Multimodality analysis confers a prognostic benefit of a T-cell infiltrated tumor microenvironment and peripheral immune status in patients with melanoma

Georgia M Beasley, Michael C Brown, Norma E Farrow, Karenia Landa, Rami N Al-Rohil, Maria Angelica Selim, Aaron D Therien, Sin-Ho Jung, Junheng Gao, David Boczkowski, Eda K Holl, April K S Salama, Darell D Bigner, Matthias Gromeier, Smita K Nair, Georgia M Beasley, Michael C Brown, Norma E Farrow, Karenia Landa, Rami N Al-Rohil, Maria Angelica Selim, Aaron D Therien, Sin-Ho Jung, Junheng Gao, David Boczkowski, Eda K Holl, April K S Salama, Darell D Bigner, Matthias Gromeier, Smita K Nair

Abstract

Background: We previously reported results from a phase 1 study testing intratumoral recombinant poliovirus, lerapolturev, in 12 melanoma patients. All 12 patients received anti-PD-1 systemic therapy before lerapolturev, and 11 of these 12 patients also received anti-PD-1 after lerapolturev. In preclinical models lerapolturev induces intratumoral innate inflammation that engages antitumor T cells. In the current study, prelerapolturev and postlerapolturev tumor biopsies and blood were evaluated for biomarkers of response.

Methods: The following analyses were performed on tumor tissue (n=11): (1) flow cytometric assessment of immune cell density, (2) NanoString Digital Spatial profiling of protein and the transcriptome, and (3) bulk RNA sequencing. Immune cell phenotypes and responsiveness to in vitro stimulation, including in vitro lerapolturev challenge, were measured in peripheral blood (n=12).

Results: Three patients who received anti-PD-1 therapy within 30 days of lerapolturev have a current median progression-free survival (PFS) of 2.3 years and had higher CD8+T cell infiltrates in prelerapolturev tumor biopsies relative to that of 7 patients with median PFS of 1.6 months and lower CD8+T cell infiltrates in prelerapolturev tumor biopsies. In peripheral blood, four patients with PFS 2.3 years (including three that received anti-PD-1 therapy within 30 days before lerapolturev and had higher pretreatment tumor CD8+T cell infiltrates) had significantly higher effector memory (CD8+, CCR7-, CD45RA-) but lower CD8+PD-1+ and CD4+PD-1+ cells compared with eight patients with median PFS 1.6 months. In addition, pretreatment blood from the four patients with median PFS 2.3 years had more potent antiviral responses to in vitro lerapolturev challenge compared with eight patients with median PFS 1.6 months.

Conclusion: An inflamed pretreatment tumor microenvironment, possibly induced by prior anti-PD-1 therapy and a proficient peripheral blood pretreatment innate immune response (antiviral/interferon signaling) to lerapolturev was associated with long term PFS after intratumoral lerapolturev in a small cohort of patients. These findings imply a link between intratumoral T cell inflammation and peripheral immune function.

Trial registration number: NCT03712358.

Keywords: immunity, innate; oncolytic virotherapy; tumor microenvironment.

Conflict of interest statement

Competing interests: GMB reports clinical trial funding from Istari Oncology, Delcath, Oncosec Medical, Replimune, and Checkmate Pharmaceuticals paid to Duke University. AnS receives research funding paid to the institution from Bristol Myers Squibb, Immunocore, Merck, Pfizer Advisory roles; and has served on advisory boards for Array, Novartis, Iovance, and Regeneron. MCB, DDB and MG are paid consultants of Istari Oncology, Inc. and MG and DDB are scientific advisors for Istari Oncology; DDB and MG hold equity in Istari Oncology. MCB, DDB, MG and SKN own intellectual property related to this research, which has been licensed to Istari Oncology. Duke University (Licensor of PVSRIPO) has a financial interest in Istari Oncology. All other authors declare they have no competing interests.

© 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
Flow cytometry and RNA sequencing analysis of tumor tissue. (A) Number of CD8+T cells and (B) number of CD4+T cells in tumor tissue as assessed by flow cytometry of fresh tumor tissue. (C)- Number of CD8+T cells (left) and CD4+T cells (right) in pretreatment tumor tissue. (D) CIBERSORT predicted (bulk RNA-sequencing) enrichment of CD8+T cells and active memory CD4+T cells in pretreatment tumor tissue Please see online supplemental figure S1 for full heatmap of CIBERSORT enrichment scores for each patient. Patients in red have current median PFS 2.3 years compared with patients in black text with median PFS 1.6 months. PFS, progression-free survival.
Figure 2
Figure 2
Analysis of tumor tissue sections. NanoString Digital Spatial Profiling (DSP) low power fluorescent images of pre-treatment tumor tissue; CD3=red, CD11c=yellow, CD163=green, DNA=blue. All images are ‘low power’= 2 mm field of view, 100 x total magnification. Number in left lower corner indicates patient number. Top row, patients with median PFS 1.6 months after lerapolturev. Bottom row, patients with median PFS 2.3 years after lerapolturev. The number of CD3+T cells as assessed by flow cytometry is also shown for each tumor. Below the DSP images, a corresponding H&E stain is also shown. Note for patient 11, the H&E demonstrates near 100% necrotic tissue in the pretreatment biopsy, as such this was dropped from analysis. PFS, progression-free survival.
Figure 3
Figure 3
NanoString Digital Spatial Profiling (DSP) of tumor tissue. Top left and middle panel, ‘low power’ = 2 mm field of view, 100 x total magnification, ‘high power’ = 45 μm field of view, 400 × total magnification. (A, B) Patient 2 prelerapolturev and (C, D) post-lerapolturev tumor. (A, C) Corresponding H&E. (B) DSP low power, fluorescent image, CD3=red, CD11c=yellow, CD163=green, DNA=blue. Same fluorescent markers also present in (D, F, H). B1—High power view of region 1 circled (white circle) in image B. (B2) High power view of region 2 circled (white circle) in image B. (D) DSP image low power, tumor cells can be identified as cells with irregular blue nucleus. D1—High power view of region 1 circled (white circle) in image D. D2—High power view of region 2 circled (white circle) in image D. Bottom left and middle (E, F) Patient 8 prelerapolturev and G, H postlerapolturev tumor. (E, G) Corresponding H&E stains. (F) DSP image low power. (F1) High power view of region 1 circled (white circle) in image F. (F2) High power view of region 2 circled (white circle) in image F. Tumor cells can be identified as cells with irregular blue nucleus depicted in F2, red circle. H- DSP image low power. (H1) High power view of region 1 circled (white circle) in image H. (H2) High power view of region 2 circled (white circle) in image (H. I, J) Heatmaps of selected protein expression obtained from tumor regions of interest in (I) pretreatment tumor tissue and J- CD3+compartment only. Patient identification on the y axis (right), pre-NON is pretreatment tissue collected without lerapolturev injection.
Figure 4
Figure 4
NanoString Digital Spatial Profiling (DSP) of tumor tissue. NanoString DSP images after fluorescent staining on pretreatment tissue, CD8=teal, CD4=red, CD11c=yellow, and DNA=blue. ‘Low power’ = 2 mm field of view, 100 × total magnification. Rectangles indicate ROIs selected based on enrichment for cell type or tumor. (A) Low power image patient 12. (B) Low power image patient 7. (C) Volcano plot comparing ROI on CD8+ cells only between patients 8, 9, 12, 5 and 7. Differentially expressed genes appear in blue. ROI, regions of interest.
Figure 5
Figure 5
Flow cytometry analysis of immune subsets in peripheral blood. PBMCs were analyzed for T cell memory populations and PD1 expression on CD8+ and CD4+ conventional T cells as shown at the pretreatment time point. Patients in red have median PFS of 2.3 years after lerapolturev and patients in black have median PFS 1.6 months after lerapolturev. Data brackets indicate mean ±SEM; p values are from unpaired t-test. PBMCs, peripheral blood mononuclear cells; PFS, progression-free survival.
Figure 6
Figure 6
Assessment of baseline peripheral immune cell function in response to lerapolturev. PBMCs from the pretreatment time point were challenged with laboratory grade PVSRIPO in vitro for 24 hours. Supernatant was tested for proinflammatory cytokines. Patients in red have median PFS of 2.3 years after lerapolturev and patients in black have median PFS 1.6 months after lerapolturev. Data brackets indicate mean ±SEM; p values are from unpaired t-test. See online supplemental figure S3 for complete results of cytokines and stimuli tested; online supplemental figure S4 presents baseline raw MFI and concentration of cytokines for all stimuli. GMCSF, granulocyte-macrophage colony-stimulating factor; MFI, mean fluorescence intensity; PBMCs, peripheral blood mononuclear cells; PFS, progression-free survival.

References

    1. Jacquelot N, Roberti MP, Enot DP, et al. . Predictors of responses to immune checkpoint blockade in advanced melanoma. Nat Commun 2017;8:592. 10.1038/s41467-017-00608-2
    1. Erdag G, Schaefer JT, Smolkin ME, et al. . Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res 2012;72:1070–80. 10.1158/0008-5472.CAN-11-3218
    1. Horton BL, Williams JB, Cabanov A, et al. . Intratumoral CD8+ T-cell apoptosis is a major component of T-cell dysfunction and impedes antitumor immunity. Cancer Immunol Res 2018;6:14–24. 10.1158/2326-6066.CIR-17-0249
    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. Wei SC, Levine JH, Cogdill AP, et al. . Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 2017;170:1120–33. 10.1016/j.cell.2017.07.024
    1. Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin Cancer Res 2016;22:1865–74. 10.1158/1078-0432.CCR-15-1507
    1. Taube JM, Klein A, Brahmer JR, et al. . Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 2014;20:5064–74. 10.1158/1078-0432.CCR-13-3271
    1. Siddiqui I, Schaeuble K, Chennupati V, et al. . Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 2019;50:195–211. 10.1016/j.immuni.2018.12.021
    1. Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015;348:56–61. 10.1126/science.aaa8172
    1. Herbst RS, Soria J-C, Kowanetz M, et al. . Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014;515:563–7. 10.1038/nature14011
    1. Robert C, Long GV, Brady B, et al. . Five-Year Outcomes With Nivolumab in Patients With Wild-Type BRAF Advanced Melanoma. J Clin Oncol 2020;38:3937–46. 10.1200/JCO.20.00995
    1. Ribas A, Dummer R, Puzanov I, et al. . Oncolytic virotherapy promotes intratumoral T cell infiltration and improves anti-PD-1 immunotherapy. Cell 2017;170:1109–19. 10.1016/j.cell.2017.08.027
    1. Cerullo V, Diaconu I, Romano V, et al. . An oncolytic adenovirus enhanced for toll-like receptor 9 stimulation increases antitumor immune responses and tumor clearance. Molecular Therapy 2012;20:2076–86. 10.1038/mt.2012.137
    1. Gromeier M, Lachmann S, Rosenfeld MR, et al. . Intergeneric poliovirus recombinants for the treatment of malignant glioma. Proc Natl Acad Sci U S A 2000;97:6803–8. 10.1073/pnas.97.12.6803
    1. Holl EK, Brown MC, Boczkowski D, et al. . Recombinant oncolytic poliovirus, PVSRIPO, has potent cytotoxic and innate inflammatory effects, mediating therapy in human breast and prostate cancer xenograft models. Oncotarget 2016;7:79828–41. 10.18632/oncotarget.12975
    1. Brown MC, Holl EK, Boczkowski D, et al. . Cancer immunotherapy with recombinant poliovirus induces IFN-dominant activation of dendritic cells and tumor antigen–specific CTLs. Sci Transl Med 2017;9. 10.1126/scitranslmed.aan4220
    1. Brown MC, Mosaheb MM, Mohme M, et al. . Viral infection of cells within the tumor microenvironment mediates antitumor immunotherapy via selective TBK1-IRF3 signaling. Nat Commun 2021;12:1858. 10.1038/s41467-021-22088-1
    1. Beasley GM, Nair SK, Farrow NE, et al. . Phase I trial of intratumoral PVSRIPO in patients with unresectable, treatment-refractory melanoma. J Immunother Cancer 2021;9:e002203. 10.1136/jitc-2020-002203
    1. Holl EK, Frazier VN, Landa K, et al. . Examining peripheral and tumor cellular Immunome in patients with cancer. Front Immunol 2019;10:10. 10.3389/fimmu.2019.01767
    1. Brown MC, Bryant JD, Dobrikova EY, et al. . Induction of viral, 7-methyl-guanosine cap-independent translation and oncolysis by mitogen-activated protein kinase-interacting kinase-mediated effects on the serine/arginine-rich protein kinase. J Virol 2014;88:13135–48. 10.1128/JVI.01883-14
    1. Beechem JM. High-Plex spatially resolved RNA and protein detection using digital spatial profiling: a technology designed for Immuno-oncology biomarker discovery and translational research. Methods Mol Biol 2020;2055:563–83. 10.1007/978-1-4939-9773-2_25
    1. Toki MI, Merritt CR, Wong PF, et al. . High-plex predictive marker discovery for melanoma Immunotherapy–Treated patients using digital spatial profiling. Clin Cancer Res 2019;25:5503–12. 10.1158/1078-0432.CCR-19-0104
    1. Delorey TM, Ziegler CGK, Heimberg G, et al. . COVID-19 tissue atlases reveal SARS-CoV-2 pathology and cellular targets. Nature 2021;595:107–13. 10.1038/s41586-021-03570-8
    1. Delorey TM, Ziegler CGK, Heimberg G, et al. . A single-cell and spatial atlas of autopsy tissues reveals pathology and cellular targets of SARS-CoV-2. bioRxiv 2021. doi:10.1101/2021.02.25.430130. [Epub ahead of print: 25 Feb 2021].
    1. Morpheus. Available:
    1. Spearman C. The proof and measurement of association between two things. Am J Psychol 1987;100:441–71. 10.2307/1422689
    1. Newman AM, Steen CB, Liu CL, et al. . Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat Biotechnol 2019;37:773–82. 10.1038/s41587-019-0114-2
    1. Kluger HM, Tawbi HA, Ascierto ML, et al. . Defining tumor resistance to PD-1 pathway blockade: recommendations from the first meeting of the SITC immunotherapy resistance Taskforce. J Immunother Cancer 2020;8:e000398. 10.1136/jitc-2019-000398
    1. Boukhaled GM, Gadalla R, Elsaesser HJ, et al. . Pre-encoded responsiveness to type I interferon in the peripheral immune system defines outcome of PD1 blockade therapy. Nat Immunol 2022;23:1273–83. 10.1038/s41590-022-01262-7
    1. Yost KE, Satpathy AT, Wells DK, et al. . Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med 2019;25:1251–9. 10.1038/s41591-019-0522-3
    1. Gajewski TF, Corrales L, Williams J, et al. . Cancer immunotherapy targets based on understanding the T Cell-Inflamed versus non-T Cell-Inflamed tumor microenvironment. Adv Exp Med Biol 2017;1036:19–31. 10.1007/978-3-319-67577-0_2
    1. Fu Q, Chen N, Ge C, et al. . Prognostic value of tumor-infiltrating lymphocytes in melanoma: a systematic review and meta-analysis. Oncoimmunology 2019;8:1593806. 10.1080/2162402X.2019.1593806
    1. Voabil P, de Bruijn M, Roelofsen LM, et al. . An ex vivo tumor fragment platform to dissect response to PD-1 blockade in cancer. Nat Med 2021;27:1250–61. 10.1038/s41591-021-01398-3
    1. Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol 2018;15:81–94. 10.1038/nrclinonc.2017.166
    1. Holling TM, Schooten E, van Den Elsen PJ. Function and regulation of MHC class II molecules in T-lymphocytes: of mice and men. Hum Immunol 2004;65:282–90. 10.1016/j.humimm.2004.01.005
    1. Liu N, Phillips T, Zhang M, et al. . Serine protease inhibitor 2A is a protective factor for memory T cell development. Nat Immunol 2004;5:919–26. 10.1038/ni1107
    1. Kim KH, Cho J, Ku BM, et al. . The First-Week Proliferative Response of Peripheral Blood PD-1+CD8+ T Cells Predicts the Response to Anti-PD-1 Therapy in Solid Tumors. Clin Cancer Res 2019;25:2144–54. 10.1158/1078-0432.CCR-18-1449
    1. Huang AC, Orlowski RJ, Xu X, et al. . A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med 2019;25:454–61. 10.1038/s41591-019-0357-y
    1. Gogas H, Ribas A, Chesney J. MASTERKEY-265: A phase 3, randomized, placebo (PBO)-controlled study of talimogene laherparepvec (T) plus pembrolizumab (P) for unresectable stage IIIB-IVM1C melanoma (MEL) 2021.

Source: PubMed

3
Předplatit