Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Target Oncol ; 12(4): 525-533, 2017 08.
Article in English | MEDLINE | ID: mdl-28669023

ABSTRACT

BACKGROUND: Patients with metastatic colorectal cancer (mCRC) refractory to standard therapies have a poor prognosis. In this setting, recruitment into clinical trials is warranted, and studies driven by selection according to individual tumor molecular characteristics are expected to provide added value. OBJECTIVE: We retrospectively analyzed data from patients with mCRC refractory to or following failure of standard therapies who were enrolled into phase I/II clinical studies at the Niguarda Cancer Center based on the presence of a specific molecular profile expected to represent the target of susceptibility to the experimental drug(s). PATIENTS AND METHODS: From June 2011 to May 2016, 2044 patients with mCRC underwent molecular screening. Eighty patients (3.9%) were enrolled in ad hoc studies; the median age was 60 years (range 36-86) and the median number of previous treatment lines was five (range 2-8). Molecular characteristics exploited within these studies were MGMT promoter hypermethylation (48.7%), HER2 amplification (28.8%), BRAF V600E mutation (20%), and novel gene fusions involving ALK or NTRK (2.5%). RESULTS: One patient (1%) had RECIST (Response Evaluation Criteria In Solid Tumors) complete response (CR), 13 patients (16.5%) experienced a partial response (PR), and 28 (35%) stable disease (SD). Median progression-free survival (PFS) was 2.8 months (range 2.63-3.83), with 24% of patients displaying PFS >5 months. Median growth modulation index (GMI) was 0.85 (range 0-15.61) and 32.5% of patients had GMI >1.33. KRAS exon 2 mutations were found in 38.5% of patients, and among the 78 patients with known KRAS status, those with wild-type tumors had longer PFS than those with mutated tumors (3.80 [95% CI 2.80-5.03] vs. 2.13 months [95% CI 1.77-2.87], respectively, p = 0.001). Median overall survival (OS) was 7.83 months (range 7.17-9.33) for all patients, and patients with KRAS wild-type tumors had longer OS than those with mutated tumors (7.83 [95% CI 7.33-10.80] vs. 7.18 months [95% CI 5.63-9.33], respectively, p = 0.06). CONCLUSIONS: This single-institution retrospective study indicates that in a heavily pretreated population approximately 4% of mCRC tumors display a potential actionable molecular context suitable for therapeutic intervention. Application of molecular selection is challenging but improves clinical outcome even in later lines of treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Aged , Aged, 80 and over , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Retrospective Studies
2.
ESMO Open ; 1(6): e000111, 2016.
Article in English | MEDLINE | ID: mdl-28848658

ABSTRACT

OBJECTIVE: To identify imaging markers predicting clinical outcomes to regorafenib in metastatic colorectal carcinoma (mCRC). METHODS: The RadioCORRECT study is a post hoc analysis of a cohort of patients with mCRC treated within the phase III placebo-controlled CORRECT trial of regorafenib. Baseline and week 8 contrast-enhanced CT were used to assess response by RECIST 1.1, changes in the sum of target lesion diameters (ΔSTL), lung metastases cavitation and liver metastases density. Primary and secondary objectives were to develop ex novo univariable and multivariable models to predict overall survival (OS) and progression-free survival (PFS), respectively. RESULTS: 202 patients were enrolled, 134 (66.3%) treated with regorafenib and 68 (33.7%) with placebo. In the univariate analysis, PFS predictors were lung metastases cavitation at baseline (HR 0.50, 95% CI 0.27 to 0.92, p=0.03) and at week 8 (HR 0.58, 95% CI 0.36 to 0.93, p=0.02). Baseline cavitation (HR 0.23, 95% CI 0.08 to 0.66, p=0.007), RECIST 1.1 (HR 0.23, 95% CI 0.14 to 0.4, p <0.0001) and ΔSTL (HR 1.16, 95% CI 1.06 to 1.27, p=0.002) predicted OS. We found an increase of 9% of diameter as the best threshold for discriminating OS (HR 2.64, 95% CI 1.61 to 4.34, p <0.001). In the multivariate analysis, baseline and week 8 cavitation remained significant PFS predictors. Baseline cavitation, RECIST 1.1 and ΔSTL remained predictors of OS in exploratory multivariable models. Assessment of liver metastases density did not predict clinical outcome. CONCLUSIONS: RECIST 1.1 and ΔSTL predict favourable outcome to regorafenib. In contrast to liver metastases density that failed to be a predictor, lung metastases cavitation represents a novel radiological marker of favourable outcome that deserves consideration.

4.
Oncologist ; 19(9): 966-74, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25117065

ABSTRACT

Everolimus is a valid therapeutic option for neuroendocrine tumors (NETs); however, data in a real-world setting outside regulatory trials are sparse. The aim of this study was to determine everolimus tolerability and efficacy, in relation to previous treatments, in a compassionate use program. A total of 169 patients with advanced progressive NETs treated with everolimus were enrolled, including 85 with pancreatic NETs (pNETs) and 84 with nonpancreatic NETs (non-pNETs). Previous treatments included somatostatin analogs (92.9%), peptide receptor radionuclide therapy (PRRT; 50.3%), chemotherapy (49.7%), and PRRT and chemotherapy (22.8%). Overall, 85.2% of patients experienced adverse events (AEs), which were severe (grade 3-4) in 46.1%. The most frequent severe AEs were pneumonitis (8.3%), thrombocytopenia (7.7%), anemia (5.3%), and renal failure (3.5%). In patients previously treated with PRRT and chemotherapy, a 12-fold increased risk for severe toxicity was observed, with grade 3-4 AEs reported in 86.8% (vs. 34.3% in other patients). In addition, 63.3% of patients required temporarily everolimus discontinuation due to toxicity. Overall, 27.8% of patients died during a median follow-up of 12 months. Median progression-free survival (PFS) and overall survival (OS) were 12 months and 32 months, respectively. Similar disease control rates, PFS, and OS were reported in pNETs and non-pNETs. In the real-world setting, everolimus is safe and effective for the treatment of NETs of different origins. Higher severe toxicity occurred in patients previously treated with systemic chemotherapy and PRRT. This finding prompts caution when using this drug in pretreated patients and raises the issue of planning for everolimus before PRRT and chemotherapy in the therapeutic algorithm for advanced NETs.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/pathology , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Sirolimus/analogs & derivatives , Aged , Carcinoid Tumor/drug therapy , Carcinoid Tumor/pathology , Compassionate Use Trials , Disease-Free Survival , Drug-Related Side Effects and Adverse Reactions/classification , Everolimus , Female , Humans , Male , Middle Aged , Neoplasm Staging , Neuroendocrine Tumors/pathology , Octreotide/administration & dosage , Pancreatic Neoplasms/pathology , Sirolimus/administration & dosage , Sirolimus/adverse effects
5.
Target Oncol ; 5(1): 19-28, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20383783

ABSTRACT

The introduction of KRAS testing as a diagnostic tool to select patients for epidermal growth factor receptor (EGFR)-targeted cetuximab- or panitumumab-based therapies for metastatic colorectal cancer is widely regarded as a key advance in the field of personalized cancer medicine. Oncologists are now facing emerging issues in the treatment of metastatic colorectal cancer, including: (i) the identification of additional genetic determinants of primary resistance to EGFR-targeted therapy for further improving selection of patients; (ii) the explanation of rare cases of patients carrying KRAS-mutated tumors who have been reported to respond to either cetuximab or panitumumab and (iii) the discovery of mechanisms of secondary resistance to anti-EGFR antibody therapies. Here we discuss the potential role of comprehensive dissection of the key oncogenic nodes in the EGFR signaling cascade to predict resistance and sensitivity to EGFR monoclonal antibodies in metastatic colorectal cancer. Current data suggest that, together with KRAS mutations, the evaluation of BRAF and PIK3CA/PTEN alterations could also be useful for selecting patients with reduced chance to benefit from EGFR-targeted therapy. Furthermore, measuring EGFR gene copy number also appears relevant to positively identify responders. Up until now, each of these markers has been mainly assessed as a single event, often in retrospective analyses and patients' series. As these molecular alterations display overlapping patterns of occurrence, this adds considerable complexity to the drawing of an algorithm suitable for clinical decision-making. We suggest that in the near future comprehensive molecular analysis of the entire oncogenic pathway triggered by the EGFR should be performed, thus enhancing the prediction ability of individual markers.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carcinoma/drug therapy , Colorectal Neoplasms/drug therapy , Animals , Biomarkers, Pharmacological , Carcinoma/diagnosis , Carcinoma/genetics , Carcinoma/pathology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Decision Making , Drug Resistance, Neoplasm , ErbB Receptors/immunology , Gene Dosage/genetics , Humans , Mutation/genetics , Neoplasm Metastasis , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Signal Transduction , ras Proteins/genetics , ras Proteins/metabolism
6.
Anticancer Res ; 28(6B): 3991-2, 2008.
Article in English | MEDLINE | ID: mdl-19192661

ABSTRACT

We report on radiological abnormalities resembling recurrent tumor in adult medulloblastoma receiving intensified chemotherapy and radiotherapy. Evidence provided in this paper confirms previous reports in the pediatric population and suggests that neuroradiologist and medical oncologists should be aware of new possible radiological findings related to aggressive treatments for brain tumors.


Subject(s)
Brain Neoplasms/diagnosis , Medulloblastoma/diagnosis , Adult , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Female , Humans , Magnetic Resonance Imaging , Medulloblastoma/pathology , Medulloblastoma/therapy
7.
Gastric Cancer ; 10(4): 221-7, 2007.
Article in English | MEDLINE | ID: mdl-18095077

ABSTRACT

BACKGROUND: The aim of the study was to evaluate whether the therapy-induced reduction of the (18)F-fluorodeoxyglucose positron emission tomography (18F-FDG-PET) maximum standardized uptake value in patients with advanced gastric adenocarcinoma treated with chemotherapy plus cetuximab could predict the objective response and outcome early during the treatment. METHODS: The study was performed as a part of a phase II trial evaluating cetuximab plus the leucovorin/5-fluorouracil/irinotecan (FOLFIRI) regimen. The objective response was evaluated according to the response evaluation criteria in solid tumors (RECIST) every 6 weeks. The early metabolic response evaluated by 18F-FDG-PET was assessed according to our own evaluated cutoff value (<35%) after receiver operating characteristic (ROC) analysis. RESULTS: Twenty of 22 patients had positive baseline 18F-FDG-PET. The best RECIST response was: complete response (CR), 3; partial response (PR), 9; stable disease (SD), 8. Twelve patients (60%) were classified as metabolic responders and 8 (40%) as nonresponders. At the median follow-up time of 11 months, median time to disease progression (TTP) and overall survival (OS) for early metabolic responders versus nonresponders were 11 versus 5 months (P = 0.0016) and 16 versus 6 months (P = 0.1493), respectively. CONCLUSION: The early metabolic response evaluated by 18F-FDG-PET predicted the clinical outcome in this series of patients with advanced gastric cancer treated with chemotherapy plus cetuximab.


Subject(s)
Adenocarcinoma/diagnostic imaging , Adenocarcinoma/drug therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Positron-Emission Tomography , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Cetuximab , Clinical Trials, Phase II as Topic , ErbB Receptors , Female , Fluorodeoxyglucose F18 , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Radiopharmaceuticals , Remission Induction , Treatment Outcome
8.
Tumori ; 93(6): 611-5, 2007.
Article in English | MEDLINE | ID: mdl-18338499

ABSTRACT

Around 20-30% of patients with hepatic metastasis from colorectal cancer can undergo liver resection, but the increased response rate obtained with the addition of monoclonal antibodies to chemotherapy regimens could result in a higher rate of liver surgery. In this report we describe the case of a patient who underwent a liver resection after neoadjuvant treatment with capecitabine, oxaliplatin and bevacizumab and who achieved a complete pathological response of the liver metastasis. A preoperative CT scan demonstrated a partial response to the treatment while 18FDG-PET scan correctly evaluated the complete pathological response in the liver and detected an active interaortocaval lymph node metastasis. New specific studies are required to evaluate the imaging response in metastatic colorectal cancer patients especially after treatment with new, targeted agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/pathology , Fluorodeoxyglucose F18 , Hepatectomy , Liver Neoplasms/diagnostic imaging , Neoadjuvant Therapy/methods , Positron-Emission Tomography , Tomography, X-Ray Computed , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Bevacizumab , Capecitabine , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Liver Neoplasms/surgery , Lymphatic Metastasis/diagnostic imaging , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Positron-Emission Tomography/methods , Predictive Value of Tests , Radiopharmaceuticals
9.
Clin Colorectal Cancer ; 6(1): 38-45, 2006 May.
Article in English | MEDLINE | ID: mdl-16796790

ABSTRACT

As new improvements in the treatment of colorectal cancer have become available, it has become important to understand the benefits of new therapies or the deleterious effects stemming from the increased risk of toxicity. In particular, a more rational approach to adjuvant chemotherapy for patients with stage II/III disease should be defined by understanding which patients have a higher recurrence risk. Many studies have investigated several molecular markers, but none has been definitively associated with patient outcome. We present a review of studies that have evaluated the immunohistochemical correlation between expression of some biomarkers, such as thymidylate synthase, p53, Ki-67, Bcl-2, and microsatellite instability status expressed by Mut-L homologue 1 and Mut-S homologue 2 proteins, and the prognosis of patients with stage II/III colorectal cancer. We have evaluated studies in which > or = 100 patients were involved in an effort to ensure a representative study group. The only biomarker likely to have a prognostic value is microsatellite instability status, which correlated with a better prognosis.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/pathology , Adaptor Proteins, Signal Transducing , Carrier Proteins/analysis , Humans , Immunohistochemistry , Ki-67 Antigen/analysis , Microsatellite Repeats , MutL Protein Homolog 1 , MutL Proteins , Neoplasm Proteins/analysis , Nuclear Proteins/analysis , Prognosis , Proto-Oncogene Proteins c-bcl-2/analysis , Thymidylate Synthase/analysis , Tumor Suppressor Protein p53/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...