Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nature ; 543(7644): 248-251, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28151488

ABSTRACT

Zika virus (ZIKV) has recently emerged as a pandemic associated with severe neuropathology in newborns and adults. There are no ZIKV-specific treatments or preventatives. Therefore, the development of a safe and effective vaccine is a high priority. Messenger RNA (mRNA) has emerged as a versatile and highly effective platform to deliver vaccine antigens and therapeutic proteins. Here we demonstrate that a single low-dose intradermal immunization with lipid-nanoparticle-encapsulated nucleoside-modified mRNA (mRNA-LNP) encoding the pre-membrane and envelope glycoproteins of a strain from the ZIKV outbreak in 2013 elicited potent and durable neutralizing antibody responses in mice and non-human primates. Immunization with 30 µg of nucleoside-modified ZIKV mRNA-LNP protected mice against ZIKV challenges at 2 weeks or 5 months after vaccination, and a single dose of 50 µg was sufficient to protect non-human primates against a challenge at 5 weeks after vaccination. These data demonstrate that nucleoside-modified mRNA-LNP elicits rapid and durable protective immunity and therefore represents a new and promising vaccine candidate for the global fight against ZIKV.


Subject(s)
RNA, Messenger/administration & dosage , RNA, Messenger/chemistry , Viral Vaccines/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Female , Glycoproteins/genetics , Glycoproteins/immunology , Injections, Intradermal , Macaca mulatta/immunology , Macaca mulatta/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/chemistry , RNA Stability , RNA, Messenger/genetics , RNA, Viral/administration & dosage , RNA, Viral/chemistry , RNA, Viral/genetics , Time Factors , Vaccination , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus Infection/immunology
2.
Mol Ther ; 28(7): 1569-1584, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32359470

ABSTRACT

Influenza viruses are respiratory pathogens of public health concern worldwide with up to 650,000 deaths occurring each year. Seasonal influenza virus vaccines are employed to prevent disease, but with limited effectiveness. Development of a universal influenza virus vaccine with the potential to elicit long-lasting, broadly cross-reactive immune responses is necessary for reducing influenza virus prevalence. In this study, we have utilized lipid nanoparticle-encapsulated, nucleoside-modified mRNA vaccines to intradermally deliver a combination of conserved influenza virus antigens (hemagglutinin stalk, neuraminidase, matrix-2 ion channel, and nucleoprotein) and induce strong immune responses with substantial breadth and potency in a murine model. The immunity conferred by nucleoside-modified mRNA-lipid nanoparticle vaccines provided protection from challenge with pandemic H1N1 virus at 500 times the median lethal dose after administration of a single immunization, and the combination vaccine protected from morbidity at a dose of 50 ng per antigen. The broad protective potential of a single dose of combination vaccine was confirmed by challenge with a panel of group 1 influenza A viruses. These findings support the advancement of nucleoside-modified mRNA-lipid nanoparticle vaccines expressing multiple conserved antigens as universal influenza virus vaccine candidates.


Subject(s)
Antigens, Viral/genetics , Influenza A Virus, H1N1 Subtype/immunology , Nucleosides/chemistry , Orthomyxoviridae Infections/prevention & control , Vaccines, Synthetic/administration & dosage , Animals , Antibodies, Viral/metabolism , Antigens, Viral/chemistry , Disease Models, Animal , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Injections, Intradermal , Liposomes , Mice , NIH 3T3 Cells , Nanoparticles , Neuraminidase/chemistry , Neuraminidase/genetics , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Orthomyxoviridae Infections/immunology , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology , mRNA Vaccines
3.
Mol Ther ; 21(8): 1570-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23799535

ABSTRACT

In recent years, RNA interference (RNAi) therapeutics, most notably with lipid nanoparticle-based delivery systems, have advanced into human clinical trials. The results from these early clinical trials suggest that lipid nanoparticles (LNPs), and the novel ionizable lipids that comprise them, will be important materials in this emerging field of medicine. A persistent theme in the use of materials for biomedical applications has been the incorporation of biodegradability as a means to improve biocompatibility and/or to facilitate elimination. Therefore, the aim of this work was to further advance the LNP platform through the development of novel, next-generation lipids that combine the excellent potency of the most advanced lipids currently available with biodegradable functionality. As a representative example of this novel class of biodegradable lipids, the lipid evaluated in this work displays rapid elimination from plasma and tissues, substantially improved tolerability in preclinical studies, while maintaining in vivo potency on par with that of the most advanced lipids currently available.


