In a study of neoantigen-specific T cell therapeutic efficacy, a cellular therapy model involving activated MISTIC T cells and interleukin 2 was utilized in lymphodepleted mice with tumors. Our comprehensive approach to understanding treatment response involved employing flow cytometry, single-cell RNA sequencing, and a concurrent whole-exome and RNA sequencing analysis.
Characterizing the isolated 311C TCR revealed a high affinity for mImp3, yet a complete absence of cross-reactivity with wild-type molecules. The MISTIC mouse was engineered to furnish a reservoir of mImp3-specific T cells. The infusion of activated MISTIC T cells, part of an adoptive cellular therapy model, caused rapid intratumoral infiltration and remarkably potent antitumor effects, ultimately leading to long-term cures in a majority of GL261-bearing mice. Retained neoantigen expression was evident in the subset of mice that failed to respond to adoptive cell therapy, accompanied by intratumoral MISTIC T-cell dysfunction. MISTIC T cell therapy's effectiveness was diminished in mice harboring tumors exhibiting diverse mImp3 expression, illustrating the obstacles to precision treatment in human tumors of a mixed lineage.
In a preclinical glioma model, we developed and characterized the first TCR transgenic targeting an endogenous neoantigen, revealing the therapeutic promise of adoptively transferred neoantigen-specific T cells. For research into anti-tumor T-cell responses in glioblastoma, both fundamentally and translationally, the MISTIC mouse offers a robust, novel platform.
A preclinical glioma model hosted the generation and characterization of the first TCR transgenic against an endogenous neoantigen. We then validated the therapeutic potential of neoantigen-specific T cells, which were adoptively transferred. A powerful and novel platform, the MISTIC mouse, enables basic and translational research on antitumor T-cell responses within glioblastoma.
Unfortunately, some patients diagnosed with locally advanced/metastatic non-small cell lung cancer (NSCLC) experience a poor outcome when treated with anti-programmed cell death protein 1 (PD-1)/anti-programmed death-ligand 1 (PD-L1) therapies. Combining this agent with complementary agents could yield better results. A multicenter phase 1b open-label trial investigated the concurrent use of sitravatinib, a spectrum-selective tyrosine kinase inhibitor, and the anti-PD-1 antibody, tislelizumab.
Enrolled in the study were patients with locally advanced or metastatic NSCLC, specifically Cohorts A, B, F, H, and I, each containing 22 to 24 participants (N=22-24). The A and F cohorts comprised patients who had been given systemic therapy prior to study enrollment, demonstrating anti-PD-(L)1 resistance/refractoriness in either non-squamous (cohort A) or squamous (cohort F) disease. Cohort B's patient population comprised individuals who had received prior systemic therapy, presenting with anti-PD-(L)1-naive non-squamous disease. Prior systemic therapy for metastatic disease and anti-PD-(L)1/immunotherapy were absent in patients from cohorts H and I, who further exhibited PD-L1-positive non-squamous (cohort H) or squamous (cohort I) tissue types. Patients were administered sitravatinib 120mg orally, once daily, in conjunction with tislelizumab 200mg intravenously, every three weeks, up to study termination, disease advancement, unacceptable toxicity, or death. In all treated patients (N=122), the safety and tolerability profile formed the primary endpoint. Progression-free survival (PFS), and investigator-assessed tumor responses were secondary endpoints evaluated in the study.
The median duration of observation was 109 months, with a spread from a minimum of 4 months to a maximum of 306 months. IU1 Treatment-related adverse events (TRAEs) were observed in a high percentage, 984%, of patients, and 516% of them experienced Grade 3 TRAEs. Discontinuation of either medication, due to TRAEs, occurred in 230% of the patient population. In cohorts A, F, B, H, and I, the response rates were as follows: 87% (n=2/N=23, 95% confidence interval: 11% to 280%), 182% (n=4/N=22, 95% CI: 52% to 403%), 238% (n=5/N=21, 95% CI: 82% to 472%), 571% (n=12/N=21, 95% CI: 340% to 782%), and 304% (n=7/N=23, 95% CI: 132% to 529%), respectively. Cohort A failed to demonstrate a median response duration, whereas other cohorts displayed response times varying from 69 to 179 months. The percentage of patients achieving disease control spanned a remarkable range of 783% to 909%. Cohort A demonstrated a median PFS of 42 months, while cohort H exhibited a median PFS of 111 months, highlighting substantial differences in treatment efficacy.
Among patients diagnosed with locally advanced or metastatic non-small cell lung cancer (NSCLC), the combination of sitravatinib and tislelizumab demonstrated a generally well-tolerated treatment regimen, presenting no new safety concerns and maintaining safety profiles in line with the established safety characteristics of these individual therapies. Objective responses were consistently found in every studied cohort, notably including patients unexposed to systemic or anti-PD-(L)1 therapies, or individuals with anti-PD-(L)1-resistant/refractory disease. Subsequent investigation in specific NSCLC populations is suggested based on the supporting findings.
NCT03666143: A summary of the study.
The significance of NCT03666143 is of interest.
For patients with relapsed/refractory B-cell acute lymphoblastic leukemia, murine chimeric antigen receptor T (CAR-T) cell therapy has shown positive clinical effects. Nevertheless, the potential for the murine single-chain variable fragment domain to elicit an immune response might hinder the long-term survival of CAR-T cells, potentially causing a relapse.
The safety and effectiveness of autologous and allogeneic humanized CD19-targeted CAR-T cells (hCART19) were assessed in a clinical trial of patients with relapsed/refractory B-cell acute lymphoblastic leukemia (R/R B-ALL). Fifty-eight patients, aged between 13 and 74 years, participated in and received treatment between February 2020 and March 2022. Key performance indicators for the analysis included complete remission (CR) rate, overall survival (OS), event-free survival (EFS), and safety.
By day 28, a remarkable 931% (54 out of 58) of patients achieved a complete remission (CR) or complete remission with incomplete count recovery (CRi), with 53 displaying minimal residual disease negativity. Over a median follow-up duration of 135 months, the estimated one-year overall survival and event-free survival rates were calculated as 736% (95% confidence interval: 621% to 874%) and 460% (95% confidence interval: 337% to 628%), respectively. The median overall survival and event-free survival times were 215 months and 95 months, respectively. Analysis revealed no substantial enhancement in human antimouse antibodies post-infusion (p=0.78). In the blood, B-cell aplasia persisted for a duration of 616 days, demonstrating a longer timeframe than observed in our preceding mCART19 trial. The reversible nature of toxicities extended to severe cytokine release syndrome, occurring in 36% (21 out of 58) of patients, and severe neurotoxicity, observed in 5% (3 patients from 58). In contrast to the prior mCART19 trial, patients receiving hCART19 demonstrated prolonged event-free survival without a concomitant rise in toxicity. In addition, our findings suggest that patients who completed consolidation therapy, including allogeneic hematopoietic stem cell transplants or CD22-targeted CAR-T cell treatments following hCART19 therapy, exhibited a greater event-free survival (EFS) duration compared to patients without such consolidation therapy.
R/R B-ALL patients demonstrate that hCART19 exhibits favorable short-term effectiveness and manageable toxicity.
An important clinical trial, NCT04532268, merits attention.
Regarding the clinical trial NCT04532268.
The ubiquitous phenomenon of phonon softening in condensed matter systems is frequently accompanied by charge density wave (CDW) instabilities and anharmonicity. BioMonitor 2 Superconductivity, charge density waves, and phonon softening exhibit a complex interplay that is a subject of vigorous discussion. This work examines the consequences of anomalous soft phonon instabilities on superconductivity, based on a recently developed theoretical framework that considers phonon damping and softening within the Migdal-Eliashberg theory. From model calculations, a sharp dip in the phonon dispersion relation, either acoustic or optical (including the occurrence of Kohn anomalies, frequently linked to CDWs), signifies phonon softening and thus leads to a substantial increase in the electron-phonon coupling constant. This, in alignment with the optimal frequency concept of Bergmann and Rainer, can under certain conditions, produce a substantial increase in the superconducting transition temperature Tc. In short, our data supports the possibility that high-temperature superconductivity may be attainable through the use of momentum-confined soft phonon anomalies.
As a second-line treatment for acromegaly, Pasireotide long-acting release (LAR) has received regulatory approval. For patients with uncontrolled IGF-I levels, a starting dose of 40mg of pasireotide LAR administered every four weeks is recommended, with a possible subsequent increase to 60mg monthly. Genetics behavioural Three patients benefiting from a pasireotide LAR de-escalation strategy are showcased in this presentation. Every 28 days, a 61-year-old female patient with resistant acromegaly was given pasireotide LAR 60mg as a treatment. A reduction in pasireotide LAR therapy, starting at 40mg and diminishing to 20mg, occurred upon IGF-I's entry into the lower age range. During 2021 and 2022, IGF-I levels maintained a consistent position inside the normal range. Three neurosurgical procedures were undertaken on a 40-year-old female patient, whose acromegaly proved resistant to treatment. As part of the PAOLA study in 2011, she received pasireotide LAR 60mg as a treatment. In light of the sustained IGF-I overcontrol and radiological stability, a dosage reduction of the therapy to 40mg was implemented in 2016, followed by a further decrease to 20mg in 2019. The patient's hyperglycemia was addressed through the administration of metformin. In 2011, a 37-year-old male patient, struggling with resistant acromegaly, underwent treatment with pasireotide LAR 60mg. Therapy dosage was adjusted downward to 40mg in 2018, a consequence of managing IGF-I levels excessively, and subsequently reduced to 20mg in 2022.