Macrophages and cathepsin proteases blunt chemotherapeutic response in breast cancer

Tanaya Shree, Oakley C Olson, Benelita T Elie, Jemila C Kester, Alfred L Garfall, Kenishana Simpson, Katherine M Bell-McGuinn, Emily C Zabor, Edi Brogi, Johanna A Joyce, Tanaya Shree, Oakley C Olson, Benelita T Elie, Jemila C Kester, Alfred L Garfall, Kenishana Simpson, Katherine M Bell-McGuinn, Emily C Zabor, Edi Brogi, Johanna A Joyce

Abstract

The microenvironment is known to critically modulate tumor progression, yet its role in regulating treatment response is poorly understood. Here we found increased macrophage infiltration and cathepsin protease levels in mammary tumors following paclitaxel (Taxol) chemotherapy. Cathepsin-expressing macrophages protected against Taxol-induced tumor cell death in coculture, an effect fully reversed by cathepsin inhibition and mediated partially by cathepsins B and S. Macrophages were also found to protect against tumor cell death induced by additional chemotherapeutics, specifically etoposide and doxorubicin. Combining Taxol with cathepsin inhibition in vivo significantly enhanced efficacy against primary and metastatic tumors, supporting the therapeutic relevance of this effect. Additionally incorporating continuous low-dose cyclophosphamide dramatically impaired tumor growth and metastasis and improved survival. This study highlights the importance of integrated targeting of the tumor and its microenvironment and implicates macrophages and cathepsins in blunting chemotherapeutic response.

Figures

Figure 1.
Figure 1.
Cathepsin levels in mammary tumors increase following Taxol treatment. (A) Tumor volume curves for PyMT transgenic mice (left) and orthotopically implanted mice (right) treated with one dose of either vehicle or Taxol (50 mg/kg) on day 0, as described in the Materials and Methods. n = 16–17 for PyMT mice; n = 5–8 for orthotopic model of TS1 PyMT cell line; (*) P < 0.05; (ns) not significant. (B) Taxol increased cathepsin levels at the tumor site. Implanted tumors were harvested from mice 7 d after vehicle or Taxol treatment, and whole-tumor lysates were assayed for cathepsin proteins. Quantitation of high-molecular-weight (HMW) and lower-molecular-weight (LMW) active cathepsin B (left) and cathepsin C and cathepsin L (right) are shown above the corresponding Western blots. Quantitation of the loading control actin is in Supplemental Figure S2. n = 6–7 mice per group; (*) P < 0.05; (**) P < 0.01; (***) P < 0.0001. (C) Cathepsin transcripts also increased after Taxol treatment. qRT–PCR analysis for cathepsin genes in lysates from implanted tumors, relative to an endogenous control gene, Ubiquitin C, and normalized to vehicle. n = 7–8 mice per group; (*) P < 0.05, (**) P < 0.01 by the Mann-Whitney test. (D,E) qRT–PCR analysis of cathepsin transcripts in tumor cells (D) or macrophages (E) in culture following treatment with DMSO control or Taxol showed that expression does not change significantly. n = 3 independent experiments in each case.
Figure 2.
Figure 2.
Chemotherapy induces a specific increase in macrophage infiltration in breast tumors. (A) Macrophages accumulate in tumors after Taxol treatment. Representative images of orthotopic tumors stained for the macrophage marker Iba1 7 d after vehicle or Taxol treatment. Bars, 100 μm. (B) Quantitation of intratumoral Iba1+ cells in tumors of vehicle- and Taxol-treated mice via image analysis. n = 12 vehicle, 9 Taxol; (***) P < 0.0001. All data in B–F are from whole tumors isolated 7 d after vehicle or Taxol treatment. (C,D) qRT–PCR from whole-tumor lysates for transcripts of CD68 (macrophages) and CD45 (all leukocytes) 7 d after vehicle or Taxol treatment (n = 7–8 tumors); (*) P < 0.05. (E) Quantitation by image analysis of the percentage of CD45+Iba1− cells of total DAPI+ cells in tumors after treatment with vehicle or Taxol (n = 6 mice per group). (F) Representative images of tumor sections from mice injected with the cathepsin ABP after vehicle or Taxol treatment and costained with the macrophage marker Iba1. (G) Macrophage numbers increase in breast cancer patients following neoadjuvant chemotherapy. Representative images of matched samples pre- and post-treatment from patients 2, 3, 5, and 6 stained with CD68 or a pan-cytokeratin (CK) antibody to visualize tumor cells. Positively stained cells are labeled in brown. Rare CD68+ cells in the pretreatment biopsies are indicated by white arrowheads. Patient information can be found in Supplemental Table S1. Bars: F,G, 50 μm.
Figure 3.
Figure 3.
Macrophage-supplied cathepsins protect tumor cells from cell death. (A) Schematic of coculture assay. (B) Percentage of DAPI+ (dead) tumor cells in mono- or coculture of the TS1 cell line with wild-type (WT) BMDMs 48 h after Taxol (50 nM) or DMSO treatment. Addition of the cathepsin inhibitor JPM (10 μM) abrogated the protective effect of BMDMs following Taxol treatment. (C) BMDMs also protected from Taxol-induced cell death in additional PyMT-derived tumor cell lines, TS2, Met-1, and AT-3, with experimental conditions as in B. (D) Percentage of TS1 tumor cell death in monoculture and in cocultures with wild-type or cathepsin B-, cathepsin S-, cathepsin C-, or cathepsin L-null BMDMs 48 h after treatment. Deletion of cathepsin B or cathepsin S significantly reduced macrophage-conferred protection. (E) Macrophage-conferred protection is largely contact-independent. Cell death in TS1 tumor cells cultured alone or in the presence of wild-type BMDM-CM or CAF-CM 48 h after treatment with Taxol or DMSO control. Addition of JPM (10 μM) to the CM abrogated the BMDM-conferred protection. For all graphs, data are from three independent experiments, each done in triplicate. (*) P < 0.05; (**) P < 0.01; (***) P < 0.001; (ns) not significant.
Figure 4.
Figure 4.
Cathepsin inhibition sensitizes tumors to Taxol treatment. (A) Trial schematic showing dosing of MTD Taxol, JPM, and CLD Cyclo in the 5-wk regression trial (from 9–14 wk of age). (B) Tumor volume curves for PyMT transgenic mice treated with Taxol, JPM, and Taxol+JPM, compared with vehicle, based on twice-weekly external palpation caliper measurements. n = 17 vehicle, 16 JPM, 16 Taxol, 16 Taxol+JPM. P-values in B–D derive from area-under-the-curve (AUC) analyses, with pairwise comparisons between indicated groups, although some of these effects did not reach statistical significance when adjusted for multiple comparisons. (C) Tumor volume curves for Cyclo- and Cyclo+JPM-treated mice compared with vehicle-treated mice. n = 14 Cyclo, 14 Cyclo+JPM. (D) Tumor volume curves for Taxol+Cyclo- and Triple-treated mice compared with vehicle-treated mice. n = 16 Taxol+Cyclo, 16 Triple. (E) End-stage tumor volumes based on measurements of excised tumors. (*) P < 0.05, (**) P < 0.01, (***) P < 0.0001, unpaired t-tests. Comparisons are with the vehicle group unless otherwise indicated. n = 14–15 per treatment group.
Figure 5.
Figure 5.
Combination treatments reduce lung metastasis and increase survival. (A) Total lung metastasis burden for indicated treatment groups, as determined by the percentage of total lung area covered by metastases for each mouse. Each data point in the scatter plots represents the mean value for an individual animal; horizontal lines represent the median with the interquartile range. (*) P < 0.05, (**) P < 0.01, Mann-Whitney test; n = 13 vehicle, 13 JPM, 13 Taxol, 10 Taxol+JPM, 12 Taxol+Cyclo, 13 Triple. (B) Graph showing the number of metastases per mouse per unit area. (C) Graph showing the average size (area in square microns) of metastatic lesions. (D) Survival trial schematic showing dosing of MTD Taxol, JPM, and CLD Cyclo. (E, left) Kaplan-Meier long-term survival curves of mice in the treatment groups indicated. Late-stage survival data beginning at the 70th day of treatment with only mice alive on that day are shown at the right. n = 15–21 per treatment group. P-values from log-rank test comparisons of survival data can be found in Supplemental Table S4.
Figure 6.
Figure 6.
Cathepsin-mediated chemoprotection is relevant to other chemotherapies and additional in vivo models. (A) Percentage of DAPI+ (dead) tumor cells in mono- or coculture of the TS1 cell line with BMDMs 48 h after etoposide (20 μM), doxorubicin (300 nM), gemcitabine (400 nM), carboplatin (50 μM), or DMSO treatment. Coculture with BMDMs was protective against treatment with etoposide and doxorubicin, but not gemcitabine or carboplatin. As with Taxol, the protective effect observed in BMDM coculture was significantly abrogated by the cathepsin inhibitor JPM (10 μM). (B) Cell death in tumor cells cultured alone or in the presence of wild-type (WT) BMDM-CM or CAF-CM 48 h after treatment with etoposide, doxorubicin, gemcitabine, carboplatin, or DMSO control. For all graphs in A and B, data are from three independent experiments, each performed in triplicate. (*) P < 0.05; (**) P < 0.01; (***) P < 0.001; (ns) not significant. (C) FVB/n mice orthotopically implanted with TS1 cells were treated with etoposide (10 mg/kg per week). Addition of JPM (100 mg/kg per day) significantly improved response to etoposide. (*) P < 0.05. The etoposide/JPM combination treatment also significantly reduced tumor growth compared with the vehicle-treated group on day 14 (endpoint for the vehicle- and JPM-treated mice due to tumor burden limits). n = 8–10 mice per group. (D) Athy/Nu mice orthotopically implanted with MDA-231 cells were treated with MTD Taxol (25mg/kg per week) or vehicle control. Addition of JPM (100 mg/kg per day) significantly improved response to Taxol in this preclinical xenograft model. (*) P < 0.05 compared with Taxol alone; (***) P < 0.001 compared with vehicle; n = 9–12 mice per group. Triangles below the graphs in C and D indicate the time points at which etoposide and Taxol, respectively, were administered.

References

    1. Abboud-Jarrous G, Atzmon R, Peretz T, Palermo C, Gadea BB, Joyce JA, Vlodavsky I 2008. Cathepsin L is responsible for processing and activation of proheparanase through multiple cleavages of a linker segment. J Biol Chem 283: 18167–18176
    1. Ahn GO, Tseng D, Liao CH, Dorie MJ, Czechowicz A, Brown JM 2010. Inhibition of Mac-1 (CD11b/CD18) enhances tumor response to radiation by reducing myeloid cell recruitment. Proc Natl Acad Sci 107: 8363–8368
    1. Bell-McGuinn KM, Garfall AL, Bogyo M, Hanahan D, Joyce JA 2007. Inhibition of cysteine cathepsin protease activity enhances chemotherapy regimens by decreasing tumor growth and invasiveness in a mouse model of multistage cancer. Cancer Res 67: 7378–7385
    1. Blansfield JA, Caragacianu D, Alexander HR III, Tangrea MA, Morita SY, Lorang D, Schafer P, Muller G, Stirling D, Royal RE, et al. 2008. Combining agents that target the tumor microenvironment improves the efficacy of anticancer therapy. Clin Cancer Res 14: 270–280
    1. Bogyo M, Verhelst S, Bellingard-Dubouchaud V, Toba S, Greenbaum D 2000. Selective targeting of lysosomal cysteine proteases with radiolabeled electrophilic substrate analogs. Chem Biol 7: 27–38
    1. Borowsky AD, Namba R, Young LJ, Hunter KW, Hodgson JG, Tepper CG, McGoldrick ET, Muller WJ, Cardiff RD, Gregg JP 2005. Syngeneic mouse mammary carcinoma cell lines: Two closely related cell lines with divergent metastatic behavior. Clin Exp Metastasis 22: 47–59
    1. Cailleau R, Mackay B, Young RK, Reeves WJ Jr 1974. Tissue culture studies on pleural effusions from breast carcinoma patients. Cancer Res 34: 801–809
    1. DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF, Gallagher WM, Wadhwani N, Keil SD, Junaid SA, et al. 2011. Leukocyte complexity in breast cancer predicts overall survival and functionally regulates response to chemotherapy. Cancer Discov 1: 52–65
    1. Emmenegger U, Man S, Shaked Y, Francia G, Wong JW, Hicklin DJ, Kerbel RS 2004. A comparative analysis of low-dose metronomic cyclophosphamide reveals absent or low-grade toxicity on tissues highly sensitive to the toxic effects of maximum tolerated dose regimens. Cancer Res 64: 3994–4000
    1. Gocheva V, Zeng W, Ke D, Klimstra D, Reinheckel T, Peters C, Hanahan D, Joyce JA 2006. Distinct roles for cysteine cathepsin genes in multistage tumorigenesis. Genes Dev 20: 543–556
    1. Gocheva V, Chen X, Peters C, Reinheckel T, Joyce JA 2010a. Deletion of cathepsin H perturbs angiogenic switching, vascularization and growth of tumors in a mouse model of pancreatic islet cell cancer. Biol Chem 391: 937–945
    1. Gocheva V, Wang HW, Gadea BB, Shree T, Hunter KE, Garfall AL, Berman T, Joyce JA 2010b. IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion. Genes Dev 24: 241–255
    1. Greenbaum D, Medzihradszky KF, Burlingame A, Bogyo M 2000. Epoxide electrophiles as activity-dependent cysteine protease profiling and discovery tools. Chem Biol 7: 569–581
    1. Guy CT, Cardiff RD, Muller WJ 1992. Induction of mammary tumors by expression of polyomavirus middle T oncogene: A transgenic mouse model for metastatic disease. Mol Cell Biol 12: 954–961
    1. Halangk W, Lerch MM, Brandt-Nedelev B, Roth W, Ruthenbuerger M, Reinheckel T, Domschke W, Lippert H, Peters C, Deussing J 2000. Role of cathepsin B in intracellular trypsinogen activation and the onset of acute pancreatitis. J Clin Invest 106: 773–781
    1. Joyce JA, Pollard JW 2009. Microenvironmental regulation of metastasis. Nat Rev Cancer 9: 239–252
    1. Joyce JA, Baruch A, Chehade K, Meyer-Morse N, Giraudo E, Tsai FY, Greenbaum DC, Hager JH, Bogyo M, Hanahan D 2004. Cathepsin cysteine proteases are effectors of invasive growth and angiogenesis during multistage tumorigenesis. Cancer Cell 5: 443–453
    1. Kerbel RS, Kamen BA 2004. The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer 4: 423–436
    1. Kohler C 2007. Allograft inflammatory factor-1/Ionized calcium-binding adapter molecule 1 is specifically expressed by most subpopulations of macrophages and spermatids in testis. Cell Tissue Res 330: 291–302
    1. Kozin SV, Kamoun WS, Huang Y, Dawson MR, Jain RK, Duda DG 2010. Recruitment of myeloid but not endothelial precursor cells facilitates tumor regrowth after local irradiation. Cancer Res 70: 5679–5685
    1. Lin EY, Jones JG, Li P, Zhu L, Whitney KD, Muller WJ, Pollard JW 2003. Progression to malignancy in the polyoma middle T oncoprotein mouse breast cancer model provides a reliable model for human diseases. Am J Pathol 163: 2113–2126
    1. Locher C, Conforti R, Aymeric L, Ma Y, Yamazaki T, Rusakiewicz S, Tesniere A, Ghiringhelli F, Apetoh L, Morel Y, et al. 2010. Desirable cell death during anticancer chemotherapy. Ann N Y Acad Sci 1209: 99–108
    1. Man S, Bocci G, Francia G, Green SK, Jothy S, Hanahan D, Bohlen P, Hicklin DJ, Bergers G, Kerbel RS 2002. Antitumor effects in mice of low-dose (metronomic) cyclophosphamide administered continuously through the drinking water. Cancer Res 62: 2731–2735
    1. Mohamed MM, Sloane BF 2006. Cysteine cathepsins: Multifunctional enzymes in cancer. Nat Rev Cancer 6: 764–775
    1. Nardin A, Lefebvre ML, Labroquere K, Faure O, Abastado JP 2006. Liposomal muramyl tripeptide phosphatidylethanolamine: Targeting and activating macrophages for adjuvant treatment of osteosarcoma. Curr Cancer Drug Targets 6: 123–133
    1. Palermo C, Joyce JA 2008. Cysteine cathepsin proteases as pharmacological targets in cancer. Trends Pharmacol Sci 29: 22–28
    1. Pham CT, Ley TJ 1999. Dipeptidyl peptidase I is required for the processing and activation of granzymes A and B in vivo. Proc Natl Acad Sci 96: 8627–8632
    1. Pietras K, Hanahan D 2005. A multitargeted, metronomic, and maximum-tolerated dose ‘chemo-switch’ regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer. J Clin Oncol 23: 939–952
    1. Ponomarev V, Doubrovin M, Serganova I, Vider J, Shavrin A, Beresten T, Ivanova A, Ageyeva L, Tourkova V, Balatoni J, et al. 2004. A novel triple-modality reporter gene for whole-body fluorescent, bioluminescent, and nuclear noninvasive imaging. Eur J Nucl Med Mol Imaging 31: 740–751
    1. Pyonteck SM, Gadea BB, Wang HW, Gocheva V, Hunter KE, Tang LH, Joyce JA 2011. Deficiency of the macrophage growth factor CSF-1 disrupts pancreatic neuroendocrine tumor development. Oncogene doi: 10.1038/onc.2011.337
    1. Qian BZ, Pollard JW 2010. Macrophage diversity enhances tumor progression and metastasis. Cell 141: 39–51
    1. Reiser J, Adair B, Reinheckel T 2010. Specialized roles for cysteine cathepsins in health and disease. J Clin Invest 120: 3421–3431
    1. Roth W, Deussing J, Botchkarev VA, Pauly-Evers M, Saftig P, Hafner A, Schmidt P, Schmahl W, Scherer J, Anton-Lamprecht I, et al. 2000. Cathepsin L deficiency as molecular defect of furless: Hyperproliferation of keratinocytes and pertubation of hair follicle cycling. FASEB J 14: 2075–2086
    1. Sanchez-Munoz A, Perez-Ruiz E, Ribelles N, Marquez A, Alba E 2008. Maintenance treatment in metastatic breast cancer. Expert Rev Anticancer Ther 8: 1907–1912
    1. Schurigt U, Sevenich L, Vannier C, Gajda M, Schwinde A, Werner F, Stahl A, von Elverfeldt D, Becker AK, Bogyo M, et al. 2008. Trial of the cysteine cathepsin inhibitor JPM-OEt on early and advanced mammary cancer stages in the MMTV-PyMT-transgenic mouse model. Biol Chem 389: 1067–1074
    1. Sevenich L, Schurigt U, Sachse K, Gajda M, Werner F, Muller S, Vasiljeva O, Schwinde A, Klemm N, Deussing J, et al. 2010. Synergistic antitumor effects of combined cathepsin B and cathepsin Z deficiencies on breast cancer progression and metastasis in mice. Proc Natl Acad Sci 107: 2497–2502
    1. Shaked Y, Henke E, Roodhart JM, Mancuso P, Langenberg MH, Colleoni M, Daenen LG, Man S, Xu P, Emmenegger U, et al. 2008. Rapid chemotherapy-induced acute endothelial progenitor cell mobilization: Implications for antiangiogenic drugs as chemosensitizing agents. Cancer Cell 14: 263–273
    1. Sharma A, Straubinger RM 1994. Novel taxol formulations: Preparation and characterization of taxol-containing liposomes. Pharm Res 11: 889–896
    1. Shi GP, Villadangos JA, Dranoff G, Small C, Gu L, Haley KJ, Riese R, Ploegh HL, Chapman HA 1999. Cathepsin S required for normal MHC class II peptide loading and germinal center development. Immunity 10: 197–206
    1. Stewart TJ, Abrams SI 2007. Altered immune function during long-term host–tumor interactions can be modulated to retard autochthonous neoplastic growth. J Immunol 179: 2851–2859
    1. Xian X, Hakansson J, Stahlberg A, Lindblom P, Betsholtz C, Gerhardt H, Semb H 2006. Pericytes limit tumor cell metastasis. J Clin Invest 116: 642–651
    1. Yang R, Niepel M, Mitchison TK, Sorger PK 2010. Dissecting variability in responses to cancer chemotherapy through systems pharmacology. Clin Pharmacol Ther 88: 34–38

Source: PubMed

3
購読する