Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell ; 187(10): 2521-2535.e21, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38697107

ABSTRACT

Cancer immunotherapy remains limited by poor antigenicity and a regulatory tumor microenvironment (TME). Here, we create "onion-like" multi-lamellar RNA lipid particle aggregates (LPAs) to substantially enhance the payload packaging and immunogenicity of tumor mRNA antigens. Unlike current mRNA vaccine designs that rely on payload packaging into nanoparticle cores for Toll-like receptor engagement in immune cells, systemically administered RNA-LPAs activate RIG-I in stromal cells, eliciting massive cytokine/chemokine response and dendritic cell/lymphocyte trafficking that provokes cancer immunogenicity and mediates rejection of both early- and late-stage murine tumor models. In client-owned canines with terminal gliomas, RNA-LPAs improved survivorship and reprogrammed the TME, which became "hot" within days of a single infusion. In a first-in-human trial, RNA-LPAs elicited rapid cytokine/chemokine release, immune activation/trafficking, tissue-confirmed pseudoprogression, and glioma-specific immune responses in glioblastoma patients. These data support RNA-LPAs as a new technology that simultaneously reprograms the TME while eliciting rapid and enduring cancer immunotherapy.


Subject(s)
Immunotherapy , Lipids , RNA , Tumor Microenvironment , Animals , Dogs , Female , Humans , Mice , Antigens, Neoplasm/immunology , Brain Neoplasms/therapy , Brain Neoplasms/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Glioblastoma/therapy , Glioblastoma/immunology , Glioma/therapy , Glioma/immunology , Immunotherapy/methods , Mice, Inbred C57BL , Neoplasms/therapy , Neoplasms/immunology , RNA/chemistry , RNA/therapeutic use , RNA, Messenger/metabolism , RNA, Messenger/genetics , Lipids/chemistry
2.
bioRxiv ; 2023 Mar 23.
Article in English | MEDLINE | ID: mdl-36993158

ABSTRACT

To prospectively determine whether brain tumors will respond to immune checkpoint inhibitors (ICIs), we developed a novel mRNA vaccine as a viral mimic to elucidate cytokine release from brain cancer cells in vitro. Our results indicate that cytokine signatures following mRNA challenge differ substantially from ICI responsive versus non-responsive murine tumors. These findings allow for creation of a diagnostic assay to quickly assess brain tumor immunogenicity, allowing for informed treatment with ICI or lack thereof in poorly immunogenic settings.

3.
medRxiv ; 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36993772

ABSTRACT

Messenger RNA (mRNA) has emerged as a remarkable tool for COVID-19 prevention but its use for induction of therapeutic cancer immunotherapy remains limited by poor antigenicity and a regulatory tumor microenvironment (TME). Herein, we develop a facile approach for substantially enhancing immunogenicity of tumor-derived mRNA in lipid-particle (LP) delivery systems. By using mRNA as a molecular bridge with ultrapure liposomes and foregoing helper lipids, we promote the formation of 'onion-like' multi-lamellar RNA-LP aggregates (LPA). Intravenous administration of RNA-LPAs mimics infectious emboli and elicits massive DC/T cell mobilization into lymphoid tissues provoking cancer immunogenicity and mediating rejection of both early and late-stage murine tumor models. Unlike current mRNA vaccine designs that rely on payload packaging into nanoparticle cores for toll-like receptor engagement, RNA-LPAs stimulate intracellular pathogen recognition receptors (RIG-I) and reprogram the TME thus enabling therapeutic T cell activity. RNA-LPAs were safe in acute/chronic murine GLP toxicology studies and immunologically active in client-owned canines with terminal gliomas. In an early phase first-in-human trial for patients with glioblastoma, we show that RNA-LPAs encoding for tumor-associated antigens elicit rapid induction of pro-inflammatory cytokines, mobilization/activation of monocytes and lymphocytes, and expansion of antigen-specific T cell immunity. These data support the use of RNA-LPAs as novel tools to elicit and sustain immune responses against poorly immunogenic tumors.

4.
Front Immunol ; 12: 705580, 2021.
Article in English | MEDLINE | ID: mdl-34421912

ABSTRACT

Evading T cell surveillance is a hallmark of cancer. Patients with solid tissue malignancy, such as glioblastoma (GBM), have multiple forms of immune dysfunction, including defective T cell function. T cell dysfunction is exacerbated by standard treatment strategies such as steroids, chemotherapy, and radiation. Reinvigoration of T cell responses can be achieved by utilizing adoptively transferred T cells, including CAR T cells. However, these cells are at risk for depletion and dysfunction as well. This review will discuss adoptive T cell transfer strategies and methods to avoid T cell dysfunction for the treatment of brain cancer.


Subject(s)
Brain Neoplasms , Glioblastoma , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Glioblastoma/immunology , Glioblastoma/therapy , Humans , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use
5.
Front Oncol ; 11: 691246, 2021.
Article in English | MEDLINE | ID: mdl-34150663

ABSTRACT

Immunotherapy has revolutionized care for many solid tissue malignancies, and is being investigated for efficacy in the treatment of malignant brain tumors. Identifying a non-invasive monitoring technique such as metabolomics monitoring to predict patient response to immunotherapy has the potential to simplify treatment decision-making and to ensure therapy is tailored based on early patient response. Metabolomic analysis of peripheral immune response is feasible due to large metabolic shifts that immune cells undergo when activated. The utility of this approach is under investigation. In this review, we discuss the metabolic changes induced during activation of an immune response, and the role of metabolic profiling to monitor immune responses in the context of immunotherapy for malignant brain tumors. This review provides original insights into how metabolomics monitoring could have an important impact in the field of tumor immunotherapy if achievable.

6.
Neuro Oncol ; 22(9): 1249-1261, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32391559

ABSTRACT

In oncology, "immunotherapy" is a broad term encompassing multiple means of utilizing the patient's immune system to combat malignancy. Prominent among these are immune checkpoint inhibitors, cellular therapies including chimeric antigen receptor T-cell therapy, vaccines, and oncolytic viruses. Immunotherapy for glioblastoma (GBM) has had mixed results in early trials. In this context, the past, present, and future of immune oncology for the treatment of GBM was discussed by clinical, research, and thought leaders as well as patient advocates at the first annual Remission Summit in 2019. The goal was to use current knowledge (published and unpublished) to identify possible causes of treatment failures and the best strategies to advance immunotherapy as a treatment modality for patients with GBM. The discussion focuses on past failures, current limitations, failure analyses, and proposed best practices moving forward.


Subject(s)
Brain Neoplasms , Glioblastoma , Oncolytic Viruses , Adult , Brain Neoplasms/therapy , Glioblastoma/therapy , Humans , Immunotherapy
7.
Galen Med J ; 9: e1730, 2020.
Article in English | MEDLINE | ID: mdl-34466580

ABSTRACT

BACKGROUND: It has been well-documented that the Fc receptor-like (FCRL) molecule contributes to the pathogenesis of certain autoimmune disorders. FCRL molecules belong to the immunoglobulin superfamily produced by B cells. Also, these molecules induce activating or inhibitory signals of B cells. According to this information and also considering the critical role of immune reactions in organ transplantation, the following experiment was performed to analyze the gene expression level of FCRLs in peripheral blood mononuclear cells of kidney transplant recipients. MATERIALS AND METHODS: Blood samples were obtained from 32 renal transplant patients on days 1, 3, and 7 post-transplantations. Patients were divided into two groups according to the presence or absence of rejection. Also, 24 age-matched healthy subjects were enrolled as control group. After total RNA extraction from peripheral blood mononuclear cells (PBMC) and cDNA synthesis, the gene expression levels of FCRL1, FCRL2, and FCRL4 in each group were measured by real-time polymerase chain reaction. RESULTS: Our results showed that FCRL1 expression levels in kidney transplant patients were significantly less than healthy controls. The overall FCRL2 expression level was not significantly different between them. However, at days 1 and 7, following transplantation in the non-rejected group FCRL2 level was significantly higher than the control group. Comparing the FCRL4 gene expression levels of both groups with healthy controls showed a significant decrease in the third and seventh days post-transplantation. CONCLUSION: It can be concluded that mononuclear cells, mainly B cells, have an essential role to play in kidney transplantation.

8.
Ultrason Sonochem ; 68: 104460, 2020 Nov.
Article in English | MEDLINE | ID: mdl-30712851

ABSTRACT

The aim of this study was ultrasound assisted removal of Ceftriaxone sodium (CS) based on CCD model. Using sonochemical synthesized Bi2WO6 implanted on graphitic carbon nitride/Multiwall carbon nanotube (g-C3N4/MWCNT/Bi2WO6). For this purpose g-C3N4/MWCNT/Bi2WO6 was synthesized and characterized using diverse approaches including XRD, FE-SEM, XPS, EDS, HRTEM, FT-IR. Then, the contribution of conventional variables including pH, CS concentration, adsorbent dosage and ultrasound contact time were studied by central composite design (CCD) under response surface methodology (RSM). ANOVA was employed to the variable factors, and the most desirable operational conditions mass provided. Drug adsorption yield of 98.85% obtained under these defined conditions. Through conducting five experiments, the proper prediction of the optimum point were examined. The respective results showed that RSD% was lower than 5% while the t-test confirmed the high quality of fitting. Langmuir isotherm equation fits the experimental data best and the removal followed pseudo-second order kinetics. The estimation of the experimentally obtained maximum adsorption capacities was 19.57 mg.g- of g-C3N4/MWCNT/Bi2WO6 for CS. Boundary layer diffusion explained the mechanism of removal via intraparticle diffusion.

9.
Cancer Res ; 80(3): 499-509, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31723000

ABSTRACT

Anti-VEGF therapy prolongs recurrence-free survival in patients with glioblastoma but does not improve overall survival. To address this discrepancy, we investigated immunologic resistance mechanisms to anti-VEGF therapy in glioma models. A screening of immune-associated alterations in tumors after anti-VEGF treatment revealed a dose-dependent upregulation of regulatory T-cell (Treg) signature genes. Enhanced numbers of Tregs were observed in spleens of tumor-bearing mice and later in tumors after anti-VEGF treatment. Elimination of Tregs with CD25 blockade before anti-VEGF treatment restored IFNγ production from CD8+ T cells and improved antitumor response from anti-VEGF therapy. The treated tumors overexpressed the glutamate/cystine antiporter SLC7A11/xCT that led to elevated extracellular glutamate in these tumors. Glutamate promoted Treg proliferation, activation, suppressive function, and metabotropic glutamate receptor 1 (mGlutR1) expression. We propose that VEGF blockade coupled with glioma-derived glutamate induces systemic and intratumoral immunosuppression by promoting Treg overrepresentation and function, which can be pre-emptively overcome through Treg depletion for enhanced antitumor effects. SIGNIFICANCE: Resistance to VEGF therapy in glioblastoma is driven by upregulation of Tregs, combined blockade of VEGF, and Tregs may provide an additive antitumor effect for treating glioblastoma.


Subject(s)
Bevacizumab/pharmacology , Drug Resistance, Neoplasm , Glioblastoma/immunology , Glutamic Acid/metabolism , T-Lymphocytes, Regulatory/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/pharmacology , Apoptosis , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Female , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/immunology
10.
World Neurosurg ; 135: e510-e519, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31863896

ABSTRACT

BACKGROUND: Patient safety indicators (PSIs) and hospital-acquired conditions (HACs) are reported quality measures. We compared their prevalence in patients with secretory and nonsecretory pituitary adenoma using the National (Nationwide) Inpatient Sample (NIS), Healthcare Cost and Utilization Project, Agency for Healthcare Research and Quality. METHODS: The NIS was queried for hospitalizations 2002-2014 involving pituitary adenomas. Prevalence of PSI, HAC, and 9 pituitary-related complications was determined using International Classification of Diseases, Ninth Revision codes. Patient risk factors were evaluated through multivariate analysis. RESULTS: A total of 20,743 patients with nonsecretory tumor and 3385 patients with secretory tumor were identified. Among patients with nonsecretory tumor, 3.79% experienced any PSI or HAC. Of patients with secretory tumor, 2.54% had any PSI or HAC. Before adjusting for covariation, secretory patients were less likely to have any PSI or HAC (odds ratio [OR], 0.652; P = 0.0002), experience any pituitary-related complication (OR, 0.804; P < 0.0001), have a poor outcome (hazard ratio [HR], 0.435; P < 0.0001), and die during hospitalization (HR, 0.293; P = 0.0015). Secretory patients had significantly shorter mean hospital length of stay (secretory/nonsecretory percent difference, -11.95%; P < 0.0001). However, inverse propensity score-weighted ORs comparing the groups' outcomes showed that there was no significant difference in the prevalence of any PSIs and HACs (OR, 0.963; P = 0.8570), pituitary-related complications (OR, 0.894; P = 0.1321), poor outcomes (HR, 0.990; P = 0.9287), in-hospital death (HR, 0.663; P = 0.2967), and length of stay (percent difference, -2.31%; P = 0.2967) between groups. CONCLUSIONS: Lack of significant difference in outcome measures after controlling for covariation is consistent with our finding that patients with nonsecretory tumor have more comorbidities on presentation for treatment. PSIs and HACs have limited ability to measure complications specific to pituitary tumors.


Subject(s)
Adenoma/complications , Pituitary Neoplasms/complications , Adult , Female , Hospitalization , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Risk Factors
11.
J Immunother Cancer ; 7(1): 321, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31753028

ABSTRACT

BACKGROUND: Dendritic cell (DC) vaccine efficacy is directly related to the efficiency of DC migration to the lymph node after delivery to the patient. We discovered that a naturally occurring metabolite, sarcosine, increases DC migration in human and murine cells resulting in significantly improved anti-tumor efficacy. We hypothesized that sarcosine induced cell migration was due to chemokine signaling. METHODS: DCs were harvested from the bone marrow of wild type C57BL/6 mice and electroporated with tumor messenger RNA (mRNA). Human DCs were isolated from peripheral blood mononuclear cells (PBMCs). DCs were treated with 20 mM of sarcosine. Antigen specific T cells were isolated from transgenic mice and injected intravenously into tumor bearing mice. DC vaccines were delivered via intradermal injection. In vivo migration was evaluated by flow cytometry and immunofluorescence microscopy. Gene expression in RNA was investigated in DCs via RT-PCR and Nanostring. RESULTS: Sarcosine significantly increased human and murine DC migration in vitro. In vivo sarcosine-treated DCs had significantly increased migration to both the lymph nodes and spleens after intradermal delivery in mice. Sarcosine-treated DC vaccines resulted in significantly improved tumor control in a B16F10-OVA tumor flank model and improved survival in an intracranial GL261-gp100 glioma model. Gene expression demonstrated an upregulation of CXCR2, CXCL3 and CXCL1 in sarcosine- treated DCs. Further metabolic analysis demonstrated the up-regulation of cyclooxygenase-1 and Pik3cg. Sarcosine induced migration was abrogated by adding the CXCR2 neutralizing antibody in both human and murine DCs. CXCR2 neutralizing antibody also removed the survival benefit of sarcosine-treated DCs in the tumor models. CONCLUSION: Sarcosine increases the migration of murine and human DCs via the CXC chemokine pathway. This platform can be utilized to improve existing DC vaccine strategies.


Subject(s)
Cancer Vaccines/immunology , Cell Movement/drug effects , Cell Movement/immunology , Dendritic Cells/drug effects , Dendritic Cells/physiology , Receptors, CXCR/metabolism , Sarcosine/pharmacology , Signal Transduction/drug effects , Adoptive Transfer , Animals , Biomarkers , Disease Models, Animal , Humans , Immunotherapy , Mice , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Oxidative Stress , Receptors, CXCR/genetics
12.
Nat Commun ; 10(1): 4016, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31488817

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy targeting solid tumors has stagnated as a result of tumor heterogeneity, immunosuppressive microenvironments, and inadequate intratumoral T cell trafficking and persistence. Early (≤3 days) intratumoral presentation of CAR T cells post-treatment is a superior predictor of survival than peripheral persistence. Therefore, we have co-opted IL-8 release from tumors to enhance intratumoral T-cell trafficking through a CAR design for maximal antitumor activity in solid tumors. Here, we demonstrate that IL-8 receptor, CXCR1 or CXCR2, modified CARs markedly enhance migration and persistence of T cells in the tumor, which induce complete tumor regression and long-lasting immunologic memory in pre-clinical models of aggressive tumors such as glioblastoma, ovarian and pancreatic cancer.


Subject(s)
Glioblastoma/immunology , Immunotherapy, Adoptive , Interleukin-8/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Disease Models, Animal , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mice, Inbred NOD , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
13.
Transplantation ; 103(6): 1121-1130, 2019 06.
Article in English | MEDLINE | ID: mdl-30801518

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) have protolerogenic effects in renal transplantation, but they induce long-term regulatory T cells (Treg)-dependent graft acceptance only when infused before transplantation. When given posttransplant, MSCs home to the graft where they promote engraftment syndrome and do not induce Treg. Unfortunately, pretransplant MSC administration is unfeasible in deceased-donor kidney transplantation. METHODS: To make MSCs a therapeutic option also for deceased organ recipients, we tested whether MSC infusion at the time of transplant (day 0) or posttransplant (day 2) together with inhibition of complement receptors prevents engraftment syndrome and allows their homing to secondary lymphoid organs for promoting tolerance. We analyzed intragraft and splenic MSC localization, graft survival, and alloimmune response in mice recipients of kidney allografts and syngeneic MSCs given on day 0 or on posttransplant day 2. C3a receptor (C3aR) or C5a receptor (C5aR) antagonists were administered to mice in combination with the cells or were used together to treat MSCs before infusion. RESULTS: Syngeneic MSCs given at day 0 homed to the spleen increased Treg numbers and induced long-term graft acceptance. Posttransplant MSC infusion, combined with a short course of C3aR or C5aR antagonist or administration of MSCs pretreated with C3aR and C5aR antagonists, prevented intragraft recruitment of MSCs and graft inflammation, inhibited antidonor T-cell reactivity, but failed to induce Treg, resulting in mild prolongation of graft survival. CONCLUSIONS: These data support testing the safety/efficacy profile of administering MSCs on the day of transplant in deceased-donor transplant recipients and indicate that complement is crucial for MSC recruitment into the kidney allograft.


Subject(s)
Complement Inactivating Agents/administration & dosage , Graft Rejection/prevention & control , Graft Survival/drug effects , Kidney Transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Receptors, Complement/antagonists & inhibitors , Transplantation Tolerance/drug effects , Animals , Drug Administration Schedule , Female , Graft Rejection/immunology , Mesenchymal Stem Cells/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Complement/immunology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/immunology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Time Factors , Transplantation, Homologous , Transplantation, Isogeneic
14.
Neuro Oncol ; 21(6): 730-741, 2019 06 10.
Article in English | MEDLINE | ID: mdl-30668768

ABSTRACT

BACKGROUND: The changes induced in host immunity and the tumor microenvironment by chemotherapy have been shown to impact immunotherapy response in both a positive and a negative fashion. Temozolomide is the most common chemotherapy used to treat glioblastoma (GBM) and has been shown to have variable effects on immune response to immunotherapy. Therefore, we aimed to determine the immune modulatory effects of temozolomide that would impact response to immune checkpoint inhibition in the treatment of experimental GBM. METHODS: Immune function and antitumor efficacy of immune checkpoint inhibition were tested after treatment with metronomic dose (MD) temozolomide (25 mg/kg × 10 days) or standard dose (SD) temozolomide (50 mg/kg × 5 days) in the GL261 and KR158 murine glioma models. RESULTS: SD temozolomide treatment resulted in an upregulation of markers of T-cell exhaustion such as LAG-3 and TIM-3 in lymphocytes which was not seen with MD temozolomide. When temozolomide treatment was combined with programmed cell death 1 (PD-1) antibody therapy, the MD temozolomide/PD-1 antibody group demonstrated a decrease in exhaustion markers in tumor infiltrating lymphocytes that was not observed in the SD temozolomide/PD-1 antibody group. Also, the survival advantage of PD-1 antibody therapy in a murine syngeneic intracranial glioma model was abrogated by adding SD temozolomide to treatment. However, when MD temozolomide was added to PD-1 inhibition, it preserved the survival benefit that was seen by PD-1 antibody therapy alone. CONCLUSION: The peripheral and intratumoral immune microenvironments are distinctively affected by dose modulation of temozolomide.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain Neoplasms/immunology , Glioblastoma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Temozolomide/pharmacology , Animals , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , Tumor Cells, Cultured , Tumor Microenvironment
15.
Oncoimmunology ; 8(1): e1514921, 2019.
Article in English | MEDLINE | ID: mdl-30546954

ABSTRACT

Controversy surrounds the role of cytomegalovirus (CMV) in glioblastoma (GBM). However, several studies have shown that CMV nucleic acids and proteins are present within GBM tumor tissue. CMV has been implicated in GBM pathogenesis by affecting tumor stem cell factors, angiogenesis and immune pathways. Anti-viral therapy has not been found to definitively improve outcomes for patients with GBM. Several studies have leveraged CMV by targeting CMV antigens using ex-vivo expanded T cells or dendritic cell vaccines. The initial results from these studies are promising and larger studies are underway.

16.
Nano Lett ; 18(10): 6195-6206, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30259750

ABSTRACT

Translation of nanoparticles (NPs) into human clinical trials for patients with refractory cancers has lagged due to unknown biologic reactivities of novel NP designs. To overcome these limitations, simple well-characterized mRNA lipid-NPs have been developed as cancer immunotherapeutic vaccines. While the preponderance of RNA lipid-NPs encoding for tumor-associated antigens or neoepitopes have been designed to target lymphoid organs, they remain encumbered by the profound intratumoral and systemic immunosuppression that may stymie an activated T cell response. Herein, we show that systemic localization of untargeted tumor RNA (derived from whole transcriptome) encapsulated in lipid-NPs, with excess positive charge, primes the peripheral and intratumoral milieu for response to immunotherapy. In immunologically resistant tumor models, these RNA-NPs activate the preponderance of systemic and intratumoral myeloid cells (characterized by coexpression of PD-L1 and CD86). Addition of immune checkpoint inhibitors (ICIs) (to animals primed with RNA-NPs) augments peripheral/intratumoral PD-1+CD8+ cells and mediates synergistic antitumor efficacy in settings where ICIs alone do not confer therapeutic benefit. These synergistic effects are mediated by type I interferon released from plasmacytoid dendritic cells (pDCs). In translational studies, personalized mRNA-NPs were safe and active in a client-owned canine with a spontaneous malignant glioma. In summary, we demonstrate widespread immune activation from tumor loaded RNA-NPs concomitant with inducible PD-L1 expression that can be therapeutically exploited. While immunotherapy remains effective for only a subset of cancer patients, combination therapy with systemic immunomodulating RNA-NPs may broaden its therapeutic potency.


Subject(s)
Glioma/drug therapy , Immunotherapy , Lipids/administration & dosage , Nanoparticles/administration & dosage , Precision Medicine , Animals , B7-2 Antigen/antagonists & inhibitors , B7-2 Antigen/genetics , B7-2 Antigen/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Disease Models, Animal , Dogs , Glioma/immunology , Glioma/pathology , Glioma/veterinary , Humans , Lipids/chemistry , Lipids/immunology , Lymphocyte Activation/immunology , Nanoparticles/chemistry , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , Transcriptome/genetics
17.
Neuro Oncol ; 20(12): 1566-1572, 2018 11 12.
Article in English | MEDLINE | ID: mdl-29733389

ABSTRACT

Temozolomide is the most widely used chemotherapy for patients with glioblastoma (GBM) despite the fact that approximately half of treated patients have temozolomide resistance and all patients eventually fail therapy. Due to the limited efficacy of existing therapies, immunotherapy is being widely investigated for patients with GBM. However, initial immunotherapy trials in GBM patients have had disappointing results as monotherapy. Therefore, combinatorial treatment strategies are being investigated. Temozolomide has several effects on the immune system that are dependent on mode of delivery and the dosing strategy, which may have unpredicted effects on immunotherapy. Here we summarize the immune modulating role of temozolomide alone and in combination with immunotherapies such as dendritic cell vaccines, T-cell therapy, and immune checkpoint inhibitors for patients with GBM.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Immunomodulation/drug effects , Temozolomide/therapeutic use , Brain Neoplasms/immunology , Glioblastoma/immunology , Humans , Prognosis
18.
Front Mol Neurosci ; 11: 82, 2018.
Article in English | MEDLINE | ID: mdl-29643764

ABSTRACT

Background: Malignant gliomas are heterogeneous brain tumors with the potential for aggressive disease progression, as influenced by suppressive immunoediting. Given the success and enhanced potential of immune-checkpoint inhibitors in immunotherapy, we focused on the connections between genetic alterations affected by IDH1 mutations and immunological landscape changes and PDL-1 expression in gliomas. Methods: Paired surgically resected tumors from lower-grade gliomas (LGGs) and glioblastomas (GBM) were investigated, and a genetic analysis of patients' primary tumor samples culled from TCGA datasets was performed. Results: The results demonstrate that when compared with IDH1-mutant tumors, IDH1 wildtype tumors represent an immunosuppression landscape and elevated levels of PD-L1 expression. DNA hypo-methylation of the PD-L1 gene, as well as high gene and protein expressions, were observed in the wildtype tumors. We also found that quantitative levels of IDH1 mutant proteins were positively associated with recurrence-free survival (RFS). A key product of the IDH1 mutation (2-hydroxyglutarate) was found to transiently increase DNA methylation and suppress PD-L1 expression. Conclusions: IDH1 mutations impact the immune landscape of gliomas by affecting immune infiltrations and manipulating checkpoint ligand PD-L1 expression. Applications of immune checkpoint inhibitors may be beneficial for chemoradiation-insensitive IDH1-wildtype gliomas.

19.
Immunol Invest ; 44(5): 427-37, 2015.
Article in English | MEDLINE | ID: mdl-26107743

ABSTRACT

Mesenchymal stem cells (MSCs) can modulate dendritic cells (DCs) activation and induce tolerogenic characteristics in DCs. All mechanisms involved in MSCs-induced tolerogenic DCs are not fully understood. MicroRNAs (miRs) play important role in maturation and function of DCs. In this study, we investigated the effects of MSCs culture supernatant (C.S.) on expression of miR-155 and miR-23b in mice DCs. BALB/c mice spleens were used for DCs isolation. MSCs were isolated from the mice bone marrow and cultured in DMEM media. When MSCs expanded to sixth passage, C.S. was collected after 12, 24 and 48 h. Quantitative polymerase chain reaction (QPCR) was used to determine the expression of miR-155 and miR-23b in DCs treated with C.S. after 6 and 12 h. Secretion of IL-23 and TGF- ß were detected in DCs treated with C.S. by ELISA after 24 h. miR-23b expression was significantly increased in DCs treated with 12 h C.S. for 12 h compared to negative controls. miR-155 expression did not change in DCs treated with C.S. after 6 and 12 h. miR-23b expression was significantly increased in DCs treated with 12 h C.S. for 12 h, compared to those treated with C.S. for 6 h. Similarly, miR-23b expression was increased in DCs treated with 24 h C.S. for 12 h when compared to those treated for 6 h. Production of TGF-ß and IL-23 were not influenced by C.S. In conclusion, miR-23b is considered to be one of the mechanisms involved in tolerogenic DCs induction by C.S. in a time-dependent manner.


Subject(s)
Culture Media, Conditioned/pharmacology , Dendritic Cells/drug effects , Gene Expression Regulation/drug effects , Immunologic Factors/pharmacology , Mesenchymal Stem Cells/metabolism , MicroRNAs/biosynthesis , Adipogenesis , Animals , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Immunologic Factors/isolation & purification , Immunologic Factors/metabolism , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Osteogenesis , Specific Pathogen-Free Organisms , Spleen/cytology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...