Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
1.
Cancers (Basel) ; 16(1)2023 Dec 20.
Article de Anglais | MEDLINE | ID: mdl-38201470

RÉSUMÉ

OBJECTIVE: While PLD-Trabectedin is an approved treatment for relapsed platinum-sensitive ovarian cancer, its efficacy and tolerability has so far not been tested extensively in patients who progress after poly ADP-ribose polymerase inhibitor (PARPi) treatment. METHODOLOGY: This multicenter, retrospective analysis had the objective of comparing patients receiving PLD-Trabectedin after being treated with PARP-I (cases) with PARPi-naïve patients. Descriptive and survival analyses were performed for each group. RESULTS: Data from 166 patients were collected, composed of 109 cases and 57 controls. In total, 135 patients were included in our analyses, composing 46 controls and 89 cases. The median PFS was 11 months (95% IC 10-12) in the control group vs. 8 months (95% IC 6-9) in the case group (p value 0.0017). The clinical benefit rate was evaluated, with an HR for progression of 2.55 (1.28-5.06) for the case group (p value 0.008), persisting when adjusted for BRCA and line with treatment. We compared hematological toxicity, gastro-intestinal toxicity, hand-foot syndrome (HFS), fatigue, and liver toxicity, and no statistically significant disparity was noted, except for HFS with a p value of 0.006. The distribution of G3 and G4 toxicities was also equally represented. CONCLUSION: The MITO39 study showed a statistically significant difference in terms of PFS, suggesting that previous exposure to PARPi might inhibit the efficacy of PLD-Trabectedin. Regarding tolerability, no remarkable disparity was noted; PLD-Trabectedin was confirmed to be a well-tolerated scheme in both groups. To our knowledge, these are the first data regarding this topic, which we deem to be of great relevance in the current landscape.

2.
Front Oncol ; 12: 960387, 2022.
Article de Anglais | MEDLINE | ID: mdl-36249001

RÉSUMÉ

Background: A virtual reality experience (VRE) could represent a viable non-pharmacological intervention to reduce and better manage the main factors of psychophysical distress related to the diagnosis and treatment of cancer. Aim: The "Patient's Dream" study was a two-arm randomized controlled trial conducted at the Regina Elena National Cancer Institute - IRCCS (Rome, Italy) from April 2019 to January 2020 to evaluate VRE impact in patients affected by breast or ovarian cancer. Before starting the first cycle of chemotherapy (CT), patients were randomized to receive the VRE (VRE arm) as "distraction therapy" or to entertain themselves with conventional means (control arm). The primary aims were the assessment of psychological distress, anxiety and quality of life between the two study arms. Secondary endpoints were the perceived time during the first course of CT and the acute and late toxicity. Results: Fourty-four patients were enrolled, 22 patients were randomly assigned to the VRE arm and 22 to the control arm. Collected data underline the absence of prevalent disturbs of anxiety and depression in both groups. Nevertheless, even if the state anxiety values before and after CT decreased in both groups, this reduction was statistically significant over time only in the VRE arm. The duration of therapy perceived by patients undergoing distraction therapy was significantly shorter when compared to the control group. The use of VRE during the first CT cycle appeared to reduce asthenia outcomes. Conclusion: Obtained data suggest that the VRE positively influenced the levels of state anxiety among cancer patients and support the continuous research on VRE as a distraction intervention, with the aim to meet the clinical need for effective nonpharmacologic adjunctive therapies. Clinical trial registration: https://clinicaltrials.gov/ct2/show/NCT05234996, identifier NCT05234996.

3.
Int J Mol Sci ; 21(22)2020 Nov 10.
Article de Anglais | MEDLINE | ID: mdl-33182542

RÉSUMÉ

Breast cancer was one of the first malignancies to benefit from targeted therapy, i.e., treatments directed against specific markers. Inhibitors against HER2 are a significant example and they improved the life expectancy of a large cohort of patients. Research on new biomarkers, therefore, is always current and important. AXL, a member of the TYRO-3, AXL and MER (TAM) subfamily, is, today, considered a predictive and prognostic biomarker in many tumor contexts, primarily breast cancer. Its oncogenic implications make it an ideal target for the development of new pharmacological agents; moreover, its recent role as immune-modulator makes AXL particularly attractive to researchers involved in the study of interactions between cancer and the tumor microenvironment (TME). All these peculiarities characterize AXL as compared to other members of the TAM family. In this review, we will illustrate the biological role played by AXL in breast tumor cells, highlighting its molecular and biological features, its involvement in tumor progression and its implication as a target in ongoing clinical trials.


Sujet(s)
Tumeurs du sein/physiopathologie , Protéines proto-oncogènes/physiologie , Récepteurs à activité tyrosine kinase/physiologie , Antinéoplasiques/usage thérapeutique , Marqueurs biologiques tumoraux/antagonistes et inhibiteurs , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/physiologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Mouvement cellulaire/génétique , Mouvement cellulaire/physiologie , Résistance aux médicaments antinéoplasiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/génétique , Transition épithélio-mésenchymateuse/physiologie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Protéines et peptides de signalisation intercellulaire/composition chimique , Protéines et peptides de signalisation intercellulaire/génétique , Protéines et peptides de signalisation intercellulaire/physiologie , Thérapie moléculaire ciblée/méthodes , Invasion tumorale/génétique , Invasion tumorale/physiopathologie , Inhibiteurs de protéines kinases/usage thérapeutique , Maturation post-traductionnelle des protéines , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/génétique , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Récepteurs à activité tyrosine kinase/génétique , Microenvironnement tumoral/génétique , Microenvironnement tumoral/physiologie , Axl Receptor Tyrosine Kinase
4.
Clin Breast Cancer ; 20(6): e761-e770, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-32580907

RÉSUMÉ

INTRODUCTION: p53 and antiapoptotic B-cell leukemia/lymphoma 2 (BLC2) have been proposed as prognostic markers for early breast cancer (BC), although their relationship with conventional parameters and patient prognosis, as well as their distribution within the molecular BC subtypes remains uncertain. PATIENTS AND METHODS: In this observational study, we analyzed the immunohistochemical expression of p53 and BLC2 in 1099 early BC patients surgically treated between 2000 and 2006 and followed for at least 5 years, also considering their association with pathologic factors and molecular subtypes, as well as their influence on disease-free survival. RESULTS: p53 and BLC2 are distributed differently across molecular subtypes (P < .0001); in particular, p53 positivity and BLC2 negativity seems to be associated with more aggressive conventional tumor phenotypes. Moreover, BLC2 negativity seems to be a significant discriminating factor for disease-free survival (P = .003) according to Kaplan-Meier analysis, while p53 seems to have no discriminating effect. Among patients with discordant p53/BLC2 phenotype, the combination p53+BLC2- seems to be associated with the worst outcomes (P = .007) and significantly influenced the clinical course of node-negative patients treated only with hormone therapy (P = .004). CONCLUSION: These two biomarkers, in addition to conventional pathologic factors and molecular subtype, could help define the risk and outcome of BC.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Tumeurs du sein/mortalité , Récidive tumorale locale/épidémiologie , Protéines proto-oncogènes c-bcl-2/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antinéoplasiques hormonaux/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Marqueurs biologiques tumoraux/analyse , Région mammaire/anatomopathologie , Région mammaire/chirurgie , Tumeurs du sein/diagnostic , Tumeurs du sein/anatomopathologie , Tumeurs du sein/thérapie , Traitement médicamenteux adjuvant/méthodes , Survie sans rechute , Femelle , Études de suivi , Humains , Immunohistochimie , Estimation de Kaplan-Meier , Mastectomie , Adulte d'âge moyen , Récidive tumorale locale/anatomopathologie , Pronostic , Protéines proto-oncogènes c-bcl-2/analyse , Récepteurs à la progestérone , Protéine p53 suppresseur de tumeur/analyse , Jeune adulte
5.
Future Oncol ; 16(22): 1629-1637, 2020 Aug.
Article de Anglais | MEDLINE | ID: mdl-32501121

RÉSUMÉ

Aim: To investigate the toxicity of nab-paclitaxel (wNP)/nonpegylated liposome-encapsulated doxorubicin (wNPLD) combination in HER2-negative metastatic breast cancer (MBC) patients as first-line treatment. Materials & methods: Phase I, single-arm study in metastatic breast cancer patients naive to previous chemotherapy for advanced disease. A 3 + 3 dose-escalation design was used to determine the safety. Primary endpoints were the identification of dose-limiting toxicity and maximum tolerated dose. Results: In total, 12 patients (mean age: 52 years; median metastatic sites: 2) were enrolled and 97 cycles were completed. Maximum tolerated dose was wNP + wNPLD 25 mg/m2. The most common adverse events were neutropenia, nausea, diarrhea and mucositis. The objective response rate was 68% (response mean duration: 12.6 months). Conclusion: wNP/wNPLD combination constitutes an active regimen with mild toxicity.


Sujet(s)
Albumines/administration et posologie , Tumeurs du sein/traitement médicamenteux , Doxorubicine/analogues et dérivés , Paclitaxel/administration et posologie , Récepteur ErbB-2/analyse , Adulte , Sujet âgé , Albumines/effets indésirables , Tumeurs du sein/anatomopathologie , Doxorubicine/administration et posologie , Doxorubicine/effets indésirables , Femelle , Humains , Dose maximale tolérée , Adulte d'âge moyen , Métastase tumorale , Paclitaxel/effets indésirables , Polyéthylène glycols/administration et posologie , Polyéthylène glycols/effets indésirables
6.
Breast ; 41: 137-143, 2018 Oct.
Article de Anglais | MEDLINE | ID: mdl-30092500

RÉSUMÉ

BACKGROUND: We reported the results of an Italian large retrospective analysis that evaluated the effectiveness and safety of T-DM1 in 'field-practice' breast cancer patients. We performed a sub-analysis to investigate the clinical activity of T-DM1 in patients with brain metastases (BMs). METHODS: The records of 87 adult women with HER2-positive breast cancer and BMs treated with T-DM1 were reviewed. Their clinical outcomes were compared with those of 216 patients without central nervous system (CNS) involvement. RESULTS: Response to T-DM1 treatment in BMs was available for 53 patients in the BM group (60.9%): two patients reported a complete response (3.8%), 11 patients obtained partial response (20.7%; overall response rate: 24.5%), 16 patients had a stable disease (30.1%). Regarding extracranial disease, a total of 77 and 191 patients were evaluable for response in BM group and non-BM group, respectively. The overall response rate was 35.1% in the BM group and 38.3% in the non-BM group; disease control rate was 53.3% and 66.6%, respectively. At a median follow-up of 16 months (range: 1-55), median cumulative progression-free survival (PFS) was 7 months (95% CI: 5.4-8.6) in the BM group and 8 months (95% CI: 5.7-10.3) in the non-BM group. In the second-line setting, PFS was 5 (95% CI: 3.1-6.9) versus 11 (95% CI: 7.1-14.9) months (p = 0.01). Overall survival was 14 months (95% CI: 12.2-15.8) in the BM group and 32 months (95% CI: 24.4-39.6) in the non-BM group (p < 0.0001). CONCLUSIONS: T-DM1 is active in breast cancer patients with BMs.


Sujet(s)
Antinéoplasiques immunologiques/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du sein/traitement médicamenteux , Maitansine/analogues et dérivés , Trastuzumab/usage thérapeutique , Ado-trastuzumab emtansine , Adulte , Sujet âgé , Tumeurs du cerveau/mortalité , Tumeurs du cerveau/secondaire , Tumeurs du sein/mortalité , Tumeurs du sein/anatomopathologie , Femelle , Humains , Maitansine/usage thérapeutique , Adulte d'âge moyen , Récepteur ErbB-2 , Études rétrospectives , Analyse de survie , Résultat thérapeutique
7.
Oncology ; 94 Suppl 1: 29-33, 2018.
Article de Anglais | MEDLINE | ID: mdl-30041177

RÉSUMÉ

BACKGROUND: Brain metastases develop in approximately 10-25% of patients with metastatic breast cancer (MBC) and are associated with a very poor prognosis. CASE REPORT: We report the case of a 40-year-old woman with MBC and associated lung, bone, liver, and brain metastases, who experienced a time to progression of several months with eribulin after whole-brain radiotherapy (WBRT), 2 lines of chemotherapy, and 1 line of hormonal therapy, maintaining a good toxicity profile. DISCUSSION: Eribulin, in association with local treatment such as WBRT, can be well tolerated and effective in achieving a long progression-free survival and a good control of brain metastases in patients with MBC who have received multiple lines of treatment. The vascular remodeling properties of eribulin, combined with brain radiotherapy, might facilitate the passage of eribulin across the blood brain barrier, improving brain response. CONCLUSION: Our anecdotal experience suggests that eribulin may have a potentially beneficial effect on brain metastases while maintaining a good systemic control of the disease in patients with MBC.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Tumeurs du cerveau/traitement médicamenteux , Encéphale/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Furanes/usage thérapeutique , Cétones/usage thérapeutique , Adulte , Encéphale/anatomopathologie , Tumeurs du cerveau/anatomopathologie , Tumeurs du sein/anatomopathologie , Survie sans rechute , Femelle , Humains
8.
J Exp Clin Cancer Res ; 35: 104, 2016 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-27357210

