DKK1 augments breast cancer progression.
To determine the role of DKK1 in breast cancer progression, we first measured DKK1 serum levels in C57BL/6 mice orthotopically injected with luminal B, ER+, hormone-resistant PyMT-BO1 cell line into the mammary fat pad (MFP). Serum DKK1 levels were significantly increased 14 days post inoculation (Fig. 1A), recapitulating the elevated levels observed in breast cancer patients17. Next, we administered the DKK1-neutralizing monoclonal antibody mDKN01 (10mg/kg), every other day following tumor inoculation and found a significant reduction in primary tumor growth compared to isotype control (IgG) (Fig. 1B). To further investigate the role of DKK1 during tumor dissemination, we injected the firefly luciferase-conjugated PyMT-BO1 cell line into albino C57BL/6 mice either intracardiacally (i.c.) or in the tibias (i.t.), followed by treatment with mDKN01. Bioluminescence imaging (BLI) showed significantly reduced tumor growth at all sites, including to visceral organs (Fig. 1C, D) and bone (Fig. 1E, F).
DKK1 levels were also significantly increased 14 days post inoculation in BALB/c mice orthotopically injected with the triple-negative 4T1 tumor line (Fig. 1G). Administration of mDKN01 (10mg/kg) led to a significant reduction in primary tumor growth compared to IgG control (Fig. 1H). Similarly, DKK1 neutralization significantly reduced the growth of the luminal B, ER/PR+, hormone-sensitive EO771 breast cancer cell line (Supp Fig. 1A). These results demonstrate the involvement of DKK1 in supporting tumor progression and the therapeutic benefit of a DKK1 targeting agent in various breast cancer subtypes.
DKK1 is expressed in the tumor microenvironment and in bone.
In breast cancer, high levels of circulating DKK1 correlate with reduced survival and metastatic dissemination, especially to the bone15, 17. To assess expression of DKK1 at tumor site, we analyzed a single-cell RNA seq dataset from 26 primary human breast tumors, including 11 ER+ (Luminal A or B), 5 HER2+, and 10 triple-negative breast cancers (TNBC) (GSE17607830). UMAP visualization resulted in 4 different clusters (endothelial cells, immune cells, epithelial cells, and cancer-associated fibroblasts (CAFs)), annotated by using canonical and signature-based markers31 (Supp Fig. 2A, B). DKK1 was found to be mainly expressed in the cancer epithelial cells in the HER2+ subtype, and to a less extent in TNBC, while barely detectable in the normal epithelial cells (Fig. 2A). To our surprise, we also detected DKK1 expression, albeit limited, in the stromal compartment, where DKK1 was mainly detected within CAF clusters in TNBC, and to a lower extent also in the HER2+ and ER+ subtypes (Fig. 2B). Further subtype analysis based on previous breast cancer CAF classification32, 33 (Supp Fig. 2C, D), showed the highest DKK1 expression in ACTA2+COL1a1highPDGFRa+ myofibroblasts (Fig. 2C). Based on these findings, we performed multiplex immunohistochemistry (IHC) using human breast cancer tissue microarrays containing TNBC, HER2+ and ER+ subtypes. DKK1 staining was observed in some cancer epithelial cells, identified by the co-expression with a pan-cytokeratin (PanCK) marker, and in PDGRFa+ and aSMA+ stromal populations in all tumor types (Fig. 2D-F), while it was not detected in the terminal duct lobular unit of normal breast tissue (Supp Fig. 2E).
Next, we investigated DKK1 expression in the PyMT, 4T1, and EO771 tumor models. While Dkk1 was barely detectable in the tumor cell lines in vitro (Supp Fig. 1D), Dkk1 transcripts were detected in the tumor mass 14 days post-inoculation (Supp Fig. 1B, E). Similar to the patient samples, IHC showed DKK1 staining in stromal cells with an elongated, fibroblast-like morphology in the PyMT orthotopic tumors (Fig. 2G) and in the spontaneous MMTV-PyMT tumors (Fig. 2I). Further, co-staining with CAF markers, showed DKK1 colocalization with elongated aSMA+ cells and partial colocalization with the more ubiquitous CAF marker COL1a1 (Fig. 2H, J). In concordance with its undetectable transcript expression, DKK1 staining was not present in the murine cancer lines.
Because in homeostatic conditions DKK1 is known to be highly expressed in bone by osteoblast-lineage cells34, we also evaluated expression of bone-derived Dkk1 in mice bearing primary breast tumors. We found increased expression of Dkk1 in crushed bones devoid of marrow cells of tumor bearing mice compared to no tumor controls (Supp Fig. 1C, F). These results indicate the existence of different sources of DKK1, with local production of DKK1 at tumor site by either tumor cells and/or CAFs, and distal production of DKK1 by bone cells.
Bone and CAF-derived DKK1 contribute to systemic and local increases in DKK1 levels during tumor progression.
To determine the role of bone versus CAF-derived DKK1 during tumor progression, we generated mouse models with targeted deletion of DKK1 in osteoblasts and fibroblasts. To specifically delete Dkk1 from osteoblasts, we crossed Dkk1fl/fl mice with the doxycycline-repressible Sp7 Cre line (herein referred to as Sp7-Dkk1cKO)35. Because Dkk1 deletion leads to embryonic lethality, we fed moms and pups with a doxycycline-containing diet until weaning to suppress the transgene activation, and orthotopically injected the PyMT tumor cells when mice reached 6–8 weeks of age. Strikingly, Sp7-Dkk1cKO mice showed a significant reduction in primary tumor growth (Fig. 3A). While Dkk1 expression at tumor site was not reduced compared to littermate controls, DKK1 levels in circulation were drastically reduced (Fig. 3B-D), indicating that bone-derived DKK1 contributes to systemic elevation of DKK1 during tumor progression.
Based on the expression of DKK1 in aSMA+ cells in the tumor microenvironment, next, we crossed Dkk1fl/fl mice with the inducible aSMA CreERT2 line (referred to as aSMA-Dkk1cKO). We induced Cre activation by intraperitoneal administration of tamoxifen for 5 consecutive days (100mg/kg per dose) to 10–12 weeks old mice. As a control, we crossed the aSMACreERT2 mice with the Rosa26-LSL-tdTomato line (herein referred to as aSMA-tdT mice) and injected PyMT cells into their MFP to confirm the presence of aSMA-tdT+ cells exclusively at the tumor site, but not in the bone (Supp Fig. 3A). Intriguingly, aSMA-Dkk1cKO also showed to a significant reduction in primary tumor growth compared to littermate controls (Fig. 3E), despite showing efficient deletion of Dkk1 only in the tumor mass but no changes in the bone and circulation (Fig. 3F-H). Multiplex IHC further confirmed expression of DKK1 or lack thereof in aSMA+ cells (Fig. 3I). To exclude the possibility of tamoxifen directly affecting tumor growth, we performed an MTT assay by culturing the PyMT tumor cells with different concentrations of tamoxifen (0, 1, 2, and 10 µM) for 24 and 48 hours. We did not observe any changes in cell density at all time points and doses tested (Supp Fig. 3B). To exclude the possibility that tamoxifen could affect PyMT tumor growth through suppression of estrogen, we injected the PyMT cells subcutaneously (SQ) into tamoxifen treated male mice and confirmed a significant reduction in tumor growth in aSMA-Dkk1cKO animals compared to controls (Supp Fig. 3C).
To further investigate the role of CAF-derived DKK1 during tumor progression, we co-injected Dkk1-deficient (Dkk1cKO) or sufficient (Dkk1WT) CAFs with PyMT tumor cells (1:1 ratio) into the MFP of naïve WT recipient mice. CAFs were isolated based on tdT expression from primary tumors in aSMA-Dkk1cKO-tdT and aSMA-Dkk1WT-tdT mice. Mice injected with tumor cells alone were used as controls. Highlighting the importance of local production of DKK1, mice co-injected with Dkk1 deficient CAFs showed smaller tumor size compared to mice co-injected with Dkk1 sufficient CAFs (Fig. 3J).
All together these results demonstrate that there are at least two sources of DKK1, one released by bone cells contributing to DKK1 levels in circulation and possibly exerting systemic pro-tumorigenic effects, and one from CAFs at tumor site, possibly affecting the local tumor microenvironment; abrogating DKK1 expression by either of the sources is sufficient to reduce tumor growth.
DKK1 does not directly modulate the tumor cells.
To assess whether DKK1 exerts direct effects on tumor cell proliferation, we cultured the PyMT, 4T1, and EO771 tumor cells in the presence of recombinant DKK1 (rDKK1) at various concentrations (0, 50, 100, and 200 ng/ml) for 24, 48, and 72 hours and performed an MTT assay. For all three cell lines, rDKK1 did not increase cell density compared to unstimulated cells at all time points and doses tested (Supp Fig. 4A-C) nor induced any significant changes in cell cycle and survival (Supp Fig. 4D, E).
To better understand how DKK1 promotes tumor progression in vivo, we performed bulk RNA sequencing of GFP-H2B-mApple-Thy1.1+ PyMT-BO1 cells isolated from orthotopic tumors in WT mice receiving IgG or mDKN01, after exclusion of Ter119+ erythrocytes and CD45+ immune cells (Fig. 4A, Supp Fig. 4F). Out of 16,363 genes sequenced, only 134 genes were differentially expressed (DEGs, p < 0.05 and |fold change|>2) between the two groups (Fig. 4B). KEGG pathway enrichment analysis confirmed no differences in pathways related to cell viability or cell cycle but rather showed changes in pathways related to immune responses (Fig. 4C). Gene set enrichment analysis (GSEA) further showed hallmarks of anti-tumor immune responses being upregulated in the mDKN01-treated tumors compared to IgG, including interferon-gamma response, interferon alpha response, TNFa signaling via NFkB, and IL-2/STAT5 signaling (Fig. 4D). These results suggest that expression of DKK1 at tumor site might contribute to an immune suppressive environment, rather than directly affecting tumor growth.
Local production of DKK1 at tumor site affects tumor immune infiltration.
To determine if DKK1 modulates the immune landscape of the tumor microenvironment, we profiled the tumor-infiltrating immune populations from IgG or mDKN01-treated mice via flow cytometry, 16 days post tumor inoculation. We found a significant increase in the number of CD45+ cells per grams of tumors following mDKN01 administration, with CD4+ and CD8+ T cells, F4/80+ macrophages and NK cells being the most increased subsets (Fig. 5A, Supp Fig. 5A, B).
To determine whether local production of DKK1 at tumor site limits the infiltration of immune populations, we performed IHC to determine the localization of CD45+ cells in PyMT tumors isolated from IgG or mDKN01 treated mice (Fig. 5B), aSMA-Dkk1WT or aSMA-Dkk1cKO animals (Fig. 5C) and from WT mice co-injected with tumor cells together with aSMA-Dkk1WT or aSMA-Dkk1cKO CAFs (Fig. 5D). CD45+ cells primarily resided at the edges of the tumor mass in all control groups. In contrast, the presence of CD45+ immune populations in the central regions of the tumor mass was readily evident in animals following DKK1 neutralization or deletion of DKK1 in the CAFs. These results suggest that CAF-derived DKK1 can limit the infiltration of immune cells at tumor site, regardless of the elevated levels of DKK1 in circulation.
DKK1 targets NK cells to support tumor progression.
To identify the immune populations targeted by DKK1, we injected PyMT cells into the MFP of female NSG mice, which lack T, B, and NK cells, and administered IgG or mDKN01. Strikingly, the anti-tumor effects of mDKN01 were fully abrogated in this mouse model (Fig. 5E). Next, we adopted a selective immune cell depletion approach. First, we depleted T cells by administering anti-CD4 and anti-CD8 antibodies, delivered every 4 days starting 2 days before tumor inoculation, using 500µg for the first dose and 250µg for the subsequent doses, in mice orthotopically injected with PyMT cells and concomitantly receiving IgG or mDKN01. As expected, depletion of T cells only slightly increased tumor burden compared to the IgG control group (Fig. 5F, Supp Fig. 5C), confirming the low involvement of T cells in the PyMT tumor model36. Furthermore, mDKN01 treatment significantly reduced tumor growth in mice depleted of T cells to levels comparable to mDKN01 as single agent, suggesting that T cells are not targeted by DKK1.
Next, we depleted NK cells by administering the anti-NK1.1 antibody (100µg/dose) once a week starting 2 days before tumor inoculation, in mice orthotopically injected with PyMT cells receiving IgG or mDKN01. Unlike the T cells, depletion of NK cells significantly increased tumor burden compared to control mice and completely abrogated the anti-tumor effects of mDKN01 (Fig. 5G, Supp Fig. 5D). These results indicate that DKK1 promotes tumor growth by impacting NK cells, but not T cells, in the PyMT breast cancer model.
DKK1 suppresses perforin-mediated NK cell cytotoxicity.
To determine whether DKK1 directly affects NK cell functionality, we performed ex vivo killing assays to quantify NK cell-mediated killing of PyMT tumor cells in the presence and absence of rDKK1. NK cells were isolated from spleens of poly I:C treated mice and co-cultured with the cell trace violet (CTV)-labeled PyMT tumor cells for 4 hours. Tumor cell death was assessed by the expression of 7-AAD via flow cytometry (Fig. 6A, Supp Fig. 6). rDKK1 significantly suppressed NK cell mediated PyMT killing at all tested effector (NK cells) to target (PyMT) ratios (Fig. 6B). To assess integrated killing over time, we performed a 48-hour IncuCyte Live Cell assay using a 2:1 effector (NK cells) to target (H2B-mApple-Thy1.1+ PyMT-BO1 cells) ratio (Fig. 6A). Addition of rDKK1 resulted in a significantly higher number of tumor cells at the end of assay, indicating impaired NK cell killing of PyMT tumor cells (Fig. 6C).
To further investigate whether CAFs at the tumor site suppress NK cell functions by producing DKK1, we co-cultured tdT+ CAFs isolated from orthotopic tumors in aSMA-Dkk1WT-tdT mice, together with NK cells and CTV-labeled PyMT tumor cells in the presence of IgG or mDKN01 (Fig. 6D). Increasing the number of CAFs reduced the NK-mediated killing of PyMT tumor cells. While DKK1 neutralization had no effects in the absence of CAFs, it restored NK cell functionality against PyMT cells in the presence of CAFs (Fig. 6E). These results demonstrate that CAF-derived DKK1 suppresses NK cell tumoricidal activities in the tumor microenvironment.
To determine how DKK1 impacts NK cells at the transcriptional level, we performed bulk RNA sequencing of NK cells sorted from the spleen of poly I:C treated mice using the CD45+CD3−NK1.1+ markers and stimulated ex vivo with rDKK1 for 4 hours. Out of 14,197 genes detected, 327 genes were differentially expressed in the rDKK1-stimulated versus unstimulated NK cells (Fig. 6F). KEGG pathway analysis showed enrichment in signaling pathways related to NK cell development (Notch signaling pathway) and function (HIF-1, Rap1, mTOR and pathways involved in the regulation of actin cytoskeleton) (Fig. 6G). Furthermore, GSEA analysis showed reductions in genes related to anti-tumor immune responses including the IL-2/STAT5 pathway, the interferon-gamma response and the PI3K/AKT/mTOR signaling (Fig. 6H). Accordingly, the rDKK1-exposed NK cells showed decreased gene expression of cytokine receptors Ifngr1, Ifnar1, Il2r, Il15ra and Il18r1, adhesion molecules involved in the maintenance of the immunological synapse Itgal, Cd244a, and Cd226 and activating receptors Ncr1, Klrk1 and Cd226 (Fig. 6I). We also observed that the expression of Prf1, a key effector molecule used by NK cells to make pores and transfer cytotoxic granules to the target cells to induce their specific killing, was also decreased in the NK cells exposed to rDKK1.
To further investigate whether DKK1 impairs NK cell-mediated cytotoxicity via perforin downregulation, we injected the PyMT cells into the MFP of mice lacking perforin (Prf1−/−) treated with IgG or mDKN01. Strikingly, the anti-tumor effects of mDKN01 were fully abrogated (Fig. 6J), similarly to what observed in the NSG model (Fig. 5E). Since T cell depletion did not alter the anti-tumor effects of mDKN01 (Fig. 5F), this suggests that loss of perforin primarily impacts NK cell driven responses in this context. Thus, these data demonstrate that local production of DKK1 at tumor site promotes tumor growth by suppressing perforin-mediated NK cell cytotoxicity.
DKK1 directly suppresses human NK cell functions.
To investigate whether DKK1 exerts similar suppressive effects on human NK cells (hNK cells), we collected peripheral blood from healthy donors and isolated hNK cells to assess their ability to kill the MDA-MB-231 human breast cancer line in the absence or presence of recombinant human DKK1 (rhDKK1). Consistent with the murine NK cell findings (Fig. 6B, C), rhDKK1 significantly decreased hNK cell killing of MDA-MB-231 cells (Fig. 7A) and of the NK-sensitive K562 target cell line (Fig. 7B).
We next hypothesized that one mechanism through which DKK1 suppresses NK cell function is by modulating NK cell activating receptor expression. Indeed, rhDKK1 led to a significant decrease in NK activating receptors such as NKG2D, NKp30, and NKp46 (Fig. 7C, Supp Fig. 7A). Based on these findings, we considered whether DKK1 may be impacting the ability of NK cells to interact with the tumor cells. Murine NK cells, isolated from the spleen of poly I:C treated mice, and mCherry+ PyMT cells were plated at 2:1 ratio in the presence or absence of rDKK1 (200ng/ml) for 3 hours, followed by F-actin staining and analyzed by high magnification confocal microscopy. We only evaluated interactions between tumor and NK cells in proximity to each other and excluded cells that were more than 10µm apart. In control conditions, 70% of NK cells were found in direct association with the tumor cells (8/11) (Fig. 7D top). In the presence of rDKK1, only 33% of NK cells were in close contact with the tumor cells (3/9), while the majority of NK cells were in adjacent regions interacting with the tumor cells through a long protrusion (Fig. 7D bottom).
Finally, to explore the possibility that DKK1 might modulate the expression of NK cell activating and/or inhibitory ligands on the tumor cells, we stimulated the MDA-MD-231 breast cancer line with rhDKK1 but noted no changes in activating and inhibitory NK ligand levels via flow cytometry (Supp Fig. 7B, C) and qRT-PCR (Supp Fig. 7D). These results were also confirmed in a microarray dataset from human triple-negative breast cancer samples (GSE2165337). Although gene expression levels of DKK1 were significantly higher in TNBC samples compared to normal breast tissue, only HLA-E, whose binding to NKG2A has been shown to disrupt actin formation at the immunological synapse and negatively affect NK function38, showed increased expression in TNBC (Fig. 7E). Expression of the NK activating ligands PVR, MICA/B, PVRL2, CD58, and NCR3LG1 were not statistically different. Collectively, these findings suggest that in human breast cancer, DKK1 directly suppresses NK cells, rather than exhibiting direct effects on tumor cells.
DKK1 levels correlate with metastatic progression and reduced cytotoxic NK cells in breast cancer patients.
Finally, to determine whether DKK1 levels correlate with tumor progression and immune suppression in breast cancer patients, we analyzed DKK1 plasma levels and the activation status of NK cells in the blood of 15 patients with stage IV, HER2−, ER+ breast cancer and skeletal disease, at time of diagnosis and after 15 to 18 months of standard-of-care endocrine therapy-based regimens with denosumab as antiresorptive therapy. Skeletal metastases were monitored by routine surveillance imaging with CT and bone scintigraphy scans. 7 patients were classified as stable as they had no radiographic evidence of skeletal metastatic progression during the study period. 8 patients were classified as progressive as they demonstrated radiographic evidence of new or progressive skeletal lesions during the study period (Fig. 7F, Table. 1). Although DKK1 levels in circulation were not significantly different in the stable and progressive patients at baseline, patients with progressive skeletal metastases demonstrated a significant increase in DKK1 compared to time of diagnosis (Fig. 7G).
To assess whether patients with increased DKK1 had reduced and/or dysfunctional NK cells, we measured the number of CD3−CD56+ NK cells in circulation and found no changes between the stable and progressive patients (Supp Fig. 7E, F). However, the percentage of circulating CD16+CD56dim NK cells, which represent the more cytotoxic subset39, were reduced in patients with progressive disease (Fig. 7H) as the disease progressed. These cells also showed lower expression of perforin and granzyme B as the disease progressed. These results suggest that patients with progressive bone metastatic disease experience systemic immune suppression accompanied by increased DKK1 in circulation and decreased cytotoxic NK cells.