Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Oxid Med Cell Longev ; 2022: 5479491, 2022.
Article in English | MEDLINE | ID: mdl-35154567

ABSTRACT

BACKGROUND: Radioimmunotherapy has become one of the most promising strategies for cancer treatment. Preclinical and clinical studies have demonstrated that antiangiogenic therapy can improve the efficacy of immunotherapy and sensitize radiotherapy through a variety of mechanisms. However, it is undefined whether angiogenesis inhibitors can enhance the effect of radioimmunotherapy. In this study, we aim to explore the role of anlotinib (AL3818) on the combination of radiotherapy and immune checkpoint inhibitors in Lewis lung carcinoma mouse. METHODS: C57BL/6 mouse subcutaneous tumor model was used to evaluate the ability of different treatment regimens in tumor growth control. Immune response and immunophenotyping including the quantification and activation were determined by flow cytometry, multiplex immunofluorescence, and multiplex immunoassay. RESULTS: Triple therapy (radiotherapy combined with anti-PD-L1 and anlotinib) increased tumor-infiltrating lymphocytes and reversed the immunosuppressive effect of radiation on the tumor microenvironment in mouse model. Compared with radioimmunotherapy, the addition of anlotinib also boosted the infiltration of CD8+ T cells and M1 cells and caused a decrease in the number of MDSCs and M2 cells in mice. The levels of IFN-gamma and IL-18 were the highest in the triple therapy group, while the levels of IL-23, IL-13, IL-1 beta, IL-2, IL-6, IL-10, and Arg-1 were significantly reduced. NF-κB, MAPK, and AKT pathways were downregulated in triple therapy compared with radioimmunotherapy. Thus, the tumor immune microenvironment was significantly improved. As a consequence, triple therapy displayed greater benefit in antitumor efficacy. CONCLUSION: Our findings indicate that anlotinib might be a potential synergistic treatment for radioimmunotherapy to achieve better antitumor efficacy in NSCLC patients by potentiating the tumor immune microenvironment.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/radiotherapy , Immune Checkpoint Inhibitors/administration & dosage , Indoles/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Quinolines/administration & dosage , Radioimmunotherapy/methods , Tumor Microenvironment/immunology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Carcinoma, Lewis Lung/immunology , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Female , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/radiation effects , Mice , Mice, Inbred C57BL , Radiotherapy Dosage , Signal Transduction/drug effects , Signal Transduction/immunology , Signal Transduction/radiation effects , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects
2.
J Pathol ; 256(2): 223-234, 2022 02.
Article in English | MEDLINE | ID: mdl-34731491

ABSTRACT

Radiation and bacillus Calmette-Guérin (BCG) instillations are used clinically for treatment of urothelial carcinoma, but the precise mechanisms by which they activate an immune response remain elusive. The role of the cGAS-STING pathway has been implicated in both BCG and radiation-induced immune response; however, comparison of STING pathway molecules and the immune landscape following treatment in urothelial carcinoma has not been performed. We therefore comprehensively analyzed the local immune response in the bladder tumor microenvironment following radiotherapy and BCG instillations in a well-established spontaneous murine model of urothelial carcinoma to provide insight into activation of STING-mediated immune response. Mice were exposed to the oral carcinogen, BBN, for 12 weeks prior to treatment with a single 15 Gy dose of radiation or three intravesical instillations of BCG (1 × 108 CFU). At sacrifice, tumors were staged by a urologic pathologist and effects of therapy on the immune microenvironment were measured using the NanoString Myeloid Innate Immunity Panel and immunohistochemistry. Clinical relevance was established by measuring immune biomarker expression of cGAS and STING on a human tissue microarray consisting of BCG-treated non-muscle-invasive urothelial carcinomas. BCG instillations in the murine model elevated STING and downstream STING-induced interferon and pro-inflammatory molecules, intratumoral M1 macrophage and T-cell accumulation, and complete tumor eradication. In contrast, radiotherapy caused no changes in STING pathway or innate immune gene expression; rather, it induced M2 macrophage accumulation and elevated FoxP3 expression characteristic of immunosuppression. In human non-muscle-invasive bladder cancer, STING protein expression was elevated at baseline in patients who responded to BCG therapy and increased further after BCG therapy. Overall, these results show that STING pathway activation plays a key role in effective BCG-induced immune response and strongly indicate that the effects of BCG on the bladder cancer immune microenvironment are more beneficial than those induced by radiation. © 2021 The Pathological Society of Great Britain and Ireland.


Subject(s)
Antineoplastic Agents/administration & dosage , BCG Vaccine/administration & dosage , Immunity, Innate/drug effects , Immunity, Innate/radiation effects , Immunotherapy , Membrane Proteins/immunology , Radiation Dosage , Urinary Bladder Neoplasms/therapy , Urothelium/drug effects , Urothelium/radiation effects , Administration, Intravesical , Animals , Female , Humans , Inflammation Mediators/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/radiation effects , Membrane Proteins/metabolism , Mice, Inbred C57BL , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/radiation effects , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Urothelium/immunology , Urothelium/metabolism
3.
Cancer Lett ; 522: 238-254, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34571084

ABSTRACT

The response rate of anti-PD therapy in most cancer patients remains low. Therapeutic drug and tumor-infiltrating lymphocytes (TILs) are usually obstructed by the stromal region within tumor microenvironment (TME) rather than distributed around tumor cells, thus unable to induce the immune response of cytotoxic T cells. Here, we constructed the cationic thermosensitive lipid nanoparticles IR780/DPPC/BMS by introducing cationic NIR photosensitizer IR-780 iodide (IR780) modified lipid components, thermosensitive lipid DPPC and PD-1/PD-L1 inhibitor BMS202 (BMS). Upon laser irradiation, IR780/DPPC/BMS penetrated into deep tumor, and reduced cancer-associated fibroblasts (CAFs) around tumor cells to remodel the spatial distribution of TILs in TME. Interestingly, the cationic IR780/DPPC/BMS could capture released tumor-associated antigens (TAAs), thereby enhancing the antigen-presenting ability of DCs to activate cytotoxic T lymphocytes. Moreover, IR780/DPPC/BMS initiated gel-liquid crystal phase transition under laser irradiation, accelerating the disintegration of lipid bilayer structure and leading to the responsive release of BMS, which would reverse the tumor immunosuppression state by blocking PD-1/PD-L1 pathway for a long term. This combination treatment can synergistically exert the antitumor immune response and inhibit the tumor growth and metastasis.


Subject(s)
B7-H1 Antigen/immunology , Liposomes/pharmacology , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/immunology , Acetamides/chemistry , Acetamides/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Immune Tolerance/drug effects , Immunotherapy/methods , Indoles/chemistry , Indoles/pharmacology , Liposomes/chemistry , Low-Level Light Therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/radiation effects , Nanoparticles/chemistry , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/radiotherapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Pyridines/chemistry , Pyridines/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/radiation effects , Tumor Microenvironment/drug effects
4.
Int J Radiat Oncol Biol Phys ; 111(1): 284-296, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33933481

ABSTRACT

PURPOSE: Dexamethasone is commonly given during radiation therapy (RT) to manage toxicities. Our study examines if dexamethasone coadministration with RT inhibits the RT-induced antitumor T cell response in mouse. METHODS AND MATERIALS: Intramuscularly implanted MC38 tumors were irradiated with 15 Gy after establishing for 7 days. Tumor bearing mice were administered dexamethasone using multiple schedules and doses. Peripheral lymphocyte reduction was monitored by complete blood count and intratumoral and tumor draining lymph node (tdLN) populations by flow cytometry. Effector phenotype and function of ex vivo stimulated tumor-infiltrating lymphocytes (TILs) and naïve splenocytes as well as in vivo TILs with or without dexamethasone were monitored by flow cytometry and ELISA. RESULTS: Long course high dose, short course high dose, and short course human equivalent dose dexamethasone reduced peripheral lymphocytes yet did not inhibit survival after irradiation. Short course high dose administration decreased TIL and tdLN lymphocyte activation as well as tdLN mass but did not affect TIL frequencies or change tdLN cell population composition. Dexamethasone inhibited effector function of ex vivo stimulated naïve splenocytes and TILs, but magnitude of IFN-γ secretion was consistently higher in TILs regardless of dexamethasone dose. In vivo analysis of TILs after irradiation and HE dexamethasone treatment showed that TILs had a similar effector phenotype compared with vehicle controls. CONCLUSIONS: Dexamethasone reduces blood and tdLN lymphocytes. Dexamethasone also suppresses TIL activation/effector function yet does not affect survival in irradiated MC38 tumor bearing mice, which depend on RT-induced immune responses for therapy efficacy. Additional study in human subjects is warranted.


Subject(s)
Colorectal Neoplasms/radiotherapy , Dexamethasone/pharmacology , Lymphocytes, Tumor-Infiltrating/radiation effects , Animals , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Dexamethasone/administration & dosage , Interferon-gamma/biosynthesis , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Phenotype
5.
Future Oncol ; 17(13): 1683-1694, 2021 May.
Article in English | MEDLINE | ID: mdl-33726502

ABSTRACT

Radiation therapy (RT) in some cases results in a systemic anticancer response known as the abscopal effect. Multiple hypotheses support the role of immune activation initiated by RT-induced DNA damage. Optimal radiation dose is necessary to promote the cGAS-STING pathway in response to radiation and initiate an IFN-1 signaling cascade that promotes the maturation and migration of dendritic cells to facilitate antigen presentation and stimulation of cytotoxic T cells. T cells then exert a targeted response throughout the body at areas not subjected to RT. These effects are further augmented through the use of immunotherapeutic drugs resulting in increased T-cell activity. Tumor-infiltrating lymphocyte presence and TREX1, KPNA2 and p53 signal expression are being explored as prognostic biomarkers.


Subject(s)
Chemoradiotherapy/methods , Dendritic Cells/immunology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/radiotherapy , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cell Movement/radiation effects , Clinical Trials as Topic , DNA Damage/immunology , DNA Damage/radiation effects , Dendritic Cells/radiation effects , Exodeoxyribonucleases/analysis , Exodeoxyribonucleases/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/radiation effects , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/mortality , Phosphoproteins/analysis , Phosphoproteins/metabolism , Prognosis , Progression-Free Survival , Radiotherapy Dosage , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/metabolism , alpha Karyopherins/analysis , alpha Karyopherins/metabolism
6.
Int J Radiat Oncol Biol Phys ; 110(5): 1341-1349, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33647370

ABSTRACT

Outcomes for triple negative breast cancer (TNBC) are poor and may be improved by increasing CD8+ tumor infiltrating lymphocytes (TIL) to augment antitumor immunity. Radiation (RT) can promote immunogenic cell death with increased antitumor T cell activity but also stimulates suppressive regulatory T cells (Tregs). Because metabolic alterations affect immune homeostasis and prior studies show caloric restriction (CR) combined with RT improves preclinical TNBC outcomes, we hypothesized that CR augments RT, in part, by altering intratumoral immunity. Using an in vivo model of TNBC, we treated mice with ad libitum (AL) diet, radiation, a CR diet, or CR + RT, and demonstrated an immune suppressive environment with a significant increase in CD4+ CD25+Foxp3+ Tregs after RT but not in CR-fed mice. CD8:Treg ratio in CR + RT TIL increased 4-fold compared with AL + RT mice. In vivo CD8 depletion was performed to assess the role of effector T cells in mitigating the effects of CR, and it was found that in mice undergoing CR, depletion of CD8 T cells resulted in increased tumor progression and decreased median survival compared with isotype control-treated mice. In addition, PD-1 expression on CD3+CD8+ T cells within the tumor microenvironment was significantly increased in CR + RT versus AL + RT treated mice as per immunofluorescence. Serum from breast cancer patients undergoing RT alone or CR and RT was collected pre- and postintervention, and a cytokine array demonstrated that patients treated with CR + RT had notable decreases in immunosuppressive cytokines such as IL-2Rγ, IL-10Rß, and TGF-ß2 and 3 compared with patients receiving RT alone. In conclusion, combining CR with RT decreases intratumoral Tregs, increases CD8:Treg, and increases PD-1 expression via a process dependent on CD8 T cells in a TNBC model. Breast cancer patients undergoing CR concurrently with RT also had significant reduction in immunosuppressive cytokine levels compared with those receiving RT alone.


Subject(s)
Caloric Restriction , Lymphocytes, Tumor-Infiltrating/radiation effects , T-Lymphocytes, Regulatory/radiation effects , Triple Negative Breast Neoplasms/radiotherapy , Tumor Microenvironment/radiation effects , Adult , Aged , Animals , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/radiation effects , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/radiation effects , Combined Modality Therapy/methods , Disease Progression , Female , Flow Cytometry , Forkhead Transcription Factors , Humans , Interleukin Receptor Common gamma Subunit/blood , Interleukin-10 Receptor beta Subunit/blood , Interleukin-2 Receptor alpha Subunit , Lymphocyte Depletion/methods , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Middle Aged , Programmed Cell Death 1 Receptor/metabolism , Random Allocation , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta2/blood , Transforming Growth Factor beta3/blood , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/mortality , Tumor Microenvironment/immunology
7.
Oncol Rep ; 45(3): 1273-1283, 2021 03.
Article in English | MEDLINE | ID: mdl-33432367

ABSTRACT

The immune landscape of head and neck squamous cell carcinoma in pretreated areas remains poorly documented. We aimed to assess the tumor microenvironment for biomarkers of antitumor immune responses in tumors in previously irradiated areas compared with de novo tumors. This retrospective monocentric study analyzed 100 paraffin­embedded surgical samples of invasive head and neck squamous cell carcinoma (oral cavity, oropharynx, larynx, hypopharynx) from patients who underwent surgery between January 2010 and November 2017. We compared the immune microenvironment in 50 de novo tumors and 50 tumors recurring within irradiated areas. We used immunohistochemistry to assess p16 status, CD3+/CD8+ tumor­infiltrating lymphocytes (TILs), and programmed death­ligand 1 (PD­L1) expression on tumor and immune cells in stromal and intratumoral components. CD3+ TIL counts were significantly lower in intratumoral and stromal components (P=0.003 and P=0.020, respectively) in the irradiated area cohort; there was no significant difference between CD8+ TIL counts in the two cohorts. The percentage of tumors with PD­L1+ tumor cells (tumor proportion score ≥1%) was significantly lower within the irradiated area cohort than the de novo cohort (56.0% vs. 86.0%, P<0.001). There were also significantly fewer tumors with PD­L1+ immune cells in the irradiated area cohort. Predominantly, tumors from the irradiated area cohort had microenvironments classified as 'adaptive immune resistance'. There was persistence of cytotoxic cells in tumors in the irradiated areas but lower PD­L1 expression and CD3+ TIL counts than in the de novo tumors. This offers an initial hypothesis to explain why these lesions are less responsive to immunotherapy, even though they may still have antitumor capacities. Assessment of immune response biomarkers in patients treated with immunotherapy in randomized trials is required.


Subject(s)
Biomarkers, Tumor/metabolism , Head and Neck Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Recurrence, Local/therapy , Squamous Cell Carcinoma of Head and Neck/therapy , Aged , B7-H1 Antigen/analysis , B7-H1 Antigen/metabolism , Biomarkers, Tumor/analysis , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Lymphocytes, Tumor-Infiltrating/radiation effects , Male , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Radiotherapy, Adjuvant , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
8.
J Surg Oncol ; 123(1): 117-126, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33084061

ABSTRACT

BACKGROUND AND OBJECTIVES: Limited literature exists examining the immune microenvironment in liposarcoma, particularly with regard to the impact of radiotherapy. A major problem is the lack of scoring system for the tumour-infiltrating lymphocytes (TILs) in sarcoma. This study aims to describe the immune environment pre- and postradiotherapy and identify the optimal immune infiltrate scoring system for sarcoma. METHODS: Thirty-nine paired tissue samples (pre- and postradiotherapy) from patients with liposarcoma were scored by two pathologists for TILs using pre-existing systems (for breast cancer and melanoma) and compared for interobserver reliability. Immunohistochemical staining was performed for various immune markers. RESULTS: The TIL scoring system for breast cancer yielded perfect agreement (κ = 1.000). 21% of patients had increased TILs after radiotherapy, 87.5% of whom had dedifferentiated liposarcoma. Immune suppressor expression was increased frequently after radiotherapy (CD68 increased in 59.4%, PD-L1 increased in 25%). Immune effector expression (CD8) was unchanged in 84.4%. CONCLUSIONS: Breast cancer TIL scoring is reproducible in liposarcoma and has high interobserver reliability. Radiotherapy was observed to have a limited impact on immune effectors but seemed to have more impact in upregulating immune suppressors, suggesting radiotherapy may contribute to disease control through immunomodulatory effects. Dedifferentiated liposarcoma represents a uniquely responsive subtype.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Liposarcoma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Recurrence, Local/immunology , Radiotherapy/methods , Tumor Microenvironment/immunology , Adolescent , Adult , Aged , CD8-Positive T-Lymphocytes/radiation effects , Female , Follow-Up Studies , Humans , Liposarcoma/pathology , Liposarcoma/radiotherapy , Lymphocytes, Tumor-Infiltrating/radiation effects , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Prognosis , Retrospective Studies , Tumor Microenvironment/radiation effects , Young Adult
9.
Int J Radiat Oncol Biol Phys ; 110(2): 510-520, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33383126

ABSTRACT

PURPOSE: The establishment of a preclinical model of the abscopal effect on hepatocellular carcinoma (HCC) and evaluation of whether the hypofractionated radiation therapy (RT) multitumor Hepa1-6 mouse HCC model could be used to suppress nonradiated tumor mass was performed in this study. METHODS AND MATERIALS: Hepa1-6 mouse liver cancer cell lines were used to form tumors. Immunogenicity was analyzed using ELISpot and immune cell labeled antibody. Interferon (IFN) ß expression was confirmed through polymerase chain reaction. RESULTS: After investigation, the intratumoral transcription of type Ⅰ IFN increased by 2-fold. The antitumor immune response to Hepa 1-6 cells induced by radiation was increased. Moreover, the influx of activated CD8+ T cells was increased in nonirradiated tumors. The number of dendritic cells and activation status were evaluated by flow cytometry on the second day after irradiation. Flow cytometry revealed a significantly increased dendritic cell population expressing the CD11c molecule in tumor-draining lymph nodes. Furthermore, because irradiation leads to adaptation of immune resistance of tumor cells against RT, we sought to elucidate a potent tool to overcome the resistance and confirm the ability of PD-L1 antibody to survive late RT resistance. CONCLUSIONS: The immunologic mechanism of the abscopal effect was revealed and the application of PD-L1 inhibitor successfully performed as a breakthrough in late RT resistance in the Hepa1-6 tumor model.


Subject(s)
Carcinoma, Hepatocellular/therapy , Immune Checkpoint Inhibitors/therapeutic use , Liver Neoplasms/therapy , Radiation Tolerance/immunology , Radiosurgery/methods , Animals , Antineoplastic Agents , B7-H1 Antigen/administration & dosage , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , CD11 Antigens/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Combined Modality Therapy/methods , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/radiation effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Interferon Type I/metabolism , Interferon-beta/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Lymph Nodes/metabolism , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/radiation effects , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Radiation Dose Hypofractionation , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/radiation effects
10.
Int J Mol Sci ; 21(20)2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33096928

ABSTRACT

Most gliomas are associated with a fatal prognosis and remain incurable because of their infiltrative growth. Consequently, the addition of immunotherapy to conventional therapy may improve patient outcomes. Here, we analyzed T-cell infiltration and, therefore, a major prerequisite for successful immunotherapy in a series of primary (n = 78) and recurrent (n = 66) isocitrate dehydrogenase (IDH)-mutant glioma and their changes following treatment with radio- and/or chemotherapy. After multicolor immunofluorescence staining, T cells were counted in entire tumor sections using a software-based setup. Newly diagnosed diffuse IDH-mutant gliomas displayed a median T-cell infiltration of 0.99 T cells/mm2 (range: 0-48.97 CD3+ T cells/mm2), which was about two-fold increased for CD3+, helper, and cytotoxic T cells in recurrent glioma. Furthermore, T-cell infiltration of recurrent tumors was associated with the type of adjuvant treatment of the primary tumor. Interestingly, only glioma patients solely receiving radiotherapy presented consistently with increased T-cell infiltration in their recurrent tumors. This was confirmed in a subset of 27 matched pairs. In conclusion, differences in the T-cell infiltration of primary and recurrent gliomas were demonstrated, and evidence was provided for a beneficial long-term effect on T-cell infiltration upon treatment with radiotherapy.


