Network


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

Hotspot


Dive into the research topics where Lorenzo D'Ambrosio is active.

Publication


Featured researches published by Lorenzo D'Ambrosio.


Lancet Oncology | 2015

Sorafenib and everolimus for patients with unresectable high-grade osteosarcoma progressing after standard treatment: a non-randomised phase 2 clinical trial

Giovanni Grignani; Emanuela Palmerini; Virginia Ferraresi; Lorenzo D'Ambrosio; Rossella Bertulli; Sebastian Dorin Asaftei; Angela Tamburini; Ymera Pignochino; Dario Sangiolo; Emanuela Marchesi; Federica Capozzi; R. Biagini; Marco Gambarotti; Franca Fagioli; Paolo G. Casali; Piero Picci; Stefano Ferrari; Massimo Aglietta

BACKGROUND Results of previous study showed promising but short-lived activity of sorafenib in the treatment of patients with unresectable advanced and metastatic osteosarcoma. This treatment failure has been attributed to the mTOR pathway and might therefore be overcome with the addition of mTOR inhibitors. We aimed to investigate the activity of sorafenib in combination with everolimus in patients with inoperable high-grade osteosarcoma progressing after standard treatment. METHODS We did this non-randomised phase 2 trial in three Italian Sarcoma Group centres. We enrolled adults (≥18 years) with relapsed or unresectable osteosarcoma progressing after standard treatment (methotrexate, cisplatin, and doxorubicin, with or without ifosfamide). Patients received 800 mg sorafenib plus 5 mg everolimus once a day until disease progression or unacceptable toxic effects. The primary endpoint was 6 month progression-free survival (PFS). All analyses were intention-to-treat. This trial is registered with ClinicalTrials.gov, number NCT01804374. FINDINGS We enrolled 38 patients between June 16, 2011, and June 4, 2013. 17 (45%; 95% CI 28-61) of 38 patients were progression free at 6 months. Toxic effects led to dose reductions, or short interruptions, or both in 25 (66%) of 38 patients and permanent discontinuation for two (5%) patients. The most common grade 3-4 adverse events were lymphopenia and hypophosphataemia each in six (16%) patients, hand and foot syndrome in five (13%), thrombocytopenia in four (11%), and fatigue, oral mucositis, diarrhoea, and anaemia each in two (5%). One patient (3%) had a grade 3 pneumothorax that required trans-thoracic drainage, and that recurred at the time of disease progression. This was reported as a serious adverse event related to the study drugs in both instances. No other serious adverse events were reported during the trial. There were no treatment-related deaths. INTERPRETATION Although the combination of sorafenib and everolimus showed activity as a further-line treatment for patients with advanced or unresectable osteosarcoma, it did not attain the prespecified target of 6 month PFS of 50% or greater. FUNDING Italian Sarcoma Group.


Clinical Cancer Research | 2013

The Combination of Sorafenib and Everolimus Abrogates mTORC1 and mTORC2 Upregulation in Osteosarcoma Preclinical Models

Ymera Pignochino; Carmine Dell'Aglio; Marco Basiricò; Federica Capozzi; Marco Soster; Serena Marchiò; Stefania Bruno; Loretta Gammaitoni; Dario Sangiolo; Erica Torchiaro; Lorenzo D'Ambrosio; Franca Fagioli; Stefano Ferrari; Marco Alberghini; Piero Picci; Massimo Aglietta; Giovanni Grignani

Purpose: The multikinase inhibitor sorafenib displays antitumor activity in preclinical models of osteosarcoma. However, in sorafenib-treated patients with metastatic-relapsed osteosarcoma, disease stabilization and tumor shrinkage were short-lived and drug resistance occurred. We explored the sorafenib treatment escape mechanisms to overcome their drawbacks. Experimental Design: Immunoprecipitation, Western blotting, and immunohistochemistry were used to analyze the mTOR pathway [mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2)]. Cell viability, colony growth, and cell migration were evaluated in different osteosarcoma cell lines (MNNG-HOS, HOS, KHOS/NP, MG63, U-2OS, SJSA-1, and SAOS-2) after scalar dose treatment with sorafenib (10–0.625 μmol/L), rapamycin-analog everolimus (100–6.25 nmol/L), and combinations of the two. Cell cycle, reactive oxygen species (ROS) production, and apoptosis were assessed by flow cytometry. Nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice injected with MNNG-HOS cells were used to determine antitumor and antimetastatic effects. Angiogenesis and vascularization were evaluated in vitro by exploiting endothelial branching morphogenesis assays and in vivo in xenografted mice and chorioallantoic membranes. Results: After sorafenib treatment, mTORC1 signaling was reduced (downstream target P-S6), whereas mTORC2 was increased (phospho-mTOR Ser2481) in MNNG-HOS xenografts compared with vehicle-treated mice. Combining sorafenib with everolimus resulted in complete abrogation of both mTORC1 [through ROS-mediated AMP-activated kinase (AMPK) activation] and mTORC2 (through complex disassembly). The sorafenib/everolimus combination yielded: (i) enhanced antiproliferative and proapoptotic effects, (ii) impaired tumor growth, (iii) potentiated antiangiogenesis, and (iv) reduced migratory and metastatic potential. Conclusion: mTORC2 activation is an escape mechanism from sorafenib treatment. When sorafenib is combined with everolimus, its antitumor activity is increased by complete inhibition of the mTOR pathway in the preclinical setting. Clin Cancer Res; 19(8); 2117–31. ©2013 AACR.


Cancer Research | 2014

Cytokine-Induced Killer Cells Eradicate Bone and Soft-Tissue Sarcomas

Dario Sangiolo; Giulia Mesiano; Loretta Gammaitoni; Valeria Leuci; Maja Todorovic; Lidia Giraudo; Cristina Cammarata; Carmine Dell'Aglio; Lorenzo D'Ambrosio; Alberto Pisacane; Ivana Sarotto; Sara Miano; Ivana Ferrero; Fabrizio Carnevale-Schianca; Ymera Pignochino; Francesco Sassi; Andrea Bertotti; Wanda Piacibello; Franca Fagioli; Massimo Aglietta; Giovanni Grignani

Unresectable metastatic bone sarcoma and soft-tissue sarcomas (STS) are incurable due to the inability to eradicate chemoresistant cancer stem-like cells (sCSC) that are likely responsible for relapses and drug resistance. In this study, we investigated the preclinical activity of patient-derived cytokine-induced killer (CIK) cells against autologous bone sarcoma and STS, including against putative sCSCs. Tumor killing was evaluated both in vitro and within an immunodeficient mouse model of autologous sarcoma. To identify putative sCSCs, autologous bone sarcoma and STS cells were engineered with a CSC detector vector encoding eGFP under the control of the human promoter for OCT4, a stem cell gene activated in putative sCSCs. Using CIK cells expanded from 21 patients, we found that CIK cells efficiently killed allogeneic and autologous sarcoma cells in vitro. Intravenous infusion of CIK cells delayed autologous tumor growth in immunodeficient mice. Further in vivo analyses established that CIK cells could infiltrate tumors and that tumor growth inhibition occurred without an enrichment of sCSCs relative to control-treated animals. These results provide preclinical proof-of-concept for an effective strategy to attack autologous sarcomas, including putative sCSCs, supporting the clinical development of CIK cells as a novel class of immunotherapy for use in settings of untreatable metastatic disease.


Journal of Clinical Oncology | 2011

Complete Resolution of Life-Threatening Bleomycin-Induced Pneumonitis After Treatment With Imatinib Mesylate in a Patient With Hodgkin's Lymphoma: Hope for Severe Chemotherapy-Induced Toxicity?

Fabrizio Carnevale-Schianca; Susanna Gallo; Delia Rota-Scalabrini; Dario Sangiolo; Marco Fizzotti; Daniela Caravelli; Antonio Capaldi; Giancarlo Anselmetti; Erica Palesandro; Lorenzo D'Ambrosio; Valentina Coha; Remo Obert; Massimo Aglietta; Giovanni Grignani

Introduction Bleomycin-induced pneumonitis (BIP) is a scarcely manageable pulmonary toxicity that occurs in approximately 20% of patients who are affected with cancers that are treated with bleomycin-containing regimens. Generally, the clinical picture is extremely complicated and therapy is based on steroids. The overall response rate to such therapy is limited. Recent insights in BIP pathogenesis have indicated that a key feature is deregulated mechanisms of tissue repair that are driven by profibrotic cytokines. Here we describe a patient with lifethreatening BIP who was completely cured with imatinib mesylate (IM) after steroids and all other therapies had failed.


