Aby wyświetlić tę treść, wymagana jest subskrypcja JoVE. Zaloguj się lub rozpocznij bezpłatny okres próbny.
Method Article
* Wspomniani autorzy wnieśli do projektu równy wkład.
Studying tumor microenvironment may identify prognostic or predictive biomarkers of clinical response to immunotherapy. Presented here, is an innovative method based on in situ fluorescence multispectral imaging to analyze and count automatically various subpopulations of CD8+ T cells. This reproducible and reliable technique is suitable for large cohort analyses.
Immune cells are important components of the tumor microenvironment and influence tumor growth and evolution at all stages of carcinogenesis. Notably, it is now well established that the immune infiltrate in human tumors can correlate with prognosis and response to therapy. The analysis of the immune infiltrate in the tumor microenvironment has become a major challenge for the classification of patients and the response to treatment.
The co-expression of inhibitory receptors such as Program Cell Death Protein 1 (PD1; also known as CD279), Cytotoxic T Lymphocyte Associated Protein 4 (CTLA-4), T-Cell Immunoglobulin and Mucin Containing Protein-3 (Tim-3; also known as CD366), and Lymphocyte Activation Gene 3 (Lag-3; also known as CD223), is a hallmark of T cell exhaustion. We developed a multiparametric in situ immunofluorescence staining to identify and quantify at the cellular level the co-expression of these inhibitory receptors. On a retrospective series of frozen tissue of renal cell carcinomas (RCC), using a fluorescence multispectral imaging technology coupled with an image analysis software, it was found that co-expression of PD-1 and Tim-3 on tumor infiltrating CD8+ T cells is correlated with a poor prognosis in RCC. To our knowledge, this represents the first study demonstrating that this automated multiplex in situ technology may have some clinical relevance.
In the past few years, immunotherapy has emerged as a very promising treatment for many types of cancers, including RCC. Particularly, immunotherapy based on the inhibition of inhibitory checkpoints like PD-1 and CTLA-4 has been reported to be clinically effective1,2,3,4,5,6. Monoclonal antibodies against CTLA-4, PD-1, or Program Death Ligand 1 (PD-L1) are already approved in several cancers and lead to long lasting clinical responses in more than 20% of patients7. Nevertheless, not all patients are responders, the cost of the treatment is high, and these treatments are toxic, leading to potential serious autoimmune-like side effects. Therefore, the current challenge is to identify predictive markers to those new immunotherapies. The rate of mutations in the tumor, the expression of PD-L1, or the levels of intratumoral CD8+ T cell infiltration have been reported to correlate with clinical response. However, this association is still too weak to recommend the use of these clinical biomarkers in clinical practice except for the companion test for PD-L1 before the administration of Pembrolizumab in non-small cell lung cancer (NSCLC) patients8,9,1011,12. It has been demonstrated that the co-expression of many inhibitory receptors like PD-1, Tim-3, Lag-3, and CTLA-4, induces a cell exhaustion phenotype and resistance to therapy13,14,15. Since peripheral blood is not representative of the tumor microenvironment, it is of high interest to analyze the phenotypic features of the cells in situ. PD-1 and Tim-3 co-expressing T cells are known to be functionally impaired cells in several contexts13,16,17. In this study, the prognostic impact of the co-expression of the two inhibitory receptors PD-1 and Tim-3 on CD8+ T cells was assessed.
Up until now, studying the co-expression of multiple markers on tumor infiltrating lymphocytes (TILs) has mainly been performed by flow cytometry analysis, making it necessary to work on fresh tumors and therefore precluding retrospective analyses. With conventional in situ staining, only one staining at a time can be performed, and the characterization of the cell type that co-expresses the markers is not possible. For example, PD-L1 is expressed by many cell types of the tumoral microenvironment, making it difficult to define by conventional immunohistochemistry analysis which cells expressing PD-L1 are the more relevant for correlative studies. In this work, we developed an innovative in situ multiparametric immunofluorescence method with computer-counting to correlate the co-expression of PD-1 and Tim-3 by tumor infiltrating CD8+ T-cells with clinical outcomes in RCC. This technique has several advantages, including the possibility to analyze at a single cell level and multiple markers at the same time using a multispectral camera that can capture restricted intervals >10 nm through liquid crystal filters18. Moreover, the procedure is automatic which enables an inter-operator reproducibility and a shortened analysis compared to manual techniques19. In the cancer field, several studies reported convincing multiple stainings of immune molecules like PD-1, PD-L1, and CD8 in Merkel-cell carcinoma, lung cancer, and head and neck cancer20,21,22,23. The automated cell count is possible with training by the user (phenotyping step). The fluorescence is measured in the different cell compartments (nuclei, cytoplasm, and membrane).
Here, different subsets of tumor-infiltrating CD8+ T cells expressing PD-1 and/or Tim-3 in a large cohort of RCC were counted and the results were correlated with clinical gravity scores and survival parameters. It was also possible to analyze membrane fluorescence intensity at the cellular resolution with Mean Fluorescence Intensity (MFI) data like in cytometry. As far as we know, this represents the first study reporting prognostic results using this multispectral imaging based count technique.
Access restricted. Please log in or start a trial to view this content.
This study was conducted in accordance with the Declaration of Helsinki and was approved by the local ethics committee (CPP Ile de France nr. 2012-05-04). Informed consent was obtained from the participant included in the cohorts.
1. Tissue Material
2. In Situ Immunofluorescence Staining of TILs
3. Fluorescence Analysis and Automated Cell Count
Access restricted. Please log in or start a trial to view this content.
Using the general protocol described above, we aimed to quantify intratumoral CD8+ T cells co-expressing the inhibitory receptors PD-1 and Tim-3 in frozen tissues from patients with RCC, and to correlate the results with clinical outcomes25.
Optimization of the CD8/PD-1/Tim-3 Staining:
Different species of antibodies ...
Access restricted. Please log in or start a trial to view this content.
Modifications and Troubleshooting:
The tissue quality is an important parameter; it can be easily checked by hematoxylin and eosin coloration.
One advantage of the technique is the possibility of multiplexed stainings, but to avoid fluorophore spillover, it recommended to choose emission wavelengths with delta of 10 nm minimum. Here, because we had 4 stainings including DAPI, we chose to spread out the wavelengths (DAPI: 460 nm, AF488: 519 nm, Cyani...
Access restricted. Please log in or start a trial to view this content.
All authors have no conflict of interest to declare.
This work was supported by grants from Institut National du Cancer (INCA) (ET), Ligue contre le Cancer (ET), Université Sorbonne Paris Cité (ET), ANR (Selectimmunco) (ET), Labex Immuno-Oncology (ET), SIRIC CARPEM (CG, ET). EdG was funded by a fellowship of Fondation ARC. EV and CD were funded by a fellowship of APHP (bourse année recherche). ChB is funded by a fellowship of Université Sorbonne Paris Cité (contrat doctoral). The authors thank Bristol Myers Squibb for their funding in this project. The authors thank the department of Pathology of Hopital Européen Georges Pompidou and Necker (Laurianne Chambolle, Elodie Michel and Gisèle Legall). The authors thank the Histology platform of PARCC, Hopital Européen Georges Pompidou (Corinne Lesaffre).
Access restricted. Please log in or start a trial to view this content.
Name | Company | Catalog Number | Comments |
Vectra 3 Automated Quantitative Pathology Imaging | Perkin Elmer | CLS142338 | |
inForm cell analysis 2.1. | Perkin Elmer | CLS135781 | |
R software | https://www.r-project.org | ||
Dakopen delimiting pen | Dako | S2002 | |
Tris Buffer Salin TBS Tablets | Takara, Bio Inc. | TAKT9141Z | pH7.6 100 tablet |
Tris Buffer Salin Tween 20 TBS(+Tween20) | Takara, Bio Inc. | TAKT9142Z | pH 7.6 100 tablets |
Biotin blocking system | Dako | X0590 | Avidin 0.1% and Biotin 0.01% |
normal donkey serum | Jackson Immunoresearch | 017-000-001 | 5% vol./vol. concentration |
Fluoroshield with DAPI | Sigma-aldrich | F6057 | 1.5 µg/mL concentration |
Knittel glass coverslip | Knittel Gläser, | 100039 | 24x60 mm 100 cover slips |
Rabbit anti-CD8 Clone P17-V | novus | NBP1-79055 | use at 4µg/mL |
Mouse anti-PD-1 Clone NAT | abcam | ab52587 | use at 2 µg/mL |
Goat anti-Tim-3 | R&D | AF2365 | use at 3 µg/mL |
Rabbit anti-PD-L1 Clone SP142 | Roche | 7309457001 | use at 1 µg/mL |
Mouse AF647 labeled pan- Keratin Clone C11 | Cell Signalling | 4528 | use at 0.5 µg/mL |
Goat anti-human gal9 | R&D | AF2045 | use at 0.3 µg/mL |
Cyan 5 conjugated donkey anti-rabbit | Jackson Immunoresearch | 711-175-152 | use at 5 µg/mL |
Biotinylated F(ab’2) donkey anti-mouse IgG | Jackson Immunoresearch | 715-066-150 | use at 3 µg/mL |
Alexa Fluor488 conjugated donkey anti-goat IgG | abcam | ab150133 | use at 5 µg/mL |
Cy3 labeled streptavidin | Amersham | PA43001 | use at 3 µg/mL |
negative control mouse IgG1 | Dako | X0931 | use at 2 µg/mL |
IgG from goat serum | Sigma-aldrich | I5256 | use at 3 µg/mL |
IgG from rabbit serum | Sigma-aldrich | I5006 | use at 4µg/mL |
Access restricted. Please log in or start a trial to view this content.
Zapytaj o uprawnienia na użycie tekstu lub obrazów z tego artykułu JoVE
Zapytaj o uprawnieniaThis article has been published
Video Coming Soon
Copyright © 2025 MyJoVE Corporation. Wszelkie prawa zastrzeżone