In this issue of Blood, Chaturvedi et al examined the T-cell phenotype in a cohort of patients with hemophagocytic lymphohistiocytosis (HLH) vs a cohort with sepsis. They found that, in HLH, there is expansion of a subset of peripheral blood T cells that express CD38 and HLA-DR.1  This profile is most striking for CD8+ T cells, which also exhibit an effector memory phenotype and type 1 cytokine bias. HLH and other cytokine storm syndromes have emerged as areas of importance and interest in the hematology, as well as in the critical care, community. Additionally, there has been growing recognition of the phenotypic overlap between HLH and sepsis, with some sepsis patients even being described as having features of HLH or macrophage activation syndrome (MAS; defined as liver dysfunction, hyperferritinemia, and hematologic failure).2  However, phenotypic overlap does not necessarily mean pathobiological similarity.

In HLH, an increased proportion of CD38high/HLA-DR+ T cells produce cytokines (eg, TNF and IFN-γ) that drive the activation of innate immune cells, such as macrophages, natural killer cells, and dendritic cells. In early sepsis, there is limited to no evidence of CD8 T-cell activation, nor is there increased expression of CD38 or HLA-DR on T cells. The figure was created with BioRender.com.

In HLH, an increased proportion of CD38high/HLA-DR+ T cells produce cytokines (eg, TNF and IFN-γ) that drive the activation of innate immune cells, such as macrophages, natural killer cells, and dendritic cells. In early sepsis, there is limited to no evidence of CD8 T-cell activation, nor is there increased expression of CD38 or HLA-DR on T cells. The figure was created with BioRender.com.

Close modal

CD8+ T cells have always been in the limelight when contemplating the pathogenesis of primary (genetic, inherited) HLH; however, CD8+ T cells have often been ignored in the study of the mechanisms involved in sepsis, with the exception of the pathologic immune system downregulation known as immunoparalysis that often follows severe sepsis.2,3  In murine models of primary HLH, significant and prolonged CD8+ T-cell activation resulting from continued stimulation by activated antigen-presenting cells leads to the profound release of proinflammatory cytokines, such as interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), interleukin-2 (IL-2), and IL-6.4,5  Collectively, these cytokines mediate much of the morbidity and mortality associated with HLH.

Although immunologic data from murine models do not always translate fully in human diseases, the findings of Chaturvedi et al mirror what has been described in primary HLH murine models. Specifically, in the HLH patient samples examined, there was evidence of significant T-cell activation with increased proportions of a unique subset of CD38high/HLA-DR+ cells, with this phenotype more striking for CD8+ cells vs CD4+ cells (see figure). These CD38high/HLA-DR+ CD8+ T cells exhibited an effector memory phenotype marked by type 1 differentiation and increased release of IFN-γ and TNF-α. They were proliferative and exhibited reduced CD5+ expression, suggesting persistent antigen stimulation. Notably, CD38high/HLA-DR+ CD8+ T cells were not restricted to the blood; they were present in other tissues, such as cerebrospinal fluid, lymph nodes, and bone marrow. Although the proportions of CD38high/HLA-DR+ CD8+ cells did not correlate with clinical or laboratory markers of HLH activity at diagnosis, they decreased following HLH treatment, consistent with a reduction in disease activity. Strikingly, the proportions and phenotype of CD38high/HLA-DR+ T cells in patients with sepsis were essentially no different from those in healthy controls.

These findings are congruent with previous findings of reduced HLA-DR expression on monocytes and T cells in sepsis, which has been associated with increased sepsis severity and, in some cases, the development of immunoparalysis.3,6  Notably, when using a cutoff >7% circulating CD38high/HLA-DR+ CD8+ T cells, HLH and early severe sepsis were accurately distinguished from each other with a sensitivity of 100% and specificity of 89%. The findings of Chaturvedi et al are compelling given the challenge of diagnosing HLH and distinguishing it from sepsis, particularly in patients in the critical care unit. With further clinical validation, the assessment of CD38high/HLA-DR+ CD8+ T-cell proportions in the blood may enable a rapid and more accurate differentiation of HLH from sepsis. If so, the use of flow cytometry to characterize T cells could lead to earlier therapeutic interventions. Likewise, it will be a useful tool for assessing disease response. The information presented is relevant when considering frontline treatments for HLH, such as etoposide plus dexamethasone or anti-thymocyte globulin, which ablate activated T cells.7-9  Their findings confirm that T-cell activation is a central feature of murine and human HLH, supporting the use of these agents in the treatment of HLH.

In an earlier study, Amman et al characterized T-cell activation within different HLH subgroups. They found elevated proportions of HLA-DR+ T cells in patients with primary or virally triggered secondary HLH.10  However, T-cell HLA-DR expression was not significantly upregulated in patients with nonvirally induced secondary HLH, mostly patients with MAS.10  These findings are not surprising given the predominance of innate immune system activation in MAS, which is similar to that observed in sepsis.4  These differences suggest that the characteristics of the T-cell activation in secondary HLH may vary depending on the underlying trigger. Thus, possible differences in T-cell HLA-DR expression and other markers, such as CD38 expression, should be evaluated further in different HLH subgroups.

Despite their clinical similarities, it is important to consider immunologic nuances between HLH and sepsis, especially as novel targeted therapies are developed and tested. In light of the study by Chaturvedi et al, therapies targeting T cells might be harmful if given to patients with early sepsis and might exacerbate the later development of immunoparalysis. On the other hand, therapies targeting cytokines released primarily by innate immune cells might be insufficient in the management of HLH. The question then arises, how does a provider marry the broadening knowledge of disease pathophysiology and therapeutic options when confronted with a patient presenting with hyperinflammation with an unclear underlying etiology? Flow cytometric assessment of CD38 and HLA-DR expression on CD8+ T cells might provide data to assist in making the correct diagnosis. Alternatively, one could consider initial treatment with agents that target cytokines more broadly, such as dexamethasone with or without the addition of a JAK inhibitor. Looking to the future, clinical trials should be designed to produce strong clinical and outcome data, as well as robust immunologic information that will better elucidate the underlying disease pathophysiology and direct the best targeted therapy.

Conflict-of-interest disclosure: M.R.H. and K.E.N. receive research support from Incyte Corporation.

1.
Chaturvedi
V
,
Marsh
RA
,
Zoref-Lorenz
A
, et al
.
T-cell activation profiles distinguish hemophagocytic lymphohistiocytosis and early sepsis
.
Blood
.
2021
;
137
(
17
):
2337
-
2346
.
2.
Carcillo
JA
,
Berg
RA
,
Wessel
D
, et al;
Eunice Kennedy Shriver National Institute of Child Health and Human Development Collaborative Pediatric Critical Care Research Network
.
A multicenter network assessment of three inflammation phenotypes in pediatric sepsis-induced multiple organ failure
.
Pediatr Crit Care Med
.
2019
;
20
(
12
):
1137
-
1146
.
3.
Hall
MW
,
Knatz
NL
,
Vetterly
C
, et al
.
Immunoparalysis and nosocomial infection in children with multiple organ dysfunction syndrome
.
Intensive Care Med
.
2011
;
37
(
3
):
525
-
532
.
4.
Albeituni
S
,
Verbist
KC
,
Tedrick
PE
, et al
.
Mechanisms of action of ruxolitinib in murine models of hemophagocytic lymphohistiocytosis
.
Blood
.
2019
;
134
(
2
):
147
-
159
.
5.
Terrell
CE
,
Jordan
MB
.
Perforin deficiency impairs a critical immunoregulatory loop involving murine CD8(+) T cells and dendritic cells
.
Blood
.
2013
;
121
(
26
):
5184
-
5191
.
6.
Ditschkowski
M
,
Kreuzfelder
E
,
Rebmann
V
, et al
.
HLA-DR expression and soluble HLA-DR levels in septic patients after trauma
.
Ann Surg
.
1999
;
229
(
2
):
246
-
254
.
7.
Bergsten
E
,
Horne
A
,
Aricó
M
, et al
.
Confirmed efficacy of etoposide and dexamethasone in HLH treatment: long-term results of the cooperative HLH-2004 study
.
Blood
.
2017
;
130
(
25
):
2728
-
2738
.
8.
Johnson
TS
,
Terrell
CE
,
Millen
SH
,
Katz
JD
,
Hildeman
DA
,
Jordan
MB
.
Etoposide selectively ablates activated T cells to control the immunoregulatory disorder hemophagocytic lymphohistiocytosis
.
J Immunol
.
2014
;
192
(
1
):
84
-
91
.
9.
Mahlaoui
N
,
Ouachée-Chardin
M
,
de Saint Basile
G
, et al
.
Immunotherapy of familial hemophagocytic lymphohistiocytosis with antithymocyte globulins: a single-center retrospective report of 38 patients
.
Pediatrics
.
2007
;
120
(
3
):
e622
-
e628
.
10.
Ammann
S
,
Lehmberg
K
,
Zur Stadt
U
, et al;
HLH study of the GPOH
.
Primary and secondary hemophagocytic lymphohistiocytosis have different patterns of T-cell activation, differentiation and repertoire
.
Eur J Immunol
.
2017
;
47
(
2
):
364
-
373
.
Sign in via your Institution