Skip to main content
Erschienen in: Experimental Hematology & Oncology 1/2024

Open Access 01.12.2024 | Research

Targeting focal adhesion kinase boosts immune response in KRAS/LKB1 co-mutated lung adenocarcinoma via remodeling the tumor microenvironment

verfasst von: Meng Qiao, Fei Zhou, Xinyu Liu, Tao Jiang, Haowei Wang, Xuefei Li, Chao Zhao, Lei Cheng, Xiaoxia Chen, Shengxiang Ren, Zaiqi Wang, Caicun Zhou

Erschienen in: Experimental Hematology & Oncology | Ausgabe 1/2024

Abstract

Background

KRAS mutation is one of the most common oncogenic drivers in NSCLC, however, the response to immunotherapy is heterogeneous owing to the distinct co-occurring genomic alterations. KRAS/LKB1 co-mutated lung adenocarcinoma displays poor response to PD-1 blockade whereas the mechanism remains undetermined.

Methods

We explored the specific characteristics of tumor microenvironment (TME) in KL tumors using syngeneic KRASG12DLKB1−/− (KL) and KRASG12DTP53−/− (KP) lung cancer mouse models. The impact of focal adhesion kinase (FAK) inhibitor on KL lung tumors was investigated in vitro and in vivo through evaluation of both KL cell lines and KL lung cancer mouse models.

Results

We identified KL tumors as “immune-cold” tumors with excessive extracellular matrix (ECM) collagen deposition that formed a physical barrier to block the infiltration of CD8+T cells. Mechanistically, abundant activated cancer-associated fibroblasts (CAFs) resulted from FAK activation contributed to the formation of the unique TME of KL tumors. FAK inhibition with a small molecular inhibitor could remodel the TME by inhibiting CAFs activation, decreasing collagen deposition and further facilitating the infiltration of anti-tumor immune cells, including CD8+ T cells, DC cells and M1-like macrophages into tumors, hence, converting “immune-cold” KL tumors into “immune-hot” tumors. The combined FAK inhibitor and PD-1 blockade therapy synergistically retarded primary and metastatic tumor growth of KL tumors.

Conclusions

Our study identified FAK as a promising intervention target for KL tumors and provided basis for the combination of FAK inhibitor with PD-1 blockade in the management of KL lung cancers.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s40164-023-00471-6.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
NSCLC
Non-small cell lung cancer
TME
Tumor microenvironment
KL
KRASG12DLKB1/
KP
KRASG12DTP53/
LKB1
Liver kinase B1
FAK
Focal adhesion kinase
ECM
Extracellular matrix
CAF
Cancer-associated fibroblasts
ICIs
Immune checkpoint inhibitors
PD1
Programmed death-1
ORR
Objective response rate
DC
Dendritic cells
TANs
Tumor associated neutrophils
PFS
Progression-free survival
OS
Overall-survival
TAMs
Tumor-associated macrophage
GSEA
Gene set enrichment analysis
FAP
Fibroblast activating protein
NK
Nature killer
MHC
Major histocompatibility complex
Foxp3
Forkhead box P3
M-MDSC
Monocytic-myeloid-derived suppressor cells
PMN-MDSC
Polymorphonucler-myeloid derived suppressor cells
BP
Biological process
CC
Cellular component
MF
Molecular function
TIL
Tumor infiltrating lymphocyte
SMA
Smooth muscle actin
IHC
Immunohistochemistry
TGF-β
Transforming growth factor-beta
DMEM
Dulbecco’s modified eagle's medium
HE
Hematoxylin–eosin
Treg
Regulatory T cell
SEM
Standard error of mean

Background

KRAS mutation is one of the most common oncogenic drivers in non-small cell lung cancer (NSCLC), accounting for ~ 25% of Caucasians and ~ 10% of Asians [1, 2]. Recently, immune checkpoint inhibitors (ICIs) that target programmed cell death protein-1 (PD-1)/PD ligand-1 (PD-L1) axis have significantly prolonged survival outcomes of patients with metastatic NSCLC, especially for patients with KRAS mutations [3].
Notably, not all patients with KRAS mutations benefit from ICIs, largely because of tumor heterogeneity-a consequence of co-occurring genomic alterations. Skoulidis et al. divided KRAS-mutant lung adenocarcinoma into three subgroups on account of concomitant co-mutations, namely STK11/LKB1 mutations (KL subgroup), TP53 mutations (KP subgroup), and (KRAS-only subgroup) and revealed that KL subgroup responded poorly to ICIs, with an objective response rate (ORR) of only 7.4% [4]. As STK11/LKB1 mutations occur in approximately one-third of KRAS-mutated lung adenocarcinomas [5], to uncover the potential mechanism is in high demand and is critical for developing effective combination strategies.
Several lines of evidence have demonstrated that loss of LKB1 could (i) up-regulate c-MYC expression by promoting MZF1 transcription and increase the secretion of IL-23 and CCL9, hence, inhibit the recruitment of CD3+ T cells [6]; (ii) activate the WNT/β-catenin pathway in tumors cell to promote CCL4 production and thereby inhibiting recruitment of CD103+ dendritic cells (DCs) [7]; (iii) increase the expression of IL-6 which can recruit more tumor associated neutrophils (TANs) [8]; and (iv) silence STING transcription to result in insensitivity to cytoplasmic double-strand DNA detection, further facilitating immune escape [9]. All these findings, in view of intrinsic features of tumor cells or immune cells, partially explain why KL tumors develop primary resistance to ICI monotherapy. Of note, KL tumors were also refractory to combination of ICIs and chemotherapy [10]. Therefore, other important components in the tumor microenvironment (TME), particularly extracellular matrix (ECM) and cancer-associated fibroblasts (CAFs) which are also now recognized to play essential roles in antitumor immunity raised our research interest [11]. Therapeutic targeting of ECM or CAFs have recently represented a promising strategy in the anticancer arsenal. Intriguingly, a previous study demonstrated that KL lung cancer model had high levels of collagen deposition compared to KRAS-only model, while the mechanistic explanation was not clearly illustrated [12]. Considering the pivotal role of ECM and CAFs in TME composition, it is of significance to unravel the potential mechanism of their impact on anti-tumor immunity.
Here, using syngeneic KRASG12DLKB1−/− lung cancer mouse models, our study unveiled pivotal roles of ECM and CAFs in the formation of “immune-cold” KL tumors. We found that more ECM collagen deposited in KL tumors compared with KP tumors, resulting in forming a physical barrier to inhibit the infiltration of CD8+T cells, mechanistically owing to abundant CAFs with fibroblastic focal adhesion kinase (FAK) activation in the TME. Furthermore, using a novel FAK inhibitor (IN10018, received fast track designation from the U.S. Food and Drug Administration and Breakthrough Therapy Designation by the China National Medical Products Administration) to inhibit the activation of CAFs could break the physical barrier, effectively recruit effector immune cells into the TME, thus boosting anticancer immune response in KL lung cancer mouse models. Importantly, the combination of FAK inhibitor and PD-1 blockade could significantly retard primary and metastatic tumor growth of KL tumors.

Methods

Human data sharing

The OAK and POPLAR trials were available as supplementary information in Gandara DR et al. The MSKCC studies are available at www.​cbioportal.​org. The patient survival data and genetic aberrations were extracted for subgroup analysis.

Cell lines and reagents

Mouse KL cell line was a gift of Ji et al. Somatic KRAS activation and Lkb1 loss were induced in the lungs of genetically engineered male mice (KrasG12D/+/Lkb1L/L with the C57BL6/J genetic background) by nasal inhalation of Adeno-Cre (2 × 106 PFU). After 12 weeks Adeno-Cre treatment, tumors from mice of defined genotypes were dissected and cut into small pieces and cultured in RPMI-1640 + 10% FBS + 1% penicillin–streptomycin. The medium was changed every other day until cells outgrew and stable KL cell lines formed. Mouse KP cell line was provided by InxMed. A549 and H1299 cell lines were purchased from American Type Culture Collection (ATCC). L929 cell line was kindly provided by Stem Cell Bank, Chinese Academy of Sciences. Cells were maintained in DMEM or RPMI1640 (HyClone) supplemented with 10% fetal bovine serum (FBS) (Gibco) and 1% penicillin–streptomycin (Gibco). The small molecule FAK inhibitor IN10018 was provided by InxMed. Anti-PD-1 mAb (#BE0146, clone: RMP1-14), IgG control (#BE0089, clone: 2A3), InVivoPure™ pH 7.0 Dilution Buffer (#IP0070) and InVivoPure™ pH 6.5 Dilution Buffer (#IP0065) were purchase from BioXcell (West Lebanon, New Hampshire, USA).

Plasmid and siRNA transfection

Human-STK11 and mouse-STK11 were generated by PCR, cloned into pHBLV-vector and transfected into cells using lipofectamine3000. siRNAs were commercially synthesized (Ribo-Bio Co) and transfected into cells using riboFECT CP Transfection Kit (Ribo-Bio Co). siRNA sequences targeting human genes were as below: siSTK11-1, TGAGGAGGTTACGGCACAA.

Quantitative RT-PCR (qRT-PCR)

Total RNAs were isolated from cells using RNAiso Plus (TAKARA) and reverse-transcribed to cDNA using RevertAid First Strand cDNA Synthesis Kit (Thermo Scientific). Diluted cDNA was subject to qRT-PCR analysis using TB Green™ Premix Ex Taq™ II (TAKARA). Relative expression levels were calculated by the comparative Ct approach. qRT-PCR primers were listed as follows: m-FAP-Forward, 5′-GTCACCTGATCGGCAATTTGT-3′ and m-FAP-Reverse, 5′- CCCCATTCTGAAGGTCGTAGAT-3′; m-actin-Forward, 5′- TCCTGTGGCATCCACGAAACTACA-3′ and m-actin-Reverse, 5′- ACCAGACAGCACTGTGTTGGCATA-3′.

Western blotting

Protein samples were extracted from cells using RIPA lysis buffer (Beyotime). The quantification of protein level was measured with BCA kit (Beyotime). The samples were boiled in 5X loading buffer, separated by SDS-PAGE, transferred to PVDF membranes (Millipore). After blocking, the membrane was incubated overnight at 4 °C with primary antibodies as followings: LKB1 (#3047, Cell Signaling Technology; 1:1000 dilution), FAK (#610,087, BD Pharmingen; 1:1000 dilution), p-FAK (#81,298, Abcam; 1:1000 dilution), GAPDH (#5174, Cell Signaling Technology; 1:1000 dilution). The membrane was then washed with TBST and incubated with secondary antibody for 1 h at room temperature in the dark. The targeted proteins were detected by ECL reagent (Tanon) and the exposure was performed by ChemiDoc XRS (Bio-Rad).

Cell counting kit-8 (CCK-8) assay

KL cells were split into 96-well plate (3 × 103 cells, 100 μL per well, six replicates). The culture medium was changed into medium with different dose of FAK inhibitor on the other day. After incubating for 24 h, 48 h and 72 h, 10 μL of the CCK-8 (Dojindo) was added to each well of the plate and continued to incubate for 2 h. Absorbance was measured at a wavelength of 450 nm with microplate reader (Bio-Tek).

Cell apoptosis assay

The rates of cell apoptosis were measured by an Annexin V-FITC/PI Apoptosis Detection Kit (Yeasen biotech Co., Ltd.). Briefly, cells were harvested and washed with cold PBS twice. After being resuspended in 100 μL of binding buffer, cells were incubated with Annexin V and PI for 15 min in the dark. Then the apoptosis data was acquired on Beckman Cytoflex (Beckman Coulter). The results were analyzed with FlowJo 10.6.1 software (TreeStar).

RNA-seq and data analysis

The mouse tumor tissue was harvested for total RNA extraction with TRIzol (Invitrogen). RNA integrity was assessed using the RNA Nano 6000 Assay Kit of the Bioanalyzer 2100 system (Agilent Technologies). A total amount of 1 μg RNA per sample was used as input material for the RNA sample preparations. The cDNA libraries were generated with NEBNext Ultra RNA Library Prep Kit for Illumina (New England Biolabs Inc). The clustering of the index-coded samples was performed on a cBot Cluster Generation System using TruSeq PE Cluster Kit v3-cBot-HS (Illumia) according to the manufacturer’s instructions. After cluster generation, the library preparations were sequenced on an Illumina Novaseq platform and 150 bp paired-end reads were generated. Gene expression levels were quantified as normalized FPKM (fragments per kilobase of exon per million mapped fragments). GSEA was performed using local version of the GSEA analysis tool (http://​www.​broadinstitute.​org/​gsea/​index.​jsp). KEGG pathways and GO enrichment analysis of differentially expressed genes were implemented by clusterProfiler R package. The Reactome database brings together the various reactions and biological pathways of human model species. The corrected P value less than 0.05 were considered significantly enriched by differential expressed genes in Reactome database. Heatmaps were produced by Novomagic (https://​magic.​novogene.​com). FPKM RNA sequencing data was uploaded to GEO (GSE244452).

In vivo tumor formation assay

A total of 2 × 106 KP or KL cells were suspended in 200 μL phosphate-buffered saline (PBS) and then injected subcutaneously into 6 week old male C57BL/6 mice in the right flank. The tumor volumes were measured by caliper and calculated with a formula of π/6 × long diameter × short diameter × short diameter. Mice were sacrificed when tumor volume reached 700–800 mm3. Tumors from KP or KL cell-bearing mice were excised for further analysis.

In vivo drug response and tumor growth assays

To explore the efficacy of PD-1 inhibitor in KP and KL mouse model, 2 × 106 KP or KL cells were suspended in 200 μL PBS and were injected subcutaneously in the right flank of syngeneic recipient 6–8 week old male mice. Mice bearing tumors between 150 and 200 mm3 were randomly assigned to intraperitoneal treatment with six doses of 200 μg anti-PD-1 (clone RMPI-14; BioXCell) or isotype control antibody (clone 2A3; BioXCell) administered every 3 days (N = 5–8 mice per group). The body weights and tumor volumes were monitored and recorded twice a week.
To explore the impact of FAK inhibitor and combination therapy in KL mouse models, 2 × 106 KL cells were suspended in 200 μL PBS and were injected subcutaneously in the right flank of syngeneic recipient male mice. FAK inhibitor treatment or vehicle control reagent (0.5% Natrosol 250 HX dissolved in distilled water) was initiated at the first day when tumor volume reached at indicated size and administered at 25 mg/kg by oral gavage every day. Anti-PD-1 (clone RMPI-14; BioXCell) or isotype control antibody (clone 2A3; BioXCell) were administered at 200μg/mouse in total of six doses intraperitoneally. Tumor volume was measured with a formula of 0.5 × long diameter × short diameter × short diameter. At indicated day, tumor tissues or spleen were harvested for analysis.
All the animal studies were approved by the Institutional Committee for Animal Care and Use, Tongji University School of Medicine, and were performed in accordance with the institutional guidelines.

Histologic analysis

4 μm paraffin-embedded tissue were deparaffinized and rehydrated, stained with hematoxyline and eosin (H&E), IHC, IF, Masson's Trichrome Kit (SenBeiJia Biological Technology Co., Ltd.) and Picro-Sirius Red (SenBeiJia Biological Technology Co., Ltd.) following manufacturer protocol. For IHC, slides were incubated overnight in a humidified chamber at 4 °C with primary antibodies as followings: LKB1 (#13,031, Cell Signaling Technology; 1:250 dilution), Collagen I (#BA0325, Boster; 1:250 dilution), Collagen III (#ab7778, Abcam; 1:200 dilution), CD8 (#98,941, Cell Signaling Technology; 1:400 dilution), HIF-1α (#ab114977, Abcam; 1:250 dilution), p-FAK (Tyr397) (#44624G, Invitrogen; 1:200 dilution). The whole slides were scanned with Pannoramic 250 (3D HISTECH). The intensities of brown-colored precipitate stained with DAB were measured and quantified with IOD or cell intensity by Image Pro Plus 6 (Media Cybernetics). For IF, the FAP/α-SMA was used for activated CAF analysis (FAP: #ab53066, Abcam, 1:200 dilution; α-SMA: #ab7817, Abcam, 1:1000 dilution). CD31 (#77,699, Cell Signaling Technology; 1:100 dilution) was used for vascular analysis. The whole slides were scanned with Pannoramic MIDI (3D HISTECH).

Flow cytometry

The single-cell suspensions of tumor tissue were generated with tumor dissociation kit, mouse (Miltenyi Biotec) according to manufacturer’s instructions and mouse spleen were mechanically dissociated. After re-suspended in FACS buffer (PBS, 2%FBS), the cells were blocked with anti-mouse CD16/CD32 (eBioscience) 10 min prior to staining with surface antibodies at 4 °C. Surface markers were then added along with 10 μL Brilliant Stain Buffer (BD Bioscience) in a final staining volume to 100 μL. For intracellular staining, cells were fixed and permeabilized with Transcription Factor Buffer Set (BD Bioscience) followed by staining with intracellular antibodies. Fluorochrome-conjugated antibodies for flow cytometry analysis were listed in Additional file 1: Table S1. The flow cytometry data was acquired on Beckman Cytoflex (Beckman Coulter) and the results were analyzed with FlowJo 10.6.1 software (TreeStar).

Cytokine assays

The intratumoral CCL2, CCL5, CCL7, CXCL1, CXCL2 and VEGF were measured using Luminex liquid suspension chip. Mouse Premixed Multi-Analyte Kit (#LXSAMSM-06, R&D systems) was used in accordance with manufacturer’s instructions. Briefly, the lysate of tumor tissue was embedded with microbeads for 2 h, and then incubated with detection antibody for 1 h. Subsequently, streptavidin-PE was added into each well for 30 min, and values were read using the Bio-Plex MAGPIX System (Bio-Rad). The intratumoral CXCL9 (#MCX900, R&D systems) and CXCL5 (#ab100719, Abcam) were analyzed with commercial ELISA Kit according to the manufacturer's instructions.

Statistical analysis

Results were presented as mean ± SEM. The log-rank test was used for comparison of survival outcomes downloaded from OAK, POPLAR and MSKCC with Kaplan–Meier method. Unpaired Student’s t-test was used to compare the statistical significance between tested and control groups. For comparison of tumor growth between two groups in vivo, two-way analysis of variance (ANOVA) with Tukey’s post hoc test was performed (control versus FAK inhibitor group in Fig. 6A; control versus anti-PD1 and FAK inhibitor versus FAK inhibitor + anti-PD1 group in Fig. 7C) [13].Survival time of mice was defined from the day of tumor cell inoculation until the tumor volume reached 1000 mm3.Statistical analyses were performed using GraphPad Prism 8 (GraphPad Software). Statistical significance was determined as indicated in the figure legends. Significance was set to P < 0.05 and represented as *P < 0.05, **P < 0.01, ***P < 0.001.

Results

KL lung adenocarcinoma displayed a poor response to ICI therapy

Firstly, we investigated the impact of KRAS/LKB1 co-mutations on survival outcomes in PD-1/PD-L1 inhibitor-treated lung adenocarcinoma patients from POPLAR and OAK trials and confirmed that KL subgroup had significantly shorter progression-free survival (PFS) compared with other KRAS-mutated patients (median PFS:1.3 vs. 4.1 months, p = 0.0025). Integrating the data from MSKCC cohort, we further demonstrated that overall survival (OS) also tended to be shorter in KL group (median OS: 9.0 vs. 15.0 months, p = 0.065) (Fig. 1A).
Previous studies reported that among all subtypes of KRAS-driven lung cancers, tumors with frequently inactivated TP53 were most sensitive to PD-1 blockade [4]. Therefore, to explore the underlying mechanisms for the distinct therapeutic response, we used KRASG12DLKB1−/− (hereafter referred to “KL”, previously established from a spontaneously arising lung adenocarcinoma in the KrasG12DLkb1fl/fl mouse model, (Additional file 3: Fig. S1A) and KRASG12DTP53−/− (hereafter referred to “KP”) murine lung adenocarcinoma cell lines [14, 15] for further analyses. Isogenic cell lines upon confirmation of STK11 knockout (by immunoblotting for the LKB1 protein) (Additional file 3: Fig. S1B) were implanted into the right flank of syngeneic recipient mice and cohorts of tumor-bearing mice were randomized to be treated with anti-PD-1 antibody or IgG control (Fig. 1B). The expression of LKB1 by IHC in KP or KL tumors further confirmed that STK11 was successfully knocked out (Fig. 1C and D). Tumor growth curve suggested that anti-PD-1 antibody significantly attenuated tumor growth in subcutaneous KP mouse model (Fig. 1C) but showed no efficacy in KL mouse model (Fig. 1D). No significant differences in body weight were found between anti-PD-1 antibody and control group in KP or KL mouse model. These results suggested that KL tumors were resistant to anti-PD1 antibody and our KL mouse model was reliable for subsequent research.

KL tumors revealed immunosuppressive TME with fewer immune-active cells and more immunosuppressive cells infiltration

We next sought to elucidate how LKB1-deficiency impacted the TME. Firstly, comprehensive immune cell populations between KP and KL tumors via flow cytometry analysis were compared. In general, the infiltrated quantity of T cells (CD3) and B cells (CD19) were significantly decreased in KL tumors as compared with KP tumors (Fig. 2A and B) while no significant difference was observed regarding NK cells (CD3NK1.1+) (Fig. 2C). Moreover, KL tumors had significantly fewer CD8+T cells, CD4+T cells and Foxp3+Tregs infiltration (Fig. 2D–F). In addition, DCs (CD45+CD11c+MHC-II+) and tumor-associated macrophage (TAMs)[16], regardless of M1-like macrophage (CD11b+F4/80+CD86+) or M2-like macrophage (CD11b+F4/80+CD206+) were also less infiltrated in KL tumors than KP tumors (Fig. 2G and I). Tumor-associated neutrophils (TANs) and polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs, CD45+CD11b+Ly6G+Ly6Clow), which suppressed T cells function to help form immunosuppressive TME [17], were significantly highly infiltrated in KL tumors (Fig. 2H and J). Taken together, our data demonstrated that KL tumors revealed immunosuppressive TME, with fewer immune-active cells infiltration but with more immunosuppressive cells infiltration, including PMN-MDSC and TANs.

KL tumors had excessive ECM deposition and collagen, which block T cells infiltration into tumor nest

To uncover the mechanisms that contributed to the formation of immunosuppressive TME of KL tumors, RNA-seq-based transcriptome profiling of KL and KP subcutaneous tumors was analyzed. Briefly, 3497 genes were up-regulated and 3952 genes were down-regulated in KL tumors compared with KP tumors. Furthermore, GO, Reactome and gene set enrichment analysis (GSEA) were performed and showed that genes encoding for ECM-related components were significantly enriched (Fig. 3A–C). The ECM provided a physical scaffold for all cells in the TME and is the major component. Collagen has been implicated in cancer progression, metastasis, and drug resistance. As expected, the transcriptomic data showed that collagen-formation related genes were mostly upregulated in KL tumors. Among top 20 collagen-related genes, COL3A1 and COL1A1 were significantly upregulated in KL tumors (Fig. 3D, Additional file 2: Table S2). Consistently, Sirius Red and Masson staining confirmed the increased intra-tumoral collagen deposition and specifically, collagen I and III, which are encoded by COL1A1 and COL3A1, in KL tumors (Fig. 3E ). To elucidate the impact of collagen deposition on the distribution of immune cells, we further analyzed the spatial correlation between collagen deposition and CD8+ T cells infiltration. The results showed that abundant CD8+ T cells were present at sites of collagen deposition (Fig. 3F). Therefore, we analyzed the distribution of CD8+ T cells in two regions (tumor nest and stroma, including tumor margin and intratumor stroma). In tumor nest, CD8+ T cells were significantly less infiltrated in KL tumors, however, at the tumor margin where collagen deposited, CD8+ T cells infiltration were much higher in KL tumors (Fig. 3G).
The above results revealed that (i) compared with KP tumors, KL tumor had excessive ECM deposition; (ii) excessive collagen deposition blocked CD8+ T cells infiltrating into tumor nest to exert killing function; and (iii) CD8+ T cells could barely infiltrate into tumor nest owing to the collagen deposition and were accumulated in the margin of KL tumors.

Activated CAFs were abundant in KL tumors and fibroblastic FAK is hyperactivated

CAFs are the predominant stromal cells in the TME [18]. Activated CAFs, which highly express α-SMA and fibroblast activating protein (FAP), usually accumulate at the edge of tumor nest, secret collagen fibers to surround the malignant cells with dense desmoplastic stroma, hence facilitate drug resistance via blocking anti-tumor drugs and immune cells into tumor nest [19, 20]. Therefore, we questioned whether CAFs were significantly activated in KL tumors. Firstly, using TIMER2.0 database (http://​timer.​cistrome.​org/​), we observed a positive correlation between the expression of dominant genes encoding collagen fibers and markers for activated CAFs (Fig. 4A and B). Consistently, our RNA-seq data also showed the more CAFs were infiltrated in KL tumors (Fig. 4C). To investigate whether KL tumor cells directly impacted the activation of fibroblasts, KL and KP tumor cells were co-cultured with mouse fibroblast (L929) in vitro. The results showed that co-culture with KL tumor cells significantly increased expression of FAP protein, suggesting that KL tumor cells could activate CAFs (Fig. 4D). Immunofluorescence further confirmed that activated CAFs were highly infiltrated in KL tumors (Fig. 4E). Next, we performed KEGG pathway analysis of dissected KL and KP tumors to explore the mechanisms of abundant activated CAFs. Interestingly, focal adhesions-related genes were enriched in KL tumors (Fig. 4F). Focal adhesions are integrated points of cellular membrane which link to extracellular matrix via integrin heterodimers [21, 22]. FAK is recruited to the sites of focal adhesions and get auto-phosphorylated upon ECM-induced integrin receptor activation [23]. Previous studies demonstrated that FAK activation was closely associated with stromal fibrosis and immunosuppressive TME [24, 25]. Further co-localization analysis in tumor specimens demonstrated that FAK was hyperactivated in CAFs (Fig. 4G). Given the fact that FAK activation within pancreatic cancer CAFs was associated with ECM synthesis and deposition [26], we studied whether fibroblastic FAK hyperactivation could lead to CAFs activation and collagen deposition. KL tumors-bearing mouse were treated with FAK inhibitor (IN10018, InxMed, 25 mg/kg) or natrosol as control and tumor components were assessed at different time points (Day 3, Day 7, Day 14) (Fig. 5A). As indicated, FAK inhibitor effectively inhibited FAK phosphorylation (Fig. 5B). Further GO analysis revealed that FAK inhibitor had significant impact on ECM-related genes (FAK inhibitor vs control at Day 14, FAK inhibitor at Day14 vs FAK inhibitor at Day 7) (Fig. 5C). Immunofluorescence and IHC results also demonstrated FAK inhibitor could significantly decrease fibrosis and CAFs activation (Fig. 5D-G).
Angiogenesis is one of the hallmarks of cancer and blood vessels are also the important components of the TME [11, 27]. Notably, previous evidence showed that FAK pathway was associated with angiogenesis and tumor growth [2830]. Therefore, we questioned whether the use of FAK inhibitor has an impact on TME via inhibiting tumor angiogenesis in KL tumors. Indeed, GO analysis and immunofluorescence staining revealed that angiogenesis-associated genes were highly enriched in KL tumors (Additional file 4: Fig. S2A, B). Although FAK inhibitor slightly alleviated hypoxia at Day 7, FAK inhibitor had no significant impact on angiogenesis (Additional file 4: Fig. S2C, D).
In summary, these findings suggested that (i) abundant activated CAFs resulted in excessive collagen deposition in KL tumors; and (ii) fibroblastic FAK activity contributed CAFs activation and hence, induced collagen formation.

Inhibition of FAK suppressed tumor progression via boosting immune response in KL tumors

Therapeutic benefits of pharmacological FAK inhibition in previous preclinical models have been confirmed to directly inhibit FAK expressed by neoplastic cells and thus, reduced tumor growth, invasion and metastasis [31, 32]. To illustrate the cytotoxic effects of FAK inhibition on tumor cells, we utilized human NSCLC cell lines with LKB1-mutant (A549), LKB1-WT (H1299), mouse lung cancer cell line with LKB1-wild type (KP) and mouse lung cancer cell line with LKB1-loss (KL). However, knockdown of LKB1 in LKB1-wild type cell lines and upregulation of LKB1 in LKB1-mutant or LKB1-loss cell lines did not significantly influence the activation of FAK pathway (Additional file 5: Fig. S3A). Additionally, FAK inhibitor had minimal impact on cell viability and no obvious effects on apoptosis of KL cells in vitro (Additional file 5: Fig. S3B, C). These results provided the evidence that FAK inhibitor did not directly exert cytotoxic effects on lung tumor cells.
Next, we investigated the anti-tumor effect of FAK inhibition on tumor growth in KL mouse model and found that tumor growth was significantly attenuated (Fig. 6A). Therefore, we speculated FAK inhibitor might exert anti-tumor effect via remodeling the TME. Interestingly, FAK inhibitor significantly promoted proliferation of entire immune cells (CD45+Ki67+, Additional file 6: Fig. S4A). Moreover, FAK inhibitor significantly increased the proportions of T cells (on Day 7 and Day 14), B cells (on Day 3 and Day 7) and NK cells (on Day 3 and Day 14) (Fig. 6B–D). Specifically, a simultaneous increase in CD8+ T cells was observed with FAK inhibition on Day 3, Day 7 and Day 14 (Fig. 6E). Interestingly, on Day 14, more Foxp3+Tregs were also infiltrated in FAK inhibitor groups (Fig. 6F). Regarding myeloid cells, FAK inhibition facilitated more DCs infiltration on Day 7 and Day 14 (Fig. 6H). The quantity of M-MDSC were temporarily elevated with the treatment of FAK inhibitor on day 3 and day 7. In contrary, less PMN-MDSCs were infiltrated with FAK inhibition on day 14 (Fig. 6G). Although the total quantity of macrophages in two groups were not significantly altered, FAK inhibitor changed the proportions of macrophage phenotype, with more infiltration of M1-like macrophage and less infiltration of M2-like macrophage (Fig. 6I). The dynamic changes of immune cells in spleen were generally consistent with tumors upon FAK inhibition (Additional file 7: Fig. S5).
We also examined the dynamic change levels of related cytokines in tumor tissues. Firstly, RNA-seq demonstrated the corresponding changed immune cell-related cytokines, including CXCL9, CCL5, CCL2, VEGFA, CXCL5, CXCL1, CXCL2, CCL7 et al., were also significantly altered (Additional file 8: Fig. S6A). Specifically, CXCL1 was significantly reduced on Day 3 and Day 7 and CXCL2 was reduced on Day 7 with FAK inhibition, Meanwhile, CXCL9 was upregulated on Day 7 (Additional file 8: Fig. S6B).
The above findings suggested that FAK inhibitor effectively suppressed tumor growth via boost immune response, converting “immune-cold” tumors into “immune-hot” tumors. Upon FAK inhibition, more lymphocytes were infiltrated, especially CD8+ T cells. Meanwhile, several immunosuppressive cells, including PMN-MDSC and M2-like macrophage, were less infiltrated. Interestingly, FAK inhibitor could also upregulate PD-L1 expression on non-immune cells (CD45PD-L1+, Additional file 6: Fig. S4B), implying a potential synergetic anti-tumor effect with anti-PD-1 antibody.

Combining FAK inhibition and PD-1 blockade optimized anti-tumor effects in KL mouse models

Based on our previous findings, we further examined the anti-tumor effect of combining FAK inhibition and PD-1 blockade in KL mouse models. Tumor-bearing KL models and the following schedules were shown in Fig. 7A. PD-1 blockade had no impact on FAK phosphorylation (Fig. 7B). As expected, FAK inhibitor monotherapy or plus PD-1 blockade could both attenuate tumor growth compared with either PD-1 blockade group or control. Importantly, as compared with FAK inhibitor alone, the combination therapy resulted in a synergetic anti-tumor effect (Fig. 7C). Furthermore, the combination of FAK inhibitor and PD-1 blockade also led to prolonged time to reach endpoint (the time when tumor volume reached 1000 mm3) (Fig. 7D). Notably, on Day 30, histological analyses of lung tissues from subcutaneous mouse model showed that tumor cells had metastasized to lung, suggesting KL tumor cells were aggressive and invasive (Fig. 7E). Interestingly, FAK inhibitor with and without PD-1 blockade could also effectively decrease the metastatic area in lung tissue and inhibit lung metastases compared with control of anti-PD1 monotherapy group (Fig. 7F). As expected, FAK inhibitor and the combination therapy also significantly decreased collagen deposition and inhibited CAFs activation (Fig. 8A–C). Moreover, vessel density was similar among the four groups (Additional file 9: Fig. S7).
We then investigated the impact of different treatment regimens on the TME. The results showed that the proportions of T cells, specifically CD8+T cells were significantly increased in the combination group (Additional file 10: Fig. S8A and Fig. 8D). Additionally, CD8/Treg ratio was numerically elevated in the combination group compared with FAK inhibitor monotherapy group (p = 0.08) (Fig. 8D). Further IHC staining showed that FAK inhibitor in combination with PD-1 blockade could increase CD8+ T cells infiltration in both subcutaneous and lung metastatic tumor tissues (Fig. 8E and F). The proportions of B cells, NK cells and CD8+GZMB+ T cells were also significantly increased with FAK inhibition while there was no significant difference between FAK inhibitor monotherapy and the combination therapy (Additional file 10: Fig. S8B–D). In terms of Tregs, the combination therapy numerically reduced Tregs infiltration compared with FAK inhibitor monotherapy group (Additional file 10: Fig. S8E). Regarding myeloid cells, the combination group or FAK inhibitor significantly promoted DCs infiltration and reduce PMN-MDSC and macrophages infiltration (Additional file 10: Fig. S8F–H).
Collectively, these results demonstrated FAK inhibitor could overcome primary resistance of KL tumors to PD-1 blockade.

Discussion

The failure of PD-1 blockade therapy in KL lung adenocarcinoma emphasized the demand to uncover the characteristics of the TME in KL tumors and develop novel combinational therapeutic strategy. Utilizing KL and KP mouse model, we identified KL tumor as “immune-cold” tumor with excessive ECM collagen deposition that formed a physical barrier to block the infiltration of CD8+T cells. Mechanistically, abundant activated CAFs resulted from fibroblastic FAK activation contributed to the formation of the unique TME of KL tumors. Upon FAK inhibition to inhibit the activation of CAFs, our study demonstrated that FAK inhibitor could break the physical barrier via decreasing collagen deposition, and further facilitate the infiltration of CD8+ T cells, DC cells and M1-like macrophages into tumors, hence, convert “immune-cold” KL tumors into “immune-hot” tumors. The synergetic anti-tumor effect of FAK inhibitor and PD-1 blockade in KL mouse models provided new sights for developing effective combination strategies that reestablish antitumor immunity in KL tumors (Fig. 9).
It was reported that high stromal density could provide a barrier to the delivery of cytotoxic agents and has been postulated to limit T cells access to the tumor site in pancreatic cancer [33]. Similarly, in the preliminary exploration of the TME in KL lung adenocarcinoma, we found that dense collagen deposited and formed a physical barrier to prohibit the infiltration of immune cells to KL tumors. Also, a previous study demonstrated that the KL lung cancer model showed high levels of collagen deposition compared to KRAS-only model [12]. Owing to this physical barrier, the infiltration of immune cells, including CD8+ T cells, were generally decreased in KL tumors. Therefore, our findings might at least partially explain why KL tumors were not only refractory to ICI monotherapy, but also poorly responded to the combination of ICIs and chemotherapy [10].
Notably, not all immune cell infiltration was reduced in KL tumors. In our KL lung cancer mouse model, the infiltration of PMN-MDSC was significantly increased, therefore, the physical barrier cannot fully explain the formation of unique TME of KL tumors. Interestingly, CAFs, as the most abundant type of stromal cells in the TME, played a critical role in collagen formation and were hyperactivated in KL tumors. CAFs were not only the main source of collagen, but also secreted a variety of proteins, cytokines, chemokines to shape the TME [34]. Activated CAFs secreted factors such as IL-6, CCL2, and CXCL12 to affect T cell function [35]. CAFs could also secrete CXCL1 and CXCL2 to recruit suppressive immune cells PMN-MDSC into tumors [36]. Therefore, we believed that the abnormal activation of CAFs in KL tumors might be the key determiner to orchestrate the “immune-cold” tumors.
Focal adhesions are mediators of cell-ECM junctions that activate the intracellular FAK pathway [37]. In pancreatic cancer, the activation of FAK pathway of tumor cells can lead to extracellular fibrosis and immunosuppressive TME [33]. Also, FAK activation in squamous tumor cells increased the infiltration of Tregs via promoting the secretion of TGF-β and CCL5 and inhibiting the killing function of CD8+ T cells [38]. Previous studies have shown that LKB1 could inhibit the downstream signaling of FAK by phosphorylating MARK1, thereby inhibiting the formation of ECM collagen, and LKB1 deficiency can lead to the activation of the FAK pathway owing to the imbalance of oxidative stress levels [39, 40]. However, in our KL lung cancer model, LKB1 was not related to activation of FAK pathway in tumor cells. Meanwhile, fibroblastic FAK was hyperactivated and FAK inhibitor could significantly suppress the activation of CAFs and reduce the deposition of ECM collagen. There was evidence that the activation of intracellular FAK-Src pathway in CAFs could result in activation of fibroblasts by upregulating the expression of α-SMA [41]. Furthermore, in pancreatic cancer, inhibition of the FAK pathway in CAFs can effectively inhibit the activation of CAFs, thereby inhibiting tumor metastases, regulating the recruitment of immune cells, and promoting the degradation of extracellular collagen [26], which was consistent with our findings. However, Demircioglu et al. found that complete knockout of FAK protein in CAFs could facilitate the glycolysis of pancreatic cancer cells, thereby promoting the tumor growth [24]. Notably, the role of the intracellular FAK pathway depends on the helper of the scaffold protein and the activity of the kinase. Our current study used IN10018, a potent and highly selective ATP-competitive FAK inhibitor, to inhibit FAK activity but still retained FAK as a scaffold protein activity, representing a new option for inhibiting the activation of CAFs and remodeling the TME.
On the basis of FAK inhibitor could break the fibrotic barrier, we further demonstrated that FAK inhibitor could effectively suppress tumor growth via converting “cold” tumors into “hot” tumors, rather than cytotoxic effects directly on tumor cells. With FAK inhibition, more lymphocytes were infiltrated, especially CD8+T cells and DCs. Meanwhile, several immunosuppressive cells, including PMN-MDSC and M2-like macrophage, were less infiltrated. Zaghdoudi et al. also found that inhibition of FAK in CAFs could significantly promote the recruitment of M1-like macrophages and reduce the M2-like macrophages [26]. Besides a direct impact on the ECM collagen deposition, FAK inhibitor could also remodel the TME through regulating cytokines secretion, such as CXCL1 and CXCL2. Previous study showed that CAFs could mediate resistance to CSF-1R therapy by secreting CXCL1 and CXCL2 to recruit PMN-MDSCs, and the combined inhibition of CXCR2 and CSF-1R could overcome tumor resistance [36]. In addition, the level of CXCL9 was also elevated after application of FAK inhibitor in our study. CXCL9 is mainly secreted by M1-like macrophages and DCs to facilitate CD8+ T cells infiltration [42]. Therefore, we speculated that infiltrated DCs and M1-like macrophages upon FAK inhibition may further secrete CXCL9 to promote the infiltration of CD8+ T cells.
Previous study demonstrated that combing FAK inhibition and KRAS G12C inhibitors could improve the treatment outcomes for KRAS G12C mutant cancers through the regulation of the FAK-YAP signaling [43]. Given the fact that FAK inhibitor could remodel the TME in the current study, we next utilized KL mouse model to investigate the feasibility and efficacy of combining FAK inhibitor and PD-1 blockade. As expected, FAK inhibitor could overcome primary resistance of KL tumors to PD-1 blockade and a synergetic anti-tumor effect of FAK inhibitor and PD-1 blockade in KL tumors was observed. Notably, the combination therapy could also effectively inhibit lung metastases. Similarly, in PD-L1+ triple negative breast cancer cells lines, FAK inhibitor and PD-L1 antibody also exerted synergetic anti-tumor effects [44, 45]. Inhibition of the FAK pathway in CAFs could also significantly inhibited lung metastases in breast cancer [25].
Although the current study provided the new insight of therapeutic strategy for KL lung cancer tumors, however, several limitations and further investigations need to be acknowledged: (i) KP tumors responded well to PD-1 blockade, however, the impact of FAK inhibitor on KP tumors remained unclear. Peng et al. reported that collagen promoted anti-PD-1/PD-L1 acquired resistance in KP tumors [46]. Therefore, FAK inhibitor which could destroy stromal fibrosis might be a promising candidate in the treatment of KP tumor after its resistance to PD-1 blockade; (ii) Activated fibroblastic FAK was observed in KL tumors, however, how LKB1-deficiency KRAS-mutated tumor cells activated CAF and resulted in hyperactivating FAK pathway was not determined. It was reported that tumor-secreted LOXL-2 activated fibroblast through FAK signaling in breast cancer [41]. Secreted LOXL2 was also a novel therapeutic target that promoted gastric cancer metastasis via the FAK pathway [47]. Therefore, in KL tumors, whether LOXL-2 is a key regulator contributing to activated FAK pathway in CAF needs to be further investigated; and (iii) TME features in subcutaneous tumor models might be distinct from that in orthotopic tumors. Therefore, our findings should be verified in orthotopic mouse models in further investigations.
In summary, our findings emphasized the critical role of ECM collagen and CAFs in the formation of “immune-cold” KL tumors. FAK inhibition with a small molecular inhibitor could inhibit the activation of CAFs, destroy the stromal fibrosis barrier, and thus to convert the nature of KL tumors from “cold” to “hot”. Our study provided new sights for developing effective combination strategies that re-establish antitumor immunity in KL tumors in the future studies.

Acknowledgements

We thank Hongbin Ji for his support and providing KL mouse cell line, Baoyuan Zhang for technical support, Jun Zhang, Fei Li and Haipeng Liu for helpful insight for this project, Xiaojun Yang and Liang Tang for equipment maintenance, Xiaomin Cheng for establishment of animal models, Qianlu Huang and Yinqiao Shen for helping perform flow cytometry.

Declarations

All the animal studies were approved by the Institutional Committee for Animal Care and Use, Shanghai Pulmonary Hospital, and were performed in accordance with the institutional guidelines.
All authors agreed on the manuscript.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat El Osta B, Behera M, Kim S, Berry LD, Sica G, Pillai RN, et al. Characteristics and outcomes of patients with metastatic KRAS-mutant lung adenocarcinomas: the lung cancer mutation consortium experience. J Thorac Oncol. 2019;14(5):876–89.PubMedPubMedCentralCrossRef El Osta B, Behera M, Kim S, Berry LD, Sica G, Pillai RN, et al. Characteristics and outcomes of patients with metastatic KRAS-mutant lung adenocarcinomas: the lung cancer mutation consortium experience. J Thorac Oncol. 2019;14(5):876–89.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Dearden S, Stevens J, Wu YL, Blowers D. Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology (mutMap). Ann Oncol. 2013;24(9):2371–6.PubMedPubMedCentralCrossRef Dearden S, Stevens J, Wu YL, Blowers D. Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology (mutMap). Ann Oncol. 2013;24(9):2371–6.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Lee CK, Man J, Lord S, Cooper W, Links M, Gebski V, et al. Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non-small cell lung carcinoma: a systematic review and meta-analysis. JAMA Oncol. 2018;4(2):210–6.PubMedCrossRef Lee CK, Man J, Lord S, Cooper W, Links M, Gebski V, et al. Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non-small cell lung carcinoma: a systematic review and meta-analysis. JAMA Oncol. 2018;4(2):210–6.PubMedCrossRef
4.
Zurück zum Zitat Skoulidis F, Goldberg ME, Greenawalt DM, Hellmann MD, Awad MM, Gainor JF, et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 2018;8(7):822–35.PubMedPubMedCentralCrossRef Skoulidis F, Goldberg ME, Greenawalt DM, Hellmann MD, Awad MM, Gainor JF, et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 2018;8(7):822–35.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511(7511):543–50.CrossRef Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511(7511):543–50.CrossRef
6.
Zurück zum Zitat Kortlever RM, Sodir NM, Wilson CH, Burkhart DL, Pellegrinet L, Brown Swigart L, et al. Myc cooperates with ras by programming inflammation and immune suppression. Cell. 2017;171(6):1301-1315 e1314.PubMedPubMedCentralCrossRef Kortlever RM, Sodir NM, Wilson CH, Burkhart DL, Pellegrinet L, Brown Swigart L, et al. Myc cooperates with ras by programming inflammation and immune suppression. Cell. 2017;171(6):1301-1315 e1314.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature. 2015;523(7559):231–5.PubMedCrossRef Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature. 2015;523(7559):231–5.PubMedCrossRef
8.
Zurück zum Zitat Koyama S, Akbay EA, Li YY, Aref AR, Skoulidis F, Herter-Sprie GS, et al. STK11/LKB1 deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T-cell activity in the lung tumor microenvironment. Cancer Res. 2016;76(5):999–1008.PubMedPubMedCentralCrossRef Koyama S, Akbay EA, Li YY, Aref AR, Skoulidis F, Herter-Sprie GS, et al. STK11/LKB1 deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T-cell activity in the lung tumor microenvironment. Cancer Res. 2016;76(5):999–1008.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Kitajima S, Ivanova E, Guo S, Yoshida R, Campisi M, Sundararaman SK, et al. Suppression of STING associated with LKB1 loss in KRAS-driven lung cancer. Cancer Discov. 2019;9(1):34–45.PubMedCrossRef Kitajima S, Ivanova E, Guo S, Yoshida R, Campisi M, Sundararaman SK, et al. Suppression of STING associated with LKB1 loss in KRAS-driven lung cancer. Cancer Discov. 2019;9(1):34–45.PubMedCrossRef
10.
Zurück zum Zitat Gandara DR, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat Med. 2018 Sep;24(9):1441-1448. Gandara DR, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat Med. 2018 Sep;24(9):1441-1448.
11.
Zurück zum Zitat Bejarano L, Jordao MJC, Joyce JA. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 2021;11(4):933–59.PubMedCrossRef Bejarano L, Jordao MJC, Joyce JA. Therapeutic targeting of the tumor microenvironment. Cancer Discov. 2021;11(4):933–59.PubMedCrossRef
12.
Zurück zum Zitat Gilbert-Ross M, Konen J, Koo J, Shupe J, Robinson BS, Wiles BS, et al. Targeting adhesion signaling in KRAS, LKB1 mutant lung adenocarcinoma. JCI Insight. 2017;2(5): e90487.PubMedPubMedCentralCrossRef Gilbert-Ross M, Konen J, Koo J, Shupe J, Robinson BS, Wiles BS, et al. Targeting adhesion signaling in KRAS, LKB1 mutant lung adenocarcinoma. JCI Insight. 2017;2(5): e90487.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Reticker-Flynn NE, Zhang W, Belk JA, Basto PA, Escalante NK, Pilarowski GO, et al. Lymph node colonization induces tumor-immune tolerance to promote distant metastasis. Cell. 2022;185(11):1924-1942 e1923.PubMedPubMedCentralCrossRef Reticker-Flynn NE, Zhang W, Belk JA, Basto PA, Escalante NK, Pilarowski GO, et al. Lymph node colonization induces tumor-immune tolerance to promote distant metastasis. Cell. 2022;185(11):1924-1942 e1923.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Tong X, Chen Y, Zhu X, Ye Y, Xue Y, Wang R, et al. Nanog maintains stemness of Lkb1-deficient lung adenocarcinoma and prevents gastric differentiation. EMBO Mol Med. 2021;13(9): e14795.PubMedPubMedCentralCrossRef Tong X, Chen Y, Zhu X, Ye Y, Xue Y, Wang R, et al. Nanog maintains stemness of Lkb1-deficient lung adenocarcinoma and prevents gastric differentiation. EMBO Mol Med. 2021;13(9): e14795.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Wang Z, Fang Z, Chen G, Liu B, Xu J, Li F, et al. Chromobox 4 facilitates tumorigenesis of lung adenocarcinoma through the Wnt/beta-catenin pathway. Neoplasia. 2021;23(2):222–33.PubMedCrossRef Wang Z, Fang Z, Chen G, Liu B, Xu J, Li F, et al. Chromobox 4 facilitates tumorigenesis of lung adenocarcinoma through the Wnt/beta-catenin pathway. Neoplasia. 2021;23(2):222–33.PubMedCrossRef
16.
Zurück zum Zitat Zhu S, Yi M, Wu Y, Dong B, Wu K. Roles of tumor-associated macrophages in tumor progression: implications on therapeutic strategies. Exp Hematol Oncol. 2021;10(1):60.PubMedPubMedCentralCrossRef Zhu S, Yi M, Wu Y, Dong B, Wu K. Roles of tumor-associated macrophages in tumor progression: implications on therapeutic strategies. Exp Hematol Oncol. 2021;10(1):60.PubMedPubMedCentralCrossRef
17.
18.
Zurück zum Zitat Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 2010;17(2):135–47.PubMedCrossRef Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 2010;17(2):135–47.PubMedCrossRef
19.
Zurück zum Zitat Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci USA. 2013;110(50):20212–7.PubMedPubMedCentralCrossRef Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci USA. 2013;110(50):20212–7.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Yang X, Lin Y, Shi Y, Li B, Liu W, Yin W, et al. FAP promotes immunosuppression by cancer-associated fibroblasts in the tumor microenvironment via STAT3-CCL2 signaling. Cancer Res. 2016;76(14):4124–35.PubMedCrossRef Yang X, Lin Y, Shi Y, Li B, Liu W, Yin W, et al. FAP promotes immunosuppression by cancer-associated fibroblasts in the tumor microenvironment via STAT3-CCL2 signaling. Cancer Res. 2016;76(14):4124–35.PubMedCrossRef
21.
Zurück zum Zitat Lee BY, Timpson P, Horvath LG, Daly RJ. FAK signaling in human cancer as a target for therapeutics. Pharmacol Ther. 2015;146:132–49.PubMedCrossRef Lee BY, Timpson P, Horvath LG, Daly RJ. FAK signaling in human cancer as a target for therapeutics. Pharmacol Ther. 2015;146:132–49.PubMedCrossRef
22.
23.
Zurück zum Zitat Lietha D, Cai X, Ceccarelli DF, Li Y, Schaller MD, Eck MJ. Structural basis for the autoinhibition of focal adhesion kinase. Cell. 2007;129(6):1177–87.PubMedPubMedCentralCrossRef Lietha D, Cai X, Ceccarelli DF, Li Y, Schaller MD, Eck MJ. Structural basis for the autoinhibition of focal adhesion kinase. Cell. 2007;129(6):1177–87.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Demircioglu F, Wang J, Candido J, Costa ASH, Casado P, de Luxan DB, et al. Cancer associated fibroblast FAK regulates malignant cell metabolism. Nat Commun. 2020;11(1):1290.PubMedPubMedCentralCrossRef Demircioglu F, Wang J, Candido J, Costa ASH, Casado P, de Luxan DB, et al. Cancer associated fibroblast FAK regulates malignant cell metabolism. Nat Commun. 2020;11(1):1290.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Wu HJ, Hao M, Yeo SK, Guan JL. FAK signaling in cancer-associated fibroblasts promotes breast cancer cell migration and metastasis by exosomal miRNAs-mediated intercellular communication. Oncogene. 2020;39(12):2539–49.PubMedPubMedCentralCrossRef Wu HJ, Hao M, Yeo SK, Guan JL. FAK signaling in cancer-associated fibroblasts promotes breast cancer cell migration and metastasis by exosomal miRNAs-mediated intercellular communication. Oncogene. 2020;39(12):2539–49.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Zaghdoudi S, Decaup E, Belhabib I, Samain R, Cassant-Sourdy S, Rochotte J, et al. FAK activity in cancer-associated fibroblasts is a prognostic marker and a druggable key metastatic player in pancreatic cancer. EMBO Mol Med. 2020;12(11): e12010.PubMedPubMedCentralCrossRef Zaghdoudi S, Decaup E, Belhabib I, Samain R, Cassant-Sourdy S, Rochotte J, et al. FAK activity in cancer-associated fibroblasts is a prognostic marker and a druggable key metastatic player in pancreatic cancer. EMBO Mol Med. 2020;12(11): e12010.PubMedPubMedCentralCrossRef
27.
28.
Zurück zum Zitat Lechertier T, Reynolds LE, Kim H, Pedrosa AR, Gomez-Escudero J, Munoz-Felix JM, et al. Pericyte FAK negatively regulates Gas6/Axl signalling to suppress tumour angiogenesis and tumour growth. Nat Commun. 2020;11(1):2810.PubMedPubMedCentralCrossRef Lechertier T, Reynolds LE, Kim H, Pedrosa AR, Gomez-Escudero J, Munoz-Felix JM, et al. Pericyte FAK negatively regulates Gas6/Axl signalling to suppress tumour angiogenesis and tumour growth. Nat Commun. 2020;11(1):2810.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Tavora B, Batista S, Reynolds LE, Jadeja S, Robinson S, Kostourou V, et al. Endothelial FAK is required for tumour angiogenesis. EMBO Mol Med. 2016;8(10):1229.PubMedPubMedCentralCrossRef Tavora B, Batista S, Reynolds LE, Jadeja S, Robinson S, Kostourou V, et al. Endothelial FAK is required for tumour angiogenesis. EMBO Mol Med. 2016;8(10):1229.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Zhou F, Sun J, Ye L, Jiang T, Li W, Su C, et al. Fibronectin promotes tumor angiogenesis and progression of non-small-cell lung cancer by elevating WISP3 expression via FAK/MAPK/ HIF-1alpha axis and activating wnt signaling pathway. Exp Hematol Oncol. 2023;12(1):61.PubMedPubMedCentralCrossRef Zhou F, Sun J, Ye L, Jiang T, Li W, Su C, et al. Fibronectin promotes tumor angiogenesis and progression of non-small-cell lung cancer by elevating WISP3 expression via FAK/MAPK/ HIF-1alpha axis and activating wnt signaling pathway. Exp Hematol Oncol. 2023;12(1):61.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Hochwald SN, Nyberg C, Zheng M, Zheng D, Wood C, Massoll NA, et al. A novel small molecule inhibitor of FAK decreases growth of human pancreatic cancer. Cell Cycle. 2009;8(15):2435–43.PubMedCrossRef Hochwald SN, Nyberg C, Zheng M, Zheng D, Wood C, Massoll NA, et al. A novel small molecule inhibitor of FAK decreases growth of human pancreatic cancer. Cell Cycle. 2009;8(15):2435–43.PubMedCrossRef
32.
Zurück zum Zitat Stokes JB, Adair SJ, Slack-Davis JK, Walters DM, Tilghman RW, Hershey ED, et al. Inhibition of focal adhesion kinase by PF-562,271 inhibits the growth and metastasis of pancreatic cancer concomitant with altering the tumor microenvironment. Mol Cancer Ther. 2011;10(11):2135–45.PubMedPubMedCentralCrossRef Stokes JB, Adair SJ, Slack-Davis JK, Walters DM, Tilghman RW, Hershey ED, et al. Inhibition of focal adhesion kinase by PF-562,271 inhibits the growth and metastasis of pancreatic cancer concomitant with altering the tumor microenvironment. Mol Cancer Ther. 2011;10(11):2135–45.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat Med. 2016;22(8):851–60.PubMedPubMedCentralCrossRef Jiang H, Hegde S, Knolhoff BL, Zhu Y, Herndon JM, Meyer MA, et al. Targeting focal adhesion kinase renders pancreatic cancers responsive to checkpoint immunotherapy. Nat Med. 2016;22(8):851–60.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Higashino N, Koma YI, Hosono M, Takase N, Okamoto M, Kodaira H, et al. Fibroblast activation protein-positive fibroblasts promote tumor progression through secretion of CCL2 and interleukin-6 in esophageal squamous cell carcinoma. Lab Invest. 2019;99(6):777–92.PubMedCrossRef Higashino N, Koma YI, Hosono M, Takase N, Okamoto M, Kodaira H, et al. Fibroblast activation protein-positive fibroblasts promote tumor progression through secretion of CCL2 and interleukin-6 in esophageal squamous cell carcinoma. Lab Invest. 2019;99(6):777–92.PubMedCrossRef
36.
Zurück zum Zitat Kumar V, Donthireddy L, Marvel D, Condamine T, Wang F, Lavilla-Alonso S, et al. Cancer-associated fibroblasts neutralize the anti-tumor effect of CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. Cancer Cell. 2017;32(5):654-668 e655.PubMedPubMedCentralCrossRef Kumar V, Donthireddy L, Marvel D, Condamine T, Wang F, Lavilla-Alonso S, et al. Cancer-associated fibroblasts neutralize the anti-tumor effect of CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. Cancer Cell. 2017;32(5):654-668 e655.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Tan X, Yan Y, Song B, Zhu S, Mei Q, Wu K. Focal adhesion kinase: from biological functions to therapeutic strategies. Exp Hematol Oncol. 2023;12(1):83.PubMedPubMedCentralCrossRef Tan X, Yan Y, Song B, Zhu S, Mei Q, Wu K. Focal adhesion kinase: from biological functions to therapeutic strategies. Exp Hematol Oncol. 2023;12(1):83.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Serrels A, Lund T, Serrels B, Byron A, McPherson RC, von Kriegsheim A, et al. Nuclear FAK controls chemokine transcription, tregs, and evasion of anti-tumor immunity. Cell. 2015;163(1):160–73.PubMedPubMedCentralCrossRef Serrels A, Lund T, Serrels B, Byron A, McPherson RC, von Kriegsheim A, et al. Nuclear FAK controls chemokine transcription, tregs, and evasion of anti-tumor immunity. Cell. 2015;163(1):160–73.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Konen J, Wilkinson S, Lee B, Fu H, Zhou W, Jiang Y, et al. LKB1 kinase-dependent and -independent defects disrupt polarity and adhesion signaling to drive collagen remodeling during invasion. Mol Biol Cell. 2016;27(7):1069–84.PubMedPubMedCentralCrossRef Konen J, Wilkinson S, Lee B, Fu H, Zhou W, Jiang Y, et al. LKB1 kinase-dependent and -independent defects disrupt polarity and adhesion signaling to drive collagen remodeling during invasion. Mol Biol Cell. 2016;27(7):1069–84.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Kline ER, Shupe J, Gilbert-Ross M, Zhou W, Marcus AI. LKB1 represses focal adhesion kinase (FAK) signaling via a FAK-LKB1 complex to regulate FAK site maturation and directional persistence. J Biol Chem. 2013;288(24):17663–74.PubMedPubMedCentralCrossRef Kline ER, Shupe J, Gilbert-Ross M, Zhou W, Marcus AI. LKB1 represses focal adhesion kinase (FAK) signaling via a FAK-LKB1 complex to regulate FAK site maturation and directional persistence. J Biol Chem. 2013;288(24):17663–74.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Barker HE, Bird D, Lang G, Erler JT. Tumor-secreted LOXL2 activates fibroblasts through FAK signaling. Mol Cancer Res. 2013;11(11):1425–36.PubMedCrossRef Barker HE, Bird D, Lang G, Erler JT. Tumor-secreted LOXL2 activates fibroblasts through FAK signaling. Mol Cancer Res. 2013;11(11):1425–36.PubMedCrossRef
42.
Zurück zum Zitat Dangaj D, Bruand M, Grimm AJ, Ronet C, Barras D, Duttagupta PA, et al. Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors. Cancer Cell. 2019;35(6):885-900 e810.PubMedPubMedCentralCrossRef Dangaj D, Bruand M, Grimm AJ, Ronet C, Barras D, Duttagupta PA, et al. Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors. Cancer Cell. 2019;35(6):885-900 e810.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Zhang B, Zhang Y, Zhang J, Liu P, Jiao B, Wang Z, et al. Focal adhesion kinase (FAK) inhibition synergizes with KRAS G12C inhibitors in treating cancer through the regulation of the FAK-YAP signaling. Adv Sci (Weinh). 2021;8(16): e2100250.PubMedCrossRef Zhang B, Zhang Y, Zhang J, Liu P, Jiao B, Wang Z, et al. Focal adhesion kinase (FAK) inhibition synergizes with KRAS G12C inhibitors in treating cancer through the regulation of the FAK-YAP signaling. Adv Sci (Weinh). 2021;8(16): e2100250.PubMedCrossRef
45.
Zurück zum Zitat Mohan N, Hosain S, Zhao J, Shen Y, Luo X, Jiang J, et al. Atezolizumab potentiates Tcell-mediated cytotoxicity and coordinates with FAK to suppress cell invasion and motility in PD-L1(+) triple negative breast cancer cells. Oncoimmunology. 2019;8(9): e1624128.PubMedPubMedCentralCrossRef Mohan N, Hosain S, Zhao J, Shen Y, Luo X, Jiang J, et al. Atezolizumab potentiates Tcell-mediated cytotoxicity and coordinates with FAK to suppress cell invasion and motility in PD-L1(+) triple negative breast cancer cells. Oncoimmunology. 2019;8(9): e1624128.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Peng DH, Rodriguez BL, Diao L, Chen L, Wang J, Byers LA, et al. Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8(+) T cell exhaustion. Nat Commun. 2020;11(1):4520.PubMedPubMedCentralCrossRef Peng DH, Rodriguez BL, Diao L, Chen L, Wang J, Byers LA, et al. Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8(+) T cell exhaustion. Nat Commun. 2020;11(1):4520.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Peng L, Ran YL, Hu H, Yu L, Liu Q, Zhou Z, et al. Secreted LOXL2 is a novel therapeutic target that promotes gastric cancer metastasis via the Src/FAK pathway. Carcinogenesis. 2009;30(10):1660–9.PubMedCrossRef Peng L, Ran YL, Hu H, Yu L, Liu Q, Zhou Z, et al. Secreted LOXL2 is a novel therapeutic target that promotes gastric cancer metastasis via the Src/FAK pathway. Carcinogenesis. 2009;30(10):1660–9.PubMedCrossRef
48.
Zurück zum Zitat Sturm G, Finotello F, Petitprez F, Zhang JD, Baumbach J, Fridman WH, et al. Comprehensive evaluation of transcriptome-based cell-type quantification methods for immuno-oncology. Bioinformatics. 2019;35(14):i436–45.PubMedPubMedCentralCrossRef Sturm G, Finotello F, Petitprez F, Zhang JD, Baumbach J, Fridman WH, et al. Comprehensive evaluation of transcriptome-based cell-type quantification methods for immuno-oncology. Bioinformatics. 2019;35(14):i436–45.PubMedPubMedCentralCrossRef
Metadaten
Titel
Targeting focal adhesion kinase boosts immune response in KRAS/LKB1 co-mutated lung adenocarcinoma via remodeling the tumor microenvironment
verfasst von
Meng Qiao
Fei Zhou
Xinyu Liu
Tao Jiang
Haowei Wang
Xuefei Li
Chao Zhao
Lei Cheng
Xiaoxia Chen
Shengxiang Ren
Zaiqi Wang
Caicun Zhou
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
Experimental Hematology & Oncology / Ausgabe 1/2024
Elektronische ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-023-00471-6

Weitere Artikel der Ausgabe 1/2024

Experimental Hematology & Oncology 1/2024 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.