Pathophysiology and Immunology of Chronic Graft-Versus-Host Disease


Cell subset

Risk of cGVHD

References

Effector memory CD4EM T cells (CCR7neg CD62Llow)

Increase in this T-cell subset co-related with increased risk of cGVHD

Yamashita et al. [7]

CD8+ Central Memory T cells

Increase in this T-cell subset co-related with increased risk of cGVHD

Yamashita et al. [8]

CD4+CD27 – T cells

Increase in this T-cell subset co-related with increased risk of cGVHD

Fukunaga et al. [9]

Tregs CD4+CD25+

Increase in this T-cell subset co-related with decreased risk of cGVHD

Zorn et al. [10]

Host APCs

If activated with co-stimulatory molecules CD86/CD80 – co-related with increased risk of cGVHD

Schlomchik et al. [11]

CD3-CD16+/56+ NK-cell

Lower numbers in patients with cGVHD

Abrahamsen et al. [12]


APCs antigen-presenting cells, CD cluster of differentiation, NK natural killer




Table 2.2
Inflammatory biomarkers co-related with development of cGVHD








































Inflammatory marker

Risk of cGVHD

References

TNFα

Levels increased in acute and cGVHD

Dander et al. [13]

IL-6 and IL-8

Levels increased in acute and cGVHD

Dander et al. [13]

IFNγ, IL-12

Increased levels co-relate with cGVHD

Rozmus et al. [12]

Day 7 IL-15 level

Low day 7 IL-15 levels correspond to subsequent cGVHD

Pratt et al. [14]

CXCL9 levels

Levels increased at onset of cGVHD

Kitko et al. [15]

ST2, CXCL9, MMP3, Osteopentin

Elevated serum levels at day 100 co-relate with cGVHD

Yu et al. [16]

BAFF (B-cell activating factor)

Levels elevated in cGVHD

Sarantopoulous et al. [17]


CXCL9 chemokine (C-X-C motif) ligand 9, IFNγ interferon gamma, IL interleukin, MMP3 matrix metalloproteinase, ST2 suppression of tumorigenicity 2, TNFα tumor necrosis factor alpha



T Cells



T-Cell Differentiation Status


Immune cell subsets have been studied extensively in patients to determine their predictive value in chronic GVHD. Most studies have focused on CD4+ (post-thymic) T cell subsets. Human peripheral blood CD4+ T cells are classified into three broad populations: (1) naive CD45RA+CCR7+ and two memory subsets; (2) CD45RA-CCR7+ (central memory); and (3) CD45RA-CCR7- (effector memory). Chemokine receptor CCR7 is required for migration of T cells into secondary lymphoid organs such as lymph nodes and the spleen. CD62L expression guides lymphocytes into lymphoid tissue and is tightly linked to CCR7 expression on memory CD4+ T cells. Yamashita et al. [7] studied relative proportions of effector memory CD4EM T cells (CCR7-CD62Llow in patients with established cGVHD and compared these to CD4EM T cells in healthy controls and patients with no clinical signs of cGVHD. Chronic GVHD patients had a significantly higher percentage of CCR7-CD62Llow cells compared with healthy controls (35.5 % vs 13.8 % respectively; P < .0001) or stem cell transplantation patients without cGVHD (35.5 % vs 21.7 % respectively; P <.01) in the total CD4+ population.

Changes in relative ratios of CD4 and CD8 T cell subsets and decrease in CD4+ central memory T cells has been noted in patients with chronic GVHD. Yamashita et al. [8] reported changes in T cell subsets in 37 patients who developed cGVHD after allogeneic HSCT. Specifically, an increase in central memory CD8+ cells with concomitant decline in CD4+ cells was noted. This immune cell pattern was not seen in patients who did not develop cGVHD post-HSCT or in patients who responded to immunotherapy with photopheresis. This finding indicates that the ratio of central and effector memory T cell subsets is altered in cGVHD and successful treatment leads to normalization of this ratio.

Fukunaga et al. [9] reported a unique subset of T cells, CD4+CD27−, which are seen in peripheral blood in increased frequency in patients with cGVHD (39.5 % compared to < 10 % in healthy adults). These T cells have shortened telomere length, increased susceptibility to activation-induced cell death and decreased clonal diversity. This depletion of central memory CD4+ T cell pool increases patients’ susceptibility to recurrent infections, thereby increasing infectious morbidity. Patients who have decreasing numbers of CD4+CD27+ cells post-allogeneic HCT should be monitored closely for infectious complications and should remain on appropriate antimicrobial prophylaxis until immune recovery occurs post-HSCT.


CD4 T-Cell Cytokine Subsets (Th1, Th2, Th 17)


The CD4 T helper cells can be classified in to Th1 and Th2 based on cytokine secretion profiling, with Th1 cells secreting IFN-γ, IL-2 and TNFα. Th2 cells produce IL-4,IL-5,IL-6,IL-10 and IL-13. Th1 cells are responsible for delayed type hypersensitivity and are important in defense from infectious microorganisms. When exposed to foreign antigens, the APCs (macrophages and dendritic cells [DCs]) migrate to lymphoid organs and present antigens to naïve T cells and produce proinflammatory cytokines, such as IL-12, which leads to Th1 type response. Human and animal studies have shown that acute GVHD is a clinical syndrome caused by an imbalance between Th1 (pro-inflammatory cytokines) and Th2 response (anti-inflammatory cytokines) [1820]. Chronic GVHD is a T cell mediated allo-reactive process and Th2 response dominates in cGVHD [21, 22]. Another subset of CD4 T cells which produce IL-17 (Th17) have been also implicated in development of cGVHD. Dander et al. [13] studied the role of IL-17 producing CD4+ T cells (Th17) in cGVHD in serum of 51 patients post-allo HSCT with clinical manifestations of cGVHD and compared this to 15 healthy donors (HD). Patients with cGVHD showed an increase of Th17 population compared with HD (mean SFU = 178/25,000 cells, n = 18, ANOVA P < 0.001). Importantly, by analyzing the proportion of Th17 cells according to the activation status of cGVHD (active vs. inactive phases), the authors were able to demonstrate that patients with active cGVHD show an increase of Th17 population (mean SFU = 237/25,000 cells, n = 13, ANOVA P < 0.001). Inflammatory cytokines produced by Th17 cells such as IL-6, TNF- α and IL-8 were also significantly elevated in patients with active cGVHD.

Therapeutic approaches: Based on the critical role played by CD4+ T cells in propagating cGVHD, attempts have been made to reduce these alloreactive T cells by inhibition of the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) pathway. Common gamma chain signaling via JAK pathway leads to up-regulation of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21—this cytokine storm causes T cell activation, lineage commitment, and survival signaling. Patients with steroid refractory cGVHD who were treated with the commercially available JAK1/2 inhibitor ruxolitinib exhibited excellent clinical responses with an overall response rate of 85.4 %. These responses were independent of organ involvement [23]. Additional clinical trials are in progress to further study the role of this important class of medications in prevention of cGVHD (NCT02806375, NCT01790295).


Regulatory T Cells (Tregs)


Regulatory T cells, or Tregs, constitute 5–10 % of circulating CD4+ T cells, and suppress auto- and allo-reactive T cell clones. Tregs have also been associated with clinical symptoms of cGVHD. Immunophenotypically, Tregs are CD4+ and CD25+ and express forkhead transcription factor FOXP3. Zorn et al. [10] evaluated CD4+CD25+ Tregs in 30 patients with cGVHD after allogeneic HSCT, 27 patients without active cGVHD, and 26 healthy controls. They also evaluated T cell receptor excision circles (TREC) by peripheral blood polymerase chain reaction (PCR) as a marker of thymic activity in post-HCT patients. Patients with active cGVHD had significantly lower expression of FOXP3 when compared with patients without cGVHD (P = .009) or healthy donors (P = .01). Moreover, patients with active cGVHD had significantly lower expression of FOXP3 when compared with patients without cGVHD (P = .009) or healthy donors (P = .01). Patients with or without cGVHD showed a significant decrease in TRECs compared with healthy donors (P < .001), supporting that thymic function is substantially impaired following allogeneic HSCT.

Therapeutic approaches: Based on studies showing the role of IL-2 as a growth factor for Tregs, clinical trials have been reported with use of low-dose IL-2 in cGVHD setting with promising response rates [24, 25]. Additional combinations of IL-2 with Tregs (NCT01937468) and photopheresis (NCT02340676) are ongoing to see if therapeutic response can be augmented above that seen with IL-2 monotherapy.



Antigen-Presenting Cells


In addition to T cells, antigen-presenting cells (APCs), such as macrophages, dendritic cells, and B cells, play a critical role in initiating and propagating immune responses associated with cGVHD. Schlomchik et al. [11] demonstrated that host APCs are radio- and chemo-resistant post-conditioning regimens, and are critical for antigen presentation to incoming donor T cells, thereby proving the antigenic and co-stimulatory signals for T-cell activation and expansion leading to aGVHD. T-cell activation in the context of APC requires (1) antigen presentation with MHC class II molecules (MHC restriction) and (2) signal transduction via co-stimulatory molecules such as CD 80 and CD86. APCs are able to recognize the presence of microorganisms through the detection of conserved pathogen-associated molecular patterns (PAMP) and rapidly initiate tailored responses to these danger signals. Blockage of PAMP inducible co-stimulatory molecules such as CD40 or B7.1/B7.2 is effective in decreasing incidence of GVHD. She et al. [26] described the role of B cells primed for TLR9 (toll-like receptor 9), the response of which may play a role in pathophysiology of cGVHD. TLR9 expression has a significant correlation with expression of CD86 and CD80—furthermore, expression of these surface proteins was used as surrogate for TLR9 expression in this trial. A significantly greater percentage of B cells from and early cGVHD group (3–8 months post-HSCT; n = 19, 56.3 %) were capable of up-regulating CD86, compared to 6-month non-cGVHD controls (n = 9, 15.8 %; P = .0004) in response to PS-modified CpG. To confirm the B cell responses were mediated by TLR pathway, mRNA expression levels were checked in purified B cells by RT-PCR. There was a significant correlation (n = 12, r2 = 0 .65, P = .002) between PS-modified CpG 2006 response and B cell TLR9 mRNA levels. Anderson et al. [27] further clarified the role of donor and host APCs in cGVHD. Both donor and host APCs can elicit cGVHD phenotype in setting of CD80/86 co-stimulation—in the absence of this co-stimulatory signal, no cGVHD developed. This process is CD4+ T cell mediated. Their findings show that donor APCs can cause late cGVHD in a CD4 cell dependent mechanism in setting of appropriate co-stimulatory signals. This finding provides additional therapeutic targets for prevention of cGVHD.


B Cells and Humoral Immunity



B Cell and B-Cell Activating Factor (BAFF)


The role of donor B cells in mediating chronic GVHD by antibody-mediated targeting of recipient tissues was first reported by Sarantopoulous et al. [17]. B-cell activating factor (BAFF) is known to be a key regulator of normal B-cell homeostasis in humans [28] and high BAFF levels have been found in patients with a variety of autoimmune diseases. A total of 104 patients who had undergone allogeneic HSCT between 1994 and 2005 for hematologic malignancies were studied. Enzyme-linked immunosorbent assay (ELISA) was used to measure plasma BAFF levels, and flow cytometry was used to assess BAFF receptor expression on B cells in patients with or without chronic GVHD. These plasma samples were collected prospectively at predetermined time points and BAFF levels were correlated with clinical outcomes. BAFF levels were significantly higher in patients with active chronic GVHD compared with those without disease (P = 0.02 and 0.0004, respectively). Patients treated with glucocorticoids showed reduction in BAFF levels, suggesting this correlation with disease severity. Furthermore, it was noted that BAFF levels were high post-HSCT and declined in patients who never developed chronic GVHD. In contrast, BAFF levels remained elevated in patients who developed clinical manifestations of cGVHD. Six-month BAFF levels ≥ 10 ng/mL were strongly associated with subsequent development of chronic GVHD (P < 0.0001). Following transplant, plasma BAFF levels correlated inversely with BAFF receptor expression on B cells (P = 0.01), suggesting that soluble BAFF affected B-cells through this receptor [29].

Fujii et al. [30] demonstrated variation in biomarker levels based on early (3–8 months post-HSCT) versus late ≥ 9 months post-HCT). Soluble B cell activation factor (sBAFF), anti-dsDNA antibody, soluble interleukin-2 receptor alpha (sIL-2Rα), and soluble CD13 (sCD13) were elevated in patients with early-onset cGVHD compared with controls. Soluble B-cell activation factor and anti-dsDNA were elevated in patients with late-onset cGVHD. This previous finding suggests that the pathophysiology of cGVHD is heterogeneous with different mechanisms operative at different time-points following HSCT.


Auto-antibodies


The role of stimulatory auto-antibodies in platelet-derived growth factor receptors (PDGFR) was studied by Svegliati et al. [31] in sclerotic cGVHD. Based on clinical evidence of agonistic antibodies toward PDGFR in patients with systemic sclerosis, they tested 39 post-HCT patients with cGVHD for presence of stimulatory auto-antibodies to the PDGFR. They detected the presence of stimulatory antibodies to the PDGFR in all patients with extensive cGVHD, but none in the patients without cGVHD. Their finding also supports the use of tyrosine kinase inhibitors (such as imatinib) as therapeutic agents in scleroderma by virtue of its anti-PDGFRA activity. Wechalekar et al. [32] studied, in 13 HCT recipients, the presence of auto-antibodies in rheumatoid factor (RF), antinuclear antibody (ANA), double-stranded DNA (dsDNA), antimitochondrial antibody, antismooth muscle antibody (AntiSm), antiendomysial, antireticulin antibodies, antithyroid peroxidase antibodies, and an extractable nuclear antigen screen. All the patients with antibodies had cGVHD, whereas none of the patients without cGVHD had any auto-antibodies (P = 0.025). Three of the patients (23 %) had only one autoantibody and three others (23 %) had more than one auto-antibody. ANA was positive in three patients (23.3 %), dsDNA in four patients (30.7 %), RF in one patient (7.6 %), and anti-smooth muscle in two patients (15.3 %). In the present study, auto-antibodies were detected predominantly in patients with the presence of cGVHD. They also appeared to be more frequent in an unmanipulated graft and less frequent in patients with a T-cell depleted allograft.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Sep 3, 2017 | Posted by in Dermatology | Comments Off on Pathophysiology and Immunology of Chronic Graft-Versus-Host Disease

Full access? Get Clinical Tree

Get Clinical Tree app for offline access