Abstract
Background A variety of immune pathways can lead to graft-versus-host disease. A better understanding of the type of immune response causing graft-versus-host disease in defined clinical hematopoietic stem cell transplant settings is required to inform development of methods for monitoring patients and providing them tailored care.Design and Methods Twenty-five patients were recruited presenting with myeloid malignancies and treated with a reduced intensity conditioning transplant regimen with graft-versus-host disease prophylaxis comprising in vivo lymphocyte depletion with alemtuzumab and cyclosporin. A prospective study was performed of lymphocyte subset reconstitution in peripheral blood in relation to the incidence of graft-versus-host disease.Results Acute graft-versus-host disease was associated with significantly higher numbers of natural killer cells and donor-derived effector CD4 T cells (CD45RO+ CD27−) early (day 30) after transplantation (p=0.04 and p=0.02, respectively). This association was evident before the emergence of clinical pathology in six out of seven patients. Although numbers of regulatory CD4 T cells (CD25high Foxp3+) were similar at day 30 in all patients, a significant deficit in those who developed acute graft-versus-host disease was apparent relative to effector CD4 T cells (median of 41 effectors per regulatory cell compared to 12 to 1 for patients without graft-versus-host disease) (p=0.03). By day 180, a functional regulatory CD4 T-cell population had expanded significantly in patients who developed chronic graft-versus-host disease, reversing the imbalance (median of 3 effectors per regulatory cell compared to 9.6 to 1 for patients without graft-versus-host disease) (p=0.018) suggesting no overt absence of immune regulation in the late onset form of the disease.Conclusions Imbalance of effector and regulatory CD4 T cells is a signature of graft-versus-host disease in this transplantation protocol.Introduction
Graft-versus-host disease (GvHD) can be a life-threatening complication of allogeneic hematopoietic stem cell transplantation (HSCT). Both the incidence and severity of GvHD are reduced by lymphocyte-depletion strategies, such as use of the anti-CD52 cytolytic mono-clonal antibody called alemtuzumab (Campath 1H), and pharmacological immunosuppression. These non-specific approaches, however, leave patients vulnerable to infection and at increased risk of leukemia relapse due to loss of a beneficial graft-versus-leukemia (GvL) immune response. The problem of disease relapse is especially pertinent for patients treated with reduced intensity conditioning regimens that utilize lower doses of chemotherapy or irradiation and rely, to varying extents, on the GvL response to eliminate disease.1 Treatment regimens ideally should be based on prudent use of immunosuppression sufficient to prevent GvHD without excessive compromise of the GvL response. At present, the diagnosis of GvHD is mainly based on clinical features. The disease is, therefore, recognized only after emergence of pathology and treatment is reactionary. A better understanding of the immunological mechanisms involved in GvHD is required for the development of predictive diagnostic methods. This would allow segregation of patients according to risk and judicious tailoring of immunosuppression.
GvHD is caused by alloreactive donor lymphocytes that attack patient’s cells due to recognition of foreign minor histocompatibility antigens or major histocompatibility antigens in the case of HLA-mismatched transplants.2 The disease has distinct acute and chronic forms. The physiology of late-onset, chronic GvHD is distinct from that of the early, acute form of disease, the cause of the pathology is poorly understood and the disease does not respond well to conventional therapies.3
A variety of immune effector pathways can lead to GvHD pathology. The αβ T cells are principle players, and depletion of either CD4 or CD8 T cells can reduce clinical GvHD4,5 but few studies have examined the role of T-cell subsets. Chronic GvHD has been associated with an increase in CD4 effector memory,6 increase in CD8 central memory and decrease in CD4 central memory cells.7 In acute GvHD, a correlation was found with prevalence of naïve and central memory CD4 T cells in the donor allograft.8 Naïve human T cells appear more alloreactive than memory T cells in vitro9 but pathogen-specific memory T cells that cross-react with alloantigens have been implicated in the development of GvHD.10–12 Natural killer (NK) cells also exhibit alloreactivity, but clinical studies indicate an ambiguous role in GvHD. Some reports describe an association of NK cell allo-disparities with an increased incidence of GvHD13,14 but others found no impact15,16 and some studies have shown rapid NK recovery can have a beneficial effect on GvHD.17,18 Involvement of B cells is indicated by a correlation between chronic GvHD and increased levels of antibodies specific for minor histocompatibility antigens19 together with the demonstration that some patients respond to B-cell depletion therapy.20
Failure of regulatory mechanisms may also play a role in GvHD. Studies using mouse models have shown regulatory CD4 T cells can exert a beneficial immune suppressive effect in GvHD.21–24 Results of clinical studies to assess the role of regulatory T cells in GvHD are less clear. In some studies a deficit of these cells was found in GvHD,25–27 in other studies no difference was recorded between patients with or without disease28,29 and yet other studies indicated that regulatory T cells may be increased in patients with chronic GvHD.30,31
The redundancy of mechanisms potentially involved in GvHD hinders development of new diagnostic methods and therapies. The type of immune response likely reflects the immune status of the patient and donor, the transplant preparative regimen and underlying diseases. Consequently there is a need to understand the mechanistic basis of GvHD that develops for individual treatment protocols. We performed a longitudinal study of lymphocyte subset reconstitution after allogeneic HSCT in 25 patients, all presenting with myeloid disease, who received peripheral blood stem cell transplants after a uniform reduced-intensity conditioning regimen comprising the chemotoxic agents fludarabine and busul-phan.32 GvHD prophylaxis was based on in vivo immune depletion with alemtuzumab together with cyclosporin. After this preparative treatment the incidence of acute GvHD was 36% and that of chronic GvHD was 22%.33 The dynamics of naïve, memory and effector T cells, regulatory CD4 T cells, NK and B-cell recovery were correlated with GvHD incidence to indicate the key players driving the disease. The information gained provides the essential basis for identifying patients at risk of GvHD and improving disease control by selecting treatments appropriate to the type of immune response involved.
Design and Methods
Patients and transplant regimen
A prospective study was performed of 25 patients who underwent allogeneic HSCT for myeloid malignancies between September 2005 and September 2006 at King’s College Hospital. The transplant preparative regimen consisted of fludarabine (30 mg/m daily, administered intravenously from day -9 to day -5), busulphan (3.2 mg/kg body weight, administered intravenously in four divided doses from day -3 to day -2), and alemtuzumab (20 mg/day intravenously on days -8 to day -4). Unselected allogeneic peripheral blood stem cells were infused on day 0. Intravenous cyclosporin was started from day -1 as GvHD prophylaxis at a dose adjusted to achieve plasma trough levels of 150–200 ng/L for all patients. Oral cyclosporin was substituted when a good oral intake was achieved and rapidly tapered to discontinuation from day 60 in the absence of GvHD. Acute and chronic GvHD were graded using standard criteria.34,35 Recombinant granulocyte colony-stimulating factor (G-CSF) was administered subcutaneously or intravenously from day +7 until neutrophil engraftment. The patients’ characteristics are shown in Table 1. Clinical data were censored at May 2007. Peripheral blood samples were collected immediately prior to conditioning for the transplant and at days 30, 60, 90, 180, 270 and 360 after transplantation. Samples of peripheral blood were also collected from 11 healthy age-matched individuals (median age 51 years; range, 41–56 years). King’s College Hospital Research Ethics Committee approved the use of the patients’ samples and The Royal Free Hospital Research Ethics Committee approved the use of the samples from healthy volunteers. Written informed consent was obtained from all participants.
Immunophenotypic analysis
Lymphocyte subsets were enumerated in whole peripheral blood using fluorochrome-labeled monoclonal antibodies to CD4 (clone SK3), CD8 (SK1), CD25 (2A3), CD27 (M-T271), CD45RO (UCHL1), CD56 (B159), (BD Biosciences) and CD3 (OKT3), CD19 (HIB19), CD31 (WM59), CD45RA (HI100), CD62L (Dreg 56), FoxP3 (PCH101), and rat IgG2a isotype control (eBR2a) (eBioscience). Cells in 200 μL peripheral blood were stained for surface markers and erythrocytes were removed using FACS lysing solution (BD Biosciences). Intracellular Foxp3 staining was performed after permeabilization (BD Biosciences Cytofix/Cytoperm solution) according to the manufacturer’s instructions. Eight-color analysis was performed by flow cytometry using a BD FACSCanto II (BD Biosciences) and results analyzed with FlowJo software (TreeStar). NK cells were defined as CD3 CD56. B cells were defined as CD19. CD3 CD4 and CD3 CD8 T-cell subsets were defined as CD45ROCD27 naïve, CD45RO CD27 CD62L central memory, CD45RO CD27 CD62L effector memory, CD45RO CD27 effectors and CD45RO CD27 terminal effectors. CD4 regulatory T cells were defined as CD4 CD25, Foxp3. CD4 T-cell recent thymic emigrants were defined as CD4 CD45RA CD31 CD62L. Cell subset numbers were calculated from percentage values based on an absolute lymphocyte count of the blood sample obtained using an automated leukocyte counter.
Chimerism
Peripheral blood mononuclear cells were purified by density gradient centrifugation on Lympholyte-H (Cedarlane Laboratories) and CD4 T-cell subsets isolated using a FACSAria sorter after surface staining with CD3, CD4, CD45RO and CD27 antibodies. Purity of the populations was >95%. Cells were lysed with proteinase K (0.2 mg/mL in 1 mM EDTA, 20 mM Tris-HCl pH 8.0, 1% Tween-20). Donor and recipient composition was determined by polymerase chain reaction amplification of informative alleles from 15 polymorphic short tandem repeat loci and the sex-determining amelogenin loci (Powerplex; Promega Corp, Madison, WI, USA). Products were separated by capillary electrophoresis using an ABI 3130XL DNA sequencer and results analyzed using Genemapper 4.0 software (Applied Biosystems). Quantification was based on area under the peaks. The sensitivity of this methodology was previously shown to be 5% by cell dilution studies.36
T-cell function
Suppressive activity of CD4 CD25 regulatory T cells from patients was measured by their ability to inhibit proliferation of CD4 CD25 T cells from a healthy volunteer. Cell subsets were purified by immunomagnetic cell sorting using a regulatory T-cell isolation kit and an AutoMACS separator (Miltenyi Biotec). CD4 CD25 cells (5,000) were cultured in RPMI 1640 medium containing 10% human AB serum and stimulated with anti-CD3/CD28 antibody coated beads at a ratio of one bead to five cells (Dynabeads human Treg expander, Invitrogen) in the presence or absence of CD4 CD25 cells (5,000). After 5 days, 1 μCi of [H]-labeled thymidine was added and cells harvested after 16 h. Thymidine incorporation was measured using a liquid scintillation counter. Percentage suppression was calculated using the equation 100-[(counts per minute CD25plus CD25 co-culture/counts per minute CD25 alone) x 100].
Statistical methods
Statistical analyses were performed using SPSS 14.0 for Windows software (SPSS Inc.). Non-parametric statistical comparisons were performed using the Mann-Whitney U test. Differences were considered statistically significant when p values were less than 0.05.
Results
The residual T-cell population after lymphocyte depletion with alemtuzumab is dominated by effector and memory subsets
Alemtuzumab induced profound depletion of all lymphocyte subsets. Serial analysis of peripheral blood to monitor immune reconstitution after treatment showed that NK cells were the main lymphocyte population at day 30, accounting for a median of 24% compared to 12% in healthy age-matched volunteers (data not shown). The numbers of NK cells reached the normal range at 90 days whereas B-cell recovery was slowest commencing day 90 (data not shown). Figure 1A shows that the number of CD8 T cells recovered rapidly, reaching the normal range by 9 months, whereas the numbers of CD4 T cells were still below the normal range at 12 months. T-cell subset composition was investigated by analyzing expression of CD45RO and the co-stimulatory molecule CD27, which is lost during antigen-induced differentiation.37,38 The composition of both CD4 and CD8 T-cell populations was skewed toward effectors with a deficit of naïve cells relative to the composition in age-matched healthy volunteers (Figure 1B). Recovery of naïve T cells in patients was not detectable until 6 months and the majority of naïve CD4 T cells expressed CD31 (range, 75–98%) (Figure 1C), which is a marker of recent thymic origin39 indicating that they arose from renewed thymopoiesis.
Higher numbers of natural killer cells and donor-derived effector CD4 T cells at day 30 after transplantation correlate with acute graft-versus-host disease
The incidence of acute GvHD in the cohort was 28% (n=7) and that of chronic GvHD was 16% (n=4). All cases of GvHD developed prior to administration of donor lymphocytes to treat falling donor chimerism or disease relapse. The characteristics of the individuals who developed and those who did not develop GvHD were equivalent (Table 1). Comparison of lymphocyte subset recovery after transplantation with development of GvHD showed a significant correlation between acute GvHD and higher numbers of NK cells (p=0.04) and CD4 T cells (p=0.02) at day 30 (Table 2). The larger CD4 T population in patients who developed acute GvHD was mainly due to higher numbers of cells with effector (CD45RO CD27 CD62L) (p=0.02) and effector memory (CD45RO CD27 CD62L) (p=0.03) phenotypes (Table 2).
This finding indicates that effector CD4 T cells may be key players in GvHD, in which case they are likely to be of donor origin. The transplant preparative regimen used frequently produces a period of prolonged mixed T-cell chimerism after transplantation.36 The origin of effector CD4 T cells present at day 30 in patients who developed acute GvHD was, therefore, determined by isolating the population and evaluating the percentage composition of donor and patient cells by genetic profiling of pertinent polymorphic loci. Effector CD4 T cells (CD45RO CD27) from four patients who developed acute GvHD were predominantly of donor origin (86% for patient 2, 100% for patient 14, 50% for patient 25, 60% for patient 28) whereas the donor component of the memory CD4 T-cell population (CD45RO CD27) was lower (84% for patient 2, 63% for patient 14, 28% for patient 25, 10% for patient 28). Attempts to determine the donor composition of the CD4 T-cell subset at day 30 for patients who did not develop GvHD were not successful because of insufficient numbers of cells. Analysis, at later time points, of four patients who did not develop GvHD showed that they had lower percentages of donor-derived cells and this was more pronounced in the effector than the memory CD4 T-cell population, contrasting with the findings in patients who did develop GvHD (donor-derived cells accounted for 50% of the memory and 38% of the effector populations at day 60 in patient 4, for 10% of the memory and 0% of the effector populations at day 60 in patient 7, for 16% and 5% at day 60 in patient 11 and for 55% and 60% at day 90 in patient 15). These results provide support for donor-derived effector CD4 T cells being key players driving GvHD in this cohort of patients.
The median onset of acute GvHD symptoms was 63 days (range, 12–120). Given this variation in time of disease onset, representative examples of NK and T cell population dynamics during GvHD progression are shown individually for three patients (Figure 2). The association of high NK cell numbers with GvHD was only seen at the early, day 30 time-point. Thereafter, the NK cell population size remained relatively static in patients who developed GvHD while continued expansion was seen in patients without GvHD (Figure 2A). The initial expansion of the effector CD4 T cell population at day 30 continued in all patients who developed GvHD suggesting that these cells are the key players mediating pathology in this cohort of patients (Figure 2B upper panel). In patients 12 and 14, the numbers of effector CD4 T cells overshot the normal range in the healthy age-matched volunteers, but patient 25 illustrates that the expansion can be more modest. There was no consistent increase in effector CD8 T cell numbers in patients with GvHD (Figure 2B lower panel). A large increase in the number of effector CD8 T cells was observed in patient 12, perhaps associated with a concurrent episode of cytomegalovirus reactivation, but numbers for patients 14 and 25 were below the average for patients who did not develop GvHD. No correlation between B-cell reconstitution and GvHD was seen at any time point (data not shown).
The frequency of regulatory CD4 T cells relative to effector CD4 T cells is reduced at day 30 in patients who develop acute graft-versus-host disease
Regulatory CD4 T cells suppress the activity of effector T cells and, consequently, could have a beneficial role in the control of GvHD. Enumeration of CD4 CD25 Foxp3 T cells showed that the numbers of these putative regulatory cells at the early, day 30 time-point were similar in patients who did and did not develop acute GvHD (Figure 3A left panel). However, a significant deficit in the group that developed acute GvHD was apparent when numbers were considered relative to effector CD4 T cells at day 30. The median ratio of effector CD4 T cells per regulatory CD4 T cell in the acute GvHD group was 41 to 1 compared to 12 to 1 for patients without GvHD (p=0.03) (Figure 3A right panel).
The regulatory CD4 T-cell population expands in patients with chronic graft-versus-host disease
At later time points, the CD4 CD25 Foxp3 T-cell population gradually increased in all patients with no difference in numbers (Figure 3B) or percentage composition (data not shown) between patients with and without GvHD up to and including day 90. At day 90, the difference in the median ratio of effector CD4 T cells to CD4 CD25 Foxp3 T cells in the patients with or without GvHD had narrowed to 15 to 1, and 13.5 to 1 respectively (data not shown). Beyond day 100, there was a striking increase in the CD4 CD25 Foxp3 T-cell numbers in four patients who developed chronic GvHD (indicated by large white triangles in Figure 3B). These cells constituted a higher percentage of the CD4 T-cell population (range, 13–31%) at day 180 compared to the median of 5% in healthy age-matched volunteers and 4.03% among patients who did not develop GvHD. The number and percentage composition of CD4 CD25 Foxp3 cells at day 180 and beyond was not significantly different in patients without GvHD and in the five patients who had acute but not chronic GvHD (data not shown).
There are no unique markers for regulatory CD4 T cells and expression of intracellular Foxp3 by activated human CD4 T cells40–42 potentially confounds enumeration of regulatory T cells in inflammatory diseases such as GvHD. To substantiate our interpretation of regulatory CD4 T-cell recovery, CD27 was analyzed as an additional marker in some samples. CD27 is constitutively expressed on functional regulatory T cells43,44 but is lost on effector T cells.37,38 At day 30, the median ratio of effector CD4 T cells per CD4 CD25 Foxp3 CD27 T cell in five patients who subsequently developed acute GvHD was 58 to 1 whereas it was 6.8 to 1 (p=0.03) for three patients who did not develop GvHD, providing additional support that regulatory CD4 T cells are deficient relative to effector CD4 T cells early after transplantation in patients who develop acute GvHD. Analysis of CD4 CD25 Foxp3 CD27 T-cell dynamics in patients 12, 14 and 25 (Figure 3C) revealed that expansion of the regulatory CD4 T-cell population during chronic GvHD comprises a delayed wave following the spike in effector CD4 T-cell numbers. We verified that regulatory CD4 T-cell numbers were increased in patients with chronic GvHD by analysis of samples from an additional six patients with disease at day 180 (Figure 3D). Effector CD4 T-cell numbers were similar in all patients but significantly more regulatory CD4 T cells were present in the ten patients with chronic GvHD (p=0.008). Consequently the median ratio of effector per regulatory CD4 T cell in the chronic GvHD group was 3 to 1 compared to 9.6 to 1 for patients without GvHD (p=0.018).
Definition of regulatory status requires demonstration of functional suppressive activity. Expansion of the putative regulatory CD4 T-cell population in chronic GvHD patients provided sufficient numbers of cells for assessment of their ability to inhibit in vitro proliferation of CD4 CD25 cells. Effector T-cell function was variable among patients, probably reflecting the extent and type of immunosuppression administered to control GvHD. Therefore criss-cross assays were performed in which CD4 CD25 cells from patients were co-cultured with CD4 CD25 cells from a healthy volunteer representing a uniform response with which to assess suppressive activity. Results in Figure 3E show that CD4 CD25 cells from three patients with chronic GvHD all inhibited proliferation and, therefore, possessed regulatory function. We attribute the relatively low percentage suppression to some loss of regulatory activity after freezing and thawing cells as this has been observed in comparative studies performed with samples from healthy volunteers (data not shown).
Discussion
The incidence of GvHD in HSCT patients treated using a reduced intensity conditioning regimen with alemtuzumab and cyclosporin for GvHD prophylaxis was shown to correlate with higher numbers of effector CD4 T cells. A significant association was detected early (day 30) after transplantation, preceding the emergence of clinical pathology, for six of seven patients who developed acute GvHD. No correlation with CD8 T cells and GvHD was observed. There was no gross deficiency of regulatory CD4 T cells with numbers up to day 100 being similar in patients who developed GvHD and those who did not. However, the ratio of effector to regulatory CD4 T cells was significantly lower at day 30 in patients who developed acute GvHD suggesting that a diminished quantitative potential to control activity of the larger population of effectors early after transplantation may contribute to disease development. This result emphasizes the need for knowledge of the global immune response and the relationship between populations in order to understand the basis of the pathology.
At later time points, the gap between effector and regulatory CD4 T cell numbers closed in patients who developed GvHD. The kinetics of reconstitution of these cell populations in patients who recovered from acute GvHD and did not progress to develop chronic GvHD aligned with the pattern seen in patients who never developed GvHD. In contrast, the regulatory CD4 T-cell population expanded in patients who developed chronic GvHD, producing an imbalance relative to effectors that appeared to favor regulation. This observation seems counter-intuitive. Inflammatory conditions are typically associated with reduced regulatory T cells and this has indeed been reported in the context of clinical chronic GvHD.25,26 However, some reports have described relatively high numbers of putative regulatory CD25 CD4 T cells in patients with chronic GvHD,30,31 similar to our observations made using a reinforced definition of regulatory cells by considering Foxp3 and CD27 status and demonstration of suppressive activity. The longitudinal analysis of effector and regulatory CD4 T-cell dynamics performed in our study revealed that the expansion of regulatory T cells in chronic GvHD patients in this cohort occurred as a delayed wave after the earlier expansion of effector CD4 T cells (Figure 3C). Seen in this context, the CD4 T-cell-mediated immune response in these GvHD patients is consistent with evidence of the sequential co-evolution of memory and regulatory CD4 T cells in the periphery in response to antigen stimulus. An initial phase of CD4 T-cell expansion that mediates pathology is typically followed by recovery dependent upon generation in the periphery of regulatory CD25 Foxp3 CD4 T cells from the memory population.45 Regulatory CD4 T cells from patients in our study did not express CD31 (data not shown) indicating they were not thymus-derived but may indeed have developed de novo from the effector CD4 T-cell population. It is also noteworthy that three of the four patients with chronic GvHD with expanded regulatory CD4 T-cell populations subsequently experienced leukemia relapse. This trend, with its implication that anti-leukemia immunity may be suppressed, has been reported previously after HSCT for chronic myeloid leukemia.46
Correlation of higher effector CD4 T-cell numbers prior to emergence of GvHD pathology, expansion of the population during disease development and imbalance relative to regulatory CD4 T cells shows that acute GvHD pathology in these patients is primarily mediated by CD4 T cells. Alemtuzumab, used in the treatment regimen, is a humanized form of the monoclonal antibody Campath specific for CD52 which is expressed by lymphocytes, monocytes and some subtypes of dendritic cells and results in extensive depletion of all lymphocyte subsets. The T-cell population that recovered seems to have originated from residual mature cells because very few naïve T cells were present and there was little evidence of thymopoiesis prior to day 180. Studies of lymphocyte repopulation after lymphocyte depletion with alemtuzumab in the clinical setting of autoimmune disease,47,48 and solid organ transplantation49,50 all report a predominance of memory CD4 T cells in the residual population. In vitro studies suggest that alemtuzumab is less efficient at depleting memory CD4 T cells.49,50 Differential susceptibility of T-cell subsets to alemtuzumab-mediated elimination may, therefore, underlie the skewed CD4 T-cell population that emerges in patients who develop GvHD.
The other lymphocyte population whose numbers, at day 30, correlated with acute GvHD in the patients was NK cells, perhaps suggesting that these cells play a role in initiation of disease. It has been reported that rapid NK cell recovery can be associated with a lower incidence of GvHD,18 attributed to elimination of the patients’ antigen-presenting cells by alloreactive donor-derived NK cells. This would remove both the allostimulus for donor T cells and the potential reservoir of malignant cells, explaining the also reported lower risk of leukemia relapse.18 We did not observe this beneficial effect of NK cells in our cohort of patients. This may be because complete depletion of T cells seems to be required for the positive effect of NK cells to be apparent.13 It is also possible that disease control early after transplantation in our cohort of patients was primarily determined by the chemotoxicity of the regimen used with a relatively minor role for the GvL response. Consistent with this view, the increased numbers of effector CD4 T cells and NK cells in patients who developed acute GvHD were not associated with a beneficial impact on incidence of disease relapse (data not shown) despite the related nature of the GvHD and GvL alloresponses.
The increased numbers of NK cells, effector CD4 T cells and relative deficit of regulatory CD4 T cells to effectors in patients who developed acute GvHD was evident at day 30 after transplantation. The risk of acute GvHD is thus pre-determined early after a transplant. These phenotypic characteristics preceded emergence of pathology in the majority of patients suggesting further studies are warranted to assess the prognostic utility. Furthermore the signature appears GvHD-specific because it did not correlate with other complications after HSCT such as cytomegalovirus reactivation (data not shown). Reliable clinical indicators would enable safe early withdrawal of immunosuppression for patients not at risk of GvHD, identification of patients requiring a more intensive regimen for protection against GvHD, and could be used to guide response to therapy in patients who develop disease.
Knowledge of the type of immune response that produces GvHD is also a prerequisite for the improvement and development of new strategies to treat GvHD. The relative deficit of regulatory to effector CD4 T cells at day 30 in patients who developed acute GvHD suggests a need to modify treatment protocols to favor balanced early reconstitution of these subsets. This could, perhaps, be achieved by changes to the immunosuppressive regimen that aim to preserve regulatory T cells.44 Adoptive transfer of regulatory T cells is also being evaluated as a therapy for GvHD.51 Numbers of regulatory CD4 T cells were, however, not deficient in our GvHD patients and were actually increased in those with the chronic form of disease; furthermore, these cells exhibited suppressive activity in vitro. Additional studies are, therefore, required to understand why chronic GvHD pathology persists. Experimental mouse models of GvHD indicate regulatory T-cell suppressive activity has greatest efficacy when the alloresponse has not fully developed.52,53 The type of regulatory T cell is also likely to be important. CD103 regulatory T cells have been shown to suppress active chronic GvHD in mice,54 perhaps because they can traffic to sites of pathology.
Studies correlating immune reconstitution following HSCT with GvHD development have typically focused on tracking one lymphocyte subset, often in a cohort of patients treated with a variety of conditioning regimens. In contrast, we have undertaken a longitudinal study characterizing recovery of the mixture of lymphocyte populations in patients all treated with the same regimen that incorporates the widely used combination of alemtuzumab and cyclosporin for GvHD prophylaxis. In addition to identifying the key players in the immune response that causes GvHD in this treatment setting, our study revealed the importance of the balance between effector and regulatory CD4 T-cell populations in determining immune reactivity. Global immune profiling studies of this type provide information essential for improvements in diagnosis and treatment of GvHD.
Acknowledgments:
we thank Gary Warnes (Institute of Cellular and Molecular Medicine, The Royal London and Queen Mary’s Hospital, London), Ayad Eddaoudi (Cancer Research UK, London Research Institute, London) and David Darling (Department of Haematological Medicine, King’s College London Denmark Hill Campus, London) for cell sorting by flow cytometry.
Footnotes
- Authorship and Disclosures KM designed the study, planned and performed experiments, analyzed data and edited the manuscript. ZL designed the study, collected patients’ samples and clinical data, and edited the manuscript. BA planned and performed experiments, analyzed data and edited the manuscript. LP, AA and SK planned and performed experiments and analyzed data. AP contributed to research discussion and reviewed the manuscript. GL planned experiments, analyzed data and edited the manuscript. JAM and GJM designed the study and contributed to research discussion. LDB designed the study, planned and performed experiments, analyzed data and wrote the manuscript.
- The authors reported no potential conflict of interest.
- Funding: KM was funded by the Anthony Nolan Trust. BA was supported by a Clinical Research Fellowship from the Medical Research Council. SK was supported by King’s College Hospital Joint Research Committee.
- Received November 4, 2008.
- Revision received February 10, 2009.
- Accepted March 3, 2009.
References
- Slavin S, Nagler A, Naparstek E, Kapelushnik Y, Aker M, Cividalli G. 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; 91:756-63. PubMedGoogle Scholar
- Reddy P, Ferrara JL. Immunobiology of acute graft-versus-host disease. Blood Rev. 2003; 17:187-94. PubMedhttps://doi.org/10.1016/S0268-960X(03)00009-2Google Scholar
- Shlomchik WD, Lee SJ, Couriel D, Pavletic SZ. Transplantation’s greatest challenges: advances in chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2007; 13(1 Suppl 1):2-10. PubMedhttps://doi.org/10.1016/j.bbmt.2006.10.020Google Scholar
- Champlin R, Ho W, Gajewski J, Feig S, Burnison M, Holley G. Selective depletion of CD8+ T lymphocytes for prevention of graft-versus-host disease after allogeneic bone marrow transplantation. Blood. 1990; 76:418-23. PubMedGoogle Scholar
- Gallardo D, Garcia-Lopez J, Sureda A, Canals C, Ferra C, Cancelas JA. Low-dose donor CD8+ cells in the CD4-depleted graft prevent allogeneic marrow graft rejection and severe graft-versus-host disease for chronic myeloid leukemia patients in first chronic phase. Bone Marrow Transplant. 1997; 20:945-52. PubMedhttps://doi.org/10.1038/sj.bmt.1701008Google Scholar
- Yamashita K, Choi U, Woltz PC, Foster SF, Sneller MC, Hakim FT. Severe chronic graft-versus-host disease is characterized by a preponderance of CD4(+) effector memory cells relative to central memory cells. Blood. 2004; 103:3986-8. PubMedhttps://doi.org/10.1182/blood-2003-09-3286Google Scholar
- Yamashita K, Horwitz ME, Kwatemaa A, Nomicos E, Castro K, Sokolic R. Unique abnormalities of CD4(+) and CD8(+) central memory cells associated with chronic graft-versus-host disease improve after extracorporeal photopheresis. Biol Blood Marrow Transplant. 2006; 12(Suppl 2):22-30. PubMedGoogle Scholar
- Yakoub-Agha I, Saule P, Depil S, Micol JB, Grutzmacher C, Boulanger-Villard F. A high proportion of donor CD4+ T cells expressing the lymph node-homing chemokine receptor CCR7 increases incidence and severity of acute graft-versus-host disease in patients undergoing allogeneic stem cell transplantation for hematological malignancy. Leukemia. 2006; 20:1557-65. PubMedhttps://doi.org/10.1038/sj.leu.2404308Google Scholar
- Foster AE, Marangolo M, Sartor MM, Alexander SI, Hu M, Bradstock KF. Human CD62L− memory T cells are less responsive to alloantigen stimulation than CD62L+ naive T cells: potential for adoptive immunotherapy and allodepletion. Blood. 2004; 104:2403-9. PubMedhttps://doi.org/10.1182/blood-2003-12-4431Google Scholar
- Burrows SR, Khanna R, Burrows JM, Moss DJ. An alloresponse in humans is dominated by CTL cross-reactive with a single EBV CTL epitope: implications for graft versus host disease. J Exp Med. 1994; 179:1155-61. PubMedhttps://doi.org/10.1084/jem.179.4.1155Google Scholar
- Koelle DM, Chen HB, McCurkan CM, Petersdorf EW. Herpes simplex virus type 2-specific CD8 cytotoxic T lymphocyte cross-reactivity against prevalent HLA class I alleles. Blood. 2002; 99:3844-7. PubMedhttps://doi.org/10.1182/blood.V99.10.3844Google Scholar
- Elkington R, Khanna R. Cross-recognition of human alloantigen by cytomegalovirus glycoprotein-specific CD4+ cytotoxic T lymphocytes: implications for graft-versus-host disease. Blood. 2005; 105:1362-4. PubMedhttps://doi.org/10.1182/blood-2004-07-2602Google Scholar
- Sun JY, Dagis A, Gaidulis L, Miller MM, Rodriguez R, Parker P. Detrimental effect of natural killer cell alloreactivity in T-replete hematopoietic cell transplantation (HCT) for leukemia patients. Biol Blood Marrow Transplant. 2007; 13:197-205. PubMedhttps://doi.org/10.1016/j.bbmt.2006.09.009Google Scholar
- Miller JS, Cooley S, Parham P, Farag SS, Verneris MR, McQueen KL. Missing KIR ligands are associated with less relapse and increased graft-versus-host disease (GVHD) following unrelated donor allogeneic HCT. Blood. 2007; 109:5058-61. PubMedhttps://doi.org/10.1182/blood-2007-01-065383Google Scholar
- Giebel S, Locatelli F, Lamparelli T, Velardi A, Davies S, Frumento G. Survival advantage with KIR ligand incompatibility in hematopoietic stem cell transplantation from unrelated donors. Blood. 2003; 102:814-9. PubMedhttps://doi.org/10.1182/blood-2003-01-0091Google Scholar
- Beelen DW, Ottinger HD, Ferencik S, Elmaagacli AH, Peceny R, Trenschel R. Genotypic inhibitory killer immunoglobulin-like receptor ligand incompatibility enhances the long-term antileukemic effect of unmodified allogeneic hematopoietic stem cell transplantation in patients with myeloid leukemias. Blood. 2005; 105:2594-600. PubMedhttps://doi.org/10.1182/blood-2004-04-1441Google Scholar
- Scholl S, Mugge LO, Issa MC, Kasper C, Pachmann K, Hoffken K. Impact of early NK cell recovery on development of GvHD and CMV reactivation in dose-reduced regimen prior to allogeneic PBSCT. Bone Marrow Transplant. 2005; 35:183-90. PubMedhttps://doi.org/10.1038/sj.bmt.1704752Google Scholar
- Savani BN, Mielke S, Adams S, Uribe M, Rezvani K, Yong AS. Rapid natural killer cell recovery determines outcome after T-cell-depleted HLA-identical stem cell transplantation in patients with myeloid leukemias but not with acute lymphoblastic leukemia. Leukemia. 2007; 21:2145-52. PubMedhttps://doi.org/10.1038/sj.leu.2404892Google Scholar
- Miklos DB, Kim HT, Miller KH, Guo L, Zorn E, Lee SJ. Antibody responses to H-Y minor histocompatibility antigens correlate with chronic graft-versus-host disease and disease remission. Blood. 2005; 105:2973-8. PubMedhttps://doi.org/10.1182/blood-2004-09-3660Google Scholar
- Cutler C, Miklos D, Kim HT, Treister N, Woo SB, Bienfang D. Rituximab for steroid-refractory chronic graft-versus-host disease. Blood. 2006; 108:756-62. PubMedhttps://doi.org/10.1182/blood-2006-01-0233Google Scholar
- Taylor PA, Lees CJ, Blazar BR. The infusion of ex vivo activated and expanded CD4(+)CD25(+) immune regulatory cells inhibits graft-versus-host disease lethality. Blood. 2002; 99:3493-9. PubMedhttps://doi.org/10.1182/blood.V99.10.3493Google Scholar
- Hoffmann P, Ermann J, Edinger M, Fathman CG, Strober S. Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med. 2002; 196:389-99. PubMedhttps://doi.org/10.1084/jem.20020399Google Scholar
- Cohen JL, Trenado A, Vasey D, Klatzmann D, Salomon BL. CD4(+) CD25(+) immunoregulatory T cells: new therapeutics for graft-versus-host disease. J Exp Med. 2002; 196:401-6. PubMedhttps://doi.org/10.1084/jem.20020090Google Scholar
- Zhang C, Todorov I, Zhang Z, Liu Y, Kandeel F, Forman S. Donor CD4+ T and B cells in transplants induce chronic graft-versus-host disease with autoimmune manifestations. Blood. 2006; 107:2993-3001. PubMedhttps://doi.org/10.1182/blood-2005-09-3623Google Scholar
- Miura Y, Thoburn CJ, Bright EC, Phelps ML, Shin T, Matsui EC. Association of Foxp3 regulatory gene expression with graft-versus-host disease. Blood. 2004; 104:2187-93. PubMedhttps://doi.org/10.1182/blood-2004-03-1040Google Scholar
- Zorn E, Kim HT, Lee SJ, Floyd BH, Litsa D, Arumugarajah S. Reduced frequency of FOXP3+ CD4+CD25+ regulatory T cells in patients with chronic graft-versus-host disease. Blood. 2005; 106:2903-11. PubMedhttps://doi.org/10.1182/blood-2005-03-1257Google Scholar
- Rezvani K, Mielke S, Ahmadzadeh M, Kilical Y, Savani BN, Zeilah J. High donor FOXP3-positive regulatory T-cell (Treg) content is associated with a low risk of GVHD following HLA-matched allogeneic SCT. Blood. 2006; 108:1291-7. PubMedhttps://doi.org/10.1182/blood-2006-02-003996Google Scholar
- Meignin V, Peffault de Latour R, Zuber J, Regnault A, Mounier N, Lemaitre F. Numbers of Foxp3-expressing CD4+CD25high T cells do not correlate with the establishment of long-term tolerance after allogeneic stem cell transplantation. Exp Hematol. 2005; 33:894-900. PubMedhttps://doi.org/10.1016/j.exphem.2005.05.001Google Scholar
- Arimoto K, Kadowaki N, Ishikawa T, Ichinohe T, Uchiyama T. FOXP3 expression in peripheral blood rapidly recovers and lacks correlation with the occurrence of graft-versus-host disease after allogeneic stem cell transplantation. Int J Hematol. 2007; 85:154-62. PubMedhttps://doi.org/10.1532/IJH97.06160Google Scholar
- Clark FJ, Gregg R, Piper K, Dunnion D, Freeman L, Griffiths M. Chronic graft-versus-host disease is associated with increased numbers of peripheral blood CD4+CD25high regulatory T cells. Blood. 2004; 103:2410-6. PubMedhttps://doi.org/10.1182/blood-2003-06-2073Google Scholar
- Sanchez J, Casano J, Alvarez MA, Roman-Gomez J, Martin C, Martinez F. Kinetic of regulatory CD25high and activated CD134+ (OX40) T lymphocytes during acute and chronic graft-versus-host disease after allogeneic bone marrow transplantation. Br J Haematol. 2004; 126:697-703. PubMedhttps://doi.org/10.1111/j.1365-2141.2004.05108.xGoogle Scholar
- Ho AY, Pagliuca A, Kenyon M, Parker JE, Mijovic A, Devereux S. Reduced-intensity allogeneic hematopoietic stem cell transplantation for myelodysplastic syndrome and acute myeloid leukemia with multilineage dysplasia using fludarabine, busulphan, and alemtuzumab (FBC) conditioning. Blood. 2004; 104:1616-23. PubMedhttps://doi.org/10.1182/blood-2003-12-4207Google Scholar
- Lim ZY, Ho AY, Ingram W, Kenyon M, Pearce L, Czepulkowski B. Outcomes of alemtuzumab-based reduced intensity conditioning stem cell transplantation using unrelated donors for myelodysplastic syndromes. Br J Haematol. 2006; 135:201-9. PubMedhttps://doi.org/10.1111/j.1365-2141.2006.06272.xGoogle Scholar
- Sullivan KM, Agura E, Anasetti C, Appelbaum F, Badger C, Bearman S. Chronic graft-versus-host disease and other late complications of bone marrow transplantation. Semin Hematol. 1991; 28:250-9. PubMedGoogle Scholar
- Sullivan KM. Graft versus host disease. Blackwell: Sciences, Malden, MA; 1999. Google Scholar
- Lim Z, Pearce L, Ho AYL, Barber L, Ingram W, Usai M. Delayed attainment of full donor chimerism following alemtuzumab based reduced intensity conditioning haematopoietic stem cell transplantation for AML and MDS is associated with improved outcomes. Br J Haematol. 2007; 138:517-26. PubMedhttps://doi.org/10.1111/j.1365-2141.2007.06676.xGoogle Scholar
- Appay V, Dunbar PR, Callan M, Klenerman P, Gillespie GM, Papagno L. Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nat Med. 2002; 8:379-85. PubMedhttps://doi.org/10.1038/nm0402-379Google Scholar
- Fritsch RD, Shen X, Sims GP, Hathcock KS, Hodes RJ, Lipsky PE. Stepwise differentiation of CD4 memory T cells defined by expression of CCR7 and CD27. J Immunol. 2005; 175:6489-97. PubMedhttps://doi.org/10.4049/jimmunol.175.10.6489Google Scholar
- Kimmig S, Przybylski GK, Schmidt CA, Laurisch K, Mowes B, Radbruch A. Two subsets of naive T helper cells with distinct T cell receptor excision circle content in human adult peripheral blood. J Exp Med. 2002; 195:789-94. PubMedhttps://doi.org/10.1084/jem.20011756Google Scholar
- Gavin MA, Torgerson TR, Houston E, DeRoos P, Ho WY, Stray-Pedersen A. Single-cell analysis of normal and FOXP3-mutant human T cells: FOXP3 expression without regulatory T cell development. Proc Natl Acad Sci USA. 2006; 103:6659-64. PubMedhttps://doi.org/10.1073/pnas.0509484103Google Scholar
- Tran DQ, Ramsey H, Shevach EM. Induction of FOXP3 expression in naive human CD4+FOXP3 T cells by T-cell receptor stimulation is transforming growth factor-beta dependent but does not confer a regulatory phenotype. Blood. 2007; 110:2983-90. PubMedhttps://doi.org/10.1182/blood-2007-06-094656Google Scholar
- Wang J, Ioan-Facsinay A, van der Voort EI, Huizinga TW, Toes RE. Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur J Immunol. 2007; 37:129-38. PubMedhttps://doi.org/10.1002/eji.200636435Google Scholar
- Ruprecht CR, Gattorno M, Ferlito F, Gregorio A, Martini A, Lanzavecchia A. Coexpression of CD25 and CD27 identifies FoxP3+ regulatory T cells in inflamed synovia. J Exp Med. 2005; 201:1793-803. PubMedhttps://doi.org/10.1084/jem.20050085Google Scholar
- Coenen JJ, Koenen HJ, van Rijssen E, Hilbrands LB, Joosten I. Rapamycin, and not cyclosporin A, preserves the highly suppressive CD27+ subset of human CD4+CD25+ regulatory T cells. Blood. 2006; 107:1018-23. PubMedhttps://doi.org/10.1182/blood-2005-07-3032Google Scholar
- Vukmanovic-Stejic M, Zhang Y, Cook JE, Fletcher JM, McQuaid A, Masters JE. Human CD4+ CD25hi Foxp3+ regulatory T cells are derived by rapid turnover of memory populations in vivo. J Clin Invest. 2006; 116:2423-33. PubMedhttps://doi.org/10.1172/JCI28941Google Scholar
- Nadal E, Garin M, Kaeda J, Apperley J, Lechler R, Dazzi F. Increased frequencies of CD4(+)CD25(high) T(regs) correlate with disease relapse after allogeneic stem cell transplantation for chronic myeloid leukemia. Leukemia. 2007; 21:472-9. PubMedhttps://doi.org/10.1038/sj.leu.2404522Google Scholar
- Brett S, Baxter G, Cooper H, Johnston JM, Tite J, Rapson N. Repopulation of blood lymphocyte sub-populations in rheumatoid arthritis patients treated with the depleting humanized monoclonal antibody, CAMPATH-1H. Immunology. 1996; 88:13-9. PubMedhttps://doi.org/10.1046/j.1365-2567.1996.d01-650.xGoogle Scholar
- Cox AL, Thompson SA, Jones JL, Robertson VH, Hale G, Waldmann H. Lymphocyte homeostasis following therapeutic lymphocyte depletion in multiple sclerosis. Eur J Immunol. 2005; 35:3332-42. PubMedhttps://doi.org/10.1002/eji.200535075Google Scholar
- Pearl JP, Parris J, Hale DA, Hoffmann SC, Bernstein WB, McCoy KL. Immunocompetent T-cells with a memory-like phenotype are the dominant cell type following antibody-mediated T-cell depletion. Am J Transplant. 2005; 5:465-74. PubMedhttps://doi.org/10.1111/j.1600-6143.2005.00759.xGoogle Scholar
- Trzonkowski P, Zilvetti M, Friend P, Wood KJ. Recipient memory-like lymphocytes remain unresponsive to graft antigens after CAMPATH-1H induction with reduced maintenance immunosuppression. Transplantation. 2006; 82:1342-51. PubMedhttps://doi.org/10.1097/01.tp.0000239268.64408.84Google Scholar
- Hoffmann P, Boeld TJ, Eder R, Albrecht J, Doser K, Piseshka B. Isolation of CD4+CD25+ regulatory T cells for clinical trials. Biol Blood Marrow Transplant. 2006; 12:267-74. PubMedhttps://doi.org/10.1016/j.bbmt.2006.01.005Google Scholar
- Jones SC, Murphy GF, Korngold R. Post-hematopoietic cell transplantation control of graft-versus-host disease by donor CD425 T cells to allow an effective graft-versus-leukemia response. Biol Blood Marrow Transplant. 2003; 9:243-56. PubMedhttps://doi.org/10.1053/bbmt.2003.50027Google Scholar
- Nguyen VH, Zeiser R, Dasilva DL, Chang DS, Beilhack A, Contag CH. In vivo dynamics of regulatory T-cell trafficking and survival predict effective strategies to control graft-versus-host disease following allogeneic transplantation. Blood. 2007; 109:2649-56. PubMedhttps://doi.org/10.1182/blood-2006-08-044529Google Scholar
- Zhao D, Zhang C, Yi T, Lin CL, Todorov I, Kandeel F. In vivo-activated CD103+CD4+ regulatory T cells ameliorate ongoing chronic graft-versus-host disease. Blood. 2008; 112:2129-38. PubMedhttps://doi.org/10.1182/blood-2008-02-140277Google Scholar