Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
EMBO J ; 37(23)2018 12 03.
Article in English | MEDLINE | ID: mdl-30322894

ABSTRACT

Metabolic reprogramming has been described in rapidly growing tumors, which are thought to mostly contain fast-cycling cells (FCCs) that have impaired mitochondrial function and rely on aerobic glycolysis. Here, we characterize the metabolic landscape of glioblastoma (GBM) and explore metabolic specificities as targetable vulnerabilities. Our studies highlight the metabolic heterogeneity in GBM, in which FCCs harness aerobic glycolysis, and slow-cycling cells (SCCs) preferentially utilize mitochondrial oxidative phosphorylation for their functions. SCCs display enhanced invasion and chemoresistance, suggesting their important role in tumor recurrence. SCCs also demonstrate increased lipid contents that are specifically metabolized under glucose-deprived conditions. Fatty acid transport in SCCs is targetable by pharmacological inhibition or genomic deletion of FABP7, both of which sensitize SCCs to metabolic stress. Furthermore, FABP7 inhibition, whether alone or in combination with glycolysis inhibition, leads to overall increased survival. Our studies reveal the existence of GBM cell subpopulations with distinct metabolic requirements and suggest that FABP7 is central to lipid metabolism in SCCs and that targeting FABP7-related metabolic pathways is a viable therapeutic strategy.


Subject(s)
Drug Resistance, Neoplasm , Fatty Acids/metabolism , Glioblastoma/metabolism , Glycolysis , Mitochondria/metabolism , Oxidative Phosphorylation , Animals , Cell Line, Tumor , Fatty Acid-Binding Protein 7/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/pathology , Neoplasm Proteins/metabolism , Tumor Suppressor Proteins/metabolism
2.
J Neurooncol ; 151(1): 29-39, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32757093

ABSTRACT

INTRODUCTION: Brain tumors remain especially challenging to treat due to the presence of the blood-brain barrier. The unique biophysical properties of nanomaterials enable access to the tumor environment with minimally invasive injection methods such as intranasal and systemic delivery. METHODS: In this review, we will discuss approaches taken in NP delivery to brain tumors in preclinical neuro-oncology studies and ongoing clinical studies. RESULTS: Despite recent development of many promising nanoparticle systems to modulate immunologic function in the preclinical realm, clinical work with nanoparticles in malignant brain tumors has largely focused on imaging, chemotherapy, thermotherapy and radiation. CONCLUSION: Review of early preclinical studies and clinical trials provides foundational safety, feasibility and toxicology data that can usher a new wave of nanotherapeutics in application of immunotherapy and translational oncology for patients with brain tumors.


Subject(s)
Brain Neoplasms , Nanoparticles , Adjuvants, Immunologic/therapeutic use , Blood-Brain Barrier , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Drug Delivery Systems , Humans , Immunologic Factors/therapeutic use
3.
Mol Ther ; 27(4): 837-849, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30448196

ABSTRACT

With the presence of the blood-brain barrier (BBB), successful immunotherapeutic drug delivery to CNS malignancies remains a challenge. Immunomodulatory agents, such as cytokines, can reprogram the intratumoral microenvironment; however, systemic cytokine delivery has limited access to the CNS. To bypass the limitations of systemically administered cytokines, we investigated if RNA-modified TĀ cells could deliver macromolecules directly to brain tumors. The abilities of TĀ cells to cross the BBB and mediate direct cytotoxic killing of intracranial tumors make them an attractive tool as biological carriers. Using TĀ cell mRNA electroporation, we demonstrated that activated TĀ cells can be modified to secrete granulocyte macrophage colony-stimulating factor (GM-CSF) protein while retaining their inherent effector functions inĀ vitro. GM-CSF RNA-modified TĀ cells effectively delivered GM-CSF to intracranial tumors inĀ vivo and significantly extended overall survival in an orthotopic treatment model. Importantly, GM-CSF RNA-modified TĀ cells demonstrated superior anti-tumor efficacy as compared to unmodified TĀ cells alone or in combination with systemic administration of recombinant GM-CSF. Anti-tumor effects were associated with increased IFN-ƎĀ³ secretion locally within the tumor microenvironment and systemic antigen-specific TĀ cell expansion. These findings demonstrate that RNA-modified TĀ cells may serve as a versatile platform for the effective delivery of biological agents to CNS tumors.


Subject(s)
Brain Neoplasms/therapy , Cell- and Tissue-Based Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Immunotherapy, Adoptive/methods , RNA/genetics , T-Lymphocytes/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Survival/genetics , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Green Fluorescent Proteins/metabolism , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Transfection/methods , Tumor Microenvironment/genetics
4.
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
5.
Int J Mol Sci ; 19(10)2018 Sep 23.
Article in English | MEDLINE | ID: mdl-30249040

ABSTRACT

Cancer vaccines may be harnessed to incite immunity against poorly immunogenic tumors, however they have failed in therapeutic settings. Poor antigenicity coupled with systemic and intratumoral immune suppression have been significant drawbacks. RNA encoding for tumor associated or specific epitopes can serve as a more immunogenic and expeditious trigger of anti-tumor immunity. RNA stimulates innate immunity through toll like receptor stimulation producing type I interferon, and it mediates potent adaptive responses. Since RNA is inherently unstable, delivery systems have been developed to protect and deliver it to intended targets in vivo. In this review, we discuss liposomes as RNA delivery vehicles and their role as cancer vaccines.


Subject(s)
Cancer Vaccines/administration & dosage , Immunotherapy , Liposomes/administration & dosage , Neoplasms/immunology , Neoplasms/therapy , RNA/administration & dosage , Animals , Cancer Vaccines/chemistry , Drug Delivery Systems , Humans , Liposomes/chemistry , RNA/chemistry
6.
Cancer Immunol Immunother ; 65(2): 205-11, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26759007

ABSTRACT

Regulatory B cells that secrete IL-10 (IL-10(+) Bregs) represent a suppressive subset of the B cell compartment with prominent anti-inflammatory capacity, capable of suppressing cellular and humoral responses to cancer and vaccines. B lymphocyte stimulator (BLyS) is a key regulatory molecule in IL-10(+) Breg biology with tightly controlled serum levels. However, BLyS levels can be drastically altered upon chemotherapeutic intervention. We have previously shown that serum BLyS levels are elevated, and directly associated, with increased antigen-specific antibody titers in patients with glioblastoma (GBM) undergoing lymphodepletive temozolomide chemotherapy and vaccination. In this study, we examined corresponding IL-10(+) Breg responses within this patient population and demonstrate that the IL-10(+) Breg compartment remains constant before and after administration of the vaccine, despite elevated BLyS levels in circulation. IL-10(+) Breg frequencies were not associated with serum BLyS levels, and ex vivo stimulation with a physiologically relevant concentration of BLyS did not increase IL-10(+) Breg frequency. However, BLyS stimulation did increase the frequency of the overall B cell compartment and promoted B cell proliferation upon B cell receptor engagement. Therefore, using BLyS as an adjuvant with therapeutic peptide vaccination could promote humoral immunity with no increase in immunosuppressive IL-10(+) Bregs. These results have implications for modulating humoral responses in human peptide vaccine trials in patients with GBM.


Subject(s)
B-Cell Activating Factor/blood , B-Lymphocytes, Regulatory/immunology , Glioblastoma/blood , Glioblastoma/immunology , Lymphocyte Count , Antibodies/blood , Antibodies/immunology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , B-Lymphocytes, Regulatory/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Glioblastoma/mortality , Glioblastoma/therapy , Humans , Immunotherapy/methods , Interleukin-10/metabolism , Lymphocyte Activation/immunology , Temozolomide , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
7.
Pediatr Blood Cancer ; 63(10): 1856-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27273469

ABSTRACT

Patients with common variable immunodeficiency (CVID) have a higher incidence of autoimmune disease, which may mark the disease onset; however, anemia secondary to pure red cell aplasia is an uncommon presenting feature. Here, we describe a case of CVID-like humoral immune deficiency in a child who initially presented with red cell aplasia and ultimately developed progressive bone marrow failure. Although bone marrow transplantation (BMT) has been associated with high mortality in CVID, our patient was successfully treated with a matched sibling BMT and engrafted with >98% donor chimerism and the development of normal antibody titers to diphtheria and tetanus toxoids.


Subject(s)
Bone Marrow Transplantation , Common Variable Immunodeficiency/therapy , Red-Cell Aplasia, Pure/complications , Child , Common Variable Immunodeficiency/immunology , Humans , Immunity, Humoral , Male , Red-Cell Aplasia, Pure/immunology
8.
Cancer Immunol Immunother ; 64(4): 419-27, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25555571

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive malignancy associated with profound host immunosuppression mediated in part by FoxP3 expressing regulatory CD4+ T lymphocytes (Tregs) that down-regulate anti-tumor immunity. In order to assess whether FoxP3 was an independent driver differentially expressed in primary versus recurrent GBMs, we stained resected primary and recurrent GBM tumors for CD3, CD4, CD8 and FoxP3 expression using standard immunohistochemistry. Slides were scanned with a high-resolution scanner (ScanScope CS; Aperio), and image analysis software (Aperio ScanScope) was used to enumerate lymphocyte subpopulations allowing for high-throughput analysis and bypassing manual selection bias. As shown in previous studies, enumeration of individual lymphocyte populations did not correlate with clinical outcomes in patients with GBM. However, the CD4+ to regulatory FoxP3+ T cell ratio was diminished in recurrent disease, and increased CD3 and CD8+ to regulatory T cell ratios showed a positive correlation with survival outcomes in primary GBM. These results suggest that while absolute numbers of tumor infiltrating lymphocytes may not be informative for predicting clinical outcomes in patients with GBM, the effective balance of CD3, CD4 and CD8+ T cells to immunosuppressive FoxP3+ regulatory cells may influence clinical outcomes in this patient population.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/mortality , Forkhead Transcription Factors/metabolism , Glioblastoma/mortality , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Recurrence, Local/mortality , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Brain Neoplasms/immunology , Brain Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Follow-Up Studies , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Survival Rate
9.
Nat Rev Clin Oncol ; 21(7): 489-500, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38760500

ABSTRACT

mRNA vaccines have been revolutionary in terms of their rapid development and prevention of SARS-CoV-2 infections during the COVID-19 pandemic, and this technology has considerable potential for application to the treatment of cancer. Compared with traditional cancer vaccines based on proteins or peptides, mRNA vaccines reconcile the needs for both personalization and commercialization in a manner that is unique to each patient but not beholden to their HLA haplotype. A further advantage of mRNA vaccines is the availability of engineering strategies to improve their stability while retaining immunogenicity, enabling the induction of complementary innate and adaptive immune responses. Thus far, no mRNA-based cancer vaccines have received regulatory approval, although several phase I-II trials have yielded promising results, including in historically poorly immunogenic tumours. Furthermore, many earlyĀ phase trials testing a wide range of vaccine designs are currently ongoing. In this Review, we describe the advantages of cancer mRNA vaccines and advances in clinical trials using both cell-based and nanoparticle-based delivery methods, with discussions of future combinations and iterations that might optimize the activity of these agents.


Subject(s)
COVID-19 , Cancer Vaccines , Neoplasms , mRNA Vaccines , Humans , Cancer Vaccines/therapeutic use , Cancer Vaccines/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/prevention & control , Neoplasms/genetics , COVID-19/prevention & control , COVID-19/immunology , SARS-CoV-2/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use , RNA, Messenger/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/immunology , Clinical Trials as Topic
10.
J Hematol Oncol ; 17(1): 4, 2024 01 08.
Article in English | MEDLINE | ID: mdl-38191498

ABSTRACT

Myelodysplastic neoplasms (MDS) define clonal hematopoietic malignancies characterized by heterogeneous mutational and clinical spectra typically seen in the elderly. Curative treatment entails allogeneic hematopoietic stem cell transplant, which is often not a feasible option due to older age and significant comorbidities. Immunotherapy has the cytotoxic capacity to elicit tumor-specific killing with long-term immunological memory. While a number of platforms have emerged, therapeutic vaccination presents as an appealing strategy for MDS given its promising safety profile and amenability for commercialization. Several preclinical and clinical trials have investigated the efficacy of vaccines in MDS; these include peptide vaccines targeting tumor antigens, whole cell-based vaccines and dendritic cell-based vaccines. These therapeutic vaccines have shown acceptable safety profiles, but consistent clinical responses remain elusive despite robust immunological reactions. Combining vaccines with immunotherapeutic agents holds promise and requires further investigation. Herein, we highlight therapeutic vaccine trials while reviewing challenges and future directions of successful vaccination strategies in MDS.


Subject(s)
Hematologic Neoplasms , Myelodysplastic Syndromes , Vaccines , Aged , Humans , Myelodysplastic Syndromes/therapy , Immunotherapy , Vaccination
11.
Mol Ther Methods Clin Dev ; 32(1): 101192, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38327807

ABSTRACT

The COVID-19 pandemic has caused about seven million deaths worldwide. Preventative vaccines have been developed including Spike gp mRNA-based vaccines that provide protection to immunocompetent patients. However, patients with primary immunodeficiencies, patients with cancer, or hematopoietic stem cell transplant recipients are not able to mount robust immune responses against current vaccine approaches. We propose to target structural SARS-CoV-2 antigens (i.e., Spike gp, Membrane, Nucleocapsid, and Envelope) using circulating human antigen-presenting cells electroporated with full length SARS-CoV-2 structural protein-encoding mRNAs to activate and expand specific TĀ cells. Based on the Th1-type cytokine and cytolytic enzyme secretion upon antigen rechallenge, we were able to generate SARS-CoV-2 specific TĀ cells in up to 70% of unexposed unvaccinated healthy donors (HDs) after 3 subsequent stimulations and in 100% of recovered patients (RPs) after 2 stimulations. By means of SARS-CoV-2 specific TCRƟ repertoire analysis, TĀ cells specific to Spike gp-derived hypomutated regions were identified in HDs and RPs despite viral genomic evolution. Hence, we demonstrated that SARS-CoV-2 mRNA-loaded antigen-presenting cells are effective activating and expanding COVID19-specific TĀ cells. This approach represents an alternative to patients who are not able to mount adaptive immune responses to current COVID-19 vaccines with potential protection across new variants that have conserved genetic regions.

12.
Cancer Immunol Immunother ; 62(6): 983-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23591978

ABSTRACT

B lymphocyte stimulator (BLyS) is a cytokine involved in differentiation and survival of follicular B cells along with humoral response potentiation. Lymphopenia is known to precipitate dramatic elevation in serum BLyS; however, the use of this effect to enhance humoral responses following vaccination has not been evaluated. We evaluated BLyS serum levels and antigen-specific antibody titers in 8 patients undergoing therapeutic temozolomide (TMZ)-induced lymphopenia, with concomitant vaccine against a tumor-specific mutation in the epidermal growth factor receptor (EGFRvIII). Our studies demonstrate that TMZ-induced lymphopenia corresponded with spikes in serum BLyS that directly preceded the induction of anti-EGFRvIII antigen-specific antibody titers, in some cases as high as 1:2,000,000. Our data are the first clinical observation of BLyS serum elevation and greatly enhanced humoral immune responses as a consequence of chemotherapy-induced lymphopenia. These observations should be considered for the development of future vaccination strategies in the setting of malignancy.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , B-Cell Activating Factor/blood , Cancer Vaccines/immunology , Dacarbazine/analogs & derivatives , Glioblastoma/immunology , Lymphocyte Depletion , Antibodies/blood , Antibody Specificity/immunology , Antineoplastic Agents, Alkylating/adverse effects , Dacarbazine/adverse effects , Dacarbazine/therapeutic use , Glioblastoma/therapy , Humans , Lymphopenia/blood , Lymphopenia/chemically induced , Temozolomide
13.
Acta Biomater ; 172: 466-479, 2023 12.
Article in English | MEDLINE | ID: mdl-37788737

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has demonstrated remarkable success as an immunotherapy for hematological malignancies, and its potential for treating solid tumors is an active area of research. However, limited trafficking and mobility of T cells within the tumor microenvironment (TME) present challenges for CAR T cell therapy in solid tumors. To gain a better understanding of CAR T cell function in solid tumors, we subjected CD70-specific CAR T cells to a challenge by evaluating their immune trafficking and infiltration through a confined 3D microchannel network in a bio-conjugated liquid-like solid (LLS) medium. Our results demonstrated successful CAR T cell migration and anti-tumor activity against CD70-expressing glioblastoma and osteosarcoma tumors. Through comprehensive analysis of cytokines and chemokines, combined with in situ imaging, we elucidated that immune recruitment occurred via chemotaxis, and the effector-to-target ratio plays an important role in overall antitumor function. Furthermore, through single-cell collection and transcriptomic profiling, we identified differential gene expression among the immune subpopulations. Our findings provide valuable insights into the complex dynamics of CAR T cell function in solid tumors, informing future research and development in this promising cancer treatment approach. STATEMENT OF SIGNIFICANCE: The use of specialized immune cells named CAR T cells to combat cancers has demonstrated remarkable success against blood cancers. However, this success is not replicated in solid tumors, such as brain or bone cancers, mainly due to the physical barriers of these solid tumors. Currently, preclinical technologies do not allow for reliable evaluation of tumor-immune cell interactions. To better study these specialized CAR T cells, we have developed an innovative in vitro three-dimensional model that promises to dissect the interactions between tumors and CAR T cells at the single-cell level. Our findings provide valuable insights into the complex dynamics of CAR T cell function in solid tumors, informing future research and development in this promising cancer treatment approach.


Subject(s)
Bone Neoplasms , Neoplasms , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/genetics , T-Lymphocytes , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Antigens, Neoplasm , Neoplasms/metabolism , Bone Neoplasms/metabolism , Cell Communication , Tumor Microenvironment
14.
bioRxiv ; 2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36865164

ABSTRACT

Cancer immunotherapy offers lifesaving treatments for cancers, but the lack of reliable preclinical models that could enable the mechanistic studies of tumor-immune interactions hampers the identification of new therapeutic strategies. We hypothesized 3D confined microchannels, formed by interstitial space between bio-conjugated liquid-like solids (LLS), enable CAR T dynamic locomotion within an immunosuppressive TME to carry out anti-tumor function. Murine CD70-specific CAR T cells cocultured with the CD70-expressing glioblastoma and osteosarcoma demonstrated efficient trafficking, infiltration, and killing of cancer cells. The anti-tumor activity was clearly captured via longterm in situ imaging and supported by upregulation of cytokines and chemokines including IFNg, CXCL9, CXCL10, CCL2, CCL3, and CCL4. Interestingly, target cancer cells, upon an immune attack, initiated an "immune escape" response by frantically invading the surrounding microenvironment. This phenomenon however was not observed for the wild-type tumor samples which remained intact and produced no relevant cytokine response. Single cells collection and transcriptomic profiling of CAR T cells at regions of interest revealed feasibility of identifying differential gene expression amongst the immune subpopulations. Complimentary 3D in vitro platforms are necessary to uncover cancer immune biology mechanisms, as emphasized by the significant roles of the TME and its heterogeneity.

15.
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.

16.
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.

17.
Neuromolecular Med ; 24(1): 8-12, 2022 03.
Article in English | MEDLINE | ID: mdl-34101090

ABSTRACT

Glioblastoma (GBM) is the most common primary brain tumor in adults and is universally lethal with a median survival of less than two years with standard therapy. RNA-based immunotherapies have significant potential to establish a durable treatment response for malignant brain tumors including GBM. RNA offers clear advantages over antigen-focused approaches but cannot often be directly administered due to biological instability. This review will focus on utilization of RNA dendritic cell vaccines and RNA nanoparticle therapies in the treatment of GBM. RNA-pulsed dendritic cell vaccines have been shown to be safe in a small phase I clinical trial and RNA-loaded nanoparticle vaccines will soon be underway in GBM patients (NCT04573140).


Subject(s)
Brain Neoplasms , Cancer Vaccines , Glioblastoma , Adult , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cancer Vaccines/therapeutic use , Clinical Trials, Phase I as Topic , Dendritic Cells/pathology , Glioblastoma/genetics , Glioblastoma/therapy , Humans , Immunotherapy , RNA/genetics , RNA/therapeutic use
18.
Cells ; 11(12)2022 06 20.
Article in English | MEDLINE | ID: mdl-35741103

ABSTRACT

The promising outcomes of chimeric antigen receptor (CAR) T cell therapy in hematologic malignancies potentiates its capability in the fight against many cancers. Nevertheless, this immunotherapy modality needs significant improvements for the treatment of solid tumors. Researchers have incrementally identified limitations and constantly pursued better CAR designs. However, even if CAR T cells are armed with optimal killer functions, they must overcome and survive suppressive barriers imposed by the tumor microenvironment (TME). In this review, we will discuss in detail the important role of TME in CAR T cell trafficking and how the intrinsic barriers contribute to an immunosuppressive phenotype and cancer progression. It is of critical importance that preclinical models can closely recapitulate the in vivo TME to better predict CAR T activity. Animal models have contributed immensely to our understanding of human diseases, but the intensive care for the animals and unreliable representation of human biology suggest in vivo models cannot be the sole approach to CAR T cell therapy. On the other hand, in vitro models for CAR T cytotoxic assessment offer valuable insights to mechanistic studies at the single cell level, but they often lack in vivo complexities, inter-individual heterogeneity, or physiologically relevant spatial dimension. Understanding the advantages and limitations of preclinical models and their applications would enable more reliable prediction of better clinical outcomes.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Animals , Cell Movement , Immunotherapy, Adoptive/methods , Neoplasms/pathology , T-Lymphocytes , Tumor Microenvironment
19.
Nat Cancer ; 3(1): 11-24, 2022 01.
Article in English | MEDLINE | ID: mdl-35121998

ABSTRACT

Pediatric central nervous system tumors are the most common solid malignancies in childhood, and aggressive therapy often leads to long-term sequelae in survivors, making these tumors challenging to treat. Immunotherapy has revolutionized prospects for many cancer types in adults, but the intrinsic complexity of treating pediatric patients and the scarcity of clinical studies of children to inform effective approaches have hampered the development of effective immunotherapies in pediatric settings. Here, we review recent advances and ongoing challenges in pediatric brain cancer immunotherapy, as well as considerations for efficient clinical translation of efficacious immunotherapies into pediatric settings.


Subject(s)
Brain Neoplasms , Central Nervous System Neoplasms , Brain Neoplasms/therapy , Central Nervous System Neoplasms/therapy , Child , Humans , Immunologic Factors , Immunotherapy/adverse effects , Survivors
SELECTION OF CITATIONS
SEARCH DETAIL