Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
1.
Ann Oncol ; 35(3): 293-301, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38092228

RÉSUMÉ

BACKGROUND: Sacituzumab govitecan (SG), a novel antibody-drug conjugate (ADC) targeting TROP2, is approved for pre-treated metastatic triple-negative breast cancer (mTNBC). We conducted an investigator-initiated clinical trial evaluating neoadjuvant (NA) SG (NCT04230109), and report primary results. PATIENTS AND METHODS: Participants with early-stage TNBC received NA SG for four cycles. The primary objective was to assess pathological complete response (pCR) rate in breast and lymph nodes (ypT0/isN0) to SG. Secondary objectives included overall response rate (ORR), safety, event-free survival (EFS), and predictive biomarkers. A response-guided approach was utilized, and subsequent systemic therapy decisions were at the discretion of the treating physician. RESULTS: From July 2020 to August 2021, 50 participants were enrolled (median age = 48.5 years; 13 clinical stage I disease, 26 stage II, 11 stage III). Forty-nine (98%) completed four cycles of SG. Overall, the pCR rate with SG alone was 30% [n = 15, 95% confidence interval (CI) 18% to 45%]. The ORR per RECIST V1.1 after SG alone was 64% (n = 32/50, 95% CI 77% to 98%). Higher Ki-67 and tumor-infiltrating lymphocytes (TILs) were predictive of pCR to SG (P = 0.007 for Ki-67 and 0.002 for TILs), while baseline TROP2 expression was not (P = 0.440). Common adverse events were nausea (82%), fatigue (76%), alopecia (76%), neutropenia (44%), and rash (48%). With a median follow-up time of 18.9 months (95% CI 16.3-21.9 months), the 2-year EFS for all participants was 95%. Among participants with a pCR with SG (n = 15), the 2-year EFS was 100%. CONCLUSIONS: In the first NA trial with an ADC in localized TNBC, SG demonstrated single-agent efficacy and feasibility of response-guided escalation/de-escalation. Further research on optimal duration of SG as well as NA combination strategies, including immunotherapy, are needed.


Sujet(s)
Anticorps monoclonaux humanisés , Camptothécine/analogues et dérivés , Immunoconjugués , Tumeurs du sein triple-négatives , Humains , Adulte d'âge moyen , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Traitement néoadjuvant , Antigène KI-67 , Antigènes néoplasiques/génétique , Immunoconjugués/effets indésirables
2.
Ann Oncol ; 31(5): 590-598, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-32245699

RÉSUMÉ

BACKGROUND: Little is known about mechanisms of resistance to poly(adenosine diphosphate-ribose) polymerase inhibitors (PARPi) and platinum chemotherapy in patients with metastatic breast cancer and BRCA1/2 mutations. Further investigation of resistance in clinical cohorts may point to strategies to prevent or overcome treatment failure. PATIENTS AND METHODS: We obtained tumor biopsies from metastatic breast cancer patients with BRCA1/2 deficiency before and after acquired resistance to PARPi or platinum chemotherapy. Whole exome sequencing was carried out on each tumor, germline DNA, and circulating tumor DNA. Tumors underwent RNA sequencing, and immunohistochemical staining for RAD51 foci on tumor sections was carried out for functional assessment of intact homologous recombination (HR). RESULTS: Pre- and post-resistance tumor samples were sequenced from eight patients (four with BRCA1 and four with BRCA2 mutation; four treated with PARPi and four with platinum). Following disease progression on DNA-damaging therapy, four patients (50%) acquired at least one somatic reversion alteration likely to result in functional BRCA1/2 protein detected by tumor or circulating tumor DNA sequencing. Two patients with germline BRCA1 deficiency acquired genomic alterations anticipated to restore HR through increased DNA end resection: loss of TP53BP1 in one patient and amplification of MRE11A in another. RAD51 foci were acquired post-resistance in all patients with genomic reversion, consistent with reconstitution of HR. All patients whose tumors demonstrated RAD51 foci post-resistance were intrinsically resistant to subsequent lines of DNA-damaging therapy. CONCLUSIONS: Genomic reversion in BRCA1/2 was the most commonly observed mechanism of resistance, occurring in four of eight patients. Novel sequence alterations leading to increased DNA end resection were seen in two patients, and may be targetable for therapeutic benefit. The presence of RAD51 foci by immunohistochemistry was consistent with BRCA1/2 protein functional status from genomic data and predicted response to later DNA-damaging therapy, supporting RAD51 focus formation as a clinically useful biomarker.


Sujet(s)
Tumeurs du sein , Tumeurs de l'ovaire , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Humains , Tumeurs de l'ovaire/traitement médicamenteux , Platine/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique
3.
Ann Oncol ; 31(5): 626-633, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-32205017

RÉSUMÉ

BACKGROUND: This phase Ib study evaluated the safety, tolerability, pharmacokinetics, and preliminary efficacy of the oral AKT inhibitor ipatasertib and chemotherapy or hormonal therapy in patients with advanced or metastatic solid tumors to determine combined dose-limiting toxicities (DLTs), maximum tolerated dose, and recommended phase II doses and schedules. PATIENTS AND METHODS: The clinical study comprised four combination treatment arms: arm A (with docetaxel), arm B [with mFOLFOX6 (modified leucovorin, 5-fluorouracil, and oxaliplatin)], arm C (with paclitaxel), and arm D (with enzalutamide). Primary endpoints were safety and tolerability; secondary endpoints were pharmacokinetics, clinical activity per Response Evaluation Criteria in Solid Tumors v1.1, and prostate-specific antigen levels. RESULTS: In total, 122 patients were enrolled. Common adverse events were diarrhea, nausea, vomiting, decreased appetite, and fatigue. The safety profiles of the combination regimens were consistent with those of the background regimens, except for diarrhea, hyperglycemia, and rash, which were previously observed with ipatasertib treatment. The only combination DLT across all treatment arms was one event of grade 3 dehydration (ipatasertib 600 mg and paclitaxel). Recommended phase II doses for ipatasertib were 600 mg (and mFOLFOX6) and 400 mg (and paclitaxel), respectively. The maximum assessed dose of ipatasertib 600 mg combined with docetaxel or enzalutamide was well tolerated. Coadministration with enzalutamide (a cytochrome P450 3A inducer) resulted in approximately 50% lower ipatasertib exposure. CONCLUSIONS: Ipatasertib in combination with chemotherapy or hormonal therapy was well tolerated with a safety profile consistent with that of ATP-competitive AKT inhibitors. CLINICAL TRIAL NUMBER: NCT01362374.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Humains , Mâle , Dose maximale tolérée , Tumeurs/traitement médicamenteux , Pipérazines , Pyrimidines/usage thérapeutique
4.
Ann Oncol ; 30(8): 1289-1297, 2019 08 01.
Article de Anglais | MEDLINE | ID: mdl-31147675

RÉSUMÉ

BACKGROUND: This hypothesis-generating trial evaluated neoadjuvant ipatasertib-paclitaxel for early triple-negative breast cancer (TNBC). PATIENTS AND METHODS: In this randomized phase II trial, patients with early TNBC (T ≥ 1.5 cm, N0-2) were randomized 1 : 1 to receive weekly paclitaxel 80 mg/m2 with ipatasertib 400 mg or placebo (days 1-21 every 28 days) for 12 weeks before surgery. Co-primary end points were pathologic complete response (pCR) rate (ypT0/TisN0) in the intention-to-treat (ITT) and immunohistochemistry phosphatase and tensin homolog (PTEN)-low populations. Secondary end points included pCR rate in patients with PIK3CA/AKT1/PTEN-altered tumors and pre-surgery response rates by magnetic resonance imaging (MRI). RESULTS: pCR rates with ipatasertib versus placebo were 17% versus 13%, respectively, in the ITT population (N = 151), 16% versus 13% in the immunohistochemistry PTEN-low population (N = 35), and 18% versus 12% in the PIK3CA/AKT1/PTEN-altered subgroup (N = 62). Rates of overall and complete response (CR) by MRI favored ipatasertib in all three populations (CR rate 39% versus 9% in the PIK3CA/AKT1/PTEN-altered subgroup). Ipatasertib was associated with more grade ≥3 adverse events (32% versus 16% with placebo), especially diarrhea (17% versus 1%). Higher cycle 1 day 8 (C1D8) immune score was significantly associated with better response only in placebo-treated patients. All ipatasertib-treated patients with low immune scores and a CR had PIK3CA/AKT1/PTEN-altered tumors. CONCLUSIONS: Adding ipatasertib to 12 weeks of paclitaxel for early TNBC did not clinically or statistically significantly increase pCR rate, although overall response rate by MRI was numerically higher with ipatasertib. The antitumor effect of ipatasertib was most pronounced in biomarker-selected patients. Safety was consistent with prior experience of ipatasertib-paclitaxel. A T-cell-rich environment at C1D8 had a stronger association with improved outcomes in paclitaxel-treated patients than seen for baseline tumor-infiltrating lymphocytes. This dependency may be overcome with the addition of AKT inhibition, especially in patients with PIK3CA/AKT1/PTEN-altered tumors. CLINICALTRIALS.GOV: NCT02301988.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Traitement néoadjuvant/méthodes , Paclitaxel/administration et posologie , Pipérazines/administration et posologie , Pyrimidines/administration et posologie , Tumeurs du sein triple-négatives/thérapie , Adulte , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Marqueurs biologiques tumoraux/antagonistes et inhibiteurs , Marqueurs biologiques tumoraux/génétique , Région mammaire/imagerie diagnostique , Région mammaire/anatomopathologie , Région mammaire/chirurgie , Survie sans rechute , Méthode en double aveugle , Calendrier d'administration des médicaments , Femelle , Mutation gain de fonction , Humains , Imagerie par résonance magnétique , Mastectomie , Adulte d'âge moyen , Traitement néoadjuvant/effets indésirables , Stadification tumorale , Paclitaxel/effets indésirables , Sélection de patients , Pipérazines/effets indésirables , Placebo/administration et posologie , Placebo/effets indésirables , Pyrimidines/effets indésirables , Tumeurs du sein triple-négatives/mortalité , Tumeurs du sein triple-négatives/anatomopathologie
5.
Ann Oncol ; 29(1): 154-161, 2018 01 01.
Article de Anglais | MEDLINE | ID: mdl-29045554

