71 results on '"Maria Vieito"'
Search Results
2. Supplementary Figure from A Novel EGFRvIII T-Cell Bispecific Antibody for the Treatment of Glioblastoma
- Author
-
Joan Seoane, Pablo Umaña, Josep Tabernero, Maria Vieito, Joan Carles, Paolo Nuciforo, Josep González, Alberto Di Somma, Luis Reyes-Figueroa, Estela Pineda, Christian Klein, Marina Bacac, Sara Colombetti, Johannes Sam, Franz Osl, Joerg Mueller, Stefanie Briner, Marine Richard, Virginie Steinhart, Thomas Pöschinger, Linda Fahrni, Xavier Guardia-Reyes, Simona Casalino, Marta Cicuendez, Esteban Cordero, Francisco Martínez-Ricarte, Alba Martínez-Moreno, Isabel Cuartas, Anne Freimoser-Grundschober, Valeria Nicolini, Alexandra Arias, Ester Bonfill-Teixidor, Ester Planas-Rigol, Inja Waldhauer, and Raffaella Iurlaro
- Abstract
Supplementary Figure from A Novel EGFRvIII T-Cell Bispecific Antibody for the Treatment of Glioblastoma
- Published
- 2023
3. Figure S1-S5 and Table S1-S3 from Capturing Hyperprogressive Disease with Immune-Checkpoint Inhibitors Using RECIST 1.1 Criteria
- Author
-
Elena Garralda, Raquel Perez-Lopez, Rodrigo Dienstmann, Josep Tabernero, Jordi Rodón, Enriqueta Felip, Joan Carles, Eva Muñoz-Couselo, Ana Oaknin, Elena Elez, Maria Alsina, Mafalda Oliveira, Roger Berché, Guillermo Villacampa, Jose Mateos, Javier Ros, Gemma Mur, Irene Braña, Maria Vieito, Analia Azaro, Cristina Viaplana, Maria Ochoa de Olza, Cinta Hierro, Alonso García-Ruiz, Juan Martin-Liberal, and Ignacio Matos
- Abstract
Figure S1. Flowchart of study selection process with ICIs treatment. Figure S2. Flowchart of study selection process with TAs treatment. Figure S3. Overall Survival comparing HPD with non-HPD in patients with progression disease as best response in the ICIs and TAs cohorts ( RECIST 1.1 progression disease Landmark Analysis). Figure S4. Overall Survival in second line (a) and third (b) setting HPD vs non-HPD progressors by RECIST criteria in ICIs cohort. Figure S5. Concordance between RECIST and TGR criteria in ICIs cohort. Table S1. Computed tomography (CT) acquisition protocol parameters. Table S2. Clinical variables in patients treated with TAs. Table S3. Multivariate model in patients with progression disease as best response in ICIs group (using TGR).
- Published
- 2023
4. Data from Capturing Hyperprogressive Disease with Immune-Checkpoint Inhibitors Using RECIST 1.1 Criteria
- Author
-
Elena Garralda, Raquel Perez-Lopez, Rodrigo Dienstmann, Josep Tabernero, Jordi Rodón, Enriqueta Felip, Joan Carles, Eva Muñoz-Couselo, Ana Oaknin, Elena Elez, Maria Alsina, Mafalda Oliveira, Roger Berché, Guillermo Villacampa, Jose Mateos, Javier Ros, Gemma Mur, Irene Braña, Maria Vieito, Analia Azaro, Cristina Viaplana, Maria Ochoa de Olza, Cinta Hierro, Alonso García-Ruiz, Juan Martin-Liberal, and Ignacio Matos
- Abstract
Purpose:Most hyperprogression disease (HPD) definitions are based on tumor growth rate (TGR). However, there is still no consensus on how to evaluate this phenomenon.Patients and Methods:We investigated two independent cohorts of patients with advanced solid tumors treated in phase I trials with (i) programmed cell death 1 (PD-1)/PD-L1 antibodies in monotherapy or combination and (ii) targeted agents (TA) in unapproved indications. A Response Evaluation Criteria in Solid Tumors (RECIST) 1.1–based definition of hyperprogression was developed. The primary endpoint was the assessment of the rate of HPD in patients treated with ICIs or TAs using both criteria (RECIST and TGR) and the impact on overall survival (OS) in patients who achieved PD as best response.Results:Among 270 evaluable patients treated with PD-1/PD-L1 inhibitors, 29 PD-1/PD-L1–treated patients (10.7%) had HPD by RECIST definition. This group had a significantly lower OS (median of 5.23 months; 95% CI, 3.97–6.45) when compared with the non-HPD progressor group (median, 7.33 months; 95% CI, 4.53–10.12; HR = 1.73, 95% CI, 1.05–2.85; P = 0.04). In a subset of 221 evaluable patients, 14 (6.3%) were categorized as HPD using TGR criteria, differences in median OS (mOS) between this group (mOS 4.2 months; 95% IC, 2.07–6.33) and non-HPD progressors (n = 44) by TGR criteria (mOS 6.27 months; 95% CI, 3.88–8.67) were not statistically significant (HR 1.4, 95% IC, 0.70–2.77; P = 0.346). Among 239 evaluable patients treated with TAs, 26 (10.9%) were classified as having HPD by RECIST and 14 using TGR criteria in a subset of patients. No differences in OS were observed between HPD and non-HPD progressors treated with TAs.Conclusions:HPD measured by TGR or by RECIST was observed in both cohorts of patients; however, in our series, there was an impact on survival only in the immune-checkpoint inhibitor cohort when evaluated by RECIST. We propose a new way to capture HPD using RECIST criteria that is intuitive and easy to use in daily clinical practice.
- Published
- 2023
5. BET inhibitor trotabresib in heavily pretreated patients with solid tumors and diffuse large B-cell lymphomas
- Author
-
Victor Moreno, Maria Vieito, Juan Manuel Sepulveda, Vladimir Galvao, Tatiana Hernández-Guerrero, Bernard Doger, Omar Saavedra, Carmelo Carlo-Stella, Jean-Marie Michot, Antoine Italiano, Massimo Magagnoli, Cecilia Carpio, Antonio Pinto, Rafael Sarmiento, Barbara Amoroso, Ida Aronchik, Ellen Filvaroff, Bishoy Hanna, Xin Wei, Zariana Nikolova, Irene Braña, Institut Català de la Salut, [Moreno V, Hernández-Guerrero T, Doger B] START Madrid-FJD, Hospital Universitario Fundación Jimenez Diaz, Madrid, Spain. [Vieito M, Galvao V, Saavedra O, Carpio C, Braña I] Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Sepulveda JM] Hospital Universitario 12 de Octubre, Madrid, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Multidisciplinary ,Otros calificadores::/uso terapéutico [Otros calificadores] ,General Physics and Astronomy ,Cèl·lules B - Tumors - Tractament ,Medicaments antineoplàstics - Ús terapèutic ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,General Chemistry ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,General Biochemistry, Genetics and Molecular Biology ,Neoplasms::Neoplasms by Histologic Type::Lymphoma::Lymphoma, Non-Hodgkin::Lymphoma, B-Cell::Lymphoma, Large B-Cell, Diffuse [DISEASES] ,neoplasias::neoplasias por tipo histológico::linfoma::linfoma no Hodgkin::linfoma de células B::linfoma de células B grandes difuso [ENFERMEDADES] ,Other subheadings::/therapeutic use [Other subheadings] ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] - Abstract
B-cell lymphoma; Cancer therapy; CNS cancer Limfoma de cèl·lules B; Teràpia del càncer; Càncer del SNC Linfoma de células B; Terapia del cáncer; Cáncer del SNC Bromodomain and extraterminal proteins (BET) play key roles in regulation of gene expression, and may play a role in cancer-cell proliferation, survival, and oncogenic progression. CC-90010-ST-001 (NCT03220347) is an open-label phase I study of trotabresib, an oral BET inhibitor, in heavily pretreated patients with advanced solid tumors and relapsed/refractory diffuse large B-cell lymphoma (DLBCL). Primary endpoints were the safety, tolerability, maximum tolerated dose, and RP2D of trotabresib. Secondary endpoints were clinical benefit rate (complete response [CR] + partial response [PR] + stable disease [SD] of ≥4 months’ duration), objective response rate (CR + PR), duration of response or SD, progression-free survival, overall survival, and the pharmacokinetics (PK) of trotabresib. In addition, part C assessed the effects of food on the PK of trotabresib as a secondary endpoint. The dose escalation (part A) showed that trotabresib was well tolerated, had single-agent activity, and determined the recommended phase 2 dose (RP2D) and schedule for the expansion study. Here, we report long-term follow-up results from part A (N = 69) and data from patients treated with the RP2D of 45 mg/day 4 days on/24 days off or an alternate RP2D of 30 mg/day 3 days on/11 days off in the dose-expansion cohorts (parts B [N = 25] and C [N = 41]). Treatment-related adverse events (TRAEs) are reported in almost all patients. The most common severe TRAEs are hematological. Toxicities are generally manageable, allowing some patients to remain on treatment for ≥2 years, with two patients receiving ≥3 years of treatment. Trotabresib monotherapy shows antitumor activity, with an ORR of 13.0% (95% CI, 2.8–33.6) in patients with R/R DLBCL (part B) and an ORR of 0.0% (95% CI, 0.0–8.6) and a CBR of 31.7% (95% CI, 18.1–48.1) in patients with advanced solid tumors (part C). These results support further investigation of trotabresib in combination with other anticancer agents. This study was sponsored by Celgene, a Bristol Myers Squibb Company. The study sponsor was involved in the study design, analysis of data, and writing the manuscript. Medical writing and editorial assistance were provided by Bernard Kerr, PGDipSci, and Agata Shodeke, of Spark, funded by Bristol Myers Squibb.
- Published
- 2023
6. Olutasidenib (FT-2102) in patients with relapsed or refractory IDH1-mutant glioma: A multicenter, open-label, phase Ib/II trial
- Author
-
Macarena I de la Fuente, Howard Colman, Mark Rosenthal, Brian A Van Tine, Danijela Levacic, Tobias Walbert, Hui K Gan, Maria Vieito, Mohammed M Milhem, Kathryn Lipford, Sanjeev Forsyth, Sylvie M Guichard, Yelena Mikhailov, Alexander Sedkov, Julie Brevard, Patrick F Kelly, Hesham Mohamed, Varun Monga, Institut Català de la Salut, [de la Fuente MI] Sylvester Comprehensive Cancer Center and Department of Neurology, University of Miami, Miami, Florida, USA. [Colman H] Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA. [Rosenthal M] Peter MacCallum Cancer Centre Melbourne, Victoria, Australia. [Van Tine BA] Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA. [Levacic D] Baylor and Scott White Vasicek Cancer Center, Baylor University Temple, Temple, Texas, USA. [Walbert T] Henry Ford Cancer Institute, Henry Ford Health System and Wayne State University, Detroit, Michigan, USA. [Vieito M] Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Cancer Research ,Gliomes - Tractament ,Neoplasms::Neoplasms by Histologic Type::Neoplasms, Germ Cell and Embryonal::Neuroectodermal Tumors::Neoplasms, Neuroepithelial::Glioma [DISEASES] ,Oncology ,Otros calificadores::/uso terapéutico [Otros calificadores] ,Medicaments antineoplàstics - Ús terapèutic ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,Other subheadings::/therapeutic use [Other subheadings] ,Neurology (clinical) ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,neoplasias::neoplasias por tipo histológico::neoplasias de células germinales y embrionarias::tumores neuroectodérmicos::neoplasias neuroepiteliales::glioma [ENFERMEDADES] - Abstract
Background Olutasidenib (FT-2102) is a highly potent, orally bioavailable, brain-penetrant and selective inhibitor of mutant isocitrate dehydrogenase 1 (IDH1). The aim of the study was to determine the safety and clinical activity of olutasidenib in patients with relapsed/refractory gliomas harboring an IDH1R132X mutation. Methods This was an open-label, multicenter, nonrandomized, phase Ib/II clinical trial. Eligible patients (≥18 years) had histologically confirmed IDH1R132X-mutated glioma that relapsed or progressed on or following standard therapy and had measurable disease. Patients received olutasidenib, 150 mg orally twice daily (BID) in continuous 28-day cycles. The primary endpoints were dose-limiting toxicities (DLTs) (cycle 1) and safety in phase I and objective response rate using the Modified Response Assessment in Neuro-Oncology criteria in phase II. Results Twenty-six patients were enrolled and followed for a median 15.1 months (7.3‒19.4). No DLTs were observed in the single-agent glioma cohort and the pharmacokinetic relationship supported olutasidenib 150 mg BID as the recommended phase II dose. In the response-evaluable population, disease control rate (objective response plus stable disease) was 48%. Two (8%) patients demonstrated a best response of partial response and eight (32%) had stable disease for at least 4 months. Grade 3‒4 adverse events (≥10%) included alanine aminotransferase increased and aspartate aminotransferase increased (three [12%], each). Conclusions Olutasidenib 150 mg BID was well tolerated in patients with relapsed/refractory gliomas harboring an IDH1R132X mutation and demonstrated preliminary evidence of clinical activity in this heavily pretreated population.
- Published
- 2022
7. Contributors
- Author
-
Samira Aghajani, Mazaher Ahmadi, Nobuyoshi Akimitsu, Iban Aldecoa, José Luís Alves, Veronica Aran, Ivan Archilla, Oscar Arrieta, Geetanjali Arora, C.S. Bal, Carmen Balaña, Joana Balça-Silva, Marcos Barbosa, João Basso, Amir Barzegar Behrooz, Mattia Bramini, Francesco Bruno, Célia Cabral, Andrés F. Cardona, Cristina Carrato, Simona Casalino, Rita Cascão, Valentina Castagnola, Sara Castañer Llanes, Miguel Castelo-Branco, Courtney Clark, Vanessa Coelho-Santos, Alexander B. Cook, Bárbara Costa, Bruno M. Costa, Gustavo Costa, Gabriella Costabile, Tânia F.G.G. Cova, Seyed Mohammad Hossein Dabiri, Ahmad Daher, Jéssica Delgado, Ana María Díaz-Lanza, Marta Domenech, Eva María Domínguez-Martín, Luiz Gustavo Dubois, Jason T. Duskey, Thomas Efferth, Claudia C. Faria, Ana Fortuna, Juan Esteban Garcia-Robledo, Saeid Ghavami, Alfredo Gimeno, Célia M.F. Gomes, Joana F. Guerreiro, Ankit Halder, Ainhoa Hernandez, Electra Eduina Hernández Santana, Santosh Kesari, Antonio Lopez-Rueda, Tayyebeh Madrakian, Mariana Magalhães, Bruno Manadas, Cláudia Martins, Diana Matias, Filipa Mendes, Maria Mendes, Donald W. Miller, Teresa Moran, Ana Moreira, Montse Moreno, Matteo Moschetta, Andrés Mosquera, Vivaldo Moura-Neto, Arani Mukherjee, Sandra C.C. Nunes, Laura Oleaga, Camila Ordoñez, Catarina Pacheco, Alberto A.C.C. Pais, António Paulo, Alessia Pellerino, Mariana Pereira, Estela Pineda, Catarina I.G. Pinto, Prasoon Prakash, Edoardo Pronello, Josep Puig, Teresa Ribalta, Patrícia Rijo, Katerina Rojas, Roberta Rudà, Luca Saba, Bruno Sarmento, Michele Schlich, Shreoshi Sengupta, Fernando Silva, Francisco Silva, Kumaravel Somasundaram, João Sousa, Sanjeeva Srivastava, Giovanni Tosi, Martina Trevisani, Nuno Vale, Carla Varela, Ayushi Verma, Maria Vieito, Carla Vitorino, Rui Vitorino, Tavia Walsh, and Vinith Yathindranath
- Published
- 2023
8. The role of vaccines in glioblastoma—Updated clinical results
- Author
-
Maria Vieito, Simona Casalino, Montse Moreno, and Katerina Rojas
- Published
- 2023
9. A Pan-Cancer Clinical Platform to Predict Immunotherapy Outcomes and Prioritize Immuno-Oncology Combinations in Early-Phase Trials
- Author
-
Alberto Hernando, Maria Vila-Casadesús, Yacine Bareche, Alberto Gonzalez-Medina, Francesco Mancuso, Deborah Lo Giacco, Agatha Martin, Omar Saavedra, Irene Brana, Maria Vieito, Roberta Fasani, John Stagg, Farnoosh Abbas-Aghababazadeh, Benjamin Haibe-Kains, Roger Berche, Nadia Saoudi Gonzalez, Claudia Valverde, Eva Muñoz-Couselo, Cristina Suarez, Marc Diez, Maria Elena Elez, Jaume Capdevila, Ana Oaknin, Cristina Saura, Teresa Macarulla, Joan Carles Galceran, Enriqueta Felip, Rodrigo Dienstmann, Philippe L. Bedard, Paolo Nuciforo, Joan Seoane, Josep Tabernero, Elena Garralda, and Ana Vivancos
- Published
- 2023
10. 642 EO2401 microbiome derived therapeutic vaccine + nivolumab, with/without standard continuous, or low-dose symptom directed, bevacizumab, in recurrent glioblastoma: phase 1–2 EOGBM1–18/ROSALIE study
- Author
-
David Reardon, Ahmed Idbaih, Maria Vieito, Ghazaleh Tabatabai, Agostina Stradella, Francois Ghiringhelli, Michael Burger, Iris Mildenberger, Ulrich Herrlinger, Macarena González, Marta GilMartin, Mirjam Renovanz, Mehdi Touat, Patrick Wen, Antje Wick, Cecile Gouttefangeas, Ana Maia, Chistophe Bonny, Jan Fagerberg, and Wolfgang Wick
- Published
- 2022
11. 589 Xevinapant plus nivolumab in patients with advanced solid tumors who progressed on prior anti–PD-1/PD-L1 treatment: results of a dose-optimization, exploratory phase 1b/2 trial
- Author
-
Emiliano Calvo Aller, Glenn Hanna, Maria Vieito Villar, Caroline Even, Victor Moreno, Chul Kim, Shuchi Gulati, Daniel Morgensztern, Ana Acuna-Villaorduna, Philippe Cassier, Dennie Jones, Florilene Bouisset, Daniela Sahlender, Elisabeth Rouits, Dany Spaggiari, Lars Damstrup, and Carlos-Alberto Gomez-Roca
- Published
- 2022
12. 641 Strong immune response to therapeutic vaccination with EO2401 microbiome derived therapeutic vaccine + nivolumab: interim report of the EOGBM1–18/ROSALIE study
- Author
-
Ana Maia, Hélène Toussaint, Joao Gamelas Magalhaes, David Reardon, Ahmed IDBAIH, Maria Vieito, Ghazaleh Tabatabai, Agostina Stradella, François Ghiringhelli, Michael C Burger, Iris Mildenberger, Ulrich Herrlinger, Macarena Gonzalez, Alice Hervieu, Marta GilMartin, Mirjam Renovanz, Mehdi Touat, Patrick Y Wen, Antje Wick, Laurent Chene, Cécile Gouttefangeas, Christophe Bonny, Jan Fagerberg, and Wolfgang Wick
- Published
- 2022
13. Trotabresib (CC-90010) in combination with adjuvant temozolomide or concomitant temozolomide plus radiotherapy in patients with newly diagnosed glioblastoma
- Author
-
Maria Vieito, Matteo Simonelli, Filip de Vos, Victor Moreno, Marjolein Geurts, Elena Lorenzi, Marina Macchini, Martin J van den Bent, Gianluca Del Conte, Maja de Jonge, Maria Cruz Martín-Soberón, Barbara Amoroso, Tania Sanchez-Perez, Marlene Zuraek, Bishoy Hanna, Ida Aronchik, Ellen Filvaroff, Henry Chang, Cristina Mendez, Marina Arias Parro, Xin Wei, Zariana Nikolova, Juan Manuel Sepulveda, Neurology, Medical Oncology, Institut Català de la Salut, [Vieito M] Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. [Simonelli M] Department of Biomedical Sciences, Humanitas University, Milan, Italy. IRCCS Humanitas Research Hospital, Milan, Italy. [de Vos F] Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands. [Moreno V] START Madrid-FJD, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain. [Geurts M] Erasmus MC Cancer Institute, Rotterdam, the Netherlands. [Lorenzi E] IRCCS Humanitas Research Hospital, Milan, Italy, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Glioblastoma multiforme - Tractament ,terapéutica::terapéutica::farmacoterapia::protocolos antineoplásicos::terapéutica::farmacoterapia::protocolos de quimioterapia antineoplásica combinada [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Other subheadings::/therapy [Other subheadings] ,Therapeutics::Radiotherapy [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,General Medicine ,Neoplasms::Neoplasms by Histologic Type::Neoplasms, Germ Cell and Embryonal::Neuroectodermal Tumors::Neoplasms, Neuroepithelial::Glioma::Astrocytoma::Glioblastoma [DISEASES] ,Glioblastoma multiforme - Radioteràpia ,Quimioteràpia combinada ,neoplasias::neoplasias por tipo histológico::neoplasias de células germinales y embrionarias::tumores neuroectodérmicos::neoplasias neuroepiteliales::glioma::astrocitoma::glioblastoma [ENFERMEDADES] ,terapéutica::radioterapia [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Therapeutics::Therapeutics::Drug Therapy::Antineoplastic Protocols::Therapeutics::Drug Therapy::Antineoplastic Combined Chemotherapy Protocols [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Temozolomide ,Pharmacokinetics ,Trotabresib ,BET inhibitors ,Glioblastoma ,Otros calificadores::/terapia [Otros calificadores] - Abstract
Background Standard-of-care treatment for newly diagnosed glioblastoma (ndGBM), consisting of surgery followed by radiotherapy (RT) and temozolomide (TMZ), has improved outcomes compared with RT alone; however, prognosis remains poor. Trotabresib, a novel bromodomain and extraterminal inhibitor, has demonstrated antitumor activity in patients with high-grade gliomas. Methods In this phase Ib, dose-escalation study (NCT04324840), we investigated trotabresib 15, 30, and 45 mg combined with TMZ in the adjuvant setting and trotabresib 15 and 30 mg combined with TMZ+RT in the concomitant setting in patients with ndGBM. Primary endpoints were to determine safety, tolerability, maximum tolerated dose, and/or recommended phase II dose (RP2D) of trotabresib. Secondary endpoints were assessment of preliminary efficacy and pharmacokinetics. Pharmacodynamics were investigated as an exploratory endpoint. Results The adjuvant and concomitant cohorts enrolled 18 and 14 patients, respectively. Trotabresib in combination with TMZ or TMZ+RT was well tolerated; most treatment-related adverse events were mild or moderate. Trotabresib pharmacokinetics and pharmacodynamics in both settings were consistent with previous data for trotabresib monotherapy. The RP2D of trotabresib was selected as 30 mg 4 days on/24 days off in both settings. At last follow-up, 5 (28%) and 6 (43%) patients remain on treatment in the adjuvant and concomitant settings, respectively, with 1 patient in the adjuvant cohort achieving complete response. Conclusions Trotabresib combined with TMZ in the adjuvant setting and with TMZ+RT in the concomitant setting was safe and well tolerated in patients with ndGBM, with encouraging treatment durations. Trotabresib 30 mg was established as the RP2D in both settings.
- Published
- 2022
14. First-in-human phase I/II, open-label study of the anti-OX40 agonist INCAGN01949 in patients with advanced solid tumors
- Author
-
Elizabeth J Davis, Juan Martin-Liberal, Rebecca Kristeleit, Daniel C Cho, Sarah P Blagden, Dominik Berthold, Dana B Cardin, Maria Vieito, Rowan E Miller, Prashanth Hari Dass, Angela Orcurto, Kristen Spencer, John E Janik, Jason Clark, Thomas Condamine, Jennifer Pulini, Xuejun Chen, Janice M Mehnert, Institut Català de la Salut, [Davis EJ] Vanderbilt University Medical Center, Nashville, Tennessee, USA. [Martin-Liberal J, Vieito M] Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Kristeleit R] Research Department of Oncology, University College London, London, UK. [Cho DC] Perlmutter Cancer Center, NYU Langone Health, NYU Grossman School of Medicine, New York, USA. [Blagden SP] Department of Oncology, University of Oxford, Oxford, UK. [Berthold D] Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Pharmacology ,Cancer Research ,Maximum Tolerated Dose ,Càncer - Tractament ,Immunology ,Medicaments antineoplàstics - Ús terapèutic ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,diagnóstico::pronóstico::resultado del tratamiento [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,Antibodies, Monoclonal ,Antineoplastic Agents ,Receptors, OX40 ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Diagnosis::Prognosis::Treatment Outcome [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Neoplasms [DISEASES] ,neoplasias [ENFERMEDADES] ,Oncology ,Neoplasms ,Avaluació de resultats (Assistència sanitària) ,Molecular Medicine ,Immunology and Allergy ,Humans ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] - Abstract
BackgroundOX40 is a costimulatory receptor upregulated on antigen-activated T cells and constitutively expressed on regulatory T cells (Tregs). INCAGN01949, a fully human immunoglobulin G1κ anti-OX40 agonist monoclonal antibody, was designed to promote tumor-specific immunity by effector T-cell activation and Fcγ receptor-mediated Treg depletion. This first-in-human study was conducted to determine the safety, tolerability, and preliminary efficacy of INCAGN01949.MethodsPhase I/II, open-label, non-randomized, dose-escalation and dose-expansion study conducted in patients with advanced or metastatic solid tumors. Patients received INCAGN01949 monotherapy (7–1400 mg) in 14-day cycles while deriving benefit. Safety measures, clinical activity, pharmacokinetics, and pharmacodynamic effects were assessed and summarized with descriptive statistics.ResultsEighty-seven patients were enrolled; most common tumor types were colorectal (17.2%), ovarian (8.0%), and non-small cell lung (6.9%) cancers. Patients received a median three (range 1–9) prior therapies, including immunotherapy in 24 patients (27.6%). Maximum tolerated dose was not reached; one patient (1.1%) receiving 350 mg dose reported dose-limiting toxicity of grade 3 colitis. Treatment-related adverse events were reported in 45 patients (51.7%), with fatigue (16 (18.4%)), rash (6 (6.9%)), and diarrhea (6 (6.9%)) being most frequent. One patient (1.1%) with metastatic gallbladder cancer achieved a partial response (duration of 6.3 months), and 23 patients (26.4%) achieved stable disease (lasting >6 months in one patient). OX40 receptor occupancy was maintained over 90% among all patients receiving doses of ≥200 mg, while no treatment-emergent antidrug antibodies were detected across all dose levels. Pharmacodynamic results demonstrated that treatment with INCAGN01949 did not enhance proliferation or activation of T cells in peripheral blood or reduce circulating Tregs, and analyses of tumor biopsies did not demonstrate any consistent increase in effector T-cell infiltration or function, or decrease in infiltrating Tregs.ConclusionNo safety concerns were observed with INCAGN01949 monotherapy in patients with metastatic or advanced solid tumors. However, tumor responses and pharmacodynamic effects on T cells in peripheral blood and post-therapy tumor biopsies were limited. Studies evaluating INCAGN01949 in combination with other therapies are needed to further evaluate the potential of OX40 agonism as a therapeutic approach in patients with advanced solid tumors.Trial registration numberNCT02923349.
- Published
- 2022
15. SEOM clinical guideline for management of adult medulloblastoma (2020)
- Author
-
R. Luque, M. Benavides, L. Egaña, Pedro Pérez-Segura, S. del Barco, Joaquín M. Sepúlveda, Maria Martinez-Garcia, Maria Vieito, Estela Pineda, and J. A. García-Gómez
- Subjects
Adult ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adult Medulloblastoma ,medicine.medical_treatment ,Clinical Guides in Oncology ,Antineoplastic Agents ,Medical Oncology ,Postoperative Complications ,Internal medicine ,Epidemiology ,medicine ,Humans ,Adults ,Molecular Targeted Therapy ,Cerebellar Neoplasms ,Societies, Medical ,Medulloblastoma ,Chemotherapy ,Evidence-Based Medicine ,Radiotherapy ,business.industry ,Palliative Care ,Consolidation Chemotherapy ,General Medicine ,Guideline ,Prognosis ,medicine.disease ,Combined Modality Therapy ,Treatment ,Clinical trial ,Tolerability ,Spain ,Vincristine ,Retreatment ,Cisplatin ,Neoplasm Recurrence, Local ,business - Abstract
Recent advances in molecular profiling, have reclassified medulloblastoma, an undifferentiated tumor of the posterior fossa, in at least four diseases, each one with differences in prognosis, epidemiology and sensibility to different treatments. The recommended management of a lesion with radiological characteristics suggestive of MB includes maximum safe resection followed by a post-surgical MR 1.5 cm2, presence of micro- or macroscopic dissemination, and age > 3 years as well as pathological (presence of anaplastic or large cell features) and molecular findings (group, 4, 3 or p53 SHH mutated subgroup) determine the risk of relapse and should guide adjuvant management. Although there is evidence that both high-risk patients and to a lesser degree, standard-risk patients benefit from adjuvant craneoespinal radiation followed by consolidation chemotherapy, tolerability is a concern in adult patients, leading invariably to dose reductions. Treatment after relapse is to be considered palliative and inclusion on clinical trials, focusing on the molecular alterations that define each subgroup, should be encouraged. Selected patients can benefit from surgical rescue or targeted radiation or high-dose chemotherapy followed by autologous self-transplant. Even in patients that are cured by chemorradiation presence of significant sequelae is common and patients must undergo lifelong follow-up.
- Published
- 2021
16. A Novel EGFRvIII T-Cell Bispecific Antibody for the Treatment of Glioblastoma
- Author
-
Raffaella Iurlaro, Inja Waldhauer, Ester Planas-Rigol, Ester Bonfill-Teixidor, Alexandra Arias, Valeria Nicolini, Anne Freimoser-Grundschober, Isabel Cuartas, Alba Martínez-Moreno, Francisco Martínez-Ricarte, Esteban Cordero, Marta Cicuendez, Simona Casalino, Xavier Guardia-Reyes, Linda Fahrni, Thomas Pöschinger, Virginie Steinhart, Marine Richard, Stefanie Briner, Joerg Mueller, Franz Osl, Johannes Sam, Sara Colombetti, Marina Bacac, Christian Klein, Estela Pineda, Luis Reyes-Figueroa, Alberto Di Somma, Josep González, Paolo Nuciforo, Joan Carles, Maria Vieito, Josep Tabernero, Pablo Umaña, and Joan Seoane
- Subjects
ErbB Receptors ,Cancer Research ,Oncology ,Brain Neoplasms ,Cell Line, Tumor ,T-Lymphocytes ,Antibodies, Bispecific ,Receptors, Antigen, T-Cell ,Animals ,Cytokines ,Humans ,Glioblastoma - Abstract
T-cell bispecific antibodies (TCB) are engineered molecules that bind both the T-cell receptor and tumor-specific antigens. Epidermal growth factor receptor variant III (EGFRvIII) mutation is a common event in glioblastoma (GBM) and is characterized by the deletion of exons 2–7, resulting in a constitutively active receptor that promotes cell proliferation, angiogenesis, and invasion. EGFRvIII is expressed on the surface of tumor cells and is not expressed in normal tissues, making EGFRvIII an ideal neoantigen target for TCBs. We designed and developed a novel 2+1 EGFRvIII-TCB with optimal pharmacologic characteristics and potent antitumor activity. EGFRvIII-TCB showed specificity for EGFRvIII and promoted tumor cell killing as well as T-cell activation and cytokine secretion only in patient-derived models expressing EGFRvIII. Moreover, EGFRvIII-TCB promoted T-cell recruitment into intracranial tumors. EGFRvIII-TCB induced tumor regression in GBM animal models, including humanized orthotopic GBM patient-derived xenograft models. Our results warrant the clinical testing of EGFRvIII-TCB for the treatment of EGFRvIII-expressing GBMs.
- Published
- 2022
17. Olutasidenib (FT-2102) in patients with relapsed or refractory IDH1-mutant glioma: a multicenter, open-label, phase 1b/2 trial
- Author
-
Macarena I, de la Fuente, Howard, Colman, Mark, Rosenthal, Brian A, Van Tine, Danijela, Levacic, Tobias, Walbert, Hui K, Gan, Maria, Vieito, Mohammed M, Milhem, Kathryn, Lipford, Sanjeev, Forsyth, Sylvie M, Guichard, Yelena, Mikhailov, Alexander, Sedkov, Julie, Brevard, Patrick F, Kelly, Hesham, Mohamed, and Varun, Monga
- Abstract
Olutasidenib (FT2102) is a highly potent, orally bioavailable, brain-penetrant and selective inhibitor of mutant isocitrate dehydrogenase 1 (IDH1). The aim of the study was to determine the safety and clinical activity of olutasidenib in patients with relapsed/refractory gliomas harboring an IDH1 R132X mutation.This was an open-label, multicenter, non-randomized, phase 1b/2 clinical trial. Eligible patients (≥18 years) had histologically confirmed IDH1 R132Xmutated glioma that relapsed or progressed on or following standard therapy and had measurable disease. Patients received olutasidenib, 150 mg orally twice daily (BID) in continuous 28-day cycles. The primary endpoints were dose-limiting toxicities (DLTs) (cycle 1) and safety in phase 1 and objective response rate using the Modified Response Assessment in Neuro-Oncology criteria in phase 2.Twenty-six patients were enrolled and followed for a median 15.1 months (7.3‒19.4). No DLTs were observed in the single-agent glioma cohort and the pharmacokinetic relationship supported olutasidenib 150 mg BID as the recommended phase 2 dose. In the response-evaluable population, disease control rate (objective response plus stable disease) was 48%. Two (8%) patients demonstrated a best response of partial response and eight (32%) had stable disease for at least 4 months. Grade 3‒4 adverse events (≥10%) included alanine aminotransferase increased and aspartate aminotransferase increased (three [12%], each).Olutasidenib 150 mg BID was well tolerated in patients with relapsed/refractory gliomas harboring an IDH1 R132X mutation and demonstrated preliminary evidence of clinical activity in this heavily pretreated population.
