Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 22(1): 524, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822345

RESUMEN

BACKGROUND: Olfactory neuroblastoma is a rare malignancy of the anterior skull base typically treated with surgery and adjuvant radiation. Although outcomes are fair for low-grade disease, patients with high-grade, recurrent, or metastatic disease oftentimes respond poorly to standard treatment methods. We hypothesized that an in-depth evaluation of the olfactory neuroblastoma tumor immune microenvironment would identify mechanisms of immune evasion in high-grade olfactory neuroblastoma as well as rational targetable mechanisms for future translational immunotherapeutic approaches. METHODS: Multispectral immunofluorescence and RNAScope evaluation of the tumor immune microenvironment was performed on forty-seven clinically annotated olfactory neuroblastoma samples. A retrospective chart review was performed and clinical correlations assessed. RESULTS: A significant T cell infiltration was noted in olfactory neuroblastoma samples with a stromal predilection, presence of myeloid-derived suppressor cells, and sparse natural killer cells. A striking decrease was observed in MHC-I expression in high-grade olfactory neuroblastoma compared to low-grade disease, representing a mechanism of immune evasion in high-grade disease. Mechanistically, the immune effector stromal predilection appears driven by low tumor cell MHC class II (HLA-DR), CXCL9, and CXCL10 expression as those tumors with increased tumor cell expression of each of these mediators correlated with significant increases in T cell infiltration. CONCLUSION: These data suggest that immunotherapeutic strategies that augment tumor cell expression of MHC class II, CXCL9, and CXCL10 may improve parenchymal trafficking of immune effector cells in olfactory neuroblastoma and augment immunotherapeutic responses.


Asunto(s)
Quimiocina CXCL10 , Quimiocina CXCL9 , Estesioneuroblastoma Olfatorio , Antígenos HLA-DR , Inmunoterapia , Microambiente Tumoral , Humanos , Estesioneuroblastoma Olfatorio/terapia , Estesioneuroblastoma Olfatorio/patología , Estesioneuroblastoma Olfatorio/inmunología , Quimiocina CXCL10/metabolismo , Inmunoterapia/métodos , Femenino , Masculino , Persona de Mediana Edad , Quimiocina CXCL9/metabolismo , Microambiente Tumoral/inmunología , Antígenos HLA-DR/metabolismo , Anciano , Neoplasias Nasales/terapia , Neoplasias Nasales/patología , Neoplasias Nasales/inmunología , Adulto , Regulación Neoplásica de la Expresión Génica
2.
BMC Immunol ; 23(1): 5, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35148673

RESUMEN

BACKGROUND: Determining T cell responses to naturally processed and presented antigens is a critical immune correlate to determine efficacy of an investigational immunotherapeutic in clinical trials. In most cases, minimal epitopes and HLA restriction elements are unknown. RESULTS: Here, we detail the experimental use of ex vivo expanded autologous B cells as antigen presenting cells to overcome the limitation of unknown HLA restriction, and the use of electroporated full length mRNA encoding full length parental proteins to ensure that any observed T cell responses are specific for antigens that are naturally processed and presented. CONCLUSIONS: This technique can serve as useful experimental approach to determine the induction or enhancement of specific responses to naturally processed and presented antigens on HLA class I molecules in peripheral blood or tumor infiltrating T cells.


Asunto(s)
Presentación de Antígeno , Células Presentadoras de Antígenos , Antígenos de Neoplasias , Linfocitos B , Epítopos , Epítopos de Linfocito T , Linfocitos T
3.
Am J Otolaryngol ; 35(6): 747-52, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25097181

RESUMEN

PURPOSE: To compare post-operative complication rates between inpatient and outpatient endoscopic airway surgery in patients with laryngotracheal stenosis. Secondary objectives included characterization of a cohort of patients with this disease. METHODS: Retrospective review of patients with laryngotracheal stenosis in a tertiary care laryngology practice over a 5-year period. RESULTS: Ninety-one patients underwent 223 endoscopic airway surgeries. Of 114 outpatient interventions, 1 patient (0.8%) sought emergent medical care following discharge for respiratory distress. Of 109 procedures resulting in admission, no patients required transfer to a higher level of care, endotracheal intubation or placement of a surgical airway. There was no statistically significant difference in complication rates between patients treated as outpatients or inpatients (p=0.33, chi square). There were no cardiopulmonary events. There were no pneumothoraces despite frequent use of jet ventilation. The most common etiologic category was idiopathic (58%), followed by granulomatosis with polyangiitis (16%) and history of tracheotomy (12%). Most patients with idiopathic disease were female (p<0.001, Fisher's exact test). CONCLUSION: Patients undergoing endoscopic surgery for airway stenosis rarely have post-operative complications, and outpatient surgery appears to be a safe alternative to post-operative admission and observation.


Asunto(s)
Procedimientos Quirúrgicos Ambulatorios , Endoscopía , Laringoestenosis/cirugía , Estenosis Traqueal/cirugía , Adulto , Endoscopía/efectos adversos , Femenino , Hospitalización , Humanos , Masculino , Complicaciones Posoperatorias/epidemiología , Estudios Retrospectivos , Medición de Riesgo
4.
Head Neck ; 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38622975

RESUMEN

BACKGROUND: Neutrophilic cells are among the most abundant immune populations within the head and neck tumor microenvironment (TME) and harbor multiple mechanisms of immunosuppression. Despite these important features, neutrophilic cells may be underrepresented in contemporary studies that aim to comprehensively characterize the immune landscape of the TME due to discrepancies in tissue processing and analysis techniques. Here, we review the role of pathologically activated neutrophilic cells within the TME and pitfalls of various approaches used to study their frequency and function in clinical samples. METHODS: The literature was identified by searching PubMed for "immune landscape" and "tumor immune microenvironment" in combination with keywords describing solid tumor malignancies. Key publications that assessed the immune composition of solid tumors derived from human specimens were included. The tumor and blood processing methodologies in each study were reviewed in depth and correlated with the reported abundance of neutrophilic cells. RESULTS: Neutrophilic cells do not survive cryopreservation, and many studies fail to identify and study neutrophilic cell populations due to cryopreservation of clinical samples for practical reasons. Additional single-cell transcriptomic studies filter out neutrophilic cells due to low transcriptional counts. CONCLUSIONS: This report can help readers critically interpret studies aiming to comprehensively study the immune TME that fail to identify and characterize neutrophilic cells.

5.
Oral Oncol ; 150: 106705, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38280289

RESUMEN

OBJECTIVES: Tumor infiltrating neutrophils suppress T cell function, but whether neutrophils in circulation contribute to systemic immunosuppression is unclear. We aimed to study whether peripheral neutrophils that accumulate with tumor progression contribute to systemic immunosuppression, and if observed suppression of systemic anti-tumor immunity could be reversed with complete surgical tumor removal. MATERIALS AND METHODS: Syngeneic murine oral cancers were established in immunocompetent mice. Proteomic and functional immune assays were used to study plasma cytokine concentration, peripheral immune frequencies, and systemic anti-tumor immunity with and without complete primary tumor resection. RESULTS: Ly6G+ neutrophilic cells, but not other myeloid cell types, accumulated in the periphery of mice with progressing tumors. This accumulation positively associated with plasma G-CSF concentration. Circulating neutrophils were functionally immunosuppressive. Complete surgical tumor removal reversed the observed neutrophilia, with neutrophil frequencies returning to baseline in 21 days. Multiple independent functional assays revealed enhanced systemic anti-tumor immunity in mice following tumor resection compared to tumor-bearing mice, and the observed enhanced systemic immunity could be reproduced with selective neutrophil depletion. CONCLUSIONS: Complete primary tumor resection can reverse neutrophilia that develops during tumor progression and result in enhanced systemic anti-tumor immunity. Primary tumor removal relieves neutrophil-driven systemic immunosuppression and may itself contribute to the clinical benefit observed with neoadjuvant immunotherapy.


Asunto(s)
Terapia de Inmunosupresión , Proteómica , Animales , Ratones , Línea Celular Tumoral , Inmunoterapia , Tolerancia Inmunológica , Microambiente Tumoral
6.
Laryngoscope ; 134(7): 3238-3244, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38436434

RESUMEN

OBJECTIVES: To characterize the distribution of immune cell subsets within laryngeal papillomas and to study the function of potentially immunosuppressive neutrophilic and regulatory T cells (Tregs). METHODS: Fresh clinical papilloma specimens were collected at the time of surgery and studied with multiparameter flow cytometry. Papilloma infiltrating neutrophilic cells and Tregs were sorted and studied functionally with ex vivo T cell suppression assays. RESULTS: Flow cytometric analysis of fresh laryngeal papillomas samples from 18 adult patients with recurrent respiratory papillomatosis revealed patterns in immune constituency between patients. Clearly divergent phenotypes based primarily on the degree of neutrophilic and T cell infiltration were identified. Relative neutrophilic cell enrichment and T cell depletion were observed in 50% of samples and neutrophilic cell depletion and T cell enrichment were observed in the others. Greater papilloma neutrophilic cell enrichment was positively associated with the number of clinically indicated interventions required in the 12 months prior to sample collection, linking papilloma neutrophil inflammation to disease severity. Functional assays revealed the ability of both papilloma infiltrating neutrophilic and Tregs to suppress T cell function at roughly equal magnitudes, but substantially increased infiltration of neutrophilic cells compared to Tregs across samples. CONCLUSION: Neutrophilic cells are an important contributor to immunosuppression within the respiratory papilloma microenvironment. Given these data and the association between greater neutrophilic cell infiltration and lack of clinical response to therapeutic vaccination, additional study of strategies aimed at limiting neutrophilic cell infiltration or function within papillomas is warranted. LEVEL OF EVIDENCE: 4 Laryngoscope, 134:3238-3244, 2024.


Asunto(s)
Citometría de Flujo , Neutrófilos , Papiloma , Fenotipo , Linfocitos T Reguladores , Humanos , Neutrófilos/inmunología , Neutrófilos/patología , Linfocitos T Reguladores/inmunología , Masculino , Adulto , Papiloma/patología , Papiloma/inmunología , Femenino , Persona de Mediana Edad , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/patología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/patología , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/inmunología , Microambiente Tumoral/inmunología , Anciano
7.
Cancer Med ; 13(7): e7146, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38581118

RESUMEN

BACKGROUND: De-escalation strategies for newly-diagnosed p16-positive oropharyngeal squamous cell carcinoma (p16+ OPSCC), aim to reduce treatment-related morbidity without compromising disease control. One strategy is neoadjuvant cisplatin and docetaxel chemotherapy (NAC + S) before transoral robotic surgery, with pathology-based risk-adapted adjuvant treatment. METHODS: We examined the recurrence-free survival (RFS) for patients who received NAC + S. RESULTS: Comparing outcomes in 103 patients between 2008 and 2023, 92% avoided adjuvant treatment and showed significantly higher 2-year recurrence-free survival (RFS) compared to those with adjuvant treatment (95.9% vs. 43.8%, p = 0.0049) CONCLUSION: Our findings suggest that pathology-based risk-adapted omission of adjuvant treatment following NAC + S does not appear to elevate recurrence risk and that NAC may identify patients with favorable tumor biology, yielding a 2-year RFS probability exceeding 95% without adjuvant treatment. Further, the study identifies a patient subset experiencing disease recurrence despite triple modality therapy. Despite limitations, including a retrospective design and modest sample size, the data advocate for controlled NAC + S studies.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias Orofaríngeas , Procedimientos Quirúrgicos Robotizados , Humanos , Terapia Neoadyuvante , Estudios Retrospectivos , Procedimientos Quirúrgicos Robotizados/efectos adversos , Carcinoma de Células Escamosas/cirugía , Recurrencia Local de Neoplasia/prevención & control , Neoplasias Orofaríngeas/cirugía , Quimioterapia Adyuvante , Neoplasias de Cabeza y Cuello/etiología
8.
Front Oncol ; 14: 1376622, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38741774

RESUMEN

Introduction: Cancer stem cells (CSCs), a group of tumor-initiating and tumor-maintaining cells, may be major players in the treatment resistance and recurrence distinctive of chordoma. Characterizing CSCs is crucial to better targeting this subpopulation. Methods: Using flow cytometry, six chordoma cell lines were evaluated for CSC composition. In vitro, cell lines were stained for B7H6, HER2, MICA-B, ULBP1, EGFR, and PD-L1 surface markers. Eighteen resected chordomas were stained using a multispectral immunofluorescence (mIF) antibody panel to identify CSCs in vivo. HALO software was used for quantitative CSC density and spatial analysis. Results: In vitro, chordoma CSCs express more B7H6, MICA-B, and ULBP1, assessed by percent positivity and mean fluorescence intensity (MFI), as compared to non-CSCs in all cell lines. PD- L1 percent positivity is increased by >20% in CSCs compared to non-CSCs in all cell lines except CH22. In vivo, CSCs comprise 1.39% of chordoma cells and most are PD-L1+ (75.18%). A spatial analysis suggests that chordoma CSCs cluster at an average distance of 71.51 mm (SD 73.40 mm) from stroma. Discussion: To our knowledge, this study is the first to identify individual chordoma CSCs and describe their surface phenotypes using in vitro and in vivo methods. PD-L1 is overexpressed on CSCs in chordoma human cell lines and operative tumor samples. Similarly, potential immunotherapeutic targets on CSCs, including B7H6, MICA-B, ULBP1, EGFR, and HER2 are overexpressed across cell lines. Targeting these markers may have a preferential role in combating CSCs, an aggressive subpopulation likely consequential to chordoma's high recurrence rate.

9.
Ann Otol Rhinol Laryngol ; 122(11): 672-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24358626

RESUMEN

We performed a retrospective chart review to examine and describe our clinical experience of use of the Lichtenberger technique to place silicone elastomer keels after lysis of existing webs or for prevention of webs following anterior commissure surgery in adults. Twenty-two patients were identified for inclusion, ranging in age from 24 to 80 years. For 18 patients with existing glottic webs,the surgical procedure involved laryngoscopy with complete lysis of the anterior glottic web by laser or sharp technique, followed by placement of a square of silicone elastomer that is sutured in place with the Lichtenberger needle holder and left in place for 3 to 5 weeks. The procedure was well tolerated, and successfully corrected the web in all but 2 cases. For 4 patients, the procedure was performed prophylactically at the time of anterior commissure surgery considered high-risk for web formation. The procedure does not require a tracheotomy, and patients can maintain a normal diet and have functional phonation while the keel is in place. This approach to treating anterior glottic webs offers several advantages over traditional open thyrotomy with keel placement and should be considered to treat or prevent anterior glottic webs.


Asunto(s)
Glotis/cirugía , Enfermedades de la Laringe/cirugía , Laringoscopía/métodos , Implantación de Prótesis/métodos , Elastómeros de Silicona , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Estudios de Seguimiento , Humanos , Enfermedades de la Laringe/diagnóstico , Laringoestenosis/diagnóstico , Laringoestenosis/prevención & control , Masculino , Persona de Mediana Edad , Diseño de Prótesis , Estudios Retrospectivos , Adulto Joven
10.
Oral Oncol ; 146: 106570, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37738775

RESUMEN

INTRODUCTION: Resident memory T (TRM) cells are embedded in peripheral tissue and capable of acting as sentinels that can respond quickly to repeat pathogen exposure as part of an endogenous anti-microbial immune response. Recent evidence suggests that chronic antigen exposure and other microenvironment cues may promote the development of TRM cells within solid tumors as well, and that this TRM phenotype can sequester tumor-specific T cells into tumors and out of circulation resulting in limited systemic antitumor immunity. Here, we perform a review of the published English literature and describe tissue-specific mediators of TRM cell differentiation in states of infection and malignancy with special focus on the role of TGF-ß and how targeting TGF-ß signaling could be used as a therapeutical approach to promote tumor systemic immunity. DISCUSSION: The presence of TRM cells with antigen specificity to neoepitopes in tumors associates with positive clinical prognosis and greater responsiveness to immunotherapy. Recent evidence indicates that solid tumors may act as reservoirs for tumor specific TRM cells and limit their circulation - possibly resulting in impaired systemic antitumor immunity. TRM cells utilize specific mechanisms to egress from peripheral tissues into circulation and other peripheral sites, and emerging evidence indicates that immunotherapeutic approaches may initiate these processes and increase systemic antitumor immunity. CONCLUSIONS: Reversing tumor sequestration of tumor-specific T cells prior to surgical removal or radiation of tumor may increase systemic antitumor immunity. This finding may underlie the improved recurrence free survival observed with neoadjuvant immunotherapy in clinical trials.


Asunto(s)
Memoria Inmunológica , Neoplasias , Humanos , Células T de Memoria , Terapia Neoadyuvante , Inmunoterapia , Neoplasias/terapia , Factor de Crecimiento Transformador beta , Microambiente Tumoral
11.
Oral Oncol ; 138: 106309, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36682187

RESUMEN

OBJECTIVES: Biomarkers are needed to identify patients likely to respond to neoadjuvant immunotherapy (NIT) prior to receiving definitive treatment. MATERIALS AND METHODS: We hypothesized that expression of tumor cell HLA class I would correlate with pathologic response (PR) following NIT for primary untreated head and neck cancer. Multispectral immunofluorescence of pre- and post-treatment biopsy specimens from a neoadjuvant study of bintrafusp alfa, a dual TGF-ß and PD-L1 inhibitor, was performed. RESULTS: Discordant expression of tumor cell HLA class I and PD-L1 measured by multispectral immunofluorescence was observed with most positive tumor cells expressing HLA class I or PD-L1 but not both. Spatial analysis revealed colocalization between tumor parenchyma T cells and HLA class I positive tumors cells, but no clear colocalization between T cells and PD-L1 positive tumor cells. Greater pre-treatment tumor cell HLA class I expression, but not PD-L1 expression or tumor T cell infiltration, correlated with the development of a PR. Additionally, increased tumor cell HLA class I expression after NIT compared to before NIT correlated with development of a PR, whereas inconsistent changes in PD-L1 and T cell infiltration were observed after treatment in all patients. CONCLUSIONS: These data provide the rationale for the study of tumor cell HLA class I expression in larger prospective studies powered to determine the performance of biomarkers of PR in newly diagnosed HNSCC patients receiving NIT.


Asunto(s)
Neoplasias de Cabeza y Cuello , Antígenos de Histocompatibilidad Clase I , Humanos , Terapia Neoadyuvante , Estudios Prospectivos , Biomarcadores , Inmunoterapia , Antígeno B7-H1/metabolismo
12.
Head Neck ; 45(9): 2294-2302, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37480219

RESUMEN

BACKGROUND: Treatment of patients with newly diagnosed HPV-associated oropharyngeal squamous cell carcinoma (OPSCC) with neoadjuvant chemotherapy (NAC) results in a high rate of 5-year recurrence free survival with few patients requiring adjuvant treatment. We hypothesized that NAC enhances primary tumor HPV-specific T cell responses. METHODS: HPV-specific responses in tumor infiltrating lymphocytes (TILs) before and after NAC were determined using autologous co-culture assays. RESULTS: Greater HPV16-specific TIL responses, sometimes polyclonal, were observed after NAC compared to before in 8 of 10 patients (80%) with PCR-verified HPV16-positive tumors. A significant association was observed between net-negative change in HPV-specific TIL response and disease relapse (p = 0.04, Mann-Whitney test), whereas pathologic complete response at time of surgery did not correlate with recurrence. CONCLUSIONS: NAC induces HPV-specific tumor T cell responses in patients with newly diagnosed HPV-associated OPSCC; whereas lack of an increase following NAC may associate with risk of relapse.


Asunto(s)
Neoplasias de Cabeza y Cuello , Neoplasias Orofaríngeas , Infecciones por Papillomavirus , Humanos , Linfocitos T , Pronóstico , Terapia Neoadyuvante/métodos , Infecciones por Papillomavirus/complicaciones , Recurrencia Local de Neoplasia , Neoplasias Orofaríngeas/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/complicaciones , Neoplasias de Cabeza y Cuello/complicaciones
13.
Cancers (Basel) ; 15(4)2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36831373

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) remains a prevalent diagnosis with current treatment options that include radiotherapy and immune-mediated therapies, in which tumor necrosis factor-α (TNFα) is a key mediator of cytotoxicity. However, HNSCC and other cancers often display TNFα resistance due to activation of the canonical IKK-NFκB/RELA pathway, which is activated by, and induces expression of, cellular inhibitors of apoptosis proteins (cIAPs). Our previous studies have demonstrated that the IAP inhibitor birinapant sensitized HNSCC to TNFα-dependent cell death in vitro and radiotherapy in vivo. Furthermore, we recently demonstrated that the inhibition of the G2/M checkpoint kinase WEE1 also sensitized HNSCC cells to TNFα-dependent cell death, due to the inhibition of the pro-survival IKK-NFκB/RELA complex. Given these observations, we hypothesized that dual-antagonist therapy targeting both IAP and WEE1 proteins may have the potential to synergistically sensitize HNSCC to TNFα-dependent cell death. Using the IAP inhibitor birinapant and the WEE1 inhibitor AZD1775, we show that combination treatment reduced cell viability, proliferation and survival when compared with individual treatment. Furthermore, combination treatment enhanced the sensitivity of HNSCC cells to TNFα-induced cytotoxicity via the induction of apoptosis and DNA damage. Additionally, birinapant and AZD1775 combination treatment decreased cell proliferation and survival in combination with radiotherapy, a critical source of TNFα. These results support further investigation of IAP and WEE1 inhibitor combinations in preclinical and clinical studies in HNSCC.

14.
Front Immunol ; 14: 1200970, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37638000

RESUMEN

Introduction: Amplification of human chromosome 3q26-29, which encodes oncoprotein ΔNp63 among other isoforms of the p63 family, is a feature common to squamous cell carcinomas (SCCs) of multiple tissue origins. Along with overexpression of ΔNp63, activation of the protooncogene, RAS, whether by overexpression or oncogenic mutation, is frequently observed in many cancers. In this study, analysis of transcriptome data from The Cancer Genome Atlas (TCGA) demonstrated that expression of TP63 mRNA, particularly ΔNp63 isoforms, and HRAS are significantly elevated in advanced squamous cell carcinomas of the head and neck (HNSCCs), suggesting pathological significance. However, how co-overexpressed ΔNp63 and HRAS affect the immunosuppressive tumor microenvironment (TME) is incompletely understood. Methods: Here, we established and characterized an immune competent mouse model using primary keratinocytes with retroviral-mediated overexpression of ΔNp63α and constitutively activated HRAS (v-rasHa G12R) to evaluate the role of these oncogenes in the immune TME. Results: In this model, orthotopic grafting of wildtype syngeneic keratinocytes expressing both v-rasHa and elevated levels of ΔNp63α consistently yield carcinomas in syngeneic hosts, while cells expressing v-rasHa alone yield predominantly papillomas. We found that polymorphonuclear (PMN) myeloid cells, experimentally validated to be immunosuppressive and thus representing myeloid-derived suppressor cells (PMN-MDSCs), were significantly recruited into the TME of carcinomas arising early following orthotopic grafting of ΔNp63α/v-rasHa-expressing keratinocytes. ΔNp63α/v-rasHa-driven carcinomas expressed higher levels of chemokines implicated in recruitment of MDSCs compared to v-rasHa-initiated tumors, providing a heretofore undescribed link between ΔNp63α/HRAS-driven carcinomas and the development of an immunosuppressive TME. Conclusion: These results support the utilization of a genetic carcinogenesis model harboring specific genomic drivers of malignancy to study mechanisms underlying the development of local immunosuppression.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Células Supresoras de Origen Mieloide , Humanos , Animales , Ratones , Carcinoma de Células Escamosas/genética , Inmunosupresores , Carcinoma de Células Escamosas de Cabeza y Cuello , Modelos Animales de Enfermedad , Microambiente Tumoral/genética
15.
Cancer Cell ; 41(5): 887-902.e5, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-37059104

RESUMEN

Neoadjuvant immunotherapies (NITs) have led to clinical benefits in several cancers. Characterization of the molecular mechanisms underlying responses to NIT may lead to improved treatment strategies. Here we show that exhausted, tumor-infiltrating CD8+ T (Tex) cells display local and systemic responses to concurrent neoadjuvant TGF-ß and PD-L1 blockade. NIT induces a significant and selective increase in circulating Tex cells associated with reduced intratumoral expression of the tissue-retention marker CD103. TGF-ß-driven CD103 expression on CD8+ T cells is reversed following TGF-ß neutralization in vitro, implicating TGF-ß in T cell tissue retention and impaired systemic immunity. Transcriptional changes implicate T cell receptor signaling and glutamine metabolism as important determinants of enhanced or reduced Tex treatment response, respectively. Our analysis illustrates physiological and metabolic changes underlying T cell responses to NIT, highlighting the interplay between immunosuppression, tissue retention, and systemic anti-tumor immunity and suggest antagonism of T cell tissue retention as a promising neoadjuvant treatment strategy.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias de Cabeza y Cuello , Humanos , Terapia Neoadyuvante , Neoplasias de Cabeza y Cuello/terapia , Neoplasias de Cabeza y Cuello/metabolismo , Inmunoterapia , Factor de Crecimiento Transformador beta/metabolismo , Adaptación Fisiológica , Linfocitos Infiltrantes de Tumor
16.
Cell Death Differ ; 30(5): 1382-1396, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37055579

RESUMEN

TNFα is a key mediator of immune, chemotherapy and radiotherapy-induced cytotoxicity, but several cancers, including head and neck squamous cell carcinomas (HNSCC), display resistance to TNFα due to activation of the canonical NFκB pro-survival pathway. However, direct targeting of this pathway is associated with significant toxicity; thus, it is vital to identify novel mechanism(s) contributing to NFκB activation and TNFα resistance in cancer cells. Here, we demonstrate that the expression of proteasome-associated deubiquitinase USP14 is significantly increased in HNSCC and correlates with worse progression free survival in Human Papillomavirus (HPV)- HNSCC. Inhibition or depletion of USP14 inhibited the proliferation and survival of HNSCC cells. Further, USP14 inhibition reduced both basal and TNFα-inducible NFκB activity, NFκB-dependent gene expression and the nuclear translocation of the NFκB subunit RELA. Mechanistically, USP14 bound to both RELA and IκBα and reduced IκBα K48-ubiquitination leading to the degradation of IκBα, a critical inhibitor of the canonical NFκB pathway. Furthermore, we demonstrated that b-AP15, an inhibitor of USP14 and UCHL5, sensitized HNSCC cells to TNFα-mediated cell death, as well as radiation-induced cell death in vitro. Finally, b-AP15 delayed tumor growth and enhanced survival, both as a monotherapy and in combination with radiation, in HNSCC tumor xenograft models in vivo, which could be significantly attenuated by TNFα depletion. These data offer new insights into the activation of NFκB signaling in HNSCC and demonstrate that small molecule inhibitors targeting the ubiquitin pathway warrant further investigation as a novel therapeutic avenue to sensitize these cancers to TNFα- and radiation-induced cytotoxicity.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Inhibidor NF-kappaB alfa/genética , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/genética , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/radioterapia , FN-kappa B , Muerte Celular , Línea Celular Tumoral , Ubiquitina Tiolesterasa/genética
17.
Sci Transl Med ; 15(719): eadj0740, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37878675

RESUMEN

Recurrent respiratory papillomatosis (RRP) is a rare, debilitating neoplastic disorder caused by chronic infection with human papillomavirus (HPV) type 6 or 11 and characterized by growth of papillomas in the upper aerodigestive tract. There is no approved medical therapy, and patients require repeated debulking procedures to maintain voice and airway function. PRGN-2012 is a gorilla adenovirus immune-therapeutic capable of enhancing HPV 6/11-specific T cell immunity. This first-in-human, phase 1 study (NCT04724980) of adjuvant PRGN-2012 treatment in adult patients with severe, aggressive RRP demonstrates the overall safety and clinically meaningful benefit observed with PRGN-2012, with a 50% complete response rate in patients treated at the highest dose. Responders demonstrate greater expansion of peripheral HPV-specific T cells compared with nonresponders. Additional correlative studies identify an association between reduced baseline papilloma HPV gene expression, greater interferon responses and expression of CXCL9 and CXCL10, and greater papilloma T cell infiltration in responders. Conversely, nonresponders were characterized by greater HPV and CXCL8 gene expression, increased neutrophilic cell infiltration, and reduced T cell papilloma infiltration. These results suggest that papilloma HPV gene expression may regulate interferon signaling and chemokine expression profiles within the tumor microenvironment that cooperate to govern clinical response to therapeutic HPV vaccination in patients with respiratory papillomatosis.


Asunto(s)
Papiloma , Infecciones por Papillomavirus , Infecciones del Sistema Respiratorio , Adulto , Humanos , Infecciones por Papillomavirus/terapia , Infecciones por Papillomavirus/patología , Microambiente Tumoral , Infecciones del Sistema Respiratorio/terapia , Interferones , Papiloma/terapia , Papiloma/patología , Vacunación
18.
Cell Rep ; 42(7): 112823, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37463106

RESUMEN

Cancers often display immune escape, but the mechanisms are incompletely understood. Herein, we identify SMYD3 as a mediator of immune escape in human papilloma virus (HPV)-negative head and neck squamous cell carcinoma (HNSCC), an aggressive disease with poor response to immunotherapy with pembrolizumab. SMYD3 depletion induces upregulation of multiple type I interferon (IFN) response and antigen presentation machinery genes in HNSCC cells. Mechanistically, SMYD3 binds to and regulates the transcription of UHRF1, encoding for a reader of H3K9me3, which binds to H3K9me3-enriched promoters of key immune-related genes, recruits DNMT1, and silences their expression. SMYD3 further maintains the repression of immune-related genes through intragenic deposition of H4K20me3. In vivo, Smyd3 depletion induces influx of CD8+ T cells and increases sensitivity to anti-programmed death 1 (PD-1) therapy. SMYD3 overexpression is associated with decreased CD8 T cell infiltration and poor response to neoadjuvant pembrolizumab. These data support combining SMYD3 depletion strategies with checkpoint blockade to overcome anti-PD-1 resistance in HPV-negative HNSCC.


Asunto(s)
Neoplasias de Cabeza y Cuello , N-Metiltransferasa de Histona-Lisina , Interferón Tipo I , Infecciones por Papillomavirus , Carcinoma de Células Escamosas de Cabeza y Cuello , Humanos , Proteínas Potenciadoras de Unión a CCAAT , Linfocitos T CD8-positivos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , N-Metiltransferasa de Histona-Lisina/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Ubiquitina-Proteína Ligasas
19.
Head Neck ; 44(10): E31-E37, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35815785

RESUMEN

BACKGROUND: Immune checkpoint blockade can provide clinical benefit for patients with advanced cancer. Here, we report durable disease control over many years following PD-L1 blockade through induction of a viral antigen-specific T cell response in an adult patient with recurrent respiratory papillomatosis. METHODS: Antigen-specific T cell response assays, single cell RNA-sequencing, and RNA-scope was used to study clinical tissues. RESULTS: An HPV6 E2-specific T cell clone restricted to HLA-B*55, present at low frequency in the pre-treatment papilloma, significantly expanded after six doses of PD-L1 blockade and remained present and functional at the site of initial response in the larynx as a tissue resident memory T cell for 4 years. An associated reduction in E2 target gene was observed following treatment. CONCLUSIONS: Although demonstrated in a single exceptional responder, these results highlight that immune checkpoint blockade may induce durable, viral antigen-specific immunity of sufficient magnitude to control disease in patients with nonmalignant disorders.


Asunto(s)
Antígeno B7-H1 , Papiloma , Adulto , Antígenos Virales , Humanos , Inhibidores de Puntos de Control Inmunológico , Infecciones por Papillomavirus , ARN , Infecciones del Sistema Respiratorio
20.
Oral Oncol ; 132: 106012, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35820346

RESUMEN

Preclinical models of cancer have long been paramount to understanding tumor development and advancing the treatment of cancer. Creating preclinical models that mimic the complexity and heterogeneity of human tumors is a key challenge in the advancement of cancer therapy. About ten years ago, we created the mouse oral carcinoma (MOC) cell line models that were derived from 7, 12-dimethylbenz(a) anthracene (DMBA)-induced mouse oral squamous cell cancers. This model has been used in numerous investigations, including studies on tumor biology and therapeutics. We have seen remarkable progress in cancer immunology in recent years, and these cell lines, which are syngeneic to C57BL/6 background, have also been used to study the anti-tumor immune response. Herein, we aim to review the MOC model from its development and characterization to its use in non-immunological and immunological preclinical head and neck squamous cell carcinoma (HNSCC) studies. Integrating and refining these MOC model studies and extending findings to other systems will provide crucial insights for translational approaches aimed at improving head and neck cancer treatment.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias de la Boca/patología , Estudios Retrospectivos , Carcinoma de Células Escamosas de Cabeza y Cuello
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA