ZEB1 transcription factor promotes immune escape in melanoma

Maud Plaschka, Valentin Benboubker, Maxime Grimont, Justine Berthet, Laurie Tonon, Jonathan Lopez, Myrtille Le-Bouar, Brigitte Balme, Garance Tondeur, Arnaud de la Fouchardière, Lionel Larue, Alain Puisieux, Yenkel Grinberg-Bleyer, Nathalie Bendriss-Vermare, Bertrand Dubois, Christophe Caux, Stéphane Dalle, Julie Caramel, Maud Plaschka, Valentin Benboubker, Maxime Grimont, Justine Berthet, Laurie Tonon, Jonathan Lopez, Myrtille Le-Bouar, Brigitte Balme, Garance Tondeur, Arnaud de la Fouchardière, Lionel Larue, Alain Puisieux, Yenkel Grinberg-Bleyer, Nathalie Bendriss-Vermare, Bertrand Dubois, Christophe Caux, Stéphane Dalle, Julie Caramel

Abstract

Background: The efficacy of immunotherapies in metastatic melanoma depends on a robust T cell infiltration. Oncogenic alterations of tumor cells have been associated to T cell exclusion. Identifying novel cancer cell-intrinsic non-genetic mechanisms of immune escape, the targeting of which would reinstate T cell recruitment, would allow to restore the response to anti-programmed cell death protein 1 (PD-1) antibody therapy. The epithelial-to-mesenchymal transition (EMT)-inducing transcription factor ZEB1 is a major regulator of melanoma cell plasticity, driving resistance to mitogen-activated protein kinase (MAPK) targeted therapies. We thus wondered whether ZEB1 signaling in melanoma cells may promote immune evasion and resistance to immunotherapy.

Methods: We evaluated the putative correlation between ZEB1 expression in melanoma cells and the composition of the immune infiltrate in a cohort of 60 human melanoma samples by combining transcriptomic (RNA-sequencing) and seven-color spatial multi-immunofluorescence analyses. Algorithm-based spatial reconstitution of tumors allowed the quantification of CD8+, CD4+ T cells number and their activation state (PD-1, Ki67). ZEB1 gain-of-function or loss-of-function approaches were then implemented in syngeneic melanoma mouse models, followed by monitoring of tumor growth, quantification of immune cell populations frequency and function by flow cytometry, cytokines secretion by multiplex analyses. Chromatin-immunoprecipitation was used to demonstrate the direct binding of this transcription factor on the promoters of cytokine-encoding genes. Finally, the sensitivity to anti-PD-1 antibody therapy upon ZEB1 gain-of-function or loss-of-function was evaluated.

Results: Combined spatial and transcriptomic analyses of the immune infiltrates in human melanoma samples demonstrated that ZEB1 expression in melanoma cells is associated with decreased CD8+ T cell infiltration, independently of β-catenin pathway activation. ZEB1 ectopic expression in melanoma cells impairs CD8+ T cell recruitment in syngeneic mouse models, resulting in tumor immune evasion and resistance to immune checkpoint blockade. Mechanistically, we demonstrate that ZEB1 directly represses the secretion of T cell-attracting chemokines, including CXCL10. Finally, Zeb1 knock-out, by promoting CD8+ T cell infiltration, synergizes with anti-PD-1 antibody therapy in promoting tumor regression.

Conclusions: We identify the ZEB1 transcription factor as a key determinant of melanoma immune escape, highlighting a previously unknown therapeutic target to increase efficacy of immunotherapy in melanoma.

Trial registration number: NCT02828202.

