34 results on '"Ornella Cutaia"'
Search Results
2. Epigenetic Immune Remodeling of Mesothelioma Cells: A New Strategy to Improve the Efficacy of Immunotherapy
- Author
-
Maria Fortunata Lofiego, Sara Cannito, Carolina Fazio, Francesca Piazzini, Ornella Cutaia, Laura Solmonese, Francesco Marzani, Carla Chiarucci, Anna Maria Di Giacomo, Luana Calabrò, Sandra Coral, Michele Maio, Alessia Covre, and on behalf of the EPigenetic Immune-Oncology Consortium Airc (EPICA) Investigators
- Subjects
epigenetic drugs ,DNA methylation ,immunotherapy ,malignant pleural mesothelioma ,Genetics ,QH426-470 ,Biotechnology ,TP248.13-248.65 - Abstract
Malignant pleural mesothelioma (MPM) is an aggressive malignancy with a severe prognosis, and with a long-standing need for more effective therapeutic approaches. However, treatment with immune checkpoint inhibitors is becoming an increasingly effective strategy for MPM patients. In this scenario, epigenetic modifications may negatively regulate the interplay between immune and malignant cells within the tumor microenvironment, thus contributing to the highly immunosuppressive contexture of MPM that may limit the efficacy of immunotherapy. Aiming to further improve prospectively the clinical efficacy of immunotherapeutic approaches in MPM, we investigated the immunomodulatory potential of different classes of epigenetic drugs (i.e., DNA hypomethylating agent (DHA) guadecitabine, histone deacetylase inhibitors VPA and SAHA, or EZH2 inhibitors EPZ-6438) in epithelioid, biphasic, and sarcomatoid MPM cell lines, by cytofluorimetric and real-time PCR analyses. We also characterized the effects of the DHA, guadecitabine, on the gene expression profiles (GEP) of the investigated MPM cell lines by the nCounter platform. Among investigated drugs, exposure of MPM cells to guadecitabine, either alone or in combination with VPA, SAHA and EPZ-6438 demonstrated to be the main driver of the induction/upregulation of immune molecules functionally crucial in host-tumor interaction (i.e., HLA class I, ICAM-1 and cancer testis antigens) in all three MPM subtypes investigated. Additionally, GEP demonstrated that treatment with guadecitabine led to the activation of genes involved in several immune-related functional classes mainly in the sarcomatoid subtype. Furthermore, among investigated MPM subtypes, DHA-induced CDH1 expression that contributes to restoring the epithelial phenotype was highest in sarcomatoid cells. Altogether, our results contribute to providing the rationale to develop new epigenetically-based immunotherapeutic approaches for MPM patients, potentially tailored to the specific histologic subtypes.
- Published
- 2021
- Full Text
- View/download PDF
3. Immunomodulatory Properties of DNA Hypomethylating Agents: Selecting the Optimal Epigenetic Partner for Cancer Immunotherapy
- Author
-
Carolina Fazio, Alessia Covre, Ornella Cutaia, Maria Fortunata Lofiego, Patrizia Tunici, Carla Chiarucci, Sara Cannito, Gianluca Giacobini, James N. Lowder, Roberta Ferraldeschi, Pietro Taverna, Anna Maria Di Giacomo, Sandra Coral, and Michele Maio
- Subjects
DNA hypomethylating agent ,epigenetics ,cancer ,immune phenotype ,immunotherapy ,Therapeutics. Pharmacology ,RM1-950 - Abstract
DNA hypomethylating agents (DHAs) play a well-acknowledged role in potentiating the immunogenicity and the immune recognition of neoplastic cells. This immunomodulatory activity of DHAs is linked to their ability to induce or to up-regulate on neoplastic cells the expression of a variety of immune molecules that play a crucial role in host-tumor immune interactions. To further investigate the clinical potential of diverse epigenetic compounds when combined with immunotherapeutic strategies, we have now compared the tumor immunomodulatory properties of the first generation DHAs, azacytidine (AZA) and decitabine (DAC) and of the next generation DHA, guadecitabine. To this end, human melanoma and hematological cancer cells were treated in vitro with 1 μM guadecitabine, DAC or AZA and then studied by molecular and flow cytometry analyses for changes in their baseline expression of selected immune molecules involved in different mechanism(s) of immune recognition. Results demonstrated a stronger DNA hypomethylating activity of guadecitabine and DAC, compared to AZA that associated with stronger immunomodulatory activities. Indeed, the mRNA expression of cancer testis antigens, immune-checkpoint blocking molecules, immunostimulatory cytokines, involved in NK and T cell signaling and recruiting, and of genes involved in interferon pathway was higher after guadecitabine and DAC compared to AZA treatment. Moreover, a stronger up-regulation of the constitutive expression of HLA class I antigens and of Intercellular Adhesion Molecule-1 was observed with guadecitabine and DAC compared to AZA. Guadecitabine and DAC seem to represent the optimal combination partners to improve the therapeutic efficacy of immunotherapeutic agents in combination/sequencing clinical studies.
- Published
- 2018
- Full Text
- View/download PDF
4. Soluble NKG2D ligands are biomarkers associated with the clinical outcome to immune checkpoint blockade therapy of metastatic melanoma patients
- Author
-
Cristina Maccalli, Diana Giannarelli, Carla Chiarucci, Ornella Cutaia, Gianluca Giacobini, Wouter Hendrickx, Giovanni Amato, Diego Annesi, Davide Bedognetti, Maresa Altomonte, Riccardo Danielli, Luana Calabrò, Anna Maria Di Giacomo, Francesco M. Marincola, Giorgio Parmiani, and Michele Maio
- Subjects
cytotoxic t-lymphocyte antigen-4 (ctla-4) ,metastatic melanoma ,nkg2d ligands ,programmed cell death-1 (pd-1) ,t cell responses ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The introduction of immune checkpoint blockade into the clinical practice resulted in improvement of survival of a significant portion of melanoma patients. Consequently, predictive biomarkers of response are needed to optimize patient's stratification and the development of combination therapies. The aim of this study was to determine whether levels of soluble NKG2D ligands (MICA, MICB, ULBP1, 2 and 3; sNKG2DLs) in the serum of melanoma patients can serve as useful predictors of response to the treatment with immune checkpoint blockade. sNKG2DLs were measured by ELISA in baseline and post-treatment serum and these results were correlated with the clinical outcome of melanoma patients (N = 194). The same determinations were performed also in a cohort of patients (N = 65) treated with either chemotherapy, radiotherapy, or mutated BRAF inhibitors (BRAFi). Absence of soluble MICB and ULBP-1 in baseline serum correlated with improved survival (OS = 21.6 and 25.3 mo and p = 0.02 and 0.01, respectively) of patients treated with immunological therapies while detectable levels of these molecules were found in poor survivors (OS = 8.8 and 12.1 mo, respectively). Multivariate analysis showed that LDH (p
- Published
- 2017
- Full Text
- View/download PDF
5. Data from Primary Analysis and 4-Year Follow-Up of the Phase III NIBIT-M2 Trial in Melanoma Patients With Brain Metastases
- Author
-
Michele Maio, Mario Mandalà, Diana Giannarelli, Monica Valente, Luana Calabrò, Roberto Camerini, Alessia Covre, Giovanni Amato, Ornella Cutaia, Paolo Marchetti, Massimo Guidoboni, Pietro Quaglino, Michele Guida, Pier Francesco Ferrucci, Michele Del Vecchio, Vanna Chiarion-Sileni, and Anna Maria Di Giacomo
- Abstract
Purpose:Phase II trials have shown encouraging activity with ipilimumab plus fotemustine and ipilimumab plus nivolumab in melanoma brain metastases. We report the primary analysis and 4-year follow-up of the NIBIT-M2 study, the first phase III trial comparing these regimens with fotemustine in patients with melanoma with brain metastases.Patients and Methods:This phase III study recruited patients 18 years of age and older with BRAF wild-type or mutant melanoma, and active, untreated, asymptomatic brain metastases from nine centers, randomized (1:1:1) to fotemustine, ipilimumab plus fotemustine, or ipilimumab plus nivolumab. The primary endpoint was overall survival (OS).Results:From January, 2013 to September, 2018, 27, 26, and 27 patients received fotemustine, ipilimumab plus fotemustine, and ipilimumab plus nivolumab. Median OS was 8.5 months [95% confidence interval (CI), 4.8–12.2] in the fotemustine arm, 8.2 months (95% CI, 2.2–14.3) in the ipilimumab plus fotemustine arm (HR vs. fotemustine, 1.09; 95% CI, 0.59–1.99; P = 0.78), and 29.2 months (95% CI, 0–65.1) in the ipilimumab plus nivolumab arm (HR vs. fotemustine, 0.44; 95% CI, 0.22–0.87; P = 0.017). Four-year survival rate was significantly higher for ipilimumab plus nivolumab than fotemustine [(41.0%; 95% CI, 20.6–61.4) vs. 10.9% (95% CI, 0–24.4; P = 0.015)], and was 10.3% (95% CI, 0–22.6) for ipilimumab plus fotemustine. In the fotemustine, ipilimumab plus fotemustine, and ipilimumab plus nivolumab arms, respectively, 11 (48%), 18 (69%), and eight (30%) patients had treatment-related grade 3 or 4 adverse events, without treatment-related deaths.Conclusions:Compared with fotemustine, ipilimumab plus nivolumab significantly improved overall and long-term survival of patients with melanoma with asymptomatic brain metastases.
- Published
- 2023
6. Fig.S1 from Primary Analysis and 4-Year Follow-Up of the Phase III NIBIT-M2 Trial in Melanoma Patients With Brain Metastases
- Author
-
Michele Maio, Mario Mandalà, Diana Giannarelli, Monica Valente, Luana Calabrò, Roberto Camerini, Alessia Covre, Giovanni Amato, Ornella Cutaia, Paolo Marchetti, Massimo Guidoboni, Pietro Quaglino, Michele Guida, Pier Francesco Ferrucci, Michele Del Vecchio, Vanna Chiarion-Sileni, and Anna Maria Di Giacomo
- Abstract
Study design and treatment schedule. Patients in either the ipilimumab plus fotemustine arm or the ipilimumab plus nivolumab arm could continue maintenance treatment until disease progression, excessive toxicity, or patient refusal
- Published
- 2023
7. Table S1 from Primary Analysis and 4-Year Follow-Up of the Phase III NIBIT-M2 Trial in Melanoma Patients With Brain Metastases
- Author
-
Michele Maio, Mario Mandalà, Diana Giannarelli, Monica Valente, Luana Calabrò, Roberto Camerini, Alessia Covre, Giovanni Amato, Ornella Cutaia, Paolo Marchetti, Massimo Guidoboni, Pietro Quaglino, Michele Guida, Pier Francesco Ferrucci, Michele Del Vecchio, Vanna Chiarion-Sileni, and Anna Maria Di Giacomo
- Abstract
Maintenance phase treatment exposure
- Published
- 2023
8. Fig S2 from Primary Analysis and 4-Year Follow-Up of the Phase III NIBIT-M2 Trial in Melanoma Patients With Brain Metastases
- Author
-
Michele Maio, Mario Mandalà, Diana Giannarelli, Monica Valente, Luana Calabrò, Roberto Camerini, Alessia Covre, Giovanni Amato, Ornella Cutaia, Paolo Marchetti, Massimo Guidoboni, Pietro Quaglino, Michele Guida, Pier Francesco Ferrucci, Michele Del Vecchio, Vanna Chiarion-Sileni, and Anna Maria Di Giacomo
- Abstract
Swimmer plot analysis of NIBIT-M2 patients. Swimmer plot showing by study arm patients who at the time of data cut-off were alive and either still on study therapy (n=7) or off study therapy without having received subsequent treatment (n=6), and all patients who had received subsequent treatment at the time of data cut-off, regardless of whether alive or dead (n=32). Patients excluded from the plot (n=31) died without receiving any subsequent therapy.
- Published
- 2023
9. Tremelimumab plus durvalumab retreatment and 4-year outcomes in patients with mesothelioma: a follow-up of the open label, non-randomised, phase 2 NIBIT-MESO-1 study
- Author
-
Maresa Altomonte, Diana Giannarelli, Michele Maio, Claudio Rosati, Carolina Fazio, Aldo Morra, Alessia Covre, Giulia Rossi, Giuseppe Palmieri, Luana Calabrò, Maria Grazia Daffinà, Milena Casula, Anna Maria Di Giacomo, and Ornella Cutaia
- Subjects
Pulmonary and Respiratory Medicine ,Mesothelioma ,medicine.medical_specialty ,Durvalumab ,Lung Neoplasms ,Population ,Antibodies, Monoclonal ,Antibodies, Monoclonal, Humanized ,Follow-Up Studies ,Humans ,Retreatment ,Antibodies ,NO ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Monoclonal ,medicine ,Clinical endpoint ,030212 general & internal medicine ,education ,Humanized ,education.field_of_study ,business.industry ,medicine.disease ,030228 respiratory system ,Response Evaluation Criteria in Solid Tumors ,Peritoneal mesothelioma ,business ,Tremelimumab ,Progressive disease ,medicine.drug - Abstract
Summary Background The NIBIT-MESO-1 study demonstrated the efficacy and safety of tremelimumab combined with durvalumab in patient with unresectable mesothelioma followed up for a median of 52 months [IQR 49–53]. Here, we report 4-year survival and outcomes after retreatment, and the role of tumour mutational burden (TMB) in identifying patients who might have a better outcome in response to combined therapy. Methods NIBIT-MESO-1 was an open-label, non-randomised, phase 2 trial of patients with unresectable pleural or peritoneal mesothelioma who received intravenous tremelimumab (1 mg/kg bodyweight) and durvalumab (20 mg/kg bodyweight) every 4 weeks for four doses, followed by maintenance intravenous durvalumab at the same dose and schedule for nine doses. In this follow-up study, patients with disease progression following initial clinical benefit—ie, a partial repsonse or stable disease—were eligible for retreatment and with the same doses and schedules for tremelimumab and durvalumab as used in the NIBIT-MESO-1 trial. The primary endpoint, immune-related objective response rate, was evaluated per immune-related modified Response Evaluation Criteria in Solid Tumors (RECIST) or immune-related RECIST 1.1 criteria for patients with pleural or peritoneal malignant mesothelioma, respectively. Key secondary endpoints were overall survival and safety, and TMB was also evaluated post hoc in patients who had tumour tissue available before treatment. The intention-to-treat population was used for analysis of all efficacy endpoints. This study is registered with ClinicalTrials.gov , number NCT02588131 . Findings 40 patients were enrolled in the NIBIT-MESO-1 study between Oct 30, 2015, and Oct 12, 2016. At data cut-off, April 30, 2020, five (13%) of 40 patients were alive, and 35 (88%) patients had died of progressive disease. At a median follow-up of 52 months (IQR 49–53), median overall survival was 16·5 months (95% CI 13·7–19·2). Survival was 20% (eight of 40 patients) at 36 months and 15% (six of 40 patients) and 48 months. 17 (43%) of 40 patients met the criteria for enrolment in the retreatment study and were retreated with at least one dose of tremelimumab and durvalumab. No immune-related objective responses were observed in the 17 retreated patients. Seven (41%) of 17 patients achieved immune-related stable disease. From the start of retreatment to a median follow-up of 24 months (22·0–25·0), median overall survival was 12·5 months (95% CI 0·0–25·8), and survival at 12 months was 52·9%, at 18 months was 35·3%, and at 24 months was 23·5%. There were no grade 3–4 immune-related adverse events in the retreatment cohort. In a post-hoc analysis of 28 patients for whom tumour tissue before treatment was available, patients with a TMB higher than the median value of 8·3 mutations per Mb had a higher median overall survival compared with patients with TMB below the median value, but this difference was non-significant. Moreover, when patients were additionally stratified for ICI retreatment (n=13), there was a significant difference in survival between those with a TMB higher than the median of 8·3 mutations per Mb and those with TMB lower than the median in the retreated cohort (41·3 months vs 17·4 months; p=0·02). Interpretation Tremelimumab combined with durvalumab was associated with long-term survival in patients with mesothelioma. Retreatment was safe and resulted in clinically meaningful outcomes, thus suggesting its potential application in the clinical practice of mesothalioma patients. Funding NIBIT Foundation, Fondazione AIRC, AstraZeneca.
- Published
- 2021
10. 844 Immunomodulatory activity of epigenetic drugs combinations in mesothelioma: laying the ground for new immunotherapeutic strategies
- Author
-
Sara Cannito, Health Biology, Ornella Cutaia, Carolina Fazio, Maria Fortunata Lofiego, Francesca Piazzini, Laura Solmonese, Luana Calabrò, Michele Maio, and Alessia Covre
- Subjects
0301 basic medicine ,EZH2 ,Human leukocyte antigen ,Biology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Sarcomatoid Mesothelioma ,medicine.disease ,lcsh:RC254-282 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,ULBP2 ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Cancer/testis antigens ,Mesothelioma ,Epigenetics - Abstract
Background Growing evidence are demonstrating the therapeutic efficacy of immune checkpoint inhibitors (ICI) in mesothelioma; however, a limited percentage of patients benefits from this therapeutic approach. Epigenetic modifications play a relevant role in negatively regulating the cross-talk between neoplastic and immune cells, and in contributing to the highly immunosuppressive mesothelioma microenvironment. A better understanding of mesothelioma epigenetic landscape could open the path to novel and potentially more effective approaches combining ICI and epigenetic drugs. We investigated the immunomodulatory potential of epigenetic agents by comparing the activity of DNA hypomethylating agents (DHA) with histone deacetylases inhibitors (HDACi) and EZH2 inhibitors (EZH2i), alone or combined with DHA, in mesothelioma cells. Methods Four mesothelioma cell lines were treated with the DHA guadecitabine 1μM, or with the HDACi, Valproic Acid (VPA) 1mM, or the EZH2i, EPZ-6438 1μM, alone or combined with guadecitabine. We investigated the expression of HLA class I molecules by flow-cytometry and of PD-L1, cancer testis antigens (CTA: NY-ESO, MAGE-A1), Natural Killer Group 2 member D Ligands (NKG2DLs: MIC-A, MIC-B, ULBP2) and EMT-regulating cadherins (CDH1, CDH2) by quantitative Real-Time PCR. Fold change (FC) expression for each treatment vs untreated cells was reported as mean values (FCm) among investigated cell lines. A positive modulation of the expression was considered if FCm>1.5. Results Guadecitabine upregulated the expression of HLA class I antigens (FCm=1.75), PD-L1 (FCm=2.38), NKG2DLs (MIC-A FCm=1.96, MIC-B FCm=2.57, and ULBP2 FCm=3.56), and upregulated/induced CTA expression. Similarly, VPA upregulated HLA class I antigens (FCm=1.67), PD-L1 (FCm=3.17), NKG2DLs (MIC-A FCm=1.78, MIC-B FCm=3.04, and ULBP2 FCm=3.75) expression; however, CTA expression was modulated only in 1 mesothelioma cell line. Conversely, EPZ-6438 up-regulated only NY-ESO-1 and MIC-B expression in 1 mesothelioma cell line. The addition of both VPA and EPZ-6483 to guadecitabine strengthened its immunomodulatory activity. Specifically, guadecitabine plus VPA or EPZ-6438 upregulated the expression of HLA class I antigens FCm=2.55 or 2.69, PD-L1 FCm=8.04 or 2.65, MIC-A FCm=3.81 or 2.26, MIC-B FCm=8.00 or 3.03, ULBP2 FCm=6.24 or 4.53, respectively. Higher levels of CTA upregulation/induction were observed with combination treatments vs guadecitabine alone. Cadherins modulation was mesothelioma histotype-related: CDH1 expression was induced in the 2 constitutively-negative sarcomatoid mesothelioma cells by guadecitabine alone or combined with VPA or EPZ-6438; CDH2 expression was upregulated by VPA alone (FCm=1.53) or plus guadecitabine (FCm=2.54). Conclusions Combination of DHA-based immunotherapies with other classes of epigenetic drugs could be an effective strategy to be pursued in the mesothelioma clinic.
- Published
- 2020
11. NK- and T-cell subsets in malignant mesothelioma patients: Baseline pattern and changes in the context of anti-CTLA-4 therapy
- Author
-
Milad Tannazi, Valeria Ventura, Rosa Sottile, Patrick Micke, Carla Chiarucci, Rossana Tallerico, Paolo Frumento, Victor Pontén, Klas Kärre, Alessia Covre, Maria H. Johansson, Ornella Cutaia, Michele Maio, Costanza Maria Cristiani, Ennio Carbone, Luana Calabrò, Cinzia Garofalo, Sottile, R, Tannazi, M, Johansson, Mh, Cristiani, Cm, Calabró, L, Ventura, V, Cutaia, O, Chiarucci, C, Covre, A, Garofalo, C, Pontén, V, Tallerico, R, Frumento, P, Micke, P, Maio, M, Kärre, K, and Carbone, E
- Subjects
Male ,Mesothelioma ,Cancer Research ,Lung Neoplasms ,Cell ,NK cells ,Kaplan-Meier Estimate ,0302 clinical medicine ,T-Lymphocyte Subsets ,Monoclonal ,Killer Cells ,CTLA-4 Antigen ,Humanized ,Cells, Cultured ,anti-CTLA-4 therapy ,Tumor ,Cultured ,biology ,immunoprofiling ,malignant mesothelioma ,T cells ,Antibodies, Monoclonal, Humanized ,Antineoplastic Agents ,CD56 Antigen ,Cell Line, Tumor ,Cytokines ,Female ,Gene Expression Regulation, Neoplastic ,Humans ,K562 Cells ,Killer Cells, Natural ,Middle Aged ,Prognosis ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Natural ,Antibody ,medicine.drug ,T cell ,Cells ,Antibodies ,Cell Line ,03 medical and health sciences ,Immune system ,medicine ,Neoplastic ,business.industry ,Mesothelioma, Malignant ,medicine.disease ,Immune checkpoint ,Gene Expression Regulation ,Cancer research ,biology.protein ,business ,Tremelimumab ,CD8 - Abstract
Malignant mesothelioma (MM) is a highly aggressive form of cancer with limited treatment options. Although the role of NK cells has been studied in many solid tumors, the pattern of NK-cell subsets and their recognition of mesothelioma cells remain to be explored. We used RNA expression data of MM biopsies derived from the cancer genome atlas to evaluate the immune cell infiltrates. We characterized the phenotype of circulating NK and T cells of 27 MM patients before and after treatment with an anti-CTLA-4 antibody (tremelimumab). These immune cell profiles were compared to healthy controls. The RNA expression data of the MM biopsies indicated the presence of NK cells in a subgroup of patients. We demonstrated that NK cells recognize MM cell lines and that IL-15 stimulation improved NK cell-mediated lysis in vitro. Using multivariate projection models, we found that MM patients had a perturbed ratio of CD56bright and CD56dim NK subsets and increased serum concentrations of the cytokines IL-10, IL-8 and TNF-α. After tremelimumab treatment, the ratio between the CD56bright and CD56dim subsets shifted back towards physiological levels. Furthermore, the improved overall survival was correlated with low TIM-3+ CD8+ T-cell frequency, high DNAM-1+ CD56dim NK-cell frequency and high expression levels of NKp46 on the CD56dim NK cells before and after immune checkpoint blockade. Together, our observations suggest that NK cells infiltrate MM and that they can recognize and kill mesothelioma cells. The disease is associated with distinct lymphocytes patterns, some of which correlate with prognosis or are affected by treatment with tremelimumab.
- Published
- 2019
12. Guadecitabine plus ipilimumab in unresectable melanoma: the NIBIT-M4 clinical trial
- Author
-
Alessia Covre, Giovanni Amato, Michele Maio, Francesca Finotello, James N. Lowder, Amelie Nemc, Santa Monterisi, Zlatko Trajanoski, Florent Petitprez, Anna Maria Di Giacomo, Sandra Coral, Andrea Lazzeri, Laetitia Lacroix, Ornella Cutaia, Riccardo Danielli, Luca Sigalotti, Wolf H. Fridman, Monica Valente, Carolina Fazio, Mohammad Azab, Andrea Anichini, Diana Giannarelli, Christoph Bock, Dietmar Rieder, Clelia Miracco, Catherine Sautès-Fridman, and Aram Oganesian
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Programmed Cell Death 1 Receptor ,Ipilimumab ,CD8-Positive T-Lymphocytes ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Neoplasm Metastasis ,Adverse effect ,Survival rate ,Melanoma ,Aged ,Neoplasm Staging ,Aged, 80 and over ,business.industry ,Cancer ,DNA Methylation ,Middle Aged ,medicine.disease ,3. Good health ,Clinical trial ,Gene Expression Regulation, Neoplastic ,Survival Rate ,030104 developmental biology ,Treatment Outcome ,Oncology ,Tolerability ,030220 oncology & carcinogenesis ,Azacitidine ,Female ,Patient Safety ,business ,medicine.drug - Abstract
Purpose: The immunomodulatory activity of DNA hypomethylating agents (DHAs) suggests they may improve the effectiveness of cancer immunotherapies. The phase Ib NIBIT-M4 trial tested this hypothesis using the next-generation DHA guadecitabine combined with ipilimumab. Patients and Methods: Patients with unresectable stage III/IV melanoma received escalating doses of guadecitabine 30, 45, or 60 mg/m2/day subcutaneously on days 1 to 5 every 3 weeks, and ipilimumab 3 mg/kg intravenously on day 1 every 3 weeks, starting 1 week after guadecitabine, for four cycles. Primary endpoints were safety, tolerability, and MTD of treatment; secondary were immune-related (ir) disease control rate (DCR) and objective response rate (ORR); and exploratory were changes in methylome, transcriptome, and immune contextures in sequential tumor biopsies, and pharmacokinetics. Results: Nineteen patients were treated; 84% had grade 3/4 adverse events, and neither dose-limiting toxicities per protocol nor overlapping toxicities were observed. Ir-DCR and ir-ORR were 42% and 26%, respectively. Median CpG site methylation of tumor samples (n = 8) at week 4 (74.5%) and week 12 (75.5%) was significantly (P < 0.05) lower than at baseline (80.3%), with a median of 2,454 (week 4) and 4,131 (week 12) differentially expressed genes. Among the 136 pathways significantly (P < 0.05; Z score >2 or ←2) modulated by treatment, the most frequently activated were immune-related. Tumor immune contexture analysis (n = 11) demonstrated upregulation of HLA class I on melanoma cells, an increase in CD8+, PD-1+ T cells and in CD20+ B cells in posttreatment tumor cores. Conclusions: Treatment of guadecitabine combined with ipilimumab is safe and tolerable in advanced melanoma and has promising immunomodulatory and antitumor activity.
- Published
- 2019
- Full Text
- View/download PDF
13. Abstract 4488: Immunomodulatory activity of epigenetic drugs: Laying the ground for new combined immunotherapeutic strategies for glioblastoma
- Author
-
Giuseppe Oliveri, Maria Fortunata Lofiego, Carla Chiarucci, Elisabetta Gambale, Alessia Covre, Sara Cannito, Ornella Cutaia, Francesca Piazzini, Michele Maio, Monica Valente, Carolina Fazio, Clelia Miracco, Anna Maria Di Giacomo, and Sandra Coral
- Subjects
Cancer Research ,business.industry ,medicine.medical_treatment ,Cancer ,Immunotherapy ,Human leukocyte antigen ,medicine.disease ,Immune system ,Oncology ,Hypomethylating agent ,Glioma ,Cancer cell ,Cancer research ,Cancer/testis antigens ,Medicine ,business - Abstract
Rationale: Glioblastoma multiforme (GBM) is the most common and deadliest primary brain tumour in adults, with a very dismal prognosis and limited effective therapies. Immunotherapy may play a role in treating GBM, though the initial clinical results are still poor, and novel combined treatment(s) could possibly improve its efficacy. Manipulating the epigenome has been considered a therapeutic opportunity in GBM, since epigenetic modifications regulate glioma proliferation, metastasis, invasion, and prognosis. In addition, epigenetic alterations may negatively regulate the cross-talk between GBM and immune cells in GBM microenvironment (TME), thus limiting the efficacy of immunotherapy. We previously demonstrated the immunomodulatory potential of epigenetic drugs in cancer cells, and translated this notion in the clinic to improve efficacy of immunotherapy of melanoma patients (Di Giacomo AM, CCR 2019). Accordingly, this study aimed to evaluate the immunomodulatory potential of epigenetic drugs in GBM, and to provide the scientific rationale for new combinatorial therapeutic approaches in GBM patients. Methods: Patient-derived GBM (pGBM) cell lines (n=10) were established from surgically-removed GBM lesions and treated with the DNA hypomethylating agent guadecitabine (1 µM); commercially available human GBM (cGBM) cell lines (n=3) were treated with guadecitabine (1 µM) and/or with the EZH2-inhibitor GSK126 (10 µM). Expression of HLA class I and ICAM-1 by GBM cells was investigated by flow-cytometry; cancer testis antigens (CTA, i.e., NY-ESO, MAGE-A1 and MAGE-A3) and the epithelial-mesenchymal transition (EMT)-regulating cadherins (i.e., CDH1 and CDH2) expression was analyzed by quantitative Real-Time PCR. Results: A positive modulation of immune profile was observed in all investigated guadecitabine-treated GBM cells. Specifically, in untreated vs guadecitabine-treated cells, MFI mean values ± SD of HLA class I were 402±326 vs 442±303 in pGBM cells, and 277±127 vs 381±138 in cGBM cells; MFI mean values ± SD of ICAM-1 were 53±72 vs 92±116 in pGBM cells, and 5±6 vs 26±17 in cGBM cells. Though a slight increment was induced by GSK126 for HLA class I (312±169) and ICAM-1 (9±11) expression in cGBM, an additive effect was detected with GSK126+guadecitabine (405±261 for HLA class I and 35±22 for ICAM-1) in cGBM cells. Guadecitabine induced NY-ESO-1, MAGE-A1, and MAGE-A3 expression in 9/10, 6/10, and 5/8 pGBM cells, respectively and in all cGBM cells. Levels of CTA expression were increased in cGBM by GSK126+guadecitabine vs guadecitabine with mean Fold Change (FC) of 1.5, 1.6 and 1.6 for NY-ESO-1, MAGE-A1 and MAGE-A3 expression. Finally, CDH1 expression was induced in 1/3 and in 2/3 cGBM cells by guadecitabine and guadecitabine+GSK126, respectively; CDH2-positive expression was upregulated by guadecitabine (mean FC=2.45) but not by guadecitabine+GSK126. Conclusions: Immunomodulatory properties of epigenetic drugs in GBM should be exploited to improve the efficacy of GBM immunotherapy. Citation Format: Maria Fortunata Lofiego, Francesca Piazzini, Carolina Fazio, Ornella Cutaia, Carla Chiarucci, Sara Cannito, Elisabetta Gambale, Monica Valente, Anna Maria Di Giacomo, Sandra Coral, Clelia Miracco, Giuseppe Oliveri, Michele Maio, Alessia Covre. Immunomodulatory activity of epigenetic drugs: Laying the ground for new combined immunotherapeutic strategies for glioblastoma [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4488.
- Published
- 2020
14. Circulating Levels of PD-L1 in Mesothelioma Patients from the NIBIT-MESO-1 Study: Correlation with Survival
- Author
-
Alessia Covre, Gianluca Giacobini, Michele Maio, Maria Fortunata Lofiego, Sandra Coral, Carla Chiarucci, Giovanni Amato, Sara Cannito, Riccardo Danielli, Anna Maria Di Giacomo, Ornella Cutaia, Diana Giannarelli, Luana Calabrò, Carolina Fazio, and Maria Grazia Daffinà
- Subjects
PD-L1 ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Durvalumab ,medicine.medical_treatment ,lcsh:RC254-282 ,Article ,NO ,immune checkpoint inhibitors ,03 medical and health sciences ,0302 clinical medicine ,Biomarker ,Cancer immunotherapy ,Immune checkpoint inhibitors ,Mesothelioma ,Soluble PD-L1 ,Antigen ,Internal medicine ,medicine ,cancer immunotherapy ,biology ,business.industry ,Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,mesothelioma ,030220 oncology & carcinogenesis ,biology.protein ,biomarker ,Biomarker (medicine) ,business ,soluble PD-L1 ,Tremelimumab ,medicine.drug - Abstract
Targeting of the programmed cell death protein (PD)-1/programmed death-ligand 1 (PD-L1) axis has shown a significant clinical impact in several tumor types. Accordingly, our phase II NIBIT-MESO-1 study demonstrated an improved clinical efficacy in mesothelioma patients treated with the anti-PD-L1 durvalumab combined with the anti-cytotoxic T-lymphocyte antigen (CTLA)-4 tremelimumab, as compared to tremelimumab alone. Due to the promising therapeutic activity of immune check-point inhibitors (ICIs) in mesothelioma patients, the identification of biomarkers predictive of response to treatment is of crucial relevance. The prognostic role of soluble PD-L1 (sPD-L1) proposed in cancer patients prompted us to investigate this protein in sera from mesothelioma patients (n = 40) enrolled in the NIBIT-MESO-1 study. A significant (p <, 0.001) increase in sPD-L1 levels was detected in patients after the first cycle and during therapy vs. baseline. A longer overall survival (OS) was observed in patients with sPD-L1 concentrations below (at baseline, d1C2, d1C5 (p <, 0.01)) or FC values above (p <, 0.05 at d1C2, d1C3, d1C5) their statistically calculated optimal cut-offs. On the basis of these initial results, the specific role of CTLA-4-, PD-L1-, or PD-1-targeting on sPD-L1 release was then investigated in sera from 81 additional ICI-treated solid cancer patients. Results showed a significant (p <, 0.001) increase of sPD-L1 levels during therapy compared to baseline only in anti-PD-L1-treated patients, supporting the specific involvement of PD-L1 targeting in the release of its soluble form. Our findings suggest that sPD-L1 represents a predictive biomarker of clinical response to anti-PD-L1 cancer immunotherapy.
- Published
- 2020
15. Immunomodulatory properties of DNA hypomethylating agents: Selecting the optimal epigenetic partner for cancer immunotherapy
- Author
-
Sandra Coral, Ornella Cutaia, Anna Maria Di Giacomo, Michele Maio, Alessia Covre, Carla Chiarucci, Roberta Ferraldeschi, Sara Cannito, Maria Fortunata Lofiego, Pietro Taverna, Gianluca Giacobini, James N. Lowder, Carolina Fazio, and Patrizia Tunici
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T cell ,Immune phenotype ,Decitabine ,Human leukocyte antigen ,DNA hypomethylating agent ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer ,Epigenetics ,Immunotherapy ,Cancer immunotherapy ,medicine ,Pharmacology (medical) ,Original Research ,Pharmacology ,lcsh:RM1-950 ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Cancer/testis antigens ,medicine.drug - Abstract
DNA hypomethylating agents (DHAs) play a well-acknowledged role in potentiating the immunogenicity and the immune recognition of neoplastic cells. This immunomodulatory activity of DHAs is linked to their ability to induce or to up-regulate on neoplastic cells the expression of a variety of immune molecules that play a crucial role in host-tumor immune interactions. To further investigate the clinical potential of diverse epigenetic compounds when combined with immunotherapeutic strategies, we have now compared the tumor immunomodulatory properties of the first generation DHAs, azacytidine (AZA) and decitabine (DAC) and of the next generation DHA, guadecitabine. To this end, human melanoma and hematological cancer cells were treated in vitro with 1 μM guadecitabine, DAC or AZA and then studied by molecular and flow cytometry analyses for changes in their baseline expression of selected immune molecules involved in different mechanism(s) of immune recognition. Results demonstrated a stronger DNA hypomethylating activity of guadecitabine and DAC, compared to AZA that associated with stronger immunomodulatory activities. Indeed, the mRNA expression of cancer testis antigens, immune-checkpoint blocking molecules, immunostimulatory cytokines, involved in NK and T cell signaling and recruiting, and of genes involved in interferon pathway was higher after guadecitabine and DAC compared to AZA treatment. Moreover, a stronger up-regulation of the constitutive expression of HLA class I antigens and of Intercellular Adhesion Molecule-1 was observed with guadecitabine and DAC compared to AZA. Guadecitabine and DAC seem to represent the optimal combination partners to improve the therapeutic efficacy of immunotherapeutic agents in combination/sequencing clinical studies.
- Published
- 2018
16. SRC Family Kinase Inhibition Through a New Pyrazolo[3,4-d]Pyrimidine Derivative as a Feasible Approach for Glioblastoma Treatment
- Author
-
Nadia Casini, Francesca Pentimalli, Iris Maria Forte, Elisa Ceccherini, Ornella Cutaia, Elena Dreassi, Antonio Giordano, Maurizio Botta, Luca Esposito, Claudio Zamperini, Silvia Schenone, Maria Sole Polito, and Paola Indovina
- Subjects
Cell growth ,Kinase ,Cell ,Cell Biology ,Biology ,Pharmacology ,Biochemistry ,medicine.anatomical_structure ,Apoptosis ,Cell culture ,medicine ,Cytotoxic T cell ,Src family kinase ,Clonogenic assay ,Molecular Biology - Abstract
Glioblastoma (GB) is the most common and aggressive primary tumor of the central nervous system. The current standard of care for GB consists of surgical resection, followed by radiotherapy combined with temozolomide chemotherapy. However, despite this intensive treatment, the prognosis remains extremely poor. Therefore, more effective therapies are urgently required. Recent studies indicate that SRC family kinases (SFKs) could represent promising molecular targets for GB therapy. Here, we challenged four GB cell lines with a new selective pyrazolo[3,4-d]pyrimidine derivative SFK inhibitor, called SI221. This compound exerted a significant cytotoxic effect on GB cells, without significantly affecting non-tumor cells (primary human skin fibroblasts), as evaluated by MTS assay. We also observed that SI221 was more effective than the well-known SFK inhibitor PP2 in GB cells. Notably, despite the high intrinsic resistance to apoptosis of GB cells, SI221 was able to induce this cell death process in all the GB cell lines, as observed through cytofluorimetric analysis and caspase-3 assay. SI221 also exerted a long-term inhibition of GB cell growth and was able to reduce GB cell migration, as shown by clonogenic assay and scratch test, respectively. Moreover, through in vitro pharmacokinetic assays, SI221 proved to have a high metabolic stability and a good potential to cross the blood brain barrier, which is an essential requirement for a drug intended to treat brain tumors. Therefore, despite the need of developing strategies to improve SI221 solubility, our results suggest a potential application of this selective SFK inhibitor in GB therapy. J. Cell. Biochem. 116: 856–863, 2015. © 2014 Wiley Periodicals, Inc.
- Published
- 2015
17. Fourteenth Meeting of the Network Italiano per la Bioterapia dei Tumori (NIBIT) on Cancer Bio-Immunotherapy, Siena, Italy, October 13-15, 2016
- Author
-
Vincenzo Russo, Alessia Covre, Carolina Fazio, Maria Fortunata Lofiego, Maresa Altomonte, Ornella Cutaia, Gianluca Giacobini, Andrea Lazzeri, Patrizia Tunici, Michele Maio, Carla Chiarucci, Antonello Lamboglia, and Erica Bertocci
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Tumor microenvironment ,Checkpoint blockade agents ,business.industry ,medicine.medical_treatment ,Immunology ,Cancer ,Cancer vaccines ,Immunotherapy ,NIBIT ,Targeted therapies ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Immunology and Allergy ,business ,030215 immunology - Published
- 2017
18. Soluble NKG2D ligands are biomarkers associated with the clinical outcome to immune checkpoint blockade therapy of metastatic melanoma patients
- Author
-
Wouter Hendrickx, Giovanni Amato, Luana Calabrò, Gianluca Giacobini, Francesco M. Marincola, Ornella Cutaia, Michele Maio, Diego Annesi, Giorgio Parmiani, Carla Chiarucci, Davide Bedognetti, Cristina Maccalli, Anna Maria Di Giacomo, Diana Giannarelli, Riccardo Danielli, and Maresa Altomonte
- Subjects
0301 basic medicine ,Oncology ,lcsh:Immunologic diseases. Allergy ,medicine.medical_specialty ,Metastatic melanoma ,Cytotoxic T-lymphocyte antigen-4 (CTLA-4) ,metastatic melanoma ,NKG2D ligands ,programmed cell death-1 (PD-1) ,T cell responses ,medicine.medical_treatment ,Immunology ,Nkg2d ligands ,lcsh:RC254-282 ,t cell responses ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,nkg2d ligands ,medicine ,Immunology and Allergy ,Original Research ,Chemotherapy ,business.industry ,Melanoma ,programmed cell death-1 (pd-1) ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immune checkpoint ,Blockade ,Radiation therapy ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cohort ,business ,lcsh:RC581-607 ,cytotoxic t-lymphocyte antigen-4 (ctla-4) - Abstract
The introduction of immune checkpoint blockade into the clinical practice resulted in improvement of survival of a significant portion of melanoma patients. Consequently, predictive biomarkers of response are needed to optimize patient's stratification and the development of combination therapies. The aim of this study was to determine whether levels of soluble NKG2D ligands (MICA, MICB, ULBP1, 2 and 3; sNKG2DLs) in the serum of melanoma patients can serve as useful predictors of response to the treatment with immune checkpoint blockade. sNKG2DLs were measured by ELISA in baseline and post-treatment serum and these results were correlated with the clinical outcome of melanoma patients (N = 194). The same determinations were performed also in a cohort of patients (N = 65) treated with either chemotherapy, radiotherapy, or mutated BRAF inhibitors (BRAFi). Absence of soluble MICB and ULBP-1 in baseline serum correlated with improved survival (OS = 21.6 and 25.3 mo and p = 0.02 and 0.01, respectively) of patients treated with immunological therapies while detectable levels of these molecules were found in poor survivors (OS = 8.8 and 12.1 mo, respectively). Multivariate analysis showed that LDH (p
- Published
- 2017
19. MA05.07 Efficacy and Safety of Re-Treatment with Tremelimumab and Durvalumab Within the NIBIT-MESO-1 Study
- Author
-
A.M. Di Giacomo, Maria Grazia Daffinà, Guido Rossi, Giovanni Amato, Michele Maio, Luana Calabrò, Diana Giannarelli, Ornella Cutaia, Aldo Morra, and C. Rosati
- Subjects
Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,Durvalumab ,business.industry ,Internal medicine ,medicine ,business ,Tremelimumab ,medicine.drug - Published
- 2019
20. Safety and immunobiological activity of guadecitabine sequenced with ipilimumab in metastatic melanoma patients: The phase Ib NIBIT-M4 study
- Author
-
Anna Maria Di Giacomo, Andrea Anichini, Francesca Finotello, Ornella Cutaia, Christoph Bock, Luca Sigalotti, Laetitia Lacroix, Alessia Covre, Florent Petitprez, Carolina Fazio, Diana Giannarelli, Wolf H. Fridman, Mohammad Azab, Zlatko Trajanoski, James N. Lowder, Catherine Sautès-Fridman, Michele Maio, Dietmar Rieder, and Sandra Coral
- Subjects
Cancer Research ,Guadecitabine ,Metastatic melanoma ,guadecitabine ,business.industry ,Cancer ,Ipilimumab ,medicine.disease ,Oncology ,guadecitabine, ipilimumab, melanoma ,melanoma ,Cancer research ,Medicine ,ipilimumab ,business ,medicine.drug - Abstract
2549 Background: DNA hypomethylating agents show broad immuno-modulatory activity in neoplastic cells, and may improve the effectiveness of cancer immunotherapies. The phase 1b NIBIT-M4 trial investigated a previously unexplored therapeutic strategy using the next-generation DNA hypomethylating agent guadecitabine sequenced with ipilimumab for the treatment of advanced melanoma. Methods: Patients with unresectable Stage III/IV melanoma received escalating doses of guadecitabine 30, 45 or 60 mg/m2 subcutaneously on Days 1–5 every three weeks, and ipilimumab 3 mg/kg intravenously on Day 1 every three weeks, starting one week after guadecitabine, for four cycles. Primary endpoints were the safety, tolerability and maximum tolerated dose of treatment; secondary endpoints included immune-related disease control and objective response. Genome-wide methylation, RNA sequencing, and immunohistochemistry analyses were performed on tumor samples collected at baseline, W4 and W12. (NCT02608437). Results: 19 patients were treated and evaluable for safety and efficacy. The most common treatment-related adverse events of any grade were myelotoxicity (n = 17; 89%) and immune-related adverse events (n = 12; 63%). Grade 3 or 4 myelotoxicity occurred in 15 (79%) patients. There were no dose limiting toxicities. Rates of immune-related disease control and objective response were 8/19 (42%) and 5/19 (26%), respectively. Exploratory analyses of tumour samples (n = 8) showed that median CpG site methylation at Week 4 (74.5%) and Week 12 (75.5%) was significantly lower (p < 0.05) than at baseline (80.3%), with a median of 2454 (Week 4) and 4131 (Week 12) differentially expressed genes identified compared to baseline; among the 136 pathways significantly modulated by treatment, the most frequently activated were immune-related. Tumour immune contexture analysis (n = 11) demonstrated up-regulation of Human Leukocyte Antigen (HLA) class I molecules on melanoma cells, and an increase in CD8+, PD-1+ T cells and in CD20+ B cells in post-treatment tumour core specimens. Conclusions: Sequential guadecitabine and ipilimumab is safe and tolerable in patients with metastatic melanoma, and has promising immunological and anti-tumour activity. Clinical trial information: NCT02608437.
- Published
- 2019
21. ICOS Expression as Immunologic Marker in Immune Activating Monoclonal Antibodies
- Author
-
Riccardo, Danielli, Ornella, Cutaia, Carolina, Fazio, Erika, Bertocci, Ester, Fonsatti, Michele, Maio, and Luana, Calabrò
- Subjects
Inducible T-Cell Co-Stimulator Protein ,Antibodies, Monoclonal ,Humans ,Cell Separation ,Lymphocytes ,Ipilimumab ,Biomarkers ,Cells, Cultured - Abstract
Compelling experimental evidences point to monitoring of the absolute number of circulating ICOS-positive T cells as an early predictive marker of clinical activity of anti-CTLA-4 antibodies in cancer patients. Here, we report available data focusing on this issue and operative procedures to detect ICOS expression on circulating peripheral blood mononuclear cells during CTLA-4 blockade therapy.
- Published
- 2016
22. ICOS expression as immunologic marker in immune activating monoclonal antibodies
- Author
-
Ornella Cutaia, Ester Fonsatti, Erika Bertocci, Riccardo Danielli, Luana Calabrò, Carolina Fazio, and Michele Maio
- Subjects
0301 basic medicine ,medicine.drug_class ,CTLA-4 ,Flow cytometry ,ICOS ,Immunomodulating antibodies ,Solid tumors ,Monoclonal antibody ,Peripheral blood mononuclear cell ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Predictive marker ,biology ,business.industry ,Cancer ,medicine.disease ,Blockade ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Antibody ,business - Abstract
Compelling experimental evidences point to monitoring of the absolute number of circulating ICOS-positive T cells as an early predictive marker of clinical activity of anti-CTLA-4 antibodies in cancer patients. Here, we report available data focusing on this issue and operative procedures to detect ICOS expression on circulating peripheral blood mononuclear cells during CTLA-4 blockade therapy.
- Published
- 2016
23. Abstract CT059: Epigenetic tumor remodelling to improve the efficacy of immune checkpoint blockade: the NIBIT-M4 clinical trial
- Author
-
Michele Maio, Dietmar Rieder, Zlatko Trajanoski, Francesca Finotello, Carolina Fazio, Anna Maria Di Giacomo, Catherine Sautès-Fridman, Wolf H. Fridman, Christoph Bock, Alessia Covre, Ornella Cutaia, Luca Sigalotti, Florent Petitprez, Laetitia Lacroix, and James N. Lowder
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Immunogenicity ,Ipilimumab ,Immune checkpoint ,Blockade ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Interferon ,Internal medicine ,Cancer cell ,medicine ,IRF7 ,business ,030215 immunology ,medicine.drug - Abstract
Introduction: The anti-CTLA-4 monoclonal antibody (mAb) ipilimumab (Ipi) has firstly shown the therapeutic potential of targeting immune checkpoints in metastatic melanoma (MM) patients (pts), though with limited objective clinical responses, ranging from 10 to 13% (Hodi, NEJM 2010; Ascierto, TLO 2017). Targeting of CTLA-4 is of interest due to its role in the priming phase of the immune response; thus, anti-CTLA-4 mAb are being tested in different combinations to improve their clinical efficacy also by overcoming tumor resistance to treatment. We have thoroughly demonstrated that epigenetic remodelling of cancer cells with DNA hypomethylating agents (DHA) improves tumor immunogenicity and immune recognition (Maio M, CCR 2015). These findings identified DHA as highly intriguing “partner agents” to improve the therapeutic index of checkpoint blockers, including CTLA-4 mAb. This hypothesis is being tested in the ongoing NIBIT-M4 study (NCT02608437). Methods: The NIBIT-M4 is a phase 1b, dose-escalation study sequencing treatment-naïve/pre-treated, unresectable Stage III/IV MM pts, with the next generation DHA guadecitabine followed by Ipi. The dose escalation follows a 3+3 design: guadecitabine (at 30, 45 or 60 mg/m2) is administered s.c. on Week (W) 0, 3, 6, 9, days 1-5 and Ipi i.v. at 3 mg/kg on W1, 4, 7 and 10 q21d. Tumor assessment per immune-related response criteria is performed at screening (W0), W12, 18, 24 and q12 weeks. Patients-wise genome-wide methylation, RNA sequencing, whole-exome sequencing, and immunohistochemistry (IHC) analyses were performed on tumor samples collected at W0, 4 and 12. Canonical pathways and upstream regulators were investigated with IPA. Preliminary results: Fourteen out of the 19 planned pts have been enrolled so far, with no dose limiting toxicities. Among the 11 evaluable pts, the objective response rate is 27%. A reduction in tumor CpG sites methylation, from W0 (median 81.18%) to W4 (median 76.54%) and W12 (median 76.89%), was identified in the initial 8 pts enrolled. Transcriptomic analyses identified 3,897 ± 3,577 and 5,414 ± 3,569 differentially expressed genes (DEG) (q 1.0) at W4 and W12 compared to W0, respectively. Among the 137 pathways that were significantly modulated in all tumor samples at either W4 and/or W12, the most frequently activated ones were immune related (i.e., Th1 and Th2, iCOS-iCOSL and CD28 Signaling in T Helper Cells, and Interferon (IFN) Signaling). In addition, the top predicted upstream regulators were genes involved in the interferon IFN pathway (i.e., IFNG, STAT1, IRF7, IFNA2, IRF1). Though limited by the current sample size investigated, a greater number of significantly (p Conclusions: Epigenetic tumor immune-remodeling seems to be a promising strategy to improve the efficacy of CTLA-4 blockade, to be further pursued also with other immunomodulating mAb. *AMDG and AC equally contributed. Citation Format: Anna M. Di Giacomo, Alessia Covre, Francesca Finotello, Dietmar Rieder, Luca Sigalotti, Carolina Fazio, Ornella Cutaia, Christoph Bock, Florent Petitprez, Laetitia Lacroix, James N. Lowder, Wolf H. Fridman, Catherine Sautès-Fridman, Zlatko Trajanoski, Michele Maio. Epigenetic tumor remodelling to improve the efficacy of immune checkpoint blockade: the NIBIT-M4 clinical trial [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr CT059.
- Published
- 2018
24. SRC family kinase inhibition through a new pyrazolo[3,4-d]pyrimidine derivative as a feasible approach for glioblastoma treatment
- Author
-
Elisa, Ceccherini, Paola, Indovina, Claudio, Zamperini, Elena, Dreassi, Nadia, Casini, Ornella, Cutaia, Iris Maria, Forte, Francesca, Pentimalli, Luca, Esposito, Maria Sole, Polito, Silvia, Schenone, Maurizio, Botta, and Antonio, Giordano
- Subjects
G2 Phase Cell Cycle Checkpoints ,Pyrimidines ,src-Family Kinases ,c-Src kinase inhibitors ,Cell Movement ,Cell Line, Tumor ,Glioblastoma ,Humans ,Pyrazoles ,Apoptosis ,Protein Kinase Inhibitors ,Cell Proliferation - Abstract
Glioblastoma (GB) is the most common and aggressive primary tumor of the central nervous system. The current standard of care for GB consists of surgical resection, followed by radiotherapy combined with temozolomide chemotherapy. However, despite this intensive treatment, the prognosis remains extremely poor. Therefore, more effective therapies are urgently required. Recent studies indicate that SRC family kinases (SFKs) could represent promising molecular targets for GB therapy. Here, we challenged four GB cell lines with a new selective pyrazolo[3,4-d]pyrimidine derivative SFK inhibitor, called SI221. This compound exerted a significant cytotoxic effect on GB cells, without significantly affecting non-tumor cells (primary human skin fibroblasts), as evaluated by MTS assay. We also observed that SI221 was more effective than the well-known SFK inhibitor PP2 in GB cells. Notably, despite the high intrinsic resistance to apoptosis of GB cells, SI221 was able to induce this cell death process in all the GB cell lines, as observed through cytofluorimetric analysis and caspase-3 assay. SI221 also exerted a long-term inhibition of GB cell growth and was able to reduce GB cell migration, as shown by clonogenic assay and scratch test, respectively. Moreover, through in vitro pharmacokinetic assays, SI221 proved to have a high metabolic stability and a good potential to cross the blood brain barrier, which is an essential requirement for a drug intended to treat brain tumors. Therefore, despite the need of developing strategies to improve SI221 solubility, our results suggest a potential application of this selective SFK inhibitor in GB therapy.
- Published
- 2015
25. Efficacy and safety of an intensified schedule of tremelimumab for chemotherapy-resistant malignant mesothelioma: An open-label, single-arm, phase 2 study
- Author
-
Marica Lenoci, Giovanni Amato, Ester Fonsatti, Ornella Cutaia, Riccardo Danielli, Diego Annesi, Luana Calabrò, Diana Giannarelli, Michele Maio, Maresa Altomonte, Anna Maria Di Giacomo, Carolina Fazio, and Aldo Morra
- Subjects
Adult ,Male ,Mesothelioma ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Lung Neoplasms ,Pleural Neoplasms ,Phases of clinical research ,Antineoplastic Agents ,Antibodies, Monoclonal, Humanized ,NO ,Internal medicine ,medicine ,Clinical endpoint ,Humans ,media_common.cataloged_instance ,European union ,Aged ,media_common ,business.industry ,Mesothelioma, Malignant ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,Surgery ,Regimen ,Treatment Outcome ,Drug Resistance, Neoplasm ,Response Evaluation Criteria in Solid Tumors ,Female ,business ,Tremelimumab ,Progressive disease ,medicine.drug - Abstract
Summary Background CTLA4 blockade by tremelimumab 15 mg/kg every 90 days provided preliminary evidence of activity in patients with pretreated malignant mesothelioma; however, retrospective exposure–response analysis of data from patients with melanoma suggested that this schedule could result in underexposure to tremelimumab. We therefore investigated the efficacy and safety of an intensified schedule of tremelimumab in patients with advanced malignant mesothelioma. Methods In this open-label, single-arm, phase 2 study, participants aged 18 years or older with unresectable, advanced malignant mesothelioma (measurable in accordance with the Response Evaluation Criteria in Solid Tumors [RECIST]), a life expectancy of 3 months or more, an Eastern Cooperative Oncology Group performance status of 2 or less, and who had failed a first-line platinum-based regimen were enrolled at the University Hospital of Siena, Siena, Italy. Participants received tremelimumab 10 mg/kg once every 4 weeks for six doses, then every 12 weeks until disease progression, unacceptable toxic effects, or refusal to continue treatment. The primary endpoint was the proportion of patients achieving an immune-related objective response (complete or partial), assessed in all patients who received at least one dose of the study drug. This study is registered with the European Union Clinical Trials Register, number 2012-002762-12, and ClinicalTrials.gov, number NCT01655888. Findings Between July 30, 2012, and July 15, 2013, we enrolled 29 patients with a median age of 65 years (range 42–78), stage III (n=11) or IV (n=18) disease, and an Eastern Cooperative Oncology Group performance status of 0–1 (n=23) or 2 (n=6). Malignant mesothelioma histology was epithelioid (n=21, including one peritoneal), biphasic (n=6), sarcomatoid (n=1), or undefined (n=1). Patients received a median of six doses of tremelimumab (range 1–13). After a median follow-up of 21·3 months (IQR 18·7–25·9), four immune-related-partial responses were recorded, one at the first tumour assessment (after about 12 weeks) and three at the second tumour assessment (about 24 weeks), with two responses occurring after initial progressive disease and one response after initial stable disease. 15 (52%) of patients achieved disease control, with a median duration of 10·9 months (95% CI 8·2–13·6). According to modified RECIST, one patient (3%) achieved a partial response and 11 (38%) patients achieved disease control rate. Grade 1–2 treatment-related adverse events occurred in 26 (90%) patients and grade 3–4 adverse events in two (7%) patients. The most common treatment-related adverse events were gastrointestinal, dermatological, and fever. Interpretation Our results suggest that the intensified schedule of tremelimumab investigated seems to have clinical and immunological activity in patients with advanced malignant mesothelioma, and a good safety profile. The same intensified schedule is now being investigated in an ongoing randomised, double-blind, placebo-controlled, phase 2b study. Funding Associazione Italiana per la Ricerca sul Cancro, Istituto Toscano Tumori, and MedImmune.
- Published
- 2015
26. Intralesional administration of L19-IL2/L19-TNF in stage III or stage IVM1a melanoma patients: results of a phase II study
- Author
-
Ornella Cutaia, Giuliano Elia, Clelia Miracco, Mario Santinami, Carolina Fazio, Roberto Patuzzo, Annabella Di Florio, Dario Neri, Andrea Maurichi, Andrea Lazzeri, Riccardo Danielli, G. Gallino, Michele Maio, Anna Maria Di Giacomo, and Leonardo Giovannoni
- Subjects
Male ,Pathology ,Cancer Research ,Skin Neoplasms ,medicine.medical_treatment ,Phases of clinical research ,Bystander effect ,Immunocytokines ,Intralesional delivery ,Melanoma ,Neoadjuvant ,Adult ,Aged ,Disease-Free Survival ,Drug Synergism ,Drug Therapy, Combination ,Female ,Follow-Up Studies ,Humans ,Immunotherapy ,Injections, Intralesional ,Middle Aged ,Neoplasm Metastasis ,Neoplasm Staging ,Prospective Studies ,Recombinant Fusion Proteins ,Young Adult ,Oncology ,Immunology ,Immunology and Allergy ,Medicine (all) ,Stage (cooking) ,3. Good health ,Intralesional ,Combination ,medicine.medical_specialty ,medicine.drug_class ,Monoclonal antibody ,Injections ,Drug Therapy ,medicine ,Stage IIIC ,business.industry ,Cancer ,medicine.disease ,Cutaneous melanoma ,Cancer research ,business - Abstract
The intratumoral injection of cytokines, in particular IL2, has shown promise for cutaneous melanoma patients with unresectable disease or continuous recurrence despite surgery. We recently reported that the intralesional injection of L19-IL2, an immunocytokine combining IL2 and the human monoclonal antibody fragment L19, resulted in efficient regional control of disease progression, increased time to distant metastasis and evidence of effect on circulating immune cell populations. We have also shown in preclinical models of cancer a remarkable synergistic effect of the combination of L19-IL2 with L19-TNF, a second clinical-stage immunocytokine, based on the same L19 antibody fused to TNF. Here, we describe the results of a phase II clinical trial based on the intralesional administration of L19-IL2 and L19-TNF in patients with stage IIIC and IVM1a metastatic melanoma, who were not candidate to surgery. In 20 efficacy-evaluable patients, 32 melanoma lesions exhibited complete responses upon intralesional administration of the two products, with mild side effects mainly limited to injection site reactions. Importantly, we observed complete responses in 7/13 (53.8 %) non-injected lesions (4 cutaneous, 3 lymph nodes), indicating a systemic activity of the intralesional immunostimulatory treatment. The intralesional administration of L19-IL2 and L19-TNF represents a simple and effective method for the local control of inoperable melanoma lesions, with a potential to eradicate them or make them suitable for a facile surgical removal of the residual mass.
- Published
- 2014
27. Immune correlates of metastatic melanoma patients treated with ipilimumab in combination with fotemustine in the phase II NIBIT-M1 study
- Author
-
Anna Maria Di Giacomo, Ester Fonsatti, Carla Chiarucci, Giorgio Parmiani, Ornella Cutaia, Cristina Maccalli, Diana Giannarelli, Filippo Capocefalo, Michele Maio, and Hugues J. M. Nicolay
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,Melanoma patient ,Metastatic melanoma ,business.industry ,education ,Immunology ,Ipilimumab ,Immune correlates ,Internal medicine ,Peripheral blood lymphocyte ,Poster Presentation ,medicine ,Molecular Medicine ,Immunology and Allergy ,Fotemustine ,business ,medicine.drug - Abstract
Meeting abstracts Ipilimumab (IPI) in combination with fotemustine (FTM) has shown a promising clinical activity in metastatic melanoma (MM) patients (pts) enrolled in the NIBIT-M1 trial (Di Giacomo, et al., Lancet Oncology, 2012). This study investigated changes in immunological parameters in the
- Published
- 2013
28. Tremelimumab for patients with chemotherapy-resistant advanced malignant mesothelioma: an open-label, single-arm, phase 2 trial
- Author
-
Luana Calabrò, Anna Maria Di Giacomo, Maresa Altomonte, Giovanni Amato, Ester Fonsatti, Diana Giannarelli, Ornella Cutaia, Luciano Mutti, Aldo Morra, Riccardo Danielli, and Michele Maio
- Subjects
Male ,Mesothelioma ,medicine.medical_specialty ,Pleural Neoplasms ,Antineoplastic Agents ,Antibodies, Monoclonal, Humanized ,Gastroenterology ,anti-ctla4 ,NO ,Internal medicine ,Clinical endpoint ,Humans ,Medicine ,CTLA-4 Antigen ,Survival rate ,Aged ,Neoplasm Staging ,Salvage Therapy ,business.industry ,Antibodies, Monoclonal ,Middle Aged ,Prognosis ,medicine.disease ,Peritoneal Malignant Mesothelioma ,Surgery ,Survival Rate ,Oncology ,Tolerability ,Drug Resistance, Neoplasm ,Response Evaluation Criteria in Solid Tumors ,Female ,business ,Tremelimumab ,Progressive disease ,Follow-Up Studies ,medicine.drug - Abstract
Summary Background Monoclonal antibodies to cytotoxic T-lymphocyte antigen 4 (CTLA4) have therapeutic activity in different tumour types. We aimed to investigate the efficacy, safety, and immunological activity of the anti-CTLA4 monoclonal antibody, tremelimumab, in advanced malignant mesothelioma. Methods In our open-label, single-arm, phase 2 study, we enrolled patients aged 18 years or older with measurable, unresectable malignant mesothelioma and progressive disease after a first-line platinum-based regimen. Eligible patients had to have a life expectancy of 3 months or more, an Eastern Cooperative Oncology Group performance status of 2 or less, and no history of autoimmune disease. Patients received tremelimumab 15 mg/kg intravenously once every 90 days until progressive disease or severe toxicity. The primary endpoint was the proportion of patients who achieved an objective response (complete or partial response), with a target response rate of 17% according to the modified Response Evaluation Criteria in Solid Tumors (RECIST) for pleural malignant mesothelioma or standard RECIST 1.0 for peritoneal malignant mesothelioma. Analyses were done according to intention to treat. This trial is registered with EudraCT, number 2008-005171-95, and ClinicalTrials.gov, number NCT01649024. Findings Between May 27, 2009, and Jan 10, 2012, we enrolled 29 patients. All patients received at least one dose of tremelimumab (median two doses, range one to nine). No patients had a complete response and two patients (7%) had a durable partial response (one lasting 6 months and one lasting 18 months); one partial response occurred after initial progressive disease. Thus, the study did not reach its primary endpoint. However, we noted disease control in nine (31%) patients and a median progression-free survival of 6·2 months (95% CI 1·3–11·1) and a median overall survival of 10·7 months (0·0–21·9). 27 patients (93%) had at least one grade 1–2 treatment-emergent adverse event (mainly cutaneous rash, pruritus, colitis, or diarrhoea), and four patients (14%) had at least one grade 3–4 treatment-emergent adverse event (two gastrointestinal, one neurological, two hepatic, and one pancreatic). Interpretation Although the effect size was small in our phase 2 trial, tremelimumab seemed to have encouraging clinical activity and an acceptable safety and tolerability profile in previously treated patients with advanced malignant mesothelioma. Funding Associazione Italiana per la Ricerca sul Cancro, Istituto Toscano Tumori, Pfizer, and Fondazione Buzzi Unicem.
- Published
- 2013
29. Long-term survival and immunological parameters in metastatic melanoma patients who responded to ipilimumab 10 mg/kg within an expanded access programme
- Author
-
Ornella Cutaia, Diana Giannarelli, Luana Calabrò, Anna Maria Di Giacomo, Erica Bertocci, Maresa Altomonte, Clelia Miracco, Carolina Fazio, Ester Fonsatti, Maurizio Biagioli, Riccardo Danielli, Isabella Pesce, and Michele Maio
- Subjects
Oncology ,Adult ,CD4-Positive T-Lymphocytes ,Male ,Cancer Research ,medicine.medical_specialty ,Immunology ,Ipilimumab ,Antineoplastic Agents ,CD8-Positive T-Lymphocytes ,Metastatic melanoma ,Antibodies ,NO ,Expanded access programme ,Inducible T-Cell Co-Stimulator Protein ,Young Adult ,Long-term survival ,Internal medicine ,Monoclonal ,Long term survival ,medicine ,Immunology and Allergy ,Humans ,Lymphocyte Count ,Young adult ,Neoplasm Metastasis ,Melanoma ,Aged ,Neoplasm Staging ,Retrospective Studies ,Predictive marker ,Antibodies, Monoclonal ,Female ,Middle Aged ,Treatment Outcome ,business.industry ,Biomarker ,Retrospective cohort study ,medicine.disease ,Expanded access ,Biomarker (medicine) ,business ,medicine.drug - Abstract
Ipilimumab can result in durable clinical responses among patients with advanced melanoma. However, no predictive marker of clinical activity has yet been identified. We provide preliminary data describing the correlation between immunological parameters and response/survival among patients with advanced melanoma who received ipilimumab 10 mg/kg in an expanded access programme.Patients received ipilimumab 10 mg/kg every 3 weeks (Q3W) for four doses (induction) and Q12W from week 24 (W24) as maintenance therapy. Tumor assessments were conducted Q12W. Expression of inducible T cell costimulator (ICOS) on CD4(+) and CD8(+) T cells was assessed at baseline, W7, W12 and W24, and the ratio between absolute neutrophils (N) and lymphocytes (L) determined at baseline, W4, W7 and W10.Median overall survival among 27 patients was 9.6 months (95 % CI 3.2-16.1), with 3- and 4-year survival rates of 20.4 %. Five patients survived4 years. Patients with an increase in the number of circulating ICOS(+) T cells at W7 were more likely to experience disease control and have improved survival. An N/L ratio below the median at W7 and W10 was also associated with better survival compared with an N/L ratio above the median.Ipilimumab can induce long-term survival benefits in heavily pretreated patients with metastatic melanoma. Changes in the number of circulating ICOS(+) T cells or N/L ratio during ipilimumab treatment may represent early markers of response. However, given the limited sample size, further investigation is required.
- Published
- 2013
30. Expression and regulation of B7-H3 immunoregulatory receptor, in human mesothelial and mesothelioma cells: immunotherapeutic implications
- Author
-
Erica Bertocci, Francesca Colizzi, Pier Giorgio Natali, Anna Maria Di Giacomo, Luciano Mutti, Michele Maio, Riccardo Danielli, Luana Calabrò, Ester Fonsatti, Alessia Covre, Ornella Cutaia, and Luca Sigalotti
- Subjects
Mesothelioma ,Cell type ,B7 Antigens ,Physiology ,T cell ,medicine.medical_treatment ,Pleural Neoplasms ,Clinical Biochemistry ,Biology ,Cancer Vaccines ,Flow cytometry ,Immunophenotyping ,Neuroblastoma ,Antigen ,Antigens, CD ,Cell Line, Tumor ,medicine ,Humans ,Receptors, Immunologic ,medicine.diagnostic_test ,Antibodies, Monoclonal ,Epithelial Cells ,Cell Biology ,Immunotherapy ,Molecular biology ,Pleural Effusion ,medicine.anatomical_structure ,Hypomethylating agent ,Cell culture - Abstract
No treatment prolongs the survival of malignant mesothelioma (MM) patients. Since MM elicits anti-tumor host's immune responses, immunotherapy represents a promising strategy for its control. Immunomodulatory antibodies against components of the B7 family of immunomodulatory molecules that regulate T cell activation are being investigated in human malignancies including MM. The expression of B7-H3, a new component of the B7 family was investigated in primary cultures of human mesothelial cells (HMC) and in MM cell lines by flow cytometry and molecular analyses, and in MM tissues by immunohistochemistry. The role of DNA hypomethylating agents in modulating levels of B7-H3 expression in MM cells was also studied. Reverse transcriptase-polymerase chain reaction (RT-PCR) demonstrated that B7-H3 mRNA was consistently detectable in mesothelial and MM cells investigated; however, real-time quantitative RT-PCR analyses showed highly heterogeneous levels of B7-H3 mRNA among investigated MM cells. The analysis of B7-H3 protein expression indicated that comparable levels of B7-H3 were expressed on both cell types. Treatment with the DNA hypomethylating agent 5-aza-2'-deoxycytidine did not significantly affect the expression of B7-H3 mRNA in MM cells. In vivo, while B7-H3 was expressed in all 13 tumor biopsies of the epithelial variant, with high levels in 54% of cases, it was rarely detectable in spindle type MM in which 1/5 biopsies weakly expressed B7-H3. These findings suggest that B7-H3 is a promising target for new immunotherapeutic strategies in MM, with particular emphasis in the epithelial variant.
- Published
- 2011
31. CD40 expression by human melanocytic lesions and melanoma cell lines and direct CD40 targeting with the therapeutic anti-CD40 antibody CP-870.893
- Author
-
Pier Giorgio Natali, Ester Fonsatti, Michele Maio, Michael Chiou, Luana Calabrò, Anusha Kalbasi, Ornella Cutaia, William D. Tap, Erica Bertocci, Antoni Ribas, Bartosz Chmielowski, and Maresa Altomonte
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Biopsy ,Immunology ,Cell Separation ,Biology ,Antibodies, Monoclonal, Humanized ,Flow cytometry ,NO ,Cell Line, Tumor ,medicine ,CD40 ,melanoma ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,immunotherapy ,CD40 Antigens ,Neoplasm Metastasis ,Pharmacology ,medicine.diagnostic_test ,Melanoma ,Gene Expression Profiling ,Cancer ,Antibodies, Monoclonal ,Immunotherapy ,medicine.disease ,Flow Cytometry ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,Histone Deacetylase Inhibitors ,Cell culture ,Cancer research ,biology.protein ,Melanocytes ,Antibody - Abstract
Anti-CD40 antibodies are in clinical development in patients with metastatic melanoma, a cancer that has been reported earlier to express CD40. The antitumor activity of anti-CD40 antibodies may be mediated by direct cytotoxic effects on CD40-positive melanoma cells or indirectly through modulation of host cells. In these studies, biopsies of patients with primary and metastatic melanoma, short-term cultures, and established melanoma cell lines were analyzed for CD40 expression using a combination of methods including immunohistochemistry, flow cytometry, and gene expression profiling, and the cytotoxic effects of the anti-CD40 antibody CP-870,893 on melanoma cell lines were tested using cell viability assays. CD40 was expressed at a higher frequency in metastatic melanoma lesions compared with primary melanomas. There was a variable expression of CD40 in synchronous and metachronous melanoma metastases. Expression of CD40 was present in slightly over half of a large panel of short-term primary melanoma cultures, with a wide range of expression levels by flow cytometry. Similar results were obtained in established melanoma cell lines when analyzed at the mRNA level or by surface protein staining. In approximately one-third of cell lines, the expression of CD40 could be up-regulated with a histone deacetylase inhibitor. Treatment with increasing concentrations of CP-870,893 had no antitumor activity against either CD40-positive or negative melanoma cell lines. In conclusion, approximately one-third to one-half of melanomas expresses CD40 at variable levels. Direct exposure to a CD40-activating antibody does not lead to antitumor activity in melanoma cell lines, suggesting that the antitumor effects of these antibodies in the clinic may be indirectly mediated.
- Published
- 2010
32. Abstract LB-228: Pharmacokinetics of Tremelimumab, a fully human anti-CTLA-4 monoclonal antibody, in subjects with unresectable malignant mesothelioma
- Author
-
Ornella Cutaia, Carolina Fazio, Diego Annesi, Luana Calabrò, Ramy Ibrahim, Rajesh Narwal, Alessandra Di Pietro, Michele Maio, Paul B. Robbins, and Ester Fonsatti
- Subjects
Volume of distribution ,Cancer Research ,education.field_of_study ,business.industry ,Population ,Pharmacology ,medicine.disease ,NONMEM ,Oncology ,Pharmacokinetics ,medicine ,Trough level ,Dosing ,education ,business ,Tremelimumab ,Progressive disease ,medicine.drug - Abstract
Purpose: Tremelimumab is a fully human IgG2 monoclonal antibody specific for cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4/CD152). Tremelimumab blocks the interaction of CTLA-4 with its ligands B7.1 and B7.2, and enhances human T-cell activation, as demonstrated by increased interleukin-2 production in in-vitro/in-vivo studies. The primary objectives of this analysis were to assess the preliminary population pharmacokinetics (PK) of tremelimumab in mesothelioma and identify the impact of potential patient/disease characteristics on PK variability. Methods: Tremelimumab serum concentration-time data were collected from a second-line, single-arm phase-2 study (NCT01649024/NCT01655888) designed to evaluate clinical activity along with safety and tolerability of multiple IV doses of tremelimumab in patients with malignant mesothelioma. Two dosing regimens were tested: 15 mg/kg every 90 days (Q90D) up to 4 doses in a 12 month period until progressive disease or unacceptable toxicity and, 10 mg/kg every 4 weeks (Q4W) for 6 months followed by 10 mg/kg every 12 weeks (Q12W) until progressive disease or unacceptable toxicity. A total of 40 subjects provided evaluable PK data at various prespecified time points following both dosing regimens. Tremelimumab serum concentrations were determined using a validated enzyme-linked immunosorbent assay (ELISA) with a lower limit of quantitation (LLOQ) of 0.156 µg/mL. PK analysis was performed using a non-linear mixed effects modeling approach with NONMEM 7.2 software. Results: Tremelimumab PK exposure in mesothelioma subjects was similar to previous melanoma studies following 15 mg/kg Q90D. Following the 15 mg/kg Q90D, PK exposure was below the target trough level of ∼30 µg/mL for about half of the dosing interval with almost all subjects below the LLOQ at the end of the 90-day dosing interval. Following more frequent dosing of 10 mg/kg Q4W, PK exposure was increased and maintained at or above the target level in the majority of subjects over the entire dosing interval. Tremelimumab PK was best described using a 2-compartment linear model with first order elimination. Following IV dosing, the typical clearance (CL) and central volume of distribution (Vc) were 0.2 L/day and 3.5 L, respectively. The between-subject variability for CL and Vc were 22% and 7%, respectively. The estimated typical PK parameters were similar to other monoclonal antibodies without target mediated elimination. The baseline body weight and Eastern Cooperative Oncology Group (ECOG) performance status were identified as significant covariates for CL, whereas only baseline body weight was significant covariate for volume of distribution. Conclusions: A 2-compartment population PK model adequately described tremelimumab PK in subjects with mesothelioma. PK parameter estimates were similar in both mesothelioma and melanoma populations. Tremelimumab 10 mg/kg Q4W yielded higher exposure than 15 mg/kg Q90D with a majority of subjects maintaining concentrations above the target trough level of ∼30 µg/mL over the dosing interval. Preliminary covariate screening identified body weight and ECOG as influential factors for PK parameters. These correlations will be further validated using data from larger clinical trials. Citation Format: Luana Calabro', Rajesh Narwal, Paul B. Robbins, Alessandra Di Pietro, Ornella Cutaia, Ester Fonsatti, Diego Annesi, Carolina Fazio, Ramy Ibrahim, Michele Maio. Pharmacokinetics of Tremelimumab, a fully human anti-CTLA-4 monoclonal antibody, in subjects with unresectable malignant mesothelioma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr LB-228. doi:10.1158/1538-7445.AM2014-LB-228
- Published
- 2014
33. A phase 2 single-arm study with tremelimumab at an optimized dosing schedule in second-line mesothelioma patients
- Author
-
Riccardo Danielli, Maresa Altomonte, Luana Calabrò, Ester Fonsatti, Michele Maio, Ornella Cutaia, Diana Giannarelli, Carolina Fazio, Aldo Morra, and Anna Maria Di Giacomo
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,business.industry ,medicine.disease ,Monoclonal antibody ,Second line ,Antigen ,Internal medicine ,Immunology ,medicine ,Cytotoxic T cell ,Dosing ,Mesothelioma ,business ,Tremelimumab ,Single Arm Study ,medicine.drug - Abstract
7531 Background: We reported encouraging clinical activity of tremelimumab (treme), a fully human IgG 2 monoclonal antibody specific for cytotoxic T lymphocyte-associated antigen 4, at 15 mg/kg q3 ...
- Published
- 2014
34. A phase II study of intratumoral application of L19IL2/L19TNF in melanoma patients in clinical stage III or stage IV M1a with presence of injectable cutaneous and/or subcutaneous lesions
- Author
-
Michele Maio, Anna Maria Di Giacomo, Leonardo Giovannoni, Dario Neri, Annabella Di Florio, Pier Adelchi Ruffini, Ornella Cutaia, Andrea Lazzeri, Riccardo Danielli, Giuliano Elia, G. Gallino, Roberto Patuzzo, Mario Santinami, and Carolina Fazio
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Oncology ,business.industry ,Melanoma ,medicine ,Phases of clinical research ,Stage (cooking) ,medicine.disease ,business ,Stage iv - Abstract
TPS9103^ Background: Preclinical studies in three different murine models of subcutaneous tumors have shown that a single intratumoral injection of a combination of two immunocytokines, L19-IL2 and...
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.