Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 13.255
Filtrer
1.
Head Neck Pathol ; 18(1): 73, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39110300

RÉSUMÉ

PURPOSE: Our aim was to assess the ability of simultaneous immunohistochemical staining (IHC) for p16 and p53 to accurately subclassify head and neck squamous cell carcinomas (HNSCC) as HPV-associated (HPV-A) versus HPV-independent (HPV-I) and compare p53 IHC staining patterns to TP53 mutation status, p16 IHC positivity and HPV status. METHODS: We stained 31 HNSCCs for p53 and p16, and performed next-generation sequencing (FoundationOne©CDx) on all cases and HPV in-situ hybridization (ISH) when sufficient tissue was available (n = 23). p53 IHC staining patterns were assessed as wildtype (wt) or abnormal (abn) patterns i.e. overexpression, null or cytoplasmic staining. RESULTS: In a majority of cases (28/31) interpretation of p16 and p53 IHC was straightforward; 10 were considered HPV-A (p16+/p53wt) and 18 cases were HPV-I (p16-/p53abn). In the remaining three tumours the unusual immunophenotype was resolved by molecular testing, specifically (i) subclonal p16 staining and wild type p53 staining in a tumour positive for HPV and with no TP53 mutation (HPV-A), (ii) negative p16 and wild type p53 staining with a TP53 mutation and negative for HPV (HPV-I), and (iii) equivocally increased p16 staining with mutant pattern p53 expression, negative HPV ISH and with a TP53 mutation (HPV-I). CONCLUSION: Performing p16 and p53 IHC staining simultaneously allows classification of most HNSCC as HPV-A (p16 +, p53 wild type (especially basal sparing or null-like HPV associated staining patterns, which were completely specific for HPV-A SCC) or HPV-I (p16 -, p53 mutant pattern expression), with the potential for limiting additional molecular HPV or mutational testing to selected cases only.


Sujet(s)
Marqueurs biologiques tumoraux , Inhibiteur p16 de kinase cycline-dépendante , Tumeurs de la tête et du cou , Immunohistochimie , Infections à papillomavirus , Carcinome épidermoïde de la tête et du cou , Protéine p53 suppresseur de tumeur , Humains , Inhibiteur p16 de kinase cycline-dépendante/analyse , Protéine p53 suppresseur de tumeur/analyse , Carcinome épidermoïde de la tête et du cou/virologie , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Marqueurs biologiques tumoraux/analyse , Adulte d'âge moyen , Infections à papillomavirus/complications , Mâle , Femelle , Sujet âgé , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/virologie , Adulte , Sujet âgé de 80 ans ou plus
2.
Technol Cancer Res Treat ; 23: 15330338241272051, 2024.
Article de Anglais | MEDLINE | ID: mdl-39113534

RÉSUMÉ

Head and neck malignancies are a significant global health concern, with head and neck squamous cell carcinoma (HNSCC) being the sixth most common cancer worldwide accounting for > 90% of cases. In recent years, there has been growing recognition of the potential role of alternative splicing (AS) in the etiology of cancer. Increasing evidence suggests that AS is associated with various aspects of cancer progression, including tumor occurrence, invasion, metastasis, and drug resistance. Additionally, AS is involved in shaping the tumor microenvironment, which plays a crucial role in tumor development and response to therapy. AS can influence the expression of factors involved in angiogenesis, immune response, and extracellular matrix remodeling, all of which contribute to the formation of a supportive microenvironment for tumor growth. Exploring the mechanism of AS events in HNSCC could provide insights into the development and progression of this cancer, as well as its interaction with the tumor microenvironment. Understanding how AS contributes to the molecular changes in HNSCC cells and influences the tumor microenvironment could lead to the identification of new therapeutic targets. Targeted chemotherapy and immunotherapy strategies tailored to the specific AS patterns in HNSCC could potentially improve treatment outcomes and reduce side effects. This review explores the concept, types, processes, and technological advancements of AS, focusing on its role in the initiation, progression, treatment, and prognosis of HNSCC.


Sujet(s)
Épissage alternatif , Carcinome épidermoïde de la tête et du cou , Microenvironnement tumoral , Humains , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Microenvironnement tumoral/génétique , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , Régulation de l'expression des gènes tumoraux , Marqueurs biologiques tumoraux/génétique
3.
Cancer Immunol Immunother ; 73(10): 207, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39105870