Subject(s)
Brain Neoplasms/radiotherapy , Glioma/radiotherapy , Lymphocytes, Tumor-Infiltrating/radiation effects , Adult , Aged , Aged, 80 and over , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Female , Glioma/genetics , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Lymphocytes, Tumor-Infiltrating/pathology , Male , Matched-Pair Analysis , Middle Aged , Mutation , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/radiation effects , Tumor Microenvironment/radiation effects , Young Adult
11.
Eur J Cancer ; 140: 28-36, 2020 11.
Article in English | MEDLINE | ID: mdl-33039811

ABSTRACT

BACKGROUND: A history of radiotherapy and chemoradiotherapy (CRT) reportedly increases the efficacy of the PD-1 blockade in patients with advanced non-small cell lung cancer (NSCLC). We investigated the efficacy of anti-PD-(L)1 therapy after CRT failure and how CRT changes the status of PD-L1 expression on tumours and on tumour-infiltrated lymphocytes (TILs). METHODS: We retrospectively reviewed patients with unresectable locally advanced NSCLC (LA-NSCLC) who were treated with CRT between 2007 and 2018 and evaluated the efficacy of the PD-(L)1 blockade after CRT failure. We also compared the PD-L1 (clone: 22C3) expression levels and the tumoral and stromal distributions of CD8-positive TILs using paired formalin-fixed, paraffin-embedded specimens obtained before and after CRT. RESULTS: We identified 422 patients and 65 patients who had relapsed after CRT received anti-PD-(L)1 therapy. The objective response rate (ORR) and the progression-free survival (PFS) after anti-PD-(L)1 therapy were 48% and 8.7 months (95% CI, 4.5-13), respectively. The RR and PFS did not differ according to the pre-CRT PD-L1 expression levels. PD-L1 expression changed in 16 of the 18 patients between before and after CRT, but a specific trend was not seen (increased, 9 patients; decreased, 7 patients; no change, 2 patients). In contrast, the density of tumoral CD8-positive TILs increased after CRT treatment (pre-CRT median, 110/mm2 versus post-CRT median, 470/mm2; p = 0.025). CONCLUSIONS: Anti-PD-(L)1 therapy was effective in patients with LA-NSCLC who had progressed after CRT regardless of their pre-CRT PD-L1 expression. The efficacy of anti-PD-(L)1 therapy for patients with NSCLC with CRT failure was superior to that of standard second-line treatment for patients with advanced NSCLC.


Subject(s)
Antineoplastic Agents/therapeutic use , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Neoplasm Recurrence, Local/therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/radiation effects , Carcinoma, Non-Small-Cell Lung/metabolism , Chemoradiotherapy/methods , Female , Humans , Lung Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/radiation effects , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Progression-Free Survival , Retrospective Studies
12.
J Hematol Oncol ; 13(1): 105, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32723363

ABSTRACT

Immune checkpoint inhibitors targeting programmed cell death 1 (PD-1), programmed cell death ligand-1 (PD-L1), and others have shown potent clinical efficacy and have revolutionized the treatment protocols of a broad spectrum of tumor types, especially non-small-cell lung cancer (NSCLC). Despite the substantial optimism of treatment with PD-1/PD-L1 inhibitors, there is still a large proportion of patients with advanced NSCLC who are resistant to the inhibitors. Preclinical and clinical trials have demonstrated that radiotherapy can induce a systemic antitumor immune response and have a great potential to sensitize refractory "cold" tumors to immunotherapy. Stereotactic body radiation therapy (SBRT), as a novel radiotherapy modality that delivers higher doses to smaller target lesions, has shown favorable antitumor effects with significantly improved local and distant control as well as better survival benefits in various solid tumors. Notably, research has revealed that SBRT is superior to conventional radiotherapy, possibly because of its more powerful immune activation effects. Thus, PD-1/PD-L1 inhibitors combined with SBRT instead of conventional radiotherapy might be more promising to fight against NSCLC, further achieving more favorable survival outcomes. In this review, we focus on the underlying mechanisms and recent advances of SBRT combined with PD-1/PD-L1 inhibitors with an emphasis on some future challenges and directions that warrant further investigation.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/radiotherapy , Immune Checkpoint Inhibitors/therapeutic use , Immunity/radiation effects , Lung Neoplasms/radiotherapy , Radiosurgery , Antigens, Neoplasm/immunology , Antineoplastic Agents/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/physiology , Biomarkers , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Chemotaxis, Leukocyte/radiation effects , Clinical Trials as Topic , Combined Modality Therapy , Drug Resistance, Neoplasm , Humans , Immune Checkpoint Inhibitors/adverse effects , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/radiation effects , Multicenter Studies as Topic , Neoplasm Proteins/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/physiology , Progression-Free Survival , Radiation Dose Hypofractionation , Radiosurgery/adverse effects , Radiosurgery/methods , Radiotherapy Planning, Computer-Assisted , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/radiation effects , Treatment Outcome , Tumor Escape
13.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32581061

