Immune cell topography predicts response to PD-1 blockade in cutaneous T cell lymphoma

Darci Phillips, Magdalena Matusiak, Belén Rivero Gutierrez, Salil S Bhate, Graham L Barlow, Sizun Jiang, Janos Demeter, Kimberly S Smythe, Robert H Pierce, Steven P Fling, Nirasha Ramchurren, Martin A Cheever, Yury Goltsev, Robert B West, Michael S Khodadoust, Youn H Kim, Christian M Schürch, Garry P Nolan, Darci Phillips, Magdalena Matusiak, Belén Rivero Gutierrez, Salil S Bhate, Graham L Barlow, Sizun Jiang, Janos Demeter, Kimberly S Smythe, Robert H Pierce, Steven P Fling, Nirasha Ramchurren, Martin A Cheever, Yury Goltsev, Robert B West, Michael S Khodadoust, Youn H Kim, Christian M Schürch, Garry P Nolan

Abstract

Cutaneous T cell lymphomas (CTCL) are rare but aggressive cancers without effective treatments. While a subset of patients derive benefit from PD-1 blockade, there is a critically unmet need for predictive biomarkers of response. Herein, we perform CODEX multiplexed tissue imaging and RNA sequencing on 70 tumor regions from 14 advanced CTCL patients enrolled in a pembrolizumab clinical trial (NCT02243579). We find no differences in the frequencies of immune or tumor cells between responders and non-responders. Instead, we identify topographical differences between effector PD-1+ CD4+ T cells, tumor cells, and immunosuppressive Tregs, from which we derive a spatial biomarker, termed the SpatialScore, that correlates strongly with pembrolizumab response in CTCL. The SpatialScore coincides with differences in the functional immune state of the tumor microenvironment, T cell function, and tumor cell-specific chemokine recruitment and is validated using a simplified, clinically accessible tissue imaging platform. Collectively, these results provide a paradigm for investigating the spatial balance of effector and suppressive T cell activity and broadly leveraging this biomarker approach to inform the clinical use of immunotherapies.

Conflict of interest statement

G.P.N. and Y.G. are co-founders and stockholders of Akoya Biosciences, Inc. and inventors on patent US9909167 (On-slide staining by primer extension). D.P., C.M.S., and G.P.N. are inventors on pending patent US62971722 (Spatial method to predict immunotherapy outcome in cancer), filed by Stanford University. C.M.S. is a scientific advisor to Enable Medicine, LLC. Y.H.K. received research funding from Merck & Co. The other authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. Discrimination of malignant and reactive…
Fig. 1. Discrimination of malignant and reactive CD4+ T cells in the CTCL TME.
a Workflow for sample preparation, CODEX, RNAseq, and computational analyses. b Kaplan-Meier overall survival curve, comparing responders and nonresponders (hazard ratio 0.0969 responder/nonresponder; p value calculated by log-rank test). c Pretreatment IHC protein marker expression per patient in responders and nonresponders (mean, red bar). P values calculated by two-sided Wilcoxon’s rank-sum tests (p = not significant (n.s.) for all comparisons). d Representative pretreatment IHC images for select markers from a responder (top) and nonresponder (bottom). e CODEX antibody panel (see also Supplementary Fig. 1e). f Identification of 21 cell types by clustering (see also Supplementary Fig. 2a, b). g Visual verification of reactive (blue crosses) versus malignant (red crosses) CD4+ T cells in CTCL tissue. Scale bars, 20 µm. h, Mean expression of select markers on all malignant (red bars, mean ± s.e.m.) relative to reactive (blue line) CD4+ T cells per tissue microarray spot (pink circles); cores were excluded if they contained <5 CD4+ T cells. P values calculated by two-sided Wilcoxon’s rank-sum tests. i Cell size, measured in pixels/cell, of all malignant (red square) and reactive (blue square) CD4+ T cells (mean ± s.e.m.). P value calculated by a two-sided Wilcoxon’s rank-sum test. j Ranking genes most predictive of tumor cells per tissue microarray spot using an L1-regularized linear model. Red-colored genes have positive predictive coefficients (i.e., most likely to represent tumor cells); gray-colored genes have negative predictive coefficients (i.e., less likely to represent tumor cells). Known CTCL marker genes are highlighted in yellow. Source data are provided as a Source Data file.
Fig. 2. Characterization of the CTCL TME…
Fig. 2. Characterization of the CTCL TME pre- and postpembrolizumab treatment.
a, b Top panels: Representative CODEX seven-color overlay images from a responder (left) and nonresponder (right) pretreatment. Scale bar, 50 µm. Insets, corresponding H&E images; scale bars, 50 µm. Bottom panels: corresponding cell type maps. c Upper pie chart: overall frequencies of tumor, immune and auxiliary cell types. Lower pie chart: overall frequencies of all immune cell types, including CD4+ T cells, CD8+ T cells, Tregs, M1 macrophages, M2 macrophages, and other (B cells, dendritic cells, Langerhans cells, mast cells, neutrophils, and plasma cells). d Cell type frequencies of CD4+ T cell, CD8+ T cell, Treg, M1 macrophage, and M2 macrophage as a percentage of all immune cells per tissue microarray spot across patient groups (mean, red bar). P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons (p = not significant (n.s.) for all comparisons). e, f Pretreatment IFN-γ (e) and TGF-β (f) gene scores per tissue microarray spot in responders and nonresponders. For all box plots: box center line, median; box limits, upper and lower quartiles; box whiskers, 1.5x the interquartile range (IQR). P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons. g, h Immune activation (g) and immunosuppression (h) gene scores, computed on bulk RNA-seq data, per tissue microarray spot across patient groups. Boxes, median ± upper and lower quartiles; whiskers, 1.5x IQR. P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons. Source data are provided as a Source Data file.
Fig. 3. Cellular neighborhoods reveal differences in…
Fig. 3. Cellular neighborhoods reveal differences in the spatial TME organization in responders and nonresponders.
a Cellular neighborhood (CN) analysis schematic. [1] Selection of computational parameters, including the window size (five in this schematic) and the number of CNs to be computed (five in this schematic). [2] Assignment of an index cell (i, center of window) to a given CN based on the composition of cell types within its corresponding window the clustering of windows. [3] Heatmap of cell type distribution for each CN and assignment of CN names. [4] Visualization of CNs as a Voronoi diagram. b Identification of 10 conserved CNs in the CTCL TME using a window size of 10. c Representative Voronoi diagram of the 10 CNs in a responder post-treatment, with the corresponding H&E and seven color fluorescent CODEX images. Scale bar, 20 µm. de Voronoi diagrams of CNs in a responder (d) and nonresponder (e) post-treatment, highlighting CN-5 (tumor and dendritic cells), CN-8 (tumor and CD4+ T cells) and CN-10 (Treg enriched). fh Frequencies of CN-5 (f), CN-8 (g) and CN-10 (h) per tissue microarray spot across patient groups (mean, red bar). P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons. ik Frequencies of ICOS+ CD4+ T cell (i), Ki-67+ CD4+ T cell (j) and ICOS+ Treg (k) as a percentage of all immune cells per tissue microarray spot across patient groups (mean, red bar). P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons. Source data are provided as a Source Data file.
Fig. 4. Spatial relationship between CD4 +…
Fig. 4. Spatial relationship between CD4+ T cells, Tregs and tumor cells predicts pembrolizumab response in CTCL.
aSpatialScore schematic. The SpatialScore is calculated by taking the ratio of the physical distance between each CD4+ T cell and its nearest tumor cell (distance “right”) relative to its nearest Treg (distance “left”). [1] A lower SpatialScore (i.e., CD4+ T cells closer to tumor cells than Tregs) suggests increased T cell effector activity. [2] A higher spatial score (i.e., CD4+ T cells closer to Tregs than tumor cells) suggests increased T cell suppression. bcSpatialScore calculated per cell for all CD4+ T cells (b) and PD-1+ CD4+ T cells (c) across patient groups (mean ± s.e.m.). P values calculated with a linear mixed-effect model taking a patient identifier as a random effect. d GZMB protein expression on PD-1+ CD4+ T cells by CODEX per tissue microarray spot (mean fluorescence intensity (arbitrary units, a.u.), red bar). P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons. e CODEX images showing contact between a tumor cell (cross) and GZMB-expressing PD-1+ CD4+ T cell (arrow) in responder patient 13 post-treatment. Scale bars, 10 µm. f Cytotoxicity gene scores, computed on bulk RNA-seq data, per tissue microarray spot. Boxes, median ± upper and lower quartiles; whiskers, 1.5x IQR. P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons. gh Pre- to post-treatment changes in tumor therapy resistance gene scores, computed on bulk RNA-seq data, per patient in responders (g) and nonresponders (h). Boxes, median ± upper and lower quartiles; whiskers, 1.5x IQR. P values were calculated by two-sided Wilcoxon’s signed-rank tests. i Ki-67+ tumor cell frequencies per tissue microarray spot (mean, red bar). P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons. jSpatialScore calculated from Vectra mIHC data per cell for all PD-1+CD4+ T cells (mean ± s.e.m.). P values calculated with a linear mixed-effect model taking a patient identifier as a random effect. k Vectra mIHC images (left panels) and corresponding spatial plots (right panels)from responder patient 13 (R) and nonresponder patient 14 (NR) pretreatment. Scale bars, 20 µm. lSpatialScore calculated from Vectra mIHC data per patient in responders and nonresponders pretreatment(mean, red bar). P value calculated by a two-sided Wilcoxon’s rank-sum test, with no adjustments for multiple hypotheses. Source data are provided as a Source Data file.
Fig. 5. CXCL13 is a key driver…
Fig. 5. CXCL13 is a key driver of pembrolizumab response in CTCL.
a Seven genes from bulk RNAseq data predictive of the SpatialScore. b Normalized bulk CXCL13 gene expression per tissue microarray spot. Boxes, median ± upper and lower quartiles; whiskers, 1.5x IQR. P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons. c CXCL13 protein expression by IHC per tissue microarray spot (mean, red bar). P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons. d Representative CXCL13 IHC images from responder patient 9 (left panels) and nonresponder patient 14 (right panels). Scale bars, 20 µm. efCXCL13 expression in single-cell transcriptomes from CTCL skin tumors (Gaydosik et al.). e Normalized expression of CXCL13 in single cells; excluded cells with CXCL13 log1p normalized read counts < 0.5. Boxes, median ± upper and lower quartiles; whiskers, 1.5x IQR. f Proportion of CXCL13-expressing cells per cell type. g CIBERSORTx workflow schematic. A CSx deconvolution signature matrix was generated from single-cell transcriptomes (Gaydosik et al.) (left) and applied to CTCL bulk transcriptomes obtained with laser-capture microdissection (LCM) and Smart-3Seq (right) to enumerate cell type fractions and resolve gene expression profiles. h Heatmap correlation of CSx-resolved and CODEX-identified cell type frequencies; Spearman coefficients are on the diagonal. ij Differential expression of CSx-resolved tumor cell genes in responders (j) and nonresponders (k) pre- and post-treatment. P values calculated with a linear mixed-effect model with Benjamini-Hochberg correction (significantly different genes (p < 0.1), red; CXCL13 highlighted yellow). k Vectra mIHC image (top left), corresponding tumor cell depiction (top right), corresponding CXCL13 IHC image (bottom left), and corresponding overlay image of CXCL13 staining and tumor cells (bottom right) in responder patient 9 post-treatment. Scale bars, 20 µm. l Normalized CSx-resolved CXCL13 expression in tumor cells per tissue microarray spot. Boxes, median ± upper and lower quartiles; whiskers, 1.5x IQR. P values calculated with a linear mixed-effect model with Bonferroni’s corrections for multiple comparisons. mn Pre- to post-treatment changes in normalized CXCL13 gene expression from CSx-resolved tumor genes per patient in responders (m) and nonresponders (n). Boxes, median ± upper and lower quartiles; whiskers, 1.5x IQR. P values calculated by two-sided Wilcoxon’s signed-rank tests. o Correlation of CSx-resolved tumor cell CXCL13 expression and bulk CXCR5 expression per tissue microarray spot. Data evaluated with two-sided Spearman test. Source data are provided as a Source Data file.
