Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
J Clin Invest ; 132(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35552271

ABSTRACT

The inability of CD8+ effector T cells (Teffs) to reach tumor cells is an important aspect of tumor resistance to cancer immunotherapy. The recruitment of these cells to the tumor microenvironment (TME) is regulated by integrins, a family of adhesion molecules that are expressed on T cells. Here, we show that 7HP349, a small-molecule activator of lymphocyte function-associated antigen-1 (LFA-1) and very late activation antigen-4 (VLA-4) integrin cell-adhesion receptors, facilitated the preferential localization of tumor-specific T cells to the tumor and improved antitumor response. 7HP349 monotherapy had modest effects on anti-programmed death 1-resistant (anti-PD-1-resistant) tumors, whereas combinatorial treatment with anti-cytotoxic T lymphocyte-associated protein 4 (anti-CTLA-4) increased CD8+ Teff intratumoral sequestration and synergized in cooperation with neutrophils in inducing cancer regression. 7HP349 intratumoral CD8+ Teff enrichment activity depended on CXCL12. We analyzed gene expression profiles using RNA from baseline and on treatment tumor samples of 14 melanoma patients. We identified baseline CXCL12 gene expression as possibly improving the likelihood or response to anti-CTLA-4 therapies. Our results provide a proof-of-principle demonstration that LFA-1 activation could convert a T cell-exclusionary TME to a T cell-enriched TME through mechanisms involving cooperation with innate immune cells.


Subject(s)
Lymphocyte Function-Associated Antigen-1 , Melanoma , CD8-Positive T-Lymphocytes , CTLA-4 Antigen , Humans , Immunotherapy/methods , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocytes, Tumor-Infiltrating , Melanoma/drug therapy , Melanoma/genetics , Programmed Cell Death 1 Receptor , T-Lymphocytes/metabolism , Tumor Microenvironment
2.
J Immunother Cancer ; 7(1): 136, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31118108

ABSTRACT

BACKGROUND: ONC201 is a small molecule antagonist of DRD2, a G protein-coupled receptor overexpressed in several malignancies, that has prolonged antitumor efficacy and immunomodulatory properties in preclinical models. The first-in-human trial of ONC201 previously established a recommended phase II dose (RP2D) of 625 mg once every three weeks. Here, we report the results of a phase I study that evaluated the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of weekly ONC201. METHODS: Patients ≥ 18 years old with an advanced solid tumor refractory to standard treatment were enrolled. Dose escalation proceeded with a 3 + 3 design from 375 mg to 625 mg of ONC201. One cycle, also the dose-limiting toxicity (DLT) window, was 21 days. The primary endpoint was to determine the RP2D of weekly ONC201, which was confirmed in an 11-patient dose expansion cohort. RESULTS: Twenty patients were enrolled: three at 375 mg and 17 at 625 mg of ONC201. The RP2D was defined as 625 mg with no DLT, treatment discontinuation, or dose modifications due to drug-related toxicity. PK profiles were consistent with every-three-week dosing and similar between the first and fourth dose. Serum prolactin and caspase-cleaved cytokeratin-18 induction were detected, along with intratumoral integrated stress response activation and infiltration of granzyme B+ Natural Killer cells. Induction of immune cytokines and effectors was higher in patients who received ONC201 once weekly versus once every three weeks. Stable disease of > 6 months was observed in several prostate and endometrial cancer patients. CONCLUSIONS: Weekly, oral ONC201 is well-tolerated and results in enhanced immunostimulatory activity that warrants further investigation. TRIAL REGISTRATION: NCT02250781 (Oral ONC201 in Treating Patients With Advanced Solid Tumors), NCT02324621 (Continuation of Oral ONC201 in Treating Patients With Advanced Solid Tumors).


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Receptors, Dopamine D2/immunology , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Humans , Middle Aged
3.
Sci Rep ; 8(1): 12519, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30131591

ABSTRACT

Bacillus Calmette-Guerin (BCG) is the only vaccine against TB and has limited protection efficacy, which wanes past adolescence. Multifunctional CD8+ T cells (IFN-γ+/TNF-α+/IL-2+) are associated with lower reactivation risk and enhanced control of active Mtb infection. Since boosting with BCG is contraindicated, booster vaccines that augment T cell immunity in the lungs of BCG-vaccinated individuals are urgently needed. We developed a vaccination strategy based on self-assembling peptide nanofibers presenting Mtb-specific CD8+ or CD4+ T cell epitopes that induce high frequency and antigen-specific effector memory T cells producing IFN-γ and IL-2. Intranasal immunization with peptide nanofibers was well tolerated in mice leading to increased antigen-specific CD8+ T cell population in the lungs. Co-assembled nanofibers of CD8+ T cell epitopes and toll-like receptor 2 (TLR2) agonists induced a 8-fold expansion in multifunctional CD8+ T cell populations in the lungs of vaccinated mice. Aerosol challenge with Mtb in BCG-primed and nanofiber-boosted mice provided an additional 0.5-log CFU reduction in lung bacterial load and indicating enhanced protection compared to BCG alone. Together, these data suggest that heterologous prime-boost with BCG and peptide nanofiber vaccines induces cell mediated immunity in the lung, reduces bacterial burden, and is a potentially safer alternative for boosting BCG-primed immunity.


Subject(s)
Antigens, Bacterial/chemistry , Epitopes, B-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/administration & dosage , Mycobacterium tuberculosis/immunology , Peptides/administration & dosage , T-Lymphocytes/immunology , Administration, Intranasal , Animals , BCG Vaccine/administration & dosage , BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Immunization, Secondary , Infusions, Parenteral , Interferon-gamma/metabolism , Interleukin-2/metabolism , Mice , Nanofibers , Peptides/chemical synthesis , Peptides/immunology
4.
Oncogene ; 37(27): 3672-3685, 2018 07.
Article in English | MEDLINE | ID: mdl-29622797

ABSTRACT

BMP receptor inhibitors induce death of cancer cells through the downregulation of antiapoptotic proteins XIAP, pTAK1, and Id1-Id3. However, the current most potent BMP receptor inhibitor, DMH2, does not downregulate BMP signaling in vivo because of metabolic instability and poor pharmacokinetics. Here we identified the site of metabolic instability of DMH2 and designed a novel BMP receptor inhibitor, JL5. We show that JL5 has a greater volume of distribution and suppresses the expression of Id1 and pTak1 in tumor xenografts. Moreover, we demonstrate JL5-induced tumor cell death and tumor regression in xenograft mouse models without immune cells and humanized with adoptively transferred human immune cells. In humanized mice, JL5 additionally induces the infiltration of immune cells within the tumor microenvironment. Our studies show that the BMP signaling pathway is targetable in vivo and BMP receptor inhibitors can be developed as a therapeutic to treat cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolones/pharmacology , Tumor Microenvironment/drug effects , A549 Cells , Adoptive Transfer , Animals , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inhibitor of Differentiation Protein 1/biosynthesis , Lung Neoplasms/pathology , MAP Kinase Kinase Kinases/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyrazoles/therapeutic use , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Quinolones/chemistry , Quinolones/therapeutic use , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
J Clin Invest ; 128(6): 2325-2338, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29533922

ABSTRACT

ONC201 is a first-in-class, orally active antitumor agent that upregulates cytotoxic TRAIL pathway signaling in cancer cells. ONC201 has demonstrated safety and preliminary efficacy in a first-in-human trial in which patients were dosed every 3 weeks. We hypothesized that dose intensification of ONC201 may impact antitumor efficacy. We discovered that ONC201 exerts dose- and schedule-dependent effects on tumor progression and cell death signaling in vivo. With dose intensification, we note a potent anti-metastasis effect and inhibition of cancer cell migration and invasion. Our preclinical results prompted a change in ONC201 dosing in all open clinical trials. We observed accumulation of activated NK+ and CD3+ cells within ONC201-treated tumors and that NK cell depletion inhibits ONC201 efficacy in vivo, including against TRAIL/ONC201-resistant Bax-/- tumors. Immunocompetent NCR1-GFP mice, in which NK cells express GFP, demonstrated GFP+ NK cell infiltration of syngeneic MC38 colorectal tumors. Activation of primary human NK cells and increased degranulation occurred in response to ONC201. Coculture experiments identified a role for TRAIL in human NK-mediated antitumor cytotoxicity. Preclinical results indicate the potential utility for ONC201 plus anti-PD-1 therapy. We observed an increase in activated TRAIL-secreting NK cells in the peripheral blood of patients after ONC201 treatment. The results offer what we believe to be a unique pathway of immune stimulation for cancer therapy.


Subject(s)
Cell Proliferation/drug effects , Colorectal Neoplasms , Heterocyclic Compounds, 4 or More Rings/pharmacology , Killer Cells, Natural/immunology , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/immunology , Animals , Cell Death/drug effects , Cell Death/immunology , Cell Survival/drug effects , Cell Survival/immunology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Female , HCT116 Cells , Humans , Imidazoles , Killer Cells, Natural/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Pyridines , Pyrimidines , Signal Transduction/drug effects , Signal Transduction/immunology , Xenograft Model Antitumor Assays
6.
Nanomedicine (Lond) ; 12(23): 2693-2706, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29098928

ABSTRACT

The major goal of immunity is maintaining host survival. Toward this, immune cells recognize and eliminate targets that pose a danger. Primarily, these are external invaders (pathogens) and internal invaders (cancers). Their recognition relies on distinguishing foreign components (antigens) from self-antigens. Since cancer cells are the host's own cells that are harmfully altered, they are difficult to distinguish from normal self. Furthermore, the antigens least resembling the host are often sequestered in parts of the tumor least accessible to immune responses. Therefore, to sufficiently boost immunity, these tumor antigens must be exposed to the immune system. Toward this, nanoparticles provide an innovating means of tumor antigen presentation and are destined to become an integral part of cancer immunotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Cancer Vaccines/therapeutic use , Nanoparticles/chemistry , Neoplasms/therapy , Animals , Antigen Presentation , Autoantigens/metabolism , Drug Delivery Systems/methods , Humans , Immunotherapy , Immunotherapy, Active , Molecular Targeted Therapy , Neoplasms/immunology , Surface Properties
7.
ACS Biomater Sci Eng ; 3(2): 126-143, 2017 Feb 13.
Article in English | MEDLINE | ID: mdl-33450791

ABSTRACT

Natural and synthetic biomaterials are increasingly being used for the development of vaccines and immunotherapies as alternatives to traditional live-attenuated formulations due to their improved safety profiles and no risk of reversion to virulence. Polymeric materials in particular enjoy attention due to the ease of fabrication, control over physicochemical properties, and their wide range of immunogenicity. While the majority of studies focus on inducing protective antibody responses, in recent years, materials-based strategies for the delivery of antigens and immunomodulators to improve CD8+T cell immunity against infectious and non-infectious diseases have gained momentum. Notably, platforms based on polymeric nanoparticles, liposomes, micelles, virus-like particles, self-assembling peptides and peptidomimetics, and multilayer thin films show considerable promise in preclinical studies. In this Review, we first introduce the concepts of CD8+T cell activation, effector and memory functions, and cytotoxic activity, followed by vaccine design for eliciting robust and protective long-lived CD8+T cell immunity. We then discuss different materials-based vaccines developed in the past decade to elicit CD8+T cell responses based on molecular composition or fabrication methods and conclude with a summary and glimpse at the future trends in this area.

8.
Vaccine ; 34(26): 2976-2981, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27026149

ABSTRACT

Chikungunya virus (CHIKV) is an arthritogenic alphavirus that during the last decade has significantly expanded its geographical range and caused large outbreaks of human disease around the world. Although mortality rates associated with CHIKV outbreaks are low, acute and chronic illnesses caused by CHIKV represent a significant burden of disease largely affecting low and middle income countries. This report summarizes the current status of vaccine development for CHIKV.


Subject(s)
Chikungunya Fever/prevention & control , Viral Vaccines/therapeutic use , Animals , Biomedical Research/trends , Chikungunya virus , Clinical Trials as Topic , Disease Models, Animal , Humans
9.
FASEB J ; 30(6): 2171-86, 2016 06.
Article in English | MEDLINE | ID: mdl-26917740

ABSTRACT

Recent data shows that fibroblast growth factor 14 (FGF14) binds to and controls the function of the voltage-gated sodium (Nav) channel with phenotypic outcomes on neuronal excitability. Mutations in the FGF14 gene in humans have been associated with brain disorders that are partially recapitulated in Fgf14(-/-) mice. Thus, signaling pathways that modulate the FGF14:Nav channel interaction may be important therapeutic targets. Bioluminescence-based screening of small molecule modulators of the FGF14:Nav1.6 complex identified 4,5,6,7 -: tetrabromobenzotriazole (TBB), a potent casein kinase 2 (CK2) inhibitor, as a strong suppressor of FGF14:Nav1.6 interaction. Inhibition of CK2 through TBB reduces the interaction of FGF14 with Nav1.6 and Nav1.2 channels. Mass spectrometry confirmed direct phosphorylation of FGF14 by CK2 at S228 and S230, and mutation to alanine at these sites modified FGF14 modulation of Nav1.6-mediated currents. In 1 d in vitro hippocampal neurons, TBB induced a reduction in FGF14 expression, a decrease in transient Na(+) current amplitude, and a hyperpolarizing shift in the voltage dependence of Nav channel steady-state inactivation. In mature neurons, TBB reduces the axodendritic polarity of FGF14. In cornu ammonis area 1 hippocampal slices from wild-type mice, TBB impairs neuronal excitability by increasing action potential threshold and lowering firing frequency. Importantly, these changes in excitability are recapitulated in Fgf14(-/-) mice, and deletion of Fgf14 occludes TBB-dependent phenotypes observed in wild-type mice. These results suggest that a CK2-FGF14 axis may regulate Nav channels and neuronal excitability.-Hsu, W.-C. J., Scala, F., Nenov, M. N., Wildburger, N. C., Elferink, H., Singh, A. K., Chesson, C. B., Buzhdygan, T., Sohail, M., Shavkunov, A. S., Panova, N. I., Nilsson, C. L., Rudra, J. S., Lichti, C. F., Laezza, F. CK2 activity is required for the interaction of FGF14 with voltage-gated sodium channels and neuronal excitability.


Subject(s)
Casein Kinase II/metabolism , Fibroblast Growth Factors/metabolism , Neurons/physiology , Voltage-Gated Sodium Channels/physiology , Animals , Casein Kinase II/genetics , Female , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Enzymologic , HEK293 Cells , Hippocampus/cytology , Hippocampus/physiology , Humans , Male , Mice , Mice, Knockout , Patch-Clamp Techniques
10.
J Mater Chem B ; 4(9): 1640-1649, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-32263017

ABSTRACT

To combat mucosal pathogens that cause gastrointestinal (GI) infections, local mucosal immunity is required which is best achieved through oral vaccination. Oral delivery of vaccines is also a safe and convenient alternative to injected vaccines due to its non-invasive nature and high compliance rate for all ages. However, the lack of effective and safe mucosal adjuvants, the selective permeability of the mucus barrier, and the harsh GI environment continue to pose a significant challenge for oral vaccine development. Microparticle-based strategies are attractive for oral vaccination due to their ability to efficiently penetrate the mucus barrier and have the added advantage of protecting the antigen in the harsh gastric environment. In this work, self-adjuvanting peptide nanofiber-CaCO3 composite microparticles were prepared and investigated for oral vaccine delivery. Compared to polymeric microparticles, inorganic CaCO3 microparticles have unique advantages due to the biocompatibility of CaCO3 as a natural mineral, mild preparation conditions, and its porous structure that is suitable for loading other materials. Particle size distribution, nanofiber loading efficiency, morphology, and degradation in simulated gastric fluid were characterized. The composite microparticles were efficient at penetrating the mucus barrier and were localized to immune inductive sites and elicited the production of mucosal antibody responses, particularly the protective IgA isotype following oral administration. The magnitude of the mucosal immune response was comparable to the gold-standard adjuvant cholera toxin B (CTB). Our results indicate that OVA-KFE8/CaCO3 composite microparticles are efficient self-adjuvanting oral vaccine delivery vehicles for induction of mucosal antibody responses.

11.
ACS Biomater Sci Eng ; 1(7): 601-609, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-33434976

ABSTRACT

d-Amino acid analogs of peptides and proteins are attractive for applications in biotechnology and medicine due to their reduced proteolytic sensitivity. Here, we report that self-assembling peptide nanofibers composed of d-amino acids act as immune adjuvants, and investigate their ability to induce antibody responses in comparison to their l-amino acid counterparts. The model antigenic peptide OVA (chicken egg ovalbumin aa 323-339) from chicken egg ovalbumin, known to elicit antibody responses in mice, was linked to an l- or d-amino acid self-assembling peptide domain to generate enantiomeric nanofibers displaying the same epitope. The chiral nature of the fusion peptides was confirmed by circular dichrosim spectroscopy and transmission electron microscopy studies indicated that OVA-bearing enantiomers self-assembled into nanofibers with similar morphologies. In mice, d-amino acid peptide nanofibers displaying OVA elicited stronger antibody responses, equivalent levels of CD4+ T cell responses, and long-term antigen-presentation in vivo compared to l-amino acid nanofibers. Our findings indicate that self-assembling peptides composed of d-amino acids are strong immune adjuvants and that biomaterial stereochemistry can be used as a design tool to program adaptive immune responses for vaccine development.

12.
Vaccine ; 32(10): 1174-80, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24308959

ABSTRACT

Vaccines that elicit robust CD8⁺ T cell responses are desirable for protection against infectious diseases and cancers. However, most vaccine adjuvants fail to elicit robust CD8⁺ T cell responses without inflammation and associated toxicity. We recently reported that self-assembling peptides that form nanofibers in physiological buffers elicited strong adjuvant-free and antigen-specific antibody responses in mice. However, whether or not such nanofibers likewise can elicit strong CD8⁺ T cell responses is unknown. Here, we demonstrate that the self-assembling peptide Q11 conjugated to a CD8⁺ T cell epitope of ovalbumin (Q11-OVA), elicits strong antigen-specific primary and recall responses, and in a vaccination regimen protects against subsequent infection. Importantly, we show that these antigenic peptide nanofibers do not persist as an inflammatory antigen depot at the injection site. Our results demonstrate for the first time that self-assembling peptides may be useful as carriers for vaccines where CD8⁺ T cell-mediated protection is needed.


Subject(s)
Adjuvants, Immunologic , CD8-Positive T-Lymphocytes/immunology , Nanofibers , Peptides/immunology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Ovalbumin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL