Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38963227

RESUMEN

The electrochemical reduction of nitrate (NO3-) ions to ammonia (NH3) provides an alternative method to eliminate harmful NO3- pollutants in water as well as to produce highly valuable NH3 chemicals. The NH3 yield rate however is still limited to the µmol h-1 cm-2 range when dealing with dilute NO3- concentrations found in waste streams. Copper (Cu) has attracted much attention because of its unique ability to effectively convert NO3- to NH3. We have reported a simple and scalable electrochemical method to produce a Cu foil having its surface covered with a porous Cu nanostructure enriched with (100) facets, which efficiently catalyzes NO3- to NH3. The Cu(100)-rich electrocatalyst showed a very high NH3 production rate of 1.1 mmol h-1 cm-2 in NO3- concentration as low as 14 mM NO3-, which is 4-5 times higher than the best-reported values. Increasing the NO3- concentration (140 mM) resulted in an NH3 production yield rate of 3.34 mmol h-1 cm-2. The durability test conducted for this catalyst foil in a flow cell system showed greater than 100 h stability with a Faradaic efficiency greater than 98%, demonstrating its potential to be used on an industrially relevant scale. Further, density functional theory (DFT) calculations have been performed to understand the better catalytic activity of Cu(100) compared to Cu(111) facets toward NO3-RR.

2.
Nat Cancer ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844817

RESUMEN

Many individuals with cancer are resistant to immunotherapies. Here, we identify the gene encoding the pyrimidine salvage pathway enzyme cytidine deaminase (CDA) among the top upregulated metabolic genes in several immunotherapy-resistant tumors. We show that CDA in cancer cells contributes to the uridine diphosphate (UDP) pool. Extracellular UDP hijacks immunosuppressive tumor-associated macrophages (TAMs) through its receptor P2Y6. Pharmacologic or genetic inhibition of CDA in cancer cells (or P2Y6 in TAMs) disrupts TAM-mediated immunosuppression, promoting cytotoxic T cell entry and susceptibility to anti-programmed cell death protein 1 (anti-PD-1) treatment in resistant pancreatic ductal adenocarcinoma (PDAC) and melanoma models. Conversely, CDA overexpression in CDA-depleted PDACs or anti-PD-1-responsive colorectal tumors or systemic UDP administration (re)establishes resistance. In individuals with PDAC, high CDA levels in cancer cells correlate with increased TAMs, lower cytotoxic T cells and possibly anti-PD-1 resistance. In a pan-cancer single-cell atlas, CDAhigh cancer cells match with T cell cytotoxicity dysfunction and P2RY6high TAMs. Overall, we suggest CDA and P2Y6 as potential targets for cancer immunotherapy.

3.
Nat Med ; 30(6): 1667-1679, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38773341

RESUMEN

An important challenge in the real-world management of patients with advanced clear-cell renal cell carcinoma (aRCC) is determining who might benefit from immune checkpoint blockade (ICB). Here we performed a comprehensive multiomics mapping of aRCC in the context of ICB treatment, involving discovery analyses in a real-world data cohort followed by validation in independent cohorts. We cross-connected bulk-tumor transcriptomes across >1,000 patients with validations at single-cell and spatial resolutions, revealing a patient-specific crosstalk between proinflammatory tumor-associated macrophages and (pre-)exhausted CD8+ T cells that was distinguished by a human leukocyte antigen repertoire with higher preference for tumoral neoantigens. A cross-omics machine learning pipeline helped derive a new tumor transcriptomic footprint of neoantigen-favoring human leukocyte antigen alleles. This machine learning signature correlated with positive outcome following ICB treatment in both real-world data and independent clinical cohorts. In experiments using the RENCA-tumor mouse model, CD40 agonism combined with PD1 blockade potentiated both proinflammatory tumor-associated macrophages and CD8+ T cells, thereby achieving maximal antitumor efficacy relative to other tested regimens. Thus, we present a new multiomics and spatial map of the immune-community architecture that drives ICB response in patients with aRCC.


Asunto(s)
Linfocitos T CD8-positivos , Carcinoma de Células Renales , Antígenos HLA , Inmunoterapia , Neoplasias Renales , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/terapia , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Humanos , Neoplasias Renales/inmunología , Neoplasias Renales/terapia , Neoplasias Renales/genética , Neoplasias Renales/patología , Animales , Inmunoterapia/métodos , Linfocitos T CD8-positivos/inmunología , Ratones , Antígenos HLA/inmunología , Antígenos HLA/genética , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Aprendizaje Automático , Antígenos CD40/inmunología , Antígenos CD40/genética , Macrófagos Asociados a Tumores/inmunología , Transcriptoma , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Femenino
4.
Cell ; 187(11): 2690-2702.e17, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38723627

RESUMEN

The quality and quantity of tumor-infiltrating lymphocytes, particularly CD8+ T cells, are important parameters for the control of tumor growth and response to immunotherapy. Here, we show in murine and human cancers that these parameters exhibit circadian oscillations, driven by both the endogenous circadian clock of leukocytes and rhythmic leukocyte infiltration, which depends on the circadian clock of endothelial cells in the tumor microenvironment. To harness these rhythms therapeutically, we demonstrate that efficacy of chimeric antigen receptor T cell therapy and immune checkpoint blockade can be improved by adjusting the time of treatment during the day. Furthermore, time-of-day-dependent T cell signatures in murine tumor models predict overall survival in patients with melanoma and correlate with response to anti-PD-1 therapy. Our data demonstrate the functional significance of circadian dynamics in the tumor microenvironment and suggest the importance of leveraging these features for improving future clinical trial design and patient care.


Asunto(s)
Linfocitos T CD8-positivos , Inmunoterapia , Linfocitos Infiltrantes de Tumor , Ratones Endogámicos C57BL , Microambiente Tumoral , Animales , Humanos , Ratones , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Relojes Circadianos , Ritmo Circadiano , Células Endoteliales/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Melanoma/terapia , Melanoma/patología , Microambiente Tumoral/inmunología
5.
J Plast Reconstr Aesthet Surg ; 92: 75-78, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38513343

RESUMEN

BACKGROUND: Rhinoplasty enhances facial symmetry and functionality. However, the accurate and reliable quantification of nasal defects pre-surgery remains an ongoing challenge. AIM: This study introduces a novel approach for defect quantification using 2D images and artificial intelligence, providing a tool for better preoperative planning and improved surgical outcomes. MATERIALS AND METHODS: A pre-trained AI model for facial landmark detection was utilised on a dataset of 250 images of male patients aged 18 to 24 who underwent rhinoplasty for cosmetic nasal deformity correction. The analysis concentrated on 36 different distances between the facial landmarks. These distances were normalised using min-max scaling to counter image size and quality variations. Post-normalisation, statistical parameters, including mean, median, and standard deviation, were calculated to identify and quantify nasal defects. RESULTS: The methodology was tested and validated using images from different ethnicities and regions, showing promising potential as a beneficial surgical aid. The normalised data produced reliable quantifications of nasal defects (average 76.2%), aiding in preoperative planning and improving surgical outcomes and patient satisfaction. APPLICATIONS: The developed method can be extended to other facial plastic surgeries. Furthermore, it can be used to create app-based software, assist medical education, and improve patient-doctor communication. CONCLUSION: This novel method for defect quantification in rhinoplasty using AI and image processing holds significant potential in improving surgical planning, outcomes, and patient satisfaction, marking an essential step in the fusion of AI and plastic surgery.


Asunto(s)
Rinoplastia , Humanos , Rinoplastia/métodos , Masculino , Adulto Joven , Adolescente , Puntos Anatómicos de Referencia , Nariz/anomalías , Nariz/cirugía , Cuidados Preoperatorios/métodos , Inteligencia Artificial
6.
Cell Rep Med ; 5(1): 101377, 2024 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-38232703

RESUMEN

Current immunotherapies provide limited benefits against T cell-depleted tumors, calling for therapeutic innovation. Using multi-omics integration of cancer patient data, we predict a type I interferon (IFN) responseHIGH state of dendritic cell (DC) vaccines, with efficacious clinical impact. However, preclinical DC vaccines recapitulating this state by combining immunogenic cancer cell death with induction of type I IFN responses fail to regress mouse tumors lacking T cell infiltrates. Here, in lymph nodes (LNs), instead of activating CD4+/CD8+ T cells, DCs stimulate immunosuppressive programmed death-ligand 1-positive (PD-L1+) LN-associated macrophages (LAMs). Moreover, DC vaccines also stimulate PD-L1+ tumor-associated macrophages (TAMs). This creates two anatomically distinct niches of PD-L1+ macrophages that suppress CD8+ T cells. Accordingly, a combination of PD-L1 blockade with DC vaccines achieves significant tumor regression by depleting PD-L1+ macrophages, suppressing myeloid inflammation, and de-inhibiting effector/stem-like memory T cells. Importantly, clinical DC vaccines also potentiate T cell-suppressive PD-L1+ TAMs in glioblastoma patients. We propose that a multimodal immunotherapy and vaccination regimen is mandatory to overcome T cell-depleted tumors.


Asunto(s)
Glioblastoma , Vacunas , Humanos , Animales , Ratones , Linfocitos T CD8-positivos , Antígeno B7-H1 , Macrófagos , Células Dendríticas , Ganglios Linfáticos/metabolismo , Vacunas/metabolismo
7.
Immunol Rev ; 321(1): 71-93, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37937803

RESUMEN

The cellular stress and immunity cycle is a cornerstone of organismal homeostasis. Stress activates intracellular and intercellular communications within a tissue or organ to initiate adaptive responses aiming to resolve the origin of this stress. If such local measures are unable to ameliorate this stress, then intercellular communications expand toward immune activation with the aim of recruiting immune cells to effectively resolve the situation while executing tissue repair to ameliorate any damage and facilitate homeostasis. This cellular stress-immunity cycle is severely dysregulated in diseased contexts like cancer. On one hand, cancer cells dysregulate the normal cellular stress responses to reorient them toward upholding growth at all costs, even at the expense of organismal integrity and homeostasis. On the other hand, the tumors severely dysregulate or inhibit various components of organismal immunity, for example, by facilitating immunosuppressive tumor landscape, lowering antigenicity, and increasing T-cell dysfunction. In this review we aim to comprehensively discuss the basis behind tumoral dysregulation of cellular stress-immunity cycle. We also offer insights into current understanding of the regulators and deregulators of this cycle and how they can be targeted for conceptualizing successful cancer immunotherapy regimen.


Asunto(s)
Neoplasias , Humanos , Inmunoterapia , Comunicación Celular , Microambiente Tumoral
8.
Cell Discov ; 9(1): 114, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37968259

RESUMEN

CD8+ T cell activation via immune checkpoint blockade (ICB) is successful in microsatellite instable (MSI) colorectal cancer (CRC) patients. By comparison, the success of immunotherapy against microsatellite stable (MSS) CRC is limited. Little is known about the most critical features of CRC CD8+ T cells that together determine the diverse immune landscapes and contrasting ICB responses. Hence, we pursued a deep single cell mapping of CRC CD8+ T cells on transcriptomic and T cell receptor (TCR) repertoire levels in a diverse patient cohort, with additional surface proteome validation. This revealed that CRC CD8+ T cell dynamics are underscored by complex interactions between interferon-γ signaling, tumor reactivity, TCR repertoire, (predicted) TCR antigen-specificities, and environmental cues like gut microbiome or colon tissue-specific 'self-like' features. MSI CRC CD8+ T cells showed tumor-specific activation reminiscent of canonical 'T cell hot' tumors, whereas the MSS CRC CD8+ T cells exhibited tumor unspecific or bystander-like features. This was accompanied by inflammation reminiscent of 'pseudo-T cell hot' tumors. Consequently, MSI and MSS CRC CD8+ T cells showed overlapping phenotypic features that differed dramatically in their TCR antigen-specificities. Given their high discriminating potential for CD8+ T cell features/specificities, we used the single cell tumor-reactive signaling modules in CD8+ T cells to build a bulk tumor transcriptome classification for CRC patients. This "Immune Subtype Classification" (ISC) successfully distinguished various tumoral immune landscapes that showed prognostic value and predicted immunotherapy responses in CRC patients. Thus, we deliver a unique map of CRC CD8+ T cells that drives a novel tumor immune landscape classification, with relevance for immunotherapy decision-making.

9.
Cancer Immunol Res ; 11(12): 1611-1629, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37933083

RESUMEN

Forkhead box P3 (Foxp3)-expressing regulatory T cells (Treg) are the guardians of controlled immune reactions and prevent the development of autoimmune diseases. However, in the tumor context, their increased number suppresses antitumor immune responses, indicating the importance of understanding the mechanisms behind their function and stability. Metabolic reprogramming can affect Foxp3 regulation and, therefore, Treg suppressive function and fitness. Here, we performed a metabolic CRISPR/Cas9 screen and pinpointed novel candidate positive and negative metabolic regulators of Foxp3. Among the positive regulators, we revealed that targeting the GDP-fucose transporter Slc35c1, and more broadly fucosylation (Fuco), in Tregs compromises their proliferation and suppressive function both in vitro and in vivo, leading to alteration of the tumor microenvironment and impaired tumor progression and protumoral immune responses. Pharmacologic inhibition of Fuco dampened tumor immunosuppression mostly by targeting Tregs, thus resulting in reduced tumor growth. In order to substantiate these findings in humans, tumoral Tregs from patients with colorectal cancer were clustered on the basis of the expression of Fuco-related genes. FucoLOW Tregs were found to exhibit a more immunogenic profile compared with FucoHIGH Tregs. Furthermore, an enrichment of a FucoLOW signature, mainly derived from Tregs, correlated with better prognosis and response to immune checkpoint blockade in melanoma patients. In conclusion, Slc35c1-dependent Fuco is able to regulate the suppressive function of Tregs, and measuring its expression in Tregs might pave the way towards a useful biomarker model for patients with cancer. See related Spotlight by Silveria and DuPage, p. 1570.


Asunto(s)
Melanoma , Linfocitos T Reguladores , Humanos , Inmunidad , Tolerancia Inmunológica , Factores de Transcripción Forkhead/genética , Microambiente Tumoral
11.
Genes Immun ; 24(5): 270-279, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37759086

RESUMEN

There is an urgent need for new and better biomarker modalities to estimate the risk of recurrence within the luminal-like breast cancer (BC) population. Molecular diagnostic tests used in the clinic lack accuracy in identifying patients with early luminal BC who are likely to develop metastases. This study provides proof of concept that various liquid biopsy read-outs could serve as valuable candidates to build a multi-modal biomarker model distinguishing, already at diagnosis, between early metastasizing and non-metastasizing patients. All these blood biomarkers (chemokines, microRNAs, leukemia inhibitory factor, osteopontin, and serum-induced functional myeloid signaling responses) can be measured in baseline plasma/serum samples and could be added to the existing prognostic factors to improve risk stratification and more patient-tailored treatment in early luminal BC.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Humanos , Femenino , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Biomarcadores de Tumor
12.
13.
Oncoimmunology ; 12(1): 2219591, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37284695

RESUMEN

Immunogenic cell death (ICD) refers to an immunologically distinct process of regulated cell death that activates, rather than suppresses, innate and adaptive immune responses. Such responses culminate into T cell-driven immunity against antigens derived from dying cancer cells. The potency of ICD is dependent on the immunogenicity of dying cells as defined by the antigenicity of these cells and their ability to expose immunostimulatory molecules like damage-associated molecular patterns (DAMPs) and cytokines like type I interferons (IFNs). Moreover, it is crucial that the host's immune system can adequately detect the antigenicity and adjuvanticity of these dying cells. Over the years, several well-known chemotherapies have been validated as potent ICD inducers, including (but not limited to) anthracyclines, paclitaxels, and oxaliplatin. Such ICD-inducing chemotherapeutic drugs can serve as important combinatorial partners for anti-cancer immunotherapies against highly immuno-resistant tumors. In this Trial Watch, we describe current trends in the preclinical and clinical integration of ICD-inducing chemotherapy in the existing immuno-oncological paradigms.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Muerte Celular , Muerte Celular Inmunogénica , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Citocinas/metabolismo
14.
Sci Transl Med ; 15(691): eadd1016, 2023 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-37043555

RESUMEN

Clinically relevant immunological biomarkers that discriminate between diverse hypofunctional states of tumor-associated CD8+ T cells remain disputed. Using multiomics analysis of CD8+ T cell features across multiple patient cohorts and tumor types, we identified tumor niche-dependent exhausted and other types of hypofunctional CD8+ T cell states. CD8+ T cells in "supportive" niches, like melanoma or lung cancer, exhibited features of tumor reactivity-driven exhaustion (CD8+ TEX). These included a proficient effector memory phenotype, an expanded T cell receptor (TCR) repertoire linked to effector exhaustion signaling, and a cancer-relevant T cell-activating immunopeptidome composed of largely shared cancer antigens or neoantigens. In contrast, "nonsupportive" niches, like glioblastoma, were enriched for features of hypofunctionality distinct from canonical exhaustion. This included immature or insufficiently activated T cell states, high wound healing signatures, nonexpanded TCR repertoires linked to anti-inflammatory signaling, high T cell-recognizable self-epitopes, and an antiproliferative state linked to stress or prodeath responses. In situ spatial mapping of glioblastoma highlighted the prevalence of dysfunctional CD4+:CD8+ T cell interactions, whereas ex vivo single-cell secretome mapping of glioblastoma CD8+ T cells confirmed negligible effector functionality and a promyeloid, wound healing-like chemokine profile. Within immuno-oncology clinical trials, anti-programmed cell death protein 1 (PD-1) immunotherapy facilitated glioblastoma's tolerogenic disparities, whereas dendritic cell (DC) vaccines partly corrected them. Accordingly, recipients of a DC vaccine for glioblastoma had high effector memory CD8+ T cells and evidence of antigen-specific immunity. Collectively, we provide an atlas for assessing different CD8+ T cell hypofunctional states in immunogenic versus nonimmunogenic cancers.


Asunto(s)
Glioblastoma , Neoplasias Pulmonares , Humanos , Linfocitos T CD8-positivos , Glioblastoma/metabolismo , Multiómica , Receptores de Antígenos de Linfocitos T/metabolismo
15.
J Orthop ; 37: 46-52, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36974096

RESUMEN

Objective: To evaluate and compare clinical efficacy and effect on specific serum biomarker with serial injections of growth factor concentrate (GFC) for knee osteoarthritis (KOA) in a randomized triple blinded placebo controlled interventional study. Methods: Final assessment was done on 58 patients. Patients with Kellgren-Lawrence grade II, III knee osteoarthritis were administered monthly intraarticular injections(3 injections) of GFC(n = 31) or saline(n = 27) and evaluated clinically with visual analogue scale(VAS) and Western Ontario and McMaster Universities Arthritis Index(WOMAC) at 3,6 and 12 months post therapy. Biochemical analysis was done with serum biomarker of cartilage degeneration, Collagen 2-1 (Coll2-1), estimated at baseline and at final follow up. Results: Both the groups exhibited statistically significant improvements (P < 0.05) in VAS at 3,6 and 12 months. WOMAC improvement reached statistical significance for GFC group at every evaluation (P < 0.001) but only at 12 months in NS group (P = 0.029). The improvements were clinically meaningful only in GFC group throughout follow up (Minimal clinically important differences >12% of baseline in WOMAC and >2 cm difference in mean for VAS). Intergroup comparison revealed GFC to be much better for both scores at every evaluation (95% CI of 0.2-1.5,[P = 0.008], 1.4-2.9,[P < 0.0001], and 2.7-4.2,[P < 0.0001] for VAS, 7.3-16.0 [P < 0.001], 11.6-21.9 [P < 0.001] and 18.1-31.1[P < 0.001] for WOMAC). Statistically significant decrease in serum Coll2-1 levels were observed for GFC group only. No serious complications were seen. Conclusion: Serial(three) monthly GFC injections result in clinically meaningful improvement of subjective pain and function outcome scores, sustaining up to 12 months in KOA grade II and III. GFC also lead to significant reduction in serum levels of cartilage degradation biomarker coll2-1.

16.
Nat Commun ; 14(1): 217, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36639382

RESUMEN

Despite their low abundance in the tumor microenvironment (TME), classical type 1 dendritic cells (cDC1) play a pivotal role in anti-cancer immunity, and their abundance positively correlates with patient survival. However, their interaction with CD4+ T-cells to potentially enable the cytotoxic T lymphocyte (CTL) response has not been elucidated. Here we show that contact with activated CD4+ T-cells enables human ex vivo cDC1, but no other DC types, to induce a CTL response to cell-associated tumor antigens. Single cell transcriptomics reveals that CD4+ T-cell help uniquely optimizes cDC1 in many functions that support antigen cross-presentation and T-cell priming, while these changes don't apply to other DC types. We robustly identify "helped" cDC1 in the TME of a multitude of human cancer types by the overlap in their transcriptomic signature with that of recently defined, tumor-infiltrating DC states that prove to be positively prognostic. As predicted from the functional effects of CD4+ T-cell help, the transcriptomic signature of "helped" cDC1 correlates with tumor infiltration by CTLs and Thelper(h)-1 cells, overall survival and response to PD-1-targeting immunotherapy. These findings reveal a critical role for CD4+ T-cell help in enabling cDC1 function in the TME and may establish the helped cDC1 transcriptomic signature as diagnostic marker in cancer.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Neoplasias/metabolismo , Presentación de Antígeno , Linfocitos T Citotóxicos , Células Dendríticas , Linfocitos T Colaboradores-Inductores/metabolismo , Microambiente Tumoral
17.
Immunol Cell Biol ; 101(2): 112-129, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36479949

RESUMEN

FOXP3-expressing regulatory T cells (Treg ) are indispensable for immune homeostasis and tolerance, and in addition tissue-resident Treg have been found to perform noncanonical, tissue-specific functions. For optimal tolerogenic function during inflammatory disease, Treg are equipped with mechanisms that assure lineage stability. Treg lineage stability is closely linked to the installation and maintenance of a lineage-specific epigenetic landscape, specifically a Treg -specific DNA demethylation pattern. At the same time, for local and directed immune regulation Treg must possess a level of functional plasticity that requires them to partially acquire T helper cell (TH ) transcriptional programs-then referred to as TH -like Treg . Unleashing TH programs in Treg , however, is not without risk and may threaten the epigenetic stability of Treg with consequently pathogenic ex-Treg contributing to (auto-) inflammatory conditions. Here, we review how the Treg -stabilizing epigenetic landscape is installed and maintained, and further discuss the development, necessity and lineage instability risks of TH 1-, TH 2-, TH 17-like Treg and follicular Treg .


Asunto(s)
Tolerancia Inmunológica , Linfocitos T Reguladores , Factores de Transcripción Forkhead
18.
J Med Phys ; 48(4): 338-344, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38223791

RESUMEN

Introduction: Conventionally, the fattened beam is being used in radiotherapy for routine clinical cases even after introduction of intensity-modulated radiotherapy with incorporation of multi-leaf collimator system. With the removal of the flattening filter (FF) from the beam's path average energy of the photon gets reduced resulting in reduced scatter, reduction in treatment time, and reduced neutron contamination for high-energy beam and ultimately resulting in treatment plan quality deviations. This study aims to investigate the usefulness of the FF-free (FFF) beam for routine head-and-neck cancer (HNC) cases treated with volumetric arc therapy (VMAT) and dosimetrically compares the result with the FF beam. Materials and Methods: In this study, 20 patients treated on HalcyonTM (unmatched 6 megavoltage [6MV] FFF beam) medical linear accelerator with VMAT of different HNC selected and for comparison with 6MV FF beam, 20 equivalent treatment plans are created for TruebeamTM configuration and the plans have been evaluated for target coverage, doses to the organ at risk (OAR), and other dose quality indices. Results: Comparable target coverage, doses to OARs except for rest right parotid (P = 0.02) between 6MV FFF beam and 6MV FF beam is observed. Insignificant differences in conformity index, homogeneity index, and gradient index have been observed. Higher monitor unit (MU) (P ≤ 0.001) and lesser beam on time (BOT) (P = 0.003) have been observed in 6MV FFF. Conclusion: 6MV FFF beam provides comparable target coverage and improved dose-sparing effect to most of the OARs. 6MV FFF beam has lesser BOT, but on the other hand number of MUs is higher as compared to 6MV-FF plans.

19.
Cells ; 11(23)2022 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-36497148

RESUMEN

Tumour-associated macrophages (TAMs) are essential players in the tumour microenvironment (TME) and modulate various pro-tumorigenic functions such as immunosuppression, angiogenesis, cancer cell proliferation, invasion and metastasis, along with resistance to anti-cancer therapies. TAMs also mediate important anti-tumour functions and can clear dying cancer cells via efferocytosis. Thus, not surprisingly, TAMs exhibit heterogeneous activities and functional plasticity depending on the type and context of cancer cell death that they are faced with. This ultimately governs both the pro-tumorigenic and anti-tumorigenic activity of TAMs, making the interface between TAMs and dying cancer cells very important for modulating cancer growth and the efficacy of chemo-radiotherapy or immunotherapy. In this review, we discuss the interface of TAMs with cancer cell death from the perspectives of cell death pathways, TME-driven variations, TAM heterogeneity and cell-death-inducing anti-cancer therapies. We believe that a better understanding of how dying cancer cells influence TAMs can lead to improved combinatorial anti-cancer therapies, especially in combination with TAM-targeting immunotherapies.


Asunto(s)
Neoplasias , Macrófagos Asociados a Tumores , Humanos , Macrófagos/metabolismo , Microambiente Tumoral , Neoplasias/metabolismo , Inmunoterapia
20.
Cell Death Dis ; 13(12): 1062, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36539408

RESUMEN

Gliomas, the most frequent type of primary tumor of the central nervous system in adults, results in significant morbidity and mortality. Despite the development of novel, complex, multidisciplinary, and targeted therapies, glioma therapy has not progressed much over the last decades. Therefore, there is an urgent need to develop novel patient-adjusted immunotherapies that actively stimulate antitumor T cells, generate long-term memory, and result in significant clinical benefits. This work aimed to investigate the efficacy and molecular mechanism of dendritic cell (DC) vaccines loaded with glioma cells undergoing immunogenic cell death (ICD) induced by photosens-based photodynamic therapy (PS-PDT) and to identify reliable prognostic gene signatures for predicting the overall survival of patients. Analysis of the transcriptional program of the ICD-based DC vaccine led to the identification of robust induction of Th17 signature when used as a vaccine. These DCs demonstrate retinoic acid receptor-related orphan receptor-γt dependent efficacy in an orthotopic mouse model. Moreover, comparative analysis of the transcriptome program of the ICD-based DC vaccine with transcriptome data from the TCGA-LGG dataset identified a four-gene signature (CFH, GALNT3, SMC4, VAV3) associated with overall survival of glioma patients. This model was validated on overall survival of CGGA-LGG, TCGA-GBM, and CGGA-GBM datasets to determine whether it has a similar prognostic value. To that end, the sensitivity and specificity of the prognostic model for predicting overall survival were evaluated by calculating the area under the curve of the time-dependent receiver operating characteristic curve. The values of area under the curve for TCGA-LGG, CGGA-LGG, TCGA-GBM, and CGGA-GBM for predicting five-year survival rates were, respectively, 0.75, 0.73, 0.9, and 0.69. These data open attractive prospects for improving glioma therapy by employing ICD and PS-PDT-based DC vaccines to induce Th17 immunity and to use this prognostic model to predict the overall survival of glioma patients.


Asunto(s)
Neoplasias Encefálicas , Glioma , Fotoquimioterapia , Animales , Ratones , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patología , Glioma/genética , Glioma/terapia , Glioma/patología , Transcriptoma , Sistema Nervioso Central/patología , Proteínas Cromosómicas no Histona/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...