Fig. 6. Proposed mechanisms of pembrolizumab response…
Fig. 6. Proposed mechanisms of pembrolizumab response in CTCL.
Cartoon depicting proposed mechanisms of pembrolizumab response in therapy responders (top panel) and nonresponders (bottom panel) pre- and post-treatment. The functional immune status of the TME is represented by blue shading when activated and pink shading when suppressed. In nonresponders, the TME is continually immunosuppressed and persistently exhausted PD-1+CD4+ T cells are in closer proximity to potently suppressive ICOS+ Tregs. Moreover, the tumor cells of nonresponders are less susceptible to pembrolizumab therapy. In contrast, responders have a neutral functional immune state pretreatment, which facilitates the baseline spatial organization that underlies a lower SpatialScore and immune cell priming and activation. This enables the transition from exhausted PD-1+ CD4+ T cells to effector/cytotoxic GZMBhi PD-1+ CD4+ T cells following pembrolizumab therapy. Additionally, responder tumor cells are susceptible to PD-1 blockade and overexpress CXCL13 post-treatment, which chemoattracts effector PD-1+ CD4+ T cells toward tumor cells, providing a mechanism for the sustained clinical response seen in responders.

References

    1. Willemze R, et al. The 2018 update of the WHO-EORTC classification for primary cutaneous lymphomas. Blood. 2019;133:1703–1714.
    1. Scarisbrick JJ, et al. Cutaneous Lymphoma International Consortium Study of Outcome in Advanced Stages of Mycosis Fungoides and Sezary Syndrome: Effect of Specific Prognostic Markers on Survival and Development of a Prognostic Model. J. Clin. Oncol. 2015;33:3766–3773.
    1. Wilcox RA. Mogamulizumab: 2 birds, 1 stone. Blood. 2015;125:1847–1848.
    1. Postow MA, Callahan MK, Wolchok JD. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. 2015;33:1974–1982.
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–1355.
    1. Gong J, Chehrazi-Raffle A, Reddi S, Salgia R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J. Immunother. Cancer. 2018;6:8.
    1. Sharpe AH, Pauken KE. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 2018;18:153–167.
    1. Ribas A. Tumor immunotherapy directed at PD-1. N. Engl. J. Med. 2012;366:2517–2519.
    1. Ungewickell A, et al. Genomic analysis of mycosis fungoides and Sezary syndrome identifies recurrent alterations in TNFR2. Nat. Genet. 2015;47:1056–1060.
    1. Lesokhin AM, et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J. Clin. Oncol. 2016;34:2698–2704.
    1. Kantekure K, et al. Expression patterns of the immunosuppressive proteins PD-1/CD279 and PD-L1/CD274 at different stages of cutaneous T-cell lymphoma/mycosis fungoides. Am. J. Dermatopathol. 2012;34:126–128.
    1. Querfeld C, et al. Primary T Cells from Cutaneous T-cell Lymphoma Skin Explants Display an Exhausted Immune Checkpoint Profile. Cancer Immunol. Res. 2018;6:900–909.
    1. Khodadoust MS, et al. Pembrolizumab in Relapsed and Refractory Mycosis Fungoides and Sezary Syndrome: A Multicenter Phase II Study. J. Clin. Oncol. 2020;38:20–28.
    1. Jackson HW, et al. The single-cell pathology landscape of breast cancer. Nature. 2020;578:615–620.
    1. Schurch CM, et al. Coordinated Cellular Neighborhoods Orchestrate Antitumoral Immunity at the Colorectal Cancer Invasive Front. Cell. 2020;182:1341–1359 e1319.
    1. Black S, et al. CODEX multiplexed tissue imaging with DNA-conjugated antibodies. Nat. Protoc. 2021;16:3802–3835.
    1. Goltsev Y, et al. Deep Profiling of Mouse Splenic Architecture with CODEX Multiplexed Imaging. Cell. 2018;174:968–981 e915.
    1. Kennedy-Darling J, et al. Highly multiplexed tissue imaging using repeated oligonucleotide exchange reaction. Eur. J. Immunol. 2021;51:1262–1277.
    1. Phillips D, et al. Highly Multiplexed Phenotyping of Immunoregulatory Proteins in the Tumor Microenvironment by CODEX Tissue Imaging. Front Immunol. 2021;12:687673.
    1. Foley JW, et al. Gene expression profiling of single cells from archival tissue with laser-capture microdissection and Smart-3SEQ. Genome Res. 2019;29:1816–1825.
    1. Newman AM, et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat. Biotechnol. 2019;37:773–782.
    1. Olsen EA, et al. Clinical end points and response criteria in mycosis fungoides and Sezary syndrome: a consensus statement of the International Society for Cutaneous Lymphomas, the United States Cutaneous Lymphoma Consortium, and the Cutaneous Lymphoma Task Force of the European Organisation for Research and Treatment of Cancer. J. Clin. Oncol. 2011;29:2598–2607.
    1. Olsen EA, et al. Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T-cell lymphoma. J. Clin. Oncol. 2007;25:3109–3115.
    1. Watanabe R. Protective and pathogenic roles of resident memory T cells in human skin disorders. J. Dermatol Sci. 2019;95:2–7.
    1. de Masson, A. et al. High-throughput sequencing of the T cell receptor beta gene identifies aggressive early-stage mycosis fungoides. Sci. Transl. Med.10, eaar5894 (2018).
    1. Pulitzer M. Cutaneous T-cell Lymphoma. Clin. Lab Med. 2017;37:527–546.
    1. Iwahara K, Hashimoto K. T-cell subsets and nuclear contour index of skin-infiltrating T-cells in cutaneous T-cell lymphoma. Cancer. 1984;54:440–446.
    1. Picchio MC, et al. CXCL13 is highly produced by Sezary cells and enhances their migratory ability via a synergistic mechanism involving CCL19 and CCL21 chemokines. Cancer Res. 2008;68:7137–7146.
    1. Campbell JJ, Clark RA, Watanabe R, Kupper TS. Sezary syndrome and mycosis fungoides arise from distinct T-cell subsets: a biologic rationale for their distinct clinical behaviors. Blood. 2010;116:767–771.
    1. Suga H, et al. The role of IL-32 in cutaneous T-cell lymphoma. J. Invest Dermatol. 2014;134:1428–1435.
    1. Qu K, et al. Chromatin accessibility landscape of cutaneous T cell lymphoma and dynamic response to HDAC inhibitors. Cancer Cell. 2017;32:27–41 e24.
    1. Jariwala N, et al. TIGIT and Helios Are Highly Expressed on CD4(+) T Cells in Sezary Syndrome Patients. J. Invest Dermatol. 2017;137:257–260.
    1. Capriotti E, et al. Expression of T-plastin, FoxP3 and other tumor-associated markers by leukemic T-cells of cutaneous T-cell lymphoma. Leuk. Lymphoma. 2008;49:1190–1201.
    1. Ascierto ML, et al. An immunologic portrait of cancer. J. Transl. Med. 2011;9:146.
    1. Galon J, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960–1964.
    1. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer. 2019;19:133–150.
    1. Tumeh PC, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571.
    1. Binnewies M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018;24:541–550.
    1. Ayers M, et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 2017;127:2930–2940.
    1. Mariathasan S, et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554:544–548.
    1. Iglesia MD, et al. Prognostic B-cell signatures using mRNA-seq in patients with subtype-specific breast and ovarian cancer. Clin. Cancer Res. 2014;20:3818–3829.
    1. Schuck, A. M. A. R., & Dennis P. In Community Change: Theories, Practice, and Evidence Promoting change through community level interventions Ch. 2, 61-140 (The Aspen Institute, 2006).
    1. Sim GC, et al. IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J. Clin. Invest. 2014;124:99–110.
    1. Tu JF, et al. Regulatory T cells, especially ICOS(+) FOXP3(+) regulatory T cells, are increased in the hepatocellular carcinoma microenvironment and predict reduced survival. Sci. Rep. 2016;6:35056.
    1. Liu H, et al. Reduced cytotoxic function of effector CD8+ T cells is responsible for indoleamine 2,3-dioxygenase-dependent immune suppression. J. Immunol. 2009;183:1022–1031.
    1. Keren L, et al. A Structured Tumor-Immune Microenvironment in Triple Negative Breast Cancer Revealed by Multiplexed Ion Beam Imaging. Cell. 2018;174:1373–1387 e1319.
    1. Carey CD, et al. Topological analysis reveals a PD-L1-associated microenvironmental niche for Reed-Sternberg cells in Hodgkin lymphoma. Blood. 2017;130:2420–2430.
    1. Bagot M, et al. Isolation of tumor-specific cytotoxic CD4+ and CD4+CD8dim+ T-cell clones infiltrating a cutaneous T-cell lymphoma. Blood. 1998;91:4331–4341.
    1. Echchakir H, et al. Cutaneous T cell lymphoma reactive CD4+ cytotoxic T lymphocyte clones display a Th1 cytokine profile and use a fas-independent pathway for specific tumor cell lysis. J. Invest Dermatol. 2000;115:74–80.
    1. Haabeth OA, et al. How Do CD4(+) T Cells Detect and Eliminate Tumor Cells That Either Lack or Express MHC Class II Molecules? Front Immunol. 2014;5:174.
    1. Kitano S, et al. Enhancement of tumor-reactive cytotoxic CD4+ T cell responses after ipilimumab treatment in four advanced melanoma patients. Cancer Immunol. Res. 2013;1:235–244.
    1. Marshall NB, et al. NKG2C/E Marks the Unique Cytotoxic CD4 T Cell Subset, ThCTL, Generated by Influenza Infection. J. Immunol. 2017;198:1142–1155.
    1. Oh DY, et al. Intratumoral CD4(+) T Cells Mediate Anti-tumor Cytotoxicity in Human Bladder Cancer. Cell. 2020;181:1612–1625 e1613.
    1. Shin J, et al. Lesional gene expression profiling in cutaneous T-cell lymphoma reveals natural clusters associated with disease outcome. Blood. 2007;110:3015–3027.
    1. Kari L, et al. Classification and prediction of survival in patients with the leukemic phase of cutaneous T cell lymphoma. J. Exp. Med. 2003;197:1477–1488.
    1. Brown DC, Gatter KC. Ki67 protein: the immaculate deception? Histopathology. 2002;40:2–11.
    1. Abel EJ, et al. Analysis and validation of tissue biomarkers for renal cell carcinoma using automated high-throughput evaluation of protein expression. Hum. Pathol. 2014;45:1092–1099.
    1. Griffin GK, et al. Spatial signatures identify immune escape via PD-1 as a defining feature of T-cell/histiocyte-rich large B-cell lymphoma. Blood. 2021;137:1353–1364.
    1. Borcherding N, et al. Single-Cell Profiling of Cutaneous T-Cell Lymphoma Reveals Underlying Heterogeneity Associated with Disease Progression. Clin. Cancer Res. 2019;25:2996–3005.
    1. Buus TB, et al. Single-cell heterogeneity in Sezary syndrome. Blood Adv. 2018;2:2115–2126.
    1. Fotheringham S, et al. Genome-wide loss-of-function screen reveals an important role for the proteasome in HDAC inhibitor-induced apoptosis. Cancer Cell. 2009;15:57–66.
    1. Gaydosik AM, et al. Single-Cell Lymphocyte Heterogeneity in Advanced Cutaneous T-cell Lymphoma Skin Tumors. Clin. Cancer Res. 2019;25:4443–4454.
    1. Vaque JP, et al. PLCG1 mutations in cutaneous T-cell lymphomas. Blood. 2014;123:2034–2043.
    1. Wolk K, et al. Deficient cutaneous antibacterial competence in cutaneous T-cell lymphomas: role of Th2-mediated biased Th17 function. Clin. Cancer Res. 2014;20:5507–5516.
    1. Politis DN, Romano JP. Large Sample Confidence Regions Based on Subsamples under Minimal Assumptions. Ann. Stat. 1994;22:2031–2050, 2020.
    1. Gardner MJ, Altman DG. Confidence intervals rather than P values: estimation rather than hypothesis testing. Br. Med J. (Clin. Res Ed.) 1986;292:746–750.
    1. Ho J, Tumkaya T, Aryal S, Choi H, Claridge-Chang A. Moving beyond P values: data analysis with estimation graphics. Nat. Methods. 2019;16:565–566.
    1. Campbell DJ, Koch MA. Phenotypical and functional specialization of FOXP3+ regulatory T cells. Nat. Rev. Immunol. 2011;11:119–130.
    1. Gobert M, et al. Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome. Cancer Res. 2009;69:2000–2009.
    1. Kazanietz MG, Durando M, Cooke M. CXCL13 and Its Receptor CXCR5 in Cancer: Inflammation, Immune Response, and Beyond. Front Endocrinol. (Lausanne) 2019;10:471.
    1. Burkle A, et al. Overexpression of the CXCR5 chemokine receptor, and its ligand, CXCL13 in B-cell chronic lymphocytic leukemia. Blood. 2007;110:3316–3325.
    1. Gandini S, Massi D, Mandala M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: A systematic review and meta-analysis. Crit. Rev. Oncol. Hematol. 2016;100:88–98.
    1. Lu S, et al. Comparison of Biomarker Modalities for Predicting Response to PD-1/PD-L1 Checkpoint Blockade: A Systematic Review and Meta-analysis. JAMA Oncol. 2019;5:1195–1204.
    1. Giraldo NA, et al. Multidimensional, quantitative assessment of PD-1/PD-L1 expression in patients with Merkel cell carcinoma and association with response to pembrolizumab. J. Immunother. Cancer. 2018;6:99.
    1. Johnson DB, et al. Quantitative Spatial Profiling of PD-1/PD-L1 Interaction and HLA-DR/IDO-1 Predicts Improved Outcomes of Anti-PD-1 Therapies in Metastatic Melanoma. Clin. Cancer Res. 2018;24:5250–5260.
    1. Tang H, Qiao J, Fu YX. Immunotherapy and tumor microenvironment. Cancer Lett. 2016;370:85–90.
    1. Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 2017;17:559–572.
    1. Klein S, et al. PD-1 blockade activates conventional CD4 T cells and the innate immune response during glioblastoma eradication. J. Immunol. 2018;200:57.59–57.59.
    1. Carstens JL, et al. Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer. Nat. Commun. 2017;8:15095.
    1. Ribas A, Tumeh PC. The future of cancer therapy: selecting patients likely to respond to PD1/L1 blockade. Clin. Cancer Res. 2014;20:4982–4984.
    1. Gu-Trantien, C. et al. CXCL13-producing TFH cells link immune suppression and adaptive memory in human breast cancer. JCI Insight2, e91487 (2017).
    1. Bindea G, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013;39:782–795.
    1. Ignacio RMC, et al. Chemokine Network and Overall Survival in TP53 Wild-Type and Mutant Ovarian Cancer. Immune Netw. 2018;18:e29.
    1. Yang, M. et al. CXCL13 shapes immunoactive tumor microenvironment and enhances the efficacy of PD-1 checkpoint blockade in high-grade serous ovarian cancer. J Immunother Cancer9, e001136 (2021).
    1. Thommen DS, et al. A transcriptionally and functionally distinct PD-1(+) CD8(+) T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat. Med. 2018;24:994–1004.
    1. Goswami, S. et al. ARID1A mutation plus CXCL13 expression act as combinatorial biomarkers to predict responses to immune checkpoint therapy in mUCC. Sci. Transl. Med.12, e001136 (2020).
    1. Jenkins RW, et al. Ex Vivo Profiling of PD-1 Blockade Using Organotypic Tumor Spheroids. Cancer Disco. 2018;8:196–215.
    1. Kamekura R, et al. Circulating PD-1(+)CXCR5(−)CD4(+) T cells underlying the immunological mechanisms of IgG4-related disease. Rheumatol. Adv. Pr. 2018;2:rky043.
    1. Basu A, Yearley JH, Annamalai L, Pryzbycin C, Rini B. Association of PD-L1, PD-L2, and Immune Response Markers in Matched Renal Clear Cell Carcinoma Primary and Metastatic Tissue Specimens. Am. J. Clin. Pathol. 2019;151:217–225.
    1. Seo YD, et al. Mobilization of CD8(+) T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer. Clin. Cancer Res. 2019;25:3934–3945.
    1. Parra ER, et al. Validation of multiplex immunofluorescence panels using multispectral microscopy for immune-profiling of formalin-fixed and paraffin-embedded human tumor tissues. Sci. Rep. 2017;7:13380.
    1. Goksuluk D, Korkmaz S, Zararsiz G, Karaagaoglu A. E. easyROC: an interactive web-tool for ROC curve analysis using R language environment. R. J. 2016;8:213–226.
    1. Samusik N, Good Z, Spitzer MH, Davis KL, Nolan GP. Automated mapping of phenotype space with single-cell data. Nat. Methods. 2016;13:493–496.
    1. Kuznetsova A, Brockhoff PB, Christensen RHB. lmerTest Package: Tests in Linear Mixed Effects Models. J. Stat. Softw. 2017;82:26.
    1. Quail MA, et al. A large genome center’s improvements to the Illumina sequencing system. Nat. Methods. 2008;5:1005–1010.
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.
    1. Bates D, Mächler M, Bolker B, Walker S. Fitting Linear Mixed-Effects Models Usinglme4. J. Stat. Softw. 2015;67:48.
    1. Stuart T, et al. Comprehensive Integration of Single-Cell Data. Cell. 2019;177:1888–1902 e1821.
    1. Blondel VD, Guillaume J-L, Lambiotte R, Lefebvre E. Fast unfolding of communities in large networks. J. Stat. Mech.: Theory Exp. 2008;2008:P10008.
    1. McInnes, L., Healy, J. & Melville, J. Umap: Uniform manifold approximation and projection for dimension reduction. arXiv preprint arXiv:1802.03426 (2018).
    1. Wickham, H. ggplot2: elegant graphics for data analysis. (Springer, 2016).
    1. Slowikowski, K. ggrepel: Repulsive text and label geoms for ‘ggplot2’. R package version 0.65 (2016).

Source: PubMed

3
Abonnieren