ABSTRACT

BACKGROUND: Prostate cancer (PCa) has a profoundly immunosuppressive microenvironment and is commonly immune excluded with few infiltrative lymphocytes and low levels of immune activation. High-dose radiation has been demonstrated to stimulate the immune system in various human solid tumors. We hypothesized that localized radiation therapy, in the form of high dose-rate brachytherapy (HDRBT), would overcome immune suppression in PCa. METHODS: To investigate whether HDRBT altered prostate immune context, we analyzed preradiation versus postradiation human tissue from a cohort of 24 patients with localized PCa that received HDRBT as primary treatment (RadBank cohort). We performed Nanostring immune gene expression profiling, digital spatial profiling, and high-throughput immune cell multiplex immunohistochemistry analysis. We also resolved tumor and nontumor zones in spatial and bioinformatic analyses to explore the immunological response. RESULTS: Nanostring immune profiling revealed numerous immune checkpoint molecules (eg, B7-H3, CTLA4, PDL1, and PDL2) and TGFß levels were increased in response to HDRBT. We used a published 16-gene tumor inflammation signature (TIS) to divide tumors into distinct immune activation states (high:hot, intermediate and low:cold) and showed that most localized PCa are cold tumors pre-HDRBT. Crucially, HDRBT converted 80% of these 'cold'-phenotype tumors into an 'intermediate' or 'hot' class. We used digital spatial profiling to show these HDRBT-induced changes in prostate TIS scores were derived from the nontumor regions. Furthermore, these changes in TIS were also associated with pervasive changes in immune cell density and spatial relationships-in particular, between T cell subsets and antigen presenting cells. We identified an increased density of CD4+ FOXP3+ T cells, CD68+ macrophages and CD68+ CD11c+ dendritic cells in response to HDRBT. The only subset change specific to tumor zones was PDL1- macrophages. While these immune responses were heterogeneous, HDRBT induced significant changes in immune cell associations, including a gained T cell and HMWCK+ PDL1+ interaction in tumor zones. CONCLUSION: In conclusion, we showed HDRBT converted "cold" prostate tumors into more immunologically activated "hot" tissues, with accompanying spatially organized immune infiltrates and signaling changes. Understanding and potentially harnessing these changes will have widespread implications for the future treatment of localized PCa, including rational use of combination radio-immunotherapy.


Subject(s)
Biomarkers/analysis , Brachytherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/radiotherapy , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Aged , Humans , Lymphocytes, Tumor-Infiltrating/radiation effects , Male , Middle Aged , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , T-Lymphocytes/radiation effects , Tumor Microenvironment/radiation effects
14.
PLoS One ; 15(4): e0231507, 2020.
Article in English | MEDLINE | ID: mdl-32287292

ABSTRACT

INTRODUCTION: Irradiation can induce multiple inhibitory and stimulatory effects on the immune system. In recent studies, it has been noted that administration of radiation with various doses and fractionation plans may influence on immune responses in microenvironment of tumor. But in radiobiology, the Biologically Effective Dose (BED) formula has been designed for calculating isoeffect doses in different regimens of daily clinical practice. In other words, BED has also been used to predict the effects of fractionation schedules on tumor cells. METHODS: In our study, three different regimens with BEDs of 40 gray (Gy) were analyzed in BALB/c mice. These included conventional fractionated radiotherapy (RT) (3Gyx10), high-dose hypofractionated RT (10Gyx2), and single ablative high-dose RT (15Gyx1). RESULTS: As BED predicts, all three similarly decreased tumor volumes and increased survival times relative to controls, but after high dose exposure in ablative group, the expression of IFNγ was increased following high infiltration of CD8 cells into the tumor microenvironment. When anti-PDL-1 was combined with RT, single ablative high-dose radiation enhanced antitumor activity by increasing IFNγ in tumors and CD8+ tumor-infiltrating lymphocytes; as a result, this combining therapy had enhanced antitumor activity and lead to control tumor volume effectively and improve significantly survival rate and finally the recurrence of tumor was not observed. CONCLUSION: Results show distinct radiation doses and fractionation schemes with same BED have different immunogenic response and these findings can provide data helping to design regimens of radiation combined with immune checkpoint blockers (ICBs).


Subject(s)
Lymphocytes, Tumor-Infiltrating/radiation effects , Neoplasms/radiotherapy , Radiobiology/methods , Animals , B7-H1 Antigen/radiation effects , CD8-Positive T-Lymphocytes/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Dose Fractionation, Radiation , Female , Interferon-gamma/radiation effects , Interferon-gamma/therapeutic use , Mice , Mice, Inbred BALB C , Radiation Dose Hypofractionation/standards , Relative Biological Effectiveness , Tumor Burden , Tumor Microenvironment/radiation effects
15.
Int J Radiat Oncol Biol Phys ; 108(1): 286-294, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32004580

ABSTRACT

PURPOSE: Despite good preclinical evidence, clinical data on the effect of neoadjuvant chemoradiation therapy (CRT) on expression of immune markers in esophageal cancer are limited. This study aimed to evaluate the changes in indoleamine 2,3-dioxygenase 1 (IDO1) expression, programmed cell death-ligand 1 (PD-L1) expression, and CD8+ tumor-infiltrating lymphocyte status after neoadjuvant CRT and the prognostic significance in esophageal squamous cell carcinoma (ESCC). METHODS AND MATERIALS: Between 2003 and 2017, 138 patients with ESCC who underwent neoadjuvant CRT and esophagectomy without achieving pathologic complete response were included for analysis. Both pre-CRT biopsies and post-CRT surgical specimens were available in 82 patients. Immunohistochemistry of IDO1, PD-L1, and CD8 density were analyzed. RESULTS: Among 82 paired samples, the expression levels of IDO1 and PD-L1 and CD8 density increased significantly after neoadjuvant CRT (P < .01 for all). Patients with high IDO1 expression after CRT had poorer overall survival (P = .001) and recurrence-free survival (P < .001) than those with low IDO1 expression. High post-CRT CD8 density was significantly correlated with more favorable overall survival (P = .01) and recurrence-free survival (P = .008). Neither pre- nor post-CRT PD-L1 expression was an independent prognostic factor for survival. Stratification analysis revealed that patients with combined low IDO1 expression and high CD8 density after CRT were significantly associated with better survival than other subgroups. The major findings were reproducible in an independent validation cohort. CONCLUSIONS: IDO1 and PD-L1 expression and CD8 density increased significantly after neoadjuvant CRT in ESCC. The post-CRT IDO1 expression and CD8 density could serve as prognostic biomarkers for survival.


Subject(s)
Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/therapy , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Neoadjuvant Therapy , Adult , CD8 Antigens/metabolism , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/immunology , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/diagnosis , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/metabolism , Female , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/radiation effects , Male , Middle Aged , Prognosis
16.
Strahlenther Onkol ; 196(8): 725-735, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31953603

ABSTRACT

PURPOSE: To evaluate radiotherapy-induced changes in the expression of programmed death ligand 1 (PD-L1), programmed death 1 (PD-1), and human leukocyte antigen class I (HLA-1) in patients with uterine cervical cancer, as well as infiltration of CD8+ and Forkhead box P3+ (FoxP3+) T lymphocytes into tumor tissue and the prognostic value of these parameters. MATERIALS AND METHODS: We performed immunohistochemical analysis of pre-radiotherapy biopsies and corresponding post-radiotherapy resected tissues in 104 uterine cervical cancer patients undergoing preoperative chemoradiotherapy or radiotherapy alone. We scored the expression of various proteins to distinguish positive from negative samples. RESULTS: PD-L1-expressing tumor cells (PD-L1 TC) increased significantly after chemoradiotherapy (p = 0.043). CD8+ T cell infiltration (p = 0.002) and FoxP3+ T cell infiltration (p = 0.003) decreased significantly after chemoradiotherapy. Expression of PD­1, PD-L1-expressing immune cells (PD-L1 IC), and HLA­1 did not change after chemoradiotherapy. In biopsy specimens obtained before chemoradiotherapy or radiotherapy, greater infiltration of CD8+ T cells (p = 0.001) and FoxP3+ T cells (p = 0.003) were significant predictors of better overall survival (OS). In surgical specimens obtained after chemoradiotherapy or radiotherapy, greater infiltration of PD-L1 TC was the only significant predictor of better OS (p < 0.001) and was related to a significantly lower probability of out-of-field recurrence (p = 0.005). CONCLUSION: Chemoradiotherapy induced an immunologic shift that increased PD-L1 TC. Chemoradiotherapy has immunological effects that can influence the results of treatment for uterine cervical cancer.


Subject(s)
B7-H1 Antigen/analysis , Carcinoma, Squamous Cell/radiotherapy , Gene Expression Regulation, Neoplastic/radiation effects , HLA Antigens/analysis , Neoadjuvant Therapy , Neoplasm Proteins/analysis , T-Lymphocyte Subsets/immunology , Uterine Neoplasms/radiotherapy , Adult , Aged , Antineoplastic Agents, Alkylating/therapeutic use , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/surgery , Carcinoma, Squamous Cell/therapy , Chemoradiotherapy, Adjuvant , Cisplatin/therapeutic use , Female , Forkhead Transcription Factors/analysis , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/radiation effects , Middle Aged , Prognosis , Proportional Hazards Models , ROC Curve , Retrospective Studies , T-Lymphocyte Subsets/chemistry , Treatment Outcome , Uterine Neoplasms/immunology , Uterine Neoplasms/surgery , Uterine Neoplasms/therapy
17.
Strahlenther Onkol ; 196(5): 465-473, 2020 May.
Article in English | MEDLINE | ID: mdl-31828392

ABSTRACT

PURPOSE: Considering the effects of P53 binding protein 1 (53BP1) expression and T lymphocyte infiltration density on tumor radiosensitivity, we investigated the relation of 53BP1 expression and immunoscore based on T lymphocyte infiltration density with the efficacy of neoadjuvant chemoradiotherapy (CRT) for rectal cancer. METHODS: Fifty-five patients with rectal cancer receiving neoadjuvant CRT followed by surgery were enrolled. The 53BP1 expression level and the density of CD3+, CD8+, and CD45RO+ T lymphocytes in the tumor tissues were examined by immunohistochemistry, and the relation of these findings to the rates of tumor regression, disease-free survival (DFS), and overall survival (OS) was analyzed. RESULTS: The levels of 53BP1 and the CD3/CD8 immunoscore were closely correlated with the response to CRT (p < 0.05), with an area under the receiver operating characteristic curve for CRT efficacy prediction of 0.626 and 0.717, respectively. Further survival analysis revealed that high 53BP1 expression effectively prolonged 2­year DFS compared with low 53BP1 expression (87.5% [95%CI 77.3-97.7] vs. 53.3% [95%CI 28.1-78.6]; p < 0.05), while the effect of immunoscore on survival was restricted by the expression status of 53BP1. Cox multivariate analysis confirmed 53BP1 as an independent prognostic factor in DFS. CONCLUSION: The pretreatment levels of 53BP1 and the immunoscore based on CD3+/CD8+ T cell infiltration density in tumor tissues are effective predictors for the CRT response, and 53BP1 has a more pronounced impact on prognosis.


