Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maria Maddalena Laterza is active.

Publication


Featured researches published by Maria Maddalena Laterza.


World Journal of Gastroenterology | 2014

Treatment of gastric cancer

Michele Orditura; Gennaro Galizia; V. Sforza; Valentina Gambardella; Alessio Fabozzi; Maria Maddalena Laterza; Francesca Andreozzi; Jole Ventriglia; B. Savastano; Andrea Mabilia; Eva Lieto; Fortunato Ciardiello; Ferdinando De Vita

The authors focused on the current surgical treatment of resectable gastric cancer, and significance of peri- and post-operative chemo or chemoradiation. Gastric cancer is the 4(th) most commonly diagnosed cancer and the second leading cause of cancer death worldwide. Surgery remains the only curative therapy, while perioperative and adjuvant chemotherapy, as well as chemoradiation, can improve outcome of resectable gastric cancer with extended lymph node dissection. More than half of radically resected gastric cancer patients relapse locally or with distant metastases, or receive the diagnosis of gastric cancer when tumor is disseminated; therefore, median survival rarely exceeds 12 mo, and 5-years survival is less than 10%. Cisplatin and fluoropyrimidine-based chemotherapy, with addition of trastuzumab in human epidermal growth factor receptor 2 positive patients, is the widely used treatment in stage IV patients fit for chemotherapy. Recent evidence supports the use of second-line chemotherapy after progression in patients with good performance status.


International Journal of Cancer | 2016

HER2 loss in HER2-positive gastric or gastroesophageal cancer after trastuzumab therapy : Implication for further clinical research

Filippo Pietrantonio; Marta Caporale; Federica Morano; Mario Scartozzi; Annunziata Gloghini; F. De Vita; Elisa Giommoni; Lorenzo Fornaro; Giuseppe Aprile; Davide Melisi; Rosa Berenato; Alessia Mennitto; Chiara C. Volpi; Maria Maddalena Laterza; Valeria Pusceddu; Lorenzo Antonuzzo; Enrico Vasile; Elena Ongaro; F. Simionato; F. de Braud; Valter Torri; M. Di Bartolomeo

Mechanisms of acquired resistance to trastuzumab‐based treatment in gastric cancer are largely unknown. In this study, we analyzed 22 pairs of tumor samples taken at baseline and post‐progression in patients receiving chemotherapy and trastuzumab for advanced HER2‐positive [immunohistochemistry (IHC) 3+ or 2+ with in‐situ hybridization (ISH) amplification] gastric or gastroesophageal cancers. Strict clinical criteria for defining acquired trastuzumab resistance were adopted. Loss of HER2 positivity and loss of HER2 over‐expression were defined as post‐trastuzumab IHC score <3+ and absence of ISH amplification, and IHC “downscoring” from 2+/3+ to 0/1+, respectively. HER2 IHC was always performed, while ISH was missing in 3 post‐progression samples. Patients with initial HER2 IHC score 3+ and 2+ were 14 (64%) and 8 (36%), respectively. Loss of HER2 positivity and HER2 over‐expression was observed in 32 and 32% samples, respectively. The chance of HER2 loss was not associated with any of the baseline clinicopathological variables. The only exception was in patients with initial IHC score 2+ versus 3+, for both endpoints of HER2 positivity (80 vs. 14%; p = 0.008) and HER2 over‐expression (63 vs. 14%; p = 0.025). As already shown in breast cancer, loss of HER2 may be observed also in gastric cancers patients treated with trastuzumab‐based chemotherapy in the clinical practice. This phenomenon may be one of the biological reasons explaining the failure of anti‐HER2 second‐line strategies in initially HER2‐positive disease.


International Journal of Surgery | 2016

Pancreatic neuroendocrine tumors: Nosography, management and treatment.

Michele Orditura; Angelica Petrillo; Jole Ventriglia; A. Diana; Maria Maddalena Laterza; Alessio Fabozzi; B. Savastano; Elisena Franzese; Giovanni Conzo; Luigi Santini; Fortunato Ciardiello; Ferdinando De Vita

Pancreatic neuroendocrine tumors (pNETs) represent about 7% of all NETs, 8.7% of gastroenteropancreatic NETs (GEP-NETs) and 1-2% of all pancreatic neoplasms. In the last two decades, the increased diagnosis of pNETs has generated great interest and the development of different classifications, grading and staging systems. Recently, several trials were performed in order to improve the knowledge of biomarkers and imaging and to provide an early diagnosis, but their role is still under debate. Nowadays, surgery represents the only curative approach for pNETs. Approximately 90% of pNETs are silent and non-functional; therefore, most patients are diagnosed in late stage and present metastatic (60%) or locally unresectable advanced disease (21%) with a poor prognosis. Not many therapeutic options are available for pNETs, with different treatments for G1-G2 and G3 tumors, because these diseases are still rare and trials are made up of few series of patients. At present, medical treatments is controversial. On these bases, we believe that a multidisciplinary team composed of surgeons, oncologists, endocrinologists, radiation oncologists, radiologists, pathologists and medicals nuclear is required. This paper presents a review of present state-of-the-art in the field of pNETs.


Current Oncology | 2014

Effect of preoperative chemoradiotherapy on outcome of patients with locally advanced esophagogastric junction adenocarcinoma—a pilot study

Michele Orditura; Gennaro Galizia; N. Di Martino; E. Ancona; Carlo Castoro; R. Pacelli; Floriana Morgillo; S. Rossetti; V. Gambardella; Antonio Farella; Maria Maddalena Laterza; A. Ruol; Alessio Fabozzi; V. Napolitano; Francesco Iovino; Eva Lieto; L Fei; Giovanni Conzo; Fortunato Ciardiello; F. De Vita

BACKGROUND To date, few studies of preoperative chemotherapy or chemoradiotherapy (crt) in gastroesophageal junction (gej) cancer have been statistically powered; indeed, gej tumours have thus far been grouped with esophageal or gastric cancer in phase iii trials, thereby generating conflicting results. METHODS We studied 41 patients affected by locally advanced Siewert type i and ii gej adenocarcinoma who were treated with a neoadjuvant crt regimen [folfox4 (leucovorin-5-fluorouracil-oxaliplatin) for 4 cycles, and concurrent computed tomography-based three-dimensional conformal radiotherapy delivered using 5 daily fractions of 1.8 Gy per week for a total dose of 45 Gy], followed by surgery. Completeness of tumour resection (performed approximately 6 weeks after completion of crt), clinical and pathologic response rates, and safety and outcome of the treatment were the main endpoints of the study. RESULTS All 41 patients completed preoperative treatment. Combined therapy was well tolerated, with no treatment-related deaths. Dose reduction was necessary in 8 patients (19.5%). After crt, 78% of the patients showed a partial clinical response, 17% were stable, and 5% experienced disease progression. Pathology examination of surgical specimens demonstrated a 10% complete response rate. The median and mean survival times were 26 and 36 months respectively (95% confidence interval: 14 to 37 months and 30 to 41 months respectively). On multivariate analysis, TNM staging and clinical response were demonstrated to be the only independent variables related to long-term survival. CONCLUSIONS In our experience, preoperative chemoradiotherapy with folfox4 is feasible in locally advanced gej adenocarcinoma, but shows mild efficacy, as suggested by the low rate of pathologic complete response.


Current Oncology | 2013

Serum insulin-like growth factor 1 correlates with the risk of nodal metastasis in endocrine-positive breast cancer

Floriana Morgillo; F. De Vita; Giuliano Antoniol; Michele Orditura; Pasquale Pio Auriemma; M. R. Diadema; Eva Lieto; B. Savastano; Lucia Festino; Maria Maddalena Laterza; Alessio Fabozzi; Jole Ventriglia; Angelica Petrillo; Fortunato Ciardiello; Alfonso Barbarisi; Francesco Iovino

Increased insulin-like growth factor (igf) signalling has been observed in breast cancer, including endocrine-responsive cancers, and has been linked to disease progression and recurrence. In particular, igf-1 has the ability to induce and promote lymphangiogenesis through the induction of vascular endothelial growth factor C (vegfc). In the present study, we analyzed serum and tumour samples from 60 patients with endocrine-positive breast cancer to determine the expression and the possible relationship of circulating igf-1, igf binding protein 3 (igfbp3), and vegfc with the presence of lymphatic metastasis and other immunohistochemical parameters. The analysis revealed a clear and significant correlation between high basal levels of igf-1, igfbp3, and vegfc and lymph node metastasis in endocrine-responsive breast cancer. In addition, expression of those molecules was significantly higher in breast cancer patients than in healthy control subjects. Those findings may enable more accurate prediction of prognosis in patients with breast cancer.


Frontiers in Pharmacology | 2016

Axitinib after Sunitinib in metastatic renal cancer: Preliminary results from Italian "Real-World" SAX Study

Carmine D'Aniello; Maria Grazia Vitale; Azzurra Farnesi; Lorenzo Calvetti; Maria Maddalena Laterza; Carla Cavaliere; Chiara Della Pepa; Vincenza Conteduca; Anna Crispo; Ferdinando De Vita; Francesco Grillone; Enrico Ricevuto; Michele De Tursi; Rocco De Vivo; Marilena Di Napoli; Sabrina Chiara Cecere; Gelsomina Iovane; Alfonso Amore; Raffaele Piscitelli; Giuseppe Quarto; Salvatore Pisconti; Gennaro Ciliberto; Piera Maiolino; Paolo Muto; Sisto Perdonà; Massimiliano Berretta; Emanuele Naglieri; Luca Galli; Giacomo Cartenì; Ugo De Giorgi

Axitinib is an oral angiogenesis inhibitor, currently approved for treatment of metastatic renal cell carcinoma (mRCC) after failure of prior treatment with Sunitinib or cytokine. The present study is an Italian Multi-Institutional Retrospective Analysis that evaluated the outcomes of Axitinib, in second-line treatment of mRCC. The medical records of 62 patients treated with Axitinib, were retrospectively reviewed. The Progression Free Survival (PFS), the Overall Survival (OS), the Objective Response Rate (ORR), the Disease Control Rate (DCR), and the safety profile of axitinib and sunitinib–axitinib sequence, were the primary endpoint. The mPFS was 5.83 months (95% CI 3.93–7.73 months). When patients was stratified by Heng score, mPFS was 5.73, 5.83, 10.03 months according to poor, intermediate, and favorable risk group, respectively. The mOS from the start of axitinib was 13.3 months (95% CI 8.6–17.9 months); the observed ORR and DCR were 25 and 71%, respectively. When stratified patients by subgroups defined by duration of prior therapy with Sunitinib (≤ vs. >median duration), there was a statistically significant difference in mPFS with 8.9 (95% CI 4.39–13.40 months) vs. 5.46 months (95% CI 4.04–6.88 months) for patients with a median duration of Sunitinib >13.2 months. DCR and ORR to previous Sunitinib treatment was associated with longer statistically mPFS, 7.23 (95% CI 3.95–10.51 months, p = 0.01) and 8.67 (95% CI 4.0–13.33 months, p = 0.008) vs. 2.97 (95% CI 0.65–5.27 months, p = 0.01) and 2.97 months (95% CI 0.66–5.28 months, p = 0.01), respectively. Overall Axitinib at standard schedule of 5 mg bid, was well-tolerated. The most common adverse events of all grades were fatig (25.6%), hypertension (22.6%), gastro-intestinal disorders (25.9%), and hypothyroidism (16.1%). The sequence Sunitinib–Axitinib was well-tolerated without worsening in side effects, with a median OS of 34.7 months (95% CI 18.4–51.0 months). Our results are consistent with the available literature; this retrospective analysis confirms that Axitinib is effective and safe in routine clinical practice.


Clinical Cancer Research | 2017

Biomarkers of Primary Resistance to Trastuzumab in HER2-Positive Metastatic Gastric Cancer Patients: the AMNESIA Case-Control Study.

Filippo Pietrantonio; Giovanni Fucà; Federica Morano; Annunziata Gloghini; Simona Corso; Giuseppe Aprile; Federica Perrone; Ferdinando De Vita; Elena Tamborini; Gianluca Tomasello; Ambra Vittoria Gualeni; Elena Ongaro; Adele Busico; Elisa Giommoni; Chiara C. Volpi; Maria Maddalena Laterza; Salvatore Corallo; Michele Prisciandaro; Maria Antista; Alessandro Pellegrinelli; Lorenzo Castagnoli; Serenella M. Pupa; Giancarlo Pruneri; Filippo de Braud; Silvia Giordano; Chiara Cremolini; Maria Di Bartolomeo

Purpose: Refining the selection of HER2-positive metastatic gastric cancer patient candidates for trastuzumab is a challenge of precision oncology. Preclinical studies have suggested several genomic mechanisms of primary resistance, leading to activation of tyrosine kinase receptors other than HER2 or downstream signaling pathways. Experimental Design: We carried out this multicenter, prospective, case-control study to demonstrate the negative predictive impact of a panel of candidate genomic alterations (AMNESIA panel), including EGFR/MET/KRAS/PI3K/PTEN mutations and EGFR/MET/KRAS amplifications. Hypothesizing a prevalence of candidate alterations of 30% and 0% in resistant and sensitive HER2-positive patients, respectively, 20 patients per group were needed. Results: AMNESIA panel alterations were significantly more frequent in resistant (11 of 20, 55%) as compared with sensitive (0% of 17) patients (P < 0.001), and in HER2 IHC 2+ (7 of 13, 53.8%) than 3+ (4 of 24, 16.7%) tumors (P = 0.028). Patients with tumors bearing no candidate alterations had a significantly longer median progression-free [5.2 vs. 2.6 months; HR, 0.34; 95% confidence interval (CI), 0.07–0.48; P = 0.001] and overall survival (16.1 vs. 7.6 months; HR, 0.38; 95% CI, 0.09–0.75; P = 0.015). The predictive accuracy of the AMNESIA panel and HER2 IHC was 76% and 65%, respectively. The predictive accuracy of the combined evaluation of the AMNESIA panel and HER2 IHC was 84%. Conclusions: Our panel of candidate genomic alterations may be clinically useful to predict primary resistance to trastuzumab in patients with HER2-positive metastatic gastric cancer and should be further validated with the aim of molecularly stratifying HER2-addicted cancers for the development of novel treatment strategies. Clin Cancer Res; 24(5); 1082–9. ©2017 AACR.


Frontiers in Pharmacology | 2017

Sequential Treatment with Pazopanib and Everolimus in Metastatic Renal Cell Carcinoma

Sabrina Rossetti; Carmine D'Aniello; Gelsomina Iovane; Sarah Scagliarini; Maria Maddalena Laterza; Fernando De Vita; Clementina Savastano; Giacomo Cartenì; Maria Assunta Porricelli; Massimiliano Berretta; Salvatore Pisconti; Gaetano Facchini; Carla Cavaliere

In metastatic renal cell carcinoma, complete response to first-line antiangiogenic agents is rare and resistance to therapy often develops. Protocols for sequential treatment with angiogenesis and mTOR inhibitors are under evaluation to improve outcomes. In this observational, real-world study, patients received a first-line therapy with pazopanib until discontinuation for disease progression or toxicity, then a second-line with everolimus. Primary endpoints were overall survival (OS) for sequence, progression free survival (PFS) for each agent, and safety. Thirty-one patients were included in the analysis: 73.3% of patients underwent nephrectomy before treatment, 25.8% had at least three comorbidities. At the beginning of therapy, the median age was 68 years, with more than 60% of patients older than 65 years. The median OS for sequence was 26.5 months (95% CI 17.4-nc); median PFS was 10.6 months (95% CI 6.3–12.1) with pazopanib and 5.3 months (95% CI 3.8–6.7) with everolimus. The median persistence in pazopanib therapy was 8.1 months (Interquartile Range IQR 5.3–12.7), with 31% of patients who required dose reduction, while persistence in everolimus was 4.4 months (IQR 3.4–6.5). Sequence was well tolerated with a different profile of adverse events for each agent. These data confirmed that pazopanib was effective, even in reduced dosing, and well tolerated and suggested that everolimus may represent an opportunity to continue a therapy when patients cannot further tolerate angiogenesis inhibitors or develop a resistance.


Tumori | 2013

Gefitinib in non-small cell lung carcinoma: a case report of an unusual side effect and complete response in advanced disease

Maria Maddalena Laterza; Bruno Chiurazzi; MariaFiorella Brangi; Ferdinando Riccardi; Giacomo Cartenì

Gefitinib is a tyrosine kinase inhibitor, indicated in advanced non-small cell lung cancer in all lines of treatment for patients harboring EGFR mutations. It has a favorable toxicity profile but may induce unexpected adverse effects, such as an infiammatory reaction in the bladder. We report a rare case of hemorrhagic cystitis, an unusual side effect, in a patient with non-small cell lung cancer treated with gefitinib, which did not compromise the clinical response.


International Journal of Molecular Sciences | 2018

What’s New in Gastric Cancer: The Therapeutic Implications of Molecular Classifications and Future Perspectives

G. Tirino; L. Pompella; Angelica Petrillo; Maria Maddalena Laterza; Annalisa Pappalardo; Marianna Caterino; Michele Orditura; Fortunato Ciardiello; Gennaro Galizia; Ferdinando De Vita

Despite some remarkable innovations and the advent of novel molecular classifications the prognosis of patients with advanced gastric cancer (GC) remains overall poor and current clinical application of new advances is disappointing. During the last years only Trastuzumab and Ramucirumab have been approved and currently used as standard of care targeted therapies, but the systemic management of advanced disease did not radically change in contrast with the high number of molecular drivers identified. The Cancer Genome Atlas (TCGA) and Asian Cancer Research Group (ACRG) classifications paved the way, also for GC, to that more contemporary therapeutic approach called “precision medicine” even if tumor heterogeneity and a complex genetic landscape still represent a strong barrier. The identification of specific cancer subgroups is also making possible a better selection of patients that are most likely to respond to immunotherapy. This review aims to critically overview the available molecular classifications summarizing the main druggable molecular drivers and their possible therapeutic implications also taking advantage of new technologies and acquisitions.

Collaboration


Dive into the Maria Maddalena Laterza's collaboration.

Top Co-Authors

Avatar

Angelica Petrillo

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Jole Ventriglia

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

B. Savastano

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

F. De Vita

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

G. Tirino

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Michele Orditura

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Fortunato Ciardiello

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Alessio Fabozzi

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

Ferdinando De Vita

Seconda Università degli Studi di Napoli

View shared research outputs
Top Co-Authors

Avatar

A. Diana

Seconda Università degli Studi di Napoli

View shared research outputs
Researchain Logo
Decentralizing Knowledge