RÉSUMÉ

BACKGROUND: HHLA2 (human endogenous retrovirus-H long terminal repeat-associating protein 2) represents a recently identified member of the B7 immune checkpoint family, characterized by limited expression in normal tissues but notable overexpression in various cancer types. Nevertheless, the precise function and interaction with immune cells remain poorly understood, particularly in laryngeal squamous cell carcinoma (LSCC). This investigation endeavored to elucidate the biological significance of HHLA2 within the tumor microenvironment of human LSCC tissues and delineate the clinical relevance and functional roles of HHLA2 in LSCC pathogenesis. METHODS: Through multiplexed immunohistochemistry analyses conducted on tissue microarrays sourced from LSCC patients (n = 72), the analysis was executed to assess the expression levels of HHLA2, density and spatial patterns of CD68+HLA-DR+CD163- (M1 macrophages), CTLA-4+CD4+FoxP3+ (CTLA-4+Treg cells), CTLA-4+CD4+FoxP3- (CTLA-4+Tcon cells), exhausted CD8+T cells, and terminally exhausted CD8+T cells in LSCC tissues. Survival analysis was conducted to evaluate the prognostic significance of HHLA2 and these immune checkpoints or immune cell populations, employing COX regression analysis to identify independent prognostic factors. RESULTS: Kaplan-Meier (K-M) survival curves revealed a significant association between HHLA2 expression and overall survival (OS) in LSCC. Elevated levels of HHLA2 were linked to reduced patient survival, indicating its potential as a prognostic marker (HR: 3.230, 95%CI 0.9205-11.34, P = 0.0067). Notably, increased infiltration of CD68+ cells (total macrophages), STING+CD68+HLA-DR+CD163- (STING+M1 macrophages), CTLA-4+CD4+FoxP3+, CTLA-4+CD4+FoxP3-, PD-1+LAG-3+CD8+T cells, and PD-1+LAG-3+TIM-3+CD8+T cells strongly linked to poorer survival outcomes (P < 0.05). A discernible trend was observed between the levels of these immune cell populations, STING+CD68+ (STING+ total macrophages), CD68+HLA-DR+CD163-, STING+CD68+CD163+HLA-DR- (STING+M2 macrophages), PD-1+LAG-3-CD8+T cells, PD-1+TIM-3+CD8+T cells, and PD-1+LAG-3+TIM-3-CD8+T cells and prognosis. Importantly, multivariate COX analysis identified HHLA2 as an independent predictive factor for OS in LSCC patients (HR = 3.86, 95% CI 1.08-13.80, P = 0.038). This underscored the potential of HHLA2 as a critical marker for predicting patient outcomes in LSCC. CONCLUSIONS: HHLA2 emerged as a detrimental prognostic biomarker for assessing OS in LSCC patients. Relative to other immune checkpoints, HHLA2 exhibited heightened predictive efficacy for the prognosis of LSCC patients.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du larynx , Lymphocytes TIL , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Mâle , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Femelle , Tumeurs du larynx/immunologie , Tumeurs du larynx/mortalité , Tumeurs du larynx/anatomopathologie , Pronostic , Adulte d'âge moyen , Marqueurs biologiques tumoraux/métabolisme , Sujet âgé , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/mortalité , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde/immunologie , Carcinome épidermoïde/mortalité , Carcinome épidermoïde/anatomopathologie , Immunoglobulines
4.
Head Neck Pathol ; 18(1): 77, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39141262

RÉSUMÉ

BACKGROUND: Nucleotide-binding domain-like receptor protein 3 (NLRP3), an inflammasome, is reported to be dysregulated or aberrantly expressed in chronic inflammation, leading to a myriad of inflammatory disorders, autoimmune diseases, and cancer. This study aimed to explore the expression and role of NLRP3 protein and the secreted cytokine IL-ß1 in oral squamous cell carcinoma (OSCC) and potentially malignant oral disorders (PMOD). MATERIAL & METHODS: Tissue NLRP3 expression was quantified using sandwich ELISA in 30 cases each of OSCC, PMOD, and normal oral mucosa. Serum IL-ß1 level was also measured by ELISA to determine their correlation. In surgically treated OSCC cases, pathological parameters such as tumor size, depth of invasion (DOI), pTNM stage, and perineural & lymphovascular invasion were assessed and correlated with NLRP3 & IL-ß1 levels to investigate their roles in tumor progression, invasion, and metastasis. RESULTS: Tissue NLRP3 expression was markedly elevated in OSCC, with significant IL-ß1 levels observed in the serum of both OSCC and PMOD cases. Both markers showed a pronounced increase with the severity of dysplasia, indicating a strong association (p = 0.003%). The expression levels of tissue NLRP3 and serum IL-ß1 were positively correlated with DOI and tumor size. Furthermore, their elevated levels, alongside higher histological grades, indicate roles in the dedifferentiation and progression of tumor cells. CONCLUSION: The findings indicated that increased expression of NLRP3 and IL-ß1 in PMOD correlates with higher transformation rates, along with tumor progression and dedifferentiation in OSCC. Consequently, these markers hold promise as valuable targets for prognostic assessment, diagnostics, and therapeutic strategies in OSCC.


Sujet(s)
Marqueurs biologiques tumoraux , Interleukine-1 bêta , Tumeurs de la bouche , Protéine-3 de la famille des NLR contenant un domaine pyrine , Humains , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/diagnostic , Mâle , Femelle , Adulte d'âge moyen , Marqueurs biologiques tumoraux/analyse , Pronostic , Adulte , Sujet âgé , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/diagnostic , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde/diagnostic , Carcinome épidermoïde/anatomopathologie
5.
BMC Cancer ; 24(1): 989, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39123141

RÉSUMÉ

BACKGROUND: Ketogenic interventions like short-term fasting show potential as complementary therapies to enhance the effectiveness of chemotherapy for cancer. However, the specific effects of fasting on head and neck squamous cell carcinoma (HNSCC) cells and healthy oral mucosa cells during these treatments are not well understood. This study investigates whether short-term fasting can differentially impact HNSCC cell survival and viability compared to healthy keratinocytes while undergoing standard chemotherapy regimens. METHODS: This study investigated the effects of fasting on cell viability in HN5 cell line and healthy oral keratinocyte cells. The HN5 cell line, derived from human tongue squamous cell carcinoma, and primary human keratinocytes isolated from the basal layer of gingival epithelium were divided into three groups: (1) control, (2) treated with the standard chemotherapeutic agent cisplatin, and (3) treated with cisplatin under fasting conditions achieved through 48-hour glucose restriction mimicking the blood glucose levels of fasted individuals. Cell proliferation was assessed at 48 and 72 h using the MTT assay, a colorimetric method based on mitochondrial dehydrogenase activity. Flow cytometry analysis with specific apoptosis and necrosis markers distinguished between early and late apoptotic, necrotic, and viable cells. RESULTS: Cell viability in HN5 and healthy keratinocyte cells decreased in cisplatin with low glucose groups compared to cisplatin and control groups. The same results were observed for healthy keratinocyte cells; only a decrease in cell viability in cisplatin groups compared to control groups was observed, which was not statistically significant. Cell apoptosis in HN5 and healthy keratinocyte cells increased in cisplatin with low glucose groups compared to cisplatin and control groups. In healthy keratinocyte cells, the cisplatin with low glucose group showed an impressive increase in necrosis, late apoptosis, and early apoptosis and a significant decrease in live cells compared with other groups. CONCLUSION: This study revealed that short-term fasting chemotherapy significantly improved HNSCC cell line apoptosis and necrosis.


Sujet(s)
Antinéoplasiques , Apoptose , Carcinome épidermoïde , Prolifération cellulaire , Survie cellulaire , Cisplatine , Jeûne , Kératinocytes , Humains , Cisplatine/pharmacologie , Cisplatine/effets indésirables , Lignée cellulaire tumorale , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/effets indésirables , Prolifération cellulaire/effets des médicaments et des substances chimiques , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/anatomopathologie , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/métabolisme
6.
Med Oncol ; 41(9): 225, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39136825

RÉSUMÉ

We have read the original article titled "P4HA2 contributes to head and neck squamous carcinoma progression and EMT through PI3K/AKT signaling pathway" by Yan-Ling Wu et al., which was published in the Medical Oncology journal, with great interest. This study provides valuable insights into the involvement of P4HA2 in the progression of head and neck squamous cell carcinoma (HNSCC), highlighting its potential as an oncogenic factor that promotes epithelial-mesenchymal transition (EMT), motility, invasion, and proliferation of cancer cells through the PI3K/AKT signaling pathway. While this work enhances our understanding of the role of P4HA2 in HNSCC, there are certain aspects that remain unexplored. These areas could be further investigated in future research to obtain a more comprehensive understanding. Specifically, the study did not investigate other signaling pathways or molecular mechanisms through which P4HA2 may impact the development of HNSCC. By exploring these molecular pathways, it may be possible to identify specific targets for pharmaceutical intervention to inhibit the production of P4HA2. Examining these aspects in future research would significantly contribute to our understanding of the role of P4HA2 in HNSCC and its potential as a therapeutic target. We appreciate the authors for their significant contribution and eagerly await future studies that expand upon these findings.


Sujet(s)
Transition épithélio-mésenchymateuse , Tumeurs de la tête et du cou , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Carcinome épidermoïde de la tête et du cou , Humains , Protéines proto-oncogènes c-akt/métabolisme , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Phosphatidylinositol 3-kinases/métabolisme , Évolution de la maladie , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme
7.
Nat Commun ; 15(1): 6923, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39134540

RÉSUMÉ

The combination of radiotherapy/chemoradiotherapy and immune checkpoint blockade can result in poor outcomes in patients with locally advanced head and neck squamous cell carcinoma (HNSCC). Here, we show that combining ATR inhibition (ATRi) with radiotherapy (RT) increases the frequency of activated NKG2A+PD-1+ T cells in animal models of HNSCC. Compared with the ATRi/RT treatment regimen alone, the addition of simultaneous NKG2A and PD-L1 blockade to ATRi/RT, in the adjuvant, post-radiotherapy setting induces a robust antitumour response driven by higher infiltration and activation of cytotoxic T cells in the tumour microenvironment. The efficacy of this combination relies on CD40/CD40L costimulation and infiltration of activated, proliferating memory CD8+ and CD4+ T cells with persistent or new T cell receptor (TCR) signalling, respectively. We also observe increased richness in the TCR repertoire and emergence of numerous and large TCR clonotypes that cluster based on antigen specificity in response to NKG2A/PD-L1/ATRi/RT. Collectively, our data point towards potential combination approaches for the treatment of HNSCC.


Sujet(s)
Protéines mutées dans l'ataxie-télangiectasie , Antigène CD274 , Immunothérapie , Carcinome épidermoïde de la tête et du cou , Microenvironnement tumoral , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des radiations , Animaux , Humains , Souris , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/radiothérapie , Carcinome épidermoïde de la tête et du cou/thérapie , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Immunothérapie/méthodes , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/thérapie , Tumeurs de la tête et du cou/radiothérapie , Lignée cellulaire tumorale , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Sous-famille C des récepteurs de cellules NK de type lectine/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Lymphocytes T CD8+/immunologie , Femelle , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/effets des radiations , Souris de lignée C57BL , Antigènes CD40/métabolisme , Antigènes CD40/immunologie , Antigènes CD40/antagonistes et inhibiteurs
8.
Front Immunol ; 15: 1390873, 2024.
Article de Anglais | MEDLINE | ID: mdl-39136017

RÉSUMÉ

Background: In view of improving biomarkers predicting the efficacy of immunotherapy for head and neck squamous cell carcinoma (R/M HNSCC), this multicenter retrospective study aimed to identify clinical, tumor microenvironmental, and genomic factors that are related to therapeutic response to the anti- Programmed cell death protein 1 (PD-1) antibody, nivolumab, in patients with R/M HNSCC. Methods: The study compared 53 responders and 47 non-responders, analyzing formalin-fixed paraffin-embedded samples using 14-marker multiplex immunohistochemistry and targeted gene sequencing. Results: Of 100 patients included, responders had significantly lower smoking and alcohol index, higher incidence of immune related adverse events, and higher PD-1 ligand (PD-L1) expression in immune cells as well as PD-L1 combined positive score (CPS) than non-responders. The frequency of natural killer cells was associated with nivolumab response in patients with prior cetuximab use, but not in cetuximab-naïve status. Age-stratified analysis showed nivolumab response was linked to high CPS and lymphoid-inflamed profiles in patients aged ≥ 65. In contrast, lower NLR in peripheral blood counts was associated with response in patients aged < 65. Notably, TP53 mutation-positive group had lower CPS and T cell densities, suggesting an immune-excluded microenvironment. Patients with altered tumor suppressor gene pathways, including TP53, CDKN2A, and SMAD4 mutations, had lower CPS, higher smoking index, and were associated with poor responses. Conclusion: Nivolumab treatment efficacy in HNSCC is influenced by a combination of clinical factors, age, prior treatment, immune environmental characteristics, and gene mutation profiles.


Sujet(s)
Tumeurs de la tête et du cou , Nivolumab , Carcinome épidermoïde de la tête et du cou , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Nivolumab/usage thérapeutique , Nivolumab/effets indésirables , Mâle , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/génétique , Femelle , Sujet âgé , Adulte d'âge moyen , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/génétique , Études rétrospectives , Marqueurs biologiques tumoraux/génétique , Antinéoplasiques immunologiques/usage thérapeutique , Antinéoplasiques immunologiques/effets indésirables , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Résultat thérapeutique , Adulte , Antigène CD274/génétique , Sujet âgé de 80 ans ou plus , Mutation , Génomique/méthodes
9.
Front Immunol ; 15: 1408173, 2024.
Article de Anglais | MEDLINE | ID: mdl-39136024

RÉSUMÉ

Introduction: The human leukocyte antigen complex (HLA) is essential for inducing specific immune responses to cancer by presenting tumor-associated peptides (TAP) to T cells. Overexpressed tumor associated antigens, mainly cancer-testis antigens (CTA), are outlined as essential targets for immunotherapy in oropharyngeal squamous cell carcinoma (OPSCC). This study assessed the degree to which presentation, gene expression, and antibody response (AR) of TAP, mainly CTA, are correlated in OPSCC patients to evaluate their potential as immunotherapy targets. Materials and methods: Snap-frozen tumor (NLigand/RNA=40), healthy mucosa (NRNA=6), and healthy tonsils (NLigand=5) samples were obtained. RNA-Seq was performed using Illumina HiSeq 2500/NovaSeq 6000 and whole exome sequencing (WES) utilizing NextSeq500. HLA ligands were isolated from tumor tissue using immunoaffinity purification, UHPLC, and analyzed by tandem MS. Antibodies were measured in serum (NAb=27) utilizing the KREX™ CT262 protein array. Data analysis focused on 312 proteins (KREX™ CT262 panel + overexpressed self-proteins). Results: 183 and 94 of HLA class I and II TAP were identified by comparative profiling with healthy tonsils. Genes from 26 TAP were overexpressed in tumors compared to healthy mucosa (LFC>1; FDR<0.05). Low concordance (r=0.25; p<0.0001) was found between upregulated mRNA and class I TAP. The specific mode of correlation of TAP was found to be dependent on clinical parameters. A lack of correlation was observed both between mRNA and class II TAP, as well as between class II tumor-unique TAP (TAP-U) presentation and antibody response (AR) levels. Discussion: This study demonstrates that focusing exclusively on gene transcript levels fails to capture the full extent of TAP presentation in OPSCC. Furthermore, our findings reveal that although CTA are presented at relatively low levels, a few CTA TAP-U show potential as targets for immunotherapy.


Sujet(s)
Antigènes néoplasiques , Tumeurs de l'oropharynx , Humains , Tumeurs de l'oropharynx/immunologie , Tumeurs de l'oropharynx/génétique , Antigènes néoplasiques/immunologie , Antigènes néoplasiques/génétique , Mâle , Femelle , Adulte d'âge moyen , Présentation d'antigène/immunologie , Sujet âgé , Régulation de l'expression des gènes tumoraux , Production d'anticorps/génétique , Production d'anticorps/immunologie , Adulte , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/génétique , , Multi-omique
10.
Article de Chinois | MEDLINE | ID: mdl-39107124

RÉSUMÉ

Objective: To assess the efficacy of neoadjuvant treatment with PD-1 (programmed cell death protein 1) inhibitors combined with paclitaxel (albumin-conjugated) and cisplatin (TP regimen) for locally advanced hypopharyngeal squamous cell carcinoma and laryngeal organ function preservation. Methods: Data of 53 patients, including 51 males and 2 females, aged 38-70 years old, who were diagnosed with locally advanced hypopharyngeal squamous carcinoma confirmed by histology and enhanced CT at the Cancer Prevention and Control Center of Sun Yat-sen University during the initial treatment from January 1, 2019 to January 15, 2023, were retrospectively analyzed. All patients received neoadjuvant therapy with PD-1 inhibitors combined with albumin-bound paclitaxel (260 mg/m2) and cisplatin (60 mg/m2) for 3 to 4 cycles. The main outcome measures were larynx dysfunction-free survival (LDFS), overall survival (OS), and progression-free survival (PFS). Survival curves were plotted using the Kaplan-Meier method, and Cox multifactorial analysis was further performed if Cox univariate analysis was statistically significant. Results: The overall efficiency was 90.6% (48/53). The 1-year and 2-year LDFS rates were 83.8% (95%CI: 74.0% to 94.8%) and 50.3% (95%CI: 22.1% to 91.6%), the 1-year and 2-year OS rates were 95.2% (95%CI: 88.9% to 100.0%) and 58.2% (95%CI: 25.6% to 81.8%), and the 1-year and 2-year PFS rates were 83.9% (95%CI: 74.2% to 94.9%) and 53.5% (95%CI: 32.1% to 89.1%). Adverse events associated with the neoadjuvant therapy were mainly myelosuppression (45.3%), gastrointestinal reactions (37.7%) and hypothyroidism (20.8%). Conclusion: The neoadjuvant treatment of locally advanced hypopharyngeal squamous cell carcinoma using PD-1 inhibitors combined with paclitaxel and cisplatin can provide with a higher survival rate with a improved laryngeal organ function preservation rate.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Cisplatine , Tumeurs de l'hypopharynx , Traitement néoadjuvant , Paclitaxel , Humains , Mâle , Adulte d'âge moyen , Femelle , Cisplatine/usage thérapeutique , Paclitaxel/usage thérapeutique , Paclitaxel/administration et posologie , Adulte , Sujet âgé , Tumeurs de l'hypopharynx/thérapie , Tumeurs de l'hypopharynx/traitement médicamenteux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Études rétrospectives , Albumines/usage thérapeutique , Albumines/administration et posologie , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/thérapie , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/thérapie , Carcinome épidermoïde/anatomopathologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs
11.
Support Care Cancer ; 32(9): 575, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107440

RÉSUMÉ

PURPOSE: Patients with head/neck squamous cell carcinomas (HNSCC) experience significant tumor- and treatment-related side effects. No efficient summary measure capturing the totality of side effect burden currently exists. We examined associations between a single patient-reported outcome (PRO) item evaluating side effect bother (FACT GP5, "I am bothered by side effects of treatment") with overall side effects in HNSCC. METHODS: We performed a retrospective secondary analysis of development of the Functional Assessment of Cancer Therapy (FACT) Head/Neck Symptom Index (FHNSI-10), which included completing FACT-HN (including Head/Neck Cancer Subscale (HNCS) and Trial Outcome Index (TOI)) and the pain intensity numeric rating scale (NRS). We calculated Spearman's correlations between GP5 and these measures of patient-reported global health, head/neck side effects, and pain intensity NRS. A correlation of > 0.4 was considered sufficient evidence of association. RESULTS: Ninety-seven patients completed baseline and 85 completed 3-month follow-up surveys. GP5 was highly correlated with FACT-HN total score (baseline r = 0.66, 3 months r = 0.67) and FHNSI-10 (baseline r = 0.63, 3 months r = 0.65). GP5 correlated with multiple FACT-HN subscales including FACT-G, physical well-being, functional well-being, HNCS, and TOI (range baseline r = 0.53-0.77, range 3 months r = 0.49-0.77). Worsening GP5 score was associated with worsening overall HNCS (p = 0.002), worsening FHNSI-10 score (p < 0.001), and worsening mean pain intensity (p < 0.001). CONCLUSION: GP5 exhibited validity within HNSCC, exhibiting substantial correlations with a number of HNSCC-related PRO measures including FACT-HN and FHNSI-10. Worsening GP5 was associated with worsening HNCS, FHNSI summary score, and pain intensity. GP5 has promise as a summary indicator of symptom and side effect bother in HNSCC.


Sujet(s)
Tumeurs de la tête et du cou , Mesures des résultats rapportés par les patients , Carcinome épidermoïde de la tête et du cou , Humains , Mâle , Femelle , Études rétrospectives , Adulte d'âge moyen , Sujet âgé , Adulte , Mesure de la douleur/méthodes , Enquêtes et questionnaires , Sujet âgé de 80 ans ou plus , Qualité de vie , Études de suivi
12.
J Cancer Res Clin Oncol ; 150(8): 379, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093329

RÉSUMÉ

PURPOSE: There is no agreed-upon standard option for patients with locally advanced head and neck squamous cell carcinoma (LA HNSCC) unfit for cisplatin-based regimens. Therefore, we performed a systematic review to explore alternative options for this population. METHODS: We searched PubMed, Cochrane, and Embase databases for observational studies and clinical trials (CTs) assessing treatment options for LA HNSCC cisplatin-ineligible patients. This study was registered in PROSPERO under the number CRD42023483156. RESULTS: This systematic review included 24 studies (18 observational studies and 6 CTs), comprising 4450 LA HNSCC cisplatin-ineligible patients. Most patients were treated with cetuximab-radiotherapy [RT] (50.3%), followed by carboplatin-RT (31.7%). In seven studies reporting median overall survival (OS) in patients treated with cetuximab-RT, it ranged from 12.8 to 46 months. The median OS was superior to 40 months in two studies assessing carboplatin-RT, and superior to 15 months in two studies assessing RT alone. For other regimens such as nimotuzumab-RT, docetaxel-RT, and carboplatin-RT plus paclitaxel the median OS was 21, 25.5, and 28 months, respectively. CONCLUSIONS: Our systematic review supports the use of a variety of therapy combinations for LA HNSCC cisplatin-ineligible patients. We highlight the urgent need for clinical studies assessing treatment approaches in this population.


Sujet(s)
Cisplatine , Tumeurs de la tête et du cou , Carcinome épidermoïde de la tête et du cou , Humains , Cisplatine/administration et posologie , Cisplatine/usage thérapeutique , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/thérapie , Carcinome épidermoïde de la tête et du cou/mortalité , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/thérapie , Tumeurs de la tête et du cou/mortalité , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Chimioradiothérapie/méthodes , Cétuximab/usage thérapeutique , Cétuximab/administration et posologie , Carboplatine/administration et posologie , Antinéoplasiques/usage thérapeutique
13.
Cancer Immunol Immunother ; 73(10): 187, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093451

RÉSUMÉ

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) typically present with a complex anatomical distribution, often accompanied by insidious symptoms. This combination contributes to its high incidence and poor prognosis. It is now understood that the immune features of cellular components within the tumor ecosystem and their complex interactions are critical factors influencing both tumor progression and the effective immune response. METHODS: We obtained single-cell RNA sequencing data of 26,496 cells from three tumor tissues and five normal tissues and performed subsequent analyses. Immunohistochemical staining on tumor sections was used to validate the presence of malignant cells. Additionally, we included bulk RNA sequencing data from 502 HNSCC patients. Kaplan-Meier analysis and the log-rank test were employed to assess predictors of patient outcomes. RESULTS: We identified three epithelial subclusters exhibiting immune-related features. These subclusters promoted the infiltration of T cells, dendritic cells, and monocytes into the tumor microenvironment. Additionally, cancer-associated fibroblasts displayed tumor-promoting and angiogenesis characteristics, contrasting with the predominant antigen-presenting and inflammatory roles observed in fibroblasts from normal tissues. Furthermore, tumor endothelial subsets exhibited a double-sided effect, promoting tumor progression and enhancing the effectiveness of immune response. Finally, follicular helper T cells and T helper 17 cells were found to be significantly correlated with improved outcomes in HNSCC patients. These CD4+ T cell subpopulations could promote the anti-tumor immune response by recruiting and activating B and T cells. CONCLUSION: Our findings provide deeper insights into the immune features of the tumor ecosystem and reveal the prognostic significance of follicular helper T cells and T helper 17 cells. These findings may pave the way for the development of therapeutic approaches.


Sujet(s)
Tumeurs de la tête et du cou , Lymphocytes TIL , Analyse de l'expression du gène de la cellule unique , Carcinome épidermoïde de la tête et du cou , Cellules Th17 , Microenvironnement tumoral , Femelle , Humains , Mâle , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , Lymphocytes TIL/immunologie , Pronostic , RNA-Seq/méthodes , Analyse de l'expression du gène de la cellule unique/méthodes , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Lymphocytes T auxiliaires folliculaires/immunologie , Cellules Th17/immunologie , Microenvironnement tumoral/immunologie
14.
Medicine (Baltimore) ; 103(31): e39184, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093745

RÉSUMÉ

BACKGROUND: Increasing evidence has shown that hypoxia is a biomarker of tumor proliferation and metastasis. This research aimed to identify a hypoxia-associated gene prognostic index (HAGPI) in head and neck squamous cell carcinoma (HNSCC) and based on HAGPI-defined subgroups to predict prognosis and response to immune checkpoint inhibitors therapy. METHODS: RNA-sequencing transcriptomic data for patients with HNSCC were downloaded from The Cancer Genome Atlas (TCGA). Protein-protein interaction network analysis was performed to select hypoxia-related hub genes. Univariate and multivariate cox regression analyses were used to identify hub genes to develop the HAGPI. Afterward expression data were imported into CIBERSORT to evaluate the relative proportion of 22 immune cells and compared the relative proportions of immune cells between the 2 HAGPI subgroups. The relationship between immunopheno score (IPS) and HAGPI was validated for immune checkpoint inhibitors (ICIs) response in TCGA cohorts. RESULTS: The HAGPI was constructed based on HS3ST1, HK1, PGK1, STC2, SERPINE1, PKLR genes. In high-HAGPI patients, the primary and secondary endpoint events in TCGA and GEO cohorts were significantly lower than low-HAGPI groups (P < .05). HAGPI-high patients exhibited a poorer prognosis than HAGPI-low patients did. The abundance of M2 macrophages and NK cell were significantly enhanced in the high-HAGPI while T cells regulatory and T cells CD8, were markedly elevated in the low-HAGPI. Meanwhile, patients in the low-HAGPI patients had higher levels of immunosuppressant expression and less aggressive phenotypes. Furthermore, IPS analysis showed that the low-HAGPI group with higher IPS represented a more immunogenic phenotype. CONCLUSION: The current study developed and verified a HAPGI model that can be considered as an independent prognostic biomarker and elucidated the tumor immune microenvironment of HNSCC.


Sujet(s)
Tumeurs de la tête et du cou , Inhibiteurs de points de contrôle immunitaires , Carcinome épidermoïde de la tête et du cou , Humains , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/mortalité , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mâle , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/mortalité , Pronostic , Femelle , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Appréciation des risques/méthodes , Cartes d'interactions protéiques/génétique , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Transcriptome , Hypoxie , Sujet âgé
15.
Radiat Oncol ; 19(1): 104, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095864

RÉSUMÉ

BACKGROUND: Radiotherapy (RT) in head and neck squamous cell cancer (HNSCC) often leads to sticky saliva and xerostomia (SSX). Dose sparing of salivary glands (SG) reduces occurrence of SSX but few studies investigated the relationship between RT dose to SG substructures and SSX. We therefore investigated this hypothesis, focusing on the parotid duct (PD). METHODS: Retrospective data was collected from 99 HNSCC patients treated at our center with (chemo-)radiotherapy (CRT). PD and other organs-at-risk (OAR) were (re-)contoured and DVHs were generated without re-planning. SSX was graded according to CTCAE v.4.03 and evaluated at acute, subacute, and two late timepoints. RESULTS: Most patients presented with loco-regionally advanced disease. In 47% of patients, up-front neck dissection preceded CRT. Weighted mean dose was 28.6 Gy for bilateral parotid glands (PG), and 32.0 Gy for PD. Acute SSX presented as grades 0 (35.3%), I (41.4%), II (21.2%) and III (2.0%). There was no association of OARs and SSX ≥ grade 2 in univariable logistic regression (LR). Multivariable LR showed statistically significant relationship of acute SSX with: PG weighted mean dose (OR 0.84, p = 0.004), contralateral PG mean dose (OR 1.14, p = 0.02) and contralateral PD planning OAR (PD PRV) mean dose (OR 1.84, p = 0.03). CONCLUSIONS: There was an association of acute SSX with dose exposure of PD PRV in multivariable regression, only. Due to statistical uncertainties and the retrospective nature of this analysis, further studies are required to confirm or reject the hypothesis.


Sujet(s)
Tumeurs de la tête et du cou , Organes à risque , Glande parotide , Dosimétrie en radiothérapie , Carcinome épidermoïde de la tête et du cou , Xérostomie , Humains , Xérostomie/étiologie , Études rétrospectives , Mâle , Femelle , Adulte d'âge moyen , Glande parotide/effets des radiations , Sujet âgé , Tumeurs de la tête et du cou/radiothérapie , Carcinome épidermoïde de la tête et du cou/radiothérapie , Organes à risque/effets des radiations , Adulte , Sujet âgé de 80 ans ou plus , Salive/effets des radiations , Lésions radiques/étiologie , Carcinome épidermoïde/radiothérapie , Radiothérapie conformationnelle avec modulation d'intensité/effets indésirables , Radiothérapie conformationnelle avec modulation d'intensité/méthodes , Planification de radiothérapie assistée par ordinateur/méthodes
16.
Nat Commun ; 15(1): 6755, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39117659

RÉSUMÉ

Histone lysine methyltransferase 2D (KMT2D) is the most frequently mutated epigenetic modifier in head and neck squamous cell carcinoma (HNSCC). However, the role of KMT2D in HNSCC tumorigenesis and whether its mutations confer any therapeutic vulnerabilities remain unknown. Here we show that KMT2D deficiency promotes HNSCC growth through increasing glycolysis. Additionally, KMT2D loss decreases the expression of Fanconi Anemia (FA)/BRCA pathway genes under glycolytic inhibition. Mechanistically, glycolytic inhibition facilitates the occupancy of KMT2D to the promoter/enhancer regions of FA genes. KMT2D loss reprograms the epigenomic landscapes of FA genes by transiting their promoter/enhancer states from active to inactive under glycolytic inhibition. Therefore, combining the glycolysis inhibitor 2-DG with DNA crosslinking agents or poly (ADP-ribose) polymerase (PARP) inhibitors preferentially inhibits tumor growth of KMT2D-deficient mouse HNSCC and patient-derived xenografts (PDXs) harboring KMT2D-inactivating mutations. These findings provide an epigenomic basis for developing targeted therapies for HNSCC patients with KMT2D-inactivating mutations.


Sujet(s)
Glycolyse , Carcinome épidermoïde de la tête et du cou , Animaux , Humains , Souris , Glycolyse/génétique , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Protéine BRCA1/métabolisme , Protéine BRCA1/génétique , Protéine BRCA1/déficit , Protéine BRCA2/génétique , Protéine BRCA2/métabolisme , Protéine BRCA2/déficit , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Anémie de Fanconi/génétique , Anémie de Fanconi/métabolisme , Régulation de l'expression des gènes tumoraux , Tests d'activité antitumorale sur modèle de xénogreffe , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Femelle , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Transduction du signal , Régions promotrices (génétique)/génétique , Protéine de la leucémie myéloïde-lymphoïde
17.
Int J Nanomedicine ; 19: 7185-7200, 2024.
Article de Anglais | MEDLINE | ID: mdl-39050876

RÉSUMÉ

Introduction: Traditional surgical resection, radiotherapy, and chemotherapy have been the treatment options for patients with head and neck squamous cell carcinoma (HNSCC) over the past few decades. Nevertheless, the five-year survival rate for patients has remained essentially unchanged, and research into treatments has been relatively stagnant. The combined application of photothermal therapy (PTT) and immunotherapy for treating HNSCC has considerable potential. Methods: Live-dead cell staining and CCK-8 assays proved that Fe3O4 nanoparticles are biocompatible in vitro. In vitro, cellular experiments utilized flow cytometry and immunofluorescence staining to verify the effect of Fe3O4 nanoparticles on the polarisation of tumor-associated macrophages. In vivo, animal experiments were conducted to assess the inhibitory effect of Fe3O4 nanoparticles on tumor proliferation under the photothermal effect in conjunction with BMS-1. Tumour tissue sections were stained to observe the effects of apoptosis and the inhibition of tumor cell proliferation. The histological damage to animal organs was analyzed by hematoxylin and eosin (H&E) staining. Results: The stable photothermal properties of Fe3O4 nanoparticles were validated by in vitro cellular and in vivo animal experiments. Fe3O4 photothermal action not only directly triggered immunogenic cell death (ICD) and enhanced the immunogenicity of the tumor microenvironment but also regulated the expression of tumor-associated macrophages (TAMs), up-regulating CD86 and down-regulating CD206 to inhibit tumor growth. The PD-1/PD-L1 inhibitor promoted tumor suppression, and reduced tumor recurrence and metastasis. In vivo studies demonstrated that the photothermal action exhibited a synergistic effect when combined with immunotherapy, resulting in significant suppression of primary tumors and an extension of survival. Conclusion: In this study, we applied Fe3O4 photothermolysis in a biomedical context, combining photothermolysis with immunotherapy, exploring a novel pathway for treating HNSCC and providing a new strategy for effectively treating HNSCC.


Sujet(s)
Tumeurs de la tête et du cou , Immunothérapie , Thérapie photothermique , Carcinome épidermoïde de la tête et du cou , Macrophages associés aux tumeurs , Animaux , Thérapie photothermique/méthodes , Carcinome épidermoïde de la tête et du cou/thérapie , Carcinome épidermoïde de la tête et du cou/immunologie , Souris , Tumeurs de la tête et du cou/thérapie , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/anatomopathologie , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/immunologie , Immunothérapie/méthodes , Humains , Lignée cellulaire tumorale , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/composition chimique , Association thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antigène CD274/antagonistes et inhibiteurs , Apoptose/effets des médicaments et des substances chimiques
18.
BMC Cancer ; 24(1): 904, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39068410

RÉSUMÉ

The lncRNA NEAT1 has been shown to promote the progression of several cancers, containing laryngeal squamous cell carcinoma (LSCC). However, the precise mechanism by which it promotes LSCC progression remains unclear. In this study, we verified the high expression of lncRNA NEAT1 in LSCC tissues and cells using RT-qPCR. Analysis of clinical data exhibited that high expression of lncRNA NEAT1 was associated with a history of smoking, worse T stage, lymph node metastasis, and later TNM stage in patients with LSCC. The promotion effect of lncRNA NEAT1 on LSCC cell proliferation, migration, invasion, and tumor growth in vivo was verified by CCK-8, plate clone formation, Transwell, and nude mouse tumorigenicity assays. Bioinformatics prediction and double luciferase reporter gene assay verified the binding of miR-411-3p to lncRNA NEAT1 and FZD3 mRNA, and inhibition of miR-411-3p reversed the inhibitory effect of lncRNA NEAT1 on FZD3 expression in LSCC cells. We also verified that lncRNA NEAT1-mediated FZD3 activation in the Wnt pathway affects LSCC development. In conclusion, we demonstrate that lncRNA NEAT1 promotes the progression of LSCC, and propose that the lncRNA NEAT1/miR-411-3p/FZD3 axis may be an effective target for LSCC therapy.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du larynx , microARN , ARN long non codant , Voie de signalisation Wnt , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , microARN/génétique , microARN/métabolisme , Tumeurs du larynx/génétique , Tumeurs du larynx/anatomopathologie , Tumeurs du larynx/métabolisme , Voie de signalisation Wnt/génétique , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Animaux , Souris , Mâle , Femelle , Lignée cellulaire tumorale , Invasion tumorale/génétique , Récepteurs Frizzled/génétique , Récepteurs Frizzled/métabolisme , Souris nude , Adulte d'âge moyen , Carcinome épidermoïde/génétique , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/métabolisme
19.
BMC Complement Med Ther ; 24(1): 287, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39068492

RÉSUMÉ

BACKGROUND: Oral squamous cell carcinoma (OSCC) is the most common malignancy of the head and neck. Zeng-Sheng-Ping, composed of Sophora tonkinensis Gagnep., Bistorta officinalis Delarbre, Sonchus arvensis L., Prunella vulgaris L., Dioscorea bulbifera L., and Dictamnus dasycarpus Turcz., was regarded as an anti-cancer drug with significant clinical efficacy, but was discontinued due to liver toxicity. Our research group developed a modified Zeng-Sheng-Ping (ZSP-M) based on original Zeng-Sheng-Ping that exhibited high efficiency and low toxicity in preliminary investigations, although its pharmacodynamic mechanism is still unclear. Here, we aimed to elucidate the pharmacodynamic material basis of ZSP-M and investigate its chemopreventive effect on OSCC by modulating tumor associated macrophages (TAMs). METHODS: Components of ZSP-M were characterized using ultra-performance liquid chromatography-mass spectrometry. Chemopreventive effect induced by ZSP-M against experimental oral cancer was investigated using the 4-nitroquinoline N-oxide precancerous lesion mouse model. RNA sequencing analysis was used to gain a global transcriptional view of the effect of ZSP-M treatment. A cell co-culture model was used to study the targeted effect of ZSP-M on TAMs and the biological properties of OSCC cells and to detect changes in TAM phenotypes. The binding of ZSP-M active compounds to TNF alpha induced protein 6 (TNFAIP6) protein was analyzed by molecular docking and dynamic simulation. RESULTS: Forty main components of ZSP-M were identified, the most abundant of which were flavonoids. ZSP-M inhibited the degree of epithelial dysplasia in precancerous lesions by inhibiting the expression of the TNFAIP6 and CD163 proteins in the precancerous lesions of the tongue. ZSP-M inhibited proliferation, colony formation, migration and invasion of SCC7 cells by targeting TAMs. ZSP-M reduced the expression of CD163+ cells, inhibited the expression of TNFAIP6 protein, Arg1 mRNA and Il10 mRNA in TAMs, and reduced IL-10 cytokine release in the co-culture environment. This effect was maintained after the addition of recombinant TNFAIP6 protein. Computer simulations showed that trifolirhizin and maackiain are well-connected to TNFAIP6. CONCLUSIONS: ZSP-M counteracts the immunosuppressive action of TAMs by specific targeting of TNFAIP6, thereby exerting chemopreventive activity of OSCC.


Sujet(s)
Tumeurs de la bouche , Macrophages associés aux tumeurs , Animaux , Souris , Tumeurs de la bouche/traitement médicamenteux , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Carcinome épidermoïde/traitement médicamenteux , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Humains , Lignée cellulaire tumorale , Mâle , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux
20.
Sci Rep ; 14(1): 15962, 2024 07 10.
Article de Anglais | MEDLINE | ID: mdl-38987626

RÉSUMÉ

The presence of cancer stem cells (CSCs) contributes significantly to treatment resistance in various cancers, including head and neck squamous cell carcinoma (HNSCC). Despite this, the relationship between cancer stemness and immunity remains poorly understood. In this study, we aimed to identify potential immunotherapeutic targets and sensitive drugs for CSCs in HNSCC. Using data from public databases, we analyzed expression patterns and prognostic values in HNSCC. The stemness index was calculated using the single-sample gene set enrichment analysis (ssgsea) algorithm, and weighted gene co-expression network analysis (WGCNA) was employed to screen for key stemness-related modules. Consensus clustering was then used to group samples for further analysis, and prognosis-related key genes were identified through regression analysis. Our results showed that tumor samples from HNSCC exhibited higher stemness indices compared to normal samples. WGCNA identified a module highly correlated with stemness, comprising 187 genes, which were significantly enriched in protein digestion and absorption pathways. Furthermore, we identified sensitive drugs targeting prognostic genes associated with tumor stemness. Notably, two genes, HLF and CCL11, were found to be highly associated with both stemness and immunity. In conclusion, our study identifies a stemness-related gene signature and promising drug candidates for CSCs of HNSCC. Additionally, HLF and CCL11, which are associated with both stemness and immunity, represent potential targets for immunotherapy in HNSCC.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Tumeurs de la tête et du cou , Cellules souches tumorales , Carcinome épidermoïde de la tête et du cou , Humains , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/immunologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Pronostic , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/anatomopathologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Réseaux de régulation génique , Analyse de profil d'expression de gènes , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE