Introduction
Neutrophils are the most abundant leukocytes present in the blood and function as a first-line defense against pathogens (Kobayashi and DeLeo,
2009; Hidalgo and Casanova-Acebes,
2021). Despite their usually short lifespan, it is now recognized that neutrophils participate in the establishment of chronic diseases such as cancer and support the spread of metastases to distant tissues (Gentles et al,
2015; Shaul and Fridlender,
2019; Coffelt et al,
2015; Yang et al,
2020). In non-small cell lung cancer (NSCLC), a high neutrophil-to-lymphocyte (NLR) ratio and high density of intra-tumoral neutrophils are prognostic for poor patient survival (Kargl et al,
2017; Faget et al,
2017). The tumor-promoting functions of neutrophils are many and versatile (Jaillon et al,
2020; Siwicki and Pittet,
2021). They can, for instance, stimulate tumor progression through the production of reactive oxygen species (ROS) (Canli et al,
2017; Zhong et al,
2021) or the secretion of angiogenic factors (Scapini et al,
2004; Nozawa et al,
2006; Kuang et al,
2011). In parallel, they can prevent the action of cytotoxic T cells through the secretion of arginase, ROS and nitric oxide (NO) (Munder et al
2006; Mensurado et al,
2018; Casbon et al,
2015), thus creating a favorable environment for tumor growth.
Strategies to deplete neutrophils in vivo, while providing important biological information in laboratory models (Boivin et al,
2020), are not translatable to humans as they could induce neutropenia, a major life-threatening condition where the body is unable to fight infections. For this reason, neutrophils have not been considered promising targets for treating cancer. There is now accumulating evidence, however, for molecular and phenotypic diversity of neutrophils in solid tumors. In a mouse model of lung cancer, analysis of the neutrophil compartment revealed that a subset of cells, marked by surface expression of the sialic acid-binding protein SiglecF, exhibit pro-tumor activity (Engblom et al,
2017; Pfirschke et al,
2020), suggesting that tumors can re-educate these cells for their benefit. We now know that there is great neutrophil heterogeneity in NSCLC (Zilionis et al,
2019a; Salcher et al,
2022), which should allow the identification of targeted therapies that disrupt selected tumor-associated neutrophils (TANs) without impacting the basal pool of circulating neutrophils, necessary for the host innate immune defense.
Physiologically, neutrophils patrol the organism for a short time before their death and clearance; homeostasis is maintained by daily replenishment of neutrophils from the bone marrow. This ensures an efficient set of pathogen-killing cells, while at the same time preventing the deleterious effects they could have on healthy tissue, such as in the establishment of chronic inflammation (Hidalgo and Casanova-Acebes,
2021b). Nevertheless, it has been shown that neutrophil death is delayed within inflamed tissues upon secretion of inflammatory mediators (Fox et al,
2010). Surprisingly, we and others have recently shown that SiglecF
+ neutrophils survive within the lung tumor microenvironment for more than 6–8 days (Ancey et al,
2021a; Pfirschke et al,
2020; Boivin et al,
2021), suggesting that these cells undergo profound intrinsic changes when exposed to the tumor microenvironment, enabling adaptation and prolonged survival. For example, a metabolic shift toward increased glucose usage, triggered by glucose transporter Glut1 upregulation in TANs, contributes to their ageing and tumor support (Ancey et al,
2021a; Bodac and Meylan,
2021).
Apoptosis is an extensively studied, regulated form of cell death that is triggered and modulated through two main axes known as the extrinsic and intrinsic pathways (Galluzzi et al,
2018). The latter is tightly regulated by the balance of pro- and anti-apoptotic proteins of the B cell lymphoma protein 2 (Bcl-2) family (Hatok and Racay,
2016), amongst which Bcl-xL has an anti-apoptotic role. It binds to the pro-apoptotic effectors Bak and Bax, preventing their clustering and the permeabilization of the mitochondrial outer membrane, which otherwise leads to the release of cytochrome c and downstream activation of caspases. In cancer, Bcl-xL was linked to tumor cell proliferation and resistance to chemotherapy, including in lung cancer (Stover et al,
2019; Shen et al,
2018). While the expression of Bcl-xL in neutrophils remained controversial for some time, recent studies show an induction of the protein in inflammatory conditions where neutrophil apoptosis is delayed, like sepsis (Guo et al,
2006) and rheumatoid arthritis (Carrington et al,
2021).
Here, we demonstrate that TANs’ death is prevented by GM-CSF-dependent Bcl-xL induction. A-1331852, an orally available, potent, and highly selective Bcl-xL inhibitor, reduces TAN survival and burden while sparing peripheral blood neutrophils in the KrasFrt-STOP-Frt-G12D/WT; p53Frt/Frt (hereafter called KP) mouse model of lung adenocarcinoma (LUAD), and its long-term use diminishes lung tumor growth.
Discussion
The importance of the tumor microenvironment (TME) in cancer progression is widely recognized (Hanahan,
2022). Processes such as matrix remodeling, new vessel formation and immune cell modulation contribute to tumor cell survival and proliferation. Immune cells need to adapt to the challenges imposed by the TME, such as insufficient nutrients and oxygen. These conditions eventually impact their survival and functionality and are often linked to the acquisition of pro-tumorigenic properties (Strauss et al,
2021; Ramel et al,
2021). For instance, positron emission tomography imaging has shown that myeloid cells in tumors consume high amounts of glucose (Reinfeld et al,
2021), and our previous research demonstrated that TANs rely on increased glucose metabolism to survive in the TME and differentiate into tumor-promoting SiglecF
+ cells (Ancey et al,
2021a). The prolonged survival of TANs may be a common characteristic in LUAD and other solid malignancies. In a mouse model of lung squamous cell carcinoma, for example, the presence of SiglecF
+ TANs (Mollaoglu et al,
2018) suggests an increased lifespan, which remains to be established. Similarly, TANs in human hepatocellular carcinoma (HCC) exhibit enhanced survival ex vivo, attributed to increased autophagy (Li et al,
2015).
In addition to metabolic adaptations, the survival and behavior of TANs are influenced by a variety of molecules secreted within the TME (Duits and de Visser,
2021). While assessing the modulation of neutrophil survival by tumor-secreted factors in vivo is difficult, several ex vivo studies demonstrated that it is stimulated upon incubation with supernatants of various tumor cell lines, including head and neck cancer (Wu et al,
2011; Dumitru et al,
2012). In the present research, we show that lung tumor-secreted GM-CSF enhances neutrophil survival. GM-CSF, which was reported to extend neutrophil survival in non-cancer contexts (Colotta et al,
1992; Lee et al,
1993), was also shown to regulate the functionality of neutrophils in different mouse models of cancer. For example, tumor-derived GM-CSF activates neutrophils and enhances PD-L1 expression via JAK-STAT3 in gastric cancer (Wang et al,
2017), while the same signaling pathway endows neutrophils with immunosuppressive activity in metastatic head and neck cancer (Pylaeva et al,
2022). In addition, breast tumor cells stimulate arginase-1 expression in myeloid cells through GM-CSF secretion, thus creating an immunosuppressive microenvironment (Su et al,
2021). Oncogenic KrasG12D was shown to stimulate the production of GM-CSF by pancreatic ductal cells, which led to the recruitment of CD8 T-cell-suppressing myeloid cells in vivo and subsequent cancer outgrowth (Pylayeva-Gupta et al,
2012). Interestingly, SiglecF
+ neutrophils were shown to be induced by GM-CSF in a mouse model of renal fibrosis (Ryu et al,
2022). Altogether, these studies and our findings position GM-CSF as a critical factor in promoting the survival of neutrophils and their phenotypic conversion toward tumor-supportive cells.
Beyond the specific role GM-CSF has on neutrophils as described in the present work, this growth factor has been shown to modulate cancer progression in opposite directions. CTLA-4 blockade-based anti-tumor immunity was augmented upon prior vaccination with autologous and irradiated tumor cells engineered to secrete GM-CSF, in patients suffering from metastatic melanoma or ovarian carcinoma (Hodi et al,
2008). However, in a mouse model of pancreatic ductal adenocarcinoma, the production of GM-CSF by tumor cells triggered the proliferation of c-Kit
+Lineage
neg splenocytes and their differentiation into immunosuppressive myeloid cells, which were then recruited to the tumors and enhanced their growth (Bayne et al,
2012). In our study, we identify that GM-CSF acts directly and locally in the tumor microenvironment to sustain TAN survival.
While BH3 mimetics were developed to trigger tumor cell death, leading to the clinical approval of Venetoclax against several hematological malignancies (Lasica and Anderson,
2021), our data suggest their repurposing against non-malignant, tumor-supportive innate cells. Of note, Navitoclax and A-1331852 were shown to prime NSCLC cell lines for apoptosis when combined with chemotherapeutic agents (Kim et al,
2017; Potter et al,
2021). Thus, combined administration of a Bcl-xL inhibitor and chemotherapy could counteract disease progression through direct tumor cell killing and indirect, TAN-dependent targeting. In our mouse model, known to be refractory to immune checkpoint blockade (Pfirschke et al,
2016), we failed to sensitize tumors to anti-PD-1 upon Bcl-xL blockade. One possible explanation is that T cells, which reside mainly at the tumor periphery and often in tertiary lymphoid-like structures in the KP model (DuPage et al,
2011; Joshi et al,
2015; Faget et al,
2017), did not infiltrate the tumor mass upon Bcl-xL blockade. Thus, additional stimuli should be employed to trigger tumor homing of effector T cells. The utilization of BH3 mimetics in cancer patients suffers limitations from secondary effects. While Navitoclax has shown promise as potential cancer therapy for solid tumors (Tse et al,
2008; Leverson et al,
2015), its clinical application is limited, in part due to induced neutropenia when combined with chemotherapies (Leverson et al,
2015). Comparatively, the use of Bcl-xL-selective inhibitors in rats did not result in neutropenia and did not suppress human ex vivo granulopoiesis (Leverson et al,
2015). Our results align with these findings, as none of the mice treated with A-1331852 became neutropenic, even after 3 weeks of treatment. One anticipated issue is, however, the on-target toxicity Bcl-xL blockade has on platelets (Debrincat et al,
2015), which was observed with the use of Navitoclax in the clinics (Ploumaki et al,
2023). Since then, the development of proteolysis-targeting chimeras (PROTACs) to prevent the uptake of Bcl-xL-specific mimetics by thrombocytes effectively attenuated platelet depletion (Khan et al,
2019; Negi and Voisin-Chiret,
2022). Because we demonstrated that small amounts of A-1331852 are sufficient to inhibit tumor cell-enhanced human neutrophil survival, low-dose treatments or the use of PROTACs could provide beneficial effects for the patients with reduced or clinically manageable thrombocytopenia.
There is accumulating evidence for tumor-promoting roles of neutrophils in cancer. Nevertheless, neutrophils can also participate in tumor elimination. In early-stage human lung cancer, neutrophils mediate the tumor-killing capacities of T cells (Eruslanov et al,
2014), and neutrophils from healthy blood donors appear to exhibit tumor cell-killing activity in vitro (Yan et al,
2014). Recently, a neutrophil response has been described for successful tumor control upon immunotherapy in lung, colon (Gungabeesoon et al,
2023) and melanoma tumor mouse models (Hirschhorn et al,
2023). Because patients under chemotherapy as frontline treatment are at risk of developing neutropenia (Blayney and Schwartzberg,
2022), G-CSF is often administered to foster neutrophil production and their egress from the bone marrow (Mehta et al,
2015), (Lambertini et al,
2015). Concurrent administration of G-CSF with radiotherapy was shown to enhance neutrophil tumor-killing ability in mice (Takeshima et al,
2016). Our results with combined G-CSF administration and Bcl-xL blockade suggest a qualitative change of the tumor-infiltrating neutrophil pool, whereby old, tumor-supportive TANs are replaced by younger, possibly tumor-antagonizing neutrophils. Thus, understanding and targeting specific pathways of TAN ageing could become an interesting option to disrupt tumor support and favor the anti-tumor activity of these cells.
Neutrophil targeting is difficult to accomplish in vivo, but the plasticity of these cells in cancer progression may hold the key to more precise and specific targeting. Our present work proposes a safe approach to selectively target pro-tumoral TANs while preserving the healthy pool of neutrophils, which can be harnessed for their capacities to fight cancer.
Methods
Mouse model
Kras
Frt-STOP-Frt-G12D/WT;Tp53
Frt/Frt (KP) mice, which were generated by crossing Kras
Frt-STOP-Frt-G12D/WT (RRID:IMSR_JAX:008653) (Young et al,
2011) and Tp53
Frt/Frt (RRID:IMSR_JAX:017767) (Lee et al,
2012) mice, were generously provided by D.G. Kirsch from Duke University Medical Center. These mice were bred in a mixed 129-C57BL/6 background. Mice were kept at 2–5 per cage and provided normal diet.
Tumor initiation and follow-up
To activate the oncogenic KrasG12D and delete Tp53, tumors were initiated in 12–14-week-old mice through the intratracheal administration of 107 plaque-forming units (PFU) of a commercially available adenoviral CMV-Flp vector (Ad5CMVFlpo, obtained from University of Iowa Viral Vector Core Facility) per mouse, to activate the oncogenic KrasG12D and induce the deletion of Tp53. To monitor the progression of tumors, mice were anesthetized using isoflurane and kept anesthetized throughout the scanning process. Lung images were captured using an X-Ray microtomography machine (µCT) (Quantum FX; PerkinElmer) with a voxel size of 50 μm, using retrospective respiratory gating. The volume of individual tumors was obtained using Osirix MD (Pixmeo, RRID:SCR_013618) and following the protocol described in bio-protocol.org/prep390.
Mouse treatments
Both genders were used for the experiments. Treatment with A-1331852 was initiated at 16 weeks after tumor initiation. Treatment exclusion criteria were based on the health score sheets. The Bcl-xL inhibitor A-1331852 (Cat#: HY-19741, MedChemExpress) was given at a dose of 25 mg/kg. The dual Bcl-2 and Bcl-xL inhibitor Navitoclax (ABT-263, Cat#: HY-10087, MedChemExpress) was given at a dose of 100 mg/kg and The Bcl-2-specific inhibitor Venetoclax (ABT-199, Cat#: HY-15531 MedChemExpress) at a dose of 50 mg/kg. All the compounds for in vivo treatments were formulated in 10% DMSO, 40% PEG-300, 5% Tween Tween-80 and 45% saline and were administered daily orally.
For the BrdU assay, 2 mg of freshly prepared BrdU (Merck, 10280879001) was prepared into 100 μL of PBS and injected intraperitoneally. For immunotherapy treatment, anti-mouse PD-1 (clone 29 F.1A12, Bio X Cell, BE0273, RRID:AB_2687796) was intraperitoneally injected at a dose of 200 μg/mouse three times a week for 2 weeks. For G-CSF treatment, recombinant mouse G-CSF (PeproTech, 250-05) was intraperitoneally injected daily at a dose of 10 μg for 3 weeks. Neutrophil depletion was performed using a double antibody approach with anti-Ly6G antibody (clone 1A8, #BP0075-1) injection followed by anti-rat Kappa immunoglobulin (clone MAR 18.5, #BE0122), according to a published method (Boivin et al,
2020). The antibodies and corresponding isotype controls (#BP0290 and #BP0089) were purchased from BioXCell.
Immunohistochemistry and immunofluorescence on KP mouse slides
After dissection, lungs with tumors were isolated, the lobes were separated, fixed with 3.7% formaldehyde solution (Sigma) overnight, and then paraffin-embedded. Blocks of tissues were cut into sections of 4 µm of thickness. For immunohistochemistry (IHC), slides were dewaxed and antigen retrieval was performed using 10 mM sodium citrate. Blocking was performed with 5% goat serum. The slides were then stained with primary antibodies overnight at 4 °C, washed and incubated with secondary antibodies for 1 h at room temperature. Antibody used for IHC is anti-Ki67 (1:100, ThermoFisher Scientific, MA5-14520). After washing, slides were incubated for 40 min with anti-rabbit Immpress horseradish peroxidase (HRP, Vector Laboratories, RRID:AB_2336529). The positive cells were then revealed using 3-3’-diaminobenzidine (DAB) substrate. Harris hematoxylin counterstain of the nuclei was subsequently performed.
For immunofluorescence, after dewaxing and antigen retrieval, slides were blocked with 5% goat serum and incubated with the following primary antibodies: anti-Bcl-xL (1:100, Abcam, ab32370) and anti-MPO (1:400, R&D SYSTEMS, Cat#: AF3667). Secondary antibodies are anti-rabbit Alexa 488 (1:500, ThermoFisher Scientific, A-21206) and anti-goat Alexa 568 (1:500, ThermoFisher Scientific, A-11057). For both IHC and immunofluorescence, tissue slides were scanned with the VS120-SL Olympus slide scanner at ×20 magnification, and quantification of positive cells on IHCs was performed using the QuPath software.
To assess the infiltration of different T-cell subpopulations in tumors, multiplexing (4plex) immunofluorescence was performed by the EPFL histology core facility using the fully automated Ventana Discovery ULTRA (Roche Diagnostics) and with the Ventana solutions. Briefly, paraffin sections were dewaxed, rehydrated and incubated sequentially with the following antibodies: anti-pan-cytokeratin (1:100, Novusbio, NBP600-579), anti-CD8 (1:100, Dako, M7103), anti-CD4 (1:100, Invitrogen Cat#: 14-0042-82) and anti-Foxp3 (1:50, Invitrogen Cat#: 14-5773). The slides were incubated with primary antibodies for one hour at 37 °C and then with a secondary antibody anti-rabbit ImmPRESSTM HRP (1:200, Vector Laboratories, Cat#: MP-7401). They were then sequentially revealed with the following TSA kits: the FAM (Roche Diagnostics, 07988150001), Red 610 (Roche Diagnostics, 07988176001), Rhodamine-6G (Roche Diagnostics, 07988168001) and Cyanine 5 (Roche Diagnostics, 07551215001) and counterstained with DAPI for nuclear staining.
Tumor and cell line-derived supernatant production
KP tumors were dissociated into single-cell suspensions as described previously (Faget et al,
2017). Briefly, individual tumors were digested by mechanical dissociation using the GentleMACS tissue dissociator (Miltenyi) coupled with enzymatic digestion using DNase I (0.02 mg/ml) and collagenase (1 mg/mL), resuspended in DMEM without FBS. Single-cell suspensions were then resuspended in complete DMEM (10% FBS and 1% PenStrep) and were plated at a density of 10
6 cells/mL. After 24 h of incubation, the supernatant (SN) was collected and passed through 0.22-µm filters. The supernatants were kept at −80 °C until use. For SN produced from SV2 cells, 10
6 cells/mL were cultured in DMEM, and SN was retrieved and filtered after 24 h.
GM-CSF measurement
GM-CSF presence in the supernatant derived from digested tumors was measured with the enzyme-linked immunosorbent assay ELISA MAX™ Standard Set Mouse GM-CSF (432201, Biolegend), following the manufacturer’s protocol.
Flow cytometry
Single-cell suspensions obtained from tumors with mechanical and enzymatic digestion were resuspended in FACS buffer (2%FCS and 2 mmol/L EDTA) with FcR-block (anti-CD16/32, BioLegend) and stained with antibodies (Appendix Table
S2), in darkness and for 20 min on ice. For intracellular staining of Bcl-xL, cells were fixed in BD Cytofix/Cytoperm (BD #554714) for 30 min on ice, then washed and incubated with Permeabilization buffer (Invitrogen #00–8333-56). For quantitative assessment of neutrophils in tumors, 15 µL of counting beads (CountBright
TM, Invitrogen, Cat#: C36950) were added to the sample before acquisition. For the experiment in Fig.
3E, acquisitions were performed with the full-spectrum analyzer Cytek Aurora (Cytek Biosciences). For the experiments, acquisitions were performed using the LSRII SORP (Becton Dickinson), a 5-laser and 18-detector analyzer at the EPFL Flow Cytometry Core Facility. Data analysis was performed using FlowJo (FlowJo LLC ©). Flow cytometry gating strategy is indicated in Appendix Fig.
S1.
Cell lines
The human cell line A549 (RRID:CVCL_0023) was purchased from ATCC and cultured in RPMI medium supplemented with 10% FBS. The murine cell lines SV2 and T5 were generated in our laboratory from a single KrasLox-STOP-Lox-G12D/WT; p53Flox/Flox and a KrasFrt-STOP-Frt-G12D/WT; p53Frt/Frt lung tumor, from a male and a female mouse, respectively. Briefly, tumors were digested into single-cell suspensions and cultured for 25 passages before experimentation in DMEM supplemented with 10% FBS and 1% PenStrep. Cells were cultured at 37 °C with 5% CO2. Mycoplasma tests were performed to ensure mycoplasma-free cell cultures.
Tumor cell viability assay and clonogenic assay
SV2 lung tumor cells were seeded at 3000 cells per well in 96-well plates and treated with A-1331852 diluted in tenfold steps (range) and cell viability was measured using PrestoBlue (ThermoFisher Scientific, Cat#: A13261) after 48 h and 72 h of incubation. To test the proliferative capacity of tumor cells, a clonogenic assay was performed by incubating 100, 200, or 400 single SV2 cells into six-well plates with increasing doses of A-1331852 and letting them grow for 1 week. Incubation with crystal violet solution enabled to stain for the colonies formed, which were then counted manually.
Mouse neutrophil isolation and survival assay
Neutrophils from different tissues were isolated using anti-Ly6G MicroBeads UltraPure magnetic beads (clone REA526, Miltenyi Biotec, 130-120-337) according to the manufacturer’s instructions. For bone marrow neutrophils, bone marrow was collected from femurs and tibia by flushing of the bone with PBS and filtered through a 40-µm filter. For healthy lung neutrophils, lungs from healthy mice were prepared the same way as lung tumors. For the survival assays, freshly isolated bone marrow neutrophils (105 cells) were incubated in 96-well plates and in 200 µL of tumor or SV2 cell-derived SN for 24 h. Neutralization of GM-CSF was done by pre-incubation of the SN with 10 ng/mL of anti-GM-CSF (PreproTech, Cat#500-P65) before adding it to the neutrophils. For the survival assay with GM-CSF, neutrophils were incubated with 10 ng/mL of recombined murine GM-CSF (PreproTech, Cat#315-03). For the experiments related to pathways inhibitors, Stattic (STAT3 inhibitor, MedChemExpress, Cat#: HY-13818), Ruxolitinib (JAK1/2 inhibitor, MedChemExpress, Cat#: HY-50856), MLN120B (IKKß inhibitor, MedChemExpress, Cat#: HY-15473) and LY294002 (PI3K inhibitor, MedChemExpress, Cat#: HY-10108) were resuspended in DMSO and diluted at the concentrations indicated in the figure legends. TANs were isolated from single-cell tumor suspensions using the anti-Ly6G MicroBeads UltraPure magnetic beads. Isolated TANs were stained with anti-SiglecF-Pe-Vio615 antibody (Cat#: 130-112-172) for 15 min on ice, before incubation, to specifically determine the effect of A-1331852 on SiglecF+ and SiglecF- TANs. For viability determination of Ly6G- cells from KP tumors, TANs were isolated from single-cell tumor suspensions using anti-Ly6G positive selection with magnetic beads. The remaining Ly6G negative fraction was collected and incubated with 10 nM of A-1331852 for 18 h.
The viable cells were measured by flow cytometry using the LIVE/DEADTM fixable blue dead stain kit (Invitrogen, Cat#: L23105) staining for dead cell exclusion.
Detection of apoptotic neutrophils
Measurement of apoptotic neutrophils was done by double staining with Annexin V (FITC) and 7-AAD (detection kit, BioLegend, #640922) and analyzed by flow cytometry. TNF (Preprotech: Cat#: 315-01 A) was used at 5 ng/mL as a positive control for neutrophil apoptosis. Briefly, 100,000 neutrophils were resuspended in 100 µL of Annexin V binding buffer and 5 µL of Annexin V-FITC and 5 µL of 7-AAD were added. The samples were kept in the dark for 15 min at room temperature and analyzed by flow cytometry within one hour. Additionally, cleaved-caspase-3 (Asp175, Cell Signalling, rabbit mAb #9664) was detected by western blot.
RNA isolation and real-time PCR
TRIzol Reagent (Invitrogen, 15596018) was utilized to extract the total RNA following the manufacturer’s instructions. The High-Capacity cDNA Reverse Transcription Kit (ThermoFisher Scientific, 4368814) was employed to synthesize cDNAs from 1 μg total RNA. Real-time PCR was carried out using 5 ng of cDNA with the Taqman universal PCR master mix (ThermoFisher Scientific, 4324018) and the following Taqman probes: Bcl2l1: Mm00437783_m1; Bcl2a1: Mm03646861_mH; Bcl-2: Mm00477631_m1; Mcl1: Mm01257351_g1; SiglecF: Mm00523987_m1; Ifit3: Mm01704846_s1; Ifnb1: Mm00439552_s1; Irf3: Mm00516784_m1; Dxd58: Mm01216853_m1 and the gene expression level normalization was done with Rpl30: Mm01611464_g1 or with Hprt: Mm00446968_m1.
Mouse mRNA sequencing and human TANs single-cell sequencing
Human gene expression data
Five public transcriptome datasets (Appendix Table
S3) have been combined to assess the overall survival of LUAD patients. To ensure data consistency and allow meaningful comparisons, background subtraction and normalization were performed on each dataset using the robust multi-array average (RMA) method from the affy package (Bolstad et al,
2003). In addition, to address potential batch effects that might arise due to differences in data collection and processing protocols, we employed the ComBat method from the sva package (Johnson et al,
2007). This method utilizes an empirical Bayes framework to adjust for known batch effects, harmonizing the combined dataset. The median expression of
CSF2 was then used to stratify patients into high or low groups and their survival probability was compared using a log-rank test.
Western blot
Protein extracts from isolated neutrophils were obtained through lysis and sonication of the cells in RIPA buffer (20 mmol/L Tris pH8, 50 mmol/L NaCl, 0.5% sodium deoxycholate, 0.1% SDS, 1 mmol/L Na3VO4, protease inhibitor cocktail (Roche, 11836145001). Protein quantification was done using the Bradford assay. The following antibodies were used: anti-Bcl-xL (1:1000, Abcam, ab32370) and γ-tubulin (1:3000, Invitrogen, Cat#MA1-850).
Immunofluorescence on human LUAD samples
Briefly, the formalin-fixed paraffin-embedded tissue sections were deparaffinized, rehydrated, and were subjected to antigen retrieval in EnVision FLEX Target Retrieval High pH Solution (Tris/EDTA, pH9.0; Agilent) at 95 °C for 20 min using PT Link (Agilent Technologies, Santa Clara, CA, USA). The sections were then permeabilized with 0.1% Triton X-100 in PBS for 15 min and blocked with 5% V/V donkey serum (Jackson ImmunoResearch, 017-000-121). The primary antibodies, anti-MPO (R&D SYSTEMS, AF3667) and anti-Bcl-xL (Abcam, ab32370), were added for overnight incubation at 4 °C. The primary antibodies were washed with washing solution 0.05% Tween-20 in PBS followed by incubation with secondary antibodies, Alexa Fluor 488-conjugated Donkey anti-Goat (Jackson ImmunoResearch, 705-545-003) and Cy3-conjugated Donkey anti-Rabbit (Jackson ImmunoResearch, 711-165-152) diluted in donkey serum, for 1 h at room temperature. The secondary antibodies were washed and counterstained with DAPI for nuclear inspection. The slides were scanned using Axioscan7 (Carl Zeiss SA, Oberkochen, Germany) and double-positive cells were analyzed using QuPath’s (v 0.3.2) classifier.
Human blood neutrophil isolation and survival assay
Peripheral blood neutrophils were isolated from whole-blood samples of healthy donors. Neutrophils were isolated from the blood using a Polymorphprep (Progen, Cat# 1895) gradient and the remaining red blood cells were lysed with ACK-buffer. The purity was assessed to be >95% with Quick-Fix staining of CytoSpinned samples. The viability of neutrophils incubated with A549 cell line supernatant was assessed by flow cytometry using the LIVE/DEADTM fixable blue dead stain kit (Invitrogen, Cat#: L23105). The viability of human neutrophils upon treatment with A-1331852 was determined using PrestoBlue.
Statistics
The sample size for mouse experiments was based on a previous study (Faget et al,
2017). Before treatments, μCT scans to visualize tumors enabled to randomize mice. Tumor volume measurements were performed blindly. The figure legends contain information on the statistical details of the experiments, including the number of repeats performed and the statistical tests used. The normality of distribution was tested using the Kolmogorov–Smirnov Test. Multiple comparisons of normally distributed samples were carried out using ANOVA, and Student
t tests were used for two independent samples. For nonparametrically distributed samples, the Kruskal–Wallis ANOVA with the Bonferroni correction for multiple comparisons, and Mann–Whitney
U test for two independent samples. Statistical analysis was performed using Prism 9 software.
Study approval
All mouse experiments were performed with the permission of the Veterinary Authority of the Canton de Vaud, Switzerland, License number VD2931. The human blood samples used for fundamental research were anonymized prior to transmission by the Transfusion Interrégionale CRS, which waived the need for a cantonal ethics authorization. The human lung tumor sections were archived at the Institut Jules Bordet biobank and obtained with the agreement from the local ethics committee. The experiments on human material conformed to the principles set out in the WMA Declaration of Helsinki and the Department of Health and Human Services Belmont Report.
Graphics
Figures
1A,E,
3D,E,
4A,
5A,B,
EV3F,
EV4A, Appendix Fig.
S2A, and synopsis graphics were created with
BioRender.com.