Keywords: immunotherapy; lymphocytes; melanoma; tumor escape; tumor microenvironment; tumor-infiltrating.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
High ZEB1 expression in tumor cells is associated with decreased CD8+ T cell infiltration in a cohort of human melanoma samples. (A) Schematic diagram of the RNA-seq and spatial multi-immunofluorescence (multi-IF) analyses of human cutaneous melanoma samples. (B) Representative pictures of multi-immunofluorescence opal staining for ZEB1 (red), SOX10 (white), CD8 (green), CD4 (orange), Ki67 (magenta) and PD-1 (yellow). Blue: DAPI. Scale bar=100 µm. ZEB1-expressing CD8, CD4, and stromal cells are indicated with green, orange and red arrows, respectively. (C) Schematic representation of digital image analyses pipeline and tumor reconstruction. The entire tumor sections were selected for the analysis. Each cell was detected based on the nucleus and associated with a phenotype based on the expression of SOX10 (melanoma cell), CD8 (CD8+ T cells), and CD4 (CD4+ T cells). For each cell, both the location and immunofluorescence intensities of each markers were stored in a so-called phenotype matrix. Tumors were then reconstructed, and quantification was performed using the R software. (D) Whole tumor regions were spatially reconstituted using the R software. Melanoma cells are represented using a color gradient (green, orange, red, and black) corresponding to ZEB1 nuclear intensity in melanoma cells (respectively <1; (1;3); (3;10); >10). Spatial reconstitution of CD8 localization is represented with blue dots. The black insets correspond to representative pictures of multi-IF opal staining for ZEB1 (red), CD8 (green), and SOX10 (white) of a ZEB1 low/CD8 infiltrated (upper panel) and a ZEB1 high/CD8 excluded (lower panel) tumor. Blue: DAPI. Note that in the ZEB1 low tumor, ZEB1 high cells are not melanoma SOX10-positive cells. Scale bars=100 µm. (E) Stacked bar representing the percentage of CD8 excluded (light blue) and CD8 infiltrated (dark blue) tumors within ZEB1 low and ZEB1 high tumors. (F) Percentage of CD8 T cells as quantified by IF in ZEB1 low (green: n=28) and ZEB1 high (red: n=10) tumors (mean with SD, Mann-Whitney test).
Figure 2
Figure 2
ZEB1-mediated T cell recruitment defect is independent of β-catenin pathway activation. (A) β-catenin ssGSEA score calculated from the RNA-seq data, using the previously described score from Spranger et al based on seven genes: EFNB3, APC2, TCF1, C-MYC, TCF12, VEGFA and CTNNB1. Tumors were classified as ZEB1 low (n=38) and ZEB1 high (n=12) following the IF analysis (mean with SD, Mann-Whitney test). (B) Single cell RNA-seq from Jerby-Arnon et al. X-axis: β-catenin single-sample gene set enrichment analysis (ssGSEA) score calculated on melanoma cells. Y-axis: ZEB1 expression (TPM) in melanoma cells. R²=0.0005748, p value=0.2817. (C) Pie charts representing the proportion of nuclear β-catenin (dark orange) and cytoplasmic β-catenin (light orange) tumors within ZEB1high melanoma (n=12). Representative pictures of cytoplasmic (on the left) and nuclear (on the right) β-catenin staining (in red). DAPI in blue. Scale bars=100 µm and 200 µm, respectively. (D) Representative picture of cytoplasmic β-catenin staining in a ZEB1high tumor: β-catenin (red), ZEB1 (yellow) and SOX10 (cyan). DAPI in blue. Scale bar=50 µm. RNA-seq, RNA-sequencing.
Figure 3
Figure 3
ZEB1 ectopic expression in melanoma cells increases tumor growth in immunocompetent mice. (A) Br16M3 and NR6.1 murine melanoma cells were infected with retroviruses expressing ZEB1. Western blot analyses of ZEB1. GAPDH was used as a loading control. (B) 3×106 Br16M3 (left) or NR6.1 (right) control (green) or ZEB1-overexpressing (red) cells were injected subcutaneously into C57BL6 immunocompetent mice (upper panel) or into immunodeficient RAG2 KO mice (lower panel). The mean tumor volume is represented (±SEM, Mann-Whitney test). Br16M3: n=17–19 and n=4 for immunocompetent and RAG2 knock-out (KO) mice respectively; NR6.1: n=5 for both immunocompetent and RAG2 KO mice.
Figure 4
Figure 4
ZEB1 ectopic expression in melanoma cells impairs the recruitment of CD8+ T lymphocytes. (A) FACS analyses of CD45 infiltration 2 weeks after injection into immunocompetent C57BL6 mice of Br16M3 (left) or NR6.1 (right) models: control (blue); ZEB1 ectopic expression (red), n=4–5 per group. Bar chart representing the number of CD45+ cells/g of tumor (mean with SD, Mann-Whitney test). (B and C) FACS analyses of CD8+ and CD4+ infiltration in Br16M3 and NR6.1 control and ZEB1-overexpressing tumors. Bar charts representing the number of CD8+ (B) or CD4+ (C) cells per gram of tumor. (D) Representative pictures of H&E coloration, ZEB1 or CD8 immunostaining in Br16M3 (left) and NR6.1 (right) control and ZEB1-overexpressing tumors collected 2 weeks after injection into C57BL6 mice. Purple chromogen was used for IHC visualization in pigmented NR6.1 tumors instead of brown DAB for non-pigmented Br16M3 tumors. Scale bars=50 µm. Arrows indicate purple positive CD8+ T cells. (E) Quantification of CD8+ T cell infiltration in Br16M3 and NR6.1 control versus ZEB1-overexpressing tumors. Bar chart representing the number of CD8+ T cells per mm² of tumor (n=5–6, mean with SD, Mann-Whitney test). (F) FACS analyses of FOXP3+ CD4+ regulatory T cells (Treg) infiltration in Br16M3 and NR6.1 control and ZEB1-overexpressing tumors. Bar charts representing the percentage of Tregs among CD4+ T cells in Br16M3 control (blue) and ZEB1-overexpressing (red) tumors, 14 (left) and 21 (right) days after injection (mean with SD, Mann-Whitney test). (G) Bar chart representing the ratio between the number of CD8+ T cells and the number of Tregs in Br16M3 control and ZEB1-overexpressing tumors (mean with SD, Mann-Whitney test).
Figure 5
Figure 5
Melanoma cell-intrinsic ZEB1 signaling decreases the production of T cell-attracting chemokines. (A) Heatmap representing the relative quantity of 35 analytes measured using the Meso Scale Diagnostics (MSD) technology in supernatants of dilacerated control (n=5) or ZEB1-overexpressing (n=8) Br16M3 tumors. TGF-β isoforms were also quantified with the same technology in n=6 and n=8 control or ZEB1-overexpressing tumors, respectively. Mann-Whitney test. (B) Bar charts representing the concentration of CXCL10, CCL4, TGF-β1 and 2 (pg/mL), in the supernatant of dilacerated Br16M3 control (n=5 or 6) and ZEB1-overexpressing (n=8) tumors (mean with SD, Mann-Whitney test). (C) Chromatin immunoprecipitation assay of ZEB1 showing binding to the promoter of Cxcl10 and Ccl4 in Br16M3 cells. The fraction of chromatin bound to the promoter, with IgG control or anti-ZEB1 antibody is represented as a percentage of input (n>=3) (mean with SD, t-test). (D) NR6.1 control and ZEB1-overexpressing murine melanoma cells were infected with control or Cxcl10-expressing lentiviruses. qPCR analyses of CXCL10 mRNA expression in NR6.1 control (CT), ZEB1 control (Z1 CT) and ZEB1-overexpressing CXCL10 (Z1 CXCL10) (n=1). (E) 2.5×106 NR6.1 ZEB1 control (Z1 CT) and ZEB1 CXCL10 (Z1 CXCL10) cells were injected subcutaneously into C57BL6 immunocompetent mice (n=5 per group). The mean tumor volume is represented (±SEM, Mann-Whitney test). (F) FACS analyses of CD45 and CD8 infiltration 17 days after injection into immunocompetent C57BL6 mice of NR6.1 ZEB1 control (n= 4) and ZEB1 CXCL10 (n= 5). Bar chart representing the number of CD45+ cells and CD8+ T cells/g of tumor (mean with SD, Mann-Whitney test). (G) Quantification of CD8+ T cell infiltration in NR6.1 ZEB1 control and ZEB1 CXCL10 tumors. Bar chart representing the number of CD8+ T cells per mm² of tumor (n=5 per group, mean with SD, Mann-Whitney test).
Figure 6
Figure 6
Zeb1 knock-out in melanoma cells strongly reduces tumor growth and favors CD8+ T cell infiltration. (A) Br42M6 murine melanoma cells were infected with lentiviruses expressing the Cas9 and guide RNA (sgRNA) targeting ZEB1 or scramble sgRNA. 42C1 is a scramble clone, and 42Z6 is a Zeb1 knock-out clone. Western blot analyses of ZEB1. GAPDH was used as a loading control. (B) 2.5×106 42C1 (orange) or 42Z6 (blue) cells were injected subcutaneously into C57BL6 immunocompetent mice (left, n=6–10 per group) or into RAG KO mice (right, n=3 per group). The mean tumor volume is represented (±SD, Mann-Whitney test). (C) Representative pictures of ZEB1 and CD8 immunostaining (brown, DAB) in 42C1 (scramble) and 42Z6 (ZEB1 KO) tumors. Scale bars=50 µm. Quantification of CD8 infiltration in 42C1 (scramble) and 42Z6 (ZEB1 KO) tumors. Bar chart representing the number of CD8+ T cell per mm² of tumor (mean with SD, Mann-Whitney test).
Figure 7
Figure 7
ZEB1 overexpression in melanoma cells triggers resistance while knock-out of Zeb1 improves the response to immune checkpoint blockade. (A) NR6.1 murine melanoma cells were infected with retroviruses expressing ZEB1. 2×106 NR6.1 control (red) or ZEB1-overexpressing (blue) cells were injected subcutaneously into C57BL6 immunocompetent mice (five per group) and treated with either anti-PD-1 (dashed lines) or control isotype (iso, solid lines) 5, 7, and 9 days after injection. The mean tumor volume for 5 mice is represented (±SD, Student’s t-test). (B) NR6.1 murine melanoma Kaplan-Meier survival curves with log-rank test, mice were euthanized when tumor volume reached 1500mm3 or diameter >15mm. (C) 2×106 control 25C19 (orange) or Zeb1 KO 25Z19 (blue) cells were injected subcutaneously into C57BL6 immunocompetent mice (five per group) and treated with either anti-PD-1 (dashed lines) or control isotype (iso, solid lines) 5, 7, and 9 days after injection. The mean tumor volume for 5 mice is represented (±SD, Student’s t-test). (D) Br25F4 murine melanoma Kaplan-Meier survival curves with log-rank test, mice were euthanized when tumor volume reached 1500 mm3 or diameter >15mm. (*P<0.05, **p<0.01, ***p<0.001, ****p<0.0001).

References

    1. Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. . Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med Overseas Ed 2017;377:1345–56. 10.1056/NEJMoa1709684
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. . Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. New England Journal of Medicine 2019;381:1535–46. 10.1056/NEJMoa1910836
    1. Hugo W, Shi H, Sun L, et al. . Non-genomic and immune evolution of melanoma acquiring MAPKi resistance. Cell 2015;162:1271–85. 10.1016/j.cell.2015.07.061
    1. Arozarena I, Wellbrock C. Phenotype plasticity as enabler of melanoma progression and therapy resistance. Nat Rev Cancer 2019;19:377–91. 10.1038/s41568-019-0154-4
    1. Rambow F, Marine J-C, Goding CR. Melanoma plasticity and phenotypic diversity: therapeutic barriers and opportunities. Genes Dev 2019;33:1295–318. 10.1101/gad.329771.119
    1. Sharma P, Hu-Lieskovan S, Wargo JA, et al. . Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168:707–23. 10.1016/j.cell.2017.01.017
    1. Abril-Rodriguez G, Torrejon DY, Liu W, et al. . Pak4 inhibition improves PD-1 blockade immunotherapy. Nature Cancer 2020;1:46–58. 10.1038/s43018-019-0003-0
    1. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015;523:231–5. 10.1038/nature14404
    1. Peng W, Chen JQ, Liu C, et al. . Loss of PTEN promotes resistance to T cell–mediated immunotherapy. Cancer Discov 2016;6:202–16. 10.1158/-15-0283
    1. Zingg D, Arenas-Ramirez N, Sahin D, et al. . The histone methyltransferase EZH2 controls mechanisms of adaptive resistance to tumor immunotherapy. Cell Rep 2017;20:854–67. 10.1016/j.celrep.2017.07.007
    1. Cerezo-Wallis D, Contreras-Alcalde M, Troulé K, et al. . Midkine rewires the melanoma microenvironment toward a tolerogenic and immune-resistant state. Nat Med 2020;26:1865–77. 10.1038/s41591-020-1073-3
    1. Rambow F, et al. . Toward minimal residual Disease-Directed therapy in melanoma toward minimal residual Disease-Directed therapy in melanoma. Cell 2018:1–13.
    1. Tirosh I, Izar B, Prakadan SM, et al. . Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 2016;352:189–96. 10.1126/science.aad0501
    1. Hoek KS, Goding CR. Cancer stem cells versus phenotype-switching in melanoma. Pigment Cell Melanoma Res 2010;23:746–59. 10.1111/j.1755-148X.2010.00757.x
    1. Verfaillie A, Imrichova H, Atak ZK, et al. . Decoding the regulatory landscape of melanoma reveals TEADS as regulators of the invasive cell state. Nat Commun 2015;6:1–16. 10.1038/ncomms7683
    1. Wouters J, Kalender-Atak Z, Minnoye L, et al. . Robust gene expression programs underlie recurrent cell states and phenotype switching in melanoma. Nat Cell Biol 2020;22:986–98. 10.1038/s41556-020-0547-3
    1. Goding CR, Arnheiter H. MITF — the first 25 years 2019:1–25.
    1. Puisieux A, Brabletz T, Caramel J. Oncogenic roles of EMT-inducing transcription factors. Nat Cell Biol 2014;16:488–94. 10.1038/ncb2976
    1. Caramel J, Ligier M, Puisieux A. Pleiotropic roles for ZEB1 in cancer. Cancer Res 2018;78:30–5. 10.1158/0008-5472.CAN-17-2476
    1. Caramel J, Papadogeorgakis E, Hill L, et al. . A switch in the expression of embryonic EMT-Inducers drives the development of malignant melanoma. Cancer Cell 2013;24:466–80. 10.1016/j.ccr.2013.08.018
    1. Richard G, Dalle S, Monet M-A, et al. . ZEB1-mediated melanoma cell plasticity enhances resistance to MAPK inhibitors. EMBO Mol Med 2016;8:1143–61. 10.15252/emmm.201505971
    1. Terry S, Savagner P, Ortiz-Cuaran S, et al. . New insights into the role of EMT in tumor immune escape. Mol Oncol 2017;11:824–46. 10.1002/1878-0261.12093
    1. Dongre A, Rashidian M, Reinhardt F, et al. . Epithelial-To-Mesenchymal transition contributes to immunosuppression in breast carcinomas. Cancer Res 2017;77:3982–9. 10.1158/0008-5472.CAN-16-3292
    1. Tang Y, Durand S, Dalle S, et al. . EMT-Inducing transcription factors, drivers of melanoma phenotype switching, and resistance to treatment. Cancers 2020;12:2154. 10.3390/cancers12082154
    1. Bruneel K, Verstappe J, Vandamme N, et al. . Intrinsic balance between ZEB family members is important for melanocyte homeostasis and melanoma progression. Cancers 2020;12:2248–25. 10.3390/cancers12082248
    1. Jerby-Arnon L, Shah P, Cuoco MS, et al. . A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 2018;175:984–97. 10.1016/j.cell.2018.09.006
    1. Dhomen N, Reis-Filho JS, da Rocha Dias S, et al. . Oncogenic BRAF induces melanocyte senescence and melanoma in mice. Cancer Cell 2009;15:294–303. 10.1016/j.ccr.2009.02.022
    1. Ackermann J, Frutschi M, Kaloulis K, et al. . Metastasizing melanoma formation caused by expression of activated N-Ras Q61K on an INK4a-deficient background. Cancer Res 2005;65:4005–11. 10.1158/0008-5472.CAN-04-2970
    1. Scott CL, Omilusik KD. ZEBs: novel players in immune cell development and function. Trends Immunol 2019;40:431–46. 10.1016/j.it.2019.03.001
    1. Spranger S, Gajewski TF. Tumor-Intrinsic oncogene pathways mediating immune avoidance. Oncoimmunology 2016;5:e1086862. 10.1080/2162402X.2015.1086862
    1. El Kharbili M, Agaësse G, Barbollat-Boutrand L, et al. . Tspan8-β-catenin positive feedback loop promotes melanoma invasion. Oncogene 2019;38:3781–93. 10.1038/s41388-019-0691-z
    1. Cooper ZA, Juneja VR, Sage PT, et al. . Response to BRAF inhibition in melanoma is enhanced when combined with immune checkpoint blockade. Cancer Immunol Res 2014;2:643–54. 10.1158/2326-6066.CIR-13-0215
    1. Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol 2017;17:559–72. 10.1038/nri.2017.49
    1. Meeth K, Wang JX, Micevic G, et al. . The YUMM lines: a series of congenic mouse melanoma cell lines with defined genetic alterations. Pigment Cell Melanoma Res 2016;29:590–7. 10.1111/pcmr.12498
    1. Kalbasi A, Ribas A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat Rev Immunol 2020;20:25–39. 10.1038/s41577-019-0218-4
    1. Katsura A, Tamura Y, Hokari S, et al. . ZEB1-regulated inflammatory phenotype in breast cancer cells. Mol Oncol 2017;11:1241–62. 10.1002/1878-0261.12098
    1. Dangaj D, Bruand M, Grimm AJ, et al. . Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors. Cancer Cell 2019;35:885–900. 10.1016/j.ccell.2019.05.004
    1. Spranger S, Dai D, Horton B, et al. . Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 2017;31:711–23. 10.1016/j.ccell.2017.04.003
    1. Mariathasan S, Turley SJ, Nickles D, et al. . TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018;554:544–8. 10.1038/nature25501
    1. Kudo-Saito C, Shirako H, Takeuchi T, et al. . Cancer metastasis is accelerated through immunosuppression during snail-induced EMT of cancer cells. Cancer Cell 2009;15:195–206. 10.1016/j.ccr.2009.01.023
    1. Landsberg J, Kohlmeyer J, Renn M, et al. . Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature 2012;490:412–6. 10.1038/nature11538
    1. Boshuizen J, Vredevoogd DW, Krijgsman O, et al. . Reversal of pre-existing NGFR-driven tumor and immune therapy resistance. Nat Commun 2020;11:1–13. 10.1038/s41467-020-17739-8
    1. Riesenberg S, Groetchen A, Siddaway R, et al. . MITF and c-Jun antagonism interconnects melanoma dedifferentiation with pro-inflammatory cytokine responsiveness and myeloid cell recruitment. Nat Commun 2015;6:8755. 10.1038/ncomms9755
    1. Shibue T, Weinberg RA. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat Rev Clin Oncol 2017;14:611–29. 10.1038/nrclinonc.2017.44
    1. Hugo W, Zaretsky JM, Sun L, et al. . Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 2016;165:35–44. 10.1016/j.cell.2016.02.065
    1. Liu D, Schilling B, Liu D, et al. . Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat Med 2019;25:1916–27. 10.1038/s41591-019-0654-5
    1. Fu R, Li Y, Jiang N, et al. . Inactivation of endothelial ZEB1 impedes tumor progression and sensitizes tumors to conventional therapies. Journal of Clinical Investigation 2020;130:1252–70. 10.1172/JCI131507
    1. Boshuizen J, Koopman LA, Krijgsman O, et al. . Cooperative targeting of melanoma heterogeneity with an Axl antibody-drug conjugate and BRAF/MEK inhibitors. Nat Med 2018;24:203–12. 10.1038/nm.4472
    1. Tsoi J, Robert L, Paraiso K, et al. . Multi-stage differentiation defines melanoma subtypes with differential vulnerability to drug-induced iron-dependent oxidative stress. Cancer Cell 2018;33:890–904. 10.1016/j.ccell.2018.03.017
    1. Viswanathan VS, Ryan MJ, Dhruv HD, et al. . Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 2017;547:453–7. 10.1038/nature23007
    1. Skrypek N, Goossens S, De Smedt E, et al. . Epithelial-to-mesenchymal transition: epigenetic reprogramming driving cellular plasticity. Trends in Genetics 2017;33:943–59. 10.1016/j.tig.2017.08.004
    1. Morel D, Jeffery D, Aspeslagh S, et al. . Combining epigenetic drugs with other therapies for solid tumours — past lessons and future promise. Nat Rev Clin Oncol 2020;17:91–107. 10.1038/s41571-019-0267-4
    1. Wang L, Leite de Oliveira R, Huijberts S, et al. . An acquired vulnerability of drug-resistant melanoma with therapeutic potential. Cell 2018;173:1413–25. 10.1016/j.cell.2018.04.012

Source: PubMed

3
订阅