<<

Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

CLINICAL RESEARCH | TRANSLATIONAL CANCER MECHANISMS AND THERAPY

Marrow-Infiltrating Regulatory T Cells Correlate with the Presence of Dysfunctional CD4þPD-1þ Cells and Inferior Survival in Patients with Newly Diagnosed Multiple Myeloma Nouf Alrasheed1, Lydia Lee1, Ehsan Ghorani1,2, Jake Y. Henry1,2, Lucia Conde3, Melody Chin1, Daria Galas-Filipowicz1, Andrew J.S. Furness1,2, Selina J. Chavda1, Huw Richards1, Dunnya De-Silva1, Oliver C. Cohen4, Dominic Patel5, Anthony Brooks6, Manuel Rodriguez-Justo5, Martin Pule1, Javier Herrero3, Sergio A. Quezada1,2, and Kwee L. Yong1

ABSTRACT ◥ Purpose: is described in multiple mye- (increased PD-1, LAG-3) compared with patients with lower fre- loma. While preclinical models suggest a role for altered T-cell quencies of Tregs. Analysis of CD4 and CD8 effectors revealed that in disease progression, the contribution of immune low CD4effector (CD4eff):Treg ratio and increased frequency of PD- dysfunction to clinical outcomes remains unclear. We aimed to 1–expressing CD4eff cells were independent predictors of early characterize marrow-infiltrating T cells in newly diagnosed patients relapse over and above conventional risk factors, such as genetic and explore associations with outcomes of first-line therapy. risk and depth of response. Ex vivo functional analysis and RNA Experimental Design: We undertook detailed characterization sequencing revealed that CD4 and CD8 cells from patients with þ of T cells from (BM) samples, focusing on immune greater abundance of CD4effPD-1 cells displayed transcriptional checkpoints and features of immune dysfunction, correlating with and secretory features of dysfunction. clinical features and progression-free survival. Conclusions: BM-infiltrating T-cell subsets, specifically Tregs Results: We found that patients with multiple myeloma had and PD-1–expressing CD4 effectors, negatively influence clinical greater abundance of BM regulatory T cells (Tregs) which, in turn, outcomes in newly diagnosed patients. Pending confirmation in expressed higher levels of the activation marker CD25 compared larger cohorts and further mechanistic work, these immune para- with healthy donors. Patients with higher frequencies of Tregs had meters may inform new risk models, and present potential targets shorter PFS and a distinct Treg immune checkpoint profile for immunotherapeutic strategies.

Introduction evolution, host factors, including immunological fitness and function, likely also influence clinical outcomes of treatment. Multiple myeloma is a common cancer of plasma cells (PC) which is Accumulating evidence points toward a global immune dysregula- responsible for 2% of cancer deaths (1). Despite significant progress tion in multiple myeloma including impaired presentation (3) seen with the inclusion of proteasome inhibitors and immunomod- and impaired T-cell effector function (4) with accumulation of sup- ulatory drugs (IMiD) into the mainstay of treatment regimens (2), pressive cell types (5, 6). These mechanisms appear to converge on myeloma remains almost universally incurable. Along with intrinsic disabling –driven antitumor immunity (7) and accordingly drug sensitivities of tumor cells, and genomic drivers of clonal alterations in T-cell phenotype and function have been consistently reported in models of multiple myeloma. First, T regulatory cells 1Research Department of Haematology, University College London Cancer (Treg) suppress T-cell cytotoxicity and have been reported to be an Institute, London, United Kingdom. 2Cancer Unit, Research Depart- important driver of disease progression (8). Second, there appears to be ment of Haematology, University College London Cancer Institute, London, a relative reduction in cytotoxic T cells relative to Tregs (8). Third, United Kingdom. 3Bill Lyons Informatics Centre, University College London checkpoint proteins, such as the coinhibitory receptor, PD-1, are 4 Cancer Institute, London, United Kingdom. University College London Hospitals reported to be expressed on T cells from patients with multiple NHS Foundation Trust, London, United Kingdom. 5Department of Histopathol- 6 myeloma (9, 10) with increased expression of its ligand PD-L1 on ogy, University College London, London, United Kingdom. Institute of Child fl Health, University College London, London, United Kingdom. tumor cells (11). Despite these reports, the in uence of these altera- tions to T-cell phenotype on patient outcomes remains to be clarified. Note: Supplementary data for this article are available at Clinical Cancer Data regarding Treg numbers and relationship to clinical outcomes are Research Online (http://clincancerres.aacrjournals.org/). conflicting (12–14) and reports of increased PD-1 on T cells from L. Lee and E. Ghorani contributed equally to this article. S.A. Quezada and K.L. patients with multiple myeloma have not been generally corroborated Yong contributed equally to this article. or correlated to outcome (15). Reasons for these discrepancies include Corresponding Authors: Sergio A. Quezada and Kwee L. Yong, UCL Cancer different assay systems, examination of peripheral (PB) versus Institute, 72 Huntley Street, London WC1E 6BT, United Kingdom. Phone: 4420- marrow or the use of heterogenous patient cohorts. Many studies 7679-6139; Fax: 4420-7679-6222; E-mail: [email protected] and [email protected] included relapsed refractory patients, where the host is likely to be affected by prior therapies, repeated infection, and Clin Cancer Res 2020;XX:XX–XX advanced disease. doi: 10.1158/1078-0432.CCR-19-1714 To resolve some of these issues, we investigated the marrow- 2020 American Association for Cancer Research. infiltrating T-cell populations in untreated patients with multiple

AACRJournals.org | OF1

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Alrasheed et al.

tomycin (Gibco; complete medium), at 37C with soluble anti-CD3 Translational Relevance (OKT3) and anti-CD28 (0.5 mg/mL, 15E8; Miltenyi Biotec). GolgiPlug Multiple myeloma is the second most common hematologic (1 mL/mL, BD Biosciences) was added for last 4 hours of incubation. malignancy and remains incurable. Beyond tumor biology and Cells were then stained for surface markers, CD4, CD8, CD69, and genomic features driving disease resistance, host factors includ- fixable viability dye, washed and fixed/permeabilized for staining for ing impaired immunity and frailty also contribute to poor intracellular TNFa, IFNg, IL2, and FoxP3 (Supplementary Table S2). outcomes. Despite reports of immune dysfunction in this cancer, clear evidence for the contribution to clinical outcomes remains RNA sequencing and analysis þ þ þ þ lacking. We show, for the first time, that high abundance of Treg RNA was extracted from flow sorted CD3 CD4 and CD3 CD8 þ and PD-1 CD4 effector cells in bone marrow of newly diag- cells from BM MNCs using ReliaPrep RNA Cell Miniprep System nosed patients are independent predictors of early relapse. This (Promega). cDNA libraries were prepared using the SMART-Seq v4 work supports growing literature on the importance of CD4 Ultra Low Input RNA Kit (Clontech Laboratories, Inc.). Samples were effector cells in multiple myeloma, and confirms a role for the sequenced on two lanes of the HiSeq 3000 instrument (Illumina) using PD-1/PD-L1 axis to multiple myeloma pathobiology. Our work a 75 bp paired-end run at UCL Institute of Child Health (London, UK). þ identifies Tregs and PD-1 CD4 effectors as potential therapeu- RNAseq data were processed with a modified version of the next- tic targets, and opens up avenues for further mechanistic studies flow nf-core RNAseq pipeline (https://github.com/nf-core/rnaseq). into early relapse. Pending confirmationinfuturepatient Reads were trimmed with TrimGalore v0.4.1, aligned against hg19 cohorts, such immune parameters may refine existing risk with STAR v2.5.2a, and duplicated reads were marked with Picard models, facilitating patient stratification for therapeutic strate- v2.18.9. Read counts per gene were generated with featureCounts gies targeting key CD4 populations. v1.6.2 and used for differential gene expression analysis. Gene set enrichment analysis (GSEA) was run using Gene Ontology pathways and previously reported sets of genes differentially expressed by dysfunctional CD4 (16–18) and CD8 T cells (19, 20). Human ortho- myeloma, with focus on Tregs and coinhibitory receptors seeking to logues of mouse genes were identified using Ensembl and NCBI understand the influence of these recognized suppressive T-cell popu- HomoloGene databases. lations on the clinical outcomes of first-line treatment. Statistical analysis Progression-free survival (PFS) was defined as time from start of Materials and Methods first-line therapy to first progression or death [as per International Patients and controls Myeloma Working Group criteria (21)]. Flow cytometric data were Bone marrow (BM) aspirates were obtained from patients with analyzed with FlowJo version 10 (Tree Star Inc). The percentage newly diagnosed (ND) multiple myeloma with written informed of a cell population expressing any given marker is designated as consent (Research ethics committee reference: 07/Q0502/17). Control “frequency” (of that marker) within the relevant Treg, CD4 effector, or BM aspirates (n ¼ 15) were collected from healthy volunteers under- CD8 populations. Statistical analyses were performed with GraphPad going BM harvesting with Anthony Nolan, and subjects undergoing Prism software (Prism 7). P values were calculated using Mann– BM sampling who had no hematologic diagnosis (Supplementary Whitney U test. PFS was estimated using Kaplan–Meier methods Table S1; REC reference: 15/YH/0311). All BM samples were collected with log-rank test. A multivariate Cox regression model was used to in ethylenediamine-tetraacetic acid and processed within 24 hours. evaluate the independent contribution of variables. All tests of signif- Patients were considered to have adverse risk disease if FISH dem- icance were two-sided and P values ≤0.05 considered statistically onstrated one of these: t(4;14), t(14;16), t(14,20), and del(17p). significant.

Isolation of mononuclear cells from BM aspirates BM mononuclear cells (MNC) were isolated by Ficoll Paque (GE Results Healthcare) centrifugation and cryopreserved in FBS (Gibco) contain- Patient characteristics and treatment outcomes ing 10% DMSO (Sigma-Aldrich). Aliquots were subsequently thawed Seventy-eight patients with ND multiple myeloma were identified, for staining and flow cytometry, functional studies, or RNA with median age 59 years (35–86), 64.1% were male (Supplementary sequencing. Table S3). FISH-defined genetic risk was available in 74 patients, of whom 19.2% were adverse risk. All patients commenced active treat- Flow cytometry analysis ment, most (68, 87.18%) with proteasome inhibitor regimens, and 25 Surface antigen staining was performed using the fluorochrome- (31.25%) underwent autologous stem cell transplant (ASCT). Overall conjugated CD3, CD4, PD-1, ICOS, CD25, CD33, CD11b, response rate was 87%, and 53.8% achieved complete response/very CD8, LAG-3, CD4, CD14, CD45RA, CCR7, and fixable viability dye- good partial response (CR/VGPR). With median follow-up of e780. For intracellular staining, cells were fixed/permeabilized using 22 months (1–43), median PFS was not reached. There was a trend the FoxP3 Staining Buffer Set (eBioscience), then for improved PFS with standard risk genetics (P ¼ 0.075 cf high risk), stained with FoxP3, CTLA-4, Ki-67, and GzmB. Details of all anti- ASCT (P ¼ 0.06), and in patients with deeper response (CR/VGPR vs. bodies are in Supplementary Table S2. Data acquisition was on a BD rest, P ¼ 0.09; Supplementary Fig. S1). LSR II Fortessa (BD Biosciences). BM of patients with ND multiple myeloma contains high stimulation experiments frequency of Treg cells Cryopreserved BM MNCs were thawed and cultured at 0.5 1016 We first examined the relative frequencies of T-cell subsets in the cells/mL in RPMI (Lonza), 20%FBS (Gibco), and 1% penicillin/strep- BM of patients with multiple myeloma (gating strategy in Fig. 1A).

OF2 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Dysfunctional Bone Marrow CD4 T Cells in Myeloma

Figure 1. T-cell subsets in BM of patients with ND multiple myeloma. A, Dot plots display gating strategy for CD4 effectors (CD4þFoxP3, B), and Tregs, as (CD4þFoxP3þ, A) and as (FoxP3þCD25þ, C). B, Frequency of Tregs, identified as CD4þFoxP3þ, as % of live MNCs (left), % of live CD4þ cells (middle), and identified as FoxP3þCD25þ as % of live CD4 cells (right) in healthy donors (HD) and patients with myeloma (MM). C, CD4eff:Treg ratio (left) CD8:Treg ratio (middle) and CD4:CD8 ratio (right). Medians indicated (, P < 0.01; ; P < 0.001; , P < 0.0001).

þ þ þ While the frequencies of CD3, CD4, and CD8 cells were comparable CD4 CD25 FoxP3 (3.41% of CD4 cells in multiple myeloma BM with healthy donors (HD; Supplementary Fig. S2A), the frequency vs. 1.27% in HD; P ¼ 0.001; Fig. 1B). þ þ of Treg cells (CD4 FoxP3 ) was significantly higher in BM of The balance between Tregs and effector T cells shapes the antitumor patients with multiple myeloma (0.51% of live MNCs vs. 0.07% (22). We defined CD4 effectors (CD4eff)as þ þ in HD; P < 0.0001, 3.33% of CD4 cells vs. 1.13%; P ¼ 0.0006; CD4 FoxP3 cells, and observed that the CD4eff:Treg ratio in patients Fig. 1B). This was also the case when Tregs were identified as with multiple myeloma was significantly lower when compared with

AACRJournals.org Clin Cancer Res; 2020 OF3

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Alrasheed et al.

Figure 2. Influence of Tregs on PFS. A, Frequency of Tregs (CD4þFoxP3þ cells as % of CD4) in Treglo and Treghi patients (left), PFS in Treglo and Treghi patients (middle), and representative FACS plot for patient with Treghi (top) and Treglo (bottom). , P < 0.0001. B, PFS in patients with high and low CD4eff:Treg ratio (left) and CD8:Treg ratio (right), defined as >median, and ≤median. C, IHC staining for CD138 (red), CD4 (brown), and FoxP3 (blue) from patient with Treghi (left) and Treglo (right). Magnification: 400. Treghi ¼ patients with frequency of Treg > median. Treglo ¼ patients with frequency of Treg ≤ median.

þ þ þ HD (20.83 vs. 140.2; P ¼ <0.0001), this was also the case for the also seen when Tregs were identified as CD4 FoxP3 CD25 cells CD8:Treg ratio (36.34 vs. 170.4; P ¼ <0.0001; Fig. 1C). We found (P ¼ 0.022; Supplementary Fig. S2B). We used surv_cutpoint no correlation between Treg cells, CD4eff:Treg ratio or CD8:Treg function from the “survminer” R package (https://github.com/ ratio with percentage of PCs in BM (Supplementary Fig. S3A). kassambara/survminer) to determine the optimal cut-off value for Neither did we find any correlation of CD4:CD8 ratio with PC Treg frequency, and ascertained this to be 3.31%, which is the infiltration. median value. Having noted that the ratios of effector cells to Tregs in patients Higher frequency of Treg cells is associated with a shorter PFS with multiple myeloma are low compared with HD, we next We sought to determine whether the presence of Treg cells in the examined the association with PFS. We observed that patients with BM of ND patients had any influence on clinical outcomes. We low CD4eff:Treg ratios (≤median) had significantly shorter PFS used PFS, a common primary endpointforstudiesinpatientswith compared with high CD4eff:Treg ratios (>median; HR, 4.22; 95% þ þ multiple myeloma (23). Identifying Tregs as CD4 FoxP3 cells, we CI, 1.79–10.15; P ¼ 0.005; Fig. 2B). There was a weaker association observed that patients with multiple myeloma with a high fre- of CD8:Treg ratio with PFS (P ¼ 0.067; Fig. 2B). Triple color IHC quency of Tregs (>median, Treghi)hadsignificantly shorter PFS was performed on BM trephine biopsies to confirm presence of when compared with patients with multiple myeloma with low Tregs in representative Treghi and Treglo patients (Fig. 2C). There frequency of Tregs [≤median, Treglo;HR,2.91;95%confidence were no associations between CD4 effectors, CD8 cells, or CD4:8 interval (CI), 1.21–7.04; P ¼ 0.021; Fig. 2A]. Similar findings were ratio with PFS (Supplementary Fig. S2B).

OF4 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Dysfunctional Bone Marrow CD4 T Cells in Myeloma

Activation status of Treg cells significant differences in frequencies of PD-1, LAG-3, or CTLA-4 We next examined the phenotype of marrow-infiltrating Tregs, on Tregs from patients with multiple myeloma compared with HD to better understand their influence on clinical outcomes. We (Fig. 3A), there was a greater frequency of PD-1 and LAG-3 on observed higher expression of CD25 on Tregs from patients with Tregs from Treghi patients compared with Treglo (Fig. 3C). These multiple myeloma compared with HD suggesting higher level of differences in checkpoint protein expression suggest that functional activation of multiple myeloma Tregs (Fig. 3A), as CD25 expression as well as quantitative features of marrow-infiltrating Tregs in is associated with Treg activity and suppressive function (24). In patients with multiple myeloma may be important (25, 26). We this cohort of patients with multiple myeloma, both the abundance further explored the differentiation status of BM Tregs in a separate of CD25hi cells and expression intensity of CD25 [mean fluores- cohort of patients with ND multiple myeloma, observing that the cence intensity (MFI)] was greater among Tregs compared with majority are CD45RA indicating that marrow Tregs in these CD4 effectors and CD8 T cells (Fig. 3B). While there were no patients have an activated phenotype (Fig. 3D).

Figure 3. Expression of checkpoint proteins on Tregs. A, Frequency of CD25, PD-1, LAG-3, and CTLA-4 on Tregs (gated as CD4þFoxP3þ) in HD and MM. B, CD25 expression as frequency (left) and MFI (right) on CD4 effectors, CD8, and Tregs. C, Frequency of PD-1, LAG-3, CTLA-4, and CD25 on Tregs (CD4þFoxP3þ)in Treglo (frequency of Treg ≤ median) and Treghi patients (frequency of Treg >median). Mean SEM. MM, patients with myeloma (n ¼ 78, A; n ¼ 43, B). HD, healthy donors (n ¼ 15, A; n ¼ 12, B). D, Resting (CD45RAþ) and activated (CD45RA) Tregs (CD4þFoxP3þ) in a separate cohort of patients with ND multiple myeloma (n ¼ 12; left) and representative FACS plot (right) showing gating for resting and activated Tregs. ns, not significant; , P < 0.05; , P < 0.0001.

AACRJournals.org Clin Cancer Res; 2020 OF5

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Alrasheed et al.

Expression of immune checkpoint proteins on CD4 and CD8 respectively; Fig. 4A and B) with no significant differences in ICOS or effector cells in patients wirth multiple myeloma CTLA-4 (Fig. 4A and B). In addition, a higher percentage of CD8 Next we asked whether altered Treg frequency and activation state T cells from patients with multiple myeloma expressed PD-1 (P ¼ were reflected in effector T-cell function in multiple myeloma BM. 0.045) and the cytotoxic granule GzmB compared with HD (P ¼ 0.01; Examining coinhibitory and coactivation receptors on CD4eff and CD8 Fig. 4B). There was no correlation between the frequency of any T cells, we observed that frequenciesofLAG-3andKi-67werehigheron coinhibitory or coactivation receptor on CD4eff or on CD8 T cells both CD4eff and CD8 T cells from patients with multiple myeloma with disease burden in the BM, except for frequency of LAG-3 on CD8 compared with HD (P ¼ 0.001, P ¼ 0.009, P ¼ 0.0001, P ¼ 0.0001, T cells (r ¼ 0.27, P ¼ 0.028; Supplementary Fig. S3B).

Figure 4. Coactivation and coinhibitory receptors on CD4 and CD8 effector T cells and correlation with PFS. PD-1, LAG-3, ICOS, CTLA-4, GzmB, and Ki-67 expression (% positive) on CD4 effectors (A) and CD8 T cells (B) in HD and patients with multiple myeloma. C, PFS in patients according to frequency of PD-1þ on CD4 effectors (left) þ and CD8 T cells (right). PD-1hi ¼ >median, PD-1lo ¼ ≤median. D, Expression of LAG-3, CTLA-4, and GzmB on PD-1 CD4 effectors from CD4effPD-1lo and CD4effPD-1hi patients (mean SEM), and representative FACS plots of CD4effPD-1lo (top) and CD4effPD-1hi patients (bottom; , P < 0.05; , P < 0.01; , P < 0.001; , P < 0.0001).

OF6 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Dysfunctional Bone Marrow CD4 T Cells in Myeloma

Notably, we observed a positive correlation between Treg frequency CD4eff, we observed that patients with multiple myeloma with more þ þ and the fraction of PD-1 CD4eff and CD8 cells (Supplementary CD4effPD-1 cells (>median, termed CD4effPD-1hi) had significantly þ Fig. S4), but no correlation with the frequency of any other coin- shorter PFS compared with those with less CD4effPD-1 cells (≤medi- hibitory or coactivation receptors. Accordingly, PD-1 expression on an, CD4effPD-1lo; HR, 3.98; 95% CI, 1.66–9.55; P ¼ 0.007; Fig. 4C). In CD4 effectors also correlated with PD-1 on CD8 cells (Supplementary contrast, there was no correlation between frequency of PD-1 on CD8 Fig. S4), and a positive correlation was also noted between PD-1 T cells and PFS (Fig. 4C). There was no correlation between frequency expression on Tregs and on CD4 effectors (Supplementary Fig. S4D). of LAG-3, ICOS, or CTLA4 on either CD4eff or CD8 T cells and PFS To understand the relationship between PD-1 expression and differ- (Supplementary Fig. S7A–S7C). Similarly, no correlation was found entiation status of marrow-infiltrating effector cells, we further studied between GzmB or Ki-67 or on either CD4eff or CD8 T cells and PFS a similar cohort of patients with ND multiple myeloma. Interestingly, (Supplementary Fig. S7D and S7E). while terminally differentiated effector memory cells reexpressing CD45RA comprise a large proportion of CD8 cells, this subset Coinhibitory and coactivation markers on effector T cells from comprises only a minority of CD4 effectors, with the effector memory CD4effPDhi patients (EM) subset being dominant in most patients (Supplementary Given the association with clinical outcomes, we examined þ þ Fig. S5A). PD-1 CD4 effectors were enriched for central memory the CD4effPD-1 cell fraction in multiple myeloma in more detail. þ (CM, CCR7 CD45RA ), and EM (CCR7 CD45RA ) cells when This subset coexpressed the exhaustion markers LAG-3/CTLA-4 compared with PD-1-CD4 effectors (Supplementary Fig. S5B). and the terminal differentiation marker GzmB more frequently in Finally, the frequency of monocytic myeloid-derived suppressor CD4effPD-1hi compared with CD4effPD-1lo patients (P ¼ 0.0035, cells (M-MDSC) in the BM of patients with multiple myeloma was P ¼ 0.046, P ¼ 0.034, respectively; Fig. 4D), suggesting this subset is higher when compared with HD (P ¼ 0.006; Supplementary Fig. S6A). characterized by a dysfunctionalstatethatismorepronounced The frequency of M-MDSCs showed only a weak correlation with among CD4effPD-1hi patients. þ CD4effPD-1 levels (Supplementary Fig. S6). CD4eff:Treg ratio and CD4effPD-1þ cells are independent of Frequency of CD4effPD-1þ T cells correlates with PFS known clinical and cytogenetic predictors of PFS Next we examined the association of coinhibitory receptor expres- Having identified immune features with prognostic value, we þ sion on CD4 and CD8 effectors with clinical outcomes. When we examined both CD4eff:Treg ratio and CDeffPD-1 cell frequency for divided patients into two groups based on the frequency of PD-1 on associations with known clinical prognostic parameters. We found no

Figure 5. Clinical and immune parameters influencing PFS in ND multiple myeloma. A, Forest plot showing hazard ratios and 95% CI for each parameter, by multivariate Cox regression analysis. B, Risk model based on three baseline risk factors: CD4eff:Treg ratio (≤median), CD4eff PD-1 (>median), and genetic risk (high). Group 1 ¼ 0 risk factors (n ¼ 20); Group 2 ¼ 1 risk factor (n ¼ 21); Group 3 ¼ 2 or more risk factors (n ¼ 33).

AACRJournals.org Clin Cancer Res; 2020 OF7

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Alrasheed et al.

association between ISS, genetic risk, ASCT, or response depth with increased suppressive capacity. Tregs contribute to cancer progression þ either CD4eff:Treg ratio or CD4effPD-1 cells (Supplementary Fig. S8). by directly suppressing the effector T-cell activity and here we also A multivariate Cox regression model was built including genetic risk, report that CD4eff:Treg ratio may be independently prognostic in ASCT, ISS and depth of response, and the immune features identified multiple myeloma. We are also the first to present data in multiple above. In this model, CD4eff:Treg ratio retained independent prog- myeloma correlating PD-1 expression on CD4 T cells to patient þ nostic value, along with CD4effPD-1 cells, genetic risk, ASCT, and outcomes, and to impaired cytokine production as well as transcrip- depth of response (Fig. 5A). A risk model was bulit including CD4eff: tional signatures of dysfunctional CD4 and CD8 cells. This supports a þ Treg ratio, CD4effPD-1 cells, and genetic risk, stratifying patients into growing body of evidence underpinning the role of CD4 T cells in the three risk groups based on diagnostic features. Patients with two or antitumor immune response (29), and suggests the independent more risk factors had significantly shorter PFS (Fig. 5B). importance of immune dysregulation on prognosis. Myeloma cells have been shown to promote Treg expansion Effector T cells from CD4effPDhi patients display transcriptional in vivo (8) and in vitro (30). In addition, Treg depletion improves and secretory features of dysfunction survival in a syngeneic murine model of multiple myeloma (8), To gain mechanistic insight into the potential dysfunction of indicating that this is a key immunosuppressive population that effector T cells from CD4effPD-1hi patients, we sorted CD4 and CD8 facilitates disease progression. Previous studies report higher levels cells from CD4effPD-1hi and CD4effPD-1lo patients for RNA of Tregs in PB in patients with multiple myeloma compared with age- sequencing. GSEA carried out using gene sets from previous studies matched controls (6, 12) and in BM compared with patients with of impaired CD4 function (16–18) revealed that CD4 cells from MGUS (14). One study reported that higher levels of Tregs in PB CD4effPD-1hi patients have transcriptional features of CD4 dys- correlated with shorter time to progression (14), but no study has function. Among three gene sets tested, all were enriched among systematically examined Treg numbers and phenotype in the BM of genes differentially expressed by CD4 cells from CD4effPD-1hi ND patients. Ours is the first study to examine BM-infiltrating Tregs patients, although only the Tilstra and colleagues signature reached at diagnosis and significantly extends these earlier reports because we statistical significance (P <0.001; Fig. 6A). Similarly, CD8 T cells show for the first time that the CD4eff:Treg ratio in the tumor from CD4effPD-1hi patients also displayed transcriptional features environment independently associates with clinical outcomes. We of dysfunction (Fig. 6A). We then performed GSEA to identify also observed that increased Treg numbers associated with greater pathways enriched in T cells from CD4effPD-1hi versus CD4effPD- frequencies of the checkpoint proteins, PD-1 and LAG-3 (on Tregs), 1lo patients. Pathways related to activation downstream of T-cell consistent with murine models of multiple myeloma (8). Previous receptor (TCR) signalling, proliferation, and regulation of work has confirmed the suppressive function of Tregs from BM of were enriched in CD4 cells from CD4effPD-1hi patients (Supple- patients with multiple myeloma (31, 32), while expression levels of mentary Fig. S9A). Similar pathways of activation and proliferation these checkpoint proteins is reported to associate with Treg suppres- were also upregulated in CD8 T cells from CD4effPD-1hi patients sive function in other (25, 26, 33). Further functional and (Supplementary Fig. S9A), as described previously fordysfunctional molecular studies on PD-1–expressing Tregs from BM of patients with CD8 T cells (27, 28). multiple myeloma are planned, to provide mechanistic insights. To further explore the notion that T cells from CD4effPD-1hi Tregs actively suppress cytolytic T-cell activity (8), and the ratio patients are functionally impaired, we next assessed cytokine secre- of Tregs to effector cells has been reported to correlate with survival tion by stimulating whole BM MNCs with anti-CD3 and anti-CD28 outcomes (6). In this series of patients, the high frequency of Tregs antibodies. We found that after 6-hour stimulation, there was a in the BM resulted in lower effector T cell: Treg ratios; however, trendtowardhigherTNFa,IFNg, and IL2 production in activated only the CD4eff:Treg ratio significantly correlated with PFS. In þ þ CD4 effectors (CD4 FoxP3 CD69 )fromCD4effPD-1lo patients comparison, there was only a trend of CD8:Treg ratio to outcome þ compared with CD4 effectors from CD4effPD-1hi patients; however, (P ¼ 0.067) which challenges the prevailing view that CD8 T cells only the frequency and intensity (MFI) of TNFa reached statistical are the dominant contributors to antitumor immunity (34). Indeed, significance (P ¼ 0.0043; Fig. 6B; P ¼ 0.0411; Supplementary the antitumor functions of the CD4 tumor compartment are Fig. S9B). A similar pattern was observed with activated CD8 T increasingly recognized (29) which encompasses their helper func- þ þ þ cells (CD8 CD69 )frompatientswithCD4effPD-1lo; these effec- tion for cytotoxic CD8 T cells (35) as well as the ability to directly tors produced more TNFa compared with those from CD4effPD-1hi eliminate tumor (36). In multiple myeloma, CD4-mediated cytol- patients (P ¼ 0.026; Fig. 6B), with a trend toward higher IFNg and ysis of autologous tumor cells has been demonstrated in vitro (37) IL2 production. and in a syngeneic murine myeloma model, direct CD4-mediated Collectively, these data suggest that CD4 effectors and CD8 T cytotoxicity was demonstrated even in the absence of tumor MHC cells from CD4effPD-1hi patients display transcriptional and func- II expression (38). Moreover, in a recent in vivo autograft model, tional features of dysfunction that may contribute to poorer significant reduction in tumor control was observed on depletion of outcomes. either CD4 or CD8 T cells (39). PD-1 is an early marker of the T-cell dysfunction observed in chronic infections and cancer characterized by a hierarchical loss of Discussion effector function and proliferation. Classically, analysis of this dys- We present data correlating the phenotype and function of BM CD4 functional immune state has focused on CD8 T cells (40). Studies in T-cell subsets at diagnosis to clinical outcomes of first-line treatment in small patient cohorts report increased PD-1 levels on CD8 cells in the a large cohort of patients with multiple myeloma. Specifically, we PB and BM of patients with multiple myeloma (10, 41), but we are the report for the first time that patients with a high frequency of marrow- first to show that PD-1 on CD4 cells is prognostic of clinical outcomes. infiltrating Tregs at diagnosis have poorer clinical outcomes. Beyond Despite a correlation between PD-1 on CD4 effectors and CD8 cells, numerical differences, high frequency of Tregs is accompanied by we did not find any association of CD8 parameters with clinical phenotypic changes (increased PD-1 and LAG-3) suggestive of outcomes. On the other hand, the CD8 compartment from

OF8 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Dysfunctional Bone Marrow CD4 T Cells in Myeloma

Figure 6. Effectors in CD4effPD-1hi patients are transcriptionally and functionally distinct from those in CD4effPD-1lo patients. A, GSEA dot plots showing preferential expression of genes related to dysfunction in CD4þ effectors (left) and CD8 cells (right) from CD4effPD-1hi patients, insets refer to gene sets used, NES, normalized enrichment score (B) TNFa (left), IFNg (middle), and IL2 (right) producing CD4 effectors (top) and CD8 effectors (bottom) following stimulation with anti-CD3 and anti-CD28 for 6 hours (, P < 0.05; , P < 0.01).

CD4effPD-1hi patients also (as well as CD4 effectors) manifested latory pathways (42), respectively. In keeping with upregulated TCR reduced cytokine secretion and transcriptional features of dysfunction, signalling, we found enrichment of pathways related to transcrip- þ suggesting that the presence of increased PD-1 CD4 effectors is tion and cell cycle, suggestive of cell activation. While initial reports indicative of a broader, pan-T-cell dysfunctional phenotype. of T-cell dysfunction in murine models of chronic infection indi- Examining transcriptomic profiles of T cells from CD4effPD-1hi cated a near total loss of T-cell effector function (43), it is increas- patients, we observed enrichment of pathways that are character- ingly clear from studies of solid malignancy that the effector istic of T-cell dysfunction. Among both CD4 and CD8 T cells, we potential of dysfunctional T cells is reduced but not absent and found enrichment of both TCR and nonclassical NF-kBpathways active cell proliferation is a key feature of this state (28, 44). indicative of ongoing antigen stimulation and activity of costimu- Consistent with previous reports of T-cell dysfunction (45, 46), we

AACRJournals.org Clin Cancer Res; 2020 OF9

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Alrasheed et al.

additionally observed enrichment of metabolic pathways including release, augmenting T-cell costimulation signals (50). Thus, the oxidative phosphorylation among both subsets and an expression prognostic impact of PD-1 expression on CD4 cells remains to be profile indicative of heightened sensitivity to apoptosis among CD4 confirmed in the context of lenalidomide therapy. but not CD8 T cells. In conclusion, our work demonstrates that increased Tregs Impaired cytokine production by dysfunctional T cells has been in association with dysfunctional CD4 effectors identified by high reported previously (28) and we extend this finding to BM- PD-1 expression correlate with significantly shorter PFS in patients infiltrating T cells in multiple myeloma. Here we tested T-cell with ND multiple myeloma. These data support the importance of cytokine production and found this to be reduced in both CD4 CD4 T cells as mediators of antitumor immunity in myeloma and and CD8 effectors from CD4effPD-1hi patients that reached statis- prompt further mechanistic studies to gain better understanding of tical significance only for TNFa. Larger studies that take into the biology of CD4 dysfunction and Treg function, and open up account several variables such as stimulus, duration of stimulation, therapeutic opportunities for these patients. and cell population are required to confirm these observations. We observed increased numbers of MDSCs in multiple myeloma but Disclosure of Potential Conflicts of Interest further work is needed to explore the contribution of the myeloid M. Pule is an employee/paid consultant for and holds ownership interest compartment to the immune dysfunction in untreated multiple (including patents) in Autolus Ltd. K.L. Yong reports receiving research grants myeloma marrow. fromTakeda,Amgen,Sanofi, and Celgene, and speakers bureau honoraria from In this work, we used patient BM as opposed to PB as we wished Sanofi,Janssen,andAmgen,andisanadvisoryboardmember/unpaidconsultant to examine the multiple myeloma–driving, immune changes within for Janssen. No potential conflicts of interest were disclosed by the other authors. the . Recent in vivo multiple myeloma models report differences in the immune phenotype of circulating Authors’ Contributions and BM-infiltrating T cells (8) in disease, and indicate earlier Conception and design: N. Alrasheed, M. Rodriguez-Justo, M. Pule, S.A. Quezada, changes within the BM immune microenvironment. Similarly, a K.L. Yong study in patient samples also reported functional differences Development of methodology: N. Alrasheed, L. Lee, D. Patel, M. Pule, K.L. Yong between BM and PB effector T cells (47). In addition, we found Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): N. Alrasheed, M. Chin, D. Galas-Filipowicz, A.J.S. Furness, the age of patients did not correlate with CD4eff:Treg ratio or eff S.J. Chavda, H. Richards, O.C. Cohen, D. Patel, A. Brooks CD4 PD-1 cells. However, as our cohort of healthy donors were Analysis and interpretation of data (e.g., statistical analysis, biostatistics, younger, comparisons with patients with myeloma need to be computational analysis): N. Alrasheed, E. Ghorani, J.Y. Henry, L. Conde, interpreted with caution. Anotherpointtonoteisthataminority A.J.S. Furness, S.J. Chavda, H. Richards, A. Brooks, M. Rodriguez-Justo, M. Pule, of patients (10%) had >80% BM PC infiltration, which may have J. Herrero, S.A. Quezada, K.L. Yong amplified differences in marker expression, thus our findings await Writing, review, and/or revision of the manuscript: N. Alrasheed, L. Lee, E. Ghorani, A.J.S. Furness, O.C. Cohen, D. Patel, M. Rodriguez-Justo, S.A. Quezada, confirmation in further patient cohorts. K.L. Yong Our study suggests that immune parameters in BM of untreated Administrative, technical, or material support (i.e., reporting or organizing data, patients with multiple myeloma may inform risk of relapse, and that constructing databases): M. Chin, D. Galas-Filipowicz, D. De-Silva, D. Patel combining such immune features with genetic risk in a new risk Study supervision: S.A. Quezada, K.L. Yong model identifies patients likely to have very poor outcomes. In this patient cohort, we used the median frequency of Tregs (3.31%) as a Acknowledgments cut-off value (confirmed using “survminer”). Pending confirmation The authors thank Prof. David Linch (Formerly Head of the Department of in a larger validation cohort, this measure could be used to identify Haematology at University College London) for valuable comments on this patients with inferior treatment outcomes who may benefitfrom manuscript, and Dr. Nicholas Counsell at Cancer Research UK and UCL Cancer adjunctive immune-directed therapies, for example, Treg depletion Trials Centre for statistical assistance. This study was supported by grants from the Medical Research Council (MR/S001883/1), and by King Faisal Hospital and strategies. Promising agents include IFNa/IFNb receptor antago- Research Centre, Saudi Arabia. This work was undertaken with support from the nists and the use of CD25 antibodies optimized for depletion (22). Cancer Research UK (CRUK)-UCL Centre (C416/A18088), and a Cancer Immu- Blockade of the PD-L1/PD-1 axis has already been explored in notherapy Accelerator Award (CITA-CRUK; C33499/A20265), at University multiple myeloma (48), but in the relapsed refractory setting, and it College London/University College London Hospitals, which is a National remains to be established if checkpoint blockade could overcome Institute for Health Research Biomedcial Research Centre, and a Bloodwise immune dysfunction in ND patients, for example, with high CD4 Research Centre of Excellence. S.A. Quezada is a CRUK Senior Cancer Research Fellowship (C36463/A22246) and is funded by a CRUK Biotherapeutic Program effector levels of PD-1 either as a monotherapy or in combination Grant (C36463/A20764). with Treg-depleting agents. The disappointing results of single- agent checkpoint blockade in multiple myeloma has been suggested The costs of publication of this article were defrayed in part by the payment of page to relate to T-cell senescence rather than exhaustion (49). These charges. This article must therefore be hereby marked advertisement in accordance þ authors, however, only examined CD8 T cells, thus the question of with 18 U.S.C. Section 1734 solely to indicate this fact. the effect of PD-1 blockade on CD4 effector function remains unanswered. Interestingly, only 3 of 78 patients in our cohort Received July 23, 2019; revised December 21, 2019; accepted March 24, 2020; received the IMiD lenalidomide, which acts to enhance cytokine published first March 27, 2020.

References 1. National Center for Health Statistics. Mortality Statistics, National Center 2. Kumar SK, Rajkumar SV, Dispenzieri A, Lacy MQ, Hayman SR, Buadi FK, et al. for Health Statistics (NCHS), Centers for Disease Control and Prevention; Improved survival in multiple myeloma and the impact of novel therapies. Blood 2015. 2007;111:2516–21.

OF10 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Dysfunctional Bone Marrow CD4 T Cells in Myeloma

3. Racanelli V, Leone P, Frassanito MA, Brunetti C, Perosa F, Ferrone S, et al. 23. Durie BGM, Harousseau JL, Miguel JS, Blade J, Barlogie B, Anderson K, et al. Alterations in the antigen processing-presenting machinery of transformed International uniform response criteria for multiple myeloma. Leukemia 2006; plasma cells are associated with reduced recognition by CD8þ T cells and 20:1467–73. characterize the progression of MGUS to multiple myeloma. Blood 2010;115: 24. Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, et al. Functional 1185–93. delineation and differentiation dynamics of human CD4þ T cells expressing the 4. Dosani T, Carlsten M, Maric I, Landgren O. The cellular immune system FoxP3 transcription factor. Immunity 2009;30:899–911. in myelomagenesis: NK cells and T cells in the development of myeloma 25. Stathopoulou C, Gangaplara A, Mallett G, Flomerfelt FA, Liniany LP, Knight D, [corrected] and their uses in . Blood Cancer J 2015;5: et al. PD-1 inhibitory receptor downregulates asparaginyl endopeptidase and e306. maintains Foxp3 transcription factor stability in induced regulatory T cells. 5. An G, Acharya C, Feng X, Wen K, Zhong M, Zhang L, et al. Osteoclasts promote Immunity 2018;49:247–63. immune suppressive microenvironment in multiple myeloma: therapeutic 26. Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Kuchroo VK, implication. Blood 2016;128:1590–603. et al. PD-L1 regulates the development, maintenance, and function of induced 6. Favaloro J, Brown R, Aklilu E, Yang S, Suen H, Hart D, et al. Myeloma skews regulatory T cells. J Exp Med 2009;206:3015–29. regulatory T and pro-inflammatory balance in favor of a 27. Goods BA, Hernandez AL, Lowther DE, Lucca LE, Lerner BA, Gunel M, et al. suppressive state. Leuk Lymphoma 2014;55:1090–8. Functional differences between PD-1þ and PD-1- CD4þ effector T cells in 7. Joshua D, Suen H, Brown R, Bryant C, Ho PJ, Hart D, et al. The T cell in myeloma. healthy donors and patients with glioblastoma multiforme. PLoS One 2017;12: Clin Lymphoma Myeloma Leuk 2016;16:537–42. e0181538. 8. Kawano Y, Zavidij O, Azzi J, Ghobrial IM, Kawano Y, Zwicker JI, et al. Blocking 28. Thommen DS, Koelzer VH, Herzig P, Roller A, Trefny M, Dimeloe S, et al. A IFNAR1 inhibits multiple myeloma – driven Treg expansion and immunosup- transcriptionally and functionally distinct PD-1þ CD8þ T cell pool with pression. J Clin Invest 2018;128:2487–99. predictive potential in non-small-cell lung cancer treated with PD-1 blockade. 9. Rosenblatt J, Glotzbecker B, Mills H, Vasir B, Tzachanis D, Levine JD, et al. PD-1 Nat Med 2018;24:1–11. blockade by CT-011, anti-PD-1 antibody, enhances ex vivo T-cell responses 29. Haabeth OA, Tveita AA, Fauskanger M, Schjesvold F, Berg K, Hofgaard PO, et al. to autologous /myeloma fusion vaccine. J Immunother 2011;34: How do CD4þ T cells detect and eliminate tumor cells that either lack or express 409–18. MHC class II molecules? Front Immunol 2014;5:174. 10. Gorg€ un€ G, Samur MK, Cowens KB, Paula S, Bianchi G, Anderson JE, 30. Tai Y, Lin L, Xing L, Cho S, Yu T, Acharya C, et al. APRIL signaling via TACI et al. Lenalidomide enhances immune checkpoint blockade-induced mediates by T regulatory cells in multiple myeloma: immune response in multiple myeloma. Clin Cancer Res 2015;21: therapeutic implications. Leukemia 2019;33:426–38. 4617–8. 31. Foglietta M, Castella B, Mariani S, Coscia M, Godio L, Ferracini R, et al. The bone 11. Tamura H, Ishibashi M, Yamashita T, Tanosaki S, Okuyama N, Kondo A, marrow of myeloma patients is steadily inhabited by a normal-sized pool of et al. Marrow stromal cells induce B7-H1 expression on myeloma cells, functional regulatory T cells irrespective of the disease status. Haematologica generating aggressive characteristics in multiple myeloma. Leukemia 2012; 2014;99:1605–10. 27:464–72. 32. Atanackovic D, Cao Y, Luetkens T, Panse J, Faltz C, Arfsten J, et al. CD4þ 12. Beyer M, Kochanek M, Giese T, Endl E, Weihrauch MR, Knolle PA, et al. In vivo CD25þ FOXP3þ T regulatory cells reconstitute and accumulate in the bone peripheral expansion of naive CD4 þ CD25 high FoxP3 þ regulatory T cells in marrow of patients with multiple myeloma following allogeneic stem cell patients with multiple myeloma. Blood 2006;107:3940–9. transplantation. Haematologica 2008;93:423–30. 13. Gupta R, Ganeshan P, Hakim M, Verma R, Sharma A, Kumar L. Significantly 33. Camisaschi C, Casati C, Rini F, Perego M, De Filippo A, Triebel F, et al. LAG-3 reduced population in patients with untreated multiple expression defines a subset of CD4 þ CD25 high Foxp3 þ regulatory T cells that myeloma. Leuk Res 2011;35:874–8. are expanded at tumor sites. J Immunol 2010;184:6545–51. 14. Muthu Raja KR, Rihova L, Zahradova L, Klincova M, Penka M, Hajek 34. Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, R. Increased T regulatory cells are associated with adverse clinical et al. Cancer exome analysis reveals a T cell dependent mechanism of cancer features and predict progression in multiple myeloma. PLoS One immunoediting. Nature 2013;482:400–4. 2012;7:e47077. 35. Ahrends T, Spanjaard A, Pilzecker B, Babaøa˛ N, Bovens A, Xiao Y, et al. CD4þ T 15. Sponaas A-M, Yang R, Rustad EH, Standal T, Solvang Thoresen A, Dao Vo C, cell help confers a effector program including coinhibitory et al. PD1 is expressed on exhausted T cells as well as virus specific memory receptor downregulation and increased tissue invasiveness. Immunity 2017;47: CD8þ T cells in the bone marrow of myeloma patients. Oncotarget 2018;9: 848–61. 32024–35. 36. Perez-Diez A, Joncker NT, Choi K, Chan WFN, Anderson CC, Lantz O, et al. 16. Crawford A, Angelosanto JM, Kao C, Doering TA, Odorizzi PM, Barnett BE, CD4 cells can be more efficient at tumor rejection than CD8 cells. Blood 2007; þ et al. Molecular and transcriptional basis of CD4 T cell dysfunction during 109:5346–54. chronic infection. Immunity 2014;40:289–302. 37. Zhang X, Gao L, Meng K, Han C, Li Q, Feng Z, et al. Characterization of CD4 þ T 17. Shin B, Kress RL, Kramer PA, Usmar VMD, Bellis SL, Harrington LE. Effector cell-mediated cytotoxicity in patients with multiple myeloma. Cell Immunol CD4 T cells with progenitor potential mediate chronic intestinal inflammation. 2018;327:62–7. J Exp Med 2018;215:1803–12. 38. Fauskanger M, Haabeth OAW, Skjedal FM, Bogen B, Tveita AA. Tumor killing 18. Tilstra JS, Avery L, Menk AV, Gordon RA, Smita S, Kane LP, et al. Kidney- by CD4 þ T cells is mediated via induction of inducible nitric oxide synthase- infiltrating T cells in murine lupus nephritis are metabolically and functionally dependent cytotoxicity. Front Immunol 2018;9:1684. exhausted. J Clin Invest 2018;128:4884–97. 39. Vuckovic S, Minnie SA, Smith D, Gartlan KH, Watkins TS, Markey KA, et al. 19. Guo X, Zhang Y, Zheng L, Zheng C, Song J, Zhang Q, et al. Global character- Bone marrow transplantation generates T cell – dependent control of myeloma ization of T cells in non-small-cell lung cancer by single-cell sequencing. in mice. J Clin Invest 2019;129:106–21. Nat Med 2018;24:978–85. 40. Thommen DS, Schumacher TN. T Cell Dysfunction in Cancer. Cancer Cell 2018; 20. Zheng C, Zheng L, Yoo J, Guo H, Zhang Y, Guo X. Landscape of infiltrating T 33:547–62. cells in liver cancer revealed by single-cell sequencing resource landscape of 41. Zelle-Rieser C, Thangavadivel S, Biedermann R, Brunner A, Stoitzner P, Will- infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell 2017; enbacher E, et al. T cells in multiple myeloma display features of exhaustion and 169:1342–56. senescence at the tumor site. J Hematol Oncol 2016;9:116. 21. Rajkumar SV, Harousseau J, Durie B, Anderson KC, Dimopoulos M, Kyle R, et al. 42. Gerondakis S, Fulford TS, Messina NL, Grumont RJ. NF-kB control of T cell Consensus recommendations for the uniform reporting of clinical trials: report development. Nat Immunol 2014;15:15–25. of the International Myeloma Workshop Consensus Panel 1. Blood 2011;117: 43. Zajac AJ, Blattman JN, Murali-Krishna K, Sourdive DJD, Suresh M, Altman JD, 4691–6. et al. Viral immune evasion due to persistence of activated T cells without effector 22. Arce Vargas F, Furness AJS, Solomon I, Joshi K, Mekkaoui L, Lesko MH, et al. Fc- function. J Exp Med 1998;188:2205–13. optimized anti-CD25 depletes tumor-infiltrating regulatory T cells and syner- 44. Li H, van der Leun AM, Yofe I, Lubling Y, Gelbard-Solodkin D, van Akkooi ACJ, gizes with PD-1 blockade to eradicate established tumors. Immunity 2017;46: et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated 577–86. compartment within human melanoma. Cell 2019;176:775–89.

AACRJournals.org Clin Cancer Res; 2020 OF11

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Alrasheed et al.

45. Ogando J, Saez ME, Santos J, Nuevo-Tapioles C, Gut M, Esteve-Codina A, 48. Suen H, Brown R, Yang S, Ho P, Gibson J, Joshua D. The failure of immune et al. PD-1 signaling affects cristae morphology and leads to mitochondrial checkpoint blockade in multiple myeloma with PD-1 inhibitors in a phase 1 dysfunction in human CD8þ T . J Immunother Cancer 2019;7: study. Leukemia 2015;29:1621–2. 1–17. 49. Suen H, Brown R, Yang S, Weatherburn C, Ho PJ, Woodland N, et al. Multiple 46. Horton B, Williams J, Cabanov A, Spranger S, Gajewski T. Intratumoral CD8þ myeloma causes clonal T-cell immunosenescence: identification of potential T-cell apoptosis is a major component of T-cell dysfunction and impedes anti- novel targets for promoting tumour immunity and implications for checkpoint tumor immunity. Cancer Immunol Res 2018;6:14–24. blockade. Leukemia 2016;30:1716–24. 47. Choi C, Witzens M, Bucur M, Feuerer M, Sommerfeldt N, Trojan A, et al. 50. Kronke€ J, Udeshi ND, Narla A, Grauman P, Hurst SN, Mcconkey M, et al. Enrichment of functional CD8 memory T cells specific for MUC1 in bone Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple marrow of patients with multiple myeloma. Blood 2005;105:2132–5. myeloma cells. Science 2014;343:301–5.

OF12 Clin Cancer Res; 2020 CLINICAL CANCER RESEARCH

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst March 27, 2020; DOI: 10.1158/1078-0432.CCR-19-1714

Marrow-Infiltrating Regulatory T Cells Correlate with the Presence of Dysfunctional CD4 +PD-1+ Cells and Inferior Survival in Patients with Newly Diagnosed Multiple Myeloma

Nouf Alrasheed, Lydia Lee, Ehsan Ghorani, et al.

Clin Cancer Res Published OnlineFirst March 27, 2020.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-19-1714

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2020/03/27/1078-0432.CCR-19-1714.DC1

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2020/05/29/1078-0432.CCR-19-1714. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 29, 2021. © 2020 American Association for Cancer Research.