Skip to main content

Immunohistochemical expression of the hepatocyte growth factor in chromophobe renal cell carcinoma

Abstract

Background

The prognostic value of Hepatocyte growth factor (HGF) in non-clear cell renal cell carcinoma (RCC) is still unclear. The aim of this study is to evaluate the prognostic impact of HGF expression in a large cohort of chromophobe RCC (chRCC).

Methods

Patients who underwent renal surgery due to chRCC were recruited. Clinical data was retrospectively evaluated. Tumor specimen were analyzed for HGF expression by immunohistochemistry.

Results

81 chRCC patients were eligible for analysis, thereof 37 (45.7%) patients were positive for HGF. No significant associations were found for HGF expression and clinical attributes in patients with chRCC. Kaplan-Meier analysis revealed no differences in 5-year overall survival (OS) for patients with HGF compared to HGF+ tumors (95.0% versus 90.9%; p = 0.410).

Conclusions

In chRCC HGF expression is not associated with parameters of aggressiveness or survival.

Peer Review reports

Background

Hepatocyte growth factor (HGF), also known as scatter factor, is the natural endogenous ligand of the Mesenchymal-epithelial transition factor (MET) receptor. The HGF/MET pathway is known to be associated with the appearance of several attributes of cancer and is utilized as a relevant target across many solid tumors [1]. To create active HGF, enzymes such as serum HGF activator and cellular type II transmembrane serine proteinases proteolytically cleave pro-HGF at the Arg494-Val495 bond [2]. The result is a bioactive form of HGF, a high-affinity ligand for the MET receptor: αβ-HGF. The structure of αβ-HGF shows a disulfide-linked heterodimer of an α-chain (69 kD) with β-chain subunits (34 kD) [3, 4]. Pro-HGF also binds the MET receptor, but only with very low affinity compared to αβ-HGF [5]. Only αβ-HGF can lead to overactivation of HGF/MET axis. This overactivation is known to result in tumor progression and tumorigenesis in various cancer types. Several clinical studies have addressed the role of the HGF/MET pathway in diverse types of cancer, such as gastric [6], colorectal [7], breast [8], hepatocellular [9], pancreatic [10], lung [11] and renal cancer [12]. It is a promising approach to use HGF as a prognostic marker to identify patients who gain the most benefit from HGF/MET targeted therapies. The predictive value of MET biomarkers is indisputable, as shown in several studies. High predictive values for response to foretinib and savolitinib in papillary renal cell carcinoma could be verified [13].

A broad spectrum of histopathological entities in renal cell carcinomas (RCC) is described in the 2022 World Health Organization (WHO) classification, which shows the Chromophobe renal cell carcinoma (chRCC) being the third most common (5–7%) of RCCs [14]. The chRCC shows a favourable prognosis, with high 5-year recurrence-free survival (RFS), and 10-year cancer-specific survival (CSS) [15]. Despite the fact that there is a low risk of tumor progression in patients with chRCC, some patients show development of metastatic disease with poor prognosis. There is no grading system established for this tumor subtype and due to its innate nuclear atypia, it cannot be graded by the Fuhrman grading system [14]. So far, no prognostic biomarkers exist in chRCC, leaving tumor stage, necrosis and sarcomatoid change as the most important prognostic factor for the chRCC.

Therefore, the aim of this study was to evaluate the prognostic impact of HGF in chRCC. To the best of our knowledge, this is the first study which analyzed this aspect in the third most common RCC subtype.

Methods

Patients and tumor characteristics

For this study 81 patients were included, who were diagnosed with chRCC after surgery by using the electronic pathology register between 1996 and 2014. Relevant clinical attributes relating to each tissue sample were collected with regard to tumor stage and histological subtype according to the Union internationale contre le cancer (UICC) 2010 TNM tumor staging system. Suitable specimens were selected by a pathologist (FE) and tissue micro arrays (TMA) were prepared from the primary tumor as previously described [16]. The histological subtype was confirmed by a second uropathologist (AH) after performing immunohistochemistry (IHC; CD177 and CK7). Follow up data was inquired from electronic patient charts. To confirm overall survival (OS), data was verified by the Munich Cancer Registry of the Munich Tumor Center. The Ethics Committee of the Technical University of Munich (384/13) authorized this study with due regard to the German Human Research Act and the Declaration of Helsinki.

Procedures

Expression of HGF was determined by IHC. 2 μm TMA slides were stained for HGF (R&D Systems, AF-294-NA, dilution 1:40) with a fully automated Dako Autostainer (Dako, Agilent pathology systems). For antigen retrieval a pH of 7.2 was accomplished. For the visualization of bound primary antibody EnVision Detection Kit (Dako, EnVision + System-HRP) was used. Therefore, sections were rinsed in tap water, counterstained with Mayer’s Hematoxylin solution and finally mounted. As positive control paraffin-embedded human colon tissue was used. All stained tissue samples were assessed in an anonymized way by a pathologist (FE). The evaluation was performed under a Leitz ARISTOPLAN light microscope (Leica Microsystems, Germany) with a x10 eyepiece, a 22-mm field of view and x40 objective lens (Plan FLUOTAR x40/0.70).

The staining reaction was classified on all available tumor surface according to a semi-quantitative IHC reference scale previously described [17]. HGF was localized primarily on the membrane and partly in the cytoplasm of tumor cells. Tissue with sarcomatoid change, growth pattern, and necrosis were not punched.

The staining intensity was scored from 0 to 3 (0 = no staining, 1 = weak staining (pale yellow), 2 = moderate staining (yellowish brown), 3 = strong staining (brown)) according to the H-score as already described (Fig. 1) [18,19,20]. The area of staining was evaluated in percent (0-100%), a staining intensity score was defined by multiplying the score with the stained area (Table 1) [17, 21]. To dichotomize our patient collective, we defined the median of observed distribution as the cut off, since there is no normative data in the literature regarding the staining intensity of cell membrane or cell cytoplasm. Because of the limited number of cases we used the median as a binary cutoff. Since the median was 0, an HGF staining score of 0 was defined as HGF, and a staining higher than 0 was defined as HGF+.

figure 1

Immunohistochemical staining of HGF in chromophobe renal cell carcinoma specimen. (a) positive (40x magnification) (b) negative (40x magnification)

Table 1 Pathohistological characteristics of the chRCC samples (n = 81)

Statistical analysis

The primary endpoint of the study was OS. In the absence of death, the endpoint was censored at the last date of follow-up. Follow-up was defined by the time span from the date of the surgery to the date of death or from surgery to the last known date of follow-up.

To analyze patient/tumor characteristics in relation to corresponding subgroups with or without HGF expression, several tests were used in correlation to the nature of the requested variable: Fisher’s exact tests, chi-square, independent t-test, and Mann-Whitney U-Test. Kaplan-Meier survival times were estimated, with subgroups being compared using the log-rank test. SPSS 27.0 (USA) was used for statistical assessment. Two-sided p-values below 0.05 were considered statistically significant.

Results

Patients´ characteristics and HGF expression

The median age of the cohort was 59.8 (range: 31–79) years. Of the patients 60 (74.1%), 14 (17.3%) and 7 (8.6%) presented with pT1, pT2 and pT3 tumors, respectively. 86.4% of the patients had AJCC Stage I/II. Furthermore, 6 (7.4%) of all patients presented with lymph node metastasis or synchronous distant metastasis. HGF expression was found in 37 (45.7%) of the chRCC TMA specimens, respectively (Fig. 1). No associations between HGF+ expression and patient or tumor characteristics (including lymph node metastasis, distant metastasis, Cancer Stage (AJCC) and T-stage) were identified (Table 2).

Table 2 chRCC patient´s and tumor characteristics in dependence of HGF expression

HGF expression and clinical course

Median follow-up was 40.5 (IQR: 10.8-109.3) months. At the time of last follow-up, 46 (56.8%) patients were alive, 9 (11.1%) patients died and 26 (32.1%) patients were lost to follow up. In the subgroups of HGF vs. HGF+ 25 (56.8%) vs. 21 (56.8%) patients were alive, 3 (6.8%) vs. 6 (16.2%) patients had died and 16 (36.4%) vs. 10 (27.0%) patients were lost to follow up (p = 0.342, chi square).

Kaplan-Meier analysis disclosed a 5 year- OS for HGF compared to HGF+ tumors of 95.0% compared to 90.9% (p = 0.410, log rank) (Fig. 2).

figure 2

5-year overall survival for patients with chromophobe renal cell carcinoma in dependence of HGF expression. Kaplan-Meier analysis disclosed a 5 year- OS for HGF- compared to HGF+ tumors of 95.0% compared to 90.9% (p = 0.410, log rank) (Fig. 2)

Discussion

To the best of our knowledge, this is the first study which analyzed the prognostic role of HGF expression in chRCC. In view of the fact that the HGF/MET pathway is relevant in tumor progression and tumorigenesis in diverse types of cancer [6,7,8,9,10,11,12, 22], it seems obvious that therapeutic approaches are expected to target either the ligand (HGF) or the receptor (MET). There are already available therapeutic options targeting MET (c-MET inhibitors, e.g. Cabozantinib, Crizotinib and Capmatinib) [23]. The study by Silva Paiva et al. relates to c-Met expression in renal cell carcinoma with bone metastases and shows a detailed overview of therapeutic options, targeting the c-Met pathway [22]. Despite the strong pre-clinical evidence that simultaneous targeting of both the ligand and the receptor, offers the most effective approach for reducing cancer, clinical investigations regarding this setting are rare. Recently, promising pre-clinical results have led to clinical trials in patients with refractory solid tumors using YYB101, a HGF neutralising antibody [24]. YYB102 leads to almost total inhibition of the HGF/MET pathway by binding to the alpha chain of HGF resulting in thoroughly blocking HGF from binding c-MET [24]. However, there are no clinical trials using YYB101 in RCC. Clinical trials are currently limited to Phase 1b/2a clinical trial in metastatic colorectal cancer patients in Korea [24]. The focus of our study lays on HGF as a promising prognostic marker, knowing that the HGF/MET pathway is one of the most important pro-oncogenic, pro-angiogenetic, and pro-metastatic signals in various cancer types [25]. Despite promising pre-clinical evidence regarding the HGF/MET pathway in numerous solid types of cancer, results in our study show a surprisingly low clinical impact of HGF expression in chRCC.

Several evidences have shown that HGF/Met signaling is substantial in cancer cells for the maintenance of self-renewal mechanisms and even development of chemoresistance [26]. Currently, research results indicated that HGF/Met signaling could support mechanisms for immune escape of cancer cells, so new approaches are being developed, to combine MET and programmed cell death receptor-1 (PD-1)/programmed cell death receptor-ligand 1 (PD-L1) inhibition in drug design and targeted therapy [25, 26].

There are of course several limitations evaluating these results: the study design uses a retrospective analysis. Due to the rare incidence of this tumor subtype the number of cases is limited, however, 81 chRCC cases can be considered as a large cohort. Additional limitations are the use of TMAs and the methodology of immunohistochemistry.

Conclusions

In conclusion our study aimed to evaluate HGF as a prognostic marker in chRCC, using a large group of 81 (26 lost to follow-up) patients diagnosed with chRCC. However, HGF did not qualify as a prognostic marker in chRCC for survival.

Data Availability

The datasets generated and/or analyzed during the current study are not publicly available due to medical confidentiality regarding patients’ data but are available from the corresponding author on reasonable request.

Abbreviations

AJCC:

American Joint Committee on Cancer

chRCC:

Chromophobe renal cell carcinoma

CSS:

Cancer specific survival

HGF:

hepatocyte growth factor

IHC:

Immunohistochemistry

MET:

Mesenchymal-epithelial transition factor

OS:

Overall survival

RCC:

Renal cell carcinoma

TMA:

Tissue micro arrays

UICC:

Union internationale contre le cancer

WHO:

World Health Organization

References

  1. Moosavi F, Giovannetti E, Saso L, Firuzi O. HGF/MET pathway aberrations as diagnostic, prognostic, and predictive biomarkers in human cancers. Crit Rev Clin Lab Sci. 2019;56(8):533–66.

    Article  CAS  PubMed  Google Scholar 

  2. Fukushima T, Uchiyama S, Tanaka H, Kataoka H. Hepatocyte Growth Factor Activator: A Proteinase Linking Tissue Injury with Repair. Int J Mol Sci. 2018;19(11).

  3. Zhang Y, Xia M, Jin K, Wang S, Wei H, Fan C, et al. Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol Cancer. 2018;17(1):45.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Organ SL, Tsao MS. An overview of the c-MET signaling pathway. Ther Adv Med Oncol. 2011;3(1 Suppl):7–S19.

    Article  Google Scholar 

  5. Naldini L, Vigna E, Bardelli A, Follenzi A, Galimi F, Comoglio PM. Biological activation of pro-HGF (hepatocyte growth factor) by urokinase is controlled by a stoichiometric reaction. J Biol Chem. 1995;270(2):603–11.

    Article  CAS  PubMed  Google Scholar 

  6. Kim HS, Chon HJ, Kim H, Shin SJ, Wacheck V, Gruver AM, et al. MET in gastric cancer with liver metastasis: the relationship between MET amplification and met overexpression in primary stomach tumors and liver metastasis. J Surg Oncol. 2018;117(8):1679–86.

    Article  CAS  PubMed  Google Scholar 

  7. Huang CY, Zhou QY, Hu Y, Wen Y, Qiu ZW, Liang MG, et al. Hepatocyte growth factor is a prognostic marker in patients with colorectal cancer: a meta-analysis. Oncotarget. 2017;8(14):23459–69.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Wang M, Liang L, Lei X, Multani A, Meric-Bernstam F, Tripathy D, et al. Evaluation of cMET aberration by immunohistochemistry and fluorescence in situ hybridization (FISH) in triple negative breast cancers. Ann Diagn Pathol. 2018;35:69–76.

    Article  CAS  PubMed  Google Scholar 

  9. Rimassa L, Abbadessa G, Personeni N, Porta C, Borbath I, Daniele B, et al. Tumor and circulating biomarkers in patients with second-line hepatocellular carcinoma from the randomized phase II study with tivantinib. Oncotarget. 2016;7(45):72622–33.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Neuzillet C, Couvelard A, Tijeras-Raballand A, de Mestier L, de Gramont A, Bedossa P, et al. High c-Met expression in stage I-II pancreatic adenocarcinoma: proposal for an immunostaining scoring method and correlation with poor prognosis. Histopathology. 2015;67(5):664–76.

    Article  PubMed  Google Scholar 

  11. Gao HF, Li AN, Yang JJ, Chen ZH, Xie Z, Zhang XC, et al. Soluble c-Met levels correlated with tissue c-Met protein expression in patients with Advanced Non-Small-Cell Lung Cancer. Clin Lung Cancer. 2017;18(1):85–91.

    Article  CAS  PubMed  Google Scholar 

  12. Kim JH, Kim BJ, Kim HS. Clinicopathological impacts of high c-Met expression in renal cell carcinoma: a meta-analysis and review. Oncotarget. 2017;8(43):75478–87.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Choueiri TK, Vaishampayan U, Rosenberg JE, Logan TF, Harzstark AL, Bukowski RM, et al. Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. J Clin Oncol. 2013;31(2):181–6.

    Article  CAS  PubMed  Google Scholar 

  14. Moch H, Amin MB, Berney DM, Compérat EM, Gill AJ, Hartmann A, et al. The 2022 World Health Organization classification of Tumours of the urinary system and male genital Organs-Part A: renal, Penile, and testicular tumours. Eur Urol. 2022;82(5):458–68.

    Article  PubMed  Google Scholar 

  15. Volpe A, Novara G, Antonelli A, Bertini R, Billia M, Carmignani G, et al. Chromophobe renal cell carcinoma (RCC): oncological outcomes and prognostic factors in a large multicentre series. BJU Int. 2012;110(1):76–83.

    Article  PubMed  Google Scholar 

  16. Erlmeier F, Feuchtinger A, Borgmann D, Rudelius M, Autenrieth M, Walch AK, et al. Supremacy of modern morphometry in typing renal oncocytoma and malignant look-alikes. Histochem Cell Biol. 2015;144(2):147–56.

    Article  CAS  PubMed  Google Scholar 

  17. Kurokawa Y, Matsuura N, Kawabata R, Nishikawa K, Ebisui C, Yokoyama Y, et al. Prognostic impact of major receptor tyrosine kinase expression in gastric cancer. Ann Surg Oncol. 2014;21(Suppl 4):584–90.

    Article  Google Scholar 

  18. Birks DK, Kleinschmidt-DeMasters BK, Donson AM, Barton VN, McNatt SA, Foreman NK, et al. Claudin 6 is a positive marker for atypical teratoid/rhabdoid tumors. Brain Pathol. 2010;20(1):140–50.

    Article  CAS  PubMed  Google Scholar 

  19. Gao F, Li M, Xiang R, Zhou X, Zhu L, Zhai Y. Expression of CLDN6 in tissues of gastric cancer patients: Association with clinical pathology and prognosis. Oncol Lett. 2019;17(5):4621–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Wang Q, Zhang Y, Zhang T, Han ZG, Shan L. Low claudin-6 expression correlates with poor prognosis in patients with non-small cell lung cancer. Onco Targets Ther. 2015;8:1971–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Phan LM, Fuentes-Mattei E, Wu W, Velazquez-Torres G, Sircar K, Wood CG, et al. Hepatocyte growth Factor/cMET pathway activation enhances Cancer Hallmarks in Adrenocortical Carcinoma. Cancer Res. 2015;75(19):4131–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Silva Paiva R, Gomes I, Casimiro S, Fernandes I, Costa L. c-Met expression in renal cell carcinoma with bone metastases. J Bone Oncol. 2020;25:100315.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Pothula SP, Xu Z, Goldstein D, Pirola RC, Wilson JS, Apte MV. Targeting HGF/c-MET Axis in Pancreatic Cancer. Int J Mol Sci. 2020;21(23).

  24. Kim ST, Hong JY, Park SH, Park JO, Park YW, Park N, et al. First-in-human phase I trial of anti-hepatocyte growth factor antibody (YYB101) in refractory solid tumor patients. Ther Adv Med Oncol. 2020;12:1758835920926796.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Papaccio F, Della Corte CM, Viscardi G, Di Liello R, Esposito G, Sparano F et al. HGF/MET and the Immune System: Relevance for Cancer Immunotherapy. Int J Mol Sci. 2018;19(11).

  26. Zambelli A, Biamonti G, Amato A. HGF/c-Met signalling in the Tumor Microenvironment. Adv Exp Med Biol. 2021;1270:31–44.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Ulrike Mühlthaler for her assistance with the HGF immunohistochemistry.

Funding

Open Access funding enabled and organized by Projekt DEAL. This work was supported by a grant of the Deutsche Forschungsgemeinschaft (DFG), GZ:ER 795/1–1 (Franziska Erlmeier).

Author information

Authors and Affiliations

Authors

Contributions

ME, SS and FE participated in the data interpretation and drafting of the manuscript. MM performed the statistical analysis. FE carried out the data acquisition. SZ, MA, WW and AH revised the manuscript for important intellectual content. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Marie Mikuteit.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

The study was approved by the Ethics Committee of the Technical University of Munich (384/13) and was performed in accordance with the German Human Research Act and with the Declaration of Helsinki. Informed written consent was obtained from all individual participants included in the study. Details that disclose the identity of the subjects under study were omitted.

Consent for publication

Not applicable.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Erlmeier, M., Mikuteit, M., Zschäbitz, S. et al. Immunohistochemical expression of the hepatocyte growth factor in chromophobe renal cell carcinoma. BMC Urol 23, 90 (2023). https://doi.org/10.1186/s12894-023-01263-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12894-023-01263-0

Keywords