RÉSUMÉ

Background: Homologous recombination defects in BRCA1/2-mutated tumors result in sensitivity to poly(ADP-ribose) polymerase inhibitors, which interfere with DNA damage repair. Veliparib, a potent poly(ADP-ribose) polymerase inhibitor, enhanced the antitumor activity of platinum agents and temozolomide in early phase clinical trials. This phase II study examined the safety and efficacy of intermittent veliparib with carboplatin/paclitaxel (VCP) or temozolomide (VT) in patients with BRCA1/2-mutated breast cancer. Patients and methods: Eligible patients ≥18 years with locally recurrent or metastatic breast cancer and a deleterious BRCA1/2 germline mutation were randomized 1 : 1 : 1 to VCP, VT, or placebo plus carboplatin/paclitaxel (PCP). Primary end point was progression-free survival (PFS); secondary end points included overall survival (OS) and overall response rate (ORR). Results: Of 290 randomized patients, 284 were BRCA+, confirmed by central laboratory. For VCP versus PCP, median PFS was 14.1 and 12.3 months, respectively [hazard ratio (HR) 0.789; 95% CI 0.536-1.162; P = 0.227], interim median OS 28.3 and 25.9 months (HR 0.750; 95% CI 0.503-1.117; P = 0.156), and ORR 77.8% and 61.3% (P = 0.027). For VT (versus PCP), median PFS was 7.4 months (HR 1.858; 95% CI 1.278-2.702; P = 0.001), interim median OS 19.1 months (HR 1.483; 95% CI 1.032-2.131; P = 0.032), and ORR 28.6% (P < 0.001). Safety profile was comparable between carboplatin/paclitaxel arms. Adverse events (all grades) of neutropenia, anemia, alopecia, and neuropathy were less frequent with VT versus PCP. Conclusion: Numerical but not statistically significant increases in both PFS and OS were observed in patients with BRCA1/2-mutated recurrent/metastatic breast cancer receiving VCP compared with PCP. The addition of veliparib to carboplatin/paclitaxel significantly improved ORR. There was no clinically meaningful increase in toxicity with VCP versus PCP. VT was inferior to PCP. An ongoing phase III trial is evaluating VCP versus PCP, with optional continuation single-agent therapy with veliparib/placebo if chemotherapy is discontinued without progression, in this patient population. Clinical trial information: NCT01506609.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeur du sein de l'homme/traitement médicamenteux , Tumeur du sein de l'homme/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Adulte , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Benzimidazoles/administration et posologie , Benzimidazoles/effets indésirables , Tumeurs du sein/anatomopathologie , Tumeur du sein de l'homme/anatomopathologie , Carboplatine/administration et posologie , Carboplatine/effets indésirables , Femelle , Gène BRCA1 , Gène BRCA2 , Mutation germinale , Humains , Mâle , Adulte d'âge moyen , Métastase tumorale , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/génétique , Récidive tumorale locale/anatomopathologie , Paclitaxel/administration et posologie , Paclitaxel/effets indésirables , Placebo , Méthode en simple aveugle , Témozolomide/administration et posologie , Témozolomide/effets indésirables , Jeune adulte
6.
Ann Oncol ; 26(9): 1904-1910, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-26202594

RÉSUMÉ

BACKGROUND: Increased hepatocyte growth factor/MET signaling is associated with an aggressive phenotype and poor prognosis in triple-negative breast cancer (TNBC). We evaluated the benefit of adding onartuzumab, a monoclonal anti-MET antibody, to paclitaxel with/without bevacizumab in patients with TNBC. PATIENTS AND METHODS: Women with metastatic TNBC were randomized to receive onartuzumab plus placebo plus weekly paclitaxel (OP; n = 60) or onartuzumab plus bevacizumab plus paclitaxel (OBP; n = 63) or placebo plus bevacizumab plus paclitaxel (BP; n = 62). The primary end point was progression-free survival (PFS); additional end points included overall survival (OS), objective response rate (ORR), and safety. This trial was hypothesis generating and did not have power to detect minimum clinically meaningful differences between treatment arms. RESULTS: There was no improvement in PFS with the addition of onartuzumab to BP [hazard ratio (HR), 1.08; 95% confidence interval (CI) 0.69-1.70]; the risk of a PFS event was higher with OP than with BP (HR, 1.74; 95% CI 1.13-2.68). Most patients had MET-negative tumors (88%); PAM50 subtype analysis showed basal-like tumors in 68% of samples. ORR was higher in the bevacizumab arms (OBP: 42.2%; 95% CI 28.6-57.1; BP: 54.7%; 95% CI 41.0-68.4) compared with OP (27.5%; 95% CI 15.9-40.6). Median OS was shorter with OBP (HR, 1.36; 95% CI 0.75-2.46) and OP (HR, 1.92; 95% CI 1.03-3.59), than with BP. Peripheral edema was more frequent in the onartuzumab arms (OBP, 51.8%; OP, 58.6%) versus BP (17.7%). CONCLUSION: This study did not show a clinical benefit of the addition of onartuzumab to paclitaxel with/without bevacizumab in patients with predominantly MET-negative TNBC. CLINICALTRIALSGOV: NCT01186991.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Bévacizumab/usage thérapeutique , Paclitaxel/usage thérapeutique , Tumeurs du sein triple-négatives/traitement médicamenteux , Adulte , Sujet âgé , Inhibiteurs de l'angiogenèse/effets indésirables , Inhibiteurs de l'angiogenèse/usage thérapeutique , Anticorps monoclonaux/effets indésirables , Antinéoplasiques d'origine végétale/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Bévacizumab/effets indésirables , Survie sans rechute , Femelle , Humains , Adulte d'âge moyen , Paclitaxel/effets indésirables , Placebo/usage thérapeutique
7.
Br J Cancer ; 111(11): 2058-66, 2014 11 25.
Article de Anglais | MEDLINE | ID: mdl-25290090

RÉSUMÉ

BACKGROUND: This phase 1 study evaluated the maximum tolerated dose (MTD), safety, and efficacy of bosutinib (competitive Src/Abl tyrosine kinase inhibitor) plus capecitabine. METHODS: Patients with locally advanced/metastatic breast, pancreatic, or colorectal cancers; cholangiocarcinoma; or glioblastoma received bosutinib plus capecitabine at eight of nine possible dose combinations using an 'up-down' design to determine the toxicity contour of the combination. RESULTS: Among 32 enrolled patients, none of the 9 patients receiving MTD (bosutinib 300 mg once daily plus capecitabine 1000 mg m(-2) twice daily) experienced dose-limiting toxicities (DLTs). Overall, 2 out of 31 (6%) evaluable patients experienced DLTs (grade 3 neurologic pain (n=1); grade 3 pruritus/rash and increased alanine aminotransferase (n=1)). Most common treatment-related adverse events (AEs) were diarrhoea, nausea, vomiting, palmar-plantar erythrodysesthesia (PPE), fatigue; most frequent grade 3/4 AEs: PPE, fatigue, and increased alanine/aspartate aminotransferase. Although diarrhoea was common, 91% of affected patients experienced maximum grade 1/2 events that resolved. Best overall confirmed partial response or stable disease >24 weeks (all tumour types) was observed in 6 and 13% of patients. CONCLUSIONS: In this population of patients with advanced solid tumours, bosutinib plus capecitabine demonstrated a safety profile similar to that previously reported for bosutinib or capecitabine monotherapy; limited efficacy was observed.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs/traitement médicamenteux , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Dérivés de l'aniline/administration et posologie , Dérivés de l'aniline/effets indésirables , Capécitabine , Désoxycytidine/administration et posologie , Désoxycytidine/effets indésirables , Désoxycytidine/analogues et dérivés , Femelle , Fluorouracil/administration et posologie , Fluorouracil/effets indésirables , Fluorouracil/analogues et dérivés , Humains , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Stadification tumorale , Tumeurs/anatomopathologie , Nitriles/administration et posologie , Nitriles/effets indésirables , Quinoléines/administration et posologie , Quinoléines/effets indésirables
8.
Ann Oncol ; 25(8): 1656-63, 2014 Aug.
Article de Anglais | MEDLINE | ID: mdl-24827126

RÉSUMÉ

BACKGROUND: To establish the maximum tolerated dose, determine safety/tolerability and evaluate the pharmacokinetics and preliminary efficacy of olaparib in combination with cisplatin in patients with advanced solid tumors. PATIENTS AND METHODS: Patients aged ≥ 18 years with advanced solid tumors, who had progressed on standard treatment, were assigned to a treatment cohort and received oral olaparib [50-200 mg twice daily (bid); 21-day cycle] continuously or intermittently (days 1-5 or 1-10) in combination with cisplatin (60-75 mg/m(2) intravenously) on day 1 of each cycle. RESULTS: Dose-limiting toxicities (DLTs) of grade 3 neutropenia (cisplatin 75 mg/m(2) with continuous olaparib 100 mg bid or 200 mg bid; n = 1 each) and grade 3 lipase elevation (cisplatin 75 mg/m(2) with olaparib 100 mg bid days 1-10 or 50 mg bid days 1-5; n = 1 each) were reported. Olaparib and cisplatin doses were subsequently reduced to 50 mg bid days 1-5 and 60 mg/m(2), respectively; no DLTs were reported for patients receiving this regimen. The most frequent grade ≥ 3 adverse events were neutropenia (16.7%), anemia (9.3%) and leucopenia (9.3%). Thirty patients (55.6%) received colony-stimulating factors for hematologic support. The overall objective response rate was 41% for patients with measurable disease, and 43% and 71% among patients with a BRCA1/2 mutation who had ovarian and breast cancer, respectively. CONCLUSIONS: Olaparib in combination with cisplatin 75 mg/m(2) was not considered tolerable; intermittent olaparib (50 mg bid, days 1-5) with cisplatin 60 mg/m(2) improved tolerability. Promising antitumor activity in patients with germline BRCA1/2 mutations was observed and warrants further investigation.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Cisplatine/administration et posologie , Tumeurs/traitement médicamenteux , Tumeurs de l'ovaire/traitement médicamenteux , Phtalazines/administration et posologie , Pipérazines/administration et posologie , Adulte , Sujet âgé , Tumeurs du sein/anatomopathologie , Cisplatine/effets indésirables , Évolution de la maladie , Femelle , Humains , Mâle , Adulte d'âge moyen , Tumeurs/anatomopathologie , Tumeurs de l'ovaire/anatomopathologie , Phtalazines/effets indésirables , Pipérazines/effets indésirables , Résultat thérapeutique
9.
Breast Cancer Res Treat ; 139(2): 403-10, 2013 Jun.
Article de Anglais | MEDLINE | ID: mdl-23645007

RÉSUMÉ

We aimed to evaluate the efficacy and feasibility of combining trastuzumab/vinorelbine with bevacizumab in patients with first-or second-line HER2-positive, metastatic breast cancer (MBC). Eligible patients had HER2-positive measureable MBC, with no more than one prior line of chemotherapy, and were treated with trastuzumab (4 mg/kg × 2 mg/kg weekly thereafter), vinorelbine (25 mg/m(2) weekly), and bevacizumab (10 mg/kg every 2 weeks). Co-primary endpoints were (a) the proportion of patients alive and progression-free at 1 year and (b) safety profile/feasibility. Feasibility was defined as a rate of grade 3/4 non-hematologic toxicity attributable to protocol-based therapy <20 %. Twenty-nine patients were enrolled (n = 22 first-line, n = 7 second-line). Median age was 48 years (range 37-68). The median number of cycles received was 8 (1-23) and median duration on treatment was 7.4 months (range 1-22). The study was closed early due to higher-than-expected rates of grade 3/4 non-hematologic toxicities, with 50 events in 20 patients. A total of six patients (21 %) were taken off study for treatment-related toxicity. Most common treatment-related toxicities included fatigue (n = 7), febrile neutropenia (n = 4), and headache (n = 3). At 1 year, 8/22 first-line (36 %) and 2/7 second-line (29 %) patients were alive and progression-free. Median PFS was 9.9 months and 7.8 months in the first- and second-line cohorts, respectively. Objective responses were observed in 16/22 (73 %) and 5/7 (71 %) patients in the first- and second-line settings. Although the combination of vinorelbine, trastuzumab, and bevacizumab showed notable activity in HER2-positive MBC, the proportion of first-line patients alive and progression-free at 1 year was deemed unlikely to reach the pre-defined threshold for declaring success. Additionally, unacceptable toxicity was observed, at rates greater than previously reported with vinorelbine/trastuzumab or vinorelbine/bevacizumab doublet combinations.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Récepteur ErbB-2/métabolisme , Adulte , Sujet âgé , Anticorps monoclonaux humanisés/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Bévacizumab , Tumeurs du sein/mortalité , Tumeurs du sein/anatomopathologie , Femelle , Humains , Adulte d'âge moyen , Métastase tumorale , Stadification tumorale , Trastuzumab , Résultat thérapeutique , Vinblastine/administration et posologie , Vinblastine/analogues et dérivés , Vinorelbine
10.
Mol Cell ; 6(2): 373-84, 2000 Aug.
Article de Anglais | MEDLINE | ID: mdl-10983984

RÉSUMÉ

Pleckstrin homology (PH) domains are protein modules of around 120 amino acids found in many proteins involved in cellular signaling. Certain PH domains drive signal-dependent membrane recruitment of their host proteins by binding strongly and specifically to lipid second messengers produced by agonist-stimulated phosphoinositide 3-kinases (PI 3-Ks). We describe X-ray crystal structures of two different PH domains bound to Ins(1,3,4,5)P4, the head group of the major PI 3-K product PtdIns(3,4,5)P3. One of these PH domains (from Grp1) is PtdIns(3,4,5)P3 specific, while the other (from DAPP1/PHISH) binds strongly to both PtdIns(3,4,5)P3 and its 5'-dephosphorylation product, PtdIns(3,4)P2. Comparison of the two structures provides an explanation for the distinct phosphoinositide specificities of the two PH domains and allows us to predict the 3-phosphoinositide selectivity of uncharacterized PH domains.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Inositol phosphates/métabolisme , Lipoprotéines , Phosphatidylinositol 3-kinases/composition chimique , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidyl inositols/métabolisme , Séquence d'acides aminés , Sites de fixation , Protéines du sang/composition chimique , Protéines du sang/métabolisme , Cristallographie aux rayons X , Acides gras/composition chimique , Acides gras/métabolisme , Liaison hydrogène , Modèles moléculaires , Données de séquences moléculaires , Structure secondaire des protéines , Alignement de séquences , Similitude de séquences d'acides aminés , Transduction du signal , Spécificité du substrat , Domaine d'homologie SRC
11.
J Biol Chem ; 273(46): 30497-508, 1998 Nov 13.
Article de Anglais | MEDLINE | ID: mdl-9804818

RÉSUMÉ

Pleckstrin homology (PH) domains are small protein modules involved in recruitment of signaling molecules to cellular membranes, in some cases by binding specific phosphoinositides. We describe use of a convenient "dot-blot" approach to screen 10 different PH domains for those that recognize particular phosphoinositides. Each PH domain bound phosphoinositides in the assay, but only two (from phospholipase C-delta1 and Grp1) showed clear specificity for a single species. Using soluble inositol phosphates, we show that the Grp1 PH domain (originally cloned on the basis of its phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3) binding) binds specifically to D-myo-inositol 1,3,4,5-tetrakisphosphate (Ins(1,3,4,5)P4) (the PtdIns(3,4,5)P3 headgroup) with KD = 27.3 nM, but binds D-myo-inositol 1,3,4-trisphosphate (Ins(1,3,4)P3) or D-myo-inositol 1,4,5-trisphosphate (Ins(1,4,5)P3) over 80-fold more weakly. We show that this specificity allows localization of the Grp1 PH domain to the plasma membrane of mammalian cells only when phosphatidylinositol 3-kinase (PI 3-K) is activated. The presence of three adjacent equatorial phosphate groups was critical for inositol phosphate binding by the Grp1 PH domain. By contrast, another PH domain capable of PI 3-K-dependent membrane recruitment (encoded by EST684797) does not distinguish Ins(1,3,4)P3 from Ins(1,3,4,5)P3 (binding both with very high affinity), despite selecting strongly against Ins(1,4,5)P3. The remaining PH domains tested appear significantly less specific for particular phosphoinositides. Together with data presented in the literature, our results suggest that many PH domains bind similarly to multiple phosphoinositides (and in some cases phosphatidylserine), and are likely to be regulated in vivo by the most abundant species to which they bind. Thus, using the same simple approach to study several PH domains simultaneously, our studies suggest that highly specific phosphoinositide binding is a characteristic of relatively few cases.


Sujet(s)
Phosphatidyl inositols/métabolisme , Similitude de séquences d'acides aminés , Animaux , Sites de fixation , Calorimétrie , Humains , Inositol phosphates/métabolisme , Isoenzymes/métabolisme , Cinétique , Ligands , Souris , Phosphatidylinositol 3-kinases/métabolisme , Phospholipase C delta , Rats , Récepteurs cytoplasmiques et nucléaires/métabolisme , Relation structure-activité , Type C Phospholipases/métabolisme
12.
EMBO J ; 17(18): 5374-87, 1998 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-9736615

RÉSUMÉ

Phosphatidylinositol 3-kinase (PI3K) mediates a variety of cellular responses by generating PtdIns(3,4)P2 and PtdIns(3,4,5)P3. These 3-phosphoinositides then function directly as second messengers to activate downstream signaling molecules by binding pleckstrin homology (PH) domains in these signaling molecules. We have established a novel assay in the yeast Saccharomyces cerevisiae to identify proteins that bind PtdIns(3,4)P2 and PtdIns(3,4,5)P3 in vivo which we have called TOPIS (Targets of PI3K Identification System). The assay uses a plasma membrane-targeted Ras to complement a temperature-sensitive CDC25 Ras exchange factor in yeast. Coexpression of PI3K and a fusion protein of activated Ras joined to a PH domain known to bind PtdIns(3,4)P2 (AKT) or PtdIns(3,4,5)P3 (BTK) rescues yeast growth at the non-permissive temperature of 37 degreesC. Using this assay, we have identified several amino acids in the beta1-beta2 region of PH domains that are critical for high affinity binding to PtdIns(3,4)P2 and/or PtdIns(3,4,5)P3, and we have proposed a structural model for how these PH domains might bind PI3K products with high affinity. From these data, we derived a consensus sequence which predicts high-affinity binding to PtdIns(3, 4)P2 and/or PtdIns(3,4,5)P3, and we have identified several new PH domain-containing proteins that bind PI3K products, including Gab1, Dos, myosinX, and Sbf1. Use of this assay to screen for novel cDNAs which rescue yeast at the non-permissive temperature should provide a powerful approach for uncovering additional targets of PI3K.


Sujet(s)
Protéines fongiques/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Phosphoprotéines , Saccharomyces cerevisiae/génétique , Systèmes de seconds messagers/physiologie , Séquence d'acides aminés , Protéines du sang/génétique , Membrane cellulaire , Séquence consensus , Séquence conservée , Modèles moléculaires , Mutation , Phosphatidylinositol 3-kinases/génétique , Phosphates phosphatidylinositol/métabolisme , Liaison aux protéines , Protéines de fusion recombinantes , Similitude de séquences d'acides aminés , Protéines G ras/physiologie
13.
Cell ; 87(4): 733-43, 1996 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-8929541

RÉSUMÉ

We provide evidence that a class of integrins combines with the adaptor Shc and thereby with Grb2. Coimmunoprecipitation and mutagenesis experiments indicate that the recruitment of Shc is specified by the extracellular or transmembrane domain of integrin alpha subunit and suggest that this process is mediated by caveolin. Mutagenesis and dominant-negative inhibition studies reveal that Shc is necessary and sufficient for activation of the MAP kinase pathway in response to integrin ligation. Mitogens and Shc-activating integrins cooperate to promote transcription from the Fos serum response element and transit through G1. In contrast, adhesion mediated by integrins not linked to Shc results in cell cycle arrest and apoptosis even in presence of mitogens. These findings indicate that the association of specific integrins with Shc regulates cell survival and cell cycle progression.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Protéines adaptatrices du transport vésiculaire , Antigènes CD/métabolisme , Cycle cellulaire/physiologie , Antigènes CD29/métabolisme , Protéines/métabolisme , Transduction du signal , Cellules 3T3 , Animaux , Antigènes CD/génétique , Apoptose , Cellules CHO , Calcium-Calmodulin-Dependent Protein Kinases/métabolisme , Adhérence cellulaire , Molécules d'adhérence cellulaire/métabolisme , Cricetinae , Protéines de liaison à l'ADN/génétique , Activation enzymatique , Matrice extracellulaire/métabolisme , Focal adhesion kinase 1 , Focal adhesion protein-tyrosine kinases , Protéine adaptatrice GRB2 , Régulation de l'expression des gènes , Intégrine alphaV , Antigènes CD29/génétique , Protéines membranaires/métabolisme , Souris , Protéines nucléaires/génétique , Tests aux précipitines , Liaison aux protéines , Protein-tyrosine kinases/métabolisme , Protéines recombinantes/métabolisme , Séquences d'acides nucléiques régulatrices , Facteur de réponse au sérum , Protéines adaptatrices de signalisation Shc , Protéine transformante-1 contenant un domaine d'homologie-2 de Src , Transcription génétique
15.
J Biol Chem ; 271(8): 3959-62, 1996 Feb 23.
Article de Anglais | MEDLINE | ID: mdl-8626723

RÉSUMÉ

Stimulation of the insulin receptor (IR) results in tyrosine phosphorylation of the intermediate molecules insulin receptor substrate-1 (IRS-1), IRS-2, and Shc, which then couple the IR to downstream signaling pathways by serving as binding sites for signaling molecules with SH2 domains. It has been proposed that direct binding of IRS-1, IRS-2, and Shc to an NPX-Tyr(P) motif in the juxtamembrane region of the IR is required for tyrosine phosphorylation of these molecules by the IR. In this regard, Shc and IRS-1 contain domains that are distinct from SH2 domains, referred to as the phosphotyrosine binding (PTB) or phosphotyrosine interaction (PI) domains, which bind phosphotyrosine in the context of an NPX-Tyr(P) motif. To further clarify the role of the Shc PTB/PI domain, we identified a mutation in this domain that abrogated binding of Shc to the IR in vitro. Interestingly, this mutation completely abolished Shc phosphorylation by the IR in vivo whereas mutation of the arginine in the FLVRES motif of the Shc SH2 domain did not affect Shc phosphorylation by insulin. In addition, we identified specific amino acids on the IR that are required for the IR to stimulate Shc but not IRS-1 phosphorylation in vivo. As with the PTB/PI domain Shc mutant, the ability of these mutant receptors to phosphorylate Shc correlates with the binding of the PTB/PI domain of Shc to similar sequences in vitro. These findings support a model in which binding of the PTB/PI domain of Shc directly to the NPX-Tyr(P) motif on the IR mediates Shc phosphorylation by insulin.


Sujet(s)
Phosphoprotéines/métabolisme , Phosphotyrosine/métabolisme , Récepteur trkA/immunologie , Domaine d'homologie SRC , Cellules 3T3 , Séquence d'acides aminés , Animaux , Membrane cellulaire/métabolisme , Clonage moléculaire , Humains , Substrats du récepteur à l'insuline , Protéines et peptides de signalisation intracellulaire , Souris , Données de séquences moléculaires , Mutagenèse dirigée , Mutation ponctuelle , Récepteur trkA/biosynthèse , Protéines de fusion recombinantes/métabolisme , Transduction du signal
16.
Curr Biol ; 5(11): 1296-302, 1995 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-8574587

RÉSUMÉ

BACKGROUND: In insulin-sensitive cells, such as adipocytes and skeletal muscle, the activation of phosphoinositide 3-kinase (PI 3-kinase) is thought to be critical in allowing insulin to stimulate both the uptake of glucose and the translocation of a specialized glucose transporter, GLUT4, to the plasma membrane. However, the downstream mediators that couple PI 3-kinase to GLUT4 translocation are still not known. Recent studies have shown that the GTP-binding protein Rac mediates some of the biological effects of PI 3-kinase, and these findings have led to the suggestion that Rac may be a common mediator for a variety of responses mediated by PI 3-kinase. To determine whether Rac couples PI 3-kinase to glucose uptake in adipocytes, we produced 3T3-L1 cells expressing either a constitutively active Rac1 (V12 Rac1, containing a valine residue at position 12) or a dominant-inhibitory Rac1 (N17 Rac1, containing an asparagine residue at position 17). RESULTS: The stable expression of both V12 Rac1 and N17 Rac1 led to observable phenotypes in 3T3-L1 cells; expression of V12 Rac1 resulted in constitutive formation of lamellipodia and constitutive activation of the cJun-N-terminal kinase (JNK), whereas expression of N17 Rac1 inhibited the insulin-stimulated formation of lamellipodia. However, neither basal glucose uptake nor insulin-stimulated glucose uptake was affected by the expression of either mutant Rac protein. In addition, expression of V12 Rac1 did not reverse the inhibition of insulin-stimulated glucose uptake caused by the PI 3-kinase inhibitor wortmannin. CONCLUSIONS: These findings provide direct evidence that PI 3-kinase does not use Rac to couple the insulin receptor to glucose uptake in adipocytes. Furthermore, the finding that Rac does not mediate glucose uptake in response to insulin is consistent with the idea that PI 3-kinase couples to a variety of different effector molecules in cells, and suggests that some of the specificity in the biological responses elicited by PI 3-kinase may be mediated by the activation of different effector molecules.


Sujet(s)
Adipocytes/métabolisme , Protéines G/métabolisme , Glucose/métabolisme , Mitogen-Activated Protein Kinases , Protéines du muscle , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Cellules 3T3 , Androstadiènes/pharmacologie , Animaux , Transport biologique , Calcium-Calmodulin-Dependent Protein Kinases/métabolisme , Milieux de culture sans sérum , Protéines G/génétique , Expression des gènes , Transporteur de glucose de type 4 , Insuline/pharmacologie , JNK Mitogen-Activated Protein Kinases , Souris , Transporteurs de monosaccharides/métabolisme , Phosphatidylinositol 3-kinases , Wortmannine , Protéines G rac
17.
Proc Natl Acad Sci U S A ; 92(22): 10247-51, 1995 Oct 24.
Article de Anglais | MEDLINE | ID: mdl-7479761

RÉSUMÉ

Recent experimental evidence has focused attention to the role of two molecules, insulin receptor substrate 1 (IRS-1) and phosphatidylinositol 3-kinase (PI3-kinase), in linking the insulin receptor to glucose uptake; IRS-1 knockout mice are insulin resistant, and pharmacological inhibitors of PI3-kinase block insulin-stimulated glucose uptake. To investigate the role of PI3-kinase and IRS-1 in insulin-stimulated glucose uptake we examined whether stimulation of insulin-sensitive cells with platelet-derived growth factor (PDGF) or with interleukin 4 (IL-4) stimulates glucose uptake; the activated PDGF receptor (PDGFR) directly binds and activates PI3-kinase, whereas the IL-4 receptor (IL-4R) activates PI3-kinase via IRS-1 or the IRS-1-related molecule 4PS. We found that stimulation of 3T3-L1 adipocytes with PDGF resulted in tyrosine phosphorylation of the PDGFR and activation of PI3-kinase in these cells. To examine whether IL-4 stimulates glucose uptake, L6 myoblasts were engineered to overexpress GLUT4 as well as both chains of the IL-4R (L6/IL-4R/GLUT4); when these L6/IL-4R/GLUT4 myoblasts were stimulated with IL-4, IRS-1 became tyrosine phosphorylated and associated with PI3-kinase. Although PDGF and IL-4 can activate PI3-kinase in the respective cell lines, they do not possess insulin's ability to stimulate glucose uptake and GLUT4 translocation to the plasma membrane. These findings indicate that activation of PI3-kinase is not sufficient to stimulate GLUT4 translocation to the plasma membrane. We postulate that activation of a second signaling pathway by insulin, distinct from PI3-kinase, is necessary for the stimulation of glucose uptake in insulin-sensitive cells.


Sujet(s)
Adipocytes/métabolisme , Glucose/métabolisme , Insuline/pharmacologie , Transporteurs de monosaccharides/métabolisme , Protéines du muscle , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Transduction du signal , Cellules 3T3 , Adipocytes/cytologie , Animaux , Transport biologique/effets des médicaments et des substances chimiques , Différenciation cellulaire , Membrane cellulaire/métabolisme , Désoxyglucose/métabolisme , Activation enzymatique , Facteur de croissance épidermique/pharmacologie , Transporteur de glucose de type 4 , Cinétique , Souris , Phosphatidylinositol 3-kinases , Facteur de croissance dérivé des plaquettes/pharmacologie , Récepteur à l'insuline/physiologie , Transduction du signal/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE