A crucial mode of action of trastuzumab is the labeling of HER2-positive (HER2+) tumor cells for the eradication by natural killer (NK) cells, a process called antibody-dependent cellular cytotoxicity (ADCC). However, despite widespread HER2 expression among cancer entities, only a fraction, with robust HER2 overexpression, benefits from trastuzumab therapy. ADCC requires both sufficient lymphocytic infiltration and close binding of the immune cells to the antibody-tagged tumor cells. We hypothesized that the chemokine CX3CL1 could improve both processes, as it is synthesized as a membrane-bound, adhesive form that is eventually cleaved into a soluble, chemotactic protein. Here, we show that CX3CL1 overexpression is a positive prognostic marker in breast cancer. CX3CL1 overexpression attracted tumor-suppressive lymphocytes, including NK cells, and inhibited tumor growth and lung metastasis in the syngeneic 4T1 breast cancer mouse model. In HER2+ SKBR3, MDA-MB-453, and HT-29 tumor cells, CX3CL1 overexpression increased NK cell–mediated cytotoxicity in vitro and acted synergistically with trastuzumab. Even though CX3CL1 did not further improve trastuzumab efficacy in vivo in the trastuzumab-sensitive MDA-MB-453 model, it compensated for NK-cell depletion and prolonged survival. In the HER2 low–expressing HT-29 model, however, CX3CL1 overexpression not only prolonged survival time but also overcame trastuzumab resistance in a partly NK cell–dependent manner. Taken together, these findings identify CX3CL1 as a feasible pharmacologic target to enable trastuzumab therapy in HER2 low–expressing cancers and render it a potential predictive biomarker to determine therapy responders.

The mAb trastuzumab, directed against the HER2 oncogene, has led to an unprecedented improvement in the therapy of HER2-positive early and advanced breast and metastatic gastric cancer, paving the way for many other immunotherapies to follow (1). However, despite the fact that the HER2 protein is detectable in the majority of breast cancers, only about 25% of all breast cancers express it at levels that allow successful anti-HER2 treatment. Other cancers that also express HER2 (e.g., colon cancer) do not satisfactorily benefit from an anti-HER2 therapy (2).

Apart from the inhibition of HER2 downstream proliferative signaling pathways, trastuzumab's mechanism of action encompasses labeling of the tumor cells for their recognition and destruction by natural killer (NK) cells by a process called antibody-dependent cellular cytotoxicity (ADCC; refs. 3–5). NK cells trigger specific antitumor T-cell immunity (3). The importance of these cellular immune mechanisms is clinically underscored by studies demonstrating that the number of tumor-infiltrating lymphocytes is predictive of trastuzumab sensitivity (6, 7). Thus, raising the intratumoral accumulation of NK cells and other tumor-suppressive lymphocytes might be a feasible approach to enhance trastuzumab efficacy (8).

The chemokine CX3CL1, the only member of the CX3C subfamily of chemokines, is capable of recruiting NK and T cells to the tumor microenvironment (9). It is first synthesized as a membrane-bound form that allows binding of CX3CR1-positive immune cells to CX3CL1-positive target cells; it is then cleaved, primarily by ADAM proteases (e.g., ADAM17), to exert its chemotactic function as a soluble chemokine (10). These two properties render CX3CL1 a promising molecule to improve trastuzumab-mediated immune responses in HER2-expressing tumors.

However, there are conflicting data on the functional and prognostic relevance of CX3CL1 in cancer. Although most in vivo studies support an antitumor effect through immune activation (11–13), there are also data on the involvement of CX3CL1 in early tumorigenesis, for example, in HER2-positive breast cancer, via EGF pathway transactivation (14). Overexpression of CX3CL1 is associated with enhanced immune infiltration and a better prognosis in several cancer types (15–20). In pancreatic cancer, however, the opposite effect is reported (21).

Apart from its possible role in the very early stages of tumorigenesis, the functional role of CX3CL1 in breast cancer has not yet been explored. We speculated that overexpression of CX3CL1 might be a feasible way to improve the therapeutic success of trastuzumab by enhancing both the recruitment and the binding of NK cells to the trastuzumab-tagged tumor cells. In this study, we demonstrated that CX3CL1 was a protective chemokine in an immunocompetent breast cancer mouse model and acted synergistically with trastuzumab in NK cell–mediated ADCC in vitro. In vivo, CX3CL1 did not further improve the therapeutic success in trastuzumab-sensitive HER2-overexpressing cancer. However, its overexpression alone was sufficient to enable trastuzumab therapy in HER2 low–expressing, originally trastuzumab-resistant cancer. Our results support the idea that CX3CL1 might be a predictive marker of trastuzumab sensitivity in HER2 low–expressing cancers, and that its pharmacologic induction might be a feasible adjuvant to trastuzumab therapy.

Cell lines and cell culture

Human tumor cell lines: SKBR3 (ATCC: HTB-30 breast), MDA-MB-453 (ATCC: HTB-131 breast), BT474 (ATCC: HTB-20), MCF7 (ATCC: HTB-22 breast), MDA-MB-231 (ATCC: HTB-26 breast), and HT-29 (ATCC: HTB-38 colorectal). Mouse tumor cell lines: 4T1 (ATCC: CRL-2539 breast). Cells were cultured in RPMI medium (cell lines: BT474, MCF7, MDA-MB-231, 4T1) or McCoy's Medium (GIBCO, cell lines: SKBR3, MDA-MB-453, HT-29) provided with 10% [volume for volume (v/v)] heat-inactivated FCS, 0.272 mmol/L l-asparagine, 0.550 mmol/L arginine, and 10 mmol/L HEPES buffer at 37°C and 5% (v/v) CO2. Cell passaging was performed with 0.025% trypsin/0.01% EDTA in PBS (treatment less than 2 minutes). Cell lines were authenticated by DNA profiling of eight short tandem repeat sequences and were further controlled for the presence of mouse and rat DNA (authentication service DSMZ). The human cell lines were validated before submission of this work. All the cell lines are regularly tested for Mycoplasma contamination via PCR.

Vector DNA (pCMV-6-mCX3CL1, pCMV-6-hCX3CL1, pCMV-6-Entry, Origene) was stably transfected in human and murine breast cancer cells with Lipofectin (Thermo Fisher Scientific). Transfected cells were selected with neomycin (Thermo Fisher Scientific) for 14 days, and positive clones were selected for further expansion and testing. Proliferation of cell clones was tested by seeding 1 × 104 cells in 24-well plates and counting for 4 days. Transient transfection was performed without further neomycin selection. Cells were kept in culture for 24 hours after transfection and were then used for the in vitro experiments.

Tissue samples and IHC

For the IHC studies, formalin-fixed, paraffin-embedded tumor specimens from 250 patients with early breast cancer treated at the Department of Gynecology and Obstetrics (Technical University of Munich, Munich, Germany) between 2005 and 2012 were used, stored in an interdisciplinary tissue bank (Department of Pathology, Technical University of Munich, Munich, Germany). Patients with distant metastases or after neoadjuvant chemotherapy at the time of surgery were excluded from the study. Detailed patient characteristics are given in Supplementary Table S1. The HER2-low (n = 178) and HER2-overexpressing cohorts (n = 25) were subgroups of this larger cohort. In another cohort, only HER2-overexpressing tumors treated neoadjuvantly with trastuzumab were included (n = 69). The study was approved by the Institutional Review Board of the Technical University of Munich (Munich, Germany; approval 227/17S) in accordance with the Declaration of Helsinki. Written informed consent was obtained from all patients.

Tissue microarrays were generated as described previously (22). Mouse tissue was fixed in 4% paraformaldehyde for 48 hours and paraffin embedded. Both human and murine tissue was processed to 3 μm sections. The slides were deparaffinized by xylene and hydrolyzed by a descending alcohol row (100%–50%). Heat-induced antigen retrieval was performed in citrate buffer (pH 6.0) before endogenous peroxidase activity was blocked with 3% (v/v) H2O2 (20 minutes), followed by an additional blocking step with 5% goat serum in antibody dilution solution (ZUC025-500, Zytomed). Samples were then incubated with anti-CX3CL1 (1 μg/mL in antibody dilution solution, clone AF365, R&D Systems), F4/80 antibody (1:300 in antibody dilution solution, clone D2S9R, Cell Signaling Technology), or anti-CD49b (2.5 ng/mL, BioLegend) for 1 hour at room temperature. Detection was performed using the polymer one-step system (Zytomed) and 3,3′-Diaminobenzidine (DAB; Zytomed) as substrate. Slides were counterstained with hematoxylin (Merck). Slides were washed thoroughly with TBST (0.1% Tween 20) between each step. Evaluation of the staining was performed by using a semiquantitative score based on the staining intensity of tumor tissue: absent (0), weak (1+), moderate (2+), or strong (3+; Fig. 1A). Evaluators were blinded to the clinical data. In cases with positive staining, all tumor cells were positive, and therefore, percentage of positivity was not included in our evaluation.

Figure 1.

CX3CL1 is a prognostic marker in human breast cancer, inhibits tumor growth, and enhances lymphocytic infiltration in murine breast cancer in vivo. A, CX3CL1 expression was immunohistochemically determined in human breast cancer samples and semiquantitatively scored on a four-tier scale (n = 250; cohort summary Supplementary Table S1). Bars, 50 μm. B, Distribution of CX3CL1 expression in human breast cancer according to the receptor-defined breast cancer subtypes: ERposHER2neg (n = 203), HER2pos (n = 25), and triple-negative breast cancer (TNBC; n = 22). C, Kaplan–Meier plot for PFS stratified by CX3CL1 expression among CX3CL1-positive cases (CX3CL1low, n = 67; CX3CL1medium, n = 103; and CX3CL1high, n = 55). Significance was tested with the log-rank test. D–G, 4T1 murine breast cancer cells were stably transfected with a murine CX3CL1 (Cx3cl1) expression vector (4T1-Cx3cl1+) or a control vector (4T1). A total of 1 × 104 cells were subcutaneously implanted into the mammary fat pad of Balb/c mice, and mice were sacrificed after 21 days. D, Tumor volumes over time presented as mean ± SEM. E, Tumor weight of resected tumors after 21 days. F, Pulmonary metastatic burden was quantified by determining the number of 6-thioguanine–resistant selected 4T1 colonies grown in vitro from isolated lung tissue. G, Number of different intratumoral lymphocyte subpopulations as percentage of the total cell number. Data in DG are cumulative from at least two independent experiments with at least 6 mice/group. Statistical significance (P values) was calculated using two-way ANOVA (D) or the Mann–Whitney U test (EG). Horizontal lines in EG represent means. Each dot in EG represents an individual mouse.

Figure 1.

CX3CL1 is a prognostic marker in human breast cancer, inhibits tumor growth, and enhances lymphocytic infiltration in murine breast cancer in vivo. A, CX3CL1 expression was immunohistochemically determined in human breast cancer samples and semiquantitatively scored on a four-tier scale (n = 250; cohort summary Supplementary Table S1). Bars, 50 μm. B, Distribution of CX3CL1 expression in human breast cancer according to the receptor-defined breast cancer subtypes: ERposHER2neg (n = 203), HER2pos (n = 25), and triple-negative breast cancer (TNBC; n = 22). C, Kaplan–Meier plot for PFS stratified by CX3CL1 expression among CX3CL1-positive cases (CX3CL1low, n = 67; CX3CL1medium, n = 103; and CX3CL1high, n = 55). Significance was tested with the log-rank test. D–G, 4T1 murine breast cancer cells were stably transfected with a murine CX3CL1 (Cx3cl1) expression vector (4T1-Cx3cl1+) or a control vector (4T1). A total of 1 × 104 cells were subcutaneously implanted into the mammary fat pad of Balb/c mice, and mice were sacrificed after 21 days. D, Tumor volumes over time presented as mean ± SEM. E, Tumor weight of resected tumors after 21 days. F, Pulmonary metastatic burden was quantified by determining the number of 6-thioguanine–resistant selected 4T1 colonies grown in vitro from isolated lung tissue. G, Number of different intratumoral lymphocyte subpopulations as percentage of the total cell number. Data in DG are cumulative from at least two independent experiments with at least 6 mice/group. Statistical significance (P values) was calculated using two-way ANOVA (D) or the Mann–Whitney U test (EG). Horizontal lines in EG represent means. Each dot in EG represents an individual mouse.

Close modal

ELISA

Tumor cell lines (SKBR3, MDA-MB-453, BT474, and HT-29) were seeded in 12-well plates (1.4 × 105 cells) and grown to a confluence of 80%. Afterward, cells were washed with PBS and cultivated in serum-free medium for 24 hours before test reagents were added for another 24 hours. Final concentrations were as follows: TNFα (10 ng/mL, Peprotech), TAPI-2 (7.5 μmol/L, Tocris). For determination of the transfection efficacy, cell supernatants were taken from unstarved, unstimulated cells. For measurement of human CX3CL1, mouse CX3CL1, and human CXCL10, we used the DuoSet ELISA Kits DY365, DY472, and DY266, respectively, according to the manufacturer's recommendations (R&D Systems). Triplicates of 100 μL undiluted supernatant were analyzed (Multiskan FC, Thermo Fisher Scientific). The absolute target protein concentration was calculated based on a predefined recombinant protein standard.

Western blot analysis

Tumor cells from stimulation experiments were collected and immediately lysed with RIPA buffer [1% Triton X100, complete protease inhibitor cocktail (Roche), 1 mmol/L vanadate, 1 mmol/L glycerol phosphate, 50 mmol/L sodium fluoride, 10 mmol/L sodium pyrophosphate in TBS (pH 7.4), all chemicals from Merck]. The samples were incubated for 20 minutes on ice and then treated with ultrasound (Sonopuls, Bandelin) for 20 seconds. The concentration of total protein was determined using the BCA Protein Assay Kit (Pierce, Thermo Fisher Scientific). Protein samples (30 μg) were separated by a 10% SDS-PAGE and blotted onto a 0.45 μm nitrocellulose membrane by wet blotting. After blocking (5% milk powder in TBST for 1 hour at room temperature), incubation with the primary antibodies anti-CX3CL1 (clone MAB365, R&D Systems, 1 μg/mL), anti–HER-2 (A0485, Dako, 1:1,000), or anti-GAPDH (Millipore, 0.125 μg/mL) in 2.5% milk powder/TBST was performed for 1 hour at room temperature. Secondary anti-goat (Life Technologies) or anti-mouse horseradish peroxidase (HRP)–conjugated antibody (Jackson Immuno Research) was used 1:10,000 for 1 hour at room temperature. Detection was performed using secondary HRP-conjugated antibodies and ECL reagent (Thermo Fisher Scientific) with the Bio-Rad detection system. A thorough washing step with TBST was performed between each step.

Cell lysis assay

NK cells were freshly isolated from a leukapheresis product of three different volunteer donors. CD19+ (Miltenyi Biotec) and CD3+ (Miltenyi Biotec) cells were separated from the NK cells in a negative selection approach using the cliniMACS system (Miltenyi Biotec). NK-cell enrichment was determined using FACS analysis for CD56 (BD Biosciences) and CD45 (Invitrogen). Purity of NK-cell populations was determined as described before (23, 24), for the NK cells used a purity >80% was achieved.

The NK cell–mediated killing of tumor cells was assessed using a dissociation-enhanced lanthanide fluorescent immunoassay (DELFIA, PerkinElmer) with time-resolved fluorescence detection. NK cells were stimulated for 3 days with IL2 (500 U/mL, Peprotech). Stably or transiently transfected cancer cells, as well as cells treated with 10 ng/mL TNFα or 7.5 μmol/L TAPI-2, were collected, adjusted to 5 × 105 cells, and stained with the 0.5% DELFIA BATDA fluorescent reagent in 200 μL RPMI medium without phenol red. Afterward, the cells were treated with trastuzumab (40 μg/mL, Roche), anti-CX3CL1/goat IgG (20 μg/mL, clone AF365, R&D Systems), and/or soluble CX3CL1 (10 ng/mL, amino acids 1–76, PeproTech) for 30 minutes. NK cells and tumor cells (5 × 104 cells) were coincubated for 4 hours at three different ratios (1:10, 1:5, and 1:2.5) in V-bottom 96-well plates. After incubation, the released DELFIA BATDA was detected with europium solution using a time-resolved fluorometer (Victor Wallac 3). The spontaneous lysis was subtracted, and all the values were normalized to a corresponding maximum lysis control as described by the manufacturer (DELFIA Cell cytotoxicity assay, PerkinElmer).

Animals

Eight- to 10-week-old wild-type Balb/cAnNCrl mice (strain 028), SCID mice (CB17/Icr-Prkdcscid/IcrIcoCrl, strain 236), and SCID Beige mice (CB17.Cg-PrkdcscidLystbg-J/Crl, strain 250) were obtained from Charles River Laboratory. Mice were maintained in a pathogen-free animal facility, and all animal procedures and experiments were approved by the Government of Upper Bavaria (Regierung von Oberbayern) and were in accordance with the institutional guidelines of the Technical University of Munich (Munich, Germany).

Tumor growth in vivo

Cell lines (4T1, HT-29, MDA-MB-453) were used after confirmation of Cx3cl1 overexpression and negative Mycoplasma testing. Mice were subcutaneously injected with 1 × 104 4T1 cells, 2 × 106 HT-29, or 2 × 106 MDA-MB-453 cells in PBS into the fourth mammary fat pad. Tumor cells were washed in PBS before injection. The syngeneic 4T1 mouse experiments were terminated 21 days after tumor inoculation. Mice bearing HT-29 or MDA-MB-453 tumors were culled after the tumor diameter reached 1.5 cm. The depletion of NK cells was performed by intraperitoneal injection of 100 μL anti-asialo GM1 (986-10001, FUIFILM Wako Chemicals) or 100 μL corresponding rabbit serum as control twice a week, starting 1 day after tumor inoculation. Treatment with trastuzumab was started once the tumor diameter reached 0.5 cm. The mice were treated with 10 mg/kg trastuzumab i.p. twice a week or vehicle control. In the MDA-MB-453 model, mice were treated for 3 weeks in total. In the HT-29 model, mice were treated until the end of the experiment (tumor size). The tumor diameter was measured using calipers every 72 hours, and the tumor volume was calculated (largest diameter2 × smallest diameter × 0.5). Right before the finalization of the animals, blood was taken from the vena facialis, stored on ice for 30 minutes, and centrifuged at 10,000 rcf for 10 minutes. Serum was taken and stored at −80°C for further use. Directly after the mice were euthanized, tumor tissue, spleen, kidney, and heart were removed, and stored in liquid nitrogen and 4% buffered formalin for 48 hours. Tissue for FACS analysis or colony formation was stored in PBS until further use.

qPCR

Fresh-frozen tumor tissues (20 mg) from mice were mechanically disrupted, and total RNA isolated using the RNeasy Plus Kit (Qiagen). A total of 1 μg of RNA was then used for reverse transcription (Super Script IV, Thermo Fisher Scientific). Quality of the RNA was checked by agarose gel electrophoresis and by NanoDrop (Thermo Fisher Scientific). A total of 210 ng of cDNA were used for the qRT-PCR analysis applying Universal Probe system (Roche). The following combination of primers and probes was used: Cx3cl1 (Probe#80, up: 5′-GGC TTT GCT CAT CCG CTA T-3′, down: 5′- CAG AAG CGT CTG TGC TGT GT-3′), mHprt (Probe#95, up: 5′-CCT CCT CAG ACC GCT TTT T-3′. Down: 5′-AAC CTG GTT CAT CAT CGC TAA-3′). The relative expression values were compared with the housekeeping gene (Hprt) expression using the ΔΔCT method (Lightcycler MX3005, Agilent).

Flow cytometry

For analysis of tumor infiltrates, 4T1 tumor tissues were dissected, cut into small pieces, and digested in RPMI medium containing Liberase TL (0.5 mg/mL; Roche) and Dnase I (0.25 mg/mL; Roche) for 45 minutes at 37°C. Reaction was stopped using 0.5 M EDTA solution (GIPCO). After filtration through a 70 μm cell strainer, cells were counted, and 5 × 106 cells were used for cell surface staining. The following fluorophore-conjugated antibodies were used: anti-mCD3 (clone 17A2, BioLegend, 2.5 μg/mL), anti-mCD4 (clone RM4-5, eBioscience, 1.3 μg/mL), anti-mCD8 (clone53-7.3, eBioscience, 1 μg/mL), anti-mCD49b (DX5, eBioscience, 1 μg/mL), anti-mFoxp3 (FJK-16s, eBioscience, 2.5 μg/mL), anti-mCD25 (PC61, BioLegend, 0.5 μg/mL), anti-mCD11b (M1/70, eBioscience, 0.8 μg/mL), anti-mF4/80 (BM8, eBioscience, 1.3 μg/mL), anti-mLy-6C (HK1.4, BioLegend, 1.3 μg/mL), and anti-mLy6G (1A8, BioLegend, 1.7 μg/mL). For analysis of CX3CL1 membrane expression of tumor cells, anti-CX3CL1 (clone AF365, R&D Systems, 200 μg/mL) was used together with an Alexa488 secondary fluorescent dye (Abcam, 6 ng/μL).

Detection of dead cells was performed with fixable viability dye eFluor 506 (eBioscience), and cell suspensions were incubated with mCD16/CD32 Fc-blocking antibody (553142, BD Biosciences) for 10 minutes. before the antibody staining was performed. Flow cytometry was performed using the FACS Canto 2 (BD Biosciences) together with the DIVA software. Data analysis was done with the FlowJoV10 software (Treestar).

Determination of lung metastases

Lung tissues of 4T1-inoculated mice were collected directly after finalization, and digestion was performed as described above for primary tumor tissue. The cell suspensions were then resuspended in selection RPMI medium containing 6-thioguanine (5 μg/mL; Merck) and 50 U/mL penicillin/50 μg/mL streptomycin. Increasing dilutions (1:10, 1:100, and 1:1,000) were prepared, and cells moved to 10-cm cell culture dishes containing 10 mL of selection RPMI medium. After incubation at 37°C for 2 weeks, colonies were counted after fixation with 100% methanol.

Published dataset analysis

For further exploratory survival analyses, we used a publicly available Affymetrix dataset (retrievable at www.kmplot.com; refs. 25, 26). All cases of breast cancer were included in the analysis (n = 3951) using best cutoff as selected by the program.

Statistical analysis

Analysis of in vitro data was performed using a one-way ANOVA. Kaplan–Meier estimates of event-free survival were compared by log-rank tests. Hypothesis testing of in vivo experiments, in detail the differences in immune cell populations, were done with the Mann–Whitney U tests. All tests were performed on exploratory two-sided 5% levels of significance (SPSS Statistics Software, Version 25.0, SPSS Inc.).

CX3CL1 inhibits tumor growth and metastasis in breast cancer

To unravel the role of CX3CL1 in breast cancer, we first determined its expression and localization immunohistochemically in the breast tumor microenvironment (TME) of 250 cases of early breast cancer (Supplementary Table S1). CX3CL1 was mainly localized in tumor and endothelial cells, and tumor cell CX3CL1 expression was semiquantitatively scored on a four-tier scale based on the staining intensity (Fig. 1A). Within individual tumors, there was hardly any heterogeneity in CX3CL1 expression. CX3CL1 expression was evenly distributed among the classical, receptor-defined breast cancer subtypes (Fig. 1B), and there was no relevant correlation with tumor size, lymph node involvement, or nuclear grading. In CX3CL1-expressing breast cancers, CX3CL1 expression was positively associated with progression-free survival (PFS; Fig. 1C). The relative 5-year PFS was 74%, 84%, and 89% for CX3CL1 IHC 1+, 2+, and 3+ cases, and the relative 10-year PFS was 61%, 69%, and 79% for CX3CL1 IHC 1+, 2+, and 3+ cases, respectively. There was no significant difference in overall survival between the CX3CL1 expression groups. Our results are in line with the prognostic value of CX3CL1 mRNA in publicly available breast cancer datasets (Supplementary Fig. S1; ref. 25).

We next sought to define the functional impact of CX3CL1 overexpression in a fully immunocompetent mouse model of breast cancer. Because our IHC studies identified tumor cells as the major source of CX3CL1 in the TME, we stably transfected murine 4T1 breast cancer cells to express murine CX3CL1 (Cx3cl1) using a vector (Supplementary Fig. S2A) and orthotopically implanted them into the mammary fat pad of Balb/c mice. Cx3cl1 overexpression did not affect cell proliferation in vitro (Supplementary Fig. S2B). However, in vivo, Cx3cl1 overexpression resulted in delayed tumor growth (Fig. 1D) and significantly less tumor weight after 21 days (Fig. 1E) compared with vector control cells (median, 0.16 vs. 0.11 g; P = 0.04). Although tumor Cx3cl1 overexpression was still measurable after 21 days (Supplementary Fig. S2C), Cx3cl1 serum concentrations were not enhanced in the mice harboring Cx3cl1-overexpressing tumors (Supplementary Fig. S2D), suggesting rather locally restricted effects of the chemokine. Nevertheless, Cx3cl1 almost completely prevented the formation of lung metastases (median number of 6-thioguanine–resistant colonies 185 vs. 0; P = 0.004; Fig. 1F). Cx3cl1-overexpressing tumors showed a significantly enhanced infiltration by CD3CD49b+ NK cells (0.35% vs. 0.45%; P = 0.06), CD3+CD4+ T cells (1.66% vs. 2.47%; P = 0.0015), and CD3+CD8+ cytotoxic T cells (0.35% vs. 0.64%; P = 0.04; Fig. 1G). However, regulatory T cells (Treg, CD3+CD4+FOXP3+) were not considerably elevated (0.55% vs. 0.71%; P = 0.22). LyG6+LyC6+ myeloid cells were significantly diminished in Cx3cl1-overexpressing tumors, whereas no change was seen in F4/80+MHC II+ macrophages or CD11b+ cells (Supplementary Fig. S2E). These results demonstrate the protective nature of CX3CL1 in breast cancer. The induction of NK-cell infiltration supported our hypothesis that CX3CL1 might be advantageous, especially for therapies dependent on the presence of NK cells, such as trastuzumab (3).

Membrane-bound and soluble CX3CL1 are expressed in HER2+ tumor cell lines

To test a possible exploitation of CX3CL1 in anti-HER2 therapy, we first examined the expression and regulation of CX3CL1 in the HER2-positive cancer cell lines SKBR3, MDA-MB-453, BT474, and HT-29. These cell lines expressed HER2 to variable degrees (Fig. 2A). All cell lines, except MDA-MB-453, secreted soluble CX3CL1 upon stimulation with TNFα, a potent inducer of CX3CL1 in noncancerous cells (Fig. 2B; ref. 27). SKBR3 and BT474 cells secreted significant amounts of CX3CL1 also in the absence of an inflammatory stimulus. The fraction of secreted CX3CL1 could be reduced upon ADAM17 (a disintegrin and metalloproteinase 17) inhibition by TAPI-2, demonstrating involvement of this protease in CX3CL1 shedding from the surface of tumor cells (Fig. 2B). This effect was not due to ADAM17-mediated modifications of the TNFα response because the secretion of CXCL10, which is also induced by TNFα (28), was not considerably altered (Supplementary Fig. S3A). MDA-MB-453 cells did not release any soluble CX3CL1, neither at baseline nor upon TNFα stimulation (Fig. 2B), although these cells were principally responsive to TNFα (Supplementary Fig. S3B). FACS analyses of vital cells to determine the membrane-bound CX3CL1 and Western blot analyses of whole-cell lysates confirmed the induction of CX3CL1 by TNFα concomitant with an increase of membrane-bound CX3CL1 upon ADAM17 inhibition in SKBR3, BT474, and HT-29 cells (Fig. 2C and D). In MDA-MB-453 cells, however, apart from intracellular baseline CX3CL1 expression, still no induction by TNFα and no regulation by ADAM17 inhibition was observed. Altogether, these data demonstrate that some HER2-positive cancer cell lines intrinsically express both membrane-bound and soluble CX3CL1, some need inflammatory stimuli to induce CX3CL1 expression, and others do not express significant amounts of CX3CL1.

Figure 2.

CX3CL1 is expressed as a membrane-bound protein, which is partly shed into the soluble chemokine in HER2-positive tumor cell lines. A, Western blot analysis of HER2 expression in different HER2-positive cancer cell lines. HER2-negative MCF7 and MDA-MB-231 cells served as negative controls. B, Impact of TNFα (10 ng/mL, 24 hours) and/or the ADAM17 inhibitor TAPI-2 (7.5 μmol/L, 24 hours) on CX3CL1 release into the cell culture supernatants of HER2-positive cancer cell lines measured by ELISA (n = 3, presented as mean ± SEM). Significance was determined by one-way ANOVA. *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, FACS analyses of membrane-bound CX3CL1 in viable HER2-positive cell lines. D, Representative immunoblot analyses of total cellular CX3CL1 expression in HT-29 and MDA-MB-453 cells under same conditions as in B.

Figure 2.

CX3CL1 is expressed as a membrane-bound protein, which is partly shed into the soluble chemokine in HER2-positive tumor cell lines. A, Western blot analysis of HER2 expression in different HER2-positive cancer cell lines. HER2-negative MCF7 and MDA-MB-231 cells served as negative controls. B, Impact of TNFα (10 ng/mL, 24 hours) and/or the ADAM17 inhibitor TAPI-2 (7.5 μmol/L, 24 hours) on CX3CL1 release into the cell culture supernatants of HER2-positive cancer cell lines measured by ELISA (n = 3, presented as mean ± SEM). Significance was determined by one-way ANOVA. *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, FACS analyses of membrane-bound CX3CL1 in viable HER2-positive cell lines. D, Representative immunoblot analyses of total cellular CX3CL1 expression in HT-29 and MDA-MB-453 cells under same conditions as in B.

Close modal

CX3CL1 enhances NK cell–mediated lysis of HER2+ cells synergistically with trastuzumab

Having characterized CX3CL1 expression and regulation in HER2-positive cancer cell lines, we next determined its impact on NK cell–mediated cytotoxicity and trastuzumab-mediated ADCC. Because preincubation of target cells with TNFα has been shown to enhance NK cell–mediated cytotoxicity in noncancerous cells (29), we examined whether this effect was also detectable in HER2-positive tumor cells and whether it was affected by CX3CL1. To this end, tumor cells were preincubated for 24 hours with or without TNFα prior to the addition of NK cells, and cytotoxicity was measured using a standard europium release assay. In both SKBR3 and HT-29 cells, TNFα led to an increase of specific tumor cell lysis by approximately 80% and 15%, respectively, which could be partially blocked by a monoclonal anti-CX3CL1 (Fig. 3A; Supplementary Fig. S4A). In the CX3CL1-negative, TNFα-responsive MDA-MB-453 cells, TNFα did not have any effect on cell lysis (Fig. 3A). These results suggest an involvement of CX3CL1 in TNFα-enhanced NK cell–mediated tumor cell lysis.

Figure 3.

CX3CL1 improves NK cell–mediated lysis of HER2-positive tumor cells and enhances their trastuzumab sensitivity. A, Four-hour Europium cytotoxicity assay showing the effect of TNFα on NK cell–mediated tumor cell lysis and its dependency on CX3CL1. Tumor cells (target cells) were incubated with TNFα (10 ng/mL) and/or a monoclonal anti-CX3CL1 or control IgG (20 μg/mL) for 30 minutes and coincubated with NK cells (effector cells) at three different effector:target ratios (E:T ratio) for 4 hours. Values were normalized to the corresponding BSA/control IgG–treated group of each experiment (three independent experiments, in triplicates). B, Effect of trastuzumab and CX3CL1 overexpression on NK cell–mediated tumor cell lysis using the same approach as in A. Tumor cells were transfected with a CX3CL1 expression vector or a respective control vector. Trastuzumab was given at a concentration of 40 μg/mL. Data are from three independent experiments (triplicates, results presented as mean ± SEM). Lower cases indicate statistical significance (P < 0.05). Significance was determined by one-way ANOVA.

Figure 3.

CX3CL1 improves NK cell–mediated lysis of HER2-positive tumor cells and enhances their trastuzumab sensitivity. A, Four-hour Europium cytotoxicity assay showing the effect of TNFα on NK cell–mediated tumor cell lysis and its dependency on CX3CL1. Tumor cells (target cells) were incubated with TNFα (10 ng/mL) and/or a monoclonal anti-CX3CL1 or control IgG (20 μg/mL) for 30 minutes and coincubated with NK cells (effector cells) at three different effector:target ratios (E:T ratio) for 4 hours. Values were normalized to the corresponding BSA/control IgG–treated group of each experiment (three independent experiments, in triplicates). B, Effect of trastuzumab and CX3CL1 overexpression on NK cell–mediated tumor cell lysis using the same approach as in A. Tumor cells were transfected with a CX3CL1 expression vector or a respective control vector. Trastuzumab was given at a concentration of 40 μg/mL. Data are from three independent experiments (triplicates, results presented as mean ± SEM). Lower cases indicate statistical significance (P < 0.05). Significance was determined by one-way ANOVA.

Close modal

Next, we wanted to know whether overexpression of CX3CL1 was per se sufficient to enhance NK cell–mediated tumor cell lysis and capable of acting synergistically with trastuzumab. CX3CL1 was stably overexpressed in the three cell lines SKBR3, HT-29, and MDA-MB-453, respectively (Supplementary Fig. S4B), which were exposed to NK cells in the presence or absence of trastuzumab (Fig. 3B). CX3CL1 enhanced NK cell–mediated cytotoxicity by approximately 10% in SKBR3 and HT-29 cells and by 35% in MDA-MB-453 cells. As expected, trastuzumab alone caused an increase in tumor cell lysis of about 50% in the two HER2-overexpressing cell lines SKBR3 and MDA-MB-453 and of nearly 20% in HER2-weak HT-29 cells. In all three cell lines, there was a synergistic effect of CX3CL1 overexpression and trastuzumab with a specific tumor cell lysis reaching almost 100% in HT-29 and MDA-MB-453 cells (Fig. 3B). This synergism could not be mimicked by the addition of soluble CX3CL1 (chemokine domain, amino acids 1–76) to tumor cells and NK cells in wild-type HT-29 and MDA-MB-453 cells, suggesting that the membrane-bound form of CX3CL1 is primarily responsible for the described CX3CL1 effects in these cell lines (Supplementary Fig. S4C). In line with this, the increase in specific tumor cell lysis by the addition of the ADAM17 inhibitor TAPI-2 could be partially abrogated by a CX3CL1-blocking antibody in HT-29 cells (Supplementary Fig. S4D). Taken together, these results showed that CX3CL1 conferred susceptibility of tumor cells to NK cell–mediated cytotoxicity and acted synergistically with trastuzumab in HER2-positive cancer cells in vitro.

CX3CL1 enables trastuzumab therapy in HER2 low–expressing cancer in vivo

Given the synergism of CX3CL1 overexpression and trastuzumab on tumor cell lysis in vitro, we next tested whether CX3CL1 would enhance trastuzumab therapy in HER2-positive cancer also in vivo. To this end, we used xenotransplantation models in Balb/SCID mice, which lack adaptive immunity but still retain NK-cell activity, thus allowing us to study NK cell–mediated effects. First, we tested the role of CX3CL1 in the trastuzumab-sensitive MDA-MB-453 tumor model, which lacks intrinsic CX3CL1 (Fig. 2B; Supplementary Fig. S5A). After tumor implantation, mice were randomized to either receive trastuzumab or vehicle, and NK-cell depletion using an anti-asialo GM1 or an IgG control. Basophils, which are also depleted by the anti-asialo GM1 antibody, do not express CX3CR1 and were therefore not considered relevant to the study of CX3CL1 effects (30). Anti-HER2 treatment (or vehicle control) was initiated after the tumor had reached a diameter of 5 mm. In vehicle-treated mice, overexpression of CX3CL1 generally resulted in a significantly prolonged survival time of the mice (median, 161 vs. 132 days; P = 0.002; Fig. 4A). The initial tumor growth was slightly accelerated in the CX3CL1-overexpressing tumors (time to therapy start 34 vs. 41 days; P = 0.14). Although we generally detected only isolated DX5+ NK cells by IHC in the MDA-MB-453 tumors (with therefore no measurable difference between the groups), NK-cell depletion significantly reduced survival in the non-CX3CL1–overexpressing group (median survival, 93 vs. 132 days; P < 0.0001). However, overexpression of CX3CL1 was able to compensate for NK-cell depletion, as there was no substantial survival difference between NK cell–depleted and nondepleted mice (median survival, 165 vs. 161 days; P = 0.63; Fig. 4A). In this model, small amounts of trastuzumab were already sufficient to induce complete tumor regression (Supplementary Fig. S5B), which is in line with prior reports in these HER2-overexpressing breast cancer models (31). Herein, NK-cell depletion had no effect on tumor growth and survival, suggesting that NK cell–mediated ADCC is less important than inhibition of HER2 signaling in these HER2-overexpressing tumors. The intratumoral frequency of macrophages, also known to express the CX3CR1 receptor, was not significantly different between the groups (Supplementary Fig. S5C).

Figure 4.

CX3CL1 enables trastuzumab treatment in HER2 low–expressing cancer in vivo. A, MDA-MB-453 cells were stably transfected with a murine CX3CL1 expression vector (Cx3cl1+) or vector control (VC), and 2 × 106 cells were implanted subcutaneously into Balb/c SCID mice (5–15 animals per group). Animals were randomized into the intraperitoneal treatment groups (trastuzumab vs. vehicle, NK-cell depletion by anti-asialo-GM1 vs. control IgG). Kaplan–Meier plot showing the survival of MDA-MB-453 inoculated animals. B, PFS of patients with HER2 low–expressing breast cancer (IHC score 1+ or 2+/FISH negative) stratified by CX3CL1 expression as determined by IHC (CX3CL1low, n = 14; CX3CL1medium, n = 86; and CX3CL1high, n = 78). C, HT-29 cells were stably transfected with a murine CX3CL1 expression vector (Cx3cl1+) or vector control, and 2 × 106 cells were subcutaneously implanted into Balb/c SCID or SCID Beige mice. Animals were randomized to receive trastuzumab or placebo (vehicle) treatment (at least 16 animals/group). NK-cell infiltration was quantified by IHC DX5 staining of formalin-fixed tumor tissue after dissection. Horizontal lines are medians. Each dot represents an individual mouse. Significance was determined using the Mann–Whitney U test. D, Tumor growth curves of the four subgroups in HT-29–bearing animals (SCID). Kaplan–Meier survival plot of Balb/c SCID mice (E) and SCID Beige mice (F) bearing Cx3cl1+ or vector control HT-29 tumors treated with or without trastuzumab. Dashed lines represent survival curves from the SCID experiment normalized to the control group of the SCID Beige model. G, Intratumoral F4/80+ cells were quantified by IHC in formalin-fixed tumor tissue after dissection. Kaplan–Meier and DX5 and F4/80 data are cumulative from at least two independent core experiments with at least 7 mice per group. Significance in A and B and E and F was tested with the log-rank test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Figure 4.

CX3CL1 enables trastuzumab treatment in HER2 low–expressing cancer in vivo. A, MDA-MB-453 cells were stably transfected with a murine CX3CL1 expression vector (Cx3cl1+) or vector control (VC), and 2 × 106 cells were implanted subcutaneously into Balb/c SCID mice (5–15 animals per group). Animals were randomized into the intraperitoneal treatment groups (trastuzumab vs. vehicle, NK-cell depletion by anti-asialo-GM1 vs. control IgG). Kaplan–Meier plot showing the survival of MDA-MB-453 inoculated animals. B, PFS of patients with HER2 low–expressing breast cancer (IHC score 1+ or 2+/FISH negative) stratified by CX3CL1 expression as determined by IHC (CX3CL1low, n = 14; CX3CL1medium, n = 86; and CX3CL1high, n = 78). C, HT-29 cells were stably transfected with a murine CX3CL1 expression vector (Cx3cl1+) or vector control, and 2 × 106 cells were subcutaneously implanted into Balb/c SCID or SCID Beige mice. Animals were randomized to receive trastuzumab or placebo (vehicle) treatment (at least 16 animals/group). NK-cell infiltration was quantified by IHC DX5 staining of formalin-fixed tumor tissue after dissection. Horizontal lines are medians. Each dot represents an individual mouse. Significance was determined using the Mann–Whitney U test. D, Tumor growth curves of the four subgroups in HT-29–bearing animals (SCID). Kaplan–Meier survival plot of Balb/c SCID mice (E) and SCID Beige mice (F) bearing Cx3cl1+ or vector control HT-29 tumors treated with or without trastuzumab. Dashed lines represent survival curves from the SCID experiment normalized to the control group of the SCID Beige model. G, Intratumoral F4/80+ cells were quantified by IHC in formalin-fixed tumor tissue after dissection. Kaplan–Meier and DX5 and F4/80 data are cumulative from at least two independent core experiments with at least 7 mice per group. Significance in A and B and E and F was tested with the log-rank test. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant.

Close modal

Supporting this lack of synergism of CX3CL1 expression and trastuzumab efficacy in MDA-MB-453, we did not find a significant association of CX3CL1 expression and PFS in patients with HER2-overexpressing (IHC 2+/FISH-positive, IHC 3+), trastuzumab-treated breast cancer (Supplementary Fig. S6A). In a second cohort of patients with HER2-overexpressing breast cancer who were treated neoadjuvantly with trastuzumab and chemotherapy, we did not observe a substantial correlation of pretherapeutic CX3CL1 expression and therapy response (Supplementary Fig. S6B). However, in the subgroup of CX3CL1-positive, HER2 low–expressing breast cancers (IHC score 1+ or 2+/FISH negative), we found a significant association of CX3CL1 expression and PFS (Fig. 4B).

We therefore tested the interaction of CX3CL1 and trastuzumab in the HER2 low–expressing HT-29 model. Stable transfection of HT-29 cells with a murine CX3CL1 expression vector (Supplementary Fig. S7A and S7B) caused a significant, about 4-fold increase, in NK-cell infiltration into the tumors (Fig. 4C). Trastuzumab, however, had no impact on NK-cell infiltration. As in the 4T1 and the MDA-MB-453 models, we again observed an accelerated initial tumor growth phase in the Cx3cl1-overexpressing tumors (median time to first measurable tumor 9 vs. 12 days; P = 0.001; time to onset of therapy 12 vs. 15 days; P = 0.03). Subsequently, Cx3cl1 overexpression led to a significantly reduced tumor growth compared with control mice (Fig. 4D), and median survival after onset of therapy was prolonged by Cx3cl1 overexpression (21 vs. 14 days; P = 0.038; Fig. 4E). Although trastuzumab had no significant effect in the absence of Cx3cl1, it significantly slowed down tumor growth and improved survival in Cx3cl1-overexpressing tumors (median survival time, 36 vs. 21 days; P < 0.0001). To further explore the contribution of NK cells to these effects, we performed the same experiment in SCID Beige mice lacking NK cells. Here, virtually no intratumoral DX5+ NK cells were detectable by IHC. Overexpression of Cx3cl1 had no impact on tumor growth or survival anymore (Fig. 4F; Supplementary Fig. S7C). Cx3cl1 and trastuzumab still had a synergistic effect, but it was significantly reduced compared with the effect seen in SCID mice (adjusted median survival difference between Cx3cl1+ and Cx3cl1+/trastuzumab 22.5 days in SCID vs. 15.5 days in SCID Beige, P < 0.0001). These results demonstrate an involvement of NK cells in the Cx3cl1-induced survival benefit and its synergism with trastuzumab in vivo. However, they also suggest other mechanisms to contribute. Because macrophages also express CX3CR1 and facilitate antibody-dependent cellular phagocytosis (32, 33), we determined their intratumoral accumulation. In contrast to our findings in the 4T1 and the MDA-MB-453 models, in both HT-29 experiments, we detected a significant increase in F4/80-positive macrophages upon Cx3cl1 overexpression, which might also participate in the observed CX3CL1/trastuzumab synergism (Fig. 4G). In summary, our data provide evidence that CX3CL1 is capable of overcoming trastuzumab resistance in HER2 low–expressing cancer and might be a suitable biomarker for the selection of patients for trastuzumab therapy in this cohort.

One of the key questions in contemporary immuno-oncology is how to transform a “cold,” less inflamed tumor into a “hot” tumor, enriched in immune cells. This is a fundamental prerequisite for the success of most immunotherapies, including immune checkpoint inhibitors and mAbs directed against tumor antigens (34). The poorly explored chemokine CX3CL1 does not only attract tumor-suppressive lymphocytes, it might also enhance their binding to the target tumor cells due to its presence as a membrane-bound form (10). In this study, we provide functional evidence for this concept, exemplified by the trastuzumab treatment of HER2-expressing cancer. We found that overexpression of CX3CL1 not only improved NK- and T-cell infiltration and inhibited tumor growth and metastatic spread, but also enhanced NK-cell killing of trastuzumab-tagged tumor cells in vitro. In vivo, CX3CL1 was sufficient to overcome trastuzumab resistance in a preclinical model of HER2 low–expressing cancer.

Previous studies demonstrate that intratumoral CX3CL1 recruits NK and T cells and impairs tumor growth in preclinical models of lymphoma, hepatocellular carcinoma, colon, and lung cancer (11, 35–38). In contrast, there are reports suggesting that CX3CL1 might foster migration and metastatic spread of CX3CR1-positive tumor cells to distant, CX3CL1-enriched metastatic sites such as brain or bone (39, 40). This, however, could also add to the protective nature of intratumoral CX3CL1 expression, as local CX3CL1 might prevent CX3CR1-positive tumor cells from leaving the primary tumor. Tardáguila and colleagues (14) demonstrate that CX3CL1 participates in the tumorigenesis of HER2-positive breast cancer via EGFR pathway transactivation, but it does not affect growth of established tumors. In line with this, we observed an accelerated initial tumor growth in the CX3CL1-overexpressing tumors in both the MDA-MB-453 and the HT-29 tumor models, and only a late separation of tumor growth curves in the 4T1 model. However, in the further course of the experiments, CX3CL1 suppressed tumor growth and improved survival in all three tumor models. In support of this, we observed a significant association of tumor cell–associated CX3CL1 and PFS in our breast cancer cohort, which could be confirmed in publicly available mRNA databases (25). Park and colleagues (20) reported similar results in their breast cancer cohort, in which CX3CL1 was positively associated with the number of CD8+ T cells, CD57+ NK cells, as well as disease-free and overall survival. Several groups report a downregulation of CX3CL1 in breast cancer compared with normal breast tissue, which might promote immune escape (14, 41–43). In summary, although CX3CL1 might play a role in EGFR-mediated breast cancer tumorigenesis, our results in different breast cancer models support a protective role in established breast cancer, which is further backed by clinical data.

Forced overexpression of CX3CL1 was sufficient to augment NK-cell cytotoxicity against tumor cells in the current work. Previously, CX3CL1 is demonstrated to induce NK cell–mediated damage of endothelial cells (44, 45). CX3CL1-overexpressing HeLa cells or soluble CX3CL1 triggers IFNγ release and lytic activity of NK-92 cells in vitro (46, 47). Our results showed that the enhancement of NK-cell cytotoxicity by TNFα, which has been described before (29), can be, at least in part, traced back to CX3CL1, as the inhibition by a monoclonal anti-CX3CL1 suppressed TNFα-mediated cytotoxicity, and TNFα had no effect in CX3CL1-negative but per se TNF-responsive MDA-MB-453 cells. However, it is still not entirely clear which CX3CL1 isoform is responsible for these effects. There is evidence for a stronger stimulation of CX3CR1 downstream processes, such as Ca2+ influx or IFNγ secretion, in NK cells by the membrane-bound CX3CL1 form in vitro (48, 49). However, the only study in cancer that differentiates between the effects of membrane-bound and soluble CX3CL1, found antitumor activity of soluble CX3CL1 and a mixed to no effect of membrane-bound chemokine in vivo (35). We approached this question by adding recombinant soluble CX3CL1 to the tumor cell–NK cell coincubation, which did not have any substantial effect. On the other hand, the increase in NK-cell lytic activity by ADAM17 inhibition was partially abrogated by CX3CL1 blockage, again implicating a role for membrane-bound CX3CL1 in this enhanced cytotoxicity. This is in line with reports showing that ADAM17 confers resistance to trastuzumab (50, 51). It is tempting to speculate that the cleavage of membrane-bound CX3CL1 might participate in this process.

Our results demonstrate that CX3CL1 acts in concert with trastuzumab by improving NK-cell attraction and cytotoxicity, which might be exemplary for other ADCC-dependent therapies. In a cohort of 53 patients with HER2-positive, trastuzumab-treated breast cancer, expression of CX3CL1 was associated with a reduced risk of relapse (52). In the trastuzumab-treated, HER2 high–expressing cohort, we did not find a correlation of trastuzumab response and CX3CL1 expression. This supports our in vivo finding of a lacking synergism in the HER2-overexpressing tumor model. However, in patients with HER2 low–expressing breast cancer, we observed a positive association with PFS. As these patients had not been treated with trastuzumab according to the current guidelines, we do not know whether CX3CL1 is also predictive for trastuzumab response. In the HER2-overexpressing MDA-MB-453 tumor model, we observed neither an improvement of trastuzumab therapy by CX3CL1 overexpression nor a dependence on NK cells because a short trastuzumab treatment already induced complete tumor regression. This could be explained by the notion that immunoregulatory effects of trastuzumab are less important in HER2-high tumors in which the mere inactivation of the HER2 receptor activity is crucial and possibly sufficient (53).

The current study focused on the role of NK cells in CX3CL1-mediated tumor suppression and trastuzumab therapy. However, our results in SCID Beige mice show that other mechanisms might also participate, for example, the enhanced recruitment of macrophages observed in the HT-29 model. Macrophages utilize the CX3CR1 receptor for chemotaxis (54) and are capable of enhancing trastuzumab action through antibody-dependent cellular phagocytosis (33). However, they have been reported to be an adverse prognostic marker in trastuzumab-treated HER2-positive breast cancer (55), possibly because their polarization state determines the impact they have on anti-HER2 treatment (56). The next step now will be to clarify their role in CX3CL1-mediated antitumor response and trastuzumab efficacy.

In conclusion, our results demonstrate that CX3CL1 overexpression improves trastuzumab efficacy in HER2 low–expressing cancer in vitro and in vivo. Besides being a potential predictor of response in these cancers that are currently not treated with trastuzumab, pharmacologic enrichment of CX3CL1 in the TME might be a feasible way to enhance the clinical efficacy of trastuzumab therapy. The next steps are now to decipher ways to increase CX3CL1 expression in cancer cells. Besides the induction of its expression, the inhibition of its proteolytic inactivation might be a promising approach (10). This could potentially open targeted anti-HER2 therapy to a large number of patients.

T.F. Dreyer reports grants from DFG during the conduct of the study. C. Stange reports grants from Deutsche Forschungsgemeinschaft during the conduct of the study. S. Seitz reports grants from DFG during the conduct of the study. A.K. Wege reports grants from German Research Foundation (Deutsche Forschungsgemeinschaft, DFG) during the conduct of the study. W. Weichert reports personal fees from Roche, MSD, Bristol Myers Squibb, AstraZeneca, Pfizer, Merck, Lilly, Boehringer, Novartis, Takeda, Bayer, Amgen, Astellas, Illumina, Siemens, Agilent, and Molecular Health for advisory boards/speaker programs and grants from Roche, MSD, Bristol Myers Squibb, and AstraZeneca (to institution) outside the submitted work. M. Kiechle reports other support from Theraws Diagnostic GmbH and AIM GmbH, and personal fees from AstraZeneca, Celgene, Eli Lilly, Exeltis, Teva, and Myriad Genetics during the conduct of the study. H. Bronger reports grants from Deutsche Forschungsgemeinschaft (DFG), German Research Foundation during the conduct of the study, as well as personal fees from Roche, AstraZeneca, GlaxoSmithKline, Clovis, Teva, PharmaMar, and Pfizer outside the submitted work. No disclosures were reported by the other authors.

T.F. Dreyer: Conceptualization, data curation, software, supervision, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing. S. Kuhn: Investigation, visualization, writing–original draft, writing–review and editing. C. Stange: Validation. N. Heithorst: Data curation, validation, investigation, writing–original draft, writing–review and editing. D. Schilling: Conceptualization, resources, formal analysis, supervision, investigation, writing–original draft, writing–review and editing. J. Jelsma: Investigation, writing–original draft, writing–review and editing. W. Sievert: Conceptualization, resources, supervision, investigation, writing–original draft, writing–review and editing. S. Seitz: Validation, writing–original draft, writing–review and editing. S. Stangl: Resources, formal analysis, investigation, writing–original draft, writing–review and editing. A. Hapfelmeier: Software, formal analysis, writing–original draft, writing–review and editing. A. Noske: Formal analysis, validation, investigation, writing–original draft, writing–review and editing. A.K. Wege: Funding acquisition, investigation, writing–original draft, writing–review and editing. W. Weichert: Resources, supervision, writing–original draft, project administration, writing–review and editing. J. Ruland: Resources, writing–original draft, project administration, writing–review and editing. M. Schmitt: Conceptualization, supervision, funding acquisition, writing–original draft, writing–review and editing. J. Dorn: Resources, funding acquisition, writing–original draft, writing–review and editing. M. Kiechle: Resources, supervision, funding acquisition, writing–original draft, project administration, writing–review and editing. U. Reuning: Resources, writing–original draft, writing–review and editing. V. Magdolen: Conceptualization, resources, data curation, formal analysis, supervision, funding acquisition, writing–original draft, project administration, writing–review and editing. G. Multhoff: Conceptualization, resources, supervision, writing–original draft, writing–review and editing. H. Bronger: Conceptualization, resources, data curation, formal analysis, supervision, funding acquisition, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing.

The authors thank Christine Huber for excellent technical assistance. This work was supported by project grants from the German Research Foundation (Deutsche Forschungsgemeinschaft, DFG; BR4733/2-1, to H. Bronger and WE3606/3-1, to A.K. Wege).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Baselga
J
. 
Treatment of HER2-overexpressing breast cancer
.
Ann Oncol
2010
;
21
:
vii36
40
.
2.
Meric-Bernstam
F
,
Johnson
AM
,
Dumbrava
EEI
,
Raghav
K
,
Balaji
K
,
Bhatt
M
, et al
Advances in HER2-targeted therapy: novel agents and opportunities beyond breast and gastric cancer
.
Clin Cancer Res
2019
;
25
:
2033
41
.
3.
Muntasell
A
,
Cabo
M
,
Servitja
S
,
Tusquets
I
,
Martinez-Garcia
M
,
Rovira
A
, et al
Interplay between natural killer cells and anti-HER2 antibodies: perspectives for breast cancer immunotherapy
.
Front Immunol
2017
;
8
:
1544
.
4.
Clynes
RA
,
Towers
TL
,
Presta
LG
,
Ravetch
JV
. 
Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets
.
Nat Med
2000
;
6
:
443
6
.
5.
Beano
A
,
Signorino
E
,
Evangelista
A
,
Brusa
D
,
Mistrangelo
M
,
Polimeni
MA
, et al
Correlation between NK function and response to trastuzumab in metastatic breast cancer patients
.
J Transl Med
2008
;
6
:
25
.
6.
Ingold Heppner
B
,
Untch
M
,
Denkert
C
,
Pfitzner
BM
,
Lederer
B
,
Schmitt
W
, et al
Tumor-infiltrating lymphocytes: a predictive and prognostic biomarker in neoadjuvant-treated HER2-positive breast cancer
.
Clin Cancer Res
2016
;
22
:
5747
54
.
7.
Salgado
R
,
Denkert
C
,
Campbell
C
,
Savas
P
,
Nuciforo
P
,
Aura
C
, et al
Tumor-infiltrating lymphocytes and associations with pathological complete response and event-free survival in HER2-positive early-stage breast cancer treated with lapatinib and trastuzumab: a secondary analysis of the NeoALTTO Trial
.
JAMA Oncol
2015
;
1
:
448
54
.
8.
Andre
F
,
Dieci
MV
,
Dubsky
P
,
Sotiriou
C
,
Curigliano
G
,
Denkert
C
, et al
Molecular pathways: involvement of immune pathways in the therapeutic response and outcome in breast cancer
.
Clin Cancer Res
2013
;
19
:
28
33
.
9.
Chen
DS
,
Mellman
I
. 
Oncology meets immunology: the cancer-immunity cycle
.
Immunity
2013
;
39
:
1
10
.
10.
Bronger
H
,
Magdolen
V
,
Goettig
P
,
Dreyer
T
. 
Proteolytic chemokine cleavage as a regulator of lymphocytic infiltration in solid tumors
.
Cancer Metastasis Rev
2019
;
38
:
417
30
.
11.
Lavergne
E
,
Combadiere
B
,
Bonduelle
O
,
Iga
M
,
Gao
JL
,
Maho
M
, et al
Fractalkine mediates natural killer-dependent antitumor responses in vivo
.
Cancer Res
2003
;
63
:
7468
74
.
12.
Xin
H
,
Kikuchi
T
,
Andarini
S
,
Ohkouchi
S
,
Suzuki
T
,
Nukiwa
T
, et al
Antitumor immune response by CX3CL1 fractalkine gene transfer depends on both NK and T cells
.
Eur J Immunol
2005
;
35
:
1371
80
.
13.
Zeng
Y
,
Huebener
N
,
Fest
S
,
Weixler
S
,
Schroeder
U
,
Gaedicke
G
, et al
Fractalkine (CX3CL1)- and interleukin-2-enriched neuroblastoma microenvironment induces eradication of metastases mediated by T cells and natural killer cells
.
Cancer Res
2007
;
67
:
2331
8
.
14.
Tardáguila
M
,
Mira
E
,
Garcia-Cabezas
MA
,
Feijoo
AM
,
Quintela-Fandino
M
,
Azcoitia
I
, et al
CX3CL1 promotes breast cancer via transactivation of the EGF pathway
.
Cancer Res
2013
;
73
:
4461
73
.
15.
Hyakudomi
M
,
Matsubara
T
,
Hyakudomi
R
,
Yamamoto
T
,
Kinugasa
S
,
Yamanoi
A
, et al
Increased expression of fractalkine is correlated with a better prognosis and an increased number of both CD8+ T cells and natural killer cells in gastric adenocarcinoma
.
Ann Surg Oncol
2008
;
15
:
1775
82
.
16.
Kehlen
A
,
Greither
T
,
Wach
S
,
Nolte
E
,
Kappler
M
,
Bache
M
, et al
High coexpression of CCL2 and CX3CL1 is gender-specifically associated with good prognosis in soft tissue sarcoma patients
.
Int J Cancer
2014
;
135
:
2096
106
.
17.
Liu
J
,
Li
Y
,
Zhu
X
,
Li
Q
,
Liang
X
,
Xie
J
, et al
Increased CX3CL1 mRNA expression level is a positive prognostic factor in patients with lung adenocarcinoma
.
Oncol Lett
2019
;
17
:
4877
90
.
18.
Mlecnik
B
,
Tosolini
M
,
Charoentong
P
,
Kirilovsky
A
,
Bindea
G
,
Berger
A
, et al
Biomolecular network reconstruction identifies T-cell homing factors associated with survival in colorectal cancer
.
Gastroenterology
2010
;
138
:
1429
40
.
19.
Ohta
M
,
Tanaka
F
,
Yamaguchi
H
,
Sadanaga
N
,
Inoue
H
,
Mori
M
. 
The high expression of Fractalkine results in a better prognosis for colorectal cancer patients
.
Int J Oncol
2005
;
26
:
41
7
.
20.
Park
MH
,
Lee
JS
,
Yoon
JH
. 
High expression of CX3CL1 by tumor cells correlates with a good prognosis and increased tumor-infiltrating CD8+ T cells, natural killer cells, and dendritic cells in breast carcinoma
.
J Surg Oncol
2012
;
106
:
386
92
.
21.
Xu
X
,
Wang
Y
,
Chen
J
,
Ma
H
,
Shao
Z
,
Chen
H
, et al
High expression of CX3CL1/CX3CR1 axis predicts a poor prognosis of pancreatic ductal adenocarcinoma
.
J Gastrointest Surg
2012
;
16
:
1493
8
.
22.
Seiz
L
,
Dorn
J
,
Kotzsch
M
,
Walch
A
,
Grebenchtchikov
NI
,
Gkazepis
A
, et al
Stromal cell-associated expression of kallikrein-related peptidase 6 (KLK6) indicates poor prognosis of ovarian cancer patients
.
Biol Chem
2012
;
393
:
391
401
.
23.
Krause
SW
,
Gastpar
R
,
Andreesen
R
,
Gross
C
,
Ullrich
H
,
Thonigs
G
, et al
Treatment of colon and lung cancer patients with ex vivo heat shock protein 70-peptide-activated, autologous natural killer cells: a clinical phase I trial
.
Clin Cancer Res
2004
;
10
:
3699
707
.
24.
Gross
C
,
Schmidt-Wolf
IG
,
Nagaraj
S
,
Gastpar
R
,
Ellwart
J
,
Kunz-Schughart
LA
, et al
Heat shock protein 70-reactivity is associated with increased cell surface density of CD94/CD56 on primary natural killer cells
.
Cell Stress Chaperones
2003
;
8
:
348
60
.
25.
Gyorffy
B
,
Lanczky
A
,
Eklund
AC
,
Denkert
C
,
Budczies
J
,
Li
Q
, et al
An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients
.
Breast Cancer Res Treat
2010
;
123
:
725
31
.
26.
Nagy
A
,
Munkacsy
G
,
Gyorffy
B
. 
Pancancer survival analysis of cancer hallmark genes
.
Sci Rep
2021
;
11
:
6047
.
27.
Ludwig
A
,
Weber
C
. 
Transmembrane chemokines: versatile ‘special agents’ in vascular inflammation
.
Thromb Haemost
2007
;
97
:
694
703
.
28.
Bronger
H
,
Singer
J
,
Windmuller
C
,
Reuning
U
,
Zech
D
,
Delbridge
C
, et al
CXCL9 and CXCL10 predict survival and are regulated by cyclooxygenase inhibition in advanced serous ovarian cancer
.
Br J Cancer
2016
;
115
:
553
63
.
29.
Wang
R
,
Jaw
JJ
,
Stutzman
NC
,
Zou
Z
,
Sun
PD
. 
Natural killer cell-produced IFN-gamma and TNF-alpha induce target cell cytolysis through up-regulation of ICAM-1
.
J Leukoc Biol
2012
;
91
:
299
309
.
30.
Iikura
M
,
Miyamasu
M
,
Yamaguchi
M
,
Kawasaki
H
,
Matsushima
K
,
Kitaura
M
, et al
Chemokine receptors in human basophils: inducible expression of functional CXCR4
.
J Leukoc Biol
2001
;
70
:
113
20
.
31.
Chen
J
,
Yang
C
,
Guo
B
,
Sena
ES
,
Macleod
MR
,
Yuan
Y
, et al
The efficacy of trastuzumab in animal models of breast cancer: a systematic review and meta-analysis
.
PLoS One
2016
;
11
:
e0158240
.
32.
Conroy
MJ
,
Lysaght
J
. 
CX3CL1 signaling in the tumor microenvironment
.
Adv Exp Med Biol
2020
;
1231
:
1
12
.
33.
Shi
Y
,
Fan
X
,
Deng
H
,
Brezski
RJ
,
Rycyzyn
M
,
Jordan
RE
, et al
Trastuzumab triggers phagocytic killing of high HER2 cancer cells in vitro and in vivo by interaction with Fcgamma receptors on macrophages
.
J Immunol
2015
;
194
:
4379
86
.
34.
Nagarsheth
N
,
Wicha
MS
,
Zou
W
. 
Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy
.
Nat Rev Immunol
2017
;
17
:
559
72
.
35.
Vitale
S
,
Cambien
B
,
Karimdjee
BF
,
Barthel
R
,
Staccini
P
,
Luci
C
, et al
Tissue-specific differential antitumour effect of molecular forms of fractalkine in a mouse model of metastatic colon cancer
.
Gut
2007
;
56
:
365
72
.
36.
Guo
J
,
Zhang
M
,
Wang
B
,
Yuan
Z
,
Guo
Z
,
Chen
T
, et al
Fractalkine transgene induces T-cell-dependent antitumor immunity through chemoattraction and activation of dendritic cells
.
Int J Cancer
2003
;
103
:
212
20
.
37.
Tang
L
,
Hu
HD
,
Hu
P
,
Lan
YH
,
Peng
ML
,
Chen
M
, et al
Gene therapy with CX3CL1/Fractalkine induces antitumor immunity to regress effectively mouse hepatocellular carcinoma
.
Gene Ther
2007
;
14
:
1226
34
.
38.
Kee
JY
,
Arita
Y
,
Shinohara
K
,
Ohashi
Y
,
Sakurai
H
,
Saiki
I
, et al
Antitumor immune activity by chemokine CX3CL1 in an orthotopic implantation of lung cancer model in vivo
.
Mol Clin Oncol
2013
;
1
:
35
40
.
39.
Andre
F
,
Cabioglu
N
,
Assi
H
,
Sabourin
JC
,
Delaloge
S
,
Sahin
A
, et al
Expression of chemokine receptors predicts the site of metastatic relapse in patients with axillary node positive primary breast cancer
.
Ann Oncol
2006
;
17
:
945
51
.
40.
Jamieson-Gladney
WL
,
Zhang
Y
,
Fong
AM
,
Meucci
O
,
Fatatis
A
. 
The chemokine receptor CX(3)CR1 is directly involved in the arrest of breast cancer cells to the skeleton
.
Breast Cancer Res
2011
;
13
:
R91
.
41.
Lacroix
M
,
Toillon
RA
,
Leclercq
G
. 
Stable ‘portrait’ of breast tumors during progression: data from biology, pathology and genetics
.
Endocr Relat Cancer
2004
;
11
:
497
522
.
42.
Perreard
L
,
Fan
C
,
Quackenbush
JF
,
Mullins
M
,
Gauthier
NP
,
Nelson
E
, et al
Classification and risk stratification of invasive breast carcinomas using a real-time quantitative RT-PCR assay
.
Breast Cancer Res
2006
;
8
:
R23
.
43.
Graham
K
,
Ge
X
,
de Las Morenas
A
,
Tripathi
A
,
Rosenberg
CL
. 
Gene expression profiles of estrogen receptor-positive and estrogen receptor-negative breast cancers are detectable in histologically normal breast epithelium
.
Clin Cancer Res
2011
;
17
:
236
46
.
44.
Gorini
S
,
Callegari
G
,
Romagnoli
G
,
Mammi
C
,
Mavilio
D
,
Rosano
G
, et al
ATP secreted by endothelial cells blocks CX(3)CL 1-elicited natural killer cell chemotaxis and cytotoxicity via P2Y(1)(1) receptor activation
.
Blood
2010
;
116
:
4492
500
.
45.
Yoneda
O
,
Imai
T
,
Goda
S
,
Inoue
H
,
Yamauchi
A
,
Okazaki
T
, et al
Fractalkine-mediated endothelial cell injury by NK cells
.
J Immunol
2000
;
164
:
4055
62
.
46.
Zhang
X
,
Wei
H
,
Chen
Q
,
Tian
Z
. 
Activation of human natural killer cells by recombinant membrane-expressed fractalkine on the surface of tumor cells
.
Oncol Rep
2007
;
17
:
1371
5
.
47.
Zhang
X
,
Wei
H
,
Wang
H
,
Tian
Z
. 
Involvement of interaction between Fractalkine and CX3CR1 in cytotoxicity of natural killer cells against tumor cells
.
Oncol Rep
2006
;
15
:
485
8
.
48.
Yoneda
O
,
Imai
T
,
Nishimura
M
,
Miyaji
M
,
Mimori
T
,
Okazaki
T
, et al
Membrane-bound form of fractalkine induces IFN-gamma production by NK cells
.
Eur J Immunol
2003
;
33
:
53
8
.
49.
Harrison
JK
,
Jiang
Y
,
Chen
S
,
Xia
Y
,
Maciejewski
D
,
McNamara
RK
, et al
Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia
.
Proc Natl Acad Sci U S A
1998
;
95
:
10896
901
.
50.
Feldinger
K
,
Generali
D
,
Kramer-Marek
G
,
Gijsen
M
,
Ng
TB
,
Wong
JH
, et al
ADAM10 mediates trastuzumab resistance and is correlated with survival in HER2 positive breast cancer
.
Oncotarget
2014
;
5
:
6633
46
.
51.
Duffy
MJ
,
Crown
J
,
Mullooly
M
. 
ADAM10 and ADAM17: new players in trastuzumab tesistance
.
Oncotarget
2014
;
5
:
10963
4
.
52.
Triulzi
T
,
Regondi
V
,
De Cecco
L
,
Ghedini
G
,
Carcangiu
ML
,
Pupa
SM
, et al
Tumor dependence on HER2 signaling as a player in immune infiltration required for trastuzumab activity
[abstract]
.
In
:
Proceedings of the 106th Annual Meeting of the American Association for Cancer Research
; 
2015
Apr 18–22
;
Philadelphia, PA. Philadelphia (PA)
:
AACR
;
Cancer Res 2015;75(15 Suppl):Abstract nr 5015
.
doi: 10.1158/1538-7445.AM2015-5015
.
53.
Pegram
M
,
Slamon
D
. 
Biological rationale for HER2/neu (c-erbB2) as a target for monoclonal antibody therapy
.
Semin Oncol
2000
;
27
:
13
9
.
54.
Panek
CA
,
Ramos
MV
,
Mejias
MP
,
Abrey-Recalde
MJ
,
Fernandez-Brando
RJ
,
Gori
MS
, et al
Differential expression of the fractalkine chemokine receptor (CX3CR1) in human monocytes during differentiation
.
Cell Mol Immunol
2015
;
12
:
669
80
.
55.
Honkanen
TJ
,
Tikkanen
A
,
Karihtala
P
,
Makinen
M
,
Vayrynen
JP
,
Koivunen
JP
. 
Prognostic and predictive role of tumour-associated macrophages in HER2 positive breast cancer
.
Sci Rep
2019
;
9
:
10961
.
56.
Xu
M
,
Liu
M
,
Du
X
,
Li
S
,
Li
H
,
Li
X
, et al
Intratumoral delivery of IL-21 overcomes anti-Her2/Neu resistance through shifting tumor-associated macrophages from M2 to M1 phenotype
.
J Immunol
2015
;
194
:
4997
5006
.