Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Lancet Oncol ; 23(9): 1189-1200, 2022 09.
Article in English | MEDLINE | ID: mdl-35952709

ABSTRACT

BACKGROUND: TGF-ß is an immunosuppressive cytokine that is upregulated in colorectal cancer. TGF-ß blockade improved response to chemoradiotherapy in preclinical models of colorectal adenocarcinoma. We aimed to test the hypothesis that adding the TGF-ß type I receptor kinase inhibitor galunisertib to neoadjuvant chemoradiotherapy would improve pathological complete response rates in patients with locally advanced rectal cancer. METHODS: This was an investigator-initiated, single-arm, phase 2 study done in two medical centres in Portland (OR, USA). Eligible patients had previously untreated, locally advanced, rectal adenocarcinoma, stage IIA-IIIC or IV as per the American Joint Committee on Cancer; Eastern Cooperative Oncology Group status 0-2; and were aged 18 years or older. Participants completed two 14-day courses of oral galunisertib 150 mg twice daily, before and during fluorouracil-based chemoradiotherapy (intravenous fluorouracil 225 mg/m2 over 24 h daily 7 days per week during radiotherapy or oral capecitabine 825 mg/m2 twice per day 5 days per week during radiotherapy; radiotherapy consisted of 50·4-54·0 Gy in 28-30 fractions). 5-9 weeks later, patients underwent response assessment. Patients with a complete response could opt for non-operative management and proceed to modified FOLFOX6 (intravenous leucovorin 400 mg/m2 on day 1, intravenous fluorouracil 400 mg/m2 on day 1 then 2400 mg/m2 over 46 h, and intravenous oxaliplatin 85 mg/m2 on day 1 delivered every 2 weeks for eight cycles) or CAPEOX (intravenous oxaliplatin 130 mg/m2 on day 1 and oral capecitabine 1000 mg/m2 twice daily for 14 days every 3 weeks for four cycles). Patients with less than complete response underwent surgical resection. The primary endpoint was complete response rate, which was a composite of pathological complete response in patients who proceeded to surgery, or clinical complete response maintained at 1 year after last therapy in patients with non-operative management. Safety was a coprimary endpoint. Both endpoints were assessed in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT02688712, and is active but not recruiting. FINDINGS: Between Oct 19, 2016, and Aug 31, 2020, 38 participants were enrolled. 25 (71%) of the 35 patients who completed chemoradiotherapy proceeded to total mesorectal excision surgery, five (20%) of whom had pathological complete responses. Ten (29%) patients had non-operative management, three (30%) of whom ultimately chose to have total mesorectal excision. Two (67%) of those three patients had pathological complete responses. Of the remaining seven patients in the non-operative management group, five (71%) had clinical complete responses at 1 year after their last modified FOLFOX6 infusion. In total, 12 (32% [one-sided 95% CI ≥19%]) of 38 patients had a complete response. Common grade 3 adverse events during treatment included diarrhoea in six (16%) of 38 patients, and haematological toxicity in seven (18%) patients. Two (5%) patients had grade 4 adverse events, one related to chemoradiotherapy-induced diarrhoea and dehydration, and the other an intraoperative ischaemic event. No treatment-related deaths occurred. INTERPRETATION: The addition of galunisertib to neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer improved the complete response rate to 32%, was well tolerated, and warrants further assessment in randomised trials. FUNDING: Eli Lilly via ExIST program, The Providence Foundation.


Subject(s)
Adenocarcinoma , Neoplasms, Second Primary , Rectal Neoplasms , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Capecitabine , Chemoradiotherapy/adverse effects , Diarrhea/etiology , Fluorouracil , Humans , Neoadjuvant Therapy/adverse effects , Neoplasm Staging , Neoplasms, Second Primary/pathology , Oxaliplatin , Pyrazoles , Quinolines , Rectal Neoplasms/drug therapy , Rectal Neoplasms/pathology , Transforming Growth Factor beta
2.
J Immunol ; 204(12): 3416-3424, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32341058

ABSTRACT

Radiation therapy is capable of directing adaptive immune responses against tumors by stimulating the release of endogenous adjuvants and tumor-associated Ags. Within the tumor, conventional type 1 dendritic cells (cDC1s) are uniquely positioned to respond to these signals, uptake exogenous tumor Ags, and migrate to the tumor draining lymph node to initiate cross-priming of tumor-reactive cytotoxic CD8+ T cells. In this study, we report that radiation therapy promotes the activation of intratumoral cDC1s in radioimmunogenic murine tumors, and this process fails to occur in poorly radioimmunogenic murine tumors. In poorly radioimmunogenic tumors, the adjuvant polyinosinic-polycytidylic acid overcomes this failure following radiation and successfully drives intratumoral cDC1 maturation, ultimately resulting in durable tumor cures. Depletion studies revealed that both cDC1 and CD8+ T cells are required for tumor regression following combination therapy. We further demonstrate that treatment with radiation and polyinosinic-polycytidylic acid significantly expands the proportion of proliferating CD8+ T cells in the tumor with enhanced cytolytic potential and requires T cell migration from lymph nodes for therapeutic efficacy. Thus, we conclude that lack of endogenous adjuvant release or active suppression following radiation therapy may limit its efficacy in poorly radioimmunogenic tumors, and coadministration of exogenous adjuvants that promote cDC1 maturation and migration can overcome this limitation to improve tumor control following radiation therapy.


Subject(s)
Dendritic Cells/immunology , Neoplasms/immunology , Neoplasms/radiotherapy , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Movement/immunology , Cross-Priming/immunology , Immunotherapy, Adoptive/methods , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C/immunology , Radiotherapy/methods
3.
Cancer Immunol Immunother ; 70(4): 989-1000, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33097963

ABSTRACT

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


Subject(s)
Carcinoma, Pancreatic Ductal/radiotherapy , Gamma Rays , Myeloid-Derived Suppressor Cells/immunology , Oligonucleotides, Antisense/genetics , Pancreatic Neoplasms/radiotherapy , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Female , Humans , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Mice, Nude , Myeloid-Derived Suppressor Cells/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Prognosis , STAT3 Transcription Factor/genetics , T-Lymphocytes, Regulatory/immunology , Tumor Cells, Cultured , Tumor Microenvironment
4.
J Immunol ; 200(1): 177-185, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29150567

ABSTRACT

Although prophylactic vaccines provide protective humoral immunity against infectious agents, vaccines that elicit potent CD8 T cell responses are valuable tools to shape and drive cellular immunity against cancer and intracellular infection. In particular, IFN-γ-polarized cytotoxic CD8 T cell immunity is considered optimal for protective immunity against intracellular Ags. Suppressor of cytokine signaling (SOCS)1 is a cross-functional negative regulator of TLR and cytokine receptor signaling via degradation of the receptor-signaling complex. We hypothesized that loss of SOCS1 in dendritic cells (DCs) would improve T cell responses by accentuating IFN-γ-directed immune responses. We tested this hypothesis using a recombinant Listeria monocytogenes vaccine platform that targets CD11c+ DCs in mice in which SOCS1 is selectively deleted in all CD11c+ cells. Unexpectedly, in mice lacking SOCS1 expression in CD11c+ cells, we observed a decrease in CD8+ T cell response to the L. monocytogenes vaccine. NK cell responses were also decreased in mice lacking SOCS1 expression in CD11c+ cells but did not explain the defect in CD8+ T cell immunity. We found that DCs lacking SOCS1 expression were functional in driving Ag-specific CD8+ T cell expansion in vitro but that this process was defective following infection in vivo. Instead, monocyte-derived innate TNF-α and inducible NO synthase-producing DCs dominated the antibacterial response. Thus, loss of SOCS1 in CD11c+ cells skewed the balance of immune response to infection by increasing innate responses while decreasing Ag-specific adaptive responses to infectious Ags.


Subject(s)
Bacterial Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Suppressor of Cytokine Signaling 1 Protein/metabolism , Adaptive Immunity , Animals , CD11c Antigen/metabolism , CD8-Positive T-Lymphocytes/microbiology , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Immunity, Innate , Interferon-gamma/metabolism , Killer Cells, Natural/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Suppressor of Cytokine Signaling 1 Protein/genetics
5.
Hepatol Res ; 47(7): 702-714, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27501850

ABSTRACT

AIM: Small, solitary hepatocellular carcinoma is curable with stereotactic radiation or other methods of tumor ablation, however, regional and systemic tumor recurrence occurs in over 70% of patients. Here we describe the ability of immunoradiotherapy to induce an antitumor immune response and delay the growth of tumors in immunocompetent mice. METHODS: A syngeneic hepatocellular carcinoma cell line (Hep-55.1c) was injected directly into the livers of C57BL/6 mice using ultrasound guidance, then tumors were treated with stereotactic radiation using a Small Animal Radiation Research Platform with computed tomography guidance. RESULTS: Delivery of three doses of 250 µg anti-programmed cell death protein-1 (αPD-1) antibody concurrently with 30 Gy stereotactic body radiation therapy in three fractions reduced the growth rate of tumors and improved survival (P < 0.05). Combined treatment was associated with increased CD8+ cytotoxic T cells in the tumor; depletion of CD8 T cells eliminated the efficacy of combined treatment. Combined treatment also induced expression of programmed cell death-1 ligand expression on tumor-infiltrating macrophages, and the tumors grew rapidly after αPD-1 treatment was discontinued. CONCLUSIONS: Tumor response to stereotactic radiation can be augmented by concurrent treatment with αPD-1. The efficacy of this combination therapy was transient, however, and treatment induced markers of adaptive immune resistance. These data are promising, but also indicate that mechanisms of immune resistance will need to be durably overcome for this combination to generate lasting immunity to protect against tumor recurrence.

6.
J Immunol ; 190(7): 3246-55, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23460736

ABSTRACT

Little is known of the regulation of IL-23 secretion in dendritic cells (DC) despite its importance for human Th17 responses. In this study, we show for first time, to our knowledge, that the ataxia telangiectasia mutated (ATM) pathway, involved in DNA damage sensing, acts as an IL-23 repressor. Inhibition of ATM with the highly selective antagonist KU55933 markedly increased IL-23 secretion in human monocyte-derived DC and freshly isolated myeloid DC. In contrast, inhibiting the closely related mammalian target of rapamycin had no effect on IL-23. Priming naive CD4(+) T cells with ATM-inhibited DC increased Th17 responses over and above those obtained with mature DC. Although ATM blockade increased the abundance of p19, p35, and p40 mRNA, IL-12p70 secretion was unaffected. To further examine a role for ATM in IL-23 regulation, we exposed DC to low doses of ionizing radiation. Exposure of DC to x-rays resulted in ATM phosphorylation and a corresponding depression of IL-23. Importantly, ATM inhibition with KU55933 prevented radiation-induced ATM phosphorylation and abrogated the capacity of x-rays to suppress IL-23. To explore how ATM repressed IL-23, we examined a role for endoplasmic reticulum stress responses by measuring generation of the spliced form of X-box protein-1, a key endoplasmic reticulum stress transcription factor. Inhibition of ATM increased the abundance of X-box protein-1 mRNA, and this was followed 3 h later by increased peak p19 transcription and IL-23 release. In summary, ATM activation or inhibition, respectively, inhibited or augmented IL-23 release. This novel role of the ATM pathway represents a new therapeutic target in autoimmunity and vaccine development.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Gene Expression Regulation , Interleukin-23/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , Dendritic Cells/immunology , Endoplasmic Reticulum Stress , Enzyme Activation/radiation effects , Gene Expression Regulation/radiation effects , Humans , Interleukin-23/metabolism , Lymphocyte Activation/immunology , Regulatory Factor X Transcription Factors , Signal Transduction/radiation effects , Th17 Cells/immunology , Th17 Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
8.
Sci Rep ; 14(1): 11909, 2024 05 24.
Article in English | MEDLINE | ID: mdl-38789721

ABSTRACT

T cells recirculate through tissues and lymphatic organs to scan for their cognate antigen. Radiation therapy provides site-specific cytotoxicity to kill cancer cells but also has the potential to eliminate the tumor-specific T cells in field. To dynamically study the effect of radiation on CD8 T cell recirculation, we used the Kaede mouse model to photoconvert tumor-infiltrating cells and monitor their movement out of the field of radiation. We demonstrate that radiation results in loss of CD8 T cell recirculation from the tumor to the lymph node and to distant sites. Using scRNASeq, we see decreased proliferating CD8 T cells in the tumor following radiation therapy resulting in a proportional enrichment in exhausted phenotypes. By contrast, 5 days following radiation increased recirculation of T cells from the tumor to the tumor draining lymph node corresponds with increased immunosurveillance of the treated tumor. These data demonstrate that tumor radiation therapy transiently impairs systemic T cell recirculation from the treatment site to the draining lymph node and distant untreated tumors. This may inform timing therapies to improve systemic T cell-mediated tumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lymph Nodes/radiation effects , Lymph Nodes/pathology , Lymph Nodes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/radiotherapy , Neoplasms/immunology , Neoplasms/pathology , Cell Tracking/methods , Cell Line, Tumor , Mice, Inbred C57BL , Fluorescence
9.
Clin Dev Immunol ; 2013: 281958, 2013.
Article in English | MEDLINE | ID: mdl-23653658

ABSTRACT

Radiation therapy is showing potential as a partner for immunotherapies in preclinical cancer models and early clinical studies. As has been discussed elsewhere, radiation provides debulking, antigen and adjuvant release, and inflammatory targeting of effector cells to the treatment site, thereby assisting multiple critical checkpoints in antitumor adaptive immunity. Adaptive immunity is terminated by inflammatory resolution, an active process which ensures that inflammatory damage is repaired and tissue function is restored. We discuss how radiation therapy similarly triggers inflammation followed by repair, the consequences to adaptive immune responses in the treatment site, and how the myeloid response to radiation may impact immunotherapies designed to improve control of residual cancer cells.


Subject(s)
Inflammation/radiotherapy , Myeloid Cells/radiation effects , Neoplasms/radiotherapy , Adaptive Immunity/radiation effects , Animals , Antigens, Neoplasm/immunology , Clinical Trials as Topic , Disease Models, Animal , Humans , Immunomodulation , Immunotherapy , Myeloid Cells/immunology , Neoplasms/immunology , Wound Healing/radiation effects
10.
Methods Cell Biol ; 174: 55-63, 2023.
Article in English | MEDLINE | ID: mdl-36710051

ABSTRACT

The response to radiation therapy incorporates both the direct impacts of radiation on cancer cells as well as the immune consequences that can help or hinder control of residual disease. Understanding the response of an individual patient's cancer to radiation, and the impact of radiation on the immune cell subsets present in the tumor prior to radiation therapy, can help identify potential predictors of outcome. Here, we describe a methodological approach to using an explant tumor model to characterize and study the immune cell subsets in murine tumors following exposure to ex vivo radiation treatment. The broader tumor environment incorporates distinct microenvironments consisting of tumor stroma and cancer cell nests, with limited interchange between these zones. Ex vivo analysis of tumor explants ensures that these environments remain intact and allows patient-specific response assessments with a broader range of treatment conditions to find optimal conditions and immunotherapy combinations. While this protocol describes the treatment of murine tumors, with minor variations the same protocol can be used to study and characterize various immune populations following radiation therapy in human tumors.


Subject(s)
Neoplasms , Humans , Animals , Mice , Neoplasms/radiotherapy , Immunotherapy/methods , Tumor Microenvironment
11.
Int Rev Cell Mol Biol ; 378: 61-104, 2023.
Article in English | MEDLINE | ID: mdl-37438021

ABSTRACT

Dendritic cells perform critical functions in bridging innate and adaptive immunity. Their ability to sense adjuvant signals in their environment, migrate on maturation, and cross-present cell-associated antigens enables these cells to carry antigen from tissue sites to lymph nodes, and thereby prime naïve T cells that cannot enter tissues. Despite being an infrequent cell type in tumors, we discuss how dendritic cells impact the immune environment of tumors and their response to cancer therapies. We review how radiation therapy of tumors can impact dendritic cells, through transfer of cell associated antigens to dendritic cells and the release of endogenous adjuvants, resulting in increased antigen presentation in the tumor-draining lymph nodes. We explore how tumor specific factors can result in negative regulation of dendritic cell function in the tumor, and the impact of direct radiation exposure to dendritic cells in the treatment field. These data suggest an important role for dendritic cell subpopulations in activating new T cell responses and boosting existing T cell responses to tumor associated antigens in tumor draining lymph nodes following radiation therapy. It further justifies a focus on the needs of the lymph node T cells to improve systemic anti-immunity following radiation therapy.


Subject(s)
Adaptive Immunity , Antigen Presentation , Dendritic Cells
12.
Front Oral Health ; 4: 1180869, 2023.
Article in English | MEDLINE | ID: mdl-37496754

ABSTRACT

Although treatment modalities for head and neck cancer have evolved considerably over the past decades, survival rates have plateaued. The treatment options remained limited to definitive surgery, surgery followed by fractionated radiotherapy with optional chemotherapy, and a definitive combination of fractionated radiotherapy and chemotherapy. Lately, immunotherapy has been introduced as the fourth modality of treatment, mainly administered as a single checkpoint inhibitor for recurrent or metastatic disease. While other regimens and combinations of immunotherapy and targeted therapy are being tested in clinical trials, adapting the appropriate regimens to patients and predicting their outcomes have yet to reach the clinical setting. Radiotherapy is mainly regarded as a means to target cancer cells while minimizing the unwanted peripheral effect. Radiotherapy regimens and fractionation are designed to serve this purpose, while the systemic effect of radiation on the immune response is rarely considered a factor while designing treatment. To bridge this gap, this review will highlight the effect of radiotherapy on the tumor microenvironment locally, and the immune response systemically. We will review the methodology to identify potential targets for therapy in the tumor microenvironment and the scientific basis for combining targeted therapy and radiotherapy. We will describe a current experience in preclinical models to test these combinations and propose how challenges in this realm may be faced. We will review new players in targeted therapy and their utilization to drive immunogenic response against head and neck cancer. We will outline the factors contributing to head and neck cancer heterogeneity and their effect on the response to radiotherapy. We will review in-silico methods to decipher intertumoral and intratumoral heterogeneity and how these algorithms can predict treatment outcomes. We propose that (a) the sequence of surgery, radiotherapy, chemotherapy, and targeted therapy should be designed not only to annul cancer directly, but to prime the immune response. (b) Fractionation of radiotherapy and the extent of the irradiated field should facilitate systemic immunity to develop. (c) New players in targeted therapy should be evaluated in translational studies toward clinical trials. (d) Head and neck cancer treatment should be personalized according to patients and tumor-specific factors.

13.
Sci Rep ; 13(1): 8634, 2023 05 27.
Article in English | MEDLINE | ID: mdl-37244938

ABSTRACT

Radiation therapy induces immunogenic cell death in cancer cells, whereby released endogenous adjuvants are sensed by immune cells to direct adaptive immune responses. TLRs expressed on several immune subtypes recognize innate adjuvants to direct downstream inflammatory responses in part via the adapter protein MyD88. We generated Myd88 conditional knockout mice to interrogate its contribution to the immune response to radiation therapy in distinct immune populations in pancreatic cancer. Surprisingly, Myd88 deletion in Itgax (CD11c)-expressing dendritic cells had little discernable effects on response to RT in pancreatic cancer and elicited normal T cell responses using a prime/boost vaccination strategy. Myd88 deletion in Lck-expressing T cells resulted in similar or worsened responses to radiation therapy compared to wild-type mice and lacked antigen-specific CD8+ T cell responses from vaccination, similar to observations in Myd88-/- mice. Lyz2-specific loss of Myd88 in myeloid populations rendered tumors more susceptible to radiation therapy and elicited normal CD8+ T cell responses to vaccination. scRNAseq in Lyz2-Cre/Myd88fl/fl mice revealed gene signatures in macrophages and monocytes indicative of enhanced type I and II interferon responses, and improved responses to RT were dependent on CD8+ T cells and IFNAR1. Together, these data implicate MyD88 signaling in myeloid cells as a critical source of immunosuppression that hinders adaptive immune tumor control following radiation therapy.


Subject(s)
CD8-Positive T-Lymphocytes , Pancreatic Neoplasms , Mice , Animals , Myeloid Differentiation Factor 88/metabolism , Monocytes/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/radiotherapy , Mice, Knockout , Adjuvants, Immunologic/metabolism , Mice, Inbred C57BL , Pancreatic Neoplasms
14.
Sci Rep ; 13(1): 6277, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37072485

ABSTRACT

Tissue resident memory (Trm) CD8 T cells infiltrating tumors represent an enriched population of tumor antigen-specific T cells, and their presence is associated with improved outcomes in patients. Using genetically engineered mouse pancreatic tumor models we demonstrate that tumor implantation generates a Trm niche that is dependent on direct antigen presentation by cancer cells. However, we observe that initial CCR7-mediated localization of CD8 T cells to tumor draining lymph nodes is required to subsequently generate CD103+ CD8 T cells in tumors. We observe that the formation of CD103+ CD8 T cells in tumors is dependent on CD40L but independent of CD4 T cells, and using mixed chimeras we show that CD8 T cells can provide their own CD40L to permit CD103+ CD8 T cell differentiation. Finally, we show that CD40L is required to provide systemic protection against secondary tumors. These data suggest that CD103+ CD8 T cell formation in tumors can occur independent of the two-factor authentication provided by CD4 T cells and highlight CD103+ CD8 T cells as a distinct differentiation decision from CD4-dependent central memory.


Subject(s)
Immunologic Memory , Neoplasms , Animals , Mice , CD40 Ligand , Neoplasms/pathology , CD8-Positive T-Lymphocytes , Lymphocyte Activation
15.
Immunology ; 136(4): 437-47, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22578109

ABSTRACT

The treatment of high-grade tumours must consider a tumour environment dominated by cells that support cancer growth. In addition to directing angiogenesis and invasion, alternatively activated macrophages in the tumour provide protection from adaptive immunity and permit tumour growth. Agonist antibodies to the tumour necrosis factor receptor family member OX40 are an effective therapy for cancer in a range of murine models; however, as with many immune therapies, αOX40 therapy is less effective as the tumour grows and develops an immune suppressive environment. We demonstrate that αOX40 directly activates T cells and that this T-cell activation alters macrophage differentiation in the tumour environment. We demonstrate that macrophages in the tumour limit the efficacy of αOX40 therapy, and that combining αOX40 therapy with inhibitors of arginase significantly enhances survival of tumour-bearing mice. These data demonstrate that macrophages in the tumour environment limit the effectiveness of OX40-based immunotherapy, and combination therapies that target both the cell-mediated immune response and the suppressive tumour environment will be required for translation of effective immunotherapies to patients with established tumours.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Macrophages/immunology , Neoplasms, Experimental/therapy , Receptors, OX40/immunology , Animals , Antibodies, Monoclonal/immunology , Arginase/antagonists & inhibitors , Cell Differentiation , Interferon-gamma/metabolism , Interleukin-12/pharmacology , Interleukin-18/pharmacology , Lymphocyte Activation , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Receptors, OX40/agonists , Tumor Microenvironment/immunology
16.
Neoplasia ; 31: 100808, 2022 09.
Article in English | MEDLINE | ID: mdl-35691060

ABSTRACT

In this review we consider what appears to be a paradox in immunotherapies based around radiation therapy. The paradox is based on three main points. 1. That T cells are needed for radiation's efficacy; 2. That tumor-specific T cells are enriched in the field of treatment; and 3. That radiation kills T cells in the treatment field. We discuss evidence of the effect of radiation on T cells in the field given their ongoing movement in and out of tissues and the tumor, and how the movement of T cells impacts the treated primary tumor and untreated distant metastases. Given this evidence, we revisit the paradox to understand how the extraordinary efficacy of radiation and immunity in preclinical models is dependent on this radiation sensitive cell.


Subject(s)
Neoplasms , T-Lymphocytes , Humans , Immunotherapy
17.
Front Oral Health ; 3: 902160, 2022.
Article in English | MEDLINE | ID: mdl-35937775

ABSTRACT

The clinical response to cancer therapies involves the complex interplay between the systemic, tumoral, and stromal immune response as well as the direct impact of treatments on cancer cells. Each individual's immunological and cancer histories are different, and their carcinogen exposures may differ. This means that even though two patients with oral tumors may carry an identical mutation in TP53, they are likely to have different pre-existing immune responses to their tumors. These differences may arise due to their distinct accessory mutations, genetic backgrounds, and may relate to clinical factors including previous chemotherapy exposure and concurrent medical comorbidities. In isolation, their cancer cells may respond similarly to cancer therapy, but due to their baseline variability in pre-existing immune responses, patients can have different responses to identical therapies. In this review we discuss how the immune environment of tumors develops, the critical immune cell populations in advanced cancers, and how immune interventions can manipulate the immune environment of patients with pre-malignancies or advanced cancers to improve therapeutic outcomes.

18.
Sci Rep ; 12(1): 14954, 2022 Sep 02.
Article in English | MEDLINE | ID: mdl-36056093

ABSTRACT

Multiple preclinical studies have shown improved outcomes when radiation therapy is combined with immune modulating antibodies. However, to date, many of these promising results have failed to translate to successful clinical studies. This led us to explore additional checkpoint and co-stimulatory pathways that may be regulated by radiation therapy. Here, we demonstrate that radiation increases the expression of inducible T cell co-stimulator (ICOS) on both CD4 and CD8 T cells in the blood following treatment. Moreover, when we combined a novel ICOS agonist antibody with radiation we observed durable cures across multiple tumor models and mouse strains. Depletion studies revealed that CD8 T cells were ultimately required for treatment efficacy, but CD4 T cells and NK cells also partially contributed to tumor control. Phenotypic analysis showed that the combination therapy diminished the increased infiltration of regulatory T cells into the tumor that typically occurs following radiation alone. Finally, we demonstrate in a poorly immunogenic pancreatic tumor model which is resistant to combined radiation and anti-PD1 checkpoint blockade that the addition of this novel ICOS agonist antibody to the treatment regimen results in tumor control. These findings identify ICOS as part of a T cell pathway that is modulated by radiation and targeting this pathway with a novel ICOS antibody results in durable tumor control in preclinical models.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Animals , Antibodies/metabolism , CD4-Positive T-Lymphocytes , Inducible T-Cell Co-Stimulator Protein/metabolism , Mice , Neoplasms/metabolism , T-Lymphocytes, Regulatory
19.
Life Sci Alliance ; 5(9)2022 09.
Article in English | MEDLINE | ID: mdl-35487695

ABSTRACT

Radiation therapy generates extensive cancer cell death capable of promoting tumor-specific immunity. Within the tumor, conventional dendritic cells (cDCs) are known to carry tumor-associated antigens to the draining lymph node (TdLN) where they initiate T-cell priming. How radiation influences cDC migration is poorly understood. Here, we show that immunological efficacy of radiation therapy is dependent on cDC migration in radioimmunogenic tumors. Using photoconvertible mice, we demonstrate that radiation impairs cDC migration to the TdLN in poorly radioimmunogenic tumors. Comparative transcriptional analysis revealed that cDCs in radioimmunogenic tumors express genes associated with activation of endogenous adjuvant signaling pathways when compared with poorly radioimmunogenic tumors. Moreover, an exogenous adjuvant combined with radiation increased the number of migrating cDCs in these poorly radioimmunogenic tumors. Taken together, our data demonstrate that cDC migration play a critical role in the response to radiation therapy.


Subject(s)
Dendritic Cells , Lymph Nodes , Animals , Mice , T-Lymphocytes
20.
J Vasc Surg ; 54(4): 1215-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21871773

ABSTRACT

BACKGROUND: The benefit of carotid endarterectomy (CEA) over best medical therapy was established using intra-arterial angiography (IAA) for patient selection. Its cost, availability, and risk together with the emergence of newer imaging modalities have led to its replacement in the routine assessment of internal carotid artery (ICA) stenosis. The relative performance of these methods should dictate the optimum imaging strategy in symptomatic patients. METHODS: A previous meta-analysis (NIHR Health Technology Assessment Programme) was reviewed. Medline and PubMed search was performed for relevant publications since 2006 together with a review of the references in retrieved publications. RESULTS: Compared to IAA, the sensitivity and specificity for noninvasive imaging of a ≥70% to 99% ICA stenosis are duplex ultrasound (DUS): 0.89 (0.85-0.92) and 0.84 (0.77-0.89); time-of-flight magnetic resonance angiography (TOF-MRA): 0.88 (0.82-0.92) and 0.84 (0.76-0.97); contrast-enhanced MRA (CE-MRA): 0.94 (0.88-0.97) and 0.93 (0.89-0.96); and computed tomography angiography: 0.77 (0.68-0.84) and 0.95 (0.91-0.97), respectively. A policy of initial DUS followed by confirmatory CE-MRA best matches patient selection by arteriography. Single modality imaging for 50% to 69% ICA stenoses suggests reduced reliability resulting in more inappropriate operations. CONCLUSIONS: DUS is the optimum screening tool due to its sensitivity and specificity, availability, and low cost. When CEA appears indicated, confirmatory imaging with CE-MRA is the most reliable and cost-effective method of investigation.


Subject(s)
Carotid Stenosis/diagnosis , Diagnostic Imaging , Carotid Stenosis/complications , Carotid Stenosis/economics , Carotid Stenosis/therapy , Contrast Media , Cost-Benefit Analysis , Diagnostic Imaging/economics , Diagnostic Imaging/methods , Evidence-Based Medicine , Health Care Costs , Humans , Magnetic Resonance Angiography , Predictive Value of Tests , Sensitivity and Specificity , Severity of Illness Index , Tomography, X-Ray Computed , Ultrasonography, Doppler, Duplex
SELECTION OF CITATIONS
SEARCH DETAIL