Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 573
1.
J Nanobiotechnology ; 20(1): 525, 2022 Dec 10.
Article En | MEDLINE | ID: mdl-36496387

Glioblastoma (GBM) is the most aggressive brain tumor, which owns the characteristics of high recurrence, low survival rate and poor prognosis because of the existence of blood brain barrier (BBB) and complicated brain tumor microenvironment. Currently, immunotherapy has attracted much attention on account of favorable therapeutic effect. In this study, we designed a cRGD-modified cancer cell membrane (CM) coated calcium carbonate nanoparticle to deliver interleukin-12 messenger RNA (IL-12 mRNA@cRGD-CM-CaCO3 NPs). The cRGD-modified CM as the shell can endow the nanoparticles with BBB crossing and tumor homing/homotypic targeting effect in the brain tumor microenvironment. IL-12 mRNA-loaded calcium carbonate nanoparticles as the core allow synergistic immunotherapy of necroptosis-induced immune response and IL-12 mRNA transfection under ultrasound irradiation. The as-prepared biomimetic nanoparticles showed superior target and immunotherapeutic outcomes, suggesting that this biomimetic nanoplatform provides a feasible strategy for promoting BBB-penetrating and antitumor immunity.


Brain Neoplasms , Glioblastoma , Nanoparticles , Humans , Brain Neoplasms/drug therapy , Calcium Carbonate , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/metabolism , Interleukin-12/administration & dosage , Interleukin-12/therapeutic use , RNA, Messenger , Tumor Microenvironment
2.
Nat Commun ; 12(1): 5151, 2021 08 26.
Article En | MEDLINE | ID: mdl-34446712

Poorly inflamed carcinomas do not respond well to immune checkpoint blockade. Converting the tumour microenvironment into a functionally inflamed immune hub would extend the clinical benefit of immune therapy to a larger proportion of cancer patients. Here we show, by using comprehensive single-cell transcriptome, proteome, and immune cell analysis, that Entinostat, a class I histone deacetylase inhibitor, facilitates accumulation of the necrosis-targeted recombinant murine immune-cytokine, NHS-rmIL12, in experimental mouse colon carcinomas and poorly immunogenic breast tumours. This combination therapy reprograms the tumour innate and adaptive immune milieu to an inflamed landscape, where the concerted action of highly functional CD8+ T cells and activated neutrophils drive macrophage M1-like polarization, leading to complete tumour eradication in 41.7%-100% of cases. Biomarker signature of favourable overall survival in multiple human tumor types shows close resemblance to the immune pattern generated by Entinostat/NHS-rmIL12 combination therapy. Collectively, these findings provide a rationale for combining NHS-IL12 with Entinostat in the clinical setting.


Benzamides/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Immunoglobulin G/administration & dosage , Interleukin-12/administration & dosage , Pyridines/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Adaptive Immunity/drug effects , Animals , Breast Neoplasms/mortality , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Colonic Neoplasms/mortality , Drug Therapy, Combination , Female , Humans , Immunity, Innate/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Tumor Microenvironment/drug effects
3.
ACS Appl Mater Interfaces ; 13(10): 12454-12462, 2021 Mar 17.
Article En | MEDLINE | ID: mdl-33683872

Infection and delayed wound healing are two major serious complications related to traumatic injuries and cause a significant burden to patients and society. Most currently available drug delivery materials typically carry a single drug, lack protection from drug loading, and face challenges in on-demand and precisely controlled drug release. Here, we report a flower (Cirsium arvense)-inspired capsule-integrated multilayer nanofilm (FICIF), synthesized using a layer-by-layer self-assembly, for programmed multiple drug co-delivery for trauma (open fracture as an example) treatments. Our approach allows polypeptide multilayer nanofilms and innovative impregnated capsules to assemble hierarchical reservoirs with specific drug binding sites, shielding protection capability, and ordered packing structures. The resultant FICIF nanocarriers enable sustained and on-demand co-delivery of a unique immune-tuning cytokine (interleukin 12p70) and a growth factor (bone morphogenetic protein 2) in clinical use, resulting in extraordinary anti-infection (3 orders of magnitude improved bacterial killing) and bone regeneration (5 times enhanced bone healing) in treating infected rat femur fractures. The successful synthesis of these biomimetic high-performance delivery nanocoatings is expected to serve as a source of inspiration for the development of biomaterials for various clinical applications.


Anti-Bacterial Agents/administration & dosage , Bone Morphogenetic Protein 2/administration & dosage , Delayed-Action Preparations/chemistry , Interleukin-12/administration & dosage , Nanocapsules/chemistry , Peptides/chemistry , Animals , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Biomimetic Materials/chemistry , Bone Morphogenetic Protein 2/therapeutic use , Bone and Bones/injuries , Bone and Bones/microbiology , Cell Line , Cirsium/chemistry , Humans , Interleukin-12/therapeutic use , Rats
4.
Nat Commun ; 12(1): 444, 2021 01 19.
Article En | MEDLINE | ID: mdl-33469002

Glioblastoma multiforme (GBM) is the most common and aggressive form of primary brain cancer, for which effective therapies are urgently needed. Chimeric antigen receptor (CAR)-based immunotherapy represents a promising therapeutic approach, but it is often impeded by highly immunosuppressive tumor microenvironments (TME). Here, in an immunocompetent, orthotopic GBM mouse model, we show that CAR-T cells targeting tumor-specific epidermal growth factor receptor variant III (EGFRvIII) alone fail to control fully established tumors but, when combined with a single, locally delivered dose of IL-12, achieve durable anti-tumor responses. IL-12 not only boosts cytotoxicity of CAR-T cells, but also reshapes the TME, driving increased infiltration of proinflammatory CD4+ T cells, decreased numbers of regulatory T cells (Treg), and activation of the myeloid compartment. Importantly, the immunotherapy-enabling benefits of IL-12 are achieved with minimal systemic effects. Our findings thus show that local delivery of IL-12 may be an effective adjuvant for CAR-T cell therapy for GBM.


Brain Neoplasms/therapy , Glioblastoma/therapy , Immunoconjugates/administration & dosage , Immunotherapy, Adoptive/methods , Interleukin-12/administration & dosage , Animals , Brain/diagnostic imaging , Brain/immunology , Brain/pathology , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor/transplantation , Disease Models, Animal , ErbB Receptors/immunology , Female , Glioblastoma/diagnostic imaging , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Immunoconjugates/immunology , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/immunology , Injections, Intralesional/methods , Interleukin-12/immunology , Magnetic Resonance Imaging, Interventional , Mice , Receptors, Chimeric Antigen/immunology , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology
5.
Nanotechnology ; 32(9): 095107, 2021 Feb 26.
Article En | MEDLINE | ID: mdl-33238258

In the past two decades, protein drugs have evolved to become the most successful and important strategy in cancer therapy. However, systematical administration of protein drugs may cause serious side effects. In order to prepare a new promising hydrophilic drugs carrier, we constructed a PEGylated hyaluronic acid nanogel (NI-MAHA-PEG nanogel) with hypoxia and enzymatic responsiveness, which can selectively release hydrophilic drugs interleukin-12 (IL-12) on demand in a tumor microenvironment. We observed that release of IL-12 from nanogels by hypoxia-responsive stimulation, nanogels have anti-tumor effects on melanoma. Compared with physiological conditions, the IL-12 release rate has achieved remarkable growth under hypoxic conditions. Similarly, the drug release rate increased significantly with the addition of 500 U ml-1 hyaluronidase. We provide a novel strategy to allow efficient delivery, on-demand release, and enhanced access of proteins to hypoxic tumor regions. The rational design of this nanogels drug delivery system can further explore the use of various drugs to treat many cancers.


Hyaluronic Acid/chemistry , Interleukin-12/administration & dosage , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Animals , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Interleukin-12/chemistry , Interleukin-12/pharmacology , Mice , Nanogels , Polyethylene Glycols/chemistry , Xenograft Model Antitumor Assays
6.
Front Immunol ; 11: 575597, 2020.
Article En | MEDLINE | ID: mdl-33178203

Interleukin-12 (IL-12) is a potent, pro-inflammatory type 1 cytokine that has long been studied as a potential immunotherapy for cancer. Unfortunately, IL-12's remarkable antitumor efficacy in preclinical models has yet to be replicated in humans. Early clinical trials in the mid-1990's showed that systemic delivery of IL-12 incurred dose-limiting toxicities. Nevertheless, IL-12's pleiotropic activity, i.e., its ability to engage multiple effector mechanisms and reverse tumor-induced immunosuppression, continues to entice cancer researchers. The development of strategies which maximize IL-12 delivery to the tumor microenvironment while minimizing systemic exposure are of increasing interest. Diverse IL-12 delivery systems, from immunocytokine fusions to polymeric nanoparticles, have demonstrated robust antitumor immunity with reduced adverse events in preclinical studies. Several localized IL-12 delivery approaches have recently reached the clinical stage with several more at the precipice of translation. Taken together, localized delivery systems are supporting an IL-12 renaissance which may finally allow this potent cytokine to fulfill its considerable clinical potential. This review begins with a brief historical account of cytokine monotherapies and describes how IL-12 went from promising new cure to ostracized black sheep following multiple on-study deaths. The bulk of this comprehensive review focuses on developments in diverse localized delivery strategies for IL-12-based cancer immunotherapies. Advantages and limitations of different delivery technologies are highlighted. Finally, perspectives on how IL-12-based immunotherapies may be utilized for widespread clinical application in the very near future are offered.


Antineoplastic Agents/administration & dosage , Genetic Therapy , Immunotherapy , Interleukin-12/administration & dosage , Neoplasms/therapy , Animals , Antineoplastic Agents/adverse effects , Drug Carriers , Drug Compounding , Gene Transfer Techniques , Genetic Therapy/adverse effects , Genetic Vectors , Humans , Immunotherapy/adverse effects , Interleukin-12/adverse effects , Interleukin-12/genetics , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Treatment Outcome , Tumor Microenvironment
7.
MAbs ; 12(1): 1836713, 2020.
Article En | MEDLINE | ID: mdl-33136526

Antibody-based delivery of bioactive molecules represents a promising strategy for the improvement of cancer immunotherapy. Here, we describe the generation and characterization of R6N, a novel fully human antibody specific to the alternatively spliced domain D of Tenascin C, which is highly expressed in the stroma of primary tumors and metastasis. The R6N antibody recognized its cognate tumor-associated antigen with identical specificity in mouse and human specimens. Moreover, the antibody was able to selectively localize to solid tumors in vivo as evidenced by immunofluorescence-based biodistribution analysis. Encouraged by these results, we developed a novel fusion protein (termed mIL12-R6N) consisting of the murine interleukin 12 fused to the R6N antibody in homodimeric tandem single-chain variable fragment arrangement. mIL12-R6N exhibited potent antitumor activity in immunodeficient mice bearing SKRC52 renal cell carcinoma, as well as in immunocompetent mice bearing SMA-497 glioma. The experiments presented in this work provide a rationale for possible future applications for the R6N antibody for the treatment of cancer patients.


Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Interleukin-12/administration & dosage , Neoplasms, Experimental , Tenascin/antagonists & inhibitors , Alternative Splicing , Animals , Humans , Mice , Molecular Targeted Therapy/methods , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies
8.
Cancer Lett ; 492: 31-43, 2020 11 01.
Article En | MEDLINE | ID: mdl-32758617

Immunogenic chemotherapy has been shown to be effective against several cancer types. Here, we identified trametinib as an inducer of immunogenic cell death (ICD), and found that it exerts beneficial antitumor effects if used in combination with interleukin (IL)-12 in a Kras-mutant mouse model of spontaneous lung cancer. Tumor cells treated with trametinib showed the hallmarks of ICD, including cell-surface expression of calreticulin, release of high mobility group box 1 (HMGB1) from the nucleus, and activation of dendritic cells. Tumor-bearing mice treated with both trametinib and IL-12 showed increased infiltration and proliferation of T cells within the tumor, as well as increased effector function of NK cells and T cells, indicating that this therapeutic combination can improve the quality of the immune response. Taken together, our results provide a potential new therapeutic regimen for the treatment of KRAS-mutant lung adenocarcinomas.


Interleukin-12/administration & dosage , Lung Neoplasms/drug therapy , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Animals , Cell Line, Tumor , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Pyridones/administration & dosage , Pyrimidinones/administration & dosage , T-Lymphocytes/immunology
9.
Clin Cancer Res ; 26(23): 6284-6298, 2020 12 01.
Article En | MEDLINE | ID: mdl-32817076

PURPOSE: While immune checkpoint inhibitors such as anti-PD-L1 are rapidly becoming the standard of care in the treatment of many cancers, only a subset of treated patients have long-term responses. IL12 promotes antitumor immunity in mouse models; however, systemic recombinant IL12 had significant toxicity and limited efficacy in early clinical trials. EXPERIMENTAL DESIGN: We therefore designed a novel intratumoral IL12 mRNA therapy to promote local IL12 tumor production while mitigating systemic effects. RESULTS: A single intratumoral dose of mouse (m)IL12 mRNA induced IFNγ and CD8+ T-cell-dependent tumor regression in multiple syngeneic mouse models, and animals with a complete response demonstrated immunity to rechallenge. Antitumor activity of mIL12 mRNA did not require NK and NKT cells. mIL12 mRNA antitumor activity correlated with TH1 tumor microenvironment (TME) transformation. In a PD-L1 blockade monotherapy-resistant model, antitumor immunity induced by mIL12 mRNA was enhanced by anti-PD-L1. mIL12 mRNA also drove regression of uninjected distal lesions, and anti-PD-L1 potentiated this response. Importantly, intratumoral delivery of mRNA encoding membrane-tethered mIL12 also drove rejection of uninjected lesions with very limited circulating IL12p70, supporting the hypothesis that local IL12 could induce a systemic antitumor immune response against distal lesions. Furthermore, in ex vivo patient tumor slice cultures, human IL12 mRNA (MEDI1191) induced dose-dependent IL12 production, downstream IFNγ expression and TH1 gene expression. CONCLUSIONS: These data demonstrate the potential for intratumorally delivered IL12 mRNA to promote TH1 TME transformation and robust antitumor immunity.See related commentary by Cirella et al., p. 6080.


Colorectal Neoplasms/prevention & control , Interleukin-12/administration & dosage , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/prevention & control , RNA, Messenger/administration & dosage , Th1 Cells/immunology , Tumor Microenvironment/immunology , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Female , Humans , Interleukin-12/genetics , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , RNA, Messenger/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
J Immunother Cancer ; 8(1)2020 06.
Article En | MEDLINE | ID: mdl-32554612

BACKGROUND: While prophylactic human papillomavirus (HPV) vaccines will certainly reduce the incidence of HPV-associated cancers, these malignancies remain a major health issue. PDS0101 is a liposomal-based HPV therapeutic vaccine consisting of the immune activating cationic lipid R-DOTAP and HLA-unrestricted HPV16 peptides that has shown in vivo CD8+ T cell induction and safety in a phase I study. In this report, we have employed the PDS0101 vaccine with two immune modulators previously characterized in preclinical studies and which are currently in phase II clinical trials. Bintrafusp alfa (M7824) is a first-in-class bifunctional fusion protein composed of the extracellular domains of the transforming growth factor-ß receptor type II (TGFßRII) fused to a human IgG1 monoclonal antibody blocking programmed cell death protein-1 ligand (PDL1), designed both as a checkpoint inhibitor and to bring the TGFßRII 'trap' to the tumor microenvironment (TME). NHS-interleukin-12 (NHS-IL12) is a tumor targeting immunocytokine designed to bring IL-12 to the TME and thus enhance the inflammatory Th1 response. METHODS: We employed TC-1 carcinoma (expressing HPV16 E6 and E7 and devoid of PDL1 expression) in a syngeneic mouse model in monotherapy and combination therapy studies to analyze antitumor effects and changes in immune cell types in the spleen and the TME. RESULTS: As a monotherapy, the PDS0101 vaccine generated HPV-specific T cells and antitumor activity in mice bearing HPV-expressing mEER oropharyngeal and TC-1 lung carcinomas. When used as a monotherapy in the TC-1 model, NHS-IL12 elicited antitumor effects as well as an increase in CD8+ T cells in the TME. When used as a monotherapy, bintrafusp alfa did not elicit antitumor effects or any increase in T cells in the TME. When all three agents were used in combination, maximum antitumor effects were observed, which correlated with increases in T cells and T-cell clonality in the TME. CONCLUSION: These studies provide the rationale for the potential clinical use of combinations of agents that can (1) induce tumor-associated T-cell responses, (2) potentiate immune responses in the TME and (3) reduce immunosuppressive entities in the TME.


Cancer Vaccines/immunology , Carcinoma/therapy , Immune Checkpoint Inhibitors/administration & dosage , Papillomavirus Infections/therapy , Papillomavirus Vaccines/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Carcinoma/immunology , Carcinoma/pathology , Carcinoma/virology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Human papillomavirus 16/immunology , Humans , Immunoconjugates/administration & dosage , Immunogenicity, Vaccine , Immunoglobulin G/administration & dosage , Immunotherapy, Active/methods , Interleukin-12/administration & dosage , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Papillomavirus Vaccines/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Repressor Proteins/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
11.
Adv Exp Med Biol ; 1257: 155-168, 2020.
Article En | MEDLINE | ID: mdl-32483738

Interleukin(IL)-12 is a protein that activates T cells and macrophages to kill tumor cells. However, despite this cytokine showing strong antitumor activity in preclinical settings, translation to patients has been slowed by toxic side effects, poor distribution to peripheral tissues, and improper dosing regimens. Osteosarcoma (OS) is an aggressive primary tumor of bone that has shown particular responsiveness to recombinant (r)IL-12 in preclinical models. Poly(lactic-co-glycolic) acid (PLGA) nanospheres, an FDA-approved drug delivery vector, may be a viable delivery vector for transporting biologically active IL-12 to tissues without disturbing normal homeostasis. In this chapter, we explore the potential for using IL-12-loaded nanospheres (IL-12-NS, <1 µm in diameter) to treat cancer, describe the synthesis process, and examine a typical protein release profile while providing insight and future directions of nanoscale tumor immunotherapeutics.


Bone Neoplasms , Immunotherapy , Interleukin-12 , Nanocapsules , Osteosarcoma , Bone Neoplasms/therapy , Humans , Immunotherapy/trends , Interleukin-12/administration & dosage , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Osteosarcoma/therapy
12.
Adv Exp Med Biol ; 1257: 169-178, 2020.
Article En | MEDLINE | ID: mdl-32483739

This chapter discusses a novel target of osteosarcoma (OS), cell-surface vimentin (CSV), and a novel generation of interleukin-12 (IL12), CSV-targeted IL12, for treating OS tumor metastasis. Vimentin is a known intracellular structural protein for mesenchymal cells but is also documented in tumor cells. Our recent study definitively revealed that vimentin can be translocated to the surface of very aggressive tumor cells, such as metastatic cells. This CSV property allows investigators to capture circulating tumor cells (CTCs) across any type of tumor, including OS. CTCs are known as the seeds of metastasis; therefore, targeting these cells using CSV is a logical approach for use in a metastatic OS setting. Interestingly, we found that the peptide VNTANST can bind to CSV when fused to the p40 subunit encoding the DNA of IL12. Systemic delivery of this CSV-targeted IL12 immune therapy inhibited OS metastasis and relapse in a mouse tumor model as detailed in this chapter. This CSV-targeted delivery of IL12 also reduced toxicity of IL12. In summary, this chapter details a novel approach for safe IL12 immune therapy via targeting CSV.


Bone Neoplasms , Immunotherapy , Interleukin-12 , Sarcoma , Vimentin , Animals , Bone Neoplasms/therapy , Immunotherapy/trends , Interleukin-12/administration & dosage , Mice , Neoplasm Recurrence, Local , Sarcoma/therapy , Vimentin/metabolism
13.
Arch Virol ; 165(9): 1959-1968, 2020 Sep.
Article En | MEDLINE | ID: mdl-32519007

Newcastle disease (ND), caused by virulent Newcastle disease virus (NDV) strains, has been one of the most problematic diseases affecting the poultry industry worldwide. Conventional vaccines provide effective protection for birds to survive ND outbreaks, but they may not completely suppress NDV shedding. NDV strains circulate on farms for a long time after the initial infection and cause potential risks. A new vaccine with fast clearance ability and low viral shedding is needed. In this study, we used interleukin-12 (IL-12) as an adjuvant and electroporation (EP) as an advanced delivery system to improve a DNA vaccine candidate. The fusion (F) protein gene from an NDV strain of the prevalent genotype VII.1.1 was cloned to prepare the vaccine. Chickens immunized with the F gene DNA vaccine co-delivered with an IL-12-expressing plasmid DNA showed higher neutralizing antibody levels and stronger concanavalin-A-induced lymphocyte proliferation than those treated with the F gene DNA vaccine alone. The co-delivered vaccine provided 100% protection, and less viral shedding and a shorter release time were observed in challenged chickens than when the F gene DNA vaccine was administered alone. The use of F gene DNA combined with IL-12 delivered by electroporation is a promising approach for vaccination against ND.


Adjuvants, Immunologic/administration & dosage , Interleukin-12/immunology , Newcastle Disease/prevention & control , Newcastle disease virus/immunology , Poultry Diseases/prevention & control , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Chickens , Electroporation , Interleukin-12/administration & dosage , Newcastle Disease/immunology , Newcastle Disease/virology , Newcastle disease virus/genetics , Newcastle disease virus/physiology , Poultry Diseases/immunology , Poultry Diseases/virology , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virus Shedding
14.
Clin Cancer Res ; 26(12): 2827-2837, 2020 06 15.
Article En | MEDLINE | ID: mdl-32376655

PURPOSE: Tumors with low frequencies of checkpoint positive tumor-infiltrating lymphocytes (cpTIL) have a low likelihood of response to PD-1 blockade. We conducted a prospective multicenter phase II trial of intratumoral plasmid IL-12 (tavokinogene telseplasmid; "tavo") electroporation combined with pembrolizumab in patients with advanced melanoma with low frequencies of checkpoint positive cytotoxic lymphocytes (cpCTL). PATIENTS AND METHODS: Tavo was administered intratumorally days 1, 5, and 8 every 6 weeks while pembrolizumab (200 mg, i.v.) was administered every 3 weeks. The primary endpoint was objective response rate (ORR) by RECIST, secondary endpoints included duration of response, overall survival and progression-free survival. Toxicity was evaluated by the CTCAE v4. Extensive correlative analysis was done. RESULTS: The combination of tavo and pembrolizumab was well tolerated with adverse events similar to those previously reported with pembrolizumab alone. Patients had a 41% ORR (n = 22, RECIST 1.1) with 36% complete responses. Correlative analysis showed that the combination enhanced immune infiltration and sustained the IL-12/IFNγ feed-forward cycle, driving intratumoral cross-presenting dendritic cell subsets with increased TILs, emerging T cell receptor clones and, ultimately, systemic cellular immune responses. CONCLUSIONS: The combination of tavo and pembrolizumab was associated with a higher than expected response rate in this poorly immunogenic population. No new or unexpected toxicities were observed. Correlative analysis showed T cell infiltration with enhanced immunity paralleling the clinical activity in low cpCTL tumors.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Case-Control Studies , Female , Follow-Up Studies , Humans , Interleukin-12/administration & dosage , Male , Melanoma/pathology , Middle Aged , Prognosis , Prospective Studies
15.
PLoS Negl Trop Dis ; 14(3): e0007813, 2020 03.
Article En | MEDLINE | ID: mdl-32196487

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne disease caused by SFTS virus (SFTSV) infection. Despite a gradual increase of SFTS cases and high mortality in endemic regions, no specific viral therapy nor vaccine is available. Here, we developed a single recombinant plasmid DNA encoding SFTSV genes, Gn and Gc together with NP-NS fusion antigen, as a vaccine candidate. The viral antigens were fused with Fms-like tyrosine kinase-3 ligand (Flt3L) and IL-12 gene was incorporated into the plasmid to enhance cell-mediated immunity. Vaccination with the DNA provides complete protection of IFNAR KO mice upon lethal SFTSV challenge, whereas immunization with a plasmid without IL-12 gene resulted in partial protection. Since we failed to detect antibodies against surface glycoproteins, Gn and Gc, in the immunized mice, antigen-specific cellular immunity, as confirmed by enhanced antigen-specific T cell responses, might play major role in protection. Finally, we evaluated the degree of protective immunity provided by protein immunization of the individual glycoprotein, Gn or Gc. Although both protein antigens induced a significant level of neutralizing activity against SFTSV, Gn vaccination resulted in relatively higher neutralizing activity and better protection than Gc vaccination. However, both antigens failed to provide complete protection. Given that DNA vaccines have failed to induce sufficient immunogenicity in human trials when compared to protein vaccines, optimal combinations of DNA and protein elements, proper selection of target antigens, and incorporation of efficient adjuvant, need to be further investigated for SFTSV vaccine development.


Adjuvants, Immunologic/pharmacology , Antigens, Viral/immunology , Bunyaviridae Infections/prevention & control , Interleukin-12/administration & dosage , Phlebovirus/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antigens, Viral/genetics , Disease Models, Animal , Female , Immunity, Cellular , Interleukin-12/pharmacology , Mice, Knockout , Phlebovirus/genetics , Plasmids/administration & dosage , T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
16.
J Immunother Cancer ; 8(1)2020 03.
Article En | MEDLINE | ID: mdl-32217766

BACKGROUND: Local recurrence and remote metastasis are major challenges to overcome in order to improve the survival of patients with cancer after surgery. Oncolytic viruses are a particularly attractive option for prevention of postsurgical disease as they offer a non-toxic treatment option that can directly target residual tumor deposits and beneficially modulate the systemic immune environment that is suppressed post surgery and allows residual disease escape from control. Here, we report that a novel Vaccinia virus (VV), VVΔTKΔN1L (with deletion of both thymidine kinase (TK) and N1L genes) armed with interleukin 12 (IL-12), can prolong postoperative survival when used as a neoadjuvant treatment in different murine and hamster surgical models of cancer. METHODS: A tumor-targeted replicating VV with deletion of TK gene and N1L gene (VVΔTKΔN1L) was created. This virus was armed rationally with IL-12. The effect of VVΔTKΔN1L and VVΔTKΔN1L-IL12 on modulation of the tumor microenvironment and induction of tumor-specific immunity as well the feasibility and safety as a neoadjuvant agent for preventing recurrence and metastasis after surgery were assessed in several clinically relevant models. RESULTS: VVΔTKΔN1L can significantly prolong postoperative survival when used as a neoadjuvant treatment in three different surgery-induced metastatic models of cancer. Efficacy was critically dependent on elevation of circulating natural killer cells that was achieved by virus-induced cytokine production from cells infected with N1L-deleted, but not N1L-intact VV. This effect was further enhanced by arming VVΔTKΔN1L with IL-12, a potent antitumor cytokine. Five daily treatments with VVΔTKΔN1L-IL12 before surgery dramatically improved postsurgical survival. VVΔTKΔN1L armed with human IL-12 completely prevented tumor recurrence in surgical models of head and neck cancer in Syrian hamsters. CONCLUSIONS: These data provide a proof of concept for translation of the regime into clinical trials. VVΔTKΔN1L-IL12 is a promising agent for use as an adjuvant to surgical treatment of solid tumors.


Immunity/immunology , Lung Neoplasms/prevention & control , Neoplasm Recurrence, Local/prevention & control , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Pancreatic Neoplasms/prevention & control , Vaccinia virus/genetics , Adjuvants, Immunologic/administration & dosage , Animals , Apoptosis , Cell Proliferation , Female , Humans , Interleukin-12/administration & dosage , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/surgery , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Tumor Cells, Cultured , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
17.
Methods Mol Biol ; 2121: 115-127, 2020.
Article En | MEDLINE | ID: mdl-32147791

Innate lymphoid cells (ILCs) are lymphocytes with critical roles in homeostasis, inflammation, and immunity to pathogens. ILCs are rare relative to other immune cell populations and are primarily defined by lack of expression of markers associated with other immune cell lineages and are predominantly found in mucosal tissues like the gut, lung and skin. They are classified into distinct subsets, ILC1, ILC2, and ILC3, which mirror subsets of CD4+ helper T cells. ILC subsets have distinct cytokine and transcription factor profiles which align with their biological functions, although recently it has emerged that ILC subsets are not phenotypically fixed and exhibit considerable heterogeneity and plasticity in different contexts. Here, we describe protocols for the maintenance, expansion, and induction of plasticity in mouse and human ILC2s. The resulting cells can be used for molecular interrogation of ILC function and biology, both in vivo and in vitro.


Cell Plasticity/immunology , Cytokines/administration & dosage , Cytokines/pharmacology , Immunity, Innate , Leukocytes, Mononuclear/cytology , Lung/cytology , Lymphocyte Subsets/cytology , Animals , Cell Count , Cell Lineage , Cell Plasticity/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Interleukin-12/administration & dosage , Interleukin-12/pharmacology , Interleukin-18/administration & dosage , Interleukin-1beta/pharmacology , Interleukin-2/pharmacology , Interleukin-33/administration & dosage , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lung/drug effects , Lung/immunology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Mice
18.
CEN Case Rep ; 9(3): 204-209, 2020 08.
Article En | MEDLINE | ID: mdl-32100251

Ustekinumab (UST), an interleukin (IL)-12/IL-23-blocking monoclonal antibody, is a novel therapeutic option for Crohn's disease (CD). We describe a 24-year-old man with CD who showed an abrupt decline in renal function after administration of UST. Twenty-nine months previously, the patient was diagnosed with CD, and abnormal urinalysis findings in health checkup were coincidentally found at that time. Three months previously, treatment for CD was switched from infliximab to UST because of therapy-resistant severe diarrhea and bloody stools. A single dose of UST (260 mg) was initially intravenously administered, followed by single subcutaneous administration (90 mg) 2 months later. Thereafter, the patient exhibited rapid renal dysfunction with significant urinary abnormalities, although his gastrointestinal symptoms had completely disappeared. He was admitted to our hospital for further examination and treatment. Renal pathologic findings were compatible with crescentic glomerulonephritis consisting of almost fibro-cellular crescents. Immunofluorescent study showed IgA and C3 deposition in the glomerular mesangial area and IgA subclass staining revealed predominant IgA1 with concomitant mild IgA2 deposition. Furthermore, galactose-deficient IgA1 (Gd-IgA1) was also positive in the mesangial area. In addition, serum-Gd-IgA1 level was moderately increased. UST treatment was stopped and he responded to intensive steroid therapy with a parallel reduction of serum creatinine and Gd-IgA1 levels without flare of gastrointestinal symptoms. To our knowledge, this is the first case of immunoglobulin A nephropathy (IgAN) in patient with CD that might be aggravated by UST treatment. We presume that inhibition of IL-12/23 signaling with UST may cause to form crescentic IgAN by enhancing Gd-IgA1 production.


Crohn Disease/drug therapy , Galactose/deficiency , Glomerulonephritis, IGA/chemically induced , Interleukin-12/adverse effects , Kidney/physiopathology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/therapeutic use , Administration, Intravenous , Antibodies, Monoclonal/adverse effects , Complement C3/metabolism , Galactose/blood , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Humans , Immunoglobulin A/blood , Immunoglobulin A/metabolism , Injections, Subcutaneous , Interleukin-12/administration & dosage , Interleukin-12/therapeutic use , Kidney/pathology , Male , Steroids/administration & dosage , Steroids/therapeutic use , Treatment Outcome , Withholding Treatment , Young Adult
19.
Cell Immunol ; 349: 104042, 2020 03.
Article En | MEDLINE | ID: mdl-32061376

Cytotoxic T lymphocytes (CTLs) play a major role in cancer immunotherapy. A potent tumor immunotherapy may not only require activation of anti-tumor effector cells but also rely on the use of cytokines to create a controlled environment for the development of anti-tumor T cells. In this study, we fabricated a dual-target immunonanoparticle, e.g. poly(d,l-lactide-co-glycolide) nanoparticle, by loading Interleukin-12 (IL-12) and modifying with CD8 and Glypican-3 antibodies on the surface. Our results demonstrate that the fabricated targeting immunonanoparticles bind specifically to the two target cells of interest, i.e. CD8+ T cells and HepG-2 cells via the antibody-antigen interactions and form T cell-HepG-2 cell clusters, which enhances the cytotoxicity of T cells. IL-12-containing dual-target immunonanoparticles delivered IL-12 specifically to CD8+ T cells, and favored the expansion, activation and cytotoxic activity of CD8+ T lymphocytes. These results suggest that dual-target IL-12-encapsulated nanoparticles are a promising platform for cancer immunotherapy.


CD8-Positive T-Lymphocytes/drug effects , Immunotherapy/methods , Interleukin-12/administration & dosage , Nanocapsules/administration & dosage , T-Lymphocytes, Cytotoxic/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Division/drug effects , Cytotoxicity, Immunologic , Drug Delivery Systems , Drug Screening Assays, Antitumor , Drug Stability , Hep G2 Cells , Humans , Immunophenotyping , Interferon-gamma Release Tests , Interleukin-12/pharmacology , Lymphocyte Activation/drug effects , Polylactic Acid-Polyglycolic Acid Copolymer , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Rosette Formation , T-Lymphocytes, Cytotoxic/immunology
20.
J Immunother Cancer ; 8(1)2020 01.
Article En | MEDLINE | ID: mdl-31959727

BACKGROUND: Interleukin-12 (IL-12) is a potent, proinflammatory cytokine that holds promise for cancer immunotherapy, but its clinical use has been limited by its toxicity. To minimize systemic exposure and potential toxicity while maintaining the beneficial effects of IL-12, we developed a novel IL-12-based therapeutic system that combines tumor-specific T-cell-mediated delivery of IL-12 with membrane-restricted IL-12 localization and inducible IL-12 expression. METHODS: Therapeutic T cells targeting a tumor antigen were genetically engineered to express membrane-anchored IL-12 (aIL-12). Expression, function, and shedding of the aIL-12 molecule was assessed in vitro. Tumor treatment efficacy was assessed in vivo with T cell receptor (TCR) transgenic murine tumor models and a tumor xenograft model. Key outcomes were change in tumor size, circulating levels of IL-12 and other cytokines, and survival. Toxicity was assessed via change in body weight. Tumor growth curve measurements were compared using repeated-measures two-way analyses of variance. RESULTS: Retroviral gene transfer resulted in cell membrane expression of aIL-12 by transduced T cells. In each of two transgenic murine tumor models, tumor-specific T cells constitutively expressing aIL-12 demonstrated increased antitumor efficacy, low circulating IL-12 and interferon-γ, and no weight loss. Expression of aIL-12 via a NFAT-inducible promoter resulted in coordinate expression of aIL-12 with T cell activation. In an OT-I TCR transgenic murine tumor model, the NFAT-inducible aIL-12 molecule improved tumor treatment and did not result in detectable levels of IL-12 in serum or in weight loss. In a human tumor xenograft model, the NFAT-inducible aIL-12 molecule improved antitumor responses by human T cells coexpressing a tumor-specific engineered TCR. Serum IL-12 levels were undetectable with the NFAT-inducible construct in both models. CONCLUSION: Expression of aIL-12 by tumor-targeting therapeutic T cells demonstrated low systemic exposure and improved efficacy. This treatment strategy may have broad applications to cellular therapy with tumor-infiltrating lymphocytes, chimeric antigen receptor T cells, and TCR T cells.


Antigens, Neoplasm/immunology , Immunotherapy, Adoptive/methods , Interleukin-12/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Receptors, Antigen, T-Cell/immunology , Animals , Cell Line, Tumor , Cell Membrane/genetics , Cell Membrane/metabolism , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Interferon-gamma/metabolism , Interleukin-12/administration & dosage , Interleukin-12/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Xenograft Model Antitumor Assays
...