Vessel co-option mediates resistance to anti-angiogenic therapy in liver metastases

Sophia Frentzas, Eve Simoneau, Victoria L Bridgeman, Peter B Vermeulen, Shane Foo, Eleftherios Kostaras, Mark Nathan, Andrew Wotherspoon, Zu-Hua Gao, Yu Shi, Gert Van den Eynden, Frances Daley, Clare Peckitt, Xianming Tan, Ayat Salman, Anthoula Lazaris, Patrycja Gazinska, Tracy J Berg, Zak Eltahir, Laila Ritsma, Jacco Van Rheenen, Alla Khashper, Gina Brown, Hanna Nystrom, Malin Sund, Steven Van Laere, Evelyne Loyer, Luc Dirix, David Cunningham, Peter Metrakos, Andrew R Reynolds, Sophia Frentzas, Eve Simoneau, Victoria L Bridgeman, Peter B Vermeulen, Shane Foo, Eleftherios Kostaras, Mark Nathan, Andrew Wotherspoon, Zu-Hua Gao, Yu Shi, Gert Van den Eynden, Frances Daley, Clare Peckitt, Xianming Tan, Ayat Salman, Anthoula Lazaris, Patrycja Gazinska, Tracy J Berg, Zak Eltahir, Laila Ritsma, Jacco Van Rheenen, Alla Khashper, Gina Brown, Hanna Nystrom, Malin Sund, Steven Van Laere, Evelyne Loyer, Luc Dirix, David Cunningham, Peter Metrakos, Andrew R Reynolds

Abstract

The efficacy of angiogenesis inhibitors in cancer is limited by resistance mechanisms that are poorly understood. Notably, instead of through the induction of angiogenesis, tumor vascularization can occur through the nonangiogenic mechanism of vessel co-option. Here we show that vessel co-option is associated with a poor response to the anti-angiogenic agent bevacizumab in patients with colorectal cancer liver metastases. Moreover, we find that vessel co-option is also prevalent in human breast cancer liver metastases, a setting in which results with anti-angiogenic therapy have been disappointing. In preclinical mechanistic studies, we found that cancer cell motility mediated by the actin-related protein 2/3 complex (Arp2/3) is required for vessel co-option in liver metastases in vivo and that, in this setting, combined inhibition of angiogenesis and vessel co-option is more effective than the inhibition of angiogenesis alone. Vessel co-option is therefore a clinically relevant mechanism of resistance to anti-angiogenic therapy and combined inhibition of angiogenesis and vessel co-option might be a warranted therapeutic strategy.

Conflict of interest statement

Competing finanical interests None of the authors declared any competing financial interests.

Figures

Figure 1. Correlation between HGP and pathological…
Figure 1. Correlation between HGP and pathological response in patients treated preoperatively with bevacizumab
a. Diagrams illustrating the morphology of the normal liver or the morphology of the tumor-liver interface in liver metastases with a desmoplastic, pushing or replacement HGP. b. The HGPs and the pathological response to bev-chemo were scored in 59 CRCLMs from 33 patients treated preoperatively with bev-chemo at RM. Graph shows % HGP (replacement, desmoplastic, pushing) scored in each individual lesion and the data are grouped by pathological response score: >75%, 50–75%, 25–49% or <25% viable tumor. Median number of lesions examined per patient was 1 (range = 1 to 4 lesions per patient). c–e. Examples of H&E-stained lesions from the RM cohort are shown. Arrows point to examples of replacement HGP areas. Arrowheads point to examples of desmoplastic HGP areas. Asterisks indicate areas of infarct-like necrosis. f. The HGPs and the pathological response to bev-chemo were scored in 128 CRCLMs from 59 patients treated with bev-chemo at MUHC. Graph shows % HGP (replacement, desmoplastic, pushing) scored in each individual lesion and the data are grouped by pathological response score: >75%, 50–75%, 25–49% or <25% viable tumor. Median number of lesions examined per patient was 2 (range = 1 to 12 lesions per patient). The χ2-test was used to determine statistical significance (see 2 x 2 contingency tables in panels b and f). Scale bars, 1 mm.
Figure 2. Correlation between HGP and morphological…
Figure 2. Correlation between HGP and morphological response on CT in patients treated preoperatively with bevacizumab
a–f. CT scans of patients treated preoperatively with bev-chemo. Examples of optimal (OR), partial (PR) or absent (AR) morphological response are shown. a,b. OR; in the pre-treatment image a lesion in liver segment VII (arrowhead) is scored as group-3 (a); the same lesion imaged after 4 cycles of bevacizumab in combination with CAPOX is now scored as group-1 (b). c,d. PR; in the pre-treatment image a lesion in liver segment II (arrowhead) is scored as group-3 (c); the same lesion imaged after 4 cycles of bevacizumab in combination with CAPOX is now scored as group-2 (d). e,f. AR; in the pre-treatment image a lesion in liver segment VI (arrowhead) is scored as group-3 (e); the same lesion imaged after 6 cycles of bevacizumab in combination with FOLFIRI is still scored as group-3 (f). g. Morphological response and HGP were scored in 52 liver metastases from 31 patients treated preoperatively with bev-chemo at RM. Graph shows the % HGP scored in each individual lesion (replacement, desmoplastic, pushing). Lesions are grouped according to response: AR, PR or OR. Lesions scored as AR were classed as poor responders, whilst those scored as PR or OR were classed as good responders. Median number of lesions examined per patient was 1 (range = 1 to 4 lesions per patient). The χ2 test was used to determine statistical significance (see 2 x 2 contingency table in panel g).
Figure 3. Cancer cells infiltrate the hepatic…
Figure 3. Cancer cells infiltrate the hepatic plates and co-opt sinusoidal blood vessels in the replacement HGP
a. An area of normal liver is shown. Staining is for hepatocyte specific antigen (HSA, green) to detect hepatocytes and collagen-3 (col-3, red) to detect liver sinusoidal blood vessels (SV). b–d. Staining for cancer cells (CK, red) and hepatocytes (HSA, green) at the tumor-liver interface (b,c) and within the tumor mass (d) in a replacement HGP liver metastasis of colorectal cancer. Examples of displaced hepatocytes are marked (arrowheads). e–g. Staining for cytokeratin 20 (CK20, brown) to identify cancer cells and CD31 to identify blood vessels (blue) in replacement HGP liver metastases of colorectal cancer. Arrows and arrowheads indicate examples of liver sinusoidal blood vessels where one end of the vessel is physically located in the liver parenchyma (arrows), whilst the other end is surrounded by cancer cells (arrowheads). Asterisk, tumor. Lv, normal liver. SV, sinusoidal blood vessel. Scale bars, 25 μM.
Figure 4. The replacement HGP occurs in…
Figure 4. The replacement HGP occurs in progressive disease and is associated with a poor outcome in patients treated with bevacizumab
a. Left: HGPs in untreated CRCLMs (n = 32 lesions from 19 MUHC patients). Middle: HGPs in pre-existing CRCLMs (n = 128 lesions from 59 MUHC patients). Right: HGPs in new CRCLMs (n = 35 lesions from 13 MUHC patients). Graphs show % replacement (R), % desmoplastic (D) and % pushing (P) HGP per lesion ± SEM. b. Kaplan-Meier estimates of OS for 62 MUHC patients treated preoperatively with bev-chemo stratified into two groups: predominant replacement HGP (26 patients) or predominant desmoplastic HGP (35 patients). c. Kaplan-Meier estimates of OS for 29 MUHC patients treated preoperatively with chemotherapy alone stratified into two groups: predominant replacement HGP (12 patients) or predominant desmoplastic HGP (16 patients). d. Kaplan-Meier estimates of OS for 51 MUHC patients with a predominant desmoplastic HGP stratified into two groups: desmoplastic HGP treated with bev-chemo (35 patients) or desmoplastic HGP treated with chemotherapy alone (16 patients). e. Kaplan-Meier estimates of OS for 38 MUHC patients with a predominant replacement HGP stratified into two groups: replacement HGP treated with bev-chemo (26 patients) or replacement HGP treated with chemotherapy alone (12 patients). Kruskall-Walls test (a) or the Log-Rank test (b–e) were used to determine statistical significance. Hazard ratios were calculated using Cox-regression. * P<0.001.
Figure 5. The replacement HGP predominates in…
Figure 5. The replacement HGP predominates in breast cancer liver metastases
a. The HGPs were examined in breast cancer liver metastases (BCLMs) from 17 patients. Graph shows the % HGP (replacement, desmoplastic, pushing) scored in each case. The cases are grouped by intrinsic subtype of breast cancer. Lum A, luminal A. Lum B (HER2-), luminal B HER2 negative. Lum B (HER2+), luminal B HER2 positive. TN, triple negative. b–g. Morphology of the replacement growth pattern of BCLMs.Diagram of the tumor-liver interface in the replacement HGP (b). H&E-stained human BCLM sample illustrating the tumor-liver interface (c). Co-staining for hepatocyte specific antigen (HSA) to label hepatocytes and pan-cytokeratin (CK) to label cancer cells confirms that breast cancer cells infiltrate the liver parenchyma and replace hepatocytes in BCLM (d). Co-staining for alpha smooth muscle actin (αSMA) to label fibroblasts and CK to label cancer cells confirms the absence of a desmoplastic stroma at the tumor-liver interface in BCLM (e). Co-staining for collagen-3 (col-3) to label sinusoidal vessels and CK to label cancer cells shows that the vascular architecture of the adjacent liver is preserved at the tumor-liver interface in BCLM (f). Co-staining for CD31 to label blood vessels and cytokeratin 19 (CK19) to label cancer cells confirms the infiltrative pattern of tumor growth that facilitates vessel co-option in BCLM (g). Asterisk, cancer cells; Lv, normal liver. Scale bars, 50 μM.
Figure 6. Inhibition of vessel co-option and…
Figure 6. Inhibition of vessel co-option and angiogenesis is more effective than targeting angiogenesis alone
a,b. Areas of replacement (a) and desmoplastic (b) HGP are shown in a preclinical (HT29 cell line) orthotopic model of advanced liver metastasis. Staining shown is for: H&E, CK and HSA, CK and col-3, CK and αSMA or cytokeratin 20 (CK20) and CD31, as indicated. c,d. Characterization of parental HT29 cells (parent) and HT29 cells transduced with control non-targeting shRNA (control shRNA) or ARPC3-targeting shRNAs (shARPC3-1, shARPC3-2 or shARPC3-3). In c, ARPC3 expression was determined by western blotting (see also Supplementary Data Set 1). Graph shows ARPC3 expression relative to parental HT29 cells ± SEM (n = 3 independent western blots). In d, cell motility was measured by time-lapse microscopy. Graph shows cell velocity (μm per minute) relative to parental HT29 cells ± SEM (n = 30 tracked cells per group pooled from 2 independent experiments). e. Quantification of the HGPs in control- and ARPC3-knockdown tumors. Graph shows the % replacement (R), % desmoplastic (D) and % pushing (P) HGP per group ± SEM (n = 6 mice per group). f–h. Tumors with normal ARPC3 levels (control shRNA) or ARPC3 knockdown (shARPC3-3) were established in the livers of mice, followed by treatment with B20-4.1.1 plus capecitabine (BC) or vehicle alone (Vh) for two weeks followed by histopathological analysis. Graph in f shows the % HGP per group ± SEM (n = 8 mice per group). Graph in g shows liver tumor burden expressed in terms of lesion area ± SEM (n = 8 mice per group). Graph in h shows tumor vessel density in terms of vessels per mm2 ± SEM (n = 8 mice per group). For statistical analysis, Student’s t-test (panels c,g,h) or Mann Whitney U-test (panels d,e,f) were used. *P<0.05, **P<0.01 ***P<0.001, ****P<0.0001. n.s., no significant difference. Asterisk, cancer cells; DS, desmoplastic stroma; Lv, normal liver. Scale bars, 50 μM.

References

    1. Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov. 2004;3:391–400.
    1. Kerbel RS. Tumor angiogenesis. N Engl J Med. 2008;358:2039–2049.
    1. Hurwitz H, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. The New England journal of medicine. 2004;350:2335–2342.
    1. Cunningham D, et al. Bevacizumab plus capecitabine versus capecitabine alone in elderly patients with previously untreated metastatic colorectal cancer (AVEX): an open-label, randomised phase 3 trial. Lancet Oncol. 2013;14:1077–1085.
    1. Ebos JM, Kerbel RS. Antiangiogenic therapy: impact on invasion, disease progression, and metastasis. Nat Rev Clin Oncol. 2011;8:210–221.
    1. Vasudev NS, Reynolds AR. Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis. 2014;17:471–494.
    1. Pezzella F, et al. Angiogenesis in primary lung cancer and lung secondaries. Eur J Cancer. 1996;32A:2494–2500.
    1. Vermeulen PB, et al. Liver metastases from colorectal adenocarcinomas grow in three patterns with different angiogenesis and desmoplasia. The Journal of pathology. 2001;195:336–342.
    1. Dome B, Hendrix MJ, Paku S, Tovari J, Timar J. Alternative vascularization mechanisms in cancer: Pathology and therapeutic implications. Am J Pathol. 2007;170:1–15.
    1. Donnem T, et al. Vessel co-option in primary human tumors and metastases: an obstacle to effective anti-angiogenic treatment? Cancer Med. 2013;2:427–436.
    1. Jayson GC, Kerbel R, Ellis LM, Harris AL. Antiangiogenic therapy in oncology: current status and future directions. Lancet. 2016
    1. Adams RB, et al. Selection for hepatic resection of colorectal liver metastases: expert consensus statement. HPB (Oxford) 2013;15:91–103.
    1. Van den Eynden GG, et al. The multifaceted role of the microenvironment in liver metastasis: biology and clinical implications. Cancer research. 2013;73:2031–2043.
    1. Stessels F, et al. Breast adenocarcinoma liver metastases, in contrast to colorectal cancer liver metastases, display a non-angiogenic growth pattern that preserves the stroma and lacks hypoxia. Br J Cancer. 2004;90:1429–1436.
    1. Gruenberger B, et al. Bevacizumab, capecitabine, and oxaliplatin as neoadjuvant therapy for patients with potentially curable metastatic colorectal cancer. J Clin Oncol. 2008;26:1830–1835.
    1. Chaudhury P, et al. Perioperative chemotherapy with bevacizumab and liver resection for colorectal cancer liver metastasis. HPB. 2010;12:37–42.
    1. Wong R, et al. A multicentre study of capecitabine, oxaliplatin plus bevacizumab as perioperative treatment of patients with poor-risk colorectal liver-only metastases not selected for upfront resection. Ann Oncol. 2011;22:2042–2048.
    1. Chun YS, et al. Association of computed tomography morphologic criteria with pathologic response and survival in patients treated with bevacizumab for colorectal liver metastases. JAMA. 2009;302:2338–2344.
    1. Shindoh J, et al. Optimal morphologic response to preoperative chemotherapy: an alternate outcome end point before resection of hepatic colorectal metastases. J Clin Oncol. 2012;30:4566–4572.
    1. Boonsirikamchai P, et al. CT findings of response and recurrence, independent of change in tumor size, in colorectal liver metastasis treated with bevacizumab. AJR American journal of roentgenology. 2011;197:W1060–1066.
    1. Van den Eynden GG, et al. The histological growth pattern of colorectal cancer liver metastases has prognostic value. Clin Exp Metastasis. 2012;29:541–549.
    1. Nurnberg A, Kitzing T, Grosse R. Nucleating actin for invasion. Nature reviews. 2011;11:177–187.
    1. Otsubo T, et al. Involvement of Arp2/3 complex in the process of colorectal carcinogenesis. Modern pathology : an official journal of the United States and Canadian Academy of Pathology Inc. 2004;17:461–467.
    1. Iwaya K, et al. Correlation between liver metastasis of the colocalization of actin-related protein 2 and 3 complex and WAVE2 in colorectal carcinoma. Cancer science. 2007;98:992–999.
    1. Kopetz S, et al. Synergistic activity of the SRC family kinase inhibitor dasatinib and oxaliplatin in colon carcinoma cells is mediated by oxidative stress. Cancer research. 2009;69:3842–3849.
    1. Nyati MK, et al. The potential of 5-fluorocytosine/cytosine deaminase enzyme prodrug gene therapy in an intrahepatic colon cancer model. Gene therapy. 2002;9:844–849.
    1. Gray MJ, et al. Therapeutic targeting of Id2 reduces growth of human colorectal carcinoma in the murine liver. Oncogene. 2008;27:7192–7200.
    1. Liang WC, et al. Cross-species vascular endothelial growth factor (VEGF)-blocking antibodies completely inhibit the growth of human tumor xenografts and measure the contribution of stromal VEGF. J Biol Chem. 2006;281:951–961.
    1. Miller KD, et al. Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol. 2005;23:792–799.
    1. Miller K, et al. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. The New England journal of medicine. 2007;357:2666–2676.
    1. Miles DW, et al. Phase III study of bevacizumab plus docetaxel compared with placebo plus docetaxel for the first-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol. 2010;28:3239–3247.
    1. Robert NJ, et al. RIBBON-1: randomized, double-blind, placebo-controlled, phase III trial of chemotherapy with or without bevacizumab for first-line treatment of human epidermal growth factor receptor 2-negative, locally recurrent or metastatic breast cancer. J Clin Oncol. 2011;29:1252–1260.
    1. Brufsky AM, et al. RIBBON-2: a randomized, double-blind, placebo-controlled, phase III trial evaluating the efficacy and safety of bevacizumab in combination with chemotherapy for second-line treatment of human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol. 2011;29:4286–4293.
    1. Naresh KN, Nerurkar AY, Borges AM. Angiogenesis is redundant for tumour growth in lymph node metastases. Histopathology. 2001;38:466–470.
    1. Jeong HS, et al. Investigation of the Lack of Angiogenesis in the Formation of Lymph Node Metastases. J Natl Cancer Inst. 2015;107
    1. Colpaert CG, et al. Cutaneous breast cancer deposits show distinct growth patterns with different degrees of angiogenesis, hypoxia and fibrin deposition. Histopathology. 2003;42:530–540.
    1. Breast-Cancer-Progression-Working-Party. Evidence for novel non-angiogenic pathway in breast-cancer metastasis. Lancet. 2000;355:1787–1788.
    1. Szabo V, et al. Mechanism of tumour vascularization in experimental lung metastases. The Journal of pathology. 2015;235:384–396.
    1. Carbonell WS, Ansorge O, Sibson N, Muschel R. The vascular basement membrane as “soil” in brain metastasis. PLoS One. 2009;4:e5857.
    1. Bugyik E, et al. Lack of angiogenesis in experimental brain metastases. J Neuropathol Exp Neurol. 2011;70:979–991.
    1. Valiente M, et al. Serpins promote cancer cell survival and vascular co-option in brain metastasis. Cell. 2014;156:1002–1016.
    1. Jain RK, et al. Biomarkers of response and resistance to antiangiogenic therapy. Nat Rev Clin Oncol. 2009;6:327–338.
    1. Kuczynski EA, et al. Co-option of Liver Vessels and Not Sprouting Angiogenesis Drives Acquired Sorafenib Resistance in Hepatocellular Carcinoma. J Natl Cancer Inst. 2016;108
    1. Rubenstein JL, et al. Anti-VEGF antibody treatment of glioblastoma prolongs survival but results in increased vascular cooption. Neoplasia. 2000;2:306–314.
    1. Kusters B, et al. Vascular endothelial growth factor-A(165) induces progression of melanoma brain metastases without induction of sprouting angiogenesis. Cancer research. 2002;62:341–345.
    1. Leenders WP, et al. Antiangiogenic therapy of cerebral melanoma metastases results in sustained tumor progression via vessel co-option. Clin Cancer Res. 2004;10:6222–6230.
    1. Paez-Ribes M, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009;15:220–231.
    1. de Groot JF, et al. Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro Oncol. 2010;12:233–242.
    1. Lu KV, et al. VEGF Inhibits Tumor Cell Invasion and Mesenchymal Transition through a MET/VEGFR2 Complex. Cancer Cell. 2012;22:21–35.
    1. Sennino B, et al. Suppression of Tumor Invasion and Metastasis by Concurrent Inhibition of c-Met and VEGF Signaling in Pancreatic Neuroendocrine Tumors. Cancer Discov. 2012;2:270–287.
    1. Depner C, et al. EphrinB2 repression through ZEB2 mediates tumour invasion and anti-angiogenic resistance. Nature communications. 2016;7:12329.
    1. Bland JM, Altman DG. Statistical methods for assessing agreement between two methods of clinical measurement. Lancet. 1986;1:307–310.
    1. Ribero D, et al. Bevacizumab improves pathologic response and protects against hepatic injury in patients treated with oxaliplatin-based chemotherapy for colorectal liver metastases. Cancer. 2007;110:2761–2767.
    1. Chang HH, Leeper WR, Chan G, Quan D, Driman DK. Infarct-like necrosis: a distinct form of necrosis seen in colorectal carcinoma liver metastases treated with perioperative chemotherapy. The American journal of surgical pathology. 2012;36:570–576.
    1. Goldhirsch A, et al. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol. 2013;24:2206–2223.
    1. Hammond ME, et al. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer (unabridged version) Archives of pathology & laboratory medicine. 2010;134:e48–72.
    1. Wolff AC, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol. 2013;31:3997–4013.
    1. Gourlaouen M, Welti JC, Vasudev NS, Reynolds AR. Essential role for endocytosis in the growth factor-stimulated activation of ERK1/2 in endothelial cells. J Biol Chem. 2013;288:7467–7480.
    1. Grambsch PM, Therneau TM. Proportional Hazards Tests and Diagnostics Based on Weighted Residuals. Biometrika. 1994;81:515–526.

Source: PubMed

3
订阅