RÉSUMÉ

The discovery of human epidermal growth factor receptor 2 (HER2) and its role in the biology of breast cancer and the subsequent development of HER2-targeted therapies, have dramatically improved clinical outcomes for women with early-stage and advanced HER2-positive breast cancer (BC).HER-2 targeted therapies represent a major step forward in achieving the goal of delivering individualized targeted therapy for BC, and trastuzumab was the first anti-HER-2 strategy to be approved for treatment of HER-2 positive BC. This review discusses the treatment of metastatic HER2-positive BC and describes efficacy and safety of novel anti-HER2 target therapies in first-line metastatic settings and the future challenges include refining such treatments, reducing toxicity and simultaneously developing innovative therapies. Furthermore, combinations of trastuzumab and drugs targeting the downstream pathway are described.In the next future will be possible to use an ample armamentarium of combination therapies directed against HER2 and key signaling components integrated in the HER network. This approach will allow clinicians to tailor the management of the individual patient on the basis of tumor- specific biomarker profiles.There is an urgent need for prospective biomarker-driven trials to identify patients for whom targeting is cost-effective.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Récepteur ErbB-2/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Femelle , Humains , Métastase tumorale , Récepteur ErbB-2/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Trastuzumab/usage thérapeutique
10.
Drug Des Devel Ther ; 9: 6177-83, 2015.
Article de Anglais | MEDLINE | ID: mdl-26640370

RÉSUMÉ

BACKGROUND: There is a deep need to improve the care of metastatic breast cancer (MBC) patients, since even today it remains an incurable disease. Taxanes are considered the most effective cytotoxic drugs for the treatment of MBC, both in monotherapy and in combined schedules, but the need for synthetic solvents contributes to the severe toxicities and may have a negative impact on the efficacy. Nanoparticle albumin-bound paclitaxel (Nab-paclitaxel) is a colloidal suspension of paclitaxel and human serum albumin initially developed to avoid the toxicities associated with conventional taxanes. PATIENTS AND METHODS: The aim of this prospective, single-center open-label, noncomparative study was to evaluate the efficacy and safety of nab-paclitaxel in MBC patients pretreated with taxanes. The patients were treated with nab-paclitaxel as a single agent, 260 mg/m(2) on day 1 of each 3-week cycle or 125 mg/m(2) weekly. The primary endpoint was the overall response rate (ORR). Secondary objectives were duration of response, clinical benefit rate, progression-free survival (PFS), overall survival, and safety. RESULTS: A total of 42 patients (median age 48 years, median Eastern Cooperative Oncology Group performance status 0, triple-negative MBC 19%, all pretreated with a taxane-based therapy, mainly in advanced disease) were enrolled in the study. The ORR was 23.8%, including one complete response (2.4%) and nine partial responses (21.4%); the disease control rate was 50%. The median duration of response was 7.2 months. After a median follow-up of 9 months, the median PFS was 4.6 months. ORR and PFS were similar irrespective of the previous chemotherapy lines, metastatic sites, and biomolecular expression. Nab-paclitaxel was well tolerated, and the most frequent treatment-related toxicities were mild to moderate (grades 1-2). CONCLUSION: This real-life study shows that nab-paclitaxel has a significant antitumor activity and a manageable safety profile in patients pretreated with taxanes and experiencing a treatment failure after at least one line of chemotherapy.


Sujet(s)
Albumines/usage thérapeutique , Antinéoplasiques d'origine végétale/usage thérapeutique , Paclitaxel/usage thérapeutique , Tumeurs du sein triple-négatives/traitement médicamenteux , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Albumines/effets indésirables , Antinéoplasiques d'origine végétale/effets indésirables , Survie sans rechute , Femelle , Études de suivi , Humains , Adulte d'âge moyen , Nanoparticules , Métastase tumorale , Paclitaxel/effets indésirables , Études prospectives , Taux de survie , Taxoïdes/usage thérapeutique , Tumeurs du sein triple-négatives/anatomopathologie , Jeune adulte
11.
BMC Cancer ; 14: 947, 2014 Dec 13.
Article de Anglais | MEDLINE | ID: mdl-25495785

RÉSUMÉ

BACKGROUND: The aim of this multicenter, retrospective study was to evaluate the efficacy and safety of metronomic oral cyclophosphamide (MOC) in heavily treated, relapsed ovarian cancer (ROC) patients. METHODS: oral cyclophosphamide (Endoxan®, Baxter, Italy) was administered at the dose of 50 mg daily, continuously. Treatment-related toxicity and response to treatment were assessed by the NCI-CTC criteria, and RECIST criteria, respectively. Progression-free (PFS), and overall survival (OS) were also assessed. RESULTS: 54 patients were analyzed: 20 patients (37.0%) were considered primarily platinum refractory/resistant, while 34 patients (63.0%) were defined as platinum sensitive; 79.6% of patients had received ≥2 previous lines before starting MOC. The objective response rate (ORR) was 20.4%. Eleven patients (20.4%) experienced stable disease and 8 of them had a response duration ≥6 months. A total of 32 patients (59.2.%) progressed during treatment. Median PFS was 4 months, and the 12-month PFS rate was 19.6%; median OS was 13 months, and the 12-month OS rate was 51.5% . Patients responding to MOC showed a more favorable PFS (median = 17 months) compared to patients with stabilization (median = 6 months) or progression of disease (median = 3 months) (p value = 0.0001). Median OS of responding patients was 30 months compared to 11 months in cases achieving stabilization, or progression of disease (median = 8 months) (p value = 0.0001). Only 1 patient experienced grade 3 anemia. Non-hematological grade 3 toxicity was registered in 2 patients. CONCLUSIONS: MOC could provide a valid alternative in terms of risk/benefit ratio in the palliative treatment of heavily treated ROC patients.


Sujet(s)
Antinéoplasiques alcoylants/administration et posologie , Cyclophosphamide/administration et posologie , Récidive tumorale locale/traitement médicamenteux , Tumeurs de l'ovaire/traitement médicamenteux , Administration métronomique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antinéoplasiques alcoylants/effets indésirables , Cyclophosphamide/effets indésirables , Femelle , Humains , Adulte d'âge moyen , Tumeurs de l'ovaire/anatomopathologie , Études rétrospectives , Thérapie de rattrapage/méthodes , Analyse de survie , Résultat thérapeutique
12.
Expert Opin Pharmacother ; 14(6): 699-706, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23472669

RÉSUMÉ

OBJECTIVE: Lapatinib, a dual HER2/EGFR tyrosine kinase inhibitor (TKI), associated to capecitabine represents the treatment of choice in HER2-positive metastatic breast cancer (BC) patients in progression after trastuzumab-based therapy. Though lapatinib-based therapy prolongs the time to progression, its efficacy is often limited by the development of drug resistance. It is aimed to evaluate novel biomarkers predictive of lapatinib response, we analyzed EGFR protein and gene status in a series of 50 metastatic HER2-positive BC patients. METHODS: Lapatinib was given at 1250 mg/day continuously and capecitabine at 2000 mg/m(2)/day every 3 weeks. EGFR protein expression and gene copy number (GCN) were assessed by immunohistochemistry and FISH, respectively. Receiver operating curve (ROC) analysis identified the value of > 3.36 EGFR copies/nucleus as the cut-off point able to discriminate responders versus non-responders. RESULTS: A statistical significant correlation between EGFR GCN value > 3.36 and response to lapatinib (p = 0.01) was found. Cox regression analysis further supported these findings evidencing that HER2 score 3+ and EGFR GCN increase are positive predictor factors of lapatinib response. CONCLUSIONS: Though further investigations are needed to confirm these findings, EGFR GCN could be a suitable screening to identify the subset of BC patients particularly responsive to the dual TKI lapatinib.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs du sein/traitement médicamenteux , Récepteurs ErbB/génétique , Quinazolines/pharmacologie , Adulte , Sujet âgé , Marqueurs biologiques/métabolisme , Tumeurs du sein/anatomopathologie , Résistance aux médicaments antinéoplasiques , Femelle , Dosage génique , Humains , Lapatinib , Adulte d'âge moyen , Métastase tumorale , Modèles des risques proportionnels , Études prospectives , Inhibiteurs de protéines kinases/pharmacologie , Courbe ROC , Récepteur ErbB-2/métabolisme , Analyse de régression
13.
Expert Opin Pharmacother ; 14(5): 629-41, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23414148

RÉSUMÉ

INTRODUCTION: Nausea and vomiting are well recognized in different clinical situations, suggesting that no single mechanism is likely to be responsible for their production. Chemotherapy-induced nausea and vomiting (CINV) can have a negative impact on quality of life and this may lead to a refusal of curative therapy or to a decline in palliative benefits offered by cytotoxic treatment. Palonosetron is a new agent in the class of 5-HT3 receptor antagonists (5-HT3RAs), and differs from the other agents by its higher receptor-binding affinity and longer half-life. These pharmacological properties have resulted in improved antiemetic activity in clinical trials, particularly in the treatment of delayed CINV following moderate emetogenic chemotherapy (MEC). AREA COVERED: A systematic review of the medical literature was completed to inform this update. MEDLINE, the Cochrane Collaboration Library and meeting materials from ASCO and MASCC were all searched. EXPERT OPINION: Palonosetron was the only serotonin receptor antagonist approved for prevention of delayed CINV caused by MEC and its use was incorporated in guideline recommendations. To date, several treatment settings such as multiple day chemotherapy require further studies to improve emesis related to therapy.


Sujet(s)
Antiémétiques/usage thérapeutique , Antinéoplasiques/effets indésirables , Isoquinoléines/usage thérapeutique , Nausée/prévention et contrôle , Quinuclidines/usage thérapeutique , Antagonistes des récepteurs 5-HT3 de la sérotonine/usage thérapeutique , Vomissement/prévention et contrôle , Animaux , Antiémétiques/administration et posologie , Antiémétiques/effets indésirables , Antiémétiques/pharmacocinétique , Association de médicaments , Humains , Isoquinoléines/administration et posologie , Isoquinoléines/effets indésirables , Isoquinoléines/pharmacocinétique , Nausée/induit chimiquement , Antagonistes du récepteur de la neurokinine-1 , Palonosétron , Guides de bonnes pratiques cliniques comme sujet , Quinuclidines/administration et posologie , Quinuclidines/effets indésirables , Quinuclidines/pharmacocinétique , Radiothérapie/effets indésirables , Antagonistes des récepteurs 5-HT3 de la sérotonine/administration et posologie , Antagonistes des récepteurs 5-HT3 de la sérotonine/effets indésirables , Antagonistes des récepteurs 5-HT3 de la sérotonine/pharmacocinétique , Substance P/antagonistes et inhibiteurs , Résultat thérapeutique , Vomissement/induit chimiquement
14.
Anticancer Res ; 33(1): 21-8, 2013 Jan.
Article de Anglais | MEDLINE | ID: mdl-23267124

RÉSUMÉ

Mammalian target of rapamycin (mTOR) is a crucial mediator of tumor progression and may be a promising target in a significant proportion of patients with breast cancer. More specifically, the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mTOR pathway plays a critical role in multiple cellular functions including metabolism, proliferation, growth and survival. This pathway is higly active in many types of cancer and is linked to resistance to many types of therapy. Direct blockade of the mTOR pathway is a new area in breast cancer therapy, with the potential to modulate growth factor- and estrogen-dependent and estrogen-independent pathways, which contribute to the pathogenesis and progression of tumors. Thus, inhibitors of mTOR are of interest as potential therapeutic agents for patients with breast cancer, everolimus and temsirolimus being the main representatives of this category. This review of the literature analyzes the available data emerging from trials and evaluates the efficacy and safety of mTOR inhibitors in all subtypes of breast cancer.


Sujet(s)
Tumeurs du sein , Tumeurs hormonodépendantes , Sérine-thréonine kinases TOR , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Essais cliniques comme sujet , Résistance aux médicaments antinéoplasiques/génétique , Oestrogènes/métabolisme , Évérolimus , Femelle , Humains , Thérapie moléculaire ciblée , Tumeurs hormonodépendantes/métabolisme , Tumeurs hormonodépendantes/anatomopathologie , Tumeurs hormonodépendantes/thérapie , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Récepteur ErbB-2/métabolisme , Sirolimus/analogues et dérivés , Sirolimus/usage thérapeutique , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Sérine-thréonine kinases TOR/génétique , Sérine-thréonine kinases TOR/métabolisme
15.
Gynecol Oncol Case Rep ; 1(1): 24-5, 2011.
Article de Anglais | MEDLINE | ID: mdl-24371596

RÉSUMÉ

► Growing teratoma syndrome (GTS) with unusual liver locations are described after fertility preserving surgery and chemotherapy treatment for mixed malignant ovarian germ cell tumors (MGCT). ► It's a rare syndrome of mixed malignant ovarian germ cell tumors and in both cases enlarged and growing liver masses appeared during cisplatin-etoposide-bleomicin (BEP) chemotherapy. ► Radiological exams (CT scan and MRI) were suggestive for secondary metastasis and serum markers became negative during chemotherapy.

16.
Recenti Prog Med ; 101(7-8): 277-82, 2010.
Article de Italien | MEDLINE | ID: mdl-20842953

RÉSUMÉ

Genetic factors could alter drug metabolism and activity and could predict drug toxicity and/or efficacy. Several chemotherapy agents are administered in different schedules for the treatment of different cancer histotypes. The most used drug in the treatment of gastro-intestinal, head and neck and breast neoplasms is the 5-fluorouracil (5-FU). Capecitabine is a prodrug of 5-FU. Cisplatin based chemotherapy is administered in the treatment of lung, genitourinary tract, head and neck, occult neoplasms, mesothelioma and melanoma. Taxanes are used in lung, breast, head and neck, genitourinary tract neoplasms and sarcomas. Determination of polymorphisms in metabolizing enzymes before the administration of chemotherapy could offer new strategies for optimizing the treatment of individual patients.


Sujet(s)
Antinéoplasiques d'origine végétale/toxicité , Antinéoplasiques/usage thérapeutique , Résistance aux médicaments antinéoplasiques , Tumeurs/traitement médicamenteux , Pharmacogénétique , Toxicologie , Animaux , Antimétabolites antinéoplasiques/métabolisme , Antimétabolites antinéoplasiques/usage thérapeutique , Antimétabolites antinéoplasiques/toxicité , Antinéoplasiques/métabolisme , Camptothécine/analogues et dérivés , Camptothécine/métabolisme , Camptothécine/toxicité , Cisplatine , Dihydrouracil dehydrogenase (NADP) , Antienzymes/toxicité , Fluorouracil/métabolisme , Fluorouracil/toxicité , Génotype , Irinotécan , Tumeurs/mortalité , Polymorphisme génétique , Analyse de survie , Taxoïdes/usage thérapeutique , Thymidylate synthase/génétique , Facteurs temps
17.
Ther Drug Monit ; 31(4): 482-8, 2009 Aug.
Article de Anglais | MEDLINE | ID: mdl-19571774

RÉSUMÉ

5-Fluorouracil (5-FU) is a major chemotherapy drug used for the treatment of tumors. It is catabolized mainly by dihydropyrimidine dehydrogenase, and patients with a complete or partial deficiency of dihydropyrimidine dehydrogenase activity are at risk of developing severe 5-FU-associated toxicity. The aim of this study was to demonstrate that intact peripheral blood mononuclear cells (PBMCs) can be an effective model to evaluate the degradation rate of 5-FU. We developed a sensitive and specific liquid chromatography-tandem mass spectrometry method to measure in vitro the rate of 5-FU degradation by intact PBMC. 5-FU degradation rate was determined by measuring the decrease of a fixed amount of 5-FU (10 microg/mL) added to a solution of PBMC, after 2 hours incubation, expressed as nanogram per milliliter of 5-FU degraded per minute x 10(6) cells. Freshly prepared intact PBMC can degrade efficiently in vitro-added 5-FU. The assay consists of 3 steps: (1) PBMC isolation from peripheral blood, (2) PBMC incubation with 5-FU in vitro for different times, and (3) determination of 5-FU amount to calculate the degradation rate. 5-FU was analyzed by a Q Trap 2000 triple quadrupole/ion trap mass spectrometer in the multiple-reaction-monitoring modes. The chromatographic separation was accomplished using a C18 column with a run time of 16 minutes. By analyzing samples from 39 patients with no 5-FU toxicity, the mean 5-FU degradation rate was 1.85 +/- 0.50 ng x mL(-1) x min(-1) x 10(6) cells. The assessment of a test to measure 5-FU degradation rate in PBMC of patients before 5-FU administration could represent a prescreening method for evaluating the possible toxicity of this drug as an aid to set up a personalized medicine approach for each patient.


Sujet(s)
Fluorouracil/métabolisme , Agranulocytes/métabolisme , Sujet âgé , Chromatographie en phase liquide à haute performance , Femelle , Humains , Mâle , Adulte d'âge moyen , Période réfractaire psychologique , Spectrométrie de masse en tandem
18.
Cancer ; 104(6): 1237-45, 2005 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-16078261

RÉSUMÉ

BACKGROUND: Gemcitabine infusion at the fixed dose rate of 10 mg/m(2) per minute (FDR-gemcitabine) has pharmacokinetic advantages and may result in improved therapeutic efficacy. METHODS: Between April 2002 and September 2003, 40 patients with advanced-stage pancreatic adenocarcinoma (PDAC; n = 27) or biliary tree carcinoma (BTC; n = 13) were treated with weekly FDR-gemcitabine (1000 mg/m(2)). The primary end point was the response rate. The secondary end points were progression-free and overall survival (PFS and OS), tumor marker response, and clinical benefit response (CBR). RESULTS: The overall response rate (ORR) on an intent-to-treat basis was 15% (95% confidence interval [95% CI], 4-26%). A positive CBR was obtained in 14 of 29 (48%) patients. Seventeen of 25 (68%) patients had a reduction in carbohydrate antigen 19-9 (CA 19-9) of > 25%. The median time to treatment failure and the median PFS were 17 weeks (95% CI, 13-22 weeks) and 19 weeks (95% CI, 15-23 weeks), respectively. The median OS was 40 weeks (95% CI, 36-45 weeks) and the 1-year actuarial survival rate was 25.8%. Multivariate analysis showed that a performance status score of 0-1 at study entry and locally advanced disease were the only independent predictors of longer PFS and OS, whereas a reduction in CA 19-9 serum levels > 75% was an independent predictor of longer PFS, but had no impact on OS. Toxicity was mild with Grade 3-4 neutropenia (according to the National Cancer Institute-Common Toxicity Criteria [version 2.0]) in 18 of 427 treatment weeks (4.2%), and Grade 3 anemia and thrombocytopenia in 6 of 427 treatment weeks (1.4%) and 9 of 427 treatment weeks (2.1%), respectively, and asymptomatic Grade 3-4 transaminase elevation in 21 of 427 treatment weeks (4.9%). CONCLUSIONS: FDR-gemcitabine at the weekly dose of 1000 mg/m(2) demonstrated promising activity, despite negligible toxicity, in patients with advanced-stage PDAC and BTC.


Sujet(s)
Adénocarcinome/traitement médicamenteux , Tumeurs des voies biliaires/traitement médicamenteux , Désoxycytidine/analogues et dérivés , Tumeurs du pancréas/traitement médicamenteux , Tumeurs des voies biliaires/mortalité , Antigène CA 19-9/sang , Désoxycytidine/administration et posologie , Désoxycytidine/effets indésirables , Désoxycytidine/usage thérapeutique , Femelle , Humains , Mâle , Tumeurs du pancréas/mortalité , Taux de survie ,
19.
Cancer ; 101(1): 133-8, 2004 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-15221998

RÉSUMÉ

BACKGROUND: Cyclooxygenase-2 (COX-2) is up-regulated frequently and may constitute a promising therapeutic target in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS: Patients with advanced PDAC who had progressive disease after gemcitabine-based chemotherapy were eligible for this pilot study. Treatment was comprised of oral celecoxib (400 mg twice daily) and protracted intravenous (i.v.) infusion 5-fluorouracil (5-FU) (200 mg/m(2) per day), both given continuously for a maximum of 9 treatment months, in the absence of disease progression or unacceptable toxicity. Patients were examined weekly for toxicity and were restaged every 6-8 weeks for tumor assessment. RESULTS: Seventeen patients entered the study. Asymptomatic transaminase elevation was the most common toxicity and reached NCI-CTC (version 3.0) Grade 3-4 in 4 of 133 treatment weeks. No other hematologic or nonhematologic toxicity > Grade 2 was observed. Four patients discontinued celecoxib due to upper gastrointestinal tract toxicity. Two confirmed partial responses (durations of 23 weeks and 68 weeks, respectively) and 2 patients with stable disease (durations of 10 weeks and 13 weeks, respectively) were observed for an overall response rate of 12% (95% confidence interval, 0-27%) in the intent-to-treat population. A significant decrease (> or = 50%) in serum CA 19.9 levels was observed in 3 of 9 evaluable patients. The median time to disease progression was 8 weeks, and the median overall survival was 15 weeks. CONCLUSIONS: The combination of oral celecoxib and 5-FU by protracted i.v. infusion was found to be feasible and well tolerated, and was capable of inducing durable objective responses, even in patients with far advanced, gemcitabine-resistant/refractory PDAC. Further exploration of COX-2 inhibitor/fluropyrimidine combinations is warranted.


Sujet(s)
Adénocarcinome/traitement médicamenteux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du pancréas/traitement médicamenteux , Adénocarcinome/anatomopathologie , Adulte , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Antigène carcinoembryonnaire/sang , Antigène carcinoembryonnaire/effets des médicaments et des substances chimiques , Célécoxib , Évolution de la maladie , Femelle , Fluorouracil/administration et posologie , Fluorouracil/effets indésirables , Humains , Perfusions veineuses , Mâle , Adulte d'âge moyen , Tumeurs du pancréas/anatomopathologie , Projets pilotes , Pyrazoles , Sulfonamides/administration et posologie , Sulfonamides/effets indésirables , Résultat thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...