- Published
- 2022
18. Phase I prognostic online (PIPO): A web tool to improve patient selection for oncology early phase clinical trials
- Author
-
Teresa Macarulla, A. Pedrola, O. Saavedra, Javier Ros, Cristina Saura, Cristina Viaplana, Elena Garralda, R. Berché, Elena Elez, Enriqueta Felip, I. Gardeazabal, Maria Ochoa de Olza, Irene Brana, Rodrigo Dienstmann, Joan Carles, Juan Martin-Liberal, Guzman Alonso, Guillermo Villacampa, Jordi Rodon, Cinta Hierro, Eva Muñoz-Couselo, A. Hernando-Calvo, Analia Azaro, Maria Vieito, Vladimir Galvao, Josep Tabernero, Ignacio Matos, Ana Oaknin, Institut Català de la Salut, [Matos I, Carles J] Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain. [Villacampa G, Berché R, Pedrola A, Viaplana C, Dienstmann R] Oncology Data Science (OdysSey) Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Hierro C, Martin-Liberal J, Braña I, Azaro A, Vieito M, Saavedra O, Gardeazabal I, Hernando-Calvo A, Alonso G, Galvao V, Ochoa de Olza M, Ros J, Muñoz-Couselo E, Elez E, Rodon J, Saura C, Macarulla T, Oaknin A, Felip E, Garralda E] Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Tabernero J] Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Department of Medicine, UVic-UCC, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Male ,Oncology ,Information Science::Informatics::Medical Informatics::Medical Informatics Applications [INFORMATION SCIENCE] ,Cancer Research ,medicine.medical_specialty ,Prognostic variable ,Oncologia ,Phases of clinical research ,Ciencias de la información::informática::informática médica::aplicaciones de la informática médica [CIENCIA DE LA INFORMACIÓN] ,Medicina - Informàtica ,Medical Oncology ,Web tool ,neoplasias [ENFERMEDADES] ,Patient Portals ,Internal medicine ,medicine ,Humans ,Selection (genetic algorithm) ,técnicas de investigación::métodos::diseño de la investigación::selección de los pacientes [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Aged ,Clinical Trials as Topic ,business.industry ,Patient Selection ,Middle Aged ,Prognosis ,Clinical trial ,Neoplasms [DISEASES] ,Cohort ,Prognostic model ,Investigative Techniques::Methods::Research Design::Patient Selection [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Female ,Early phase ,business ,Internet-Based Intervention ,Assaigs clínics - Abstract
Immunotherapy; Phase 1 trials; Prognostic model Inmunoterapia; Ensayos de fase 1; Modelo pronóstico Immunoteràpia; Assajos de fase 1; Model pronòstic Purpose Patient selection in phase 1 clinical trials (Ph1t) continues to be a challenge. The aim of this study was to develop a user-friendly prognostic calculator for predicting overall survival (OS) outcomes in patients to be included in Ph1t with immune checkpoint inhibitors (ICIs) or targeted agents (TAs) based on clinical parameters assessed at baseline. Methods Using a training cohort with consecutive patients from the VHIO phase 1 unit, we constructed a prognostic model to predict median OS (mOS) as a primary endpoint and 3-month (3m) OS rate as a secondary endpoint. The model was validated in an internal cohort after temporal data splitting and represented as a web application. Results We recruited 799 patients (training and validation sets, 558 and 241, respectively). Median follow-up was 21.2 months (m), mOS was 10.2 m (95% CI, 9.3–12.7) for ICIs cohort and 7.7 m (95% CI, 6.6–8.6) for TAs cohort. In the multivariable analysis, six prognostic variables were independently associated with OS – ECOG, number of metastatic sites, presence of liver metastases, derived neutrophils/(leukocytes minus neutrophils) ratio [dNLR], albumin and lactate dehydrogenase (LDH) levels. The phase 1 prognostic online (PIPO) calculator showed adequate discrimination and calibration performance for OS, with C-statistics of 0.71 (95% CI 0.64–0.78) in the validation set. The overall accuracy of the model for 3m OS prediction was 87.2% (95% CI 85%–90%). Conclusions PIPO is a user-friendly objective and interactive tool to calculate specific survival probabilities for each patient before enrolment in a Ph1t. The tool is available at https://pipo.vhio.net/. The research leading to these results has received funding from “la Caixa” Foundation (LCF/PR/CE07/50610001). Cellex Foundation for providing research facilities and equipment. This work was supported by the Accelerator Award (UpSMART) from Fundacion Científica – Asociacion Espanola Contra el Cancer (FC -AECC)/ Associazione Italiana per la Ricerca sul Cancro (AIRC) /Cancer Research United Kingdom (CRUK).
- Published
- 2021
19. CTIM-17. EO2401 THERAPEUTIC VACCINE FOR PATIENTS WITH RECURRENT GLIOBLASTOMA: PHASE 1/2 ROSALIE STUDY (NCT04116658)
- Author
-
David A Reardon, Ahmed Idbaih, Maria Vieito, Ghazaleh Tabatabai, Agostina Stradella, François Ghiringhelli, Michael C Burger, Iris Mildenberger, Macarena González, Alice Hervieu, Marta Gil Martin, Mirjam Renovanz, Mehdi Touat, Patrick Y Wen, Antje Wick, Cécile Gouttefangeas, Ana Maia, Christophe Bonny, Jan Fagerberg, and Wolfgang Wick
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
EO2401 includes microbial-derived, synthetically produced HLA-A2 restricted peptides with molecular mimicry to antigens (IL13Rα2, BIRC5 and FOXM1) upregulated in glioblastoma, and the CD4 helper peptide UCP2. Patients with glioblastoma at first progression received EO2401 (300µg/peptide, q2weeks x4 then q4weeks), EO2401+nivolumab (3mg/kg q2weeks), or E02401+nivolumab+bevacizumab (10mg/kg q2weeks). Cohort-1 included EO2401x2 then EO2401+nivolumab. EO2401+nivolumab was evaluated in Cohort-2a, as adjuvant treatment in Cohort-2b, and as neoadjuvant/adjuvant treatment in Cohort-2c. Cohort-3 assessed EO2401+nivolumab+bevacizumab. Part 1 included 40 patients (Cohort-1/3, Cohort-2a/23, Cohort-2b/3, Cohort-3/11). Part 2 allowed low-dose-bevacizumab (5mg/kg q2weeks) for symptomatic edema and enrolled 38 patients (Cohort-1/18, Cohort-2a/15, Cohort-2b/3; and recruiting Cohort-2c/2 target 6, Cohort-3/0 target 15).Safety assessment of part 1 showed EO2401+nivolumab+/-bevacizumab to be well tolerated with EO2401 associated toxicity limited to local administration site reactions (48%; all grade 1-2). The nivolumab-/bevacizumab-toxicity was consistent with historical single-agent data. Strong CD8 T cell ELISPOT responses against the 3 vaccine peptides and cross-reactivity against targeted antigens was demonstrated in the majority of evaluable patients. Immune response was confirmed with tetramer staining of specific CD8 either ex vivo or after in vitro stimulation. For part 1, median progression-free survival (mPFS), and median survival (mOS) for EO2401+nivolumab (Cohorts-1/2/2b, n=29 median follow-up [mFU] 14.0 months) were 1.8 and 10.6 months. Patients on EO2401+nivolumab+bevacizumab (n = 11 mFU 9.6 m) had mPFS 5.5 months and 9 patients alive 7-12.4 months. Objective Response Rate/Disease Control Rate for EO2401+nivolumab and EO2401+nivolumab+bevacizumab was 10%/34% and 55%/82%.Median treatment duration for Cohort-2a part 1 was 6.1 weeks (1/23 on treatment), while it was 10.0 weeks (8/15 on treatment) for Cohort-2a part 2. Overall, in part 2, 36% received low-dose-bevacizumab.EO2401 generated strong immune responses and was well tolerated. Addition of standard bevacizumab to EO2401+nivolumab improved PFS and tumor response. Symptom driven low-dose-bevacizumab supported longer treatment durations. Outcome of study part 2 will be presented.
- Published
- 2022
20. CTNI-21. TROTABRESIB (CC-90010) IN COMBINATION WITH CONCOMITANT TEMOZOLOMIDE PLUS RADIOTHERAPY AND ADJUVANT TEMOZOLOMIDE IN PATIENTS WITH NEWLY DIAGNOSED GLIOBLASTOMA: UPDATED RESULTS FROM A PHASE 1B/2 STUDY
- Author
-
Maria Vieito, Juan Manuel Sepulveda, Victor Moreno, Filip de Vos, Marjolein Geurts, Elena Lorenzi, Marina Macchini, Martin van den Bent, Gianluca Del Conte, Maria Cruz Martín-Soberón, Petter Brandal, Maria Martinez Garcia, Barbara Amoroso, Tania Sanchez-Perez, Marlene Zuraek, Bishoy Hanna, Ellen Filvaroff, Henry Chang, Marina Arias Parro, Xin Wei, Yu Liu, Zariana Nikolova, and Matteo Simonelli
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Trotabresib, a novel bromodomain and extraterminal protein inhibitor, has demonstrated antitumor activity and blood–brain barrier penetration in patients with high-grade gliomas, and enhanced the antiproliferative effects of temozolomide in preclinical models. CC-90010-GBM-002 (NCT04324840) is a phase 1b/2 study investigating the addition of trotabresib to standard-of-care (SOC) concomitant temozolomide plus radiotherapy and adjuvant temozolomide, followed by maintenance trotabresib, in patients with newly diagnosed glioblastoma. The design of the dose escalation (part A) has been described previously (Vieito M, et al. SNO 2021. Abstract CTNI-51). Primary objectives of part A were to establish the safety, tolerability, and maximum tolerated dose/recommended phase 2 dose (RP2D) of trotabresib. In part A, addition of trotabresib to SOC was safe and well tolerated in the concomitant (N = 14) and adjuvant (N = 18) cohorts; the most frequent grade 3/4 treatment-related adverse event was thrombocytopenia (7/14 and 9/18 patients, respectively). The RP2D for trotabresib was 30 mg/day 4 days on/24 days off in both settings. At data cutoff (February 20, 2022), median duration of treatment was 34 weeks (concomitant cohort) and 33 weeks (adjuvant cohort); progression-free survival data are not yet mature. Trotabresib plasma pharmacokinetics and pharmacodynamics were consistent with monotherapy. At last follow-up, 6 and 5 patients remained on treatment in the concomitant and adjuvant dose-escalation cohorts, respectively, including 1 patient in cycle 20 with ongoing complete response. The ongoing randomized phase 2 dose expansion (part B; planned N = 162) is comparing concomitant trotabresib at the RP2D + SOC followed by adjuvant trotabresib at the RP2D + SOC, followed by maintenance trotabresib 45 mg/day 4 days on/24 days off, versus SOC alone in patients with newly diagnosed IDH–wild-type glioblastoma. Key objectives are to compare progression-free and overall survival, safety, and tolerability. Longer follow-up from part A and the first disclosure of data from part B will be presented.
- Published
- 2022
21. Safety and efficacy results from the expansion phase of the first-in-human study evaluating TGFβ inhibitor SAR439459 alone and combined with cemiplimab in adults with advanced solid tumors
- Author
-
Debbie Robbrecht, Bernard Doger, Jean-Jacques Grob, Oliver Edgar Bechter, Maria J. de Miguel, Maria Vieito, Dirk Schadendorf, Giuseppe Curigliano, Ivan Borbath, Marcus O. Butler, Alejo Rodriguez-Vida, Wilson H. Miller, Tun Tun Lin, Nina Masson, Clemence Pouzin, Rui Wang, Brigitte Demers, Amele Amrate, Giovanni Abbadessa, and Matteo Simonelli
- Subjects
Cancer Research ,Oncology - Abstract
2524 Background: SAR439459 (SAR459) is a human anti-TGFβ monoclonal antibody that neutralizes all isoforms of TGFβ. In preclinical models, combining SAR459 with an anti-PD-1 showed improved anti-tumor activity compared to SAR459 single agent. In the dose escalation, acceptable tolerability was observed, the MTD was not reached and the preliminary RP2D was 22.5mg/kg Q3W when combined with cemiplimab (CEMI; S. Williamson et al. J Clin Oncol 39, 2021[suppl 15; #2510]). Reduction of plasma TGFβ was ≥90% at doses ≥0.25mg/kg Q2W, with a trend of a decrease in intra-tumoral TGFβ (D. Robbrecht et al. JITC 2021;9 [suppl 2; #250]). Here we report safety and efficacy results of the dose expansion. Methods: The expansion phase of this open-label, phase 1/1b study aimed to determine the optimal dose of SAR459 (7.5 mg/kg or 22.5 mg/kg Q3W) in patients (pts) with advanced melanoma (MEL) resistant to anti-PD(L)1 therapy (Part 2A); and the ORR (confirmed responses) in all treated pts with SAR459 22.5 mg/kg + CEMI 350 mg Q3W in pts with MEL, Non-small Cell Lung Cancer (NSCLC), or Hepatocellular Carcinoma (HCC), resistant to anti-PD(L)1; as well as in pts with mesenchymal Colorectal Cancer (CRC) or Urothelial Cancer (UC), anti-PD(L)1 naïve (Part 2B). Results: From October 2019 to September 2021, 109 pts with ECOG PS 0-1 enrolled in Part 2A (14) and Part 2B (95). Overall, the median age was 63 years and 83% of pts received up to 3 prior treatment lines for advanced disease (range 1-8). Based on preliminary data, the ORR in Part 2B was 8% (Table). No significant association between clinical response and plasma TGFb level at baseline or modulation upon treatment was observed. The correlation between tumor TGFb level and clinical benefit is inconclusive due to limited number of tumor biopsies. No response was observed in Part 2A. Overall, 100% of pts had at least one treatment emergent adverse event (AE), 67% were G≥3, 34% related G≥3, 17% G5, and 4% related G5. The limited number of patients treated with SAR459 alone at the RP2D did not allow to demonstrate added toxicity due to the combination. Overall, 51 pts (47%) reported hemorrhagic AE of any grade, 8 pts (7%) had G≥3 and 5 pts (5%) had fatal outcome. The rate of bleeding and severe hemorrhagic AE was higher in HCC pts compared to the other cohorts: 11/14 (79%) pts had a hemorrhagic AE, of which 3 (21%) G≥3 and fatal. An exploratory analysis showed a trend for higher frequency of any grade SAR459-related and fatal hemorrhagic AE in patients with higher exposure. Conclusions: The NCT03192345 study was discontinued due to a lack of efficacy, and a high bleeding risk particularly in pts with HCC. Clinical trial information: NCT03192345. [Table: see text]
- Published
- 2022
22. Safety and efficacy of tusamitamab ravtansine (SAR408701) in long-term treated patients with nonsquamous non–small cell lung cancer (NSQ NSCLC) expressing carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5)
- Author
-
Charles Ricordel, Fabrice Barlesi, Sophie Cousin, Byoung Chul Cho, Emiliano Calvo, Tae Min Kim, Carole Helissey, Jin-Soo Kim, Maria Vieito, Valentina Boni, François Ghiringhelli, Mustapha Chadjaa, Nina Masson, Christine Soufflet, and Anas Gazzah
- Subjects
Cancer Research ,Oncology - Abstract
9039 Background: Tusamitamab ravtansine (tusa) is a novel antibody-drug conjugate that selectively targets CEACAM5, a cell surface glycoprotein highly expressed in several tumor types. In previously reported results from an open-label Phase 1/2 study (NCT02187848), tusa showed promising antitumor activity in patients (pts) with heavily pretreated NSQ NSCLC and high CEACAM5 expression (Gazzah A et al. J Clin Oncol. 2020;38[15 suppl]:9505). Herein we report results for pts treated for ≥ 12 mo with NSQ NSCLC and high or moderate CEACAM5 expression. Methods: In the Phase 1/2 study, 92 pts with heavily pretreated NSQ NSCLC and high (n = 64) or moderate (n = 28) CEACAM5 expression (≥ 2+ intensity in ≥ 50% of tumor cells or in ≥ 1% to < 50% of tumor cells, respectively) were treated with tusa 100 mg/m2 Q2W. As of Jan 2020, among CEACAM5 high expressors, 13 had confirmed partial response (PR) and 28 had stable disease (SD); among moderate expressors, 2 had PR and 15 had SD. We focus here on pts treated ≥ 12 mo as of Dec 2021. Results: A total of 24 pts were treated for ≥ 6 mo, 15 pts for ≥ 9 mo, 11 pts for ≥ 12 mo, 6 pts for ≥ 24 mo, and 2 pts for ≥ 42 mo. At the data cutoff, 5 pts remained on treatment, 1 for > 3.5 y. For pts treated ≥ 12 mo median (range) treatment duration was 26.6 (12.1–45.3) mo. Of 15 pts with PR in the prior analysis, as of Dec 2021 PR was still observed in 10 pts (67%) treated for ≥ 6 mo, 8 pts (53%) for ≥ 9 mo, and 7 pts (47%) for ≥ 12 mo. For the 11 pts treated ≥ 12 mo, 7 had PR and 4 had SD. Of the 11 pts treated ≥ 12 mo, 9 had high CEACAM5 expression and 2 had moderate CEACAM5 expression; most had prior treatment with an anti-PD1/PD-L1. Pts treated for ≥ 12 mo had better ECOG performance status and fewer prior treatments than the overall group. Of pts treated for ≥ 12 mo, PR occurred irrespective of CEACAM5 expression level. Only 1 pt treated for ≥ 12 mo discontinued due to a treatment emergent adverse event (TEAE) (breast cancer). Corneal events (keratitis/keratopathy) were the most frequent TEAEs, occurring in 8 pts (73%); 4 pts (36%) with Grade ≥ 3; 7 pts had subsequent treatment modification (delay or delay/reduced dose). No corneal TEAE was serious or led to treatment discontinuation. Conclusions: Almost half (47%) of pts who achieved a PR were treated for ≥ 1 y, suggesting that response to tusa in heavily pretreated pts was durable and frequently sustained. No patient discontinued long-term treatment because of drug-related toxicity. Corneal toxicity in these pts was manageable with dose modification (delay/reduction). The observed long-term clinical benefit and safety profile of tusa support its further clinical development; a Phase 3 study is ongoing to evaluate tusa monotherapy in previously treated pts with high CEACAM5 expressing NSQ NSCLC. Clinical trial information: NCT02187848.
- Published
- 2022
23. 521P Dose escalation study of two different alternative dosing schedules of tusamitamab ravtansine (tusa, SAR408701) in patients (pts) with advanced solid tumors
- Author
-
L. Charbonnier, Y-J. Bang, P. Bedard, C. Soufflet, M-H. Ryu, A. Gazzah, N. Masson, S. Yoruk, J. Tabernero, Maria Vieito, and N. Fagniez
- Subjects
Oncology ,medicine.medical_specialty ,Dosing schedules ,business.industry ,Internal medicine ,Dose escalation ,medicine ,In patient ,Hematology ,business - Published
- 2021
24. Author Correction: The Molecular Tumor Board Portal supports clinical decisions and automated reporting for precision oncology
- Author
-
Maria Vieito, David Tamborero, Adrià Lopez-Fernandez, Jonas Bergh, Peter Horak, and Carlos Caldas
- Subjects
Cancer Research ,Oncology - Published
- 2022
25. Phase I study of CC-90010, a reversible, oral BET inhibitor in patients with advanced solid tumors and relapsed/refractory non-Hodgkin's lymphoma
- Author
-
B. Hanna, I. Aronchik, T. Sánchez-Pérez, Bernard Doger, Tatiana Hernandez-Guerrero, O. Ferrero, R. Sarmiento, Juan Manuel Sepúlveda, J. De Alvaro, Irene Brana, J. Di Martino, Marlene Zuraek, O. Saavedra, Manisha Lamba, Maria Vieito, E. Filvaroff, Victor Moreno, M. Arias, Zariana Nikolova, Institut Català de la Salut, [Moreno V, Hernández-Guerrero T, Doger B] START Madrid-FJD, Hospital Fundación Jimenez Diaz, Madrid, Spain. [Sepulveda JM] Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain. [Vieito M, Saavedra O, Braña I] Department of Gene Expression and Cancer, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Adult ,0301 basic medicine ,medicine.medical_specialty ,Maximum Tolerated Dose ,Anemia ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Antineoplastic Agents ,Neoplasms::Neoplasms by Histologic Type::Lymphoma::Lymphoma, Non-Hodgkin [DISEASES] ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Gastroenterology ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Refractory ,Internal medicine ,Hodgkin, Malaltia de - Tractament ,medicine ,Carcinoma ,Humans ,Adverse effect ,Aged ,Aged, 80 and over ,business.industry ,Lymphoma, Non-Hodgkin ,Hematology ,Middle Aged ,medicine.disease ,Lymphoma ,Non-Hodgkin's lymphoma ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Lymphoma, Large B-Cell, Diffuse ,Neoplasm Recurrence, Local ,business ,neoplasias::neoplasias por tipo histológico::linfoma::linfoma no Hodgkin [ENFERMEDADES] - Abstract
Inhibidor de BET; Limfoma no Hodgkin; Tumors sòlids Inhibidor de BET; Linfoma no Hodgkin; Tumores sólidos BET inhibitor; Non-Hodgkin's lymphoma; Solid tumors Background Bromodomain and extra-terminal (BET) proteins are epigenetic readers that regulate expression of genes involved in oncogenesis. CC-90010 is a novel, oral, reversible, small-molecule BET inhibitor. Patients and methods CC-90010-ST-001 (NCT03220347; 2015-004371-79) is a phase I dose-escalation and expansion study of CC-90010 in patients with advanced or unresectable solid tumors and relapsed/refractory (R/R) non-Hodgkin's lymphoma (NHL). We report results from the dose escalation phase, which explored 11 dose levels and four dosing schedules, two weekly (2 days on/5 days off; 3 days on/4 days off), one biweekly (3 days on/11 days off), and one monthly (4 days on/24 days off). The primary objectives were to determine the safety, maximum tolerated dose (MTD) and/or recommended phase II dose (RP2D) and schedule. Secondary objectives were to evaluate signals of early antitumor activity, pharmacokinetics, and pharmacodynamics. Results This study enrolled 69 patients, 67 with solid tumors and two with diffuse large B-cell lymphoma (DLBCL). The median age was 57 years (range, 21–80) and the median number of prior regimens was four (range, 1–9). Treatment-related adverse events (TRAEs) were mostly mild and manageable; grade 3/4 TRAEs reported in more than two patients were thrombocytopenia (13%), anemia, and fatigue (4% each). Six patients had dose-limiting toxicities. MTDs were 15 mg (2 days on/5 days off), 30 mg (3 days on/11 days off), and 45 mg (4 days on/24 days off). The RP2D and schedule selected for expansion was 45 mg (4 days on/24 days off). As of 8 October 2019, one patient with grade 2 astrocytoma achieved a complete response, one patient with endometrial carcinoma had a partial response, and six patients had prolonged stable disease ≥11 months. Conclusions CC-90010 is well tolerated, with single-agent activity in patients with heavily pretreated, advanced solid tumors. This work was supported by Celgene Corporation, Summit, NJ, USA. (no grant number).
- Published
- 2020
26. Grade I meningioma with disseminated bone disease: a rare clinical phenomenon
- Author
-
A. Valdivia, Maria Vieito, Oriol Mirallas, and D. Marmolejo
- Subjects
Male ,medicine.medical_specialty ,Bone disease ,Antineoplastic Agents, Hormonal ,medicine.medical_treatment ,Octreotide ,Pelvic Pain ,Zoledronic Acid ,Pelvis ,Meningioma ,03 medical and health sciences ,0302 clinical medicine ,Fatal Outcome ,medicine ,Meningeal Neoplasms ,Humans ,Disseminated disease ,Unusual Presentation of More Common Disease/Injury ,Bone Density Conservation Agents ,Radiotherapy ,business.industry ,Pelvic pain ,Neurooncology ,Estrogen Antagonists ,General Medicine ,Middle Aged ,medicine.disease ,Radiation therapy ,Tamoxifen ,030220 oncology & carcinogenesis ,Disease Progression ,Drug Therapy, Combination ,Radiology ,Neurosurgery ,medicine.symptom ,Bone Diseases ,Neoplasm Grading ,business ,Tomography, X-Ray Computed ,030217 neurology & neurosurgery ,Grade I Meningioma - Abstract
Meningioma, the second most common primary tumour of the central nervous system, is classified into three different grades based on their characteristics. Each tumour grade includes different molecular subtype, growth potential, and thus, different prognosis. Grade I meningioma is the most common subtype with a benign course, in which systemic dissemination rarely occurs. We present the case of a 48-year-old male patient with a history of grade I meningioma who was referred 3 years after the initial diagnosis to our centre due to pelvic pain. Computed tomography (CT) images showed new pelvic bone lesions whose histopathological report was compatible with a grade I meningioma. Neither hormonal therapy concomitant with octreotide nor hydroxiurea treatments were effective. Very little is known about this entity’s prevalence and treatment when disseminated disease occurs. Thus, we think it is important to increase the positive and negative clinical experiences in this setting.
- Published
- 2020
27. Impact of HER2 status in resected gastric or gastroesophageal junction adenocarcinoma in a Western population
- Author
-
Jose Ramón Antunez Lopez, Nieves Martinez Lago, Ihab Abdulkader Nallib, Maria Elena Padin Iruegas, Rafael Varela Ponte, Maria Vieito Villar, Rafael López, and Juan José Carrera Alvarez
- Subjects
Cancer Research ,medicine.medical_specialty ,Population ,Gastroesophageal Junction Adenocarcinoma ,Gastroenterology ,Trastuzumab ,HER2 ,Internal medicine ,medicine ,In patient ,skin and connective tissue diseases ,education ,neoplasms ,education.field_of_study ,Her2 expression ,business.industry ,Research ,gastric cancer ,DuoCISH ,medicine.disease ,Confidence interval ,Oncology ,immunohistochemistry ,Adenocarcinoma ,Immunohistochemistry ,prognosis ,business ,medicine.drug - Abstract
Background HER2 status is a predictive biomarker of response to trastuzumab in advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma. However, there is relatively little known about the role of HER2 in resected gastric or GEJ adenocarcinoma in the Western population. Methods Retrospective, observational, single centre study of patients with gastric or GEJ adenocarcinoma undergoing surgery with curative intent between January 2007 and June 2014 in the University Hospital Complex of Santiago de Compostela. The expression of HER2 was determined by immunohistochemistry (IHC) using DAKO-HercepTest™ and gene amplification with DuoCISH using a DAKO-DuoCISH kit. The study of HER2 expression and amplification was carried out in all the patients and it was correlated with classic clinicopathological parameters, survival and recurrence pattern. Results 106 patients were included. HER2 expression was as follows: 71.7% HER2 negative, 21.7% HER2 equivocal and 6.6% HER2 positive, or with HER2 overexpression. 13.2% of patients (14/106) had HER2 amplification by DuoCISH. A significant association was seen between overexpression and amplification of HER2 (p < 0.001). HER2 positivity was associated with the intestinal subtype (p = 0.010) and a low grade of differentiation (p = 0.018). Likewise, HER2 was significantly associated with a worse prognosis: overall survival (OS) 32.3 months HER2 positive versus 93.9 months HER2 negative (HR 0.42; confidence interval 95% 0.18–0.93; p = 0.028); and the presence of distant metastasis without accompanying locoregional recurrence (p = 0.048). Conclusion HER2 status defines a subgroup with differentiated clinicopathological characteristics, worse prognosis and distant dissemination, without accompanying locoregional recurrence, in patients with resected gastric or GEJ adenocarcinoma operated on in a Western population.
- Published
- 2020
28. P14.58 Efficacy and safety of lomustine versus fotemustine as first and second line treament in relapsed glioblastoma patients
- Author
-
C Fabregat, Maria Vieito, S. Del Barco, Ainhoa Hernandez, C Panciroli, Noelia Vilarino, C. Balana, D Garcia-Illescas, Montserrat Domenech, and Carlos Mesia
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,O-6-methylguanine-DNA methyltransferase ,Lomustine ,medicine.disease ,Second line ,Internal medicine ,medicine ,Adjuvant therapy ,Fotemustine ,Neurology (clinical) ,Progression-free survival ,business ,Adverse effect ,Glioblastoma ,medicine.drug - Abstract
BACKGROUND Glioblastoma (GB) is the most aggressive primary brain tumour. Despite the survival benefit associated with adjuvant therapy, most of patients (pts) relapse after initial therapy. Nitrosoureas (NU) are the standard treatment at relapse in Europe. Both fotemustine (FM) (Addeo schema) and lomustine (LM) (administered orally every 6 weeks) are used in this context. MATERIAL AND METHODS This retrospective cohort study included pts diagnosed with GB treated with NU at relapse in four Catalonia hospitals from 2010 to 2020. Clinical and pathological data were collected from medical records. We analysed 6months-progression-free survival (6m-PFS), progression-free survival (PFS) and overall survival (OS) from the start of NU to progression or death respectively. Differences in toxicity grade using CTCAE v5.0 were analysed globally as ‘non-toxicity’, ‘mild toxicity (grade 1 or 2)’ and ‘high toxicity (grade 3 or 4)’. RESULTS We identified 236 GB pts with a median age of 58 years old. 29% of the pts presented MGMT promotor methylation and only 3%(n=7) had IDH mutation. After a median follow-up of 20 months, 94% of the pts were dead at the time of the analyses. At first line, 83 pts were treated with FM and 18 with LM. Pts treated with FM had better performance status (PS) than those treated with LM (p=.010). Median PFS was 2 months and 6m-PFS was 12% vs 6% in FM and LM group respectively (p=.87). Median OS was 3 months with LM vs 6 months with FM, with no statistically significant differences even adjusted for prognostic factors (p=.79 HR:0.9 CI 95% 0.41–1.96).At second line, 78 were treated with FM and 24 with LM, no differences between groups. Median PFS was 2 months in both groups and median OS was 3 vs 5 months for pts treated with LM vs FM respectively, with no significant differences. 6m-PFS was 13% for LM vs 0% for the FM group (p=.39).Pts received a mean of 1.7 cycles (every 6 weeks) and 4.1 cycles (every 2 weeks) in LM and FM group, respectively. Thrombocytopenia was the most common serious side-effect, with a higher proportion of grade 1–2 toxicity in the FM group (p=.03) that also required more treatment delays (p=.01). CONCLUSION Despite being retrospective study and a few pts were treated with LM, there were no differences neither in PFS nor in OS in pts treated with LM vs FM at first or second line. Higher G1-2 thrombocytopenia was shown in the FM group probably due to a higher number of hematology samples collected.
- Published
- 2021
29. Abstract CT033: Inducible T cell costimulatory (ICOS) receptor agonist, feladilimab (FE), alone and in combination (combo) with pembrolizumab (PE): Results from INDUCE-1 relapsed/refractory (R/R) melanoma expansion cohorts (EC)
- Author
-
Maria Vieito Villar, Courtney Henry, Sapna Yadavilli, Victor Moreno, Antoine Italiano, Omid Hamid, Marc S. Ballas, Marco Balas, Michael Chisamore, Frans L. Opdam, Michele Maio, Xiao Ji, Jeffrey S. Weber, Axel Hoos, David C. Turner, Jose Manuel Trigo, and Catherine E. Ellis
- Subjects
Agonist ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.drug_class ,Melanoma ,medicine.medical_treatment ,T cell ,Cancer ,Pembrolizumab ,Immunotherapy ,medicine.disease ,Gastroenterology ,Immune checkpoint ,medicine.anatomical_structure ,Oncology ,Refractory ,Internal medicine ,medicine ,business - Abstract
Background: INDUCE-1 is a first-in-human trial investigating FE, an IgG4 ICOS agonist non-T-cell depleting mAb, in monotherapy (mono) and combo with PD-1 inhibitor PE. ICOS is highly expressed in melanoma, an immunotherapy responsive tumor, and is a biomarker of response to anti-CTLA-4 treatment; in nonclinical models, ICOS agonism has enhanced activity in combo with immune checkpoint blockade (ICB). Preliminary data from INDUCE-1 R/R melanoma ECs are presented to support FE expansion in ICB experienced (exp) R/R melanoma patients (pts). Methods: Eligible pts had non-uveal R/R melanoma, ≤5 prior lines of systemic therapy, achieved response or stable disease (SD) on prior anti-PD-1/L1 treatment, and no prior immunotherapy-related Grade ≥3 toxicities leading to treatment discontinuation. In FE mono and FE + PE 200 mg combo ECs, pts were randomized to FE 0.3 or 1 mg/kg; treatment was given every 3 wks up to 35 cycles until disease progression or unacceptable toxicity. Safety and efficacy were assessed. Biomarkers in tumor biopsies were analyzed. Results: By 30 Mar 2020, 39 pts in the mono EC were evaluable (21 pts at 0.3; 18 pts at 1 mg/kg FE); all pts were anti-PD-1/L1 exp; 21/39 (54%) were anti-CTLA-4 exp; 7/39 (18%) had ≥4 prior lines of therapy. Of the 39 pts, 29 (74%) discontinued treatment due to progression. Response rate was 10% (4/39; 1 complete response, 3 partial responses); 8/39 (21%) pts had SD and 2/8 (25%) pts with SD had tumor size reductions (RECIST v1.1) at ≥19%; 5 pts (13%) had SD at ≥18 wks. Duration of response (DoR) was 2.0+ to 6.3 mo. BRAF and NRAS status did not impact clinical activity. Anti-CTLA-4 exp pts had a numerically higher response rate than anti-CTLA-4 naïve pts. In the combo EC, 17 anti-PD-1/L1 exp pts were evaluable (6 pts at 0.3; 11 pts at 1 mg/kg FE); 12/17 (71%) were anti-CTLA-4 exp; 15/17 (88%) discontinued treatment, all due to progression. Response rate was 18% (3/17), DoR was 7.9+ to 8.2 mo, and disease control rate (response or SD) was 53% (9/17); 1 pt with SD had tumor size reduction of 10%. Ongoing analyses with mono EC showed a 28.6% (2/7) response rate, DoR of 2.0+ to 6.3 mo, and mOS of ~26 mo in pts with ICOS high tumor-infiltrating T cells. Sample size limits comparison; however, clinical activity was similar between FE doses and between BRAF+ and BRAF− pts. Adverse events (AEs) were consistent with AEs reported for PE. Conclusions: FE is the first ICOS agonist with reported single-agent activity in ICB exp R/R melanoma, supporting ICOS as a target. FE + PE in combo shows promising clinical activity and manageable safety in R/R melanoma. Continued survival follow-up of ECs is warranted. Updated efficacy and PK/PD data to be presented. Funding: Study 204691 (NCT02723955) funded by GSK in collaboration with Merck Sharp & Dohme Corp, a subsidiary of Merck & Co, Inc, Kenilworth, NJ, USA. Citation Format: Michele Maio, Jeffrey S. Weber, Maria Vieito Villar, Frans L. Opdam, Victor Moreno, Omid Hamid, José Trigo, Michael Chisamore, Marco Balas, Sapna Yadavilli, David C. Turner, Courtney Henry, Xiao Ji, Catherine Ellis, Marc Ballas, Axel Hoos, Antoine Italiano. Inducible T cell costimulatory (ICOS) receptor agonist, feladilimab (FE), alone and in combination (combo) with pembrolizumab (PE): Results from INDUCE-1 relapsed/refractory (R/R) melanoma expansion cohorts (EC) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr CT033.
- Published
- 2021
30. Improving circulating tumor cells enumeration and characterization to predict outcome in first line chemotherapy mCRPC patients
- Author
-
Luis León-Mateos, Antonio Gómez-Tato, Laura Muinelo-Romay, Urbano Anido, Rafael López, Helena Casas, Manuel Abreu, Maria Vieito, Alicia Abalo, and Miguel Abal
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Pathology ,Population ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Circulating tumor cell ,Internal medicine ,medicine ,Progression-free survival ,Liquid biopsy ,education ,education.field_of_study ,business.industry ,prostate cancer ,medicine.disease ,taxanes resistance ,030104 developmental biology ,Docetaxel ,circulating tumour cells (CTCs) ,Cabazitaxel ,030220 oncology & carcinogenesis ,Personalized medicine ,business ,prognostic markers ,Research Paper ,medicine.drug - Abstract
// Luis Leon-Mateos 1, * , Helena Casas 2, * , Alicia Abalo 2, 3 , Maria Vieito 6 , Manuel Abreu 2, 3 , Urbano Anido 3 , Antonio Gomez-Tato 4 , Rafael Lopez 2, 3, 5 , Miguel Abal 3 and Laura Muinelo-Romay 3 1 Axencia Galega de Conecemento en Saude (ACIS), SERGAS, Santiago de Compostela, Spain 2 Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), CIBERONC, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain 3 Translational Medical Oncology Group, Health Research Institute of Santiago (IDIS), CIBERONC, Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain 4 School of Mathematics, University of Santiago de Compostela (Campus Vida), Santiago de Compostela, Spain 5 Roche-Chus Joint Unit for Precision Oncology, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), Santiago de Compostela, Spain 6 Research Unit for Molecular Therapy of Cancer, CNS Tumors, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain * Co first-authors Correspondence to: Luis Leon-Mateos, email: Luis.Leon.Mateos@sergas.es Laura Muinelo-Romay, email: lmuirom@gmail.com Keywords: prostate cancer, circulating tumour cells (CTCs), prognostic markers, taxanes resistance Received: January 17, 2017 Accepted: May 02, 2017 Published: May 19, 2017 ABSTRACT Introduction: There is a critical need of new surrogate markers for improving the therapeutic selection and monitoring of metastatic prostate cancer patients. Nowadays clinical management of these patients is been driven by biochemical and clinical parameters without enough accuracy to allow a real personalized medicine. The present study was conducted to go insight the molecular profile of circulating tumor cells (CTCs) isolated from advanced metastatic castration-resistant prostate cancer (mCRPC) with the aim of identifying prognostic marker with potential utility for therapy selection and monitoring. Materials and Methods: CTCs isolation was carried out in peripheral blood samples from 29 mCRPC patients that undergo systemic chemotherapy based on taxanes (docetaxel/cabazitaxel) and 19 healthy controls using in parallel CellSearch and an alternative EpCAM-based immunoisolation followed by RT-qPCR analysis to characterize the CTC population. A panel of 17 genes related with prostate biology, hormone regulation, stem properties, tumor aggressiveness and taxanes responsiveness was analysed to identify an expression signature characterizing the CTCs. Results: Patients with ≥ 5 CTCs/7.5ml of peripheral blood at baseline and during the treatment showed lower progression free survival (PFS) and overall survival (OS). Changes of CTCs levels during the treatment were also associated with the patient’s outcome. These results confirmed previous data obtained using CellSearch in mCRPC. In addition, we found a CTC profile mainly characterized by the expression of relevant genes for the hormone dependent regulation of PCa such as AR and CYP19 together with genes strongly implicated in PCa progression and resistance development such as BIRC5 , TUB1A , GDF15 , RAB7 and SPINK1 . Our gene-expression profiling also permitted the identification of valuable prognostic biomarkers. Thus, high levels of AR, CYP19 and GDF15 were associated with poor PFS rates while AR, GDF15 and BIRC5 were also found as reliable predictors of OS. Besides, a logistic model using KLK3 and BIRC5 showed a high specificity and sensitivity compared to CellSearch to discriminate patients with a more aggressive evolution. Conclusions: The molecular characterization of CTCs from advanced mCRPC patients provided with a panel of specific biomarkers, including genes related to taxanes resistance, with a promising applicability as “liquid biopsy” for the management of these patients.
- Published
- 2017
31. 527O CC-90010, a reversible, oral bromodomain and extra-terminal (BET) inhibitor in patients (Pts) with advanced solid tumours (STs) and relapsed/refractory (R/R) non-Hodgkin lymphoma: Updated results of a phase I study
- Author
-
Maria Vieito, Carmelo Carlo-Stella, Antoine Italiano, G. Musuraca, B. Hanna, Virtudes Moreno, Bernard Doger, J.M. Sepulveda Sanchez, Irene Brana, E. Filvaroff, Tatiana Hernandez-Guerrero, I. Aronchik, J-M. Michot, Zariana Nikolova, T. Sánchez-Pérez, Barbara Amoroso, S. Mora, O. Saavedra, Ana Rita Martins Pinto, and R. Sarmiento
- Subjects
BET inhibitor ,Oncology ,business.industry ,Relapsed refractory ,Cancer research ,Medicine ,Hodgkin lymphoma ,In patient ,Hematology ,business ,Phase i study ,Bromodomain - Published
- 2020
32. 573P Efficacy of immunotherapy (IT) after prior immune checkpoint inhibitors (ICIs) exposure
- Author
-
Rodrigo Dienstmann, M. Diez Garcia, Ignacio Matos, Irene Brana, Helena Verdaguer, O. Saavedra Santa Gadea, M. González, C. Ortiz Velez, Elena Garralda, I. Baraibar Argota, Vladimir Galvao, R. Berché, A. Hernando-Calvo, A. Valdivia, Maria Vieito, E. Muñoz Couselo, A. Callejo, G. Alonso Casal, J. Tabernero, and A.B. Azaro Pedrazzoli
- Subjects
Oncology ,business.industry ,medicine.medical_treatment ,Immune checkpoint inhibitors ,Cancer research ,medicine ,Hematology ,Immunotherapy ,business - Published
- 2020
33. 918MO Molecular enrichment and outcomes based on ESCAT levels in metastatic salivary gland tumours (mSGT) patients (pts) treated in early clinical trials
- Author
-
Margarita Alberola, A.B. Azaro Pedrazzoli, A. Hernando-Calvo, Maria Vieito, A. De Pablo, Sergi Benavente, Jordi Temprana-Salvador, Elena Garralda, Rodrigo Dienstmann, Jordi Giralt, Enriqueta Felip, Irene Brana, Vladimir Galvao, Susana Aguilar, R. Berché, Coro Bescos, G. Alonso Casal, Juan Lorente, M.A. Rezqallah Aron, and O. Saavedra Santa Gadea
- Subjects
Clinical trial ,Oncology ,medicine.medical_specialty ,medicine.anatomical_structure ,Salivary gland ,business.industry ,Internal medicine ,Medicine ,Hematology ,business - Published
- 2020
34. LIF regulates CXCL9 in tumor-associated macrophages and prevents CD8+ T cell tumor-infiltration impairing anti-PD1 therapy
- Author
-
Carolina Marques, Juan Sahuquillo, Elena Garralda, Ester Bonfill-Teixidor, Isabel Cuartas, Josep Tabernero, Estela Pineda, Irene Brana, Maria Vieito, Francisco Martínez-Ricarte, Francesc Graus, Laura Escudero, Carlota Rubio-Perez, Mónica Pascual-García, Raffaella Iurlaro, Massimo Squatrito, Joan Seoane, Ester Planas-Rigol, Alexandra Arias, Paolo Nuciforo, Ada Sala-Hojman, Carmen Espejo, Leire Pedrosa, Isabel Huber-Ruano, European Research Council, Asociación Española Contra el Cáncer, Instituto de Salud Carlos III, FERO Foundation, Fundación La Caixa, Fundación BBVA, Cellex Foundation, Institut Català de la Salut, [Pascual-García M, Bonfill-Teixidor E] Vall d'Hebron Institut d'Oncologia, Barcelona, Spain. Hospital Universitari Vall d'Hebron, Barcelona, Spain. CIBERONC, Madrid, Spain. [Planas-Rigol E, Rubio-Perez C, Iurlaro R, Arias A, Cuartas I, Sala-Hojman A, Escudero L, Huber-Ruano I, Nuciforo P, Braña I, Garralda E, Vieito M] Vall d'Hebron Institut d'Oncologia, Barcelona, Spain. Hospital Universitari Vall d'Hebron, Barcelona, Spain. [Martínez-Ricarte F, Espejo C, Sahuquillo J] Vall d’Hebron Institut de Recerca, Barcelona, Spain. Hospital Universitari Vall d'Hebron, Barcelona, Spain. Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain. [Tabernero J] Vall d'Hebron Institut d'Oncologia, Barcelona, Spain. Hospital Universitari Vall d'Hebron, Barcelona, Spain. CIBERONC, Madrid, Spain. Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain. [Seoane J] Vall d'Hebron Institut d'Oncologia, Barcelona, Spain. Hospital Universitari Vall d'Hebron, Barcelona, Spain. CIBERONC, Madrid, Spain. Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain. Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain., Hospital Universitari Vall d'Hebron, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Amino Acids, Peptides, and Proteins::Peptides::Intercellular Signaling Peptides and Proteins::Cytokines::Leukemia Inhibitory Factor [CHEMICALS AND DRUGS] ,General Physics and Astronomy ,02 engineering and technology ,Mice, SCID ,CD8-Positive T-Lymphocytes ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Chemokine CXCL9 ,Leukemia Inhibitory Factor ,Medicaments antineoplàstics ,Epigenesis, Genetic ,Neoplasms ,Tumor Microenvironment ,Cytotoxic T cell ,Otros calificadores::Otros calificadores::/inmunología [Otros calificadores] ,lcsh:Science ,reproductive and urinary physiology ,Chemokine CCL2 ,Cancer ,Otros calificadores::Otros calificadores::/tratamiento farmacológico [Otros calificadores] ,Multidisciplinary ,Chemistry ,021001 nanoscience & nanotechnology ,Citocines - Immunologia ,3. Good health ,medicine.anatomical_structure ,embryonic structures ,CXCL9 ,Neoplasias [ENFERMEDADES] ,0210 nano-technology ,Infiltration (medical) ,Cancer microenvironment ,endocrine system ,Cells::Blood Cells::Leukocytes::Leukocytes, Mononuclear::Lymphocytes::T-Lymphocytes::CD8-Positive T-Lymphocytes [ANATOMY] ,Science ,T cell ,CCL2 ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,Other subheadings::Other subheadings::/immunology [Other subheadings] ,medicine ,Animals ,Humans ,células::células sanguíneas::leucocitos::leucocitos mononucleares::linfocitos::linfocitos T::linfocitos T CD8-positivos [ANATOMÍA] ,Cèl·lules T - Immunologia ,Macrophages ,Tumors - Tractament ,General Chemistry ,Immunotherapy ,medicine.disease ,Antibodies, Neutralizing ,Immune checkpoint ,Neoplasms [DISEASES] ,Mice, Inbred C57BL ,030104 developmental biology ,Cancer research ,lcsh:Q ,aminoácidos, péptidos y proteínas::péptidos::péptidos y proteínas de señalización intercelular::citocinas::factor inhibidor de la leucemia [COMPUESTOS QUÍMICOS Y DROGAS] ,Immunologic Memory ,CD8 ,Neoplasm Transplantation - Abstract
Cancer response to immunotherapy depends on the infiltration of CD8+ T cells and the presence of tumor-associated macrophages within tumors. Still, little is known about the determinants of these factors. We show that LIF assumes a crucial role in the regulation of CD8+ T cell tumor infiltration, while promoting the presence of protumoral tumor-associated macrophages. We observe that the blockade of LIF in tumors expressing high levels of LIF decreases CD206, CD163 and CCL2 and induces CXCL9 expression in tumor-associated macrophages. The blockade of LIF releases the epigenetic silencing of CXCL9 triggering CD8+ T cell tumor infiltration. The combination of LIF neutralizing antibodies with the inhibition of the PD1 immune checkpoint promotes tumor regression, immunological memory and an increase in overall survival., LIF is a pleiotropic cytokine that promotes an immunosuppressive microenvironment and has critical functions in embryonic development. Here, the authors show that LIF regulates CD8+ T cell tumor infiltration in cancer by repressing CXCL19 and promoting the presence of protumoral macrophages and thatLIF inhibition, via neutralizing antibodies, promotes T cell infiltration and synergizes with immune checkpoint inhbitors resulting in tumor regression and immunological memory.
- Published
- 2019
35. Molecular profiling and targeted agents in recurrent, metastatic salivary gland tumor (R/M SGT) patients (pts) treated at two academic centers
- Author
-
Eoghan Ruadh Malone, Aaron R. Hansen, Omar Saavedra Santa Gadea, Enriqueta Felip, Lillian L. Siu, Angela Rodriguez, Maria Vieito, Coro Bescos, Ilan Weinreb, Katherine Lajkosz, Anneli Eliason, Alejandra Rezqallah, Susana Aguilar, Alberto Hernando-Calvo, J.D. Assaf, Sarah Jennings, Juan Lorente, Irene Brana, Anna Spreafico, and Elena Garralda
- Subjects
Oncology ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,medicine.medical_specialty ,Salivary gland tumor ,business.industry ,Internal medicine ,Cancer centre ,Medicine ,business ,Selection (genetic algorithm) - Abstract
6081 Background: Treatment selection based on actionable alterations (AAs) is an appealing strategy for pts with R/M SGT. The GEMS-001 study (NCT02069730) at Princess Margaret Cancer Centre (PM) and the Vall D´Hebron Institute of Oncology (VHIO) pre-screening program facilitate the identification of AAs for R/M SGT pts and treatment selection. Methods: We analyzed R/M SGT treated at PM and VHIO from 2015 to 2020. Clinicopathological features, molecular alterations and treatment modalities were correlated with outcomes. The primary endpoint was overall response rate (ORR) by RECIST 1.1. Clinical benefit rate (CBR) was defined by pts with partial response or stable disease ≥4 months. Clinical actionability of multigene panel testing (NGS) and immunohistochemistry (IHC) were assessed as per institutional molecular tumor boards or investigators. Pts were opportunistically matched to available therapies from each center. Results: In total 206 pts were enrolled. On IHC, HER2 overexpression was present in 9%, Androgen Receptor (AR) 33%, Estrogen/Progesterone Receptor (ER/PR) 11% and ALK overexpression 0%. On NGS, PIK3CA mutation (mut) was in 9%, NTRK fusion 6%, NOTCH1-3 mut 5%, HRAS mut 6%, ERBB2/3 alterations (alt) 4% and FGFR1-4 alt 3%. Up to 92 pts (45%) displayed at least 1 AA and 36 pts (18%) had ≥2 AAs. A total of 60 pts (29%) were matched to AAs. Of those matched, median age was 60 years (range 33-84), M:F 21:39, 95% ECOG≤1 with a median number of prior treatment lines 0 (range 0-3), and their AAs included 26 AR, 9 HER2 or ERBB2 overexpression, 9 PIK3CA mut, 3 NTRK fusion, 3 FGFR1-3 alt and 10 other AAs (2 ER/PR overexpression, 2 EGFR mut, 1 c-kit mut, 1 BAP1 mut, 1 Non-V600 BRAF mut, 1 CDKN2A mut, 1 CHEK2 mut and 1 PTCH1 mut). Overall, ORR was 27% for the matched population. See table for outcomes. Conclusions: In our cohort, almost one third of the population received therapies matched to AAs. Our results suggest that targeted therapies have promising activity in pts with R/M SGT supporting comprehensive molecular and IHC profiling in treatment determination.[Table: see text]
- Published
- 2021
36. Inducible T-cell co-stimulatory (ICOS) receptor agonist, feladilimab (fela), alone and in combination (combo) with pembrolizumab (P): Results from INDUCE-1 urothelial carcinoma (UC) expansion cohorts (ECs)
- Author
-
Catherine E. Ellis, Michael Chisamore, Eric Angevin, Marc S. Ballas, Erminia Massarelli, Debra Rogan, Victor Moreno, Hui K Gan, Axel Hoos, Maria Vieito, Antoine Italiano, Riccardo Danielli, Arjun Vasant Balar, Courtney Henry, Frans L. Opdam, Xiao Ji, and Francesco Ricci
- Subjects
Agonist ,Cancer Research ,medicine.drug_class ,business.industry ,T cell ,Pembrolizumab ,Monoclonal antibody ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Receptor ,business ,Urothelial carcinoma - Abstract
4519 Background: INDUCE-1 is a first-in-human trial evaluating fela, an IgG4 ICOS agonist non-T-cell depleting mAb, as monotherapy (mono) and in combo with P. ECs include tumor types, such as UC, with high ICOS expression and immunotherapy-favorable features. Fela induced IFNγ, increased PD-1/L1 expression, and enhanced antitumor activity in combo with PD-1 blockade nonclinically. We report preliminary efficacy, safety, and biomarker data of fela ± P in INDUCE-1 UC ECs. Methods: Eligible patients (pts) had recurrent/metastatic (R/M) UC of the upper or lower urinary tract, ≤6 prior systemic therapy lines in the advanced setting, measurable disease, and no active autoimmune disease. Pts received 0.3 or 1 mg/kg fela (mono EC; anti-PD-1/L1–experienced [exp] pts) or 0.3 mg/kg fela + 200 mg P (combo EC; anti-PD-1/L1–naïve pts) every 3 wks, up to 35 cycles until disease progression or unacceptable toxicity. Disease was assessed every 9 wks through wk 54, then every 12 wks. Archival and/or fresh biopsy tumor tissue was collected for biomarker analyses and safety assessed. Results: By Nov 6 2020, 13 anti-PD-1/L1–exp and 32 anti-PD-1/L1–naïve pts were evaluable in the mono and combo ECs, respectively. In the mono EC, median age was 69 yrs (range: 47–82), 92% of pts were male, and 85% received ≥2 prior therapy lines in the metastatic setting. In the combo EC, median age was 70 yrs (range: 42–84), 75% of pts were male, and 72% received ≥1 prior therapy line in the metastatic setting. In the mono EC, median duration of follow-up (mDoF) was 10.6 mo (range: 1.1–22.8); overall response rate (ORR) was 8% (1 partial response [PR]; 95% CI: 0.2, 36.0) with a duration of response (DoR) of 6.1 mo; disease control rate (DCR [response or stable disease for ≥9 wks]) was 23% (95% CI: 5.0, 53.8), and median overall survival (mOS) was 14.5 mo (95% CI: 2.8, NR), with 74% of pts alive at 6 mo. In the combo EC, mDoF was 9.6 mo (range: 0.9–28.3); ORR was 22% (7 PRs; 95% CI: 9.3, 40.0) with a median DoR of 8.3 months (range: 3.5–23.3+); DCR was 63% (95% CI: 43.7, 78.9), and mOS was 10.7 mo (95% CI: 5.2, 18.1), with 64% of pts alive at 6 mo. Grade ≥3 treatment-related AEs were reported for 0% and 9% of pts in the mono (N = 16) and combo (N = 44) safety populations, respectively. PD-L1 expression and ICOS-specific biomarkers are being evaluated, with promising trends observed in enrichment of clinical activity in preliminary analyses. Conclusions: Fela is the first ICOS agonist with reported single-agent activity in anti-PD-1/L1–exp relapsed/refractory UC. Fela + P in combo shows promising clinical activity and manageable safety in PD-1/L1–naïve R/M UC. Further study is warranted. Updated data to be presented. Funding: Study 204691 (NCT02723955) funded by GlaxoSmithKline in collaboration with Merck Sharp & Dohme Corp, a subsidiary of Merck & Co, Inc, Kenilworth, NJ, USA. Clinical trial information: NCT02723955.
- Published
- 2021
37. Evaluating the role of immune-checkpoint inhibitor (ICI) combinations in patients (pts) with unselected 'cold' tumors enrolled in early clinical trials (CT)
- Author
-
Teresa Macarulla, Ignacio Matos, Omar Saavedra Santa Gadea, Guzman Alonso, Josep Tabernero, Honey Kumar Oberoi, Elena Elez, Jaume Capdevila, Irene Brana, R. Berché, Rodrigo Dienstmann, Carmen Sandoval García, Analia Azaro, Alberto Hernando-Calvo, Javier Ros, Maria Vieito, Vladimir Galvao, Elena Garralda, Natassia Ann Wornham, and Juan Francisco Grau Bejar
- Subjects
Oncology ,Response rate (survey) ,Clinical trial ,Cancer Research ,medicine.medical_specialty ,business.industry ,Internal medicine ,Immune checkpoint inhibitors ,medicine ,In patient ,business - Abstract
2597 Background: In order to improve the expected response rate (ORR) of less than 10% in cold tumors, several ICI combinations are being evaluated in clinical trials. However, most of these trials don’t require any biomarker and pts are included based solely in histology. We aimed to assess the benefit of ICI combinations in pts with unselected cold tumors included in early CT. Methods: ICI naïve pts with cold tumors treated from 2015 to 2021 with ICI combinations in early CT at VHIO were reviewed. Clinico-pathological data and anti-tumor activity were extracted from a prospective database. ORR was defined as per RECIST v1.1 and clinical benefit rate (CBR) as complete/partial response (CR/PR) + stable disease (SD) for ≥ 4 months (m). Kaplan Meier estimates of progression-free survival (PFS) and overall survival (OS) were calculated and a Cox model according to LIPI (Lung Immune Prognostic Index = baseline LDH and derived neutrophil to lymphocyte ratio) was constructed. Immune-related adverse events (irAE) were classified as per CTCAE v.4.03. Hyperprogressive disease (HPD) was evaluated using RECIST v1.1 (Matos et al, 2020). Results: Out of 97 pts, median age was 62y, 61% had ECOG 0 and 29.8% had LIPI 0 (good prognostic score). Most pts had microsatellite stable (MSS) colorectal cancer (60.8%) or ovarian cancer (14.4%). Regimens included anti-PD1/L1 + another ICI in 69% (most commonly anti-LAG3 [26,8%] and CD40 agonist [20.9%]), anti-PD1/L1 + other molecule in 21.7% (most commonly SHP2 inhibitor [33.3%] and anti p53-HDM2 [28.5%]) and bispecific antibodies in 9.3% (anti-PD1/L1 + anti-LAG3 or CD137 agonist). No patient achieved a response. CBR was 15.3% (11 pts with MSS colorectal cancer, 2 ovarian cancer, 1 olfactory neuroblastoma, 1 paraganglioma). 33 pts (34%) presented irAE, 15 pts (15.5%) had irAE ≥ G2, 4 pts (4.1%) had G3 irAE (dry mouth, hypertransaminasemia, myocarditis and neutrophils count decreased) and 1 patient (1%) had G4 hyperglicemia. 58 pts (59.7%) had progressive disease (PD) as best response, 19 of these pts (32.7%) presented irAE. Overall, 20 pts (20.6%) met definition of HPD, representing 34.4% of pts with PD as best response. Median PFS for overall and CBR population were 1.9 m (CI95% 1.7-2.0) and 5.9 m (5.4-NR), respectively. Median OS for overall population was 7.6 m (5.9-9.5), with a trend for improved OS if LIPI good score vs. others (12.6 m vs. 6.2 m, hazard ratio 1.9, (CI 95% 1.1-3.3), p = 0.02). Among hyperprogressors, median OS was 5.33 m (3.39 - NR) and significantly worse LIPI scores (intermediate [1] or poor [2]) were observed as compared to pts with CBR (75% vs 53.3% p = 0.001). Conclusions: ICI combinations demonstrated very limited activity in pts with unselected cold tumors. However, the risk for irAE and HPD remain substantial. Further drug-biomarker co-development strategies are urgently needed to increase the risk benefit ratio for these pts.
- Published
- 2021
38. ItRECIST adapted efficacy assessment in solid tumors treated with intratumoral immunotherapy
- Author
-
A. Hernando-Calvo, Maria Vieito, Elena Elez, Xavier Serres-Creixams, Gemma Mur-Bonet, Vladimir Galvao, Teresa Macarulla, Josep Tabernero, Mafalda Oliveira, Omar Saavedra Santa Gadea, Guillem Cunill-Macià, Guzman Alonso, Honey Kumar Oberoi, Enriqueta Felip, Iosune Baraibar, Eva Muñoz-Couselo, Ignacio Matos, Marc Diez, Elena Garralda, and Irene Brana
- Subjects
Cancer Research ,Oncology ,business.industry ,Intratumoral Therapy ,Immune checkpoint inhibitors ,medicine.medical_treatment ,medicine ,Cancer research ,Immunotherapy ,business - Abstract
2557 Background: The development of human intratumoral therapy (HIT-IT) has surged as a promising strategy to overcome resistance to checkpoint inhibitors (CPI), promoting a stronger tumor-specific immune response while reducing systemic exposure. A broad variety of agents (i.e: oncolytic viruses, toll-like receptors agonists) administered both in superficial- and deep-seated lesions are being currently tested in clinical trials (CT). Due to the local intervention on tumors, radiological assessment by standard RECIST is challenging and new methods of response that capture and integrate the local and systemic response to HIT-IT are needed. We aimed to evaluate the feasibility and clinical utility of itRECIST (Goldmacher et al., 2020) in patients (pts) treated with HIT-IT in early phase CT. Methods: Retrospective analysis of a cohort of pts with different solid tumor types enrolled in CT including HIT-IT in our institution between August’18 and January’21. Clinical characteristics were collected. Efficacy in target-injected (T-I) and target-non-injected (T-NI) lesions was assessed by objective response rate (ORR) and disease control rate (DCR), as per itRECIST. Overall disease ORR and DCR were assessed per RECIST 1.1/iRECIST. Treatment-related adverse events (TRAEs) were assessed with CTCAE v.5.0. ORR was calculated with Clopper-Pearson method. Survival analysis was made using Kaplan-Meier method. Results: A total of 37 pts were included. Median age was 66 years, 19 pts (51%) were male, all pts had ECOG 0-1. 24 pts (65%) were CPI-naïve. Median previous lines of therapy was 2 (range [r]: 0-11). All pts (100%) received minimum 1 dose of HIT-IT. 6 pts (16%) were treated with monotherapy and 31 pts (84%) in combination with CPI. Median HIT-IT and CPI doses administered were 4 (r: 1-9) and 2 (r: 1-13), respectively. Injected lesions: cutaneous (16.2%), subcutaneous (21.6%), lymph node (32.4%), liver (29.7%). Median size of T-I lesions was 40 mm (r: 19-260). At data cutoff, 32 pts were evaluable. Median follow-up was 14.4 weeks (r: 1.0-81.1). Per RECIST 1.1, overall ORR was 6% (95% CI, 5-7) and DCR was 38% (95% CI, 21-56). Per itRECIST, ORR was 19% (95% CI, 7-36) and DCR was 63% (95% CI, 44-79) in T-I lesions (n = 32), and 10% (95% CI, 22-27) and 48% (95% CI, 29-67) in T-NI lesions (n = 29). Mean decrease in responding T-I and T-NI lesions was -47% (r: -21 to -100) and -41% (r: -26 to -59), respectively. No non-target (NT) lesion was injected. Median progression-free survival was 7.4 weeks (95% CI, 6.6 – 8.2). Median overall survival was 10.0 months (95% CI, 2.3 – 17.7). Incidence of TRAE was 58% (grade 1-2 IT-related pyrexia 43%; grade 3-4, 5%). No treatment-related deaths were recorded. Conclusions: ItRECIST is feasible to implement and adds precision to the radiological assessment of local and distant anti-tumor activity of HIT-IT. No safety issues were detected in our cohort.
- Published
- 2021
39. First-in-human study of PM14 in patients with advanced solid tumors
- Author
-
Eva Banus, Leyre Agreda, Lourdes Llanero, Antonio Nieto, Rubin Lubomirov, Gema Corral, Martin Cullell-Young, Rastilav Bahleda, Christophe Massard, Maria Vieito, Ali Zeaiter, Elena Y Cristoveanu, Cristian Fernandez, Salvador Fudio, Honey Kumar Oberoi, Santiago Ponce Aix, Luis Paz-Ares, Carmen Kahatt, and Elena Garralda
- Subjects
Antitumor activity ,Cancer Research ,business.industry ,First in human ,chemistry.chemical_compound ,Oncology ,chemistry ,Transcription (biology) ,Cancer research ,Medicine ,In patient ,business ,Gene ,DNA - Abstract
3078 Background: PM14 is a new chemical entity that forms DNA adducts which specifically inhibit RNA synthesis and block active transcription of protein-coding genes. Antitumor activity has been demonstrated in vitro in several cell lines (e.g. lung, kidney, prostate), and in vivo in mice bearing xenografted human-derived tumors (soft tissue sarcoma, small cell lung cancer, ovarian, gastric, breast and renal cancer). Methods: Open-label, dose-escalating, phase I trial of PM14 administered as a 3-hour infusion i.v. every 3 weeks (q3wk) in patients (pts) with advanced solid tumors, adequate organ function and ECOG PS score of 0-1. Two schedules were explored: Schedule A (Day 1 [D1], Day 8 [D8]) and Schedule B (D1). Results: 37 pts were treated (Schedule A/B: 28/9 pts). Baseline characteristics of pts (A/B): median age 56/47 years; male 57%/56%; ECOG PS 0: 57%/56%; median of prior lines (range): 3 (1-8)/4 (1-10). Most common tumor types (A + B): STS (n=7 pts), ovarian (n=6), pancreatic (n=4), prostate cancer (n=3). The maximum tolerated dose was 4.5 mg/m2 for A (dose-limiting toxicities [DLTs]: D8 omission due to lack of recovery of lab parameters for re-treatment [n=2 pts]) and 5.6 mg/m2 (DLTs: G4 febrile neutropenia [n=1], G4 transaminase increase [n=1]) for B. The recommended dose (RD) was 3.0 mg/m2 on D1,D8 (A), and 4.5 mg/m2 on D1 (B). No DLTs were present at the RDs. Most common toxicities were hematological abnormalities and transaminase increase. Main toxicities at the RDs are shown below. Antitumor activity comprised stable disease ≥4 months in 7 heavily pretreated pts (6 in A; 1 in B) at all dose levels. Linear pharmacokinetics were observed for PM14 at tested doses (0.25-5.6 mg/m²), with geometric mean (CV%) total plasma clearance 5.9 L/h (88%), volume of distribution 128 L (81%) and median (range) terminal half-life 15.9 h (7.5-34.3 h). Less than 1.6% of administered dose was recovered in urine. Conclusions: RDs were determined for two PM14 schedules in pts with advanced solid tumors. At the RDs, PM14 is well tolerated and has a manageable safety profile. An expansion phase in specific tumor types, with an optional Bayesian continual reassessment method for RD fine-tuning, is ongoing with both schedules.[Table: see text]
- Published
- 2021
40. Abstract 2953: Adapting a molecular prescreening program to detect notch pathway alterations in the context of early drug development
- Author
-
Elena Garralda, Susana Aguilar, Cristina Saura, Teresa Macarulla, Lorena Fariñas, Irene Brana, Analia Azaro, Enriqueta Felip, Maria Vieito, Zighereda Ogbah, Rodrigo Dienstmann, Joan Carles, O. Saavedra, Elena Elez, Ignacio Matos, and Ana Vivancos
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Colorectal cancer ,Notch signaling pathway ,Cancer ,Context (language use) ,Biology ,medicine.disease ,Molecular biology ,Metastasis ,Exon ,Breast cancer ,Oncology ,medicine ,Gene - Abstract
Introduction: Gene alterations in NOTCH signaling pathway have a prevalence ranging from Methods: From Jan/2017 to Dec/2018, 1,697 patients (pts) had their FFPE tumor samples (either primary or metastasis) analyzed for mutations (mut) using a custom developed Amplicon-Seq panel of 59 cancer-related genes (including NOTCH1 [hotspots 5% exon coverage] and NOTCH4 [hotspots 3% exon coverage]) that was run in Illumina MiSeq (v1). Additionally, 502 samples were analyzed for Copy Number Alterations (CNA) using a panel of 44 genes (including NOTCH1-4) using NanoString nCounter (copy number between 4-6 copies were validated by FISH). From Jun/2018 to Jun/2019 an expanded NOTCH Amplicon-Seq panel v2 (including additional exons in NOTCH1 plus NOTCH2 and NOTCH3 hotspots regions) replaced the prior v1 panel and 618 samples were sequenced. Results: Colorectal cancer (CRC) (n= 404 [24%]; n=241 [39 %]) and breast cancer (BC) (n=283 [17%]; n= 194 [31%]) were the most frequent tumor types for v1 and v2 panel cohorts, respectively. NOTCH1-4 mut were detected in 11 cases (0.64%). The highest rate of NOTCH pathway alterations (NOTCH1-4 mut plus CN gain or loss) was detected in BC (n=14 [7.7%]) with a clear enrichment in triple negative subtype. Higher prevalence of NOTCH1-4 mut was detected using expanded v2 panel, with 17 NOTCH1-4 mut cases (2.75%). Head & neck (H&N) (n=3 [5%]); BC (n=6 [3%]); and CRC (n=6 [2.5%]) were most common tumors. From all NOTCH1-4 gene mut detected, 8 were known actionable oncogenic driver mut and 9 were variants of unknown significance. When comparing our latest v2 results with GENIE database (mostly large NGS panels with exon capture), we found a similar prevalence in NOTCH1-4 mut in BC and CRC, but lower prevalence was seen in H&N, biliary and gastric tumors. We enrolled 4 pt in clinical trials with NOTCH inhibitors (3 NOTCH1 actionable mut pt (0.48%), 1 NOTCH3 actionable mut pt (0.16%). Conclusion: Institutional efforts to increase coverage of NOTCH pathway genes improved the detection of actionable NOTCH1-4 mut. Differences in prevalence as compared to GENIE dataset may be attributed to the small number of samples tested with our v2 panel and larger panels covering all exons of NOTCH1-4 genes in GENIE cohort. Therefore, we keep evolving our NOTCH prescreening program for clinical trial enrichment. Citation Format: Analía Azaro, Susana Aguilar, Zighereda Ogbah, Omar Saavedra, María Vieito, Ignacio Matos, Irene Braña, Enriqueta Felip, Joan Carles, Lorena Fariñas, Elena Elez, Teresa Macarulla, Cristina Saura, Rodrigo Dienstmann, Elena Garralda, Ana Vivancos. Adapting a molecular prescreening program to detect notch pathway alterations in the context of early drug development [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 2953.
- Published
- 2020
41. Abstract CT147: Phase 1 dose escalation of MSC-1, a humanized anti-LIF monoclonal antibody, in patients with advanced solid tumors
- Author
-
Elena Garralda, Marc Oliva, Irene Brana, Robin M. Hallett, Joan Seoane, Enda Moran, Dorotea Maetzel, Maria Vieito Villar, Patricia Giblin, Nehal Lakhani, Alison M. Schram, Josep Tabernero, Anna Spreafico, Lillian L. Siu, Daniel D. Von Hoff, Kimberly Hoffmann, Robert Wasserman, Judit Anido, Erkut Borazanci, and Adrianne Kelly
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Cancer ,Immunosuppression ,medicine.disease ,Denosumab ,Gastrointestinal disorder ,Tolerability ,Cancer stem cell ,Internal medicine ,medicine ,business ,Leukemia inhibitory factor ,medicine.drug - Abstract
Leukemia Inhibitory Factor (LIF) is a pleiotropic cytokine, which is highly expressed in a subset of tumors and correlates with poor prognosis. LIF is hypothesized to contribute to tumor microenvironment immunosuppression and regulation of cancer stem cells. MSC-1 is a first-in-class humanized IgG1 monoclonal antibody that potently and selectively inhibits LIF. In pre-clinical models, MSC-1 decreases tumor growth through inhibition of STAT3 signaling, promoting immune stimulatory macrophages and increasing tumor infiltration of CD8 T and NK cells. The Phase Ia clinical study employed an accelerated 3 + 3 escalation design to explore safety and tolerability, dose-limiting toxicities (DLTs), preliminary efficacy, define a recommended phase II dose (RP2D), and evaluate exploratory tumor biomarkers. Eligible patients had advanced relapsed/refractory solid tumors and received treatment with MSC-1 intravenously (75mg-1500 mg) once every 3-weeks as a single agent until disease progression. The three highest dose cohorts were expanded to further assess safety, PK, engagement of LIF in the periphery, and assess immuno-regulatory tumor activity in matched pre- and on-treatment tumor biopsies. Forty-one patients (pts) were enrolled (14 in dose escalation; 27 in expanded cohorts) with the last pt completing the study on September 23, 2019. The most common tumor types were pancreatic (13), colorectal (5), head & neck (4) and ovarian (4). Pts had received a median of 3 prior lines of therapy. All pts experienced at least one adverse event (AE). The most common considered drug-related AEs were fatigue (N=8, 20%) and gastrointestinal disorder (N=8, 20%), and there was 1 considered drug-related SAE (Gr 2 osteonecrosis of jaw in a head and neck cancer patient who previously received radiation to the area and denosumab). There were no Dose Limiting Toxicities observed during the first cycle of treatment and 2 pts discontinued treatment due to AEs. The PK profile of MSC-1 was linear with an estimated terminal half-life of ∼13 days and benign anti-drug antibody profile. There was evidence of durable peripheral saturation of LIF binding demonstrating high level of target engagement. Results supported the selection of a RP2D of 1500 mg Q3W. Prolonged stable disease (≥ 16 weeks) was observed in 9 pts. Analysis of paired biopsies collected from matched metastatic lesions supported MSC-1 mediated STAT3 signaling inhibition, stimulatory (M1) to suppressive (M2) macrophage skewing in the majority of paired biopsies evaluated and increased CD8 T-cell infiltration in a subset of samples. Single agent MSC-1 was well tolerated in doses ranged from 75 mg to 1500 mg IV OD in patients with advanced solid tumors, showed promising activity as an anti-cancer therapy, and is Phase 1b/2 ready for combination with other agents. The updated final safety, efficacy, PK, LIF stabilization analyses, and tumor biopsy data will be presented. Citation Format: Alison Schram, Erkut Borazanci, Irene Brana, Maria Vieito Villar, Elena Garralda, Anna Spreafico, Marc Oliva, Nehal Lakhani, Robert Wasserman, Kimberly Hoffmann, Robin Hallett, Judit Anido, Dorotea Maetzel, Patricia Giblin, Enda Moran, Adrianne Kelly, Joan Seoane, Daniel D. Von Hoff, Lillian Siu, Josep Tabernero. Phase 1 dose escalation of MSC-1, a humanized anti-LIF monoclonal antibody, in patients with advanced solid tumors [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 CT147.
- Published
- 2020
42. Global Gene Expression Characterization of Circulating Tumor Cells in Metastasic Castration-Resistant Prostate Cancer Patients
- Author
-
Urbano Anido, Laura Muinelo-Romay, Mercedes Suárez-Cunqueiro, Helena Casas, Óscar Rapado-González, Maria Vieito, Luis León-Mateos, Rafael López-López, Alicia Abalo, Miguel Abal, Antonio Gómez-Tato, Universidade de Santiago de Compostela. Departamento de Cirurxía e Especialidades Médico-Cirúrxicas, Institut Català de la Salut, [León-Mateos L] Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain. Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain. [Abalo A, Casas H] Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain. [Anido U] Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS), 15706 Santiago de Compostela, Spain. [Rapado-González Ó] Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain. Liquid Biopsy Analysis Unit, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), 15706 Santiago de Compostela, Spain. Department of Surgery and Medical Surgical Specialties, Medicine and Dentistry School, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain. [Vieito M] Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Vall d’Hebron Hospital Universitari, Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
circulating tumor cells (CTCs) ,Neoplasms::Neoplastic Processes::Neoplasm Metastasis::Neoplastic Cells, Circulating [DISEASES] ,Population ,lcsh:Medicine ,Disease ,Article ,03 medical and health sciences ,Prostate cancer ,Expression arrays ,0302 clinical medicine ,Circulating tumor cell ,Metàstasi ,Prostate ,Biopsy ,medicine ,education ,Circulating tumor cells (CTCs) ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,Pròstata - Càncer ,medicine.diagnostic_test ,business.industry ,lcsh:R ,Castration-resistant prostate cancer (CRPC) ,General Medicine ,medicine.disease ,Neoplasms::Neoplasms by Site::Urogenital Neoplasms::Genital Neoplasms, Male::Prostatic Neoplasms::Prostatic Neoplasms, Castration-Resistant [DISEASES] ,medicine.anatomical_structure ,neoplasias::procesos neoplásicos::metástasis neoplásica::células neoplásicas circulantes [ENFERMEDADES] ,tumor markers ,Tumor progression ,neoplasias::neoplasias por localización::neoplasias urogenitales::neoplasias de los genitales masculinos::neoplasias de la próstata::neoplasias prostáticas resistentes a la castración [ENFERMEDADES] ,castration-resistant prostate cancer (CRPC) ,Tumor markers ,030220 oncology & carcinogenesis ,Cancer research ,Biomarker (medicine) ,business ,expression arrays - Abstract
Càncer de pròstata resistent a la castració (CRPC); Cèl·lules tumorals circulants (CTC); Marcadors tumorals Cáncer de próstata resistente a la castración (CRPC); Células tumorales circulantes (CTC); Marcadores tumorales Castration-resistant prostate cancer (CRPC); Circulating tumor cells (CTCs); Tumor markers Background: Current therapeutic options in the course of metastatic castration-resistant prostate cancers (mCRPC) reinforce the need for reliable tools to characterize the tumor in a dynamic way. Circulating tumor cells (CTCs) have emerged as a viable solution to the problem, whereby patients with a variety of solid tumors, including PC, often do not have recent tumor tissue available for analysis. The biomarker characterization in CTCs could provide insights into the current state of the disease and an overall picture of the intra-tumor heterogeneity. Methods: in the present study, we applied a global gene expression characterization of the CTC population from mCRPC (n = 9), with the goal to better understand the biology of these cells and identify the relevant molecules favoring this tumor progression. Results: This analysis allowed the identification of 50 genes specifically expressed in CTCs from patients. Six of these markers (HOXB13, QKI, MAOA, MOSPD1, SDK1, and FGD4), were validated in a cohort of 28 mCRPC, showing clinical interest for the management of these patients. Of note, the activity of this CTC signature was related to the regulation of MYC, a gene strongly implicated in the biology of mCRPC. Conclusions: Overall, our results represent new evidence on the great value of CTCs as a non-invasive biopsy to characterize PC. This work was partially financed with the “liquid Biopsy crowdfunding, 2017”. L.M-L. is supported by AECC.
- Published
- 2020
43. A phase Ib/II study of olutasidenib in patients with relapsed/refractory IDH1 mutant gliomas: Safety and efficacy as single agent and in combination with azacitidine
- Author
-
Sylvie Guichard, Sanjeev Forsyth, Danijela Levaci, Macarena I. de la Fuente, Mohammed M. Milhem, Patrick Kelly, Mark Rosenthal, Tobias Walbert, Yelena Mikhailov, Hui K Gan, Maria Vieito, Kate Lipford, Blythe Thomson, Brian A. Van Tine, Varun Monga, Howard Colman, and Alexander Sedkov
- Subjects
Cancer Research ,IDH1 ,business.industry ,Mutant ,Azacitidine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Isocitrate dehydrogenase ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Relapsed refractory ,Cancer research ,medicine ,In patient ,Single agent ,business ,DNA ,030215 immunology ,medicine.drug - Abstract
2505 Background: Isocitrate dehydrogenase 1 mutations (mIDH1) are present in >70% of patients with Grade II/III gliomas resulting in production and accumulation of (R)-2-hydroxyglutarate causing DNA hypermethylation and promoting tumorigenesis. Olutasidenib is an oral, potent, brain penetrant (Kpuu=0.4 in intact rodent), and selective inhibitor of mutated IDH1 protein. Methods: Patients (pts) with relapsed/refractory (R/R) mIDH1 gliomas received olutasidenib 150 mg BID, orally either as single agent (SA) or in combination (CO) with azacitidine in a dose confirmation phase Ib followed by efficacy evaluation phase II study (NCT: 03684811). Results: As of 31-Oct-2019, 29 pts with R/R mIDH1 glioma were treated with olutasidenib as SA (n=24) or CO (n=5). The median age was 45 yrs (range: 23-64) & 62% were male. WHO Glioma Grade (Gr) at study entry was: II (17%), III (52%) & IV (31%). Median number of prior treatments was 2 (1-5); 86% had received prior temozolomide. mIDH1 status was locally determined (IHC, NGS or PCR): R132H (86%), R132L (7%), R132C (3.5%) & unspecified (3.5%). The median duration of olutasidenib treatment for SA & CO was 4.8 (1-11.4) & 1 (0.2-2.3) months, respectively. Fifteen pts discontinued (disease progression [n=12], AE [n=1], withdrew consent [n=1], other [n=1]). For SA, the most common (>25%) TEAEs (all grades, regardless of attribution) were: fatigue (50%), nausea (50%), diarrhea (33%), ALT increase (29%) & headache (29%). For CO, TEAEs that occurred in ≥ 2 pts were: nausea (n=4), fatigue (n=2), neutropenia (n=2), ALT increase (n=2) & AST increase (n=2). There were 2 protocol defined DLTs in the CO cohort, 1 pt with Gr 4 ALT, Gr 3 AST & Gr 3 GGT elevations & 1 pt with Gr 3 ALT elevation. No pts experienced a TEAE of QTcF prolongation. SA best responses are shown in Table; CO pts are too early for response assessment. The median PFS for SA was 8.3 months. Twenty (87%) and 11 (48%) pts were alive and progression-free at 6 & 12 months, respectively. Conclusions: SA olutasidenib at 150 mg BID demonstrates acceptable safety and tolerability with preliminary clinical activity in glioma pts. Evaluation of CO is ongoing. Updated safety and clinical activity, as well as evaluations of serum/CSF PK/PD will be provided. Clinical trial information: NCT03684811 . [Table: see text]
- Published
- 2020
44. Efficacy and safety of the antibody-drug conjugate (ADC) SAR408701 in patients (pts) with non-squamous non-small cell lung cancer (NSQ NSCLC) expressing carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5)
- Author
-
Carole Helissey, Byoung Chul Cho, Valentina Boni, Jin-Soo Kim, Maria Vieito, Marie Hospitel, Charles Ricordel, Sophie Cousin, Tae Min Kim, Fabrice Barlesi, Anas Gazzah, Semra Yoruk, Mustapha Chadjaa, and Emiliano Calvo
- Subjects
Cancer Research ,Antibody-drug conjugate ,biology ,Cell adhesion molecule ,business.industry ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Carcinoembryonic antigen ,Oncology ,Non squamous ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,biology.protein ,In patient ,Non small cell ,Lung cancer ,business ,Clin oncol ,030215 immunology - Abstract
9505 Background: We report updated safety and efficacy of DM4-conjugated anti-CEACAM5 ADC from the expansion part of the first-in-human study (NCT02187848; Gazzah A et al. J Clin Oncol. 2019;37:15, 9072) in 92 NSQ NSCLC pts. Methods: CEACAM5 expression was assessed by immunohistochemistry on archived tumor samples. Two cohorts of pts have been analyzed: moderate and high expressors, with CEACAM5 expression at ≥2+ intensity between ≥1% to < 50% and ≥50% of the tumor cell population, respectively. SAR408701 was administered at 100 mg/m2 IV every 2 weeks. Tumor assessments were done every 4 cycles (8 weeks). Primary endpoint was overall response rate (ORR). Results: As of January 2020, 92 pts were treated: 28 moderate and 64 high expressors, with median age 62.5 years (31–91; 42.4% of pts ≥65), 51.1% male, 71.7% ECOG PS ≥1; median of 3 prior treatments (1–10 lines) for advanced disease, including anti-tubulin agents (60.9%) and anti-PD1/PD-L1 (75%). In the moderate expressor cohort, 2 confirmed partial responses (PR) were observed (ORR 7.1%). In the high expressor cohort, 13 pts had confirmed PRs (ORR 20.3% [95% confidence interval 12.27%–31.71%]); 27 (42.2%) had stable disease; ORR of 17.8% was observed in 45 pts who had prior anti-PD1/PD-L1. Pts had a median of 7 (1–49) cycles; median relative dose intensity was 0.98. Six pts discontinued due to treatment-emergent adverse events (TEAEs). Most frequent TEAEs (all grades) were asthenia (38.0%), keratopathy/keratitis (38.0%), peripheral neuropathy (26.1%), dyspnea (23.9%), and diarrhea (22.8%). 31 pts had dose modification due to a TEAE, including dose reduction for keratopathy/keratitis in 10 pts. Hematological toxicity included leukopenia (14.4%), neutropenia (4.4%), and thrombocytopenia (13.3%). Grade ≥3 TEAEs occurred in 47.8% of pts and were assessed as drug-related in 15.2%. Conclusions: SAR408701 shows promising antitumor activity in heavily pretreated advanced NSQ NSCLC pts with high CEACAM5 expression. SAR408701 was well tolerated, with minimal hematological toxicity compared to conventional chemotherapy; keratopathy was reversible and manageable with dose modification. These data support the activity of SAR408701 in NSQ NSCLC CEACAM5 high expressors. A phase 3 trial evaluating the activity of CEACAM5-DM4 ADC monotherapy in comparison with docetaxel in NSQ NSCLC CEACAM5 high expressors after failure of standard first line chemotherapy and anti-PD1/PD-L1 is underway. Clinical trial information: NCT02187848 .
- Published
- 2020
45. INDUCE-1: Report on safety run-in cohorts combining Inducible T-cell co-stimulatory receptor (ICOS) agonist GSK3359609 (GSK609) with platinum+5-FU chemotherapy (5-FU/plat), with or without pembrolizumab (PE), for the treatment of advanced solid tumors
- Author
-
Tatiana Hernandez-Guerrero, Michael Chisamore, Raid Aljumaily, Sonia Franco, Maria Vieito, Catherine E. Ellis, Ani Sarkis Balmanoukian, Christophe Le Tourneau, Courtney Henry, Debra Rogan, Danny Rischin, Axel Hoos, David C. Turner, Jose Manuel Trigo, Erminia Massarelli, Ruby Sung, Marc S. Ballas, and Sapna Yadavilli
- Subjects
Agonist ,Cancer Research ,Chemotherapy ,business.industry ,medicine.drug_class ,T cell ,medicine.medical_treatment ,Cell ,Pembrolizumab ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,business ,Receptor ,Head and neck ,030215 immunology - Abstract
6544 Background: The KEYNOTE-048 study (Burtness, at al. Lancet 2019;394:1915–28) led to approval of PE in combination with 5-FU/plat for first-line (1L) treatment of head and neck squamous cell carcinoma (HNSCC). The Phase I INDUCE-1 study (NCT02723955) has shown that GSK609±PE has a manageable safety profile in patients (pts) with advanced solid tumors (Hansen, at al. Annals of Oncology 2018;29[suppl_8]:viii404) and that GSK609 combined with PE has anti-tumor activity in pts with anti-PD-1/L1-naïve HNSCC (Rischin, et al. Annals of Oncol 2019;30[Supplement_5]:v454–5). To evaluate the safety of GSK609±PE in combination with 5-FU/plat, we initiated additional safety cohorts. Methods: Pts eligible for GSK609+5-FU/plat had a diagnosis of advanced selected solid tumors and ≤5 prior lines of systemic therapy. Pts eligible for GSK609+PE+5-FU/plat had a diagnosis of recurrent or metastatic 1L HNSCC deemed incurable by local therapies. 5-FU/plat was administered every 3 weeks (Q3W) for 4-6 cycles (Burtness, at al. Lancet 2019;394:1915–28); GSK609 24 or 80 mg ±PE 200 mg were administered Q3W for up to 2 years/35 cycles or until disease progression or unacceptable toxicity. Results: Twenty-nine pts were enrolled in the 5-FU/plat safety cohorts: 10 pts in the GSK609+5-FU/plat cohort and 19 pts in the GSK609+PE+5-FU/plat cohort. With GSK609+5-FU/plat, 9/10 (90%) pts experienced ≥ 1 adverse event (AE). Of 32 AEs of any grade, 9 were Grade ≥3 and 3 were serious AEs (SAEs). Two of the 3 SAEs were related to study treatment (oral mucositis and febrile pancytopenia). With GSK609+PE+5-FU/plat, 18/19 (94.7%) pts experienced ≥ 1 AE. Of 119 AEs of any grade, 24 were Grade ≥3 and 15 were SAEs. Of the 15 SAEs, 11 were related to study treatment (febrile neutropenia [n=4], colitis [n=2], diarrhea [n=1], vomiting [n=1], acute kidney injury [n=1], cardiac chest pain [n=1] and lung infection [n=1]). For all cohorts, no Grade 5 AEs were observed. For 10 pts evaluable for confirmed best overall response in all cohorts, 2 pts had partial response, 6 pts had stable disease and 2 pts were nonevaluable. No difference in GSK609 exposure was observed relative to GSK609 monotherapy. Conclusions: The safety profile of GSK609 in combination with 5-FU/plat±PE is manageable. Most AEs were Grades 1 or 2 and consistent with PE and chemotherapy toxicities. Continued follow-up to investigate long-term safety and efficacy of this combination is warranted. Clinical trial information: NCT02723955 .
- Published
- 2020
46. Abstract B050: Validation of body mass index (BMI) as a prognostic factor in patients (pts) treated with immune checkpoint inhibitors (ICI) across multiple cancer types (CT) and the impact of confounding factors (CF)
- Author
-
Elena Garralda, Cinta Hierro, Guillem Aegiles, Analia Azaro, Ignacio Matos, Alejandra Ivars, Guillermo Villacampa, Ana Oaknin, Gemma Sala, Maria Vieito, Rodrigo Dienstmann, Irene Brana, Cristina Viaplana, Eva Muñoz, I. Gardeazabal, Helena Verdaguer, Juan Martin-Liberal, Maria Alsina, A. Callejo, and O. Saavedra
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Proportional hazards model ,business.industry ,Confounding ,Cancer ,medicine.disease ,Obesity ,Metformin ,Internal medicine ,Concomitant ,medicine ,Prospective cohort study ,business ,Body mass index ,medicine.drug - Abstract
Introduction: Obesity results in PD1-mediated T-cell dysfunction in preclinical models and improved outcomes from ICI. We wanted to validate this clinical association in a prospective cohort of pts receiving PD1/L1 inhibitors across multiple CT and investigate the potential impact of CF, such as age and concomitant medications (CM) that may interact with obesity. Methods: Clinicopathological data from 310 pts treated with ICI at VHIO phase 1 Unit from Aug'12 to Jul'18 were investigated. Frequent CM included metformin (M), statins (S) and others. Associations between different variables and progression-free survival (PFS) were assessed with univariable and multivariate Cox regression models and survival data were calculated by the Kaplan-Meier method. Tumor types were stratified by proven PD1/L1 inhibitor efficacy versus unknown PD1/L1 sensitivity. Results: Out of 310 pts, median age was 59.8 years (y), 54.2% were male and most frequent tumor type was melanoma (19.4%) and lung (13.2%). In univariate models, pts with BMI>25 kg/m2 (n=147, 47%) had increased PFS (3.5 months [m], CI95% 2.8-5.5) as compared to those with BMI Citation Format: Analía Azaro, Alejandra Ivars, Ignacio Matos, Omar Saavedra, Itziar Gardeazabal, Juan Martin-Liberal, Cinta Hierro, María Vieito, Irene Braña, Cristina Viaplana, Guillermo Villacampa, Gemma Sala, María Alsina, Ana Callejo, Helena Verdaguer, Guillem Aegiles, Ana Oaknin, Eva Muñoz, Rodrigo Dienstmann, Elena Garralda. Validation of body mass index (BMI) as a prognostic factor in patients (pts) treated with immune checkpoint inhibitors (ICI) across multiple cancer types (CT) and the impact of confounding factors (CF) [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr B050. doi:10.1158/1535-7163.TARG-19-B050
- Published
- 2019
47. Molecular Diagnosis of Diffuse Gliomas through Sequencing of Cell-Free Circulating Tumor DNA from Cerebrospinal Fluid
- Author
-
Carlota Rubio-Perez, Elena Martínez-Sáez, Regina Mayor, Francesc Graus, Juan Sahuquillo, Ana Vivancos, Maria Vieito, Francisco Martínez-Ricarte, Esteban Cordero, Nuria Lopez-Bigas, Soledad Gallego, Marta Cicuendez, Maria A. Poca, Joan Seoane, Santiago Ramón y Cajal, Joan Carles, Josep Tabernero, and Estela Pineda
- Subjects
Adult ,Male ,Cancer Research ,IDH1 ,DNA Mutational Analysis ,Kaplan-Meier Estimate ,IDH2 ,Circulating Tumor DNA ,03 medical and health sciences ,0302 clinical medicine ,Diffuse Astrocytoma ,Glioma ,medicine ,Biomarkers, Tumor ,Humans ,neoplasms ,Exome sequencing ,ATRX ,In Situ Hybridization, Fluorescence ,Aged ,Aged, 80 and over ,business.industry ,Genomics ,Middle Aged ,medicine.disease ,Prognosis ,Primary tumor ,Immunohistochemistry ,Magnetic Resonance Imaging ,Oncology ,Molecular Diagnostic Techniques ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,Oligodendroglioma ,business ,030217 neurology & neurosurgery - Abstract
Purpose: Diffuse gliomas are the most common primary tumor of the brain and include different subtypes with diverse prognosis. The genomic characterization of diffuse gliomas facilitates their molecular diagnosis. The anatomical localization of diffuse gliomas complicates access to tumor specimens for diagnosis, in some cases incurring high-risk surgical procedures and stereotactic biopsies. Recently, cell-free circulating tumor DNA (ctDNA) has been identified in the cerebrospinal fluid (CSF) of patients with brain malignancies. Experimental Design: We performed an analysis of IDH1, IDH2, TP53, TERT, ATRX, H3F3A, and HIST1H3B gene mutations in two tumor cohorts from The Cancer Genome Atlas (TCGA) including 648 diffuse gliomas. We also performed targeted exome sequencing and droplet digital PCR (ddPCR) analysis of these seven genes in 20 clinical tumor specimens and CSF from glioma patients and performed a histopathologic characterization of the tumors. Results: Analysis of the mutational status of the IDH1, IDH2, TP53, TERT, ATRX, H3F3A, and HIST1H3B genes allowed the classification of 79% of the 648 diffuse gliomas analyzed, into IDH-wild-type glioblastoma, IDH-mutant glioblastoma/diffuse astrocytoma and oligodendroglioma, each subtype exhibiting diverse median overall survival (1.1, 6.7, and 11.2 years, respectively). We developed a sequencing platform to simultaneously and rapidly genotype these seven genes in CSF ctDNA allowing the subclassification of diffuse gliomas. Conclusions: The genomic analysis of IDH1, IDH2, TP53, ATRX, TERT, H3F3A, and HIST1H3B gene mutations in CSF ctDNA facilitates the diagnosis of diffuse gliomas in a timely manner to support the surgical and clinical management of these patients. Clin Cancer Res; 24(12); 2812–9. ©2018 AACR.
- Published
- 2017
48. A multimarker panel for circulating tumor cells detection predicts patient outcome and therapy response in metastatic colorectal cancer
- Author
-
María de los Ángeles Casares de Cal, Rafael López-López, Antonio Gómez-Tato, Laura Muinelo-Romay, Antonio Díaz-López, Jorge Barbazan, Miguel Abal, Amparo Cano, Sonia Candamio, and Maria Vieito
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,business.industry ,Colorectal cancer ,Cancer ,medicine.disease ,Therapy response ,Circulating tumor cell ,Internal medicine ,Medicine ,Cell isolation ,Epithelial–mesenchymal transition ,Liquid biopsy ,business ,neoplasms ,TIMP1 - Abstract
Circulating tumor cells (CTCs), proposed as major players in cancer dissemination, have demonstrated clinical prognostic significance in several cancer types. However, their predictive value remains unclear. Here we evaluated the clinical utility of six CTC markers (tissue specific and epithelial to mesenchymal transition transcripts) both as prognostic and predictive tools in metastatic colorectal cancer (mCRC) patients. CTCs were immunoisolated from blood in 50 mCRC patients at baseline and at 4 and 16 weeks after treatment onset. Expression levels of GAPDH, VIL1, CLU, TIMP1, LOXL3 and ZEB2 were determined by qualitative polymerase chain reaction and normalized to the unspecific cell isolation marker CD45. At baseline, median progression-free survival (PFS) and overall survival (OS) for patients with high CTC markers were 6.3 and 12.7 months, respectively, versus 12.7 and 24.2 for patients with low CTC markers (PFS; p = 0.0003; OS; p = 0.044). Concerning response to therapy, PFS and OS for patients with increased CTC markers along treatment were, respectively, 6.6 and 13.1 months, compared with 12.7 and 24.3 for patients presenting CTC markers reduction (PFS; p = 0.004; OS; p = 0.007). Of note, CTC markers identified therapy-refractory patients not detected by standard image techniques. Patients with increased CTC markers along treatment, but classified as responders by computed tomography, showed significantly shorter survival times (PFS: 7.8 vs. 13.2; OS: 14.4 vs. 24.4; months). In conclusion, we have generated a CTC marker panel for prognosis evaluation and the identification of patients benefiting or not from therapy in mCRC. Our methodology efficiently classified patients earlier than routine computed tomography and from a minimally invasive liquid biopsy.
- Published
- 2014
49. Phase I study of CC-90010 in patients with advanced solid tumors and relapsed/refractory non-Hodgkin lymphoma (R/R NHL)
- Author
-
Tatiana Hernandez-Guerrero, Juan De Alvaro, Manisha Lamba, Ellen Filvaroff, Olga Ferrero, Marina Arias, Juan Manuel Manuel Sepulveda Sanchez, Marlene Zuraek, Zariana Nikolova, Victor Moreno, Tania Sánchez Pérez, Irene Brana, Rafael Sarmiento, Bernard Doger, Bishoy Hanna, O. Saavedra, Ida Aronchik, Maria Vieito Villar, and Jorge DiMartino
- Subjects
Cancer Research ,business.industry ,medicine.disease_cause ,Phase i study ,Bromodomain ,Oncology ,Relapsed refractory ,medicine ,Cancer research ,Hodgkin lymphoma ,In patient ,Epigenetics ,Carcinogenesis ,business ,Gene - Abstract
3015 Background: Bromodomain and extra-terminal (BET) proteins are epigenetic readers that control expression of genes involved in cell growth and oncogenesis. CC-90010 is an oral, potent and reversible BET inhibitor that showed promising activity in lymphoma and solid tumor cell lines and reduced tumor growth in xenograft models. Methods: CC-90010-ST-001 (NCT03220347; 2015-004371-79) is a phase I, first-in-human study of CC-90010 in patients (pts) with advanced solid tumors and R/R NHL. Three schedules and 11 dose levels were evaluated (Table). Primary objectives were to determine safety, maximum-tolerated dose and/or recommended phase II dose (RP2D). Secondary objectives were the identification of early activity signals, pharmacokinetics and pharmacodynamics (PD). Results: As of 10 Dec 2018, 69 pts were enrolled, 67 with solid tumors and 2 with R/R NHL. Data shown are from all pts (N = 69). The median age was 57 y (range, 21–80), 38 (55%) were male, and the median number of prior systemic anticancer regimens was 3 (range, 1–9). The RP2Ds were dose cohorts 3A and 4B. Dose-limiting toxicities (n = 6) occurred in dose cohorts 3A, 3C, and 4B. Grade 3/4 treatment-related adverse events (TRAEs) occurred in 17 pts (25%), most commonly (≥2 pts) thrombocytopenia (7%), platelet count decreased (4%), fatigue (3%), and increased alanine aminotransferase (3%). No deaths from toxicity occurred. Two pts (endometrial carcinoma and astrocytoma) had a partial response (PR); 1 occurred after the data cutoff. Seven pts had prolonged stable disease (SD) > 9 mo. Exposures and PD marker regulation increased with dose in each dosing schedule; terminal half-life was ~ 73 h. Conclusions: Most of the TRAEs observed were mild or moderate in severity, reversible, and manageable by dose adjustments and/or supportive care. Promising ongoing anticancer activity with prolonged SD and PRs were observed. The preliminary clinical data provide the rationale for dose expansion of CC-90010 in pts with selected advanced malignancies. Clinical trial information: NCT03220347. [Table: see text]
- Published
- 2019
50. Patient survival with immune checkpoint inhibitors and targeted agents in phase 1 trials: A propensity score weighted analysis
- Author
-
Cinta Hierro, Maria Vieito Villar, Guillem Argiles, Cristina Viaplana, Juan Martin-Liberal, O. Saavedra, Helena Verdaguer, Analia Azaro, Josep Tabernero, Mafalda Oliveira, Ignacio Matos, Eva Muñoz-Couselo, Elena Garralda, I. Gardeazabal, Maria Ochoa de Olza, Alejandro Navarro, Rodrigo Dienstmann, Joan Carles, Guillermo Villacampa, and Irene Brana
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Drug development ,business.industry ,Internal medicine ,Immune checkpoint inhibitors ,Propensity score matching ,medicine ,Phase 1 trials ,Patient survival ,business - Abstract
2580 Background: There have been important changes in early drug development units with an unprecedented increase of immune-oncology (IO) trials. Currently at the Vall d’Hebron Institute Oncology (VHIO) close to 50% of our Phase 1 trials (Ph1t) portfolio includes IO drugs, while from 2011 to 2015 more than 80% of our trials assessed targeted agents (TA). We wanted to investigate whether this swift had a positive impact on patient (pts) outcome. Methods: We performed a retrospective analysis of the pts treated with IO and TA at VHIO Ph1t Unit from Jun’11 to May’18. Only patients treated with IO in ≥ 2nd line were included (and without an approved IO therapy as per standard-of-care) and those with TA classified as tiers II-III-IV by the ESMO scale for clinical actionability of molecular targets ESCAT (which also represents unapproved indications). The aim of this study was to compare overall survival (OS) for the two cohorts. Given the non-randomized nature of the study a propensity score weighting (PSW) was used to control for selection bias in treatment effect estimation. Results: Out of 545 eligible pts, 281 (51.5%) received TA and 264 (48.5%) IO, with unadjusted median OS (mOS) of 7.7 months (m) and 9.2m, respectively. In univariate analysis, OS was associated with tumor type, number of previous treatment lines, regimen (monotherapy vs combination), and clinical-laboratory prognostic factors (Vioscore: albumin < 3.5 g/dl; LDH > upper limit of normal; neutrophil/[leukocytes minus neutrophils] ratio (dNLR) > 3; more than 2 sites of metastasis; and presence of liver metastasis) (p < 0.05). After adjusting for these factors in a PSW model, the IO group showed statistically significant longer OS with HR = 0.75 (CI95% 0.65 – 0.86, p < 0.0001). The In a stratified analysis by tumor type we found no significant heterogeneity in the relative benefit of IO over TA. Conclusions: In real world data from our Ph1t population, treatment with IO was associated with longer OS than treatment with TA, even after adjusting for known prognostic factors and treatment selection biases. These results suggest that the likelihood of patient benefit with IO therapies in Ph1t is increasing.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.