Skip to main content
Top
Published in: Breast Cancer Research 1/2020

01-12-2020 | Breast Cancer | Research article

High hepatocyte growth factor expression in primary tumor predicts better overall survival in male breast cancer

Authors: Si-Qi Qiu, Johan van Rooijen, Hilde H. Nienhuis, Bert van der Vegt, Hetty Timmer-Bosscha, Elise van Leeuwen-Stok, Annemiek M. E. Walenkamp, Carolien H. M. van Deurzen, Geertruida H. de Bock, Elisabeth G. E. de Vries, Carolien P. Schröder

Published in: Breast Cancer Research | Issue 1/2020

Login to get access

Abstract

Background

Breast cancer is rare in men, but management is focused on tumor characteristics commonly found in female breast cancer. The tumor microenvironment of male breast cancer is less well understood, and insight may improve male breast cancer management. The hepatocyte growth factor (HGF)/c-MET axis and the stromal cell-derived factor-1 (CXCL12)/C-X-C chemokine receptor type 4 (CXCR4) axis are prognostic in women with breast cancer. We aimed to investigate these factors in male breast cancer and correlate them with patient survival.

Methods

From 841 Dutch males with breast cancer who were enrolled in the EORTC 10085/TBCRC/BIG/NABCG International Male Breast Cancer Program (NCT01101425) and diagnosed between 1990 and 2010, archival primary tumor samples were collected. Tissue microarrays were constructed with 3 cores per sample and used for immunohistochemical analysis of HGF, c-MET, CXCL12, and CXCR4. Overall survival (OS) of the patients without metastases (M0) was analyzed using the Kaplan-Meier method. The value of the markers regarding OS was determined using univariable and multivariable Cox regression analyses, providing hazard ratios (HRs) and 95% confidence intervals (95% CIs).

Results

Of 720 out of 841 patients, sufficient tissue was available for analysis; 487 out of 720 patients had M0 disease. Patients with high HGF expression and high CXCL12 expression had a superior OS (low vs high expression of both markers, 7.5 vs 13.0 years, hazard ratio [HR] 0.64, 95% CI 0.49–0.84, P = 0.001 [HGF]; 9.1 vs 15.3 years, HR 0.63, 95% CI 0.45–0.87, P = 0.005 [CXCL12]). Multivariate analysis identified HGF as an independent predictor for OS (HR 0.64, 95% CI 0.47–0.88, P = 0.001).

Conclusions

HGF and CXCL12 tumor expression appear to identify male breast cancer patients with a relatively good prognosis. Possibly, this could support male breast cancer-specific management strategies in the future.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Ruddy KJ, Winer EP. Male breast cancer: risk factors, biology, diagnosis, treatment, and survivorship. Ann Oncol. 2013;24:1434–43.CrossRef Ruddy KJ, Winer EP. Male breast cancer: risk factors, biology, diagnosis, treatment, and survivorship. Ann Oncol. 2013;24:1434–43.CrossRef
3.
go back to reference Giordano SH, Cohen DS, Buzdar AU, Perkins G, Hortobagyi GN. Breast carcinoma in men: a population-based study. Cancer. 2004;101:51–7.CrossRef Giordano SH, Cohen DS, Buzdar AU, Perkins G, Hortobagyi GN. Breast carcinoma in men: a population-based study. Cancer. 2004;101:51–7.CrossRef
4.
go back to reference Sirieix J, Fraisse J, Mathoulin-Pelissier S, Leheurteur M, Vanlemmens L, Jouannaud C, et al. Management and outcome of metastatic breast cancer in men in the national multicenter observational ESME program. Ann Oncol. 2018;29 Abstract 294PD_PR.CrossRef Sirieix J, Fraisse J, Mathoulin-Pelissier S, Leheurteur M, Vanlemmens L, Jouannaud C, et al. Management and outcome of metastatic breast cancer in men in the national multicenter observational ESME program. Ann Oncol. 2018;29 Abstract 294PD_PR.CrossRef
5.
go back to reference Anderson WF, Jatoi I, Tse J, Rosenberg PS. Male breast cancer: a population-based comparison with female breast cancer. J Clin Oncol. 2010;28:232–9.CrossRef Anderson WF, Jatoi I, Tse J, Rosenberg PS. Male breast cancer: a population-based comparison with female breast cancer. J Clin Oncol. 2010;28:232–9.CrossRef
6.
go back to reference Liu N, Johnson KJ, Ma CX. Male breast cancer: an updated surveillance, epidemiology, and end results data analysis. Clin Breast Cancer. 2018;18:e997–1002.CrossRef Liu N, Johnson KJ, Ma CX. Male breast cancer: an updated surveillance, epidemiology, and end results data analysis. Clin Breast Cancer. 2018;18:e997–1002.CrossRef
7.
go back to reference Miao H, Verkooijen HM, Chia KS, Bouchardy C, Pukkala E, Larønningen S, et al. Incidence and outcome of male breast cancer: an international population-based study. J Clin Oncol. 2011;29:4381–6.CrossRef Miao H, Verkooijen HM, Chia KS, Bouchardy C, Pukkala E, Larønningen S, et al. Incidence and outcome of male breast cancer: an international population-based study. J Clin Oncol. 2011;29:4381–6.CrossRef
8.
go back to reference Kornegoor R, van Diest PJ, Buerger H, Korsching E. Tracing differences between male and female breast cancer: both diseases own a different biology. Histopathology. 2015;67:888–97.CrossRef Kornegoor R, van Diest PJ, Buerger H, Korsching E. Tracing differences between male and female breast cancer: both diseases own a different biology. Histopathology. 2015;67:888–97.CrossRef
9.
go back to reference Manson QF, Hoeve ND, Buerger H, Moelans CB, van Diest PJ. PD-1 and PD-L1 expression in male breast cancer in comparison with female breast cancer. Target Oncol. 2018;13:769–77.CrossRef Manson QF, Hoeve ND, Buerger H, Moelans CB, van Diest PJ. PD-1 and PD-L1 expression in male breast cancer in comparison with female breast cancer. Target Oncol. 2018;13:769–77.CrossRef
10.
go back to reference Massarweh SA, Sledge GW, Miller DP, Mccullough D, Petkov VI. Molecular characterization and mortality from breast cancer in men. J Clin Oncol. 2018;36:1396–404.CrossRef Massarweh SA, Sledge GW, Miller DP, Mccullough D, Petkov VI. Molecular characterization and mortality from breast cancer in men. J Clin Oncol. 2018;36:1396–404.CrossRef
11.
go back to reference Ottini L. Male breast cancer: a rare disease that might uncover underlying pathways of breast cancer. Nat Rev Cancer. 2014;14:643–4.CrossRef Ottini L. Male breast cancer: a rare disease that might uncover underlying pathways of breast cancer. Nat Rev Cancer. 2014;14:643–4.CrossRef
12.
go back to reference Bendinelli P, Maroni P, Matteucci E, Desiderio MA. Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast carcinoma. Cell Death Dis. 2017;8:e2578.CrossRef Bendinelli P, Maroni P, Matteucci E, Desiderio MA. Epigenetic regulation of HGF/Met receptor axis is critical for the outgrowth of bone metastasis from breast carcinoma. Cell Death Dis. 2017;8:e2578.CrossRef
13.
go back to reference Domanska UM, Kruizinga RC, Nagengast WB, Timmer-Bosscha H, Huls G, de Vries EGE, et al. A review on CXCR4/CXCL12 axis in oncology: no place to hide. Eur J Cancer. 2013;49:219–30.CrossRef Domanska UM, Kruizinga RC, Nagengast WB, Timmer-Bosscha H, Huls G, de Vries EGE, et al. A review on CXCR4/CXCL12 axis in oncology: no place to hide. Eur J Cancer. 2013;49:219–30.CrossRef
14.
go back to reference Ho-Yen CM, Jones JL, Kermorgant S. The clinical and functional significance of c-Met in breast cancer: a review. Breast Cancer Res. 2015;17:52.CrossRef Ho-Yen CM, Jones JL, Kermorgant S. The clinical and functional significance of c-Met in breast cancer: a review. Breast Cancer Res. 2015;17:52.CrossRef
15.
go back to reference Parr C, Ali AY. Boswellia frereana suppresses HGF-mediated breast cancer cell invasion and migration through inhibition of c-Met signalling. J Transl Med. 2018;12:281.CrossRef Parr C, Ali AY. Boswellia frereana suppresses HGF-mediated breast cancer cell invasion and migration through inhibition of c-Met signalling. J Transl Med. 2018;12:281.CrossRef
16.
go back to reference Zhao X, Qu J, Hui Y, Zhang H, Sun Y, Liu X, et al. Clinicopathological and prognostic significance of c-Met overexpression in breast cancer. Oncotarget. 2017;8:56758–67.PubMedPubMedCentral Zhao X, Qu J, Hui Y, Zhang H, Sun Y, Liu X, et al. Clinicopathological and prognostic significance of c-Met overexpression in breast cancer. Oncotarget. 2017;8:56758–67.PubMedPubMedCentral
17.
go back to reference Garcia S, Dalès JP, Charafe-Jauffret E, Carpentier-Meunier S, Andrac-Meyer L, Jacquemier J, et al. Poor prognosis in breast carcinomas correlates with increased expression of targetable CD146 and c-Met and with proteomic basal-like phenotype. Hum Pathol. 2007;38:830–41.CrossRef Garcia S, Dalès JP, Charafe-Jauffret E, Carpentier-Meunier S, Andrac-Meyer L, Jacquemier J, et al. Poor prognosis in breast carcinomas correlates with increased expression of targetable CD146 and c-Met and with proteomic basal-like phenotype. Hum Pathol. 2007;38:830–41.CrossRef
18.
go back to reference Edakuni G, Sasatomi E, Satoh T, Tokunaga O, Miyazaki K. Expression of the hepatocyte growth factor/c-Met pathway is increased at the cancer front in breast carcinoma. Pathol Int. 2001;51:172–8.CrossRef Edakuni G, Sasatomi E, Satoh T, Tokunaga O, Miyazaki K. Expression of the hepatocyte growth factor/c-Met pathway is increased at the cancer front in breast carcinoma. Pathol Int. 2001;51:172–8.CrossRef
19.
go back to reference Yang H, Zhang C, Cui S. Expression of hepatocyte growth factor in breast cancer and its effect on prognosis and sensitivity to chemotherapy. Mol Med Rep. 2015;11:1037–42.CrossRef Yang H, Zhang C, Cui S. Expression of hepatocyte growth factor in breast cancer and its effect on prognosis and sensitivity to chemotherapy. Mol Med Rep. 2015;11:1037–42.CrossRef
20.
go back to reference Okuyama Kishima M, De Oliveira CEC, Banin-Hirata BK, Losi-Guembarovski R, Brajão De Oliveira K, Amarante MK, et al. Immunohistochemical expression of CXCR4 on breast cancer and its clinical significance. Anal Cell Pathol. 2015;2015:891020. Okuyama Kishima M, De Oliveira CEC, Banin-Hirata BK, Losi-Guembarovski R, Brajão De Oliveira K, Amarante MK, et al. Immunohistochemical expression of CXCR4 on breast cancer and its clinical significance. Anal Cell Pathol. 2015;2015:891020.
21.
go back to reference Pernas S, Martin M, Kaufman PA, Gil-Martin M, Gomez Pardo P, Lopez-Tarruella S, et al. Balixafortide plus eribulin in HER2-negative metastatic breast cancer: a phase 1, single-arm, dose-escalation trial. Lancet Oncol. 2018;19:812–24.CrossRef Pernas S, Martin M, Kaufman PA, Gil-Martin M, Gomez Pardo P, Lopez-Tarruella S, et al. Balixafortide plus eribulin in HER2-negative metastatic breast cancer: a phase 1, single-arm, dose-escalation trial. Lancet Oncol. 2018;19:812–24.CrossRef
22.
go back to reference Rayson D, Lupichuk S, Potvin K, Dent S, Shenkier T, Dhesy-Thind S, et al. Canadian Cancer Trials Group IND197: a phase II study of foretinib in patients with estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2-negative recurrent or metastatic breast cancer. Breast Cancer Res Treat. 2016;157:109–16.CrossRef Rayson D, Lupichuk S, Potvin K, Dent S, Shenkier T, Dhesy-Thind S, et al. Canadian Cancer Trials Group IND197: a phase II study of foretinib in patients with estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2-negative recurrent or metastatic breast cancer. Breast Cancer Res Treat. 2016;157:109–16.CrossRef
23.
go back to reference Tolaney SM, Nechushtan H, Ron IG, Schöffski P, Awada A, Yasenchak CA, et al. Cabozantinib for metastatic breast carcinoma: results of a phase II placebo-controlled randomized discontinuation study. Breast Cancer Res Treat. 2016;160:305–12.CrossRef Tolaney SM, Nechushtan H, Ron IG, Schöffski P, Awada A, Yasenchak CA, et al. Cabozantinib for metastatic breast carcinoma: results of a phase II placebo-controlled randomized discontinuation study. Breast Cancer Res Treat. 2016;160:305–12.CrossRef
24.
go back to reference Tolaney SM, Tan S, Guo H, Barry W, Van Allen E, Wagle N, et al. Phase II study of tivantinib (ARQ 197) in patients with metastatic triple-negative breast cancer. Investig New Drugs. 2015;33:1108–14.CrossRef Tolaney SM, Tan S, Guo H, Barry W, Van Allen E, Wagle N, et al. Phase II study of tivantinib (ARQ 197) in patients with metastatic triple-negative breast cancer. Investig New Drugs. 2015;33:1108–14.CrossRef
25.
go back to reference Vermeulen MA, Slaets L, Cardoso F, Giordano SH, Tryfonidis K, van Diest PJ, et al. Pathological characterisation of male breast cancer: results of the EORTC 10085/TBCRC/BIG/NABCG International Male Breast Cancer Program. Eur J Cancer. 2017;82:219–27.CrossRef Vermeulen MA, Slaets L, Cardoso F, Giordano SH, Tryfonidis K, van Diest PJ, et al. Pathological characterisation of male breast cancer: results of the EORTC 10085/TBCRC/BIG/NABCG International Male Breast Cancer Program. Eur J Cancer. 2017;82:219–27.CrossRef
26.
go back to reference Cardoso F, Bartlett JMS, Slaets L, van Deurzen CHM, van Leeuwen-Stok E, Porter P, et al. Characterization of male breast cancer: results of the EORTC 10085/TBCRC/BIG/NABCG International Male Breast Cancer Program. Ann Oncol. 2018;29:405–17.CrossRef Cardoso F, Bartlett JMS, Slaets L, van Deurzen CHM, van Leeuwen-Stok E, Porter P, et al. Characterization of male breast cancer: results of the EORTC 10085/TBCRC/BIG/NABCG International Male Breast Cancer Program. Ann Oncol. 2018;29:405–17.CrossRef
27.
go back to reference van der Vegt B, De Bock GH, Bart J, Zwartjes NG, Wesseling J. Validation of the 4B5 rabbit monoclonal antibody in determining Her2/neu status in breast cancer. Mod Pathol. 2009;22:879–86.CrossRef van der Vegt B, De Bock GH, Bart J, Zwartjes NG, Wesseling J. Validation of the 4B5 rabbit monoclonal antibody in determining Her2/neu status in breast cancer. Mod Pathol. 2009;22:879–86.CrossRef
28.
go back to reference Matte I, Lane D, Laplante C, Garde-Granger P, Rancourt C, Piché A. Ovarian cancer ascites enhance the migration of patient-derived peritoneal mesothelial cells via cMet pathway through HGF-dependent and -independent mechanisms. Int J Cancer. 2015;137:289–98.CrossRef Matte I, Lane D, Laplante C, Garde-Granger P, Rancourt C, Piché A. Ovarian cancer ascites enhance the migration of patient-derived peritoneal mesothelial cells via cMet pathway through HGF-dependent and -independent mechanisms. Int J Cancer. 2015;137:289–98.CrossRef
29.
go back to reference Han Z, Xiao Y, Wang K, Yan J, Xiao Z, Fang F, et al. Development of a SPECT tracer to image c-Met expression in a xenograft model of non-small cell lung cancer. J Nucl Med. 2018;59:1686–91.CrossRef Han Z, Xiao Y, Wang K, Yan J, Xiao Z, Fang F, et al. Development of a SPECT tracer to image c-Met expression in a xenograft model of non-small cell lung cancer. J Nucl Med. 2018;59:1686–91.CrossRef
30.
go back to reference Dmitriev P, Kiseleva E, Kharchenko O, Ivashkin E, Pichugin A, Dessen P, et al. Dux4 controls migration of mesenchymal stem cells through the Cxcr4-Sdf1 axis. Oncotarget. 2016;7:65090–108.CrossRef Dmitriev P, Kiseleva E, Kharchenko O, Ivashkin E, Pichugin A, Dessen P, et al. Dux4 controls migration of mesenchymal stem cells through the Cxcr4-Sdf1 axis. Oncotarget. 2016;7:65090–108.CrossRef
31.
go back to reference Zhang Y, Yang P, Sun T, Li D, Xu X, Rui Y, et al. MiR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol. 2013;15:284–94.CrossRef Zhang Y, Yang P, Sun T, Li D, Xu X, Rui Y, et al. MiR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol. 2013;15:284–94.CrossRef
32.
go back to reference Yamashita J, Ogawa M, Yamashita S, Nomura K, Kuramoto M, Saishoji T, et al. Immunoreactive hepatocyte growth factor is a strong and independent predictor of recurrence and survival in human breast cancer. Cancer Res. 1994;54:1630–3.PubMed Yamashita J, Ogawa M, Yamashita S, Nomura K, Kuramoto M, Saishoji T, et al. Immunoreactive hepatocyte growth factor is a strong and independent predictor of recurrence and survival in human breast cancer. Cancer Res. 1994;54:1630–3.PubMed
33.
go back to reference Mirisola V, Zuccarino A, Bachmeier BE, Sormani MP, Falter J, Nerlich A, et al. CXCL12/SDF1 expression by breast cancers is an independent prognostic marker of disease-free and overall survival. Eur J Cancer. 2009;45:2579–87.CrossRef Mirisola V, Zuccarino A, Bachmeier BE, Sormani MP, Falter J, Nerlich A, et al. CXCL12/SDF1 expression by breast cancers is an independent prognostic marker of disease-free and overall survival. Eur J Cancer. 2009;45:2579–87.CrossRef
34.
go back to reference Kobayashi T, Tsuda H, Moriya T, Yamasaki T, Kikuchi R, Ueda S, et al. Expression pattern of stromal cell-derived factor-1 chemokine in invasive breast cancer is correlated with estrogen receptor status and patient prognosis. Breast Cancer Res Treat. 2010;123:733–45.CrossRef Kobayashi T, Tsuda H, Moriya T, Yamasaki T, Kikuchi R, Ueda S, et al. Expression pattern of stromal cell-derived factor-1 chemokine in invasive breast cancer is correlated with estrogen receptor status and patient prognosis. Breast Cancer Res Treat. 2010;123:733–45.CrossRef
35.
go back to reference Liu H, Li Z, Deng M, Liu Q, Zhang T, Guo W, et al. Prognostic and clinicopathological value of CXCL12/SDF1 expression in breast cancer: a meta-analysis. Clin Chim Acta. 2018;484:72–80.CrossRef Liu H, Li Z, Deng M, Liu Q, Zhang T, Guo W, et al. Prognostic and clinicopathological value of CXCL12/SDF1 expression in breast cancer: a meta-analysis. Clin Chim Acta. 2018;484:72–80.CrossRef
36.
go back to reference Moelans CB, de Ligt J, van der Groep P, Prins P, Besselink NJ, Hoogstraat M, et al. The molecular genetic make-up of male breast cancer. Endocr Relat Cancer. 2019;26:779–94.CrossRef Moelans CB, de Ligt J, van der Groep P, Prins P, Besselink NJ, Hoogstraat M, et al. The molecular genetic make-up of male breast cancer. Endocr Relat Cancer. 2019;26:779–94.CrossRef
37.
go back to reference Mavrogonatou E, Pratsinis H, Kletsas D. The role of senescence in cancer development. Semin Cancer Biol. 2019;S1044-579X:30127–0. Mavrogonatou E, Pratsinis H, Kletsas D. The role of senescence in cancer development. Semin Cancer Biol. 2019;S1044-579X:30127–0.
Metadata
Title
High hepatocyte growth factor expression in primary tumor predicts better overall survival in male breast cancer
Authors
Si-Qi Qiu
Johan van Rooijen
Hilde H. Nienhuis
Bert van der Vegt
Hetty Timmer-Bosscha
Elise van Leeuwen-Stok
Annemiek M. E. Walenkamp
Carolien H. M. van Deurzen
Geertruida H. de Bock
Elisabeth G. E. de Vries
Carolien P. Schröder
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2020
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-020-01266-x

Other articles of this Issue 1/2020

Breast Cancer Research 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine