. Eurekasanté, Les interactions médicamenteuses

. Eurekasanté,

D. Sur,

L. Gueut and D. , Interactions médicamenteuses : mécanismes et analyses de cas

N. Mathieu, Interactions médicamenteuses : de la théorie à la réalité, UFR Sciences pharmaceutiques et biologiques, 2008.

H. Lulluman, K. Mohr, and L. Hein, Atlas de poche de pharmacologie

P. Jolliet and M. Bourin,

P. Allain, Les médicaments : Pharmacologie. 4 e éd

, ANSM. Interactions médicamenteuses

D. Sur,

P. Barriot and J. Montastruc, Médicaments et iatrogenèse : Le guide. Lavoisier Médecine sciences, vol.383, 2015.

, Collège National de Pharmacologie Médicale

. Pharmacomedicale and . Org, , 2018.

L. M. Strand, P. C. Morley, R. J. Cipolle, R. Ramsey, and G. D. Lamsam, Drug-Related Problems : Their Structure and Function, DICP Ann Pharmacother. 1 nov, vol.24, issue.11, pp.1093-1097, 1990.

Y. Koh, F. Kutty, and S. C. Li, Drug-related problems in hospitalized patients on polypharmacy: the influence of age and gender, Ther. Clin. Risk. Manag. mars, vol.1, issue.1, pp.39-48, 2005.

J. Ankri, Le risque iatrogène médicamenteux chez le sujet âgé, Gérontologie Société, vol.25, issue.103, p.198, 2002.

V. Bouvier and . Vidal, Base de données médicamenteuses pour les prescripteurs libéraux

. Vidal, Eviter les effets indésirables par interactions médicamenteuses, vol.33

C. Thériaque,

, ANSM. Thesaurus des interactions médicamenteuses, 2016.

F. Ranchon, C. Bouret, B. Charpiat, and G. Leboucher, Sécurisation de l'emploi des chimiothérapies anticancéreuses administrables par voie orale, 14 avr, vol.44, issue.1, p.44, 2009.

R. Van-leeuwen, D. Brundel, C. Neef, T. Van-gelder, R. Mathijssen et al., Prevalence of potential drug-drug interactions in cancer patients treated with oral anticancer drugs, Br. J. Cancer. 19 mars, vol.108, issue.5, pp.1071-1078, 2013.

A. Bulsink, A. Imholz, J. Brouwers, and F. Jansman, Characteristics of potential drugrelated problems among oncology patients, Int. J. Clin. Pharm. juin, vol.35, issue.3, pp.401-407, 2013.

V. Escudero-vilaplana, A. Ribed, R. M. Romero-jimenez, A. Herranz-alonso, and M. Sanjurjo-saez, Pharmacotherapy follow-up of key points in the safety of oral antineoplastic agents, Eur. J. Cancer Care. (Engl). mai, vol.26, issue.3, 2017.

A. Ribed, R. M. Romero-jiménez, V. Escudero-vilaplana, I. Iglesias-peinado, A. Herranzalonso et al., Pharmaceutical care program for onco-hematologic outpatients : safety, efficiency and patient satisfaction, Int. J. Clin. Pharm. avr, vol.38, issue.2, pp.280-288, 2016.

B. Venezia and Y. , Les thérapies ciblées orales, CAHPP, 2016.

D. Sur,

, Développement des anticancéreux oraux -Projections à court, moyen et long termes

, Les thérapies ciblées dans le traitement du cancer en 2015 -État des lieux et enjeux, 2016.

N. Chaumard-billotey, Prise en charge par les anticancéreux oraux à domicile, 2016.

D. Sur,

J. Soria, S. Vignot, C. Massard, and O. Mir, Cours de chimiothérapie antitumorale et traitement médical du cancer

V. Richard, Anticancéreux : Les points essentiels

. Pharmacomedicale and . Org, , 2017.

D. Sur,

S. Faure, Thérapies ciblées anticancéreuses, Actual. Pharm. 1 mai, vol.54, issue.546, pp.57-61, 2015.

S. R. Hubbard and J. H. Till, Protein tyrosine kinase structure and function, Annu. Rev. Biochem, vol.69, pp.373-398, 2000.

P. Blume-jensen and T. Hunter, Oncogenic kinase signalling, Nature. 17 mai, issue.411, pp.355-365, 2001.

A. Laurenty, J. Selves, and R. Guimbaud, Association française de Formation Médicale Continue en Hépato-Gastro-Entérologie

D. Hanahan and R. A. Weinberg, The Hallmarks of, Cancer. Cell. Press. 7 janv, vol.100, issue.1, pp.57-70, 2000.

S. Cohen, R. C. Levi-montalcini, and . Comments, Essays of an Information Scientist, 27 avr, vol.10, issue.17, p.106, 1987.

P. Hubert, Les facteurs de croissance de la famille de l'EGF et leurs récepteurs, Bull. Cancer. avr, vol.93, issue.7, pp.1-8, 2006.

C. L. Arteaga, The epidermal growth factor receptor : from mutant oncogene in nonhuman cancers to therapeutic target in human neoplasia, J. Clin. Oncol. 15 sept, vol.19, pp.32-40, 2001.

L. Couderc, J. Dalaine, Y. Duchartre, K. Makki, and P. Violet, Les récepteurs ERBB et leurs voies de signalisation, Cell. Biology Promotion, 2007.

D. Sur,

C. Larbouret, N. Gaborit, M. Poul, A. Pèlegrin, and T. Chardès, Le récepteur HER3 ou ERBB3 -La face cachée de la planète ERBB, Med. Sci. 1 mai, vol.31, issue.5, pp.465-468, 2015.

A. M. Petit, J. Rak, M. C. Hung, P. Rockwell, N. Goldstein et al., Neutralizing antibodies against epidermal growth factor and ErbB-2/neu receptor tyrosine kinases down-regulate vascular endothelial growth factor production by tumor cells in vitro and in vivo: angiogenic implications for signal transduction therapy of solid tumors, Am. J. Pathol. déc, vol.151, issue.6, pp.1523-1530, 1997.

C. J. Wikstrand and D. D. Bigner, Prognostic applications of the Epidermal Growth Factor Receptor and its ligand, Transforming Growth Factor-alpha, J. Natl. Cancer. Inst, vol.90, issue.11, pp.799-801, 1998.

T. Valentin, D. Bonnet, and R. Guimbaud, Thérapeutiques ciblées « pour les nuls » : modes d'action des anti-EGFR et des anti-VEGF, Hépato-Gastro & Oncol. Dig. 1 sept, vol.18, issue.1, pp.4-16, 2011.

L. M. Ellis and D. J. Hicklin, VEGF-targeted therapy: mechanisms of anti-tumour activity, Nat. Rev. Cancer. août, vol.8, issue.8, pp.579-591, 2008.

M. Shibuya, Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) Signaling in Angiogenesis, Genes & Cancer. déc, vol.2, issue.12, pp.1097-1105, 2011.

S. Shinkaruk, M. Bayle, G. Laïn, and G. Déléris, Vascular endothelial cell growth factor (VEGF), an emerging target for cancer chemotherapy, Curr. Med. Chem. mars, vol.3, issue.2, pp.95-117, 2003.

, mg gél -Indications, Vidal. SUTENT, vol.12

D. Sur,

J. Demoulin and A. Essaghir, PDGF receptor signaling networks in normal and cancer cells, Cytokine & Growth Factor Rev. juin, vol.25, issue.3, pp.273-283, 2014.

S. K. Denduluri, O. Idowu, Z. Wang, Z. Liao, Z. Yan et al., Insulin-like Growth Factor (IGF) signaling in tumorigenesis and the development of cancer drug resistance, Genes & Diseases. 1 mars, vol.2, issue.1, pp.13-25, 2015.

I. Ahmad, T. Iwata, and H. Y. Leung, Mechanisms of FGFR-mediated carcinogenesis, Biochim. Biophys. Acta. avr, vol.1823, issue.4, pp.850-860, 2012.

S. Gisselbrecht, Les récepteurs de cytokines. Hématologie, vol.9, pp.477-483, 2003.

D. Wachsmann, P. Soulas-sprauel, P. Miossec, and D. Webdling, Les communications intercellulaires : les cytokines et leur réseau

L. Guglielmo, Les cytokines et leurs récepteurs

S. A. Warren, Suppressors Of Cytokine Signalling (SOCS) in the immune system, Nat. Rev. Immunol. juin, vol.2, issue.6, pp.410-416, 2002.

K. Shuai and B. Liu, Regulation of JAK-STAT signalling in the immune system, Nat. Rev. Immunol. nov, vol.3, issue.11, pp.900-911, 2003.

A. Sears, R. Ras, and M. , Co-conspirators in Cancer, 2017.

E. Beurel, S. F. Grieco, and R. S. Jope, Glycogen synthase kinase-3 (GSK3) : regulation, actions and diseases, Pharmacol. Ther. avr, vol.148, pp.114-131, 2015.

S. Misale, R. Yaeger, S. Hobor, E. Scala, M. Janakiraman et al., Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer, Nature. 28 juin, vol.486, issue.7404, pp.532-536, 2012.

C. Dreyer, E. Raymond, and S. Faivre, La voie de signalisation PI3K/AKT/mTOR, Cancéro. Dig. sept, vol.1, issue.3, pp.187-189, 2009.

A. Majchrzak, M. Witkowska, and P. Smolewski, Inhibition of the PI3K/Akt/mTOR signaling pathway in diffuse large B-cell lymphoma : current knowledge and clinical significance, Mol. Basel. Switz. 11 sept, vol.19, issue.9, pp.14304-14315, 2014.

J. Cornillon, L. Campos, and D. Guyotat, Focal adhesion kinase (FAK), une protéine aux fonctions multiples, Med. Sci. 1 juin, vol.19, issue.6-7, pp.743-752, 2003.

V. Kölsch, P. G. Charest, and R. A. Firtel, The regulation of cell motility and chemotaxis by phospholipid signaling, J. Cell. Sci. 1 mars, vol.121, issue.5, pp.551-559, 2008.

E. Jaspard, Biochimie régulation métabolique, 2011.

H. Cave, A. Campos, and A. Jenvrin, Signalisation : mécanismes moléculaires de la transduction des signaux, 2012.

A. Jabrani, Régulation de la voie Hedgehog : étude structurale et fonctionnelle de protéines de signalisation, 2012.

S. Gupta, N. Takebe, and P. Lorusso, Targeting the Hedgehog pathway in cancer, Ther. Adv. Med. Oncol. juill, vol.2, issue.4, pp.237-250, 2010.

H. Larue, M. Simoneau, T. Aboulkassim, P. Lemieux, J. Girard et al., La voie de signalisation PATCHED/Sonic Hedgehog dans le cancer superficiel de la vessie, Med. Sci. oct, vol.19, issue.10, pp.920-925, 2003.

F. Malard and M. Mohty, Inhibiteurs du protéasome dans le myélome multiple. Hématologie, vol.21, pp.326-332, 2015.

J. Rios and S. Puhalla, PARP Inhibitors in Breast Cancer : BRCA and beyond, Oncol. J. oct, vol.25, issue.11, pp.1014-1025, 2011.

J. Ricci, Rôle de la tyrosine kinase p59Fyn et des protéines du choc thermique, dans l'apoptose induite par l'engagement des récepteurs de mort cellulaire et du récepteur T, 2000.

M. Jäättelä, Escaping cell death : survival proteins in cancer, Exp. Cell. Res. 10 avr, vol.248, issue.1, pp.30-43, 1999.

A. Arrigo, Chaperons moléculaires et repliement des protéines : l'exemple de certaines protéines de choc thermique, Med. Sci. 1 juin, vol.21, issue.6-7, pp.619-625, 2005.

D. Kerboeuf, L. Vern, and Y. , , p.16, 2018.

D. Sur,

H. Zou, Y. Li, X. Liu, and X. Wang, An APAF-1·Cytochrome C : Multimeric complex is a functional apoptosome that activates Procaspase-9, J. Biol. Chem. 23 avr, 1999.

G. Liu, E. Franssen, M. I. Fitch, and E. Warner, Patient preferences for oral versus intravenous palliative chemotherapy, J. Clin. Oncol. janv, vol.15, issue.1, pp.110-115, 1997.

L. Fallowfield, L. Atkins, S. Catt, A. Cox, C. Coxon et al., Patients' preference for administration of endocrine treatments by injection or tablets : results from a study of women with breast cancer, Ann. of Oncol. févr, vol.17, issue.2, pp.205-210, 2006.

A. Cuquel and S. Loreznzo, Mise en place d'une consultation pharmaceutique pour les patients sous chimiothérapie par voie orale : intérêts et premiers résultats

F. Binder-foucard, A. Belot, P. Delafosse, L. Remontet, A. Woronoff et al., Estimation nationale de l'incidence et de la mortalité par cancer en France entre, 1980.

, Institut National Du Cancer. La chimiothérapie orale du cancer en, 2014.

, Adherence to long-therm therapies : evidence for action

D. Sur,

T. I. Barron, R. Connolly, K. Bennett, J. Feely, and M. J. Kennedy, Early discontinuation of tamoxifen : a lesson for oncologists, Cancer. 1 mars, vol.109, issue.5, pp.832-839, 2007.

K. Ruddy, E. Mayer, and A. Partridge, Patient adherence and persistence with oral anticancer treatment, Cancer J. Clin. févr, vol.59, issue.1, pp.56-66, 2009.

M. R. Dimatteo, Variations in patients' adherence to medical recommendations : a quantitative review of 50 years of research, Med. Care. mars, vol.42, issue.3, pp.200-209, 2004.

V. Regnier-denois, J. Poirson, A. Nourissat, J. Jacquin, J. P. Guastalla et al., Adherence with oral chemotherapy : results from a qualitative study of the behaviour and representations of patients and oncologists, Eur. J. Cancer Care. juill, vol.20, issue.4, pp.520-527, 2011.

D. Levêque, C. Duval, C. Poulat, B. Palas, A. El-aatmani et al., Mécanismes des interactions pharmacocinétiques impliquant les agents anticancéreux oraux, Bull. Cancer. 1 janv, vol.102, issue.1, pp.65-72, 2015.

N. Widmer, C. Bardin, E. Chatelut, A. Paci, J. Beijnen et al., Review of therapeutic drug monitoring of anticancer drugs part two targeted therapies, Eur. J. Cancer. août, vol.50, issue.12, pp.2020-2036, 2014.

B. E. Houk, C. L. Bello, B. Poland, L. S. Rosen, G. D. Demetri et al., Relationship between exposure to sunitinib and efficacy and tolerability endpoints in patients with cancer : results of a pharmacokinetic/pharmacodynamic meta-analysis, Cancer Chemother. Pharmacol. juill, vol.66, issue.2, pp.357-371, 2010.

M. Von-mehren and N. Widmer, Correlations between imatinib pharmacokinetics, pharmacodynamics, adherence, and clinical response in advanced metastatic gastrointestinal stromal tumor (GIST) : an emerging role for drug blood level testing?, Cancer Treat. Rev. juin, vol.37, issue.4, pp.291-299, 2011.

C. Massard, J. Patard, O. Hermine, and A. Ravaud, Management of side effects of targeted therapies in renal cancer iatrogenic side effects, Bull. Cancer, issue.98, pp.79-94, 2011.

T. Eisen, C. N. Sternberg, C. Robert, P. Mulders, L. Pyle et al., Targeted therapies for renal cell carcinoma : review of adverse event management strategies, J. Natl. Cancer Inst. 18 janv, vol.104, issue.2, pp.93-113, 2012.

T. E. Hutson, J. Bellmunt, C. Porta, C. Szczylik, M. Staehler et al., Long-term safety of sorafenib in advanced renal cell carcinoma : follow-up of patients from phase III TARGET, Eur. J. Cancer. sept, vol.46, issue.13, pp.2432-2440, 2010.

R. J. Motzer, T. E. Hutson, P. Tomczak, M. D. Michaelson, R. M. Bukowski et al., Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma, J. Clin. Oncol. 1 août, vol.27, issue.22, pp.3584-3590, 2009.

R. J. Motzer, T. E. Hutson, P. Tomczak, M. D. Michaelson, R. M. Bukowski et al., Sunitinib versus interferon alfa in metastatic renal-cell carcinoma, N. Engl. J Med. 11 janv, vol.356, issue.2, pp.115-124, 2007.

J. Roigas, Clinical Management of Patients Receiving Tyrosine Kinase Inhibitors for Advanced Renal Cell Carcinoma, Eur. Urol. Suppl. 1 sept, vol.7, issue.9, pp.593-600, 2008.

M. E. Lacouture, L. M. Reilly, P. Gerami, and J. Guitart, Hand foot skin reaction in cancer patients treated with the multikinase inhibitors sorafenib and sunitinib, Ann. Oncol. nov, vol.19, issue.11, pp.1955-1961, 2008.

C. R. King, Multitargeted agents for therapeutically challenging tumor : an introduction for oncology nurses, Oncol. Nurs. Soc. News, vol.21, issue.8, pp.57-58, 2006.

K. Edmonds, D. Hull, A. Spencer-shaw, J. Koldenhof, M. Chrysou et al., Strategies for assessing and managing the adverse events of sorafenib and other targeted therapies in the treatment of renal cell and hepatocellular carcinoma : recommendations from a European nursing task group, Eur. J. Oncol. Nurs. avr, vol.16, issue.2, pp.172-184, 2012.

B. E. O'brien, V. G. Kaklamani, and A. B. Benson, The assessment and management of cancer treatment-related diarrhea, Clin Colorectal Cancer. mars, vol.4, issue.6, pp.375-381, 2005.

I. M. Andrew, K. Waterfield, A. J. Hildreth, G. Kirkpatrick, and C. Hawkins, Quantifying the impact of standardized assessment and symptom management tools on symptoms associated with cancer-induced anorexia cachexia syndrome, Palliat. Med. déc, vol.23, issue.8, pp.680-688, 2009.

B. Kaplan, Y. Qazi, and J. R. Wellen, Strategies for the management of adverse events associated with mTOR inhibitors, Transplant Rev. juill, vol.28, issue.3, pp.126-133, 2014.

M. Zangari, L. M. Fink, F. Elice, F. Zhan, D. M. Adcock et al., Thrombotic events in patients with cancer receiving antiangiogenesis agents, J. Clin. Oncol, vol.27, issue.29, pp.4865-4873, 2009.

D. A. White, P. Camus, M. Endo, B. Escudier, E. Calvo et al., Noninfectious pneumonitis after everolimus therapy for advanced cancer, Am. J. Respir. Crit. Care Med. 1 août, vol.182, issue.3, pp.396-403, 2010.

C. Porta, S. Osanto, A. Ravaud, M. Climent, U. Vaishampayan et al., Management of adverse events associated with the use of everolimus in patients with cancer, Eur. J. Cancer. juin, vol.47, issue.9, pp.1287-1298, 2011.

L. Hammond, E. Marsden, N. O'hanlon, F. King, M. C. Henman et al., Identification of risks associated with the prescribing and dispensing of oral anticancer medicines in Ireland, Int. J. Clin. Pharm. déc, vol.34, issue.6, pp.893-901, 2012.

V. Kruse, A. Somers, L. Van-bortel, D. Both, A. Van-belle et al., Sunitinib for metastatic renal cell cancer patients : observational study highlighting the risk of important drug-drug interactions, J. Clin. Pharm. Ther. juin, vol.39, issue.3, pp.259-265, 2014.

A. Thomas-schoemann, B. Blanchet, C. Bardin, G. Noé, P. Boudou-rouquette et al., Drug interactions with solid tumour-targeted therapies, Crit. Rev. Oncol. Hematol. 1 janv, vol.89, issue.1, pp.179-196, 2014.

H. Klümpen, C. F. Samer, R. Mathijssen, J. Schellens, and H. Gurney, Moving towards dose individualization of tyrosine kinase inhibitors, Cancer Treat. Rev. juin, vol.37, issue.4, pp.251-260, 2011.

N. R. Budha, A. Frymoyer, G. S. Smelick, J. Y. Jin, M. R. Yago et al., Drug absorption interactions between oral targeted anticancer agents and PPIs : is pHdependent solubility the Achilles heel of targeted therapy ?, Clin. Pharmacol. Ther. août, vol.92, issue.2, pp.203-213, 2012.

J. Zürcher, G. Waeber, J. Schlaepfer, M. Pasquier, and . Le, Rev. Médicale Suisse, vol.9, issue.395, pp.1538-1542, 2013.

. Shah-r-r, J. Morganroth, and D. Shah, Cardiovascular safety of tyrosine kinase inhibitors : with a special focus on cardiac repolarisation (QT interval), Drug Saf. mai, vol.36, issue.5, pp.295-316, 2013.

D. M. Roden, R. L. Woosley, and R. K. Primm, Incidence and clinical features of the quinidine-associated long QT syndrome : implications for patient care, Am. Heart J. juin, vol.111, issue.6, pp.1088-1093, 1986.

E. Ernst and B. R. Cassileth, The prevalence of complementary/alternative medicine in cancer : a systematic review, Cancer. 15 août, vol.83, issue.4, pp.777-782, 1998.

L. Simon, D. Prebay, A. Beretz, J. L. Bagot, A. Lobstein et al., Complementary and alternative medicines taken by cancer patients, Bull. Cancer. mai, vol.94, issue.5, pp.483-488, 2007.

A. Girard, La place du pharmacien dans le conseil en phytothérapie, Université de bordeaux. UFR des Sciences pharmaceutiques, 2018.

A. Molassiotis, P. Fernadez-ortega, D. Pud, G. Ozden, J. A. Scott et al., Use of complementary and alternative medicine in cancer patients : a European survey, Ann. Oncol. avr, vol.16, issue.4, pp.655-663, 2005.

S. Gautier, J. Béné, M. Auffret, and J. Caron, Interactions entre pamplemousse et médicaments, Phytothérapie. 1 avr, vol.12, issue.2, pp.98-104, 2014.

I. Meijerman, J. H. Beijnen, and J. Schellens, Herb-drug interactions in oncology : focus on mechanisms of induction, The Oncologist. août, vol.11, issue.7, pp.742-752, 2006.

O. Yin, N. Gallagher, A. Li, W. Zhou, R. Harrell et al., Effect of grapefruit juice on the pharmacokinetics of nilotinib in healthy participants, J. Clin. Pharmacol. févr, vol.50, issue.2, pp.188-194, 2010.

F. Petitet, Interactions pharmacocinétiques entre préparation à base de plantes et médicament : une revue de l'importance clinique, Phytothérapie. 1 juin, vol.10, issue.3, pp.170-182, 2012.
DOI : 10.1007/s10298-012-0705-2

M. Huet, Les plantes médicinales chez les malades atteints de cancers : pratiques courantes et éléments de leur évaluation, Bull. Cancer. mai, vol.100, issue.5, pp.485-495, 2013.

A. Goey, K. D. Mooiman, J. H. Beijnen, J. Schellens, and I. Meijerman, Relevance of in vitro and clinical data for predicting CYP3A4-mediated herb-drug interactions in cancer patients, Cancer Treat. Rev. nov, vol.39, issue.7, pp.773-783, 2013.

, Plan cancer 2014-2019 : priorités et objectifs

L. Foroni, Présentation de l'OMéDIT Auvergne-Rhône-Alpes, 2018.

D. Sur,

. Unicancer, Quelle prise en charge des cancers en 2020 ?

D. Sur,

H. Autorité-de-santé, Guide méthodologique : recommandations de l'éducation thérapeutique du patient, définition, finalités et organisation

. Page-|-134,

D. Sur,

H. Autorité-de-santé, Structuration d'un programme d'éducation thérapeutique du patient dans le champ des maladies chroniques

D. Sur,

, Arrêté du 14 janvier 2015 relatif au cahier des charges des programmes d'éducation thérapeutique du patient et à la composition du dossier de demande de leur autorisation et de leur renouvellement et modifiant l'arrêté du 2 août 2010 modifié relatif aux compétences requises pour dispenser ou coordonner l'éducation thérapeutique du patient, La ministre des affaires sociales, de la santé et des droits des femmes

H. Autorité-de-santé, Mise en oeuvre de l'éducation thérapeutique

D. Sur,

. Assemblée-nationale and . Sénat, LOI n° 2016-41 du 26 janvier 2016 de modernisation de notre système de santé, 2016.

J. Johnson, The effects of a patient education course on persons with a chronic illness, Cancer Nurs. avr, vol.5, issue.2, p.117, 1982.

J. M. Diekmann, An Evaluation of selected "I Can Cope" programs by registered participants. Cancer Nurs, vol.11, pp.274-282, 1988.

K. Todd, S. Roberts, and C. Black, The Living with Cancer Education Programme : development of an Australian education and support programme for cancer patients and their family and friends, Eur. J. Cancer Care. déc, vol.11, issue.4, pp.271-279, 2002.

G. Grahn, M. Danielson, and K. Ulander, Learning to live with cancer in European countries, Cancer Nurs. févr, vol.22, issue.1, pp.79-84, 1999.

M. Bruchon-schweitzer, Le coping et les stratégies d'ajustement face au stress, Concepts Stress Coping. déc, issue.67, pp.68-83, 2001.

G. Grahn, Coping with the cancer experience : developing an education and support programme for cancer patients and their significant others, Eur. J. Cancer Care, vol.5, issue.3, pp.176-181

C. Lopez-martin, G. Siles, M. , A. , J. et al., Role of clinical pharmacists to prevent drug interactions in cancer outpatients : a single-centre experience, Int. J. Clin. Pharm. déc, vol.36, issue.6, pp.1251-1259, 2014.

H. Iihara, M. Ishihara, K. Matsuura, S. Kurahashi, T. Takahashi et al., Pharmacists contribute to the improved efficiency of medical practices in the outpatient cancer chemotherapy clinic, J. Eval. Clin. Pract. août, vol.18, issue.4, pp.753-760, 2012.

J. Valgus, S. Jarr, R. Schwartz, M. Rice, and S. A. Bernard, Pharmacist-led, interdisciplinary model for delivery of supportive care in the ambulatory cancer clinic setting, J. Oncol. Pract, vol.6, issue.6, pp.1-4, 2010.

T. T. Yeoh, X. Y. Tay, P. Si, and L. Chew, Drug-related problems in elderly patients with cancer receiving outpatient chemotherapy, J. Geriatr. Oncol. juill, vol.6, issue.4, pp.280-287, 2015.

G. Nightingale, E. Hajjar, K. Swartz, J. Andrel-sendecki, and A. Chapman, Evaluation of a pharmacist-led medication assessment used to identify prevalence of and associations with polypharmacy and potentially inappropriate medication use among ambulatory senior adults with cancer, J. Clin. Oncol. 1 mai, vol.33, issue.13, pp.1453-1459, 2015.

S. Benguigui, C. Bardin, C. Borja-prats, O. Conort, M. Daouphars et al., Chimiothérapies orales en officine : Quelles sont les attentes du patient ? Quels sont les besoins des pharmaciens ? Comment évoluer ? 10 èmes Journées de la Société Française de Pharmacie Oncologique, 201514.

J. Aisner, Overview of the changing paradigm in cancer treatment : oral chemotherapy, Am. J. Health Syst. Pharm. 1 mai, vol.64, issue.9, pp.4-7, 2007.

J. Périchou, F. Ranchon, I. Carpentier, V. Schwiertz, N. Vantard et al., Cancer patients' medication knowledge : what about oral immunomodulatory drugs to treat myeloma? 10 èmes Journées de la Société Française de Pharmacie Oncologique, 201514.

M. L. Voll, K. D. Yap, W. E. Terpstra, and M. Crul, Potential drug-drug interactions between anti-cancer agents and community pharmacy dispensed drugs, Pharm. World Sci, vol.32, issue.5, pp.575-580, 2010.
DOI : 10.1007/s11096-010-9410-0

V. Schwiertz, C. Bertin, A. Henry, and B. Charpiat, Estimation du nombre et de la nature des interactions médicamenteuses concernant les médicaments anticancéreux, Bull. Cancer. mai, vol.94, issue.5, pp.477-482, 2007.

G. Grahn and J. Johnson, Learning to cope and living with cancer. Learning-needs assessment in cancer patient education, Scand. J. Caring Sci, vol.4, issue.4, pp.173-181, 1990.

K. Todd, S. Roberts, and C. Black, The Living with Cancer Education Programme