US20050142114A1 - Targeted lipid-drug formulations for delivery of drugs to myeloid and lymphoid immune cells - Google Patents

Targeted lipid-drug formulations for delivery of drugs to myeloid and lymphoid immune cells Download PDF

Info

Publication number
US20050142114A1
US20050142114A1 US10/943,758 US94375804A US2005142114A1 US 20050142114 A1 US20050142114 A1 US 20050142114A1 US 94375804 A US94375804 A US 94375804A US 2005142114 A1 US2005142114 A1 US 2005142114A1
Authority
US
United States
Prior art keywords
drug
hiv
cell
targeting
liposomes
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/943,758
Other languages
English (en)
Inventor
Robert Gieseler
Guido Marquitan
Michael Scolaro
Sean Sullivan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
RODOS BIOTARGET GmbH
Original Assignee
LET THERE BE HOP MEDICAL RESEARCH INSTITUTE
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by LET THERE BE HOP MEDICAL RESEARCH INSTITUTE filed Critical LET THERE BE HOP MEDICAL RESEARCH INSTITUTE
Priority to US10/943,758 priority Critical patent/US20050142114A1/en
Assigned to LET THERE BE HOP MEDICAL RESEARCH INSTITUTE reassignment LET THERE BE HOP MEDICAL RESEARCH INSTITUTE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SULLIVAN, SEAN M., GIESELER, ROBERT K., MARQUITAN, GUIDO, SCOLARO, MICHAEL J.
Publication of US20050142114A1 publication Critical patent/US20050142114A1/en
Assigned to FOUNDERS reassignment FOUNDERS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LET THERE BE HOPE MEDICAL RESEARCH INSTITUTE
Assigned to RODOS BIOTARGET GMBH reassignment RODOS BIOTARGET GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FOUNDERS
Priority to US14/193,498 priority patent/US20140234400A1/en
Priority to US15/154,097 priority patent/US10357457B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/168Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/085Staphylococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • A61K47/6913Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome the liposome being modified on its surface by an antibody
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators

Definitions

  • the present invention relates to the medical arts, and in particular, to targeted liposomal drug delivery.
  • My-DCs Myeloid dendritic cells
  • My-DCs belong to the most potent group of professional antigen-presenting cells, with the unique ability to induce primary cellular and humoral immune responses (reviewed in Banchereau J, Paczesny S, Blanco P, Bennett L, Pascual V, Fay J, Palucka A K, Dendritic cells: controllers of the immune system and a new promise for immunotherapy , Ann N Y Acad Sci 987:180-7 [2003]).
  • These cells, within the lymphoid organs and structures, are also an important component of the HIV reservoir, together with other major sanctuary populations, i.e. follicular dendritic cells, macrophages, resting/memory T cells, and cells within the central nervous system.
  • DC-SIGN My-DC-specific intercellular adhesion molecule 3-grabbing nonintegrin
  • DC-SIGN a novel HIV receptor on DCs that mediates HIV -1 transmission , Curr Top Microbiol Immunol 276:31-54 [2003]).
  • DC-SIGN is not only expressed by myeloid DCs, but also by subpopulations of macrophages, which are another main group of HIV reservoir cells (Soilleux E J et al., Constitutive and induced expression of DC - SIGN on dendritic cell and macrophage subpopulations in situ and in vitro , J Leukoc Biol. 71(3):445-57 [2002]).
  • DC-SIGN is an endocytic adhesion receptor.
  • DC-SIGN-attached particles are shuttled into the MHC class II antigen processing and presentation pathway and are accessed to the mechanism generating T-cell immunity (as desirable in case of any viral infection), as well as B-cell immunity (as supportive in the clearance of virus, by mechanisms secondary to the generation of antibodies, such as Fc receptor-mediated phagocytosis or, in case of cytotoxic antibodies, complement-mediated lysis)
  • T-cell immunity as desirable in case of any viral infection
  • B-cell immunity as supportive in the clearance of virus, by mechanisms secondary to the generation of antibodies, such as Fc receptor-mediated phagocytosis or, in case of cytotoxic antibodies, complement-mediated lysis
  • Schjetne K W et al Mouse C ⁇ - specific T cell clone indicates that DC - SIGN is an efficient target for antibody-mediated delivery of T cell epitopes for MHC class II presentation , Int Immunol 14(12):1423-30 [2002]; Engering, A et al., The
  • Th-cell infection by MyDCs with HIV-1 is a two-phased process that depends on the DCs' developmental stage, including both directional transport of virus to the immunological synapse, as well as active de-novo synthesis of HIV-1 from proviral DNA (Turville S G, Santos J J, Frank I et al. Immunodeficiency virus uptake, turnover, and two - phase transfer in human dendritic cells , Blood; online publication ahead of print: DOI 10.1182/blood-2003-09-3129 [2003]).
  • DC-SIGN a dendritic cell - specific HIV -1- binding protein that enhances trans - infection of T cells , Cell;100:587-97 [2000]
  • HAART Highly Active Antiretroviral Therapy
  • HIV-1 the capacity of HIV-1 to establish latent infection allows viral particles to persist in tissues despite immune responses and antiretroviral therapy (Gangne J-F, Désormeaux A, Perron S, Tremblay M. J, Bergeron M. G, Targeted delivery of indinavir to HIV -1 primary reservoirs with immunoliposomes , Biochim Biophys Acta, 1558: 198-210 [2002]). It is hypothesized that the susceptibility of dendritic cells to being infected with HIV, together with their crucial immunologic function, leads to the continuous spread of HIV.
  • Liposomes are a suitable vehicle for specifically delivering encapsulated compounds to any given cell type, provided the existence of an appropriate targeting structure. Because of its highly restricted cellular expression, DC-SIGN qualifies as such a targeting molecule.
  • DC-SIGN qualifies as such a targeting molecule.
  • lipid-drug complexes have long been seen as a potential way to improve the Therapeutic Index (TI) of drugs by increasing their localization to specific organs, tissues or cells.
  • the TI is the ratio between the median toxic dose (TD50) and the median effective dose (ED50) of a particular drug.
  • TD50 median toxic dose
  • ED50 median effective dose
  • FDA U.S. Food and Drug Administration
  • lipid constituent can vary, many formulations use synthetic products of natural phospholipid, mainly phosphatidylcholine. Most of the liposome formulations approved for human use contain phosphatidylcholine (neutral charge), with fatty acyl chains of varying lengths and degrees of saturation, as a major membrane building block. A fraction of cholesterol ( ⁇ 30 mol %) is often included in the lipid formulation to modulate rigidity and to reduce serum-induced instability caused by the binding of serum proteins to the liposome membrane.
  • liposomes Based on the head group composition of the lipid and the pH, liposomes can bear a negative, neutral, or positive charge on their surface.
  • the nature and density of charge on the surface of the liposomes influences stability, kinetics, and extent of biodistribution, as well as interaction with and uptake of liposomes by target cells.
  • Liposomes with a neutral surface charge have a lower tendency to be cleared by cells of the reticuloendothelial system (RES) after systemic administration and the highest tendency to aggregate.
  • RES reticuloendothelial system
  • negatively charged liposomes reduce aggregation and have increased stability in suspension, their nonspecific cellular uptake is increased in vivo.
  • Negatively charged liposomes containing phosphatidylserine (PS) or phosphatidylglycerol (PG) were observed to be endocytosed at a faster rate and to a greater extent than neutral liposomes (Allen T M, et al., Liposomes containing synthetic lipid derivatives of poly ( ethylene glycol ) show prolonged circulation half - lives in vivo , Biochim Biophys Acta 1066:29-36 [1991]; Lee R J, et al., Folate - mediated tumor cell targeting of liposome - entrapped doxorubicin in vitro , Biochim. Biophys. Acta 1233:134-144 [1995]).
  • PS phosphatidylserine
  • PG phosphatidylglycerol
  • Negative surface charge is recognized by a variety of receptors on various cell types, including macrophages (Allen T M et al. [1991]; Lee R J, et al., Delivery of liposomes into cultured KB cells via folate receptor - mediated endocytosis , J Biol Chem 269:3198-3204 [1994]).
  • glycolipids such as the ganglioside GM 1 or phosphotidylinositol (PI)
  • PI phosphotidylinositol
  • cationic liposomes often used as a DNA condensation reagent for intracellular DNA delivery in gene therapy, have a high tendency to interact with serum proteins; this interaction results in enhanced uptake by the RES and eventual clearance by lung, liver, or spleen This mechanism of RES clearance partly explains the low in vivo transfection efficiency. Other factors, including DNA instability, immune-mediated clearance, inflammatory response, and tissue accessibility can also contribute to low transfection efficiency in animals.
  • the surface of the liposome membrane can be modified to reduce aggregation and avoid recognition by the RES using hydrophilic polymers. This strategy is often referred to as surface hydration or steric modification. Surface modification is often done by incorporating gangliosides, such as GM 1 , or lipids that are chemically conjugated to hygroscopic or hydrophilic polymers, usually polyethyleneglycol (PEG). This technology is similar to protein PEGylation.
  • PEG polyethyleneglycol
  • PEG is conjugated to the terminal amine of phosphatidylethanolamine.
  • Plasma proteins that can interact with liposomes include albumin, lipoproteins (i.e., high-density lipoprotein [HDL], low-density lipoprotein [LDL], etc.) and cell-associated proteins. Some of these proteins (e.g., HDL) can remove phospholipids from the liposome bilayer, thereby destabilizing the liposomes. This process may potentially lead to a premature leakage or dissociation of drugs from liposomes.
  • HDL high-density lipoprotein
  • LDL low-density lipoprotein
  • liposomes an invaluable drug delivery system
  • One of the key properties that make liposomes an invaluable drug delivery system is their ability to modulate the pharmacokinetics of liposome-associated and encapsulated drugs
  • Allen T M Hansen C, Martin F, Redemann C, Yau-Young A, Liposomes containing synthetic lipid derivatives of poly ( ethylene glycol ) show prolonged circulation half - lives in vivo , Biochim Biophys Acta;1066(1):29-36 [1991]
  • Allen T M Austin G A, Chonn A, Lin L, Lee K C, Uptake of liposomes by cultured mouse bone marrow macrophages: influence of liposome composition and size
  • circulation time can be increased by reducing the liposome size and modifying the surface/steric effect with PEG derivatives.
  • liposomes with membranes engineered for sufficient stability escaping clearance by the RES are now available. Therefore, long-circulation liposomes that also significantly reduce toxicological profiles of the respective drugs can be used to maintain and extend plasma drug levels. Even though only a small fraction of liposomes eventually accumulate at target sites, prolonged circulation can indirectly enhance accumulation of liposome-associated drugs to targeted tissues.
  • Anti-HIV drugs such as nucleoside analogs (e.g., dideoxynucleoside derivatives, including 3′-azido-3′-deoxythymidine [AZT], ddC, and ddI), protease inhibitors, or phosphonoacids (e.g., phosphonoformic and phosphonoacetic acids), have previously been lipid-derivatized or incorporated into liposomes (e.g., Hostetler, K Y et al., Methods of treating viral infections using antiviral liponucleotides , Ser. No. 09/846,398, US 2001/0033862; U.S. Pat. No.
  • nucleoside analogs e.g., dideoxynucleoside derivatives, including 3′-azido-3′-deoxythymidine [AZT], ddC, and ddI
  • protease inhibitors e.g., phosphono
  • Such vectors molecules include so-called protein transduction domains (PTDs), which are derived from various viruses or from Drosophila antennapedia .
  • PTDs protein transduction domains
  • HIV Tat and its derivatives which act as PTDs (e.g., Schwarze, S. R., et al., In vivo protein transduction: delivery of a biologically active protein into the mouse , Science 285:1569-72 [1999]).
  • Anti-HIV drugs have been encapsulated in the aqueous core of immunoliposomes, which include on their external surfaces antigen-specific targeting ligands (e.g., Bergeron, M G. et al, Targeting of infectious agents bearing host cell proteins , WO 00/66173 A3; Bergeron, M G. et al., Liposomes encapsulating antiviral drugs , U.S. Pat. No. 5,773,027; Bergeron, M G.
  • antigen-specific targeting ligands e.g., Bergeron, M G. et al, Targeting of infectious agents bearing host cell proteins , WO 00/66173 A3; Bergeron, M G. et al., Liposomes encapsulating antiviral drugs , U.S. Pat. No. 5,773,027; Bergeron, M G.
  • DOXIL antibody-targeted liposome
  • Mo-DCs monocyte-derived dendritic cells
  • the present invention provides a liposomal delivery system that facilitates the targeting of active agents, such as drugs, immunomodulators, lectins or other plant-derived substances specifically to myeloid cell populations of interest.
  • active agents such as drugs, immunomodulators, lectins or other plant-derived substances specifically to myeloid cell populations of interest.
  • the present invention therefore addresses, inter alia, the need to target the reservoirs of HIV, hepatitis C virus (HCV) in myeloid cells, particularly dendritic cells and macrophages, as well as follicular dendritic cells of myeloid origin, of persons infected with HIV and those suffering from AIDS, or persons infected or co-infected with HCV and those suffering from HCV-dependent pathologic alterations of the liver.
  • the present invention may allow for indirect targeting of lymphoid cells, particularly T cells, upon their physical interaction with myeloid cells.
  • the present invention may allow for the specific elimination, or down-modulation, of malignant tumor cells or immune cells mediating autoimmunity; the enhancement of DC-dependent autologous tumor immunization; the therapeutic down-regulation of autoimmune diseases; or the DC-tropic stimulation of specific adaptive immunity (both in terms of vaccination or treatment) against common pathogens, or pathogens potentially employed as agents of bioterrorism, for which there currently exists no efficient protection.
  • the present invention may also allow for biotechnological advancement, such as, inter alia, by targeting DCs for increasing the production of monoclonal antibodies, or by allowing for the production of such immunoglobulins that cannot be induced in the absence of inductive liposomal DC targeting.
  • the present invention relates to a method of preferentially, or “actively,” targeting and delivering an active agent, such as a drug, to a mammalian immune cell, in vivo or in vitro.
  • the present invention is directed to a method of preferentially targeting a liposome to a mammalian immune cell such as a myeloid progenitor cell, a monocyte, a dendritic cell, a macrophage or a T-lymphocyte.
  • a mammalian immune cell such as a myeloid progenitor cell, a monocyte, a dendritic cell, a macrophage or a T-lymphocyte.
  • the method involves administering to the immune cell, in vitro or in vivo, a liposome comprising an active agent and further comprising an outer surface that comprises at least one targeting ligand that specifically binds a marker on the surface of the immune cell, such as CD209 (DC-SIGN), CD45RO, CD4, or HLA class II.
  • DC-SIGN CD209
  • CD45RO CD45RO
  • CD4 HLA class II
  • the present invention is also particularly directed to a method of preferentially delivering a drug to an immune cell of a mammalian subject, including a human.
  • the targeted immune cells include myeloid progenitor cells, monocytes, dendritic cells, macrophages or T-lymphocytes.
  • the method involves injecting into the mammalian subject a lipid-drug complex, for example, but not limited to a liposome that comprises the drug and further comprises an outer surface comprising at least one targeting ligand that specifically binds a marker on the surface of the immune cell, such as, but not limited, to CD209 (DC-SIGN), the immune cell being infected with, or susceptible to infection with, an infectious agent, such as, but not limited to, human immunodeficiency virus, types 1 and 2 (HIV-1; HIV-2).
  • a lipid-drug complex for example, but not limited to a liposome that comprises the drug and further comprises an outer surface comprising at least one targeting ligand that specifically binds a marker on the surface of the immune cell, such as, but not limited, to CD209 (DC-SIGN), the immune cell being infected with, or susceptible to infection with, an infectious agent, such as, but not limited to, human immunodeficiency virus, types 1 and 2 (HIV-1; HIV-
  • the present invention is also directed to inventive targeted liposomes.
  • One embodiment of the targeted liposome comprises on its external surface a targeting ligand that specifically binds CD209.
  • Another embodiment of the targeted liposome comprises on its external surface a targeting ligand that specifically binds CD209 and a targeting ligand that specifically binds CD4.
  • the inventive targeted liposomes are useful for targeting immune cells, such as dendritic cells.
  • the presence of HIV-1 in reservoir cells leads to the continuous de-novo infection of na ⁇ ve T cells within the lymphoid organs and tissues of an infected person. It has been hypothesized that eradication of such sanctuary sites may eventually eliminate HIV-1 from the individual.
  • the present invention provides a targeting system which, via targeting ligands such as the dendritic cell-specific molecule DC-SIGN, delivers chemical compounds directly into these cells.
  • the present invention is particularly, but not exclusively, of benefit for delivering antiviral drugs, packaged in immunoliposomes, to myeloid- and lymphoid-derived immune cells harboring HIV-1 or HIV-2, such as the HIV reservoir in dendritic cells.
  • Another benefit of the present invention by actively targeting immune cells, is in providing vaccination strategies against HIV (e.g., Steinman R M, Granelli-Piperno A, Pope M, Trumpfheller C, Ignatius R, Arrode G, Racz P, Tenner-Racz K, The interaction of immunodeficiency viruses with dendritic cells , Curr Top Microbiol Immunol 276:1-30 [2003]; Pope M, Dendritic cells as a conduit to improve HIV vaccines , Curr Mol Med 3:229-42 [2003]).
  • vaccination strategies against HIV e.g., Steinman R M, Granelli-Piperno A, Pope M, Trumpfheller C, Ignatius R, Arrode G, Racz P, Tenner-Racz K, The interaction of immunodeficiency viruses with dendritic cells , Curr Top Microbiol Immunol 276:1-30 [2003]; Pope M, Dendritic cells as a conduit to improve HIV vaccines
  • Additional benefits provided by the present invention include utility in the treatment of conditions involving abnormal proliferation of immune cells, e.g., primary and metastatic lymphoid cancers (lymphomas and leukemias), solid tumors or their post-surgical remnants, or autoimmune diseases, including specifically targeting immune cells in gene therapy applications.
  • the present invention also provides a way to target dendritic cells for facilitating the production of anti-infective vaccines, anti-bioterrorism vaccines, anti-cancer vaccines, or biotechnological and therapeutic tools such as monoclonal antibodies.
  • the present invention is also directed to variations on the inventive targeted delivery system.
  • Any type of cell residing within any kind of organ system such as the endocrine or the nervous systems
  • any type of anatomic entity such as the urogenital or the respiratoy tracts
  • FIG. 1 shows time-dependent targeting of calcein-labeled liposomes to Mo-DCs mediated by DC-SIGN or other targeting ligands, including bispecific combinations.
  • the column entitled “Antigen Expression” shows phenotypic expression of the respective marker(s), as tested with the mAbs only. Detection was by flow cytometry with a mAb-conjugated fluorescent dye, fluorescein-5-iothiocyanate (FITC); the column “LS Binding/Uptake” shows successful targeting and uptake, as evidenced by intracellular delivery of a liposome-encapsulated fluorescent dye, calcein.
  • FITC fluorescein-5-iothiocyanate
  • FIG. 2 shows monospecific liposomal targeting with respect to kinetics and efficacy.
  • Mature MoDCs were generated according to protocol described herein and investigated for uptake of different constructs of targeted protein A liposomes furnished with mAbs directed against CD4, CD14, CD45R0 or CD209. The MoDCs were co-incubated with the liposomes for 1, 3 or 24 h and then harvested and tested by flow cytometry. Control mAbs were used to detect cellular surface expression of the respective antigens (column headed “Marker Expression”). Empty curves indicate isotype controls; shaded curves indicate test conditions. The two panels bearing bold crosses show the highest mean fluorescence intensities, indicating the highest rates of calcein uptake.
  • FIG. 3 illustrates liposomal targeting of DCs via two cell markers (termed bispecific targeting), including time dependency of the targeting efficacy over a 24-h period.
  • Mature MoDCs were generated according to protocol described herein and investigated for uptake of different constructs of targeted Protein A liposomes bearing 2-member combinations of anti-CD4, anti-CD45R0 and anti-CD209 mabs. The MoDCs were co-incubated with the liposomes for 1, 3 or 24 h and then harvested and tested by flow cytometry. Control mAbs were used to detect cellular surface expression of the respective antigens (column headed “Marker Expression”). Empty curves indicate isotype controls; shaded curves indicate test conditions.
  • FIG. 3A shows results for the combination of anti-CD4 plus anti-CD45R0 targeting ligands.
  • FIG. 3B shows results for the combination of anti-CD4 plus anti-CD209 targeting ligands.
  • FIG. 3C shows results for the combination of anti-CD45R0 plus anti-CD209 targeting ligands.
  • FIG. 4 illustrates calculated values for targeting and surface binding of immunoliposomes applied to MoDCs.
  • FIG. 5 shows surface binding vs. internalization of targeted liposomes as determined by fluorescence microscopy.
  • Original magnifications ⁇ 1000 (panels 1 and 2 ) and ⁇ 400 (panels 3 - 8 ).
  • the present invention relates to a method of preferentially delivering an active agent, such as a drug, to a mammalian immune cell.
  • an active agent such as a drug
  • delivery is in vitro, and in other embodiments delivery of the active agent is in vivo.
  • lipid-drug complex or the liposome
  • the active agent e.g., the drug
  • the targeted immune cells include myeloid progenitor cells, monocytes, dendritic cells (DCs), macrophages, and T-lymphocytes.
  • Monocytes are one of the types of cells produced by the myeloid differentiation lineage of the bone marrow. It has been shown that DCs can likewise be derived from monocytes or other types of cells, i.e. mainly progenitor cells, generated within the myeloid lineage (e.g., Peters J H, Ruhl S, Friedrichs D, Veiled accessory cells deduced from monocytes , Immunobiology 176(1-2):154-66 [1987]; Gieseler R, Heise D, Soruri A, Schwartz P, Peters J H, In - vitro differentiation of mature dendritic cells from human blood monocytes , Dev. Immunol.
  • progenitor cells e.g., Peters J H, Ruhl S, Friedrichs D, Veiled accessory cells deduced from monocytes , Immunobiology 176(1-2):154-66 [1987]
  • Gieseler R Heise D, Soruri A, Schwartz P
  • a dendritic cell includes a “myeloid dendritic cell” (My-DC), i.e., a “myeloid lineage-derived DC”, which includes a monocyte-derived dendritic cell (Mo-DCs) as well as other DC types such as, for example, promonocyte-derived dendritic cells.
  • My-DC myeloid dendritic cell
  • Mo-DCs monocyte-derived dendritic cell
  • promonocyte-derived dendritic cells e.g., Steinbach F, Gieseler R, Soruri A, Krause B, Peters J H, Myeloid DCs deduced from monocytes, In - vitro and in - vivo data support a monocytic origin of DCs , Adv Exp Med Biol. 1997;417:27-32 [1997]).
  • a dendritic cell also includes a “lymphoid dendritic cell” (Ly-DC), i.e., a “lymphoid lineage-derived DC”; the only type of DC presently known to derive from the lymphoid lineage is the plasmacytoid dendritic cell (pc-DC) (Facchetti F, Vermi W, Mason D, Colonna M, The plasmacytoid monocyte/interferon producing cells , Virchows Arch;443(6):703-17. Epub 2003 Oct. 28 [2003]).
  • a dendritic cell also includes a follicular dendritic cell (FDC).
  • a macrophage denotes a cell class comprising various organ-resident subtypes further including macrophages more typical of lymphoid or of non-lymphoid organs and tissues (e.g., Barreda D R, Hanington P C, Belosevic M, Regulation of myeloid development and function by colony stimulating factors , Dev Comp Immunol 3;28(5):509-54 [2004]).
  • a T-lymphocyte includes, but is not limited to, a T-helper cell or a T-memory cell (Woodland D L, Dutton R W, Heterogeneity of CD 4 + and CD 8 + T cells , Curr Opin Immunol;15(3):336-42 [2003]).
  • lipid-drug complex is injected into the mammalian subject, in which the immune cell is present.
  • the immune cell is infected with, or susceptible to infection with, an infectious agent, such as a virus, a bacterium, a fungus, a protozoan, or a prion.
  • infectious agents such as HIV-1 and HIV-2 (including all their clades), HSV, EBV, CMV, Ebola and Marburg virus, HAV, HBV, HCV and HPV.
  • the immune cell is, in the presence or absence of infection, associated with the occurrence of an organ-specific or a systemic autoimmune disease.
  • diseases entities are Graves' disease; thyroid-associated ophthalmopathy (a.k.a. Graves' ophthalmopathy; a.k.a. endocrine ophthalmopathy); and multiple sclerosis (a.k.a. MS).
  • a “complex” is a mixture or adduct resulting from chemical binding or bonding between and/or among its constituents or components, including the lipid, drug, and other optional components of the inventive lipid-drug complex.
  • Chemical binding or bonding can have the nature of a covalent bond, ionic bond, hydrogen bond, van der Waal's bond, hydrophobic bond, or any combination of these bonding types linking the constituents of the complex at any of their parts or moieties, of which a constituent can have one or a multiplicity of moieties of various sorts. Not every constituent of a complex needs to be bound to every other constituent, but each constituent has at least one chemical bond with at least one other constituent of the complex.
  • examples of lipid-drug complexes include liposomes (lipid vesicles), or lipid-drug sheet disk complexes.
  • Lipid-conjugated drugs can also be a part of the lipid-drug complex in accordance with the invention
  • drugs can also be associated with a lipid or a lipid complex in the absence of any type of chemical binding or bonding, such as is provided in the case of liposomes encapsulating a soluble drug in their aqueous interior space.
  • the lipid drug complex e.g., the liposome, comprises an active agent, such as a drug.
  • the drug is any drug known to be active against cellular proliferation or active against an infectious agent of interest.
  • the active agent, or drug can be an anti-viral drug or virostatic agent, such as, interferon, a nucleoside analog, or a non-nucleoside anti-viral drug.
  • anti-viral drugs e.g., a HIV reverse protease inhibitor
  • indinavir a.k.a.
  • the anti-HIV drug can also be HIV-specific small interfering RNA (siRNA), anti-sense or sense DNA or RNA molecules.
  • the active agent is an anticancer drug, an antifungal drug, or an antibacterial drug.
  • the active agent is an immunomodulatory agent (i.e., an immunoactivator, an immunogen, an immunosuppressant, or an anti-inflammatory agent), such as cyclosporin, steroids and steroid derivatives.
  • useful drugs include therapeutic cytotoxic agents (e.g., cisplatin, carboplatin, methotrexate, 5-fluorouracil, and amphotericin), naked DNA expression vectors, therapeutic proteins, therapeutic oligonucleotides or nucleotide analogs, interferons, cytokines, or cytokine agonists or antagonists.
  • cytotoxic alkylating agent such as, but not limited to, busulfan (1,4-butanediol dimethanesulphonate; Myleran, Glaxo Wellcome), chlorambucil, cyclophosphamide, melphalan, or ethyl ethanesulfonic acid
  • busulfan 1,4-butanediol dimethanesulphonate
  • Myleran Glaxo Wellcome
  • chlorambucil chlorambucil
  • cyclophosphamide cyclophosphamide
  • melphalan or ethyl ethanesulfonic acid
  • Such drugs or agents are particularly useful in treating conditions involving pathological proliferation of immune cells, for example, lymphoid cancers or autoimmune diseases.
  • the active agent is a natural substance with therapeutic properties or benefits, such as plant-derived substances in purified or recombinant form.
  • plant-derived substances include leaf extract IDS 30, rhizome derived UDA lectin, and MHL.
  • the present invention contemplates the selective employment of natural substances that have been long acknowledged for their therapeutic properties and potentials in many cultures worldwide.
  • One of such plant-derived substances salicylic acid, which is found at varying concentrations in the bark of many trees, has served as the starter substance for one of nowadays great remedies, acetyl salicylic acid (ASS), or Aspirin, respectively.
  • ASS acetyl salicylic acid
  • Aspirin Aspirin
  • the stinging nettle Urtica dioica
  • Urtica dioica is a prominent example from the numerous plants that have been known for centuries to have great therapeutic benefits. Recent scientific investigation concerning the action of some of the components of Urtica dioica provides an opportunity for their targeted application.
  • MyDCs play an important role in the initiation of rheumatoid arthritis (RA) which is an example for a disease crossing the border between autoimmune and inflammatory conditions.
  • RA rheumatoid arthritis
  • Broer and Behnke have shown that the Urtica dioica leaf extract IDS 30 (Hox- ⁇ ), which has been recommended for adjuvant therapy of RA, prevents the phenotypic/functional maturation of MyDCs; diminishes the secretion of tumor necrosis factor- ⁇ ; and reduces the T cell-stimulating capacity of MyDCs, while it dose-dependently increases the expression of chemokine receptor 5 and CD36 as well as the endocytic capacity of these cells.
  • Hox- ⁇ Urtica dioica leaf extract IDS 30
  • IDS 30 may contribute to its therapeutic effect on T cell-mediated autoimmune/inflammatory diseases such as RA (Broer J, Behnke B, Immunosuppressant effect of IDS 30 , a stinging nettle leaf extract, on myeloid dendritic cells in vitro , J Rheumatol;29(4):659-66 [2002]).
  • RA autoimmune/inflammatory diseases
  • Lectins are another example of a natural substance that has therapeutic properties and potentials.
  • Lectins i.e., carbohydrate-binding proteins with agglutinating properties
  • Lectins are produced by a number of plants, mainly in their roots or rhizomes, as vital components of their own immune systems. Shibuya et al. first described the sugar-binding properties of the stinging nettle lectin (Shibuya N, Goldstein I J, Shafer J A, Peumans W J, Broekaert W F, (Carbohydrate binding properties of the stinging nettle ( Urtica dioica ) rhizome lectin, Arch Biochem Biophys;249(1):215-24 [1986]).
  • Urtica dioica agglutinin UAA
  • Urtica dioica agglutinin UAA
  • UDA Urtica dioica agglutinin
  • EC50 ranging from 0.3 to 9 ⁇ g/ml as well as syncytium formation between persistently HIV-1- and HIV-2-infected HUT-78 cells and CD4 + .
  • Molt/4 (clone 8) cells (EC50: 0.2-2 ⁇ g/ml).
  • UDA may act as a virion/target cell fusion inhibitor
  • Bozarini J Neyts J, Schols D, Hosoya M, Van Damme E, Peumans W, De Clercq E.
  • the mannose-specific plant lectins from Cymbidium hybrid and Epipactis helleborine and the (N-acetylglucosamine)n-specific plant lectin from Urtica dioica are potent and selective inhibitors of human immunodeficiency virus and cytomegalovirus replication in vitro. Antiviral Res18(2):191-207 [1992]).
  • Such an action may relate to UDA's superantigen nature (Galelli A, Truffa-Bachi P, Urtica dioica agglutinin. A superantigenic lectin from stinging nettle rhizome, J Immunol;151(4):1821-31 [1993]).
  • the rhizome-derived UDA lectin in addition to the leaf-derived IDS-30 extract, act therapeutically on certain autoimmune diseases.
  • This superantigen has been shown to induce a rapid deletion of a large fraction of T-cell receptor V ⁇ 8.3-expressing mature T-cells (Delcourt M, Peumans W J, Wagner M C, Truffa-Bachi P, V ⁇ -specific deletion of mature thymocytes induced by the plant superantigen Urtica dioica agglutinin, Cell Immunol;168(2):158-64 [1996]).
  • mice In mice, this activity has been demonstrated to prevent the development of systemic lupus erythematosus, as UDA-treated animals did not develop overt clinical signs of lupus and nephritis (Musette P, Galelli A, Chabre H, Callard P, Peumans W, Truffa-Bachi P, Kourilsky P, Gachelin G, Urtica dioica agglutinin, a V ⁇ 8.3-specific superantigen, prevents the development of the systemic lupus erythematosus-like pathology of MRL lpr/lpr mice, Eur J Immunol;26(8): 1707-11 [1996]).
  • Urtica dioica -derived substances as well as the constituents of many other plants, that act therapeutically, either as single molecules, or their oligomers, or in combination, on defined immune cells (such as MyDCs).
  • Pathologic conditions with which these substances interfere include infectious, neoplastic, and autoimmune diseases.
  • the liposomal system described herein may be utilized to specifically encapsulate such molecular plant components in purified or recombinant form, and address cells that have been, or will be, identified as their specific targets, so as to dramatically increase their effect and harness their potential while considerably reducing the risk of toxic side effects.
  • liposomes shuttled into intracellular compartments may deliver lectins suitable to agglutinate intracellularly stored pathogens (including HIV-1, HCV, the Ebola virus, Mycobacterium tuberculosis , and others), so as to generate large lectin-pathogen complexes that may, thus be recognized by the infected cell and, subsequently, be degraded enzymatically and/or pH-dependently.
  • pathogens including HIV-1, HCV, the Ebola virus, Mycobacterium tuberculosis , and others
  • MHL Myrianthus holstii lectin
  • MHL comprises several favorable characteristics, namely agglutination of HIV-1; no toxicity for greater than two orders of magnitude above the effective dosage in 50% of infected cells (EC 50 ); and the lack of mitogenicity for human leukocytes (Charan R D, Munro M H, O'Keefe B R, Sowder R C H, McKee T C, Currens M J, Pannell L K, Boyd M R, Isolation and characterization of Myrianthus holstii lectin, a potent HIV-1 inhibitory protein from the plant Myrianthus holstii , J Nat Prod 2000 Aug.;63(8):1170-4).
  • Compounds such as UDA, MHL and many others lectins or agglutinins, respectively, may be encapsulated within liposomes, so as to selectively unfold their properties within a given targeted cell and, more specifically, inside a specified intracellular compartment(s) of such a cell, or cell types.
  • Some embodiments of the inventive method of preferentially targeting a mammalian immune cell with a liposome relate to improved means of vaccination.
  • active targeting of dendritic cells in accordance with the invention, is used for vaccinating against cancer, or against a virus such as HIV.
  • a virus such as HIV.
  • Nair, S et al. Soluble proteins delivered to dendritic cells via pH - sensitive liposomes induce primary cytotoxic T lymphocyte responses in vitro , J. Exp. Med. 175(2):609-12 [1992]; Philip, R et al., Transgene expression in dendritic cells to induce antigen - specific cytotoxic T cells in healthy donors , Cancer Gene Ther.
  • Targeting of dendritic cells in accordance with the invention is also useful for improving vaccination strategies in general via accessing intracellular endosomal MHC class I and/or MHC class II antigen processing compartments.
  • MHC class I and/or MHC class II antigen processing compartments E.g. Zhou F and Huang L, Liposome - mediated cytoplasmic delivery of proteins: an effective means of accessing the MHC class I - restricted antigen presentation pathway , Immunomethods 1994;4(3):229-35 [1994]; Owais M et al., Use of liposomes as an immunopotentiating delivery system: in perspective of vaccine development , Scand. J Immunol.
  • the inventive method of preferentially targeting a mammalian immune cell with a liposome can also be used to target dendritic cells for facilitating the production of monoclonal antibodies.
  • a mammalian immune cell with a liposome can also be used to target dendritic cells for facilitating the production of monoclonal antibodies.
  • More than one drug can be incorporated by the lipid-drug complex, or liposome, in accordance with the inventive method, such that the lipid-drug complexes, e.g., liposomes, can incorporate a first drug and a second drug, or more drugs, in combination, as suits the particular needs of the practitioner.
  • useful liposomes can comprise a combination of an anti-HIV drug and an antifungal and/or antibacterial drug.
  • the present invention does not depend on any particular chemical or biochemical mechanism by which the useful formulations of lipid-drug complex, or liposome, are obtained or by which the drug is released to target cells.
  • lipid-drug complexes such as liposomes
  • Sullivan S M Gieseler R K H, Lenzner S, Ruppert J, Gabrysiak T G, Peters J H, Cox G, Richer, L, Martin, W J, and Scolaro, M J, Inhibition of human immunodeficiency virus -1 proliferation by liposome - encapsulated sense DNA to the 5 ′ TAT splice acceptor site , Antisense Res Develop 2:187-197 [1992]; Laverman P, Boerman O C, Oyen W J G, Corstens F H M, Storm G, In vivo applications of PEG liposomes: unexpected observations , Crit Rev Ther Drug Carrier Syst 18(6):551-66 [2001]; Oussoren C, Storm G, Liposomes to target the lymphatics by subcutaneous administration , Adv Drug Deliv Rev 50(1-2):143-56 [2001];
  • lipid film composed of phospholipids only, or in combination with cholesterol and/or other additives, is formed by evaporating the organic solvent (such as chloroform) from the lipid solution.
  • Hydrophobic drugs are added to the lipid solution prior to solvent evaporation.
  • the dry lipid film is hydrated with and isotonic aqueuous solution containing the drug by agitation (ultrasound, vortex, motorized stirrer, etc.). The lipid suspension is frozen and thawed 3-4 times.
  • the suspension is then passed through a series of polycarbonate filters containing pores of a defined diameter, such as 0.8 ⁇ m, 0.4 ⁇ m, 0.2 ⁇ m, or 0.1 p 82 m.
  • a defined diameter such as 0.8 ⁇ m, 0.4 ⁇ m, 0.2 ⁇ m, or 0.1 p 82 m.
  • unencapsulated drugs are removed by gel permeation column chromatography, dialysis or diafiltration.
  • the liposomes can be sterile-filtered (e.g., through a 0.22- ⁇ m filter).
  • a cryoprotectant such as lactose, glucose, sucrose, trehalose or maltose can be added to the sterile liposomes as long as isotonicity is maintained.
  • the liposomes can then be frozen and lyophilized and stored indefinitely as a lyophilized cake (e.g., Mayer L D, Hope M J, Cullis P R, Vesicles of variable sizes produced by a rapid extrusion procedure , Biochim Biophys Acta 858: 161-168 [1986]; Tsvetkova NM et al., Effect of sugars on headgroup mobility in freeze - dried dipalmitoylphosphatidylcholine bilayers: solid - state 31 P NMR and FTIR studies , Biophys J 75: 2947-2955 [1998]; Crowe J H, Oliver A E, Hoekstra F A, Crowe L M, Stabilization of dry membranes by mixtures of hydroxyethyl starch and glucose: the
  • the lipid suspension is prepared as described above. Freeze and thaw steps on a large scale may be a problem.
  • the diameter of the liposomes is reduced by shooting the lipid suspension as a stream either at an oncoming stream of the same lipid suspension (microfluidization) or against a steel plate (gualinization). This later technology has been used by the dairy industry for homogenization of milk. Untrapped water-soluble drugs are removed by diafiltration.
  • Hydrophobic drugs are completely entrapped and there usually is no free drug to be removed (e.g., Paavola A, Kilpelainen I, Yliruusi J, Rosenberg P, Controlled release injectable liposomal gel of ibuprofen for epidural analgesia , Int J Pharm 199: 85-93 [2000]; Zheng S, Zheng Y, Beissinger R L, Fresco R, Liposome - encapsulated hemoglobin processing methods , Biomater Artif Cells Immobilization Biotechnol 20: 355-364 [1992]).
  • Another method of drug entrapment is remote loading.
  • the drug to be entrapped must carry a charge.
  • the degree of protonation or deprotonation is controlled by the pK of the ionizable group.
  • a conjugate acid or base is trapped inside the liposomes.
  • the ionizable drug is added to the outside of the liposomes.
  • the pH is dropped such that the drug serves as a neutralizing salt of the ionizable substance trapped inside the liposomes. Due to the change in pH, the counter-ion to the entrapped ionizable molecule can diffuse out of the liposomes. This creates a gradient with sufficient energy to cause the drug to diffuse into the liposomes.
  • An example is the loading of doxorubicin into preformed liposomes.
  • a lipid film is solubilized in diethylether to a final concentration of typically about 30 mM.
  • water with entrapped drug is added to three parts ether lipid solution.
  • Energy in the form of sonication is applied forcing the suspension into a homogeneous emulsion.
  • the ether is removed by evaporation, typically yielding liposomes with about a 200 nm diameter and a high trapping efficiency.
  • Ethanol/calcium liposomes for DNA entrapment are prepared by any of the above methods (extrusion, homogenization, sonication).
  • the liposomes are mixed with plasmid DNA, or linear DNA fragments, plus 8 mM calcium chloride.
  • ethanol is added to the suspension to yield a concentration of about 40%.
  • the ethanol is removed by dialysis and the resultant liposomes are generally less than 200 im in diameter with about 75% of the DNA entrapped in the liposomes.
  • liposomes can be prepared by any of the above methods.
  • the liposomes can contain a lipid to which proteins can be crosslinked.
  • these lipids are: N-glutaryl-phosphatidylethnaolamine, N-succinyl-phospatidyethanolamine, maleimido-phenyl-butyryl-phosphatidylethanolamine, succinimidyl-acetylthioacetate-phosphatidylethanolamine, SPDP-phosphatidlyethnaolamine.
  • the glutaryl and succinimidyl phosphosphatidylethanolamine can be linked to a nucleophile, such as an amine, using cyclocarbodiimide.
  • the maleimido, acetylthioacetate and SPDP phosphatidylethanolamines can be reacted with thiols on the proteins, peptides or small molecular weight ligands of ⁇ 1000 g/mol.
  • the protein can be derivatized to the liposomes after formation. Underivatized protein can be removed by gel permeation chromatography. Peptides and low molecular weight ligands can be derivatized to the lipids and added to the organic lipid solution prior to formation of the lipid film.
  • examples of useful lipids include any vesicle-forming lipid, such as, but not limited to, phospholipids, such as phosphatidylcholine (hereinafter referred to as “PC”), both naturally occurring and synthetically prepared phosphatidic acid (hereinafter referred to as “PA”), lysophosphatidylcholine, phosphatidylserine (hereinafter referred to as “PS”), phosphatidylethanolamine (hereinafter referred to as “PE”), sphingolipids, phosphatidyglycerol (hereinafter referred to as “PG”), spingomyelin, cardiolipin, glycolipids, gangliosides or cerebrosides and the like used either singularly or intermixed such as in soybean phospholipids (e.g., Asolectin, Associated Concentrates).
  • the PC, PG, PA and PE can be derived from purified egg yolk and its hydrogenated derivatives.
  • lipids such as steroids, different cholesterol isomers, aliphatic amines such as long-chained aliphatic amines and carboxylic acids, long-chained sulfates and phosphates, diacetyl phosphate, butylated hydroxytoluene, tocopherols, retinols and isoprenoid compounds can be intermixed with the phospholipid components to confer certain desired and known properties on the formed vesicles.
  • steroids different cholesterol isomers
  • aliphatic amines such as long-chained aliphatic amines and carboxylic acids
  • long-chained sulfates and phosphates diacetyl phosphate
  • butylated hydroxytoluene butylated hydroxytoluene
  • tocopherols retinols and isoprenoid compounds
  • synthetic phospholipids containing either altered aliphatic portions such as hydroxyl groups, branched carbon chains, cycloderivatives, aromatic derivatives, ethers, amides, polyunsaturated derivatives, halogenated derivatives or altered hydrophilic portions containing carbohydrate, glycol, phosphate, phosphonate, quarternary amine, sulfate, sulfonate, carboxy, amine, sulfhydryl or imidazole groups and combinations of such groups can be either substituted or intermixed with the above-mentioned phospholipids and used in accordance with the invention.
  • Saturated synthetic PC and PG such as dipalmitoyl can also be used.
  • Other amphipathic lipids that can be used, advantageously with PC are gangliosides, globosides, fatty acids, stearylamine, long-chained alcohols and the like.
  • PEGylated lipids, monoglycerides, diglycerides, triglycerides can also be included.
  • Acylated and diacylated phospholipids are also useful.
  • useful phospholipids include egg phosphatidylcholine (“EPC”), dilauryloylphosphatidylcholine (“DLPC”), dimyristoylphosphatidylcholine (“DOPC”), dipalmitoylphosphatidylcholine (“DPPC”), distearoylphosphatidylcholine (“DSPC”), 1-myristoyl-2-palmitoylphosphatidylcholine (“MPPC”), 1-palmitoyl-2-myristoyl phosphatidylcholine (“PMPC”), 1-palmitoyl-2-stearoyl phosphatidylcholine (“PSPC”), 1-stearoyl-2-palmitoyl phosphatidylcholine (“SPPC”), dioleoylphosphatidylycholine (“DOPC”), dilauryloylphosphatidylglycerol (“DLPG”), dimyristoylphosphatidylglycerol (“DLPG”), dimyristoy
  • phosphatidylcholine and cholesterol are employed.
  • any suitable molar ratio of non-steroidal lipid to steroidal lipid e.g., cholesterol
  • the drug and lipids can be by any useful known technique, for example, by sonication, vortexing, extrusion, microfluidization, homogenization, use of a detergent (later removed, e.g., by dialysis).
  • the drug and lipid are mixed at a lipid-to-drug molar ratio of about 3:1 to about 100:1 or higher which is especially useful for drugs that are relatively more toxic, and more preferably of about 3:1 to about 10:1, and most preferably of about 5:1 to about 7:1.
  • an organic solvent can facilitate the production of the lipid-drug complex, such as a liposome.
  • the organic solvent is removed by any suitable known means of removal, such as evaporating by vacuum, or by the application of heat, for example by using a hair dryer or oven, or hot ethanol injection (e.g., Deamer, U.S. Pat. No. 4,515,736), as long as the lipid and drug components are stable at the temperature used.
  • Dialysis and/or chromatography, including affinity chromatography can also be employed to remove the organic solvent.
  • Hydrating the drug is performed with water or any biocompatible aqueous buffer, e.g., phosphate-buffered saline, HEPES, or TRIS, that maintains a physiologically balanced osmolarity.
  • aqueous buffer e.g., phosphate-buffered saline, HEPES, or TRIS.
  • Liposome rehydration can be accomplished simultaneously by removing the organic solvent or, alternatively, can be delayed until a more convenient time for using the liposomes (e.g., Papahadjopoulos et al., U.S. Pat. No. 4,235,871).
  • the shelf life of re-hydratable (“dry”) liposomes is typically about 8 months to about a year. This time span can be increased by lyophilization.
  • the lipid-drug complex is a unilamellar liposome.
  • Unilamellar liposomes provide the highest exposure of drug to the exterior of the liposome, where it may interact with the surfaces of target cells.
  • multilamellar liposomes can also be used in accordance with the present invention.
  • PEGylated liposomes is also encompassed within the present invention.
  • the lipid-drug complex further comprises an outer surface comprising at least one targeting ligand that specifically binds a marker on the surface of the immune cell.
  • targeting ligands include antibodies that specifically bind the marker of interest, such as anti-CD209/DC-SIGN-specific antibodies, or anti-CD4, anti-CD45R0, or anti-HLA class II.
  • Antibodies include whole antibodies as well as antibody fragments, with a specific target-binding capability of interest, ie., antigen-specific or hapten-specific targeting ligands.
  • Antibody fragments include, for example Fab, Fab′, F(ab′) 2 , or F(v) fragments.
  • Antibodies can also be polyclonal or monoclonal antibodies.
  • Antibodies also include antigen-specific or hapten-specific targeting ligands complexed with lipid-soluble linker moieties.
  • antibodies are coupled to the lipid-drug complex, such as a liposome-drug complex, via protein A of the Staphylococcus - aureus type, or via protein G which is typical of some other bacterial species.
  • the lipid-drug complex further comprises one or more biomembrane components that can further enhance the specific (i.e., active) targeting ability, cytotoxicity, or other therapeutic parameter of the liposome.
  • biomembrane components include a membrane-associated protein, an integral or transmembrane protein (e.g., a glycophorin or a membrane channel), a lipoprotein, a glycoprotein, a peptide toxin (e.g., bee toxin), a bacterial lysin, a Staphylococcus aureus protein A, an antibody, a specific surface receptor, or a surface receptor binding ligand.
  • Such vector molecules can include so-called protein transduction domains (PTDs) which are derived from various viruses or from Drosophila antennapedia .
  • PTDs protein transduction domains
  • the HIV Tat protein, or a derivative or fragment that acts as a PTD is also useful (e.g., Schwarze, S. R., et al., In vivo protein transduction: delivery of a biologically active protein into the mouse , Science 285:1569-72 [1999]).
  • the lipid-drug complex such as a liposome, is preferably, but not necessarily, about 30 to about 150 nanometers in diameter, and more preferably about 50 to about 80 nanometers in diameter.
  • the lipid-drug complexes can be preserved for later use by any known preservative method, such as lyophilization (e.g., Crowe et al., U.S. Pat. No. 4,857,319).
  • lyophilization e.g., Crowe et al., U.S. Pat. No. 4,857,319
  • lyophilization or other useful cryopreservation techniques involve the inclusion of a cryopreservative agent, such as a disaccharide (e.g., trehalose, maltose, lactose, glucose or sucrose).
  • a cryopreservative agent such as a disaccharide (e.g., trehalose, maltose, lactose, glucose or sucrose).
  • the lipid-drug complex e.g., a liposome
  • a subject is administered to a subject by any suitable means such as, for example by injection.
  • Administration and/or injection can be intrarterial, intravenous, intrathecal, intraocular, intradermal, subcutaneous, intramuscular, intraperitoneal, or by direct (e.g., stereotactic) injection into a particular lymphoid tissue, or into a tumor or other lesion.
  • Introduction of the lipid-drug complex into lymphatic vessels preferably, is via subcutaneous or intramuscular injection.
  • lymphoid tissue is a lymph node, such as an inguinal, mesenteric, ileocecal, or axillary lymph node, or the spleen, thymus, or mucosal-associated lymphoid tissue (e.g., in the lung, lamina intestinal of the intestinal wall, Peyer's patches of the small intestine, or lingual, palatine and pharyngeal tonsils, or Waldeyer's neck ring).
  • lymph node such as an inguinal, mesenteric, ileocecal, or axillary lymph node, or the spleen, thymus, or mucosal-associated lymphoid tissue (e.g., in the lung, lamina intestinal wall, Peyer's patches of the small intestine, or lingual, palatine and pharyngeal tonsils, or Waldeyer's neck ring).
  • Injection can also be by any non-intravenous method that drains directly, or preferentially, into the lymphatic system as opposed to the blood stream. Most preferred is subcutaneous injection, typically employing a syringe needle gauge larger than the lipid-drug complex. Intraperitoneal injection is also useful. Typically, injection of the injectate volume (generally about 1-5 cm 3 ) is into the subject's arm, leg, or belly, but any convenient site can be chosen for subcutaneous injection. Because drug subcutaneously administered, in accordance with some embodiments of the present invention, enters the lymphatic system prior to entering systemic blood circulation, benefits include
  • the frequency of injection is most preferably once per week, but more or less (e.g., monthly) frequent injections can be given as appropriate.
  • the present invention facilitates a treatment regimen that can involve a convenient weekly injection rather than multiple drug doses daily, as practiced typically in current AIDS treatment regimes. This feature may lead to improved patient compliance with the full course of treatment for some individual patients.
  • a 30 ⁇ mol lipid film composed of DOPC/Chol/DOPE-MBP (36.5:33.0:2.5 mol:mol:mol) was formed (cholesterol was purchased from Calbiochem, San Diego, Calif., USA; and DOPE and DOPE-MPB were from Avanti Polar Lipids, Alabaster, Ala., USA).
  • Lipid films were hydrated with 1 ml 50 mM calcein (Molecular Probes, Eugene, Oreg., USA) in PBS (pH 7.0), sonicated in a bath sonicator (5 min) and extruded ⁇ 5 through a 0.1 ⁇ m nucleopore filter (Avanti Polar Lipids) using a hand-held extruder.
  • the mean liposome size was determined by quasielectric light scattering with a Nicomp 380 ZLS Zeta-Potential Particle Sizer (Particle Sizing Systems, Santa Barbara, Calif., USA), yielding an average diameter of 146.7 ⁇ 31.0 nm.
  • Protein A is a bacterial cell wall component consisting of a single polypeptide chain of molecular weight 42 kDa. Protein A has the ability to specifically bind to the Fc region of immunoglobulin molecules, especially IgG.
  • One protein A molecule can bind at least 2 molecules of IgG simultaneously (S ⁇ umlaut over (j) ⁇ oquist J, Meloun B, Hjelm H, Protein A isolated from Staphylococcus aureus after digestion with lysostaphin , Eur J Biochem 29: 572-578 [1972]).
  • Protein A bearing liposomes were formed and their functionality in binding antibody-molecules was tested. Targeting of DC-SIGN and other membrane markers was achieved with Protein A liposomes pre-incubated with established antibody concentrations of either of several DC-SIGN-specific mAbs (all IgG1 ⁇ isotype), or irrelevant IgG1 ⁇ control mAb (MOPC-21/P3), or anti-bodies specific for the other membrane markers.
  • SATA succinimidylacetyl-thioacetate
  • Calcein-entrapping protein A liposomes were stored at 4° C. in the dark and used for up to 3 months. Immunoliposomes were prepared by incubation for 30 min at RT of protein A liposomes with test monoclonal antibodies (mAb; see below) or irrelevant negative control IgG (mAb MOPC-21/P3; eBioscience, San Diego, Calif., USA); Reeves, J P et al., Anti - Leu 3 a induces combining site - related anti - idiotypic antibody without inducing anti - HIV activity , AIDS Res Hum Retroviruses 7:55-63 [1991]) at a 5:1 molar ratio of mAb:protein A. The molar ratio of lipid to protein A was approximately 1000. Unbound antibody could be removed with magnetic Protein A beads (New England Biolabs, Beverly, Mass., USA). However, no significant effect on cell labeling was observed.
  • Monoclonal antibody binding to protein A liposomes was tested by Ficoll flotation. Specifically, antibodies were incubated with liposomes (30 min, RT) at the mAb:lipid ratio used for cell labeling. Polyclonal rabbit anti-mouse Ab ⁇ alkaline phosphatase (AP) was added to the incubation. The mixture was made from 20% ficoll 400 using a 30% Ficoll stock in PBS with a final volume of 0.4 ml, transferred to a microfuge tube, and 0.4 ml of 10% ficoll/PBS was layered on top and subsequently added a 0.4-ml layer of PBS. Tubes were centrifuged at 15,000 rpm for 15 min at RT.
  • AP polyclonal rabbit anti-mouse Ab ⁇ alkaline phosphatase
  • the PBS/10% ficoll interface was assayed for AP activity. Incubation with secondary Ab ⁇ AP yielded a 10-fold lower activity than incubation with primary mAb and secondary antibody, indicating that primary mAb had bound to protein A on the liposomes (results not shown).
  • protein A liposomes were preincubated with either of three different CD209-specific mAbs derived from clones 120507 (IgG2b), 120526 (IgG2a) (R&D Systems, Minneapolis, Minn., USA) and DCN46 (IgG1 ⁇ ) (BD Biosciences, San Jose, Calif., USA).
  • CD209-specific mAbs derived from clones 120507 (IgG2b), 120526 (IgG2a) (R&D Systems, Minneapolis, Minn., USA) and DCN46 (IgG1 ⁇ ) (BD Biosciences, San Jose, Calif., USA).
  • CD1a BL6; Coulter Immunotech, Miami, Fla., USA
  • CD4 SIM.4
  • NIH/McKesson cf. Acknowlegments
  • CD14 UCHM-1
  • CD45R0 UCHL1
  • CD83 HB15a17.11
  • Mature cells were harvested on day 7 of culture by pelleting non-adherent veiled cells from the supernatants and detaching weakly adherent cells with 1% EDTA in PBS for 30 min at 4° C.; strongly adherent cells were obtained by gently applying a cell scraper (TPP). All fractions were pooled, washed with PBS and kept in medium 80/20 plus 1% FBS on ice until used. For testing, cells were plated in fresh culture medium with 1% FBS at a density of 2 ⁇ 10 5 cells/well. To obtain the time-dependency of the targeting to dendritic cells, the 2 ⁇ 10 5 MoDCs per well or onset in the same medium were incubated with liposomes at 50 ⁇ M lipid at 37° C.
  • Flow cytometry can be employed: (1) to determine the phenotypes of My-DCs and T-cells at different times throughout DC differentiation and DC/T-cell co-culture (i.e., mixed leukocyte cultures or antigen-specific stimulation) with or without the DCs being infected with select M- and/or T-tropic strains of HIV-1, and/or treated with DC-SIGN-specific or control liposomes; and (2) to determine co-delivery of calcein/drug(s) to infected My-DCs or, more specifically, infected MoDCs.
  • DC/T-cell co-culture i.e., mixed leukocyte cultures or antigen-specific stimulation
  • Labeled MoDCs were analyzed on a Coulter Epics XL-MCL (Beckman Coulter, Fullerton, Calif.) flow cytometer according to the manufacturer's instructions, immediately after indirect staining with (i) primary mAbs and secondary polyclonal IgG conjugated with fluorescein-5-isothiocyanate (FITC) (eBioscience) (Gieseler, R et al., In - vitro differentiation of mature dendritic cells from human blood monocytes , Dev. Immunol 6:25-39 [1998]), (ii) incubation with the respective calcein-containing immunoliposomes, or (iii) negative controls.
  • FITC fluorescein-5-isothiocyanate
  • Targeting efficacy was determined directly after incubating DCs (or, when employed, macrophages) with the respective liposome/Protein A/mAb construct, or with liposomal negative controls employing the irrelevant isotype control antibody MOPC-21/P3. Results of negative controls employing protein A liposomes not loaded with mAbs were identical to those obtained with irrelevant control IgG. An influence via nonspecific uptake of liposomes by MyDCs could thus be excluded.
  • MFI mAb MFI mAb ⁇ MFI Co-IgG (II) and expressed as the percentage of MyDCs expressing this marker (MyDC mAb + [%])
  • ⁇ MFI ILS MFI ILS-mAb ⁇ MFI ILS-co-IgG
  • Marker expression and immunoliposomal binding and uptake do not necessarily correlate. For instance, while clearly expressing a given antigen when identified with a specific mAb, interaction of the same antigen with the much larger immunoliposomes labeled with the same mAb specificity may lead to shedding of the surface marker, which will result in a loss of signal fluorescence.
  • TE ILS MyDC ILS ⁇ 100 MyDC mAb ⁇ [ % ] ( IV ) wherein a result close to 100% indicates similar binding of an mAb and its corresponding immunoliposome; a lower result indicates loss of signal upon liposomal engagement; and a result clearly above 100% shows accumulation of liposomally delivered fluorophore, hence suggesting active uptake of the respective type of immunoliposome. Equation (IV) is easily transformed for the relative fluorescence of immunoliposomes vs.
  • RF ILS fluorescently labeled mAbs
  • PBL Peripheral Blood Leukocytes
  • MNLs Mononuclear leukocytes
  • PBS phosphate-buffered saline
  • Monocytes were isolated via negative magnetic-activated cell separation (MACS; Miltenyi, Bergisch-Gladbach, Germany and Auburn, Calif,, USA) by removing CD3 + , CD7 + , CD19 + , CD45RA + , CD56 + and mIgE + cells with mAb-coated magnetic microbeads. Negative monocyte separation had been chosen to avoid cell activation and was performed according to the manufacturer's instructions. Briefly, the procedure involved 2 washes with PBS supplemented with 0.5% bovine serum albumin (BSA; cell-culture grade, ⁇ 0.1 ng/mg endotoxin; ICN, Irvine, Calif., USA) and 2 mM EDTA (Sigma, St.
  • BSA bovine serum albumin
  • Mature antigen-presenting DCs were then obtained by adding tumor-necrosis factor (TNF)- ⁇ , leading to a DC type able to initiate both T-helper (Th)1- and Th2-dependent immunity (Caux C, Dezutter-Dambuyant C, Schmitt D & Banchereau J, GM - CSF and TNF - ⁇ cooperate in the generation of dendritic Langerhans cells , Nature; 360:258-61 [1992]; Sallusto F & Lanzavecchia A, Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony - stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha , J Exp Med; 179:1109-18 [1994]; Banchereau J & Steinman R M, Dendritic cells and the control of immunity , Nature; 392:245-52 [1998]).
  • TNF tumor-necrosis
  • DCs were generated in presence of interferon (IFN)- ⁇ (Gieseler R, Heise D, Soruri A, Schwartz P & Peters J H, In - vitro differentiation of mature dendritic cells from human blood monocytes , Develop Immunol; 6:25-39 [1998]).
  • IFN interferon
  • Such DCs appear to primarily induce Th1 cells, thus activating cytotoxic T-cells eliciting anti-tumor immunity (Soruri, A.
  • the differentiation medium was collected, centrifuged, and the pelleted DC fraction of non-adherent veiled cells was harvested.
  • adherent DCs were detached from the wells by incubating them with PBS/EDTA for 30 min at 4° C., and by successively employing a rubber policeman. Detached adherent DCs were pooled with the non-adherent fraction, adjusted to the cell numbers and incubated with the liposome concentrations indicated for each experiment.
  • myeloid dendritic cells obtained by protocols employing TNF- ⁇ or IFN- ⁇ , were analyzed flow-cytrometically for expression of CD1a, CD4, CD14, CD40, CD45RA, CD45R0, CD68, CD69, CD83, CD184, CD195, CD206, CD207, CD208, and/or CD209 (i.e., DC-SIGN) with mouse anti-human IgG1 ⁇ mAbs (MOPC-21/P3 as control).
  • CD1a CD4, CD14, CD40, CD45RA, CD45R0, CD68, CD69, CD83, CD184, CD195, CD206, CD207, CD208, and/or CD209 (i.e., DC-SIGN) with mouse anti-human IgG1 ⁇ mAbs (MOPC-21/P3 as control).
  • antigens were either stained directly with FITC-, PE-, or PC5-labeled antibodies, or were stained indirectly with unlabeled first mAbs plus secondary polyclonal IgG ⁇ FITC (available from eBioscience).
  • MOPC-21/P3 was employed as the IgG1 ⁇ isotype control. Results served three purposes, i.e.
  • DC-SIGN anti-CD209
  • other antibody at working dilution
  • 20 ⁇ l anti-CD209 (DC-SIGN) and/or other antibody at working dilution were incubated with 30 ⁇ l liposomes on a rotator for 1 h at RT.
  • Aliquots of cell suspension of at least 5 ⁇ 10 4 DCs (or, when employed, macrophages) were incubated with liposomes under saturating conditions for 3 h at RT under continuous agitation, and then examined by flow cytometry. (Tested conditions were 1 h, 3 h and 24 h. The most reliable and reproducible results were obtained by 3-h co-incubation.).
  • HIV strains were obtained from the NIH Repository (Rockville Pike, Bethesda, Md.), i.e., M-(R5-)tropic Ada-M and Ba-L; and T-(X4-)tropic HXB3, Lai, Lai/IIIB and HTLV-IIIB. HIV strains were tested for their “tissue-culture 50% infective dosage” (TCID50) according to protocols known to the art. According to the TCID50 results, viral supernatants were diluted, aliquoted and frozen at ⁇ 80° C. until employed for infection at different dose-infection kinetics.
  • TCID50 tissue-culture 50% infective dosage
  • CD4 + or CD8 + T cells were stored individually or as pools from two to four donors (for allogeneic stimulation) at ⁇ 80° C. or ⁇ 196° C., according to methods known to the art. Such cells are thawed when needed for autologous or allogeneic mixed leukocyte cultures, or for recall antigen tests.
  • liposomes were surface-labeled with Protein A so as to exchangeably bind antibodies specific for different antigens. These liposomes were entrapping calcein as a fluorescent tracer dye.
  • a range of single or combined drugs interfering with HIV propagation e.g., Viread® [tenofovir], Retrovir® [AZT], Epivir® [3-TC], Zerit® [d4T], Videx® [didanosine], Emtriva® [emtricitabine], Sustiva® [efavirenz], Viramun® [nevirapine], Rescriptor® [delavirdine], Norvir® [ritonavir], Agenerase® [amprenavir], Hivid® [ddC], lopinavir, Kaletra® [lopinavir+ritonavir], Viracept® [nelfinavir], Crixivan® [indinovir sulfate], Fortovase® [
  • Supernatants can be tested according to the manufacturer's instructions for presence of p24 by a commercially available ELISA (Abbott Laboratories).
  • Quantitative Polymerase Chain Reaction for HIV.
  • the degree of integration of HIV proviral DNA into dendritic-cell host DNA can be determined by using nested primer pairs (nested semi-qPCR) for HIV proviral sequences, such as the following:
  • the PCR reaction mixture typically includes the following: Buffer (5 ⁇ l of 10 ⁇ PCR Rxn Buffer, Invitrogen); MgCl 2 (3 ⁇ l of 50 mM MgCl 2 , Invitrogen); dNTP (1 ⁇ l of mixture of dATP, dCTP,dGTP,dTTP: 10 ⁇ M, each); Outer Primer (SEQ ID NO:1; 1 ⁇ l of 10 pmol/ ⁇ l); Outer Primer (SEQ ID NO:2; 1 ⁇ l of 10 pmol/ ⁇ l); Taq (0.2 ⁇ l of 5 Units/ ⁇ l, Platinum Taq DNA Polymerase, Invitrogen); double distilled water (37 ⁇ l); DNA sample (2 ⁇ l).
  • One standard thermal cycling profile was the following: 5 min at 95° C.; (20 s at 95° C.; 30 s at 55° C.; 30 s at 72° C.) ⁇ 25; 2 min at 72° C.; hold at 4° C.
  • PCR is generally repeated using two microliters of amplified DNA transferred from the first reaction in fresh PCR reaction mixture, except using the inner primers (SEQ ID NO:3 and SEQ ID NO:4) instead of the outer primers, and employing a different thermal cycling profile: 5 min at 95° C.; (20 s at 95° C.; 30 s at 55° C.; 30 s at 72° C.) ⁇ 35; 2 min at 72° C. (melting curve 95° C. down to 55° C. in steps of 0.5° C.).
  • DNA quantification can be achieved by comparison with a serial dilution of a DNA sample of known quantity of HIV proviral DNA.
  • a Multiplex-PCR can be performed. Briefly, a second nested PCR can be performed in the same reaction, with a LUX primer labeled with 6-carboxy-4′,5′-dichloro-2′,7′-dimethoxyfluorescein succinimidyl ester, for a human chromosome sequence (genome equivalent). This permits quantification of the total DNA content per sample. Numbers of proviral copies per human genome equivalent can be calculated from such data.
  • Peripheral blood mononuclear cells were evaluated according to their size (forward scatter) and granularity (side scatter) and thus were gated as na ⁇ ve T and B cells; activated T-cells and B-cells; and monocytes, including a small proportion of blood dendritic cells (data not shown).
  • Cultured monocyte-derived dendritic cells were tested for expression of markers delineating their developmental stage (maturity), as well as for DC subtype markers. The DCs expressed markers typical for skin and mucosal DC phenotypes that are considered to play a key role in HIV infection.
  • mucosal DCs are the first immune cell type to be directly infected by HIV (and integrate its genetic information as proviral DNA) and/or harvest HIV on their surface by DC-SIGN and/or take up HIV by any of various mechanisms to retain it in intracytoplasmic compartments (e.g., endosomes, fused phago-endosomes, or phagolysosomes). Such cells then migrate to regional and local lymph nodes where passing on HIV to other cell types, most prominently T-helper cells (i.e., “CD4 cells”) as well as other reservoir cells, including the next generation of lymph node-settling DCs. In considering all this, the DCs generated in our in-vitro system thus provide an ideal model for testing the presumptive targeting efficacy for such cells in vivo.
  • CD4 cells most prominently T-helper cells
  • MoDCs matured by 7-day culture with GM-CSF, IL-4 and subsequent TNF- ⁇ were tested by flow cytometry for expression of markers generally expressed by DCs or subpopulations thereof.
  • DC-SIGN markers delineating mature DCs in vitro and in vivo (CD40, CD45R0, CD83), as well as dendritic Langerhans cells of the epidermis (CD1a) and the intestinal (CD4) and nasal mucosa (CD14).
  • Phenotyping thus served (i) for verifying MoDCs generated in vitro as mature; (ii) for proving strong expression of DC-SIGN (CD209) as the pre-conceived target for immunoliposomal compound delivery to MyDCs; (iii) for pinpointing further potential target antigens conforming to the requirement of consistent high expression; and (iv) for determining whether the generated MoDCs expressed CD1a and/or CD14 as potential targeting structures expressed by epidermal and mucosal Langerhans cells in vivo.
  • Relative mean fluorescence intensities ( ⁇ MFI) of test conditions vs. negative controls (n 3) characterized the phenotypic profile of mature MoDCs as CD1a +++ , CD4 ⁇ , CD14 ⁇ to +++ , CD40 ++ to +++ , CD45R0 + to +++ , CD83 + and CD209 +++ [with: ( ⁇ ), test antibody congruent with negative control; ( ⁇ ), ⁇ MFI peak ⁇ 5 above negative control; (+), ⁇ MFI peak ⁇ 10 above negative control; and (+++), ⁇ MFI peak ⁇ 250 negative control].
  • expression of CD14 varied most considerably among the donors.
  • DC-SIGN (CD209) and CD1a a Langerhans-cell marker
  • FIG. 1 shows targeting of calcein-labeled liposomes to MoDCs mediated by DC-SIGN or other targeting ligands.
  • Mature MoDCs were generated in vitro for 7 days. Liposomes were incubated with either one or two monoclonal antibodies (mAbs) specific for key markers expressed by MoDCs, so as to obtain monocpecific liposomes (for CD1a, CD83, or CD209) or bispecific liposomes (for CD1a+CD83, CD1a+CD209, or CD 83+CD209) (Zhou L J, Tedder T F, CD 14 + blood monocytes can differentiate into functionally mature CD 83 + dendritic cells , Proc Natl Acad Sci USA;93(6):2588-92 [1996]; Gieseler R, Heise D, Soruri A, Schwartz P, Peters J H, In - vitro differentiation of mature dendritic cells from human blood monocytes , Develop Immunol.;6(
  • Monoclonal antibodies (mAbs) and mAb-labeled immunoliposomes tested were specific for CD4, CD45R0 and CD209 (DC-SIGN).
  • mAbs Monoclonal antibodies
  • DC-SIGN Monoclonal antibodies
  • Binding of specific mAbs visualized with FITC-labeled secondary antibody (left-hand column) revealed the degree of antigen (Ag) expression.
  • Mature MyDCs generated from the same donors were incubated for 1, 3 or 24 h with immunoliposomes at 37° C. [a preliminary experiment had proven 37° C. superior to 4° C.
  • FIG. 2 shows monospecific liposomal targeting with respect to kinetics and efficacy.
  • we here investigated the time kinetics of liposomal uptake i.e. uptake of calcein at a number of time points over a24-hour period.
  • the MoDCs expressed CD14 over a broad range of membrane densities (cf. left hand graph)
  • this phenotypic pattern was not reflected after targeting.
  • CD209 (DC-SIGN) targeting again revealed the highest rate of uptake; also, the patterns of antigen expression (left-hand graph) and targeting efficacy (3-h graph) were very similar.
  • Colmenares M Puig-Kroger A, Pello O M, Corbi A L, Rivas L, Dendritic cell ( DC )- specific intercellular adhesion molecule 3 ( ICAM -3-) grabbing nonintegrin ( DC - SIGN, CD 209), a C - type surface lectin in human DCs, is a receptor for Leishmania amastigotes , J Biol Chem 277(39):36766-69 [2002]).
  • FIG. 4A left panel and FIG. 4B (left panel) show calculated values of targeting and surface binding of monospecific immunoliposomes applied to MoDCs; the results depicted are representative of at least three independent experiments.
  • FIG. 4A Provided in FIG. 4A are percentages of MoDCs expressing select markers (FITC fluorescence), and MoDCs targeted with corresponding immunoliposomes (calcein fluorescence).
  • FITC fluorescence select markers
  • calcein fluorescence calcein fluorescence
  • FITC and calcein concentrations were equimolar in all mAb or liposome conditions, the immunoliposomal net targeting efficacy (TE ILS ) and relative fluorescence of immunoliposomes vs. mAbs (RF ILS ) could be determined (equations IV and V; FIG. 4B ).
  • mono-specific immunoliposomes targeting DC-SIGN revealed the highest TE ILS and were the only preparation showing a positive RF ILS value (indicating lipo
  • the liposomal targeting efficacy of CD209-coupled liposomes was 83.31% ( FIG. 4A , left panel), and the respective LS Binding/Uptake graph in FIG. 1 demonstrates for all cells a right shift (shaded curve), relative to the control peak (open curve). This indicates that 100% of the cells had been efficiently targeted, even when only faintly expressing DC-SIGN.
  • combinations of anti-DC-SIGN liposomes with anti-CD1a or anti-CD83 liposomes did not effect increased uptake.
  • antibody concentrations were only half of those employed when targeting with one antibody only. Therefore, further investigations were warranted to determine whether bispecific targeting might, indeed, enhance the targeting efficacy, when compared to monospecific tareting.
  • FIG. 3 illustrates liposomal targeting of DCs via two cell markers (termed “bispecific targeting”), including time dependency of the targeting efficacy over a 24-h period.
  • Bispecific targeting was carried out with all 2-member combinations, or permutations, of CD4, CD45R0 and CD209. As in FIG. 2 , best results were, here again, obtained upon 3-h incubation of cells with targeted liposomes.
  • FIG. 3A shows results for the combination of anti-CD4 plus anti-CD45R0 targeting ligands. Irrespective of the incubation time, when compared to the experiment shown in FIG. 2 , a subtractive effect on liposomal uptake was obtained. Combination of anti-CD4- and anti-CD45R0 -specific targeting, therefore, did not appear to support enhanced uptake by a double-positive cell subset, e.g. the resting T-memory cell population residing in lymphoid organs. A similar result was observed for liposomes bearing the combination of anti-CD209 plus anti-CD45R0 targeting ligands ( FIG. 3C ).
  • FIG. 3B shows results for the combination of anti-CD4 plus anti-CD209 targeting ligands.
  • the abscissa in FIG. 3B shows liposomal uptake as a logarithmic increase in fluorescence. Therefore, the improvement of uptake by combined targeting of CD4 and CD209 was at least by a factor of 2 and thus, in accordance with the invention, liposomal targeting dendritic cells employing a combination of anti-CD4 and anti-CD209 targeting ligands can be a useful option, for example, in treating HIV disease.
  • Adipocytes another HIV reservoir, can also be targeted by targeting via CD4 and CD45 (e.g., Hazan, U. et al., Human adipose cells express CD 4 , CXCR 4 , and CCR 5 receptors: a new target cell type for the immunodeficiency virus -1 ? FASEB J. 16, 1254-1256 [2002; Erratum in: FASEB J.
  • CD4 and CD45 e.g., Hazan, U. et al., Human adipose cells express CD 4 , CXCR 4 , and CCR 5 receptors: a new target cell type for the immunodeficiency virus -1 ? FASEB J. 16, 1254-1256 [2002; Erratum in: FASEB J.
  • a net targeting efficiency with a positive (+) value indicates that the percentage of cells targeted efficaciously was higher than the percentage recognized by antibody only; negative ( ⁇ ) values indicate less efficient targeting than with antibody; a value of ⁇ 100% indicates that no cells at all have been targeted. All values refer to 3-hour co-incubation of cells and targeted liposomes. The three best targeting conditions were CD209>CD83+CD209>CD1a+CD209.
  • Targeting efficacy for bispecific immunoliposomes targeting CD4+CD45R0 was 58.54%; targeting efficacy for bispecific mmunoliposomes targeting CD4+CD209 was 68.74%; targeting efficacy for bispecific immunoliposomes targeting CD45R0+CD209 was 62.21%.
  • DC-SIGN-targeted system can target different HIV reservoir populations, i.e., myeloid dendritic cells and macrophage subsets, for delivering HIV-inhibiting compounds of any or all types currently known.
  • these reservoir populations can be targeted for integrating DC-SIGN-attached viruses for successive generation of immunity as well as to remove virus from the cells' surfaces, and mother-to-child virus transfer during pregnancy can be prevented.
  • DC-SIGN is strongly expressed by mucosal and skin types of dendritic cells in humans and macaques.
  • DC- SIGN a novel HIV receptor on DCs that mediates HIV -1 transmission , Curr Top Microbiol Immunol. 2003;276:31-54 [2003]; Yu Kimata M T et al., Capture and transfer of simian immunodeficiency virus by macaque dendritic cells is enhanced by DC - SIGN , J ViroL 76(23):11827-36 [2002]).
  • DC-SIGN-targeted liposomes offers the benefit of actively targeting the first cell population infected and affected in the etiology of HIV disease.
  • DC-SIGN is further expressed by dendritic and other cells located within certain placental anatomic structures.
  • Soilleux E J et al. Placental expression of DC - SIGN may mediate intrauterine vertical transmission of HIV , J Pathol. 195(5):586-92 [2001]; Soilleux E J, Coleman N, Transplacental transmission of HIV: a potential role for HIV binding lectins , Int J Biochem Cell Biol. 2003 Mar.;35(3):283-7 [2003]; Kämmerer U et al., Unique appearance of proliferating antigen-presenting cells expressing DC - SIGN ( CD 209) in the decidua of early human pregnancy , Am J Pathol. 162(3):887-96 [2003]).
  • the inventive method offers the benefit of targeting those cells that apparently play a major role in mother-to-child HIV transfer, also termed vertical transmission.
  • Pelleted single cells were successively dissolved in 100 ⁇ l of ProLong antifade mounting medium to which was added 5 ⁇ M of the positively charged AT-binding DNA dye, 4′,6-diamidino-2-phenylindole (DAPI) (both from Molecular Probes, Eugene, Oreg., USA). Fifty ⁇ l of each preparation were transferred to poly-L-lysine-coated slides (Labscientific, Livingston, N.Y., USA), cover-slipped, sealed and kept in the dark for at least 15 min before being viewed.
  • DAPI 4′,6-diamidino-2-phenylindole
  • MoDCs were then screened with a Zeiss Axioskop microscope (Carl Zeiss, Thornwood, N.Y., USA) for surface and intracellular fluorescence of calcein (green) and DNA/nuclei (blue).
  • Digital photography was carried out with an ORCA-1 CCD camera (Hamamatsu, Bridgewater, N.J., USA).
  • Photographic processing, merging of green and blue fluorescence, as well as microtomography linking to generate film clips covering MyDCs in optical depth was performed with the Northern Elite V.6.0 software package (Empix Imaging, Cheek Towaga, N.Y., USA). Dead cells were excluded from the evaluation by nuclear staining with propidium iodide as well as by their extremely bright nuclear DAPI staining.
  • Immunoliposomes carrying mAb MOPC-21/P3 were taken as negative controls; positive controls employed anti-CD209 mAb ⁇ FITC.
  • FIG. 5 illustrates surface binding vs. internalization of targeted liposomes determined by fluorescence microscopy as described above. For discerning intracellular from outshining membrane fluorescence, we then, at steps of 0.5 ⁇ m, photographed 27 to 35 microtomographies per MoDC body. After 3-h incubation with CD209-specific liposomes (corresponding to the CD209 condition in FIG. 4B ), green calcein labeling was seen only on the cell surface and was mainly confined to larger DC-SIGN-rich lipid rafts ( FIG. 5 , panel 1 ; depicting the median optical section).
  • DC-SIGN CD209
  • mannan- or mannose-binding lectin which very likely—as a liver-derived substance (Downing, I et al., Immature dendritic cells possess a sugar - sensitive receptor for human mannan - binding lectin , Immunology 109:360-4 [2003])—constitutes a component of the small amount of fetal bovine serum employed during culture and incubation.
  • MBL is even autologously secreted by immature human MoDCs (Downing I et al., Immature dendritic cells possess a sugar - sensitive receptor for human mannan - binding lectin , Immunology 2003;109:360-4 [2003]). Furthermore, MBL, via its own C-type lectin domain, can prevent HIV-1 from binding to DC-SIGN (Spear G T et al., Inhibition of DC - SIGN - mediated trans infection of T cells by mannose - binding lectin , Immunology 2003;110:80-5 [2003]). Therefore, soluble MBL (and perhaps other unidentified molecules displaying similar characteristics) did not prevent the inventive DC-SIGN-specific liposomes from interacting with the membrane-bound C-type lectin.
  • DC-SIGN-targeted immunoliposomes i.e., including targeting ligand that specifically binds CD209 deliver their contents both to immature and mature MyDCs, and that, in addition to cytoplasmatic distribution, their contents strongly accumulate in discrete intracellular compartments ( FIG. 5 ), or endosomes, respectively.
  • Th cells interacting with DCs within lymphoid organs and tissues in the course of antigen-specific stimulation can also be reached therapeutically by this strategy (Gieseler R K, Marquitan G, Hahn M J, Perdon L A, Driessen W H P, Sullivan S M, Scolaro M J, DC - SIGN - specific liposomal targeting and selective intracellular compound delivery to human myeloid dendritic cells: implications for HIV disease , Scand J Immunol;59:415-24 [2004]; Marquitan G, Gieseler R K, Driessen W H P, Perdon L A, Hahn M J, Wader T, Sullivan S M, Scolaro M J, Intracellular compound delivery to human monocyte - derived dendritic cells by immunoliposomal targeting of the C - type lectin DC - SIGN . MACS & MORE, in press [2004]).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Dispersion Chemistry (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Botany (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Biotechnology (AREA)
  • Medical Informatics (AREA)
  • Endocrinology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Hydrogenated Pyridines (AREA)
US10/943,758 2003-09-17 2004-09-17 Targeted lipid-drug formulations for delivery of drugs to myeloid and lymphoid immune cells Abandoned US20050142114A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US10/943,758 US20050142114A1 (en) 2003-09-17 2004-09-17 Targeted lipid-drug formulations for delivery of drugs to myeloid and lymphoid immune cells
US14/193,498 US20140234400A1 (en) 2003-09-17 2014-02-28 Targeted Lipid-Drug Formulations for Delivery of Drugs to Myeloid and Lymphoid Immune Cells
US15/154,097 US10357457B2 (en) 2003-09-17 2016-05-13 Targeted lipid-drug formulations for delivery of drugs to myeloid and lymphoid immune cells

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US50376903P 2003-09-17 2003-09-17
US56737604P 2004-04-30 2004-04-30
US10/943,758 US20050142114A1 (en) 2003-09-17 2004-09-17 Targeted lipid-drug formulations for delivery of drugs to myeloid and lymphoid immune cells

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/193,498 Continuation-In-Part US20140234400A1 (en) 2003-09-17 2014-02-28 Targeted Lipid-Drug Formulations for Delivery of Drugs to Myeloid and Lymphoid Immune Cells
US14/193,498 Continuation US20140234400A1 (en) 2003-09-17 2014-02-28 Targeted Lipid-Drug Formulations for Delivery of Drugs to Myeloid and Lymphoid Immune Cells

Publications (1)

Publication Number Publication Date
US20050142114A1 true US20050142114A1 (en) 2005-06-30

Family

ID=34381085

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/943,758 Abandoned US20050142114A1 (en) 2003-09-17 2004-09-17 Targeted lipid-drug formulations for delivery of drugs to myeloid and lymphoid immune cells
US14/193,498 Abandoned US20140234400A1 (en) 2003-09-17 2014-02-28 Targeted Lipid-Drug Formulations for Delivery of Drugs to Myeloid and Lymphoid Immune Cells
US15/154,097 Active US10357457B2 (en) 2003-09-17 2016-05-13 Targeted lipid-drug formulations for delivery of drugs to myeloid and lymphoid immune cells

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/193,498 Abandoned US20140234400A1 (en) 2003-09-17 2014-02-28 Targeted Lipid-Drug Formulations for Delivery of Drugs to Myeloid and Lymphoid Immune Cells
US15/154,097 Active US10357457B2 (en) 2003-09-17 2016-05-13 Targeted lipid-drug formulations for delivery of drugs to myeloid and lymphoid immune cells

Country Status (8)

Country Link
US (3) US20050142114A1 (de)
EP (1) EP1663315B1 (de)
JP (1) JP2007505925A (de)
AT (1) ATE494011T1 (de)
CA (1) CA2539256C (de)
DE (1) DE602004030923D1 (de)
DK (1) DK1663315T3 (de)
WO (1) WO2005027979A2 (de)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040170309A1 (en) * 2003-02-27 2004-09-02 Applied Imaging Corp. Linking of images to enable simultaneous viewing of multiple objects
US20060134189A1 (en) * 2004-11-17 2006-06-22 Protiva Biotherapeutics, Inc siRNA silencing of apolipoprotein B
US20080124370A1 (en) * 2006-08-03 2008-05-29 Marx Jeffrey C Reagents, methods and systems to suppress pro-inflammatory cytokines
US20090317455A1 (en) * 2006-08-31 2009-12-24 Naoto Oku Reverse targeting lipid vesicle
US20100063131A1 (en) * 2007-03-26 2010-03-11 Hirofumi Takeuchi Prompt nucleic acid delivery carrier composition
WO2010141511A3 (en) * 2009-06-01 2011-04-21 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent rna interference, compositions and methods of use thereof
WO2011109809A2 (en) * 2010-03-05 2011-09-09 New Agriculture, Inc A novel composition of matter for delivering lipid-soluble materials, and a method for producing it
US20110256213A1 (en) * 2008-10-15 2011-10-20 The Board Of Trustees Of The University Of Illinois Phospholipid micellar and liposomal compositions and uses thereof
WO2012094004A1 (en) * 2011-01-05 2012-07-12 The United States Of America, As Represented By The Secretary Of Agriculture Fusion of peptidoglycan hydrolase enzymes to a protein transduction domain allows eradication of both extracellular and intracellular gram positive pathogens
EP2638896A1 (de) 2012-03-14 2013-09-18 Bioneer A/S Kationisches liposomales Arzneimittelabgabesystem für spezifisches Targeting humaner CD14+ Monozyten in Vollblut
WO2014153114A1 (en) * 2013-03-14 2014-09-25 Fred Hutchinson Cancer Research Center Compositions and methods to modify cells for therapeutic objectives
WO2014145839A3 (en) * 2013-03-15 2015-05-07 Htd Biosystems Inc. Liposomal vaccine adjuvants and methods of processing or using same
US9603799B2 (en) 2013-03-15 2017-03-28 Htd Biosystems Inc. Liposomal vaccine adjuvants and methods of making and using same
US10427124B2 (en) * 2013-09-19 2019-10-01 Nanyang Technological University Methods for controlling assembly of lipids on a solid support
US10731157B2 (en) 2015-08-24 2020-08-04 Halo-Bio Rnai Therapeutics, Inc. Polynucleotide nanoparticles for the modulation of gene expression and uses thereof
WO2022040435A1 (en) * 2020-08-19 2022-02-24 The Board Of Regents Of The University Of Texas System Nanodrugs for targeted drug delivery and use thereof
US11566061B2 (en) 2017-01-05 2023-01-31 Fred Hutchinson Cancer Center Systems and methods to improve vaccine efficacy
US11872195B2 (en) 2016-04-14 2024-01-16 Fred Hutchinson Cancer Center Compositions and methods to program therapeutic cells using targeted nucleic acid nanocarriers

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2388531T3 (es) 2004-03-19 2012-10-16 Rodos Biotarget Gmbh Liposomas derivatizados con hidratos de carbono para la selección como diana de dominios de reconocimiento de hidratos de carbono celulares de lectinas CTL/CTLD y suministro intracelular de compuestos terapéuticamente activos
WO2007047539A2 (en) * 2005-10-14 2007-04-26 Medtronic, Inc. Localized delivery to the lymphatic system
US20140087395A1 (en) * 2011-03-31 2014-03-27 Hiroshima University Method for distinguishing between species within the genus staphilococcus
EP3873532A1 (de) 2018-10-31 2021-09-08 Novartis AG Dc-sign-antikörper-arzneimittelkonjugate
WO2024026452A1 (en) 2022-07-29 2024-02-01 Repertoire Immune Medicines, Inc. T cell epitopes associated with type 1 diabetes

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5766902A (en) * 1993-08-20 1998-06-16 Therexsys Limited Transfection process
US20030044407A1 (en) * 1999-02-22 2003-03-06 Chang Esther H. Simplified and improved method for preparing an antibody or an antibody fragment targeted immunoliposome or polyplex for systemic administration of a therapeutic or diagnostic agent
US20040208921A1 (en) * 2003-01-14 2004-10-21 Ho Rodney J. Y. Lipid-drug formulations and methods for targeted delivery of lipid-drug complexes to lymphoid tissues
US7357930B1 (en) * 1999-05-03 2008-04-15 Infectio Recherche Inc. Methods and formulations for targeting infectious agents bearing host cell proteins

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4795739A (en) * 1987-05-29 1989-01-03 Gene Labs, Inc. Method of inhibiting HIV
JPH06504429A (ja) * 1990-10-09 1994-05-26 ネクスター・フアーマシユーテイカルズ・インコーポレイテツド ウイルス複製の阻害
AU2003210659A1 (en) * 2002-01-24 2003-09-02 Sangstat Medical Corporation Combination therapy for treatment of hiv infection
EP1495141A4 (de) * 2002-03-20 2006-03-22 Massachusetts Inst Technology Hiv-therapeutikum

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5766902A (en) * 1993-08-20 1998-06-16 Therexsys Limited Transfection process
US20030044407A1 (en) * 1999-02-22 2003-03-06 Chang Esther H. Simplified and improved method for preparing an antibody or an antibody fragment targeted immunoliposome or polyplex for systemic administration of a therapeutic or diagnostic agent
US7357930B1 (en) * 1999-05-03 2008-04-15 Infectio Recherche Inc. Methods and formulations for targeting infectious agents bearing host cell proteins
US20040208921A1 (en) * 2003-01-14 2004-10-21 Ho Rodney J. Y. Lipid-drug formulations and methods for targeted delivery of lipid-drug complexes to lymphoid tissues

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7233340B2 (en) * 2003-02-27 2007-06-19 Applied Imaging Corp. Linking of images to enable simultaneous viewing of multiple objects
US20040170309A1 (en) * 2003-02-27 2004-09-02 Applied Imaging Corp. Linking of images to enable simultaneous viewing of multiple objects
US20060134189A1 (en) * 2004-11-17 2006-06-22 Protiva Biotherapeutics, Inc siRNA silencing of apolipoprotein B
US20080124370A1 (en) * 2006-08-03 2008-05-29 Marx Jeffrey C Reagents, methods and systems to suppress pro-inflammatory cytokines
US8399427B2 (en) 2006-08-03 2013-03-19 Warsaw Orthopedic, Inc. Reagents, methods and systems to suppress pro-inflammatory cytokines
US8138160B2 (en) 2006-08-03 2012-03-20 Warsaw Orthopedic, Inc. Reagents, methods and systems to suppress pro-inflammatory cytokines
US20090317455A1 (en) * 2006-08-31 2009-12-24 Naoto Oku Reverse targeting lipid vesicle
US9315828B2 (en) * 2007-03-26 2016-04-19 Hirofumi Takeuchi Prompt nucleic acid delivery carrier composition
US20100063131A1 (en) * 2007-03-26 2010-03-11 Hirofumi Takeuchi Prompt nucleic acid delivery carrier composition
AU2008230379B2 (en) * 2007-03-26 2012-12-06 Otsuka Pharmaceutical Co., Ltd. Prompt nucleic acid delivery carrier composition
US20110256213A1 (en) * 2008-10-15 2011-10-20 The Board Of Trustees Of The University Of Illinois Phospholipid micellar and liposomal compositions and uses thereof
WO2010141511A3 (en) * 2009-06-01 2011-04-21 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent rna interference, compositions and methods of use thereof
US9957505B2 (en) 2009-06-01 2018-05-01 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent RNA interference, compositions and methods of use thereof
US9200276B2 (en) 2009-06-01 2015-12-01 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent RNA interference, compositions and methods of use thereof
WO2011109809A2 (en) * 2010-03-05 2011-09-09 New Agriculture, Inc A novel composition of matter for delivering lipid-soluble materials, and a method for producing it
US9629888B2 (en) * 2010-03-05 2017-04-25 Leafpro, Llc Composition of matter for delivering lipid-soluble materials, and a method for producing it
WO2011109809A3 (en) * 2010-03-05 2012-03-01 New Agriculture, Inc A novel composition of matter for delivering lipid-soluble materials, and a method for producing it
WO2012094004A1 (en) * 2011-01-05 2012-07-12 The United States Of America, As Represented By The Secretary Of Agriculture Fusion of peptidoglycan hydrolase enzymes to a protein transduction domain allows eradication of both extracellular and intracellular gram positive pathogens
EP2638896A1 (de) 2012-03-14 2013-09-18 Bioneer A/S Kationisches liposomales Arzneimittelabgabesystem für spezifisches Targeting humaner CD14+ Monozyten in Vollblut
WO2014153114A1 (en) * 2013-03-14 2014-09-25 Fred Hutchinson Cancer Research Center Compositions and methods to modify cells for therapeutic objectives
US10392446B2 (en) 2013-03-14 2019-08-27 Fred Hutchinson Cancer Research Center Compositions and methods to modify cells for therapeutic objectives
US9603799B2 (en) 2013-03-15 2017-03-28 Htd Biosystems Inc. Liposomal vaccine adjuvants and methods of making and using same
WO2014145839A3 (en) * 2013-03-15 2015-05-07 Htd Biosystems Inc. Liposomal vaccine adjuvants and methods of processing or using same
US10427124B2 (en) * 2013-09-19 2019-10-01 Nanyang Technological University Methods for controlling assembly of lipids on a solid support
US10731157B2 (en) 2015-08-24 2020-08-04 Halo-Bio Rnai Therapeutics, Inc. Polynucleotide nanoparticles for the modulation of gene expression and uses thereof
US11872195B2 (en) 2016-04-14 2024-01-16 Fred Hutchinson Cancer Center Compositions and methods to program therapeutic cells using targeted nucleic acid nanocarriers
US11566061B2 (en) 2017-01-05 2023-01-31 Fred Hutchinson Cancer Center Systems and methods to improve vaccine efficacy
WO2022040435A1 (en) * 2020-08-19 2022-02-24 The Board Of Regents Of The University Of Texas System Nanodrugs for targeted drug delivery and use thereof

Also Published As

Publication number Publication date
EP1663315B1 (de) 2011-01-05
EP1663315A2 (de) 2006-06-07
CA2539256C (en) 2012-04-24
WO2005027979A3 (en) 2005-09-09
ATE494011T1 (de) 2011-01-15
JP2007505925A (ja) 2007-03-15
US20160324779A1 (en) 2016-11-10
CA2539256A1 (en) 2005-03-31
DE602004030923D1 (de) 2011-02-17
WO2005027979A2 (en) 2005-03-31
US10357457B2 (en) 2019-07-23
US20140234400A1 (en) 2014-08-21
DK1663315T3 (da) 2011-04-18

Similar Documents

Publication Publication Date Title
US10357457B2 (en) Targeted lipid-drug formulations for delivery of drugs to myeloid and lymphoid immune cells
Nisini et al. The multirole of liposomes in therapy and prevention of infectious diseases
Kiaie et al. Axial pharmaceutical properties of liposome in cancer therapy: Recent advances and perspectives
US10335420B2 (en) Nanoparticle-based tumor-targeted drug delivery
US20190175504A1 (en) Carbohydrate-Derivatized Liposomes for Targeting Cellular Carbohydrate Recognition Domains of Ctl/Ctld Lectins, and Intracellular Delivery of Therapeutically Active Compounds
JP2007529558A5 (de)
US20210047620A1 (en) Langerin+ Cell Targeting
AU2020367786A1 (en) Nano-enabled immunotherapy in cancer
TW201804987A (zh) 以艾弗林受體A2(EphA2)為標靶的歐洲紫杉醇產生之奈米微脂體組成物
Clemente et al. Straight to the point: targeted mRNA-delivery to immune cells for improved vaccine design
US20220193108A1 (en) Immunotherapeutic Nanoparticles And Methods Relating Thereto
Kwong Liposome-anchored local delivery of immunomodulatory agents for tumor therapy
ES2361578T3 (es) Formulaciones de fármaco-lípido para la administración dirigida de fármacos a células inmunes mieloides y linfoides.
Zhao et al. Effect of adjuvant release rate on the immunogenicity of nanoparticle-based vaccines: a case study with a nanoparticle-based nicotine vaccine
Bouma et al. Nanobody-liposomes as novel cancer vaccine platform to efficiently stimulate T cell immunity
STOLK et al. LIPOSOMAL NANOVACCINE CONTAINING Α-GALACTOSYLCERAMIDE AND GANGLIOSIDE GM3 STIMULATES ROBUST CD8+ T CELL RESPONSES VIA CD169+ MACROPHAGES AND CDC1
Saliba Development of innovative liposome-based constructs for non-invasive cancer immunotherapy in humans
Ma Delivery of dideoxyinosine to human monocyte/macrophages

Legal Events

Date Code Title Description
AS Assignment

Owner name: LET THERE BE HOP MEDICAL RESEARCH INSTITUTE, CALIF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GIESELER, ROBERT K.;MARQUITAN, GUIDO;SCOLARO, MICHAEL J.;AND OTHERS;REEL/FRAME:016472/0247;SIGNING DATES FROM 20041207 TO 20050301

AS Assignment

Owner name: FOUNDERS,ARIZONA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LET THERE BE HOPE MEDICAL RESEARCH INSTITUTE;REEL/FRAME:024517/0291

Effective date: 20081212

AS Assignment

Owner name: RODOS BIOTARGET GMBH,GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:FOUNDERS;REEL/FRAME:024574/0292

Effective date: 20081219

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION