Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Eur J Cancer ; 202: 113983, 2024 May.
Article in English | MEDLINE | ID: mdl-38452723

ABSTRACT

BACKGROUND: Uncertainty persists regarding clinical and treatment variations crucial to consider when comparing high human papillomavirus (HPV)-prevalence oropharyngeal squamous cell carcinoma (OPSCC) cohorts for accurate patient stratification and replicability of clinical trials across different geographical areas. METHODS: OPSCC patients were included from The University of Texas MD Anderson Cancer Center (UTMDACC), USA and from The University Hospital of Copenhagen, Denmark from 2015-2020, (n = 2484). Outcomes were 3-year overall survival (OS) and recurrence-free interval (RFI). Subgroup analyses were made for low-risk OPSCC patients (T1-2N0M0) and high-risk patients (UICC8 III-IV). RESULTS: There were significantly more HPV-positive (88.2 % vs. 63.1 %), males (89.4 % vs. 74.1 %), never-smokers (52.1 % vs. 23.7 %), lower UICC8-stage (I/II: 79.3 % vs. 68 %), and fewer patients treated with radiotherapy (RT) alone (14.8 % vs. 30.3 %) in the UTMDACC cohort. No difference in the adjusted OS was observed (hazard ratio [HR] 1.21, p = 0.23), but a significantly increased RFI HR was observed for the Copenhagen cohort (HR: 1.74, p = 0.003). Subgroup analyses of low- and high-risk patients revealed significant clinical and treatment differences. No difference in prognosis was observed for low-risk patients, but the prognosis for high-risk patients in the Copenhagen cohort was worse (OS HR 2.20, p = 0.004, RFI HR 2.80, p = 0.002). CONCLUSIONS: We identified significant differences in clinical characteristics, treatment modalities, and prognosis between a Northern European and Northern American OPSCC population. These differences are important to consider when comparing outcomes and for patient stratification in clinical trials, as reproducibility might be challenging.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Oropharyngeal Neoplasms , Papillomavirus Infections , Male , Humans , Squamous Cell Carcinoma of Head and Neck/epidemiology , Squamous Cell Carcinoma of Head and Neck/therapy , Prognosis , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/therapy , Human Papillomavirus Viruses , Oropharyngeal Neoplasms/epidemiology , Oropharyngeal Neoplasms/therapy , Oropharyngeal Neoplasms/pathology , Papillomavirus Infections/complications , Papillomavirus Infections/epidemiology , Papillomavirus Infections/pathology , Prevalence , Reproducibility of Results , Denmark/epidemiology , Papillomaviridae
2.
Head Neck Pathol ; 18(1): 27, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38536624

ABSTRACT

BACKGROUND: The detection of human papillomavirus (HPV) has several implications in the diagnostic work-up and management of oropharyngeal squamous cell carcinoma (OPSCC). The choice of HPV detection assay and testing algorithms differ across institutions and vary in cost, detection targets, technical feasibility, and turnaround time. In this study, we aimed to validate the VisionArray® HPV Chip for formalin-fixed and paraffin-embedded (FFPE) samples of OPSCC using the previously applied standard pan-HPV DNA PCR assay as a reference. METHODS: The validation cohort consisted of FFPE tissue samples from patients previously diagnosed with HPV DNA-positive OPSCC (n = 80), HPV DNA-negative OPSCC (n = 21), and a benign group of tumor samples consisting of Warthin's tumors (n = 20) and branchial cleft cysts of the lateral neck (n = 14). All samples were tested with p16 immunohistochemistry, pan-HPV DNA PCR, and the VisionArray® HPV Chip. RESULTS: The overall sensitivity and specificity of the VisionArray® HPV Chip assay were 100% [95% CI 95.5%; 100.0%] and 96.3% [95% CI 87.3%; 99.6%] and the positive predictive value and negative predictive value were 97.6% [95% CI 91.5%; 99.7%] and 100% [95% CI 93.2%; 100%], respectively. CONCLUSIONS: The VisionArray® HPV Chip assay can be recommended for high-risk HPV testing in FFPE tissue samples from OPSCC, providing both a fast and simultaneous genotyping for 41 clinically relevant HPV types.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Oropharyngeal Neoplasms , Papillomavirus Infections , Humans , Squamous Cell Carcinoma of Head and Neck/diagnosis , Papillomavirus Infections/complications , Papillomavirus Infections/diagnosis , Papillomaviridae/genetics , DNA, Viral/analysis , Immunohistochemistry
3.
Viruses ; 15(1)2023 01 10.
Article in English | MEDLINE | ID: mdl-36680237

ABSTRACT

The incidence of oropharyngeal squamous cell carcinoma (OPSCC) has increased in the past decades due to carcinogenic HPV infection. As this patient group suffers from considerable mortality and treatment morbidity it is important to improve prognostic strategies in OPSCC. Inflammation plays a key role in cancer and the neutrophil-to-lymphocyte ratio (NLR) in blood has been suggested as a prognostic factor for OPSCC. This study aimed to investigate the prognostic impact of NLR on overall survival (OS) and recurrence-free survival (RFS) in a retrospective cohort of 1370 patients. Included patients had pretreatment neutrophil and lymphocyte counts available, as well as a known HPV status. Patients were treated with curative intent according to Danish national guidelines. We stratified patients in groups by NLR < 2, NLR 2−4, or NLR > 4 and analyzed the influence of the NLR tertile on OS and RFS. Kaplan−Meier curves illustrated survival probability in OS and RFS in the general cohort and were stratified by HPV status. We found that an increasing NLR was associated with inferior OS (HR = 1.5 for NLR > 4) and RFS (HR = 1.6 for NLR 2−4; HR = 1.8 for NLR > 4) in multivariable analysis. The Kaplan−Meier curves displayed inferior OS and RFS with an increasing NLR for both HPV+ and HPV− patients. In conclusion, we showed that an increasing NLR is prognostic for a worse outcome of OPSCC independently of HPV status. There are possible uses of NLR in prognostication and treatment de-escalation although further studies are warranted to determine the clinical utility.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Oropharyngeal Neoplasms , Papillomavirus Infections , Humans , Squamous Cell Carcinoma of Head and Neck , Prognosis , Retrospective Studies , Neutrophils/pathology , Papillomavirus Infections/complications , Oropharyngeal Neoplasms/diagnosis , Oropharyngeal Neoplasms/pathology , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/pathology , Lymphocytes/pathology
4.
Oncoimmunology ; 11(1): 2026020, 2022.
Article in English | MEDLINE | ID: mdl-35111385

ABSTRACT

Galectin-3 (Gal3) can be expressed by many cells in the tumor microenvironment (TME), including cancer cells, cancer-associated fibroblasts, tumor-associated macrophages, and regulatory T cells (Tregs). In addition to immunosuppression, Gal3 expression has been connected to malignant cell transformation, tumor progression, and metastasis. In the present study, we found spontaneous T-cell responses against Gal3-derived peptides in PBMCs from both healthy donors and cancer patients. We isolated and expanded these Gal3-specific T cells in vitro and showed that they could directly recognize target cells that expressed Gal3. Finally, therapeutic vaccination with a long Gal3-derived peptide epitope, which induced the expansion of Gal3-specific CD8+ T cells in vivo, showed a significant tumor-growth delay in mice inoculated with EO771.LMB metastatic mammary tumor cells. This was associated with a significantly lower percentage of both Tregs and tumor-infiltrating Gal3+ cells in the non-myeloid CD45+CD11b- compartment and with an alteration of the T-cell memory populations in the spleens of Gal3-vaccinated mice. These results suggest that by activating Gal3-specific T cells by an immune-modulatory vaccination, we can target Gal3-producing cells in the TME, and thereby induce a more immune permissive TME. This indicates that Gal3 could be a novel target for therapeutic cancer vaccines.


Subject(s)
Cancer Vaccines , Neoplasms , Animals , CD8-Positive T-Lymphocytes/metabolism , Galectin 3/metabolism , Humans , Mice , Tumor Microenvironment , Vaccination , Vaccines, Subunit
5.
J Immunother Cancer ; 10(12)2022 12.
Article in English | MEDLINE | ID: mdl-36600556

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is associated with very poor survival, making it the third and fourth leading cause of all cancer-related deaths in the USA and European Union, respectively. The tumor microenvironment (TME) in PDAC is highly immunosuppressive and desmoplastic, which could explain the limited therapeutic effect of immunotherapy in PDAC. One of the key molecules that contributes to immunosuppression and fibrosis is transforming growth factor-ß (TGFß). The aim of this study was to target the immunosuppressive and fibrotic TME in PDAC using a novel immune modulatory vaccine with TGFß-derived peptides in a murine model of pancreatic cancer. METHODS: C57BL/6 mice were subcutaneously inoculated with Pan02 PDAC cells. Mice were treated with TGFß1-derived peptides (major histocompatibility complex (MHC)-I and MHC-II-restricted) adjuvanted with Montanide ISA 51VG. The presence of treatment-induced TGFß-specific T cells was assessed by ELISpot (enzyme-linked immunospot). Changes in the immune infiltration and gene expression profile in tumor samples were characterized by flow cytometry, reverse transcription-quantitative PCR (RT-qPCR), and bulk RNA sequencing. RESULTS: Treatment with immunogenic TGFß-derived peptides was safe and controlled tumor growth in Pan02 tumor-bearing mice. Enlargement of tumor-draining lymph nodes in vaccinated mice positively correlated to the control of tumor growth. Analysis of immune infiltration and gene expression in Pan02 tumors revealed that TGFß-derived peptide vaccine increased the infiltration of CD8+ T cells and the intratumoral M1/M2 macrophage ratio, it increased the expression of genes involved in immune activation and immune response to tumors, and it reduced the expression of myofibroblast-like cancer-associated fibroblast (CAF)-related genes and genes encoding fibroblast-derived collagens. Finally, we confirmed that TGFß-derived peptide vaccine actively modulated the TME, as the ability of T cells to proliferate was restored when exposed to tumor-conditioned media from vaccinated mice compared with media from untreated mice. CONCLUSION: This study demonstrates the antitumor activity of TGFß-derived multipeptide vaccination in a murine tumor model of PDAC. The data suggest that the vaccine targets immunosuppression and fibrosis in the TME by polarizing the cellular composition towards a more pro-inflammatory phenotype. Our findings support the feasibility and potential of TGFß-derived peptide vaccination as a novel immunotherapeutic approach to target immunosuppression in the TME.


Subject(s)
Cancer Vaccines , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Mice , Animals , CD8-Positive T-Lymphocytes , Transforming Growth Factor beta , Tumor Microenvironment , Disease Models, Animal , Cell Line, Tumor , Vaccines, Subunit/therapeutic use , Mice, Inbred C57BL , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/genetics , Immunosuppressive Agents/therapeutic use , Immunity , Fibrosis , Pancreatic Neoplasms
6.
Viruses ; 13(9)2021 08 26.
Article in English | MEDLINE | ID: mdl-34578274

ABSTRACT

The aim of the study was to evaluate the diagnostic accuracy of Human Papillomavirus (HPV) techniques in oropharyngeal cancer. PubMed, EMBASE, the Cochrane Library and clinicaltrials.org were systematically searched for studies reporting methods of HPV detection. Primary outcomes were sensitivity and specificity of HPV detection. In this case, 27 studies were included (n = 5488, 41.6% HPV+). In this case, 13 studies evaluated HPV detection in tumour tissue, nine studies examined HPV detection in blood samples and five studies evaluated HPV detection in oral samples. Accuracy of HPV detection in tumour tissue was high for all detection methods, with pooled sensitivity ranging from 81.1% (95% CI 71.9-87.8) to 93.1% (95% CI 87.4-96.4) and specificity ranging from 81.1% (95% CI 71.9-87.8) to 94.9% (95% CI 79.1-98.9) depending on detection methods. Overall accuracy of HPV detection in blood samples revealed a sensitivity of 81.4% (95% CI 62.9-91.9) and a specificity of 94.8% (95% CI 91.4-96.9). In oral samples pooled sensitivity and specificity were lower (77.0% (95% CI 68.8-83.6) and 74.0% (95% CI 58.0-85.4)). In conclusion, we found an overall high accuracy for HPV detection in tumour tissue regardless of the HPV detection method used. HPV detection in blood samples may provide a promising new way of HPV detection.


Subject(s)
Carcinoma, Squamous Cell/diagnosis , DNA, Viral/analysis , Molecular Diagnostic Techniques/standards , Papillomaviridae/genetics , Papillomavirus Infections/complications , Squamous Cell Carcinoma of Head and Neck/diagnosis , Carcinoma, Squamous Cell/virology , DNA, Viral/genetics , Humans , Molecular Diagnostic Techniques/methods , Papillomaviridae/isolation & purification , Reproducibility of Results , Sensitivity and Specificity , Squamous Cell Carcinoma of Head and Neck/virology
7.
APMIS ; 129(11): 619-625, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34561896

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) is a novel method enabling genetic characterization of tumor tissue at a single-cell level. This study systematically reviewed the literature on studies using scRNA-seq to characterize head and neck squamous cell carcinomas (HNSCCs). Seven studies were included, of which two studies performed scRNA-seq on 20 patients in total, and five studies used scRNA-seq data in a subsequent clinical study. The former mentioned two studies found intra-tumoral genetic heterogeneity among malignant cells but genetic uniformity among non-malignant cells. The five latter studies used scRNA-seq data in various ways. Three studies identified biomarkers related to predicting whether a patient would benefit from immunotherapeutic treatment. One study characterized genes related to the perineural invasion. One study identified genes to be used in diagnostics. Further studies performing scRNA-seq on HNSCC are required to continue the ongoing development and use of scRNA-seq.


Subject(s)
Head and Neck Neoplasms/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Squamous Cell Carcinoma of Head and Neck/genetics , Biomarkers, Tumor/genetics , Gene Expression Profiling , Genetic Heterogeneity , Head and Neck Neoplasms/pathology , Humans , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Microenvironment/genetics
8.
Viruses ; 13(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34452393

ABSTRACT

Focal epithelial hyperplasia (FEH) or Heck's disease is a rare, benign, oral condition that is associated with infection by human papillomavirus type 13, 32 or both. The whiteish to mucosal-colored, soft, papular or nodular elevated lesions in the oral cavity are normally asymptomatic but can grow to a size or at a location where treatment is needed. The diagnosis is often based on clinical presentation and histopathology, and the HPV genotype can be determined using PCR utilizing specific primers or DNA sequencing. While FEH was reported to often affect several members of the same family and exist primarily among indigenous populations around the world, the number of reported cases within the European region is increasing. This contemporary review summarizes the main findings in relation to HPV genotypes, impact of superinfection exclusion and vaccination, transmission, diagnosis, geographical and ethnical distribution, comorbidities and treatment of FEH with an emphasis on including the most recent case reports within the field. Furthermore, we describe for the first time a FEH lesion infected with the low-risk HPV90.


Subject(s)
Alphapapillomavirus/genetics , Alphapapillomavirus/pathogenicity , Focal Epithelial Hyperplasia , Papillomavirus Infections/complications , Alphapapillomavirus/classification , Genotype , Humans , Papillomavirus Infections/prevention & control , Papillomavirus Infections/transmission , Risk Factors
9.
Cell Mol Immunol ; 18(2): 415-426, 2021 02.
Article in English | MEDLINE | ID: mdl-33408343

ABSTRACT

Transforming growth factor-beta (TGFß) is a highly potent immunosuppressive cytokine. Although TGFß is a tumor suppressor in early/premalignant cancer lesions, the cytokine has several tumor-promoting effects in advanced cancer; abrogation of the antitumor immune response is one of the most important tumor-promoting effects. As several immunoregulatory mechanisms have recently been shown to be targets of specific T cells, we hypothesized that TGFß is targeted by naturally occurring specific T cells and thus could be a potential target for immunomodulatory cancer vaccination. Hence, we tested healthy donor and cancer patient T cells for spontaneous T-cell responses specifically targeting 38 20-mer epitopes derived from TGFß1. We identified numerous CD4+ and CD8+ T-cell responses against several epitopes in TGFß. Additionally, several ex vivo responses were identified. By enriching specific T cells from different donors, we produced highly specific cultures specific to several TGFß-derived epitopes. Cytotoxic CD8+ T-cell clones specific for both a 20-mer epitope and a 9-mer HLA-A2 restricted killed epitope peptide were pulsed in HLA-A2+ target cells and killed the HLA-A2+ cancer cell lines THP-1 and UKE-1. Additionally, stimulation of THP-1 cancer cells with cytokines that increased TGFß expression increased the fraction of killed cells. In conclusion, we have shown that healthy donors and cancer patients harbor CD4+ and CD8+ T cells specific for TGFß-derived epitopes and that cytotoxic T cells with specificity toward TGFß-derived epitopes are able to recognize and kill cancer cell lines in a TGFß-dependent manner.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Transforming Growth Factor beta/metabolism , Case-Control Studies , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Transforming Growth Factor beta/genetics
10.
Oncoimmunology ; 9(1): 1771142, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32923127

ABSTRACT

One way that tumors evade immune destruction is through tumor and stromal cell expression of arginine-degrading enzyme arginase-2 (ARG2). Here we describe the existence of pro-inflammatory effector T-cells that recognize ARG2 and can directly target tumor and tumor-infiltrating cells. Using a library of 34 peptides covering the entire ARG2 sequence, we examined reactivity toward these peptides in peripheral blood mononuclear cells from cancer patients and healthy individuals. Interferon-γ ELISPOT revealed frequent immune responses against several of the peptides, indicating that ARG2-specific self-reactive T-cells are natural components of the human T-cell repertoire. Based on this, the most immunogenic ARG2 protein region was further characterized. By identifying conditions in the microenvironment that induce ARG2 expression in myeloid cells, we showed that ARG2-specific CD4T-cells isolated and expanded from a peripheral pool from a prostate cancer patient could recognize target cells in an ARG2-dependent manner. In the 'cold' in vivo tumor model Lewis lung carcinoma, we found that activation of ARG2-specific T-cells by vaccination significantly inhibited tumor growth. Immune-modulatory vaccines targeting ARG2 thus are a candidate strategy for cancer immunotherapy.


Subject(s)
Arginase , Vaccines , Humans , Immunity , Leukocytes, Mononuclear , Male , T-Lymphocytes/immunology
11.
Cancer Immunol Immunother ; 68(11): 1901-1907, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31690955

ABSTRACT

L-arginine depletion by regulatory cells and cancer cells expressing arginase-1 (Arg-1) is a vital contributor to the immunosuppressive tumor microenvironment in patients with cancer. We have recently described the existence of pro-inflammatory effector T cells that recognize Arg-1. Hence, Arg-1-specific self-reactive T cells are a naturally occurring part of the memory T-cell repertoire of the human immune system. Here, we further characterize a highly immunogenic epitope from Arg-1. We describe frequent T-cell-based immune responses against this epitope in patients with cancer, as well as in healthy donors. Furthermore, we show that Arg-1-specific T cells expand in response to the TH2 cytokine interleukin (IL)-4 without any specific stimulation. Arg-1-specific memory TH1 cells that respond to increased IL-4 concentration may, therefore, drive the immune response back into the TH1 pathway. Arg-1-specific T cells thus appear to have an important function in immune regulation. Because Arg-1 plays an important role in the immunosuppressive microenvironment in most cancers, an immune modulatory vaccination approach can readily be employed to tilt the balance away from immune suppression in these settings.


Subject(s)
Arginase/immunology , Cancer Vaccines/administration & dosage , Epitopes, T-Lymphocyte/immunology , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Animals , Cancer Vaccines/immunology , Humans , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...