Network


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

Hotspot


Dive into the research topics where Claudia Omarini is active.

Publication


Featured researches published by Claudia Omarini.


Annals of Oncology | 2013

Loss of HER2 positivity and prognosis after neoadjuvant therapy in HER2-positive breast cancer patients

Valentina Guarneri; M. V. Dieci; Elena Barbieri; Federico Piacentini; Claudia Omarini; Guido Ficarra; Stefania Bettelli; Pierfranco Conte

BACKGROUND Emerging literature data are showing that a change in human epidermal growth factor receptor (HER2) status adversely affects breast cancer patients prognosis. The aim of this study was to evaluate the prognostic impact of HER2 loss in patients with HER2-positive disease treated with neoadjuvant therapy with or without anti-HER2 agents. METHODS One hundred and seven consecutive HER2-positive patients were identified from a prospectively maintained database. The first cohort includes 40 patients treated with chemotherapy (CT) alone. The second cohort includes 67 patients treated with neoadjuvant CT plus anti-HER2 agents (trastuzumab and/or lapatinib). HER2 expression was evaluated by immunihistochemistry or fluorescence in situ hybridization on pretreatment core biopsy and on surgical specimen after therapy. RESULTS The rates of pathologic complete response (pCR) and breast-conserving surgery were higher in the CT + anti-HER2 cohort. A loss of HER2 expression was observed in 40% of the patients with residual disease after CT alone versus 14.7% of the patients after CT + anti-HER2 agents (P = 0.019). Patients not achieving a pCR have a significant increase in the risk of relapse when compared with those achieving a pCR (hazard ratio [HR] 9.55, P = 0.028). Patients with HER2 loss tended to have a higher risk of relapse as comparing to patients with maintained HER2 positivity (HR 2.41, P = 0.063). CONCLUSION The pCR is confirmed as a powerful predictor of long-term outcome. The rate of HER2 loss is higher in patients receiving neoadjuvant CT without anti-HER2 agents. HER2 status on residual disease after preoperative therapy can be helpful in selecting patients at different risk of relapse, to be included in prospective trial exploring further adjuvant therapy.


Journal of Clinical Pathology | 2012

Predictors of human epidermal growth factor receptor 2 fluorescence in-situ hybridisation amplification in immunohistochemistry score 2+ infiltrating breast cancer: a single institution analysis

Maria Vittoria Dieci; Elena Barbieri; Stefania Bettelli; Federico Piacentini; Claudia Omarini; Guido Ficarra; Sara Balduzzi; Massimo Dominici; Pierfranco Conte; Valentina Guarneri

Aims Eligibility for anti-human epidermal growth factor receptor 2 (HER2) treatments in breast cancer requires a correct HER2 status assessment. Testing guidelines recommend fluorescence in-situ hybridisation (FISH) for samples scored as 2+ by immunohistochemistry. This study investigates the correlation between pathological features and FISH amplification in HER2 2+ breast cancer cases. Methods 480 HER2 2+ breast cancer samples were included. The association between tumour grade, hormone receptor status, proliferation index (Ki67) and FISH amplification, using both US Food and Drug Administration (ratio ≥2) and American Society of Clinical Oncologists/College of American Pathologists cut-offs (ratio >2.2) was evaluated. Results 90.2% of the samples were hormone receptor positive. The median Ki67 value was 23.5%; 311 (64.8%) samples showed a Ki67 value of 15% or greater. Tumour grade was evaluable in 421 cases (87.7%), 268 (55.8%) being grade 3. FISH amplification rates were 27.5% (ratio ≥2.0) and 20.8% (ratio >2.2). Grade 3 tumours were more frequently amplified than grades 1–2 tumours: 34% versus 18% (ratio ≥2.0, p<0.001) and 27% versus 9% (ratio >2.2, p<0.001). Samples with Ki67 of 15% or greater showed higher amplification rates than low Ki67 samples: 31% versus 21% (ratio ≥2.0, p=0.022) and 25% versus 12% (ratio >2.2, p=0.003). The OR for FISH amplification was significant in the case of grade 3 and high Ki67 with both cut-offs. Conclusions In this study, high tumour grade and high Ki67 significantly predicted FISH amplification in 480 HER2 2+ breast cancer samples.


Cancer management and research | 2018

Neoadjuvant treatments in triple-negative breast cancer patients: where we are now and where we are going

Claudia Omarini; Giorgia Guaitoli; Stefania Pipitone; Luca Moscetti; Laura Cortesi; Stefano Cascinu; Federico Piacentini

Triple-negative breast cancer (TNBC) remains the poorest-prognosis breast cancer (BC) subtype. Gene expression profiling has identified at least six different triple-negative subtypes with different biology and sensitivity to therapies. The heterogeneous nature of TN tumors may justify the difficulty in treating this BC subtype. Several targeted agents have been investigated in clinical trials without demonstrating a clear survival benefit. Therefore, systemic chemotherapy remains the cornerstone of current clinical practice. Improving the knowledge of tumor biology is mandatory for patient management. In stages II and III, neoadjuvant systemic treatment is an effective option of care. The achievement of a pathological complete response represents an optimal surrogate for survival outcome as well as a test for tumor drug sensitivity. In this review, we provide a brief description of the main predictive biomarkers for tumor response to systemic treatment. Moreover, we review the treatment strategies investigated for TNBCs in neoadjuvant settings focusing on experimental drugs such as immunotherapy and poly [ADP-ribose] polymerase inhibitors that hold promise in the treatment of this aggressive disease. Therefore, the management of TNBC represents an urgent, current, unmet need in daily clinical practice. A key recommendation is to design biology-driven clinical trials wherein TNBC patients may be treated on the basis of tumor molecular profile.


Oncotarget | 2017

A retrospective multicentric observational study of trastuzumab emtansine in HER2 positive metastatic breast cancer: a real-world experience

Patrizia Vici; Laura Pizzuti; Andrea Michelotti; Isabella Sperduti; Lucia Mentuccia; Luigi Di Lauro; Domenico Sergi; Paolo Marchetti; Daniele Santini; Emanuela Magnolfi; Laura Iezzi; Luca Moscetti; Agnese Fabbri; Alessandra Cassano; Antonino Grassadonia; Claudia Omarini; Federico Piacentini; Andrea Botticelli; Ilaria Bertolini; Angelo Fedele Scinto; Germano Zampa; Maria Mauri; Loretta D’Onofrio; Valentina Sini; Maddalena Barba; Marcello Maugeri-Saccà; Ernesto Rossi; Elisabetta Landucci; Silverio Tomao; Antonio Maria Alberti

We addressed trastuzumab emtansine (T-DM1) efficacy in HER2+ metastatic breast cancer patients treated in real-world practice, and its activity in pertuzumab-pretreated patients. We conducted a retrospective, observational study involving 23 cancer centres, and 250 patients. Survival data were analyzed by Kaplan Meier curves and log rank test. Factors testing significant in univariate analysis were tested in multivariate models. Median follow-up was 15 months and median T-DM1 treatment-length 4 months. Response rate was 41.6%, clinical benefit 60.9%. Median progression-free and median overall survival were 6 and 20 months, respectively. Overall, no differences emerged by pertuzumab pretreatment, with median progression-free and median overall survival of 4 and 17 months in pertuzumab-pretreated (p=0.13), and 6 and 22 months in pertuzumab-naïve patients (p=0.27). Patients who received second-line T-DM1 had median progression-free and median overall survival of 3 and 12 months (p=0.0001) if pertuzumab-pretreated, and 8 and 26 months if pertuzumab-naïve (p=0.06). In contrast, in third-line and beyond, median progression-free and median overall survival were 16 and 18 months in pertuzumab-pretreated (p=0.05) and 6 and 17 months in pertuzumab-naïve patients (p=0.30). In multivariate analysis, lower ECOG performance status was associated with progression-free survival benefit (p<0.0001), while overall survival was positively affected by lower ECOG PS (p<0.0001), absence of brain metastases (p 0.05), and clinical benefit (p<0.0001). Our results are comparable with those from randomized trials. Further studies are warranted to confirm and interpret our data on apparently lower T-DM1 efficacy when given as second-line treatment after pertuzumab, and on the optimal sequence order.We addressed trastuzumab emtansine (T-DM1) efficacy in HER2+ metastatic breast cancer patients treated in real-world practice, and its activity in pertuzumab-pretreated patients. We conducted a retrospective, observational study involving 23 cancer centres, and 250 patients. Survival data were analyzed by Kaplan Meier curves and log rank test. Factors testing significant in univariate analysis were tested in multivariate models. Median follow-up was 15 months and median T-DM1 treatment-length 4 months. Response rate was 41.6%, clinical benefit 60.9%. Median progression-free and median overall survival were 6 and 20 months, respectively. Overall, no differences emerged by pertuzumab pretreatment, with median progression-free and median overall survival of 4 and 17 months in pertuzumab-pretreated (p=0.13), and 6 and 22 months in pertuzumab-naïve patients (p=0.27). Patients who received second-line T-DM1 had median progression-free and median overall survival of 3 and 12 months (p=0.0001) if pertuzumab-pretreated, and 8 and 26 months if pertuzumab-naïve (p=0.06). In contrast, in third-line and beyond, median progression-free and median overall survival were 16 and 18 months in pertuzumab-pretreated (p=0.05) and 6 and 17 months in pertuzumab-naïve patients (p=0.30). In multivariate analysis, lower ECOG performance status was associated with progression-free survival benefit (p<0.0001), while overall survival was positively affected by lower ECOG PS (p<0.0001), absence of brain metastases (p 0.05), and clinical benefit (p<0.0001). Our results are comparable with those from randomized trials. Further studies are warranted to confirm and interpret our data on apparently lower T-DM1 efficacy when given as second-line treatment after pertuzumab, and on the optimal sequence order.


American Journal of Case Reports | 2017

Metronomic Capecitabine Effectively Blocks Leptomeningeal Carcinomatosis From Breast Cancer: A Case Report and Literature Review

Michela Maur; Claudia Omarini; Federico Piacentini; Annalisa Fontana; Elisa Pettorelli; Stefano Cascinu

Patient: Female, 57 Final Diagnosis: Meningeal carcinomatosis from breast cancer Symptoms: Seizures Medication: — Clinical Procedure: — Specialty: Oncology Objective: Unusual clinical course Background: Meningeal carcinomatosis is a rare complication in breast cancer patients. At present, there are no defined guidelines for its management. The efficacy of systemic treatment seems to depend on its ability to cross the blood-brain-barrier and its interaction with tumor vasculature. Metronomic chemotherapy is a known modality of drug administration able to inhibit tumor angiogenesis. Case Report: We present a case of symptomatic leptomeningeal carcinomatosis from breast cancer successfully treated with capecitabine. Based on the hypothesis that angiogenesis contributes to neoplastic meningitis, the patient was treated with a metronomic schedule that provided long-term clinical benefit with a very low toxicity profile. Conclusions: To assess the real impact of metronomic chemotherapy in patients with meninges involvement, a phase II study will be starting soon in our institution. A review of the literature concerning the management of meningeal carcinomatosis is also presented.


Tumori | 2013

Lapatinib and renal impairment: a case report

Federico Piacentini; Claudia Omarini; Elena Barbieri

This clinical report describes durable control of disease in a postmenopausal patient receiving hemodialysis and letrozole plus lapatinib since the diagnosis of HER2-positive, estrogen receptor-positive liver metastasis from breast cancer after anastrozole plus trastuzumab failure.


Cancer Biology & Therapy | 2018

Clinical and molecular predictors of long-term response in HER2 positive metastatic breast cancer patients

Claudia Omarini; Stefania Bettelli; Cecilia Caprera; Samantha Manfredini; F. Caggia; Giorgia Guaitoli; Luca Moscetti; Angela Toss; Laura Cortesi; Shaniko Kaleci; Antonino Maiorana; Stefano Cascinu; Pier Franco Conte; Federico Piacentini

ABSTRACT Background: HER2+ metastatic breast cancer (MBC) is a poor prognosis disease, unusually curable. To date, no predictive factors have been clearly correlated with long-term response to anti-HER2 agents. Methods: 54 HER2+ MBC patients treated with HER2 targeted therapy as first line treatment were analysed: 40 with a time to progression longer than 3 years in Long Responders (LR) group and 14 with a progression disease within one year of anti-HER2 therapy in a control group named Early Progressors (EP). The expression of 770 genes and 13 molecular pathways were evaluated using Nanostring PanCancer pathway panel performed on FFPE BC tissues. Results: Considering baseline patients and tumor characteristics, EP women had more CNS spread and more metastatic burden of disease compared to LR (p > 0.05). Gene expression analysis identified 30 genes with significantly different expression in the two cohorts; five were driver genes (BRCA1, PDGFRA, AR, PHF6 and MSH2). The majority of these genes were over-expressed, mainly in LR patients, and encoded growth factors, pro- or anti-inflammatory interleukins and DNA repair factors. Only four genes were down regulated, all in EP group (TNFSF10, CACNG1, IL20RB and BRCA1). Most of these genes were involved in MAPK and PI3K pathways. MAPK pathway was differently expressed between LR and EP (p = 0.05). PI3K was the only pathway overexpressed in EP patients. Conclusions: Whole genome expression analysis comparing LR vs. EP identified a group of genes that may predict more favourable long-term outcomes. Up-regulation of MAPK and down-regulation of PI3K pathway could be a positive predictive factors. Further clinical implications are warranted. Abbreviations: BC: breast cancer; MBC: metastatic breast cancer; LR: long responder; EP: early progressor; FFPE: formalin-fixed paraffin-embedded; CNS: central nervous system; PFS: progression free survival; OS: overall survival.


Breast Journal | 2017

Osteonecrosis of the Jaw in a Breast Cancer Patient Treated with Everolimus and a Single Dose of Zoledronic Acid

Claudia Omarini; M.E. Filieri; Roberta Depenni; Giulia Grizzi; Stefano Cascinu; Federico Piacentini

To the Editor: Bone is the most common site of metastatic recurrence in estrogen-receptor-positive breast cancer (BC) (1). Endocrine therapy associated with bisphosphonates is considered the mainstay of systemic treatment in skeletal metastatic BC. In recent years, everolimus combined with exemestane has become one of the most important therapeutic strategies in case of endocrine resistance disease (2). Everolimus inhibits mammalian target of rapamycin (mTOR), a serinethreonine kinase, downstream of PI3K/AKT pathway, resulting in a reduction in cancer cells proliferation and in a drop of production of vascular endothelial growth factor (VEGF) (3). Bisphosphonates are a class of synthetic drugs commonly used to treat bone metastasis (4). One known class-related side effect of bisphosphonates is osteonecrosis of the jaw (ONJ), an aseptic necrosis of either mandibular or maxilla with painful ulceration in the mouth, mainly associated with necrotic bone. ONJ occurs in 1.5–10% of cancer patients (5). The incidence is related to the time of exposure and the number of infusions. The precise pathogenesis of ONJ remains unknown. It has been suggested that bisphosphonates-related ONJ might be attributable to their anti-angiogenic activity. Bisphosphonates act by inhibiting vessels growth and inducing a durable reduction in circulating VEGF concentration (5). Concomitant treatment with VEGF-targeted agents seems to increase the risk of ONJ (6–8). In a large retrospective analysis, ONJ occurred in 1.1% of patients receiving bisphosphonates alone compared to 2% of patients treated with bevacizumab and bisphosphonates and 0.1% of patients receiving bevacizumab alone (9). Similar incidence has been reported in two randomized placebo-controlled trials (10). Concerning anti-VEGF tyrosine kinase inhibitors, only small series are available but the incidence of ONJ seems to be higher compared with the incidence in patients receiving anti-VEGF monoclonal antibody (6,8). To date, only two case reports of bisphosphonates and everolimus related ONJ have been published, both in patients treated for renal cell carcinoma (11,12). According with our knowledge, we report the first case of ONJ after the first infusion of zoledronic acid in a patient receiving everolimus and bisphosphonates for metastatic BC. In May 2006, a 46 years old lady underwent right mastectomy followed by adjuvant chemotherapy and endocrine treatment for hormonal receptors positive, HER2 negative BC. In October 2011 she developed multiple liver and lung metastasis. She was initiated on weekly paclitaxel and subsequent transition to aromatase inhibitor maintenance therapy. In December 2012, because of progression disease, she started everolimus 10 mg/daily and exemestane. In May 2014, symptomatic bone metastasis was detected. Palliative radiotherapy on the spine bone lesions was delivered together with systemic corticosteroids treatment. In June 2014 after panoramic x-ray and dental exam, patient received the first infusion of zoledronic acid. Neither mandibular pain nor bone and mucosal lesions were present when bisphosphonates were started. 1 month later the patient presented with jaw pain, swelling and chewing problem. Oral examination revealed an area of exposed necrotic bone associated with gingival fistula in the lower right posterior region of the jaw. To support the clinical evidence of ONJ, a dental cone beam computed tomography was performed (Figure 1). The patient was referred to maxillofacial surgeon who suggested nonsurgical management. She received antibiotics, disinfectant mouthwashes and appropriate analgesia with no complete resolution of the bone lesion. On the basis of the evidence that VEGF is essential for bone formation through up-regulation of Address correspondence and reprint requests to: Claudia Omarini, MD, Department of Medical Oncology, University Hospital of ModenaVia del Pozzo, 71 – 41124, Modena, Italy, or e-mail: [email protected]


Oncotarget | 2018

Genomic alterations at the basis of treatment resistance in metastatic breast cancer: Clinical applications

Angela Toss; Federico Piacentini; Laura Cortesi; Lucia Artuso; Isabella Bernardis; Sandra Parenti; Elena Tenedini; Guido Ficarra; Antonino Maiorana; Anna Iannone; Claudia Omarini; Luca Moscetti; Massimo Cristofanilli; Massimo Federico; Enrico Tagliafico

The standard of care for breast cancer has gradually evolved from empirical treatments based on clinical-pathological characteristics to the use of targeted approaches based on the molecular profile of the tumor. Consequently, an increasing number of molecularly targeted drugs have been developed. These drugs target specific alterations, called driver mutations, which confer a survival advantage to cancer cells. To date, the main challenge remains the identification of predictive biomarkers for the selection of the optimal treatment. On this basis, we evaluated a panel of 25 genes involved in the mechanisms of targeted treatment resistance, in 16 primary breast cancers and their matched recurrences, developed during treatment. Overall, we found a detection rate of mutations higher than that described in the literature. In particular, the most frequently mutated genes were ERBB2 and those involved in the PI3K/AKT/mTOR and the MAPK signaling pathways. The study revealed substantial discordances between primary tumors and metastases, stressing the need for analysis of metastatic tissues at recurrence. We observed that 85.7% of patients with an early-stage or locally advanced primary tumor showed at least one mutation in the primary tumor. This finding could explain the subsequent relapse and might therefore justify more targeted adjuvant treatments. Finally, the mutations detected in 50% of relapsed tissues could have guided subsequent treatment choices in a different way. This study demonstrates that mutation events may be present at diagnosis or arise during cancer treatment. As a result, profiling primary and metastatic tumor tissues may be a major step in defining optimal treatments.


Breast Cancer: Targets and Therapy | 2018

Spotlight on triptorelin in the treatment of premenopausal women with early-stage breast cancer

Marta Venturelli; Giorgia Guaitoli; Claudia Omarini; Luca Moscetti

Endocrine treatment represents the cornerstone of endocrine-sensitive premenopausal early breast cancer. The estrogen blockade plays a leading role in the therapeutic management of hormone receptor-positive breast cancer together with surgery, radiotherapy, and selective antiestrogen treatments. For several years, selective estrogen receptor modulators, such as tamoxifen, have represented the mainstay of therapy. The role of amenorrhea has been extensively elucidated in the past year: the benefit observed with chemotherapy-induced amenorrhea has strengthened its therapeutic role. Luteinizing hormone-releasing hormone (LHRH) has been introduced in oncology practice to induce amenorrhea in order to increase the advantage obtained from endocrine treatment. Triptorelin is one of the most widely used LHRH analogs currently available in clinical practice. It was recently investigated in two major clinical trials that studied the role of complete estrogen blockade in the premenopausal setting. Both showed the clinical benefit due to ovarian suppression treatment, primarily in high-risk patients. Furthermore, triptorelin and other LHRH analogs have recently been investigated in the attempt to preserve the ovarian function in young patients. The medical treatment of early breast cancer is always evolving in the effort to search for safe and efficacious treatments. The role of LHRH analogs is actually well recognized as contributing to the improvement of the medical treatment of premenopausal women with early breast cancer.

Collaboration


Dive into the Claudia Omarini's collaboration.

Top Co-Authors

Avatar

Federico Piacentini

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Stefano Cascinu

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Stefania Bettelli

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Guido Ficarra

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Antonino Maiorana

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

F. Caggia

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Samantha Manfredini

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Angela Toss

University of Modena and Reggio Emilia

View shared research outputs
Top Co-Authors

Avatar

Cecilia Caprera

University of Modena and Reggio Emilia

View shared research outputs
Researchain Logo
Decentralizing Knowledge