Novel induction of CD40 expression by tumor cells with RAS/RAF/PI3K pathway inhibition augments response to checkpoint blockade

Chi Yan, Nabil Saleh, Jinming Yang, Caroline A Nebhan, Anna E Vilgelm, E Premkumar Reddy, Joseph T Roland, Douglas B Johnson, Sheau-Chiann Chen, Rebecca L Shattuck-Brandt, Gregory D Ayers, Ann Richmond, Chi Yan, Nabil Saleh, Jinming Yang, Caroline A Nebhan, Anna E Vilgelm, E Premkumar Reddy, Joseph T Roland, Douglas B Johnson, Sheau-Chiann Chen, Rebecca L Shattuck-Brandt, Gregory D Ayers, Ann Richmond

Abstract

Background: While immune checkpoint blockade (ICB) is the current first-line treatment for metastatic melanoma, it is effective for ~ 52% of patients and has dangerous side effects. The objective here was to identify the feasibility and mechanism of RAS/RAF/PI3K pathway inhibition in melanoma to sensitize tumors to ICB therapy.

Methods: Rigosertib (RGS) is a non-ATP-competitive small molecule RAS mimetic. RGS monotherapy or in combination therapy with ICB were investigated using immunocompetent mouse models of BRAFwt and BRAFmut melanoma and analyzed in reference to patient data.

Results: RGS treatment (300 mg/kg) was well tolerated in mice and resulted in ~ 50% inhibition of tumor growth as monotherapy and ~ 70% inhibition in combination with αPD1 + αCTLA4. RGS-induced tumor growth inhibition depends on CD40 upregulation in melanoma cells followed by immunogenic cell death, leading to enriched dendritic cells and activated T cells in the tumor microenvironment. The RGS-initiated tumor suppression was partially reversed by either knockdown of CD40 expression in melanoma cells or depletion of CD8+ cytotoxic T cells. Treatment with either dabrafenib and trametinib or with RGS, increased CD40+SOX10+ melanoma cells in the tumors of melanoma patients and patient-derived xenografts. High CD40 expression level correlates with beneficial T-cell responses and better survival in a TCGA dataset from melanoma patients. Expression of CD40 by melanoma cells is associated with therapeutic response to RAF/MEK inhibition and ICB.

Conclusions: Our data support the therapeutic use of RGS + αPD1 + αCTLA4 in RAS/RAF/PI3K pathway-activated melanomas and point to the need for clinical trials of RGS + ICB for melanoma patients who do not respond to ICB alone.

Trial registration: NCT01205815 (Sept 17, 2010).

Keywords: CD40; Immune checkpoint blockade; Immunogenic cell death; Melanoma; RAS/RAF/PI3K.

Conflict of interest statement

EPR is a member of the Board of Directors of Onconova, receives grant funding from Onconova, and is a paid consultant for Onconova. All other co-authors have declared that no additional conflict of interest exists.

Figures

Fig. 1
Fig. 1
Rigosertib induces melanoma cell death and inhibits melanoma tumor growth in vivo. a IC50 determined by CellTilter Blue assay at 72 h post RGS treatment. The melanoma and normal cells were seeded in 96-well plate with 3000 cells per well. b Dot plots and quantified results of the Annexin V+ and 7-AAD+ cell percentage in YUMM3.3 culture with or without RGS treatment. c Whole-cell extracts were harvested and immunoblotted. HSP90 was used as a loading control for densitometry quantification (red numbers). d, e Tumor volume and weight of YUMM3.3 melanomas in C57BL/6 female mice. Daily RGS treatment starts at day 10 post tumor cell inoculation. CR, complete regression. f Representative images and quantitative results of cleaved-caspase3 protein levels observed from day 17 post RGS treatment (300 mg/kg) of YUMM3.3 tumors by immunohistochemistry. g Gene ontology enrichment analysis of gene targets downregulated in YUMM3.3 tumors post 17 days of RGS treatment (300 mg/kg). Results for Fold Enrichment > 3 and FDR p < 0.05 are displayed. h Heatmap summarizes 21 p53-associated targets from a total of 1495 genes screened that were significantly downregulated due to RGS treatment. a-f pooled data obtained from at least two different experiments (n = 4 ~ 15) are shown. g, h were triplicates
Fig. 2
Fig. 2
Rigosertib increases the frequency of dendritic cells in tumors and tumor-draining lymph nodes (TDLN). YUMM3.3 tumors and TDLN were collected 17 days post-treatment. a Total number and density of CD45+ leukocytes in tumors were obtained by flow cytometry. b Gating strategy for dendritic cells in tumors and TDLN. c-e Frequency and mean fluorescent intensity (MFI) of dendritic cell subsets and their correlation with tumor burden. Pooled data obtained from at least two different experiments (n = 5 ~ 10) are shown
Fig. 3
Fig. 3
Rigosertib promotes T cell and NK cell responses but attenuates tumor-associated M2 macrophages in the tumor microenvironment. YUMM3.3 tumors were collected 17 days post-treatment. a, h Live CD45+ leukocytes were concatenated after downsampling to 20,000 events for subsequent high-dimensional data analysis to normalize the contribution among samples under different treatments. Samples were then analyzed in parallel by t-SNE and manually gated leukocyte populations were overlaid onto the total t-SNE map using FlowJo 10.5.3. b-e, g, i Flow cytometric and f IHC analysis of YUMM3.3 tumors at day 17 post-treatment. j Flow cytometric tSNE analysis of PBMC samples from YUMM3.3 tumor-bearing mice treated with RGS before and after CD4 and/or CD8 antibody depletion treatments. (K) Tumor weight (day 16 post RGS treatment) of YUMM3.3 tumors in C57BL/6 mice with and without CD4 and/or CD8 depletion. a-i pooled data obtained from at least two different experiments (n = 5 ~ 10) are shown. j, k were replicates (n = 10 per group)
Fig. 4
Fig. 4
Rigosertib improves T-cell responses to αPD-1/αCTLA-4 therapy. a Schematic of ICB non-responsive B16F10 melanoma model. b-d Survival of C57BL/6 female mice, day 11 tumor weight post treatment and tumor volume of B16F10 tumors. Mice reached end point and sacrificed when tumors in the experiment exceeded 15 mm in diameter or became perforated. The experiment was terminated when no mice survived in groups other than the RGS + ICB group. e Analysis of synergy. Interaction plot for tumor growth rate with 95% confidence interval by treatment. f Live CD45+ leukocytes were concatenated after downsampling to ~ 12,500 events for t-SNE analysis through flow cytometry and T-cell frequency was shown. g Tumor volume of YUMM3.3 tumors in C57BL/6 female mice. Treatment starts at day 10 post tumor cell inoculation. h Spider plots of tumor volume changes overtime for individual YUMM3.3 tumors. i IHC and j-l Flow cytometric analysis of YUMM3.3 tumors at day 17 post-treatment. Data were replicates from one experiment (n = 10 per group)
Fig. 5
Fig. 5
Rigosertib induces CD40 on melanoma cells and promotes the anti-tumor immunity. a, b Cytokine array of YUMM3.3 tumor lysate samples at day 17 post-treatment. c Live cells were concatenated after downsampling to ~ 20,000 events for t-SNE analysis through flow cytometry. d Flow cytometric analysis of YUMM3.3 tumors in ROSA reporter mice at day 14 post RGS treatment. e Mean fluorescence intensity (MFI) on the CD45−CD40+ cells isolated from day 17 YUMM3.3 tumors in C57BL/6 mice. f, g Melanoma cells were treated with indicated drugs for 48 h and CD40 expression was detected by flow cytometry. h, i Viability of YUMM3.3 and B16F10 cells was examined by flow cytometry. j Flow cytometric analysis of CD40 expression in response to treatment with IFNγ (500 U/ml, 48 h) in different clones of YUMM3.3 cells where shRNAs targeted coding sequence (CDS), 3″ untranslated region (UTR), or random sequences (Ctrl). k Tumor volume of CD40 knockdown clones in C57BL/6 mice was determined (n ≥ 4 mice per group). l Tumor weight and tumor volume of YUMM3.3 cells growing in C57BL/6-CD40 knockout male and female mice treated with RGS (300 mg/kg). a-e data were replicates from one experiment (n = 5 ~ 10 per group). f-l pooled data obtained from at least two different experiments (n = 3 ~ 6) are shown
Fig. 6
Fig. 6
RGS and BRAF/MEK inhibitors induce CD40 expression on melanoma cells and tumors of patients responsive to RAS/RAF/MEK pathway inhibition. a NRASmut melanoma PDX 1179 and PDX1214 tumor growth over time in NSG mice with IHC staining of cleaved-caspase3, CD40 and SOX10 in from mice treated with the vehicle or 300 mg/kg rigosertib (RGS). 20X and 40X images and the scale marker are shown. b IHC staining of CD40 and SOX10 in BRAFmut melanoma PDX tumors from mice treated with the vehicle or Dabrafenib + Trametinib (D + T). c Correlation analysis of T. Ratio and CD40 fold-change post treatment in PDXs shown in b. The T. Ratio, obtained from the statistical analysis of treatment difference comparisons of the tumor growth rate based on the tumor volume, for each PDX treatment comparison. d H&E and multiplex sequential IHC analysis of hematoxylin, CD40, CD80, CD11c, CD8 and SOX10 of patient melanoma sections. Cell number of SOX10+CD40+ cells was identified of 11 paired tumor sections pre- and post-BRAF inhibitor treatment (paired t-test). Pooled data (n = 4 ~ 11) from one experiment are shown
Fig. 7
Fig. 7
Correlations between CD40 expression in human melanoma and response to RAF inhibitors and ICB. a Correlations of CD40 gene expression with CD80 and ICOS-L expression from melanoma tumors in the TCGA dataset (n = 480) in cBioPortal. b Overall survival in reference to CD40, CD80 and ICOSL copy number and mutation burden in melanoma patients in TCGA-Melanoma dataset (n = 358). c Correlation matrix with Spearman’s rank correlation coefficients after data transformation analysis of CD40, CD4, CD8, IFNγ, GzmB, IL-2, CXCL9, IL-5 and IL-8 (y = (log(x-min(x) + 1))). d Drug sensitivity analysis of Barretina CellLine 2 (DNA) and Garnett CellLine (mRNA) human cancer cell line datasets in Oncomine database. e, f Analysis of gene expression levels and their correlation to ICB resistance. Gene expression in defined melanoma cells (n = 1883) were downloaded and analyzed based on single cell RNA-Seq (https://singlecell.broadinstitute.org/)

References

    1. Reddy BY, Miller DM, Tsao H. Somatic driver mutations in melanoma. Cancer. 2017;123(S11):2104–2117. doi: 10.1002/cncr.30593.
    1. Millis SZ, Ikeda S, Reddy S, Gatalica Z, Kurzrock R. Landscape of phosphatidylinositol-3-kinase pathway alterations across 19784 diverse solid tumors. JAMA Oncol. 2016;2(12):1565–1573. doi: 10.1001/jamaoncol.2016.0891.
    1. Long GV, Hauschild A, Santinami M, Atkinson V, Mandala M, Chiarion-Sileni V, Larkin J, Nyakas M, Dutriaux C, Haydon A, et al. Adjuvant dabrafenib plus trametinib in stage III BRAF-mutated melanoma. N Engl J Med. 2017;377(19):1813–1823. doi: 10.1056/NEJMoa1708539.
    1. Larkin J, Ascierto PA, Dreno B, Atkinson V, Liszkay G, Maio M, Mandala M, Demidov L, Stroyakovskiy D, Thomas L, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med. 2014;371(20):1867–1876. doi: 10.1056/NEJMoa1408868.
    1. Charles J, Martel C, de Fraipont F, Leccia MT, Robert C, Busser B. Mechanisms of resistance to anti-BRAF treatments. Ann Dermatol Venereol. 2014;141(11):671–681. doi: 10.1016/j.annder.2014.06.021.
    1. Hernandez-Davies JE, Tran TQ, Reid MA, Rosales KR, Lowman XH, Pan M, Moriceau G, Yang Y, Wu J, Lo RS, Kong M. Vemurafenib resistance reprograms melanoma cells towards glutamine dependence. J Transl Med. 2015;13(1):210. doi: 10.1186/s12967-015-0581-2.
    1. Manzano JL, Layos L, Buges C, de Los Llanos Gil M, Vila L, Martinez-Balibrea E, Martinez-Cardus A. Resistant mechanisms to BRAF inhibitors in melanoma. Ann Transl Med. 2016;4:237. doi: 10.21037/atm.2016.06.07.
    1. Wolchok J. How recent advances in immunotherapy are changing the standard of care for patients with metastatic melanoma. Ann Oncol. 2012;23(Suppl 8):viii15–viii21. doi: 10.1093/annonc/mds258.
    1. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–723. doi: 10.1056/NEJMoa1003466.
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, Cowey CL, Schadendorf D, Wagstaff J, Dummer R, Ferrucci PF, Smylie M, Hogg D, Hill A, Márquez-Rodas I, Haanen J, Guidoboni M, Maio M, Schöffski P, Carlino MS, Lebbé C, McArthur G, Ascierto PA, Daniels GA, Long GV, Bastholt L, Rizzo JI, Balogh A, Moshyk A, Hodi FS, Wolchok JD. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2019;381(16):1535–1546. doi: 10.1056/NEJMoa1910836.
    1. Marin-Acevedo JA, Chirila RM, Dronca RS. Immune checkpoint inhibitor toxicities. Mayo Clin Proc. 2019;94(7):1321–1329. doi: 10.1016/j.mayocp.2019.03.012.
    1. Dummer R, Schadendorf D, Ascierto PA, Arance A, Dutriaux C, Di Giacomo AM, Rutkowski P, Del Vecchio M, Gutzmer R, Mandala M, et al. Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017;18(4):435–445. doi: 10.1016/S1470-2045(17)30180-8.
    1. Greger JG, Eastman SD, Zhang V, Bleam MR, Hughes AM, Smitheman KN, Dickerson SH, Laquerre SG, Liu L, Gilmer TM. Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Mol Cancer Ther. 2012;11(4):909–920. doi: 10.1158/1535-7163.MCT-11-0989.
    1. Sosman JAKM, Lolkema MPJ, et al. A phase 1b/2 study of LEE011 in combination with binimetinib (MEK162) in patients with NRAS-mutant melanoma: early encouraging clinical activity. J Clin Oncol. 2014;32(15_suppl):9009. doi: 10.1200/jco.2014.32.15_suppl.9009.
    1. Xu F, He Q, Li X, Chang CK, Wu LY, Zhang Z, Liu L, Shi WH, Zhu Y, Zhao YS, et al. Rigosertib as a selective anti-tumor agent can ameliorate multiple dysregulated signaling transduction pathways in high-grade myelodysplastic syndrome. Sci Rep. 2014;4:7310. doi: 10.1038/srep07310.
    1. Hyoda T, Tsujioka T, Nakahara T, Suemori S, Okamoto S, Kataoka M, Tohyama K. Rigosertib induces cell death of a myelodysplastic syndrome-derived cell line by DNA damage-induced G2/M arrest. Cancer Sci. 2015;106(3):287–293. doi: 10.1111/cas.12605.
    1. Chapman CM, Sun X, Roschewski M, Aue G, Farooqui M, Stennett L, Gibellini F, Arthur D, Perez-Galan P, Wiestner A. ON is selectively cytotoxic for chronic lymphocytic leukemia cells through a dual mechanism of action involving PI3K/AKT inhibition and induction of oxidative stress. Clin Cancer Res. 2012;18(7):1979–1991. doi: 10.1158/1078-0432.CCR-11-2113.
    1. Athuluri-Divakar SK, Vasquez-Del Carpio R, Dutta K, Baker SJ, Cosenza SC, Basu I, Gupta YK, Reddy MV, Ueno L, Hart JR, et al. A small molecule RAS-mimetic disrupts RAS association with effector proteins to block signaling. Cell. 2016;165(3):643–655. doi: 10.1016/j.cell.2016.03.045.
    1. Prasad A, Khudaynazar N, Tantravahi RV, Gillum AM, Hoffman BS. ON (rigosertib) inhibits PI3K/Akt pathway and activates oxidative stress signals in head and neck cancer cell lines. Oncotarget. 2016;7(48):79388–79400. doi: 10.18632/oncotarget.12692.
    1. Dai YH, Hung LY, Chen RY, Lai CH, Chang KC. ON inhibits growth of diffuse large B-cell lymphoma by cytoplasmic sequestration of sumoylated C-MYB/TRAF6 complex. Transl Res. 2016;175:129–143. doi: 10.1016/j.trsl.2016.04.001.
    1. Garcia-Manero G, Fenaux P, Al-Kali A, Baer MR, Sekeres MA, Roboz GJ, Gaidano G, Scott BL, Greenberg P, Platzbecker U, et al. Rigosertib versus best supportive care for patients with high-risk myelodysplastic syndromes after failure of hypomethylating drugs (ONTIME): a randomised, controlled, phase 3 trial. Lancet Oncol. 2016;17(4):496–508. doi: 10.1016/S1470-2045(16)00009-7.
    1. Silverman LR, Greenberg P, Raza A, Olnes MJ, Holland JF, Reddy P, Maniar M, Wilhelm F. Clinical activity and safety of the dual pathway inhibitor rigosertib for higher risk myelodysplastic syndromes following DNA methyltransferase inhibitor therapy. Hematol Oncol. 2015;33(2):57–66. doi: 10.1002/hon.2137.
    1. Jimeno A, Li J, Messersmith WA, Laheru D, Rudek MA, Maniar M, Hidalgo M, Baker SD, Donehower RC. Phase I study of ON , a novel modulator of the polo-like kinase 1 pathway, in adult patients with solid tumors. J Clin Oncol. 2008;26(34):5504–5510. doi: 10.1200/JCO.2008.17.9788.
    1. O'Neil BH, Scott AJ, Ma WW, Cohen SJ, Leichman L, Aisner DL, Menter AR, Tejani MA, Cho JK, Granfortuna J, Coveler L, Olowokure OO, Baranda JC, Cusnir M, Phillip P, Boles J, Nazemzadeh R, Rarick M, Cohen DJ, Radford J, Fehrenbacher L, Bajaj R, Bathini V, Fanta P, Berlin J, McRee AJ, Maguire R, Wilhelm F, Maniar M, Jimeno A, Gomes CL, Messersmith WA. A phase II/III randomized study to compare the efficacy and safety of rigosertib plus gemcitabine versus gemcitabine alone in patients with previously untreated metastatic pancreatic cancer. Ann Oncol. 2015;26(12):2505. doi: 10.1093/annonc/mdv477.
    1. Ohnuma T, Lehrer D, Ren C, Cho SY, Maniar M, Silverman L, Sung M, Gretz HF, 3rd, Benisovich V, Navada S, et al. Phase 1 study of intravenous rigosertib (ON ), a novel benzyl styryl sulfone structure producing G2/M arrest and apoptosis, in adult patients with advanced cancer. Am J Cancer Res. 2013;3(3):323–338.
    1. Tong AW, Stone MJ. Prospects for CD40-directed experimental therapy of human cancer. Cancer Gene Ther. 2003;10(1):1–13. doi: 10.1038/sj.cgt.7700527.
    1. Kalbasi A, Fonsatti E, Natali PG, Altomonte M, Bertocci E, Cutaia O, Calabro L, Chiou M, Tap W, Chmielowski B, et al. CD40 expression by human melanocytic lesions and melanoma cell lines and direct CD40 targeting with the therapeutic anti-CD40 antibody CP-870,893. J Immunother. 2010;33(8):810–816. doi: 10.1097/CJI.0b013e3181ee73a7.
    1. von Leoprechting A, van der Bruggen P, Pahl HL, Aruffo A, Simon JC. Stimulation of CD40 on immunogenic human malignant melanomas augments their cytotoxic T lymphocyte-mediated lysis and induces apoptosis. Cancer Res. 1999;59:1287–1294.
    1. Ibraheem K, Yhmed AMA, Qayyum T, Bryan NP, Georgopoulos NT. CD40 induces renal cell carcinoma-specific differential regulation of TRAF proteins, ASK1 activation and JNK/p38-mediated, ROS-dependent mitochondrial apoptosis. Cell Death Discov. 2019;5(1):148. doi: 10.1038/s41420-019-0229-8.
    1. Georgopoulos NT, Steele LP, Thomson MJ, Selby PJ, Southgate J, Trejdosiewicz LK. A novel mechanism of CD40-induced apoptosis of carcinoma cells involving TRAF3 and JNK/AP-1 activation. Cell Death Differ. 2006;13(10):1789–1801. doi: 10.1038/sj.cdd.4401859.
    1. Eliopoulos AG, Davies C, Knox PG, Gallagher NJ, Afford SC, Adams DH, Young LS. CD40 induces apoptosis in carcinoma cells through activation of cytotoxic ligands of the tumor necrosis factor superfamily. Mol Cell Biol. 2000;20(15):5503–5515. doi: 10.1128/MCB.20.15.5503-5515.2000.
    1. Qiu X, Klausen C, Cheng JC, Leung PC. CD40 ligand induces RIP1-dependent, necroptosis-like cell death in low-grade serous but not serous borderline ovarian tumor cells. Cell Death Dis. 2015;6(8):e1864. doi: 10.1038/cddis.2015.229.
    1. Hill SC, Youde SJ, Man S, Teale GR, Baxendale AJ, Hislop A, Davies CC, Luesley DM, Blom AM, Rickinson AB, Young LS, Eliopoulos AG. Activation of CD40 in cervical carcinoma cells facilitates CTL responses and augments chemotherapy-induced apoptosis. J Immunol. 2005;174(1):41–50. doi: 10.4049/jimmunol.174.1.41.
    1. Knox PG, Davies CC, Ioannou M, Eliopoulos AG. The death domain kinase RIP1 links the immunoregulatory CD40 receptor to apoptotic signaling in carcinomas. J Cell Biol. 2011;192(3):391–399. doi: 10.1083/jcb.201003087.
    1. Pirozzi G, Lombari V, Zanzi D, Ionna F, Lombardi ML, Errico S, Ruggiero G, Manzo C. CD40 expressed on human melanoma cells mediates T cell co-stimulation and tumor cell growth. Int Immunol. 2000;12(6):787–795. doi: 10.1093/intimm/12.6.787.
    1. Forbes SA, Bhamra G, Bamford S, Dawson E, Kok C, Clements J, Menzies A, Teague JW, Futreal PA, Stratton MR: The catalogue of somatic mutations in cancer (COSMIC). Curr Protoc Hum Genet 2008, Chapter 10:Unit 10–11.
    1. Farooqi AA, Li KT, Fayyaz S, Chang YT, Ismail M, Liaw CC, Yuan SS, Tang JY, Chang HW. Anticancer drugs for the modulation of endoplasmic reticulum stress and oxidative stress. Tumour Biol. 2015;36(8):5743–5752. doi: 10.1007/s13277-015-3797-0.
    1. Ritt DA, Abreu-Blanco MT, Bindu L, Durrant DE, Zhou M, Specht SI, Stephen AG, Holderfield M, Morrison DK. Inhibition of Ras/Raf/MEK/ERK pathway signaling by a stress-induced phospho-regulatory circuit. Mol Cell. 2016;64(5):875–887. doi: 10.1016/j.molcel.2016.10.029.
    1. Aaes TL, Vandenabeele P. The intrinsic immunogenic properties of cancer cell lines, immunogenic cell death, and how these influence host antitumor immune responses. Cell Death Differ. 2021;28(3):843–860. doi: 10.1038/s41418-020-00658-y.
    1. Waithman J, Gebhardt T, Bedoui S. Skin tumor immunity: site does matter for antigen presentation by DCs. Eur J Immunol. 2016;46(3):543–546. doi: 10.1002/eji.201646293.
    1. Choi J, Beaino W, Fecek RJ, Fabian KPL, Laymon CM, Kurland BF, Storkus WJ, Anderson CJ. Combined VLA-4-targeted radionuclide therapy and immunotherapy in a mouse model of melanoma. J Nucl Med. 2018;59(12):1843–1849. doi: 10.2967/jnumed.118.209510.
    1. Meng Y, Harlin H, O'Keefe JP, Gajewski TF. Induction of cytotoxic granules in human memory CD8+ T cell subsets requires cell cycle progression. J Immunol. 2006;177(3):1981–1987. doi: 10.4049/jimmunol.177.3.1981.
    1. LaFleur MW, Muroyama Y, Drake CG, Sharpe AH. Inhibitors of the PD-1 pathway in tumor therapy. J Immunol. 2018;200(2):375–383. doi: 10.4049/jimmunol.1701044.
    1. Khan AR, Hams E, Floudas A, Sparwasser T, Weaver CT, Fallon PG. PD-L1hi B cells are critical regulators of humoral immunity. Nat Commun. 2015;6(1):5997. doi: 10.1038/ncomms6997.
    1. Guan H, Lan Y, Wan Y, Wang Q, Wang C, Xu L, Chen Y, Liu W, Zhang X, Li Y, Gu Y, Wang Z, Xie F. PD-L1 mediated the differentiation of tumor-infiltrating CD19(+) B lymphocytes and T cells in invasive breast cancer. Oncoimmunology. 2016;5(2):e1075112. doi: 10.1080/2162402X.2015.1075112.
    1. Yang J, Yan C, Vilgelm AE, Chen SC, Ayers GD, Johnson CA, Richmond A. Targeted deletion of CXCR2 in myeloid cells alters the tumor immune environment to improve antitumor immunity. Cancer Immunol Res. 2021;9(2):200–213. doi: 10.1158/2326-6066.CIR-20-0312.
    1. Cunningham AF, Flores-Langarica A, Bobat S, Dominguez Medina CC, Cook CN, Ross EA, Lopez-Macias C, Henderson IR. B1b cells recognize protective antigens after natural infection and vaccination. Front Immunol. 2014;5:535. doi: 10.3389/fimmu.2014.00535.
    1. Yang J, Hawkins OE, Barham W, Gilchuk P, Boothby M, Ayers GD, Joyce S, Karin M, Yull FE, Richmond A. Myeloid IKKbeta promotes antitumor immunity by modulating CCL11 and the innate immune response. Cancer Res. 2014;74(24):7274–7284. doi: 10.1158/0008-5472.CAN-14-1091.
    1. Yang Y, Wilson JM. CD40 ligand-dependent T cell activation: requirement of B7-CD28 signaling through CD40. Science. 1996;273(5283):1862–1864. doi: 10.1126/science.273.5283.1862.
    1. Bugeon L, Dallman MJ. Costimulation of T cells. Am J Respir Crit Care Med. 2000;162(supplement_3):S164–S168. doi: 10.1164/ajrccm.162.supplement_3.15tac5.
    1. Shattuck-Brandt RL, Chen SC, Murray E, Johnson CA, Crandall H, O'Neal JF, Al-Rohil RN, Nebhan CA, Bharti V, Dahlman KB, et al. Metastatic melanoma patient-derived xenografts respond to mdm2 inhibition as a single agent or in combination with BRAF/MEK inhibition. Clin Cancer Res. 2020;26(14):3803–3818. doi: 10.1158/1078-0432.CCR-19-1895.
    1. Chen PL, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, Miller JP, Bassett RL, Gopalakrishnan V, Wani K, de Macedo MP, Austin-Breneman JL, Jiang H, Chang Q, Reddy SM, Chen WS, Tetzlaff MT, Broaddus RJ, Davies MA, Gershenwald JE, Haydu L, Lazar AJ, Patel SP, Hwu P, Hwu WJ, Diab A, Glitza IC, Woodman SE, Vence LM, Wistuba II, Amaria RN, Kwong LN, Prieto V, Davis RE, Ma W, Overwijk WW, Sharpe AH, Hu J, Futreal PA, Blando J, Sharma P, Allison JP, Chin L, Wargo JA. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov. 2016;6(8):827–837. doi: 10.1158/-15-1545.
    1. Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, Hodi FS, Martin-Algarra S, Mandal R, Sharfman WH, et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell. 2017;171(4):934–949. doi: 10.1016/j.cell.2017.09.028.
    1. Jerby-Arnon L, Shah P, Cuoco MS, Rodman C, Su MJ, Melms JC, Leeson R, Kanodia A, Mei S, Lin JR, Wang S, Rabasha B, Liu D, Zhang G, Margolais C, Ashenberg O, Ott PA, Buchbinder EI, Haq R, Hodi FS, Boland GM, Sullivan RJ, Frederick DT, Miao B, Moll T, Flaherty KT, Herlyn M, Jenkins RW, Thummalapalli R, Kowalczyk MS, Cañadas I, Schilling B, Cartwright ANR, Luoma AM, Malu S, Hwu P, Bernatchez C, Forget MA, Barbie DA, Shalek AK, Tirosh I, Sorger PK, Wucherpfennig K, van Allen EM, Schadendorf D, Johnson BE, Rotem A, Rozenblatt-Rosen O, Garraway LA, Yoon CH, Izar B, Regev A. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell. 2018;175(4):984–997. doi: 10.1016/j.cell.2018.09.006.
    1. Baker SJ, Cosenza SC, Athuluri-Divakar S, Reddy MVR, Carpio RV, Jain R, Aggarwal AK, Reddy EP. Mechanism of action of rigosertib does not involve tubulin binding. BioRxiv 2019.
    1. Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 2016;7(2):27–31. doi: 10.4103/0976-0105.177703.
    1. Fogarty CE, Bergmann A. Killers creating new life: caspases drive apoptosis-induced proliferation in tissue repair and disease. Cell Death Differ. 2017;24(8):1390–1400. doi: 10.1038/cdd.2017.47.
    1. Diwanji N, Bergmann A. An unexpected friend - ROS in apoptosis-induced compensatory proliferation: implications for regeneration and cancer. Semin Cell Dev Biol. 2018;80:74–82. doi: 10.1016/j.semcdb.2017.07.004.
    1. Dunnill CJ, Ibraheem K, Mohamed A, Southgate J, Georgopoulos NT. A redox state-dictated signalling pathway deciphers the malignant cell specificity of CD40-mediated apoptosis. Oncogene. 2017;36(18):2515–2528. doi: 10.1038/onc.2016.401.
    1. Beatty GL, Li Y, Long KB. Cancer immunotherapy: activating innate and adaptive immunity through CD40 agonists. Expert Rev Anticancer Ther. 2017;17(2):175–186. doi: 10.1080/14737140.2017.1270208.
    1. Long KB, Gladney WL, Tooker GM, Graham K, Fraietta JA, Beatty GL. IFNgamma and CCL2 cooperate to redirect tumor-infiltrating monocytes to degrade fibrosis and enhance chemotherapy efficacy in pancreatic carcinoma. Cancer Discov. 2016;6(4):400–413. doi: 10.1158/-15-1032.
    1. Singh M, Vianden C, Cantwell MJ, Dai Z, Xiao Z, Sharma M, Khong H, Jaiswal AR, Faak F, Hailemichael Y, Janssen LME, Bharadwaj U, Curran MA, Diab A, Bassett RL, Tweardy DJ, Hwu P, Overwijk WW. Intratumoral CD40 activation and checkpoint blockade induces T cell-mediated eradication of melanoma in the brain. Nat Commun. 2017;8(1):1447. doi: 10.1038/s41467-017-01572-7.
    1. Dempke WCM, Fenchel K, Uciechowski P, Dale SP. Second- and third-generation drugs for immuno-oncology treatment-the more the better? Eur J Cancer. 2017;74:55–72. doi: 10.1016/j.ejca.2017.01.001.
    1. Irenaeus SMM, Nielsen D, Ellmark P, Yachnin J, Deronic A, Nilsson A, Norlen P, Veitonmaki N, Wennersten CS, Ullenhag GJ. First-in-human study with intratumoral administration of a CD40 agonistic antibody, ADC-1013, in advanced solid malignancies. Int J Cancer. 2019;145(5):1189–1199. doi: 10.1002/ijc.32141.
    1. Knorr DA, Dahan R, Ravetch JV. Toxicity of an fc-engineered anti-CD40 antibody is abrogated by intratumoral injection and results in durable antitumor immunity. Proc Natl Acad Sci U S A. 2018;115(43):11048–11053. doi: 10.1073/pnas.1810566115.
    1. Litchfield K, Reading JL, Puttick C, Thakkar K, Abbosh C, Bentham R, Watkins TBK, Rosenthal R, Biswas D, Rowan A, Lim E, al Bakir M, Turati V, Guerra-Assunção JA, Conde L, Furness AJS, Saini SK, Hadrup SR, Herrero J, Lee SH, van Loo P, Enver T, Larkin J, Hellmann MD, Turajlic S, Quezada SA, McGranahan N, Swanton C. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell. 2021;184(3):596–614. doi: 10.1016/j.cell.2021.01.002.
    1. Jost M, Chen Y, Gilbert LA, Horlbeck MA, Krenning L, Menchon G, Rai A, Cho MY, Stern JJ, Prota AE, Kampmann M, Akhmanova A, Steinmetz MO, Tanenbaum ME, Weissman JS. Combined CRISPRi/a-based chemical genetic screens reveal that rigosertib is a microtubule-destabilizing agent. Mol Cell. 2017;68(1):210–223. doi: 10.1016/j.molcel.2017.09.012.
    1. Jost M, Chen Y, Gilbert LA, Horlbeck MA, Krenning L, Menchon G, Rai A, Cho MY, Stern JJ, Prota AE, Kampmann M, Akhmanova A, Steinmetz MO, Tanenbaum ME, Weissman JS. Pharmaceutical-grade rigosertib is a microtubule-destabilizing agent. Mol Cell. 2020;79(1):191–198. doi: 10.1016/j.molcel.2020.06.008.
    1. Vilgelm AE, Pawlikowski JS, Liu Y, Hawkins OE, Davis TA, Smith J, Weller KP, Horton LW, McClain CM, Ayers GD, et al. Mdm2 and aurora kinase a inhibitors synergize to block melanoma growth by driving apoptosis and immune clearance of tumor cells. Cancer Res. 2015;75(1):181–193. doi: 10.1158/0008-5472.CAN-14-2405.
    1. Yang J, Kumar A, Vilgelm AE, Chen SC, Ayers GD, Novitskiy SV, Joyce S, Richmond A. Loss of CXCR4 in myeloid cells enhances antitumor immunity and reduces melanoma growth through NK cell and FASL mechanisms. Cancer Immunol Res. 2018;6(10):1186–1198. doi: 10.1158/2326-6066.CIR-18-0045.
    1. Olivo Pimentel V, Yaromina A, Marcus D, Dubois LJ, Lambin P. A novel co-culture assay to assess anti-tumor CD8(+) T cell cytotoxicity via luminescence and multicolor flow cytometry. J Immunol Methods. 2020;487:112899. doi: 10.1016/j.jim.2020.112899.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. doi: 10.1186/s13059-014-0550-8.
    1. LBenjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Statist Soc B. 1995;57(1):289-300.

Source: PubMed

3
Předplatit