Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Publication year range
1.
Mol Genet Metab ; 143(1-2): 108560, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39121792

ABSTRACT

Isolated methylmalonic acidemia/aciduria (MMA) due to MMUT enzyme deficiency is an ultra-rare pediatric disease with high morbidity and mortality, with no approved disease-altering therapies. Previous publications showed that systemic treatment with a codon-optimized mRNA encoding wild-type human MMUT (MMUT) is a promising strategy for treatment of MMA. We developed a second-generation drug product, mRNA-3705, comprised of an mRNA encoding the MMUT enzyme formulated in a lipid nanoparticle (LNP) with incorporation of enhancements over the previous clinical candidate mRNA-3704. Both drug products produced functional MMUT in rat livers when dosed IV, and showed long-term safety and efficacy in two mouse models of MMA. mRNA-3705 produced 2.1-3.4-fold higher levels of hepatic MMUT protein expression than the first-generation drug product mRNA-3704 when given at an identical dose level, which resulted in greater and more sustained reductions in plasma methylmalonic acid. The data presented herein provide comprehensive preclinical pharmacology to support the clinical development of mRNA-3705.

2.
J Inherit Metab Dis ; 47(1): 80-92, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37401651

ABSTRACT

Phenylketonuria (PKU) or hyperphenylalaninemia is considered a paradigm for an inherited (metabolic) liver defect and is, based on murine models that replicate all human pathology, an exemplar model for experimental studies on liver gene therapy. Variants in the PAH gene that lead to hyperphenylalaninemia are never fatal (although devastating if untreated), newborn screening has been available for two generations, and dietary treatment has been considered for a long time as therapeutic and satisfactory. However, significant shortcomings of contemporary dietary treatment of PKU remain. A long list of various gene therapeutic experimental approaches using the classical model for human PKU, the homozygous enu2/2 mouse, witnesses the value of this model to develop treatment for a genetic liver defect. The list of experiments for proof of principle includes recombinant viral (AdV, AAV, and LV) and non-viral (naked DNA or LNP-mRNA) vector delivery methods, combined with gene addition, genome, gene or base editing, and gene insertion or replacement. In addition, a list of current and planned clinical trials for PKU gene therapy is included. This review summarizes, compares, and evaluates the various approaches for the sake of scientific understanding and efficacy testing that may eventually pave the way for safe and efficient human application.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Humans , Mice , Animals , Phenylalanine Hydroxylase/genetics , Phenylketonurias/genetics , Phenylketonurias/therapy , Genetic Therapy/methods , Liver/pathology , DNA
3.
J Inherit Metab Dis ; 47(1): 63-79, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37530705

ABSTRACT

Organic acidemias (OA) are a group of rare autosomal recessive disorders of intermediary metabolism that result in a systemic elevation of organic acid. Despite optimal dietary and cofactor therapy, OA patients still suffer from potentially lethal metabolic instability and experience long-term multisystemic complications. Severely affected patients can benefit from elective liver transplantation, which restores hepatic enzymatic activity, improves metabolic stability, and provides the theoretical basis for the pursuit of gene therapy as a new treatment for patients. Because of the poor outcomes reported in those with OA, especially methylmalonic and propionic acidemia, multiple gene therapy approaches have been explored in relevant animal models. Here, we review the results of gene therapy experiments performed using MMA and PA mouse models to illustrate experimental paradigms that could be applicable for all forms of OA.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Liver Transplantation , Propionic Acidemia , Animals , Mice , Humans , Propionic Acidemia/genetics , Propionic Acidemia/therapy , Propionic Acidemia/complications , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/therapy , Amino Acid Metabolism, Inborn Errors/complications , Liver Transplantation/adverse effects , Genetic Therapy , Disease Models, Animal , Methylmalonic Acid
4.
Chembiochem ; 24(5): e202200658, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36594506

ABSTRACT

The identification of pseudo- and N1 -methylpseudo-uridine (Ψ and mΨ, respectively) as immunosilent uridine analogues has propelled the development of mRNA-based vaccines and therapeutics. Here, we have characterised another uridine analogue, 5-ethynyluridine (EU), which has an ethynyl moiety. We show that this uridine analogue does not cause immune activation in human macrophages, as it does not induce interleukin-6 secretion or expression of the inflammatory and antiviral genes MX1, PKR, and TAP2. Moreover, EU allows for prolonged expression, as shown with mRNA coding for yellow fluorescent protein (YFP). Side-by-side comparisons of EU with unmodified, Ψ, and mΨ revealed that EU-modified mRNA is expressed at lower levels, but confers similar stability and low immunogenicity to the other uridine analogues. Furthermore, structure analysis of modified mRNAs suggests that the observed phenotype is largely independent of RNA folding. Thus, EU is a potential candidate for RNA-based vaccines and therapeutics.


