T. Pluskal and S. Castillo, BMC Bioinform, p.395, 2010.

F. Olivon and G. Grelier, Anal. Chem, vol.89, pp.7836-7840, 2017.

F. Olivon and F. Roussi, Anal. Bioanal. Chem, vol.409, pp.5767-5778, 2017.

H. L. Röst and T. Sachsenberg, Nat. Methods, p.741, 2016.

P. Allard and T. Péresse, Anal. Chem, vol.88, pp.3317-3323, 2016.

F. Allen and A. Pon, Nucleic Acids Res, vol.42, pp.94-99, 2014.

R. R. Silva and M. Wang, PLOS Comput. Biol, p.1006089, 2018.

D. Krug-&-r and . Müller, Nat. Prod. Rep, vol.31, pp.768-783, 2014.

A. Bouslimani and L. M. Sanchez, Nat. Prod. Rep, vol.31, pp.718-729, 2014.

, Notes and references

Q. Michaudel, Y. Ishihara, and P. S. Baran, Acc. Chem. Res, vol.48, pp.712-721, 2015.

M. H. Medema and M. A. Fischbach, Nat. Chem. Biol, p.639, 2015.

M. T. Henke and N. L. Kelleher, Nat. Prod. Rep, vol.33, pp.942-950, 2016.

S. P. Gaudencio and F. Pereira, Nat. Prod. Rep, vol.32, pp.779-810, 2015.

J. Y. Yang, L. M. Sanchez, C. M. Rath, X. Liu, P. D. Boudreau et al., J. Nat. Prod, vol.76, pp.1686-1699, 2013.

R. A. Quinn, L. Nothias, O. Vining, M. Meehan, E. Esquenazi et al., Trends Pharmacol. Sci, vol.38, pp.143-154, 2017.

A. A. Aksenov, R. Silva, R. Knight, N. P. Lopes, and P. C. Dorrestein, Nat. Rev. Chem, 2017.

H. Mohimani and P. A. Pevzner, Nat. Prod. Rep, vol.33, pp.73-86, 2016.

M. Wang, J. J. Carver, V. V. Phelan, L. M. Sanchez, N. Garg et al.,

L. Murphy, C. Gerwick, Y. Liaw, H. Yang, M. Humpf et al.,

T. Metz, D. Peryea, D. Nguyen, P. Vanleer, A. Shinn et al.,

P. C. Moore, N. Dorrestein, and . Bandeira, Nat. Biotechnol, vol.34, pp.828-837, 2016.

K. Haug, R. M. Salek, P. Conesa, J. Hastings, P. De-matos et al.,

J. L. Sansone, C. Griffin, and . Steinbeck, Nucl. Acids Res, vol.41, pp.781-786, 2013.

A. E. Fox-ramos, C. Alcover, L. Evanno, A. Maciuk, M. Litaudon et al.,

E. Jullian, P. Mouray, P. M. Grellier, S. Loiseau, E. Pomel et al., J. Nat. Prod, vol.80, pp.1007-1014, 2017.

M. Janot, Tetrahedron, vol.14, pp.113-125, 1961.

H. Rapoport and R. E. Moore, J. Org. Chem, vol.27, pp.2981-2985, 1962.

E. L. Schymanski, J. Jeon, R. Gulde, K. Fenner, M. Ruff et al., Environ. Sci. Technol, vol.48, pp.2097-2098, 2014.

E. Otogo-n'nang, G. Bernadat, E. Mouray, B. Kumulungui, P. Grellier et al., Org. Lett, vol.20, pp.6596-6600, 2018.

T. Pluskal, S. Castillo, A. Villar-briones, M. Ore?i?, and . Bioinform, , p.395, 2010.

A. Boufridi, S. Petek, L. Evanno, M. A. Beniddir, C. Debitus et al., Tetrahedron Lett, vol.57, pp.4922-4925, 2016.

A. Boufridi, D. Lachkar, D. Erpenbeck, M. A. Beniddir, L. Evanno et al., Aust. J. Chem, vol.70, pp.743-750, 2016.

G. Daletos, N. J. De-voogd, W. E. Müller, V. Wray, W. Lin et al., J. Nat. Prod, vol.77, pp.218-226, 2014.

H. S. Radeke, C. A. Digits, R. L. Casaubon, and M. L. Snapper, Chem. Biol, vol.6, pp.639-647, 1999.

M. P. Nambiar and H. C. Wu, Exp. Cell Res, vol.219, pp.671-678, 1995.

L. Evanno, D. Lachkar, A. Lamali, A. Boufridi, B. Séon-méniel et al., Eur. J. Org. Chem, pp.2486-2497, 2018.

N. Bonneau, G. Chen, D. Lachkar, A. Boufridi, J. Gallard et al., Chem. Eur. J, vol.23, pp.14454-14461, 2017.

J. R. Winnikoff, E. Glukhov, J. Watrous, P. C. Dorrestein, and W. H. Gerwick, J. Antibiot, vol.67, pp.105-112, 2013.

R. M. Van-wagoner, A. K. Drummond, and J. L. Wright, Advances in Applied Microbiology, vol.61, pp.89-217, 2007.

N. A. Moss, T. Leão, M. R. Rankin, T. M. Mccullough, P. Qu et al., ACS Chem. Biol, vol.13, pp.3385-3395, 2018.

M. C. Chambers, B. Maclean, R. Burke, D. Amodei, D. L. Ruderman et al., Nat. Biotechnol, vol.30, pp.918-920, 2012.

A. M. Frank, N. Bandeira, Z. Shen, S. Tanner, S. P. Briggs et al., J. Proteome Res, vol.7, pp.113-122, 2008.

F. Olivon, G. Grelier, F. Roussi, M. Litaudon, and D. Touboul, Anal. Chem, vol.89, pp.7836-7840, 2017.

F. Olivon, F. Roussi, M. Litaudon, and D. Touboul, Anal. Bioanal. Chem, vol.409, pp.5767-5778, 2017.

J. Wolfender, J. Nuzillard, J. J. Van-der-hooft, J. Renault, and S. Bertrand, Anal. Chem, vol.91, pp.704-742, 2019.

P. Allard, T. Péresse, J. Bisson, K. Gindro, L. Marcourt et al., Anal. Chem, vol.88, pp.3317-3323, 2016.

F. Allen, A. Pon, M. Wilson, R. Greiner, and D. Wishart, Nuc. Acids Res, vol.42, pp.94-99, 2014.

J. Gu, Y. Gui, L. Chen, G. Yuan, H. Lu et al., PLoS One, 2013.

C. L. Zani and A. R. Carroll, At that time, the CFM-ID algorithm was not able to handle the fragmentation of permanently charged compounds, J. Nat. Prod, vol.80, pp.1758-1766, 2017.

M. Wang and N. Bandeira, J. Proteome Res, vol.12, pp.3944-3951, 2013.

J. A. Beutler, J. G. Jato, G. Cragg, D. F. Wiemer, J. D. Neighbors et al., , pp.301-309, 2006.

T. Pe?resse, G. Je?ze?quel, P. Allard, V. Pham, and D. T. ,

F. Huong, J. Blanchard, H. Bignon, J. Le?vaique, M. Wolfender et al., J. Nat. Prod, vol.80, pp.2684-2691, 2017.

L. C. Klein-júnior, S. Cretton, P. Allard, G. Genta-jouve, C. S. Passos et al., J. Nat. Prod, vol.80, pp.3032-3037, 2017.

R. R. Silva, M. Wang, L. Nothias, J. J. Van-der-hooft, A. M. Caraballo-rodríguez et al.,

N. P. Klassen, P. C. Lopes, and . Dorrestein, PLOS Comput. Biol, p.1006089, 2018.

C. Ruttkies, E. L. Schymanski, S. Wolf, J. Hollender, and S. Neumann, J. Cheminformatics, vol.8, p.3, 2016.

K. B. Kang, E. J. Park, R. R. Silva, H. W. Kim, P. C. Dorrestein et al., J. Nat. Prod, vol.81, pp.1819-1828, 2018.

I. Bla?enovi?, T. Kind, J. Ji, and O. Fiehn, Metabolites, vol.8, p.31, 2018.

A. C. Hartmann, D. Petras, R. A. Quinn, I. Protsyuk, F. I. Archer et al., Proc. Natl. Acad. Sci. U.S.A, vol.114, pp.11685-11690, 2017.

J. J. Kellogg, D. A. Todd, J. M. Egan, H. A. Raja, N. H. Oberlies et al., J. Nat. Prod, vol.79, pp.376-386, 2016.

F. E. Koehn and G. T. Carter, Nat. Rev. Drug. Discov, vol.4, pp.206-220, 2005.

C. B. Naman, R. Rattan, S. E. Nikoulina, J. Lee, B. W. Miller et al., J. Nat. Prod, vol.80, pp.625-633, 2017.

L. Nothias, M. Nothias-esposito, R. Silva, M. Wang, I. Protsyuk et al., J. Nat. Prod, vol.81, pp.758-767, 2018.

H. L. Röst, T. Sachsenberg, S. Aiche, C. Bielow, H. Weisser et al., Nat. Methods, vol.13, pp.741-748, 2016.

L. Nothias-scaglia, V. Dumontet, J. Neyts, F. Roussi, J. Costa et al., Fitoterapia, vol.105, pp.202-209, 2015.

F. Olivon, P. Allard, A. Koval, D. Righi, G. Genta-jouve et al., ACS Chem. Biol, vol.12, pp.2644-2651, 2017.

J. N. Anastas and R. T. Moon, Nat. Rev. Cancer, vol.13, pp.11-26, 2012.

F. Olivon, C. Apel, P. Retailleau, P. M. Allard, J. L. Wolfender et al.,

. Desrat, Org. Chem. Front, vol.5, pp.2171-2178, 2018.

D. J. Floros, P. R. Jensen, P. C. Dorrestein, and N. Koyama, Metabolomics, vol.12, p.145, 2016.

M. Crüsemann, E. C. O'neill, C. B. Larson, A. V. Melnik, D. J. Floros et al., J. Nat. Prod, vol.80, pp.588-597, 2017.

T. Luzzatto-knaan, N. Garg, M. Wang, E. Glukhov, Y. Peng et al., , 2017.

S. Roussel, S. Preys, F. Chauchard, and J. Lallemand, J. Chromatogr. A, pp.7-59, 2014.

H. Martens, S. W. Bruun, I. Adt, G. D. Sockalingum, and A. Kohler, J. Chemometr, vol.20, pp.402-417, 2006.

E. R. Britton, J. J. Kellogg, O. M. Kvalheim, and N. B. Cech, J. Nat. Prod, vol.81, pp.484-493, 2018.

L. K. Caesar, J. J. Kellogg, O. M. Kvalheim, R. A. Cech, and N. B. Cech, Planta Med, vol.84, pp.721-728, 2018.

O. M. Kvalheim, H. Chan, I. F. Benzie, and Y. Szeto, Chemometr. Intell. Lab. Syst, vol.107, pp.98-105, 2011.

M. Vallet, Q. P. Vanbellingen, T. Fu, J. Caer, S. Dellanegra et al., J. Nat. Prod, vol.80, pp.2863-2873, 2017.

F. Peypoux, J. Bonmatin, and J. Wallach, Appl. Microbiol. Biotechnol, vol.51, pp.553-563, 1999.

R. H. Baltz, J. Ind. Microbiol. Biotechnol, 2018.

K. Kleigrewe, J. Almaliti, I. Y. Tian, R. B. Kinnel, A. Korobeynikov et al., J. Nat. Prod, vol.78, pp.1671-1682, 2015.

M. Nagarajan, V. Maruthanayagam, and M. ,

. Sundararaman, J. Appl. Toxicol, vol.32, pp.153-185, 2012.

M. Maansson, N. G. Vynne, A. Klitgaard, J. L. Nybo, J. Melchiorsen et al., mSystems, vol.1, pp.28-43, 2016.

K. R. Duncan, M. Crüsemann, A. Lechner, A. Sarkar, J. Li et al., Chem. Biol, vol.22, pp.460-471, 2015.

J. R. Doroghazi, J. C. Albright, A. W. Goering, K. Ju, R. R. Haines et al., Nat. Chem. Biol, vol.10, pp.963-968, 2014.

A. W. Goering, R. A. Mcclure, J. R. Doroghazi, and J. ,

N. A. Albright, Y. Haverland, K. Zhang, R. J. Ju, W. W. Thomson et al., ACS Cent. Sci, vol.2, pp.99-108, 2016.

R. A. Mcclure, A. W. Goering, K. Ju, J. A. Baccile, and F. ,

W. W. Schroeder, R. J. Metcalf, and N. Thomson,

. Kelleher, ACS Chem. Biol, vol.11, pp.3452-3460, 2016.

E. I. Parkinson, J. H. Tryon, A. W. Goering, K. Ju, R. A. Mcclure et al., ACS Chem. Biol, vol.13, pp.1029-1037, 2018.

M. T. Henke, A. A. Soukup, A. W. Goering, R. A. Mcclure, R. J. Thomson et al., ACS Chem. Biol, vol.11, pp.2117-2123, 2016.

J. Begani, J. Lakhani, and D. Harwani, Ann. Microbiol, vol.68, pp.1-14, 2018.

H. B. Bode, D. Reimer, S. W. Fuchs, F. Kirchner, C. Dauth et al., Chem. Eur. J, vol.18, pp.2342-2348, 2012.

A. Klitgaard, J. B. Nielsen, R. J. Frandsen, M. R. Andersen, and K. F. Nielsen, Anal. Chem, vol.87, pp.6520-6526, 2015.

F. Olivon, N. Elie, G. Grelier, F. Roussi, M. Litaudon et al.,

. Touboul, Anal. Chem, vol.90, pp.13900-13908, 2018.

L. Van-der-maaten and G. Hinton, J. Mach. Learn. Res, vol.9, pp.2579-2605, 2008.

G. Wohlgemuth, S. S. Mehta, R. F. Mejia, S. Neumann, D. Pedrosa et al., Nat. Biotechnol, vol.34, pp.1099-1101, 2016.

H. Horai, M. Arita, S. Kanaya, Y. Nihei, T. Ikeda et al., J. Mass Spectrom, vol.45, pp.703-714, 2010.

D. S. Wishart, T. Jewison, A. C. Guo, M. Wilson, C. Knox et al., Nucl. Acids Res, vol.41, pp.801-807, 2013.

H. Tsugawa, T. Cajka, T. Kind, Y. Ma, B. Higgins et al., Nat. Methods, vol.12, pp.523-526, 2015.

M. A. Stravs, E. L. Schymanski, H. P. Singer, and J. Hollender, J. Mass Spectrom, vol.48, pp.89-99, 2013.

K. Skogerson, G. Wohlgemuth, D. K. Barupal, O. Fiehn, and . Bioinform, , p.321, 2011.

O. Spjuth, T. Helmus, E. L. Willighagen, S. Kuhn, M. Eklund et al., , vol.8, p.59, 2007.

X. Domingo-almenara, J. R. Montenegro-burke, H. P. Benton, and G. Siuzdak, Anal. Chem, vol.90, pp.480-489, 2018.

J. Wandy, Y. Zhu, J. J. Van-der-hooft, R. Daly, M. P. Barrett et al., Bioinformatics, vol.34, pp.317-318, 2018.

J. J. Van-der-hooft, J. Wandy, M. P. Barrett, K. E. Burgess, and S. Rogers, Proc. Natl. Acad. Sci. U.S.A, vol.113, pp.13738-13743, 2016.

S. Böcker and K. Dührkop, J. Cheminformatics, vol.8, p.5, 2016.

K. Dührkop, H. Shen, M. Meusel, J. Rousu, and S. Böcker, Proc. Natl. Acad. Sci. U.S.A, vol.112, pp.12580-12585, 2015.

H. Mohimani, A. Gurevich, A. Shlemov, A. Mikheenko, A. Korobeynikov et al., Nat. Commun, vol.9, p.4035, 2018.

H. Mohimani, A. Gurevich, A. Mikheenko, N. Garg, and L. ,

A. Nothias, K. Ninomiya, P. C. Takada, P. A. Dorrestein, and . Pevzner, Nat. Chem. Biol, vol.13, pp.30-37, 2016.

A. Gurevich, A. Mikheenko, A. Shlemov, A. Korobeynikov, H. Mohimani et al., Nat. Microbiol, vol.3, pp.319-327, 2018.

H. Mohimani, R. D. Kersten, W. Liu, M. Wang, S. O. Purvine et al., ACS Chem. Biol, vol.9, pp.1545-1551, 2014.

Y. Beauxis and G. Genta-jouve, Bioinformatics, vol.864, 2018.

S. Böcker, M. C. Letzel, Z. Lipták, and A. Pervukhin, Bioinformatics, vol.25, pp.218-224, 2009.

E. E. Bolton, Y. Wang, P. A. Thiessen, and S. H. Bryant, Annual Reports in Computational Chemistry, vol.4, pp.217-241, 2008.

C. A. Smith, E. J. Want, G. O'maille, R. Abagyan, and G. Siuzdak, Anal. Chem, vol.78, pp.779-787, 2006.

D. M. Blei, A. Y. Ng, and M. I. Jordan, J. Mach. Learn. Res, vol.3, pp.993-1022, 2003.

S. Allard, P. Allard, I. Morel, and T. Gicquel, Drug Test. Anal, 2018.

M. Köck, A. Grube, I. B. Seiple, and P. S. Baran, Angew. Chem. Int. Ed, vol.46, pp.6586-6594, 2007.

J. B. Mcalpine, S. Chen, A. Kutateladze, J. B. Macmillan, G. Appendino et al., Nat. Prod. Rep, vol.36, pp.35-107, 2019.

V. Salim-&-v.-de and . Luca, , pp.1-37, 2013.

Y. Qu and R. Simonescu, J. Nat. Prod, vol.79, issue.2, pp.3143-3147, 2016.

H. J. Knölker, , 2016.

P. M. Dewick, , 2009.

L. F. Szabó, Molecules, vol.13, pp.1875-1896, 2008.

S. E. O'connor and &. J. Maresh, Nat. Prod. Rep, vol.23, pp.532-547, 2006.

M. Kitajima and N. Kogure, Org. Lett, vol.5, pp.2075-2078, 2003.

Y. Yang and Y. Bai, Org. Lett, vol.16, pp.6216-6219, 2014.

J. Buckingham and K. H. Baggaley, , 2010.

C. H. Heathcock and M. H. Norman, J. Org. Chem, vol.55, pp.798-811, 1990.

R. T. Brown and D. M. Duckworth, Tetrahedron Lett, vol.32, 1987.

S. Benayad and K. Ahamada, Eur. J. Org. Chem, pp.1494-1499, 2016.

Q. Pan, N. R. Mustafa, K. Tang, Y. H. Choi, and R. Verpoorte, Monoterpenoid indole alkaloids biosynthesis and its regulation in Catharanthus roseus: A literature review from genes to metabolites, Phytochemistry Rev, vol.15, pp.221-250, 2016.

B. P. Pritchett and B. M. Stoltz, Enantioselective palladium-catalyzed allylic alkylation reactions in the synthesis of Aspidosperma and structurally related monoterpene indole alkaloids, Nat. Prod. Rep, vol.35, pp.559-574, 2018.

Y. Qu, Completion of the seven-step pathway from tabersonine to the anticancer drug precursor vindoline and its assembly in yeast, Proc. Natl. Acad. Sci. USA, vol.112, pp.6224-6229, 2015.

L. Caputi, Missing enzymes in the biosynthesis of the anticancer drug vinblastine in Madagascar periwinkle, Science, vol.360, pp.1235-1239, 2018.

T. T. Dang, Sarpagan bridge enzyme has substrate-controlled cyclization and aromatization modes, Nat. Chem. Biol, vol.14, pp.760-763, 2018.

K. Miettinen, The seco-iridoid pathway from Catharanthus roseus, Nat. Commun, vol.5, p.3606, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00978593

V. Salim, F. Yu, J. Altarejos, and V. Luca, Virus-induced gene silencing identifies Catharanthus roseus 7-deoxyloganic acid-7-hydroxylase, a step in iridoid and monoterpene indole alkaloid biosynthesis, Plant J, vol.76, pp.754-765, 2013.

K. Asada, A 7-deoxyloganetic acid glucosyltransferase contributes a key step in secologanin biosynthesis in Madagascar periwinkle, Plant Cell, vol.25, pp.4123-4134, 2013.

D. Lachkar, Unified biomimetic assembly of voacalgine A and bipleiophylline via divergent oxidative couplings, Nat. Chem, vol.9, p.793, 2017.

N. Otogo and E. Obiang, Pleiokomenines A and B: Dimeric aspidofractinine alkaloids tethered with a methylene group, Org. Lett, vol.19, pp.6180-6183, 2017.

N. A. Hughes and &. Rapoport, J. Am. Chem. Soc, vol.80, pp.1604-1609, 1958.

H. Rapoport and T. P. Onak, J. Am. Chem. Soc, vol.80, pp.1601-1604, 1958.

H. Rapoport and R. J. Windgassen, J. Am. Chem. Soc, vol.82, pp.4404-4414, 1960.

H. E. Rapoport-&-r and . Moore, J. Org. Chem, vol.27, pp.2981-2985, 1962.

R. E. Moore and &. Rapoport, J. Org. Chem, vol.38, pp.215-230, 1973.

V. Muñoz and M. Sauvain, J. Ethnopharmacol, vol.69, pp.127-137, 2000.

H. Ishiyama and M. Matsumoto, Heterocycles, vol.66, pp.651-658, 2005.

J. A. Lima and R. S. Costa, Pharmacol. Biochem. Behav, vol.92, pp.508-513, 2009.

F. Mbeunkui and M. H. Grace, J. Ethnopharmacol, vol.139, pp.471-477, 2012.

, Orme aux Merisiers RD 128 / 91190 Saint-Aubin, France Coordinates, energies and lowest frequency of the most stable conformer found for compound 1: Sum of electronic and thermal Free Energies, p.1258

J. Buckingham, K. H. Baggaley, A. D. Roberts, and L. F. Szabó, Dictionary of Alkaloids, with CD-ROM

J. Guillon, P. Grellier, M. Labaied, P. Sonnet, J. Léger et al., J. Med. Chem, vol.47, 1997.

Z. Cheikh-ali, J. Caron, S. Cojean, C. Bories, P. Couvreur et al., ChemMedChem, vol.10, pp.411-418, 2015.

. A. Dr, C. Fox-ramos, P. Pavesi, . E. Dr, . Poupon et al., Pharmacognosie-chimie des substances naturelles, BioCIS, Univ. Paris-Sud

. Clément, , 92290.

.. M. Dr and D. V. Litaudon, Dumontet Institut de Chimie des Substances Naturelles, CNRS, ICSN UPR 2301

. G. Dr, C. Genta-jouve, and C. Umr-cnrs-8038, , p.75006

F. Paris, E-mail: gregory.genta-jouve@parisdescartes.fr

, Unité Molécules de Communication et Adaptation des Microorganismes (UMR 7245)

). Nothias, M. Nothias-esposito, R. Silva, M. Wang, I. Protsyuk et al., Proc. Natl. Acad. Sci. U.S.A, vol.81, pp.11999-12004, 2015.

E. Otogo-n'nang, G. Bernadat, E. Mouray, B. Kumulungui, P. Grellier et al., Org. Lett, vol.20, pp.6596-6600, 2018.

R. D. Wada, T. Kersten, S. Iwai, F. Lee, T. Sakurai et al., Angew. Chem. Int. Ed, vol.57, pp.3671-3675, 2018.

R. H. Grkovic, M. Pouwer, L. Vial, A. Gambini, J. N. Noël et al., Proc. Natl. Acad. Sci. U.S.A, vol.53, pp.9753-9758, 2011.

M. T. Henke and N. L. Kelleher, Nat. Prod. Rep, vol.33, pp.942-950, 2016.

S. Böcker, Curr. Opin. Chem. Biol, vol.36, pp.1-6, 2017.

K. Dührkop, H. Shen, M. Meusel, J. Rousu, and S. Böcker, Proc. Natl. Acad. Sci. U.S.A, vol.112, pp.12580-12585, 2015.

J. J. Van-der-hooft, J. Wandy, M. P. Barrett, K. E. Burgess, and S. Rogers, Proc. Natl. Acad. Sci. U.S.A, vol.113, pp.13738-13743, 2016.

J. Watrous, P. Roach, T. Alexandrov, B. S. Heath, J. Y. Yang et al.,

J. Moore, N. Laskin, P. C. Bandeira, and . Dorrestein, Proc. Natl. Acad. Sci, vol.109, pp.1743-1752, 2012.

R. A. Quinn, L. Nothias, O. Vining, M. Meehan, E. Esquenazi et al., Trends Pharmacol. Sci, vol.38, pp.143-154, 2017.

). A. Fox-ramos, C. Alcover, L. Evanno, A. Maciuk, M. Litaudon et al., J. Nat. Prod, vol.80, pp.1007-1014, 2017.

P. Gallard, S. Retailleau, C. Petek, L. Debitus, M. A. Evanno et al., Chem. Eur. J, vol.23, p.1237, 2017.

M. Wang, J. J. Carver, V. V. Phelan, L. M. Sanchez, N. Garg et al.,

L. Murphy, C. Gerwick, Y. Liaw, H. Yang, M. Humpf et al.,

C. B. Klitgaard, C. A. Larson, P. Boya, D. Torres-mendoza, D. J. Gonzalez et al.,

K. F. Mcphail, L. Nielsen, M. Vuong, M. F. Elfeki, N. Traxler et al.,

. Bandeira, Nat. Biotechnol, vol.34, pp.828-837, 2016.

). Allard, T. Péresse, J. Bisson, K. Gindro, L. Marcourt et al., Anal. Chem, vol.88, pp.3317-3323, 2016.

C. Dorrestein, 14, e1006089; c) S. Böcker and K, PLOS Comput. Biol, 2018.

J. Dührkop, ). Cheminformatics-;-d, J. Wolfender, J. J. Nuzillard, J. Van-der-hooft et al., Anal. Chem, vol.8, pp.704-742, 2016.

). J. Wandy, Y. Zhu, J. J. Van-der-hooft, R. Daly, M. P. Barrett et al., Bioinformatics, vol.34, pp.317-318, 2018.

J. Rowley, J. Inf. Sci, vol.33, pp.163-180, 2007.

Y. Beauxis and G. Genta-jouve, Bioinformatics, vol.864, 2018.

F. Allen, A. Pon, M. Wilson, R. Greiner, and D. Wishart, Nucleic Acids Res, vol.42, pp.94-99, 2014.

C. Audoin, V. Cocandeau, O. P. Thomas, A. Bruschini, S. Holderith et al., , vol.4, pp.421-432, 2014.

P. Allard, G. Genta-jouve, and J. Wolfender, Curr. Opin. Chem. Biol, vol.36, pp.40-49, 2017.

E. Otogo-n'nang-obiang, G. Genta-jouve, J. Gallard, B. Kumulungui, E. Mouray et al., J. Nat. Prod, vol.19, pp.1075-1078, 2017.

T. Pluskal, S. Castillo, A. Villar-briones, M. Ore?i?, and . Bioinform, , p.395, 2010.

, MIADB is a MS/MS spectral database of 172 monoterpene indole alkaloids that can be accessed online on the GNPS, 2019.

I. Bla?enovi?, T. Kind, J. Ji, and O. Fiehn, , vol.8, p.31, 2018.

. Dnp, , 2019.

G. Bachelard, ;. Bla?enovi?, T. Kind, J. Ji, and O. Fiehn, References: (1), La valeur inductive de la relativité, vol.8, p.31, 1929.

;. K. Sevenet, Z. Bláha, J. Koblicová, and . Trojanek, Collect. Czech. Chem. Commun, vol.29, issue.3, pp.101-113, 1974.

M. C. Chambers, B. Maclean, R. Burke, D. Amodei, D. L. Ruderman et al.,

B. Gatto, B. Fischer, J. Pratt, S. Egertson-;-t.-pluskal, A. Castillo et al., Nat. Biotechnol, vol.30, issue.5, pp.8696-8703, 2010.

P. Shannon, A. Markiel, O. Ozier, N. S. Baliga, J. T. Wang et al., Genome Res, vol.13, pp.2498-2504, 2003.

, Orme aux Merisiers RD 128 / 91190 Saint-Aubin, France ? Determination de l'activité antileishmanienne L'activité antileishmanienne des composés isolés a été determinée par Sébastien Pomel sous la direction de Phillipe M, Loiseau à la faculté de pharmacie de l'université

, MHOM/ET/67/HU3), souche LV9, fournie par le professeur S. Croft, de la London School of Hygiene and Tropical Medicine, au Royaume-Uni. L'évaluation sur les amastigotes axéniques a été adaptée de la méthode utilisée par, L'évaluation in vitro a été réalisée sur la lignée cellulaire de L. donovani

, Cette conversion a été contrôlée en mesurant l'absorbance à des longueurs d'ondes spécifiques de la résofurine (570 nm) et de la résazurine (600 nm), en utilisant un lecteur de microplaques (Lab systems Multiskan MS). Les activités finales ont été exprimées sous forme de valeurs de CI50, Dans les cellules vivantes, la résazurine est réduite en résofurine

J. Guillon and P. Grellier, J. Med. Chem, 1997.

Z. Cheikh-ali and J. Caron, ChemMedChem, vol.10, pp.411-418, 2015.

. Bilan-des-communications-communication-orale, congrès international : 1 SINAPSIS 2017 -Berlin, Allemagne, pp.24-26

A. E. Fox-ramos, J. Gallard, V. Dumontet, M. Litaudon, P. Champy et al., Prioritizing natural product diversity in three Alstonia species endemic to New Caledonia through massive multi-informative molecular networks

, Communications orales, congrès, vol.nationaux, p.3

, PSE Young Scientists' Meeting Lille 2017 -Villeneuve d'Asqc, pp.28-29

A. E. Fox-ramos, J. Gallard, V. Dumontet, M. Litaudon, P. Champy et al., Prioritizing natural product diversity in three Alstonia species endemic to New Caledonia through massive multi-informative molecular networks

, Journée des doctorants de 2 e année (séminaire annuel de l'UMR, 2017.

A. E. Fox-ramos, P. Champy, and M. A. Beniddir, Exploration of the chemical diversity of South American and New Caledonian Gentianales with antiparasitic potential by the molecular networking technique SINAPSIS, pp.24-26, 2016.

A. E. Fox-ramos, C. Alcover, A. Maciuk, C. Duplais, G. Bernadat et al., Unveiling the secrets of forgotten plants: molecular networking-based phytochemical study of Geissospermum laeve

, th Joint Natural Products Conference 2016 -Copenhague, Danemark, pp.24-27, 2016.

A. E. Fox-ramos, C. Alcover, A. Maciuk, C. Duplais, G. Bernadat et al., Unveiling the secrets of forgotten plants: molecular networking-based phytochemical study of Geissospermum laeve, des actes des congrès ont été publiés

F. ?. Aubin, M. A. Beniddir, A. E. Fox-ramos, E. Otogo-n'nang, C. Alcover et al.,

A. E. Fox-ramos, J. Gallard, V. Dumontet, M. Litaudon, P. Champy et al., Prioritizing natural product diversity in three Alstonia species endemic to New Caledonia through massive multi-informative molecular networks

, th Young Research Fellow Meeting, 2017.

A. E. Fox-ramos, J. Gallard, V. Dumontet, M. Litaudon, P. Champy et al., Prioritizing natural product diversity in three Alstonia species endemic to New Caledonia through massive multi-informative molecular networks

, XVI ème Journée de l'École doctorale 569 « Innovation thérapeutique : du fondamental à l'appliqué, vol.17, 2016.

A. E. Fox-ramos, C. Alcover, L. Evanno, A. Maciuk, M. Litaudon et al., Unveiling the secrets of forgotten plants: molecular networking-based phytochemical study of Geissospermum laeve

A. E. Fox-ramos, C. Alcover, A. Maciuk, C. Duplais, J. Jullian et al., Étude phytochimique de Geissospermum laeve

C. Actes-de,

, th Joint Natural Products Conference 2016 -Copenhague, Danemark, pp.24-27, 2016.

A. E. Fox-ramos, C. Alcover, A. Maciuk, C. Duplais, G. Bernadat et al., Unveiling the secrets of forgotten plants: molecular networking-based phytochemical study of Geissospermum laeve (Vell.) Miers, des actes des congrès ont été publiés, p.31, 2016.

M. A. Beniddir, A. E. Fox-ramos, E. Otogo-n'nang, C. Alcover, A. Maciuk et al., Out of fashion plants at the "big data" Era: Illuminating the overlooked Apocynaceae alkaloids chemical space by molecular networking, Planta Med, vol.81, p.46, 2016.

A. E. ?-fox-ramos, C. Alcover, L. Evanno, A. Maciuk, M. Litaudon et al., Revisiting previously investigated plants: a molecular networking-based study of Geissospermum laeve, J. Nat. Prod, vol.80, issue.4, pp.1107-1014, 2017.

A. E. ?-fox-ramos, P. Le-pogam, C. Fox-alcover, E. Otogo-n'nang, G. Cauchie et al., Collected mass spectrometry data on monoterpene, 2019.

A. E. ?-fox-ramos, L. Evanno, E. Poupon, P. Champy, and M. A. Beniddir, Natural products targeting strategies involving molecular networking: Different manners, one goal, Nat. Prod. Rep, issue.2, 2019.

A. E. ?-fox-ramos, C. Pavesi, M. Litaudon, V. Dumontet, E. Poupon et al., CANPA: Computer-Assisted Natural Products Anticipation, 2019.

, Orme aux Merisiers RD 128 / 91190 Saint-Aubin, France Ces références sont celles citées en notes de bas de page dans le corps de texte, Certaines des références citées dans les articles acceptés

S. Walker, Big Data: a revolution that will transform how we live, work, and think, Int. J. Advert, vol.33, pp.181-183, 2014.

J. Hubert, J. Nuzillard, and J. Renault, Dereplication strategies in natural product research: How many tools and methodologies behind the same concept?, Phytochem Rev, vol.16, pp.55-95, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01995008

M. Wang, J. J. Carver, V. V. Phelan, L. M. Sanchez, N. Garg et al., Sharing and community curation of mass spectrometry data with Global Natural Products Social Molecular Networking, Nat. Biotechnol, p.828, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01371824

N. B. Cech and K. Yu, Mass spectrometry for natural products research, LC GC N. Am, vol.31, pp.938-947, 2013.

D. Nikoli? and . Casmi, A manual approach for dereplication of natural products using tandem mass spectrometry, Phytochem Lett, vol.21, pp.292-296, 2016.

J. Y. Yang, L. M. Sanchez, C. M. Rath, X. Liu, P. D. Boudreau et al., Molecular Networking as a dereplication strategy, J. Nat. Prod, vol.76, pp.1686-1699, 2013.

F. Olivon, N. Elie, G. Grelier, F. Roussi, M. Litaudon et al., MetGem software for the generation of molecular networks based on t-SNE algorithm, Anal. Chem, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02159533

T. Pluskal, S. Castillo, A. Villar-briones, and M. Ore?i?, MZmine 2: modular framework for processing, visualizing, and analyzing mass spectrometry-based molecular profile data, BMC Bioinform, p.395, 2010.

F. Olivon, G. Grelier, F. Roussi, M. Litaudon, and D. Touboul, MZmine 2 datapreprocessing to enhance molecular networking reliability, Anal. Chem, vol.89, pp.7836-7840, 2017.

F. Olivon, F. Roussi, M. Litaudon, and D. Touboul, Optimized experimental workflow for tandem mass spectrometry molecular networking in metabolomics, Anal. Bioanal. Chem, vol.409, pp.5767-5778, 2017.

H. L. Röst, T. Sachsenberg, S. Aiche, C. Bielow, H. Weisser et al., OpenMS: a flexible open-source software platform for mass spectrometry data analysis, Nat. Methods, p.741, 2016.

P. Allard, T. Péresse, J. Bisson, K. Gindro, L. Marcourt et al., Integration of molecular networking and in-silico MS/MS fragmentation for natural products dereplication, Anal. Chem, vol.88, pp.3317-3323, 2016.

F. Allen, A. Pon, M. Wilson, R. Greiner, and D. Wishart, CFM-ID: a web server for annotation, spectrum prediction and metabolite identification from tandem mass spectra, Nucleic Acids Res, vol.42, pp.94-99, 2014.

R. R. Silva, M. Wang, L. Nothias, J. J. Van-der-hooft, A. M. Caraballorodríguez et al.,

, Orme aux Merisiers RD 128 / 91190 Saint-Aubin, France Dorrestein, Propagating annotations of molecular networks using in silico fragmentation, PLOS Comput. Biol, p.1006089, 2018.

D. Krug and R. Müller, Secondary metabolomics: the impact of mass spectrometrybased approaches on the discovery and characterization of microbial natural products, Nat. Prod. Rep, vol.31, pp.768-783, 2014.

A. Bouslimani, L. M. Sanchez, N. Garg, and P. C. Dorrestein, Mass spectrometry of natural products: current, emerging and future technologies, Nat. Prod. Rep, vol.31, pp.718-729, 2014.

F. Olivon, P. Allard, A. Koval, D. Righi, G. Genta-jouve et al., Bioactive natural products prioritization using massive multi-informational molecular networks, ACS Chem. Biol, vol.12, pp.2644-2651, 2017.

K. Kleigrewe, J. Almaliti, I. Y. Tian, R. B. Kinnel, A. Korobeynikov et al., Combining mass spectrometric metabolic profiling with genomic analysis: a powerful approach for discovering natural products from cyanobacteria, J. Nat. Prod, vol.78, pp.1671-1682, 2015.

S. Böcker, M. C. Letzel, Z. Lipták, and A. Pervukhin, SIRIUS: decomposing isotope patterns for metabolite identification, Bioinformatics, vol.25, pp.218-224, 2008.

K. Dührkop, H. Shen, M. Meusel, J. Rousu, and S. Böcker, Searching molecular structure databases with tandem mass spectra using CSI: FingerID, Proc. Natl. Acad. Sci. USA, vol.112, pp.12580-12585, 2015.

Y. Beauxis and G. Genta-jouve, Metwork: a web server for natural products anticipation, Bioinformatics, 2018.

L. F. Szabó, Rigorous biogenetic network for a group of indole alkaloids derived from strictosidine, Molecules, vol.13, pp.1875-1896, 2008.

P. M. Dewick, Medicinal Natural Products: A Biosynthetic Approach, 2009.

M. Lahlou, 27. Sarpagandha: health benefits, dosage , side effects, The success of natural products in drug discovery, vol.4, pp.22-31, 2013.

J. Stöckigt, L. Barleben, S. Panjikar, and E. A. Loris, 3D-Structure and function of strictosidine synthase-the key enzyme of monoterpenoid indole alkaloid biosynthesis, Plant Physiol Biochem, vol.46, pp.340-355, 2008.

J. Le-men and W. I. Taylor, A uniform numbering system for indole alkaloids, vol.21, pp.508-510, 1965.

M. W. Chase, M. Christenhusz, M. Fay, J. Byng, W. Judd et al., An update of the angiosperm phylogeny group classification for the orders and families of flowering plants: APG IV, Bot. J. Linn. Soc, vol.181, pp.1-20, 2016.

D. Gülcemal, M. Masullo, O. Alanku?-Çal??kan, T. Karay?ld?r?m, S. G. ?enol et al., Monoterpenoid glucoindole alkaloids and iridoids from Pterocephalus pinardii, vol.48, pp.239-243, 2010.

X. Huang, Y. Li, Y. Su, X. Chai, and S. Yan, Monoterpene indole alkaloids and monoterpene diglycosides from the roots of Triosteum pinnatifidum, Phytochem Lett, vol.7, pp.30-34, 2014.

J. Buckingham, K. H. Baggaley, A. D. Roberts, and L. F. Szabo, Dictionary of alkaloids, with CD-ROM, 2010.

Z. Liu, J. Zhang, S. Li, M. Liu, X. Huang et al., Ervadivamines A and B, Two Unusual Trimeric Monoterpenoid Indole Alkaloids from Ervatamia divaricata, J. Org. Chem, vol.83, pp.10613-10618, 2018.

Y. Hirasawa, S. Miyama, T. Hosoya, K. Koyama, A. Rahman et al., Alasmontamine A, a first tetrakis monoterpene indole alkaloid from Tabernaemontana elegans, Org. Lett, vol.11, pp.5718-5721, 2009.

E. O. N'nang-obiang, G. Genta-jouve, J. Gallard, B. Kumulungui, E. Mouray et al., Pleiokomenines A and B: dimeric aspidofractinine alkaloids tethered with a methylene group, Org. Lett, vol.19, pp.6180-6183, 2017.

A. E. Nugroho, Y. Hirasawa, N. Kawahara, Y. Goda, K. Awang et al., Bisnicalaterine A, a vobasine? vobasine bisindole alkaloid from Hunteria zeylanica, J. Nat. Prod, vol.72, pp.1502-1506, 2009.

E. Otogo-n'nang, G. Bernadat, E. Mouray, B. Kumulungui, P. Grellier et al., Theionbrunonines A and B: dimeric vobasine alkaloids tethered by a thioether bridge from Mostuea brunonis, Org. Lett, vol.20, pp.6596-6600, 2018.

A. E. Fox-ramos, C. Alcover, L. Evanno, A. Maciuk, M. Litaudon et al., Revisiting previously investigated plants: a molecular networking-based study of Geissospermum laeve, J. Nat. Prod, vol.80, pp.1007-1014, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02105819

L. Caputi, J. Franke, S. C. Farrow, K. Chung, R. M. Payne et al., Science, vol.360, pp.1235-1239, 2018.

V. Salim and V. De-luca, Advances in Botanical Research, vol.68, pp.1-37, 2013.

Y. Qu, R. Simonescu, and V. De-luca, Monoterpene indole alkaloids from the fruit of Tabernaemontana litoralis and differential alkaloid composition in various fruit components, J. Nat. Prod, vol.79, pp.3143-3147, 2016.

H. J. Knölker, The alkaloids, 2016.

S. E. O'connor and J. J. Maresh, Chemistry and biology of monoterpene indole alkaloid biosynthesis, vol.23, pp.532-547, 2006.

M. Kitajima, N. Kogure, K. Yamaguchi, H. Takayama, and N. Aimi, Structure reinvestigation of gelsemoxonine, a constituent of Gelsemium elegans, reveals a novel, azetidine-containing indole alkaloid, Org. Lett, vol.5, pp.2075-2078, 2003.

Y. Yang, Y. Bai, S. Sun, and M. Dai, Biosynthetically inspired divergent approach to monoterpene indole alkaloids: total synthesis of mersicarpine, leuconodines B and D, leuconoxine, melodinine E, leuconolam, and rhazinilam, Org. Lett, vol.16, pp.6216-6219, 2014.

C. H. Heathcock, M. H. Norman, and D. A. Dickman, Total synthesis of (.+-.)-vallesamidine, J. Org. Chem, vol.55, pp.798-811, 1990.

R. T. Brown, D. M. Duckworth, and C. A. Santos, Biogenetically patterned synthesis of cadambine, Tetrahedron Lett, vol.32, 1987.

S. Benayad, K. Ahamada, G. Lewin, L. Evanno, and E. Poupon, Preakuammicine: A LongAwaited Missing Link in the Biosynthesis of Monoterpene Indole Alkaloids, Eur. J. Org. Chem, pp.1494-1499, 2016.

E. Otogo-n'nang, , 2018.

. Pao-pereira, Commerce équitable et durable, pp.15-24, 2018.

M. E. Endress, S. Liede-schumann, and U. Meve, An updated classification for Apocynaceae, Phytotaxa, vol.159, pp.175-194, 2014.

J. Miers, Geissospermum urceolatum, vol.55, pp.8-09, 1878.

G. Reticulatum, , pp.8-09, 2018.

J. C. Steele, N. C. Veitch, G. C. Kite, M. S. Simmonds, and D. C. Warhurst, Indole and ?-Carboline alkaloids from Geissospermum sericeum, J. Nat. Prod, vol.65, pp.85-88, 2002.

G. Allem and . Secretariat, , pp.18-27

I. R. Koch, A. Simões, A. O. Kinoshita, L. S. Spina, A. P. Castello et al., , pp.18-27

(. Geissospermum-laeve and . Vell, , pp.18-27

S. Bertani, G. Bourdy, I. Landau, J. Robinson, P. Esterre et al., Evaluation of French Guiana traditional antimalarial remedies, J. Ethnopharmacol, vol.98, pp.45-54, 2005.

P. Grenand, C. Moretti, H. Jacquemin, and M. Prévost, , p.126, 1987.

F. Mbeunkui, M. H. Grace, and M. A. Lila, Isolation and structural elucidation of indole alkaloids from Geissospermum vellosii by mass spectrometry, J. Chromatogr. B, vol.885, pp.83-89, 2012.

, Comité français de la pharmacopée « Plantes médicinales et huiles essentielles » -CP022014043

N. A. Hughes and H. Rapoport, Flavopereirine, an alkaloid from Geissospermum vellosii, J. Am. Chem. Soc, vol.80, pp.1604-1609, 1958.

H. Rapoport, T. P. Onak, N. A. Hughes, and M. G. Reinecke, Alkaloids of Geissospermum vellosii, J. Am. Chem. Soc, vol.80, pp.1601-1604, 1958.

H. Rapoport, R. J. Windgassen, N. A. Hughes, and T. P. Onak, Alkaloids of Geissospermum vellosii. Further Studies on geissospermine and the structures of the indolic cleavage products, geissoschizine and apogeissoschizine, J. Am. Chem. Soc, vol.82, pp.4404-4414, 1960.

H. Rapoport and R. E. Moore, Alkaloids of Geissospermum vellosii. Isolation and structure determinations of vellosimine, vellosiminol, and geissolosimine, J. Org. Chem, vol.27, pp.2981-2985, 1962.

R. E. Moore and H. Rapoport, Geissovelline, a new alkaloid from Geissospermum vellossi, J. Org. Chem, vol.38, pp.215-230, 1973.

V. Muñoz, M. Sauvain, G. Bourdy, J. Callapa, S. Bergeron et al., A search for natural bioactive compounds in Bolivia through a multidisciplinary approach: Part I. Evaluation of the antimalarial activity of plants used by the Chacobo Indians, J. Ethnopharmacol, vol.69, pp.127-137, 2000.

H. Ishiyama, M. Matsumoto, M. Sekiguchi, H. Shigemori, A. Ohsaki et al., Two new indole alkaloids from Aspidosperma subincanum and Geissospermum vellosii, vol.66, pp.651-658, 2005.

J. A. Lima, R. S. Costa, R. A. Epifânio, N. G. Castro, M. S. Rocha et al., Geissospermum vellosii stembark: anticholinesterase activity and improvement of scopolamine-induced memory deficits, Pharmacol. Biochem. Behav, vol.92, pp.508-513, 2009.

F. Mbeunkui, M. H. Grace, C. Lategan, P. J. Smith, I. Raskin et al., In vitro antiplasmodial activity of indole alkaloids from the stem bark of Geissospermum vellosii, J. Ethnopharmacol, vol.139, pp.471-477, 2012.

R. Baltenweck-guyot and R. Ocampo, Identification of a novel 7-desmethyl-7-acetonyl bacteriophaeophorbide c series in a recent sediment, Org. Geochem, vol.38, pp.1580-1584, 2007.

J. Monachino, A revision of the genus Alstonia (Apocynaceae), The plant list, vol.3, pp.19-28, 1949.

A. Vieillardii-van-heurck, , pp.18-27

. Alstonia-balansae-guillaumin, , pp.20-29

A. Quaternata-van-heurck, , pp.20-29

K. Sidiyasa and . Taxonomy, Blumea, vol.11, pp.1-230, 1998.

R. Alstonia, . Br, and . Secretariat, , pp.19-28

M. S. Khyade, D. M. Kasote, and N. P. Vaikos, Alstonia scholaris (L.) R. Br. and Alstonia macrophylla Wall. ex G. Don: A comparative review on traditional uses, phytochemistry and pharmacology, Journal of ethnopharmacology, vol.153, pp.1-18, 2014.

A. P. Macabeo, K. Krohn, D. Gehle, R. W. Read, J. J. Brophy et al., Indole alkaloids from the leaves of Philippine Alstonia scholaris, Phytochemistry, vol.66, pp.1158-1162, 2005.

N. Keawpradub, E. Eno-amooquaye, P. Burke, and P. Houghton, Cytotoxic activity of indole alkaloids from Alstonia macrophylla, Planta Med, vol.65, pp.311-315, 1999.

. Alstonia-balansae-guillaumin, , pp.18-27

T. Pinchon, J. Nuzillard, B. Richard, G. Massiot, L. Le-men-olivier et al., Alkaloids from Alstonia undulata, vol.29, pp.3341-3344, 1990.

D. Guillaume, A. Morfaux, B. Richard, G. Massiot, L. Le-men-olivier et al., Some alkaloids of Alstonia undulata, vol.23, pp.2407-2408, 1984.

J. Guillon, P. Grellier, M. Labaied, P. Sonnet, J. Léger et al., Synthesis, antimalarial activity, and molecular modeling of new pyrrolo

, J. Med. Chem, vol.1, 1997.

Z. Cheikh-ali, J. Caron, S. Cojean, C. Bories, P. Couvreur et al., Squalenoylcurcumin" nanoassemblies as water-dispersible drug candidates with antileishmanial activity, vol.10, pp.411-418, 2015.

M. Girardot, A. Gadea, C. Deregnaucourt, A. Deville, L. Dubost et al., Tabernaelegantinals: unprecedented cytotoxic bisindole alkaloids from Muntafara sessilifolia, Eur. J. Org. Chem, pp.2816-2823, 2012.