Molecular Cancer | 2017

PARP1 expression drives the synergistic antitumor activity of trabectedin and PARP1 inhibitors in sarcoma preclinical models

Ymera Pignochino; Federica Capozzi; Lorenzo D'Ambrosio; Carmine Dell'Aglio; Marco Basiricò; Marta Canta; Annalisa Lorenzato; Francesca Vignolo Lutati; Sandra Aliberti; Erica Palesandro; Paola Boccone; Danilo Galizia; Sara Miano; Giulia Chiabotto; Lucia Napione; Loretta Gammaitoni; Dario Sangiolo; Maria Serena Benassi; Barbara Pasini; Giovanna Chiorino; Massimo Aglietta; Giovanni Grignani

BackgroundEnhancing the antitumor activity of the DNA-damaging drugs is an attractive strategy to improve current treatment options. Trabectedin is an isoquinoline alkylating agent with a peculiar mechanism of action. It binds to minor groove of DNA inducing single- and double-strand-breaks. These kinds of damage lead to the activation of PARP1, a first-line enzyme in DNA-damage response pathways. We hypothesized that PARP1 targeting could perpetuate trabectedin-induced DNA damage in tumor cells leading finally to cell death.MethodsWe investigated trabectedin and PARP1 inhibitor synergism in several tumor histotypes both in vitro and in vivo (subcutaneous and orthotopic tumor xenografts in mice). We searched for key determinants of drug synergism by comparative genomic hybridization (aCGH) and gene expression profiling (GEP) and validated their functional role.ResultsTrabectedin activated PARP1 enzyme and the combination with PARP1 inhibitors potentiated DNA damage, cell cycle arrest at G2/M checkpoint and apoptosis, if compared to single agents. Olaparib was the most active PARP1 inhibitor to combine with trabectedin and we confirmed the antitumor and antimetastatic activity of trabectedin/olaparib combination in mice models. However, we observed different degree of trabectedin/olaparib synergism among different cell lines. Namely, in DMR leiomyosarcoma models the combination was significantly more active than single agents, while in SJSA-1 osteosarcoma models no further advantage was obtained if compared to trabectedin alone. aCGH and GEP revealed that key components of DNA-repair pathways were involved in trabectedin/olaparib synergism. In particular, PARP1 expression dictated the degree of the synergism. Indeed, trabectedin/olaparib synergism was increased after PARP1 overexpression and reduced after PARP1 silencing.ConclusionsPARP1 inhibition potentiated trabectedin activity in a PARP1-dependent manner and PARP1 expression in tumor cells might be a useful predictive biomarker that deserves clinical evaluation.


Biology of Blood and Marrow Transplantation | 2017

Post-Transplant Cyclophosphamide and Tacrolimus–Mycophenolate Mofetil Combination Prevents Graft-versus-Host Disease in Allogeneic Peripheral Blood Hematopoietic Cell Transplantation from HLA-Matched Donors

Fabrizio Carnevale-Schianca; Daniela Caravelli; Susanna Gallo; Valentina Coha; Lorenzo D'Ambrosio; Elena Vassallo; Marco Fizzotti; Francesca Nesi; Luisa Gioeni; Massimo Berger; Alessandra Polo; Loretta Gammaitoni; Paolo Becco; Lidia Giraudo; Monica Mangioni; Dario Sangiolo; Giovanni Grignani; Delia Rota Scalabrini; Antonino Sottile; Franca Fagioli; Massimo Aglietta

Allogeneic hematopoietic cell transplant (HCT) remains the only curative therapy for many hematologic malignancies but it is limited by high nonrelapse mortality (NRM), primarily from unpredictable control of graft-versus-host disease (GVHD). Recently, post-transplant cyclophosphamide demonstrated improved GVHD control in allogeneic bone marrow HCT. Here we explore cyclophosphamide in allogeneic peripheral blood stem cell transplantation (alloPBSCT). Patients with high-risk hematologic malignancies received alloPBSCT from HLA-matched unrelated/related donors. GVHD prophylaxis included combination post-HCT cyclophosphamide 50 mg/kg (days +3 and +4) and tacrolimus/mofetil mycophenolate (T/MMF) (day +5 forward). The primary objective was the cumulative incidence of acute and chronic GVHD. Between March 2011 and May 2015, 35 consecutive patients received the proposed regimen. MMF was stopped in all patients at day +28; the median discontinuation of tacrolimus was day +113. Acute and chronic GVHD cumulative incidences were 17% and 7%, respectively, with no grade IV GVHD events, only 2 patients requiring chronic GVHD immunosuppression control, and no deaths from GVHD. Two-year NRM, overall survival, event-free survival, and chronic GVHD event-free survival rates were 3%, 77%, 54%, and 49%, respectively. The graft-versus-tumor effect was maintained as 5 of 15 patients (33%) who received HCT with evidence of disease experienced further disease response. A post-transplant cyclophosphamide + T/MMF combination strategy effectively prevented acute and chronic GVHD after alloPBSCT from HLA-matched donors and achieved an unprecedented low NRM without losing efficacy in disease control or impaired development of the graft-versus-tumor effect. This trial is registered at clinicaltrials.gov as NCT02300571.


The New England Journal of Medicine | 2014

Anticoagulation for central venous catheters in patients with Cancer

Lorenzo D'Ambrosio; Massimo Aglietta; Giovanni Grignani

Data are not clear about whether patients with central venous catheters should receive anticoagulation to prevent catheter clots. A large randomized trial is needed.


Lancet Oncology | 2018

Trabectedin and olaparib in patients with advanced and non-resectable bone and soft-tissue sarcomas (TOMAS): an open-label, phase 1b study from the Italian Sarcoma Group

Giovanni Grignani; Lorenzo D'Ambrosio; Ymera Pignochino; Emanuela Palmerini; Massimo Zucchetti; Paola Boccone; Sandra Aliberti; Silvia Stacchiotti; Rossella Bertulli; Raimondo Piana; Sara Miano; Francesco Tolomeo; Giulia Chiabotto; Dario Sangiolo; Alberto Pisacane; Angelo Paolo Dei Tos; Luca Novara; Alice Bartolini; Emanuela Marchesi; Maurizio D'Incalci; Alberto Bardelli; Piero Picci; Stefano Ferrari; Massimo Aglietta

BACKGROUND Trabectedin is an alkylating drug with a unique mechanism of action causing single-strand and double-strand DNA breaks that activate DNA damage-response pathways. Based on our preclinical data, we hypothesised that poly(ADP-ribose) polymerase 1 (PARP1) inhibitors might be an ideal partner of trabectedin and aimed to assess the safety, identify the recommended phase 2 dose, and explore preliminary signs of activity of trabectedin and olaparib combination treatment in patients with bone and soft-tissue sarcoma. METHODS We did an open-label, multicentre, phase 1b study, recruiting patients from the national Italian sarcoma network aged 18 years and older with histologically confirmed bone and soft-tissue sarcoma progressing after standard treatments with Eastern Cooperative Oncology Group performance status of 1 or less. In a classic 3 + 3 design, patients received a 24 h infusion of trabectedin on day 1 and olaparib orally twice a day in 21-day cycles across six dose levels (trabectedin 0·675-1·3 mg/m2 every 3 weeks; olaparib 100-300 mg twice a day from day 1 to 21). Intermediate dose levels were permitted to improve safety and tolerability. The primary endpoint was determination of the recommended phase 2 dose (the maximum tolerated dose). Safety and antitumour activity were assessed in all patients who received at least one dose of the study drugs. We report the results of the dose-escalation and dose-expansion cohorts. The trial is still active but closed to enrolment, and follow-up for patients who completed treatment is ongoing. This trial is registered with ClinicalTrials.gov, number NCT02398058. FINDINGS Between Nov 17, 2014, and Jan 30, 2017, of 54 patients assessed for eligibility, we enrolled 50 patients: 28 patients in the dose-escalation cohort and 22 patients in the dose-expansion cohort. Patients received a median of four cycles of treatment (IQR 2-6; range 1-17 [the patients who received the highest number of cycles are still on treatment]) with a median follow-up of 10 months (IQR 5-23). Considering all dose levels, the most common grade 3-4 adverse events were lymphopenia (32 [64%] of 50 patients), neutropenia (31 [62%]), thrombocytopenia (14 [28%]), anaemia (13 [26%]), hypophosphataemia (20 [40%]), and alanine aminotransferase concentration increase (9 [18%]). No treatment-related life-threatening adverse events or deaths occurred. One (2%) patient interrupted treatment without progression without reporting any specific toxicity. Observed dose-limiting toxicities were thrombocytopenia, neutropenia for more than 7 days, and febrile neutropenia. We selected intermediate dose level 4b (trabectedin 1·1 mg/m2 every 3 weeks plus olaparib 150 mg twice a day) as the recommended phase 2 dose. Seven (14%; 95% CI 6-27) of 50 patients achieved a partial response according to Response Evaluation Criteria In Solid Tumors 1.1. INTERPRETATION Trabectedin and olaparib in combination showed manageable toxicities at active dose levels for both drugs. Preliminary data on antitumour activity are encouraging. Two dedicated phase 2 studies are planned to assess activity of this combination in both ovarian cancer (EudraCT2018-000230-35) and soft-tissue sarcomas. FUNDING Italian Association for Cancer Research, Italian Sarcoma Group, Foundation for Research on Musculoskeletal and Rare Tumors, and Italian Ministry of Health.


Ejso | 2018

Pancreaticoduodenectomy in the surgical management of primary retroperitoneal sarcoma

William W. Tseng; Denice D. Tsao-Wei; Dario Callegaro; Giovanni Grignani; Lorenzo D'Ambrosio; Sylvie Bonvalot; Cecilia G. Ethun; Kenneth Cardona; John T. Mullen; Robert J. Canter; John E. Mullinax; Ricardo J. Gonzalez; Frits van Coevorden; Markus Albertsmeier; Kiran Dhanireddy; Salvatore Lorenzo Renne; Alessandro Gronchi

BACKGROUND In retroperitoneal sarcoma (RPS), the optimal extent of resection must balance adequate disease control with potential for morbidity. We sought to study the frequency and outcomes after a Whipple procedure or pancreaticoduodenectomy (PD) in patients undergoing resection for primary RPS. METHODS Participating referral centers within the Trans-Atlantic Retroperitoneal Sarcoma Working Group provided retrospective data from January 2007 to December 2016 for patients with primary RPS who underwent PD along with the total number of consecutive resections done during the same time period. Data from participating centers were combined for analysis. RESULTS In total, 29 patients underwent PD among 2068 resections performed for primary RPS (1.4%). The predominant histologic subtypes were liposarcoma and leiomyosarcoma. All PD patients underwent concomitant resection of additional organs (median: 2, range: 1-5), including 13 patients (45%) who also received vena cava resection. Definitive evidence of microscopic invasion of the duodenum or pancreas was seen in 84% of patients. Postoperatively, 10 patients (34%) had major complications including 8 (28%) that developed a clinically-significant pancreatic leak. One postoperative death (3.4%) occurred. With a median follow-up of 4.8 years, 19 patients (66%) developed disease recurrence. The patterns of recurrence were dependent on histologic subtype. CONCLUSION Although infrequent, when PD is done for primary RPS, resection of additional organs is often required and major complication rates are moderate. The recurrence rate is overall high and the pattern of recurrence is dictated by histologic subtype.


European Journal of Cancer | 2012

350 Poly (ADP-ribose) Polymerase-1 (PARP-1) Inhibitors Potentiate Trabectedin Activity in Preclinical Models of Bone and Soft Tissue Sarcomas

Ymera Pignochino; F. Capozzi; C. Dell'Aglio; M. Basiricò; Lorenzo D'Ambrosio; Danilo Galizia; Erica Palesandro; M.S. Benassi; Massimo Aglietta; Giovanni Grignani

Background: Trabectedin (TR) is a DNA-binding alkaloid approved in Europe for the treatment of soft tissue sarcoma as second/third line therapy. TR binds to the minor groove of DNA and interferes with gene transcription and nucleotide excision repair, inducing DNA double strand breaks (DSBs), and S/G2 cell cycle arrest. There is a strong clinical interest to increase TR activity by combination with other anti-cancer drugs. PARP-1 inhibitors (PARP1-i) which disable DNA base-excision repair mechanism causing the accumulation of DSBs, look like a worth to explore TR therapeutic partners. We focused our in vitro studies on the effects of the combination of TR with the PARP1-i olaparib (OL) and veliparib (VEL). Methods:We explored DNA damage by comet assay and phospho-histone H2AX determination and PARP-1 activity by polyADPribosylation assay in a panel of sarcoma cell lines treated with TR, OL,VEL and combinations. We evaluated cell viability after 72h treatment with escalating doses of TR (0−2nM), PARP1-i (VEL:0−80mcM; OL:0−20mcM), and their constant combinations. Following colony formation, cell cycle, apoptosis and DNA damage response were checked. Results: We demonstrated that TR-induced DNA damage and PARP1 activation in sarcoma sensitive cell line. The addition of PARP1i completely blocked basal and TR-induced PARP-1 activation. DNA damage response and checkpoints (ATM, ATR, BRCA1, CHK1, CHK2, p53) are activated after treatments with single agents and combinations inducing cell cycle arrest in S/G2 phase. This block was release after 48h in single agents-treated cells but not in combinations-treated cells leading to apoptotic cell death. Colony growth assay and viability tests revealed a strong synergism of the two drugs (Combination Index leyomiosarcoma > osteosarcoma undifferentiated pleomorphic > fibrosarcoma the most resistant). Determination of molecular markers of sensitivity and in vivo antitumoral activity studies are ongoing. Conclusions: Our results validate the biological rationale to combine TR and PARP1-i in sarcomas and suggest exploring this pharmacological approach in the clinical setting.

Collaboration


Dive into the Lorenzo D'Ambrosio'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