Abstract
Classical Hodgkin lymphoma (CHL) is unusually sensitive to PD1 inhibition and PDL1 is highly expressed on CHL cells and in the tumor microenvironment. This could be interpreted as evidence of exhaustion, but paradoxically, PD1+ lymphocyte infiltration does not predict response to PD1 inhibitors and no increase in cytotoxic markers is seen after PD1 therapy as might be expected with reversal of exhaustion. In contrast to PD1, elevated PDL1 does predict response to PD1 inhibitors and recent data associate both retained CHL MHC-II expression and increased T helper (TH) T-cell receptor diversity with response, suggesting a connection to the TH compartment. We performed a phenotypic, spatial and functional assessment of T-cell exhaustion in CHL and found co-expression of an exhaustion marker and lower PD1 expression in CHL than in reactive nodes whereas the proliferative and cytokine production capacity were similar in CHL and the reactive nodes. We found no correlation between PDL1 expression and exhaustion signatures. Instead, we identified a strong association between PDL1 expression and CHL MHC-II expression, TH recruitment, and enrichment of TH1 regulatory cells. These data suggest that a dominant effect of PDL1 expression in CHL may be TH engagement and promotion of a regulatory microenvironment rather than maintenance of exhaustion.
Introduction
Classical Hodgkin lymphoma (CHL) is the only licensed indication for CD279 (PD1, programmed cell death protein 1) inhibitors in hematologic malignancies and is unusually sensitive to this therapy.1,2 PD1 inhibitors are classically thought to act by reversing T-cell exhaustion, a state which limits the effectiveness of anti-tumor immune responses.3 Effector T cells become exhausted when they are chronically stimulated by low levels of antigen.4 Exhausted cells have sustained high expression of PD1, alongside other exhaustion markers, including CD223 (LAG3), TIM3, TBC21 (TBET) and EOMES, and progressively lose their effector functions.4,5 Exhaustion is partially reversible and PD1 inhibition can reinvigorate the T-cell response leading to improved tumor clearance.
CHL cells express high levels of ligand 1 for PD1 (PDL1) and polysomy, copy gains and amplifications of the PDL1 locus are seen in upwards of 95% of cases.6,7 PDL1 expression is also prominent within other cells in the tumor microenvironment.8 However, functional data demonstrating exhaustion in CHL are limited and most studies, including studies from our own laboratory, report only low PD1 expression in the CHL microenvironment.9 Furthermore, PD1+ cell infiltration does not predict response to PD1 inhibitors in CHL.9,10 Recent studies demonstrate that during PD1 inhibitor therapy the CHL microenvironment is characterized by a rapid reduction in PDL1+ macrophages and type 1 regulatory cells (TR1) rather than cytotoxic T-cell expansion that might be expected with reversal of T-cell exhaustion.11 Another study found that expansion of singleton (putatively newly immigrant) T helper (TH) clones is associated with PD1 inhibitor response.12 This is in line with data from solid tumors and suggests that during PD1 therapy activated tumor-specific T cells are in fact newly immigrant and not derived from exhausted populations that were present before therapy.13 These studies run counter to the traditional model of PD1 inhibitors acting by reversing exhaustion and highlight a need to better understand the function of PDL1 within the CHL microenvironment.
In this study we phenotypically and functionally assessed exhaustion in CHL and its relationship to PDL1 expression. Given the evidence of a close connection between PD1 inhibition and TH cells, we further assessed the relationship between PDL1 and TH effector subsets in CHL. We found low levels of exhaustion and no evidence of a connection between an exhausted phenotype and PDL1 expression. However, we identified a strong connection between PDL1 expression and enrichment for the TH1Reg regulatory phenotype, a recently characterized subset of T regulatory cells expressing both FOXP3 and TBET which may specialize in suppression of TH1 cells that are numerous in the CHL microenvironment. These data support and expand upon recent findings that PDL1 expression in CHL may have a closer relationship to the maintenance of a protective tumor microenvironment than to the mediation of exhaustion.
Methods
Human tissue was accessed as part of study 05/Q0605/140, approved by the Regional Ethics Committee, with consent through Barts Tissue Bank and National Health Service Blood and Transplant. Cell lines were obtained from DSMZ. Samples were identified by a systematic search of adult cases at St Bartholomew’s Hospital between 1999 and 2016. The diagnosis was confirmed in all cases by an expert hematopathologist (MC) and Epstein-Barr virus (EBV) status was determined by EBV-encoded small RNA in situ hybridization (sample characteristics in Table 1). Reactive lymph nodes (RLN) from adult patients were selected as controls. Cases positive for human immunodeficiency virus or associated with malignancy were excluded. RLN histology was follicular hyperplasia (64%), progressive transformation of germinal centers (8%) or not otherwise specified (28%). RLN provide a comparative non-malignant immune response within lymphoid tissue and have been used in previous studies.9 Non-reactive nodal tissue is scarce, usually collected during staging of known malignancy and generally small with bland infiltrate and so was considered a less robust control.
Immunohistochemistry
Immunohistochemistry (IHC) and multiplex IHC studies were performed on tissue microarrays. Forty-seven CHL cases were randomly selected for the multiplex experiments and grouped on a single array. Further details are provided in the Online Supplementary Methods.
Imaging mass cytometry
Imaging mass cytometry was performed on tissue micro-arrays on a subset of 24 cases from the multiplex IHC array. Further details are provided in the Online Supplementary Methods.
Cell culture and flow cytometry
For assessment of macrophage PDL1, KMH2-conditioned medium was harvested from high-viability, low-passage cells after 24 hours at 1x106/mL in RPMI medium. Monocytes from healthy donor peripheral blood mononuclear cells were positively selected using anti-CD14 microbeads according to the manufacturer’s instructions before being seeded in 2 mL RPMI medium at 5x105/mL in six-well plates with macrophage colony-stimulating factor 50 ng/mL and granulocyte-macrophage colony-stimulating factor 1 ng/mL. On day 7, macrophages were treated with RPMI or 50% RPMI/conditioned medium. Cells were harvested at 24 hours for flow cytometry analysis. For exhaustion assays, single cell suspensions were stimulated in 200 mL RPMI medium at 1x106/mL in 96-well plates. For cytokine and Ki67 assays, cells were stimulated with phorbol myristate acetate/ionomycin with protein transport inhibitors for 4 hours at 37°C. For proliferation assays the cells were stimulated with 10 mg/mL anti-CD3 and 1 mg/mL anti-CD28 and incubated for 4 days at 37°C after staining with CFSE. For co-cultures, healthy donor naïve CD4+ T cells were separated from peripheral blood mononuclear cells with the Stemcell EasySep kit according to the manufacturer’s instructions before co-culture at 1x106/mL with 4x104/mL KMH2 for 8 days before analysis by flow cytometry. Data were acquired using a BD LSRFortessa with compensation and fluorescence minus one controls at every run. The data were then analyzed using FlowJo software. Further details are available in the Online Supplementary Methods.
Analyses
Images were analyzed in Visiopharm and R. Package details are provided in the Online Supplementary Methods. Spatial distributions were compared to the parent cell distribution (e.g., CD8+ to CD3+ cells) to avoid confounding data due to distribution of parent cell-type or complete spatial randomness where no biologically appropriate parent cell comparator was available. Tukey style boxplot marking was used to show medians and interquartile ranges.
Results
Classical Hodgkin lymphoma cells are PDL1hi and induce a PDL1+ myeloid microenvironment
We quantified PDL1 expression by IHC in 150 patients with CHL and in 28 age-matched patients with RLN. The characteristics of the cohorts are summarized in Table 1. PDL1 expression on CHL cells (CD30+) and macrophages (CD68+) was further assessed by multiplex IHC in 47 CHL cases on a single tissue microarray. PDL1 expression was elevated in CHL (with expression seen on both CHL cells and within the microenvironment) (P<0.0001) (Figure 1A). This effect was seen irrespective of EBV status (data not shown). Mean PDL1 intensity was higher in CHL cells than in macrophages (P<0.0001) (Figure 1A). CD68+ macrophage frequency was increased in regions of high CHL density (P<0.0001). PDL1 intensity increased with macrophage proximity and density to CHL cells (P<0.0001) (Figure 1B). Consistent with this, media conditioned by the KMH2 CHL cell line induced up-regulation of PDL1 on monocyte-derived macrophages in vitro (P<0.01) (Figure 1C). PDL1 upregulation in myeloid cells co-cultured with CHL cell lines is consistent with published data.14 Media conditioned with the non-Hodgkin lymphoma cell lines DHL4 and DHL6 also induced PDL1 expression on monocyte-derived macrophages, but at significantly lower levels than the CHL cell line (data not shown). Phenotyping of microenvironmental antigen-presenting cells (CD68+ macrophages, CD20+ B cells and CD14+ monocytes) by imaging mass cytometry and comparison to RLN demonstrated elevated CD206 intensity but lower B7H4 intensity (both M2 markers) in CHL than CD68+ in RLN. PDL1 expression was highest in CHL cells and CD68+ macrophages. CD14+ monocytes had higher CD16 and CD163 expression but lower mean CD14 expression than their RLN counterparts whereas CD20+ B cells were more likely to express HLADR, CX3CR1 and CD45RA (Figure 1D).
Together, these data confirm, as previously reported, that CHL cells both express high levels of PDL1 and exist in a macrophage-enriched, PDL1+ microenvironment.8,15 Further to this, our data demonstrate that microenvironmental PDL1 expression is mainly seen within the CD68+ macro-phage compartment, and that there is a correlation between macrophage number and the intensity of macrophage PDL1 expression (R=0.6, P<0.0001) (Figure 1E), suggesting that tumors promoting macrophage recruitment were also those promoting microenvironmental PDL1 expression.
PD1+ and phenotypically exhausted cells are infrequent in classical Hodgkin lymphoma, despite high PDL1 expression
After confirming that CHL cells and macrophages in the tumor microenvironment express high levels of PDL1 we sought evidence of a corresponding exhausted PD1+ T-cell population. Exhausted T cells are characterized by sustained PD1 expression and models suggest that cells with intermediate PD1 expression (PD1int) are most amenable to reversal by PD1 inhibition.3,16 We therefore quantified PD1 expression by IHC and stratified the results by PD1 expression intensity. Lymph node germinal centers (and rare residual germinal centers in 5/150 CHL cases) were excluded from analysis because of the high PD1 expression on T follicular helper cells (TFH). The analysis was performed using the most sensitive of three anti-PD1 monoclonal antibody clones tested and results were confirmed with a second clone (Online Supplementary Figure S4). PD1 expression was heterogeneous among cases, with patterns ranging from rare or no PD1 positivity to diffuse infiltration of PD1int, rosetting PD1hi cells and rare cases of marked PD1hi infiltration (Figure 2A). Overall, despite prominent CD3, CD4 and CD8 infiltration, PD1 expression was significantly less frequent in CHL than in RLN (Figure 2B). PD1+ cells were seen at lower frequency in CHL than in RLN irrespective of PD1 expression intensity (Figure 2C). No difference in PD1 expression was seen according to EBV status, but enrichment was seen in the lymphocyte-rich histological subtype, consistent with previous reports.17
Isolated PD1 expression is not specific for exhaustion.18 We therefore assessed co-expression with other markers of exhaustion by multiplex IHC whereby tissue was stained with a panel of IHC antibodies and then the IHC antibody was serially stripped off before re-staining with a new one. Stained images were virtually aligned allowing assessment of cell-marker co-expression. Multiplex IHC was chosen over imaging mass cytometry due to the larger cohort size and analysis of larger tissue areas (three tissue microarray cores versus 1x1 mm) making it preferable for rarer cell populations. PD1 expression was restricted to small lymphocytes and was expressed primarily on CD4+ cells, with some expression on CD8+ cells and minimal non-T-cell expression (Figure 2D). Since CD4 staining produced background when combined with TIM3 and LAG3 in stripping panels, after confirming PD1 T-cell restriction, CD4 was omitted from subsequent stripping panels. PD1+CD8+ and PD1+CD8– lymphocytes were significantly less frequent in CHL than in RLN controls (Figure 2E). PD1+CD8– lymphocytes co-expressed TBET or EOMES at similar levels to RLN controls while PD1+CD8+ lymphocytes co-expressed TBET and EOMES above levels in RLN (Figure 2F). Isolated PD1 expression was the dominant phenotype with very low levels of PD1/TBET/EOMES co-expression.
In summary, despite a more prominent T-cell infiltrate, PD1+ lymphocytes were less frequent in CHL than in RLN, irrespective of the intensity of PD1 expression and there was no enrichment of PD1+CD8– lymphocytes co-expressing exhaustion markers in CHL. The dominant phenotype of both PD1+CD8– and PD1+CD8+ lymphocytes in CHL was expression of PD1 in the absence of co-expression of any other exhaustion marker.
Classical Hodgkin lymphoma T cells are proliferative, cytokine-competent and contain fewer functionally exhausted cells than do reactive lymph node T cells
Proliferation, interleukin 2 (IL2) and interferon gamma (IFNγ) production in T cells from single cell suspensions from diagnostic CHL lymph nodes compared to five RLN single cell suspensions were assessed to determine functional features of TH and cytotoxic T (TC) exhaustion.3–5 Paired formalin-fixed, paraffin-embedded samples were evaluated to ensure that the single cell suspension samples were representative of the IHC cohort. No significant difference in PDL1, CD68, PD1, TBET, GATA3 or RORγT expression was seen, but FOXP3 expression was marginally higher in the single cell suspension group of samples than in the wider IHC cohort (Online Supplementary Figure S5). The characteristics of the cohorts are described in Table 1.
CHL suspensions had higher baseline Ki67+ TH frequencies compared to the RLN controls. No significant differences in proliferation or IL2 and IFNγ production in response to stimulation were seen between CHL and RLN (Figure 3A). Ki67–PD1+ (non-proliferative PD1+) CD3+CD4+ cells were significantly less frequent in CHL than in RLN, while Ki67–PD1+ CD3+CD4– cells were present at similar levels (Figure 3B). Non-cytokine-productive Ki67–PD1+ CD3+CD4+ and CD3+CD4– cells were also significantly less frequent in CHL (Figure 3B). By comparison, no significant difference was seen in numbers of PD1+Ki67+ cells (Figure 3C). On subgroup analysis, CD3+CD4– proliferation and CD3+CD4+ cytokine production capacity were significantly higher in EBV+ CHL than in EBV– CHL (Online Supplementary Figure S6). Further phenotyping by imaging mass cytometry demonstrated elevated expression of TIM3 and modest elevation of TBET in the PD1+Ki67–(CD4+CD3+) TH population, consistent with identification of an exhausted phenotype. In contrast, the proliferative Ki67+PD1–(CD8+CD3+) TC expressed higher levels of granzyme B, while the Ki67+PD1–(CD4+CD3+) TH fraction expressed increased FOXP3 and TBET. Interestingly LAG3 expression was higher in the TH proliferative fraction, perhaps reflecting the presence of TR1 cells which are known to be enriched in CHL (Figure 3D).19 Analysis of mean marker expression in immediate neighbors of phenotypically exhausted TH cells revealed higher expression of CD20, CD45RA (expressed on B cells in addition to naïve T cells) and HLADR with no relationship to CD30, PDL1 or IRF4 (CHL cell markers). In contrast, neighboring cells of phenotypic ally proliferative TH cells expressed higher levels of IRF4, CD30, PDL1 and B7H4 (Figure 3E).
PDL1 expression is not associated with PD1 expression in classical Hodgkin lymphoma
In the light of our findings of low PD1 expression and little functional evidence of exhaustion but high PDL1 expression we assessed the relationship between PD1 and PDL1 expression across CHL cases. Cases varied by both PDL1 and PD1 expression (Figure 4A), but counterintuitively, no association was detected between PD1 and total PDL1 area or PDL1 intensity in CHL, irrespective of stratification for PD1 intensity or analysis of exhaustion marker co-expression (Figure 4B). Unexpectedly, in EBV+ CHL an inverse relationship was observed between PD1 expression and PDL1 intensity (Figure 4B). Analysis of exhaustion marker co-expression demonstrated that isolated PD1 expression rather than co-expression of exhaustion markers (LAG3/TIM3 or EOMES/TBET) was responsible for this effect (Figure 4B). Therefore, compared to RLN controls, we detected significantly fewer phenotypic or functionally exhausted (non-cytokine productive, non-proliferative, PD1+) CD3+CD4+ (TH) and CD3+CD4– (TC) cells and significantly more Ki67+ CD3+CD4+ (TH) cells in the CHL group, with no overall loss in proliferative or cytokine production capacity.
PDL1 is associated with classical Hodgkin lymphoma MHC-II expression, TH cell recruitment and local TC exclusion
In the light of the above data, we considered alternative mechanisms by which PDL1 might influence the CHL microenvironment. We focused on its relationship to TH populations given that both CHL cell MHC-II expression and CD4+ T-cell receptor diversity predict PD1 inhibitor response.6,12 This is in contrast to MHC-I, whose expression does not predict PD1 inhibitor response.20
A positive association was observed between PDL1 expression and CHL MHC-II (Figure 5A), suggesting higher PDL1 expression in CHL cells retaining TH antigen-presenting capacity or deriving other survival advantage from interaction with TH cells in the tumor microenvironment. Of note, no association was identified between PD1 or exhaustion marker expression and CHL MHC II (Online Supplementary Figure S7). CD3 infiltration was increased in CHL and was positively associated with both PDL1 staining and CHL cell MHC II expression (Figure 5B). TH cells were responsible for this pattern: CD4 infiltration correlated positively with PDL1 expression, CHL cell PDL1 intensity, CD68+ macrophage PDL1 intensity and CHL MHC-II expression, while a negative relationship was seen between these markers and CD8 infiltration (Figure 5C, D). Spatial data supported these observations. CD4+ and CD8+ cells were spatially segregated within the tumor, with CD4+ cells proximal to CHL cells while CD8+ cells were frequently excluded from the tumor microenvironment (Figure 5E). The image shows CD8 versus overall CD3 distribution with PDL1 identifying tumor. FOXP3 demonstrates local TReg enrichment). Consistent with this, (CD3+CD8+) TC cells were located significantly further from CHL cells than both CD4+CD3+ TH and CD3+ T cells overall in the imaging mass cytometry analysis (Figure 5F).
We therefore identified an association between PDL1, CHL MHC-II expression and CD4+ enrichment but CD8+ cell exclusion. Given that CHL cells secrete cytokines and actively recruit TH cells this suggests that CHL cases with high PDL1 expression may be those that actively recruit and interact with TH cells.21
PDL1 and classical Hodgkin lymphoma MHC-II are associated with differentiation of TH cells to TH1Reg but not to TH1 or non-polarized TReg cells
We next assessed the relationship between PDL1 and CHL MHC-II expression and TH cell differentiation in CHL. Naïve TH cells differentiate to effector subtypes with different functional roles based upon signals during activation. TH1 and TH17 cells are considered to have anti-tumor roles while TH2 and TReg are tumor protective. TReg also differentiate into subtypes mirroring effector types and may specialize in suppression of specific effector responses (TH1Reg being more effective at suppressing a TH1 response).22 PD1 is upregulated on activation and PDL1-PD1 signaling modulates differentiation, driving development away from TH1 towards TReg subtypes.23,24,24–27 PDL1 also regulates the conversion of differentiated TH1 to TReg and stabilizes inducible TReg conversion to TH1Reg.27–30 PDL1 expression in MHC-II-positive CHL may therefore drive TH1Reg differentiation to combat the anti-tumor TH1 response. To assess this, we first quantified the master transcription factors TBET, FOXP3, RORγT and GATA3 (for TH1, TReg, TH17 and TH2, respectively) by conventional IHC. TBET+ and FOXP3+ cells were enriched in CHL while RORγT+ cells were reduced and no difference was detected in GATA3+ (data not shown). We then used a multiplex IHC panel including CD4, TBET, FOXP3, RORγT and CD8 to define TH1 (TBET+CD4+, FOXP3–RORγT–CD8–), TH1Reg (TBET+FOXP3+CD4+, RORγT-CD8-), non-polarized TReg (FOXP3+CD4+, TBET–RORγT– CD8–), TH17 (RORγT+CD4+, TBET–FOXP3–CD8–) and TH17Reg (RORγT+FOXP3+CD4+, TBET–CD8–). TH1 and TH1Reg were enriched in CHL, whereas TH17, TH17Reg and, surprisingly, nonpolarized TReg were reduced (Figure 6A).
Next, we analyzed the spatial location of these T-cell subsets relative to CHL by imaging mass cytometry. TH1Reg were located significantly closer to (CD30+IRF4+) CHL cells than both TH1 and non-polarized (TBET–) TReg (Figure 6B). CD30 and IRF4 were selected as both are immunohisto-chemical diagnostic markers of CHL. Given these findings we assessed the relationship between TH polarization and both PDL1 and CHL MHC-II expression. Retained expression of MHC-II on CHL was associated with increased TH1Reg, but no association was seen with TH1 or non-polarized TReg. Significant correlations were observed between TH1Reg and PDL1 area, CHL PDL1 intensity and macrophage PDL1 intensity, but again no association was seen with TH1 or non-polarized TReg (Figure 6C, D). On subgroup analysis TH1Reg associations with PDL1 retained significance in both EBV+ CHL and EBV– CHL and in nodular sclerosing and mixed cellularity histological subtypes (data not shown). Associations with MHC-II expression were retained in EBV– CHL and nodular sclerosing subgroups. MHC-II negative cases were too infrequent in EBV+ and mixed cellularity groups for statistical comparison. Finally, co-culture of naïve TH from healthy donors with the KMH2 CHL cell line promoted marked TH1Reg differentiation (CD3+CD4+CD25hiCD127loFOXP3+TBET+) relative to media-activated controls. (Figure 6E) This is in line with previous publications.31 These data reveal that TH1Reg (but not TH1 or non-polarized TReg) enrichment is associated with high PDL1 and retained expression of MHC-II in CHL.
Taken together these data show a lack of enrichment of PD1+ or exhausted cells in CHL and a negative or absent association between PD1 infiltration and PDL1 expression. Instead there is enrichment of Ki67+ cells and a positive association between PDL1 expression in CHL, retained MHC-II expression and both TH and TH1Reg cells (Figure 7).
Discussion
We present data suggesting a role for PDL1 in shaping the CHL tumor microenvironment. We found similar levels of T-cell exhaustion in RLN controls and CHL and no relationship between exhaustion signatures and PDL1 expression. Instead, we identified a consistent association between PDL1 and TH recruitment, TC exclusion and TH1Reg enrichment. These data support and add to recent evidence suggesting that the dominant role for PDL1 in CHL is to modulate the tumor-protective immune tumor microenvironment rather than maintaining an exhausted T-cell infiltrate.11,12
Reversal of exhaustion is the commonly accepted mechanism of action of PD1 inhibitors in both solid and hematologic malignancies. However, recent research on CHL has shown that expansion of novel T-cell clones, increased baseline TH receptor diversity and retained CHL MHC II expression but not increased TC signatures or reinvigoration of existing (putatively exhausted) clones are associated with PD1 inhibitor response.11,12 This raises questions as to whether reversal of exhaustion is the dominant mechanism of action of PD1 inhibitors in CHL. Exhaustion is assumed to be present because of high PDL1 expression and sensitivity to PD1 inhibition; however, this is largely extrapolated from solid tumors and chronic viral infection in which TC exhaustion is well characterized.3 In contrast to solid tumors, CHL cells are derived from professional antigen-presenting cells within a TH-dominated immune environment and frequently lose MHC-I expression. It is therefore plausible that the interactions with immune cells would be different from the classical tumor-exhausted effector relationship. Evidence for exhaustion in CHL is limited. PD1 expression has been noted to be lower in CHL than in reactive tissue and similar to that in diffuse large B-cell lymphoma (which is much less responsive to PD1 inhibitors).9,32 Deep phenotyping has detected a terminally differentiated TH effector subset with TH1-like characteristics.33 No study has demonstrated the presence of functional exhaustion in CHL.
Our data inform and expand upon those in the literature and on previous work from our group in a new cohort, providing a more detailed functional assessment than any other to date but again finding no evidence of exhaustion above that seen in reactive tissue. Importantly, we also demonstrated that the effects of PDL1 both between CHL cases and spatially within individual tumors are not related to exhaustion signatures. This lack of relationship highlights that PDL1 is not a reliable surrogate for exhaustion, and the conclusion that PDL1 may therefore play roles beyond maintaining exhaustion perhaps explains why PDL1 has consistently been reported to predict PD1 inhibitor response in CHL while PD1 never has.6 We went on to identify a consistent link between PDL1 expression and TH1Reg enrichment, supporting our hypothesis that a dominant role of PDL1 within the tumor microenvironment is to sculpt the immune infiltrate by maintaining a regulatory TH environment.
The link between PDL1-PD1 signaling and the development of TH1Reg is supported by a body of literature. Activated T cells upregulate PD1 and become highly sensitive to PDL1-PD1 signaling at even low levels of PD1 expression.23 Engagement of PD1 influences T-cell phenotype at multiple stages of differentiation. During activation PD1 engagement by PDL1 promotes differentiation of naïve TH cells towards TReg and limits the development of effectors such as TH1.24,25,34–36 PDL1 promotes plasticity of fully differentiated TH1 towards TReg.27,28 PDL1 also stabilizes the conversion of inducible TReg to TH1Reg.29,30,37,38 These mechanisms point to a common concept – that of PDL1 expression shifting T-cell development away from anti-tumor effectors including TH1 and promoting development of tumor-protective regulatory cells including TH1Reg. This pattern is borne out by our data. The combined association we observed between PDL1 expression, retained CHL MHC-II expression and TH1Reg enrichment suggests this may at least in part be related to CHL antigen presentation and differentiation of naïve TH to TH1Reg and it is notable that CHL MHC-II expression is also predictive of response to PD1 inhibitors.6 Future work will examine whether the link between PDL1, CHL MHC-II and TH1Reg is via a direct or indirect effect and make comparisons to other PD1-responsive tumor types. The data cited support the possibility of a direct mechanistic link but further evidence is required to demonstrate this. It is noteworthy that while a strong correlation is seen, PD1 inhibitor response is not perfectly predicted by PDL1 or MHC-II expression.6,20 It is likely that PD1 inhibition acts via multiple pathways, beyond both exhaustion and the TH compartment, and other mechanisms including invigoration of innate immunity or B-cell compartments may play important roles.11,12,15 Our findings also provide a perspective on combination therapies such as PD1 and LAG3 co-inhibition, suggesting that rather than both focusing on exhausted cells these approaches may influence two prominent elements of the protective regulatory microenvironment (FOXP3+TBET+ T H1Reg and FOXP3–LAG3+ T R1). This may be particularly appealing given that while both are present irrespective of CHL MHC-II status, TH1Reg are enriched in MHC-II-positive CHL, while TR1 are enriched in and mechanistically linked to CHL MHC-II loss.19
In conclusion, our data build on and enhance the current understanding of the roles of PDL1 within the CHL micro-environment. We provide phenotyping and functional evidence suggesting a limited role for exhaustion in the CHL microenvironment and identify a broader role for PDL1 within the microenvironment with links to MHC-II expression, TH recruitment and the regulatory cell infiltrate. These findings are informative for our understanding of CHL cell survival within the immune microenvironment and together with recent publications reinforce the concept that the reversal of exhaustion may be too narrow a lens with which to view the activity of the PD1-PDL1 axis in CHL.
Footnotes
- Received September 25, 2021
- Accepted June 9, 2022
Correspondence
Disclosures
No conflicts of interest to disclose.
Contributions
JGT and JG designed the experiments, JGT, ET, AC and MC performed the analyses. All authors contributed to writing the manuscript.
Data-sharing statement
Data and further methodological details are available on request.
Funding
This investigation was supported by grant n. 200144/Z/15/Z to JT from the Wellcome Trust and grant funding to JGG from Barts and the London Charity, from Tom and Leona Baker and from The London Clinic Charity.
References
- Bröckelmann PJ, Goergen H, Keller U. Efficacy of nivolumab and AVD in early-stage unfavorable classic Hodgkin lymphoma: the randomized phase 2 German Hodgkin Study Group NIVAHL trial. JAMA Oncol. 2020; 6(6):872-880. https://doi.org/10.1001/jamaoncol.2020.0750PubMedPubMed CentralGoogle Scholar
- Ramchandren R, Domingo-Domènech E, Rueda A. Nivolumab for newly diagnosed advanced-stage classic Hodgkin lymphoma: safety and efficacy in the phase II CheckMate 205 study. J Clin Oncol. 2019; 37(23):1997-2007. https://doi.org/10.1200/JCO.19.00315PubMedPubMed CentralGoogle Scholar
- McLane LM, Abdel-Hakeem MS, Wherry EJ. CD8 T cell exhaustion during chronic viral infection and cancer. Annu Rev Immunol. 2019;37457-495. https://doi.org/10.1146/annurev-immunol-041015-055318PubMedGoogle Scholar
- Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015; 15(8):486-499. https://doi.org/10.1038/nri3862PubMedPubMed CentralGoogle Scholar
- Crawford A, Angelosanto JM, Kao C. Molecular and transcriptional basis of CD4+ T cell dysfunction during chronic infection. Immunity. 2014; 40(2):289-302. https://doi.org/10.1016/j.immuni.2014.01.005PubMedPubMed CentralGoogle Scholar
- Roemer MGM, Redd RA, Cader FZ. 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(10):942-950. https://doi.org/10.1200/JCO.2017.77.3994PubMedPubMed CentralGoogle Scholar
- Green MR, Monti S, Rodig SJ. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood. 2010; 116(17):3268-3277. https://doi.org/10.1182/blood-2010-05-282780PubMedPubMed CentralGoogle Scholar
- Carey CD, Gusenleitner D, Lipschitz M. Topological analysis reveals a PD-L1-associated microenvironmental niche for Reed-Sternberg cells in Hodgkin lymphoma. Blood. 2017; 130(22):2420-2430. https://doi.org/10.1182/blood-2017-03-770719PubMedPubMed CentralGoogle Scholar
- Greaves P, Clear A, Owen A. Defining characteristics of classical Hodgkin lymphoma microenvironment T-helper cells. Blood. 2013; 122(16):2856-2863. https://doi.org/10.1182/blood-2013-06-508044PubMedPubMed CentralGoogle Scholar
- Ansell SM, Lesokhin AM, Borrello I. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015; 372(4):311-319. https://doi.org/10.1056/NEJMoa1411087PubMedPubMed CentralGoogle Scholar
- Reinke S, Bröckelmann PJ, Iaccarino I. Tumor and microenvironment response but no cytotoxic T-cell activation in classic Hodgkin lymphoma treated with anti-PD1. Blood. 2020; 136(25):2851-2863. https://doi.org/10.1182/blood.2020008553PubMedGoogle Scholar
- Cader FZ, Hu X, Goh WL. A peripheral immune signature of responsiveness to PD-1 blockade in patients with classical Hodgkin lymphoma. Nat Med. 2020; 26(9):1468-1479. https://doi.org/10.1038/s41591-020-1006-1PubMedPubMed CentralGoogle Scholar
- Callahan MK, Wolchok JD. Recruit or reboot? How does anti-PD-1 therapy change tumor-infiltrating lymphocytes?. Cancer Cell. 2019; 36(3):215-217. https://doi.org/10.1016/j.ccell.2019.08.009PubMedGoogle Scholar
- Kawashima M, Carreras J, Higuchi H. PD-L1/L2 protein levels rapidly increase on monocytes via trogocytosis from tumor cells in classical Hodgkin lymphoma. Leukemia. 2020; 34(9):2405-2417. https://doi.org/10.1038/s41375-020-0737-9PubMedGoogle Scholar
- Vari F, Arpon D, Keane C. Immune evasion via PD-1/PD-L1 on NK-cells and monocyte/macrophages is more prominent in Hodgkin lymphoma than DLBCL. Blood. 2018; 131(16):1809-1819. https://doi.org/10.1182/blood-2017-07-796342PubMedPubMed CentralGoogle Scholar
- Blackburn SD, Shin H, Freeman GJ, Wherry EJ. Selective expansion of a subset of exhausted CD8 T cells by alphaPD-L1 blockade. Proc Natl Acad Sci U S A. 2008; 105(39):15016-15021. https://doi.org/10.1073/pnas.0801497105PubMedPubMed CentralGoogle Scholar
- Taylor JG, Clear A, Calaminici M, Gribben JG. Programmed cell death protein-1 (PD1) expression in the microenvironment of classical Hodgkin lymphoma is similar between favorable and adverse outcome and does not enrich over serial relapses with conventional chemotherapy. Haematologica. 2019; 104(1):e42-e44. https://doi.org/10.3324/haematol.2018.204818PubMedPubMed CentralGoogle Scholar
- Nam-Cha SH, Roncador G, Sanchez-Verde L. PD-1, a follicular T-cell marker useful for recognizing nodular lymphocyte-predominant Hodgkin lymphoma. Am J Surg Pathol. 2008; 32(8):1252-1257. https://doi.org/10.1097/PAS.0b013e318165b0d6PubMedGoogle Scholar
- Aoki T, Chong LC, Takata K. Single cell transcriptome analysis reveals disease-defining T cell subsets in the tumor microenvironment of classic Hodgkin lymphoma. Cancer Discov. 2020; 10(3):406-421. https://doi.org/10.1158/2159-8290.CD-19-0680PubMedGoogle Scholar
- Roemer MGM, Redd RA, Cader FZ. Expression of major histocompatibility complex (MHC) class II, but not MHC class I, predicts outcome in patients with classical Hodgkin lymphoma (cHL) treated with nivolumab (programmed death-1 [PD-1] blockade). Blood. 2017; 130(Suppl 1):1450. Google Scholar
- Machado L, Jarrett R, Morgan S. Expression and function of T cell homing molecules in Hodgkin’s lymphoma. Cancer Immunol Immunother. 2009; 58(1):85-94. https://doi.org/10.1007/s00262-008-0528-zPubMedGoogle Scholar
- Littringer K, Moresi C, Rakebrandt N. Common features of regulatory T cell specialization during Th1 responses. Front Immunol. 2018; 9:1344. https://doi.org/10.3389/fimmu.2018.01344PubMedPubMed CentralGoogle Scholar
- Riley JL. PD-1 signaling in primary T cells. Immunol Rev. 2009; 229(1):114-125. https://doi.org/10.1111/j.1600-065X.2009.00767.xPubMedPubMed CentralGoogle Scholar
- Francisco LM, Salinas VH, Brown KE. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med. 2009; 206(13):3015-3029. https://doi.org/10.1084/jem.20090847PubMedPubMed CentralGoogle Scholar
- Chen X, Fosco D, Kline DE. PD-1 regulates extrathymic regulatory T-cell differentiation. Eur J Immunol. 2014; 44(9):2603-2616. https://doi.org/10.1002/eji.201344423PubMedPubMed CentralGoogle Scholar
- Zuazo M, Gato-Cañas M, Llorente N. Molecular mechanisms of programmed cell death-1 dependent T cell suppression: relevance for immunotherapy. Ann Transl Med. 2017; 5(19):385. https://doi.org/10.21037/atm.2017.06.11PubMedPubMed CentralGoogle Scholar
- Amarnath S, Mangus CW, Wang JCM. The PDL1-PD1 axis converts human Th1 cells Into regulatory T cells. Sci Transl Med. 2011; 3(111):111ra120. https://doi.org/10.1126/scitranslmed.3003130PubMedPubMed CentralGoogle Scholar
- Kanamori M, Nakatsukasa H, Ito M, Chikuma S, Yoshimura A. Reprogramming of Th1 cells into regulatory T cells through rewiring of the metabolic status. Int Immunol. 2018; 30(8):357-373. https://doi.org/10.1093/intimm/dxy043PubMedGoogle Scholar
- Stathopoulou C, Gangaplara A, Mallett G. Programmed death-1 receptor signaling downregulates asparaginyl endopeptidase to maintain Foxp3 stability in induced regulatory T cells. Immunity. 2018; 49(2):247-263.e7. https://doi.org/10.1016/j.immuni.2018.05.006PubMedPubMed CentralGoogle Scholar
- Tahara M, Kondo Y, Yokosawa M. T-bet regulates differentiation of forkhead box protein 3+ regulatory T cells in programmed cell death-1-deficient mice. Clin Exp Immunol. 2015; 179(2):197-209. https://doi.org/10.1111/cei.12455PubMedPubMed CentralGoogle Scholar
- Tanijiri T, Shimizu T, Uehira K. Hodgkin’s Reed-Sternberg cell line (KM-H2) promotes a bidirectional differentiation of CD4+CD25+Foxp3+ T cells and CD4+ cytotoxic T lymphocytes from CD4+ naive T cells. J Leukoc Biol. 2007; 82(3):576-584. https://doi.org/10.1189/jlb.0906565PubMedGoogle Scholar
- Péricart S, Tosolini M, Gravelle P. Profiling immune escape in Hodgkin’s and diffuse large B-cell lymphomas using the transcriptome and immunostaining. Cancers (Basel). 2018; 10(11):415. https://doi.org/10.3390/cancers10110415PubMedPubMed CentralGoogle Scholar
- Cader FZ, Schackmann RCJ, Hu X. Mass cytometry of Hodgkin lymphoma reveals a CD4+ regulatory T-cell-rich and exhausted T-effector microenvironment. Blood. 2018; 132(8):825-836. https://doi.org/10.1182/blood-2018-04-843714PubMedPubMed CentralGoogle Scholar
- Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M, Saito T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 2012; 209(6):1201-1217. https://doi.org/10.1084/jem.20112741PubMedPubMed CentralGoogle Scholar
- Hui E, Cheung J, Zhu J. T cell costimulatory receptor CD28 is a primary target for PD-1–mediated inhibition. Science. 2017; 355(6332):1428-1433. https://doi.org/10.1126/science.aaf1292PubMedPubMed CentralGoogle Scholar
- Mizuno R, Sugiura D, Shimizu K. PD-1 primarily targets TCR signal in the inhibition of functional T cell activation. Front Immunol. 2019; 10:630. https://doi.org/10.3389/fimmu.2019.00630PubMedPubMed CentralGoogle Scholar
- Cai J, Wang D, Zhang G, Guo X. The role of PD-1/PD-L1 axis In Treg development and function: implications for cancer immunotherapy. Onco Targets Ther. 2019;128437-8445. https://doi.org/10.2147/OTT.S221340PubMedPubMed CentralGoogle Scholar
- Gianchecchi E, Fierabracci A. Inhibitory receptors and pathways of lymphocytes: the role of PD-1 in Treg development and their involvement in autoimmunity onset and cancer progression. Front Immunol. 2018; 9:2374. https://doi.org/10.3389/fimmu.2018.02374PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution 4.0 International License.