Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Vaccines (Basel) ; 11(4)2023 Apr 07.
Article in English | MEDLINE | ID: mdl-37112730

ABSTRACT

Toll-like receptors (TLRs) are indispensable for the activation, maintenance and halting of immune responses. TLRs can mediate inflammation by recognizing molecular patterns in microbes (pathogen-associated molecular patterns: PAMPs) and endogenous ligands (danger-associated molecular patterns: DAMPs) released by injured or dead cells. For this reason, TLR ligands have attracted much attention in recent years in many cancer vaccines, alone or in combination with immunotherapy, chemotherapy and radiotherapy (RT). TLRs have been shown to play controversial roles in cancer, depending on various factors that can mediate tumor progression or apoptosis. Several TLR agonists have reached clinical trials and are being evaluated in combination with standard of care therapies, including RT. Despite their prolific and central role in mediating immune responses, the role of TLRs in cancer, particularly in response to radiation, remains poorly understood. Radiation is recognized as either a direct stimulant of TLR pathways, or indirectly through the damage it causes to target cells that subsequently activate TLRs. These effects can mediate pro-tumoral and anti-tumoral effects depending on various factors such as radiation dose and fractionation, as well as host genomic features. In this review, we examine how TLR signaling affects tumor response to RT, and we provide a framework for the design of TLR-based therapies with RT.

2.
Cancer Immunol Immunother ; 72(5): 1089-1102, 2023 May.
Article in English | MEDLINE | ID: mdl-36326893

ABSTRACT

BACKGROUND:  Radioresistance of HNSCCs remains a major challenge for effective tumor control. Combined radiotherapy (RT) and immunotherapy (IT) treatment improved survival for a subset of patients with inflamed tumors or tumors susceptible to RT-induced inflammation. To overcome radioresistance and improve treatment outcomes, an understanding of factors that suppress anti-tumor immunity is necessary. In this regard, regulatory T cells (Tregs) are critical mediators of immune suppression in HNSCCs. In this study, we investigated how radiation modulates Treg infiltration in tumors through the chemokine CCL20. We hypothesized that radiation induces CCL20 secretion resulting in Treg infiltration and suppression of anti-tumor immunity. METHODS:  Human and mouse HNSCC cell lines with different immune phenotypes were irradiated at doses of 2 or 10 Gy. Conditioned media, RNA and protein were collected for assessment of CCL20. qPCR was used to determine CCL20 gene expression. In vivo, MOC2 cells were implanted into the buccal cavity of mice and the effect of neutralizing CCL20 antibody was determined alone and in combination with RT. Blood samples were collected before and after RT for analysis of CCL20. Tumor samples were analyzed by flow cytometry to determine immune infiltrates, including CD8 T cells and Tregs. Mass-spectrometry was performed to analyze proteomic changes in the tumor microenvironment after anti-CCL20 treatment. RESULTS:  Cal27 and MOC2 HNSCCs had a gene signature associated with Treg infiltration, whereas SCC9 and MOC1 tumors displayed a gene signature associated with an inflamed TME. In vitro, tumor irradiation at 10 Gy significantly induced CCL20 in Cal27 and MOC2 cells relative to control. The increase in CCL20 was associated with increased Treg migration. Neutralization of CCL20 reversed radiation-induced migration of Treg cells in vitro and decreased intratumoral Tregs in vivo. Furthermore, inhibition of CCL20 resulted in a significant decrease in tumor growth compared to control in MOC2 tumors. This effect was further enhanced after combination with RT compared to either treatment alone. CONCLUSION:  Our results suggest that radiation promotes CCL20 secretion by tumor cells which is responsible for the attraction of Tregs. Inhibition of the CCR6-CCL20 axis prevents infiltration of Tregs in tumors and suppresses tumor growth resulting in improved response to radiation.


Subject(s)
Head and Neck Neoplasms , T-Lymphocytes, Regulatory , Humans , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Proteomics , Head and Neck Neoplasms/radiotherapy , Head and Neck Neoplasms/metabolism , Tumor Microenvironment , Receptors, CCR6/genetics , Receptors, CCR6/metabolism
3.
Mol Ther ; 30(3): 1149-1162, 2022 03 02.
Article in English | MEDLINE | ID: mdl-34793974

ABSTRACT

STAT3 signaling has been shown to regulate cellular function and cytokine production in the tumor microenvironment (TME). Within the head and neck squamous cell carcinoma (HNSCC) TME, we previously showed that therapeutic targeting of STAT3 in combination with radiation resulted in improved tumor growth delay. However, given the independent regulatory effects STAT3 has on anti-tumor immunity, we aimed to decipher the effects of individually targeting STAT3 in the cancer cell, regulatory T cells (Tregs), and natural killer (NK) cell compartments in driving tumor growth and resistance to therapy in HNSCCs. We utilized a CRISPR knockout system for genetic deletion of STAT3 within the cancer cell as well as two genetic knockout mouse models, FoxP3-Cre/STAT3 fl and NKp46-Cre/STAT3 fl, for Tregs and NK cell targeting, respectively. Our data revealed differences in development of resistance to treatment with STAT3 CRISPR knockout in the cancer cell, driven by differential recruitment of immune cells. Knockout of STAT3 in Tregs overcomes this resistance and results in Treg reprogramming and recruitment and activation of antigen-presenting cells. In contrast, knockout of STAT3 in the NK cell compartment results in NK cell inactivation and acceleration of tumor growth. These data underscore the complex interplay between the cancer cell and the immune TME and carry significant implications for drug targeting and design of combination approaches in HNSCCs.


Subject(s)
Head and Neck Neoplasms , STAT3 Transcription Factor/metabolism , Animals , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/therapy , Mice , Mice, Knockout , STAT3 Transcription Factor/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/therapy , T-Lymphocytes, Regulatory , Tumor Microenvironment/genetics
4.
J Immunother Cancer ; 9(4)2021 04.
Article in English | MEDLINE | ID: mdl-33883256

ABSTRACT

BACKGROUND: Numerous trials combining radiation therapy (RT) and immunotherapy in head and neck squamous cell carcinoma (HNSCC) are failing. Using preclinical immune cold models of HNSCC resistant to RT-immune checkpoint inhibitors, we investigate therapeutic approaches of overcoming such resistance by examining the differential microenvironmental response to RT. METHODS: We subjected two HPV-negative orthotopic mouse models of HNSCC to combination RT, regulatory T cells (Treg) depletion, and/or CD137 agonism. Tumor growth was measured and intratumorous and lymph node immune populations were compared among treatment groups. Human gene sets, genetically engineered mouse models DEREG and BATF3-/-, flow and time-of-flight cytometry, RNA-Seq, Treg adoptive transfer studies, and in vitro experiments were used to further evaluate the role of dendritic cells (DCs) and Tregs in these treatments. RESULTS: In MOC2 orthotopic tumors, we find no therapeutic benefit to targeting classically defined immunosuppressive myeloids, which increase with RT. In these radioresistant tumors, supplementing combination RT and Treg depletion with anti-CD137 agonism stimulates CD103+ DC activation in tumor-draining lymph nodes as characterized by increases in CD80+ and CCR7+ DCs, resulting in a CD8 T cell-dependent response. Simultaneously, Tregs are reprogrammed to an effector phenotype demonstrated by increases in interferonγ+, tumor necrosis factorα+, PI3K+, pAKT+ and Eomes+ populations as well as decreases in CTLA4+ and NRP-1+ populations. Tumor eradication is observed when RT is increased to an 8 Gy x 5 hypofractionated regimen and combined with anti-CD25+ anti-CD137 treatment. In a human gene set from oral squamous cell carcinoma tumors, high Treg number is associated with earlier recurrence. CONCLUSIONS: Regulating Treg functionality and DC activation status within the lymph node is critical for generating a T cell effector response in these highly radioresistant tumors. These findings underscore the plasticity of Tregs and represent a new therapeutic opportunity for reprogramming the tumor microenvironment in HNSCCs resistant to conventional radioimmunotherapy approaches.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Dendritic Cells/drug effects , Drug Resistance, Neoplasm , Head and Neck Neoplasms/therapy , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Radiation Dose Hypofractionation , Radiation Tolerance , Squamous Cell Carcinoma of Head and Neck/therapy , T-Lymphocytes, Regulatory/drug effects , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Combined Modality Therapy , Dendritic Cells/immunology , Dendritic Cells/metabolism , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Depletion , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Repressor Proteins/genetics , Repressor Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Burden , Tumor Microenvironment , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
5.
Cancer Res ; 81(12): 3255-3269, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33526513

ABSTRACT

Stromal fibrosis activates prosurvival and proepithelial-to-mesenchymal transition (EMT) pathways in pancreatic ductal adenocarcinoma (PDAC). In patient tumors treated with neoadjuvant stereotactic body radiation therapy (SBRT), we found upregulation of fibrosis, extracellular matrix (ECM), and EMT gene signatures, which can drive therapeutic resistance and tumor invasion. Molecular, functional, and translational analysis identified two cell-surface proteins, a disintegrin and metalloprotease 10 (ADAM10) and ephrinB2, as drivers of fibrosis and tumor progression after radiation therapy (RT). RT resulted in increased ADAM10 expression in tumor cells, leading to cleavage of ephrinB2, which was also detected in plasma. Pharmacologic or genetic targeting of ADAM10 decreased RT-induced fibrosis and tissue tension, tumor cell migration, and invasion, sensitizing orthotopic tumors to radiation killing and prolonging mouse survival. Inhibition of ADAM10 and genetic ablation of ephrinB2 in fibroblasts reduced the metastatic potential of tumor cells after RT. Stimulation of tumor cells with ephrinB2 FC protein reversed the reduction in tumor cell invasion with ADAM10 ablation. These findings represent a model of PDAC adaptation that explains resistance and metastasis after RT and identifies a targetable pathway to enhance RT efficacy. SIGNIFICANCE: Targeting a previously unidentified adaptive resistance mechanism to radiation therapy in PDAC tumors in combination with radiation therapy could increase survival of the 40% of PDAC patients with locally advanced disease.See related commentary by Garcia Garcia et al., p. 3158 GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/12/3255/F1.large.jpg.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Carcinoma, Pancreatic Ductal/radiotherapy , Epithelial-Mesenchymal Transition , Fibrosis/pathology , Gamma Rays/adverse effects , Membrane Proteins/metabolism , Pancreatic Neoplasms/radiotherapy , Radiation Injuries/pathology , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , Antifibrotic Agents/therapeutic use , Apoptosis , Carcinoma, Pancreatic Ductal/pathology , Cell Movement , Cell Proliferation , Ephrin-B2/blood , Female , Fibrosis/drug therapy , Fibrosis/etiology , Fibrosis/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/pathology , Prognosis , Radiation Injuries/drug therapy , Radiation Injuries/etiology , Radiation Injuries/metabolism , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cancer Immunol Immunother ; 70(4): 989-1000, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33097963

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a heterogeneous tumor microenvironment (TME) comprised of myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages, neutrophils, regulatory T cells, and myofibroblasts. The precise mechanisms that regulate the composition of the TME and how they contribute to radiotherapy (RT) response remain poorly understood. In this study, we analyze changes in immune cell populations and circulating chemokines in patient samples and animal models of pancreatic cancer to characterize the immune response to radiotherapy. Further, we identify STAT3 as a key mediator of immunosuppression post-RT. We found granulocytic MDSCs (G-MDSCs) and neutrophils to be increased in response to RT in murine and human PDAC samples. We also found that RT-induced STAT3 phosphorylation correlated with increased MDSC infiltration and proliferation. Targeting STAT3 using an anti-sense oligonucleotide in combination with RT circumvented RT-induced MDSC infiltration, enhanced the proportion of effector T cells, and improved response to RT. In addition, STAT3 inhibition contributed to the remodeling of the PDAC extracellular matrix when combined with RT, resulting in decreased collagen deposition and fibrotic tissue formation. Collectively, our data provide evidence that targeting STAT3 in combination with RT can mitigate the pro-tumorigenic effects of RT and improve tumor response.


Subject(s)
Carcinoma, Pancreatic Ductal/radiotherapy , Gamma Rays , Myeloid-Derived Suppressor Cells/immunology , Oligonucleotides, Antisense/genetics , Pancreatic Neoplasms/radiotherapy , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Female , Humans , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Mice, Nude , Myeloid-Derived Suppressor Cells/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Prognosis , STAT3 Transcription Factor/genetics , T-Lymphocytes, Regulatory/immunology , Tumor Cells, Cultured , Tumor Microenvironment
7.
Cells ; 9(12)2020 12 08.
Article in English | MEDLINE | ID: mdl-33302501

ABSTRACT

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has recently emerged in China and caused a disease called coronavirus disease 2019 (COVID-19). The virus quickly spread around the world, causing a sustained global outbreak. Although SARS-CoV-2, and other coronaviruses, SARS-CoV and Middle East respiratory syndrome CoV (MERS-CoV) are highly similar genetically and at the protein production level, there are significant differences between them. Research has shown that the structural spike (S) protein plays an important role in the evolution and transmission of SARS-CoV-2. So far, studies have shown that various genes encoding primarily for elements of S protein undergo frequent mutation. We have performed an in-depth review of the literature covering the structural and mutational aspects of S protein in the context of SARS-CoV-2, and compared them with those of SARS-CoV and MERS-CoV. Our analytical approach consisted in an initial genome and transcriptome analysis, followed by primary, secondary and tertiary protein structure analysis. Additionally, we investigated the potential effects of these differences on the S protein binding and interactions to angiotensin-converting enzyme 2 (ACE2), and we established, after extensive analysis of previous research articles, that SARS-CoV-2 and SARS-CoV use different ends/regions in S protein receptor-binding motif (RBM) and different types of interactions for their chief binding with ACE2. These differences may have significant implications on pathogenesis, entry and ability to infect intermediate hosts for these coronaviruses. This review comprehensively addresses in detail the variations in S protein, its receptor-binding characteristics and detailed structural interactions, the process of cleavage involved in priming, as well as other differences between coronaviruses.


Subject(s)
Angiotensin-Converting Enzyme 2/chemistry , Middle East Respiratory Syndrome Coronavirus/metabolism , SARS-CoV-2/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Binding Sites , COVID-19/pathology , COVID-19/virology , Humans , Middle East Respiratory Syndrome Coronavirus/genetics , Molecular Dynamics Simulation , Protein Structure, Tertiary , Severe acute respiratory syndrome-related coronavirus/genetics , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Spike Glycoprotein, Coronavirus/metabolism
8.
Clin Cancer Res ; 26(12): 3058-3070, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32139402

ABSTRACT

PURPOSE: SMAD4 loss causes genomic instability and the initiation/progression of head and neck squamous cell carcinoma (HNSCC). Here, we study whether SMAD4 loss sensitizes HNSCCs to olaparib (PARP inhibitor) in combination with radiotherapy (RT). EXPERIMENTAL DESIGN: We analyzed HNSCC The Cancer Genome Atlas data for SMAD4 expression in association with FANC/BRCA family gene expression. Human HNSCC cell lines were screened for sensitivity to olaparib. Isogenic HNSCC cell lines were generated to restore or reduce SMAD4 expression and treated with olaparib, radiation, or the combination. HNSCC pretreatment specimens from a phase I trial investigating olaparib were analyzed. RESULTS: SMAD4 levels correlated with levels of FANC/BRCA genes in HNSCC. HNSCC cell lines with SMAD4 homozygous deletion were sensitive to olaparib. In vivo, olaparib or RT monotherapy reduced tumor volumes in SMAD4-mutant but not SMAD4-positive tumors. Olaparib with RT dual therapy sustained tumor volume reduction in SMAD4-deficient (mutant or knockdown) xenografts, which exhibited increased DNA damage and cell death compared with vehicle-treated tumors. In vitro, olaparib alone or in combination with radiation caused lower clonogenic survival, more DNA damage-associated cell death, and less proliferation in SMAD4-deficient cells than in SMAD4-positive (endogenous SMAD4 or transduced SMAD4) cells. Applicable to clinic, 5 out of 6 SMAD4-negative HNSCCs and 4 out of 8 SMAD4-positive HNSCCs responded to a standard treatment plus olaparib in a phase I clinical trial, and SMAD4 protein levels inversely correlated with DNA damage. CONCLUSIONS: SMAD4 levels are causal in determining sensitivity to PARP inhibition in combination with RT in HNSCCs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Disease Models, Animal , Head and Neck Neoplasms/radiotherapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Smad4 Protein/deficiency , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Animals , Apoptosis , Cell Proliferation , Cetuximab/administration & dosage , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice , Mice, Nude , Phthalazines/administration & dosage , Piperazines/administration & dosage , Prognosis , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
J Vis Exp ; (146)2019 04 22.
Article in English | MEDLINE | ID: mdl-31058896

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is a debilitating and deadly disease with a high prevalence of recurrence and treatment failure. To develop better therapeutic strategies, understanding tumor microenvironmental factors that contribute to the treatment resistance is important. A major impediment to understanding disease mechanisms and improving therapy has been a lack of murine cell lines that resemble the aggressive and metastatic nature of human HNSCCs. Furthermore, a majority of murine models employ subcutaneous implantations of tumors which lack important physiological features of the head and neck region, including high vascular density, extensive lymphatic vasculature, and resident mucosal flora. The purpose of this study is to develop and characterize an orthotopic model of HNSCC. We employ two genetically distinct murine cell lines and established tumors in the buccal mucosa of mice. We optimize collagenase-based tumor digestion methods for the optimal recovery of single cells from established tumors. The data presented here show that mice develop highly vascularized tumors that metastasize to regional lymph nodes. Single-cell multiparametric mass cytometry analysis shows the presence of diverse immune populations with myeloid cells representing the majority of all immune cells. The model proposed in this study has applications in cancer biology, tumor immunology, and preclinical development of novel therapeutics. The resemblance of the orthotopic model to clinical features of human disease will provide a tool for enhanced translation and improved patient outcomes.


Subject(s)
Head and Neck Neoplasms , Neoplasms, Experimental , Squamous Cell Carcinoma of Head and Neck , Animals , Cell Line, Tumor , Feasibility Studies , Head and Neck Neoplasms/immunology , Humans , Lymph Nodes/pathology , Mice , Mice, Inbred BALB C , Mouth Mucosa/pathology , Neoplasm Transplantation , Squamous Cell Carcinoma of Head and Neck/immunology , Tumor Microenvironment
10.
J Natl Cancer Inst ; 111(12): 1339-1349, 2019 12 01.
Article in English | MEDLINE | ID: mdl-30863843

ABSTRACT

BACKGROUND: Radioresistance represents a major problem in the treatment of head and neck cancer (HNC) patients. To improve response, understanding tumor microenvironmental factors that contribute to radiation resistance is important. Regulatory T cells (Tregs) are enriched in numerous cancers and can dampen the response to radiation by creating an immune-inhibitory microenvironment. The purpose of this study was to investigate mechanisms of Treg modulation by radiation in HNC. METHODS: We utilized an orthotopic mouse model of HNC. Anti-CD25 was used for Treg depletion. Image-guided radiation was delivered to a dose of 10 Gy. Flow cytometry was used to analyze abundance and function of intratumoral immune cells. Enzyme-linked immunosorbent assay was performed to assess secreted factors. For immune-modulating therapies, anti-PD-L1, anti-CTLA-4, and STAT3 antisense oligonucleotide (ASO) were used. All statistical tests were two-sided. RESULTS: Treatment with anti-CD25 and radiation led to tumor eradication (57.1%, n = 4 of 7 mice), enhanced T-cell cytotoxicity compared with RT alone (CD4 effector T cells [Teff]: RT group mean = 5.37 [ 0.58] vs RT + αCD25 group mean =10.71 [0.67], P = .005; CD8 Teff: RT group mean = 9.98 [0.81] vs RT + αCD25 group mean =16.88 [2.49], P = .01) and induced tumor antigen-specific memory response (100.0%, n = 4 mice). In contrast, radiation alone or when combined with anti-CTLA4 did not lead to durable tumor control (0.0%, n = 7 mice). STAT3 inhibition in combination with radiation, but not as a single agent, improved tumor growth delay, decreased Tregs, myeloid-derived suppressor cells, and M2 macrophages and enhanced effector T cells and M1 macrophages. Experiments in nude mice inhibited the benefit of STAT3 ASO and radiation. CONCLUSION: We propose that STAT3 inhibition is a viable and potent therapeutic target against Tregs. Our data support the design of clinical trials integrating STAT3 ASO in the standard of care for cancer patients receiving radiation.


Subject(s)
Head and Neck Neoplasms/radiotherapy , Lymphocyte Depletion , Radioimmunotherapy/methods , STAT3 Transcription Factor/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck/radiotherapy , T-Lymphocytes, Regulatory/radiation effects , Analysis of Variance , Animals , Cytotoxicity, Immunologic , Female , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Humans , Immunologic Memory , Interleukin-2 Receptor alpha Subunit/genetics , Lymphocyte Depletion/methods , Macrophages/radiation effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Myeloid-Derived Suppressor Cells/radiation effects , Radiation Tolerance , Radiotherapy, Image-Guided , STAT3 Transcription Factor/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/genetics , Tumor Microenvironment
11.
Front Immunol ; 9: 3154, 2018.
Article in English | MEDLINE | ID: mdl-30766539

ABSTRACT

Radiation therapy has been used for many years to treat tumors based on its DNA-damage-mediated ability to kill cells. More recently, RT has been shown to exert beneficial modulatory effects on immune responses, such as triggering immunogenic cell death, enhancing antigen presentation, and activating cytotoxic T cells. Consequently, combining radiation therapy with immunotherapy represents an important area of research. Thus far, immune-checkpoint inhibitors targeting programmed death-ligand 1 (PD-L1), programmed cell death protein 1 (PD-1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have been the focus of many research studies and clinical trials. The available data suggest that such immunotherapies are enhanced when combined with radiation therapy. However, treatment resistance, intrinsic or acquired, is still prevalent. Various theories as to how to enhance these combination therapies to overcome treatment resistance have been proposed. In this review, we focus on the principles surrounding radiation therapy's positive and negative effects on the tumor microenvironment. We explore mechanisms underlying radiation therapy's synergistic and antagonistic effects on immune responses and provide a base of knowledge for radio-immunology combination therapies to overcome treatment resistance. We provide evidence for targeting regulatory T cells, tumor-associated macrophages, and cancer-associated fibroblasts in combination radio-immunotherapies to improve cancer treatment.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Drug Resistance, Neoplasm , Endothelial Cells/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Radiation Tolerance , Radiotherapy , Signal Transduction , Animals , Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Combined Modality Therapy , Cytotoxicity, Immunologic , Endothelial Cells/drug effects , Endothelial Cells/radiation effects , Extracellular Matrix/metabolism , Humans , Immunomodulation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Neoplasms/etiology , Neoplasms/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
12.
J Neuroimmunol ; 313: 118-122, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29153603

ABSTRACT

BACKGROUND: Patients with melanoma brain metastases (MBM) have been excluded from trials evaluating immunotherapy in melanoma. As such, immunotherapy's role in MBM is poorly understood, particularly in combination with radiotherapy. METHODS: The National Cancer Database was queried for patients with MBM receiving brain radiotherapy. They were classified according to immunotherapy receipt. Multivariate Cox regression was performed to identify factors associated with survival. RESULTS: Among 1287 patients, 185 received immunotherapy. Factors associated with improved survival included younger age, academic facility, lower extracranial disease burden, stereotactic radiotherapy, chemotherapy, and immunotherapy. CONCLUSIONS: Adding immunotherapy to radiotherapy for MBM is associated with improved survival.


Subject(s)
Brain Neoplasms/secondary , Brain Neoplasms/therapy , Cranial Irradiation/methods , Immunotherapy/methods , Melanoma/pathology , Adult , Age Distribution , Aged , Brain Neoplasms/mortality , Databases, Factual/statistics & numerical data , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged
13.
Cancer ; 123(4): 600-608, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27727461

ABSTRACT

BACKGROUND: Although laryngectomy is the treatment of choice for patients with T4 larynx cancer, many patients are unable or unwilling to undergo laryngectomy and instead pursue larynx-preservation strategies combining radiotherapy (RT) and chemotherapy. Herein, the authors analyzed the National Cancer Data Base to evaluate overall survival (OS) between patients treated with surgical and organ-preserving modalities. METHODS: The National Cancer Data Base was queried for patients diagnosed from 2004 through 2012 with T4M0 laryngeal cancer who underwent either laryngectomy (surgery) with adjuvant RT (SRT), chemotherapy starting concurrently within 7 days of RT (CCRT), or multiagent induction chemotherapy starting 43 to 98 days before RT (ICRT). Multivariate analysis and propensity score matching were used to explore the association between the intervention and OS. Recursive partitioning analysis was performed to identify groups benefiting from particular modalities. RESULTS: A total of 1559 patients who underwent SRT, 1597 patients who underwent CCRT, and 386 patients who underwent ICRT were included. Adjusting for covariates, CCRT was found to be associated with inferior OS compared with SRT (hazard ratio [HR], 1.55; 95% confidence interval [95% CI], 1.41-1.70 [P<.01]) and with ICRT (HR, 1.25; 95% CI, 1.07-1.45 [P<.01]). OS among the patients treated with SRT did not appear to differ significantly from that of the ICRT cohort (HR, 0.87; 95% CI, 0.73-1.03 [P = 0.10]), a finding confirmed with propensity score matching. Recursive partitioning analysis identified no subset of patients that derived an OS benefit from either approach over the other. CONCLUSIONS: OS among patients undergoing SRT was found to be superior to that of patients treated with CCRT but did not significantly differ from that of those undergoing ICRT. Because these intriguing findings require validation, SRT should remain the standard of care for patients with this disease. However, organ preservation with ICRT may be a reasonable alternative in certain patients. Cancer 2017;123:600-608. © 2016 American Cancer Society.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Laryngeal Neoplasms/drug therapy , Laryngeal Neoplasms/radiotherapy , Laryngeal Neoplasms/surgery , Adult , Aged , Cisplatin/therapeutic use , Combined Modality Therapy/adverse effects , Disease-Free Survival , Female , Humans , Laryngeal Neoplasms/epidemiology , Laryngectomy/adverse effects , Male , Middle Aged , Neoplasm Staging , Radiotherapy, Adjuvant/adverse effects
14.
J Magn Reson Imaging ; 26(1): 144-51, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17659552

ABSTRACT

PURPOSE: To determine the contribution of blood-derived macrophages to the signal loss observed in MR images of inflammatory lesions in experimental autoimmune encephalomyelitis (EAE). MATERIALS AND METHODS: A relapsing-remitting form of EAE was induced in transgenic mice that express enhanced green fluorescent protein (EGFP) specifically in hematopoietic cells of the myelomonocytic lineage. Animals were injected with Feridex, a superparamagnetic iron oxide (SPIO) nanoparticle, 24 hours prior to in vivo MRI. MRI was performed using a 1.5T whole-body scanner; a high-performance, custom-built gradient coil insert; and a 3D steady-state free precession (SSFP) imaging pulse sequence. Comparisons were made between MR images and corresponding anti-GFP and Perl's Prussian blue (PPB)-stained brain sections. RESULTS: MR images revealed the presence of discrete regions of signal loss throughout the brains of EAE animals that were administered Feridex. Histological staining showed that regions of signal loss on MR images corresponded anatomically with regions of PPB- and GFP-positive cells. CONCLUSION: This experiment provides the first direct evidence that macrophages of hematogenous origin are labeled with SPIO after intravenous administration of Feridex, and contribute to the regions of signal loss detected in MR images of EAE brain.


Subject(s)
Contrast Media/pharmacokinetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Iron/pharmacokinetics , Macrophages/cytology , Magnetic Resonance Imaging/methods , Oxides/pharmacokinetics , Animals , Dextrans , Disease Models, Animal , Female , Ferrosoferric Oxide , Image Interpretation, Computer-Assisted , Magnetite Nanoparticles , Mice , Mice, Transgenic , Microscopy, Fluorescence , Reproducibility of Results
15.
Mol Imaging ; 3(2): 85-95, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15296673

ABSTRACT

The ability to visualize cell infiltration in experimental auto-immune encephalomyelitis (EAE), a well-known animal model for multiple sclerosis in humans, was investigated using a clinical 1.5-T magnetic resonance imaging (MRI) scanner, a custom-built, high-strength gradient coil insert, a 3-D fast imaging employing steady-state acquisition (FIESTA) imaging sequence and a superparamagnetic iron oxide (SPIO) contrast agent. An "active labeling" approach was used with SPIO administered intravenously during inflammation in EAE. Our results show that small, discrete regions of signal void corresponding to iron accumulation in EAE brain can be detected using FIESTA at 1.5 T. This work provides early evidence that cellular abnormalities that are the basis of diseases can be probed using cellular MRI and supports our earlier work which indicates that tracking of iron-labeled cells will be possible using clinical MR scanners.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Ferric Compounds , Image Interpretation, Computer-Assisted , Inflammation/pathology , Magnetic Resonance Imaging/methods , Animals , Disease Models, Animal , Feasibility Studies , Female , Gadolinium , Inflammation/etiology , Multiple Sclerosis/pathology , Rats , Rats, Inbred Lew , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL