Year Year arrow
arrow-active-down-0
Publisher Publisher arrow
arrow-active-down-1
Journal Journal arrow
arrow-active-down-2
Institution
1
Institution arrow
arrow-active-down-3
Institution Country Institution Country arrow
arrow-active-down-4
Publication Type Publication Type arrow
arrow-active-down-5
Field Of Study Field Of Study arrow
arrow-active-down-6
Topics Topics arrow
arrow-active-down-7
Open Access Open Access arrow
arrow-active-down-8
Language Language arrow
arrow-active-down-9
Filter Icon Filter 1
Year Year arrow
arrow-active-down-0
Publisher Publisher arrow
arrow-active-down-1
Journal Journal arrow
arrow-active-down-2
Institution
1
Institution arrow
arrow-active-down-3
Institution Country Institution Country arrow
arrow-active-down-4
Publication Type Publication Type arrow
arrow-active-down-5
Field Of Study Field Of Study arrow
arrow-active-down-6
Topics Topics arrow
arrow-active-down-7
Open Access Open Access arrow
arrow-active-down-8
Language Language arrow
arrow-active-down-9
Filter Icon Filter 1
Export
Sort by: Relevance
Abstract 33: Development of a humanized mouse model for γδT cells

Unconventional T cells such as gamma delta (γδ) T cells have garnered significant interest in cancer immunotherapy due to their potent anti-tumor properties including strong cytotoxic capabilities in hematological malignancies and their ability to recognize a broad range of tumor antigens independent of the classical major histocompatibility complex (MHC) molecules. Given the interest in engaging these immune cell populations in cancer therapy, we established a new in vivo humanized mouse model through the adoptive transfer of human γδT cells to support this emerging area of therapeutic interest. This model was created by transplanting γδT cells sorted from PBMC donors into Fc-resolv huIL15 NOG (functional knock-out of murine Fcγ receptors to minimize the confounding impact of mouse immune cells) mice, genetically engineered to express human IL-15. We demonstrate that various γδT cell populations, including Vδ1 and Vδ2, can be successfully maintained over a long period of time, preserving the expression of critical functional biomarkers such as NKG2D, CD69 or CD16, under various engraftment conditions. Additionally, we showed that hu γδT cells display functional engagement in vivo, achieving high efficacy with epcoritamab (CD20-CD3 Ab) against disseminated tumors in the hu γδT cell adoptive transfer mouse model. Citation Format: Lydia Blot, Lopes Joaquim, Emerson Serres, Vanessa Croize, Hervé Barret, Pauline Rettman, Clément Barjon, Dorothee Bourges, Antoine Lhermitte, Valéria Fantin, Sukhvinder Sidhu, Loreley Calvet. Development of a humanized mouse model for γδT cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2025; Part 1 (Regular Abstracts); 2025 Apr 25-30; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2025;85(8_Suppl_1):Abstract nr 33.

Read full abstract
Relevant
Cite IconCite
Chat PDF IconChat PDF
Save
Abstract B029: Forging strong bidirectional preclinical and translational research for optimized drug development of Sym023 (S095018), a novel anti-TIM3 antibody

Abstract Immune checkpoint inhibitors targeting TIM3, a molecule frequently co-expressed with PD1 on exhausted T cells and myeloid cells, represent a promising approach in cancer immunotherapy. Inhibition of TIM3 in myeloid cells promotes their activation and has been shown to enhance anti-tumor responses in both monotherapy and combination with immunotherapy or chemotherapy in mouse models (PMID: 34108686). Sym023 (S095018) offers a novel strategy for targeting TIM3 with distinct properties. Beyond blocking the binding of galectin 9 and phosphatidylserine to TIM3, our preclinical studies demonstrate that Sym023 has a direct effect on dendritic cells, inducing their maturation and triggering downstream activation of T cells. In mouse models, the combination of Sym023 (S095018) with an anti-PD1 antibody significantly improved survival and tumor control. Based on these findings and existing literature, the safety and efficacy of the combination of Sym023 with anti-PD1 (sym021) was evaluated in a phase Ib/II expansion study (Sym021-02; NCT04641871) in biliary tract cancer (BTC) patients. An in-depth biomarker plan was implemented to analyze biopsies at screening and the end of cycle 1 (EOC1). On 19 tumor biopsies, 7-plex immunohistochemistry (IHC) and transcriptomic profiling (Nanostring) were performed to study the changes in immune infiltrates during the treatment course. The combination of Sym023 and Sym021 led to increased levels of IFNg and myeloid chemokines (e.g., CXCL9, CXCL10). Treatment resulted in a higher density of activated T cells within the tumor islets, along with the upregulation of cytotoxicity-related genes. Transcriptomic profiling further revealed an upregulation of antigen processing pathways, supporting the translatability of Sym023’s mechanism of action on both T cells and myeloid cells in patients. Drawing from preclinical and biomarker data from the Sym021-02 trial, a biomarker-driven analysis was conducted to identify patients most likely to respond to the Sym023 and anti-PD1 combination. Through this translational analysis, non-small cell lung cancer (NSCLC) was identified as having significant infiltration by myeloid cells and CD8+ T cells, along with high intra-tumoral TIM3 expression, positioning NSCLC as a relevant and promising indication for this combination therapy. A platform trial (NCT06162572) featuring an arm combining cemiplimab (anti-PD1) with Sym023 (S095018) was launched for first-line treatment of patients with advanced/metastatic NSCLC with high PDL1 expression. Citation Format: Julia Geronimi, Aditi Varthaman, Trine Lindsted, Franziska Uhlenbrock, Camilla Frölich, Vincent Lombardi, Christelle Cousin, Mathieu Boucher, Audrey Delmas, Pauline Darcel, Xenophon Ianopulos, Hélène Kaplon. Forging strong bidirectional preclinical and translational research for optimized drug development of Sym023 (S095018), a novel anti-TIM3 antibody [abstract]. In: Proceedings of the AACR IO Conference: Discovery and Innovation in Cancer Immunology: Revolutionizing Treatment through Immunotherapy; 2025 Feb 23-26; Los Angeles, CA. Philadelphia (PA): AACR; Cancer Immunol Res 2025;13(2 Suppl):Abstract nr B029.

Read full abstract
Relevant
Cite IconCite
Chat PDF IconChat PDF
Save
ORPHAN GPCRS – A NEW FRONTIER IN SCHIZOPHRENIA DRUG DISCOVERY

BackgroundSchizophrenia manifests as a broad and diverse symptomatology that creates a heterogenous patient population that responds to standard of care medicines to varying extents. The three characterised symptom domains; positive, negative and cognitive, represent, but are not limited to: hallucinations and delusions, negative affect, and impairments in learning and memory, respectively. Frontline drugs only effectively address the positive symptoms in ~70% of patients, yet it is the cognitive impairments associated with schizophrenia (CIAS) that pose the greatest hurdle to improve societal integration and quality of life. These issues culminate in a critical unmet medical need. There have been a number of clinical candidates and mechanisms that have sought to address CIAS, and all have failed. These failures point to two probable influences: 1. insufficient insight into the mechanisms capable of driving a change in disease symptoms; 2. the lack of stringent preclinical models and assays that derive endpoints similar to those tested in patients. Whilst schizophrenia standard of care medicines and investigational agents engage a number of G protein-coupled receptors (GPCRs), there remains an array of CNS-enriched orphan GPCRs that represent new opportunities as drug targets (Lu et al., 2023).ObjectiveOur approach employs a holistic workflow to target validation and drug discovery through applying techniques that accelerates the process whilst increasing fidelity. We have applied structure- enabled drug design, disease-relevant pharmacology and advanced rodent models to multiple orphan GPCRs to create a global understanding of the target ranging from ligand-receptor interactions to in- depth behavioural insights.MethodWe used cryogenic electron microscopy to generate molecular models of our target orphan GPCRs in complex with their cognate G proteins to gain molecular insights into ligand binding and G protein coupling – facilitating structure-based drug design. We applied multi-endpoint pharmacology in recombinant and primary native cells to provide granularity to ligand-mediated signalling sequalae. Finally, we overlayed these findings with a comprehensive behavioural and cognitive battery (including rodent cognition touchscreens) to provide a correlation between our ligand-receptor mechanisms and psychosis- and cognition-relevant in vivo behavioural outcomes.ResultsApplication of our technical workflow has provided molecular insights to accelerate ligand optimisation and deliver ligands with distinct, signal-biased pharmacological profiles. These ligands demonstrated antipsychotic activity (reversal of hyperlocomotion) and distinct cognition-enhancing profiles in mice, across working memory and attention tasks.ConclusionsDevelopment of this unique workflow has allowed the deconvolving of mechanisms of ligand-receptor interactions and signalling that accelerated development of ligands that target orphan GPCRs and display antipsychotic and pro-cognitive effects in mice.ReferencesLu Y. 2023. Molecular insights into orphan G protein-coupled receptors relevant to schizophrenia. Br J Pharmacol. doi: 10.1111/bph.16221

Read full abstract
Open Access Icon Open Access
Relevant
Cite IconCite
Chat PDF IconChat PDF
Save
Human liver cell-based assays for the prediction of hepatic bile acid efflux transporter inhibition by drugs

ABSTRACT Introduction Drug-mediated inhibition of bile salt efflux transporters may cause liver injury. In vitro prediction of drug effects toward canalicular and/or sinusoidal efflux of bile salts from human hepatocytes is therefore a major issue, which can be addressed using liver cell-based assays. Area covered This review, based on a thorough literature search in the scientific databases PubMed and Web of Science, provides key information about hepatic transporters implicated in bile salt efflux, the human liver cell models available for investigating functional inhibition of bile salt efflux, the different methodologies used for this purpose, and the modes of expression of the results. Applications of the assays to drugs are summarized, with special emphasis to the performance values of some assays for predicting hepatotoxicity/cholestatic effects of drugs. Expert opinion Human liver cell-based assays for evaluating drug-mediated inhibition of bile acid efflux transporters face various limitations, such as the lack of method standardization and validation, the present poor adaptability to high throughput approaches, and some pitfalls with respect to interpretation of bile acid biliary excretion indexes. Hepatotoxicity of drugs is additionally likely multifactorial, highlighting that inhibition of hepatic bile salt efflux by drugs provides important, but not full, information about potential drug hepatotoxicity.

Read full abstract
Relevant
Cite IconCite
Chat PDF IconChat PDF
Save
Simplified Approach to Managing Newly Diagnosed Patients with Mild-to-Moderate Hypertension in Routine Clinical Practice.

The aim of the observational SIMPLE study was to assess real-life effectiveness and safety of a single-pill combination (SPC) of perindopril arginine/amlodipine in a broad range of subjects with newly diagnosed mild-to-moderate hypertension treated in Canadian general practice. Treatment-naïve participants aged 18-65years with mild-to-moderate hypertension, whose physicians decided to initiate the perindopril/amlodipine SPC, were recruited from Canadian clinical practice from October 2017 to February 2019. Participants were followed at 3- (M3) and 6-month (M6) visits after treatment initiation. The recommended starting dose of 3.5mg/2.5mg once daily was up-titrated, at the discretion of the treating physician, if blood pressure (BP) remained above target at subsequent visits. The primary endpoint was change in mean office systolic BP (SBP) and diastolic BP (DBP) at M6. The full analysis set included 1179 participants with a mean age of 51.5±8.7years; 61% were male. Mean SBP/DBP at baseline was 153.4±11.5/94.8±7.7mmHg. Treatment was initiated at 3.5mg/2.5mg in 76.0% participants. Over the 6-month treatment period, significant (P < 0.001) decreases from baseline were observed for SBP (-22.9±14.5mmHg) and DBP (-13.4±9.1mmHg), with 70.2% of participants achieving their BP target. Across all perindopril/amlodipine SPC dose groups, 61.4% of participants achieved BP targets at M3; mean SBP was reduced by 20.8±14.7mmHg and DBP by 11.7±9.2mmHg (both P < 0.001). Analysis by baseline subgroup revealed significant BP reductions across age groups, sex, hypertension grades, and diabetes status. Participants with Grade 2 hypertension had a significantly greater decrease than those with Grade 1 (P < 0.001). Treatment with the SPC was well tolerated, and in the 6.1% with treatment-related adverse events, the majority were mild to moderate. High (99%) self-reported adherence (< 20 missed doses) in the 49.4% with available data and high physician satisfaction with treatment (82%) were reported. Data from routine Canadian clinical practice indicate that a perindopril/amlodipine SPC is associated with significant BP reductions from baseline in a broad range of participants with different cardiovascular risk factors and may represent an appropriate first-line treatment for subjects with newly diagnosed hypertension.

Read full abstract
Open Access Icon Open Access
Relevant
Cite IconCite
Chat PDF IconChat PDF
Save
The combined use of scRNA-seq and network propagation highlights key features of pan-cancer Tumor-Infiltrating T cells

Improving the selectivity and effectiveness of drugs represents a crucial issue for future therapeutic developments in immuno-oncology. Traditional bulk transcriptomics faces limitations in this context for the early phase of target discovery as resulting gene expression levels represent the average measure from multiple cell populations. Alternatively, single cell RNA sequencing can dive into unique cell populations transcriptome, facilitating the identification of specific targets. Here, we generated Tumor-Infiltrating regulatory T cells (TI-Tregs) and exhausted T cells (Tex) gene signatures from a single cell RNA-seq pan-cancer T cell atlas. To overcome noise and sparsity inherent to single cell transcriptomics, we then propagated the gene signatures by diffusion in a protein-protein interaction network using the Patrimony high-throughput computing platform. This methodology enabled the refining of signatures by rescoring genes based on their biological connectivity and shed light not only on processes characteristics of TI-Treg and Tex development and functions but also on their immunometabolic specificities. The combined use of single cell transcriptomics and network propagation may thus represent an innovative and effective methodology for the characterization of cell populations of interest and eventually the development of new therapeutic strategies in immuno-oncology.

Read full abstract
Open Access Icon Open Access
Relevant
Cite IconCite
Chat PDF IconChat PDF
Save
Russian multicenter observational HDQ study on the validation of the questionnaire for hemorrhoidal disease

AIM: to develop and validate hemorrhoidal disease (HD) specific national questionnaire.MATERIALS AND METHODS: the Russian questionnaire, which included questions on the most typical HD symptoms, was developed by leading national specialists-proctologists. The regression analysis was used to evaluate linear and nonlinear relationships between the fact of the HD presence, objectively confirmed by a doctor, and the patients’ answers to the questions from the questionnaire. The final version of the questionnaire included only questions regarding clinical characteristics typical for hemorrhoids that were significantly associated with the presence of the disease. Validation of the questionnaire was performed on both training and test samples of patients to determine its sensitivity (Se) and specificity (Sp) in identifying patients with HD.RESULTS: initially, 7 main domains reflecting the presence and severity of symptoms typical for HD were included in the questionnaire. A training sample of patients was used to search for linear and nonlinear relationships between objectively confirmed hemorrhoids and responses to questions in the initial questionnaire. The final version of the questionnaire, consisting only of questions regarding symptoms that have demonstrated to be significant predictors of hemorrhoids, was validated on a training sample (Se = 86.5%, Sp = 73.4%, and the probability of identifying a patient with HD, according to Bayes’ theorem = 66.6%). After the developed version of the questionnaire was validated on a test sample, the final screening questionnaire for HD demonstrated Se 83.7%, Sp 74.1%, and the probability of identifying patients with HD with a positive questionnaire result, calculated according to Bayes’ theorem, was 67%.CONCLUSION: the first national questionnaire for screening HD has diagnostic characteristics indicating its high accuracy in identifying patients with HD and allowing it to be recommended for use in the general population.

Read full abstract
Relevant
Cite IconCite
Chat PDF IconChat PDF
Save