Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Semin Cancer Biol ; 95: 52-74, 2023 10.
Article in English | MEDLINE | ID: mdl-37473825

ABSTRACT

Head and neck tumors (HNTs) constitute a multifaceted ensemble of pathologies that primarily involve regions such as the oral cavity, pharynx, and nasal cavity. The intricate anatomical structure of these regions poses considerable challenges to efficacious treatment strategies. Despite the availability of myriad treatment modalities, the overall therapeutic efficacy for HNTs continues to remain subdued. In recent years, the deployment of artificial intelligence (AI) in healthcare practices has garnered noteworthy attention. AI modalities, inclusive of machine learning (ML), neural networks (NNs), and deep learning (DL), when amalgamated into the holistic management of HNTs, promise to augment the precision, safety, and efficacy of treatment regimens. The integration of AI within HNT management is intricately intertwined with domains such as medical imaging, bioinformatics, and medical robotics. This article intends to scrutinize the cutting-edge advancements and prospective applications of AI in the realm of HNTs, elucidating AI's indispensable role in prevention, diagnosis, treatment, prognostication, research, and inter-sectoral integration. The overarching objective is to stimulate scholarly discourse and invigorate insights among medical practitioners and researchers to propel further exploration, thereby facilitating superior therapeutic alternatives for patients.


Subject(s)
Artificial Intelligence , Head and Neck Neoplasms , Humans , Machine Learning , Neural Networks, Computer , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/therapy , Diagnostic Imaging/methods
2.
Small ; 20(19): e2308731, 2024 May.
Article in English | MEDLINE | ID: mdl-38327169

ABSTRACT

Immunotherapy has emerged as a potent strategy in cancer treatment, with many approved drugs and modalities in the development stages. Despite its promise, immunotherapy is not without its limitations, including side effects and suboptimal efficacy. Using nanoparticles (NPs) as delivery vehicles to target immunotherapy to lymph nodes (LNs) can improve the efficacy of immunotherapy drugs and reduce side effects in patients. In this context, this paper reviews the development of LN-targeted immunotherapeutic NP strategies, the mechanisms of NP transport during LN targeting, and their related biosafety risks. NP targeting of LNs involves either passive targeting, influenced by NP physical properties, or active targeting, facilitated by affinity ligands on NP surfaces, while alternative methods, such as intranodal injection and high endothelial venule (HEV) targeting, have uncertain clinical applicability and require further research and validation. LN targeting of NPs for immunotherapy can reduce side effects and increase biocompatibility, but risks such as toxicity, organ accumulation, and oxidative stress remain, although strategies such as biodegradable biomacromolecules, polyethylene glycol (PEG) coating, and impurity addition can mitigate these risks. Additionally, this work concludes with a future-oriented discussion, offering critical insights into the field.


Subject(s)
Immunotherapy , Lymph Nodes , Nanoparticles , Neoplasms , Immunotherapy/methods , Humans , Neoplasms/therapy , Neoplasms/immunology , Nanoparticles/chemistry , Animals
3.
J Nanobiotechnology ; 22(1): 308, 2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38825711

ABSTRACT

Research into mRNA vaccines is advancing rapidly, with proven efficacy against coronavirus disease 2019 and promising therapeutic potential against a variety of solid tumors. Adjuvants, critical components of mRNA vaccines, significantly enhance vaccine effectiveness and are integral to numerous mRNA vaccine formulations. However, the development and selection of adjuvant platforms are still in their nascent stages, and the mechanisms of many adjuvants remain poorly understood. Additionally, the immunostimulatory capabilities of certain novel drug delivery systems (DDS) challenge the traditional definition of adjuvants, suggesting that a revision of this concept is necessary. This review offers a comprehensive exploration of the mechanisms and applications of adjuvants and self-adjuvant DDS. It thoroughly addresses existing issues mentioned above and details three main challenges of immune-related adverse event, unclear mechanisms, and unsatisfactory outcomes in old age group in the design and practical application of cancer mRNA vaccine adjuvants. Ultimately, this review proposes three optimization strategies which consists of exploring the mechanisms of adjuvant, optimizing DDS, and improving route of administration to improve effectiveness and application of adjuvants and self-adjuvant DDS.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines , Nanotechnology , Neoplasms , mRNA Vaccines , Humans , Cancer Vaccines/immunology , Nanotechnology/methods , Neoplasms/therapy , Neoplasms/immunology , Animals , Drug Delivery Systems/methods , COVID-19/prevention & control , Adjuvants, Vaccine , RNA, Messenger/genetics , SARS-CoV-2/immunology , Vaccines, Synthetic/immunology
4.
J Nanobiotechnology ; 22(1): 135, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553735

ABSTRACT

The deployment of imaging examinations has evolved into a robust approach for the diagnosis of lymph node metastasis (LNM). The advancement of technology, coupled with the introduction of innovative imaging drugs, has led to the incorporation of an increasingly diverse array of imaging techniques into clinical practice. Nonetheless, conventional methods of administering imaging agents persist in presenting certain drawbacks and side effects. The employment of controlled drug delivery systems (DDSs) as a conduit for transporting imaging agents offers a promising solution to ameliorate these limitations intrinsic to metastatic lymph node (LN) imaging, thereby augmenting diagnostic precision. Within the scope of this review, we elucidate the historical context of LN imaging and encapsulate the frequently employed DDSs in conjunction with a variety of imaging techniques, specifically for metastatic LN imaging. Moreover, we engage in a discourse on the conceptualization and practical application of fusing diagnosis and treatment by employing DDSs. Finally, we venture into prospective applications of DDSs in the realm of LNM imaging and share our perspective on the potential trajectory of DDS development.


Subject(s)
Drug Delivery Systems , Lymph Nodes , Humans , Lymphatic Metastasis/pathology , Lymph Nodes/diagnostic imaging , Lymph Nodes/pathology
5.
BMC Med Educ ; 24(1): 531, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38741079

ABSTRACT

BACKGROUND: An urgent need exists for innovative surgical video recording techniques in head and neck reconstructive surgeries, particularly in low- and middle-income countries where a surge in surgical procedures necessitates more skilled surgeons. This demand, significantly intensified by the COVID-19 pandemic, highlights the critical role of surgical videos in medical education. We aimed to identify a straightforward, high-quality approach to recording surgical videos at a low economic cost in the operating room, thereby contributing to enhanced patient care. METHODS: The recording was comprised of six head and neck flap harvesting surgeries using GoPro or two types of digital cameras. Data were extracted from the recorded videos and their subsequent editing process. Some of the participants were subsequently interviewed. RESULTS: Both cameras, set at 4 K resolution and 30 frames per second (fps), produced satisfactory results. The GoPro, worn on the surgeon's head, moves in sync with the surgeon, offering a unique first-person perspective of the operation without needing an additional assistant. Though cost-effective and efficient, it lacks a zoom feature essential for close-up views. In contrast, while requiring occasional repositioning, the digital camera captures finer anatomical details due to its superior image quality and zoom capabilities. CONCLUSION: Merging these two systems could significantly advance the field of surgical video recording. This innovation holds promise for enhancing technical communication and bolstering video-based medical education, potentially addressing the global shortage of specialized surgeons.


Subject(s)
COVID-19 , Video Recording , Humans , COVID-19/epidemiology , Plastic Surgery Procedures/education , Surgical Flaps , SARS-CoV-2 , Head/surgery , Neck/surgery
6.
Cancer Immunol Immunother ; 72(6): 1647-1660, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36586012

ABSTRACT

The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome plays cell- and tissue-specific roles in cancer, meaning that its activation in different tumors or cells may play different roles in tumor progression. We have previously described the tumor-promoting function of tumor-intrinsic NLRP3/IL-1ß signaling in head and neck squamous cell carcinoma (HNSCC), but its role in immune cells remains unclear. In this study, we found that NLRP3 was highly expressed in tumor-associated macrophages (TAMs) in both mouse and human HNSCC, and the expression of NLRP3 was positively correlated with the density of TAMs according to immunohistochemistry, immunofluorescence, and flow cytometry analyses. Importantly, the number of NLRP3high TAMs was related to worse overall survival in HNSCC patients. Knocking out NLRP3 inhibited M2-like macrophage differentiation in vitro. Moreover, the carcinogenic effect induced by 4-nitroquinoline-1-oxide was decreased in Nlrp3-deficient mice, which had smaller tumor sizes. Genetic depletion of NLRP3 reduced the expression of protumoral cytokines, such as IL-1ß, IL-6, IL-10, and CCL2, and suppressed the accumulation of TAMs and myeloid-derived suppressor cells (MDSCs) in mouse HNSCC. Thus, activation of NLRP3 in TAMs may contribute to tumor progression and have prognostic significance in HNSCC.


Subject(s)
Head and Neck Neoplasms , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Mice , Animals , Squamous Cell Carcinoma of Head and Neck , Tumor-Associated Macrophages/metabolism , Interleukin-1beta/metabolism , Prognosis
7.
Pharmacol Res ; 198: 106989, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37979662

ABSTRACT

Lymph node metastasis (LNM) significantly impacts the prognosis of cancer patients. Despite significant advancements in diagnostic techniques and treatment modalities, clinical challenges continue to persist in the realm of LNM. These include difficulties in early diagnosis, limited treatment efficacy, and potential side effects and injuries associated with treatment. Nanotheranostics, a field within nanotechnology, seamlessly integrates diagnostic and therapeutic functionalities. Its primary goal is to provide precise and effective disease diagnosis and treatment simultaneously. The development of nanotheranostics for LNM offers a promising solution for the stratified management of patients with LNM and promotes the advancement of personalized medicine. This review introduces the mechanisms of LNM and challenges in its diagnosis and treatment. Furthermore, it demonstrates the advantages and development potential of nanotheranostics, focuses on the challenges nanotheranostics face in its application, and provides an outlook on future trends. We consider nanotheranostics a promising strategy to improve clinical effectiveness and efficiency as well as the prognosis of cancer patients with LNM.


Subject(s)
Lymphoma , Theranostic Nanomedicine , Humans , Lymphatic Metastasis/pathology , Prognosis , Precision Medicine , Retrospective Studies , Lymph Nodes
8.
J Oral Pathol Med ; 52(8): 766-776, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37549038

ABSTRACT

BACKGROUND: Salivary gland pleomorphic adenoma (SPA) is a common neoplasm of salivary glands that displays remarkable histological diversity. Previous studies have demonstrated the involvement of gene rearrangements and cytoskeleton-remodeling-related myoepithelial cells in SPA tumorigenesis. Cytoskeleton remodeling is necessary for epithelial-mesenchymal transition (EMT), a key process in tumor progression. However, the heterogeneity of tumor cells and cytoskeleton remodeling in SPA has not been extensively investigated. METHODS: An analysis of single-cell RNA sequencing (scRNA-seq) was performed on 27 810 cells from two donors with SPA. Bioinformatic tools were used to assess differentially expressed genes, cell trajectories, and intercellular communications. Immunohistochemistry and double immunofluorescence staining were used to demonstrate FOXC1 and MYLK expression in SPA tissues. RESULTS: Our analysis revealed five distinct cell subtypes within the tumor cells of SPA, indicating a high level of intra-lesional heterogeneity. Cytoskeleton-remodeling-related genes were highly enriched in subtype 3 of the tumor cells, which showed a close interaction with mesenchymal cells. We found that tumoral FOXC1 expression was closely related to MYLK expression in the tumor cells of SPA. CONCLUSION: Tumor cells enriched with cytoskeleton-remodeling-related genes play a crucial role in SPA development, and FOXC1 may partially regulate this process.


Subject(s)
Adenoma, Pleomorphic , Salivary Gland Neoplasms , Humans , Adenoma, Pleomorphic/pathology , Salivary Gland Neoplasms/pathology , Salivary Glands/metabolism , Sequence Analysis, RNA
9.
J Cell Mol Med ; 26(24): 5955-5965, 2022 12.
Article in English | MEDLINE | ID: mdl-36448260

ABSTRACT

Tumour cell-secreted microvesicles (MVs) contribute immensely to tumour progression. However, the role of tumoral salivary MVs in oral squamous cell carcinoma (OSCC) remains unclear. Herein, we elucidated the role of non-apoptotic salivary tumoral MVs in OSCC development, especially relating to the migration ability. We purified and compared non-apoptotic salivary tumoral MVs from 63 OSCC patients and orthotopic OSCC mice model. Next, we compared the protein difference between apoptotic and non-apoptotic MVs by Western blot, proteomics and flow cytometry from saliva and CAL27 cells. Finally, we collected the non-apoptotic MVs and co-cultured with normal oral epithelial cells, the migration ability was examined by wound healing assay and Western blot assay. Our results indicated that the levels of non-apoptotic tumoral S-MVs were significantly higher in OSCC patients with T3 to T4 stages than in patients with T1 to T2 stages or healthy donors. In OSCC mice model, we found elevations of non-apoptotic tumoral MVs associated with tumoral volume. EGFR overexpression increased the generation of non-apoptotic tumoral MVs which could significantly promote normal epithelial cell migration. In conclusion, elevated levels of non-apoptotic tumoral S-MVs are associated with clinicopathologic features of OSCC patients, implying that non-apoptotic tumoral S-MVs are a potential progressive marker of OSCC.


Subject(s)
Carcinoma, Squamous Cell , Cell-Derived Microparticles , Mouth Neoplasms , Mice , Animals , Mouth Neoplasms/pathology , Carcinoma, Squamous Cell/pathology , Cell-Derived Microparticles/metabolism , Saliva/metabolism , Proteomics , Biomarkers, Tumor/metabolism
10.
J Nanobiotechnology ; 20(1): 277, 2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35701847

ABSTRACT

Cancer immunotherapy is a novel therapeutic regimen because of the specificity and durability of immune modulations to treat cancers. Current cancer immunotherapy is limited by some barriers such as poor response rate, low tumor specificity and systemic toxicities. Porous nanomaterials (PNMs) possess high loading capacity and tunable porosity, receiving intense attention in cancer immunotherapy. Recently, novel PNMs based drug delivery systems have been employed in antitumor immunotherapy to enhance tissue or organ targeting and reduce immune-related adverse events. Herein, we summarize the recent progress of PNMs including inorganic, organic, and organic-inorganic hybrid ones for cancer immunotherapy. The design of PNMs and their performance in cancer immunotherapy are discussed in detail, with a focus on how those designs can address the challenges in current conventional immunotherapy. Lastly, we present future directions of PNMs for cancer immunotherapy including the challenges and research gaps, providing new insights about the design of PNMs for efficient cancer immunotherapy with better performance as powerful weapons against tumors. Finally, we discussed the relevant challenges that urgently need to be addressed in clinical practice, coupled with corresponding solutions to these problems.


Subject(s)
Nanostructures , Neoplasms , Drug Delivery Systems , Humans , Immunologic Factors , Immunotherapy , Nanostructures/therapeutic use , Neoplasms/drug therapy , Porosity
11.
J Nanobiotechnology ; 19(1): 379, 2021 Nov 21.
Article in English | MEDLINE | ID: mdl-34802438

ABSTRACT

Matrix metalloproteinase (MMP) 2 and 9 are the family members of proteases normally up-regulated in tumor to enhance the invasion and metastatic of tumor cells, and are associated with poor outcome of head and neck squamous cell carcinomas (HNSCCs). In the present work, MMPs-degradable gelatin nanoparticles (GNPs) are simultaneously loaded with photosensitizer indocyanine green (ICG) along with signal transducer activator of transcription 3 (STAT3) inhibitor NSC74859 (NSC, N) for efficient photothermal therapy (PTT) and immunotherapy of HNSCCs. In the tumor tissue, Gel-N-ICG nanoparticle was degraded and encapsulated ICG and NSC were effectively released. Under near-infrared (NIR) irradiation, the released ICG nanoparticles enabled effective photothermal destruction of tumors, and the STAT3 inhibitor NSC elicited potent antitumor immunity for enhanced cancer therapy. Based on two HNSCC mouse models, we demonstrated that Gel-N-ICG significantly delayed tumor growth without any appreciable body weight loss. Taken together, the strategy reported here may contribute that the stimuli-responsive proteases triggered nanoplatform could reduce tumor size more effectively in complex tumor microenvironment (TME) through combination of PTT and immunotherapy.


Subject(s)
Gelatinases/metabolism , Nanoparticles , Photosensitizing Agents , Protein Inhibitors of Activated STAT , Animals , Cell Line, Tumor , Cell Survival/drug effects , Immunotherapy , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , Mice , Nanoparticles/chemistry , Nanoparticles/metabolism , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Photothermal Therapy , Protein Inhibitors of Activated STAT/chemistry , Protein Inhibitors of Activated STAT/pharmacokinetics , Protein Inhibitors of Activated STAT/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors
12.
Nano Lett ; 19(4): 2215-2222, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30543300

ABSTRACT

Zika virus (ZIKV) has emerged as a global health threat due to its unexpected causal link to devastating neurological disorders such as fetal microcephaly; however, to date, no approved vaccine or specific treatment is available for ZIKV infection. Here we develop a biomimetic nanodecoy (ND) that can trap ZIKV, divert ZIKV away from its intended targets, and inhibit ZIKV infection. The ND, which is composed of a gelatin nanoparticle core camouflaged by mosquito medium host cell membranes, effectively adsorbs ZIKV and inhibits ZIKV replication in ZIKV-susceptible cells. Using a mouse model, we demonstrate that NDs significantly attenuate the ZIKV-induced inflammatory responses and degenerative changes and thus improve the survival rate of ZIKV-challenged mice. Moreover, by trapping ZIKV, NDs successfully prevent ZIKV from passing through physiologic barriers into the fetal brain and thereby mitigate ZIKV-induced fetal microcephaly in pregnant mice. We anticipate that this study will provide new insights into the development of safe and effective protection against ZIKV and various other viruses that threaten public health.


Subject(s)
Microcephaly/prevention & control , Nanoparticles/administration & dosage , Zika Virus Infection/prevention & control , Zika Virus/drug effects , Animals , Biomimetics/methods , Cell Membrane/drug effects , Cell Membrane/virology , Culicidae/drug effects , Culicidae/virology , Disease Models, Animal , Female , Fetus , Gelatin/administration & dosage , Gelatin/chemistry , Humans , Mice , Microcephaly/pathology , Microcephaly/virology , Nanoparticles/chemistry , Pregnancy , Zika Virus/pathogenicity , Zika Virus Infection/pathology , Zika Virus Infection/virology
13.
Cell Mol Life Sci ; 75(22): 4223-4234, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29955905

ABSTRACT

The immune system plays a critical role in the establishment, development, and progression of head and neck squamous cell carcinoma (HNSCC). As treatment with single-immune checkpoint agent results in a lower response rate in patients, it is important to investigate new strategies to maintain favorable anti-tumor immune response. Herein, the combination immunotherapeutic value of CTLA4 blockade and SFKs inhibition was assessed in transgenic HNSCC mouse model. Our present work showed that tumor growth was not entirely controlled when HNSCC model mice were administered anti-CTLA4 chemotherapeutic treatment. Moreover, it was observed that Src family kinases (SFKs) were hyper-activated and lack of anti-tumor immune responses following anti-CTLA4 chemotherapeutic treatment. We hypothesized that activation of SFKs is a mechanism of anti-CTLA4 immunotherapy resistance. We, therefore, carried out combined drug therapy using anti-CTLA4 mAbs and an SFKs' inhibitor, dasatinib. As expected, dasatinib and anti-CTLA4 synergistically inhibited tumor growth in Tgfbr1/Pten 2cKO mice. Furthermore, dasatinib and anti-CTLA4 combined to reduce the number of myeloid-derived suppressor cells and Tregs, increasing the CD8+ T cell-to-Tregs ratio. We also found that combining dasatinib with anti-CTLA4 therapy significantly attenuated the expression of p-STAT3Y705 and Ki67 in tumoral environment. These results suggest that combination therapy with SFKs inhibitors may be a useful therapeutic approach to increase the efficacy of anti-CTLA4 immunotherapy in HNSCC.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CTLA-4 Antigen/immunology , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , src-Family Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/therapy , Dasatinib/therapeutic use , Disease Models, Animal , Down-Regulation/drug effects , Drug Therapy, Combination , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/therapy , Immunotherapy , Mice , Mice, Knockout , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , STAT3 Transcription Factor/metabolism , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment , src-Family Kinases/antagonists & inhibitors
14.
Int J Cancer ; 143(6): 1494-1504, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29663369

ABSTRACT

The adenosine-induced immunosuppression hampers the immune response toward tumor cells and facilitates the tumor cells to evade immunosurveillance. CD73, an ecto-5-nucleotidase, is the ectoenzyme dephosphorylating extracellular AMP to adenosine. Here, using immunocompetent transgenic head and neck squamous cell carcinoma (HNSCC) mouse model, immune profiling showed high expression of CD73 on CD4+ and CD8+ T cells was associated with an "exhausted" phenotype. Further, treatment with anti-CD73 monoclonal antibody (mAb) significantly blunted the tumor growth in the mouse model, and the blockade of CD73 reversed the "exhausted" phenotype of CD4+ and CD8+ T cells through downregulation of total expression of PD-1 and CTLA-4 on T cells. Whereas the population of CD4+ CD73hi /CD8+ CD73hi T cells expressed higher CTLA-4 and PD-1 as compared to untreated controls. In addition, the human tissue microarrays showed the expression of CD73 is upregulated on tumor infiltrating immune cells in patients with primary HNSCC. Moreover, CD73 expression is an independent prognostic factor for poor outcome in our cohort of HNSCC patients. Altogether, these findings highlight the immunoregulatory role of CD73 in the development of HNSCC and we propose that CD73 may prove to be a promising immunotherapeutic target for the treatment of HNSCC.


Subject(s)
5'-Nucleotidase/metabolism , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/immunology , Head and Neck Neoplasms/immunology , Immune Tolerance/immunology , Lymphocytes, Tumor-Infiltrating/immunology , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/genetics , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis , Biomarkers, Tumor , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , CTLA-4 Antigen/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Follow-Up Studies , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Knockout , Mice, Transgenic , PTEN Phosphohydrolase/physiology , Phenotype , Prognosis , Receptor, Transforming Growth Factor-beta Type I/physiology , Survival Rate , Tumor Cells, Cultured
15.
Int J Cancer ; 142(5): 999-1009, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29047105

ABSTRACT

Immune evasion is a hallmark feature of cancer, and it plays an important role in tumour initiation and progression. In addition, tumour immune evasion severely hampers the desired antitumour effect in multiple cancers. In this study, we aimed to investigate the role of the Notch pathway in immune evasion in the head and neck squamous cell carcinoma (HNSCC) microenvironment. We first demonstrated that Notch1 signaling was activated in a Tgfbr1/Pten-knockout HNSCC mouse model. Notch signaling inhibition using a γ-secretase inhibitor (GSI-IX, DAPT) decreased tumour burden in the mouse model after prophylactic treatment. In addition, flow cytometry analysis indicated that Notch signaling inhibition reduced the sub-population of myeloid-derived suppressor cells (MDSCs), tumour-associated macrophages (TAMs) and regulatory T cells (Tregs), as well as immune checkpoint molecules (PD1, CTLA4, TIM3 and LAG3), in the circulation and in the tumour. Immunohistochemistry (IHC) of human HNSCC tissues demonstrated that elevation of the Notch1 downstream target HES1 was correlated with MDSC, TAM and Treg markers and with immune checkpoint molecules. These results suggest that modulating the Notch signaling pathway may decrease MDSCs, TAMs, Tregs and immune checkpoint molecules in HNSCC.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Carcinoma, Squamous Cell/immunology , Diamines/pharmacology , Disease Models, Animal , Head and Neck Neoplasms/immunology , Myeloid Cells/immunology , T-Lymphocytes, Regulatory/immunology , Thiazoles/pharmacology , Animals , Apoptosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunosuppression Therapy , Mice , Mice, Knockout , Myeloid Cells/drug effects , PTEN Phosphohydrolase/physiology , Protein Serine-Threonine Kinases/physiology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/physiology , T-Lymphocytes, Regulatory/drug effects , Tumor Cells, Cultured , Tumor Escape/drug effects
16.
Mol Carcinog ; 57(3): 429-439, 2018 03.
Article in English | MEDLINE | ID: mdl-29215754

ABSTRACT

Angiogenesis is an essential event in tumor growth and metastasis, and immune system also contributes to the tumor evasion. Emerging evidences have suggested the bidirectional link between angiogenesis and immunosuppression. Myeloid-derived suppressor cell (MDSC) is a kind of immunosuppressive cells and plays an important role in this process. However, the actual regulatory mechanisms of angiogenesis and MDSCs in head and neck squamous cell carcinoma (HNSCC) were unclear. In this study, through analyzing the immunohistochemistry staining of human HNSCC tissue microarray, we found that the microvascular density (MVD) was significantly increased in HNSCC patients. We also characterized angiogenic factors p-STAT3, VEGFA, CK2, and MDSCs marker CD11b in HNSCC tissue array, and found the close expression correlation among these markers. To determine the role of JAK2/STAT3 pathway in tumor microenvironment of HNSCC, we utilized AG490 (an inhibitor of JAK2/STAT3) for further research. Results showed that inhibition of JAK2/STAT3 suppressed angiogenesis by decreasing VEGFA and HIF1-α both in vitro and vivo. Moreover, in HNSCC transgenic mouse model, inhibiting JAK2/STAT3 not only suppressed angiogenesis but also reduced MDSCs in the tumor microenvironment through suppressing VEGFA and CK2. Our findings demonstrated the close relationship between angiogenesis and MDSCs in HNSCC, and inhibition of JAK2/STAT3 could reduce tumor-induced angiogenesis and decrease MDSCs.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Janus Kinase 2/antagonists & inhibitors , Myeloid-Derived Suppressor Cells/drug effects , Neovascularization, Pathologic/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Tyrphostins/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Human Umbilical Vein Endothelial Cells , Humans , Janus Kinase 2/metabolism , Mice, Transgenic , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tyrphostins/pharmacology
17.
Angew Chem Int Ed Engl ; 57(4): 986-991, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29193651

ABSTRACT

Here, we present a platelet-facilitated photothermal tumor therapy (PLT-PTT) strategy, in which PLTs act as carriers for targeted delivery of photothermal agents to tumor tissues and enhance the PTT effect. Gold nanorods (AuNRs) were first loaded into PLTs by electroporation and the resulting AuNR-loaded PLTs (PLT-AuNRs) inherited long blood circulation and cancer targeting characteristics from PLTs and good photothermal property from AuNRs. Using a gene-knockout mouse model, we demonstrate that the administration of PLT-AuNRs and localizing laser irradiation could effectively inhibit the growth of head and neck squamous cell carcinoma (HNSCC). In addition, we found that the PTT treatment augmented PLT-AuNRs targeting to the tumor sites and in turn, improved the PTT effects in a feedback manner, demonstrating the unique self-reinforcing characteristic of PLT-PTT in cancer therapy.


Subject(s)
Blood Platelets/chemistry , Lasers , Phototherapy , Squamous Cell Carcinoma of Head and Neck/therapy , Animals , Blood Platelets/cytology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Gold/chemistry , Humans , Mice , Mice, Inbred ICR , Mice, Knockout , Microscopy, Confocal , Nanotubes/chemistry , Nanotubes/toxicity , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , RAW 264.7 Cells , Receptor, Transforming Growth Factor-beta Type I/deficiency , Receptor, Transforming Growth Factor-beta Type I/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism
18.
J Cell Mol Med ; 21(9): 2199-2210, 2017 09.
Article in English | MEDLINE | ID: mdl-28401653

ABSTRACT

Immature myeloid cells including myeloid-derived suppressor cells (MDSCs) and tumour-associated macrophages (TAMs) promote tumour growth and metastasis by facilitating tumour transformation and angiogenesis, as well as by suppressing antitumour effector immune responses. Therefore, strategies designed to reduce MDSCs and TAMs accumulation and their activities are potentially valuable therapeutic goals. In this study, we show that negative immune checkpoint molecule B7-H3 is significantly overexpressed in human head and neck squamous cell carcinoma (HNSCC) specimen as compared with normal oral mucosa. Using immunocompetent transgenic HNSCC models, we observed that targeting inhibition of B7-H3 reduced tumour size. Flow cytometry analysis revealed that targeting inhibition of B7-H3 increases antitumour immune response by decreasing immunosuppressive cells and promoting cytotoxic T cell activation in both tumour microenvironment and macroenvironment. Our study provides direct in vivo evidence for a rationale for B7-H3 blockade as a future therapeutic strategy to treat patients with HNSCC.


Subject(s)
B7 Antigens/antagonists & inhibitors , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Myeloid Cells/pathology , Animals , B7 Antigens/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Disease Models, Animal , Humans , Macrophages/pathology , Mice, Knockout , Myeloid Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Prognosis , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism
19.
Mol Cancer ; 16(1): 99, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28592285

ABSTRACT

BACKGROUND: Cancer immunotherapy offers a promising approach in cancer treatment. The adenosine A2A receptor (A2AR) could protect cancerous tissues from immune clearance via inhibiting T cells response. To date, the role of A2AR in head and neck squamous cell carcinoma (HNSCC) has not been investigated. Here, we sought to explore the expression and immunotherapeutic value of A2AR blockade in HNSCC. METHODS: The expression of A2AR was evaluated by immunostaining in 43 normal mucosae, 48 dysplasia and 165 primary HNSCC tissues. The immunotherapeutic value of A2AR blockade was assessed in vivo in genetically defined immunocompetent HNSCC mouse model. RESULTS: Immunostaining of HNSCC tissue samples revealed that increased expression of A2AR on tumor infiltrating immune cells correlated with advanced pathological grade, larger tumor size and positive lymph node status. Elevated A2AR expression was also detected in recurrent HNSCC and HNSCC tissues with induction chemotherapy. The expression of A2AR was found to be significantly correlated with HIF-1α, CD73, CD8 and Foxp3. Furthermore, the increased population of CD4+Foxp3+ regulatory T cells (Tregs), which partially expressed A2AR, was observed in an immunocompetent mouse model that spontaneously develops HNSCC. Pharmacological blockade of A2AR by SCH58261 delayed the tumor growth in the HNSCC mouse model. Meanwhile, A2AR blockade significantly reduced the population of CD4+ Foxp3+ Tregs and enhanced the anti-tumor response of CD8+ T cells. CONCLUSIONS: These results offer a preclinical proof for the administration of A2AR inhibitor on prophylactic experimental therapy of HNSCC and suggest that A2AR blockade can be a potential novel strategy for HNSCC immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Immunomodulation , Receptor, Adenosine A2A/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , 5'-Nucleotidase/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Adult , Aged , Animals , Biomarkers , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cytokines/metabolism , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Gene Expression , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Induction Chemotherapy , Lymphocyte Count , Mice , Mice, Transgenic , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , PTEN Phosphohydrolase/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Adenosine A2A/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Recurrence , Squamous Cell Carcinoma of Head and Neck , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
20.
Int J Cancer ; 140(5): 1173-1185, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-27798955

ABSTRACT

SRC family kinases (SFKs), a group of nonreceptor tyrosine kinases, modulate multiple cellular functions, such as cell proliferation, differentiation and metabolism. SFKs display aberrant activity in progressive stages of human cancers. However, the precise role of SFKs in the head and neck squamous cell carcinoma (HNSCC) signaling network is far from clear. In this study, we found that the inhibition of SFKs activity by dasatinib effectively reduced the tumor size and population of MDSCs in the HNSCC mouse model. Molecular analysis indicates that phosphorylation of LYN, rather than SRC, was inhibited by dasatinib treatment. Next, we analyzed LYN expression by immunostaining and found that it was overexpressed in the human HNSCC specimens. Moreover, LYN expression in stromal cells positively correlated with myeloid-derived suppressor cells (MDSCs) makers CD11b and CD33 in human HNSCC. The dual positive expression of LYN in epithelial and stromal cells (EPI+ SRT+ ) was associated with unfavorable overall survival of HNSCC patients. These findings indicate that SFKs may be a potential target for an effective immunotherapy of HNSCC by decreasing MDSCs and moreover, LYN will have an impact on such therapeutic strategy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Dasatinib/therapeutic use , Head and Neck Neoplasms/pathology , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , src-Family Kinases/antagonists & inhibitors , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/immunology , Dasatinib/pharmacology , Enzyme Induction/drug effects , Epithelial Cells/enzymology , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/immunology , Humans , Mice , Mice, Knockout , Molecular Targeted Therapy , Neoplasm Proteins/physiology , PTEN Phosphohydrolase/deficiency , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/deficiency , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/deficiency , Stromal Cells/enzymology , Tumor Escape , Up-Regulation , Xenograft Model Antitumor Assays , src-Family Kinases/biosynthesis , src-Family Kinases/genetics , src-Family Kinases/metabolism , src-Family Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL