graft-vs-host disease following reduced-intensity conditioning allogeneic peripheral blood stem cell transplantation.
Exp Hematol
.
2004
;
32
:
1097
-1102.
Sign in via your Institution