Immune cellular patterns of distribution affect outcomes of patients with non-small cell lung cancer

Edwin Roger Parra, Jiexin Zhang, Mei Jiang, Auriole Tamegnon, Renganayaki Krishna Pandurengan, Carmen Behrens, Luisa Solis, Cara Haymaker, John Victor Heymach, Cesar Moran, Jack J Lee, Don Gibbons, Ignacio Ivan Wistuba, Edwin Roger Parra, Jiexin Zhang, Mei Jiang, Auriole Tamegnon, Renganayaki Krishna Pandurengan, Carmen Behrens, Luisa Solis, Cara Haymaker, John Victor Heymach, Cesar Moran, Jack J Lee, Don Gibbons, Ignacio Ivan Wistuba

Abstract

Studying the cellular geographic distribution in non-small cell lung cancer is essential to understand the roles of cell populations in this type of tumor. In this study, we characterize the spatial cellular distribution of immune cell populations using 23 makers placed in five multiplex immunofluorescence panels and their associations with clinicopathologic variables and outcomes. Our results demonstrate two cellular distribution patterns-an unmixed pattern mostly related to immunoprotective cells and a mixed pattern mostly related to immunosuppressive cells. Distance analysis shows that T-cells expressing immune checkpoints are closer to malignant cells than other cells. Combining the cellular distribution patterns with cellular distances, we can identify four groups related to inflamed and not-inflamed tumors. Cellular distribution patterns and distance are associated with survival in univariate and multivariable analyses. Spatial distribution is a tool to better understand the tumor microenvironment, predict outcomes, and may can help select therapeutic interventions.

Conflict of interest statement

E.R.P. is pathology consultant of the Nucleai LTD. C.H. reports research funding to institution from Sanofi, Dragonfly, BTG, Trisalus, Iovance, and Avenge; scientific advisory board member of Briacell with stock options; personal fees from Nanobiotix and speaker fees/honorarium from SWOG and SITC outside the scope of the submitted work. J.V. H. has received research support from AstraZeneca, Bayer, GlaxoSmithKline, and Spectrum; participated in advisory committees for AstraZeneca, Boehringer Ingelheim, Exelixis, Genentech, GlaxoSmithKline, Guardant Health, Hengrui, Lilly, Novartis, Specrtum, EMD Serono, and Synta; and received royalties and/or licensing fees from Spectrum. D.G. has served on scientific advisory committees for AstraZeneca, GlaxoSmithKline, Sanofi, Eli Lilly and Janssen and has received research support from Janssen, Takeda, Ribon Therapeutics, Astellas and AstraZeneca. I.I.W. has provided consulting or advisory roles for AstraZeneca/MedImmune, Asuragen, Bayer, Bristol Myers Squibb, Genentech/Roche, GlaxoSmithKline, Guardant Health, HTG Molecular Diagnostics, Merck, MSD Oncology, OncoCyte, Novartis, Flame Inc, and Pfizer; has received grants and personal fees from Asuragen, Genentech/Roche, Bristol Myers Squibb, AstraZeneca/MedImmune, HTG Molecular, Merck, and Guardant Health; has received personal fees from GlaxoSmithKline and Oncocyte, Daiichi-Sankyo, Roche, AstraZeneca, Pfizer and Bayer; has received research funding to his institution from 4D Molecular Therapeutics, Adaptimmune, Adaptive Biotechnologies, Akoya Biosciences, Amgen, Bayer, EMD Serono, Genentech, Guardant Health, HTG Molecular Diagnostics, Iovance Biotherapeutics, Johnson & Johnson, Karus Therapeutics, MedImmune, Merck, Novartis, OncoPlex Diagnostics, Pfizer, Silicon Biosystems, Takeda, and Novartis. The other authors declare no competing interests.

© 2023. The Author(s).

Figures

Fig. 1. Representative examples of multispectral images…
Fig. 1. Representative examples of multispectral images from non-small cell lung cancer tissue microarray specimens, with their chord diagram of the markers.
Composite spectral mixing images from multiplex immunofluorescence (mIF; 20× magnification, scale bars represent 50 µm on each image) is shown for (A) panel 1: cytokeratin (CK), CD3, CD8, PD-1, PD-L1, and CD68; (B) panel 2: CK, CD3, CD8, CD45RO, granzyme B (GZB), and FOXP3; (C) panel 3: CK, CD3, PD-L1, B7-H3, B7-H4, IDO-1; and VISTA; (D) panel 4: CK, CD3, ICOS, LAG3, OX40, TIM3, and CD20; (E) panel 5: CK, Arg-1, CD11b, CD14, CD33, CD66b, and CD68. F Chord diagram visualization showing the diversity of inter-relationships between markers’ co-expression, including all the markers in the five mIF panels. Data from 225 samples was used. Experiments and quantifications related to the presented results were conducted once. The images were generated using Vectra-Polaris 1.0.13 scanner system and InForm 2.4.8 image analysis software (Akoya Biosciences). The chord diagram was generated using all tumor cores from all mIF panels by R studio software version 3.6.1. (Source data is provided as a source data file).
Fig. 2. Representative examples of multispectral images…
Fig. 2. Representative examples of multispectral images and uniform manifold approximation and projection (UMAP) to identify cell types from non-small cell lung cancer tissue.
Composite spectral mixing images from multiplex immunofluorescence (mIF; 20× magnification, scale bars represent 50 µm on each image) showing colored marker co-expression for (A) panel 1, (C) panel 2, (E) panel 3, (G) panel 4, and (I) panel 5. Color-coded UMAP visualizations show cell types identified by mIF panels: (B) 13 major cell types identified in panel 1, (D) 14 major cell types identified in panel 2, (F) 27 major cell types identified in panel 3, (H) 25 major cell types identified in panel 4, and (J) 12 major cell types identified in panel 5. Data from 225 samples was used. Experiments and quantifications related to the presented results were conducted once. mIF images were generated using Vectra-Polaris 1.0.13 scanner system and InForm 2.4.8 image analysis software (Akoya Biosciences), and UMAP visualizations were generated using the markers from each mIF panel by Python v.3.8.9. (Source data is provided as a source data file).
Fig. 3. Nearest neighbor distance G function…
Fig. 3. Nearest neighbor distance G function and theoretical Poisson curve score graphs showing different cellular patterns of distance from cytokeratin + cells (malignant cells) to CD3 + T-cells, and heat map representing distance patterns by histologic type.
A, B Representative example of the scoring system across tissue specimens and threshold to be considered part of the mixed (A) or unmixed (B) pattern. Graphs represent the scoring system (left), point pattern distributions related to the major T-cell population (middle), and G function and theoretical Poisson curve area (right). Composite spectral mixing images from multiplex immunofluorescence (mIF; 20× magnification, scale bars represent 50 µm on each image) showing a representative image of mixed pattern of macrophages PD-L1 expression (C) and PD-1+ antigen experience T-cells (D), in the bottom inside detail of the pattern (mIF; 40× magnification, scale bars represent 20 µm on each image). Unmixed pattern of CD8 + cytotoxic T-cells (E) and in their bottom inside detail of the pattern (mIF; 40× magnification, scale bars represent 20 µm on each image). F Model interaction based on the G function and theoretical Poisson curve score shows two groups of interaction between the most critical cell phenotypes observed and malignant cells. Cell phenotypes with a score of −10 to 10 were characterized as having a mixed/heterogeneous pattern indicating more interaction with malignant cells, and cell phenotypes with a score of >10 were characterized as having an unmixed/clustering pattern indicating less interaction with malignant cells. G Median distance heat map representing the 27 most common tumor-associated immune cells near malignant cells (CK + ) across the multiplex immunofluorescence panels in adenocarcinomas (ADCs = 142 samples) and squamous cell carcinomas (SCCs = 83), data from 225 samples was used. Experiments and quantifications related to the presented results were conducted once. Graphs and heat map were generated using R studio software version 3.6.1. mIF images were generated using Vectra-Polaris 1.0.13 scanner system and InForm 2.4.8 image analysis software (Akoya Biosciences). (Source data is provided as a source data file).
Fig. 4. Violin plots showing associations between…
Fig. 4. Violin plots showing associations between patterns of immune cell distribution and clinicopathologic features.
Significant cellular distribution scoring patterns between immune cells and malignant cells are shown by (A) smoker status, (B) tumor size, and (C) KRAS mutation status for lung adenocarcinoma (n = 142) specimens. D Significant cellular distribution scoring patterns between malignant cells and immune cells are shown by tumor size for lung squamous cell carcinoma (n = 83) specimens. Violin plots showing the median bar value, lower adjacent value and outside points. Kruskal–Wallis test was used in AD comparisons between groups. Data from 225 samples was used. Graphs were generated using GraphPad Prism v.9.0.0 using the 26 cell phenotypes distribution patterns and the relevant clinicopathologic information using un-adjusted P-values. (Source data is provided as a source data file).
Fig. 5. Violin plots showing the significant…
Fig. 5. Violin plots showing the significant associations between distances of immune cell populations from malignant cells and clinicopathologic features.
Significant differences in distance between malignant cells and immune cells are shown by (A) smoker status, (B) tumor size, (C) final stage, and (D) mutation status in lung adenocarcinoma (n = 142) specimens. E Significant differences in distances between malignant cells and immune cells by tumor size are shown for lung squamous cell carcinoma (n = 83) specimens. Violin plots showing the median bar value, lower adjacent value and outside points. Kruskal-Wallis test was used in A to E comparisons between groups. Data from 225 samples was used. Graphs were generated using GraphPad Prism v.9.0.0 using the 128 measurements from malignant cells to different tumor-associated immune cells (TAICs) and between important TAICs and the relevant clinicopathologic information using un-adjusted P-values. (Source data is provided as a source data file).
Fig. 6. Kaplan–Meier analysis of overall survival…
Fig. 6. Kaplan–Meier analysis of overall survival (OS) by cellular patterns of distribution and distance from malignant cells to various immune cell subpopulations.
A Composite spectral mixing images from a detail of multiplex immunofluorescence (mIF; 40× magnification scale bars represent 50 µm on each image) and illustration of the two different patterns of distribution, mixed and unmixed. B Composite spectral mixing images from a detail of multiplex immunofluorescence (mIF; 40× magnification, scale bars represent 50 µm on each image) and illustration of the distance metrics from malignant cells to different immune cells. (CG) Kaplan-Meier OS curves. Red lines indicate mixed pattern or close (≤median) distances between malignant cells and various cell phenotypes, and blue lines indicate unmixed pattern or long (>median) distances between malignant cells and various cell phenotypes. C Patients with CD66b + granulocytes (PMNs) with a mixed pattern had better OS than those with an unmixed pattern in lung adenocarcinoma specimens. Close (≤median) distances from malignant cells to (D) CD3 + CD8 + cytotoxic T-cells, (E) CD3 + CD8 + GZB + activated cytotoxic T-cells, and (F) CD68 + macrophages and long (>median) distances from malignant cells to B7-H3 + T-cells were associated with better OS in lung adenocarcinoma specimens using un-adjusted P-values. Data from 225 samples was used. Experiments and quantifications related to the presented results were conducted once. The images were generated using the Vectra-Polaris 1.0.13 scanner system and InForm 2.4.8 image analysis software (Akoya Biosciences), and Kaplan–Meier curves and log-rank test were used and generated by the R studio software version 3.6.0. using the distribution patterns and distances from the 26-cell phenotypes with un-adjusted P-values. (Source data is provided as a source data file).

References

    1. Zappa C, Mousa SA. Non-small cell lung cancer: current treatment and future advances. Transl. Lung Cancer Res. 2016;5:288–300. doi: 10.21037/tlcr.2016.06.07.
    1. Pirker R. Adjuvant chemotherapy in patients with completely resected non-small cell lung cancer. Transl. Lung Cancer Res. 2014;3:305–310.
    1. MacLean M, et al. Outcomes of neoadjuvant and adjuvant chemotherapy in stage 2 and 3 non-small cell lung cancer: an analysis of the National Cancer Database. Oncotarget. 2018;9:24470–24479. doi: 10.18632/oncotarget.25327.
    1. Wang L, Wang W. Safety and efficacy of anaplastic lymphoma kinase tyrosine kinase inhibitors in nonsmall cell lung cancer (Review) Oncol. Rep. 2021;45:13–28.
    1. Gravbrot N, et al. Therapeutic monoclonal antibodies targeting immune checkpoints for the treatment of solid tumors. Antibodies (Basel). 2019;8:51. doi: 10.3390/antib8040051.
    1. Mielgo-Rubio X, Uribelarrea EA, Cortes LQ, Moyano MS. Immunotherapy in non-small cell lung cancer: Update and new insights. J. Clin. Transl. Res. 2021;7:1–21.
    1. Cascone T, et al. Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non-small cell lung cancer: the phase 2 randomized NEOSTAR trial. Nat. Med. 2021;27:504–514. doi: 10.1038/s41591-020-01224-2.
    1. Garon EB, et al. Five-year overall survival for patients with advanced nonsmall-cell lung cancer treated with pembrolizumab: results from the phase I KEYNOTE-001 study. J. Clin. Oncol. 2019;37:2518–2527. doi: 10.1200/JCO.19.00934.
    1. Horvath L, Thienpont B, Zhao L, Wolf D, Pircher A. Overcoming immunotherapy resistance in non-small cell lung cancer (NSCLC)—novel approaches and future outlook. Mol. Cancer. 2020;19:141. doi: 10.1186/s12943-020-01260-z.
    1. Zheng X, et al. Spatial density and distribution of tumor-associated macrophages predict survival in non-small Cell Lung Carcinoma. Cancer Res. 2020;80:4414–4425. doi: 10.1158/0008-5472.CAN-20-0069.
    1. Wortman JC, et al. Spatial distribution of B cells and lymphocyte clusters as a predictor of triple-negative breast cancer outcome. NPJ Breast Cancer. 2021;7:84. doi: 10.1038/s41523-021-00291-z.
    1. Wang PC, et al. The spatial distribution of immune cell subpopulations in hepatocellular carcinoma. Cancer Sci. 2022;113:423–431. doi: 10.1111/cas.15202.
    1. Ge Z, Wu S, Zhang Z, Ding S. Mechanism of tumor cells escaping from immune surveillance of NK cells. Immunopharmacol. Immunotoxicol. 2020;42:187–198. doi: 10.1080/08923973.2020.1742733.
    1. Tang XY, et al. Clinical research on the mechanisms underlying immune checkpoints and tumor metastasis. Front Oncol. 2021;11:693321. doi: 10.3389/fonc.2021.693321.
    1. Reuben A, et al. Comprehensive T cell repertoire characterization of non-small cell lung cancer. Nat. Commun. 2020;11:603. doi: 10.1038/s41467-019-14273-0.
    1. Parra ER, et al. Immuno-profiling and cellular spatial analysis using five immune oncology multiplex immunofluorescence panels for paraffin tumor tissue. Sci. Rep. 2021;11:8511. doi: 10.1038/s41598-021-88156-0.
    1. Li H, et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell. 2019;176:775–789. doi: 10.1016/j.cell.2018.11.043.
    1. Fridman WH, et al. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome. Nat. Rev. Clin. Oncol. 2022;19:441–457. doi: 10.1038/s41571-022-00619-z.
    1. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat. Rev. Immunol. 2010;10:490–500. doi: 10.1038/nri2785.
    1. Bull JA, et al. Combining multiple spatial statistics enhances the description of immune cell localisation within tumours. Sci. Rep. 2020;10:18624. doi: 10.1038/s41598-020-75180-9.
    1. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–444. doi: 10.1038/nature07205.
    1. Barua S, et al. Spatial interaction of tumor cells and regulatory T cells correlates with survival in non-small cell lung cancer. Lung Cancer. 2018;117:73–79. doi: 10.1016/j.lungcan.2018.01.022.
    1. Schulze AB, et al. Tumor infiltrating T cells influence prognosis in stage I-III non-small cell lung cancer. J. Thorac. Dis. 2020;12:1824–1842. doi: 10.21037/jtd-19-3414a.
    1. Ordner, J., Gutierrez Amezcua, J. M., Marcus, A., Shukla, P. S. Programmed death Ligand 1 (PD-L1) expression and CD8+ tumor-infiltrating lymphocyte-based tumor immune microenvironment classification in gynecologic carcinosarcoma: prognostic impact and implications for therapy. Int. J. Gynecol. Pathol. 10.1097/PGP.0000000000000890 (2022).
    1. Le Mercier I, Lines JL, Noelle RJ. Beyond CTLA-4 and PD-1, the generation Z of negative checkpoint regulators. Front Immunol. 2015;6:418. doi: 10.3389/fimmu.2015.00418.
    1. Picarda E, Ohaegbulam KC, Zang X. Molecular pathways: targeting B7-H3 (CD276) for human cancer immunotherapy. Clin. Cancer Res. 2016;22:3425–3431. doi: 10.1158/1078-0432.CCR-15-2428.
    1. Yu N, Li X, Zheng S, Li X. B7-H4’s role “beyond the tumor”. Inflammation. 2013;36:941–947. doi: 10.1007/s10753-013-9624-0.
    1. Massari F, Santoni M, Ciccarese C, Santini D. The immunocheckpoints in modern oncology: the next 15 years. Expert Opin. Biol. Ther. 2015;15:917–921. doi: 10.1517/14712598.2015.1035251.
    1. Linch SN, McNamara MJ, Redmond WL. OX40 agonists and combination immunotherapy: putting the pedal to the metal. Front Oncol. 2015;5:34. doi: 10.3389/fonc.2015.00034.
    1. Fu T, He Q, Sharma P. The ICOS/ICOSL pathway is required for optimal antitumor responses mediated by anti-CTLA-4 therapy. Cancer Res. 2011;71:5445–5454. doi: 10.1158/0008-5472.CAN-11-1138.
    1. Willoughby J, Griffiths J, Tews I, Cragg MS. OX40: Structure and function—what questions remain? Mol. Immunol. 2017;83:13–22. doi: 10.1016/j.molimm.2017.01.006.
    1. Parra ER, et al. Image analysis-based assessment of PD-L1 and tumor-associated immune cells density supports distinct intratumoral microenvironment groups in non-small cell lung carcinoma patients. Clin. Cancer Res. 2016;22:6278–6289. doi: 10.1158/1078-0432.CCR-15-2443.
    1. Parra ER, et al. Immunohistochemical and image analysis-based study shows that several immune checkpoints are co-expressed in non-small cell lung carcinoma tumors. J. Thorac. Oncol. 2018;13:779–791. doi: 10.1016/j.jtho.2018.03.002.
    1. Croft M, So T, Duan W, Soroosh P. The significance of OX40 and OX40L to T-cell biology and immune disease. Immunol. Rev. 2009;229:173–191. doi: 10.1111/j.1600-065X.2009.00766.x.
    1. Shi X, et al. PD-L1 expression in lung adenosquamous carcinomas compared with the more common variants of non-small cell lung cancer. Sci. Rep. 2017;7:46209. doi: 10.1038/srep46209.
    1. Xia B, Herbst RS. Immune checkpoint therapy for non-small-cell lung cancer: an update. Immunotherapy. 2016;8:279–298. doi: 10.2217/imt.15.123.
    1. Shen M, Ren X. Highlights on immune checkpoint inhibitors in non-small cell lung cancer. Tumour Biol. 2017;39:1010428317695013. doi: 10.1177/1010428317695013.
    1. Schalper KA, et al. Differential expression and significance of PD-L1, IDO-1, and B7-H4 in human lung cancer. Clin. Cancer Res. 2017;23:370–378. doi: 10.1158/1078-0432.CCR-16-0150.
    1. Brahmer JR. Immune checkpoint blockade: the hope for immunotherapy as a treatment of lung cancer? Semin Oncol. 2014;41:126–132. doi: 10.1053/j.seminoncol.2013.12.014.
    1. Ceeraz S, Nowak EC, Burns CM, Noelle RJ. Immune checkpoint receptors in regulating immune reactivity in rheumatic disease. Arthritis Res Ther. 2014;16:469. doi: 10.1186/s13075-014-0469-1.
    1. Kawase A, et al. Differences between squamous cell carcinoma and adenocarcinoma of the lung: are adenocarcinoma and squamous cell carcinoma prognostically equal? Jpn J. Clin. Oncol. 2012;42:189–195. doi: 10.1093/jjco/hyr188.
    1. Ma J, Xu H, Wang S. Immunosuppressive role of myeloid-derived suppressor cells and therapeutic targeting in lung cancer. J. Immunol. Res. 2018;2018:6319649. doi: 10.1155/2018/6319649.
    1. Chesney JA, Mitchell RA, Yaddanapudi K. Myeloid-derived suppressor cells-a new therapeutic target to overcome resistance to cancer immunotherapy. J. Leukoc. Biol. 2017;102:727–740. doi: 10.1189/jlb.5VMR1116-458RRR.
    1. Lababede O, Meziane MA. The eighth edition of TNM staging of lung cancer: reference chart and diagrams. Oncologist. 2018;23:844–848. doi: 10.1634/theoncologist.2017-0659.
    1. Guo C, et al. Prognostic significance of combinations of RNA-dependent protein kinase and EphA2 biomarkers for NSCLC. J. Thorac. Oncol. 2013;8:301–308. doi: 10.1097/JTO.0b013e318282def7.
    1. Hernandez S, et al. Multiplex immunofluorescence tyramide signal amplification for immune cell profiling of paraffin-embedded tumor tissues. Front Mol. Biosci. 2021;8:667067. doi: 10.3389/fmolb.2021.667067.
    1. Parra ER. Methods to determine and analyze the cellular spatial distribution extracted from multiplex immunofluorescence data to understand the tumor microenvironment. Front Mol. Biosci. 2021;8:668340. doi: 10.3389/fmolb.2021.668340.
    1. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat Biotechnol. 37, 38–44 (2018).
    1. Baddeley, A., Rubak, E., Turner, R. Spatial Point Patterns: Methodology and Applications with R (Press CaHC, 2015).

Source: PubMed

3
Předplatit