Chemokine Analysis in Patients with Metastatic Uveal Melanoma Suggests a Role for CCL21 Signaling in Combined Epigenetic Therapy and Checkpoint Immunotherapy

Vasu R Sah, Henrik Jespersen, Joakim Karlsson, Lisa M Nilsson, Mattias Bergqvist, Iva Johansson, Ana Carneiro, Hildur Helgadottir, Max Levin, Gustav Ullenhag, Anders Ståhlberg, Roger Olofsson Bagge, Jonas A Nilsson, Lars Ny, Vasu R Sah, Henrik Jespersen, Joakim Karlsson, Lisa M Nilsson, Mattias Bergqvist, Iva Johansson, Ana Carneiro, Hildur Helgadottir, Max Levin, Gustav Ullenhag, Anders Ståhlberg, Roger Olofsson Bagge, Jonas A Nilsson, Lars Ny

Abstract

Purpose: Patients with metastatic uveal melanoma have limited therapeutic options and high mortality rate so new treatment options are needed.

Patients and methods: We previously reported that patients treated with the PD-1 inhibitor pembrolizumab and the histone deacetylase inhibitor entinostat in the PEMDAC trial, experienced clinical benefits if their tumor originated from iris or was wildtype for BAP1 tumor suppressor gene. Here we present the 2-year follow-up of the patients in the PEMDAC trial and identify additional factors that correlate with response or survival.

Results: Durable responses were observed in 4 patients, with additional 8 patients exhibiting a stable disease. The median overall survival was 13.7 months. Grade 3 adverse events were reported in 62% of the patients, but they were all manageable. No fatal toxicity was observed. Activity of thymidine kinase 1 in plasma was higher in patients with stable disease or who progressed on treatment, compared with those with partial response. Chemokines and cytokines were analyzed in plasma. Three chemokines were significantly different when comparing patients with and without response. One of the factors, CCL21, was higher in the plasma of responding patients before treatment initiation but decreased in the same patients upon treatment. In tumors, CCL21 was expressed in areas resembling tertiary lymphoid structures (TLS). High plasma levels of CCL21 and presence of TLS-like regions in the tumor correlated with longer survival.

Conclusions: This study provides insight into durable responses in the PEMDAC trial, and describes dynamic changes of chemokines and cytokines in the blood of these patients.

Significance: The most significant finding from the 2-year follow-up study of the PEMDAC trial was that high CCL21 levels in blood was associated with response and survival. CCL21 was also expressed in TLS-like regions and presence of these regions was associated with longer survival. These analyses of soluble and tumor markers can inform on predictive biomarkers needing validation and become hypothesis generating for experimental research.

© 2023 The Authors; Published by the American Association for Cancer Research.

Figures

FIGURE 1
FIGURE 1
A, Kaplan–Meier analysis showing PFS of all patients. B, Kaplan–Meier analysis showing OS of all patients except one. C, Swimmer plot showing time on treatment, time to best response, and duration of response in all patients who received at least one dose of study drug (n  =  29) are shown. D, Kaplan–Meier analysis comparing OS between patients with LDH baseline greater or lower than the upper limit of normal (ULN).
FIGURE 2
FIGURE 2
A and B, Kaplan–Meier analysis showing PFS and OS of patients with a wildtype BAP1 status and UV-damaged uveal melanoma genome. PFS (C) and OS (D) analyses comparing patients with GNAQ- or GNA11-mutated uveal melanoma. E, Volcano plot showing differentially expressed genes between short term (n = 16) and long term (alive patients, n = 4) from bulk RNA-seq. Genes with FDR-adjusted P values <0.05 were considered statistically significant. F, Individual box plots showing relevant gene signatures implicating long-term (alive patients) survival. Statistical tests were carried out using DESeq2 and FDR-adjusted P values were denoted with *, P < 0.05; **, P < 0.01; ***, P < 0.001.
FIGURE 3
FIGURE 3
A, Heatmap showing TK1 activity value (DuA) from PEMDAC patient plasma samples, grouped within response groups. Each square is a timepoint for each patient and shows TK1 levels from pretreatment to end of study, until otherwise stated. Total plasma samples analyzed for TK1 = 287. B, Longitudinal TK1 activity for individual patients, as shown in A. C and D, Kaplan–Meier analysis showing PFS and OS, respectively for pretreatment TK1 values using a threshold of 150 DuA (median TK1 for all samples = 113). Patients with nonavailability of pretreatment samples were excluded from the analysis. E, IHC of TK1 showing nuclear/cytoplasmic magenta staining in patient biopsies 2-027, 3-012, and 3-010. F, Comparison between pretreatment TK1 values for short- and long-term survivors. G, Correlation between pretreatment TK1 (DuA) and circulating tumor DNA (counts/mL) matched patient samples (n = 21). All statistical tests were unpaired two-tailed t tests, assuming equal variance, with *, P < 0.05; **, P < 0.01; ***, P < 0.001.
FIGURE 4
FIGURE 4
A, Heatmap of 71 chemokines and cytokines analyzed among all patients and their respective timepoints. Total plasma samples analyzed = 287. Each square represents a timepoint for each patient and shows response group-based levels from pretreatment until end of study, unless otherwise stated. B, Heatmap showing differential pretreatment values between PD and partial responders. Boxed chemokines are significant (Padjusted < 0.05) after FDR correction. C, Individual chemokine or cytokine values (pg/mL) compared among different response groups. Only significantly different chemokines from B are included. D, Fold change difference between pretreatment values and week 9 after start of treatment are shown for PD patients. Arrows indicate chemokines that are significant (Padjusted < 0.05) after FDR correction between patients that survived longer and those that survived shorter. E, Individual chemokine or cytokine values (pg/mL) compared among different response groups, only significant differences from D are included. Statistical tests in bar charts were unpaired two-tailed t tests (C), assuming unequal variance, or paired t tests (D) with *, P < 0.05; **, P < 0.01; ***, P < 0.001.
FIGURE 5
FIGURE 5
Kaplan–Meier analysis showing. PFS (A) and OS (B), respectively using pretreatment plasma CCL21 (pg/mL) measurements based on their median values. C, Correlation between CCL21 (pg/mL) and CD3+CCR7+CD45RA+ (% counts) matched patient samples (n = 23). D, Flow cytometry–based comparison between short- and long-term survivors for CD3+CCR7+CD45RA+ % (T naïve and T stem cell memory) analysis using pretreatment blood samples (n = 24). Statistical test was unpaired two-tailed t tests, assuming equal variance. E, Kaplan–Meier analysis showing OS using median CD3+CCR7+CD45RA+ % values.
FIGURE 6
FIGURE 6
IHC magenta showing low mag of CD20 varied staining of TLS-like (borderline-tertiary lymphoid structures) followed by high magnification images of CD20, CD3, CCL21 within serial sections for Pt 2-027 (A), 3-010 (B). Also see Supplementary Fig. S5. Low and high magnification images of CD20+ TLS-like staining for Pt. 2-026 (C) and no TLS sample Pt. 4-017 (D). Kaplan–Meier analysis showing PFS (E) and OS (F), respectively dividing patient population into two groups based on IHC, no-TLS (n = 4) and TLS-like (n = 18). Also see Supplementary Fig. S5. G, Heatmap showing top 10% genes with highest log2 fold change, among all positively and negatively significantly regulated genes comparing no-TLS and TLS-like groups using bulk RNA-seq (log2 Reads Per Kilobase per Million mapped reads [RPKM] normalized values). Arrows represent relevant TLS-based gene signatures. Genes with FDR-adjusted P values <0.05 were considered statistically significant. H, Individual box plots showing CCL19, CCL21, CCR7, and CD79A changes in expression between the groups. Statistical tests were carried out using DESeq2 and FDR-adjusted P values were denoted with *, P < 0.05; **, P < 0.01; ***, P < 0.001.

References

    1. Carlino MS, Larkin J, Long GV. Immune checkpoint inhibitors in melanoma. Lancet 2021;398:1002–14.
    1. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. . Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med 2015;373:1803–13.
    1. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. . Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372:2018–28.
    1. Kato K, Cho BC, Takahashi M, Okada M, Lin C-Y, Chin K, et al. . Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019;20:1506–17.
    1. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. . Five-year outcomes with pembrolizumab versus chemotherapy for metastatic non-small-cell lung cancer with PD-L1 tumor proportion score ≥ 50. J Clin Oncol 2021;39:2339–49.
    1. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. . Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med 2016;375:1823–33.
    1. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, et al. . Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013,369:122–33.
    1. Wang DY, Salem J-E, Cohen JV, Chandra S, Menzer C, Ye F, et al. . Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncol 2018;4:1721–8.
    1. Puzanov I, Diab A, Abdallah K, Bingham CO, Brogdon C, Dadu R, et al. . Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the society for immunotherapy of cancer (SITC) toxicity management working group. J Immunother Cancer 2017;5:95.
    1. Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, et al. . First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet 2021;398:27–40.
    1. Dyer BA, Feng CH, Eskander R, Sharabi AB, Mell LK, McHale M, et al. . Current status of clinical trials for cervical and uterine cancer using immunotherapy combined with radiation. Int J Radiat Oncol Biol Phys 2021;109:396–412.
    1. Kjeldsen JW, Lorentzen CL, Martinenaite E, Ellebaek E, Donia M, Holmstroem RB, et al. . A phase 1/2 trial of an immune-modulatory vaccine against IDO/PD-L1 in combination with nivolumab in metastatic melanoma. Nat Med 2021;27:2212–23.
    1. Dixon-Douglas JR, Patel RP, Somasundram PM, McArthur GA. Triplet therapy in melanoma – combined BRAF/MEK inhibitors and anti-PD-(L)1 antibodies. Curr Oncol Rep 2022;24:1071–9.
    1. Adusumilli PS, Zauderer MG, Rivière I, Solomon SB, Rusch VW, O'Cearbhaill RE, et al. . A phase i trial of regional mesothelin-targeted CAR T-cell therapy in patients with malignant pleural disease, in combination with the anti-PD-1 agent pembrolizumab. Cancer Discov 2021;11:2748–63.
    1. Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin J-M, Morrison RM, et al. . Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science 2021;371:595–602.
    1. Olson DJ, Eroglu Z, Brockstein B, Poklepovic AS, Bajaj M, Babu S, et al. . Pembrolizumab plus ipilimumab following anti-PD-1/L1 failure in melanoma. J Clin Oncol 2021;39:2647–55.
    1. Jager MJ, Shields CL, Cebulla CM, Abdel-Rahman MH, Grossniklaus HE, Stern MH, et al. . Uveal melanoma. Nat Rev Dis Primers 2020;6:24.
    1. Chattopadhyay C, Kim DW, Gombos DS, Oba J, Qin Y, Williams MD, et al. . Uveal melanoma: from diagnosis to treatment and the science in between. Cancer 2016;122:2299–312.
    1. Khoja L, Atenafu EG, Suciu S, Leyvraz S, Sato T, Marshall E, et al. . Meta-analysis in metastatic uveal melanoma to determine progression free and overall survival benchmarks: an international rare cancers initiative (IRCI) ocular melanoma study. Ann Oncol 2019;30:1370–80.
    1. Nathan P, Hassel JC, Rutkowski P, Baurain J-F, Butler MO, Schlaak M, et al. . Overall survival benefit with tebentafusp in metastatic uveal melanoma. N Engl J Med 2021;385:1196–206.
    1. Bagge RO, Nelson A, Shafazand A, All-Ericsson C, Cahlin C, Elander N, et al. . Isolated hepatic perfusion as a treatment for uveal melanoma liver metastases, first results from a phase III randomized controlled multicenter trial (the SCANDIUM trial). J Clin Oncol 2023. [Online ahead of print].
    1. Piulats JM, Espinosa E, de la Cruz Merino L, Varela M, Carrion LA, Martin-Algarra S, et al. . Nivolumab plus ipilimumab for treatment-naive metastatic uveal melanoma: an open-label, multicenter, phase II trial by the spanish multidisciplinary melanoma group (GEM-1402). J Clin Oncol 2021;39:586–98.
    1. Pelster MS, Gruschkus SK, Bassett R, Gombos DS, Shephard M, Posada L, et al. . Nivolumab and ipilimumab in metastatic uveal melanoma: results from a single-arm phase II study. J Clin Oncol 2021;39:599–607.
    1. Ny L, Jespersen H, Karlsson J, Alsen S, Filges S, All-Eriksson C, et al. . The PEMDAC phase 2 study of pembrolizumab and entinostat in patients with metastatic uveal melanoma. Nat Commun 2021;12:5155.
    1. Sah VR, Karlsson J, Jespersen H, Lindberg MF, Nilsson LM, Ny L, et al. . Epigenetic therapy to enhance therapeutic effects of PD-1 inhibition in therapy-resistant melanoma. Melanoma Res 2022;32:241–8.
    1. Jespersen H, Bagge RO, Ullenhag G, Carneiro A, Helgadottir H, Ljuslinder I, et al. . Concomitant use of pembrolizumab and entinostat in adult patients with metastatic uveal melanoma (PEMDAC study): protocol for a multicenter phase II open label study. BMC Cancer 2019;19:415.
    1. Schwartz JL, Tamura Y, Jordan R, Grierson JR, Krohn KA. Monitoring tumor cell proliferation by targeting DNA synthetic processes with thymidine and thymidine analogs. J Nucl Med 2003;44:2027–32.
    1. 1000 Genomes Project Consortium; Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, et al. . A global reference for human genetic variation. Nature 2015;526:68–74.
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. . STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21.
    1. Anders S, Pyl PT, Huber W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 2015;31:166–9.
    1. Bhadury J, Nilsson LM, Muralidharan SV, Green LC, Li Z, Gesner EM, et al. . BET and HDAC inhibitors induce similar genes and biological effects and synergize to kill in Myc-induced murine lymphoma. Proc Natl Acad Sci U S A 2014;111:E2721–30.
    1. Zuo S, Wang H, Li L, Pan H, Lu L. Thymidine kinase 1 drives skin cutaneous melanoma malignant progression and metabolic reprogramming. Front Oncol 2022;12:802807.
    1. Bitter EE, Townsend MH, Erickson R, Allen C, O'Neill KL. Thymidine kinase 1 through the ages: a comprehensive review. Cell Biosci 2020;10:138.
    1. Paoletti C, Barlow WE, Cobain EF, Bergqvist M, Mehta RS, Gralow JR, et al. . Evaluating serum thymidine kinase 1 in patients with hormone receptor-positive metastatic breast cancer receiving first-line endocrine therapy in the SWOG S0226 trial. Clin Cancer Res 2021;27:6115–23.
    1. Matikas A, Wang K, Lagoudaki E, Acs B, Zerdes I, Hartman J, et al. . Prognostic role of serum thymidine kinase 1 kinetics during neoadjuvant chemotherapy for early breast cancer. ESMO Open 2021;6:100076.
    1. Nisman B, Allweis T, Kaduri L, Maly B, Gronowitz S, Hamburger T, et al. . Serum thymidine kinase 1 activity in breast cancer. Cancer Biomark 2010;7:65–72.
    1. Svedman FC, Jalsenius M, Höiom V, Grozman V, Bergqvist M, Söderdahl F, et al. . Plasma thymidine kinase activity as a novel biomarker in metastatic melanoma patients treated with immune checkpoint inhibitors. Cancers 2022;14:702.
    1. Okugawa Y, Toiyama Y, Mohri Y, Tanaka K, Kawamura M, Hiro J, et al. . Elevated serum concentration of monocyte chemotactic protein 4 (MCP-4) as a novel non-invasive prognostic and predictive biomarker for detection of metastasis in colorectal cancer. J Surg Oncol 2016;114:483–9.
    1. Shields JD, Kourtis IC, Tomei AA, Roberts JM, Swartz MA. Induction of lymphoidlike stroma and immune escape by tumors that express the chemokine CCL21. Science 2010;328:749–52.
    1. Briukhovetska D, Dörr J, Endres S, Libby P, Dinarello CA, Kobold S. Interleukins in cancer: from biology to therapy. Nat Rev Cancer 2021;21:481–99.
    1. Dai S, Zeng H, Liu Z, Jin K, Jiang W, Wang Z, et al. . Intratumoral CXCL13+CD8+T cell infiltration determines poor clinical outcomes and immunoevasive contexture in patients with clear cell renal cell carcinoma. J Immunother Cancer 2021;9:e001823.
    1. Förster R, Davalos-Misslitz AC, Rot A. CCR7 and its ligands: balancing immunity and tolerance. Nat Rev Immunol 2008;8:362–71.
    1. Farnsworth RH, Karnezis T, Maciburko SJ, Mueller SN, Stacker SA. The interplay between lymphatic vessels and chemokines. Front Immunol 2019;10:518.
    1. Takeuchi H, Fujimoto A, Tanaka M, Yamano T, Hsueh E, Hoon DSB. CCL21 chemokine regulates chemokine receptor CCR7 bearing malignant melanoma cells. Clin Cancer Res 2004;10:2351–8.
    1. Yu P, Lee Y, Liu W, Chin RK, Wang J, Wang Y, et al. . Priming of naive T cells inside tumors leads to eradication of established tumors. Nat Immunol 2004;5:141–9.
    1. Thanarajasingam U, Sanz L, Diaz R, Qiao J, Sanchez-Perez L, Kottke T, et al. . Delivery of CCL21 to metastatic disease improves the efficacy of adoptive T-cell therapy. Cancer Res 2007;67:300–8.
    1. Fankhauser M, Broggi MAS, Potin L, Bordry N, Jeanbart L, Lund AW, et al. . Tumor lymphangiogenesis promotes T cell infiltration and potentiates immunotherapy in melanoma. Sci Transl Med 2017;9:eaal4712.
    1. Xiong Y, Liu L, Wang J, Xi W, Xia Y, Bai Q, et al. . Low CCL-21 expression associates with unfavorable postoperative prognosis of patients with metastatic renal cell carcinoma. Oncotarget 2017;8:25650–9.
    1. Girard J-P, Moussion C, Förster R. HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nat Rev Immunol 2012;12:762–73.
    1. Golubovskaya V, Wu L. Different subsets of T cells, memory, effector functions, and CAR-T immunotherapy. Cancers 2016;8:36.
    1. Tang H, Zhu M, Qiao J, Fu Y-X. Lymphotoxin signalling in tertiary lymphoid structures and immunotherapy. Cell Mol Immunol 2017;14:809–18.
    1. Chelvanambi M, Fecek RJ, Taylor JL, Storkus WJ. STING agonist-based treatment promotes vascular normalization and tertiary lymphoid structure formation in the therapeutic melanoma microenvironment. J Immunother Cancer 2021;9:e001906.
    1. Sautès-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer 2019;19:307–25.
    1. Cabrita R, Lauss M, Sanna A, Donia M, Larsen MS, Mitra S, et al. . Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020;577:561–5.
    1. Rodrigues M, Mobuchon L, Houy A, Fievet A, Gardrat S, Barnhill RL, et al. . Outlier response to anti-PD1 in uveal melanoma reveals germline MBD4 mutations in hypermutated tumors. Nat Commun 2018;9:1866.
    1. Johansson PA, Stark A, Palmer JM, Bigby K, Brooks K, Rolfe O, et al. . Prolonged stable disease in a uveal melanoma patient with germline MBD4 nonsense mutation treated with pembrolizumab and ipilimumab. Immunogenetics 2019;71:433–6.
    1. Johansson PA, Brooks K, Newell F, Palmer JM, Wilmott JS, Pritchard AL, et al. . Whole genome landscapes of uveal melanoma show an ultraviolet radiation signature in iris tumours. Nat Commun 2020;11:2408.
    1. Karlsson J, Nilsson LM, Mitra S, Alsen S, Shelke GV, Sah VR, et al. . Molecular profiling of driver events in metastatic uveal melanoma. Nat Commun 2020;11:1894.
    1. Svedman FC, Jalsenius M, Grozman V, Bergqvist M, Söderdahl F, Rotstein S, et al. . Dynamics of plasma thymidine kinase activity in metastatic melanoma reflects immune checkpoint inhibitor efficacy. Acta Oncol 2022;61:1116–20.
    1. Turnquist HR, Lin X, Ashour AE, Hollingsworth MA, Singh RK, Talmadge JE, et al. . CCL21 induces extensive intratumoral immune cell infiltration and specific anti-tumor cellular immunity. Int J Oncol 2007;30:631–9.
    1. Delvecchio FR, Fincham REA, Spear S, Clear A, Roy-Luzarraga M, Balkwill FR, et al. . Pancreatic cancer chemotherapy is potentiated by induction of tertiary lymphoid structures in mice. Cell Mol Gastroenterol Hepatol 2021;12:1543–65.
    1. Pylayeva-Gupta Y, Das S, Handler JS, Hajdu CH, Coffre M, Koralov SB, et al. . IL35-producing B cells promote the development of pancreatic neoplasia. Cancer Discov 2016;6:247–55.

Source: PubMed

3
Abonner