Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Front Immunol ; 12: 754436, 2021.
Article in English | MEDLINE | ID: mdl-34733287

ABSTRACT

Cancer patients with low or absent pre-existing anti-tumour immunity ("cold" tumours) respond poorly to treatment with immune checkpoint inhibitors (ICPI). In order to render these patients susceptible to ICPI, initiation of de novo tumour-targeted immune responses is required. This involves triggering of inflammatory signalling, innate immune activation including recruitment and stimulation of dendritic cells (DCs), and ultimately priming of tumour-specific T cells. The ability of tumour localised therapies to trigger these pathways and act as in situ tumour vaccines is being increasingly explored, with the aspiration of developing combination strategies with ICPI that could generate long-lasting responses. In this effort, it is crucial to consider how therapy-induced changes in the tumour microenvironment (TME) act both as immune stimulants but also, in some cases, exacerbate immune resistance mechanisms. Increasingly refined immune monitoring in pre-clinical studies and analysis of on-treatment biopsies from clinical trials have provided insight into therapy-induced biomarkers of response, as well as actionable targets for optimal synergy between localised therapies and ICB. Here, we review studies on the immunomodulatory effects of novel and experimental localised therapies, as well as the re-evaluation of established therapies, such as radiotherapy, as immune adjuvants with a focus on ICPI combinations.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Immunomodulation , Immunotherapy/methods , Neoplasms/therapy , Adjuvants, Immunologic/therapeutic use , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/radiation effects , Cancer Vaccines/therapeutic use , Combined Modality Therapy , Dendritic Cells/immunology , Drug Screening Assays, Antitumor , Drug Synergism , Herpesvirus 1, Human/physiology , Humans , Immunity, Innate/drug effects , Immunomodulating Agents/therapeutic use , Immunomodulation/drug effects , Immunomodulation/radiation effects , Mice , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/radiotherapy , Oncolytic Virotherapy , T-Lymphocyte Subsets/immunology , Tumor Microenvironment/immunology
2.
J Photochem Photobiol B ; 221: 112247, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34175580

ABSTRACT

A need exists for further research elucidating the benefits of environmentally safe photoprotective agents against ultraviolet (UV) exposure, and plant extracts represent a human-friendly alternative formulation. This study was designed to evaluate the potential use of Bellis perennis extract (BPE), from the Asteraceae family, known as the common daisy or the English daisy, in cosmeceuticals as a photoprotective factor, using an in vitro model of UVA-induced keratinocyte damage. Human skin keratinocytes (HaCaT cell line) were incubated with BPE at 0.01, 0.1, or 1% in Dulbecco's Modified Eagle Medium (DMEM), and after 15 min they were submitted to UVA radiation at 5, 10, and 15 J/cm2 doses, respectively. For comparative purposes, Polypodium leucotomos extract (PLE), known as the fern, was used as a positive control in assessing the photoprotective effect. After 24 h of UVA exposure, cell viability (MTT and LDH assays), levels of cleaved caspase-3, cyclooxygenase-2, IL-6, reactive oxygen species (ROS) and antioxidant enzyme (catalase, SOD, and glutathione peroxidase) activity were determined. UVA radiation at 5, 10, and 15 J/cm2 doses reduced cell viability to 63%, 43%, and 23%, respectively; we selected 10 J/cm2 for our purposes. After 24 h of UVA exposure, treatment with 1% BPE and 1% PLE significantly recovered cell viability (p < 0.05). Furthermore, treatment was associated with lower cleaved caspase-3 and ROS levels, higher catalase activity, and lower IL-6 levels in the treated UVA keratinocytes compared with the untreated UVA group (p < 0.01). Our results demonstrate photoprotective and immunomodulatory effects of BPE in skin keratinocytes and support its use as a bioactive agent in cosmetic formulations to prevent skin damage caused by exposure to the UV light.


Subject(s)
Asteraceae/chemistry , Immunomodulation/drug effects , Plant Extracts/pharmacology , Radiation-Protective Agents/pharmacology , Ultraviolet Rays , Asteraceae/metabolism , Caspase 3/metabolism , Catalase/metabolism , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Humans , Immunomodulation/radiation effects , Keratinocytes/cytology , Keratinocytes/metabolism , Plant Extracts/chemistry , Radiation-Protective Agents/chemistry , Reactive Oxygen Species/metabolism
3.
Front Immunol ; 12: 617365, 2021.
Article in English | MEDLINE | ID: mdl-33936033

ABSTRACT

In situ tumor ablation techniques, like radiotherapy, cryo- and heat-based thermal ablation are successfully applied in oncology for local destruction of tumor masses. Although diverse in technology and mechanism of inducing cell death, ablative techniques share one key feature: they generate tumor debris which remains in situ. This tumor debris functions as an unbiased source of tumor antigens available to the immune system and has led to the concept of in situ cancer vaccination. Most studies, however, report generally modest tumor-directed immune responses following local tumor ablation as stand-alone treatment. Tumors have evolved mechanisms to create an immunosuppressive tumor microenvironment (TME), parts of which may admix with the antigen depot. Provision of immune stimuli, as well as approaches that counteract the immunosuppressive TME, have shown to be key to boost ablation-induced anti-tumor immunity. Recent advances in protein engineering have yielded novel multifunctional antibody formats. These multifunctional antibodies can provide a combination of distinct effector functions or allow for delivery of immunomodulators specifically to the relevant locations, thereby mitigating potential toxic side effects. This review provides an update on immune activation strategies that have been tested to act in concert with tumor debris to achieve in situ cancer vaccination. We further provide a rationale for multifunctional antibody formats to be applied together with in situ ablation to boost anti-tumor immunity for local and systemic tumor control.


Subject(s)
Ablation Techniques , Immunomodulation/drug effects , Immunomodulation/radiation effects , Neoplasms/immunology , Neoplasms/therapy , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Neoplasm/immunology , Combined Modality Therapy , Humans , Immunosuppression Therapy , Immunotherapy/methods , Neoplasms/pathology , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
4.
J Immunol Res ; 2021: 6664453, 2021.
Article in English | MEDLINE | ID: mdl-33628851

ABSTRACT

BACKGROUND: The immune mechanisms underlying low-intensity ultrasound- (LIUS-) mediated suppression of inflammation and tumorigenesis remain poorly determined. METHODS: We used microarray datasets from the NCBI GEO DataSet repository and conducted comprehensive data-mining analyses, where we examined the gene expression of 1376 innate immune regulators (innatome genes (IGs) in cells treated with LIUS. RESULTS: We made the following findings: (1) LIUS upregulates proinflammatory IGs and downregulates metastasis genes in cancer cells, and LIUS upregulates adaptive immunity pathways but inhibits danger-sensing and inflammation pathways and promote tolerogenic differentiation in bone marrow (BM) cells. (2) LIUS upregulates IGs encoded for proteins localized in the cytoplasm, extracellular space, and others, but downregulates IG proteins localized in nuclear and plasma membranes, and LIUS downregulates phosphatases. (3) LIUS-modulated IGs act partially via several important pathways of reactive oxygen species (ROS), reverse signaling of immune checkpoint receptors B7-H4 and BTNL2, inflammatory cytokines, and static or oscillatory shear stress and heat generation, among which ROS is a dominant mechanism. (4) LIUS upregulates trained immunity enzymes in lymphoma cells and downregulates trained immunity enzymes and presumably establishes trained tolerance in BM cells. (5) LIUS modulates chromatin long-range interactions to differentially regulate IGs expression in cancer cells and noncancer cells. CONCLUSIONS: Our analysis suggests novel molecular mechanisms that are utilized by LIUS to induce tumor suppression and inflammation inhibition. Our findings may lead to development of new treatment protocols for cancers and chronic inflammation.


Subject(s)
Cytokines/metabolism , Immune Checkpoint Proteins/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Escape/immunology , Ultrasonic Waves , Adaptive Immunity , Cells, Cultured , Gene Expression Profiling , Humans , Hyperthermia, Induced/methods , Immune Checkpoint Proteins/genetics , Immunity, Innate , Immunomodulation/radiation effects , Models, Biological , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction/radiation effects
6.
Int J Radiat Oncol Biol Phys ; 109(4): 867-879, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33340603

ABSTRACT

PURPOSE: Phase 1 clinical trials have established low-dose, whole-lung radiation therapy (LD-RT) as safe for patients with coronavirus disease 2019 (COVID-19)-related pneumonia. By focally dampening cytokine hyperactivation, LD-RT may improve disease outcomes through immunomodulation. METHODS AND MATERIALS: Patients with COVID-19-related pneumonia were treated with 1.5 Gy whole-lung LD-RT, followed for 28 days or until hospital discharge, and compared with age- and comorbidity-matched controls meeting identical disease severity criteria. Eligible patients were hospitalized, severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2) positive, had radiographic consolidations, and required supplemental oxygen but had not rapidly declined on admission or before drug therapy or LD-RT. Efficacy endpoints were time to clinical recovery, radiographic improvement, and biomarker response. RESULTS: Ten patients received whole-lung LD-RT between April 24 and May 24, 2020 and were compared with 10 control patients blindly matched by age and comorbidity. Six controls received COVID-19 drug therapies. Median time to clinical recovery was 12 days in the control cohort compared with 3 days in the LD-RT cohort (hazard ratio 2.9, P = .05). Median time to hospital discharge (20 vs 12 days, P = .19) and intubation rates (40% vs 10%, P = .12) in the control and LD-RT cohorts were compared. Median time from admission to recovery was 10 versus 13 days (P = .13). Hospital duration average was 19 versus 22.6 days (P = .53). Average hospital days on supplemental oxygen of any duration was 13.1 versus 14.7 days (P = .69). Average days with a documented fever was 1 versus 4.3 days (P = .12). Twenty-eight-day overall survival was 90% for both cohorts. The LD-RT cohort trended toward superior rates of improved radiographs (P = .12) and delirium (P < .01). Statistically significant reductions were observed in numerous hematologic, cardiac, hepatic, and inflammatory markers. CONCLUSIONS: A prospective cohort of predominantly elderly hospitalized patients with COVID-19-related pneumonia were recovered to room air quicker than age- and comorbidity-matched controls, with trending or significant improvements in delirium, radiographs, and biomarkers, and no significant acute toxicity. Low-dose, whole-lung radiation for patients with COVID-19-related pneumonia appears safe and may be an effective immunomodulatory treatment. Larger prospective randomized trials are needed to define the efficacy of LD-RT for COVID-19.


Subject(s)
COVID-19/immunology , COVID-19/radiotherapy , Immunomodulation/radiation effects , Lung/radiation effects , Radiation Dosage , Adult , Aged , Aged, 80 and over , COVID-19/blood , COVID-19/diagnostic imaging , Female , Hospitalization , Humans , Lung/diagnostic imaging , Lung/immunology , Male , Middle Aged , Radiotherapy Dosage , Safety , Tomography, X-Ray Computed , Treatment Outcome
7.
Commun Biol ; 3(1): 783, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33335270

ABSTRACT

Thermal ablation is a standard therapy for patients with hepatocellular carcinoma (HCC). Contemporary ablation devices are imperfect, as they lack tumor specificity. An ideal ablation modality would generate thermal energy only within tumoral tissue. Furthermore, as hyperthermia is known to influence tumor immunity, such a tumor-specific ablation modality may have the ability to favorably modulate the tumor immune landscape. Here we show a clinically relevant thermal ablation modality that generates tumor-specific hyperthermia, termed molecularly targeted photothermal ablation (MTPA), that is based upon the excellent localization of indocyanine green to HCC. In a syngeneic rat model, we demonstrate the tumor-specific hyperthermia generated by MTPA. We also show through spatial and transcriptomic profiling techniques that MTPA favorably modulates the intratumoral myeloid population towards tumor immunogenicity and diminishes the systemic release of oncogenic cytokines relative to conventional ablation modalities.


Subject(s)
Carcinoma, Hepatocellular/etiology , Immunomodulation/radiation effects , Liver Neoplasms/etiology , Photothermal Therapy/methods , Animals , Biomarkers, Tumor , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cytokines/metabolism , Disease Models, Animal , Flow Cytometry , Gene Expression , Hyperthermia, Induced , Immunomodulation/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Molecular Targeted Therapy , Rats , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
8.
Sci Rep ; 10(1): 21517, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33299018

ABSTRACT

While for photon radiation hypofractionation has been reported to induce enhanced immunomodulatory effects, little is known about the immunomodulatory potential of carbon ion radiotherapy (CIRT). We thus compared the radio-immunogenic effects of photon and carbon ion irradiation on two murine cancer cell lines of different tumor entities. We first calculated the biological equivalent doses of carbon ions corresponding to photon doses of 1, 3, 5, and 10 Gy of the murine breast cancer cell line EO771 and the OVA-expressing pancreatic cancer cell line PDA30364/OVA by clonogenic survival assays. We compared the potential of photon and carbon ion radiation to induce cell cycle arrest, altered surface expression of immunomodulatory molecules and changes in the susceptibility of cancer cells to cytotoxic T cell (CTL) mediated killing. Irradiation induced a dose-dependent G2/M arrest in both cell lines irrespective from the irradiation source applied. Likewise, surface expression of the immunomodulatory molecules PD-L1, CD73, H2-Db and H2-Kb was increased in a dose-dependent manner. Both radiation modalities enhanced the susceptibility of tumor cells to CTL lysis, which was more pronounced in EO771/Luci/OVA cells than in PDA30364/OVA cells. Overall, compared to photon radiation, the effects of carbon ion radiation appeared to be enhanced at higher dose range for EO771 cells and extenuated at lower dose range for PDA30364/OVA cells. Our data show for the first time that equivalent doses of carbon ion and photon irradiation exert similar immunomodulating effects on the cell lines of both tumor entities, highlighted by an enhanced susceptibility to CTL mediated cytolysis in vitro.


Subject(s)
Heavy Ion Radiotherapy/methods , Immunomodulation/radiation effects , Photons/therapeutic use , Animals , Apoptosis/drug effects , Carbon/pharmacology , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Humans , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms
9.
Semin Radiat Oncol ; 30(2): 181-186, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32381297

ABSTRACT

Recent preclinical and clinical studies have elucidated mechanisms whereby radiation therapy influences the anti-tumor immune response. Immunogenic cell death and phenotypic changes in tumor cells surviving radiation may underlie this effect and contribute to the capacity of radiation to elicit an in situ tumor vaccine effect. In situ vaccination is a therapeutic strategy that seeks to convert a patient's own tumor into a source of enhanced antigen recognition for the purpose of augmenting a systemic anti-tumor immune response. Capitalizing on the in situ vaccine effect of radiation, several groups have demonstrated anti-tumor efficacy in preclinical models by combining radiation with immune checkpoint blockade. Local delivery of immune adjuvants and/or immune stimulatory cytokines via direct injection into the radiated tumor microenvironment may further increase the in situ vaccine capacity of radiation therapy. However, recent studies suggest that in some contexts this effect is antagonized by the presence of distant untreated sites of disease that may dampen the systemic immune response generated by in situ vaccination through a phenomenon termed concomitant immune tolerance. Concomitant immune tolerance may be overcome by delivering radiation to all sites of metastatic disease, however this is often not possible to safely achieve using external beam radiation therapy without considerable risk of lymphopenia that would negate the immune effects of in situ vaccination. For patients with widespread metastatic disease, alternative strategies may include systemic treatment with targeted radionuclide therapies alone or in combination with an external beam radiation therapy-based in situ vaccine approach.


Subject(s)
Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Combined Modality Therapy , Cytokines/immunology , Cytokines/pharmacology , Disease Models, Animal , Humans , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/pharmacology , Immunomodulation/immunology , Immunomodulation/radiation effects , Radiation Dose Hypofractionation , Vaccination
10.
Nat Immunol ; 21(6): 649-659, 2020 06.
Article in English | MEDLINE | ID: mdl-32424359

ABSTRACT

Efficient generation of germinal center (GC) responses requires directed movement of B cells between distinct microenvironments underpinned by specialized B cell-interacting reticular cells (BRCs). How BRCs are reprogrammed to cater to the developing GC remains unclear, and studying this process is largely hindered by incomplete resolution of the cellular composition of the B cell follicle. Here we used genetic targeting of Cxcl13-expressing cells to define the molecular identity of the BRC landscape. Single-cell transcriptomic analysis revealed that BRC subset specification was predetermined in the primary B cell follicle. Further topological remodeling of light and dark zone follicular dendritic cells required CXCL12-dependent crosstalk with B cells and dictated GC output by retaining B cells in the follicle and steering their interaction with follicular helper T cells. Together, our results reveal that poised BRC-defined microenvironments establish a feed-forward system that determines the efficacy of the GC reaction.


Subject(s)
Darkness , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Immunomodulation/radiation effects , Light , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Cell Communication , Chemokine CXCL12/metabolism , Mice , Mice, Transgenic , Phenotype , Single-Cell Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
11.
Eur J Immunol ; 50(5): 725-735, 2020 05.
Article in English | MEDLINE | ID: mdl-32012249

ABSTRACT

Extracorporeal photochemotherapy (ECP) that takes advantage of the immunomodulatory effects of UV light has been extensively used for many years for the treatment of several T cell-mediated diseases, including graft-versus-host disease (GvHD) and systemic scleroderma. Immune mechanisms that lead to the establishment of T cell tolerance in ECP-treated patients remain poorly known. In this study, we have tested the effect of UV/psoralen-treated BM-derived dendritic cells, referred to as ECP-BMDCs on the outcome of an antigen-specific T cell-mediated reaction, that is, contact hypersensitivity (CHS), which is mediated by CD8+ effector T cells (CD8+ Teff ). The intravenous (i.v.) injection of antigen-pulsed ECP-BMDCs in recipient C57BL/6 mice induced specific CD8+ T cells endowed with immunomodulatory properties (referred to as CD8+ TECP ), which prevented the priming of CD8+ Teff and the development of CHS, independently of conventional CD4+ regulatory T cells. CD8+ TECP mediated tolerance by inhibiting the migration and functions of skin DC and subsequently the priming of CD8+ Teff . CD8+ TECP displayed none of the phenotypes of the usual CD8+ T regulatory cells described so far. Our results reveal an underestimated participation of CD8+ T cells to ECP-induced immunomodulation that could explain the therapeutic effects of ECP in T cell-mediated diseases.


Subject(s)
Dendritic Cells/immunology , Dermatitis, Contact/therapy , Immune Tolerance , Immunomodulation/radiation effects , T-Lymphocytes, Cytotoxic/radiation effects , T-Lymphocytes, Regulatory/radiation effects , Allergens/administration & dosage , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Dendritic Cells/cytology , Dendritic Cells/transplantation , Dermatitis, Contact/immunology , Dermatitis, Contact/physiopathology , Dinitrofluorobenzene/administration & dosage , Disease Models, Animal , Female , Ficusin/administration & dosage , Humans , Mice, Inbred C57BL , Mice, Transgenic , Photopheresis/methods , Photosensitizing Agents/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Ultraviolet Rays
12.
Cancer Immunol Immunother ; 69(2): 293-306, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31953578

ABSTRACT

Cancer immunotherapies are promising treatments for many forms of cancer. Nevertheless, the response rates to, e.g., immune checkpoint inhibitors (ICI), are still in low double-digit percentage. This calls for further therapy optimization that should take into account combination of immunotherapies with classical tumor therapies such as radiotherapy. By designing multimodal approaches, immune modulatory properties of certain radiation schemes, additional immune modulation by immunotherapy with ICI and hyperthermia, as well as patient stratification based on genetic and immune constitutions have to be considered. In this context, both the tumor and its microenvironment including cells of the innate and adaptive immune system have to be viewed in synopsis. Knowledge of immune activation and immune suppression by radiation is the basis for well-elaborated addition of certain immunotherapies. In this review, the focus is set on additional immune stimulation by hyperthermia and restoration of an immune response by ICI. The impact of radiation dose and fractionation on immune modulation in multimodal settings has to be considered, as the dynamics of the immune response and the timing between radiotherapy and immunotherapy. Another big challenge is the patient stratification that should be based on matrices of biomarkers, taking into account genetics, proteomics, radiomics, and "immunomics". One key aim is to turn immunological "cold" tumors into "hot" tumors, and to eliminate barriers of immune-suppressed or immune-excluded tumors. Comprehensive knowledge of immune alterations induced by radiation and immunotherapy when being applied together should be utilized for patient-adapted treatment planning and testing of innovative tumor therapies within clinical trials.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Drug Design , Immunomodulation/drug effects , Neoplasms/etiology , Neoplasms/therapy , Animals , Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor , Combined Modality Therapy , Humans , Hyperthermia, Induced/methods , Immunity , Immunologic Factors/pharmacology , Immunomodulation/radiation effects , Immunotherapy , Neoplasms/pathology , Research Design , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
13.
Oncogene ; 39(1): 187-203, 2020 01.
Article in English | MEDLINE | ID: mdl-31467431

ABSTRACT

Radiotherapy induces immune-related responses in cancer patients by various mechanisms. Here, we investigate the immunomodulatory role of tumor-derived microparticles (TMPs)-extracellular vesicles shed from tumor cells-following radiotherapy. We demonstrate that breast carcinoma cells exposed to radiation shed TMPs containing elevated levels of immune-modulating proteins, one of which is programmed death-ligand 1 (PD-L1). These TMPs inhibit cytotoxic T lymphocyte (CTL) activity both in vitro and in vivo, and thus promote tumor growth. Evidently, adoptive transfer of CTLs pre-cultured with TMPs from irradiated breast carcinoma cells increases tumor growth rates in mice recipients in comparison with control mice receiving CTLs pre-cultured with TMPs from untreated tumor cells. In addition, blocking the PD-1-PD-L1 axis, either genetically or pharmacologically, partially alleviates TMP-mediated inhibition of CTL activity, suggesting that the immunomodulatory effects of TMPs in response to radiotherapy is mediated, in part, by PD-L1. Overall, our findings provide mechanistic insights into the tumor immune surveillance state in response to radiotherapy and suggest a therapeutic synergy between radiotherapy and immune checkpoint inhibitors.


Subject(s)
B7-H1 Antigen/genetics , Breast Neoplasms/radiotherapy , Cell-Derived Microparticles/immunology , Immunomodulation/immunology , Animals , B7-H1 Antigen/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Cell Line, Tumor , Cell-Derived Microparticles/genetics , Cell-Derived Microparticles/radiation effects , Female , Heterografts , Humans , Immune Evasion/immunology , Immune Evasion/radiation effects , Immunomodulation/radiation effects , Mice , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/radiation effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/radiation effects
14.
Clin Cancer Res ; 26(4): 910-921, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31757878

ABSTRACT

PURPOSE: Radiotherapy (RT) is frequently used for local control of solid tumors using equal dose per fraction. Recently, single high-dose radiation has been used for ablation of solid tumors. In this report, we provide a novel immunological basis for radiation dose fractionation consisting of a single high-dose radiotherapy, followed by postablation modulation (PAM) with four daily low-dose fractions (22 Gy + 0.5 Gy × 4) to reprogram the tumor microenvironment by diminishing immune suppression, enabling infiltration of effector cells and increasing efficacy of tumor control. EXPERIMENTAL DESIGN: Palpable 3LL and 4T1 tumors in C57Bl/6 and Balb/c mice were irradiated with the Small-Animal Radiation Research Platform irradiator, and tumor growth and survival were monitored. Immunomodulation of tumor and immune cells in vitro and in vivo characterization of tumor-infiltrating immune effector cells were performed by FACS. For systemic application of PAM-RT, whole-lung irradiation was administered in 4T1-bearing Balb/c mice. RESULTS: We report significant tumor growth delays and increased survival in 3LL tumor-bearing mice with PAM. Primary tumor PAM-RT increased infiltration of immune effector cells and decreased Treg in irradiated tumors and secondary lymphoid organs. In a model of murine metastatic breast cancer (4T1), we demonstrated that systemic PAM-RT to the whole lung, 12 days after primary tumor ablative radiotherapy, increased survival with suppression of pulmonary metastases. CONCLUSIONS: We provide a novel immunologic basis for radiation dose fractionation consisting of a single high dose of radiotherapy followed by daily low-dose PAM-RT fractionation to improve the immunogenic potential of ablative radiotherapy.


Subject(s)
Carcinoma, Lewis Lung/radiotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Experimental/radiotherapy , Tumor Microenvironment/immunology , Ablation Techniques/methods , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/surgery , Cell Line, Tumor , Dose Fractionation, Radiation , Female , Immunomodulation/radiation effects , Lymphocytes, Tumor-Infiltrating/radiation effects , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/surgery , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Survival Rate , Tumor Microenvironment/radiation effects
15.
PLoS One ; 14(9): e0222022, 2019.
Article in English | MEDLINE | ID: mdl-31479484

ABSTRACT

In the United States, breast cancer is one of the most common and the second leading cause of cancer-related death in women. Treatment modalities for mammary tumor are surgical removal of the tumor tissue followed by either chemotherapy or radiotherapy or both. Radiation therapy is a whole body irradiation regimen that suppresses the immune system leaving hosts susceptible to infection or secondary tumors. Boron neutron capture therapy (BNCT) in that regard is more selective, the cells that are mostly affected are those that are loaded with 109 or more 10B atoms. Previously, we have described that liposomal encapsulation of boron-rich compounds such as TAC and MAC deliver a high payload to the tumor tissue when injected intravenously. Here we report that liposome-mediated boron delivery to the tumor is inversely proportional to the size of the murine mammary (EMT-6) tumors. The plausible reason for the inverse ratio of boron and EMT-6 tumor size is the necrosis in these tumors, which is more prominent in the large tumors. The large tumors also have receding blood vessels contributing further to poor boron delivery to these tumors. We next report that the presence of boron in blood is essential for the effects of BNCT on EMT-6 tumor inhibition as direct injection of boron-rich liposomes did not provide any added advantage in inhibition of EMT-6 tumor in BALB/c mice following irradiation despite having a significantly higher amount of boron in the tumor tissue. BNCT reaction in PBMCs resulted in the modification of these cells to anti-tumor phenotype. In this study, we report the immunomodulatory effects of BNCT when boron-rich compounds are delivered systemically.


Subject(s)
Boron Neutron Capture Therapy/methods , Immunomodulation/radiation effects , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/radiotherapy , Animals , Boron/administration & dosage , Boron/blood , Boron/pharmacokinetics , Cell Line, Tumor , Cytokines/metabolism , Female , Humans , Isotopes/administration & dosage , Isotopes/blood , Isotopes/pharmacokinetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/radiation effects , Liposomes , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Necrosis , Tissue Distribution
16.
Int J Biol Macromol ; 140: 505-514, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31437508

ABSTRACT

In order to investigate the impacts of ultrasound-assisted extraction (UAE) on Flammulina velutipes polysaccharides (FVPs), the differences between FVPs extracted by ultrasound-assisted extraction (FVPU) and FVPs extracted by hot water extraction (FVPH) were compared in terms of yield, primary compositions, surface microstructure, helix-coil transition structure, molecular weight distribution, antioxidant activity, and bidirectional immunomodulatory activity. Results indicated that UAE changed the above properties of FVPs. Compared with FVPH, higher yield, protein content, and uronic acid content but lower polysaccharide and polyphenol contents were observed in FVPU. UAE changed the surface microstructure, destroyed the triple helix structure, and increased the proportion of low molecular weight polysaccharide components of FVPU. Compared with FVPH, FVPU showed a stronger reducing power and scavenging activities on DPPH radical, hydroxyl radical, and superoxide anion radical. FVPU was a better inhibitor of inflammation compared with FVPH. However, FVPH had a better immunity enhancing effect compared with FVPU. These results were attributed to the cavitation effect of ultrasonic waves on the structure of polysaccharides during the extraction process of UAE. These findings suggested that UAE was an efficient and environmentally friendly method to produce new polysaccharides from F. velutipes for the development of functional foods or nutraceuticals.


Subject(s)
Antioxidants/chemistry , Flammulina/chemistry , Polysaccharides/chemistry , Animals , Antioxidants/pharmacology , Chemical Fractionation , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Hydroxyl Radical/chemistry , Immunomodulation/drug effects , Immunomodulation/radiation effects , Inflammation/drug therapy , Inflammation/immunology , Mice , Polysaccharides/isolation & purification , Polysaccharides/pharmacology , RAW 264.7 Cells , Superoxides/chemistry , Temperature , Ultrasonic Waves , Water/chemistry
17.
Radiother Oncol ; 140: 116-124, 2019 11.
Article in English | MEDLINE | ID: mdl-31271996

ABSTRACT

BACKGROUND: Traditional concepts of radiobiology model the direct radiation-induced cellular cytotoxicity but are not focused on late and sustained effects of radiation. Recent experimental data show the close involvement of immunological processes. METHODS: Based on systematic PubMed searches, experimental data on immunological radiation effects are summarized and analyzed in a non-quantitative descriptive manner to provide a translational perspective on the immuno-modulatory impact of radiation in cancer. RESULTS: Novel experimental findings document that sustained radiation effects are ultimately mediated through systemic factors such as cytotoxic CD8+ T cells and involve a local immuno-stimulation. Increased tumor infiltration of CD8+ T cell is a prerequisite for long-term radiation effects. CD8+ T cell depletion induces radio-resistance in experimental tumors. The proposed sequence of events involves radiation-damaged cells that release HMGB1, which activates macrophages via TLR4 to a local immuno-stimulation via TNF, which contributes to maturation of DCs. The mature DCs migrate to lymph nodes where they trigger effective CD8+ T cell responses. Radiation effects are boosted, when the physiological self-terminating negative feedback of immune reactions is antagonised via blocking of TGF-ß or via checkpoint inhibition with involvement of CD8+ T cells as common denominator. CONCLUSION: The concept of immuno-radiobiology emphasizes the necessity for a functional integrity of APCs and T cells for the long-term effects of radiotherapy. Local irradiation at higher doses induces tumor infiltration of CD8+ T cells, which can be boosted by immunotherapy. More systematic research is warranted to better understand the immunological effects of escalating radiation doses.


Subject(s)
CD8-Positive T-Lymphocytes/radiation effects , Immunomodulation/radiation effects , Neoplasms/radiotherapy , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Immunotherapy , Neoplasms/immunology
18.
Hematol Oncol Clin North Am ; 33(2): 233-248, 2019 04.
Article in English | MEDLINE | ID: mdl-30832997

ABSTRACT

Radiotherapy has known immunomodulatory effects and there exists a strong preclinical rationale for combining radiotherapy with immunotherapies. Broadly, the concurrent administration of immunotherapies and radiotherapy does not seem to result in undue toxicity, even when radiotherapy is administered to definitive doses. Recently reported results from prospective clinical trials evaluating radiotherapy/ICB combinations, such as the PACIFIC trial, provide important information on safety and efficacy in the definitive setting and identify potential abscopal effects. This review details the preclinical foundation for the combination of radiotherapy and immunotherapies, summarizes the most recent clinical data available, and highlights active and future areas of study.


Subject(s)
Immunomodulation/radiation effects , Neoplasms/immunology , Neoplasms/radiotherapy , Radioimmunotherapy , Clinical Trials as Topic , Humans
19.
Front Immunol ; 10: 193, 2019.
Article in English | MEDLINE | ID: mdl-30828330

ABSTRACT

In recent decades, there has been substantial growth in our understanding of the immune system and its role in tumor growth and overall survival. A central finding has been the cross-talk between tumor cells and the surrounding environment or stroma. This tumor stroma, comprised of various cells, and extracellular matrix (ECM), has been shown to aid in suppressing host immune responses against tumor cells. Through immunosuppressive cytokine secretion, metabolic alterations, and other mechanisms, the tumor stroma provides a complex network of safeguards for tumor proliferation. With recent advances in more effective, localized treatment, radiation therapy (XRT) has allowed for strategies that can effectively alter and ablate tumor stromal tissue. This includes promoting immunogenic cell death through tumor antigen release to increasing immune cell trafficking, XRT has a unique advantage against the tumoral immune evasion mechanisms that are orchestrated by stromal cells. Current studies are underway to elucidate pathways within the tumor stroma as potential targets for immunotherapy and chemoradiation. This review summarizes the effects of tumor stroma in tumor immune evasion, explains how XRT may help overcome these effects, with potential combinatorial approaches for future treatment modalities.


Subject(s)
Neoplasms/pathology , Neoplasms/radiotherapy , Stromal Cells/radiation effects , Tumor Microenvironment/radiation effects , Animals , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/radiation effects , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Extracellular Matrix/radiation effects , Humans , Immunity , Immunomodulation/radiation effects , Neoplasms/immunology , Radiation Tolerance/immunology , Radiation Tolerance/radiation effects , Radiotherapy , Stromal Cells/immunology , Tumor Microenvironment/immunology
20.
Clin Cancer Res ; 25(10): 2969-2974, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30723144

ABSTRACT

The development of cancers and their response to radiation are intricately linked to the tumor microenvironment (TME) in which they reside. Tumor cells, immune cells, and stromal cells interact with each other and are influenced by the microbiome and metabolic state of the host, and these interactions are constantly evolving. Stromal cells not only secrete extracellular matrix and participate in wound contraction, but they also secrete fibroblast growth factors (FGF), which mediate macrophage differentiation. Tumor-associated macrophages migrate to hypoxic areas and secrete vascular endothelial growth factor (VEGF) to promote angiogenesis. The microbiome and its byproducts alter the metabolic milieu by shifting the balance between glucose utilization and fatty acid oxidation, and these changes subsequently influence the immune response in the TME. Not only does radiation exert cell-autonomous effects on tumor cells, but it influences both the tumor-promoting and tumor-suppressive components in the TME. To gain a deeper understanding of how the TME influences the response to radiation, the American Society for Radiation Oncology and the American Association of Cancer Research organized a scientific workshop on July 26-27, 2018, to discuss how the microbiome, the immune response, the metabolome, and the stroma all shift the balance between radiosensitivity and radioresistance. The proceedings from this workshop are discussed here and highlight recent discoveries in the field, as well as the most important areas for future research.


Subject(s)
Neoplasms/pathology , Neoplasms/radiotherapy , Radiation Oncology , Tumor Microenvironment/radiation effects , Disease Management , Humans , Immunomodulation/radiation effects , Neoplasms/immunology , Radiation Oncology/methods , Risk Factors , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/radiation effects , Treatment Outcome , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL