Summary:

Aoki and colleagues have utilized single-cell RNA sequencing and imaging mass cytometry to describe the landscape of reactive, nonmalignant immune-cell populations present in classic Hodgkin lymphoma (cHL), and delineate their spatial proximity to malignant Hodgkin–Reed–Sternberg cells. From this study, they have identified a LAG3-expressing Tr1-type Treg cell population as prevalent mainly in MHC-II–negative cHL, implying a potential functional relationship underlying the differential responsiveness of MHC-II–negative versus MHC-II–positive cHLs to immunotherapy.

See related article by Aoki et al., p. 406.

Classic Hodgkin lymphoma (cHL) is unique among hematopoietic cancers in that the tumor mass is primarily composed of nonmalignant immune cells, which are secondary to the much rarer malignant cells (∼1% of total cells in affected lymph nodes), termed Hodgkin–Reed–Sternberg (HRS) cells. This situation can be compared biologically to a reactive lymph node (RLN), where the presence of a pathogenor other inflammatory stimulus triggers local immune-cell migration to and proliferation in the affected lymph node. In cHL, the inflammatory stimulus is the presence of the HRS cells themselves. The HRS cells, however, not only engage nonmalignant immune cells to form a distinctive tumor microenvironment, but also communicate with the cells in this immunologic microenvironment (appropriately described as a multicellular ecosystem) in a manner that is immunosuppressive and favorable to the growth of the tumor. Immune checkpoint inhibitors, particularly the programmed death 1 (PD-1)–blocking antibodies nivolumab and pembrolizumab (1–4) have shown encouraging efficacy in refractory or relapsed cHL, implying the ability to convert the tumor-supporting microenvironment into a tumor-inhospitable one. Notably, HRS expression of PD-L1 and MHC class II has been shown to predict response to PD-1 inhibition with nivolumab (5). Therefore, study of the heterogeneity in cellular composition and immune–antigen expression in cHL could inform future directions to improve cHL treatment.

The innovative study by Aoki and colleagues (6) utilized single-cell RNA sequencing (scRNA-seq) to characterize the nonmalignant immune-cell populations present in cHL lymph nodes, and to distinguish these from cells present in RLNs from healthy donors. From 22 patients with cHL and 5 control RLNs, the authors were able to analyze transcriptomic data from more than 127,000 cells, and thereby group them into 22 phenotypic cell clusters. Treg, Th1, Th2, and Th17 clusters were all amplified in cHL versus RLNs, whereas RLNs were relatively enriched in B-cell and CD8+ T-cell groups. The authors identified a distinct LAG3+CD4+FOXP3 T-cell population in cHL, expressing the cytokines IL10 and TGFβ, which the authors consider to be Tr1-type Tregs, a cell population previously associated with EBV+ cHL (7), which has been shown to suppress effector CD8+ T-cell function in cHL (8). The authors used an HRS-derived cell line supernatant to induce LAG3+ expression in CD4+ T cells in vitro and found that these induced LAG3+ T cells suppressed proliferation of noninduced CD4+ T cells in subsequent coculture, suggesting an immunosuppressive function induced by HRS-derived soluble factors. The antiproliferative effect was confirmed in LAG3 T cells from 4 patients with cHL, cultured ex vivo with or without the patient's LAG3+ T cells: LAG3+ cells reduced both proliferation and TNF production in LAG3 T cells.

Imaging mass cytometry (9), a novel technique combining the spatial resolution of IHC with the multidimensional single-cell analysis potential of suspension mass cytometry (also known as CyTOF), was utilized to identify spatial relationships of the cells identified in scRNA-seq analysis and the HRS cells. HRS cells in MHC-II–negative cHL (where HRS cells are MHC-II negative) were found to be in direct spatial apposition to LAG3+ T cells. Because MHC-II–negative cHLs have been observed to be relatively resistant to anti–PD-1 therapy (5), this histopathologic finding may be relevant to immune checkpoint escape. LAG3 itself was more highly expressed in MHC-II–negative versus MHC-II–positive cHLs within the LAG3+ cell population, and LAG3+ cells were also more abundant in the MHC-II–negative cHLs. In contrast, neither MHC-I nor EBV status, nor histologic subtype of cHL, appeared to affect LAG3+ T-cell abundance or LAG3 expression. In contrast, FOXP3+ T cells were much more abundant in MHC-II–positive cHLs, and could be found in contact with MHC-II–positive HRS cells. In a validation cohort of 166 patients treated with first-line chemotherapy and analyzed immunohistologically, abundance of LAG3+ cells, and also of CD68+ macrophages, consistent with previous studies (10), was found to be a poor prognostic indicator; however, MHC-II negativity itself was not.

This study has several implications: that HRS cells induce LAG3 expression in a population of Tr1 Tregs, by secreted and/or contact-mediated factors; that this primarily occurs in MHC-II–negative cHLs; and that the LAG3+ T cells have an antiproliferative effect on LAG3 T cells, which in turn may contribute to local immunosuppression and potential for immune checkpoint escape. The authors advance the hypothesis that MHC-II on MHC-II–positive HRS cells binds to LAG3 on Tr1 cells and downregulates it by negative feedback (Fig. 1). It is not clear whether this hypothesized LAG3-dependent mechanism of LAG3 suppression would be stable, and indeed it is not yet clear whether MHC-II expression on HRS downregulates LAG3 expression on Tr1 cells, or vice versa. Furthermore, Aoki and colleagues observed PD-1 (PDCD1) expression in subsets of multiple lymphocyte populations, including LAG3+ T cells, and most prominently in LAG3CD4+ T cells, which are present in both MHC-II positive and MHC-II negative cHL (6). Therefore, a mechanism by which LAG3+ T cells would promote resistance to PD-1 blockade in MHC-II–negative cHL is not yet evident. Clearly, there remain substantial gaps to be filled in understanding the mechanism by which immune checkpoint escape is more frequent in MHC-II–negative cHLs (5). Furthermore, whether there are in fact independent prognostic effects of MHC-II negativity and abundant LAG3+ Tr1 cells, or whether these are interdependent, remains unclear, as does the relationship between LAG3+ cell abundance in MHC-II–negative cHLs and FOXP3+ cell abundance in MHC-II–positive cHL. By advancing a spatial resolution–based single-cell analysis of cHL, Aoki and colleagues have opened a new window into a previously shadowy room. Somewhere within may reside keys to improved outcomes for those cHLs currently refractory to immunotherapy. Substantial further studies of the same sort will be needed to identify what and where those keys are.

Figure 1.

MHC-II status of HRS cells in cHL is associated with distinct stromal Treg immunophenotypes. Schematic shows the relationship of MHC-II+ (A) and MHC-II (B) HRS cells to prevalent immunophenotypic Treg populations, as described by Aoki and colleagues (6). A, MHC-II+ HRS cells are surrounded by a reactive stroma rich in FOXP3+ Tregs (shown with yellow cytoplasm), including some in direct cell–cell apposition to the HRS cells, but many separate from them. CD8+ effector T cells (shown with green cytoplasm) are also abundant. LAG3+ Tr1-type Tregs (shown with blue cytoplasm) are rare. Because LAG3 can bind to MHC-II, it is speculated that this interaction may lead to downregulation of LAG3 expression on Tregs, and hence a depletion of cells with the LAG3+ immunophenotype. B, MHC-II HRS cells, in contrast, are surrounded by nests of LAG3+ Tr1-type Tregs. LAG3 expression is promoted by soluble factors derived from HRS cells, but contact-mediated factors may also be important to inducing the LAG3+ cell functional phenotype, because they are found predominantly in direct physical apposition to the HRS cells. LAG3+ Tr1-type Tregs were found to produce IL10 and TGFβ, which can be immunosuppressive factors. The presence of LAG3+ Tr1-type Tregs was shown to mediate an antiproliferative effect on both CD8+ effector T cells and LAG3 CD4+ T cells. Likewise, LAG3+ Tr1-type Tregs lead to suppression of TNF production by other T cells, possibly by means of secreted IL10. The anti-inflammatory effect of LAG3+ Tr1-type Tregs may underlie differences in therapeutic susceptibility of MHC-II versus MHC-II+ cHL cases.

Figure 1.

MHC-II status of HRS cells in cHL is associated with distinct stromal Treg immunophenotypes. Schematic shows the relationship of MHC-II+ (A) and MHC-II (B) HRS cells to prevalent immunophenotypic Treg populations, as described by Aoki and colleagues (6). A, MHC-II+ HRS cells are surrounded by a reactive stroma rich in FOXP3+ Tregs (shown with yellow cytoplasm), including some in direct cell–cell apposition to the HRS cells, but many separate from them. CD8+ effector T cells (shown with green cytoplasm) are also abundant. LAG3+ Tr1-type Tregs (shown with blue cytoplasm) are rare. Because LAG3 can bind to MHC-II, it is speculated that this interaction may lead to downregulation of LAG3 expression on Tregs, and hence a depletion of cells with the LAG3+ immunophenotype. B, MHC-II HRS cells, in contrast, are surrounded by nests of LAG3+ Tr1-type Tregs. LAG3 expression is promoted by soluble factors derived from HRS cells, but contact-mediated factors may also be important to inducing the LAG3+ cell functional phenotype, because they are found predominantly in direct physical apposition to the HRS cells. LAG3+ Tr1-type Tregs were found to produce IL10 and TGFβ, which can be immunosuppressive factors. The presence of LAG3+ Tr1-type Tregs was shown to mediate an antiproliferative effect on both CD8+ effector T cells and LAG3 CD4+ T cells. Likewise, LAG3+ Tr1-type Tregs lead to suppression of TNF production by other T cells, possibly by means of secreted IL10. The anti-inflammatory effect of LAG3+ Tr1-type Tregs may underlie differences in therapeutic susceptibility of MHC-II versus MHC-II+ cHL cases.

Close modal

No potential conflicts of interest were disclosed.

1.
Bair
SM
,
Strelec
LE
,
Feldman
TA
,
Ahmed
G
,
Armand
P
,
Shah
NN
, et al
Outcomes and toxicities of programmed death-1 (PD-1) inhibitors in Hodgkin lymphoma patients in the United States: a real-world, multicenter retrospective analysis
.
Oncologist
2019
;
24
:
955
62
.
2.
Chen
R
,
Zinzani
PL
,
Fanale
MA
,
Armand
P
,
Johnson
NA
,
Brice
P
, et al
Phase II study of the efficacy and safety of pembrolizumab for relapsed/refractory classic Hodgkin lymphoma
.
J Clin Oncol
2017
;
35
:
2125
32
.
3.
Armand
P
,
Engert
A
,
Younes
A
,
Fanale
M
,
Santoro
A
,
Zinzani
PL
, et al
Nivolumab for relapsed/refractory classic Hodgkin lymphoma after failure of autologous hematopoietic cell transplantation: extended follow-up of the multicohort single-arm phase II CheckMate 205 Trial
.
J Clin Oncol
2018
;
36
:
1428
39
.
4.
Chen
R
,
Zinzani
PL
,
Lee
HJ
,
Armand
P
,
Johnson
NA
,
Brice
P
, et al
Pembrolizumab in relapsed or refractory Hodgkin lymphoma: 2-year follow-up of KEYNOTE-087
.
Blood
2019
;
134
:
1144
53
.
5.
Roemer
MGM
,
Redd
RA
,
Cader
FZ
,
Pak
CJ
,
Abdelrahman
S
,
Ouyang
J
, et al
Major histocompatibility complex class II and programmed death ligand 1 expression predict outcome after programmed death 1 blockade in classic Hodgkin lymphoma
.
J Clin Oncol
2018
;
36
:
942
50
.
6.
Aoki
T
,
Chong
LC
,
Takata
K
,
Milne
K
,
Hav
M
,
Colombo
A
, et al
Single-cell transcriptome analysis reveals disease-defining T-cell subsets in the tumor microenvironment of classic Hodgkin lymphoma
.
Cancer Discov
2020
;
10
:
406
21
.
7.
Morales
O
,
Mrizak
D
,
François
V
,
Mustapha
R
,
Miroux
C
,
Depil
S
, et al
Epstein-Barr virus infection induces an increase of T regulatory type 1 cells in Hodgkin lymphoma patients
.
Br J Haematol
2014
;
166
:
875
90
.
8.
Gandhi
MK
,
Lambley
E
,
Duraiswamy
J
,
Dua
U
,
Smith
C
,
Elliott
S
, et al
Expression of LAG-3 by tumor-infiltrating lymphocytes is coincident with the suppression of latent membrane antigen-specific CD8+ T-cell function in Hodgkin lymphoma patients
.
Blood
2006
;
108
:
2280
9
.
9.
Giesen
C
,
Wang
HA
,
Schapiro
D
,
Zivanovic
N
,
Jacobs
A
,
Hattendorf
B
, et al
Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry
.
Nat Methods
2014
;
11
:
417
22
.
10.
Steidl
C
,
Lee
T
,
Shah
SP
,
Farinha
P
,
Han
G
,
Nayar
T
, et al
Tumor-associated macrophages and survival in classic Hodgkin's lymphoma
.
N Engl J Med
2010
;
362
:
875
85
.