Subject(s)
Drug Delivery Systems , Gene Transfer Techniques , Lipids/chemistry , Nanoparticles/administration & dosage , RNA, Small Interfering/genetics , Animals , Cell Line , Factor VII/genetics , Factor VII/metabolism , Gene Silencing , Genetic Therapy , Humans , Lipids/pharmacokinetics , Macaca fascicularis , Male , Mice , Nanoparticles/chemistry , Nanoparticles/toxicity , RNA Interference , RNA, Small Interfering/chemistry , Rats
4.
Angew Chem Int Ed Engl ; 51(34): 8529-33, 2012 Aug 20.
Article in English | MEDLINE | ID: mdl-22782619

ABSTRACT

Special (lipid) delivery: The role of the ionizable lipid pK(a) in the in vivo delivery of siRNA by lipid nanoparticles has been studied with a large number of head group modifications to the lipids. A tight correlation between the lipid pK(a) value and silencing of the mouse FVII gene (FVII ED(50) ) was found, with an optimal pK(a) range of 6.2-6.5. The most potent cationic lipid from this study has ED(50) levels around 0.005 mg kg(-1) in mice and less than 0.03 mg kg(-1) in non-human primates.


Subject(s)
Gene Silencing , Lipids/administration & dosage , Liver/physiology , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Amines/chemistry , Animals , Female , Genetic Therapy/methods , Humans , Kinetics , Lipids/chemistry , Liposomes/administration & dosage , Liposomes/chemistry , Liver/metabolism , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , RNA, Small Interfering/chemistry
5.
Nat Commun ; 12(1): 3460, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34103491

ABSTRACT

Lack or dysfunction of the lymphatics leads to secondary lymphedema formation that seriously reduces the function of the affected organs and results in degradation of quality of life. Currently, there is no definitive treatment option for lymphedema. Here, we utilized nucleoside-modified mRNA encapsulated in lipid nanoparticles (LNPs) encoding murine Vascular Endothelial Growth Factor C (VEGFC) to stimulate lymphatic growth and function and reduce experimental lymphedema in mouse models. We demonstrated that administration of a single low-dose of VEGFC mRNA-LNPs induced durable, organ-specific lymphatic growth and formation of a functional lymphatic network. Importantly, VEGFC mRNA-LNP treatment reversed experimental lymphedema by restoring lymphatic function without inducing any obvious adverse events. Collectively, we present a novel application of the nucleoside-modified mRNA-LNP platform, describe a model for identifying the organ-specific physiological and pathophysiological roles of the lymphatics, and propose an efficient and safe treatment option that may serve as a novel therapeutic tool to reduce lymphedema.


Subject(s)
Lymphangiogenesis/genetics , Lymphatic Vessels/pathology , Lymphedema/pathology , Nucleosides/metabolism , Vascular Endothelial Growth Factor C/genetics , Animals , Blood Vessels/pathology , Cell Proliferation/drug effects , Diphtheria Toxin/pharmacology , Disease Models, Animal , HEK293 Cells , Humans , Immunity/drug effects , Injections, Intradermal , Lipids/administration & dosage , Lipids/chemistry , Lymphatic Vessels/drug effects , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Organ Specificity , Poly C/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tamoxifen/pharmacology , Vascular Endothelial Growth Factor C/administration & dosage , Vascular Endothelial Growth Factor C/metabolism
6.
Mol Ther Nucleic Acids ; 15: 36-47, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30974332

ABSTRACT

Despite the enormous effort in the development of effective vaccines against HIV-1, no vaccine candidate has elicited broadly neutralizing antibodies in humans. Thus, generation of more effective anti-HIV vaccines is critically needed. Here we characterize the immune responses induced by nucleoside-modified and purified mRNA-lipid nanoparticle (mRNA-LNP) vaccines encoding the clade C transmitted/founder HIV-1 envelope (Env) 1086C. Intradermal vaccination with nucleoside-modified 1086C Env mRNA-LNPs elicited high levels of gp120-specific antibodies in rabbits and rhesus macaques. Antibodies generated in rabbits neutralized a tier 1 virus, but no tier 2 neutralization activity could be measured. Importantly, three of six non-human primates developed antibodies that neutralized the autologous tier 2 strain. Despite stable anti-gp120 immunoglobulin G (IgG) levels, tier 2 neutralization titers started to drop 4 weeks after booster immunizations. Serum from both immunized rabbits and non-human primates demonstrated antibody-dependent cellular cytotoxicity activity. Collectively, these results are supportive of continued development of nucleoside-modified and purified mRNA-LNP vaccines for HIV. Optimization of Env immunogens and vaccination protocols are needed to increase antibody neutralization breadth and durability.

7.
J Control Release ; 291: 106-115, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30336167

ABSTRACT

Systemic administration of lipid nanoparticle (LNP)-encapsulated messenger RNA (mRNA) leads predominantly to hepatic uptake and expression. Here, we conjugated nucleoside-modified mRNA-LNPs with antibodies (Abs) specific to vascular cell adhesion molecule, PECAM-1. Systemic (intravenous) administration of Ab/LNP-mRNAs resulted in profound inhibition of hepatic uptake concomitantly with ~200-fold and 25-fold elevation of mRNA delivery and protein expression in the lungs compared to non-targeted counterparts. Unlike hepatic delivery of LNP-mRNA, Ab/LNP-mRNA is independent of apolipoprotein E. Vascular re-targeting of mRNA represents a promising, powerful, and unique approach for novel experimental and clinical interventions in organs of interest other than liver.


Subject(s)
Apolipoproteins E/metabolism , Drug Delivery Systems , Endothelium, Vascular/metabolism , Nanoparticles/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/administration & dosage , Administration, Intravenous , Animals , Cell Line , Drug Carriers/metabolism , Drug Delivery Systems/methods , Human Umbilical Vein Endothelial Cells , Humans , Immunoconjugates/metabolism , Mice, Inbred C57BL , RNA, Messenger/pharmacokinetics , Tissue Distribution
8.
Nat Commun ; 9(1): 3361, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30135514

ABSTRACT

Currently available influenza virus vaccines have inadequate effectiveness and are reformulated annually due to viral antigenic drift. Thus, development of a vaccine that confers long-term protective immunity against antigenically distant influenza virus strains is urgently needed. The highly conserved influenza virus hemagglutinin (HA) stalk represents one of the potential targets of broadly protective/universal influenza virus vaccines. Here, we evaluate a potent broadly protective influenza virus vaccine candidate that uses nucleoside-modified and purified mRNA encoding full-length influenza virus HA formulated in lipid nanoparticles (LNPs). We demonstrate that immunization with HA mRNA-LNPs induces antibody responses against the HA stalk domain of influenza virus in mice, rabbits, and ferrets. The HA stalk-specific antibody response is associated with protection from homologous, heterologous, and heterosubtypic influenza virus infection in mice.


Subject(s)
Antibodies, Viral/immunology , Hemagglutinins/immunology , Influenza A Virus, H5N1 Subtype/immunology , Orthomyxoviridae/immunology , RNA, Messenger/chemistry , RNA, Messenger/immunology , Animals , Cells, Cultured , Dogs , Enzyme-Linked Immunosorbent Assay , Female , Ferrets , Flow Cytometry , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Phylogeny , Rabbits
9.
J Exp Med ; 215(6): 1571-1588, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29739835

ABSTRACT

T follicular helper (Tfh) cells are required to develop germinal center (GC) responses and drive immunoglobulin class switch, affinity maturation, and long-term B cell memory. In this study, we characterize a recently developed vaccine platform, nucleoside-modified, purified mRNA encapsulated in lipid nanoparticles (mRNA-LNPs), that induces high levels of Tfh and GC B cells. Intradermal vaccination with nucleoside-modified mRNA-LNPs encoding various viral surface antigens elicited polyfunctional, antigen-specific, CD4+ T cell responses and potent neutralizing antibody responses in mice and nonhuman primates. Importantly, the strong antigen-specific Tfh cell response and high numbers of GC B cells and plasma cells were associated with long-lived and high-affinity neutralizing antibodies and durable protection. Comparative studies demonstrated that nucleoside-modified mRNA-LNP vaccines outperformed adjuvanted protein and inactivated virus vaccines and pathogen infection. The incorporation of noninflammatory, modified nucleosides in the mRNA is required for the production of large amounts of antigen and for robust immune responses.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/cytology , Nucleosides/metabolism , RNA, Messenger/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, Subunit/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Neutralizing/immunology , Antibody Formation/immunology , Antigens/metabolism , Lipids/chemistry , Macaca mulatta , Nanoparticles/chemistry , Protein Subunits/metabolism , Time Factors , Vaccination
10.
NPJ Vaccines ; 2: 29, 2017.
Article in English | MEDLINE | ID: mdl-29263884

ABSTRACT

mRNA represents a promising new vaccine technology platform with high flexibility in regard to development and production. Here, we demonstrate that vaccines based on sequence optimized, chemically unmodified mRNA formulated in optimized lipid nanoparticles (LNPs) are highly immunogenic and well tolerated in non-human primates (NHPs). Single intramuscular vaccination of NHPs with LNP-formulated mRNAs encoding rabies or influenza antigens induced protective antibody titers, which could be boosted and remained stable during an observation period of up to 1 year. First mechanistic insights into the mode of action of the LNP-formulated mRNA vaccines demonstrated a strong activation of the innate immune response at the injection site and in the draining lymph nodes (dLNs). Activation of the innate immune system was reflected by a transient induction of pro-inflammatory cytokines and chemokines and activation of the majority of immune cells in the dLNs. Notably, our data demonstrate that mRNA vaccines can compete with licensed vaccines based on inactivated virus or are even superior in respect of functional antibody and T cell responses. Importantly, we show that the developed LNP-formulated mRNA vaccines can be used as a vaccination platform allowing multiple, sequential vaccinations against different pathogens. These results provide strong evidence that the mRNA technology is a valid approach for the development of effective prophylactic vaccines to prevent infectious diseases.

11.
Nat Commun ; 8: 14630, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28251988

ABSTRACT

Monoclonal antibodies are one of the fastest growing classes of pharmaceutical products, however, their potential is limited by the high cost of development and manufacturing. Here we present a safe and cost-effective platform for in vivo expression of therapeutic antibodies using nucleoside-modified mRNA. To demonstrate feasibility and protective efficacy, nucleoside-modified mRNAs encoding the light and heavy chains of the broadly neutralizing anti-HIV-1 antibody VRC01 are generated and encapsulated into lipid nanoparticles. Systemic administration of 1.4 mg kg-1 of mRNA into mice results in ∼170 µg ml-1 VRC01 antibody concentrations in the plasma 24 h post injection. Weekly injections of 1 mg kg-1 of mRNA into immunodeficient mice maintain trough VRC01 levels above 40 µg ml-1. Most importantly, the translated antibody from a single injection of VRC01 mRNA protects humanized mice from intravenous HIV-1 challenge, demonstrating that nucleoside-modified mRNA represents a viable delivery platform for passive immunotherapy against HIV-1 with expansion to a variety of diseases.


Subject(s)
Antibodies, Neutralizing/genetics , HIV-1/drug effects , Nucleosides/chemistry , RNA, Messenger/administration & dosage , Animals , Antibodies, Monoclonal/genetics , Broadly Neutralizing Antibodies , Drug Administration Schedule , Female , HIV Antibodies/biosynthesis , HIV Infections/immunology , HIV Infections/therapy , HIV-1/immunology , Humans , Immunization, Passive , Lipids/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Nanoparticles/chemistry , RNA, Messenger/chemistry , RNA, Messenger/pharmacology , RNA, Messenger/therapeutic use
12.
Nucleic Acids Res ; 30(16): 3632-41, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12177306

ABSTRACT

Antisense oligonucleotides (ODN) targeted to specific genes have shown considerable potential as therapeutic agents. The polyanionic charges carried by these molecules, however, present a barrier to efficient cellular uptake and consequently their biological effects on gene regulation are compromised. To overcome this obstacle, a rationally designed carrier system is desirable for antisense delivery. This carrier should assist antisense ODN penetrate the cell membrane and, once inside the cell, then release the ODN and make them available for target binding. We have developed a carrier formulation employing programmable fusogenic vesicles (PFV) as the antisense delivery mediator. This study investigates the intracellular fate of PFV-ODN and bioavailability of antisense ODN to cells. The subcellular distribution of PFV and ODN was examined by monitoring the trafficking of FITC-labeled ODN and rhodamine/phosphatidylethanolamine (Rh-PE)-labeled PFV using confocal microscopy. Fluorescently tagged ODN were first co-localized with the liposomal carrier in the cytoplasm, presumably in endosome/lysosome compartments, shortly after incubation of PFV-ODN with HEK 293 and 518A2 cells. Between 24 and 48 h incubation, however, separation of FITC-ODN from the carrier and subsequent accumulation in the nucleus was observed. In contrast, the Rh-PE label was localized to the cell cytoplasm. The enhanced cellular uptake achieved using the PFV carrier, compared to incubation of free ODN with cells, and subsequent release of ODN from the carrier resulted in significant down-regulation of mRNA expression. Specifically, G3139, an antisense construct targeting the apoptotic antagonist gene bcl-2, was examined in the human melanoma cell line 518A2. Upon exposure to PFV-encapsulated G3139, cells displayed a time-dependent reduction in bcl-2 message levels. The bcl-2 mRNA level was reduced by 50% after 24 h treatment and by approximately 80% after 72 h when compared to cells treated with free G3139, empty PFV or PFV-G3622, a control ODN sequence. Our results establish that ODN can be released from PFV after intracellular uptake and can then migrate to the nucleus and selectively down-regulate target mRNA.


Subject(s)
Down-Regulation , Genes, bcl-2/genetics , Genetic Therapy/methods , Liposomes/metabolism , Melanoma/genetics , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Active Transport, Cell Nucleus , Cell Line , Cell Nucleus/metabolism , Cell Survival/drug effects , Cytoplasm/metabolism , Endosomes/metabolism , Humans , Liposomes/administration & dosage , Liposomes/chemistry , Liposomes/toxicity , Lysosomes/metabolism , Melanoma/metabolism , Melanoma/pathology , Microscopy, Confocal , Oligonucleotides, Antisense/metabolism , Oligonucleotides, Antisense/toxicity , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Time Factors , Tumor Cells, Cultured
13.
J Drug Target ; 24(9): 774-779, 2016 11.
Article in English | MEDLINE | ID: mdl-27588674

ABSTRACT

For the best part of 40 years, lipids and membrane fusion have been at the center of Pieter's research. Projects range from the purely academic quest of understanding the roles of lipids in biological membranes, to the translation of this knowledge into the most advanced lipid nanoparticle (LNP) drug delivery systems in clinical trials to-date. Pieter's pioneering work in lipid polymorphism and characterizing the unique properties of unsaturated phospatidyethanolamines (PE), together with the introduction of ionizable, dialkylamino lipids to trigger membrane fusion at acidic pH, provided the foundation on which a new generation of highly potent, well-tolerated LNPs for intravenous delivery of nucleic acid therapeutics has been built. In this contribution to the special edition honoring Pieter's achievements we highlight key research conducted in Pieter's laboratory and at several biotechnology companies, some spun out of his research group, which resulted in the development of a fusogenic delivery system for siRNA therapeutics. Patisiran®, an LNP encapsulating siRNA for hepatic gene silencing, is currently in Phase III clinical trials for treatment of Transthyretin amyloidosis as are several other siRNA products employing this delivery technology. Finally, we describe more recent work in which the platform shows real promise in the rapidly growing new field of mRNA therapeutics.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Lipids/chemistry , RNA, Messenger/pharmacology , RNA, Small Interfering/pharmacology , Humans , Nanoparticles , RNA, Messenger/chemistry , RNA, Small Interfering/chemistry
14.
Methods Enzymol ; 391: 40-57, 2005.
Article in English | MEDLINE | ID: mdl-15721373

ABSTRACT

Vincristine is a dimeric Catharanthus alkaloid derived from the Madagascan periwinkle that acts by binding to tubulin and blocking metaphase in actively dividing cells. While vincristine is widely used in the treatment of a number of human carcinomas, its use is associated with dose-limiting neurotoxicity, manifested mainly as peripheral neuropathy. It is known that the therapeutic activity of vincristine can be significantly enhanced after its encapsulation in appropriately designed liposomal systems. Enhanced efficacy is also associated with a slight decrease in drug toxicity. Thus, the therapeutic index of vincristine can be enhanced significantly through the use of a liposomal delivery system. Vincristine may be encapsulated into liposomes of varying lipid composition by several techniques, including passive loading, pH gradient loading, and ionophore-assisted loading. However, most research has focused on the encapsulation of vincristine in response to a transbilayer pH gradient, which actively concentrates the drug within the aqueous interior of the liposome. This chapter details the preparation and evaluation of liposomal vincristine. Specifically, we elaborate on the components (choice of lipids, molar proportions, etc.), methods (preparation of liposomes, drug loading methods, etc.), critical design features (size, surface charge, etc.), and key biological endpoints (circulation lifetime, bioavailability, efficacy measurements) important to the development of a formulation of vincristine with enhanced therapeutic properties.


Subject(s)
Antineoplastic Agents, Phytogenic , Drug Carriers , Liposomes , Vincristine , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Humans , Hydrogen-Ion Concentration , Ionophores/metabolism , Lipids/chemistry , Liposomes/chemical synthesis , Liposomes/chemistry , Molecular Structure , Neoplasms/drug therapy , Neoplasms/metabolism , Vincristine/administration & dosage , Vincristine/chemistry , Vincristine/pharmacokinetics , Vincristine/therapeutic use
15.
Methods Enzymol ; 391: 71-97, 2005.
Article in English | MEDLINE | ID: mdl-15721375

ABSTRACT

Doxorubicin is the best known and most widely used member of the anthracycline antibiotic group of anticancer agents. It was first introduced in the 1970s, and since that time has become one of the most commonly used drugs for the treatment of both hematological and solid tumors. The therapy-limiting toxicity for this drug is cardiomyopathy, which may lead to congestive heart failure and death. Approximately 2% of patients who have received a cumulative (lifetime) doxorubicin dose of 450-500 mg?m(2) will experience this condition. An approach to ameliorating doxorubicin-related toxicity is to use drug carriers, which engender a change in the pharmacological distribution of the drug, resulting in reduced drug levels in the heart. Examples of these carrier systems include lipid-based (liposome) formulations that effect a beneficial change in doxorubicin biodistribution, with two formulations approved for clinical use. Drug approval was based, in part, on data suggesting that beneficial changes in doxorubicin occurred in the absence of decreased therapeutic activity. Preclinical (animal) and clinical (human) studies showing that liposomes can preferentially accumulate in tumors have provided a rationale for improved activity. Liposomes represent ideal drug delivery systems, as the microvasculature in tumors is typically discontinuous, having pore sizes (100-780 nm) large enough for liposomes to move from the blood compartment into the extravascular space surrounding the tumor cells. Liposomes, in the size range of 100-200 nm readily extravasate within the site of tumor growth to provide locally concentrated drug delivery, a primary role of liposomal formulation. Although other liposomal drugs have been prepared and characterized due to the potential for liposomes to improve antitumor potency of the encapsulated drug, the studies on liposomal doxorubicin have been developed primarily to address issues of acute and chronic toxicity that occur as a consequence of using this drug. It is important to recognize that research programs directed toward the development of liposomal doxorubicin occurred concurrently with synthetic chemistry programs attempting to introduce safer and more effective anthracycline analogues. Although many of these drugs are approved for use, and preliminary liposomal formulations of these analogues have been prepared, doxorubicin continues to be a mainstay of drug cocktails used in the management of most solid tumors. It will be of great interest to observe how the approved formulations of liposomal doxorubicin are integrated into combination regimes for treatment of cancer. In the meantime, we have learned a great deal about liposomes as drug carriers from over 20 years of research on different liposomal doxorubicin formulations, the very first of which were identified in the late 1970s. This chapter will discuss the various methods for encapsulation of doxorubicin into liposomes, as well as some of the important interactions between the formulation components of the drug and how this may impact the biological activity of the associated drug. This review of methodology, in turn, will highlight research activities that are being pursued to achieve better performance parameters for liposomal formulations of doxorubicin, as well as other anticancer agents being considered for use with lipid-based carriers.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Doxorubicin/metabolism , Drug Carriers , Liposomes , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/chemistry , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Doxorubicin/chemistry , Humans , Ions , Liposomes/chemistry , Molecular Structure , Phospholipids/chemistry
16.
J Control Release ; 217: 345-51, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26264835

ABSTRACT

In recent years, in vitro transcribed messenger RNA (mRNA) has emerged as a potential therapeutic platform. To fulfill its promise, effective delivery of mRNA to specific cell types and tissues needs to be achieved. Lipid nanoparticles (LNPs) are efficient carriers for short-interfering RNAs and have entered clinical trials. However, little is known about the potential of LNPs to deliver mRNA. Here, we generated mRNA-LNPs by incorporating HPLC purified, 1-methylpseudouridine-containing mRNA comprising codon-optimized firefly luciferase into stable LNPs. Mice were injected with 0.005-0.250mg/kg doses of mRNA-LNPs by 6 different routes and high levels of protein translation could be measured using in vivo imaging. Subcutaneous, intramuscular and intradermal injection of the LNP-encapsulated mRNA translated locally at the site of injection for up to 10days. For several days, high levels of protein production could be achieved in the lung from the intratracheal administration of mRNA. Intravenous and intraperitoneal and to a lesser extent intramuscular and intratracheal deliveries led to trafficking of mRNA-LNPs systemically resulting in active translation of the mRNA in the liver for 1-4 days. Our results demonstrate that LNPs are appropriate carriers for mRNA in vivo and have the potential to become valuable tools for delivering mRNA encoding therapeutic proteins.


Subject(s)
Luciferases, Firefly/metabolism , Nanoparticles/administration & dosage , Pseudouridine/analogs & derivatives , RNA, Messenger/administration & dosage , Animals , Cells, Cultured , Dendritic Cells/metabolism , Female , HEK293 Cells , Humans , Kinetics , Luciferases, Firefly/genetics , Lung/metabolism , Mice, Inbred BALB C , Nanoparticles/chemistry , Phosphatidylethanolamines/chemistry , Pseudouridine/chemistry , RNA, Messenger/chemistry , RNA, Messenger/pharmacokinetics , Transfection/methods
18.
Mol Ther Nucleic Acids ; 2: e139, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24345865

ABSTRACT

Lipid nanoparticles (LNPs) encapsulating short interfering RNAs that target hepatic genes are advancing through clinical trials, and early results indicate the excellent gene silencing observed in rodents and nonhuman primates also translates to humans. This success has motivated research to identify ways to further advance this delivery platform. Here, we characterize the polyethylene glycol lipid (PEG-lipid) components, which are required to control the self-assembly process during formation of lipid particles, but can negatively affect delivery to hepatocytes and hepatic gene silencing in vivo. The rate of transfer from LNPs to plasma lipoproteins in vivo is measured for three PEG-lipids with dialkyl chains 14, 16, and 18 carbons long. We show that 1.5 mol % PEG-lipid represents a threshold concentration at which the chain length exerts a minimal effect on hepatic gene silencing but can still modify LNPs pharmacokinetics and biodistribution. Increasing the concentration to 2.5 and 3.5 mol % substantially compromises hepatocyte gene knockdown for PEG-lipids with distearyl (C18) chains but has little impact for shorter dimyristyl (C14) chains. These data are discussed with respect to RNA delivery and the different rates at which the steric barrier disassociates from LNPs in vivo.Molecular Therapy-Nucleic Acids (2013) 2, e139; doi:10.1038/mtna.2013.66; published online 17 December 2013.

19.
Nat Biotechnol ; 28(2): 172-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20081866

ABSTRACT

We adopted a rational approach to design cationic lipids for use in formulations to deliver small interfering RNA (siRNA). Starting with the ionizable cationic lipid 1,2-dilinoleyloxy-3-dimethylaminopropane (DLinDMA), a key lipid component of stable nucleic acid lipid particles (SNALP) as a benchmark, we used the proposed in vivo mechanism of action of ionizable cationic lipids to guide the design of DLinDMA-based lipids with superior delivery capacity. The best-performing lipid recovered after screening (DLin-KC2-DMA) was formulated and characterized in SNALP and demonstrated to have in vivo activity at siRNA doses as low as 0.01 mg/kg in rodents and 0.1 mg/kg in nonhuman primates. To our knowledge, this represents a substantial improvement over previous reports of in vivo endogenous hepatic gene silencing.


Subject(s)
Drug Carriers/chemistry , Drug Compounding/methods , Drug Design , Lipids/chemistry , RNA, Small Interfering/chemistry , Transfection/methods , Cations , RNA, Small Interfering/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL