N. C. Hodgson, Merkel cell carcinoma: Changing incidence trends, J. Surg. Oncol, vol.89, pp.1-4, 2005.

K. G. Paulson, S. Y. Park, N. A. Vandeven, K. Lachance, H. Thomas et al., Merkel Cell Carcinoma: Current United States Incidence and Projected Increases based on Changing Demographics, J. Am. Acad. Derm, 2017.

T. L. Fitzgerald, S. Dennis, S. D. Kachare, N. A. Vohra, J. H. Wong et al., Dramatic Increase in the Incidence and Mortality from Merkel Cell Carcinoma in the United States, Am. Surg, vol.81, pp.802-806, 2015.

R. W. Miller and C. S. Rabkin, Merkel cell carcinoma and melanoma: Etiological similarities and differences, Cancer Epidemiol. Biomark. Prev, vol.8, pp.153-158, 1999.

J. F. Buell, J. Trofe, M. J. Hanaway, T. M. Beebe, T. G. Gross et al., Immunosuppression and merkel cell cancer, Transplant. Proc, vol.34, pp.1780-1781, 2002.

H. Feng, M. Shuda, Y. Chang, and P. S. Moore, Clonal Integration of a Polyomavirus in Human Merkel Cell Carcinoma, Science, 2008.

R. Tothill, V. Estall, and D. Rischin, Merkel cell carcinoma: Emerging biology, current approaches, and future directions, Am. Soc. Clin. Oncol. Educ. Book. Am. Soc. Clin. Oncol. Meet, 2015.

X. Sastre-garau, M. Peter, M. F. Avril, H. Laude, J. Couturier et al., Merkel cell carcinoma of the skin: Pathological and molecular evidence for a causative role of MCV in oncogenesis, J. Pathol, vol.218, pp.48-56, 2009.

D. Schrama, E. M. Sarosi, C. Adam, C. Ritter, U. Kaemmerer et al., Characterization of six Merkel cell polyomavirus-positive Merkel cell carcinoma cell lines: Integration pattern suggest that large T antigen truncating events occur before or during integration, Int. J. Cancer J. Int. Du Cancer, vol.145, pp.1020-1032, 2019.

M. Shuda, H. J. Kwun, H. Feng, Y. Chang, and P. S. Moore, Human Merkel cell polyomavirus small T antigen is an oncoprotein targeting the 4E-BP1 translation regulator, J. Clin. Investig, vol.121, pp.3623-3634, 2011.

R. Houben, M. Shuda, R. Weinkam, D. Schrama, H. Feng et al., Merkel cell polyomavirus-infected Merkel cell carcinoma cells require expression of viral T antigens, J. Virol, vol.84, pp.7064-7072, 2010.

B. D. Lemos, B. E. Storer, J. G. Iyer, J. L. Phillips, C. K. Bichakjian et al., Pathologic nodal evaluation improves prognostic accuracy in Merkel cell carcinoma: Analysis of 5823 cases as the basis of the first consensus staging system, J. Am. Acad. Derm, vol.63, pp.751-761, 2010.

M. Poulsen, Merkel cell carcinoma of skin: Diagnosis and management strategies, Drugs Aging, vol.22, pp.219-229, 2005.

T. Y. Eng, M. G. Boersma, C. D. Fuller, V. Goytia, W. E. Jones et al., A comprehensive review of the treatment of Merkel cell carcinoma, Am. J. Clin. Oncol, vol.30, pp.624-636, 2007.

P. T. Nghiem, S. Bhatia, E. J. Lipson, R. R. Kudchadkar, N. J. Miller et al., PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma, N. Engl. J. Med, vol.374, pp.2542-2552, 2016.

H. L. Kaufman, J. Russell, O. Hamid, S. Bhatia, P. Terheyden et al., Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: A multicentre, single-group, open-label, phase 2 trial, Lancet Oncol, 2016.

E. S. Kim, Avelumab: First Global Approval, vol.77, pp.929-937, 2017.

P. Nghiem, S. Bhatia, E. J. Lipson, W. H. Sharfman, R. R. Kudchadkar et al., Durable Tumor Regression and Overall Survival in Patients With Advanced Merkel Cell Carcinoma Receiving Pembrolizumab as First-Line Therapy, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.37, pp.693-702, 2019.

S. P. D'angelo, J. Russell, C. Lebbé, B. Chmielowski, T. Gambichler et al., Efficacy and Safety of First-line Avelumab Treatment in Patients With Stage IV Metastatic Merkel Cell Carcinoma: A Preplanned Interim Analysis of a Clinical Trial, JAMA Oncol, vol.12, pp.919-932, 2018.

H. L. Kaufman, J. S. Russell, O. Hamid, S. Bhatia, P. Terheyden et al., Updated efficacy of avelumab in patients with previously treated metastatic Merkel cell carcinoma after ?1 year of follow-up: JAVELIN Merkel 200, a phase 2 clinical trial, J. Immunother. Cancer, vol.6, issue.7, 2018.

J. A. Marin-acevedo, B. Dholaria, A. E. Soyano, K. L. Knutson, S. Chumsri et al., Next generation of immune checkpoint therapy in cancer: New developments and challenges, J. Hematol. Oncol, vol.11, p.39, 2018.

T. Efferth, From ancient herb to modern drug: Artemisia annua and artemisinin for cancer therapy, Semin Cancer Biol, vol.46, pp.65-83, 2017.

J. N. Burrows, K. Chibale, and T. N. Wells, The state of the art in anti-malarial drug discovery and development, Curr. Top. Med. Chem, vol.11, pp.1226-1254, 2011.

J. Wang, C. J. Zhang, W. N. Chia, C. C. Loh, Z. Li et al., Haem-activated promiscuous targeting of artemisinin in Plasmodium falciparum, Nat. Commun, vol.6, 2015.

L. Yu, J. F. Chen, X. Shuai, Y. Xu, Y. Ding et al., Artesunate protects pancreatic beta cells against cytokine-induced damage via SIRT1 inhibiting NF-kappaB activation, J. Endocrinol. Investig, vol.39, pp.83-91, 2016.

L. E. Heller and P. D. Roepe, Artemisinin-Based Antimalarial Drug Therapy: Molecular Pharmacology and Evolving Resistance, Trop. Med. Infect

T. Efferth, A. Sauerbrey, A. Olbrich, E. Gebhart, P. Rauch et al., Molecular modes of action of artesunate in tumor cell lines, Mol. Pharm, vol.64, pp.382-394, 2003.

T. Efferth, H. Dunstan, A. Sauerbrey, H. Miyachi, and C. R. Chitambar, The anti-malarial artesunate is also active against cancer, Int. J. Oncol, vol.18, pp.767-773, 2001.

F. Jiang, J. Zhou, D. Zhang, M. Liu, and Y. Chen, Artesunate induces apoptosis and autophagy in HCT116 colon cancer cells, and autophagy inhibition enhances the artesunate-induced apoptosis, Int. J. Mol. Med, vol.42, pp.1295-1304, 2018.

T. K. Våtsveen, M. R. Myhre, C. B. Steen, S. Wälchli, O. C. Lingjaerde et al., Artesunate shows potent anti-tumor activity in B-cell lymphoma, J. Hematol. Oncol, vol.11, issue.23, 2018.

C. Zhou, W. Pan, X. P. Wang, and T. S. Chen, Artesunate induces apoptosis via a Bak-mediated caspase-independent intrinsic pathway in human lung adenocarcinoma cells, J. Cell. Physiol, vol.227, pp.3778-3786, 2012.

N. Wang, G. Zeng, J. Yin, and Z. Bian, Artesunate activates the ATF4-CHOP-CHAC1 pathway and affects ferroptosis in Burkitt's Lymphoma, Biochem. Biophys. Res. Commun, vol.519, pp.533-539, 2019.

N. Eling, L. Reuter, J. Hazin, A. Hamacher-brady, and N. R. Brady, Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells, vol.2, pp.517-532, 2015.

J. Roh, E. H. Kim, H. Jang, and D. Shin, Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis, Redox Biol, vol.11, pp.254-262, 2017.

M. Youns, J. D. Hoheisel, and T. Efferth, Traditional Chinese medicines (TCMs) for molecular targeted therapies of tumours, Curr. Drug Discov. Technol, vol.7, pp.37-45, 2010.

T. Efferth, M. R. Romero, D. G. Wolf, T. Stamminger, J. J. Marin et al., The antiviral activities of artemisinin and artesunate, Clin. Infect. Dis, vol.47, pp.804-811, 2008.

R. Houben, S. Angermeyer, S. Haferkamp, A. Aue, M. Goebeler et al., Characterization of functional domains in the Merkel cell polyoma virus Large T antigen, Int. J. Cancer, vol.136, pp.290-300, 2015.

S. Hesbacher, L. Pfitzer, K. Wiedorfer, S. Angermeyer, A. Borst et al., RB1 is the crucial target of the Merkel cell polyomavirus Large T antigen in Merkel cell carcinoma cells, Oncotarget, vol.7, pp.32956-32968, 2016.

F. Valeriote and L. Van-putten, Proliferation-dependent cytotoxicity of anticancer agents: A review, Cancer Res, vol.35, pp.2619-2630, 1975.

M. M. Compton, A biochemical hallmark of apoptosis: Internucleosomal degradation of the genome, Cancer Metastasis Rev, vol.11, pp.919-933, 1992.

T. Efferth, M. Giaisi, A. Merling, P. H. Krammer, and M. Li-weber, Artesunate induces ROS-mediated apoptosis in doxorubicin-resistant T leukemia cells, PLoS ONE, vol.2, 2007.

L. Galluzzi, I. Vitale, J. M. Abrams, E. S. Alnemri, E. H. Baehrecke et al., Molecular definitions of cell death subroutines: Recommendations of the Nomenclature Committee on Cell Death, Cell Death Differ, vol.19, pp.107-120, 2012.

C. D. Bortner and J. A. Cidlowski, Uncoupling cell shrinkage from apoptosis reveals that Na+ influx is required for volume loss during programmed cell death, J. Biol. Chem, vol.278, pp.39176-39184, 2003.

D. Tang, R. Kang, T. V. Berghe, P. Vandenabeele, and G. Kroemer, The molecular machinery of regulated cell death, Cell Res, vol.29, pp.347-364, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02125072

N. Yan and J. Zhang, The Emerging Roles of Ferroptosis in Vascular Cognitive Impairment, Front. Neurosci, vol.13, 2019.

B. R. Stockwell, J. P. Angeli, H. Bayir, A. I. Bush, M. Conrad et al., Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, vol.171, pp.273-285, 2017.

S. J. Dixon, K. M. Lemberg, M. R. Lamprecht, R. Skouta, E. M. Zaitsev et al., Ferroptosis: An iron-dependent form of nonapoptotic cell death, Cell, vol.149, pp.1060-1072, 2012.

J. J. Shacka, B. J. Klocke, K. A. Roth, and . Autophagy, bafilomycin and cell death: The "a-B-cs" of plecomacrolide-induced neuroprotection, Autophagy, vol.2, pp.228-230, 2006.

Z. Xie, Y. Xie, Y. Xu, H. Zhou, W. Xu et al., Bafilomycin A1 inhibits autophagy and induces apoptosis in MG63 osteosarcoma cells, Mol. Med. Rep, vol.10, pp.1103-1107, 2014.

M. Gao, P. Monian, Q. Pan, W. Zhang, J. Xiang et al., Ferroptosis is an autophagic cell death process, Cell Res, vol.26, pp.1021-1032, 2016.

B. Zhou, J. Liu, R. Kang, D. J. Klionsky, G. Kroemer et al., Ferroptosis is a type of autophagy-dependent cell death, Semin. Cancer Biol, 2019.

M. Sato, R. Kusumi, S. Hamashima, S. Kobayashi, S. Sasaki et al., The ferroptosis inducer erastin irreversibly inhibits system x(c)-and synergizes with cisplatin to increase cisplatin's cytotoxicity in cancer cells, Sci. Rep, vol.8, p.968, 2018.

S. J. Dixon, D. N. Patel, M. Welsch, R. Skouta, E. D. Lee et al., Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis, vol.3, 2014.

M. Takahashi, T. Nagai, N. Okamura, H. Takahashi, and A. Okano, Promoting effect of beta-mercaptoethanol on in vitro development under oxidative stress and cystine uptake of bovine embryos, Biol. Reprod, vol.66, pp.562-567, 2002.

S. Doll, B. Proneth, Y. Y. Tyurina, E. Panzilius, S. Kobayashi et al., ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition, Nat. Chem. Biol, vol.13, pp.91-98, 2017.

H. Yuan, X. Li, X. Zhang, R. Kang, and D. Tang, Identification of ACSL4 as a biomarker and contributor of ferroptosis, Biochem. Biophys. Res. Commun, vol.478, pp.1338-1343, 2016.

C. Adam, A. Baeurle, J. L. Brodsky, P. Wipf, D. Schrama et al., The HSP70 modulator MAL3-101 inhibits Merkel cell carcinoma, PLoS ONE, vol.9, 2014.

M. B. Serafin, A. Bottega, T. F. Da-rosa, C. S. Machado, V. S. Foletto et al., Drug Repositioning in Oncology, Am. J. Ther, 2019.

M. K. Tiwari and S. Chaudhary, Artemisinin-derived antimalarial endoperoxides from bench-side to bed-side: Chronological advancements and future challenges, Med. Res. Rev, 2020.

A. Adeel and A. , Time to switch from quinine, Sudan. J. Paediatr, vol.12, pp.6-7, 2012.

J. Nass and T. Efferth, The activity of Artemisia spp. and their constituents against Trypanosomiasis, Phytomed. Int. J. Phytother. Phytopharm, vol.47, pp.184-191, 2018.

K. Chen, L. M. Shou, F. Lin, W. M. Duan, M. Y. Wu et al., Artesunate induces G2/M cell cycle arrest through autophagy induction in breast cancer cells, Anticancer Drugs, vol.25, pp.652-662, 2014.

Y. Li, N. N. Shan, and X. H. Sui, Research Progress on Artemisinin and Its Derivatives against Hematological Malignancies, Chin. J. Integr. Med, vol.12, pp.919-934, 2020.

B. N. Sharma, M. Marschall, S. Henriksen, and C. H. Rinaldo, Antiviral effects of artesunate on polyomavirus BK replication in primary human kidney cells, Antimicrob. Agents Chemother, vol.58, pp.279-289, 2014.

B. N. Sharma, M. Marschall, and C. H. Rinaldo, Antiviral effects of artesunate on JC polyomavirus replication in COS-7 cells, Antimicrob. Agents Chemother, vol.58, pp.6724-6734, 2014.

G. L. Disbrow, A. C. Baege, K. A. Kierpiec, H. Yuan, J. A. Centeno et al., Dihydroartemisinin is cytotoxic to papillomavirus-expressing epithelial cells in vitro and in vivo, Cancer Res, vol.65, pp.10854-10861, 2005.

B. Proneth and M. Conrad, Ferroptosis and necroinflammation, a yet poorly explored link, Cell Death Differ, vol.26, pp.14-24, 2019.

Y. Mou, J. Wang, J. Wu, D. He, C. Zhang et al., Ferroptosis, a new form of cell death: Opportunities and challenges in cancer, J. Hematol. Oncol, vol.12, 2019.

S. Krishna, S. Ganapathi, I. C. Ster, M. E. Saeed, M. Cowan et al., A Randomised, Double Blind, Placebo-Controlled Pilot Study of Oral Artesunate Therapy for Colorectal Cancer, vol.2, pp.82-90, 2014.

J. F. Deeken, H. Wang, M. Hartley, A. K. Cheema, B. Smaglo et al., A phase I study of intravenous artesunate in patients with advanced solid tumor malignancies, Cancer Chemother. Pharmacol, vol.81, pp.587-596, 2018.

C. Von-hagens, I. Walter-sack, M. Goeckenjan, B. Storch-hagenlocher, S. Sertel et al., Long-term add-on therapy (compassionate use) with oral artesunate in patients with metastatic breast cancer after participating in a phase I study (ARTIC M33/2), Phytomed. Int. J. Phytother. Phytopharm, vol.54, pp.140-148, 2019.

M. König, C. Von-hagens, S. Hoth, I. Baumann, I. Walter-sack et al., Investigation of ototoxicity of artesunate as add-on therapy in patients with metastatic or locally advanced breast cancer: New audiological results from a prospective, open, uncontrolled, monocentric phase I study, Cancer Chemother. Pharmacol, vol.77, pp.413-427, 2016.

S. T. Rosen, V. E. Gould, H. R. Salwen, C. V. Herst, M. M. Le-beau et al., Establishment and characterization of a neuroendocrine skin carcinoma cell line, Lab. Investig, vol.56, pp.302-312, 1987.

M. Van-gele, J. H. Leonard, R. N. Van, L. H. Van, B. S. Van et al., Combined karyotyping, CGH and M-FISH analysis allows detailed characterization of unidentified chromosomal rearrangements in Merkel cell carcinoma, Int. J. Cancer, vol.101, pp.137-145, 2002.

A. Guastafierro, H. Feng, M. Thant, J. M. Kirkwood, Y. Chang et al., Characterization of an early passage Merkel cell polyomavirus-positive Merkel cell carcinoma cell line, MS-1, and its growth in NOD scid gamma mice, J. Virol. Methods, vol.187, pp.6-14, 2013.