M. Adams, Y. Li, H. Khot, C. De-kock, P. Smith et al., The synthesis and antiparasitic activity of aryl- and ferrocenyl-derived thiosemicarbazone ruthenium(ii)???arene complexes, Dalton Transactions, vol.48, issue.6, pp.4677-4685, 2013.
DOI : 10.1039/c3dt32740j

R. Aird, J. Cummings, A. Ritchie, M. Muir, R. Morris et al., In vitro and in vivo activity and cross resistance profiles of novel ruthenium (II) organometallic arene complexes in human ovarian cancer, British Journal of Cancer, vol.60, issue.10, pp.1652-1657, 2002.
DOI : 10.1038/sj.bjc.6600290

E. Ashley, M. Dhorda, R. Fairhurst, C. Amaratunga, P. Lim et al., Malaria, NJ, Tracking Resistance to Artemisinin Collaboration 2014. Spread of artemisinin resistance in Plasmodium falciparum malaria, pp.411-423
DOI : 10.1056/NEJMoa1314981

URL : https://hal.archives-ouvertes.fr/inserm-00854378

M. Barbosa, R. Corrêa, K. De-oliveira, C. Rodrigues, E. J. Nascimento et al., Antiparasitic activities of novel ruthenium/lapachol complexes, Journal of Inorganic Biochemistry, vol.136, pp.33-39, 2014.
DOI : 10.1016/j.jinorgbio.2014.03.009

P. Beagley, M. Blackie, K. Chibale, C. Clarkson, R. Meijboom et al., Synthesis and antiplasmodial activity in vitro of new ferrocene???chloroquine analogues, Dalton Trans., vol.276, issue.15, pp.3046-3051, 2003.
DOI : 10.1039/B303335J

C. Bézivin, S. Tomasi, D. Fl, and J. Boustie, Cytotoxic activity of some lichen extracts on murine and human cancer cell lines, Phytomedicine, vol.10, issue.6-7, pp.499-503, 2003.
DOI : 10.1078/094471103322331458

C. Biot, Ferroquine: A New Weapon in the Fight Against Malaria, Current Medicinal Chemistry -Anti-Infective Agents, vol.3, issue.2, pp.135-147, 2004.
DOI : 10.2174/1568012043354008

C. Biot, W. Castro, C. Botté, and M. Navarro, The therapeutic potential of metal-based antimalarial agents: Implications for the mechanism of action, Dalton Transactions, vol.22, issue.Suppl 2, pp.6335-6349
DOI : 10.1039/c2dt12247b

URL : https://hal.archives-ouvertes.fr/hal-00796400

C. Biot, F. Dubar, J. Khalife, and C. Slomianny, Opening up the advantages of the ruthenocenic bioprobes of ferroquine: distribution and localization in Plasmodium falciparum-infected erythrocytes, Metallomics, vol.2, issue.6, pp.780-783, 2012.
DOI : 10.1039/c2mt20063e

C. Biot, B. Pradines, M. Sergeant, J. Gut, P. Rosenthal et al., Design, synthesis, and antimalarial activity of structural chimeras of thiosemicarbazone and ferroquine analogues, Bioorganic & Medicinal Chemistry Letters, vol.17, issue.23, pp.6434-6438, 2007.
DOI : 10.1016/j.bmcl.2007.10.003

E. Borenfreund, H. Babich, and N. Martin-alguacil, Comparisons of two in vitro cytotoxicity assays???The neutral red (NR) and tetrazolium MTT tests, Toxicology in Vitro, vol.2, issue.1, pp.1-6, 1987.
DOI : 10.1016/0887-2333(88)90030-6

V. Brabec and O. Nováková, DNA binding mode of ruthenium complexes and relationship to tumor cell toxicity, Drug Resistance Updates, vol.9, issue.3, pp.111-122, 2006.
DOI : 10.1016/j.drup.2006.05.002

A. Casini, C. Gabbiani, F. Sorrentino, M. Rigobello, A. Bindoli et al., Emerging Protein Targets for Anticancer Metallodrugs: Inhibition of Thioredoxin Reductase and Cathepsin B by Antitumor Ruthenium(II)???Arene Compounds, Journal of Medicinal Chemistry, vol.51, issue.21, pp.6773-6781, 2008.
DOI : 10.1021/jm8006678

J. Cázares-marinero, S. Top, A. Vessières, and G. Jaouen, Synthesis and antiproliferative activity of hydroxyferrocifen hybrids against triple-negative breast cancer cells, Dalton Trans., vol.41, issue.2, pp.817-830, 2014.
DOI : 10.1039/C3DT52070F

