EP1202620A1 - Anti-inflammatory therapy for inflammatory mediated infection - Google Patents

Anti-inflammatory therapy for inflammatory mediated infection

Info

Publication number
EP1202620A1
EP1202620A1 EP00932379A EP00932379A EP1202620A1 EP 1202620 A1 EP1202620 A1 EP 1202620A1 EP 00932379 A EP00932379 A EP 00932379A EP 00932379 A EP00932379 A EP 00932379A EP 1202620 A1 EP1202620 A1 EP 1202620A1
Authority
EP
European Patent Office
Prior art keywords
inflammatory
viral
agent
hiv
group
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.)
Withdrawn
Application number
EP00932379A
Other languages
German (de)
English (en)
French (fr)
Inventor
Peter A. Anton
Michael A. Poles
Janis V. Giorgi
Julie E. Elliott
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.)
University of California
Original Assignee
University of California
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 University of California filed Critical University of California
Publication of EP1202620A1 publication Critical patent/EP1202620A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/60Salicylic acid; Derivatives thereof
    • A61K31/612Salicylic acid; Derivatives thereof having the hydroxy group in position 2 esterified, e.g. salicylsulfuric acid
    • A61K31/616Salicylic acid; Derivatives thereof having the hydroxy group in position 2 esterified, e.g. salicylsulfuric acid by carboxylic acids, e.g. acetylsalicylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/60Salicylic acid; Derivatives thereof
    • A61K31/606Salicylic acid; Derivatives thereof having amino groups
    • 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
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • 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
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2026IL-4
    • 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
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2066IL-10
    • 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
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2086IL-13 to IL-16
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/02Drugs for genital or sexual disorders; Contraceptives for disorders of the vagina
    • 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
    • A61P31/12Antivirals
    • 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
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0031Rectum, anus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0034Urogenital system, e.g. vagina, uterus, cervix, penis, scrotum, urethra, bladder; Personal lubricants

Definitions

  • TECHNICAL FIELD This invention relates the use of anti-inflammatory agents for the treatment of retro virus infections. Specifically the invention concerns the treatment of HIV infections.
  • the HIV infection cycle begins with the entry ofthe virus into the target cell.
  • the human CD4 is believed to be the primary receptor on T cells recognized by HIV.
  • the binding ofthe HIV envelope glycoprotein (env) to the CD4 receptor results in the fusion of virus and cell membranes, which in turn facilitates virus entry into the host.
  • the eventual expression of env ofthe surface of the HIV-infected host cell enables this cell to fuse with uninfected CD4-positive cells, thereby spreading the virus.
  • HIV can also enter other cells such as monocytes, B cells, and dendritic cells, which can serve as viral reservoirs, even though they may not express CD4. Cytokines are known to affect HIV replication.
  • Pro-inflammatory cytokines promote HIV replication (Fauci, Nature 384:529-534, 1996), while ⁇ -chemokines inhibit the replication of obligate CCR5 utilizing viruses (Moore, et al. , J. Virol. 70:551 -562, 1996), and enhance the replication of CXCR4 utilizing viral isolates (Dolei, et al, AIDS 12:183-190, 1998).
  • helper T cells and B cells in vivo mediated by such molecules as CD4, T cell receptor, and MHC class II, is essential in the development of thymus-dependent humoral immunity.
  • CD40 ligand are central to the development and maintenance of immunity.
  • CD40 is a transmembrane glycoprotein of 45 kDa, which is member ofa family of surface molecules with homology in their intracellular domain to nerve growth factor receptor, TNF receptors, Fas, CD17, and CD30 (Armitage et al, Nature 357:80-82, 1992).
  • CD40 has been identified on immature and mature B lymphocytes, which when cross-linked by antibodies induces B cell proliferation (Vail et al, Eur. J. Immunol. 19:1464-1467, 1989), on monocytes, dendritic cells, thymic epithelial cells.
  • CD40 On antigen presenting cells (APCs) CD40 functions as a costimulatory receptor that promotes antigen-specific T-cell activation (reviewed in Clark et al, Adv. Immunol. 63:43-68, 1996).
  • a ligand for CD40 called gp39, CD40 ligand or CD40L, has also been molecularly characterized (Armitage et al, 1992, supra), and found to be expressed on activated CD4+ Th cells (Spriggs et al, J. Exp. Med. 176:1543-1550, 1992). Cells that express the gp39 protein can trigger B cell proliferation and, through other stimulatory signals, can induce antibody proliferation (Armitage et al, 1992, supra). It has also been reported that engagement of CD40 by CD40L results in enhanced expression of CD80 and CD86, and the secretion of pro-inflammatory cytokines.
  • the normal intestinal tract is characterized by a low level of mild inflammation, which is fueled by constitutive levels of locally secreted chemokines and cytokines (Shanahan and Anton, Gut Peptides, J. Walsh eds. (Raven Press, Ltd, New York, 1994, page 851; Schreiber et al, Gasrroenterology 101 :1020 (1991); MacDermott et al, Inflammatory Bowel Diseases 4, 54 (1998); Luster, N.Engl. J.
  • gastrointestinal lymphocytes are known to differ functionally and phenotypically from their peripheral blood counterparts (Allison et al, Gasrroenterology 99:421 (1990); Jarry et al, Eur. J. Immunol. 20:1097 (1990); McGowan et al, Neuroimmunomodulation 4:70 (1997)).
  • Virtually all mucosal CD4+ lymphocytes express activation markers and are of the CD45RO+ memory subset (Schieferdecker et al, J. Virol. 149:2816 (1992)).
  • the present invention is based upon the discovery that retroviruses, such as human immunodeficiency viruses (HIV), provoke an inflammatory state, in contrast to the previous view that HIV is a static or progressive immunodeficient state.
  • retroviruses such as human immunodeficiency viruses (HIV)
  • HIV human immunodeficiency viruses
  • Such inflammatory states both provide opportunistic advantages to HIV by providing recruitment of additional inflammatory cells at the site of inflammation bearing receptors used by HIV for infection, as well as activating infected inflammatory cells causing production of viral particles.
  • the present invention provides a method for inhibiting an inflammatory mediated infection of mucosal tissue by contacting the tissue with an inhibiting effective amount of an anti-inflammatory agent alone, or in combination with an anti-viral agent.
  • the inflammatory mediated infection can be caused by a virus, such as a retrovirus (e.g., a lentivirus such as an immunodeficiency virus selected from the group consisting of human immunodeficiency virus (HIN) type 1, HIV-type 2, and simian immunodeficiency virus (SIV).
  • a retrovirus e.g., a lentivirus such as an immunodeficiency virus selected from the group consisting of human immunodeficiency virus (HIN) type 1, HIV-type 2, and simian immunodeficiency virus (SIV).
  • HIN human immunodeficiency virus
  • SIV simian immunodeficiency virus
  • the contacting ofthe tissue with the anti-inflammatory agent alone, or in combination with an anti-viral agent may be in vivo, in vitro, or ex vivo.
  • the mucosal tissue is typically mammalian and preferably human.
  • mucosal tissue examples include uro-genital tissue, (e.g., vaginal tissue), gastro-intestinal tissue, a tissue ofthe lower Gl tract, and nasal-larynx tissue to name a few.
  • the anti-inflammatory agent may be administered locally or systemically, such as by topical administration, intravenous, oral or parenteral administration, respectively.
  • the anti-inflammatory agent when used in combination with an anti-viral agent, may be administered prior to, simultaneously with or after administration of the anti-viral agent.
  • the anti-inflammatory agent may be any anti- inflammatory agent such as those that cause a decrease in the recruitment of inflammatory cells, a decrease in the production of chemokines, a decrease in the production of pro-inflammatory cytokines, or inhibits the interaction ofa chemokine or cytokine receptor with its ligand.
  • anti-inflammatory agents may be administered in singlet or combination and also may be administered along with other anti-viral compounds in smglet or combination.
  • the present invention provides a method of inhibiting activation of a retrovirus by contacting a cell infected with the virus with a virus- activation inhibiting amount of an anti-inflammatory agent alone, or in combination with an anti-viral agent.
  • Activation of inflammatory cells by autocrine and paracrine effect of pro-inflammatory mediators causes a change in the transcription regulation of inflammatory cells at the site of inflammation.
  • the activation of inflammatory cells through such process in turn results in activation of latent infections of retroviruses.
  • retroviruses include lentiviruses such as the immunodeficiency viruses HIV type 1, HIV-type 2, and simian immunodeficiency virus (SIV).
  • the contacting ofthe cell with the anti-inflammatory agent alone, or in combination with an anti-viral agent may be in vivo, in vitro, or ex vivo.
  • the anti-inflammatory agent when used in combination with an anti-viral agent, may be contacted prior to, simultaneously with or after contacting with the anti-viral agent.
  • the cell may be a mucosal cell and is typically mammalian and preferably human.
  • mucosal cells include cells derived from uro-genital tissue, (e.g., vaginal tissue), gastro-intestinal tissue, tissue ofthe lower Gl tract, oral-buccal tissue and nasal-larynx tissue to name a few.
  • the invention provides a method of inhibiting an inflammatory mediated mucosal infection in a subject by contacting the subject with an effective amount of an anti-inflammatory agent alone, or in combination with an anti-viral agent.
  • the inflammatory mediated mucosal infection may be caused by a virus or another pathogen.
  • the virus may be a retrovirus, for example, a lentivirus such as the immunodeficiency virus HIV type 1, HIV-type 2, and simian immunodeficiency virus (SIV).
  • the contacting is in vivo, such contacting may be by administering the anti-inflammatory agent locally or systemically, for example by topical administration, intravenous, oral or parenteral administration, respectively.
  • the anti-inflammatory agent when used in combination with an anti-viral agent, may be administered prior to, simultaneously with or after administration ofthe anti-viral agent.
  • the subject is typically mammalian and preferably human.
  • the invention provides a method of inhibiting transmission of an inflammatory mediated mucosal infection from a subject having or at risk of having an inflammatory mediated mucosal infection to another subject by contacting the subject having or at risk of having the inflammatory mediated mucosal infection with an effective amount of an anti-inflammatory agent alone, or in combination with an anti- viral agent, thereby inhibiting transmission of an inflammatory mediated mucosal infection to the other subject.
  • the inflammatory mediated mucosal infection may be caused by a virus or by another pathogen.
  • the virus may be a retrovirus, for example, a lentivirus such as the immunodeficiency virus HIV type 1, HIV-type 2, and simian immunodeficiency virus (SIV).
  • a retrovirus for example, a lentivirus such as the immunodeficiency virus HIV type 1, HIV-type 2, and simian immunodeficiency virus (SIV).
  • the contacting to the subject having or at risk of having an inflammatory mediated mucosal infection is in vivo, such contacting may be by administering the anti- inflammatory agent locally or systemically, for example by topical administration, intravenous, oral or parenteral administration, respectively.
  • the anti-inflammatory agent when used in combination with an anti-viral agent, may be administered prior to, simultaneously with or after administration of the anti-viral agent.
  • the subject having or at risk of having an inflammatory mediated mucosal infection is typically mammalian and preferably human.
  • the present invention provides a method of inhibiting progression of an inflammatory mediated infection in a subject by contacting the subject with an effective amount of an anti-inflammatory agent alone, or in combination with an anti-viral agent.
  • the inflammatory mediated mucosal infection may be caused by a virus or another pathogen, or related to infection by the virus or other pathogen.
  • the virus may be a retrovirus, for example, a lentivirus such as the immunodeficiency virus HIV type 1, HIV-type 2, and simian immunodeficiency virus (SIV).
  • the contacting is in vivo, such contacting may be by administering the anti-inflammatory agent locally or systemically, for example by topical administration, intravenous, oral or parenteral administration, respectively.
  • the anti-inflammatory agent when used in combination with an anti-viral agent, may be administered prior to, simultaneously with or after administration ofthe anti-viral agent.
  • the subject is typically mammalian and preferably human.
  • the present invention provides a method for preventing or decreasing the probability of infection of a subject with a human immunodeficiency virus by administering to a subject at risk of an HIV infection a prophylactic effective amount of an anti-inflammatory agent alone, or in combination with an anti-viral agent, which inhibits HIV replication, activation, or progression by reducing the number of inflammatory cells present in any given tissue, such as the uro-genital, gastro-intestinal or other mucosal tissue.
  • the present invention provides a method for preventing or decreasing the probability of human immunodeficiency virus transmission from a subject with an HIV infection to another subject by administering to the infected subject an effective amount of an anti-inflammatory agent alone, or in combination with an anti-viral agent, which inhibits HIV replication, activation, or progression by reducing the number of inflammatory cells present in any given tissue, such as the uro-genital, gastro-intestinal or other mucosal tissue, thereby preventing or decreasing the probability of HIV transmission from the infected subject to another subject.
  • composition comprising at least one dose of a therapeutically effective amount of an anti-inflammatory agent, in a pharmaceutically acceptable carrier designed to be delivered to a mucosal tissue, wherein the dose is in an amount effective to inhibit or decrease the probability of immunodeficiency virus progression, infection or transmission.
  • the present invention provides an article of manufacture, comprising at least one anti-inflammatory agent and instructions for use of the agent in inhibiting an immunodeficiency virus infection.
  • An article of manufacture can include at least one anti-inflammatory agent alone, or be in a combination with an anti-viral agent.
  • Articles of manufacture include, for example, a condom, sponge, diaphragm, cervical cap, vaginal ring, suppository, and an enema.
  • Instructions for use can be included with an article of manufacture, for example, instructions for use in prophylaxis of immunodeficiency virus infection, or in preventing or inhibiting transmission of immunodeficiency virus from one subject to another.
  • FIG. 1 shows CCR5 receptor expression on CD4+ lymphocytes from blood and from gut mucosa.
  • Flow cytometry scatter plots (A, B) demonstrate lymphocyte subset analysis to quantify percentages of cells expressing CCR5 and/or CD4 in a representative subject for blood (A) and gut (B). The number on the upper right quadrant of each plot indicates the percentage of CD4+ lymphocytes in that subject that expressed CCR5.
  • Figure 2 shows the number of CCR5 receptors per cell on CD4+ lymphocytes from blood and from gut mucosa.
  • Flow cytometry histograms (A,B) demonstrate quantitation of CCR5 expression on CD4+ lymphocytes of one ofthe six subjects for blood (A) and gut (B).
  • the rightward shift of mean fluorescence index in the CCR5+ CD4+ mucosal cells illustrates the increased numbers of CCR5 receptors per CD4+ lymphocyte.
  • the number above the bars in A and B indicates the number of molecules of CCR5 expressed per CCR5+ CD4+ lymphocyte in the blood and gut of that individual.
  • Figure 3 shows CXCR4 receptor expression on CD4+ lymphocytes from blood and from gut mucosa.
  • Flow cytometry scatter plots (A,B) demonstrate lymphocyte subset analysis to quantify percentages of cells expressing CXCR4 and/or CD4 in a representative subject for blood (A) and gut (B). The number on the upper right quadrant of each plot indicates the percentage of CD4+ lymphocytes in that subject that expressed CXCR4.
  • Figure 4 shows the number of picograms of p24 produced by MMC and PBMC after infection with HIVSX or HIVNL4-3.
  • Line graphs indicate the p24 production (picograms of p24 per 104 CD4+ lymphocytes) at 18, 72, and 130 hours after a 3-hour infection with either M-tropic HJNSX (A) or T-tropic HIV ⁇ L4-3 (B).
  • the supernatants from the MMCs cultured in the presence of 20 IU/mL of IL-2 contained greater concentrations of p24 than the supernatants from either PBMC grown with (o) or without (T) 20 IU/mL of IL-2.
  • the greater p24 production from the cultured mucosal cells suggests that they are more susceptible than PBMC to replication of M-, or T-tropic HIV-1.
  • Figure 5 shows the 3 day/IL-2 culture for isolation of mucosal mononuclear cells yields increased numbers of CD45+, CD3+ CD4+ and CD8+ cells as compared to conventional collagenase/dispase digestion.
  • the mononuclear cell populations isolated by each technique do not appear to differ significantly in their T cell subset make-up.
  • Figure 6 shows pre-amplification handling of tissue biopsy samples results in a 5-10% RNA loss.
  • Seronegative samples were 'spiked' with 250 copies ofthe standard LTR sequence pre-extraction (left) and in a parallel sample post-extraction (right). Digitized quantification of 32P emission demonstrated a 5-10% difference between pre and post extraction additions of the same amount of LTR RNA.
  • Standards demonstrate an assay sensitivity of 10 copies.
  • Figure 7 shows the internal consistency between samples obtained from different sites at the same circumferential level (30 cm) in the colon. Each sample was run in duplicate. On average, there was a 0.2 log SD between samples from each individual. All subjects had undetectable plasma viral loads.
  • Figure 8 shows a quantitative measurement of HIV in rectal biopsies.
  • DNA was extracted from duplicate biopsies of subjects with undetectable plasma HIV RNA.
  • qPCR of the HIV LTR sequence was conducted and actual detected copy numbers from each ofthe 2 samples from each subject recorded (lower panel). (The 4 different subjects are labeled "Samples #1-4").
  • Samples #1-4" The 4 different subjects are labeled "Samples #1-4".
  • ⁇ -globin quantitation and standard curves were performed in triplicate on each subject's 2 samples (only one biopsy's results are displayed in the top panel. Calculated numbers of HIV DNA copies are reported per 2x106 ⁇ -globin copies (1x106 cells).
  • Figure 9 shows that the isolation process does not alter relevant receptor expression.
  • PBMC peripheral blood mononuclear cells stained directly [upper panels] with antibodies to CD4, CD8, CCR5 or CXCR4 as identified on the horizontal and vertical axes.
  • Lower panels show results of parallel staining of the same individual's PBMC following exposure to the isolation process used for mucosal mononuclear cells.
  • Figure 10 shows that the CCR5 receptor is expressed on a significantly greater percentage of mucosal CD4+ T cells compared to blood.
  • FIG 11 shows that CCR5 receptor number per cell is significantly increased on mucosal CD4 T cells compared to blood CD4 T cells in the same subject samples shown in Figure 10. Histograms demonstrate the marked rightward shift of mean channel fluorescence correlating with increased number of receptors per cell in the mucosal preparation.
  • FIGS 13A and 13B show that CCR5+ CD4:CD8 ratios in (A) blood and (B) gut decline in IBD and HIV.
  • the left panel shows relative CCR5 expression in blood from healthy, seronegative controls, seronegative inflammatory controls and subjects with stable HIV infection.
  • the changing ratios of CCR5-expressing CD4+ T cells to CCR5 -expressing CD8+ T cells are boxed underneath. Similar presentations for mucosal lymphocytes are shown in (A).
  • Figure 14 shows that the amount of p24 (indicator of HIV production) is significantly higher in mucosal mononuclear cells (MMC) than in peripheral blood mononuclear cells (PBMC).
  • Figures 15A and 15B show that CD8+ cells are increased in colon from an
  • Figures 16A and 16B show that CCR5+ cells are increased in colon of an HIV infected patient. Biopsies from (A) HIV(-) and (B) HIV(+) colon. CCR5+ cells are indicated by brown staining (darkened in black and white photo).
  • Figures 17A through 17D show the amount of (A) RANTES; (B) IFN ⁇ ; and (C) TNF in uninfected, HIV(+) with low mucosal viral load and HIV(+) with high mucosal viral load.
  • (D) shows the increased amount of CD4 cell activation (as indicated by increased HLA-DR that occurs due to HIV-induced increases in pro- inflammatory cytokines.
  • the y-axis denotes the % of CD4 cells activated and the x- axis denotes viral load.
  • Figure 18 shows increased amounts of virus (Nlegfp) produced by mucosal cells (MMC) versus blood cells (PBMC), as indicated by increased expression of green fluorescent protein when Nlegfp is replicated in the cells.
  • MMC mucosal cells
  • PBMC blood cells
  • Figure 19 shows the effect of Asacol (mesalamine) on HIV replication.
  • Cultured cells infected with HIV were treated with 5-ASA or AZT in the amounts indicated.
  • Luciferase expression which indicates the amount of HIV replication, was quantified using a luminometer. The data shown represent nine separate studies.
  • the term “inhibit” or “inhibiting” means to reduce the activity, function or property referred to by a measurable amount, such as a reduction of at least 30% or more. Where there are multiple different activities that may be inhibited (for example, preventing cell recruitment, production of pro-inflammatory mediators, cell or viral activation, viral replication, or viral progression/proliferation), the reduction of any single activity (with or without the other activities) is sufficient to fall within the scope of this definition. In addition, where a single or where multiple agents are administered to inhibit activity, the reduction by a single agent of any single activity or the reduction by a combination of agents of any single activity is sufficient to fall within the scope of this definition.
  • an “inflammation inhibiting amount” means that amount of an inflammatory agent necessary to modulate, inhibit, or suppress inflammatory responses or symptoms.
  • activation when used in reference to a virus, means an increase in the number of virus particles or viral load systemically or locally, or an increase in synthesis of viral protein or nucleic acid in a cell. The increase is typically induced by a change in a transcriptional state of an infected cell, which results in increased virus production by the cell. The increase may also occur independently of such a change in transcriptional state. Changes in a transcriptional state of an infected cell can be induced by cytokines, chemokines and other molecules that modulate cell proliferation, differentiation or mobilization (chemotaxis).
  • Changes in a transcriptional state of an infected cell can also be induced by infection by the virus or another pathogen (bacterium, fungi, mycobacterium, etc.), or induced by exposure to an immune-modulating antigen (e.g. , LPS). Activation, which typically results in increased amounts of virus, in turn can lead to increased numbers of cells infected by the virus, which is referred to as "virus spreading.”
  • the term "inflammatory mediated,” when used in reference to an infection, disease, disorder or condition, means an infection, disease, disorder or condition which progresses or spreads, or accelerates or worsens in response to an inflammatory response ofthe subject.
  • the progression can occur, for example, through an increase in target (uninfected) cells migrating to the site of inflammation where infected cells and virus are present.
  • the recruited cells provide new targets for viral infection, which results in virus spread and, in turn, progression ofthe infection.
  • Inflammatory mediated also includes the situation where an infected cell (e.g., in a latent state), when exposed to an immune modulatory molecule or other signaling molecule (e.g. , a pro- inflammatory cytokine or other molecule that increases inflammatory response), modulates the cells' transcriptional state, thereby stimulating or increasing production of the virus by the cell.
  • mucosal tissue means any tissue in which mucosal cells are found, such tissues, include, for example, gastro-intestinal tissues (e.g., the stomach, small intestine, the large intestine, the rectum), uro-genital tissue (e.g., vaginal tissue, penile tissue, urethra), nasal-larynx tissue (e.g., nasal tissue, larynx tissue), mouth (buccal tissue) to name a few.
  • gastro-intestinal tissues e.g., the stomach, small intestine, the large intestine, the rectum
  • uro-genital tissue e.g., vaginal tissue, penile tissue, urethra
  • nasal-larynx tissue e.g., nasal tissue, larynx tissue
  • mouth buccal tissue
  • immunodeficiency viral infection such as HIV infections
  • mucosal tissue e.g., mucosal tissue
  • Most reports have emphasized a state of lymphopenia or "anti-inflammation" in the mucosa paralleling that seen progressively in the blood of infected subjects. Because mucosal tissue is populated by an increased number of activated, memory, co-receptor expressing
  • CD4+ T cells in healthy uninfected individuals, the vulnerability to infection through these tissues is high.
  • mucosal immune cells most likely CD8+ T-lymphocytes and macrophages
  • secrete increased levels of pro- inflammatory chemokines and cytokines with the intent of recruiting additional T- lymphocytes to the mucosal infection site. This heightened response or
  • the present invention relates to the treatment of a wide range of retroviral infections, retroviral related diseases and disorders using any number of anti-inflammatory agents alone, or in combination with any number of antiviral agents, to prevent or inhibit recruitment of additional susceptible cells to the inflamed tissue of a subject, thereby preventing activation of inflammatory cells through inflammatory mechanisms such as activation by cytokines and other pro-inflammatory mediators by inhibiting these pathways as well as preventing transmission to other subjects (uninfected or infected) exposed to the mucosal tissue.
  • the retroviral infections capable of treatment using the methods of the present invention include retroviral disorders caused by a large number of retroviruses.
  • SIV infection may primarily be a disease ofthe mucosal immune system (Veazey et al, Science 280:427 (1998); MacDonald and Spencer, Gastrointestinal and Hepatic Immunology, R.H. Heatley, Ed. (Cambridge University Press, 1994).
  • HIV infection also involves the mucosal immune system and infectious viral particles have been recovered directly from mucosal samples and in situ studies have demonstrated that lamina intestinal T lymphocytes are among the first cells that are infected (Koteler et al, Am. J. Pathol.
  • Inflammation results from a number of individual and related cascades or reactions caused by pro-inflammatory mediators including cytokines, prostaglandins, leukorrienes, chemokines, adhesion molecules (e.g., LFA-1) and others known to those of skill in the art.
  • pro-inflammatory mediators including cytokines, prostaglandins, leukorrienes, chemokines, adhesion molecules (e.g., LFA-1) and others known to those of skill in the art.
  • chemokine receptors play a pivotal role in permitting viral entry into a CD4+ cell.
  • the ligands for these receptors, called chemokines are also important.
  • These chemokines, along with pro-inflammatory cytokines are the main stimulators of cells but also play another role in amplification ofthe inflammatory cascade.
  • These soluble inflammatory mediators are derived mainly from CD8+ T cells. Once produced they can act in a paracrine and autocrine fashion to
  • Inflammation results in stimulation of lymphocytes and macrophages (CD4+ cells). Those inflammatory cells that harbor HIV when they are stimulated will be induced to produce large amounts ofthe virus.
  • the intestine even in healthy HIV- uninfected patients, maintains a state of low-level physiologic inflammation that is necessary to protect the interior milieu from the bath of potential pathogens that contact its surface. The great majority of lymphocytes and macrophages that compose this infiltrate express are therefore stimulated.
  • the primary target of HIV, in which the virus most effectively replicates, is the stimulated CD4+ cell. This type of cell fills the gastrointestinal mucosa. Increased viral replication results in greater spread of HIV throughout the mucosa and higher mucosal HIV viral loads.
  • the predominant aim of anti-HIV therapy is to decrease the ability of HIV to replicate and therefore spread amongst CD4+ cells which are eventually destroyed by the virus. When replication of HIV is effectively reduced so too is to ability ofthe virus to develop mutations in its genetic material that result in resistance to antiviral medications.
  • Immunosuppressive activity refers to inhibiting or decreasing the ability of B and T cells to react to be recruited or become activated to a site of inflammation. NSAIDs should diminish prostaglandins (PG) synthesis. PGs are cytostatic agents.
  • LFA-1 adhesion receptor
  • ICAM-1 CD54
  • Blockage of this interaction by monoclonal antibodies to LFA-1 and ICAM-1 results in increased survival time for mice receiving a heart allograft (Isobe et al, 1992, Science 255: 1 125-1 127).
  • the gastrointestinal mucosa is one element of this lymphoid tissue and increasing evidence suggests that HIV involves the mucosa at all stages of disease. Not only is the gastrointestinal tract the route of transmission for the majority of patients, but it is the largest lymphoid organ. As mentioned above, the gastrointestinal mucosa is characterized by a state of low-level physiologic inflammation, and the majority of its lymphocytes are activated. The naturally high concentration of pro- inflammatory cytokines that are present in the mucosa appear to enhance HIV replication in this site, resulting in a high mucosal HIV viral load and successive rounds of infection of new target gastrointestinal CD4+ cells.
  • the inventors have found that the majority of gastrointestinal CD4+ T cells express the chemokine receptors that are necessary for HIV entry.
  • the vast majority of lymphocytes ofthe gastrointestinal mucosa express both CCR5 and CXCR4.
  • CCR5 lymphocytes of the gastrointestinal mucosa express both CCR5 and CXCR4.
  • MMCs mucosal mononuclear cells express higher levels of this receptor than blood derived monocytes on a per cell basis.
  • the inventors have found that the mucosal cells are more susceptible to HIV than are peripheral blood cells in vitro.
  • HIV nucleic acids can be found in the mucosa ofthe majority of HIV-infected individuals; Kotler et al. detected HIV DNA by PCR using gag-specific primers in 70% of 20 patients he investigated. The inventors have found that even patients with undetectable plasma viral loads have replicating virus in their mucosa. A high SIV viral load is seen in the gastrointestinal mucosa whether the macaque is infected via the gut or via the parenteral route suggesting that the mucosa has a high intrinsic susceptibility to HIV. After infection, these macaques exhibit a profound early (within 7 to 21 days) loss of gastrointestinal mucosal CD4+ cells. This sign of vigorous HIV activity was not mirrored in other lymphoid sites.
  • mucosal CD4+ cell depletion has been described in the colon and duodenum during both the early asymptomatic phase of chronic infection and after the onset of clinical AIDS.
  • retroviral infections for example, infections associated with immunodeficiency viruses, and more specifically HIV are associated with a state of inflammation.
  • the inflammation can be cellular, soluble, or both.
  • the inflammation can be in any number of tissues which are susceptible to infection by a virus (e.g., mucosal tissue).
  • chemokines and chemokine receptors which function as co-receptors for HIV
  • Inflammatory cells found in regions of mucosal inflammation include a majority of CD 4-positive T-lymphocytes, are ofthe activated, memory phenotype, express high levels ofthe requisite co-receptors for HIV and are the preferred target cell for HIV.
  • the co-receptors including the chemokine receptors are a normal part of the endogenous inflammatory immune response functioning as receptors for ⁇ - chemokines.
  • chemokines and their receptors when activated, trigger a marked recruitment of circulating inflammatory cells to the mucosal site, resulting in cellular and soluble inflammation.
  • Inhibiting mucosal tissue infection, which is enhanced by the presence of inflammation, by providing anti-inflammatory agents will provide an effective method in reducing the activation, progression, and spread of such mucosal tissue diseases.
  • anti-inflammatory agents pose a useful method for mitigating, controlling, or diminishing the inflammatory response caused by a retroviral infection. Because inflammation usually results in the recruitment and activation of other inflammatory cells through pro-inflammatory mediators, including, but not limited to, prostaglandins, leukotrienes, cytokines, chemokines and others recognized by those of skill in the art, reducing such recruitment and activation will reduce the available number of CD 4-positive cells available for HIV infection.
  • pro-inflammatory mediators including, but not limited to, prostaglandins, leukotrienes, cytokines, chemokines and others recognized by those of skill in the art
  • Anti-inflammatory agents useful in the invention include agents that decrease the recruitment of inflammatory cells, decrease the production of chemokines, and pro-inflammatory cytokines that foster the perpetuation ofthe inflammatory cascade, and agents that inhibit chemokine or cytokine receptors (e.g., by inhibiting interaction of a chemokine receptor with its ligand) thereby preventing propagation ofthe inflammatory message.
  • agents include "anti-inflammatory antibodies” which bind to and prevent the biological activity ofthe protein molecules described above antibody.
  • Such antibodies include antibodies designed to interact with cytokines, cytokine receptors, chemokines, chemokine receptors, which are designed or provided to a subject (e.g., a human) to reduce or prevent an inflammatory response.
  • the invention also contemplates various pharmaceutical compositions that block retroviral and immunodeficiency virus replication or cytokine secretion in response to an immunodeficiency replication.
  • the pharmaceutical compositions according to the invention are prepared by bringing an antibody, an isolated peptide, a nucleic acid sequence, or other anti-inflammatory agent or drug according to the present invention into a form suitable for administration (e.g., a pharmaceutically acceptable carrier) to a subject using carriers, excipients and additives or auxiliaries.
  • Frequently used carriers or auxiliaries include magnesium carbonate, titanium dioxide, lactose, mannitol and other sugars, talc, milk protein, gelatin, starch, vitamins, cellulose and its derivatives, animal and vegetable oils, polyethylene glycols and solvents, such as sterile water, alcohols, glycerol and polyhydric alcohols.
  • Intravenous vehicles include fluid and nutrient replenishers.
  • Preservatives include antimicrobial, anti-oxidants, chelating agents and inert gases.
  • Other pharmaceutically acceptable carriers include aqueous solutions, non-toxic excipients, including salts, preservatives, buffers and the like, as described, for instance, in Remington's Pharmaceutical Sciences, 15th ed.
  • Such pharmaceutical compositions may include one or more anti-inflammatory agents and one or more anti- viral agents in combination.
  • the invention in another embodiment, relates to a method of blocking or inhibiting replication or spread of an immunodeficiency virus or the secretion of cytokines in response to an immunodeficiency virus.
  • This method involves administering to a subject a therapeutically effective dose of a pharmaceutical composition containing the compounds of the present invention and a pharmaceutically acceptable carrier.
  • administering the pharmaceutical composition ofthe present invention may be accomplished by any means known to the skilled artisan.
  • subject is meant any mammal, preferably a human.
  • the pharmaceutical compositions are preferably prepared and administered in dose units.
  • Solid dose units are tablets, capsules, and suppositories.
  • different daily doses are necessary. Under certain circumstances, however, higher or lower daily doses may be appropriate.
  • the administration ofthe daily dose can be carried out both by single administration in the form of an individual dose unit or else several smaller dose units and also by multiple administration of subdivided doses at specific intervals.
  • the dosage should not be so large as to cause adverse side effects, such as unwanted cross-reactions, anaphylactic reactions, and the like.
  • the dosage will vary with the age, condition, sex, and extent ofthe disease or infection in the patient and can be determined by one skilled in the art.
  • the dosage can be adjusted by the individual physician in the event of any contraindications and can be readily ascertained without resort to undue experimentation.
  • the effectiveness of treatment can be determined, for example, by monitoring the level of HIV RNA or DNA viral burden in a patient infected with an immunodeficiency virus at the site of inflammation (e.g., mucosal tissue) or by other means including measurement of inflammatory mediators, cytokines, chemokines and/or CD4+ cells.
  • An decrease or stabilization in the relative number of CD4+ cells, level of cytokines, pro- inflammatory mediators or chemokines in the tissue should correlate with the level of inflammation in the individual or tissue.
  • compositions according to the invention are in general administered topically, intravenously, orally or parenterally or as implants. Rectal and vaginal administration may prove more effective as these are typically the sites of first contact and inflammation of mucosal tissue. Such sites can be contacted to prevent or inhibit infection or transmission.
  • Suitable solid or liquid pharmaceutical preparation forms are, for example, granules, powders, tablets, coated tablets, (micro)capsules, suppositories, syrups, emulsions, suspensions, creams, gels, aerosols, drops or injectable solution in ampule form and also preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavorings, sweeteners or solubilizers are customarily used as described above.
  • the pharmaceutical compositions are suitable for use in a variety of drug delivery systems.
  • compositions according to the invention may be administered locally or systemically.
  • therapeutically effective dose is meant the quantity of a compound according to the invention necessary to prevent, to cure or at least partially arrest or decrease the symptoms ofthe disease and its complications. Amounts effective for this use will, of course, depend on the severity of the disease or infection and the weight and general state ofthe subject. Typically, dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of the pharmaceutical composition, and animal models may be used to determine effective dosages for treatment of particular disorders.
  • Effectiveness ofthe dosage can by monitored by determining, for example, the level of HIV RNA or DNA or viral burden in a patient infected with an immunodeficiency virus at the site of inflammation (e.g., mucosal tissue), by a decrease in one or more symptoms associated with the infection, or by other means including measurement of amounts of inflammatory mediators, cytokines, chemokines and/or CD4+ cells, using methods well known to one of ordinary skill in the art.
  • the immunotherapeutic method of the invention includes a prophylactic method directed to those hosts at risk for the immunodeficiency virus infection. For example, the method is useful for humans at risk for HIV infection.
  • a "prophylactically effective" amount of an anti-inflammatory agent for example, refers to that amount which is capable of inhibiting HIV replication, activation, or progression by reducing, inhibiting or preventing an inflammatory response.
  • Transmission of HIV occurs by at least three known routes: sexual contact, blood (or blood product) transfusion and via the placenta. Infection via blood includes transmission among intravenous drug users. Since contact with HIV does not necessarily result in symptomatic infection, as determined by seroconversion, all humans may be potentially at risk and, therefore, should be considered for prophylactic treatment by the therapeutic method ofthe invention.
  • Anti-inflammatory compositions useful in the invention for prophylactic activity or for preventing transmission include, for example, condoms coated or lubricated with an anti-inflammatory agent, condoms comprising a capsule present on the condom that upon use ruptures to release an anti-inflammatory agent, vaginal products including diaphragms, cervical caps, sponges, and rings coated or lubricated with an anti-inflammatory agent; vaginal douches, creams, gel lubricants, suppositories, foams, or spermicidal gels which also contain an anti-inflammatory agent, and other compositions and articles of manufacture known to those of skill in the art for local delivery of an anti-inflammatory agent to a mucosal tissue contacted during sexual intercourse.
  • Such articles of manufacture are useful in preventing or inhibiting infection of a subject by another, or useful in preventing or inhibiting transmission from an infected subject to another.
  • compositions described herein and useful in a method ofthe invention can be administered to a patient prior to infection with an immunodeficiency virus (i.e., prophylactically) or at any of the stages described below, after initial infection, or after infection in order to prevent subsequent transmission.
  • an immunodeficiency virus i.e., prophylactically
  • HIV infection may run any ofthe following courses:
  • PGL generalized lymphadenopathy
  • ARC AIDS-related complex
  • anti-inflammatory agent means an agent capable of reducing, preventing or modulating an inflammatory reaction, for example, by decreasing recruitment of inflammatory cells, decreasing chemokine production, decreasing pro-inflammatory chemokine production, decreasing pro-inflammatory cytokine production, or inhibiting interaction of a chemokine or cytokine with its receptor.
  • Such agents include, for example, anti-inflammatory antibodies (e.g., anti- cytokine, anti-receptor antibodies), peptides (e.g., agonist or antagonist of inflammatory mediators, cytokines such as IL-1, or receptors such as IL-1 receptor antagonists or soluble TNF receptors), nucleic acids (e.g., nucleic acids which encode anti-inflammatory agents such as anti-inflammatory peptides, ribozymes or antisense molecules), steroids (e.g., prednisone), non-steroidal anti-inflammatory drugs (e.g., aspirin), a 5-ASA product, commonly used anti-inflammatory drugs and combinations thereof.
  • anti-inflammatory antibodies e.g., anti- cytokine, anti-receptor antibodies
  • peptides e.g., agonist or antagonist of inflammatory mediators, cytokines such as IL-1, or receptors such as IL-1 receptor antagonists or soluble TNF receptors
  • nucleic acids e.g.
  • anti-inflammatory antibodies useful in the present invention include antibodies to cytokines and their receptors, such as anti-interleukin receptors, anti-cytokine antibodies (e.g. anti-TNF antibodies, such as REMICADE® made by Centocor), anti-chemokine antibodies (see for example Olson et al, J. of Virol. 73(5):4145-4155 (1999), the disclosure of which is incorporated herein), anti- chemokine receptor antibodies (e.g., anti-CCR5 or anti-CXCR4 receptor antibodies) and combinations thereof.
  • Other antibodies include antibodies to enzymes of enzymatic pathways which produce pro-inflammatory mediators, for example antibodies to type-1 phospholipase A2 as disclosed in U.S. Patent No.
  • anti-inflammatory nucleic acids useful in the present invention include nucleic acids encoding an anti-inflammatory peptide, a ribozyme that cleaves RNA-encoding pro-inflammatory polypeptides (e.g., cytokines or chemokines), antisense molecules capable of hybridizing to nucleic acid sequence which encode pro-inflammatory mediators, such as cytokines, cytokine receptors, chemokines, chemokine receptors, other inflammatory peptides or receptors as disclosed herein, and combinations thereof, or easily identifiable to one of ordinary skill in the art.
  • pro-inflammatory polypeptides e.g., cytokines or chemokines
  • antisense molecules capable of hybridizing to nucleic acid sequence which encode pro-inflammatory mediators, such as cytokines, cytokine receptors, chemokines, chemokine receptors, other inflammatory peptides or receptors as disclosed herein, and combinations thereof, or easily identifiable to one of ordinary skill in the art.
  • anti-inflammatory peptides include, for example, LFA adhesion molecule antagonist, cytokine receptor antagonist, transcription factor, soluble TNF- ⁇ receptor polypeptide.
  • Other anti-inflammatory peptides include for example, transcription factors such as NF-kappa B (Schottelius AJ et al, Int J Colorectal Dis 1999 Feb;14(l):18-28), peptide to platelet factor 4 (U.S. Patent No. 5,776,892, which is incorporated herein), and peptides based on CD14 as disclosed in U.S. Patent No. 5,766,593 (the disclosure of which is incorporated herein).
  • anti-inflammatory cytokines include, for example, cytokines and transcription factors.
  • Anti-inflammatory cytokines include, for example, IL-13 (Watson ML, Am J Respir Cell Mol Biol 1999 May 1; 20(5): 1007-1012), IL-4 and IL-10 (Jarvelainen HA et al, Hepatology 1999 May;29(5):1503-10), IL-16 (Klimiuk PA, et al, J Immunol. 1999 Apr l;162(7):4293-4299) and other anti-inflammatory cytokines known to those of skill in the art.
  • anti-inflammatory agents useful in the present invention include agents from a wide variety of steroidal, non-steroidal, and salicylate water-soluble and water-insoluble drugs and their acid addition or metallic salts. Both organic and inorganic salts may be used provided the anti-inflammatory agent maintains its medicament value.
  • the anti-inflammatory agents may be selected from a wide range of therapeutic agents and mixtures of therapeutic agents which may be administered in sustained release or prolonged action form.
  • Non-steroidal anti-inflammatory agents include numerous compounds of diverse chemical structure. Most if not all are believed to share a common mechanism of action, and almost all are weak organic acids. This large group of compounds can be divided into two main groups, carboxylic acids (R ⁇ COOH) and enolic acids (R--COH). Further subdivisions based on chemical structure can be made.
  • the main groups of enolic acids are the pyrazolones, such as phenylbutazone, oxyphenbutazone, dipyrone and isopyrin, and the xicams, which include piroxicam and miloxicam.
  • Carboxylic acid subgroups comprise the salicylates, e.g.
  • acetylsalicylate (aspirin); propionic acids, e.g. ibuprofen and naproxen; anthranilic acids, e.g. meclofenamic acid; phenylacetic acids, e.g. acetaminophen; aminonicotinic acids, e.g. flunixin; and indolines, e.g., indomethacin.
  • NSAIDs for which the mechanism of action is known, most have been found to inhibit the formation of arachidonic acid metabolites through suppression of cyclooxygenase and lipoxygenase pathways and thus lead to a reduction in inflammation mediated by these metabolites.
  • Cyclooxygenase converts arachidonic acid to the cyclic endoperoxides, PGG2 and PGH2 (known as PGs). By action of further specific enzymes, these compounds are converted to different members ofthe family of inflammatory mediators, the eicosanoids, which includes PGE2 and PGI2.
  • the structure of cyclooxygenases varies among tissues, and NSAIDs differ in their ability to combine with each of these enzymes, which explains differences in potency and species responses.
  • Non-limiting illustrative specific examples of non-steroidal anti-inflammatory agents with brand names, generic names, standard sizes of doses, and chemical structures include the following medicaments: ibuprofen (e.g., MOTRJN® 300, 400, 600, 800 mg, ADVIL® 200 mg) ( ⁇ )-2-(p-isobutylphenyl) propionic acid; tolmetin (TOLECTIN®) 5-(p-toluoyl)- 1 -methylpyrrole-2-acetic acid; naproxen (e.g. ,
  • ALEVE®, ANAPREX®, or NAPROSYN® 250, 375, and 500 mg, 6-methoxy- ⁇ - methyl-2-naphthaleneacetic acid, (+); flurbiprofen (ANSAID®), 50 and 100 mg, 2- fluoro- ⁇ -methyl- [l,l'-biphenyl]-4-acetic acid, ( ⁇ ); sulindac (CLINORN®)150 and 250 mg, 5-fluoro-2-methyl-l-[[p-(methylsulfinyl)phenyl]-methylene]-lH-indene-3- acetic acid; diflunisal (FLOVACIL®) 250 and 500 mg, 2'4'-difluoro-4-hydroxy-
  • piroxicam FELDENE® 4-hydroxy-2-methyl-N- 2-pyridinyl-2H- 1 ,2-benzothiazine-3-carboxamide 1 , 1 -dioxide; indomethacin (INDOCIN®), 25 and 50mg, l-(4-chlorobenzoyl)-5- methoxy-2-methyl-lH-indole-3- acetic acid; etodolac (ULTRADOL®) l,8-deethyl-l,3,4,9-tetrahydropyrano-[3,4- b]indole-l -acetic acid; meclofenamate sodium (MECLOMEN®), 50 and 100 mg, N- (2,6-dichloro -M-tolyl) anthranilic acid, sodium salt, monohydrate; fenoprofen and fenoprofen calcium (
  • Non-limiting illustrative specific examples of salicylate anti-inflammatory agents include the following medicaments: bismuth subsalicylate; salsalate; salicylic acid and salicylic acid derivatives, for example, sodium thiosalicylate, choline salicylate, magnesium salicylate, diflunisal, ibuprofen, naproxen, sulindac, diflunisal, salicylsalicylic acid, choline magnesium trisalicylate, acetylsalicylic acid, salsalate, sodium salicylate and combinations thereof; 5-aminosalicylic acid (5-ASA) and 5- ASA containing product or compounds, for example: oral mesalamine (ASACOL®, made by Procter & Gamble Pharmaceuticals; PENTASA® made by Roberts Pharmaceuticals), mesalamine rectal enema or foam or suppository, sulfasalazine, balsalazide, ipsalazide, and olsalazine (DEPENT
  • Steroidal anti-inflammatory agents include glucocorticoids.
  • Non-limiting illustrative specific examples of steroidal anti-inflammatory agents include the following medicaments: flunisolide, triamcinoline, triamcinoline acetonide, beclomethasone diproprionate, betamethasone diproprionate, hydrocortisone, cortisone, dexamethasone, budesonide, prednisone, methyl prednisolone, prednisolone, esters of any of these compounds and combinations thereof.
  • Other non- limiting examples of anti-inflammatory agents include Thalidomide (made by Celgene).
  • Retroviruses are RNA viruses wherein the viral genome is RNA.
  • the genomic RNA is reverse transcribed into a DNA intermediate which is integrated very efficiently into the chromosomal DNA of infected cells.
  • the integrated DNA intermediate is referred to as a provirus.
  • the family Retroviridae are enveloped single-stranded RNA viruses typically infect mammals, such as, for example, bovines, monkeys, sheep, and humans. Retroviruses are unique among RNA viruses in that their multiplication involves the synthesis of a DNA copy of the RNA which is then integrated into the genome ofthe infected cell.
  • the Retro viridae family consists of three groups: the spumaviruses (or foamy viruses) such as the human foamy virus (HFV); the lentiviruses, as well as visna virus of sheep; and the oncoviruses (although not all viruses within this group are oncogenic).
  • the term "lentivirus” is used in its conventional sense to describe a genus of viruses containing reverse transcriptase.
  • the lentiviruses include the "immunodeficiency viruses” which include human immunodeficiency virus (HIV) type 1 and type 2 (HIV-1 and HIV-2) and simian immunodeficiency virus (SIV).
  • A-type particles represent the immature particles ofthe B- and D-type viruses seen in the cytoplasm of infected cells. These particles are not infectious.
  • B-type particles bud as mature virions from the plasma membrane by the enveloping of intracytoplasmic A-type particles.
  • B-type particles contain an eccentrically located, electron-dense core.
  • the prototype B-type virus is mouse mammary tumor virus (MMTV). No intracytoplasmic particles can be observed in cells infected by C- type viruses. Instead, mature particles bud directly from the cell surface via a crescent 'C '-shaped condensation which then closes on itself and is enclosed by the plasma membrane. Envelope glycoprotein spikes may be visible, along with a uniformly electron-dense core. Budding may occur from the surface plasma membrane or directly into intracellular vacuoles.
  • the C-type viruses are the most commonly studied and include many of the avian and murine leukemia viruses.
  • Bovine leukemia virus BLV
  • HTLV-I/II human T-cell leukemia viruses types I and II
  • D-type particles resemble B-type particles in that they show as ring-like structures in the infected cell cytoplasm, which bud from the cell surface, but the virions incorporate short surface glycoprotein spikes.
  • the electron-dense cores are also eccentrically located within the particles.
  • Mason Pfizer monkey virus MPMV is the prototype D-type virus.
  • Retroviruses are defined by the way in which they replicate their genetic material. During replication the RNA is converted into DNA. Following infection of the cell a double-stranded molecule of DNA is generated from the two molecules of RNA which are carried in the viral particle by the molecular process known as reverse transcription. The DNA form becomes covalently integrated in the host cell genome as a provirus, from which viral RNAs are expressed with the aid of cellular and/or viral factors. The expressed viral RNAs are packaged into particles and released as infectious virions.
  • the retrovirus particle is composed of two identical RNA molecules. Each genome is a positive sense, single-stranded RNA molecule, which is capped at the 5 end and polyadenylated at the 3' tail.
  • the diploid virus particle contains the two RNA strands complexed with gag proteins, viral enzymes (pol gene products) and host tRNA molecules within a ' core' structure of gag proteins. Surrounding and protecting this capsid is a lipid bilayer, derived from host cell membranes and containing viral envelope proteins. The env proteins bind to the cellular receptor for the virus and the particle typically enters the host cell via receptor-mediated endocytosis and/or membrane fusion.
  • the viral RNA is copied into DNA by reverse transcription. This is catalyzed by the reverse transcriptase enzyme encoded by the pol region and uses the host cell tRNA packaged into the virion as a primer for DNA synthesis. In this way the RNA genome is converted into DNA genome.
  • the double-stranded linear DNA produced by reverse transcription may, or may not, have to be circularized in the nucleus before integration into the host cell genome.
  • the provirus now has two identical repeats at either end, known as the long terminal repeats (LTR).
  • LTR long terminal repeats
  • the junction between the two joined LTR sequences produces the site recognized by a pol product - the integrase protein — which catalyzes integration, such that the provirus is always joined to host DNA two base pairs (bp) from the ends ofthe LTRs.
  • bp base pairs
  • RNA splicing and translation ofthe integrated viral DNA is mediated by host cell proteins.
  • Variously spliced transcripts are generated.
  • HIV- 1/2 and HTLV-I/II viral proteins are also used to regulate gene expression. The interplay between cellular and viral factors is important in the control of virus latency and the temporal sequence in which viral genes are expressed.
  • Retroviruses can be transmitted horizontally and vertically. Efficient infectious transmission of retroviruses requires the expression on the target cell of receptors, which specifically recognize the viral envelope proteins, although viruses may use receptor-independent, nonspecific routes of entry at lesser efficiency. In addition, the target cell type must be able to support all stages ofthe replication cycle after virus has bound and penetrated (nucleic acid ofthe virus has entered the cell). Vertical transmission occurs when the viral genome becomes integrated in the germ line of the host. The provirus will then be passed from generation to generation as though it were a cellular gene. Hence, endogenous proviruses become established which frequently lie latent, but which can become activated when the host is exposed to appropriate agents.
  • the antiviral agents for use in the compositions and methods of the invention can target any stage of the virus life cycle.
  • the oncoviruses (often called the RNA tumor viruses) have been subdivided into two groups of pathogens, namely the acutely transforming and slow transforming retroviruses.
  • Acutely transforming retroviruses can transform cultured cells and can cause disease rapidly in susceptible animals. These viruses usually carry an oncogene (v- onc) within the viral genome, which is directly responsible for their tumorigenicity, and which is different in each type of virus.
  • the viral oncogenes have been derived from cellular genes that the viruses have acquired, probably as a result ofthe inclusion of cellular RNA within a viral particle. Subsequent recombination between viral and cellular RNA during reverse transcription leads to the incorporation ofthe cellular sequences into the viral genome and delivery of this novel unit into the host cell DNA.
  • transduced gene normally has a central role in control of cellular growth and differentiation, the changes in coding sequence and/or control of expression that it undergoes on incorporation into the viral genome can render it oncogenic.
  • Such cellular proto-oncogenes may become oncogenic by being placed under novel, virally determined transcriptional control (both quantitatively and temporally), and/or by sustaining critical mutations to the coding sequence.
  • full cellular transformation usually requires the expression of v-onc in conjunction with other genetic and epigenetic changes within the target cell.
  • the slow transforming retroviruses typically do not contain a 'classical' oncogene.
  • the mechanism of transformation is believed rather to involve the insertion of provirus near, or in, the coding region of a cellular proto-oncogene, called insertional mutagenesis.
  • the strong promoter and enhancer sequences within the viral LTRs can exert transcriptional effects from distances of up to several kilobase pairs from the proto-oncogene.
  • the normal regulation of expression ofthe cellular gene is disrupted, and over-expression or inappropriately timed expression can contribute to transformation.
  • HTLV-I is a slow-transforming virus, causally associated with adult T-cell leukemia (ATL), but it probably promotes T-cell transformation by a different pathway involving virally encoded regulatory proteins, especially p40tax, which transactivate expression of cellular proto-oncogenes.
  • HIV-1 and 2 have also been implicated in both the direct and indirect promotion of various types of malignancy (such as Kaposi's sarcoma), which present much more frequently in AIDS patients than in the general population.
  • malignancy such as Kaposi's sarcoma
  • the direct role of HIV in malignant transformation remains doubtful as many patients who are immunosuppressed as a result of other infections or treatments (e.g. transplant recipients) also develop tumors at increased rates.
  • the D-type viruses are not etiologically associated with malignancy, although MPMV was initially associated with a mammary tumor in a rhesus monkey. D-type viruses cause immune suppression in simian primates but by an unknown mechanism. Immune suppression is also a feature of infection by the lentiviruses (e.g. EHV and SIV) and variant strains of feline leukemia virus (FeLV). n infection with HIV and FeLV large amounts of unintegrated proviral DNA have been observed, which may be related to the pathogenesis.
  • lentiviruses e.g. EHV and SIV
  • FeLV feline leukemia virus
  • HIV- 1/2 and visna virus of sheep are associated with slow progressive disease leading to immune suppression and neurological disorders.
  • HIV is the widely recognized causative agent of the acquired immunodeficiency disease syndrome (AIDS).
  • compositions and methods ofthe invention including anti-inflammatory agents (antibodies, peptides, peptidomimetics, chemical compositions, etc.) alone, or in combination with antivirals (antibodies, peptides, virus protein/enzyme inhibitors, etc.) are all useful for treating subjects either having or at risk of having an immunodeficiency virus (e.g., HIV) related disorder, or having or at risk of transmitting an HIV related disorder.
  • HIV immunodeficiency virus
  • AIDS and ARC are particular examples of such disorders.
  • HIV-associated disorders have been recognized primarily in "at risk” groups, including homosexually active males, intravenous drug users, recipients of blood or blood products, and certain populations from Central Africa and the Caribbean. The syndrome has also been recognized in heterosexual partners of individuals in all "at risk” groups and in infants of affected mothers.
  • Retroviruses have been linked to a wide range of diseases, including anemia, neurological disorders, immune suppression, and malignancy.
  • HTLV-I for example, is associated with tropical spastic paraparesis, a condition similar in some respects to multiple sclerosis.
  • antiviral agent means an agent capable of inhibiting, reducing, preventing or modulating viral infection or production at any step or stage of the viral life cycle, for example, by inhibiting, reducing, preventing an initial step of virus infection, such as virus fusion to a cell through a cell surface receptor or independent of a cell surface receptor; subsequent entry of viral nucleic acid into the cell ("entry inhibitor”); reverse transcription of viral nucleic acid (“reverse transcriptase inhibitor”); integration of reverse transcribed viral nucleic acid into the genome of the cell (“integrase inhibitor”); proviral nucleic acid transcription or replication; translation or formation of mature viral proteins; formation/assembly of infectious viral particles; decreasing budding or release of mature virions from a cell; and decreasing activity or an amount of an enzyme associated with viral fusion or infection, replication, maturation, or budding or release from a cell.
  • anti-viral agents examples include polypeptides or functional mimetics, for example, a soluble cell surface receptor peptide that binds virus, a ligand to the cell surface receptors, an antibody or an antibody fragment that binds to the cell surface receptor or the virus particle thereby preventing binding between the virus and a cell surface receptor present on a cell.
  • Viral proteins particularly attractive for anti-viral agent targeting include envelope polypeptide, gpl20 or gp41.
  • Cell surface receptors particularly attractive for anti-viral agent targeting include X4 and R5 receptors.
  • the term "functional mimetic” means a molecule that is chemically or structurally modified, but has one or more activities (i.e., functions), or even increased activity, of the unmodified molecules. Thus, in the case of a functional mimetic of an antiviral agent, the mimetic would retain one or more antiviral activities ofthe antiviral agent.
  • a particular type of functional mimetic is a polypeptide or peptide mimetic ("peptidomimetic") which is a compound that mimics the three-dimensional structure ofthe peptide from which the mimetic is intended to mimic.
  • Peptidomimetic design can be aided through three-dimensional computer modeling techniques and can be designed to have additional characteristics that enhance therapeutic application.
  • antisense molecules can be designed with non-natural nucleotides or chemically modified in order to inhibit nuclease digestion of the antisense in vivo.
  • antiviral agents are where cellular or viral enzymes important for the viral life cycle, such as viral protease, reverse transcriptase or integrase, are inhibited.
  • viral fusion inhibitors e.g., T20 and T20 analogues (Trimeris, Inc.); entry inhibitors; integrase inhibitors; protease inhibitors (e.g., saquinavir, ritonavir, indinavir, nelfinavir, amprenavir); a nucleoside reverse transcriptase inhibitor (e.g., zidovudine (AZT), stavudine (d4T), lamivudine (3TC), didanosine (DDI), zalcitabine (ddC), abacavir); a non-nucleoside reverse transcriptase inhibitor (e.g., nevirapine, delavirdine, efavirenz);
  • a nucleoside reverse transcriptase inhibitor
  • HAART highly active anti-retroviral therapy
  • HAART may reduce viral load to undetectable levels in the plasma
  • the vast majority of patients will suffer a rebound increase in plasma viremia when therapy is halted, presumably due to the reservoirs of virus that exist beyond the bloodstream.
  • This viral reservoir has been discovered in lymphoid tissue where the majority ofthe body's lymphocytes reside (98%). Since the gastrointestinal mucosa contains the majority ofthe body's lymphocytes (40-65%), it likely represents the largest reservoir of HIV, and, therefore, is a primary target for anti-HIV therapy. Given the increased inflammatory state ofthe mucosa in HIV disease, one focus of this invention entails methods to decrease mucosal inflammation.
  • a number of techniques known to those of skill in the art can be used to assess in vitro, as well as in vivo, the effect of anti-inflammatory agents on the ability to inhibit, treat, or reduce the activation or progression of retroviral infections in mucosal cells, tissues, and subjects. Such techniques are applicable to a wide range of anti-inflammatory agents and mucosal cell infections. For example, using the techniques described below, one skilled in the art would be able to assess the effect of an anti-inflammatory agent on HIV infections in mucosal tissues and mucosal tissue in a subject. For exemplification only, outlined below is a method of assessing the effect of Thalidomide and 5 -ASA containing compounds or products, however, such examples are meant to illustrate and not limit the scope of the present invention. Data are also described showing inhibition of HIV replication in cells using mesalamine (Asacol).
  • Thalidomide is a potent anti-inflammatory medication. While it owes much of its immunomodulatory effect to its ability to down-regulate TNF ⁇ production, it appears to affect multiple sites in the inflammatory cascade. Other putative anti- inflammatory mechanisms include a role in attenuating T cell proliferation, down- regulating the level of activation of lymphocytes, inhibition of lymphocyte chemotaxis, and altering the levels of pro-inflammatory cytokines. Thalidomide may prove to be a beneficial component of HIV therapy by decreasing HIV replication systemically and in the gastrointestinal mucosa. In this regard, thalidomide can be used as an adjunct to HAART in patients with undetectable plasma viral load. Treatment with thalidomide will result in a decrease in the ability of HIV to replicate in a lymphoid environment that favors HIV replication as evidenced by lower tissue levels of HIV RNA burden in treated subjects.
  • thalidomide Studies ofthe effect of thalidomide on the infectivity of cells by HIV are performed in vitro. Patients with moderate to severely active Crohn's disease (colitis) who have begun therapy with thalidomide will be identified and recruited to help differentiate non-specific inflammatory reactions from infectious effects. Mucosal and blood cells from these subjects will be used to evaluate thalidomide 's ability to decrease infectibility, in vitro.
  • the state of inflammation is enhanced in the presence of HIV infection and is characterized by elevated levels of pro-inflammatory cytokines and chemokines. These soluble mediators play a vital role in chemoattraction of chemokine-bearing CD4+ cells to the mucosa. These targets for HIV are further activated in the mucosal environment by the soluble mediators leading to enhanced HIV replication.
  • cytokines TNF ⁇ , IL-1, IL-6, ⁇ -IFN
  • MIP la, MIP lb, RANTES chemokines
  • MMCs mucosal mononuclear cell isolation, Rnase protection assay (RPA), PCR for HIV RNA and proviral DNA as well as quantitative image analysis (QIA).
  • MMCs for flow cytometry will be stained for CD45, CD4, CD8, HLA-DR, CD45RO, CXCR4, CCR5, LFA-1, and ICAM-1. Flow cytometry is performed on MMCs isolated from the mucosa and on
  • PBMCs isolated from the blood to determine the composition ofthe lymphocyte populations in these compartments at baseline.
  • the activation state and memory state ofthe lymphocytes will be noted, as will the expression of chemokine receptors and adhesion markers.
  • ⁇ -chemokines and cytokines are quantified using the Multi-probe RPA (Riboquant, Pharmingen). This sensitive and specific assay allows quantification of chemokine and cytokine mRNA from extracted RNA from frozen biopsies. The quantity ofthe individual mRNA species will be compared with GADPH as a housekeeping gene. If chemokines and cytokines cannot be quantified with the RPA, they can be measured in supernatants of stimulated cultured MMCs by ELISA.
  • QIA utilizes immunohistochemical staining of paraffin-embedded tissue to accurately determine the number and anatomical position of lymphocytes bearing CD4, CD8, CD38, HLA-DR, CD45RO CXCR4 and CCR5 in a biopsy. In addition, it permits determination of which cells in a biopsy are producing cytokines and ⁇ - chemokines and the tissue concentrations of these factors. In HTV seropositive biopsies, the tissue levels of p24 can also be determined. A computerized image analyzer and specialized software are available to assess the total tissue area for stained cells, chemokines, cytokines, and p24 levels.
  • Results will confirm that the mucosal response to HIV is characterized by increased tissue chemokine and cytokine levels. This will, in turn result in a mild increase in the mucosal inflammatory infiltrate. By investigating chemokines and cytokines, "soluble" inflammation that would be overlooked by simply examining a cellular index will be confirmed.
  • a potent immunomodulator will decrease mucosal inflammation in patients infected with HIV. This will likely manifest as a decrease in the ability of HIV to replicate in this, the largest lymphoid organ.
  • thalidomide can further improve the life expectancy ofthe millions of patients worldwide infected with this virus.
  • Subjects with plasma undetectable HIV, having undergone baseline biopsies are treated with oral thalidomide therapy at a dose of 200 grams per day for 16 weeks. After 4 and 16 weeks of therapy, the subjects will undergo repeat endoscopic biopsies and repeat phlebotomy to obtain PBMCs and the same biomolecular experiments as described above are performed.
  • RT- and DNA-PCR are performed to analyze changes between baseline and post-therapy mucosal and plasma viral load. For statistical evaluation, the solitary index of efficacy will be a decrease in mucosal HIV viral load without a concomitant increase in plasma viral load.
  • MMCs and PBMCs obtained at the baseline and 4 and 16 weeks of thalidomide therapy are cultured with known titers of M-tropic HIVsx and T-tropic HIVNL4-3. Supernatants are collected for ELISA to quantify p24 at time 0 hrs, 36hrs, 3 days, and 7 days. Paired T tests to analyze changes in susceptibility of MMCs from IBD patients before and after therapy will be performed.
  • thalidomide will alter the inflammatory environment ofthe gastrointestinal mucosa, resulting in a decrease in soluble inflammatory mediators (i.e., pro-inflammatory cytokines, chemokines), which in turn, will decrease recruitment of immune cells that HIV can infect. Reduction of inflammation by thalidomide therapy will impact p24 production in PBMCs and MMCs in the in vitro infection experiment. It is anticipated that the main benefit of thalidomide will be via decreased recruitment of additional co-receptor-bearing, CD4+ T cells as viral targets due to attenuated soluble inflammatory mediators in the mucosa, but it is possible that decreased expression of adhesion molecules may also play a role.
  • soluble inflammatory mediators i.e., pro-inflammatory cytokines, chemokines
  • Mesalamine e.g., Asacol
  • Mesalamine is a mucosal anti-inflammatory medication taken orally or by enema, foam or suppository that is available for topical therapy. It will be a beneficial component of HIV therapy by decreasing HIV replication in gastrointestinal mucosa. Efforts to minimize mucosal inflammation with Mesalamine may be therapeutically beneficial by decreasing the concentration of pro- inflammatory cytokines and chemokines in the mucosal compartment and therefore recruitment and activation of additional cellular targets for HIV. By decreasing the concentration and activity of replicating HIV, it may also slow the development of HIV resistance to commonly used antiviral medications.
  • Mesalamine can therefore be a powerful, locally active, adjunct in the treatment, inhibition of proliferation/spread within a subject having HIV, as well as inhibit or prevent HIV infection of a subject at risk of infection, and inhibit or prevent transmission of an HIV infected subject to another subject (uninfected or infected).
  • Subjects with known, moderately active infections based on clinical, endoscopic, and histologic examination but have not been receiving Mesalamine or other 5-ASA medications for 4 weeks and have not been receiving immunomodulatory medications or steroids (oral or topical) for 3 months will be studied.
  • Non-colitic control patients will be age-matched to the colitis patients and be free of symptoms referable to their colon, and without endoscopic evidence of inflammation.
  • Colitis subjects will be seen for 2 baseline endoscopic evaluations performed one- week prior and the day of initiation of therapy.
  • Non-colitic control patients will also undergo two endoscopy examinations separated by one week. The soluble and cellular inflammatory state of mucosal samples from the ulcerative colitis and non-colitic controls will be compared.
  • endoscopic biopsies (3.3 mm OD) from a standardized level (30cm) will be obtained from each patient. Ofthe biopsies, 12 will be used for MMC isolation, 1 for RPA, and 2 for QIA. MMCs for flow cytometry will be stained for CD45, CD4, CD8, CD38, HLA-DR, CD45RO, CCR5, and CXCR4. ⁇ -chemokines and cytokines will be quantified in supernatants obtained after culture of MMCs and PBMCs in IL-2 containing medium by ELISA. One million cells in 1 ml medium in 6 well plates will be used.
  • RNA samples After 18 hours of culture, 200 ml medium will be taken for measurement of RANTES, MlP-la, MlP-lb, IL-lb, IL-12, TNF- ⁇ , and IFN- ⁇ . If needed, nucleic acid will be extracted from a liquid nitrogen frozen biopsy to determine the level of mRNA for these cytokine and chemokine species using the Multi-probe RPA (Riboquant, Pharmingen). T he quantity of the individual mRNA species will be compared with GADPH as a housekeeping gene.
  • QIA utilizes immunohistochemical staining of fresh frozen or paraffin- embedded tissue to accurately determine the number and anatomical position of lymphocytes bearing CD4, CD8, CD38, HLA-DR, CD45RO, and CCR5 in a biopsy. In addition, it permits determination of which cells in a biopsy are producing cytokines and ⁇ -chemokines, the tissue concentrations of these factors, and by inference, the effect of these ligands on chemokine receptor expression of neighboring cells. A computerized image analyzer and specialized software are available to assess the total tissue area for stained cells, chemokines, and cytokines (expressed as % area).
  • ulcerative colitis is characterized by elevated levels of chemokines and cytokines "soluble inflammation" as well as the known increased cellular inflammation.
  • the mucosa in ulcerative colitis will be characterized by soluble and cellular inflammation that is significantly greater than in non-colitic controls. The ability of Mesalamine to reduce soluble and cellular inflammatory mediators is examined.
  • Treatment with Mesalamine will result in cytokine and chemokine levels being decreased, minimizing recruitment of CCR5 -bearing CD4+ cells and lateral spread of HIV among mucosal lymphocytes; Mesalamine-induced decreases in ⁇ - chemokine secretion may reduce the amount of ligands able to block the viral co- receptors, favoring the spread of HIV; Mesalamine will preferentially inhibit pro- inflammatory cytokines resulting in suppression of T cell recruitment while not significantly impairing chemokine receptor blockade. This appears to be the most beneficial outcome when treating HIV-infected patients.
  • the possible outcomes are examined by treating patients with moderately active inflammatory bowel disease (IBD) with Mesalamine for 16 weeks and examining the changes in mucosal lymphocyte subsets, expression of chemokine receptors, and mucosal cytokine and ⁇ - chemokine levels.
  • IBD inflammatory bowel disease
  • Subjects with moderately active ulcerative colitis having undergone two baseline biopsies are treated with Mesalamine at a dose of 4.8 grams per day for 16 weeks. After receiving 4 weeks of therapy and again after completion of 16 weeks of therapy, they will undergo repeat endoscopic biopsies and the experiments are repeated on the post-therapy specimens. These studies will include performance of flow cytometry to quantify lymphocyte subsets and chemokine receptor expression, RPA to quantitate ⁇ -chemokine mRNA and QIA for in situ analysis. Patients will continue to be managed on Mesalamine or as clinically indicated by their physicians.
  • the treatment of IBD patients with Mesalamine will alter the inflammatory environment ofthe gastrointestinal mucosa, resulting in a decrease in soluble and cellular inflammatory mediators. HIV infection of an activated CD4+, chemokine receptor bearing cell results in heightened HIV replication. The inflammatory environment that characterizes the gastrointestinal mucosa likely induces vigorous HIV replication. Therapy with Mesalamine, by reducing inflammation, will down-regulate the ability of HIV to replicate in the inflamed gastrointestinal mucosa.
  • MMCs and PBMCs To examine the effect of Mesalamine therapy on HIV replication in mucosal mononuclear cells, the susceptibility of MMCs and PBMCs, from untreated ulcerative colitis patients, to M-tropic and T-tropic HIV infection is examined in culture with and without the presence of graded doses of Mesalamine. Similar infectivity experiments will be conducted on the patient's cells after they have received 4 and 16 weeks of Mesalamine therapy.
  • MMCs and PBMCs obtained at the baseline time-points will be cultured with known titers of M-tropic HIVSX and T-tropic HIVNL4-3 in the presence and absence of graded, but physiologic concentrations of Mesalamine, based on known mucosal tissue concentrations. HIV replication will be compared in each of these samples by measurement of production ofthe HIV protein p24 in the culture supernatants.
  • MMCs and PBMCs from subjects after 4 and 16 weeks of therapy will be cultured with M- and T-tropic HIV without added Mesalamine. The p24 results gathered from these samples will be compared with the baseline samples.
  • the mucosal environment in HIV is inflammatory as evidenced by elevated concentrations of pro-inflammatory cytokines and chemokines.
  • Mesalamine will significantly reduce these soluble mediators and in doing so, reduce the migration into and activation of further cellular targets for HIV infection into the mucosa.
  • CXCR4 play a major role, with CCR5-tropic viruses predominating during initial infection, CXCR4-tropic viruses becoming more prevalent with advanced disease and heterozygosity of CCR5 contributing to longer survival.
  • Differential expression of CCR5 on mucosal CD4+ T lymphocytes could contribute to preferential transmission of M-tropic viruses.
  • mucosal mononuclear cells MMC were isolated from rectosigmoid endoscopic biopsies and obtained unstimulated phlebotomy samples from HIV-1 seronegative healthy individuals. We quantified co-receptor expression on CD4+ cells by flow cytometry.
  • tissue-based immune cells can be easily and safely obtained and isolated from the mucosal lining of the gut, a renewable tissue source.
  • tissue compartments for both persistence and transmission studies, techniques to easily and safely access lymphoid tissue are essential. Lymph node and tonsillar resection/aspirations have been the most commonly reported methods.
  • Studies using these approaches have already provided illuminating, concept-changing findings including evidence that HIV-1 activity persists in lymphoid tissue when plasma levels are stable or undetectable.
  • These tissue sources reveal the biologic events that occur in secondary, organized lymphoid structures during HIV-1 infection, but require invasive surgical support for tissue acquisition.
  • Endoscopic biopsies are safe, quick, painless, and provide access to the lymphoid compartment with 100% of samples containing lymphocytes.
  • the biopsies maintain architectural orientation and can be examined histologically, or can be dissociated for flow cytometric and tissue culture evaluation as described here.
  • CCR5 expression is markedly increased on human mucosal CD4+ lymphocytes, both as a percentage of total CD4+ lymphocytes and on a per cell basis compared to peripheral blood cells. These mucosal T cells support higher levels of viral replication than CD4+ lymphocytes from blood ( Figure 4).
  • CCR5 is the co-receptor most associated with HIV-1 in early infection, and the gastrointestinal tract is one of the most common sites of transmission as well as being the body's major lymphoid organ, the amount of detected CCR5 expression carries important implications for transmission, primary infection, ongoing local spread and treatment.
  • a third point of interest is the enhanced sensitivity of mucosal lymphocytes to infection with both CCR5- and CXCR4-tropic strains of HIV-1 compared with PBMC.
  • the extraordinarily high levels of HIVNL4-3 production in MMC cultures in the presence of IL-2 but without further stimulation ofthe cells suggests that mucosal T cells provide a rich milieu for replication of variants of HIV- 1 that use either of the major co-receptors for entry.
  • CCR5-using variants are preferentially transmitted despite the abundance of CD4+ lymphocytes at the site of mucosal transmission of HIV-1 and the susceptibility of these lymphocytes to infection by both types of virus remains an important question to answer.
  • tissue biopsies of gut mucosa can be used to obtain quantitative information on immunologic and virologic determinants that may influence HIV-1 transmission and pathogenesis.
  • Our results also show that both CCR5 and CXCR4 are highly expressed on mucosal CD4+ lymphocytes from the gastrointestinal tract in healthy, HIV-1 -seronegative individuals, and these mucosal cells are highly susceptible to infection in vitro, much more so than cells from the blood.
  • Patient population Six healthy individuals, three men and three women (mean age 45, range 24-68), were recruited for the study of lymphocyte phenotypes. Two additional men were recruited for the viral culture experiments (see below). Informed consent was obtained prior to undergoing elective endoscopy for a history of blood in stool or routine polyp screening. No subjects had diarrhea symptoms or history of intestinal inflammatory or infectious disorders. Hematoxylin and eosin stained biopsies taken in the same area as study biopsies revealed no pathology and were all normal appearing when reviewed in a blinded fashion by a gastrointestinal surgical pathologist. The study was approved by the UCLA Human Subjects Protection Committee.
  • MMC Mucosal mononuclear cells
  • the disrupted tissue was incubated at 37DC for 20 minutes in a shaking water bath. Following centrifugation, the tissue samples were digested with a mixture of collagenase and dispase (Boehringer Mannheim # 269638; 0.1 mg/mL in RPMI) for 1 hour at 37DC. Further disruption was achieved by sample passage through syringes with a series of decreasing needle gauges. Debris was removed using a 70 micron cell strainer (Falcon # 2350). Resulting cells were resuspended in RPMI containing 10% fetal calf serum.
  • BDIS CD4-fluorescein isothiocyanate and CD45-peridin chlorophyl protein
  • Caltag CD8-allophycocyanin
  • PE anti-CXCR4-R-phycoerythrin
  • CD45bright A well-defined and separate population of mucosal leukocytes was identified as CD45bright, and represented about 10-50% ofthe initial mononuclear sample population. Of these, 20-40% were CD4+ lymphocytes and 26-41 % were CD8+ T cells.
  • the observed CCR5 relative fluorescence intensity (RFI) was multiplied by a calibration factor, specifically 44, determined for our FACSCalibur.
  • This calibration factor is the number of molecules of PE detected per RFI channel number.
  • the RFI channel number can be multiplied by the calibration factor to estimate the number of mAb bound per cell. This calculation was not performed for CXCR4 because it was not available as a 1 : 1 conjugate.
  • peripheral blood mononuclear cells were isolated by Ficoll-Hypaque separation and then either stained and analyzed directly by flow cytometry for percent CD45, CD4, CD8, CCR5 and CXCR4 expression or processed through the mucosal isolation procedure (collagenase/dispase treatment) and then stained and analyzed for the antigens.
  • PBMC routinely isolated and those exposed to mucosal isolation enzymes showed no discernible differences in quadrant percentages for all antibodies studied.
  • MMCs from each healthy, HIV-seronegative volunteer were isolated from four endoscopic mucosal biopsies after mechanical disruption followed by 3 -day culture in Iscove's DMEM medium supplemented with 10% human serum, containing 10 mg/ml gentamycin, penicillin, streptomycin and glutamine. Interleukin-2 (IL-2, Amgen) was added at 20 IU per mL. A total of 105 mucosal mononuclear cells and PBMCs were plated in a 96-well plate in 100 microliters of medium after a 3-hour infection with 50 mg of HIVSX or HIVNL4-3. Prior to plating, the cells were washed twice to remove free virus and adherent p24.
  • IL-2 Interleukin-2
  • CD4+ percentages were determined by flow cytometry and were used to determine the number of CD4+ lymphocytes in the cultures. Co-receptor expression on the gut cells was not tested on these donors because cells yields were not sufficient.
  • RNA Extraction of RNA from rectosigmoid biopsies results in >95% recovery of tissue RNA.
  • Samples are immediately homogenized from the frozen state (using Powergen 125 tissue homogenizer), Trizol-extracted with separation of RNA and DNA containing phases. RNA is further extracted using an Rneasy column. Quality control studies have confirmed minimal RNA degradation (agarose gel electrophoresis) and no DNA contamination (PCR of RNA template).
  • the number of HIV RNA copies is quantitated using an adaptation ofthe rTTH RNA PCR kit (Perkin-Elmer) with HIV LTR specific primers 667/AA55 designed to capture unspliced/multiply spliced HIV RNA.
  • a linear standard curve is generated using a 127 bp sequence recognized by the 667/AA55 primer pairs.
  • DNA is isolated by ethanol precipitation with at least 2 washes in 0.1 M sodium citrate/ 10% ethanol buffer.
  • the same primer pairs are used for PCR amplification (667/AA55). Linear standard curves have been generated using ⁇ -globin primers.
  • HIV-RNA is reproducibly detected in rectosigmoid biopsies from subjects with undetectable plasma viral load.
  • Efforts were made to demonstrate the replicability of results from one biopsy by comparison with others concurrently obtained at the same circumferential level (30 cm) from the same patient.
  • Single biopsies (10 mg each) from subjects with undetectable plasma viral loads were frozen, RNA extracted and amplified using LTR-specific primers 667/AA55, as described above. Each biopsy yielded an average of 25 ⁇ g RNA of which usually 1/100 was used for quantitation.
  • Results in Figure 7 demonstrate the reproducibility of quantitated RNA viral burden using rectosigmoid biopsies in a sensitive assay. The data demonstrates the tissue HIV RNA viral load from 2 biopsies obtained during the baseline sigmoidoscopies for subjects with undetectable plasma viral loads.
  • HIV-DNA is reproducibly detected in rectosigmoid biopsies from subjects with undetectable plasma viral load.
  • HIV DNA was amplified using quantitative PCR with specific 667/AA55 primers for the LTR region and b-globin specific primers used for internal linear standard. Triplicates were assayed for the b-globin specific primers; duplicates ofthe HIV proviral DNA quantitation are shown in Figure 8. Although lower limit of detection is 3 copies, 10 copies were used as our lower cut-off point. The figure shows both the actual copy number quantitated and the calculated number of copies based on a b-globin- dependent cell count. These results show our technique can detect copies of proviral DNA as low as 10 in subjects with undetectable plasma viral load.
  • CCR5 co-receptor expression on mucosal CD4 T cells Isolation of mucosal mononuclear cells does not alter phenotypic expression of CD4, CD8, CCR5, and CXCR4.
  • Samples were isolated from healthy, seronegative controls' blood (peripheral blood mononuclear cells: PBMC) and intestinal mucosa (mucosal mononuclear cells: MMC) to establish baseline CCR5 and CXCR4 expression in both compartments.
  • Figure 9 demonstrates that our isolation procedure neither strips relevant receptors (CD4, CD8, CCR5, CXCR4) nor alters their surface expression.
  • Mucosal expression of CCR5 on CD4 T cells is greatly increased compared with PBMC.
  • Isolated mucosal mononuclear cells and peripheral blood mononuclear cells were obtained from healthy, seronegative control subjects and evaluated to determine the relative percentages of CD4 T lymphocytes in each compartment expressing CCR5 receptors.
  • the 2D7 CCR5 antibody is conjugated in a 1 :1 ratio with phycoerythrin; the flow cytometry instruments used are calibrated to detect 44 phycoerythrin molecules per RFI channel (based on a standardized CD4 expression and number of antibodies bound per cell). Consequently, the number of anti-CCR5 antibodies bound per cell can be translated to number of receptors per cell, assuming monovalent binding of antibody to receptor.
  • CCR5 expression on mucosal CD4 T cells is nearly exclusively on the memory subset.
  • Blood and mucosal samples from the same seronegative, healthy controls were counter stained with CD45RO antibody as an indicator ofthe memory subset to determine the relative distribution of CCR5 staining.
  • CCR5 expression on mucosal CD4 T cells remains increased compared to PBMC in HIV-infected and inflammatory samples.
  • the CD4:CD8 ratio of CCR5 -expressing T cells decreases in HIV and IBD in both blood and gut.
  • CCR5 receptor on CD4 T cells are also expressed on CD8+ T cells.
  • Figure 13 demonstrates the dramatic downward shifts in the CD4:CD8 ratios of CCR5 expressing cells, decreasing in blood and gut samples by roughly 50% in IBD and 70- 90% in HIV. This may represent a protective down regulation of CCR5 to inhibit HIV spread.
  • the primary stimulus for decreased surface expression likely relates to the inflammatory milieu.
  • Quantitation of ⁇ -chemokine tissue concentrations using QIA Quantitation of ⁇ -chemokine tissue concentrations using QIA.
  • Endoscopic biopsies that were quickly oriented on foil and snap frozen were sent to Sweden on dry ice for cryosectioning (7 ⁇ m) and quantitative immunohistochemical staining with antibodies to identify RANTES, MIP- la and MIP- lb.
  • Quantitative image analysis (QIA) was also performed using CD4, CD8, and CCR5 antibodies. Samples from a healthy, seronegative control and an HIV-infected individual with a detectable plasma viral load were studied.
  • Results are expressed as the percentage of total tissue area measure that was positively stained with the peroxidase-labeled antibody.
  • the findings in the healthy control for RANTES, MlP-la and MlP-lb were 2.04%, 1.39% and 1.65% respectively.
  • the respective value increased to 15.1%, 6.2% and 12.1%.
  • These chemokines function to recruit additional inflammatory cells into the already inflammed mucosa. This dramatic increase that HIV infection is associated with mucosal inflammation is provided in Table 1.
  • MMC Mucosal mononuclear cells
  • the majority of cells are CD8+, but the majority of lamina propria lymphocytes are CD4+.
  • biopsies of colon mucosa were analyzed for the presence of CD8+ cells.
  • the results in Figure 15 indicate that the degree of CD8+ staining (brown cells, darkened in a black/white image) in a biopsy from an HIV- infected patient is greater than in an HIV negative biopsy (compare panels A and B).
  • the majority ofthe cells in the lamina intestinal are CD8+ in an HTV- infected patient rather than CD4+.
  • the numbers of CD4+ cells are severely depleted in colon mucosum of an HIV-infected patient.
  • the b-chemokines discussed above previously shown to be elevated by HIV, recruit cells that bear the CCR5 receptor to the mucosa ( Figure 16).
  • the CCR5 receptor is the primary co-receptor that HIV uses to enter these cells.
  • HIV is ensuring that it will be able to propogate further by inducing inflammation in this environment, at least in part via production of pro-inflammatory cytokines, thereby recruiting more target cells for infection.
  • HIN is an inflammatory disease ofthe mucosa and, therefore, is amenable to treatment with anti-inflammatory agents.
  • the data show concentrations of pro-inflammatory cytokines and chemokines (mR ⁇ A), RANTES, IFN ⁇ , and TNF (RNA), present in mucosal biopsies of normal healthy control patients, HIV patients with low amounts of virus in their mucosa and patients with higher amounts of HIV in mucosa.
  • IFNy interferon-gamma
  • HIV patients with higher mucosal viral load have much higher levels of RANTES, IFN ⁇ , and TNF than HIV patients with low mucosal viral load.
  • HIV load increases activation of CD4+ cells and production of viral progeny.
  • FIG 17D shows that as mucosal viral load increases, there is an increase in the percentage of CD4+ cells in the mucosa that are activated (as indicated by increased expression of HLA-DR).
  • the mucosa cells once infected will produce more virus. This is indicated by increased expression of green fiouresence protein by a T-tropic reporter virus (Nlegfp) when replicated in a cell.
  • Nlegfp T-tropic reporter virus
  • the results indicate that the percentage of mucosal cells producing virus is more than 300 times greater than in blood cells.
  • Nlegfp T-tropic reporter virus
  • the anti-inflammatory agent 5-ASA inhibits viral replication in cells.
  • 5-ASA 5-ASA-specific antigenesized progenesized progenesized progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progeneous progene, 35, 3,000 ⁇ M.
  • 5-ASA dose ranges used are believed to represent the physiologic range seen in the gastrointestinal mucosa of patients treated with 4.8g of oral 5-ASA per day.
  • Replication incompetent HIV pNLluc ⁇ Bgl pseudotyped with either M-tropic JRFL or T-tropic LAI envelope was used for the infection. Luciferase expression was used to assess the degree of HIV replication after 4 days in culture.
  • FIG. 19 shows that the use of Asacol (mesalamine), a topical anti- inflammatory agent, inhibits HIV replication.
  • Asacol mesalamine
  • Asacol at the two lowest doses (30 and 300 micrograms) is able to inhibit HIV replication by approximately 20%.
  • the amount of suppression is higher, but this is due, in part, to cellular toxicity ofthe drug at this dose (approximately 30% cell death vs. 13-15% cell death for 30 and 300 microgram doses; AZT produces approximately 16% cell death at a dose of 5 ⁇ g).
EP00932379A 1999-05-14 2000-05-12 Anti-inflammatory therapy for inflammatory mediated infection Withdrawn EP1202620A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US13409199P 1999-05-14 1999-05-14
US134091P 1999-05-14
PCT/US2000/013142 WO2000069255A1 (en) 1999-05-14 2000-05-12 Anti-inflammatory therapy for inflammatory mediated infection

Publications (1)

Publication Number Publication Date
EP1202620A1 true EP1202620A1 (en) 2002-05-08

Family

ID=22461735

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00932379A Withdrawn EP1202620A1 (en) 1999-05-14 2000-05-12 Anti-inflammatory therapy for inflammatory mediated infection

Country Status (13)

Country Link
EP (1) EP1202620A1 (ko)
JP (1) JP2002544210A (ko)
KR (1) KR20020024587A (ko)
CN (1) CN1353573A (ko)
AU (1) AU5010800A (ko)
BR (1) BR0010546A (ko)
CA (1) CA2368593A1 (ko)
HK (1) HK1047216A1 (ko)
IL (1) IL146130A0 (ko)
MX (1) MXPA01011606A (ko)
PL (1) PL351710A1 (ko)
TR (1) TR200103271T2 (ko)
WO (1) WO2000069255A1 (ko)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUP0302068A3 (en) * 2000-07-20 2005-05-30 Lauras As Use of cox-2 inhibitors for treating or preventing immunodeficiency and pharmaceutical compositions containing them
KR20040072720A (ko) * 2002-01-10 2004-08-18 파마시아 앤드 업존 캄파니 항바이러스제와 함께 사용되는 유두종 바이러스 감염의치료를 위한 cox-2 억제제의 용도
AR039540A1 (es) * 2002-05-13 2005-02-23 Tibotec Pharm Ltd Compuestos microbicidas con contenido de pirimidina o triazina
US6790867B2 (en) * 2002-05-20 2004-09-14 Schering-Plough Animal Health Corporation Compositions and method for treating infection in cattle and swine
CN103143022A (zh) * 2002-07-30 2013-06-12 奥默罗斯公司 眼科冲洗液及方法
ITMI20031311A1 (it) * 2003-06-27 2004-12-28 Indena Spa Formulazioni per il trattamento di disturbi artrosici.
DE602004031356D1 (de) * 2003-11-21 2011-03-24 Zalicus Inc Verfahren und reagenzien zur behandlung von entzündlichen erkrankungen
CN102002476B (zh) * 2010-08-20 2013-05-08 上海市公共卫生临床中心 一种体外培养人体黏膜活组织模型及其建立方法和应用
CN102557976B (zh) * 2010-12-15 2015-06-10 辽宁盛京制药有限公司 溴芬酸有机盐及其制备方法、其组合物及用途
EP2481751A1 (en) * 2011-01-26 2012-08-01 PharmaSurgics in Sweden AB Human lactoferrin derived peptides
AU2013201465B2 (en) 2012-10-24 2016-03-03 Rayner Surgical (Ireland) Limited Stable preservative-free mydriatic and anti-inflammatory solutions for injection
CN104013959A (zh) * 2014-05-21 2014-09-03 郭和友 逆转录抑制剂和免疫抗体细胞三联合疗法
CN105456273A (zh) * 2014-09-02 2016-04-06 杭州雷索药业有限公司 奈韦拉平在制备抗炎药物中的应用
TWI705812B (zh) 2014-12-01 2020-10-01 奥默羅斯公司 用於抑制術後眼睛炎性病況的抗炎和散瞳前房溶液
CN106176739A (zh) * 2016-08-18 2016-12-07 滨州医学院 利托那韦增强糖皮质激素抗炎作用的医药新用途
CN106309445A (zh) * 2016-08-18 2017-01-11 滨州医学院 奈非那韦增强糖皮质激素抗炎作用的医药新用途
CN106176751A (zh) * 2016-08-18 2016-12-07 滨州医学院 茚地那韦增强糖皮质激素抗炎作用的医药新用途

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5605885A (en) * 1988-05-05 1997-02-25 Entremed, Inc. Method for stimulating the immune system
EP0407701A3 (en) * 1989-05-15 1992-04-22 Fujisawa Pharmaceutical Co., Ltd. Antiretroviral pyrroline and pyrrolidine sulfonic acid derivatives
US5891924A (en) * 1996-09-26 1999-04-06 Research Development Foundation Curcumin (diferuloylmethane) inhibition of NFκB activation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0069255A1 *

Also Published As

Publication number Publication date
AU5010800A (en) 2000-12-05
JP2002544210A (ja) 2002-12-24
KR20020024587A (ko) 2002-03-30
WO2000069255A1 (en) 2000-11-23
TR200103271T2 (tr) 2002-06-21
CN1353573A (zh) 2002-06-12
MXPA01011606A (es) 2003-09-10
IL146130A0 (en) 2002-07-25
HK1047216A1 (zh) 2003-02-14
CA2368593A1 (en) 2000-11-23
BR0010546A (pt) 2002-03-05
PL351710A1 (en) 2003-06-02
WO2000069255A8 (en) 2001-03-22

Similar Documents

Publication Publication Date Title
EP1202620A1 (en) Anti-inflammatory therapy for inflammatory mediated infection
Klatt Pathology of AIDS
Patterson et al. Repertoire of chemokine receptor expression in the female genital tract: implications for human immunodeficiency virus transmission
Utay et al. Role of immune activation in progression to AIDS
Nilsson et al. Early immune activation in gut-associated and peripheral lymphoid tissue during acute HIV infection
Schmidt et al. Rapid increase of mucosal CD4 T cells followed by clearance of intestinal cryptosporidiosis in an AIDS patient receiving highly active antiretroviral therapy
Kotler et al. Effect of combination antiretroviral therapy upon rectal mucosal HIV RNA burden and mononuclear cell apoptosis
US20030138399A1 (en) Anti-inflammatory therapy for inflammatory mediated infection
Shacklett et al. Abundant expression of granzyme A, but not perforin, in granules of CD8+ T cells in GALT: implications for immune control of HIV-1 infection
Shen et al. Mucosal Correlates of Protection in HIV‐1‐Exposed Sero‐negative Persons
NIU et al. Summary of the National Institutes of Health Workshop on Primary Human Immunodeficiency Virus Type 1 Infection1, 2
Johnson et al. Anti-retroviral therapy reverses HIV-associated abnormalities in lymphocyte apoptosis
Van Deventer Anti-TNF antibody treatment of Crohn's disease
Stetson et al. Eosinophilic dermatoses
Desdouits et al. HTLV-1-associated inflammatory myopathies: low proviral load and moderate inflammation in 13 patients from West Indies and West Africa
Purohit et al. Potential impact of drugs of abuse on mother-to-child transmission (MTCT) of HIV in the era of highly active antiretroviral therapy (HAART)
Rogers et al. Fetal feline immunodeficiency virus is prevalent and occult
Schito et al. A human immunodeficiency virus–transgenic mouse model for assessing interventions that block microbial-induced proviral expression
Nair et al. Association of drug abuse with inhibition of HIV-1 immune responses: studies with long-term of HIV-1 non-progressors
EP1194141B1 (en) Antiviral therapy
US20010053517A1 (en) Determining nucleic acid sequences in a biological sample
US6387959B1 (en) Antiviral therapy
Wilkinson et al. Immune interventions: The changing face of HIV and AIDS
Edzeamey et al. HIV Infection and Therapeutic Interventions: Review on HIV Infection Biology, Highly Active Antiretroviral Therapy (HAARTs), Hepatotoxicity, Nephrotoxicity and Dyslipidemia
Omollo The Effect of Hormonal Contraception on Mucosal and Peripheral Immunity: Implications for Hiv Susceptibility Among Women in Nairobi, Kenya

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20011203

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20070328