Chimeric antigen receptor macrophage therapy for breast tumours mediated by targeting the tumour extracellular matrix

Wenlong Zhang, Ling Liu, HuiFang Su, Qin Liu, Jie Shen, Hanren Dai, Wei Zheng, Yan Lu, Weijie Zhang, Yuncheng Bei, Pingping Shen, Wenlong Zhang, Ling Liu, HuiFang Su, Qin Liu, Jie Shen, Hanren Dai, Wei Zheng, Yan Lu, Weijie Zhang, Yuncheng Bei, Pingping Shen

Abstract

Background: The extracellular matrix (ECM) is essential for malignant tumour progression, as it is a physical barrier to various kinds of anticancer therapies. Matrix metalloproteinase (MMPs) can degrade almost all ECM components, and macrophages are an important source of MMPs. Studies using macrophages to treat tumours have shown that macrophages can enter tumour tissue to play a regulatory role.

Methods: We modified macrophages with a designed chimeric antigen receptor (CAR), which could be activated after recognition of the tumour antigen HER2 to trigger the internal signalling of CD147 and increase the expression of MMPs.

Results: Although CAR-147 macrophage treatment did not affect tumour cell growth in vitro compared with control treatment. However, we found that the infusion of CAR-147 macrophages significantly inhibited HER2-4T1 tumour growth in BALB/c mice. Further investigation showed that CAR-147 macrophages could reduce tumour collagen deposition and promote T-cell infiltration into tumours, which were consistent with expectations. Interestingly, the levels of the inflammatory cytokines TNF-α and IL-6, which are key factors in cytokine release syndrome, were significantly decreased in the peripheral blood in CAR-147 macrophage-transfused mice.

Conclusion: Our data suggest that targeting the ECM by engineered macrophages would be an effective treatment strategy for solid tumours.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Construction of the chimeric antigen receptor CAR-147 targeting HER2. a The design of a chimeric antigen receptor (CAR-147) for macrophages is shown. b Human HER2 surface expression on 4T1 cells was measured. c CAR-147 surface expression on Raw264.7 cells was measured. d Control Raw264.7 (CON) cells and CAR-147 Raw264.7 (147) cells were cocultured with HER2-4T1 (HER2) cells at a ratio of 1:1 for 24 h, and the expression of MMP genes was assessed by RT-qPCR (n = 6). e Control Raw264.7 (CON) cells and CAR-147 Raw264.7 (147) cells were cocultured with HER2-4T1 (HER2) cells at a ratio of 1:2 for 24 h, and the expression of MMP genes was assessed by RT-qPCR (n = 6). f Control Raw264.7 (CON) cells and CAR-147 Raw264.7 (147) cells were cocultured with HER2-4T1 (HER2) cells at a ratio of 1:2 for 24 or 48 h, and the expression of MMP genes was assessed by RT-qPCR (n = 6). g Control Raw264.7 (CON) cells or CAR-147 Raw264.7 (147) cells were cocultured with wild-type 4T1 (CON) cells or HER2-4T1 (HER2) cells at a ratio of 1:1 for 24 h, and the expression of MMP genes was assessed by RT-qPCR (n = 6). All values are expressed as the mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001
Fig. 2
Fig. 2
The effect of CAR-147 macrophages on tumour growth in vivo. a In vivo imaging of luciferase-expressing HER2-4T1 tumour-bearing mice after intravenous injection of DiR-labelled Raw264.7 cells. b Quantitative analysis of the DiR fluorescence signal in the liver (n = 5). c Quantitative analysis of the DiR fluorescence signal at the tumour site (n = 5). d The experimental schedule for tumour implantation, macrophage infusion and bioluminescence imaging (BLI) monitoring. e BLI was performed on day 7 to assess tumour burden. Control Raw264.7 (CON) cells (1 × 106), CAR-147 Raw264.7 (147) cells (1 × 106), or PBS were injected i.v. on day 8 and day 15, and mice were followed with serial BLI. f The weight of tumours from control Raw264.7 cell- or CAR-147 Raw264.7 cell-treated mice was analysed. The data presented are pooled from two independent experiments. Each symbol indicates one mouse (n = 10). g The spleen weight of tumour-bearing mice was analysed. h The body weight of tumour-bearing mice was measured every 3 days. i Blood serum samples and tumour homogenates were collected from control Raw264.7 cell- or CAR-147 Raw264.7 cell-treated mice and analysed for proinflammatory cytokine release (IL-6, IFNγ, TNFα, IL-1β, IL-12) by ELISA (n = 5). All values are expressed as the mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001
Fig. 3
Fig. 3
CAR-147 macrophages can promote T-cell infiltration into tumours. a Flow cytometric analysis and quantification of CD45+ tumour-infiltrating leucocytes (TILs) (n = 10). b Flow cytometric analysis and quantification of CD3+ T cells in the TIL population (n = 10). c Immunofluorescence analysis of CD3+ T-cell infiltration in tumour tissues from control Raw264.7 cell- and CAR-147 Raw264.7 cell-treated animals. d Flow cytometric analysis and quantification of MDSCs (Gr-1+CD11b+) in the TIL population (n = 10). e In total, 2 × 105 HER2-4T1 cells were transplanted into BALB/c nude mice. BLI was performed to assess tumour burden. f Tumour growth from control Raw264.7 cell- or CAR-147 Raw264.7 cell-treated BALB/c nude mice was analysed by measuring volumes every 2 days. g The weight of tumours from control Raw264.7 cell- or CAR-147 Raw264.7 cell-treated BALB/c nude mice was analysed. Each symbol indicates one mouse (n = 8). All values are expressed as the mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001
Fig. 4
Fig. 4
CAR-147 macrophages reduce extracellular matrix deposition. a Representative images of tumour sections stained with Masson’s trichrome to detect extracellular matrix deposition (blue). Scale bar, 100 μm. b Quantification of the extracellular matrix (n = 15). c Analysis of MMP gene expression in tumour tissues from control Raw264.7 cell- and CAR-147 Raw264.7 cell-treated animals by RT-qPCR (n = 5). All values are expressed as the mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001
Fig. 5
Fig. 5
CAR-147 macrophages facilitate T-cell infiltration into 3D human tumour spheroids. a CAR-h147 surface expression on THP-1 cells. b Detection of HER2 expression on MDA-MB-453 cells by flow cytometry. c PMA-treated CAR-h147 THP-1 cells were cocultured with MDA-MB-453 tumour cells at a ratio of 1:1 for 48 h, and the expression of MMP genes was assessed by RT-qPCR (n = 6). d Confocal microscopy images of MDA-MB-453/THP-1 spheroids treated with CFSE-labelled Jurkat T cells for 20 h. Scale bars, 100 μm. All values are expressed as the mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001

References

    1. Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ. Natural innate and adaptive immunity to cancer. Annu. Rev. Immunol. 2011;29:235–271. doi: 10.1146/annurev-immunol-031210-101324.
    1. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–1570. doi: 10.1126/science.1203486.
    1. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl Acad. Sci. USA. 1989;86:10024–10028. doi: 10.1073/pnas.86.24.10024.
    1. D’Aloia MM, Zizzari IG, Sacchetti B, Pierelli L, Alimandi M. CAR-T cells: the long and winding road to solid tumors. Cell Death Dis. 2018;9:282. doi: 10.1038/s41419-018-0278-6.
    1. Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013;14:1014–1022. doi: 10.1038/ni.2703.
    1. Silzle T, Randolph GJ, Kreutz M, Kunz-Schughart LA. The fibroblast: sentinel cell and local immune modulator in tumor tissue. Int. J. Cancer. 2004;108:173–180. doi: 10.1002/ijc.11542.
    1. Talts JF, Wirl G, Dictor M, Muller WJ, Fassler R. Tenascin-C modulates tumor stroma and monocyte/macrophage recruitment but not tumor growth or metastasis in a mouse strain with spontaneous mammary cancer. J. Cell Sci. 1999;112(Pt 12):1855–1864.
    1. Salmon H, Franciszkiewicz K, Damotte D, Dieu-Nosjean MC, Validire P, Trautmann A, et al. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J. Clin. Invest. 2012;122:899–910. doi: 10.1172/JCI45817.
    1. Martinet L, Garrido I, Filleron T, Le Guellec S, Bellard E, Fournie JJ, et al. Human solid tumors contain high endothelial venules: association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res. 2011;71:5678–5687. doi: 10.1158/0008-5472.CAN-11-0431.
    1. Wang LC, Lo A, Scholler J, Sun J, Majumdar RS, Kapoor V, et al. Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol. Res. 2014;2:154–166. doi: 10.1158/2326-6066.CIR-13-0027.
    1. Caruana I, Savoldo B, Hoyos V, Weber G, Liu H, Kim ES, et al. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat. Med. 2015;21:524–529. doi: 10.1038/nm.3833.
    1. Hynes RO, Naba A. Overview of the matrisome—an inventory of extracellular matrix constituents and functions. Cold Spring Harb. Perspect. Biol. 2012;4:a004903. doi: 10.1101/cshperspect.a004903.
    1. Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu. Rev. Cell Dev. Biol. 2001;17:463–516. doi: 10.1146/annurev.cellbio.17.1.463.
    1. Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc. Res. 2006;69:562–573. doi: 10.1016/j.cardiores.2005.12.002.
    1. Khokha R, Murthy A, Weiss A. Metalloproteinases and their natural inhibitors in inflammation and immunity. Nat. Rev. Immunol. 2013;13:649–665. doi: 10.1038/nri3499.
    1. Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity. 2016;44:450–462. doi: 10.1016/j.immuni.2016.02.015.
    1. Fallowfield JA, Mizuno M, Kendall TJ, Constandinou CM, Benyon RC, Duffield JS, et al. Scar-associated macrophages are a major source of hepatic matrix metalloproteinase-13 and facilitate the resolution of murine hepatic fibrosis. J. Immunol. 2007;178:5288–5295. doi: 10.4049/jimmunol.178.8.5288.
    1. Pellicoro A, Aucott RL, Ramachandran P, Robson AJ, Fallowfield JA, Snowdon VK, et al. Elastin accumulation is regulated at the level of degradation by macrophage metalloelastase (MMP-12) during experimental liver fibrosis. Hepatology. 2012;55:1965–1975. doi: 10.1002/hep.25567.
    1. Thomas JA, Pope C, Wojtacha D, Robson AJ, Gordon-Walker TT, Hartland S, et al. Macrophage therapy for murine liver fibrosis recruits host effector cells improving fibrosis, regeneration, and function. Hepatology. 2011;53:2003–2015. doi: 10.1002/hep.24315.
    1. Merlin S, Bhargava KK, Ranaldo G, Zanolini D, Palestro CJ, Santambrogio L, et al. Kupffer cell transplantation in mice for elucidating monocyte/macrophage biology and for potential in cell or gene therapy. Am. J. Pathol. 2016;186:539–551. doi: 10.1016/j.ajpath.2015.11.002.
    1. Suzuki T, Arumugam P, Sakagami T, Lachmann N, Chalk C, Sallese A, et al. Pulmonary macrophage transplantation therapy. Nature. 2014;514:450–454. doi: 10.1038/nature13807.
    1. Doerschuk CM. Pulmonary alveolar proteinosis and macrophage transplantation. N. Engl. J. Med. 2015;372:1762–1764. doi: 10.1056/NEJMcibr1413035.
    1. Happle C, Lachmann N, Skuljec J, Wetzke M, Ackermann M, Brennig S, et al. Pulmonary transplantation of macrophage progenitors as effective and long-lasting therapy for hereditary pulmonary alveolar proteinosis. Sci. Transl. Med. 2014;6:250ra113. doi: 10.1126/scitranslmed.3009750.
    1. Yang L, Zhang Y. Tumor-associated macrophages: from basic research to clinical application. J. Hematol. Oncol. 2017;10:58. doi: 10.1186/s13045-017-0430-2.
    1. Zhou H, Zha Z, Liu Y, Zhang H, Zhu J, Hu S, et al. Structural insights into the down-regulation of overexpressedp185(her2/neu) protein of transformed cells by the antibody chA21. J. Biol. Chem. 2011;286:31676–31683. doi: 10.1074/jbc.M111.235184.
    1. Cheng LS, Liu AP, Yang JH, Dong YQ, Li LW, Wang J, et al. Construction, expression and characterization of the engineered antibody against tumor surface antigen, p185(c-erbB-2) Cell Res. 2003;13:35–48. doi: 10.1038/sj.cr.7290149.
    1. Toole BP. Emmprin (CD147), a cell surface regulator of matrix metalloproteinase production and function. Curr. Top. Dev. Biol. 2003;54:371–389. doi: 10.1016/S0070-2153(03)54015-7.
    1. Graham A, Wood JL, O’Leary VJ, Stone D. Human (THP-1) macrophages oxidize LDL by a thiol-dependent mechanism. Free Radic. Res. 1996;25:181–192. doi: 10.3109/10715769609149923.
    1. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457–1461. doi: 10.1126/science.1171362.
    1. Miller BW, Morton JP, Pinese M, Saturno G, Jamieson NB, McGhee E, et al. Targeting the LOX/hypoxia axis reverses many of the features that make pancreatic cancer deadly: inhibition of LOX abrogates metastasis and enhances drug efficacy. EMBO Mol. Med. 2015;7:1063–1076. doi: 10.15252/emmm.201404827.
    1. Guerriero JL. Macrophages: the road less traveled, changing anticancer therapy. Trends Mol. Med. 2018;24:472–489. doi: 10.1016/j.molmed.2018.03.006.
    1. Andreesen R, Hennemann B, Krause SW. Adoptive immunotherapy of cancer using monocyte-derived macrophages: rationale, current status, and perspectives. J. Leukoc. Biol. 1998;64:419–426. doi: 10.1002/jlb.64.4.419.
    1. Tang H, Wang Y, Chlewicki LK, Zhang Y, Guo J, Liang W, et al. Facilitating T cell infiltration in tumor microenvironment overcomes resistance to PD-L1 blockade. Cancer Cell. 2016;30:500. doi: 10.1016/j.ccell.2016.08.011.
    1. Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain MCAR. T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat. Med. 2018;24:731–738. doi: 10.1038/s41591-018-0041-7.
    1. Norelli M, Camisa B, Barbiera G, Falcone L, Purevdorj A, Genua M, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat. Med. 2018;24:739–748. doi: 10.1038/s41591-018-0036-4.

Source: PubMed

3
Subscribe