, Gene expression and functional annotation clustering analysis. A: Annotation clusters of the most up-regulated genes associated with apoptosis

G. Ontology, GO) terms. B: Annotation clusters of the most down-regulated genes associated with extracellular matrix and GTP binding GO terms

, C: Real-time quantitative reverse transcription PCR of the relative aldo-keto reductase family 1. member B10 (AKR1B10) mRNA level in HK-2, DAVID bioinformatics database 6.7 version

V. Launonen, O. Vierimaa, M. Kiuru, J. Isola, S. Roth et al., Inherited susceptibility to uterine leiomyomas and renal cell cancer, Proceedings of the National Academy of Sciences, vol.68, issue.4, pp.3387-3392, 2001.
DOI : 10.1073/pnas.68.4.820

H. Lehtonen, M. Kiuru, S. Ylisaukko-oja, R. Salovaara, R. Herva et al., Increased risk of cancer in patients with fumarate hydratase germline mutation, Journal of Medical Genetics, vol.43, issue.6, pp.523-526, 2006.
DOI : 10.1136/jmg.2005.036400

URL : http://europepmc.org/articles/pmc2564537?pdf=render

W. Reed, R. Walker, and R. Horowitz, Cutaneous leiomyomata with uterine leiomyomata, Acta Derm Venereol, vol.53, pp.409-416, 1973.

I. Tomlinson, N. Alam, A. Rowan, E. Barclay, E. Jaeger et al., Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer, Nat Genet, vol.30, pp.406-410, 2002.

J. Toro, M. Nickerson, M. Wei, M. Warren, G. Glenn et al., Mutations in the Fumarate Hydratase Gene Cause Hereditary Leiomyomatosis and Renal Cell Cancer in Families in North America, The American Journal of Human Genetics, vol.73, issue.1, pp.95-106, 2003.
DOI : 10.1086/376435

URL : https://doi.org/10.1086/376435

M. Merino, C. Torres-cabala, P. Pinto, and W. Linehen, The Morphologic Spectrum of Kidney Tumors in Hereditary Leiomyomatosis and Renal Cell Carcinoma (HLRCC) Syndrome, The American Journal of Surgical Pathology, vol.31, issue.10
DOI : 10.1097/PAS.0b013e31804375b8

, Am J Surg Pathol, vol.31, pp.1578-1585, 2007.

M. Fernandez-pugnaire and V. Delgado-florencio, Familial Multiple Cutaneous Leiomyomas, Dermatology, vol.191, issue.4, pp.295-298, 1995.
DOI : 10.1159/000246578

T. Weaver, D. Levitt, M. Donnelly, P. Stevens, and L. Banaszak, The multisubunit active site of fumarase C from Escherichia coli, Nature Structural Biology, vol.6, issue.8, pp.654-662, 1995.
DOI : 10.1016/0263-7855(88)80054-7

T. Weaver, M. Lees, V. Zaitsev, I. Zaitseva, E. Duke et al., Crystal structures of native and recombinant yeast fumarase 1 1Edited by D. Rees, Journal of Molecular Biology, vol.280, issue.3, pp.431-442, 1998.
DOI : 10.1006/jmbi.1998.1862

J. Isaacs, Y. Jung, D. Mole, S. Lee, C. Torres-cabala et al., HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: Novel role of fumarate in regulation of HIF stability, Cancer Cell, vol.8, issue.2, pp.143-153, 2005.
DOI : 10.1016/j.ccr.2005.06.017

P. Pollard, J. Briere, N. Alam, J. Barwell, E. Barclay et al., Accumulation of Krebs cycle intermediates and over-expression of HIF1?? in tumours which result from germline FH and SDH mutations, Human Molecular Genetics, vol.14, issue.15, pp.2231-2239, 2005.
DOI : 10.1042/bj20031647

. Perrier-trudova, A New Hereditary Model of HLRCC Syndrome-Associated Renal Cell Carcinoma 12

P. Pollard, B. Spencer-dene, D. Shukla, K. Howarth, E. Nye et al., Targeted Inactivation of Fh1 Causes Proliferative Renal Cyst Development and Activation of the Hypoxia Pathway, Cancer Cell, vol.11, issue.4, pp.311-319, 2007.
DOI : 10.1016/j.ccr.2007.02.005

S. Sudarshan, C. Sourbier, H. Kong, K. Block, V. Romero et al., Fumarate Hydratase Deficiency in Renal Cancer Induces Glycolytic Addiction and Hypoxia-Inducible Transcription Factor 1?? Stabilization by Glucose-Dependent Generation of Reactive Oxygen Species, Molecular and Cellular Biology, vol.29, issue.15, pp.4080-4090, 2009.
DOI : 10.1128/MCB.00483-09

A. Ooi, J. Wong, D. Petillo, D. Roossien, V. Perrier-trudova et al., An Antioxidant Response Phenotype Shared between Hereditary and Sporadic Type 2 Papillary Renal Cell Carcinoma, Cancer Cell, vol.20, issue.4, pp.511-523, 2011.
DOI : 10.1016/j.ccr.2011.08.024

J. Adam, E. Hatipoglu, O. Flaherty, L. Ternette, N. Sahgal et al., Renal Cyst Formation in Fh1-Deficient Mice Is Independent of the Hif/Phd Pathway: Roles for Fumarate in KEAP1 Succination and Nrf2 Signaling, Cancer Cell, vol.20, issue.4, pp.524-537, 2011.
DOI : 10.1016/j.ccr.2011.09.006

Y. Yang, V. Valera, H. Padilla-nash, C. Sourbier, C. Vocke et al., Fumarate hydratase (FH ? /FH ? ) hereditary leiomyomatosis renal cell carcinoma: in vitro and in vivo model of an aberrant energy metabolic pathway in human cancer, Cancer Genet Cytogenet, vol.262, issue.196, pp.45-55, 2010.

Y. Yang, V. Valera, C. Sourbier, C. Vocke, M. Wei et al., A novel fumarate hydratase-deficient HLRCC kidney cancer cell line, UOK268: a model of the Warburg effect in cancer, Cancer Genetics, vol.205, issue.7-8, pp.377-390, 2012.
DOI : 10.1016/j.cancergen.2012.05.001

O. Warburg, On the Origin of Cancer Cells, Science, vol.123, issue.3191, pp.309-314, 1956.
DOI : 10.1126/science.123.3191.309

R. Kane, P. Bross, A. Farrell, and R. Pazdur, Velcade(R): U.S. FDA Approval for the Treatment of Multiple Myeloma Progressing on Prior Therapy, The Oncologist, vol.8, issue.6, pp.508-513, 2003.
DOI : 10.1634/theoncologist.8-6-508

R. Kane, R. Dagher, A. Farrell, C. Ko, R. Sridhara et al., Bortezomib for the Treatment of Mantle Cell Lymphoma, Clinical Cancer Research, vol.13, issue.18, pp.5291-5294, 2007.
DOI : 10.1158/1078-0432.CCR-07-0871

, 21

X. Yang, M. Tan, H. Kim, J. Ditlev, M. Betten et al., A Molecular Classification of Papillary Renal Cell Carcinoma, Cancer Research, vol.65, issue.13, pp.5628-5637, 2005.
DOI : 10.1158/0008-5472.CAN-05-0533

K. Livak and T. Schmittgen, Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2???????CT Method, Methods, vol.25, issue.4, pp.402-408, 2001.
DOI : 10.1006/meth.2001.1262

M. Wong, C. Tan, S. Lee, Y. Yong, A. Ooi et al., Potential genetic anticipation in hereditary leiomyomatosis-renal cell cancer (HLRCC), Familial Cancer, vol.10, issue.2, pp.281-289, 2014.
DOI : 10.1111/j.1365-2230.2004.01675.x

B. Gardie, A. Remenieras, D. Kattygnarath, J. Bombled, S. Lefèvre et al., Novel FH mutations in families with hereditary leiomyomatosis and renal cell cancer (HLRCC) and patients with isolated type 2 papillary renal cell carcinoma, Bressac-de Paillerets B, Richard S and on behalf of the French National Cancer Institute, pp.226-234, 2011.
DOI : 10.1136/jmg.2010.085068

URL : https://hal.archives-ouvertes.fr/hal-00614499

M. Wu, A. Neilson, A. Swift, R. Moran, J. Tamagnine et al., Multiparameter metabolic analysis reveals a close link between attenuated mitochondrial bioenergetic function and enhanced glycolysis dependency in human tumor cells, American Journal of Physiology-Cell Physiology, vol.292, issue.1, pp.125-136, 2007.
DOI : 10.1172/JCI13505

D. Ferrick, A. Neilson, and C. Beeson, Advances in measuring cellular bioenergetics using extracellular flux, Drug Discovery Today, vol.13, issue.5-6, pp.268-274, 2008.
DOI : 10.1016/j.drudis.2007.12.008

G. Dennis, B. Sherman, D. Hosack, Y. J. Gao, W. Lane et al., DAVID: Database for Annotation, Vizualization, and Integrated Discovery, Genome Biology, vol.4, issue.5, p.3, 2003.
DOI : 10.1186/gb-2003-4-5-p3

D. Hanahan and R. Weinberg, Hallmarks of Cancer: The Next Generation, Cell, vol.144, issue.5, pp.646-674, 2011.
DOI : 10.1016/j.cell.2011.02.013

A. Shakya, R. Cooksey, J. Cox, V. Wang, D. Mcclain et al., Oct1 loss of function induces a coordinate metabolic shift that opposes tumorigenicity, Nature Cell Biology, vol.19, issue.3, pp.320-327, 2009.
DOI : 10.1152/ajpgi.00152.2006

B. Escudier, T. Eisen, W. Stadler, C. Szczylik, S. Oudard et al., Sorafenib in Advanced Clear-Cell Renal-Cell Carcinoma, New England Journal of Medicine, vol.356, issue.2, pp.125-134, 2007.
DOI : 10.1056/NEJMoa060655

R. Motzer, T. Hutson, P. Tomczak, M. Michaelson, R. Bukowski et al., Sunitinib versus Interferon Alfa in Metastatic Renal-Cell Carcinoma, New England Journal of Medicine, vol.356, issue.2, pp.115-124, 2007.
DOI : 10.1056/NEJMoa065044

G. Hudes, M. Carducci, P. Tomczak, J. Dutcher, R. Figlin et al., Temsirolimus, Interferon Alfa, or Both for Advanced Renal-Cell Carcinoma, New England Journal of Medicine, vol.356, issue.22, pp.2271-2281, 2007.
DOI : 10.1056/NEJMoa066838

B. Escudier, A. Pluzanska, P. Koralewski, A. Ravaud, S. Bracarda et al., Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial, The Lancet, vol.370, issue.9605, pp.2103-2111, 2007.
DOI : 10.1016/S0140-6736(07)61904-7

B. Escudier, T. Eisen, C. Porta, J. Patard, V. Khoo et al., Renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up, Ann Oncol, vol.23, pp.65-71, 2012.

, ANTICANCER RESEARCH, vol.35, pp.6639-6654, 2015.

C. Parker, R. Waters, C. Leighton, J. Hancock, R. Sutton et al., Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukaemia (ALL R3): an open-label randomised trial, The Lancet, vol.376, issue.9757, 2009.
DOI : 10.1016/S0140-6736(10)62002-8

C. Lee, V. Gebski, A. Coates, A. Veillard, V. Harvey et al., Simes RJ and Australia and New Zealand Breast Cancer Trials Group (ANZBCTG)

, Trade-offs in quality of life and survival with chemotherapy for advanced breast cancer: mature results of a randomized trial comparing single-agent mitoxantrone with combination cyclophosphamide, methotrexate, 5-fluorouracil and prednisone, Springerplus, vol.2, p.391, 2013.

P. Kantoff, S. Halabi, M. Conaway, J. Picus, J. Kirshner et al., Hydrocortisone With or Without Mitoxantrone in Men With Hormone-Refractory Prostate Cancer: Results of the Cancer and Leukemia Group B 9182 Study, Journal of Clinical Oncology, vol.17, issue.8, pp.2506-2513, 1999.
DOI : 10.1200/JCO.1999.17.8.2506

E. Millefiorini, C. Gasperini, C. Pozzilli, D. Andrea, F. Bastianello et al., Randomized placebo-controlled trial of mitoxantrone in relapsing-remitting multiple sclerosis: 24-month clinical and MRI outcome, Journal of Neurology, vol.244, issue.3, pp.153-159, 1997.
DOI : 10.1007/s004150050066

D. Goodin, B. Arnason, P. Coyle, E. Frohman, and D. Paty, The use of mitoxantrone (Novantrone) for the treatment of multiple sclerosis: Report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology, Neurology, vol.61, issue.10, pp.1332-1338, 2003.
DOI : 10.1212/01.WNL.0000095425.84407.39

F. Group, Benefit of a high-dose epirubicin regimen in adjuvant chemotherapy for node-positive breast cancer patients with poor prognostic factors: 5-year follow-up results of French Adjuvant Study Group 05 randomized trial, J Clin Oncol, vol.19, pp.602-611, 2001.

M. Brien, T. Ciuleanu, H. Tsekov, Y. Shparyk, B. Cucevia et al., Phase III Trial Comparing Supportive Care Alone With Supportive Care With Oral Topotecan in Patients With Relapsed Small-Cell Lung Cancer, Journal of Clinical Oncology, vol.24, issue.34, pp.5441-5447, 2006.
DOI : 10.1200/JCO.2006.06.5821

E. Swisher, D. Mutch, J. Rader, A. Elbendary, and T. Herzog, Topotecan in Platinum- and Paclitaxel-Resistant Ovarian Cancer, Gynecologic Oncology, vol.66, issue.3, pp.480-486, 1997.
DOI : 10.1006/gyno.1997.4787

J. Sehouli, D. Stengel, P. Harter, C. Kurzeder, A. Belau et al., Topotecan Weekly Versus Conventional 5-Day Schedule in Patients With Platinum-Resistant Ovarian Cancer: A Randomized Multicenter Phase II Trial of the North-Eastern German Society of Gynecological Oncology Ovarian Cancer Study Group, Phase III trial of four cisplatin-containing doublet combinations in stage IVB, recurrent, or persistent cervical carcinoma: a Gynecologic Oncology Group study, pp.242-248, 2009.
DOI : 10.1200/JCO.2009.27.8911

M. Boland, K. Fitzgerald, O. Neill, and L. , Topoisomerase II Is Required for Mitoxantrone to Signal Nuclear Factor ??B Activation in HL60 Cells, Journal of Biological Chemistry, vol.47, issue.3, pp.25231-25238, 2000.
DOI : 10.1074/jbc.274.2.987

URL : http://www.jbc.org/content/275/33/25231.full.pdf

A. Ferrer, S. Marce, B. Bellosillo, N. Villamor, F. Bosch et al., Activation of mitochondrial apoptotic pathway in mantle cell lymphoma: high sensitivity to mitoxantrone in cases with functional DNA-damage response genes, Oncogene, vol.23, issue.55, pp.8941-8949, 2004.
DOI : 10.1016/S0014-4827(02)00032-0

S. Karl, Y. Pritschow, M. Volcic, S. Häcker, B. Baumann et al., Identification of a novel pro-apopotic function of NF-??B in the DNA damage response, Journal of Cellular and Molecular Medicine, vol.57, issue.10, pp.4239-4256, 2009.
DOI : 10.1111/j.1582-4934.2009.00888.x

M. Lioni, K. Noma, A. Snyder, A. Klein-szanto, J. Diehl et al., Bortezomib induces apoptosis in esophageal squamous cell carcinoma cells through activation of the p38 mitogen-activated protein kinase pathway, Molecular Cancer Therapeutics, vol.7, issue.9, pp.2866-2875, 2008.
DOI : 10.1158/1535-7163.MCT-08-0391

URL : http://mct.aacrjournals.org/content/molcanther/7/9/2866.full.pdf

N. Mitsiades, C. Mitsiades, V. Poulaki, D. Chauhan, G. Fanourakis et al., Molecular sequelae of proteasome inhibition in human multiple myeloma cells, Proceedings of the National Academy of Sciences, vol.504, issue.1-2, pp.14374-14379, 2002.
DOI : 10.1016/S0014-5793(01)02770-3

URL : http://www.pnas.org/content/99/22/14374.full.pdf

D. Chen, M. Frezza, S. Schmitt, J. Kanwar, and Q. Dou, Bortezomib as the First Proteasome Inhibitor Anticancer Drug: Current Status and Future Perspectives, Current Cancer Drug Targets, vol.11, issue.3, pp.239-253, 2011.
DOI : 10.2174/156800911794519752

URL : http://europepmc.org/articles/pmc3306611?pdf=render

M. Ma, H. Yang, K. Parker, S. Manyak, J. Friedman et al., The proteasome inhibitor PS-341 markedly enhances sensitivity of multiple myeloma tumor cells to chemotherapeutic agents, Clin Cancer Res, vol.9, pp.1136-1144, 2003.

S. Strauss, K. Higginbottom, S. Jüliger, L. Maharaj, P. Allen et al., The Proteasome Inhibitor Bortezomib Acts Independently of p53 and Induces Cell Death via Apoptosis and Mitotic Catastrophe in B-Cell Lymphoma Cell Lines, Cancer Research, vol.67, issue.6, pp.2783-2790, 2007.
DOI : 10.1158/0008-5472.CAN-06-3254

URL : http://cancerres.aacrjournals.org/content/canres/67/6/2783.full.pdf

C. Sourbier, V. Valera-romero, A. Giubellino, Y. Yang, S. Sudarshan et al., Increasing reactive oxygen species as a therapeutic approach to treat hereditary leiomyomatosis and renal cell carcinoma, Cell Cycle, vol.26, issue.20, pp.4183-4189, 2010.
DOI : 10.1002/jcla.1860090615

URL : http://www.tandfonline.com/doi/pdf/10.4161/cc.9.20.13458?needAccess=true