P. Chellan, K. Land, A. Shokar, A. Au, S. An et al., Di- and Trinuclear Ruthenium-, Rhodium-, and Iridium-Functionalized Pyridyl Aromatic Ethers: A New Class of Antiparasitic Agents, Organometallics, vol.32, issue.17, pp.4793-4804, 2013.
DOI : 10.1021/om400493k

P. Chellan, K. Land, A. Shokar, A. Au, S. An et al., ) pyridyl ester complexes as in vitro antiparasitic and antitumor agents, Dalton Trans., vol.338, issue.2, pp.513-526, 2014.
DOI : 10.1039/C3DT52090K

M. Chugh, V. Sundararaman, S. Kumar, V. Reddy, W. Siddiqui et al., Protein complex directs hemoglobin-to-hemozoin formation in Plasmodium falciparum, Proceedings of the National Academy of Sciences, vol.110, issue.14, pp.5392-5397, 2013.
DOI : 10.1073/pnas.1218412110

A. Citta, A. Folda, A. Bindoli, P. Pigeon, S. Top et al., Evidence for Targeting Thioredoxin Reductases with Ferrocenyl Quinone Methides. A Possible Molecular Basis for the Antiproliferative Effect of Hydroxyferrocifens on Cancer Cells, Journal of Medicinal Chemistry, vol.57, issue.21, pp.8849-8859, 2014.
DOI : 10.1021/jm5013165

URL : https://hal.archives-ouvertes.fr/hal-01230376

M. Coogan, P. Dyson, and M. Bochmann, Introduction to the Organometallics in Biology and Medicine Issue, Organometallics, vol.31, issue.16, pp.5671-5672, 2012.
DOI : 10.1021/om300737y

M. De-madureira, A. Martins, M. Gomes, J. Paiva, A. Cunha et al., Antimalarial activity of medicinal plants used in traditional medicine in S. Tom?? and Pr????ncipe islands, Journal of Ethnopharmacology, vol.81, issue.1, pp.23-29, 2002.
DOI : 10.1016/S0378-8741(02)00005-3

A. De-oliveira, E. Da-silva, D. Rocha, E. Hillard, P. Pigeon et al., Molecular mechanism of action of 2-ferroce- nyl-1,1-diphenylbut-1-ene on HL-60 leukemia cells, ChemMed- Chem, vol.11, pp.2580-2586, 2014.

A. De-oliveira, E. Hillard, P. Pigeon, D. Rocha, F. Rodrigues et al., Biological evaluation of twenty-eight ferrocenyl tetrasubstituted olefins: Cancer cell growth inhibition, ROS production and hemolytic activity, European Journal of Medicinal Chemistry, vol.46, issue.9, pp.3778-3787, 2011.
DOI : 10.1016/j.ejmech.2011.05.043

J. Debreczeni, A. Bullock, G. Atilla, D. Williams, H. Bregman et al., Ruthenium Half-Sandwich Complexes Bound to Protein Kinase Pim-1, Angewandte Chemie International Edition, vol.125, issue.10, pp.1580-1585, 2006.
DOI : 10.1002/anie.200503468

F. Denizot and R. Lang, Rapid colorimetric assay for cell growth and survival, Journal of Immunological Methods, vol.89, issue.2, pp.271-277, 1986.
DOI : 10.1016/0022-1759(86)90368-6

F. Dubar, S. Bohic, C. Slomianny, J. Morin, P. Thomas et al., In situ nanochemical imaging of label-free drugs: a case study of antimalarials in Plasmodium falciparum-infected erythrocytes, Chem. Commun., vol.145, issue.6, pp.910-912, 2012.
DOI : 10.1039/C1CC16211J

URL : https://hal.archives-ouvertes.fr/hal-00652186

F. Dubar, T. Egan, B. Pradines, D. Kuter, K. Ncokazi et al., The Antimalarial Ferroquine: Role of the Metal and Intramolecular Hydrogen Bond in Activity and Resistance, ACS Chemical Biology, vol.6, issue.3, pp.275-287, 2011.
DOI : 10.1021/cb100322v

URL : https://hal.archives-ouvertes.fr/hal-00595416

E. Arbi, M. Pigeon, P. Top, S. Rhouma, A. Aifa et al., Evaluation of bactericidal and fungicidal activity of ferrocenyl or phenyl derivatives in the diphenyl butene series, J Organomet Chem, vol.69, pp.1038-1048, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01230388

S. Fricker, Cysteine proteases as targets for metal-based drugs, Metallomics, vol.45, issue.6, pp.366-377, 2010.
DOI : 10.1039/b924677k

D. Gambino and L. Otero, Perspectives on what ruthenium-based compounds could offer in the development of potential antiparasitic drugs, Inorganica Chimica Acta, vol.393, pp.103-114, 2012.
DOI : 10.1016/j.ica.2012.05.028

G. Gasser, I. Ott, and N. Metzler-nolte, Organometallic Anticancer Compounds, Journal of Medicinal Chemistry, vol.54, issue.1, pp.3-25, 2011.
DOI : 10.1021/jm100020w

URL : http://doi.org/10.1021/jm100020w

L. Glans, A. Ehnbom, C. De-kock, A. Martínez, J. Estrada et al., Ruthenium(ii) arene complexes with chelating chloroquine analogue ligands: Synthesis, characterization and in vitro antimalarial activity, Dalton Transactions, vol.65, issue.192, pp.2764-2773, 2012.
DOI : 10.1039/c2dt12083f

M. Gobec, J. Kljun, I. Sosi?, I. Mlinari?-ra??an, M. Ur?i? et al., Structural characterization and biological evaluation of a clioquinol???ruthenium complex with copper-independent antileukaemic activity, Dalton Transactions, vol.72, issue.24, pp.9045-9051, 2014.
DOI : 10.1039/c4dt00463a

M. Görmen, P. Pigeon, S. Top, E. Hillard, M. Huché et al., Synthesis, Cytotoxicity, and COMPARE Analysis of Ferrocene and [3]Ferrocenophane Tetrasubstituted Olefin Derivatives against Human Cancer Cells, ChemMedChem, vol.693, issue.639, pp.2039-2050, 2010.
DOI : 10.1002/cmdc.201000286

M. Görmen, P. Pigeon, S. Top, A. Vessières, M. Plamont et al., Facile synthesis and strong antiproliferative activity of disubstituted diphenylmethylidenyl-[3]ferrocenophanes on breast and prostate cancer cell lines, MedChemComm, vol.48, issue.639, pp.149-151, 2010.
DOI : 10.1039/c0md00026d

P. Graf, S. Durand, A. Alvarez, C. Montalvan, M. Montoya et al., Failure of supervised chloroquine and primaquine regimen for the treatment of Plasmodium vivax in the Peruvian Amazon, Malar Res Treat, vol.2012, p.5, 2012.

D. Hamels, P. Dansette, E. Hillard, S. Top, A. Vessieres et al., Ferrocenyl Quinone Methides as Strong Antiproliferative Agents: Formation by Metabolic and Chemical Oxidation of Ferrocenyl Phenols, Angewandte Chemie International Edition, vol.36, issue.48, pp.9124-9126, 2009.
DOI : 10.1002/anie.200903768

J. Heilmann, E. Hillard, M. Plamont, P. Pigeon, M. Bolte et al., Ferrocenyl compounds possessing protected phenol and thiophenol groups: Synthesis, X-ray structure, and in vitro biological effects against breast cancer, Journal of Organometallic Chemistry, vol.693, issue.8-9, pp.1716-1722, 2008.
DOI : 10.1016/j.jorganchem.2007.12.011

URL : https://hal.archives-ouvertes.fr/hal-01230397

J. Held, S. Jeyaraj, and A. Kreidenweiss, Antimalarial compounds in Phase II clinical development, Expert Opinion on Investigational Drugs, vol.6, issue.2, pp.363-382, 2015.
DOI : 10.1128/AAC.49.9.3631-3639.2005

J. Held, C. Supan, C. Salazar, H. Tinto, L. Bonkian et al., Ferroquine and artesunate in African adults and children with Plasmodium falciparum malaria: a phase 2, multicentre, randomised, double-blind, dose-ranging, non-inferiority study, The Lancet Infectious Diseases, vol.15, issue.12, pp.10-1016
DOI : 10.1016/S1473-3099(15)00079-1

E. Hillard, P. Pigeon, A. Vessières, C. Amatore, and G. Jaouen, The influence of phenolic hydroxy substitution on the electron transfer and anti-cancer properties of compounds based on the 2-fer- rocenyl-1-phenyl-but-1-ene motif, Dalton Trans, vol.43, pp.5073-5081, 2007.

S. Kanzok, R. Schirmer, I. Turbachova, R. Iozef, and K. Becker, The Thioredoxin System of the Malaria Parasite Plasmodium falciparum. GLUTATHIONE REDUCTION REVISITED, Journal of Biological Chemistry, vol.275, issue.51, pp.40180-40186, 2000.
DOI : 10.1074/jbc.M007633200

C. Lambros and J. Vanderberg, Synchronization of Plasmodium falciparum Erythrocytic Stages in Culture, The Journal of Parasitology, vol.65, issue.3, pp.418-420, 1979.
DOI : 10.2307/3280287

H. Lee, O. Buriez, F. Chau, E. Labbé, R. Ganguly et al., Synthesis, Characterization, and Biological Properties of Osmium-Based Tamoxifen Derivatives - Comparison with Their Homologues in the Iron and Ruthenium Series, European Journal of Inorganic Chemistry, vol.76, issue.25, pp.4217-4226, 2015.
DOI : 10.1002/ejic.201500770

H. Lee, O. Buriez, E. Labbé, S. Top, P. Pigeon et al., Oxidative Sequence of a Ruthenocene-Based Anticancer Drug Candidate in a Basic Environment, Organometallics, vol.33, issue.18, pp.4940-4946, 2014.
DOI : 10.1021/om500225k

URL : https://hal.archives-ouvertes.fr/hal-01230374

M. Marques, M. Costa, S. Filho, F. Vieira, J. Nascimento et al., Plasmodium vivax Chloroquine Resistance and Anemia in the Western Brazilian Amazon, Antimicrobial Agents and Chemotherapy, vol.58, issue.1, pp.342-347, 2014.
DOI : 10.1128/AAC.02279-12

A. Martínez, T. Carreon, E. Iniguez, A. Anzellotti, A. Sánchez et al., and Low Toxicity toward Normal Mammalian Cells, Journal of Medicinal Chemistry, vol.55, issue.8, pp.3867-3877, 2012.
DOI : 10.1021/jm300070h

A. Martínez, C. Rajapakse, D. Jalloh, C. Dautriche, and R. Sánchez-delgado, The antimalarial activity of Ru???chloroquine complexes against resistant Plasmodium falciparum is related to lipophilicity, basicity, and heme aggregation inhibition ability near water/n-octanol interfaces, JBIC Journal of Biological Inorganic Chemistry, vol.26, issue.192, pp.863-871, 2009.
DOI : 10.1007/s00775-009-0498-4

D. Minick, J. Frenz, M. Patrick, and D. Brent, A comprehensive method for determining hydrophobicity constants by reversed-phase high-performance liquid chromatography, Journal of Medicinal Chemistry, vol.31, issue.10, pp.1923-1933, 1988.
DOI : 10.1021/jm00118a010

O. Miotto, J. Almagro-garcia, M. Manske, B. Macinnis, S. Campino et al., Multiple populations of artemisinin-resistant Plasmodium falciparum in Cambodia, Nature Genetics, vol.89, issue.6, pp.648-655
DOI : 10.1101/gr.094052.109

A. Nguyen, A. Vessières, E. Hillard, S. Top, P. Pigeon et al., Ferrocifens and Ferrocifenols as New Potential Weapons against Breast Cancer, CHIMIA International Journal for Chemistry, vol.61, issue.11, pp.716-724, 2007.
DOI : 10.2533/chimia.2007.716

URL : https://hal.archives-ouvertes.fr/hal-01230400

W. Wernsdorfer, Plasmodium falciparum: effect of antimalarial drugs on the production and secretion characteristics of histidine-rich protein II, Exp Parasitol, vol.102, pp.157-163, 2002.

A. Oduola, N. Weatherly, J. Bowdre, and R. Desjardins, Plasmodium falciparum: Induction of resistance to mefloquine in cloned strains by continuous drug exposure in vitro, Experimental Parasitology, vol.67, issue.2, pp.354-360, 1988.
DOI : 10.1016/0014-4894(88)90082-3

P. Pigeon, S. Top, A. Vessières, M. Huché, E. Hillard et al., Selective Estrogen Receptor Modulators in the Ruthenocene Series. Synthesis and Biological Behavior, Journal of Medicinal Chemistry, vol.48, issue.8, pp.2814-2821, 2005.
DOI : 10.1021/jm049268h

URL : https://hal.archives-ouvertes.fr/hal-00093015

A. Pizarro, A. Habtemariam, and P. Sadler, Activation Mechanisms for Organometallic Anticancer Complexes, Top Organomet Chem, vol.32, pp.21-56, 2010.
DOI : 10.1007/978-3-642-13185-1_2

M. Pomper, H. Vanbrocklin, A. Thieme, R. Thomas, D. Kiesewetter et al., 11.beta.-Methoxy-, 11.beta.-ethyl, and 17.alpha.-ethynyl-substituted 16.alpha.-fluoroestradiols: receptor-based imaging agents with enhanced uptake efficiency and selectivity, Journal of Medicinal Chemistry, vol.33, issue.12, pp.3143-3155, 1990.
DOI : 10.1021/jm00174a009

C. Rajapakse, A. Martínez, B. Naoulou, A. Jarzecki, L. Suárez et al., Synthesis, Characterization, and in vitro Antimalarial and Antitumor Activity of New Ruthenium(II) Complexes of Chloroquine, Inorganic Chemistry, vol.48, issue.3, pp.1122-1131, 2009.
DOI : 10.1021/ic802220w

K. Rieckmann, G. Campbell, L. Sax, and J. Mrema, Drug sensitivity of Plasmodium falciparum. An in vitro microtechnique, Lancet, vol.1, pp.22-23, 1978.

M. Schneider, E. Von-angerer, H. Schönenberger, R. Michel, and H. Fortmeyer, 1,1,2-Triphenylbut-1-enes: relationship between structure, estradiol receptor affinity, and mammary tumor inhibiting properties, Journal of Medicinal Chemistry, vol.25, issue.9, pp.1070-1077, 1982.
DOI : 10.1021/jm00351a013

M. Soares, J. Costa, M. De-sa, R. Santos, P. Pigeon et al., Antiparasitic and immunomodulatory activities of 1,1-bis(4-hydroxyphenyl)-2-phenyl-but-1-ene and its protected and free 2-ferrocenyl derivatives, Drug Development Research, vol.58, pp.69-75, 2010.
DOI : 10.1002/ddr.20349

URL : https://hal.archives-ouvertes.fr/hal-01230392

J. Swarts, A. Nafady, J. Roudebush, S. Trupia, and W. Geiger, One-Electron Oxidation of Ruthenocene: Reactions of the Ruthenocenium Ion in Gentle Electrolyte Media, Inorganic Chemistry, vol.48, issue.5, pp.2156-2165, 2009.
DOI : 10.1021/ic802105b

Y. Tan, P. Pigeon, S. Top, E. Labbé, O. Buriez et al., Ferrocenyl catechols: synthesis, oxidation chemistry and anti-proliferative effects on MDA-MB-231 breast cancer cells, Dalton Transactions, vol.46, issue.639, pp.7537-7549, 2012.
DOI : 10.1002/chem.201103378

URL : https://hal.archives-ouvertes.fr/hal-01230385

S. Top, I. Efremenko, M. Rager, A. Vessières, P. Yaswen et al., -Cp*Rh-Hydroxytamoxifen Complexes at the Estrogen, ER?? and ER?? Receptors, and Growth Inhibition to Breast Cancer Cells, Inorganic Chemistry, vol.50, issue.1, pp.271-284, 2011.
DOI : 10.1021/ic1019372

URL : https://hal.archives-ouvertes.fr/hal-01408995

S. Top, E. Kaloun, A. Vessières, I. Laios, G. Leclercq et al., The first titanocenyl dichloride moiety vectorised by a selective estrogen receptor modulator (SERM). Synthesis and preliminary bio-chemical behaviour, J Organomet Chem, vol.643644, pp.350-356, 2002.

S. Top, A. Vessières, G. Leclercq, J. Quivy, J. Tang et al., Synthesis, Biochemical Properties and Molecular Modelling Studies of Organometallic Specific Estrogen Receptor Modulators (SERMs), the Ferrocifens and Hydroxyferrocifens: Evidence for an Antiproliferative Effect of Hydroxyferrocifens on both Hormone-Dependent and Hormone-Independent Breast Cancer Cell Lines, Chemistry - A European Journal, vol.9, issue.21, pp.5223-5236, 2003.
DOI : 10.1002/chem.200305024

W. Trager and J. Jensen, Human malaria parasites in continuous culture, Science, vol.193, issue.4254, pp.673-675, 1976.
DOI : 10.1126/science.781840

T. Turbitt and W. Watts, Bridged ferrocenes, Journal of Organometallic Chemistry, vol.46, issue.1, pp.109-117, 1972.
DOI : 10.1016/S0022-328X(00)90481-9

M. Wenzel, M. Patra, C. Senges, I. Ott, J. Stepanek et al., Analysis of the Mechanism of Action of Potent Antibacterial Hetero-tri-organometallic Compounds: A Structurally New Class of Antibiotics, ACS Chemical Biology, vol.8, issue.7, pp.1442-1450, 2013.
DOI : 10.1021/cb4000844