logo
    Abstract:
    Multiple myeloma (MM) is incurable once osteocytic lesions have seeded at skeletal sites, but factors mediating this deadly pathogenic advance remain poorly understood. Here we report evidence of a major role for the cell adhesion molecule CD166, which we discovered to be highly expressed in MM cell lines and primary bone marrow (BM) cells from patients. CD166+ MM cells homed more efficiently than CD166- cells to the BM of engrafted immunodeficient NSG mice. CD166 silencing in MM cells enabled longer survival, a smaller tumor burden and less osteolytic lesions, as compared to mice bearing control cells. CD166 deficiency in MM cell lines or CD138+ BM cells from MM patients compromised their ability to induce bone resorption in an ex vivo organ culture system. Further, CD166 deficiency in MM cells also reduced formation of osteolytic disease in vivo after intra-tibial engraftment. Mechanistic investigation revealed that CD166 expression in MM cells inhibited osteoblastogenesis of BM-derived osteoblast progenitors by suppressing RUNX2 gene expression. Conversely, CD166 expression in MM cells promoted osteoclastogenesis by activating TRAF6-dependent signaling pathways in osteoclast progenitors. Overall, our results define CD166 as a pivotal director in MM cell homing to the BM and MM progression, rationalizing its further study as a candidate therapeutic target for MM treatment.
    Keywords:
    Homing (biology)
    ALCAM
    Osteolysis
    The interaction of recruited immune effector cells and cancer cells within tumor microenvironment (TME) shapes the fate of cancer progression and metastasis. Many cancers including breast cancer, express a specific vacuolar ATPase (a2V) on their cell surface which acidifies the extracellular milieu helping cancer cell proliferation and metastasis. To understand the role of immune cell-associated-a2V during breast tumor pathogenesis, we knocked-out a2V (KO) from the hematopoietic stem cells (HSC) and generated breast tumors in mice. The a2V-KO mice developed faster growing, larger, and metastatic breast tumors compared to control mice. Further investigation of the TME revealed a significant reduction in the presence of CD4+ and CD8+ T cells in the a2V-KO tumors. Targeted RNA-Seq of the cells of the TME demonstrated that pro-inflammatory cytokines, death receptors, death receptor ligands, and cytotoxic effectors were significantly down-regulated within the a2V-KO TME. Interestingly, analysis of immune cells in the blood, spleen, and thymus of the non-tumor bearing a2V-KO mice revealed a significant decrease in CD4+ and CD8+ T cell populations. For the first time, this study demonstrates that inhibition of V-ATPase expression in HSC leads to a decrease in CD4+ and CD8+ T cell populations and thus promotes breast tumor growth and metastasis.
    Citations (11)
    Abstract Purposes: We have been investigating the interplay between cancer cells and host immunity, and previously demonstrated that Snail expression in tumor cells accelerates metastasis not only by enhancing tumor motility but also by inducing immune dysfunction through increase of a variety of immunoregulatory cells using murine tumor models implanted with snail-transduced B16-F10 melanoma (F10-snail+). We additionally found that Snail+ tumor cells frequently metastasize into bone marrow (BM) of the mice, and pluripotent CD45-ALCAM+ mesenchymal stem cells (MSCs) consequently increased in the mice. In this study, we attempted to characterize the Snail+ tumor-induced CD45-ALCAM+ MSCs (sMSCs). Results: The sMSCs were prepared by stimulating BMCs with F10-snail+ tumor supernatant for 5-7 days, or by sorting CD45-ALCAM+ cells from BM or spleen of the F10-snail+ tumor cells 2-3 weeks after implantation. When the mixture of the sMSCs and F10-mock tumor cells was subcutaneously injected in C57BL/6 mice, tumor growth was aggressively promoted, and spontaneous tumor dissemination in BM indicating de novo bone metastasis was observed, accompanied by increase of sMSCs in BM. In the tumor milieu and spleen of these mice, functionally impaired CD8(low) T cells and immunosuppressive Treg-inducible CD11b+Gr1+ MDSCs predominantly increased. Indeed, CD8(low) T cells were generated in the culture with splenic T cells and sMSCs, and MDSCs were expanded in the culture with BM cells and sMSCs. We identified key molecules responsible for the sMSC-induced events: ALCAM, ANGPT2 and FSTL1, which significantly increased only in the sMSC but not F10-mock-derived MSCs and normal MSCs. FSTL1 inhibited bone metastasis and consequent increase of sMSCs, CD8(low) T cells and MDSCs. Blocking ALCAM with mAb inhibited both sMSC expansion and CD8(low) T-cell induction. Blocking ANGPT2 inhibited expansion of MDSCs and Tregs rather than angiogenesis. Blocking FSTL1, which is a key trigger to initiate these cascades, in combination with elimination of other sMSC-associated factors significantly induced anti-tumor immune responses, resulting in complete inhibition of tumor growth and metastasis. Conclusions: These results suggest that sMSCs play a central role in the cancer EMT-induced immune dysfunction leading to tumor progression. Snail is reported to regulate acquisition of stem-like properties in tumor cells. However, how cancer stem cells would modulate the BM microenvironment and alter anti-tumor immune responses also remains unclear. Targeting sMSCs and its associated molecules may be a promising strategy for abrogating even cancer stem cells through reprogramming and activating anti-tumor immune responses appropriately. Citation Format: Chie Kudo-Saito, Takafumi Fuwa, Kouichi Murakami. Immune dysfunction cascades caused by ALCAM+ mesenchymal stem cells toward tumor progression. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 183. doi:10.1158/1538-7445.AM2014-183
    ALCAM
    Abstract Malignant mesothelioma (MM) is a debilitating, frequently incurable cancer that exhibits a high degree of resistance to standard cytotoxic chemotherapy (CTX). Novel therapeutic approaches to treat this disease are desperately needed. In the vast majority of cases, MM is associated with prior exposure to asbestos fibers, resulting in a chronic pro-inflammatory state in pleura. As such, we hypothesized that infiltration of MM by leukocytes fosters tumorigenesis. To investigate this, we evaluated MMs resected from patients (n=16) and compared the complexity of immune cells infiltrating MM to those found in normal pleura (n=4). Using polychromatic fluorescent-activated cell sorting (FACS) on freshly resected whole tissues, we found a significant increased presence of CD11b+CD14+HLA-DR+ monocytes/macrophages in MM (37.3 ± 4.4% of total CD45+ cells) as compared to normal pleural tissue (13.8 ± 6.5% of total CD45+ cells), and a further increase in MM resected from patients treated with neoadjuvant CTX (48.8 ± 5.5% of CD45+ cells). To determine if increased presence of CD11b+CD14+HLA-DR+ leukocytes was associated with varied expression of cytokines regulating leukocyte maturation/recruitment, we examined mRNA expression of tissues/tumors. We found increased expression of Colony Stimulating Factor 1 (CSF1), and Colony-Stimulating Factor-1 Receptor (CSF-1R) mRNA, a critical cytokine-signaling axis regulating monocyte/macrophage differentiation and recruitment into tumors, in MM tumors compared to normal pleura, and even higher levels in tumors resected from patients treated with CTX. Since recent experimental data has revealed that tumor-associated macrophages (TAMs) secrete proangiogenic, prosurvival, and pro-invasive factors that foster tumor progression, we evaluated macrophage depletion in MM as a novel therapeutic strategy. We conducted studies evaluating PLX3397 (Plexxikon Inc., Berkeley, CA), a novel, orally bioavailable, small-molecule tyrosine kinase inhibitor of CSF-1R. Using a syngeneic orthotopic murine model of MM, we found that treatment of mice with PLX3397 alters the tumor immune microenvironment by decreasing TAM infiltration and increasing the proportion of CD8+ cytotoxic T lymphocytes within tumors. This reprogramming of the tumor immune microenvironment was associated with alterations in the tumor microvasculature as evidenced by a decrease in CD31+ structures, as well as a decrease in VEGFA mRNA expression. Ultimately, these changes resulted in an increase in tumor cell apoptosis (p=0.04) and a decrease in tumor burden (p=0.0008). These studies indicate that: 1) macrophages potentiate mesothelioma development, and 2) depletion of mesothelioma-associated macrophages may improve the efficacy of cytotoxic chemotherapy. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr C231.
    Suppression of bone metastasis can improve patient quality of life. Current drugs for bone metastasis have been shown to prolong progression‐free survival but not overall survival; therefore, other potential therapeutic targets for bone metastasis should be investigated. Cell‐surface antigens, such as CD 24, have been recently shown to be involved in the metastasis of various cancers. However, whether CD 24 plays a role in bone metastasis of lung cancer remains unknown. To observe metastasis of lung cancer cells by imaging technology, we introduced a near‐infrared fluorescent protein, iRFP 720, into a bone‐seeking subclone established from lung cancer cells, HARA ‐B4 cells. The anchorage‐independent growth of these cells was then evaluated by colony formation assays. We also compared cancer cell tropism to bone tissue with HARA ‐B4 cells in the presence or absence of CD 24 by cell adhesion assays. To clarify the role of CD 24 in bone metastasis, we intracardially injected CD 24‐knockdown HARA ‐B4 cells into mice and monitored metastasis through detection of iRFP 720 using an in vivo imaging system. CD 24‐knockdown HARA ‐B4 cells in vitro showed reduced anchorage‐independent growth and cancer cell tropism to bone. Bone metastasis was diminished in mice inoculated with CD 24‐knockdown HARA ‐B4 cells, which was rescued by add‐back of CD 24 in cells. Our findings indicate that iRFP 720 is effective for in vivo imaging analysis of bone metastasis and that downregulation of CD 24 suppresses bone metastasis of lung cancer cells. These findings collectively indicate that CD 24 may be considered a promising new therapeutic candidate for the prevention of bone metastasis of lung cancer.
    Citations (25)
    Cancer cells facilitate growth and metastasis by using multiple signals from the cancer-associated microenvironment. However, it remains poorly understood whether prostate cancer (PCa) cells may recruit and utilize bone marrow cells for their growth and survival. Furthermore, the regulatory mechanisms underlying interactions between PCa cells and bone marrow cells are obscure. In this study, we isolated bone marrow cells that mainly constituted populations that were positive for CD11b and Gr1 antigens from xenograft PC-3 tumor tissues from athymic nu/nu mice. We found that the tumor-infiltrated cells alone were unable to form tumor spheroids, even with increased amounts and time. By contrast, the tumor-infiltrated cells together with PCa cells formed large numbers of tumor spheroids compared with PCa cells alone. We further utilized xenograft athymic nu/nu mice bearing bone metastatic lesions. We demonstrated that PCa cells were unable to survive and give rise to colony-forming units (CFUs) in media that were used for hematopoietic cell colony-formation unit (CFU) assays. By contrast, PC-3M cells survived when bone marrow cells were present and gave rise to CFUs. Our results showed that PCa cells required bone marrow cells to support their growth and survival and establish bone metastasis in the host environment. We showed that PCa cells that were treated with either siRNA for PIP5K1α or its specific inhibitor, ISA-2011B, were unable to survive and produce tumor spheroids, together with bone marrow cells. Given that the elevated expression of PIP5K1α was specific for PCa cells and was associated with the induced expression of VEGF receptor 2 in PCa cells, our findings suggest that cancer cells may utilize PIP5K1α-mediated receptor signaling to recruit growth factors and ligands from the bone marrow-derived cells. Taken together, our study suggests a new mechanism that enables PCa cells to gain proliferative and invasive advantages within their associated host microenvironment. Therapeutic interventions using PIP5K1α inhibitors may not only inhibit tumor invasion and metastasis but also enhance the host immune system.
    Citations (5)
    Abstract Using the 4T1 model of experimental breast cancer we had recently shown that cancer induced bone disease starts before metastatic colonization and is mediated by RANKL expressed by tumor specific T cells. The role of anti-tumor B cell immune response in the context of cancer induced bone disease has never been investigated. There is evidence in the literature that B cells are good prognostic markers for metastatic breast cancer. B cells have an intimate relationship with bone cells as they differentiate from HSC present on endosteal surfaces; cross-talk with skeletal system through the RANK-RANKL-OPG signaling axis; and produce OPG, a decoy receptor of RANKL. Here we used the BALB/c derived 4T1 (metastatic) and 67NR (non-metastatic) sibling cell lines of mammary mouse carcinomas. By day 7, 14 and 21 after tumor injection, B220+ BM B cells from 67NR+ animals produce high amounts of OPG in vitro in contrast to B220+ BM B cells of 4T1+ mice. In vitro, BM B cells from 67NR+ mice, but not from 4T1+, could inhibit the RANKL dependent, anti-4T1 T cell mediated-OC differentiation ascertained by TRAP enzymatic activity, morphology and osteolytic disk assay. Transference of BM B cells from 67NR+ mice together with 4T1 tumor cells to BALB/c mice led to inhibition of osteoclastogenesis, increased numbers of bone lining cells and mesenchyme stem cell. Besides acting directly on the bone remodeling system, these B cells also modulated T cell activity evidenced by diminished RANKL and IL-17F production. All the anti-osteolytic and pro-osteogenic activity of B cells modulate and inhibits the pre-metastatic niche formation. Indeed transference of such cells to 4T1 animals inhibited LN and BM metastatic colonization. We conclude that 67NR induced OPG+ B cells can inhibit pro-osteoclastic / pre-metastatic activity of tumor induced T cells, favoring a bone metastasis free-phenotype. These findings have implications, not only for the understanding of the direct contribution of B cells in the control of bone metastasis but also it might be a promising prognostic tool for predicting cancer-induced bone metastasis. Citation Format: Bonomo A, Monteiro AC, Leal AC, Fontão AP, Spinetti E, Balduino A. OPG+ bone marrow B cells induced by non-metastatic tumors inhibit the pre-metastatic bone niche induced by T cells. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P2-05-24.