26 research outputs found

    First-in-human phase I/Ib open-label dose-escalation study of GWN323 (anti-GITR) as a single agent and in combination with spartalizumab (anti-PD-1) in patients with advanced solid tumors and lymphomas

    Get PDF
    Antibodies; Combination; NeoplasmAnticossos; Combinació; NeoplàsiaAnticuerpos; Combinación; NeoplasiaBackground GWN323 is an IgG1 monoclonal antibody (mAb) against the glucocorticoid-induced tumor necrosis factor receptor-related protein. This first-in-human, open-label phase I/Ib study aimed to investigate the safety and tolerability and to identify the recommended doses of GWN323 with/without spartalizumab, an anti-programmed cell death receptor-1 agent, for future studies. Pharmacokinetics, preliminary efficacy and efficacy biomarkers were also assessed. Methods Patients (aged ≥18 years) with advanced/metastatic solid tumors with Eastern Cooperative Oncology Group performance status of ≤2 were included. GWN323 (10–1500 mg) or GWN323+spartalizumab (GWN323 10–750 mg+spartalizumab 100–300 mg) were administered intravenously at various dose levels and schedules during the dose-escalation phase. Dose-limiting toxicities (DLTs) were assessed during the first 21 days in a single-agent arm and 42 days in a combination arm. Adverse events (AEs) were graded per National Cancer Institute-Common Toxicity Criteria for Adverse Events V.4.03 and efficacy was assessed using Response Evaluation Criteria in Solid Tumors V.1.1. Results Overall, 92 patients (single-agent, n=39; combination, n=53) were included. The maximum administered doses (MADs) in the single-agent and combination arms were GWN323 1500 mg every 3 weeks (q3w) and GWN323 750 mg+spartalizumab 300 mg q3w, respectively. No DLTs were observed with single-agent treatment. Three DLTs (6%, all grade ≥3) were noted with combination treatment: blood creatine phosphokinase increase, respiratory failure and small intestinal obstruction. Serious AEs were reported in 30.8% and 34.0%, and drug-related AEs were reported in 82.1% and 77.4% of patients with single-agent and combination treatments, respectively. Disease was stable in 7 patients and progressed in 26 patients with single-agent treatment. In combination arm patients, 1 had complete response (endometrial cancer); 3, partial response (rectal cancer, adenocarcinoma of colon and melanoma); 14, stable disease; and 27, disease progression. GWN323 exhibited a pharmacokinetic profile typical of mAbs with a dose-dependent increase in the pharmacokinetic exposure. Inconsistent decreases in regulatory T cells and increases in CD8+ T cells were observed in the combination arm. Gene expression analyses showed no significant effect of GWN323 on interferon-γ or natural killer-cell signatures. Conclusions GWN323, as a single agent and in combination, was well tolerated in patients with relapsed/refractory solid tumors. The MAD was 1500 mg q3w for single-agent and GWN323 750 mg+spartalizumab 300 mg q3w for combination treatments. Minimal single-agent activity and modest clinical benefit were observed with the spartalizumab combination.This study was supported by Novartis Oncology

    Futibatinib, an Irreversible FGFR1–4 Inhibitor, in Patients with Advanced Solid Tumors Harboring FGF/FGFR Aberrations: A Phase I Dose-Expansion Study

    Get PDF
    Futibatinib; Advanced solid tumors; AberrationsFutibatinib; Tumores sólidos avanzados; AberracionesFutibatinib; Tumors sòlids avançats; AberracionsFutibatinib, a highly selective, irreversible FGFR1–4 inhibitor, was evaluated in a large multihistology phase I dose-expansion trial that enrolled 197 patients with advanced solid tumors. Futibatinib demonstrated an objective response rate (ORR) of 13.7%, with responses in a broad spectrum of tumors (cholangiocarcinoma and gastric, urothelial, central nervous system, head and neck, and breast cancer) bearing both known and previously uncharacterized FGFR1–3 aberrations. The greatest activity was observed in FGFR2 fusion/rearrangement–positive intrahepatic cholangiocarcinoma (ORR, 25.4%). Some patients with acquired resistance to a prior FGFR inhibitor also experienced responses with futibatinib. Futibatinib demonstrated a manageable safety profile. The most common treatment-emergent adverse events were hyperphosphatemia (81.2%), diarrhea (33.5%), and nausea (30.4%). These results formed the basis for ongoing futibatinib phase II/III trials and demonstrate the potential of genomically selected early-phase trials to help identify molecular subsets likely to benefit from targeted therapy. Significance: This phase I dose-expansion trial demonstrated clinical activity and tolerability of the irreversible FGFR1–4 inhibitor futibatinib across a broad spectrum of FGFR-aberrant tumors. These results formed the rationale for ongoing phase II/III futibatinib trials in cholangiocarcinoma, breast cancer, gastroesophageal cancer, and a genomically selected disease-agnostic population

    Safety, pharmacokinetics, and antitumor activity of the anti-CEACAM5-DM4 antibody–drug conjugate tusamitamab ravtansine (SAR408701) in patients with advanced solid tumors: first-in-human dose-escalation study

    Get PDF
    Antibody–drug conjugate; Dose-escalation study; Tusamitamab ravtansineConjugado anticuerpo-fármaco; Estudio de escalada de dosis; Tusamitamab ravtansinaConjugat anticossos-fàrmac; Estudi d'escalada de dosi; Tusamitamab ravtansinaTusamitamab ravtansine (SAR408701) is an antibody–drug conjugate composed of a humanized monoclonal antibody that binds carcinoembryonic antigen-related cell adhesion molecule-5 (CEACAM5) and a cytotoxic maytansinoid that selectively targets CEACAM5-expressing tumor cells. In this phase I dose-escalation study, we evaluated the safety, pharmacokinetics, and preliminary antitumor activity of tusamitamab ravtansine in patients with solid tumors. Patients and methods Eligible patients were aged ≥18 years, had locally advanced/metastatic solid tumors that expressed or were likely to express CEACAM5, and had an Eastern Cooperative Oncology Group Performance Status of 0 or 1. Patients were treated with ascending doses of tusamitamab ravtansine intravenously every 2 weeks (Q2W). The first three dose levels (5, 10, and 20 mg/m2) were evaluated using an accelerated escalation protocol, after which an adaptive Bayesian procedure was used. The primary endpoint was the incidence of dose-limiting toxicities (DLTs) during the first two cycles, graded using National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE) v4.03 criteria. Results Thirty-one patients received tusamitamab ravtansine (range 5-150 mg/m2). The DLT population comprised 28 patients; DLTs (reversible grade 3 microcystic keratopathy) occurred in three of eight patients treated with tusamitamab ravtansine 120 mg/m2 and in two of three patients treated with 150 mg/m2. The maximum tolerated dose was identified as 100 mg/m2. Twenty-two patients (71%) experienced ≥1 treatment-related treatment-emergent adverse event (TEAE), seven patients (22.6%) experienced ≥1 treatment-related grade ≥3 TEAE, and three patients (9.7%) discontinued treatment due to TEAEs. The most common TEAEs were asthenia, decreased appetite, keratopathy, and nausea. Three patients had confirmed partial responses. The mean plasma exposure of tusamitamab ravtansine increased in a dose-proportional manner from 10 to 150 mg/m2. Conclusions Tusamitamab ravtansine had a favorable safety profile with reversible, dose-related keratopathy as the DLT. Based on the overall safety profile, pharmacokinetic data, and Bayesian model recommendations, the maximum tolerated dose of tusamitamab ravtansine was defined as 100 mg/m2 Q2W.This work was supported by Sanofi, France (no grant number)

    Phase I prognostic online (PIPO): A web tool to improve patient selection for oncology early phase clinical trials

    Get PDF
    Immunotherapy; Phase 1 trials; Prognostic modelInmunoterapia; Ensayos de fase 1; Modelo pronósticoImmunoteràpia; Assajos de fase 1; Model pronòsticPurpose 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)

    Caracterización del mecanismo de acción de los pan-inhibidores selectivos del receptor del factor de crecimiento fibroblástico e identificación de potenciales biomarcadores predictivos de respuesta mediante la utilización de xenoinjertos derivados de pacientes con alteraciones en esta vía molecular.

    Get PDF
    En cĂ ncer, la desregulaciĂł de la via del FGFR:FGF s'ha associat a un creixement tumoral descontrolat, una angiogènesi aberrant i una major capacitat de metastatitzaciĂł. En aquest context, en l'Ăşltima dècada s'ha fet un gran esforç per desenvolupar fĂ rmacs dirigits contra aquesta diana terapèutica. Les dades d'eficĂ cia incipients amb la primera generaciĂł d'inhibidors de la via, fĂ rmacs multi-tirosina quinasa (MTKI), han estat mĂ©s aviat limitats, amb un percentatge significatiu d'efectes secundaris no desitjats -off target-. Per aquest motiu, s'estan desenvolupant nous inhibidors mĂ©s selectius i potents del FGFR (FGFRinh), però encara existeix controvèrsia sobre quines d'aquestes alteracions podrien ser els millors biomarcadors predictius de resposta al bloqueig amb FGFRinh. Amb aquest projecte de tesi, pretenem contribuir a la definiciĂł d'una estratègia terapèutica òptima per als pacients amb alteracions de la via del FGFR: FGF, mitjançant una millor selecciĂł d'aquells que mĂ©s poden beneficiar-se d'aquests FGFRinh. Amb aquest objectiu, aquesta memòria per a la defensa de tesi consta de dos estudis principals. El primer es tracta d'un estudi retrospectiu d'una cohort de pacients amb cĂ ncer avançat, pre-seleccionats per ser portadors de tumors amb aberracions en els gens del FGFR i FGFs detectades dins del programa de pre-screening VHIO, i inclosos en assaigs fase 1 on s'estava testant un FGFRinh. Es presenten les dades de l'anĂ lisi exploratori de biomarcadors realitzat desprĂ©s de la correlaciĂł estadĂ­stica de les dades clĂ­nic-patològiques i d'eficĂ cia als FGFRinh disponibles. La majoria d'aquests estudis partien de la hipòtesi que qualsevol alteraciĂł molecular en la via del FGFR:FGF podia servir com a biomarcador predictiu de resposta als FGFRinh, sense importar el subtipus tumoral. No obstant això, amb aquest anĂ lisi retrospectiu pretenem identificar entre els responedors a un subgrup de pacients portadors d'una alteraciĂł especĂ­fica FGFR:FGF comuna. El nostre objectiu Ă©s delimitar amb mĂ©s precisiĂł quins d'aquests determinants moleculars sĂłn en realitat biomarcadors fiables i discernir si, en realitat, estan al mateix nivell en relaciĂł a la predicciĂł de resposta en els diferents contextos histològics (p.ex. alteracions a nivell de l'ADN -amplificacions, mutacions, traslocacions- versus nivell mRNA versus proteĂŻna). El segon estudi en canvi, es tracta d'un estudi prospectiu amb l'objectiu de generar una col·lecciĂł de mostres de pacients amb cĂ ncer avançat seleccionats molecularment per les seves alteracions en els gens de la via FGFR: FGF. Es presenten dades sobre el desenvolupament del protocol investigacional FGFR 360Âş RESISTANCE, la plataforma traslacional implementada per obtenir mostres seriades (biòpsies tumorals pre-, en curs i post-tractament, plasma i fins i tot autòpsies en calent) en diferents moments de la malaltia d'aquests pacients. Aquest valuĂłs repositori de mostres ha servit per generar xenoinjerts derivats de pacients (PDXs), utilitzats en el nostre laboratori per validar in vivo algunes de les hipòtesis prèviament observades, aixĂ­ com integrar el coneixement obtingut de l'anĂ lisi molecular de les mostres mĂ©s rellevants d'alguns responedors inusuals seleccionats. La presentaciĂł de cadascĂşn d'aquests estudis s'acompanya d'una discussiĂł dels resultats obtinguts, aixĂ­ com un sumari final de les conclusions assolides i les futures lĂ­nies de recerca associades.En cáncer, la desregulaciĂłn de la vĂ­a del FGFR:FGF se ha asociado a un crecimiento tumoral descontrolado, una angiogĂ©nesis aberrante y una mayor capacidad de metastatizaciĂłn. En este contexto, en la Ăşltima dĂ©cada se ha realizado un gran esfuerzo en desarrollar fármacos dirigidos contra esta diana terapĂ©utica. Los datos de eficacia incipientes con la primera generaciĂłn de inhibidores de la vĂ­a, fármacos multi-tirosina quinasa (MTKI), han sido más bien limitados, con un significativo porcentaje de efectos secundarios no deseados –off target-. Por este motivo, se están desarrollando nuevos inhibidores más selectivos y potentes del FGFR (FGFRinh), pero aĂşn existe controversia sobre cuáles de estas alteraciones podrĂ­an ser los mejores biomarcadores de predicciĂłn de respuesta al bloqueo con FGFRinh. Con este proyecto de tesis, pretendemos contribuir a la definiciĂłn de una estrategia terapĂ©utica Ăłptima para los pacientes con alteraciones de la vĂ­a del FGFR:FGF, mediante una mejor selecciĂłn de aquĂ©llos que más pueden beneficiarse de estos FGFRinh. Con tal objetivo, esta memoria para la defensa de tesis consta de dos estudios principales. El primero se trata de un estudio retrospectivo de una cohorte de pacientes con cáncer avanzado, pre-seleccionados por ser portadores de tumores con aberraciones en los genes del FGFR y FGFs detectadas dentro del programa de pre-screening VHIO, e incluidos en ensayos fase 1 dĂłnde se estaba testando un FGFRinh. Se presentan los datos del análisis exploratorio de biomarcadores realizado tras la correlaciĂłn estadĂ­stica de los datos clĂ­nico-patolĂłgicos y de eficacia a los FGFRinh disponibles. La mayorĂ­a de estos estudios partĂ­an de la hipĂłtesis de que cualquier alteraciĂłn molecular en la vĂ­a del FGFR:FGF podĂ­a servir como biomarcador predictivo de respuesta a los FGFRinh, independientemente del subtipo tumoral. Sin embargo, con este análisis retrospectivo pretendemos identificar entre los respondedores a un subgrupo de pacientes portadores de una alteraciĂłn especĂ­fica comĂşn FGFR:FGF. Nuestro objetivo es delimitar con mayor precisiĂłn cuáles de estos determinantes moleculares son en realidad biomarcadores fiables y discernir si, en realidad, están al mismo nivel en relaciĂłn a la predicciĂłn de respuesta en los distintos contextos histolĂłgicos (p.ej. alteraciones a nivel del ADN –amplificaciones, mutaciones, translocaciones- versus nivel mRNA versus proteĂ­na). El segundo estudio en cambio, se trata de un estudio prospectivo cuyo objetivo es generar una colecciĂłn de muestras de pacientes con cáncer avanzado seleccionados molecularmente por sus alteraciones en los genes de la vĂ­a FGFR:FGF. Se presentan datos sobre el desarrollo del protocolo investigacional FGFR 360Âş RESISTANCE, la plataforma traslacional implementada para obtener muestras seriadas (biopsias tumorales pre-, en curso y post- tratamiento, plasma e incluso autopsias en caliente) en diferentes momentos de la enfermedad de estos pacientes. Este valioso repositorio de muestras ha servido para generar xenoinjertos derivados de pacientes (PDXs), utilizados en nuestro laboratorio para validar in vivo algunas de las hipĂłtesis previamente observadas, asĂ­ como integrar el conocimiento obtenido del análisis molecular de las muestras más relevantes de algunos respondedores inusuales seleccionados. La presentaciĂłn de cada uno de estos estudios se acompaña de una discusiĂłn de los resultados obtenidos, asĂ­ como un sumario final de las conclusiones alcanzadas y las futuras lĂ­neas de investigaciĂłn asociadas.Among cancers, deregulation of the FGFR:FGF pathway has been widely associated with uncontrolled tumoral growth, aberrant angiogenesis and increased metastatic dissemination rates. In this context, several efforts have been done during the last decade in order to develop targeted agents against this molecular pathway. However, early efficacy data seen with first-generation FGFR inhibitors, namely multi-tyrosin kinase inhibitors (MTKI), has been limited, with a significant proportion of off-target effects. Hence, more potent and selective second-generation FGFR inhibitors (FGFRinh) are currently under development, although there is a lack of consensus regarding which FGFR:FGF molecular alteration could represent a reliable predictive biomarker of response to FGFR blockade. With this thesis project, we aim to contribute in the definition of an optimal therapeutic strategy for patients whose tumours harbour FGFR:FGF pathway alterations, by better selecting those patients more likely to respond to FGFRinh. With this purpose, this thesis encompasses two principal studies. The first study is a retrospective study performed in a cohort of refractory FGFR:FGF-aberrant cancer patients, pre-selected within the molecular pre-screening program at VHIO, and treated with novel FGFRinh in ongoing phase 1 clinical trials. Here we present the biomarker exploratory analysis, statistically correlating the clinic-pathological characteristics of these patients with the efficacy signals observed. The vast majority of these studies were based on the hypothesis that any molecular alteration in the FGFR:FGF pathway could serve as predictive biomarker to FGFRinh, independently of the tumor subtype. However, this retrospective analysis is seeking to identify a subgroup of patients sharing a common molecular alteration of the FGFR:FGF pathway among the responders. Our aim is to delineate more precisely which of these molecular determinants are actually reliable biomarkers and to discern if, in fact, they are at the same level in terms of predicting response in different histological contexts (eg. DNA alterations, amplifications, mutations, translocations- versus mRNA versus protein). The second study instead is a prospective study whose objective is to generate a valuable collection of biological samples from advanced cancer patients, molecularly-selected by their FGFR:FGF gene alterations. Here we present the results of the development of the investigational protocol FGFR 360Âş RESISTANCE, the traslational platform implemented to obtain serial patient samples (tumoral biopsies pre-/on-/post-treatment, blood samples and warm autopsies, if feasible) at different timepoints of their disease. This valuable repository of samples has served to generate patient-derived xenografts (PDXs), used for validating in vivo some of our previous observed hypothesis, as well as to integrate the molecular knowledge acquired from deep-sequencing the most relevant samples from selected unusual responders. The presentation of each one of these two studies is accompanied by a discussion of the results obtained, together with a final summary of the conclusions reached and the future lines of research associated.Universitat Autònoma de Barcelona. Programa de Doctorat en Medicin

    Meteors

    No full text
    Películas ganadoras de los Premios de Vídeo Escolar 1995Contiene dos documentales: Meteoros y La catedral de Tortosa. El primero propone un recorrido gráfico y descriptivo por el mundo de los meteoros. Se describen las clases de nubes, el estudio de la circulación del agua en la atmósfera, la explicación de la lluvia, etc. También se recuerdan los fenómenos que acompañan a las tormentas y la formación de los vientos y sus nombres. Se exponen algunas consecuencias de estos fenómenos atmosféricos. El segundo reportaje presenta la importancia de la ciudad de Tortosa a los largo del tiempo como punto de comunicación tradicional a través del Ebro entre el mar y las tierras de interior. Se refleja la significación histórica de la catedral y se describen sus rasgos arquitectónicos y sus elementos más significativos.CataluñaUniversitat de Barcelona. Biblioteca de Ciències de l'Educació; Passeig de la Vall d'Hebron, 171; 08035 Barcelona; +34934021035; +34934021034;ES

    Targeting KRAS in Lung Cancer Beyond KRAS G12C Inhibitors : The Immune Regulatory Role of KRAS and Novel Therapeutic Strategies

    Get PDF
    Approximately 20% of lung adenocarcinomas harbor KRAS mutations, an oncogene that drives tumorigenesis and has the ability to alter the immune system and the tumor immune microenvironment. While KRAS was considered "undruggable" for decades, specific KRAS G12C covalent inhibitors have recently emerged, although their promising results are limited to a subset of patients. Several other drugs targeting KRAS activation and downstream signaling pathways are currently under investigation in early-phase clinical trials. In addition, KRAS mutations can co-exist with other mutations in significant genes in cancer (e.g., STK11 and KEAP1) which induces tumor heterogeneity and promotes different responses to therapies. This review describes the molecular characterization of KRAS mutant lung cancers from a biologic perspective to its clinical implications. We aim to summarize the tumor heterogeneity of KRAS mutant lung cancers and its immune-regulatory role, to report the efficacy achieved with current immunotherapies, and to overview the therapeutic approaches targeting KRAS mutations besides KRAS G12C inhibitor

    Multicenter Phase I Study of Erdafitinib (JNJ-42756493) Oral Pan-Fibroblast Growth Factor Receptor Inhibitor in Patients with Advanced or Refractory Solid Tumors

    No full text
    Purpose: Here, we report results of the first phase I study of erdafitinib, a potent, oral pan-FGFR inhibitor. Patients and Methods: Patients age >= 18 years with advanced solid tumors for which standard antineoplastic therapy was no longer effective were enrolled (NCT01703481). Parts 2 to 4 employed molecular screening for activating FGFR genomic alterations. In patients with such alterations, two selected doses/schedules identified during part 1 doseescalation [9 mg once daily and 10mg intermittently (7 days on/7 days off), as previously published (Tabernero JCO 2015; 33:3401-8)] were tested. Results: The study included 187 patients. The most common treatment-related adverse events were hyperphosphatemia (64%), dry mouth (42%), and asthenia (28%), generally grade 1/2 severity. All cases of hyperphosphatemia were grade 1/2 except for 1 grade 3 event. Skin, nail, and eye changes were observed in 43%, 33%, and 28% of patients, respectively (mostly grade 1/2 and reversible after temporary dosing interruption). Urothelial carcinoma and cholangiocarcinoma were most responsive to erdafitinib, with objective response rates (ORR) of 46.2% (12/26) and 27.3% (3/11), respectively, in response-evaluable patients with FGFR mutations or fusions. All patients with urothelial carcinoma and cholangiocarcinoma who responded to erdafitinib carried FGFR mutations or fusions. Median response duration was 5.6 months for urothelial carcinoma and 11.4 months for cholangiocarcinoma. ORRs in other tumor types were <10%. Conclusions: Erdafitinib shows tolerability and preliminary clinical activity in advanced solid tumors with genomic changes in the FGFR pathway, at two different dosing schedules and with particularly encouraging responses in urothelial carcinoma and cholangiocarcinoma.Investigational compound erdafitinib (JNJ-42756493) was discovered in collaboration with Astex Pharmaceuticals. The authors thank the patients and their families; the participating centers and investigators; and Vijay Peddareddigari (formerly of Janssen Research & Development) for study conception and design; Jean-Charles Soria, a former primary investigator who contributed to study design and oversaw clinical efforts at Institut Gustave Roussy Cancer Campus and University Paris-Sud, Villejuif, France; Jayaprakash Karkera, Dana Gaffney, Katherine Bell, Gabriela Martinez Cardona, and Joseph Portale of Janssen Research & Development for development of the RT-PCR method used for central FGFR screening; and Suso Platero (formerly of Janssen Research & Development) for translational research support. Editorial support for this publication was provided by Laurie Orloski, funded by Janssen Research & Development. This study was supported by Janssen Research & Development
    corecore