Network


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

Hotspot


Dive into the research topics where Mariacristina Di Marco is active.

Publication


Featured researches published by Mariacristina Di Marco.


Case Reports in Gastroenterology | 2011

Pancreatic Ductal Adenocarcinoma Associated with Autoimmune Pancreatitis

Raffaele Pezzilli; Silvia Vecchiarelli; Mariacristina Di Marco; Carla Serra; Donatella Santini; Lucia Calculli; Dario Fabbri; Betzabé Rojas Mena; Andrea Imbrogno

Autoimmune pancreatitis (AIP), in contrast to other benign chronic pancreatic diseases, can be cured with immunosuppressant drugs, thus the differentiation of AIP from pancreatic cancer is of particular interest in clinical practice. There is the possibility that some patients with AIP may develop pancreatic cancer, and this possibility contributes to increasing our difficulties in differentiating AIP from pancreatic cancer. We herein report the case of a 70-year-old man in whom pancreatic adenocarcinoma and AIP were detected simultaneously. We must carefully monitor AIP patients for the simultaneous presence of pancreatic cancer, even when a diagnosis of AIP is confirmed.


World Journal of Gastrointestinal Oncology | 2016

State of the art biological therapies in pancreatic cancer.

Mariacristina Di Marco; Elisa Grassi; Sandra Durante; Silvia Vecchiarelli; Andrea Palloni; Marina Macchini; Riccardo Casadei; Claudio Ricci; Riccardo Panzacchi; Donatella Santini; Guido Biasco

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies with a five-year survival rate of approximately 5%. Several target agents have been tested in PDAC, but almost all have failed to demonstrate efficacy in late phase clinical trials, despite the better understanding of PDAC molecular biology generated by large cancer sequencing initiatives in the past decade. Eroltinib (a small-molecule tyrosine-kinase inhibitor of epidermal growth factor receptor) plus gemcitabine is the only schedule with a biological agent approved for advanced pancreatic cancer, but it has resulted in a very modest survival benefit in unselected patients. In our work, we report a summary of the main clinical trials (closed and ongoing) that refer to biological therapy evaluation in pancreatic cancer treatment.


Cancer Cell | 2018

β2 Adrenergic-Neurotrophin Feedforward Loop Promotes Pancreatic Cancer

Bernhard W. Renz; Ryota Takahashi; Takayuki Tanaka; M Macchini; Yoku Hayakawa; Zahra Dantes; H. Carlo Maurer; Xiaowei Chen; Zhengyu Jiang; C. Benedikt Westphalen; Matthias Ilmer; Giovanni Valenti; Sarajo Mohanta; Andreas J.R. Habenicht; Moritz Middelhoff; Timothy H. Chu; Karan Nagar; Yagnesh Tailor; Riccardo Casadei; Mariacristina Di Marco; Axel Kleespies; Richard A. Friedman; Helen Remotti; Maximilian Reichert; Daniel L. Worthley; Jens Neumann; Jens Werner; Alina C. Iuga; Kenneth P. Olive; Timothy C. Wang

Catecholamines stimulate epithelial proliferation, but the role of sympathetic nerve signaling in pancreatic ductal adenocarcinoma (PDAC) is poorly understood. Catecholamines promoted ADRB2-dependent PDAC development, nerve growth factor (NGF) secretion, and pancreatic nerve density. Pancreatic Ngf overexpression accelerated tumor development in LSL-Kras+/G12D;Pdx1-Cre (KC) mice. ADRB2 blockade together with gemcitabine reduced NGF expression and nerve density, and increased survival of LSL-Kras+/G12D;LSL-Trp53+/R172H;Pdx1-Cre (KPC) mice. Therapy with a Trk inhibitor together with gemcitabine also increased survival of KPC mice. Analysis of PDAC patient cohorts revealed a correlation between brain-derived neurotrophic factor (BDNF) expression, nerve density, and increased survival of patients on nonselective β-blockers. These findings suggest that catecholamines drive a feedforward loop, whereby upregulation of neurotrophins increases sympathetic innervation and local norepinephrine accumulation.


Molecular Medicine Reports | 2015

Characterization of pancreatic ductal adenocarcinoma using whole transcriptome sequencing and copy number analysis by single-nucleotide polymorphism array

Mariacristina Di Marco; Annalisa Astolfi; Elisa Grassi; Silvia Vecchiarelli; Marina Macchini; Valentina Indio; Riccardo Casadei; Claudio Ricci; Marielda D'Ambra; Giovanni Taffurelli; Carla Serra; Giorgio Ercolani; Donatella Santini; Antonia D'Errico; Antonio Daniele Pinna; Francesco Minni; Sandra Durante; Laura Raffaella Martella; Guido Biasco

The aim of the current study was to implement whole transcriptome massively parallel sequencing (RNASeq) and copy number analysis to investigate the molecular biology of pancreatic ductal adenocarcinoma (PDAC). Samples from 16 patients with PDAC were collected by ultrasound‑guided biopsy or from surgical specimens for DNA and RNA extraction. All samples were analyzed by RNASeq performed at 75x2 base pairs on a HiScanSQ Illumina platform. Single‑nucleotide variants (SNVs) were detected with SNVMix and filtered on dbSNP, 1000 Genomes and Cosmic. Non‑synonymous SNVs were analyzed with SNPs&GO and PROVEAN. A total of 13 samples were analyzed by high resolution copy number analysis on an Affymetrix SNP array 6.0. RNAseq resulted in an average of 264 coding non‑synonymous novel SNVs (ranging from 146‑374) and 16 novel insertions or deletions (In/Dels) (ranging from 6‑24) for each sample, of which a mean of 11.2% were disease‑associated and somatic events, while 34.7% were frameshift somatic In/Dels. From this analysis, alterations in the known oncogenes associated with PDAC were observed, including Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations (93.7%) and inactivation of cyclin‑dependent kinase inhibitor 2A (CDKN2A) (50%), mothers against decapentaplegic homolog 4 (SMAD4) (50%), and tumor protein 53 (TP53) (56%). One case that was negative for KRAS exhibited a G13D neuroblastoma RAS viral oncogene homolog mutation. In addition, gene fusions were detected in 10 samples for a total of 23 different intra‑ or inter‑chromosomal rearrangements, however, a recurrent fusion transcript remains to be identified. SNP arrays identified macroscopic and cryptic cytogenetic alterations in 85% of patients. Gains were observed in the chromosome arms 6p, 12p, 18q and 19q which contain KRAS, GATA binding protein 6, protein kinase B and cyclin D3. Deletions were identified on chromosome arms 1p, 9p, 6p, 18q, 10q, 15q, 17p, 21q and 19q which involve TP53, CDKN2A/B, SMAD4, runt‑related transcription factor 2, AT‑rich interactive domain‑containing protein 1A, phosphatase and tensin homolog and serine/threonine kinase 11. In conclusion, genetic alterations in PDCA were observed to involve numerous pathways including cell migration, transforming growth factor‑β signaling, apoptosis, cell proliferation and DNA damage repair. However, signaling alterations were not observed in all tumors and key mutations appeared to differ between PDAC cases.


Oncologist | 2016

Membrane Localization of Human Equilibrative Nucleoside Transporter 1 in Tumor Cells May Predict Response to Adjuvant Gemcitabine in Resected Cholangiocarcinoma Patients

Giovanni Brandi; Marzia Deserti; Francesco Vasuri; Andrea Farioli; Alessio Degiovanni; Andrea Palloni; Giorgio Frega; Maria Aurelia Barbera; Stefania De Lorenzo; Ingrid Garajová; Mariacristina Di Marco; Antonio Daniele Pinna; Matteo Cescon; Alessandro Cucchetti; Giorgio Ercolani; Antonietta D’Errico-Grigioni; Maria Abbondanza Pantaleo; Guido Biasco; Simona Tavolari; Guiseppe Aprile; Stefano Cereda; Lorenzo Fornaro; Francesco Leone; Sara Lonardi; Daniele Santini; Nicola Silvestris; Enrico Vasile

BACKGROUND The use of gemcitabine as an adjuvant modality for cholangiocarcinoma (CC) is increasing, but limited data are available on predictive biomarkers of response. Human equilibrative nucleoside transporter 1 (hENT-1) is the major transporter involved in gemcitabine intracellular uptake. This study investigated the putative predictive role of hENT-1 localization in tumor cells of CC patients undergoing treatment with adjuvant gemcitabine. METHODS Seventy-one consecutive patients with resected CC receiving adjuvant gemcitabine at our center were retrospectively analyzed by immunohistochemistry for hENT-1 localization in tumor cells. The main outcome measure was disease-free survival (DFS). Hazard ratios (HRs) of relapse and associated 95% confidence intervals (CIs) were obtained from proportional hazards regression models stratified on quintiles of propensity score. RESULTS Twenty-three (32.4%) cases were negative for hENT-1, 22 (31.0%) were positive in the cytoplasm only, and 26 (36.6%) showed concomitant cytoplasm/membrane staining. Patients with membrane hENT-1 had a longer DFS (HR 0.49, 95% CI 0.24-0.99, p = .046) than those who were negative or positive only in the cytoplasm of tumor cells. Notably, the association between DFS and membrane hENT-1 was dependent on the number of gemcitabine cycles (one to two cycles: HR 0.96, 95% CI 0.34-2.68; three to four cycles: HR 0.99, 95% CI 0.34-2.90; five to six cycles: HR 0.27, 95% CI 0.10-0.77). CONCLUSION hENT-1 localization on tumor cell membrane may predict response to adjuvant gemcitabine in CC patients receiving more than four cycles of chemotherapy. Further prospective randomized trials on larger populations are required to confirm these preliminary results, so that optimal gemcitabine-based chemotherapy may be tailored for CC patients in the adjuvant setting. IMPLICATIONS FOR PRACTICE Gemcitabine is becoming an increasingly used adjuvant modality in cholangiocarcinoma (CC), but limited data are available on predictive biomarkers of response. In this study, patients receiving more than four cycles of adjuvant gemcitabine and harboring Human equilibrative nucleoside transporter 1 (hENT-1, the major transporter involved in gemcitabine intracellular uptake) on tumor cell membrane had a longer disease-free survival compared with patients negative or positive for hENT-1 only in the cytoplasm of tumor cells. Overall these results may lay the basis for further prospective randomized trials based on a larger population of patients and may prove useful for tailoring appropriate gemcitabine-based chemotherapy for CC patients in the adjuvant setting.


Journal of Gastrointestinal Surgery | 2016

Is total pancreatectomy as feasible, safe, efficacious, and cost-effective as pancreaticoduodenectomy? A single center, prospective, observational study

Riccardo Casadei; Claudio Ricci; Giovanni Taffurelli; Anna Guariniello; Anthony Di Gioia; Mariacristina Di Marco; Nico Pagano; Carla Serra; Lucia Calculli; Donatella Santini; Francesco Minni

BackgroundTotal pancreatectomy is actually considered a viable option in selected patients even if large comparative studies between partial versus total pancreatectomy are not currently available. Our aim was to evaluate whether total pancreatectomy can be considered as feasible, safe, efficacious, and cost-effective as pancreaticoduodenectomy.MethodsA single center, prospective, observational trial, regarding postoperative outcomes, long-term results, and cost-effectiveness, in a tertiary referral center was conducted, comparing consecutive patients who underwent elective total pancreatectomy and/or pancreaticoduodenectomy.ResultsSeventy-three consecutive elective total pancreatectomies and 184 pancreaticoduodenectomies were compared. There were no significant differences regarding postoperative outcomes and overall survival. The quality of life, evaluated in 119 patients according to the EQ-5D-5L questionnaire, showed that there were no significant differences regarding the five items considered. The mean EQ-5D-5L score was similar in the two procedures (total pancreatectomy = 0.872, range 0.345–1.000; pancreaticoduodenectomy = 0.832, range 0.393–1.000; P = 0.320). The impact of diabetes according to the Problem Areas in Diabetes (PAID) questionnaire did not show any significant differences except for question 13 (total pancreatectomy = 0.60; pancreaticoduodenectomy = 0.19; P = 0.022). The cost-effectiveness analysis suggested that the quality-adjusted life year was not significantly different between the two procedures (total pancreatectomy = 0.910, range 0.345–1.000; pancreaticoduodenectomy = 0.910, range −0.393–1.000; P = 0.320).ConclusionsFrom this study, it seems reasonable to suggest that total pancreatectomy can be considered as safe, feasible, and efficacious as PD and acceptable in terms of cost-effectiveness.


Case Reports in Gastroenterology | 2012

Preoperative Gemcitabine and Oxaliplatin in a Patient with Ovarian Metastasis from Pancreatic Cystadenocarcinoma

Mariacristina Di Marco; Silvia Vecchiarelli; Marina Macchini; Raffaele Pezzilli; Donatella Santini; Riccardo Casadei; Lucia Calculli; Sokol Sina; Riccardo Panzacchi; Claudio Ricci; Elisa Grassi; Francesco Minni; Guido Biasco

We describe a case of clinical benefit and partial response with gemcitabine and oxaliplatin (GEMOX) in a young patient with ovarian metastasis from cystadenocarcinoma of the pancreas. A young woman complained of abdominal pain and constipation. Computed tomography (CT) and magnetic resonance imaging scans disclosed two bilateral ovarian masses with pancreatic extension. She underwent bilateral ovarian and womb resection. During surgery peritoneal carcinosis, a pancreatic mass and multiple abdominal lesions were found. The final diagnosis was mucinous pancreatic cystadenocarcinoma with ovarian and peritoneal metastases. She started chemotherapy with GEMOX (gemcitabine 1,000 mg/m2/d1 and oxaliplatin 100 mg/m2/d2 every 2 weeks). After 12 cycles of chemotherapy a CT scan showed reduction of the pancreatic mass. She underwent distal pancreatic resection, regional lymphadenectomy and splenectomy. Pathologic examination documented prominent fibrous tissue and few neoplastic cells with mucin-filled cytoplasm. Chemotherapy was continued with gemcitabine as adjuvant treatment for another 3 cycles. There is currently no evidence of disease. As reported in the literature, GEMOX is associated with an improvement in progression-free survival and clinical benefit in patients with advanced pancreatic cancer. This is an interesting case in whom GEMOX transformed inoperable pancreatic cancer into a resectable tumor.


Journal of the Pancreas | 2012

Comparing RECIST and Choi’s Criteria to Evaluate Radiological Response to Chemotherapy in Patients with Advanced Pancreatic Cancer

Silvia Vecchiarelli; Marina Macchini; Claudio Ricci; Marielda D'Ambra; Riccardo Casadei; Lucia Calculli; Fabio Ferroni; Raffaele Pezzilli; Elisa Grassi; Francesco Minni; Guido Biasco; Mariacristina Di Marco

Context Assessment of response after chemotherapy (CTH) for pancreatic cancer (PC) is currently based on RECIST criteria. In 2007 Choi et al . published a new classification system. Objectives To evaluate the accuracy of the two classification systems for radiological response to CTH in patients affected by advanced PC. Methods From 2006 to 2011, 61 untreated patients affected by advanced pancreatic adenocarcinoma underwent palliative CTH. Thirty-seven (60.7 %) had a locally advanced PC and 24 (39.3%) a metastatic disease. All patients were treated with a bemcitabine-based CTH. We assessed radiological response after three months of first-line therapy applying both RECIST criteria and Choi’s criteria, which consider changes both in size and in density at CT. We evaluated the accuracy in restaging, comparing the class of response with overall survival (OS). OS was calculated with Kaplan-Meier method. The concordance with the two classification systems was evaluated with Kendall’s test. The accuracy in restaging was assessed through log rank test. Results At restaging, using RECIST criteria, we registered 6 (9.8%) patients with partial response (PR), 32 (52.5%) with stable disease (SD), and 23 (37.7%) with disease progression (PD). Instead Choi’s criteria assessed 18 PR (29.5%), 12 SD (19.7%) and 31 PD (50.8%). The concordance test showed that the two systems matched (P<0.001). Comparing each classification with OS, we observed that patients with different prognosis were better stratified with Choi’s criteria. Using RECIST criteria we did not found any significant difference in OS between patients with PR (12 months), SD (16 months) and PD (10 months). Using Choi’s criteria we found that OS in patients with PR was similar to patients with SD with 16 and 19 months (P=0.634). Patients with PR had an OS significantly higher than patients with PD (16 vs . 9 months; P=0.009; RR=2.3). Conclusions Choi’s criteria seem to better assess radiological response of CTH in PC patients than RECIST criteria. Due to the small number of patients, larger prospective studies are needed.


International Journal of Surgery | 2017

Prospective validation of a preoperative risk score model based on pancreatic texture to predict postoperative pancreatic fistula after pancreaticoduodenectomy

Riccardo Casadei; Claudio Ricci; Giovanni Taffurelli; Carlo Alberto Pacilio; Mariacristina Di Marco; Nico Pagano; Carla Serra; Lucia Calculli; Donatella Santini; Francesco Minni

BACKGROUND In 2015, basing on objective preoperative factors related to pancreas remnant texture (body mass index, Wirsung duct size and preoperative diagnosis), we proposed a score model to predict the risk of postoperative pancreatic fistula after partial pancreatectomies. The aim of the present study was to prospectively validate this preoperative predictive risk score for postoperative pancreatic fistula after pancreaticoduodenectomy. METHODS Prospective study of consecutive patients who underwent pancreaticoduodenectomy in which a preoperative risk score, based on factors related to the pancreatic texture, was calculated. The risk score model was tested by comparison with subjective intraoperative assessment of the pancreas remnant texture and drain amylase value on postoperative day 1. Sensitivity, specificity, positive and negative likelihood ratio and area under the curve were calculated. RESULTS Eighty-four patients who underwent pancreaticoduodnectomy were analyzed. Clinically relevant pancreatic fistulas rate was 40.6%. The risk score model with a cut-off of 6 increased the odds of pancreatic fistula approximately 3 fold but it was not independently related to it. On the contrary, considering a cut-off of 5, the risk score model increased the odds of pancreatic fistula 11-16 fold and it was independently related to it. The new risk score model and pancreatic texture had high sensitivity (97% and 88%, respectively) and low specificity (34% and 60%, respectively) while the amylase drain value had low sensitivity (44%) and high specificity (92%). CONCLUSIONS The preoperative risk score model with a cut-off of 5 was a useful predictor of clinically relevant pancreatic fistula after pancreaticoduodenectomy. The drain amylase value represents a complementary factor to the risk score in predicting a pancreatic fistula.


Oncotarget | 2016

Copy number gain of chromosome 3q is a recurrent event in patients with intraductal papillary mucinous neoplasm (IPMN) associated with disease progression

Sandra Durante; Silvia Vecchiarelli; Annalisa Astolfi; Elisa Grassi; Riccardo Casadei; Donatella Santini; Riccardo Panzacchi; Claudio Ricci; Salvatore Serravalle; Giuseppe Tarantino; Mirella Falconi; Gabriella Teti; Valentina Indio; Andrea Pession; Francesco Minni; Guido Biasco; Mariacristina Di Marco

Background Intraductal papillary mucinous neoplasm (IPMN) is the most common cystic preneoplastic lesion of pancreatic cancer. We used an approach coupling high resolution cytogenetic analysis (Affymetrix Oncoscan FFPE Array) with clinically-oriented bioinformatic interpretation of data to understand the most relevant alterations of precursor lesions at different stages to identify new diagnostic markers. Results We identified multiple copy number alterations, particularly in lesions with severe dysplasia, with 7 IPMN with low-intermediate dysplasia carrying a nearly normal karyotype and 13 IPMN with complex Karyotype (> 4 alterations), showing high grade dysplasia. A specific gain of chromosome arm 3q was found in IPMN with complex Karyotype (92%). This gain of 3q is particularly interesting for the presence of oncogenes such as PIK3CA, GATA2 and TERC that are part of pathways that deregulate cell growth and promote disease progression. Quantitative PCR and FISH analysis confirmed the data. Further demonstration of the overexpression of the PIK3CA gene supports the identification of this alteration as a possible biomarker in the early identification of patients with IPMN at higher risk for disease progression. Materials and methods High resolution cytogenetic analysis was performed in 20 formalin fixed paraffin embedded samples of IPMN by Oncoscan FFPE assay. Results were validated by qPCR and FISH analysis. Conclusions The identification of these markers at an early stage of disease onset could help to identify patients at risk for cancer progression and new candidates for a more specific targeted therapy.

Collaboration


Dive into the Mariacristina Di Marco's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge