Skip to main content
Top
Published in: BMC Cancer 1/2017

Open Access 01-12-2017 | Research article

The acceleration of glucose accumulation in renal cell carcinoma assessed by FDG PET/CT demonstrated acquisition of resistance to tyrosine kinase inhibitor therapy

Authors: Noboru Nakaigawa, Keiichi Kondo, Daiki Ueno, Kazuhiro Namura, Kazuhide Makiyama, Kazuki Kobayashi, Koichi Shioi, Ichiro Ikeda, Takeshi Kishida, Tomohiro Kaneta, Ryogo Minamimoto, Ukihide Tateishi, Tomio Inoue, Masahiro Yao

Published in: BMC Cancer | Issue 1/2017

Login to get access

Abstract

Background

Tyrosine-kinase inhibitor (TKI) targeting angiogenesis improves the prognosis of patients with metastatic renal cell carcinoma (RCC), but its effect is temporary. In order to understand the mechanism by which RCC acquires resistance to TKI, we investigated the change of glucose accumulation in RCC by FDG PET/CT when they demonstrated progression disease (PD) against TKI.

Methods

We monitored the FDG accumulation in RCC of 38 patients treated with TKI by 162 PET/CT sequentially until they were judged to demonstrate PD. Standardized uptake value (SUV), a simplified index of tissue FDG accumulation rate, was measured, and the sequential changes of max SUVmax (the highest SUV in an individual patient) was analyzed. Additionally, the expression of glucose transporter 1 (GLUT-1) and associated proteins in 786-O cells cultured under hypoxia were analyzed.

Results

The 10 patients with RCC which FDG accumulation was accelerated after beginning of TKI treatment demonstrated PD soon. The other 28 patients with RCC which FDG accumulation was suppressed by TKI showed longer progression-free survival (3.6 months vs 6.5 months, P = 0.0026), but this suppression in most cases (96%) was temporary and FDG accumulation was accelerated when tumor demonstrated PD. Interestingly, the FDG accumulation at PD was higher than that before TKI treatment in the half cases. The acceleration of FDG accumulation was suppressed by following treatment by mammalian target of rapamycin (mTOR) inhibitor. Additionally, in vitro assay demonstrated that the expression of GLUT-1 was increased in the RCC cells surviving under hypoxia condition via mTOR pathway.

Conclusions

The acceleration of glucose accumulation dependent on mTOR in RCC assessed by FDG PET/CT demonstrated acquisition of resistance to TKI. FDG PET/CT had potential as an assessment method monitoring not only the initial response but also following status of RCC during TKI treatment.

Trial registration

UMIN0000008141, 11 Jun 2012. This trial was retrospectively registered.
Literature
1.
2.
go back to reference Chow WH, Devesa SS, Warren JL, Fraumeni Jr JF. Rising incidence of renal cell carcinoma in the United States. JAMA. 1999;281:1628–31.CrossRefPubMed Chow WH, Devesa SS, Warren JL, Fraumeni Jr JF. Rising incidence of renal cell carcinoma in the United States. JAMA. 1999;281:1628–31.CrossRefPubMed
3.
go back to reference Janzen NK, Kim HL, Figlin RA, Belldegrun AS. Surveillance after radical or partial nephrectomy for localized renal cell carcinoma and management of recurrent disease. Urol Clin North Am. 2003;30:843–52.CrossRefPubMed Janzen NK, Kim HL, Figlin RA, Belldegrun AS. Surveillance after radical or partial nephrectomy for localized renal cell carcinoma and management of recurrent disease. Urol Clin North Am. 2003;30:843–52.CrossRefPubMed
5.
go back to reference Negrier S, Escudier B, Lasset C, Douillard JY, Savary J, Chevreau C, et al. Recombinant human interleukin-2, recombinant human interferon alfa-2a, or both in metastatic renal cell carcinoma: Groupe Français D’Immunotherapie. N Engl J Med. 1998;338:1272–8.CrossRefPubMed Negrier S, Escudier B, Lasset C, Douillard JY, Savary J, Chevreau C, et al. Recombinant human interleukin-2, recombinant human interferon alfa-2a, or both in metastatic renal cell carcinoma: Groupe Français D’Immunotherapie. N Engl J Med. 1998;338:1272–8.CrossRefPubMed
6.
go back to reference Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal cell carcinoma. N Engl J Med. 2007;356:115–24.CrossRefPubMed Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal cell carcinoma. N Engl J Med. 2007;356:115–24.CrossRefPubMed
7.
go back to reference Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear cell renal cell carcinoma. N Engl J Med. 2007;356:125–34.CrossRefPubMed Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear cell renal cell carcinoma. N Engl J Med. 2007;356:125–34.CrossRefPubMed
8.
go back to reference Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Oudard S, et al. Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J Clin Oncol. 2009;27:3584–90.CrossRefPubMedPubMedCentral Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Oudard S, et al. Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J Clin Oncol. 2009;27:3584–90.CrossRefPubMedPubMedCentral
9.
go back to reference Namura K, Minamimoto R, Yao M, Makiyama K, Murakami T, Sano F, et al. Impact of maximum standardized uptake value (SUVmax) evaluated by 18-Fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography (18 F-FDG-PET/CT) on survival for patients with advanced renal cell carcinoma: a preliminary report. BMC Cancer. 2010;10:667.CrossRefPubMedPubMedCentral Namura K, Minamimoto R, Yao M, Makiyama K, Murakami T, Sano F, et al. Impact of maximum standardized uptake value (SUVmax) evaluated by 18-Fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography (18 F-FDG-PET/CT) on survival for patients with advanced renal cell carcinoma: a preliminary report. BMC Cancer. 2010;10:667.CrossRefPubMedPubMedCentral
10.
go back to reference Nakaigawa N, Kondo K, Tateishi U, Minamimoto R, Kaneta T, Namura K, et al. FDG PET/CT as a prognostic biomarker in the era of molecular-targeting therapies: max SUVmax predicts survival of patients with advanced renal cell carcinoma. BMC Cancer. 2016;16:67.CrossRefPubMedPubMedCentral Nakaigawa N, Kondo K, Tateishi U, Minamimoto R, Kaneta T, Namura K, et al. FDG PET/CT as a prognostic biomarker in the era of molecular-targeting therapies: max SUVmax predicts survival of patients with advanced renal cell carcinoma. BMC Cancer. 2016;16:67.CrossRefPubMedPubMedCentral
11.
go back to reference Ueno D, Yao M, Tateishi U, Minamioto R, Makiyama K, Hayashi N, et al. Early assessment by FDG-PET/CT of patients with advanced renal cell carcinoma treated with tyrosine kinase inhibitors is predictive of disease course. BMC Cancer. 2012;12:162.CrossRefPubMedPubMedCentral Ueno D, Yao M, Tateishi U, Minamioto R, Makiyama K, Hayashi N, et al. Early assessment by FDG-PET/CT of patients with advanced renal cell carcinoma treated with tyrosine kinase inhibitors is predictive of disease course. BMC Cancer. 2012;12:162.CrossRefPubMedPubMedCentral
12.
go back to reference Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–47.CrossRefPubMed Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–47.CrossRefPubMed
13.
go back to reference Lyrdal D, Boijsen M, Suurküla M, Lundstam S, Stierner U. Evaluation of sorafenib treatment in metastatic renal cell carcinoma with 2-fluoro-2-deoxyglucose positron emission tomography and computed tomography. Nucl Med Commun. 2009;30:519–24.CrossRefPubMed Lyrdal D, Boijsen M, Suurküla M, Lundstam S, Stierner U. Evaluation of sorafenib treatment in metastatic renal cell carcinoma with 2-fluoro-2-deoxyglucose positron emission tomography and computed tomography. Nucl Med Commun. 2009;30:519–24.CrossRefPubMed
14.
go back to reference Vercellino L, Bousquet G, Baillet G, Barré E, Mathieu O, Just PA, et al. 18 F-FDG PET/CT imaging for an early assessment of response to sunitinib in metastatic renal carcinoma: preliminary study. Cancer Biother Radiopharm. 2009;24:137–44.CrossRefPubMed Vercellino L, Bousquet G, Baillet G, Barré E, Mathieu O, Just PA, et al. 18 F-FDG PET/CT imaging for an early assessment of response to sunitinib in metastatic renal carcinoma: preliminary study. Cancer Biother Radiopharm. 2009;24:137–44.CrossRefPubMed
15.
go back to reference Kayani I, Avril N, Bomanji J, Chowdhury S, Rockall A, Sahdev A, et al. Sequential FDG-PET/CT as a biomarker of response to Sunitinib in metastatic clear cell renal cancer. Clin Cancer Res. 2011;17:6021–8.CrossRefPubMed Kayani I, Avril N, Bomanji J, Chowdhury S, Rockall A, Sahdev A, et al. Sequential FDG-PET/CT as a biomarker of response to Sunitinib in metastatic clear cell renal cancer. Clin Cancer Res. 2011;17:6021–8.CrossRefPubMed
16.
go back to reference Sudarshan S, Karam JA, Brugarolas J, Thompson RH, Uzzo R, Rini B, et al. Metabolism of kidney cancer: from the lab to clinical practice. Eur Urol. 2013;63:244–51.CrossRefPubMed Sudarshan S, Karam JA, Brugarolas J, Thompson RH, Uzzo R, Rini B, et al. Metabolism of kidney cancer: from the lab to clinical practice. Eur Urol. 2013;63:244–51.CrossRefPubMed
17.
go back to reference Linehan WM, Rouault TA. Molecular pathways: fumarate hydratase-deficient kidney cancer--targeting the Warburg effect in cancer. Clin Cancer Res. 2013;19:3345–52.CrossRefPubMedPubMedCentral Linehan WM, Rouault TA. Molecular pathways: fumarate hydratase-deficient kidney cancer--targeting the Warburg effect in cancer. Clin Cancer Res. 2013;19:3345–52.CrossRefPubMedPubMedCentral
18.
go back to reference Jiménez-Valerio G, Martínez-Lozano M, Bassani N, Vidal A, Ochoa-de-Olza M, Suárez C, et al. Resistance to antiangiogenic therapies by metabolic symbiosis in renal cell carcinoma PDX models and patients. Cell Rep. 2016;15(6):1134–43.CrossRefPubMedPubMedCentral Jiménez-Valerio G, Martínez-Lozano M, Bassani N, Vidal A, Ochoa-de-Olza M, Suárez C, et al. Resistance to antiangiogenic therapies by metabolic symbiosis in renal cell carcinoma PDX models and patients. Cell Rep. 2016;15(6):1134–43.CrossRefPubMedPubMedCentral
19.
go back to reference Allen E, Miéville P, Warren CM, Saghafinia S, Li L, Peng MW, Hanahan D. Metabolic symbiosis enables adaptive resistance to anti-angiogenic therapy that is dependent on mTOR signaling. Cell Rep. 2016;15(6):1144–60.CrossRefPubMedPubMedCentral Allen E, Miéville P, Warren CM, Saghafinia S, Li L, Peng MW, Hanahan D. Metabolic symbiosis enables adaptive resistance to anti-angiogenic therapy that is dependent on mTOR signaling. Cell Rep. 2016;15(6):1144–60.CrossRefPubMedPubMedCentral
20.
go back to reference Pisarsky L, Bill R, Fagiani E, Dimeloe S, Goosen RW, Hagmann J, Hess C, Christofori G. Targeting metabolic symbiosis to overcome resistance to anti-angiogenic therapy. Cell Rep. 2016;15(6):1161–74.CrossRefPubMedPubMedCentral Pisarsky L, Bill R, Fagiani E, Dimeloe S, Goosen RW, Hagmann J, Hess C, Christofori G. Targeting metabolic symbiosis to overcome resistance to anti-angiogenic therapy. Cell Rep. 2016;15(6):1161–74.CrossRefPubMedPubMedCentral
22.
go back to reference Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–92.CrossRefPubMedPubMedCentral Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–92.CrossRefPubMedPubMedCentral
Metadata
Title
The acceleration of glucose accumulation in renal cell carcinoma assessed by FDG PET/CT demonstrated acquisition of resistance to tyrosine kinase inhibitor therapy
Authors
Noboru Nakaigawa
Keiichi Kondo
Daiki Ueno
Kazuhiro Namura
Kazuhide Makiyama
Kazuki Kobayashi
Koichi Shioi
Ichiro Ikeda
Takeshi Kishida
Tomohiro Kaneta
Ryogo Minamimoto
Ukihide Tateishi
Tomio Inoue
Masahiro Yao
Publication date
01-12-2017
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2017
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-3044-0

Other articles of this Issue 1/2017

BMC Cancer 1/2017 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