Subject(s)
Antiviral Agents , Vaccines , Humans , RNA, Messenger/genetics , RNA, Messenger/chemistry , Uridine
5.
J Surg Res ; 289: 116-120, 2023 09.
Article in English | MEDLINE | ID: mdl-37104922

ABSTRACT

INTRODUCTION: Therapies based on exogenous messenger RNA (mRNA) administration have emerged as a powerful novel strategy for the actual or potential treatment of an assortment of diseases, including congenital surgical pathologies. We sought to determine whether the minimally invasive transamniotic route could be an alternative for prenatal mRNA delivery. METHODS: Pregnant Sprague-Dawley dams underwent laparotomy followed by volume-matched intra-amniotic injections in all their fetuses (n = 120) of either a suspension of a custom firefly luciferase mRNA encapsulated by a lipid- and synthetic cationic polymer-based composite, or of a suspension of the same encapsulation components without mRNA, on gestational day 17 (E17; term = E21-22). On E18, E19, E20, and E21, samples from 14 fetal anatomical sites and maternal serum were procured for the screening of mRNA incorporation by host cells by measurement of luciferase activity via microplate luminometry. Statistical analysis was by Mann-Whitney U-test, including Bonferroni-adjustment. RESULTS: Overall survival was 87.5% (105/120). Controlled by the encapsulating composite without mRNA, luciferase activity was detected in the animals that received encapsulated mRNA in the following fetal annexes: amniotic fluid, amnion, chorion, umbilical cord, and placenta (P = 0.033 to <0.001), as well as in the following fetal sites: liver, stomach, intestines, and lungs (P = 0.043-0.002). CONCLUSIONS: Packaged exogenous mRNA can be incorporated by the fetus at least at select anatomical sites after simple intra-amniotic administration in a rodent model. The pattern and chronology of mRNA incorporation are compatible with transplacental hematogenous routing, as well as with fetal swallowing/aspiration. Further study of transamniotic mRNA administration is warranted.


Subject(s)
Amniotic Fluid , Mesenchymal Stem Cell Transplantation , Pregnancy , Animals , Female , Amnion , Placenta , Luciferases
6.
Proc Natl Acad Sci U S A ; 117(7): 3405-3414, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32005712

ABSTRACT

Drug targeting to inflammatory brain pathologies such as stroke and traumatic brain injury remains an elusive goal. Using a mouse model of acute brain inflammation induced by local tumor necrosis factor alpha (TNFα), we found that uptake of intravenously injected antibody to vascular cell adhesion molecule 1 (anti-VCAM) in the inflamed brain is >10-fold greater than antibodies to transferrin receptor-1 and intercellular adhesion molecule 1 (TfR-1 and ICAM-1). Furthermore, uptake of anti-VCAM/liposomes exceeded that of anti-TfR and anti-ICAM counterparts by ∼27- and ∼8-fold, respectively, achieving brain/blood ratio >300-fold higher than that of immunoglobulin G/liposomes. Single-photon emission computed tomography imaging affirmed specific anti-VCAM/liposome targeting to inflamed brain in mice. Intravital microscopy via cranial window and flow cytometry showed that in the inflamed brain anti-VCAM/liposomes bind to endothelium, not to leukocytes. Anti-VCAM/LNP selectively accumulated in the inflamed brain, providing de novo expression of proteins encoded by cargo messenger RNA (mRNA). Anti-VCAM/LNP-mRNA mediated expression of thrombomodulin (a natural endothelial inhibitor of thrombosis, inflammation, and vascular leakage) and alleviated TNFα-induced brain edema. Thus VCAM-directed nanocarriers provide a platform for cerebrovascular targeting to inflamed brain, with the goal of normalizing the integrity of the blood-brain barrier, thus benefiting numerous brain pathologies.


Subject(s)
Antibodies/administration & dosage , Blood-Brain Barrier/drug effects , Encephalitis/drug therapy , Endothelium, Vascular/drug effects , Nanomedicine/methods , Animals , Blood-Brain Barrier/immunology , Encephalitis/genetics , Encephalitis/immunology , Endothelium, Vascular/immunology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Mice , Receptors, Transferrin/genetics , Receptors, Transferrin/immunology , Thrombomodulin/genetics , Thrombomodulin/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
7.
Int J Mol Sci ; 24(13)2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37446400

ABSTRACT

mRNA-based therapeutics have revolutionized the world of molecular therapy and have proven their potential in the vaccination campaigns for SARS-CoV2 and clinical trials for hereditary disorders. Preclinical studies have mainly focused on in vitro and rodent studies. However, research in rodents is costly and labour intensive, and requires ethical approval for all interventions. Zebrafish embryonic disease models are not always classified as laboratory animals and have been shown to be extremely valuable for high-throughput drug testing. Zebrafish larvae are characterized by their small size, optical transparency and high number of embryos, and are therefore also suited for the study of mRNA-based therapeutics. First, the one-cell stage injection of naked mRNA can be used to assess the effectivity of gene addition in vivo. Second, the intravascular injection in older larvae can be used to assess tissue targeting efficiency of (packaged) mRNA. In this review, we describe how zebrafish can be used as a steppingstone prior to testing mRNA in rodent models. We define the procedures that can be employed for both the one-cell stage and later-stage injections, as well as the appropriate procedures for post-injection follow-up.


Subject(s)
COVID-19 , Zebrafish , Animals , Zebrafish/genetics , RNA, Messenger/genetics , RNA, Viral , SARS-CoV-2/genetics , Embryo, Nonmammalian
8.
Small ; 18(44): e2204436, 2022 11.
Article in English | MEDLINE | ID: mdl-36098251

ABSTRACT

This study presents the first messenger RNA (mRNA) therapy for metastatic ovarian cancer and cachexia-induced muscle wasting based on lipid nanoparticles that deliver follistatin (FST) mRNA predominantly to cancer clusters following intraperitoneal administration. The secreted FST protein, endogenously synthesized from delivered mRNA, efficiently reduces elevated activin A levels associated with aggressive ovarian cancer and associated cachexia. By altering the cancer cell phenotype, mRNA treatment prevents malignant ascites, delays cancer progression, induces the formation of solid tumors, and preserves muscle mass in cancer-bearing mice by inhibiting negative regulators of muscle mass. Finally, mRNA therapy provides synergistic effects in combination with cisplatin, increasing the survival of mice and counteracting muscle atrophy induced by chemotherapy and cancer-associated cachexia. The treated mice develop few nonadherent tumors that are easily resected from the peritoneum. Clinically, this nanomedicine-based mRNA therapy can facilitate complete cytoreduction, target resistance, improve resilience during aggressive chemotherapy, and improve survival in advanced ovarian cancer.


Subject(s)
Nanoparticles , Ovarian Neoplasms , Humans , Female , Cachexia/drug therapy , Cachexia/metabolism , Follistatin/metabolism , Follistatin/pharmacology , Follistatin/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Ovarian Neoplasms/complications , Ovarian Neoplasms/therapy , Muscle, Skeletal/metabolism
9.
J Inherit Metab Dis ; 45(5): 872-888, 2022 09.
Article in English | MEDLINE | ID: mdl-35766386

ABSTRACT

Hereditary methylmalonic acidemia (MMA) caused by deficiency of the enzyme methylmalonyl-CoA mutase (MMUT) is a relatively common and severe organic acidemia. The recalcitrant nature of the condition to conventional dietary and medical management has led to the use of elective liver and combined liver-kidney transplantation in some patients. However, liver transplantation is intrinsically limited by organ availability, the risks of surgery, procedural and life-long management costs, transplant comorbidities, and a remaining underlying risk of complications related to MMA despite transplantation. Here, we review pre-clinical studies that present alternative approaches to solid organ transplantation as a treatment for MMUT MMA, including adeno-associated viral gene addition therapy, mRNA therapy, and genome editing, with and without nuclease enhancement.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Methylmalonyl-CoA Mutase , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/therapy , Dependovirus/genetics , Genomics , Humans , Methylmalonic Acid
10.
BMC Biotechnol ; 21(1): 20, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33691652

ABSTRACT

Over the past decade, therapeutic messenger RNAs (mRNAs) have emerged as a highly promising new class of drugs for protein replacement therapies. Due to the recent developments, the incorporation of modified nucleotides in synthetic mRNAs can lead to maximizing protein expression and reducing adverse immunogenicity. Despite these stunning improvements, mRNA therapy is limited by the need for the development of safe and efficient carriers to protect the mRNA integrity for in vivo applications. Recently, leading candidates for in vivo drug delivery vehicles are cell-derived exosomes, which have fewer immunogenic responses. In the current study, the key hurdles facing mRNA-based therapeutics, with an emphasis on recent strategies to overcoming its immunogenicity and instability, were highlighted. Then the immunogenicity and toxicity of exosomes derived from various cell sources were mentioned in detail. Finally, an overview of the recent strategies in using exosomes for mRNA delivery in the treatment of multiple diseases was stated.


Subject(s)
Drug Delivery Systems/methods , Exosomes/genetics , RNA, Messenger/genetics , Animals , Drug Delivery Systems/instrumentation , Exosomes/chemistry , Exosomes/immunology , Humans , RNA Stability , RNA, Messenger/chemistry , RNA, Messenger/immunology
11.
Anal Biochem ; 627: 114259, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34033800

ABSTRACT

mRNA-based therapeutics have progressed significantly over the past decade and hold great potential to treat several diseases, including but not limited to cancer, infectious diseases, and genetic disorders. Many mRNA therapeutics are encapsulated in lipid nanoparticles (LNPs), which stabilize the mRNA and, following patient administration, transfect target cells to deliver the therapeutic mRNA. Developing reliable and robust in vitro bioassays is critical for expediting early screening of LNP-mRNA formulations, as well as identifying and developing lead candidate drug products. In this study, high-content fluorescent imaging was used to monitor LNP-mRNA transfections in both two- and three-dimensional HepG2 cell cultures. This technique allowed for continuous and simultaneous measurement of multiple LNP-mRNA transfection parameters, including cellular cytotoxicity, mRNA delivery, and mRNA translation/protein expression. Moreover, high-content imaging demonstrated acceptable accuracy and precision when quantifying the potency of mRNA delivery and subsequent translation and protein expression, thereby establishing the method as fit-for-purpose. Furthermore, concurrent evaluation of two different LNP formulations showed high-content imaging is sensitive at detecting pharmacological differences among varying LNP formulations. This study details the efficiency and reliability of LNP-mRNA characterizations that can be accomplished with high-content imaging.


Subject(s)
Genetic Therapy/methods , Lipids/chemistry , Nanoparticles/chemistry , Neoplasms/metabolism , RNA, Messenger/pharmacology , Diagnostic Imaging , Fluorescence , Hep G2 Cells , Humans , Neoplasms/therapy , Optical Imaging/methods , RNA, Messenger/chemistry , Reproducibility of Results , Transfection
12.
Mol Ther ; 28(1): 304-312, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31604675

ABSTRACT

Classic galactosemia (CG) is a potentially lethal inborn error of galactose metabolism that results from deleterious mutations in the human galactose-1 phosphate uridylyltransferase (GALT) gene. Previously, we constructed a GalT-/- (GalT-deficient) mouse model that exhibits galactose sensitivity in the newborn mutant pups, reduced fertility in adult females, impaired motor functions, and growth restriction in both sexes. In this study, we tested whether restoration of hepatic GALT activity alone could decrease galactose-1 phosphate (gal-1P) and plasma galactose in the mouse model. The administration of different doses of mouse GalT (mGalT) mRNA resulted in a dose-dependent increase in mGalT protein expression and enzyme activity in the liver of GalT-deficient mice. Single intravenous (i.v.) dose of human GALT (hGALT) mRNA decreased gal-1P in mutant mouse liver and red blood cells (RBCs) within 24 h with low levels maintained for over a week. Repeated i.v. injections increased hepatic GalT expression, nearly normalized gal-1P levels in liver, and decreased gal-1P levels in RBCs and peripheral tissues throughout all doses. Moreover, repeated dosing reduced plasma galactose by 60% or more throughout all four doses. Additionally, a single intraperitoneal dose of hGALT mRNA overcame the galactose sensitivity and promoted the growth in a GalT-/- newborn pup.


Subject(s)
Disease Models, Animal , Galactose/blood , Galactosemias/therapy , RNA, Messenger/administration & dosage , UTP-Hexose-1-Phosphate Uridylyltransferase/administration & dosage , Animals , Animals, Newborn , Cells, Cultured , Erythrocytes/drug effects , Erythrocytes/metabolism , Female , Fibroblasts/metabolism , Galactosemias/pathology , Galactosephosphates/metabolism , Humans , Injections, Intraperitoneal , Injections, Intravenous , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Signal Transduction/drug effects , Transfection , Treatment Outcome , UTP-Hexose-1-Phosphate Uridylyltransferase/genetics
13.
Mol Ther ; 27(4): 785-793, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30611663

ABSTRACT

Myocardial infarction (MI) and heart failure (HF) are the leading causes of death in the United States and in most other industrialized nations. MI leads to a massive loss of cardiomyocytes (CMs), which are replaced with non-CM cells, leading to scarring and, in most cases, HF. The adult mammalian heart has a low intrinsic regenerative capacity, mainly because of cell-cycle arrest in CMs. No effective treatment promoting heart regeneration is currently available. Recent efforts to use DNA-based or viral gene therapy approaches to induce cardiac regeneration post-MI or in HF conditions have encountered major challenges, mostly because of the poor and uncontrolled delivery of the introduced genes. Modified mRNA (modRNA) is a safe, non-immunogenic, efficient, transient, local, and controlled nucleic acid delivery system that can overcome the obstacles to DNA-based or viral approaches for cardiac gene delivery. We here review the use of modRNA in cardiac therapy, to induce cardioprotection and vascular or cardiac regeneration after MI. We discuss the current challenges in modRNA-based cardiac treatment, which will need to be overcome for the application of such treatment to ischemic heart disease.


Subject(s)
Enzyme Replacement Therapy/methods , Genetic Therapy/methods , Heart Failure/therapy , Myocardial Infarction/therapy , RNA, Messenger/genetics , Animals , Drug Delivery Systems , Genetic Therapy/adverse effects , Humans , Myocytes, Cardiac/metabolism , Nanoparticles , Regeneration , Transfection
14.
J Hepatol ; 71(2): 422-433, 2019 08.
Article in English | MEDLINE | ID: mdl-31102718

ABSTRACT

Porphyrias are rare inherited disorders caused by specific enzyme dysfunctions in the haem synthesis pathway, which result in abnormal accumulation of specific pathway intermediates. The symptoms depend upon the chemical characteristics of these substances. Porphyrins are photoreactive and cause photocutaneous lesions on sunlight-exposed areas, whereas accumulation of porphyrin precursors is related to acute neurovisceral attacks. Current therapies are suboptimal and mostly address symptoms rather than underlying disease mechanisms. Advances in the understanding of the molecular bases and pathogenesis of porphyrias have paved the way for the development of new therapeutic strategies. In this Clinical Trial Watch we summarise the basic principles of these emerging approaches and what is currently known about their application to porphyrias of hepatic origin or with hepatic involvement.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Bone Marrow Transplantation/methods , Cholestyramine Resin/therapeutic use , Genetic Therapy/methods , Liver Transplantation/methods , Porphyrias, Hepatic/drug therapy , Porphyrias, Hepatic/surgery , Pyrrolidines/therapeutic use , Receptor, Melanocortin, Type 1/agonists , alpha-MSH/analogs & derivatives , 5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/pharmacology , Acetylgalactosamine/therapeutic use , Heme/biosynthesis , Humans , Liver/metabolism , Porphyrias, Hepatic/classification , Porphyrias, Hepatic/pathology , Porphyrins/metabolism , Pyrrolidines/pharmacology , alpha-MSH/therapeutic use
15.
RNA Biol ; 15(7): 914-922, 2018.
Article in English | MEDLINE | ID: mdl-29923457

ABSTRACT

Arginase I (ARG1) deficiency is an autosomal recessive urea cycle disorder, caused by deficiency of the enzyme Arginase I, resulting in accumulation of arginine in blood. Current Standard of Care (SOC) for ARG1 deficiency in patients or those having detrimental mutations of ARG1 gene is diet control. Despite diet and drug therapy with nitrogen scavengers, ~25% of patients suffer from severe mental deficits and loss of ambulation. 75% of patients whose symptoms can be managed through diet therapy continue to suffer neuro-cognitive deficits. In our research, we demonstrate in vitro and in vivo that administration of ARG1 mRNA increased ARG1 protein expression and specific activity in relevant cell types, including ARG1-deficient patient cell lines, as well as in wild type mice for up to 4 days. These studies demonstrate that ARG1 mRNA treatment led to increased functional protein expression of ARG1 and subsequently an increase in urea. Hence, ARG1 mRNA therapy could be a potential treatment option to develop for patients.


Subject(s)
Arginase/metabolism , Arginine/metabolism , Biological Therapy/methods , Hyperargininemia/therapy , RNA, Messenger/administration & dosage , Animals , Arginase/genetics , HeLa Cells , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mutation , Urea/metabolism
16.
Heliyon ; 10(5): e26971, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38486748

ABSTRACT

mRNA-based therapeutics have revolutionized medicine and the pharmaceutical industry. The recent progress in the optimization and formulation of mRNAs has led to the development of a new therapeutic platform with a broad range of applications. With a growing body of evidence supporting the use of mRNA-based drugs for precision medicine and personalized treatments, including cancer immunotherapy, genetic disorders, and autoimmune diseases, this emerging technology offers a rapidly expanding category of therapeutic options. Furthermore, the development and deployment of mRNA vaccines have facilitated a prompt and flexible response to medical emergencies, exemplified by the COVID-19 outbreak. The establishment of stable and safe mRNA molecules carried by efficient delivery systems is now available through recent advances in molecular biology and nanotechnology. This review aims to elucidate the advancements in the clinical applications of mRNAs for addressing significant health-related challenges such as cancer, autoimmune diseases, genetic disorders, and infections and provide insights into the efficacy and safety of mRNA therapeutics in recent clinical trials.

17.
Cancer Lett ; 598: 217111, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38972347

ABSTRACT

Intratumoral delivery of mRNA encoding immunostimulatory molecules can initiate a robust, global antitumor response with little side effects by enhancing local antigen presentation in the tumor and the tumor draining lymph node. Neoantigen-based mRNA nanovaccine can inhibit melanoma growth in mice by intratumoral injection. Myeloid-derived suppressor cells (MDSCs) suppress antitumor immune responses by secreting immunosuppressive agents, such as reactive oxygen species (ROS). Suppression of STAT3 activity by stattic may reduce MDSC-mediated immunosuppression in the TME and promote the antitumor immune responses. In this study, in vitro transcribed mRNA encoding tumor antigen survivin was prepared and injected intratumorally in BALB/c mice bearing subcutaneous colon cancer tumors. In vivo studies demonstrated that intratumoral survivin mRNA therapy could induce antitumor T cell response and inhibit tumor growth of colon cancer. Depletion of CD8+ T cells could significantly inhibit survivin mRNA-induced antitumor effects. RT-qPCR and ELISA analysis indicated that survivin mRNA treatment led to increased expression of receptor activator nuclear factor-κB ligand (RANKL). In vitro experiment showed that MDSCs could be induced from mouse bone marrow cells by RANKL and RANKL-induced MDSCs could produce high level of ROS. STAT3 inhibitor stattic suppressed activation of STAT3 and NF-κB signals, thereby inhibiting expansion of RANKL-induced MDSCs. Combination therapy of survivin mRNA and stattic could significantly enhance antitumor T cell response, improve long-term survival and reduce immunosuppressive tumor microenvironment compared to each monotherapy. In addition, combined therapy resulted in a significantly reduced level of tumor cell proliferation and an obviously increased level of tumor cell apoptosis in CT26 colon cancer-bearing mice, which could be conducive to inhibit the tumor growth and lead to immune responses to released tumor-associated antigens. These studies explored intratumoral mRNA therapy and mRNA-based combined therapy to treat colon cancer and provide a new idea for cancer therapy.


Subject(s)
Colonic Neoplasms , Cyclic S-Oxides , Mice, Inbred BALB C , RNA, Messenger , STAT3 Transcription Factor , Survivin , Animals , Survivin/genetics , Survivin/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , RNA, Messenger/genetics , Colonic Neoplasms/therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/genetics , Cyclic S-Oxides/pharmacology , Cell Line, Tumor , Injections, Intralesional , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/drug effects , Tumor Microenvironment/drug effects , Mice , Female , Reactive Oxygen Species/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Humans , Tumor Burden/drug effects , Signal Transduction
18.
Int Urol Nephrol ; 2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38613662

ABSTRACT

Fabry Disease (FD) is a genetic disease caused by a deficiency in the activity of lysosomal galactosidase A (α-GalA), an enzyme responsible for the catabolism of globotriaosylceramide (Gb3). Since lysosomes are present throughout the body and play a crucial role in catabolism and recycling of cytosolic compounds, FD can affect multiple organs and result in various symptoms, including renal, cardiovascular, neurological, cutaneous, and ophthalmic manifestations. Due to the nonspecific symptoms and the rarity of FD, it is often diagnosed late in life. However, introducing targeted therapies such as enzyme replacement therapy (ERT) and chaperone therapy has significantly improved FD's natural history and prognosis by restoring α-GalA enzyme activity. Despite the advancements, there are limitations to the currently available therapies, which has prompted research into new potential treatments for FD, including alternative forms of enzyme replacement therapy, substrate reduction therapy, mRNA therapy, and genetic therapy. In this review, we analyze the epidemiology, pathophysiology, and treatment of FD, with particular emphasis on promising therapeutic opportunities that could shift the treatment of this rare disease from a standardized to a personalized approach soon.

19.
Mol Ther Nucleic Acids ; 35(3): 102258, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39045516

ABSTRACT

Ocular neurodegenerative diseases like glaucoma lead to progressive retinal ganglion cell (RGC) loss, causing irreversible vision impairment. Neuroprotection is needed to preserve RGCs across debilitating conditions. Nerve growth factor (NGF) protein therapy shows efficacy, but struggles with limited bioavailability and a short half-life. Here we explore a novel approach to address this deficiency by utilizing circular RNA (circRNA)-based therapy. We show that circRNAs exhibit an exceptional capacity for prolonged protein expression and circRNA-expressed NGF protects cells from glucose deprivation. In a mouse optic nerve crush model, lipid nanoparticle (LNP)-formulated circNGF administered intravitreally protects RGCs and axons from injury-induced degeneration. It also significantly outperforms NGF protein therapy without detectable retinal toxicity. Furthermore, single-cell transcriptomics revealed LNP-circNGF's multifaceted therapeutic effects, enhancing genes related to visual perception while reducing trauma-associated changes. This study signifies the promise of circRNA-based therapies for treating ocular neurodegenerative diseases and provides an innovative intervention platform for other ocular diseases.

20.
Pharmaceutics ; 16(5)2024 May 19.
Article in English | MEDLINE | ID: mdl-38794346

ABSTRACT

Over the past decade, mRNA-based therapy has displayed significant promise in a wide range of clinical applications. The most striking example of the leap in the development of mRNA technologies was the mass vaccination against COVID-19 during the pandemic. The emergence of large-scale technology and positive experience of mRNA immunization sparked the development of antiviral and anti-cancer mRNA vaccines as well as therapeutic mRNA agents for genetic and other diseases. To facilitate mRNA delivery, lipid nanoparticles (LNPs) have been successfully employed. However, the diverse use of mRNA therapeutic approaches requires the development of adaptable LNP delivery systems that can control the kinetics of mRNA uptake and expression in target cells. Here, we report effective mRNA delivery into cultured mammalian cells (HEK293T, HeLa, DC2.4) and living mouse muscle tissues by liposomes containing either 1,26-bis(cholest-5-en-3ß-yloxycarbonylamino)-7,11,16,20-tetraazahexacosane tetrahydrochloride (2X3) or the newly applied 1,30-bis(cholest-5-en-3ß-yloxycarbonylamino)-9,13,18,22-tetraaza-3,6,25,28-tetraoxatriacontane tetrahydrochloride (2X7) cationic lipids. Using end-point and real-time monitoring of Fluc mRNA expression, we showed that these LNPs exhibited an unusually delayed (of over 10 h in the case of the 2X7-based system) but had highly efficient and prolonged reporter activity in cells. Accordingly, both LNP formulations decorated with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] (DSPE-PEG2000) provided efficient luciferase production in mice, peaking on day 3 after intramuscular injection. Notably, the bioluminescence was observed only at the site of injection in caudal thigh muscles, thereby demonstrating local expression of the model gene of interest. The developed mRNA delivery systems hold promise for prophylactic applications, where sustained synthesis of defensive proteins is required, and open doors to new possibilities in mRNA-based therapies.

SELECTION OF CITATIONS
SEARCH DETAIL