Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.429
Filtrer
1.
Zhonghua Yi Xue Za Zhi ; 104(26): 2373-2377, 2024 Jul 09.
Article de Chinois | MEDLINE | ID: mdl-38978359

RÉSUMÉ

Standard treatment for patients with locally advanced head and neck squamous cell carcinoma (LAHNSCC) whose larynx could not be preserved surgically consists mainly of concurrent chemoradiotherapy (CCRT). Induction chemotherapy (ICT) followed by radiotherapy or chemoradiotherapy is also an alternative option. However, whether ICT could provide survival benefits for patients with LAHNSCC, besides its role in laryngeal preservation and selecting the treatment modality, is still controversial. The article summarizes the current position of ICT for LAHNSCC and discusses the standard regimen of ICT, its role in larynx preservation, its ability to predicting the result of chemoradiotherapy, clinical outcomes regarding the survival benefits after ICT, its role in the treatment deintensification for human papillomavirus-positive LAHNSCC and its application in the era of immunotherapy.


Sujet(s)
Carcinome épidermoïde , Chimioradiothérapie , Tumeurs de la tête et du cou , Chimiothérapie d'induction , Carcinome épidermoïde de la tête et du cou , Humains , Tumeurs de la tête et du cou/thérapie , Tumeurs de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/thérapie , Carcinome épidermoïde/thérapie , Carcinome épidermoïde/traitement médicamenteux
2.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000120

RÉSUMÉ

Head and neck squamous cell carcinoma (HNSCC) affects squamous cells in the head and neck region and is currently ranked as the sixth most common cancer worldwide. NF-E2-related factor 2 (NRF2) plays a crucial role in cellular protection and defence mechanisms and NRF2 over-expression has been linked to various cancers; however, its role in the response of HNSCC cells remains elusive. We investigated the effects of ML385, a selective NRF2 inhibitor, on HNSCC to understand the underlying molecular mechanisms, and to assess the potential of ML385 as a therapeutic agent. We treated HNSCC cell lines with ML385 and observed a significant reduction in the expression of NRF2 and its downstream target, heme oxygenase-1 (HO-1), using Western blotting. We evaluated its effects on various cellular processes, including cell proliferation, cloning, migration, and wound healing, in HNSCC cell lines. ML385 treatment substantially reduced NRF2 expression, promoting a decrease in the investigated cellular activities. Additionally, we examined changes in the expression of cell-cycle-related proteins and found that ML385 induced cell cycle arrest at the G1/S phase in HNSCC cell lines. Our findings suggest that ML385 can regulate cell cycle progression, inhibit HNSCC growth, and have potential as a therapeutic agent for HNSCC.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Tumeurs de la tête et du cou , Facteur-2 apparenté à NF-E2 , Carcinome épidermoïde de la tête et du cou , Humains , Facteur-2 apparenté à NF-E2/métabolisme , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , 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 , Mouvement cellulaire/effets des médicaments et des substances chimiques , Heme oxygenase-1/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Acétamides , Benzodioxoles
3.
BMC Cancer ; 24(1): 838, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39003442

RÉSUMÉ

BACKGROUND: The National Comprehensive Cancer Network (NCCN) guideline recommends consideration of weekly cisplatin as an alternative option for patients with head and neck cancer undergoing definitive chemoradiation. However, in a recent phase III trial (ConCERT), 20% of patients treated with weekly cisplatin could not receive a total of 200 mg/m2, and the association of low adherence to weekly cisplatin and cancer control outcomes remains unclear. To fill this knowledge gap, we performed an observational cohort study of patients with head and neck cancer undergoing definitive chemoradiation with weekly cisplatin. METHODS: Our institutional database was queried for patients with non-metastatic head and neck cancer who underwent definitive chemoradiation with weekly cisplatin (40 mg/m2) between November 2007 and April 2023. Adherence to weekly cisplatin was defined as receiving at least 5 cycles with a total cumulative dose of 200 mg/m2. Survival outcomes were evaluated using Kaplan-Meier method, log-rank tests, Cox proportional hazard multivariable (MVA) analyses. Logistic MVA was performed to identify variables associated with low adherence to weekly cisplatin. Fine-Gray MVA was performed to analyze failure outcomes with death as a competing event. RESULTS: Among 119 patients who met our criteria, 51 patients (42.9%) had low adherence to weekly cisplatin. Median follow up was 19.8 months (interquartile range 8.8-65.6). Low adherence to weekly cisplatin was associated with worse overall survival (adjusted hazards ratio [aHR] 2.94, 95% confidence interval [CI] 1.58-5.47, p < 0.001) and progression-free survival (aHR 2.32, 95% CI 1.29-4.17, p = 0.005). It was also associated with worse distant failure (aHR 4.55, 95% CI 1.19-17.3, p = 0.03), but not locoregional failure (aHR 1.61, 95% CI 0.46-5.58, p = 0.46). KPS < 90 was the only variable associated with low adherence to weekly cisplatin (adjusted odds ratio [aOR] 2.67, 95% CI 1.10-6.65, p = 0.03). CONCLUSION: Our study suggested that over 40% of patients underwent fewer than 5 weekly cisplatin cycles and that low adherence to weekly cisplatin was an independent, adverse prognostic factor for worse survival and distant failure outcomes. Those with reduced adherence to weekly cisplatin were more likely to have poor performance status. Further studies are warranted to improve the adherence to chemotherapy and outcomes.


