1. Introduction
Allogeneic hematopoietic stem cell transplantation (HSCT) is a valuable curative therapy for hematological malignancies. Haploidentical allogeneic HSCT with high-dose post-transplant cyclophosphamide (PTCy) represents an emerging alternative option for patients with hematologic malignancies when a HLA identical sibling or a matched related donor is not available [
1,
2]. PTCy effectively eradicates alloreactive T cells after haploidentical stem cell infusion while hematopoietic stem cells are spared, thereby aiding in prevention of acute and chronic graft-versus-host disease (GVHD) [
3,
4,
5]. Despite of the use of PTCy, however, patients continue to suffer from non-relapse mortality (NRM). While typical manifestations of acute GVHD and other NRM are thought to involve the skin, gastrointestinal tract, and liver, more recent reports have suggested that symptoms arising from central nervous system derangements may be a significant complication of immune therapies such as HSCT. Of these, a recent case report identified autoimmune limbic encephalitis (LE) as a complication of allogeneic HSCT [
6]. LE is an inflammatory disease of the medial temporal lobes mediated by autoㅌantibodies such as antibodies to N-methyl-D-aspartate (NMDA)-type glutamate receptor (GluR) or human herpesvirus 6 (HHV-6), and has often been reported as a paraneoplastic syndrome [
7,
8,
9]. The diagnosis of LE is often difficult due to broad range of clinical symptoms and overlapping features with other neurologic disorders, limiting the characterization on risk factors for the development of LE following allogeneic HSCT. As such, there is little data describing early changes following transplantation that precede the subacute progression of LE. Studies have shown, however, that the immediate post-transplantation course is often complicated by systemic inflammation associated with high levels of pro-inflammatory cytokines such as IL-6, TNF-α, and IFN-γ, termed cytokine-release syndrome (CRS) [
6]. Imus et al. reported that 17% of patients undergoing haploidentical HSCT had severe CRS of grade 3 to 5 [
10,
11]. CRS has been strongly linked to not only acute GVHD but also highly morbid neurologic symptoms collectively referred to as immune effector cell associated neurologic syndrome (ICANS) [
12,
13]. The progression of inflammation may be related to dysfunction in regulatory T (Treg) cells. Treg cells are a highly immunosuppressive population of CD4+ T cells characterized by high and stable expression of the interleukin (IL)-2 receptor α chain (CD25) and the master transcription factor Forkhead box protein P3 (FOXP3) [
14,
15]. Treg cells are known to be impaired in disorders of inflammation, including autoimmune diseases and GVHD [
16]. Transfer of Treg cells has been shown to alleviate GVHD symptoms in both humans and animal models [
17], while other studies have suggested that the rapid expansion of Treg cells within the first month of HSCT is critical for GVHD prevention [
18]. However, the mechanisms underlying potential Treg dysfunction following HSCT remain poorly understood. In this study, we retrospectively analyzed available clinical and laboratory data from 35 adult patients who were treated with haploidentical HSCT with PTCy for hematologic malignancies, of which four developed limbic encephalitis. We also examine the reconstitution kinetics of Treg cells and their subsets within the first 21 days of HSCT in order to elucidate potential mechanisms of early systemic inflammation that may contribute to the development of LE. Our findings demonstrate that current immunomodulatory therapies in haploidentical HSCT may be insufficient and encourage further investigation into means of ameliorating CRS and Treg dysfunction for the prevention of acute GVHD and early neurotoxicity.
3. Discussion
In this study, we characterized early biologic markers of systemic inflammation in patients who developed limbic encephalitis among other signs of acute GVHD following haploidentical HSCT with PTCy. On clinical exam, LE patients were more likely to have severe GVHD, higher CRS grade and CRP levels, and prolonged, more extreme fevers within the first three weeks of transplant. Further investigation revealed that elevated levels of pro-inflammatory cytokines and impairment of early reconstitution of regulatory T cells may be associated with the development of more severe inflammatory adverse effects such as LE and acute GVHD. All LE patients presented with seizures within the first 30 days of transplant. As such, it was important to characterize early changes that may have contributed to the development of symptoms, particularly given the lack of human studies investigating that time frame. Early fever and high levels of CRP and inflammatory cytokines IL-6, TNF-α, and IFN-γ as seen in our LE patients in the absence of infection strongly resemble cytokine release syndrome, which may result in multiorgan dysfunction, involving the brain, lung, and kidney [
6,
29]. CRS is well described in recipients of chimeric antigen receptor (CAR) T cell infusion, and more recently, other tumor therapies that function by activation of immune effector cells [
6,
30]. Based on these previous reports, supraphysiologic cytokines are likely released by donor derived alloreactive immune effector cells as well as endogenous monocytes [
31]. In support of this, levels of pro-inflammatory cytokines (
Figure 3) and clinical symptoms such as fever, vasodilation, and dyspnea (
Figure 2A,
Table S1) were highest on post-transplant day 3 prior to administration of PTCy, which is given to suppress alloreactive T cells. Interestingly, we did not observe significant differences in CMV sero-positivity, prior transplant, HCT-CI score, or donor-recipient sex mismatch in patients that developed CRS versus those that did not as reported in a retrospective multicenter study, although this may be due to small sample size [
32]. CRS, however, may be independently associated with neurotoxicity. Indeed, Imus et al. demonstrated that 13 out of 25 severe CRS patients developed encephalopathy [
10]. Other studies have suggested that diffusion of cytokines and transmigration of the subsequently activated immune cells into the cerebrospinal fluid (CSF) and central nervous system (CNS) produces neurotoxicity with a high degree of mortality [
33,
34]. This neurotoxicity may manifest as wide range of neurologic symptoms including delirium, encephalopathy, aphasia, lethargy, agitation, and seizures, and is referred to by several titles in the literature such as cell-mediated neurotoxicity syndrome (ICANS) and CAR-related encephalopathy syndrome (CRES). Currently it is difficult to determine whether LE should be included within these syndromes, but nevertheless LE may be associated with CRS and ICANS often referred to as neurotoxicity. In its severe forms, ICANS may be associated with life-threatening features such as seizures. In most cases, CRS precedes ICANS and it is understood as an initiating factor for ICANS, although concurrent manifestation of CRS and ICANS may happen [
35]. Of note, while limbic encephalitis has been reported association with human herpes virus (HHV)-6 after allogenic HSCT, all patients in this study tested negative on PCR for HHV-6 [
36].
Among the studied cytokines, IL-6 is already being considered as a potential biomarker for acute GVHD following allogenic HSCT, where elevated IL-6 levels were significantly associated with worsening outcomes including severe CRS, acute GVHD, and reduced overall survival [
37,
38]. In our study, IL-6 levels were elevated in patients who developed LE even before HSCT (
Figure 3C). It has been reported that conditioning regimens leads to host tissue damage and elevated levels of inflammatory cytokines including IL-6, which is important in the initiation phase of acute GVHD pathophysiology [
39]. LE patients showed significantly elevated IL-6 not only at baseline, but also on post-transplant days 3 and 5 compared to non-LE patients. In a study with humanized NSG mice using a patient-derived leukaemic cell line, depletion of phagocytic cells prior to CAR T cell transplantation was enough to abrogate IL-6 production and CRS and, furthermore, single-cell analysis of leukocytes isolated during CRS identified monocyte lineage cells to be the source of IL-6[
40]. Through its involvement on B-cell differentiation and auto-antibody production, differentiation of Th17 cells, inhibition of Treg differentiation, or differentiation of CD8+ cytotoxic T cells, IL-6 has been reported to possibly induce and enhance autoimmune neuronal tissue damage [
41,
42,
43]. Given that IL-6 has been strongly implicated in neuroinflammation [
44], IL-6 may be a promising biomarker for screening patients at greater risk of developing neurotoxic sequelae. In addition, treatment of CRS patients in blinatumomab and CAR T cell infusion trials with the IL-6 receptor antagonist tocilizumab was shown to produce rapid clinical stabilization [
45,
46,
47,
48]. Similar therapy with tocilizumab should be further studied in HSCT to prevent severe CRS and early CNS complications.
In addition to elevated IL-6, we also observed a significant decrease in frequency of Treg cells in LE patients by day 5 despite an initial expansion of naïve Treg cells on day 3, suggestive of suppressed Treg cells differentiation. IL-6 is known not only to inhibit TGF-β-induced T cell differentiation into regulatory T cells [
49], but also downregulate Foxp3 expression on Treg cells [
50]. Non-LE patients, on the other hand, had significant expansion of Treg cells by post-transplant day 21. Given that Treg cells are critical for suppressing inflammatory responses and have been implicated in several autoimmune diseases [
51], this early lack of reconstitution of Treg cells in LE patients may have heralded the onset of acute GVHD and neurologic symptoms. Indeed, we show that reduced Treg cells frequency was associated with greater severity of acute GVHD (
Figure 4C), consistent with previous reports stating that protection against GVHD depends not only on the depletion of donor alloreactive T cells but also the rapid and robust recovery of donor Treg cells to initiate and maintain alloimmune regulation [
52,
53,
54,
55,
56,
57]. Further studies have shown Treg cells from patients with acute GVHD exhibited multiple dysfunctions, including Foxp3 expression instability and increased apoptosis [
58]. The underlying mechanism among early induction of IL-6, Treg cells suppression or dysfunction, and early neurotoxic clinical outcomes such as LE should be carefully investigated in future studies.
Despite significant associations between clinical outcomes and biologic markers that may guide future HSCT research and protocol, this study has some limitations. The small sample size, low frequency of LE occurrence, and variability between patient presentation for sample collection on specific days post-transplant may mask more significant associations. Among 31 non-LE patients, PBMC samples from 10 non-LE patients could be available for the analysis of cytokines and Treg cells. We also lacked the means for autoantibody testing at this institute, which may provide additional insight into the mechanisms of LE development in future HSCT studies.
The development of LE as severe inflammatory adverse effects after haplo-SCT may be regulated by early increased levels of pro-inflammatory cytokines such as IL-6, TNF-α, and IFN-γ and impairment of early reconstitution of regulatory T cells. The central nervous system (CNS) has been reported to be one of the nonclassical GVHD target organs that is sterile [
59] and furthermore clinical manifestations of acute GVHD in the CNS consists of signs of encephalitis [
60,
61,
62]. Following allo-HSCT in mice, the CNS was significantly infiltrated by donor T cell and allo-HSCT recipients with GVHD manifested cell death of neurons as a direct target of alloreactive T cells [
63]. Interestingly, blockade of the IL-6 signaling resulted in marked inhibition of donor T cell accumulation, inflammatory cytokine gene expression, and host microglial cell expansion in brain [
64]. Previous preclinical and clinical studies suggest that the CNS may be targeted by donor T cells-involved alloimmune reactions [
61,
63,
65,
66]. In addition, MHC class I protein was reported to be expressed in hippocampal neurons [
67,
68], which suggest that the limbic system could be damaged by alloimmune reactions based on HLA mismatch, leading to the development of LE. In our study, LE patients show frequently specific HLA types, that is HLA B*40, 02 and HLA DRB1*08, 02 (
Supplementary Table S2), suggesting that patients with these HLA types might be more susceptible to the autoimmune attack of immune effector cells. Development of LE is possibly associated with alloimmune reaction to various proteins in the limbic system through auto-antibody formation of B cells, which may not be appropriately regulated because of the decreased function of Treg. These processes could be augmented by elevated pro-inflammatory cytokines such as IL-6, TNF-α, and IFN-γ.
In conclusion, patients that developed LE within the first 30 days of HSCT were likely to have high serum IL-6 among other inflammatory cytokines, coupled with suppression of regulatory T cell differentiation that may have allowed for rampant and unchecked inflammation. This association along with potential impact of anti-IL6 therapies should be validated in a larger and geographically and clinically diverse population in future multicenter studies.
Author Contributions
Conceptualization, J.K., I.-C.S.; Formal analysis, B.Y.H., M.-W.L., S.C., Y.J., T.T.D.P. ; Funding acquisition, I.-C.S.; Investigation, B.Y.H., M.-W.L., S.C., Y.J., T.T.D.P., S.K., J.S.K., D.-Y.J., Y.J., J.-H.P., J.K., I.-C.S.; Methodology, B.Y.H., M.-W.L., S.C., Y.J., T.T.D.P., Y.J., J.-H.P.; Project administration, S.K., J.S.K., D.-Y.J., Y.J., J.-H.P. ; Resources, S.K., J.S.K., D.-Y.J., Y.J., J.-H.P., J.K., I.-C.S. ; Supervision, J.K., I.-C.S.; Validation, S.K., J.S.K., D.-Y.J., J.K., I.-C.S. ; Visualization, B.Y.H., M.-W.L., S.C., Y.J., T.T.D.P.; Writing-original draft, B.Y.H., M.-W.L., J.K., I.-C.S.; Writing-review & editing, J.K., I.-C.S. All authors have read and agreed to the published version of the manuscript.
Figure 1.
LE patients show characteristic MRI findings and reduced overall survival. (A) T2-weight fluid attenuated inversion recovery (FLAIR) images in LE patients (1-4) demonstrated bilateral and symmetrical high signal intensity in both medial temporal lobes, insulae, and medial frontal lobes. (B) Survival curve in patients diagnosed with (red) and without (blue) LE, Statistical difference by log-rank test (N=35, p=0.001).
Figure 1.
LE patients show characteristic MRI findings and reduced overall survival. (A) T2-weight fluid attenuated inversion recovery (FLAIR) images in LE patients (1-4) demonstrated bilateral and symmetrical high signal intensity in both medial temporal lobes, insulae, and medial frontal lobes. (B) Survival curve in patients diagnosed with (red) and without (blue) LE, Statistical difference by log-rank test (N=35, p=0.001).
Figure 2.
LE patients have significantly higher Tmax and plasma CRP levels. (A) Median Tmax in LE vs non-LE groups based on recorded patient body temperatures up to 21 days post-transplantation. (B) Plasma CRP levels before transplant (BT) and days 3, 5, and 21 after haplo-identical HSCT. N=31 non-LE patients, N=4 LE patients. Statistical difference by two tailed t-test. *p < 0.05, **p < 0.01, ***p < 0.001. LP = leukapheresis.
Figure 2.
LE patients have significantly higher Tmax and plasma CRP levels. (A) Median Tmax in LE vs non-LE groups based on recorded patient body temperatures up to 21 days post-transplantation. (B) Plasma CRP levels before transplant (BT) and days 3, 5, and 21 after haplo-identical HSCT. N=31 non-LE patients, N=4 LE patients. Statistical difference by two tailed t-test. *p < 0.05, **p < 0.01, ***p < 0.001. LP = leukapheresis.
Figure 3.
Plasma inflammatory cytokine levels are increased in LE patients. (A) IFN-gamma (IFN-γ), (B) TNF-alpha (TNF-α), and (C) IL-6 measured before transplant (BT) and on days 3, 5, and 21 after haplo-identical HSCT. Statistical difference by two tailed t-test. *p < 0.05, **p < 0.01, ***p < 0.001.
Figure 3.
Plasma inflammatory cytokine levels are increased in LE patients. (A) IFN-gamma (IFN-γ), (B) TNF-alpha (TNF-α), and (C) IL-6 measured before transplant (BT) and on days 3, 5, and 21 after haplo-identical HSCT. Statistical difference by two tailed t-test. *p < 0.05, **p < 0.01, ***p < 0.001.
Figure 4.
Proportion of Tregs are reduced in LE patients and inversely related to severity of acute graft-versus-host disease. (A) Flow cytometry gating strategy, with CD4+ T cells divided into three populations: a) CD25-Foxp3-, b) CD25+Foxp3-, c) CD25hiFoxp3+. (B) Proportions of these CD4+ subsets among CD3+ T cells were compared between non-LE and LE patients before (BT) and on days 3, 5, and 21 post-HCST. Statistical difference by two tailed t-test. *p < 0.05, **p < 0.01, ***p < 0.001. (C) Spearman correlation analysis between CD4+ subsets (CD25-Foxp3-, top; CD25+Foxp3-, mid; CD25hiFoxp3+, bottom) and severity of acute GVHD at three time points (BT, left; post-HCST day 5, mid; post-HCST day 21, right). Spearman correlation coefficient (r) and p value were indicated.
Figure 4.
Proportion of Tregs are reduced in LE patients and inversely related to severity of acute graft-versus-host disease. (A) Flow cytometry gating strategy, with CD4+ T cells divided into three populations: a) CD25-Foxp3-, b) CD25+Foxp3-, c) CD25hiFoxp3+. (B) Proportions of these CD4+ subsets among CD3+ T cells were compared between non-LE and LE patients before (BT) and on days 3, 5, and 21 post-HCST. Statistical difference by two tailed t-test. *p < 0.05, **p < 0.01, ***p < 0.001. (C) Spearman correlation analysis between CD4+ subsets (CD25-Foxp3-, top; CD25+Foxp3-, mid; CD25hiFoxp3+, bottom) and severity of acute GVHD at three time points (BT, left; post-HCST day 5, mid; post-HCST day 21, right). Spearman correlation coefficient (r) and p value were indicated.
Figure 5.
LE patients show concomitant increase in conventional effector T cells. (A) Flow cytometry gating strategy with division into subsets I-VI based on CD45RA and Foxp3 as indicated. (B) Proportions of subsets compared between non-LE and LE patients before transplant (BT) and on days 3, 5, and 21 post-HCST. Statistical difference by two tailed t-test. *p < 0.05, **p < 0.01, ***p < 0.001. (C, D) Spearman correlation analysis between severity of acute GVHD and (C) Treg subsets (I, top; II, mid; III, bottom) (C) and remaining subsets (IV, top; V, mid; VI, bottom) (D). Spearman correlation coefficient (r) and p value were indicated.
Figure 5.
LE patients show concomitant increase in conventional effector T cells. (A) Flow cytometry gating strategy with division into subsets I-VI based on CD45RA and Foxp3 as indicated. (B) Proportions of subsets compared between non-LE and LE patients before transplant (BT) and on days 3, 5, and 21 post-HCST. Statistical difference by two tailed t-test. *p < 0.05, **p < 0.01, ***p < 0.001. (C, D) Spearman correlation analysis between severity of acute GVHD and (C) Treg subsets (I, top; II, mid; III, bottom) (C) and remaining subsets (IV, top; V, mid; VI, bottom) (D). Spearman correlation coefficient (r) and p value were indicated.
Table 1.
Patients’ characteristics (N=35).
Table 1.
Patients’ characteristics (N=35).
Limbic Encephalitis |
All patients (N=35) |
P value |
Patients with Treg analysis (N=13) |
P value |
Patients with LE (N=4) |
Patients without LE (N=31) |
Patients with LE (N=3) |
Patients without LE (N=10) |
Age, median (range) |
54 (28-60) |
58 (42-72) |
0.429*
|
56 (52-60) |
60 (42-72) |
0.533*
|
Gender, M:F |
3, 1 |
14, 17 |
0.261§
|
2, 1 |
4, 6 |
0.453§
|
Disease AML ALL MDS |
2 (50%) 2 (50%) 0 (0.0%) |
19 (61.3%) 5 (16.1%) 7 (22.6%) |
0.222§
|
2 (66.7%) 1 (33.3%) 0 (0.0%) |
6 (60%) 2 (20%) 2 (20%) |
0.562§
|
Poor Risk status♣
|
4 (100%) |
18 (58.1%) |
0.102§
|
3 (100%) |
7 (70%) |
0.290§
|
Disease status 1st CR 2nd CR MDS Refractory |
4 (100%) 0 (0.0%) 0 (0.0%) 0 (0.0%) |
19 (61.3%) 4 (12.9%) 7 (22.6%) 1 (3.2%) |
0.502§
|
3(100%) 0 (0.0%) 0 (0.0%) 0 (0.0%) |
5 (50%) 2 (20%) 2 (20%) 1 (10%) |
0.466§
|
Conditioning MAC RIC |
2 (50%) 2 (50%) |
17 (57.8%) 14 (45.2%) |
0.855§
|
1 (33.3%) 2 (66.7%) |
6 (60%) 4 (40%) |
0.453§
|
HCT-CI 0 1 2 3- |
4 (100%) 0 (0.0%) 0 (0.0%) 0 (0.0%) |
15 (48.4%) 10 (32.3%) 2 (6.5%) 4 (12.9%) |
0.433§
|
3 (100%) 0 (0.0%) 0 (0.0%) 0 (0.0%) |
6 (60%) 3 (30%) 0 (0.0%) 1 (10%) |
0.370§
|
BMI |
28 (23.4 – 29.4) |
22.9 (18-27.4) |
0.070*
|
26.9 (23.4-29.1) |
23.7 (18-32) |
0.197*
|
Donor Gender, M;F |
4, 0 |
24, 7 |
0.288§
|
3, 0 |
8, 2 |
0.377§
|
Donor Age |
40 (21-60) |
37.5 (50-55) |
0.477*
|
28 (21-52) |
38 (20-55) |
0.559*
|
TNC (x 108 cells/kg) |
12.5 (9.9-14.7) |
11.6 (5.9-20.4) |
0.858*
|
12.4 (9.9-12.6) |
10.9 (5.9-20.4) |
0.897*
|
CD3+cell (x108 cells/kg) |
3.2 (1.7-5.1) |
2.4 (1.3-4.9) |
0.903*
|
3.0 (1.7-3.3) |
2.2 (1.0-4.9) |
0.837*
|
CD34+cell (x106 cells/kg) |
5.2 (2.2-9.6) |
6.7 (4.8-17.7) |
0.081*
|
5.9 (4.5-9.6) |
6.8 (4.8-17.7) |
0.612*
|
CRS grade, median (range) |
2 (2-3) |
1 (0-1) |
0.004§
|
2 (2-2) |
1 (0-1) |
0.004§
|
VOD |
2 (50%) |
3 (9.7%) |
0.030§
|
1 (33.3%) |
0 (0%) |
0.026§
|
CMV reactivation |
2 (50%) |
24 (77.4%) |
0.238§
|
2 (66.7%) |
9 (90%) |
0.201§
|
F/U duration, mo Median (range) |
1.7 (1.5-2.0) |
5 (1 – 16) |
0.096*
|
2.0 (1.5-2.0) |
5.5 (1.0-16.0) |
0.089*
|
Table 2.
Characteristics of 4 patients with LE.
Table 2.
Characteristics of 4 patients with LE.
Patients Number |
Age |
Sex |
Disease |
Initial manifestation |
Day of LE diagnosis after HSCT |
HHV-6 status |
Grade of CRS |
CSF analysis |
EEG |
Hepatic VOD |
Grade of Acute GVHD |
1 |
52 |
F |
AML |
Seizure |
33 |
Negative |
2 |
Not done |
Not done |
No |
3 |
2 |
60 |
M |
AML |
Seizure |
26 |
Negative |
2 |
Not done |
Epileptic wave from right anterior temporal lobe |
Yes |
4 |
3 |
28 |
M |
ALL |
Seizure |
15 |
Negative |
3 |
WBC < 5/µL |
Slow-wave activity with low to medium voltage |
Yes |
4 |
4 |
56 |
M |
ALL |
Seizure |
24 |
Negative |
2 |
WBC < 5/µL |
Slow-wave activity with low to medium voltage |
No |
3 |