Abstract
Background and objectives Immunotherapies targeting PD-1 and CTLA-4 are key components of the treatment of metastatic clear cell renal cell carcinoma (mccRCC). However, they have distinct safety profiles and resistance to treatment can occur. We assess soluble TIM-3 (sTIM-3) in the plasma of mccRCC patients as a potential theranostic biomarker, as well as its source and biological significance.
Methods We analyzed the association of sTIM-3 with overall survival (OS), tumor response, and common clinical and biological factors across two mccRCC cohorts treated with anti-PD-1 (n = 27), anti- PD-1 or anti-PD-1 + anti-CTLA-4 (n = 124). The origin and role of sTIM-3 are studied on tumor and blood samples, using multiplex immunohistochemistry and flow cytometry as well as a syngeneic tumor model with antitumor vaccination. We also reanalyzed publicly available single-cell transcriptomic (scRNAseq) data and mass cytometry data.
Key findings and limitations sTIM-3 is elevated in the plasma of patients with mccRCC and shows distinct associations with survival on anti-PD-1 vs anti-PD-1 + anti-CTLA-4. sTIM-3 is independent from other clinical and biological factors. Myeloid immune cells appear as the prominent source of sTIM-3, which may indicate their dysfunctional role in the antitumor immune response. Future investigations are warranted in patients treated with anti-PD-1 + antiangiogenic therapies. Further functional studies are needed to confirm its theranostic value and clarify its role in the immune response.
Conclusions and clinical implications sTIM-3 appears to be a promising biomarker for optimizing treatment strategies in ccRCC as well as a potential therapeutic target.
Introduction
Immune checkpoint inhibitors (ICI) were introduced in clear cell renal cell carcinoma (ccRCC) as second line in the metastatic setting (mccRCC) with the approval of nivolumab monotherapy, an antibody blocking the-programmed cell death protein-1 (PD-1) 1. Anti-PD-1 were later combined in the first line with ipilimumab, an anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) or antiangiogenic tyrosine kinase inhibitors (TKI) 2. The International Metastatic RCC Database Consortium (IDMC) score is the only validated theranostic biomarker for mccRCC and previously helped selecting nivolumab + ipilimumab (N+I) over sunitinib 3,4. No predictive biomarkers exist to discern patients who may achieve maximal efficacy with N+I rather than other anti-PD-1-based regimens.
T cell immunoglobulin and mucin domain-containing molecule 3 (TIM-3) is an immunomodulatory transmembrane protein classically ascribed as exhaustion marker on T cells, conferring them dysfunctionality 5. In ccRCC, TIM-3-expressing T cells have been associated with a poor prognosis 6,7 and resistance to anti-PD-1 as monotherapy 8,9 or combined with antiangiogenics 10. Clinical trials targeting TIM-3 in mccRCC are underway 11,12. Soluble isoforms of TIM-3 (sTIM-3) are engendered through proteolytic cleavage 13,14. Blood-based biomarkers possess the advantages of ready accessibility and proximity to baseline measurements. We assessed whether plasma sTIM-3 is associated with resistance of mccRCC to ICI, given the detrimental value of TIM-3 expression in the TME 15 and sTIM-3 association with prognosis regardless of treatment 16.
Here we demonstrate acute sTIM-3 increase induced via antitumor immunization in mice. We assess chronic plasma sTIM-3 elevation in mccRCC patients, its association with outcome under ICI. We investigate sources of sTIM-3 with analyses of tumor specimens and peripheral blood mononuclear cells (PBMC) from mccRCC patients.
Material and methods
Patient cohorts and sample collection
ICI-treated mccRCC: Colcheckpoint (nivolumab monotherapy after prior antiangiogenic) and BIONIKK (first line nivolumab monotherapy or N+I 17) independent cohorts approved by the French Health authorities and ethics committee [CPP Ile-de-France 8 (ref.16.10.69) and CPP Ouest I SI CNRIPH n18.11.21.67518 respectively]. Healthy adult human samples were drawn from blood donation. Plasma and PBMC were collected just before ICI initiation for mccRCC. Multiplex immunohistochemistry (IHC) was performed on formalin-fixed, paraffin-embedded (FFPE) primary tumors. Participants provided informed written consent.
Mouse models
TC-1 cells were obtained from TC Wu’s laboratory (John Hopkins Hospital) and injected into anesthetized animals. Mice were vaccinated with 200µg G15F peptide from the HPV-16 E7 protein (Genosphere Biotechnologies) and 2µg of α-galactosyl-ceramide (α-Gal-Cer) adjuvant (Funakoshi Co.).
CD8+ T cells depletion was done in vivo by intraperitonel injections of a depleting antibody (100µg/100µl anti-mouse CD8α rat IgG2bκ, clone 2.43 InVivoMab™, BioXCell).
Experiments were conducted on 8-week-old female C57BL/6J mice (Janvier Labs) and approved by the ethics committee of the University Paris Cité (CEEA34).
sTIM-3 quantification
sTIM-3 was quantified in human plasma samples using the Luminex® Multiplex ProcartaPlexTM Human Immuno-Oncology Checkpoint Marker Panel 1 (14-Plex) kit (Thermofisher®), following the manufacturer’s instructions. Measures were acquired on a BioPlex-200 (BioRad®).
sTIM-3 was quantified in mouse serum by ELISA (Mouse Tim-3 SimpleStep ELISA® kit - ab255721 Abcam®), absorbance was measured on a Spectrostar microplate reader and converted to concentrations using the BMG Labtech software.
Multiplex IHC
FFPE slides were incubated with specific antibodies (details in Supp.Table 2.A) and stained in a BOND- RX automate (Leica®) using OpalTM secondary antibodies and fluorescent reagents. Specificity of TIM-3 was confirmed on additional ccRCC samples with the corresponding isotype control. Images were acquired in a Vectra Polaris scanner (Akoya/PerkinElmer) and analyzed with HALOTM (v.3.6.4, IndicaLab®).
Flow cytometry
PBMC were isolated by ficollTM density gradient centrifugation and frozen. PBMC were thawed, marked for viability (Zombie NIR, BioLegend®) and stained with antibodies against human markers (details in Supp.Table 2.B). Acquisition was performed on a Navios 10 colors cytometer (Beckman Coulter®), analyses were performed in Kaluza® (v 2.1). Thresholds for TIM-3 and CD14 positivity were set with corresponding control isotypes.
Single-cell RNA sequencing (scRNAseq) data analyses
ScRNAseq data from ccRCC and non-cancerous human kidneys were obtained from publicly available datasets 18–22 and analyzed in R (v.4.1.1) using Seurat (v.5.0.3). Cells were annotated through SingleR (v.1.8.1) and manually. Differential gene expression (DEG) and Gene set enrichment (GSEA) analyses were performed using MAST (v.1.20.0) and enrichR (v.3.2). Pseudobulk analysis of scRNAseq data was performed with DESeq2 (v.1.34.0) on aggregated transcripts counts.
Statistical analysis
Comparisons were performed for quantitative continuous variables using Mann-Whitney U (a.k.a. Wilcoxon ranks sum), Kruskal–Wallis, paired Wilcoxon, Friedman and Student’s t tests. Overall survival (OS) probabilities were estimated by the Kaplan-Meier method, differences were quantified by Cox hazard ratios (HR) and tested using the log-rank. Two-tailed p-values <0.05 were considered statistically significant. Analyzes were performed using the R software (v.4.3.2; R Foundation for Statistical Computing, Vienna, Austria).
Results
Plasma sTIM-3 is elevated in mccRCC
Plasma sTIM-3 levels were increased in treatment-naive mccRCC patients (median = 110.33 pg/L, IQR 65.36 to 151.10) compared to healthy adults (median = 46.10 pg/L, IQR 40.81 to 50.42, p < 10-3) (Fig.1.A).
To assess sTIM-3 at the early stage of the tumor response, we used a syngeneic TC-1 mouse tumor model, spontaneously poorly immunogenic but for which a specific anti-tumor immune response is elicited via vaccination. Control mice solely inoculated with TC-1 cells (day 0 – D0) showed no significant variation of plasma sTIM-3 18 days (D18) after the graft (D0 to D18 mean variation = -29.7 pg/L), whereas vaccinated mice exhibited a significant increase (D0 to D18 mean variation = +735.2 pg/L, p = 0.015 control versus vaccine) (Supp. Fig.2), suggesting a prominent role for the immune response in the production of sTIM-3.
sTIM-3 is associated with OS in mccRCC treated with ICI
High baseline sTIM-3 (above the median within each cohort) was associated with poor OS on nivolumab monotherapy in both the Colcheckpoint (n = 27, HR for death sTIM-3-high vs sTIM-3-low = 2.67, p = 0.03) and BIONIKK (n = 45, HR sTIM-3-high vs sTIM-3-low = 2.36, p = 0.04) cohorts (Fig.1.B & C).
There was no difference according to sTIM-3 stratification in patients from the BIONIKK trial treated with N+I (Supp.Fig.3.A). Furthermore, we found that sTIM-3-low participants of the nivolumab monotherapy and N+I arms had comparable OS, whereas for sTIM-3-high participants, N+I was significantly superior to nivolumab (Supp.Fig.3.B). To verify that results in the N+I group were not confounded by IMDC scores (Supp.Table 1.C), a Cox model including IMDC and sTIM-3 categories was constructed. Still, the adjusted analysis evidenced no relationship between sTIM-3 category and OS under N+I (HR high vs low = 1.0, 95% IC 0.40 - 2.72) (Supp.Fig.4).
This suggests that sTIM-3 can distinguish mccRCC patients likely to achieve prolonged survival under anti-PD-1 monotherapy, while the anti-PD-1 + anti-CTLA4 combination retains efficacy in sTIM-3-high patients.
sTIM-3 is independent from clinical and biological prognostic markers
IMDC categorization was not associated with sTIM-3 (Fig.2.A) and sTIM-3 was neither correlated with systemic inflammation markers included in the score (neutrophils and platelets counts, decreased hemoglobin) nor with other biomarkers (Suppl.Fig.5 and Supp.Fig.7).
Unlike CRP (Supp.Fig.6), sTIM-3 was not associated with tumor burden proxies (number of metastatic sites or presence of the primary tumor, Fig.2.B&C). This suggests another source than tumor cells for sTIM-3, consistent with the mouse model, and that sTim-3 does not simply reflect a higher tumor burden or systemic inflammation.
Plasma sTIM-3 is likely produced by myeloid cells in ccRCC
A. In situ multiplex IHC of ccRCC tumors
We assessed the expression of TIM-3 by tumor cells - pancytokeratin and/or PAX8 positive - and tested its association with plasma sTIM-3 in a subset of BIONIKK participants (n = 22) (Fig.3.A).
There was a high intra-tumor and inter-samples variability of TIM-3 staining of tumor cells (Fig.3.B), with a median of 0.34% (IQR 0.05% to 5.61%) positive tumor cells. Tumors with >1% TIM-3 positive tumor cells were classified as TIM-3 positive. TIM-3 positivity was neither associated with higher levels of plasma sTIM-3 (Fig.3.C), nor with survival under nivolumab monotherapy (Supp.Fig.8).
TIM-3-positive non-tumor cells did not appear to be correlated to plasma sTIM-3 (Spearman correlation = -0.26, p = 0.25, Supp.Fig.9), leading us to investigate circulating immune cells.
B. scRNAseq of ccRCC tumors
The main hypothesis for the production of sTIM-3 in human is its shedding from double-positive HAVCR2+ADAM+ cells, co-expressing HAVCR2 (encoding TIM-3) and one or both of the metalloproteinases (ADAM10 and ADAM17) for which a cleaving activity of membrane TIM-3 is reported 13,14. We quantified the expression of these genes in several scRNAseq datasets, results are shown on Obradovic et al. dataset (Fig.4).
Although some CD8+ and NK lymphocytes clusters had high transcription levels of HAVCR2, myeloid clusters (Supp.Fig.10,11 & 12) were identified as the most enriched lineage in HAVCR2+ADAM+ cells, with even distributions of the 3 different double-positive types (Fig.7.A & B; Supp.Fig.10 & 12). HAVCR2 expression was detected in renal tissue clusters but they contained marginal proportions of HAVCR2+ADAM+ cells (Fig.5.A & B). Consistent findings were reproduced on 3 other ccRCC scRNAseq datasets.
HAVCR2+ADAM+ enriched monocytes clusters were common to paired tumors and PBMC in a dataset of 3 ccRCC patients 20. Consistent with our flow cytometry data, HAVCR2 expression was mainly found on monocytes and NK clusters in PBMC (Supp.Fig.13).
Principal component analysis (PCA) on the top 200 most variable genes showed a clear separation of double-positive and non-double-positive pseudobulk samples, suggesting a distinct transcriptomic state for HAVCR2+ADAM+ myeloid cells (Supp.Fig.13).
Among top DEG, the putative pro-tumor markers TREM2 and APOE were found increased in HAVCR2+ADAM+ myeloid cells (Khantakova, Brioschi, et Molgora 2022; Bancaro et al. 2023), while NLRP3 was decreased, which may be linked to decreased APC functionality (Dixon et al. 2021). GSEA suggested a downregulation of proteins synthesis in HAVCR2+ADAM+ myeloid cells (Supp.Fig.14), as wells as cellular respiration (oxidative phosphorylation).
These results favor myeloid cells as major source of sTIM3 in mccRCC. sTIM-3 could reflect the presence of dysfunctional HAVCR2+ADAM+ myeloid cells, with globally decreased anabolic activity 23.
C. PBMC cytometry of ccRCC patients
We quantified TIM-3-positive (TIM3+) cells in PBMC from mccRCC patients (Fig.5.A), their median proportion was 22.7% (IQR 14.9% to 27.3%) (Fig.5.B). CD3-TIM3+ myeloid cells were the most common type of TIM3+ PBMC (median 61.9% of TIM3+ PBMC), followed by CD3-TIM3+ lymphoid cells. Surprisingly, T cells (CD3+TIM3+) accounted for a minority of TIM3+ PBMC (Fig.5.C). Given their intermediate CD4 positivity (Supp.Fig.15) and that their proportion among TIM3+ PBMC was the same as CD14+TIM3+ PBMC on a separate panel (Supp.Fig.16), we hypothesized that most CD3-TIM3+ myeloid cells had a monocytic origin. CD3-TIM3+ lymphoid PBMC were most likely NK cells, given their CD3-CD8low phenotype and that B cells (quantified with CD19 on another panel) were not found in TIM3+ PBMC. These results are consistent with data from the literature on peripheral blood cells from healthy donors, showing TIM-3 detection on a high proportion of monocytes, on a moderate proportion of NK cells, and low or absent detection on T cells and granulocytes (Kikushige et al. 2010; Möller-Hackbarth et al. 2013).
D. Lymphocytes depletion in TC-1 syngeneic mouse model
Serum sTIM-3 increase upon antitumor vaccination was found at earlier time-points in a second experiment with the TC-1 mouse model (Supp.Fig.20.A). Given our initial hypothesis of sTIM-3 release by T cells following vaccination, control vaccinated mice were compared to mice vaccinated and depleted in CD8+ T cells with an anti-CD8α depleting monoclonal antibody. Effective depletion was confirmed by the quasi-disappearance of T cells in tumors from anti-CD8-treated mice analyzed by flow cytometry (not shown). However sTIM-3 levels were not significantly modified by the depletion (Supp.Fig.20.B). The timing of sTIM-3 increase and its sustainability in spite of CD8+ T cells elimination favor the hypothesis of the early release of sTIM-3 by innate immune cells, possibly myeloid cells.
Discussion
Plasma sTIM-3 is a promising biomarker associated with ICI efficacy in mccRCC, with differential effects on nivolumab versus N+I and, independent from common clinical or inflammatory markers. An association with survival of patients under current anti-PD-1 + TKI regimens is also plausible and will be investigated in future research. If a differential effect on N+I compared to anti-PD-1 + TKI is confirmed, this could make sTIM-3 a theranostic biomarker. Defining a sTIM-3-high population for which the addition of the anti-CTLA-4 is needed to overcome the resistance to anti-PD-1 would facilitate the indication of N+I, for which the benefit-risk balance is delicate. sTIM-3 is also promising for other cancers where anti-PD-1 monotherapies are challenged by combinations, without strong decisional criteria (e.g. melanoma, lung cancer).
TIM-3 is known for its inhibitory role at the membrane of T cells, its shedding into sTIM-3 by activated CD8+ T cells was reported 14. Unexpectedly, our study suggests myeloid cells as major source of sTIM- 3 in mccRCC. TIM-3 was found on tumor-associated macrophages (TAM) and associated with PFS, in another cohort of ccRCC, and induced on monocytes co-cultured with RCC cell-lines. However, the patients were not treated with ICI and monocytes were obtained from healthy volunteers 24. LPS- activated monocytes from healthy donors and myeloid leukemia cells shed sTIM-3 upon activation of ADAMs 13,25. We show in mccRCC that myeloid cells represent the majority of TIM-3-positive PBMC and the most enriched lineage in HAVCR2-expressing cells within the TME. A low proportion of TIM-3- positive lymphoid cells in cytometry could be due to shedding of the marker 13,14. Nevertheless, scRNAseq shows more co-expression of ADAM10/17 in myeloid cells, thus, a bias on membrane TIM-3 detection is more likely to occur for them. TIM-3 was detected at the apical border of some healthy and cancerous proximal tubule cells in IHC (Fig.5). One hypothesis is that sTIM-3 undergoes glomerular filtration with re-uptake of the antigen by these cells26. In our analysis of Chevrier et al. mass cytometry dataset (Supp.Fig.15), TIM-3 appeared fairly expressed by immune cells but with null median signal intensities for non-immune cells in most samples (Supp.Fig.15.B&C). Finally, granulocytes are quasi- absent from PBMC and from the scRNAseq datasets but their role as source of sTIM-3 cannot be excluded.
A secreted sTIM-3 isoform comes from alternatively splicing of murine TIM-3 mRNA 27. Nonetheless, mouse embryonic fibroblasts shed transfected human TIM-3 and murine TIM-2 (TIM family) via ADAM10/17, supporting the relevance of studying sTIM-3 in this species 13,28.
Regarding the biological meaning of sTIM-3 in mccRCC patients, there are two (non-exclusive) possibilities: sTIM-3 could have per se a pro-tumor effect 25,29. On the other hand, plasma sTIM-3 could be a marker of pro-tumor processes or cell types, such as ADAM10/17 activity in the TME 30,31. We show that HAVCR2+ADAM+ myeloid cells have an increased transcription of TREM2, which is proposed as a pro-tumor macrophage marker 32. Vanmeerbeek et al. have identified TAM co-expressing TIM-3 and VSIR, associated with resistance to ICI, HAVCR2 was upregulated in myeloid cells from ICI-unresponsive patients33. The release of sTIM-3 may also reflect a deleterious state of over-activation of myeloid APC, which are required to prime T cells and sustain the response to ICI 34–36. GSEA suggest that HAVCR2+ADAM+ myeloid APC have globally impaired transcriptional and translational activities (Supp.Fig.13) 37,23.
Understanding the biological meaning of plasma sTIM-3 will be crucial for TIM-3-targeted therapies. According to the relevant hypothesis, strategies to treat sTIM-3 high patients would be either to block the putative deleterious action of sTIM-3, to inhibit ADAMs’ activities, to deplete TIM-3+ pro-tumor myeloid cells or to restore the function of paralyzed APC.
Conclusion
Plasma sTIM-3 is elevated in mccRCC and independent from clinical and inflammatory prognostic markers. It is a promising blood-based biomarker associated with ICI efficacy in this setting.
The immune myeloid compartment is the predominant candidate as source of plasma sTIM-3 in mccRCC.
Further functional studies will precise the biological role of sTIM-3 in mccRCC and ensuing therapeutic targets.
Supplementary figures
Supplementary tables
Funding
With financial support from ITMO Cancer of Aviesan within the framework of the 2021-2030 Cancer Control Strategy, on funds administered by Inserm. ITMO Cancer was not involved in the design and conduct of the study, management and analysis of the data, or approval of the manuscript.
Disclosure of interest
Ivan Pourmir certifies that all conflicts of interest, including specific financial interests and relationships and affiliations relevant to the subject matter or materials discussed in the manuscript (eg, employment/affiliation, grants or funding, consultancies, honoraria, stock ownership or options, expert testimony, royalties, or patents filed, received, or pending), are the following: Virginie Verkarre has received payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events and support for attending meetings and/or travel from MSD. Yann-Alexandre Vano has received consulting fees from BMS, Ipsen, Eisai, MSD, Pfizer; research grants from BMS, Ipsen. Stephane Oudard has received consulting fees, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events and support for attending meetings and/or travel from Pfizer, Novartis, Ipsen, Eisai, BMS, Merck; has participated in data safety monitoring board or advisory board from Roche, Ipsen, Eisai.
The remaining authors declare no potential conflict of interest with the present work.
Data sharing statement
Requests for anonymized patient data will be examined on an individual basis by relevant administrating committees of the Colcheckpoint and BIONIKK cohorts data. Processed data from animal experiments are available on request from the corresponding authors.
Acknowledgment
The authors thank Marine Sroussi, Aurélien De Reynies, Pierre Gestraud, Lorette Noiret and Aleksandar Obradovic for their discussions and advice on scRNAseq data analyzes; Stéphane Chevrier for his advice on mass cytometry data exploitation; Florian Da Silva and Chloé Broudin for their help on tumor samples logistics; Jean-Philippe Empana for his advice on survival models; Clémence Granier for inspiring scientific discussions.