Sujet(s)
Chimioradiothérapie , Cisplatine , Tumeurs de la tête et du cou , Carcinome épidermoïde de la tête et du cou , Humains , Cisplatine/administration et posologie , Cisplatine/usage thérapeutique , Mâle , Femelle , Adulte d'âge moyen , Études rétrospectives , Sujet âgé , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/mortalité , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/mortalité , Tumeurs de la tête et du cou/anatomopathologie , Chimioradiothérapie/méthodes , Adhésion au traitement médicamenteux/statistiques et données numériques , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/administration et posologie , Résultat thérapeutique , Calendrier d'administration des médicaments , Adulte , Estimation de Kaplan-Meier
4.
Sci Rep ; 14(1): 15962, 2024 Jul 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
5.
Oncotarget ; 15: 444-458, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38985143

RÉSUMÉ

OBJECTIVE: Patients with relapsed or metastatic head and neck squamous cell carcinoma (HNSCC) after primary local therapy have low response rates with cetuximab, systemic chemotherapy or check point inhibitor therapy. Novel combination therapies with the potential to improve outcomes for patients with HNSCC is an area of high unmet need. METHODS: This is a phase II single-arm clinical trial of locally advanced or metastatic HNSCC patients treated with a combination of soluble EphB4-human serum albumin (sEphB4-HSA) fusion protein and pembrolizumab after platinum-based chemotherapy with up to 2 prior lines of treatment. The primary endpoints were safety and tolerability and the primary efficacy endpoint was overall response rate (ORR). Secondary endpoints included progression free survival (PFS) and overall survival (OS). HPV status and EphrinB2 expression were evaluated for outcome. RESULTS: Twenty-five patients were enrolled. Median follow up was 40.4 months (range 9.8 - 40.4). There were 6 responders (ORR 24%). There were 5 responders in the 11 HPV-negative and EphrinB2 positive patients, (ORR 45%) with 2 of these patients achieving a complete response (CR). The median PFS in HPV-negative/EphrinB2 positive patients was 3.2 months (95% CI 1.1, 7.3). Median OS in HPV-negative/EphrinB2 positive patients was 10.9 months (95% CI 2.0, 13.7). Hypertension, transaminitis and fatigue were the most common toxicities. DISCUSSION: The combination of sEphB4-HSA and pembrolizumab has a favorable toxicity profile and favorable activity particularly among HPV-negative EphrinB2 positive patients with HNSCC.


Sujet(s)
Anticorps monoclonaux humanisés , Éphrine B2 , Tumeurs de la tête et du cou , Récepteur EphB4 , Carcinome épidermoïde de la tête et du cou , Humains , Anticorps monoclonaux humanisés/usage thérapeutique , Femelle , Mâle , Adulte d'âge moyen , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/anatomopathologie , Sujet âgé , Éphrine B2/métabolisme , Adulte , 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 , Récepteur EphB4/métabolisme , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Infections à papillomavirus/virologie , Résultat thérapeutique , Protéines de fusion recombinantes/usage thérapeutique , Sujet âgé de 80 ans ou plus
6.
Cancer Med ; 13(12): e7346, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38923758

RÉSUMÉ

OBJECTIVES: Previous studies have demonstrated that obesity may impact the efficacy of anti-PD1 therapy, but the underlying mechanism remains unclear. In this study, our objective was to determine the prognostic value of obesity in patients with oral tongue squamous cell carcinoma (OTSCC) treated with pembrolizumab and establish a subtype based on fatty acid metabolism-related genes (FAMRGs) for immunotherapy. MATERIALS AND METHODS: We enrolled a total of 56 patients with OTSCC who underwent neoadjuvant anti-PD1 therapy. Univariate and multivariate Cox regression analyses, Kaplan-Meier survival analysis, and immunohistochemistry staining were performed. Additionally, we acquired the gene expression profiles of pan-cancer samples and conducted GSEA and KEGG pathway analysis. Moreover, data from TCGA, MSigDB, UALCAN, GEPIA and TIMER were utilized to construct the FAMRGs subtype. RESULTS: Our findings indicate that high Body Mass Index (BMI) was significantly associated with improved PFS (HR = 0.015; 95% CI, 0.001 to 0.477; p = 0.015), potentially attributed to increased infiltration of PD1 + T cells. A total of 91 differentially expressed FAMRGs were identified between the response and non-response groups in pan-cancer patients treated with immunotherapy. Of these, 6 hub FAMRGs (ACSL5, PLA2G2D, PROCA1, IL4I1, UBE2L6 and PSME1) were found to affect PD-1 expression and T cell infiltration in HNSCC, which may impact the efficacy of anti-PD1 therapy. CONCLUSION: This study demonstrates that obesity serves as a robust prognostic predictor for patients with OTSCC undergoing neoadjuvant anti-PD1 therapy. Furthermore, the expression of 6 hub FAMRGs (ACSL5, PLA2G2D, PROCA1, IL4I1, UBE2L6 and PSME1) plays a pivotal role in the context of anti-PD1 therapy and deserves further investigation.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Traitement néoadjuvant , Obésité , Tumeurs de la langue , Humains , Tumeurs de la langue/traitement médicamenteux , Tumeurs de la langue/métabolisme , Tumeurs de la langue/immunologie , Tumeurs de la langue/anatomopathologie , Tumeurs de la langue/mortalité , Tumeurs de la langue/génétique , Femelle , Mâle , Traitement néoadjuvant/méthodes , Obésité/métabolisme , Obésité/complications , Adulte d'âge moyen , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Pronostic , Sujet âgé , 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/mortalité , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/thérapie , Anticorps monoclonaux humanisés/usage thérapeutique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Adulte , Indice de masse corporelle , Marqueurs biologiques tumoraux/métabolisme
7.
Lancet Oncol ; 25(7): 888-900, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38851207

RÉSUMÉ

BACKGROUND: VEGF, a key mediator of angiogenesis and resistance to immunotherapy, is overexpressed in head and neck squamous cell carcinoma (HNSCC). We aimed to determine the recommended phase 2 dose of ramucirumab, a selective VEGFR2 inhibitor, given with pembrolizumab and the objective response rate of this combination as first-line treatment for recurrent or metastatic HNSCC. METHODS: In this single-centre, phase 1/2 trial, which was done at Washington University (St Louis, MO, USA), eligible patients were aged 18 years or older with incurable recurrent or metastatic HNSCC and an Eastern Cooperative Oncology Group performance status of 0-2. Patients in phase 2 were required to have had no previous systemic therapy for recurrent or metastatic disease. In a dose de-escalation phase 1 design, patients received ramucirumab (starting dose 10 mg/kg given intravenously) and pembrolizumab (200 mg intravenously) on day 1 of each 21-day cycle. The recommended phase 2 dose of ramucirumab was defined as the highest dose at which one or fewer of three patients had dose-limiting toxicity during cycle one (primary endpoint of phase 1). In a Simon's two-stage phase 2 design, patients received the recommended phase 2 dose of ramucirumab and pembrolizumab. Tumour response (primary endpoint of phase 2) was assessed by Response Evaluation Criteria in Solid Tumours (version 1.1). We hypothesised that there would be an objective response rate of 32% or higher (null ≤13%). Eight or more responses among 33 evaluable patients (those with at least one response assessment) was evidence for activity (80% power; one-sided α=0·05). Analyses were done per protocol. The trial is registered with ClinicalTrials.gov, NCT03650764, and is closed to enrolment. FINDINGS: Between June 18, 2019, and Feb 11, 2021, three patients enrolled and were treated in phase 1 and 37 patients in phase 2. Median age of all patients was 64 years (IQR 59-72). 36 (90%) of 40 patients were men and four (10%) were women, and 36 (90%) patients were White, three (8%) were Black or African American, and one (3%) was Asian. In phase 1, no dose-limiting toxicity event occurred. The recommended phase 2 dose of ramucirumab was 10 mg/kg. Median follow-up for patients on phase 2 was 14·8 months (IQR 4·9-31·0). In phase 2, 18 (55%; 95% CI 38-70) of 33 evaluable patients had an objective response, including confirmed complete response in 11 patients, confirmed partial response in six patients, and unconfirmed partial response in one patient. The most common grade 3 or worse adverse events were dysphagia (14 [38%] of 37 patients), lung infection (11 [30%]), lymphocyte count decrease (ten [27%]), hypophosphataemia (nine [24%]), and hypertension (eight [22%]). No treatment-related deaths were recorded. INTERPRETATION: Ramucirumab and pembrolizumab were safe to administer to patients with recurrent or metastatic HNSCC, and the objective response rate with this combination as first-line treatment for recurrent or metastatic HNSCC was favourable. Further studies of ramucirumab and pembrolizumab in patients with recurrent or metastatic HNSCC are warranted. FUNDING: Lilly and the Joseph Sanchez Foundation.


Sujet(s)
Anticorps monoclonaux humanisés , Protocoles de polychimiothérapie antinéoplasique , Tumeurs de la tête et du cou , Récidive tumorale locale , , Carcinome épidermoïde de la tête et du cou , Humains , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/effets indésirables , Mâle , Femelle , Adulte d'âge moyen , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/anatomopathologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs , Adulte , Dose maximale tolérée
8.
Front Immunol ; 15: 1353435, 2024.
Article de Anglais | MEDLINE | ID: mdl-38827739

RÉSUMÉ

Objective: This retrospective study analyzed the efficacy of PD-1 inhibitors combined with albumin-bound paclitaxel and cisplatin (TP regimen) in the treatment of recurrent and metastatic hypopharyngeal/laryngeal squamous cell carcinoma (RMHSCC/RMLSCC). Methods: Patients diagnosed and treated at the Sun Yat-sen University Cancer Center from August 1, 2020, to August 15, 2023, with histologically confirmed RMHSCC/RMLSCC were included. All patients received PD-1 inhibitors combined with albumin-bound paclitaxel (260mg/m2) and cisplatin (60mg/m2) for 3-4 cycles. The primary endpoints were overall survival (OS) and progression-free survival (PFS). Results: A total of 50 patients with RMHSCC/RMLSCC who received TP+PD-1 inhibitor therapy were included, with an objective response rate (ORR) of 56.0% (28/50). The 1-year and 2-year OS rates were 80.2% (95% CI: 69.3%-92.9%) and 68.6% (95% CI: 52.6%-89.5%), respectively, while the 1-year and 2-year PFS rates were 44.7% (95% CI: 31.9%-62.5%) and 26.0% (95% CI: 12.6%-53.4%), respectively. Treatment-related adverse events mainly included rash, myelosuppression, gastrointestinal reactions, and hypothyroidism. Conclusion: In the treatment of RMHSCC/RMLSCC with TP + PD-1 inhibitors, survival rates of patients can be improved while ensuring the safety of the treatment regimen.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Cisplatine , Tumeurs de l'hypopharynx , Inhibiteurs de points de contrôle immunitaires , Tumeurs du larynx , Récidive tumorale locale , Paclitaxel , Humains , Paclitaxel/administration et posologie , Paclitaxel/usage thérapeutique , Paclitaxel/effets indésirables , Mâle , Femelle , Adulte d'âge moyen , Cisplatine/administration et posologie , Cisplatine/usage thérapeutique , Cisplatine/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Sujet âgé , Tumeurs de l'hypopharynx/mortalité , Tumeurs de l'hypopharynx/traitement médicamenteux , Tumeurs de l'hypopharynx/anatomopathologie , Études rétrospectives , Adulte , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/mortalité , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Inhibiteurs de points de contrôle immunitaires/administration et posologie , Tumeurs du larynx/traitement médicamenteux , Tumeurs du larynx/mortalité , Tumeurs du larynx/anatomopathologie , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/mortalité , Résultat thérapeutique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Métastase tumorale
9.
Cancer Med ; 13(11): e7370, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38847087

RÉSUMÉ

OBJECTIVES: Certain low-level immune-related adverse events (irAEs) have been associated with survival benefits in patients with various solid tumors on immune checkpoint inhibitors (ICIs). We aimed to investigate the association between irAEs and response to neoadjuvant ICIs in patients with head and neck squamous cell carcinoma (HNSCC) and to identify differences in circulating cytokine levels based on irAE status. METHODS: This was a retrospective cohort study including three neoadjuvant clinical trials from July 2017 to January 2022: NCT03238365 (nivolumab ± tadalafil), NCT03854032 (nivolumab ± BMS986205), NCT03618654 (durvalumab ± metformin). The presence and type of irAEs, pathologic treatment response, and survival were compared. Canonical linear discriminant analysis (LDA) was performed to identify combinations of circulating cytokines predictive of irAEs using plasma sample multiplex assay. RESULTS: Of 113 participants meeting inclusion criteria, 32 (28.3%) developed irAEs during treatment or follow-up. Positive p16 status was associated with irAEs (odds ratio [OR] 2.489; 95% CI 1.069-6.119; p = 0.043). irAEs were associated with pathologic treatment response (OR 3.73; 95% CI 1.34-10.35; p = 0.011) and with higher OS in the combined cohort (HR 0.319; 95% CI 0.113-0.906; p = 0.032). Patients with irAEs within the nivolumab cohort had significant elevations of select cytokines pre-treatment. Canonical LDA identified key drivers of irAEs among all trials, which were highly predictive of future irAE status. CONCLUSIONS: irAEs are associated with response to neoadjuvant ICI therapy in HNSCC and can serve as clinical indicators for improved clinical outcomes. irAEs can be predicted by concentrations of several circulating cytokines prior to treatment.


Sujet(s)
Cytokines , Tumeurs de la tête et du cou , Inhibiteurs de points de contrôle immunitaires , Traitement néoadjuvant , 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/sang , Carcinome épidermoïde de la tête et du cou/immunologie , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mâle , Traitement néoadjuvant/effets indésirables , Traitement néoadjuvant/méthodes , Femelle , Adulte d'âge moyen , Études rétrospectives , Cytokines/sang , Sujet âgé , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/sang , Tumeurs de la tête et du cou/immunologie , Nivolumab/effets indésirables , Nivolumab/usage thérapeutique
10.
Anticancer Res ; 44(7): 2921-2931, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38925841

RÉSUMÉ

BACKGROUND/AIM: Human papillomavirus (HPV)-positive head and neck squamous cell carcinoma (HNSCC) is clinically and immunologically distinct from HPV-negative HNSCC. Herein, we investigated the presence of tumor antigens HPV E6/E7 and wild-type p53-specific T-cell responses, and the impact of immune checkpoint blockade in patients with HPV-positive HNSCC. MATERIALS AND METHODS: Peripheral blood mononuclear cells (PBMCs) from patients with HPV-positive HNSCC were stimulated with HPV E6/E7 or wild-type p53-derived peptide mixture and evaluated using the interferon-γ enzyme-linked immunosorbent spot assay. Flow cytometry was performed to analyze the proportion of T-cell subsets and T cells expressing immune checkpoint molecules. RESULTS: HPV E6/E7-specific T cells were detected in 22 (95.7%) of 23 patients, whereas wild-type p53-specific T cells were detected in 3 (15.0%) of 20 patients. Seven (43.8%) of 16 patients exhibited wild-type p53-specific T-cell responses, as determined using whole proteins instead of peptides. Immune checkpoint blockade enhanced wild-type p53-specific T-cell responses in 9 (45.0%) of 20 patients. Flow cytometric analysis of PBMCs revealed that responders exhibiting enhanced wild-type p53-specific T-cell responses following immune checkpoint blockade had a significantly higher proportion of Ki-67+CD4+ T cells, Ki-67+CD8+ T cells, regulatory T cells, PD-1+CD4+ T cells, and TIM-3+CD4+ T cells than non-responders. CONCLUSION: Our findings indicate that tumor antigen-specific T cells are present in the peripheral blood of patients with HPV-positive HNSCC. Blockade of checkpoint pathways can enhance T-cell responses in certain patients, probably via activated T cells, Tregs, and/or exhausted CD4+ T cells.


Sujet(s)
Tumeurs de la tête et du cou , Inhibiteurs de points de contrôle immunitaires , Infections à papillomavirus , Carcinome épidermoïde de la tête et du cou , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/virologie , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/virologie , Infections à papillomavirus/immunologie , Infections à papillomavirus/virologie , Antigènes néoplasiques/immunologie , Protéines des oncogènes viraux/immunologie , Protéine p53 suppresseur de tumeur/immunologie , Adulte , Agranulocytes/immunologie , Agranulocytes/métabolisme , Papillomaviridae/immunologie , Lymphocytes T/immunologie , Virus des Papillomavirus humains
11.
Asian Pac J Cancer Prev ; 25(6): 1935-1943, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38918654

RÉSUMÉ

OBJECTIVE: The 2x2 factorial design is an effective method that allows for multiple comparisons, especially in the context of interactions between different interventions, without substantially increasing the required sample size. In view of the considerable preclinical evidence for Curcumin and Metformin in preventing the development and progression of head and neck squamous cell carcinoma (HNSCC), this study describes the protocol of the clinical trial towards applying the drug combination in prevention of second primary tumors. METHODS: We have applied the trial design to a large phase IIB/III double-blind, multi-centric, placebo-controlled, randomized clinical trial to determine the safety and efficacy of Metformin and Curcumin in the prevention of second primary tumours (SPT) of the aerodigestive tract following treatment of HNSCC (n=1,500) [Clinical Registry of India, CTRI/2018/03/012274]. Patients recruited in this trial will receive Metformin (with placebo), Curcumin (with placebo), Metformin, and Curcumin or placebo alone for a period of 36 months. The primary endpoint of this trial is the development of SPT, while the secondary endpoints are toxicities associated with the agents, incidence of recurrence, and identifying potential biomarkers. In this article, we discuss the 2x2 factorial design and how it applies to the head and neck cancer chemoprevention trial. CONCLUSION: 2x2 factorial design is an effective trial design for chemoprevention clinical trials where the effectiveness of multiple interventions needs to be tested parallelly.


Sujet(s)
Curcumine , Tumeurs de la tête et du cou , Metformine , Seconde tumeur primitive , Humains , Metformine/usage thérapeutique , Curcumine/usage thérapeutique , Tumeurs de la tête et du cou/prévention et contrôle , Tumeurs de la tête et du cou/traitement médicamenteux , Méthode en double aveugle , Seconde tumeur primitive/prévention et contrôle , Mâle , Femelle , Carcinome épidermoïde de la tête et du cou/prévention et contrôle , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte d'âge moyen , Adulte , Études de suivi , Pronostic , Plan de recherche , Sujet âgé , Essais contrôlés randomisés comme sujet
12.
Int J Mol Sci ; 25(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38891817

RÉSUMÉ

(1) Head and neck squamous cell carcinoma (HNSCC) is common, while treatment is difficult, and mortality is high. Kinase inhibitors are promising to enhance the effects of radiotherapy. We compared the effects of the PARP inhibitors talazoparib and niraparib and that of the DNA-PKcs inhibitor AZD7648, combined with ionizing radiation. (2) Seven HNSCC cell lines, including Cal33, CLS-354, Detroit 562, HSC4, RPMI2650 (HPV-negative), UD-SCC-2 and UM-SCC-47 (HPV-positive), and two healthy fibroblast cell lines, SBLF8 and SBLF9, were studied. Flow cytometry was used to analyze apoptosis and necrosis induction (AnnexinV/7AAD) and cell cycle distribution (Hoechst). Cell inactivation was studied by the colony-forming assay. (3) AZD7648 had the strongest effects, radiosensitizing all HNSCC cell lines, almost always in a supra-additive manner. Talazoparib and niraparib were effective in both HPV-positive cell lines but only consistently in one and two HPV-negative cell lines, respectively. Healthy fibroblasts were not affected by any combined treatment in apoptosis and necrosis induction or G2/M-phase arrest. AZD7648 alone was not toxic to healthy fibroblasts, while the combination with ionizing radiation reduced clonogenicity. (4) In conclusion, talazoparib, niraparib and, most potently, AZD7648 could improve radiation therapy in HNSCC. Healthy fibroblasts tolerated AZD7648 alone extremely well, but irradiation-induced effects might occur. Our results justify in vivo studies.


Sujet(s)
Apoptose , Indazoles , Phtalazines , Pipéridines , Inhibiteurs de poly(ADP-ribose) polymérases , Radiosensibilisants , Carcinome épidermoïde de la tête et du cou , Humains , Phtalazines/pharmacologie , Indazoles/pharmacologie , Pipéridines/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Lignée cellulaire tumorale , Radiosensibilisants/pharmacologie , 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/radiothérapie , Apoptose/effets des médicaments et des substances chimiques , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/radiothérapie , DNA-activated protein kinase/antagonistes et inhibiteurs , DNA-activated protein kinase/métabolisme
13.
ESMO Open ; 9(6): 103476, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38833968

RÉSUMÉ

BACKGROUND: An important unmet need for new treatment options remains for patients with recurrent or metastatic head and neck squamous cell carcinoma (R/M-HNSCC) previously treated with both platinum-based chemotherapy and anti-programmed cell death protein 1 (PD-1) antibody. Retrospective studies suggest that previous treatment with immune checkpoint inhibitor might augment the efficacy of subsequent chemotherapy. Here, we conducted a phase II trial aimed to evaluate the efficacy and safety of paclitaxel plus biweekly cetuximab for patients in this setting. PATIENTS AND METHODS: This was a single-arm, multicenter, phase II trial. Key eligibility criteria were R/M-HNSCC, and previous treatment with both platinum-based chemotherapy and PD-1 antibody. Paclitaxel plus biweekly cetuximab consisted of weekly paclitaxel 100 mg/m2 (days 1, 8, 15) and biweekly cetuximab 500 mg/m2 (days 1, 15) with a cycle of 28 days until progression or unacceptable toxicity. Primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS), overall survival (OS), disease control rate (DCR), and adverse events (AEs) (Common Terminology Criteria for Adverse Events version 5.0). RESULTS: Between August 2020 and August 2022, 35 patients were enrolled, of whom 33 were assessable for response. ORR was 69.6% (95% confidence interval 51.2% to 84.4%). With a median follow-up period for survivors of 16.6 months, median PFS and OS were 5.5 and 13.3 months, respectively. DCR was 93.7%. Twenty-three patients (65%) experienced grade 3 or 4 AEs, including neutropenia (34%), infection (14%), leukopenia (11%), mucositis (8%), and pneumonitis (8%). Eight patients discontinued study treatment due to treatment-related AEs, and no treatment-related death was observed. CONCLUSIONS: Paclitaxel plus biweekly cetuximab showed highly encouraging efficacy and manageable toxicities in R/M-HNSCC patients previously treated with both platinum-based chemotherapy and PD-1 antibody. This combination therapy warrants further investigation in this setting.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Cétuximab , Tumeurs de la tête et du cou , Paclitaxel , Humains , Cétuximab/administration et posologie , Cétuximab/usage thérapeutique , Cétuximab/pharmacologie , Paclitaxel/usage thérapeutique , Paclitaxel/administration et posologie , Paclitaxel/pharmacologie , Mâle , Adulte d'âge moyen , Femelle , Sujet âgé , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Adulte , Récidive tumorale locale/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/administration et posologie
14.
In Vivo ; 38(4): 1891-1899, 2024.
Article de Anglais | MEDLINE | ID: mdl-38936932

RÉSUMÉ

BACKGROUND: Treatment options are limited, and the prognosis is poor for patients with platinum-resistant recurrent metastatic (R/M) head and neck squamous cell carcinoma (HNSCC). This study evaluated the efficacy and safety of a paclitaxel and ifosfamide (TI) regimen in patients with R/M HNSCC whose disease had progressed following platinum-based therapy. PATIENTS AND METHODS: In this retrospective study, we included 53 patients with R/M HNSCC who underwent at least one cycle of TI-based therapy, post platinum failure, between February 2020 and August 2023. Some patients received the TI regimen in combination with immunotherapy and/or cetuximab. Key metrics assessed included the objective response rate (ORR), disease control rate, and progression-free as well as overall survival. RESULTS: The study observed an ORR of 15.8% and a disease control rate of 36.8%. The median progression-free survival for the entire cohort was 3.3 months, and the median overall survival was 9.6 months. Notably, the combination of TI with immunotherapy yielded a higher ORR of 30.8%, compared to 14.3% with TI alone. The most prevalent grade 1-2 adverse events were anemia (81%), weight loss (68%) and hypernatremia (55%). CONCLUSION: The TI-based regimen demonstrated favorable efficacy and safety profile in treating R/M HNSCC. Enhanced outcomes may be attainable when combining it with immunotherapy. This study suggests that TI-based therapy could serve as a potential salvage option for this specific patient group.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Résistance aux médicaments antinéoplasiques , Tumeurs de la tête et du cou , Ifosfamide , Récidive tumorale locale , Paclitaxel , Thérapie de rattrapage , Humains , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/mortalité , Paclitaxel/administration et posologie , Paclitaxel/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/anatomopathologie , Adulte , Ifosfamide/usage thérapeutique , Ifosfamide/administration et posologie , Études rétrospectives , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/mortalité , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Platine/usage thérapeutique , Métastase tumorale , Sujet âgé de 80 ans ou plus , Résultat thérapeutique
15.
Chin Med Sci J ; 39(2): 111-121, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38887993

RÉSUMÉ

Objective To identify nivolumab resistance-related genes in patients with head and neck squamous cell carcinoma (HNSCC) using single-cell and bulk RNA-sequencing data. Methods The single-cell and bulk RNA-sequencing data downloaded from the Gene Expression Omnibus database were analyzed to screen out differentially expressed genes (DEGs) between nivolumab resistant and nivolumab sensitive patients using R software. The Least Absolute Shrinkage Selection Operator (LASSO) regression and Recursive Feature Elimination (RFE) algorithm were performed to identify key genes associated with nivolumab resistance. Functional enrichment of DEGs was analyzed with Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. The relationships of key genes with immune cell infiltration, differentation trajectory, dynamic gene expression profiles, and ligand-receptor interaction were explored. Results We found 83 DEGs. They were mainly enriched in T-cell differentiation, PD-1 and PD-L1 checkpoint, and T-cell receptor pathways. Among six key genes identified using machine learning algorithms, only PPP1R14A gene was differentially expressed between the nivolumab resistant and nivolumab sensitive groups both before and after immunotherapy (P < 0.05). The high PPP1R14A gene expression group had lower immune score (P < 0.01), higher expression of immunosuppressive factors (such as PDCD1, CTLA4, and PDCD1LG2) (r > 0, P < 0.05), lower differentiation of infiltrated immune cells (P < 0.05), and a higher degree of interaction between HLA and CD4 (P < 0.05). Conclusions PPP1R14A gene is closely associated with resistance to nivolumab in HNSCC patients. Therefore, PPP1R14A may be a target to ameliorate nivolumab resistance of HNSCC patients.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Tumeurs de la tête et du cou , Immunothérapie , Nivolumab , Analyse de séquence d'ARN , Carcinome épidermoïde de la tête et du cou , Humains , 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/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/génétique , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/thérapie , Nivolumab/usage thérapeutique , Analyse sur cellule unique , Régulation de l'expression des gènes tumoraux
16.
Cell Mol Life Sci ; 81(1): 282, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38943031

RÉSUMÉ

Cetuximab resistance has been a major challenge for head and neck squamous cell carcinoma (HNSCC) patients receiving targeted therapy. However, the mechanism that causes cetuximab resistance, especially microRNA (miRNA) regulation, remains unclear. Growing evidence suggests that miRNAs may act as "nuclear activating miRNAs" for targeting promoter regions or enhancers related to target genes. This study elucidates a novel mechanism underlying cetuximab resistance in HNSCC involving the nuclear activation of KDM7A transcription via miR-451a. Herein, small RNA sequencing, quantitative real-time polymerase chain reaction (qRT‒PCR) and fluorescence in situ hybridization (FISH) results provided compelling evidence of miR-451a nuclear enrichment in response to cetuximab treatment. Chromatin isolation via RNA purification, microarray analysis, and bioinformatic analysis revealed that miR-451a interacts with an enhancer region in KDM7A, activating its expression and further facilitating cetuximab resistance. It has also been demonstrated that the activation of KDM7A by nuclear miR-451a is induced by cetuximab treatment and is AGO2 dependent. Logistic regression analyses of 87 HNSCC samples indicated the significance of miR-451a and KDM7A in the development of cetuximab resistance. These discoveries support the potential of miR-451a and KDM7A as valuable biomarkers for cetuximab resistance and emphasize the function of nuclear-activating miRNAs.


Sujet(s)
Cétuximab , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Tumeurs de la tête et du cou , microARN , Carcinome épidermoïde de la tête et du cou , Humains , microARN/génétique , microARN/métabolisme , Cétuximab/pharmacologie , Résistance aux médicaments antinéoplasiques/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/anatomopathologie , Carcinome épidermoïde de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Jumonji Domain-Containing Histone Demethylases/génétique , Jumonji Domain-Containing Histone Demethylases/métabolisme , Protéines Argonaute/génétique , Protéines Argonaute/métabolisme , Animaux , Souris , Noyau de la cellule/métabolisme , Noyau de la cellule/génétique , Femelle , Souris nude
17.
Biomed Pharmacother ; 176: 116906, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38876051

RÉSUMÉ

Targeting metabolic reprogramming may be an effective strategy to enhance cancer treatment efficacy. Glutamine serves as a vital nutrient for cancer cells. Inhibiting glutamine metabolism has shown promise in preventing tumor growth both in vivo and in vitro through various mechanisms. Therefore, this review collates recent scientific literature concerning the correlation between glutamine metabolism and cancer treatment. Novel treatment modalities based on amino acid transporters, metabolites, and glutaminase are discussed. Moreover, we demonstrate the relationship between glutamine metabolism and tumor proliferation, drug resistance, and the tumor immune microenvironment, offering new perspectives for the clinical treatment of head and neck squamous cell carcinoma, particularly for combined therapies. Identifying innovative approaches for enhancing the efficacy of glutamine-based metabolic therapy is crucial to improving HNSCC treatment.


Sujet(s)
Glutamine , Tumeurs de la tête et du cou , Carcinome épidermoïde de la tête et du cou , Glutamine/métabolisme , Humains , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/anatomopathologie , Animaux , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Glutaminase/métabolisme , Glutaminase/antagonistes et inhibiteurs , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Thérapie moléculaire ciblée
18.
Int J Biol Sci ; 20(8): 2790-2813, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904007

RÉSUMÉ

Coenzyme Q0 (CoQ0), a quinone derivative from Antrodia camphorata, has antitumor capabilities. This study investigated the antitumor effect of noncytotoxic CoQ0, which included NLRP3 inflammasome inhibition, anti-EMT/metastasis, and metabolic reprogramming via HIF-1α inhibition, in HNSCC cells under normoxia and hypoxia. CoQ0 suppressed hypoxia-induced ROS-mediated HIF-1α expression in OECM-1 and SAS cells. Under normoxia and hypoxia, the inflammatory NLRP3, ASC/caspase-1, NFκB, and IL-1ß expression was reduced by CoQ0. CoQ0 reduced migration/invasion by enhancing epithelial marker E-cadherin and suppressing mesenchymal markers Twist, N-cadherin, Snail, and MMP-9, and MMP-2 expression. CoQ0 inhibited glucose uptake, lactate accumulation, GLUT1 levels, and HIF-1α-target gene (HK-2, PFK-1, and LDH-A) expressions that are involved in aerobic glycolysis. Notably, CoQ0 reduced ECAR as well as glycolysis, glycolytic capability, and glycolytic reserve and enhanced OCR, basal respiration, ATP generation, maximal respiration, and spare capacity in OECM-1 cells. Metabolomic analysis using LC-ESI-MS showed that CoQ0 treatment decreased the levels of glycolytic intermediates, including lactate, 2/3-phosphoglycerate, fructose 1,6-bisphosphate, and phosphoenolpyruvate, and increased the levels of TCA cycle metabolites, including citrate, isocitrate, and succinate. HIF-1α silencing reversed CoQ0-mediated anti-metastasis (N-Cadherin, Snail, and MMP-9) and metabolic reprogramming (GLUT1, HK-2, and PKM-2) under hypoxia. CoQ0 prevents cancer stem-like characteristics (upregulated CD24 expression and downregulated CD44, ALDH1, and OCT4) under normoxia and/or hypoxia. Further, in IL-6-treated SG cells, CoQ0 attenuated fibrosis by inhibiting TGF-ß and Collagen I expression and suppressed EMT by downregulating Slug and upregulating E-cadherin expression. Interesting, CoQ0 inhibited the growth of OECM-1 tumors in xenografted mice. Our results advocate CoQ0 for the therapeutic application against HNSCC.


Sujet(s)
Transition épithélio-mésenchymateuse , Sous-unité alpha du facteur-1 induit par l'hypoxie , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Carcinome épidermoïde de la tête et du cou , Ubiquinones , Humains , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Ubiquinones/analogues et dérivés , Ubiquinones/pharmacologie , Animaux , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Souris , Inflammasomes/métabolisme , Effet Warburg en oncologie/effets des médicaments et des substances chimiques , Souris nude , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/traitement médicamenteux
19.
Cancer Res Commun ; 4(7): 1748-1764, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38916448

RÉSUMÉ

Immune checkpoint inhibitors are effective first-line therapy for solid cancers. However, low response rate and acquired resistance over time has led to the need for additional therapeutic options. Here, we evaluated synergistic antitumor efficacy of EGFR × MET targeting bispecific antibody, amivantamab with PD-L1 immunotherapy, pembrolizumab in head and neck squamous cell carcinoma (HNSCC) and lung squamous cell carcinoma tumor-bearing humanized patient-derived xenograft (PDX) models. We demonstrated that pembrolizumab or amivantamab alone was ineffective and that combination treatment induced a significant reduction of tumor growth in both models (P < 0.0001 and P < 0.01, respectively). It appeared that combination of amivantamab and pembrolizumab significantly enhanced infiltration of granzyme B-producing CD8 T cells was in the TME of HNSCC PDX (P < 0.01) and enhanced neoantigen-associated central memory CD8 T cells in circulating immune cells. Analysis of single-cell RNA transcriptomics suggested that the tumor cells dramatically upregulated EGFR and MET in response to PD-L1 immunotherapy, potentially creating a metabolic state fit for tumor persistence in the tumor microenvironment (TME) and rendered pembrolizumab ineffective. We demonstrated that EGFRHIGHMETHIGH subcluster displayed an increased expression of genes implicated in production of lactate [SLC16A3 and lactate dehydrogenase A (LDHA)] compared to the EGFRLOWMETLOW cluster. Accumulation of lactate in the TME has been associated with immunosuppression by hindering the infiltration of tumor killing CD8 T and NK cells. This study proved that amivantamab reduced glycolytic markers in the EGFRHIGHMETHIGH subcluster including SLC16A3 and LDHA and highlighted remodeling of the TME by combination treatment, providing rationale for additional therapy of amivantamab with PD-1 immunotherapy. SIGNIFICANCE: Amivantamab in synergy with pembrolizumab effectively eradicated EGFRHIGHMETHIGH tumor subcluster in the tumor microenvironment of head and neck squamous cell carcinoma and overcame resistance against anti-PD-1 immunotherapy.


Sujet(s)
Anticorps monoclonaux humanisés , Tumeurs du poumon , Carcinome épidermoïde de la tête et du cou , Microenvironnement tumoral , Humains , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/administration et posologie , 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/anatomopathologie , Animaux , Souris , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Antigène CD274/métabolisme , Lignée cellulaire tumorale
20.
J Physiol Pharmacol ; 75(2): 205-213, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38736267

RÉSUMÉ

Camptothecin (CPT), a naturally occurring alkaloid derived from the Camptotheca acuminate plant, exerts anti-tumor properties. However, its specific impact on head and neck squamous cell carcinoma (HNSCC) remains uncertain. The study was to explore the action and mechanism of CPT on HNSCC cells. First, two HNSCC cell lines (FaDu and TU686) and a normal immortalized keratinocyte (HEK001) cell line, were exposed to a spectrum of CPT concentrations (ranging from 10 to 50 µM) for durations of 24 h and 48 h. Cell viability, proliferation, migration, and invasion were assessed by CCK-8 assay, EdU incorporation assay, wound healing assay and transwell assay. Subsequently, si-RAB27A or negative control (NC) was introduced into FaDu and TU686 cells through transfection, and the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway was manipulated with L740Y-P, an activator of this pathway. The expression of proliferating cell nuclear antigen (PCNA), E-cadherin, PI3K/AKT signaling factors and RAB27A were determined by Western blot analysis. RAB27A was detected by immunofluorescence assay. It was found that CPT significantly hindered the viability, proliferation (p<0.01), migration (p<0.001), and invasion (p<0.001) of FaDu and TU686 cells. At the molecular level, administration of CPT caused a decline in the expression of PCNA, P-PI3K, P-AKT, and RAB27A, alongside an elevation in E-cadherin levels within HNSCC cells (p<0.05, p<0.01 and p<0.001). Reducing RAB27A expression enhanced the suppressive impacts of CPT on HNSCC cell viability (p<0.05 and p<0.01), migration (p<0.001) and invasion (p<0.01), these effects that were reversed upon treatment with L740Y-P in HNSCC cells (p<0.001). In summary, our study highlights the efficacy of CPT in HNSCC, demonstrating its influence on cell processes via the RAB27A-mediated PI3K/AKT pathway.


Sujet(s)
Tumeurs de la tête et du cou , Protéines proto-oncogènes c-akt , Transduction du signal , Carcinome épidermoïde de la tête et du cou , Protéines rab27 liant le GTP , Humains , Antinéoplasiques d'origine végétale/pharmacologie , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines rab27 liant le GTP/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , 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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...