Subject(s)
Chemoradiotherapy, Adjuvant , Gene Expression Regulation, Neoplastic/genetics , Neoadjuvant Therapy , Rectal Neoplasms/genetics , Rectal Neoplasms/therapy , Tumor Suppressor p53-Binding Protein 1/genetics , Adult , Aged , Combined Modality Therapy , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Immunohistochemistry , Leukocyte Count , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/pathology , Lymphocytes, Tumor-Infiltrating/radiation effects , Male , Middle Aged , Neoplasm Staging , Proctectomy , Radiation Tolerance , Rectal Neoplasms/mortality , Rectal Neoplasms/pathology , Retrospective Studies , Survival Rate , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , T-Lymphocytes/radiation effects , Treatment Outcome , Tumor Suppressor p53-Binding Protein 1/analysis , Young Adult
18.
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
19.
Cancer Immunol Res ; 8(2): 217-229, 2020 02.
Article in English | MEDLINE | ID: mdl-31831632

ABSTRACT

Because the incidence of breast cancer increases decades after ionizing radiation exposure, aging has been implicated in the evolution of the tumor microenvironment and tumor progression. Here, we investigated radiation-induced carcinogenesis using a model in which the mammary glands of 10-month-old BALB/c mice were transplanted with Trp53-null mammary tissue 3 days after exposure to low doses of sparsely ionizing γ-radiation or densely ionizing particle radiation. Mammary transplants in aged, irradiated hosts gave rise to significantly more tumors that grew more rapidly than those in sham-irradiated mice, with the most pronounced effects seen in mice irradiated with densely ionizing particle radiation. Tumor transcriptomes identified a characteristic immune signature of these aggressive cancers. Consistent with this, fast-growing tumors exhibited an immunosuppressive tumor microenvironment with few infiltrating lymphocytes, abundant immunosuppressive myeloid cells, and high COX-2 and TGFß. Only irradiated hosts gave rise to tumors lacking cytotoxic CD8+ lymphocytes (defined here as immune desert), which also occurred in younger irradiated hosts. These data suggest that host irradiation may promote immunosuppression. To test this, young chimera mice were fed chow containing a honeybee-derived compound with anti-inflammatory and immunomodulatory properties, caffeic acid phenethyl ester (CAPE). CAPE prevented the detrimental effects of host irradiation on tumor growth rate, immune signature, and immunosuppression. These data indicated that low-dose radiation, particularly densely ionizing exposure of aged mice, promoted more aggressive cancers by suppressing antitumor immunity. Dietary intervention with a nontoxic immunomodulatory agent could prevent systemic effects of radiation that fuel carcinogenesis, supporting the potential of this strategy for cancer prevention.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diet , Inflammation/diet therapy , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Experimental/prevention & control , Neoplasms, Radiation-Induced/prevention & control , Age Factors , Animals , CD8-Positive T-Lymphocytes/radiation effects , Dose-Response Relationship, Radiation , Female , Inflammation/etiology , Inflammation/pathology , Lymphocytes, Tumor-Infiltrating/radiation effects , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/immunology , Transcriptome , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
20.
Cancer Immunol Immunother ; 68(11): 1805-1817, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31628526

ABSTRACT

Antibodies targeting the T-cell immune checkpoint cytotoxic T-lymphocyte antigen-4 (CTLA4) enhance the effectiveness of radiotherapy for melanoma patients, but many remain resistant. To further improve response rates, we explored combining anti-CTLA4 blockade with antisense suppression of CD47, an inhibitory receptor on T cells that limit T-cell receptor signaling and killing of irradiated target cells. Human melanoma data from The Cancer Genome Atlas revealed positive correlations between CD47 mRNA expression and expression of T-cell regulators including CTLA4 and its counter receptors CD80 and CD86. Antisense suppression of CD47 on human T cells in vitro using a translational blocking morpholino (CD47 m) alone or combined with anti-CTLA4 enhanced antigen-dependent killing of irradiated melanoma cells. Correspondingly, the treatment of locally irradiated B16F10 melanomas in C57BL/6 mice using combined blockade of CD47 and CTLA4 significantly increased the survival of mice relative to either treatment alone. CD47 m alone or in combination with anti-CTLA4 increased CD3+ T-cell infiltration in irradiated tumors. Anti-CTLA4 also increased CD3+ and CD8+ T-cell infiltration as well as markers of NK cells in non-irradiated tumors. Anti-CTLA4 combined with CD47 m resulted in the greatest increase in intratumoral granzyme B, interferon-γ, and NK-cell marker mRNA expression. These data suggest that combining CTLA4 and CD47 blockade could provide a survival benefit by enhancing adaptive T- and NK-cell immunity in irradiated tumors.


Subject(s)
CD47 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Ipilimumab/administration & dosage , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/mortality , T-Lymphocytes, Cytotoxic/immunology , Animals , CD47 Antigen/genetics , CD47 Antigen/immunology , Combined Modality Therapy , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/radiation effects , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Radiation Dosage , Survival Rate , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/radiation effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL