WO2003032895A2 - Methods and pharmaceutical compositions for stimulating the immune system and/or treating cancer - Google Patents

Methods and pharmaceutical compositions for stimulating the immune system and/or treating cancer Download PDF

Info

Publication number
WO2003032895A2
WO2003032895A2 PCT/IL2002/000803 IL0200803W WO03032895A2 WO 2003032895 A2 WO2003032895 A2 WO 2003032895A2 IL 0200803 W IL0200803 W IL 0200803W WO 03032895 A2 WO03032895 A2 WO 03032895A2
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical composition
group
metallocene
amount
ferrocene
Prior art date
Application number
PCT/IL2002/000803
Other languages
French (fr)
Other versions
WO2003032895A3 (en
Inventor
Abraham Novogrodsky
Original Assignee
Notox Ltd.
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 Notox Ltd. filed Critical Notox Ltd.
Publication of WO2003032895A2 publication Critical patent/WO2003032895A2/en
Publication of WO2003032895A3 publication Critical patent/WO2003032895A3/en

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/295Iron group metal compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/315Zinc compounds

Definitions

  • the present invention relates to a methods and pharmaceutical compositions useful in inducing immune stimulation and/or treating cancer. More particularly, the present invention relates to methods and pharmaceutical compositions for inducing immune stimulation and/or for treating cancer, which comprise H 2 0 2 and/or an H 2 0 2 generating agent, such as metallocenes (e.g., ferrocene) and their derivatives.
  • H 2 0 2 and/or an H 2 0 2 generating agent such as metallocenes (e.g., ferrocene) and their derivatives.
  • Biological response modifiers such as IL-2, IFN- ⁇ IFN- ⁇ and TNF- ⁇ , are all possible candidates. Clinical use of some of these compounds resulted in limited response and has been associated with marked toxicity (1-5).
  • Agonistic immune stimulatory antibodies such as anti-CD3, were also evaluated for their anti-tumor activity in humans, with limited success (6,7).
  • BCG Bacillus Calmette-Guerin
  • the protooncogene p21 ras has been identified as a key molecular switch involved in regulating T cell activation triggered by different mitogens (15-19).
  • Metallocenes such as ferrocene are stable iron-containing compounds which generate free radicals. Metallocenes were so far never suggested as immune stimulants and/or an anti- cancer agents.
  • metallocenes such as, for example, ferrocene
  • ferrocene could stimulate the immune system and/or provide for anti-cancer therapy. While reducing the present invention to practice this hypothesis was tested in various systems.
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy comprising determining an immune stimulation/cancer therapy dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to immune stimulation/cancer therapy within a predetermined range of concentrations.
  • the immune stimulation is determined in vivo.
  • immune stimulation is determined in vitro.
  • the immune stimulation is determined with respect to macrophages.
  • the immune stimulation is determined with respect to lymphocytes. According to still further features in the described preferred embodiments, the immune stimulation is determined by a cell proliferation assay.
  • the cell proliferation assay includes a thymidine incorporation assay.
  • the immune stimulation is determined by a cellular response assay.
  • the cell response assay is selected from the group consisting of an NO production assay, a TNF- ⁇ production assay and an oxygen burst assay.
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy comprising determining a p21 ras signal transduction pathway component activity dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to p21 ras signal transduction pathway component activity within a predetermined range of concentrations.
  • the p21 ras signal transduction pathway component is selected from the group consisting of p21 ras and NF- ⁇ B.
  • method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy comprising determining p21 ras oxidation dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to p21 ras oxidation within a predetermined range of concentrations;
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy the method comprising determining a GTPase activity dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to the GTPase activity within a predetermined range of concentrations; and
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy comprising determining an
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy comprising determining H 2 0 2 generation catalyzed by the metallocene derivative, and determining whether the rate is within a predetermined range.
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy comprising monitoring H 2 0 2 generation catalyzed by the metallocene derivative, and determining whether the generation is above a predetermined value.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer comprising administering to a subject in need thereof at least one agent in an amount so as to induce H 0 2 generation at a concentration effective in inducing immune stimulation and or providing cancer therapy.
  • each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount so as to induce H 2 0 2 generation at a concentration effective in inducing immune stimulation following administration thereof.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial comprising administering to a subject in need thereof at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight.
  • a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer comprising administering to a subject in need thereof at least one agent in an amount effective in activating a p21 ras signal transduction pathway component.
  • a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in activating a p21 ras signal transduction pathway component following administration thereof.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer comprising administering to a subject in need thereof at least one agent in an amount effective in inducing p21 ras oxidation.
  • a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing p21 ras oxidation following administration thereof.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer comprising administering to a subject in need thereof at least one agent in an amount effective in inducing GTPase activity.
  • a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing GTPase activity following administration thereof.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer comprising administering to a subject in need thereof at least one agent in an amount effective in inducing NF- ⁇ B activity.
  • a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing NF- ⁇ B activity following administration thereof.
  • the at least one agent comprises at least one ferrocene and/or metallocene derivative.
  • the amount is between 0.025 - 10 mg/kg body weight.
  • the amount is between 0.1 - 5 mg/kg body weight. According to still further features in the described preferred embodiments the amount is between 0.2 - 3 mg/kg body weight.
  • the at least one agent is administered orally and hence the pharmaceutical composition is formulated for oral administration.
  • the at least one agent is administered by injection and hence pharmaceutical composition is formulated for administration by injection.
  • the cancer is selected from the group consisting of all malignant tumors, including, but not limited to, carcinoma, sarcoma, lymphoma or hematological malignancies, hence, the pharmaceutical composition is packaged and identified for the treatment of all malignant tumors, including, carcinomas, sarcomas, lymphomas or hematological malignancies.
  • the disease, syndrome or condition is selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency, hence, the pharmaceutical composition is packaged and identified for the treatment of a disease, syndrome or condition selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency.
  • M is a metal selected from the group consisting of Fe (iron), Cr
  • Ri-Rg are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
  • the method further comprising co-administering at least one additional agent having immune stimulation activity.
  • the pharmaceutical composition further comprises at least one additional agent having immune stimulation activity, such as, but not limited to, a cytokine (e.g., IL-2, TNF, interferon), a chemokine, an immune-stimulatory antibody (e.g., anti-CD3, anti-CTLA-4) or an anti-tumor vaccine.
  • the method further comprising co-administering at least one additional agent having anti-cancer activity.
  • the pharmaceutical composition further comprises at least one additional agent having anti-cancer activity, such as, but not limited to, chemotherapeutic agents, anti- metabolites, signal transduction inhibitors (e.g., protein tyrosine kinase inhibitors), anti-angiogenesis compounds, antibodies (e.g., anti-receptors, anti-tumor determinants), hormones and hormone antagonists, retinoids, steroids, pro-apoptotic agents, differentiating agents or radiation.
  • chemotherapeutic agents e.g., anti-metabolites, signal transduction inhibitors (e.g., protein tyrosine kinase inhibitors), anti-angiogenesis compounds, antibodies (e.g., anti-receptors, anti-tumor determinants), hormones and hormone antagonists, retinoids, steroids, pro-apoptotic agents, differentiating agents or radiation.
  • a method of inducing immune stimulation and/or treating cancer in a body cavity comprising administering into the body cavity H 2 0 in an amount effective in immune stimulation.
  • the amount is of 10-2000 ml of 50-2500 ⁇ M H 2 0 2 administerred in a single or multiple administrations).
  • the body cavity can be, for example, the peritoneal cavity or the pleural cavity.
  • a method of inducing immune stimulation and/or treating cancer comprising exposing immune cells ex vivo to at least one ferrocene and/or metallocene derivative, and administering the cells to a subject in need thereof.
  • a method of inducing immune stimulation and/or treating cancer comprising exposing immune cells ex vivo to H 2 0 2 in a concentration effective in inducing immune stimulation and administering the cells to a subject in need of thereof.
  • the present invention successfully addresses the shortcomings of the presently known configurations by providing new methodologies with which to induce immune stimulation and/or treat cancer.
  • FIGs. 1A-B demonstrate stimulation by ferrocene and IL-2 of H-thymidine incorporation in mouse splenocytes, in vitro.
  • 1A Mouse splenocytes were incubated in the present of ferrocene, at different concentrations, for 3 days. 3 H-Thymidine incorporation during the last 20 hours of incubation was determined. Results are expressed as means CPM+SD of triplicate cultures.
  • IB Same as 1A, but the cells were cultured in the presence of IL-2 (50 U/ml).
  • FIG. 2 demonstrates stimulation by ferrocene of 3 H-thymidine incorporation in mouse splenocytes, in vivo.
  • Mice were injected intraperitoneally with ferrocene at different doses. Three days later, splenocytes were isolated and incorporation of H-thymidine during 30 hours, was determined. Two mice were included in each group. Results are expressed as means CPM ⁇ SD of triplicate cultures from each mouse.
  • FIG. 3 demonstrates activation by ferrocene of mouse peritoneal macrophages, in vitro.
  • Peritoneal macrophage monolayers were prepared (as described in the Examples section that follows) and co-incubated with splenic lymphocytes in the presence of ferrocene at different concentrations. The ratio of lymphocytes to macrophages was 10:1. After incubation for 3 days, the non- adherent lymphocytes were removed by washing with PBS. Production of
  • H 2 0 2 as a measurement of oxygen burst was determined in the presence of phorbol myristate acetate (PMA) (100 ng/ml), using the aminotriazole-catalase inhibition test (see Examples section that follows). Results are expressed as means ⁇ SD of triplicate cultures.
  • PMA phorbol myristate acetate
  • FIG. 4 demonstrates activation by ferrocene of mouse peritoneal macrophages, in vivo.
  • Mice were injected intraperitoneally with ferrocene at different doses (per mouse). After 3 days, peritoneal macrophages were obtained and oxygen burst was determined in the presence and absence of PMA (100 ng/ml).
  • PMA 100 ng/ml
  • mice were injected with 1 ml of NaI0 (5 mM). NaI0 4 was previously shown to activate peritoneal macrophages, in vivo (39). Four mice were included in each experimental group and the results are expressed as means +SD.
  • 5A-C demonstrate activation by ferrocene of mouse peritoneal macrophages in vivo: TNF- ⁇ production (5 A), oxygen burst (5B), nitric oxide (5C) production.
  • Mice were injected intraperitoneally with ferrocene (1 ⁇ g/mouse) or with NaI0 4 (0.15 M, 1 ml), as a positive control.
  • ferrocene (1 ⁇ g/mouse
  • NaI0 4 (0.15 M, 1 ml
  • FIG. 6 demonstrates anti-tumor effect of ferrocene administered intraperitoneally by a single injection (Ti) at different doses in B-16 melanoma bearing mice. Ferrocene was injected intraperitoneally one day post B-16 melanoma inoculation (Ti) via the tail vein. Mice were sacrificed 25 days post tumor inoculation and the pair of lungs from each mouse was weighed. 4-6 mice were included in each experimental group. Results are expressed as the means of the weights of the pairs of lungs of each mouse +SD.
  • FIG. 7 demonstrates anti-tumor effect of ferrocene at different doses, administered intraperitoneally by multiple injections.
  • Mice were inoculated with B-16 melanoma (To). Ferrocene was administered intraperitoneally 3 times (Ti, T 8 and T ⁇ 5 ) each, at a dose specified in the Figure. Mice were sacrificed 25 days post tumor inoculation and the pair of lungs from each mouse was weighed. 4-6 mice were included in each group. Results are expressed as the means of the weights of the pair of lungs of each mouse ⁇ SD.
  • FIG. 8 demonstrates the anti-tumor effect of ferrocene. Photographs were taken of pairs of lungs from the experiments depicted in Figures 6 and 7.
  • FIGs. 9A-B demonstrate the effect of ferrocene, administered in the drinking water (1 ⁇ g/ml) on established lung metastases in B-16 melanoma bearing mice. Mice, inoculated with B-16 melanoma were supplied with drinking water (ad libidum) containing 1 ⁇ g/ml ferrocene. A mouse drinks approximately 5 ml water/day, so the overall dose of ferrocene was approximately 0.25 mg/kg body weight/day.
  • mice One group of mice was supplied with ferrocene in the drinking water for 1 week followed by replacing it with plain water, the second and third groups were supplied with ferrocene in the drinking water for 2 and 3 weeks, respectively. Mice were sacrificed 25 days post tumor inoculation and tumor load was assessed by weighing the lungs. 4-6 mice were included in each group. Results are expressed as the means of the weight of the pair of lungs of each mouse ⁇ SD (9A). Photographs of lungs of the mice that were sacrificed 25 days post tumor inoculation (9B).
  • FIGs. 10A-B demonstrate anti-tumor effect of ferrocene administered in drinking water at different concentrations.
  • Table 1 Photographs of lungs taken 25 days post tumor inoculation. Mice were supplied with drinking water containing ferrocene for 1 week (10A) or 2 weeks (10B).
  • FIG. 11 demonstrates the effect of ferrocene administered in drinking water (1 ⁇ g/ml) for one week on survival of B-16 melanoma bearing mice.
  • FIG. 12 demonstrates anti -tumor effect of ferrocene in mice bearing Lewis lung carcinoma. Ferrocene was administered intraperitoneally 3 times (Ti, T 8 and T 15 ), each at a dose of 0.025, 0.05 and 0.2 mg/kg body weight. Mice were sacrificed 28 days post tumor inoculation and tumor load was assessed by weighing the lungs. Four mice were included in each group and the results are expressed as the means of lungs' weight ⁇ SD.
  • FIG. 13 demonstrates anti-tumor effect of combined treatment with ferrocene and IL-2 in B16 melanoma bearing mice. Ferrocene was administered intraperitoneally at a single dose on day T[. IL-2 was administered intraperitoneally 3 times on days T b T 2 and T 2 each, at a dose of 25 x 10 4 U/kg body weight). 5-6 mice were included in each group. Mice were sacrificed 25 days post tumor inoculation and tumor load was assessed by weighing the lungs. Results are expressed as the means of lungs' weight ⁇ SD.
  • FIGs. 14A-B demonstrate an adoptive transfer experiments: Anti-tumor effect of immunocytes from ferrocene-treated mice.
  • 14A - Donor cells were obtained from B-16 melanoma bearing mice which were treated on day T 0 with a single injection of ferrocene (0.2 mg/kg body weight). Cells were collected from donor mice 25 days post tumor inoculation. Splenocytes (10 cells/mouse)
  • peritoneal mononuclear cells (5 x 10 cells/mouse) were administered intravenously to B16 melanoma inoculated recipient mice on day T 4 .
  • Recipient mice were sacrificed 25 day post tumor inoculation and tumor load was assessed by weighing the lungs.
  • recipient mice were included in each experiment and results are expressed as means ⁇ SD.
  • 14B Photographs of lungs from recipient mice treated with donor ("macrophages”) peritoneal mononuclear cells .
  • FIG. 15 demonstrates an adoptive transfer experiments: Anti-tumor effect of immunocytes from ferrocene-treated mice.
  • Donor cells were obtained from B-16 melanoma bearing mice which were treated on day T 0 with a single injection of ferrocene (0.2 mg/kg body weight). Cells were collected from donor mice 25 days post tumor inoculation. Splenocytes (10 8 cells/mouse) and peritoneal mononuclear cells (“macrophages”) (5 x 10 6 cells/mouse) were administered intravenously to B16 melanoma inoculated recipient mice on day T . Recipient mice were sacrificed 25 day post tumor inoculation and tumor load was assessed by weighing the lungs. 2-4 recipient mice were included in each experiment. Photographs of lungs from recipient mice treated with donor (“macrophages”) peritoneal mononuclear cells.
  • FIG. 16 demonstrate production of H 2 0 2 in ferrocene solution. H 2 0 2 production was measured by the aminotriazole-catalase inhibition test (see Examples section that follows).
  • FIG. 17 demonstrates the effect of N-acetyl cysteine (NAC) on the anti-tumor effect of ferrocene in B-16 melanoma bearing mice. Ferrocene was injected intraperitoneally at a single dose of 0.25 mg/kg body weight (optimal) or 2.5mg/kg body weight (supraoptimal), on day T N-acetylcysteine was injected at a dose of 500 mg/kg body weight, twice a day, for 3 days (Ti, T 2 , T 3 ). Mice were sacrificed 25 days post tumor inoculation and the tumor load was assessed by weighing the lungs. Five mice were included in each group and the results are expressed as means of lungs' weight ⁇ SD.
  • NAC N-acetyl cysteine
  • FIG. 18 demonstrates that ferrocene induces phosphorylation of ERK1/2 in wild type Jurkat cells and not in C118S Jurkat cells.
  • Jurkat cells wild type and C118S
  • RPMI 1640 containing 0.5 % FCS.
  • Ferrocene was added at a final concentration of 0.05, 0.2, 1 and 5 ⁇ M.
  • PHA was added at a final concentration of 20 ⁇ g/ml.
  • cells were centrifuged and suspended in lysis buffer.
  • Western blot analysis for the detection of activated (phosphorylated) ERK1/2 was done as described in the Examples section that follows.
  • FIG. 19 demonstrates the effects of ferrocene on NF- ⁇ B activity.
  • Cells were treated with the indicated concentrations of ferrocene for 4 hours prior to isolation of nuclei and assay for NF- ⁇ B binding activity as described in the Examples section that follows. Arrows indicate the protein/DNA complex.
  • FIG. 20 demonstrates the anti-tumor effect of H 2 0 2 administered intraperitoneally on established lung metastases in B-16 melanoma-bearing mice.
  • H 2 0 2 was administered intraperitoneally, twice a day, each, at a dose of 5 ⁇ moles/kg body weight, on day T 1? T 2 , and T 3 (1 cycle), or on days T T 3 , T 8 -T ⁇ o and T 15 -T] 7 (3 cycles).
  • Mice were sacrificed 25 days post tumor inoculation and tumor load was assessed by lungs' weight. Four mice were included in each group. Results are expressed as means of lungs' weight ⁇ SD.
  • FIGs. 21A-B demonstrate the anti-tumor effect of H 2 0 2 intraperitoneally.
  • the experimental design was similar to that depicted in Figure 20, except that 6 mice were included in each group. Scoring of tumor load was done by counting and measuring the size of lungs' metastases as follows: Lesion Size Score
  • FIG. 22 shows photographs of lungs from mice inoculated with B-16 melanoma. Experimental data is outlined in Figures 21A-B.
  • the present invention is of methods and pharmaceutical compositions which can be used for inducing immune stimulation and/or treating cancer.
  • the present invention is of methods and pharmaceutical compositions for inducing immune stimulation and/or for treating cancer, which comprise H 2 0 2 and/or an H 2 0 2 generating agent, such as metallocenes (e.g., ferrocene) and their derivatives.
  • H 2 0 2 and/or an H 2 0 2 generating agent such as metallocenes (e.g., ferrocene) and their derivatives.
  • Oxidizing agents have been shown to regulate protein function by modification of cysteine residues.
  • Their protein targets include Ras, calcium dependent potassium channels, N-methyl-D-aspartate and caspases (28).
  • Ras is a key element of various signaling pathways and is implicated in the regulation of proliferation and differentiation by tyrosine kinase and G protein-coupled receptors (29).
  • p21 ras is a common signaling target of reactive free radicals and cellular redox stress (21) and the immune-stimulatory properties of agents that generate free radicals, including iron-containing compounds such as hemin was described (9-13). Hemin, by itself, does not exhibit anti-tumor activity but shows this effect in combination with IL-2 (14).
  • ferrocene While conceiving the present invention, it was hypothesized that metallocenes, stable metal-containing compounds, and metallocene derivatives could stimulate the immune system and/or provide for anti-cancer therapy. While reducing the present invention to practice this hypothesis was tested in various systems. Hence, it is demonstrated herein that ferrocene, induces in vitro and in vivo activation of mouse splenocytes and peritoneal macrophages. Ferrocene also has a marked anti-tumor effect in mice bearing B-16 melanoma and Lewis lung carcinoma.
  • the immune stimulatory and anti-tumor effects of ferrocene follow a bell shaped curve, namely ferrocene at high, supraoptimal concentrations, is far less effective.
  • the inhibiting effects of ferrocene at high concentrations may be due to oxidation of thiol moieties and other susceptible molecules that are not relevant to the stimulatory process.
  • Reactive oxygen species have been implicated in damaging biomolecular components including lipids, proteins and DNA.
  • Anti-tumor effects of agents known to stimulate the immune-system have been previously reported (1-5). They include IL-2, cytokines and agonistic antibodies such as anti CD3 (6,7) and anti BAT monoclonal antibody (34). Of interest are the findings described herein that H 2 0 2 administered intraperitoneally, at a low dose, has an anti-tumor effect. H 2 0 2 -induced activation of the MAPK and NFKB pathways has been previously reported (35-37). Hydrogen peroxide was also reported to activate T lymphocytes (38). In several independent assays treatment with ferrocene showed a bell-shaped (optimum) response curve.
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy comprises determining an immune stimulation/cancer therapy dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to immune stimulation/cancer therapy within a predetermined range of concentrations.
  • immune stimulation can be determined in vivo or in vitro (e.g., ex vivo).
  • the immune stimulation can be determined with respect to macrophages and/or lymphocytes, by a cell proliferation assay, such as a thymidine incorporation assay or by a cellular response assay, such as an NO production assay, a TNF- ⁇ production assay and/or an oxygen burst assay.
  • a cell proliferation assay such as a thymidine incorporation assay
  • a cellular response assay such as an NO production assay, a TNF- ⁇ production assay and/or an oxygen burst assay.
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy comprises determining a p21 ras signal transduction pathway component activity dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to p21 ras signal transduction pathway component activity within a predetermined range of concentrations.
  • the p21 ras signal transduction pathway component is p21 ras and/or NF- ⁇ B.
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy comprising determining p21 ras oxidation dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to p21 ras oxidation within a predetermined range of concentrations; (ii) a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising determining a GTPase activity dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to the GTPase activity within a predetermined range of concentrations; and (iii) a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising
  • ferrocene and metallocene derivatives are effective immunostimulant as they catalyze the generation of H 2 0 2 which reacts with susceptible molecules, or react directly with thiol moeties such as CI 18 in p21 ras .
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy comprises monitoring the rate of H 2 0 2 generation catalyzed by the metallocene derivative, and determining whether the rate is within a predetermined range.
  • Kinetic parameters determination assay is a multi data points assay and hence less amenable for highthroughput screening.
  • a single data point assay (a highthroughput assay) for H 2 0 2 generation catalyzed by metallocene derivatives can be developed by those of skills in the art so as to be used in determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy.
  • a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy is provided.
  • the method according to this aspect of the invention comprises monitoring H 2 0 2 generation catalyzed by the metallocene derivative, and determining whether the generation is above a predetermined value.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer comprises administering to a subject in need thereof at least one agent in an amount so as to induce H 2 0 2 generation at a concentration effective in inducing immune stimulation and or providing cancer therapy.
  • each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one agent in an amount so as to induce H 2 0 2 generation at a concentration effective in inducing immune stimulation following administration thereof.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial comprises administering to a subject in need thereof at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight.
  • each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer comprises administering to a subject in need thereof at least one agent in an amount effective in activating a p21 ras signal transduction pathway component.
  • each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one agent in an amount effective in activating a p21 ras signal transduction pathway component following administration thereof.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer comprises administering to a subject in need thereof at least one agent in an amount effective in inducing p21 ras oxidation.
  • each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one agent in an amount effective in inducing p21 ras oxidation following administration thereof.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer comprises administering to a subject in need thereof at least one agent in an amount effective in inducing GTPase activity.
  • each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one agent in an amount effective in inducing GTPase activity following administration thereof.
  • a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer comprises administering to a subject in need thereof at least one agent in an amount effective in inducing NF- ⁇ B activity.
  • each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one agent in an amount effective in inducing NF- ⁇ B activity following administration thereof.
  • the agent(s) used in the methods and compositions described above comprises at least one ferrocene and/or metallocene derivative and the amount thereof in a dose-unit is 0.025 - 10 mg/kg body weight, which translates to 0.25 - 1000 mg per dose-unit for individuals weighting 10-100 kg.
  • the amount of the ferrocene and/or metallocene derivative in a dose-unit is 0.1 - 5 mg/kg body weight, which translates to 1 - 500 mg per dose-unit for individuals weighting 10-100 kg.
  • the amount of the ferrocene and/or metallocene derivative in a dose-unit is 0.2 - 3 mg/kg body weight, which translates to 2 - 300 mg per dose-unit for individuals weighting 10-100 kg.
  • the agent(s) used according to the present invention for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or for treating cancer are administered orally or by injection.
  • the pharmaceutical compositions described herein are preferably formulated for oral administration or administration by injection.
  • Other modes of application are further described herein.
  • the present invention offers treatment for any and all cancers. These include, for example, carcinomas (e.g., colon, lung, breast, prostate, ovary), sarcomas (e.g., soft tissue, bone), lymphomas (e.g., Hodgkin lymphoma, non-Hodgkin lymphoma), hematological malignancies (e.g., myelogenous leukemia, lymphocytic leukenia).
  • carcinomas e.g., colon, lung, breast, prostate, ovary
  • sarcomas e.g., soft tissue, bone
  • lymphomas e.g., Hodgkin lymphoma, non-Ho
  • the pharmaceutical composition is preferably packaged and identified for the treatment of carcinomas (e.g., colon, lung, breast, prostate, ovary), sarcomas (e.g., soft tissue, bone), lymphomas (e.g., Hodgkin , non-Hodgkin), hematological malignancies (e.g., myelogenous leukemia, lymphocytic leukenia).
  • carcinomas e.g., colon, lung, breast, prostate, ovary
  • sarcomas e.g., soft tissue, bone
  • lymphomas e.g., Hodgkin , non-Hodgkin
  • hematological malignancies e.g., myelogenous leukemia, lymphocytic leukenia.
  • the present invention further offers treatment for acquired immune deficiency, such as AIDS, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency (e.g., SKID).
  • acquired immune deficiency such as AIDS, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency (e.g., SKID).
  • the pharmaceutical composition can be packaged and identified for the treatment of a disease, syndrome or condition, such as an acquired immune deficiency, a radiotherapy induced immune deficiency, a chemotherapy induced immune deficiency and a congenital immune deficiency.
  • the metallocene or metallocene derivatives used in the methods and pharmaceutical compositions of the present invention has a a general formula:
  • M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and R]-R 8 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryl and aryloxy.
  • alkyl refers to a saturated aliphatic hydrocarbon including straight chain and branched chain groups.
  • the alkyl group has 1 to 10 carbon atoms.
  • a numerical range; e.g., "1-10” is stated herein, it means that the group, in this case the alkyl group, may contain 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. More preferably, it is a medium size alkyl having 1 to 6 carbon atoms. Most preferably, it is a lower alkyl having 1 to 4 carbon atoms.
  • the alkyl group may be substituted or unsubstituted. When substituted, the substituent group can be, for example, cycloalkyl, aryl, hydroxy, alkoxy, aryloxy, cyano, halo, carbonyl, nitro, silyl, or amino.
  • a "cycloalkyl” group refers to an all-carbon monocyclic or fused ring (i.e., rings which share an adjacent pair of carbon atoms) group wherein one of more of the rings does not have a completely conjugated pi-electron system.
  • examples, without limitation, of cycloalkyl groups are cyclopropane, cyclobutane, cyclopentane, cyclopentene, cyclohexane, cyclohexadiene, cycloheptane, cycloheptatriene, and adamantane.
  • a cycloalkyl group may be substituted or unsubstituted. When substituted, the substituent group can be, for example, cycloalkyl, aryl, hydroxy, alkoxy, aryloxy, cyano, halo, carbonyl, nitro, silyl, or amino.
  • alkenyl refers to an alkyl group which consists of at least two carbon atoms and at least one carbon-carbon double bond.
  • alkynyl refers to an alkyl group which consists of at least two carbon atoms and at least one carbon-carbon triple bond.
  • alkoxy refers to both an -O-alkyl and an -O-cycloalkyl group, as defined herein.
  • aryl refers to an all-carbon monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups having a completely conjugated pi-electron system. Examples, without limitation, of aryl groups are phenyl, naphthalenyl and anthracenyl. The aryl group may be substituted or unsubstituted. When substituted, the substituent group can be, for example, halo, trihalomethyl, cycloalkyl, aryl, hydroxy, alkoxy, aryloxy, cyano, carbonyl, nitro, silyl, or amino.
  • An "aryloxy” group refers to an -O-aryl as defined herein.
  • halo refers to flourine, chlorine, bromine or iodine.
  • trihalomethyl refers to a -CX 3 group wherein X is a halo group as defined herein.
  • a hydroxy group refers to a -OH group.
  • An "Amino” group refers to an -NH 2 group.
  • a “nitro” group refers to an -N0 2 group.
  • a “silyl” group refers to a -Si(R') 3 , where R' is hydrogen, alkyl, cycloalkyl or aryl, as defined herein.
  • the method further comprises co-administering at least one additional agent having immune stimulation and/or anti-cancer activity.
  • the pharmaceutical composition further comprises at least one additional agent having immune stimulation and/or anti-cancer activity.
  • Agents having immune stimulation activity include, for example, cytokines (e.g., IL-2, TNF, interferon), chemokines, immune-stimulatory antibodies (e.g., anti-CD3, anti-CTLA-4) or an anti-tumor vaccine.
  • Agents having anti-cancer activity include, for example, chemotherapeutic agents, anti-metabolites, signal transduction inhibitors (e.g., protein tyrosine kinase inhibitors), anti-angiogenesis compounds, antibodies (e.g., anti-receptors, anti-rumor determinants), hormones and hormone antagonists, retinoids, steroids, pro-apoptotic agents, differentiating agents or radiation.
  • signal transduction inhibitors e.g., protein tyrosine kinase inhibitors
  • anti-angiogenesis compounds e.g., antibodies (e.g., anti-receptors, anti-rumor determinants), hormones and hormone antagonists, retinoids, steroids, pro-apoptotic agents, differentiating agents or radiation.
  • ferrocene activity with respect to immune stimulation and/or cancer therapy is mediated, at least in part, by H 2 0 2 generation.
  • a method of inducing immune stimulation and/or treating cancer in a body cavity comprises administering into the body cavity of a subject in need thereof H 2 0 2 in an amount effective in immune stimulation.
  • the amount is of 10-2000 ml of 50-2500 ⁇ M H 2 0 2 in a single or multiple administrations.
  • the body cavity can be, for example, the peritoneal cavity or the pleural cavity.
  • a method of inducing immune stimulation and/or treating cancer comprises exposing immune cells ex vivo to at least one ferrocene and/or metallocene derivative, and administering the cells to a subject in need thereof.
  • a method of inducing immune stimulation and/or treating cancer comprises exposing immune cells ex vivo to H 2 0 2 in a concentration effective in inducing immune stimulation and administering the cells to a subject in need of thereof.
  • a "pharmaceutical composition” refers to a preparation of one or more of metallocene or metallocene derivatives (including salts and acids thereof) described herein, with other chemical components such as physiologically suitable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • active ingredient includes compounds directly accountable for a biological effect.
  • physiologically acceptable carrier and “pharmaceutically acceptable carrier” which may be interchangeably used refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of a compound.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols. Techniques for formulation and administration of drugs may be found in "Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intravenous, inrtaperitoneal, intrapleural, intranasal, or intraocular injections.
  • the liposomes will be targeted to and taken up selectively by the tumor.
  • compositions of the present invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the compounds of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets-, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient.
  • Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • Pharmaceutical compositions, which can be used orally include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.
  • the compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • compositions described herein may be formulated for parenteral administration, e.g., by bolus injection or continuos infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative.
  • the compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile, pyrogen-free water
  • the preparation of the present invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
  • a preparation of the present invention may also be formulated for local administration, such as a depot preparation.
  • Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the preparation may be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives such as sparingly soluble salts.
  • suitable polymeric or hydrophobic materials for example, as an emulsion in an acceptable oil
  • ion exchange resins for example, as an emulsion in an acceptable oil
  • sparingly soluble derivatives such as sparingly soluble salts.
  • Formulations for topical administration may include, but are not limited to, lotions, suspensions, ointments gels, creams, drops, liquids, sprays emulsions and powders.
  • the pharmaceutical composition is designed for a slow release of the active ingredients.
  • the composition includes particles including a slow release carrier (typically, a polymeric carrier), such as, for example, polylactic acid, and the active ingredients.
  • a slow release carrier typically, a polymeric carrier
  • Slow release biodegradable carriers are well known in the art. These are materials that may form particles that may capture therein an active compound(s) and slowly degrade/dissolve under a suitable environment (e.g., aqueous, acidic, basic, etc.) and thereby degrade/dissolve in body fluids and release the active compound(s) therein.
  • the particles are preferably nanoparticles (i.e., in the nanometer range, e.g., in the range of about 1 to about 500 nm in diameter, preferably about 50-200 nm in diameter, most preferably about 100 nm in diameter).
  • the pharmaceutical compositions herein described may also comprise suitable solid of gel phase carriers or excipients. Examples of such carriers or excipients include, but are not limited to, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin and polymers such as polyethylene glycols.
  • compositions suitable for use in context of the present invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an amount of preparation effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the therapeutically effective amount or dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC 50 as determined in cell culture (i.e., the concentration of the test compound, which achieves a half-maximal activity).
  • Toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the IC 50 and the LD 50 (lethal dose causing death in 50 % of the tested animals) for a subject compound.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl, et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 p.l). Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the therapeutic effects, termed the minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each preparation, but can be estimated from in vitro data; e.g., the concentration necessary to achieve 50-90 % activity may be ascertained using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using the MEC value. Preparations should be administered using a regimen, which maintains plasma levels above the MEC for 10-90 % of the time, preferable between 30-90 % and most preferably 50-90 %.
  • the effective local concentration of the drug may not be related to plasma concentration. In such cases, other procedures known in the art can be employed to determine the effective local concentration.
  • dosing can also be a single administration of a slow release composition described hereinabove, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
  • compositions to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • compositions of the present invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accompanied by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert.
  • Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. Suitable conditions are listed hereinabove.
  • Ferrocene (98 % pure ) was obtained from Sigma, Israel (Cat. No.
  • mice C57BL female mice, 6-8 weeks of age, were obtained from the Animal
  • B-16 melanoma and 3LL Two mouse tumors grown in C57BL mice were used: B-16 melanoma and 3LL (Lewis lung carcinoma). B-16 and 3LL were obtained from large stocks of frozen cells and were grown in cultures. Culture medium included RPMI 1640, supplemented with 5 % FCS. The B-16 and 3LL cell stocks were prepared from in vivo subcutaneous growth in C57BL mice. To generate the stocks, B-16 cells were injected intravenously at 5 x 10 4 cells/mouse, whereas 3LL cells were injected intravenously at 2 x 10 5 cells/mouse. Isolation of cells:
  • Mouse splenocytes Spleens were removed aseptically and crushed with the blunt end of a syringe in PBS. The cells were then centrifuged, washed twice with PBS and resuspended in RPMI 1640, supplemented with 0.1 mM non-essential amino acids, 1 mM sodium pyruvate, 5 x 10 5 2-mercaptoethanol, 100 ⁇ g/ml streptomycin, 100 U/ml penicillin, 0.03 % fresh glutamine, and 5 % heat-inactivated fetal calf serum.
  • Mouse peritoneal macrophages C57BL female mice were anesthetized by ether and their peritoneal cavity washed with 3-5 ml of Ca ++ and Mg ++ free PBS. The washing fluid was centrifuged and the cells in the pellet were suspended in RPMI 1640 containing 20 % FCS and distributed into microwells. After incubation for 2 hours at 37 °C, the non-adherent cells were removed by washing (3 times) with Ca " " " , Mg 1-1" free PBS.
  • A9 cells (a TNF- ⁇ sensitive cell line) were incubated for 24 hours in serum free DMEM medium containing cycloheximide (50 ⁇ g/ml) in the presence of experimental aliquots. Recombinant TNF- ⁇ was used to generate a standard curve. Cytotoxicity was assessed using the neutral red vital staining assay. Nitric oxide assay:
  • Nitric oxide levels in experimental aliquots was determined colorimetrically using a Griess reagent, essentially as described (27).
  • Western blot analysis Cells were solubilized with lysis buffer containing 50 mM Tris-HCl (pH 7.5), 0.5 % TRITON X-100, 3 mM EGTA, 12 mM ⁇ -glycerophosphate, 150 mM sodium chloride, 50 mM sodium fluoride, 1 mM sodium vanadate, 2 mM DTT, 1 mM phenylmethylsulfonyl fluoride, 1 mM aprotinin and 0.1 % 2-mercaptoethanol.
  • the cell lysates were then applied to a 10 % polyacrylamide gel.
  • the eletcrophoresed proteins were transblotted onto a nitrocellulose membrane.
  • the membranes were incubated with a mouse monoclonal anti-MAP kinase, activated (Diphosphorylated ERK1 and ERK2), clone MAPK-YT (Sigma M-8159).
  • the membrane was then incubated with HRP-conjugated goat anti-mouse antibody (Jackson) and developed with the enhanced chemiluminescence mixture.
  • GTPase assay :
  • the GTPase assay was carried out as follows: Pure recombinant P21 ras (1 ⁇ M) was added to assay tubes on ice containing 50 ⁇ l of NaCl, 200 mM; Tris, 25 mM; MgCl 2 , 10 mM; and EGTA; 0.5 mM, pH 7.4. To this was added 10 ⁇ l of each of DTT, 20 mM; ATP, 10 mM; adenosine 5'-( ⁇ , ⁇ -imido) triphosphate, 5 mM; creatine phosphate, 100 mM; and creatine kinase, 500 U/ml.
  • PBMC cells (5 x 10 6 /ml) in 1 ml were treated for 4 hours at 37 °C in RPMI 1640 containing 5 % heat inactivated FCS and the indicated treatments.
  • Cells were pelleted, washed once with cold PBS and resuspended in 250 ⁇ l of 10 mM HEPES, pH 7.9, 10 mM KC1, 1.5 mM MgCl 2 , 1 mM EDTA, 0.5 mM DTT, 0.5 mM PMSF, and 10 % glycerol. Cells were left on ice for 10 minutes and Nonidet P-40 was added to a final concentration of 0.04 %.
  • NO nitric oxide
  • Ferrocene stimulated H-thymidine incorporation in mouse splenocytes upon incubation for 3 days, in vitro (Figure 1A). Maximal stimulation was obtained by ferrocene at a dose of 0.05-0.2 ⁇ M. Ferrocene further enhanced 30 stimulation of splenocytes that had been treated with interleukin-2 ( Figure IB). Ferrocene also stimulates H-thymidine incorporation upon administration in vivo. Splenocytes that were isolated from mice that had been injected 3 days earlier with ferrocene (0.05-1 mg/kg body weight) showed increased H-thymidine incorporation above controls ( Figure 2). Purified peritoneal macrophages were not stimulated by ferrocene in vitro.
  • Ferrocene activated peritoneal macrophages in vitro upon their incubation in the presence of mouse lymphocytes (Figure 3). Macrophage activation was assessed by oxygen burst in the presence of phorbol myristate acetate (PMA). Ferrocene also stimulated mouse peritoneal macrophages upon administration in vivo. Peritoneal macrophages were isolated 3 days following a single intraperitoneal ferrocene injection and their activation was assessed by measuring oxygen burst in the presence of PMA (Figure 4).
  • PMA phorbol myristate acetate
  • mice peritoneal macrophages were also assessed by. production of TNF- ⁇ , NO and eliciting an oxygen burst (H 2 0 2 ) ( Figures 5A-C).
  • Mice were injected intraperitoneally with ferrocene (50 ⁇ g/kg body weight).
  • ferrocene 50 ⁇ g/kg body weight.
  • macrophages were isolated and TNF- ⁇ and NO levels were determined in the supernatants after incubation in vitro for 6 hours in the presence of LPS (10 ⁇ g/ml).
  • Oxygen burst was assessed by determination of H 2 O 2 production in the presence of PMA Anti-tumor properties of ferrocene:
  • mice were included in each group. Mice were sacrificed 25 days post tumor inoculation and the lungs were weighed. Weight of lungs in B16 inoculated mice above controls represents tumor load.
  • Ferrocene also has an anti-tumor effect upon administration in drinking water.
  • the effect of ferrocene in drinking water on established lung metastases in B-16 melanoma bearing mice is depicted in Table 1, Figure 9A and Figure 9B. Maximal anti-tumor effect was attained upon administration of ferrocene in drinking water (1 ⁇ g/ml) for 1 week. Administration for 2 weeks was less effective and for 3 weeks had no anti-tumor effect.
  • the anti-tumor effect on B-16 melanoma of ferrocene in drinking water (at different concentrations) administered for one and two weeks was investigated.
  • the most effective anti-tumor protocols were the administration of ferrocene in drinking water for 2 weeks at a concentration of 0.5 ⁇ g/ml and administration in drinking water for 1 week at a concentration of 2 ⁇ g/ml.
  • Other protocols were also effective to different degrees (Table 1 and Figures lOA and lOB).
  • mice were inoculated with B-16 melanoma and treated at day T 2 with ferrocene (200 ⁇ g/kg body weight). Mice were sacrificed at T 25 splenocytes and peritoneal cells (containing 50 % lymphocytes and 50 % macrophages) were isolated and injected intravenously into B-16 inoculated mice (TI).
  • FIG. 14A and 14B the peritoneal mononuclear cells from ferrocene-treated mice were most effective in eliciting an anti-tumor effect.
  • Figure 15 represents an additional experiment illustrating the anti-tumor effect of peritoneal mononuclear cells from ferrocene-treated mice.
  • Mechanisms of action of ferrocene It is postulated that the primary molecular target of ferrocene is p21 ras which has been identified as a key molecule in T lymphocyte activation (15-19). p21 ras has previously been shown to be activated by cellular redox stress (21, 22).
  • H 2 0 2 was measured using the aminotriazole-catalase inhibition assay (see
  • N-acetylcysteine (NAC), a free radical scavenger, on ferrocene-induced anti-tumor effect (B-16 melanoma) was investigated.
  • Ferrocene at an optimal dose of 0.25 mg/kg (250 ⁇ g/kg) body weight or at a supraoptimal dose of 2.5 mg/kg body weight) was injected intraperitoneally to C57BL mice which had been inoculated with B16-melanoma on day Ti.
  • NAC was injected intraperitoneally at a dose of 500 mg/kg body weight twice a day for 3 consecutive days (T l3 T 2 , T 3 )
  • p21 ras cysl l ⁇ is the primary target site for oxidation by ferrocene
  • the stimulatory effect of ferrocene on ERK1/2 was investigated in wild type Jurkat cells and in Jurkat T cells in which p21 ras cysl l ⁇ was replaced by a serine residue (referred to as p21 ras C118S, see Materials and Experimental Methods), which is not susceptible to redox-stress modification.
  • ferrocene activated ERK1/2 in wild type Jurkat cells but failed to do so in Jurkat CI 18S.
  • PHA which does not activate p21 ras via redox stress, phosphorylated ERK1/2 in both cells.
  • Levels of activated ERK1/2 in the cell lysates were determined by Western blot analysis using anti-phosphorylated ERK1/2.
  • Ferrocene stimulates GTPase activity catalyzed by pure recombinant p21 ras in a bell-shaped pattern (Table 2). Ferrocene, at a high concentration (50 nM) was inhibitory.
  • Ferrocene also activates and translocates NFKB in human PBM cells ( Figure 19), a pathway that is mediated by ras. Binding of NFKB to a DNA fragment was demonstrated in an electromobility shift assay as described in Materials and Experimental Methods. Anti-tumor effect ofH 2 0 2 , administered intraperitoneally
  • H 2 0 2 Ferrocene generates hydrogen peroxide (H 2 0 2 ) upon auto-oxidation (see Figure 16). Following the observation that ferrocene elicits an anti-tumor effect, whether H 2 0 2 itself will share this property was investigated. H 2 0 2 was injected intraperitoneally, twice a day at TI, T2 and T3 (1 cycle) or at day T r T 3 , T 8 -T ⁇ o and T 15 -T 17 (3 cycles), each time at a dose of 5 ⁇ M/kg body weight. Mice were sacrificed 25 days post tumor inoculation and tumor load was assessed.
  • Lander HM An essential role for free radicals and derived species in signal transduction. FASEB 11:118-24, 1997.

Abstract

A method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer. The method comprises administering to a subject in need thereof at least one agent in an amount so as to induce H2O2 generation at a concentration effective in inducing immune stimulation and or providing cancer therapy.

Description

METHODS AND PHARMACEUTICAL COMPOSITIONS FOR STIMULATING THE IMMUNE SYSTEM AND/OR TREATING
CANCER
FIELD AND BACKGROUND OF THE INVENTION
The present invention relates to a methods and pharmaceutical compositions useful in inducing immune stimulation and/or treating cancer. More particularly, the present invention relates to methods and pharmaceutical compositions for inducing immune stimulation and/or for treating cancer, which comprise H202 and/or an H202 generating agent, such as metallocenes (e.g., ferrocene) and their derivatives.
Compounds that stimulate the immune system in a non specific manner, have potential for treating tumors that are resistant to conventional therapy.
Biological response modifiers such as IL-2, IFN-α IFN-γ and TNF-α, are all possible candidates. Clinical use of some of these compounds resulted in limited response and has been associated with marked toxicity (1-5).
Agonistic immune stimulatory antibodies, such as anti-CD3, were also evaluated for their anti-tumor activity in humans, with limited success (6,7).
Bacillus Calmette-Guerin (BCG) preparations known to activate the immune system are effective upon administration intravesically against urinary bladder transitional cell carcinoma (TCC) (8).
The immune-stimulatory properties of agents that generate free radicals, including iron-containing compounds such as hemin were previously described (9-13). Hemin, by itself, does not exhibit anti-tumor activity but shows this effect in combination with IL-2 (14).
The protooncogene p21ras has been identified as a key molecular switch involved in regulating T cell activation triggered by different mitogens (15-19).
A breakthrough in understanding the stimulatory activity mechanism of reactive free radicals occurred when it was discovered that p21ras is the primary target of these agents (including iron-containing compounds) and cellular redox stress.
These studies of p21ras have identified a single cysteine residue, cysl 18, which is modified by free radicals. This modification triggers the activation of p21ras and leads to downstream signaling events such as the activation of mitogen-activated protein kinase, activation of phosphatidylinositol 3 '-kinase (PI-3K) and transcription factor activation (20-24).
Metallocenes, such as ferrocene are stable iron-containing compounds which generate free radicals. Metallocenes were so far never suggested as immune stimulants and/or an anti- cancer agents.
While conceiving the present invention, it was hypothesized that metallocenes, such as, for example, ferrocene, could stimulate the immune system and/or provide for anti-cancer therapy. While reducing the present invention to practice this hypothesis was tested in various systems.
SUMMARY OF THE INVENTION
According to one aspect of the present invention there is provided a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising determining an immune stimulation/cancer therapy dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to immune stimulation/cancer therapy within a predetermined range of concentrations. According to further features in preferred embodiments of the invention described below, the immune stimulation is determined in vivo.
According to still further features in the described preferred embodiments, immune stimulation is determined in vitro.
According to still further features in the described preferred embodiments, the immune stimulation is determined with respect to macrophages.
According to still further features in the described preferred embodiments, the immune stimulation is determined with respect to lymphocytes. According to still further features in the described preferred embodiments, the immune stimulation is determined by a cell proliferation assay.
According to still further features in the described preferred embodiments, the cell proliferation assay includes a thymidine incorporation assay.
According to still further features in the described preferred embodiments, the immune stimulation is determined by a cellular response assay. According to still further features in the described preferred embodiments, the cell response assay is selected from the group consisting of an NO production assay, a TNF-α production assay and an oxygen burst assay.
According to another aspect of the present invention there is provided a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising determining a p21ras signal transduction pathway component activity dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to p21ras signal transduction pathway component activity within a predetermined range of concentrations. Preferably, the p21ras signal transduction pathway component is selected from the group consisting of p21ras and NF-κB.
Hence, according to yet additional aspects of the present invention there is provided (i) method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising determining p21ras oxidation dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to p21ras oxidation within a predetermined range of concentrations; (ii) A method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising determining a GTPase activity dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to the GTPase activity within a predetermined range of concentrations; and (iii) A method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising determining an NF-κB activity dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to the NF-κB activity within a predetermined range of concentrations.
According to still another aspect of the present invention there is provided a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising determining H202 generation catalyzed by the metallocene derivative, and determining whether the rate is within a predetermined range.
According to an additional aspect of the present invention there is provided a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising monitoring H202 generation catalyzed by the metallocene derivative, and determining whether the generation is above a predetermined value. According to yet an additional aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, the method comprising administering to a subject in need thereof at least one agent in an amount so as to induce H 02 generation at a concentration effective in inducing immune stimulation and or providing cancer therapy. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount so as to induce H202 generation at a concentration effective in inducing immune stimulation following administration thereof.
According to a further aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial, the method comprising administering to a subject in need thereof at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight.
According to yet a further aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, the method comprising administering to a subject in need thereof at least one agent in an amount effective in activating a p21ras signal transduction pathway component. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in activating a p21ras signal transduction pathway component following administration thereof.
According to still a further aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, the method comprising administering to a subject in need thereof at least one agent in an amount effective in inducing p21ras oxidation. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing p21ras oxidation following administration thereof.
According to still a further aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, the method comprising administering to a subject in need thereof at least one agent in an amount effective in inducing GTPase activity. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing GTPase activity following administration thereof.
According to another aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, the method comprising administering to a subject in need thereof at least one agent in an amount effective in inducing NF-κB activity. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing NF-κB activity following administration thereof.
According to further features in preferred embodiments of the invention described below, the at least one agent comprises at least one ferrocene and/or metallocene derivative.
According to still further features in the described preferred embodiments the amount is between 0.025 - 10 mg/kg body weight.
According to still further features in the described preferred embodiments the amount is between 0.1 - 5 mg/kg body weight. According to still further features in the described preferred embodiments the amount is between 0.2 - 3 mg/kg body weight.
According to still further features in the described preferred embodiments the at least one agent is administered orally and hence the pharmaceutical composition is formulated for oral administration.
According to still further features in the described preferred embodiments the at least one agent is administered by injection and hence pharmaceutical composition is formulated for administration by injection.
According to still further features in the described preferred embodiments the cancer is selected from the group consisting of all malignant tumors, including, but not limited to, carcinoma, sarcoma, lymphoma or hematological malignancies, hence, the pharmaceutical composition is packaged and identified for the treatment of all malignant tumors, including, carcinomas, sarcomas, lymphomas or hematological malignancies. According to still further features in the described preferred embodiments the disease, syndrome or condition is selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency, hence, the pharmaceutical composition is packaged and identified for the treatment of a disease, syndrome or condition selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency.
According to still further features in the described preferred embodiments the metallocene or metallocene derivative has a general formula:
Figure imgf000009_0001
wherein, M is a metal selected from the group consisting of Fe (iron), Cr
(chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni
(nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
Ri-Rg are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
According to still further features in the described preferred embodiments, the method further comprising co-administering at least one additional agent having immune stimulation activity. Accordingly, the pharmaceutical composition, further comprises at least one additional agent having immune stimulation activity, such as, but not limited to, a cytokine (e.g., IL-2, TNF, interferon), a chemokine, an immune-stimulatory antibody (e.g., anti-CD3, anti-CTLA-4) or an anti-tumor vaccine.
According to still further features in the described preferred embodiments, the method further comprising co-administering at least one additional agent having anti-cancer activity. Accordingly, the pharmaceutical composition, further comprises at least one additional agent having anti-cancer activity, such as, but not limited to, chemotherapeutic agents, anti- metabolites, signal transduction inhibitors (e.g., protein tyrosine kinase inhibitors), anti-angiogenesis compounds, antibodies (e.g., anti-receptors, anti-tumor determinants), hormones and hormone antagonists, retinoids, steroids, pro-apoptotic agents, differentiating agents or radiation. According to another aspect of the present invention there is provided a method of inducing immune stimulation and/or treating cancer in a body cavity, the method comprising administering into the body cavity H20 in an amount effective in immune stimulation. Preferably, the amount is of 10-2000 ml of 50-2500 μM H202 administerred in a single or multiple administrations). The body cavity can be, for example, the peritoneal cavity or the pleural cavity.
According to still another aspect of the present invention there is provided a method of inducing immune stimulation and/or treating cancer, the method comprising exposing immune cells ex vivo to at least one ferrocene and/or metallocene derivative, and administering the cells to a subject in need thereof.
According to yet another aspect of the present invention there is provided a method of inducing immune stimulation and/or treating cancer, the method comprising exposing immune cells ex vivo to H202 in a concentration effective in inducing immune stimulation and administering the cells to a subject in need of thereof.
The present invention successfully addresses the shortcomings of the presently known configurations by providing new methodologies with which to induce immune stimulation and/or treat cancer.
BRIEF DESCRIPTION OF THE DRAWINGS
The invention is herein described, by way of example only, with reference to the accompanying drawings. With specific reference now to the drawings in detail, it is stressed that the particulars shown are by way of example and for purposes of illustrative discussion of the preferred embodiments of the present invention only, and are presented in the cause of providing what is believed to be the most useful and readily understood description of the principles and conceptual aspects of the invention. In this regard, no attempt is made to show structural details of the invention in more detail than is necessary for a fundamental understanding of the invention, the description taken with the drawings making apparent to those skilled in the art how the several forms of the invention may be embodied in practice.
In the drawings:
FIGs. 1A-B demonstrate stimulation by ferrocene and IL-2 of H-thymidine incorporation in mouse splenocytes, in vitro. 1A - Mouse splenocytes were incubated in the present of ferrocene, at different concentrations, for 3 days. 3H-Thymidine incorporation during the last 20 hours of incubation was determined. Results are expressed as means CPM+SD of triplicate cultures. IB - Same as 1A, but the cells were cultured in the presence of IL-2 (50 U/ml).
FIG. 2 demonstrates stimulation by ferrocene of 3H-thymidine incorporation in mouse splenocytes, in vivo. Mice were injected intraperitoneally with ferrocene at different doses. Three days later, splenocytes were isolated and incorporation of H-thymidine during 30 hours, was determined. Two mice were included in each group. Results are expressed as means CPM ±SD of triplicate cultures from each mouse.
FIG. 3 demonstrates activation by ferrocene of mouse peritoneal macrophages, in vitro. Peritoneal macrophage monolayers were prepared (as described in the Examples section that follows) and co-incubated with splenic lymphocytes in the presence of ferrocene at different concentrations. The ratio of lymphocytes to macrophages was 10:1. After incubation for 3 days, the non- adherent lymphocytes were removed by washing with PBS. Production of
H202 as a measurement of oxygen burst was determined in the presence of phorbol myristate acetate (PMA) (100 ng/ml), using the aminotriazole-catalase inhibition test (see Examples section that follows). Results are expressed as means ± SD of triplicate cultures.
FIG. 4 demonstrates activation by ferrocene of mouse peritoneal macrophages, in vivo. Mice were injected intraperitoneally with ferrocene at different doses (per mouse). After 3 days, peritoneal macrophages were obtained and oxygen burst was determined in the presence and absence of PMA (100 ng/ml). As a positive control, mice were injected with 1 ml of NaI0 (5 mM). NaI04 was previously shown to activate peritoneal macrophages, in vivo (39). Four mice were included in each experimental group and the results are expressed as means +SD. FIGs. 5A-C demonstrate activation by ferrocene of mouse peritoneal macrophages in vivo: TNF-α production (5 A), oxygen burst (5B), nitric oxide (5C) production. Mice were injected intraperitoneally with ferrocene (1 αg/mouse) or with NaI04 (0.15 M, 1 ml), as a positive control. Three days later the peritoneal cavity was washed with PBS and macrophage monolayers (106 cells/6 mm plate) were obtained as described in the Examples section that follows. LPS (10 μg/ml) was added, and after 6 hours, supernatants were collected and TNF-α (5 A) and nitric oxide (5C) levels were determined as described in the Examples section that follows. Oxygen burst (H202 production, 5B) was determined in the absence and presence of PMA. FIG. 6 demonstrates anti-tumor effect of ferrocene administered intraperitoneally by a single injection (Ti) at different doses in B-16 melanoma bearing mice. Ferrocene was injected intraperitoneally one day post B-16 melanoma inoculation (Ti) via the tail vein. Mice were sacrificed 25 days post tumor inoculation and the pair of lungs from each mouse was weighed. 4-6 mice were included in each experimental group. Results are expressed as the means of the weights of the pairs of lungs of each mouse +SD.
FIG. 7 demonstrates anti-tumor effect of ferrocene at different doses, administered intraperitoneally by multiple injections. Mice were inoculated with B-16 melanoma (To). Ferrocene was administered intraperitoneally 3 times (Ti, T8 and Tι5) each, at a dose specified in the Figure. Mice were sacrificed 25 days post tumor inoculation and the pair of lungs from each mouse was weighed. 4-6 mice were included in each group. Results are expressed as the means of the weights of the pair of lungs of each mouse ±SD.
FIG. 8 demonstrates the anti-tumor effect of ferrocene. Photographs were taken of pairs of lungs from the experiments depicted in Figures 6 and 7. FIGs. 9A-B demonstrate the effect of ferrocene, administered in the drinking water (1 μg/ml) on established lung metastases in B-16 melanoma bearing mice. Mice, inoculated with B-16 melanoma were supplied with drinking water (ad libidum) containing 1 μg/ml ferrocene. A mouse drinks approximately 5 ml water/day, so the overall dose of ferrocene was approximately 0.25 mg/kg body weight/day. One group of mice was supplied with ferrocene in the drinking water for 1 week followed by replacing it with plain water, the second and third groups were supplied with ferrocene in the drinking water for 2 and 3 weeks, respectively. Mice were sacrificed 25 days post tumor inoculation and tumor load was assessed by weighing the lungs. 4-6 mice were included in each group. Results are expressed as the means of the weight of the pair of lungs of each mouse ±SD (9A). Photographs of lungs of the mice that were sacrificed 25 days post tumor inoculation (9B).
FIGs. 10A-B demonstrate anti-tumor effect of ferrocene administered in drinking water at different concentrations. For experimental details see Table 1 below. Photographs of lungs taken 25 days post tumor inoculation. Mice were supplied with drinking water containing ferrocene for 1 week (10A) or 2 weeks (10B).
FIG. 11 demonstrates the effect of ferrocene administered in drinking water (1 μg/ml) for one week on survival of B-16 melanoma bearing mice.
FIG. 12 demonstrates anti -tumor effect of ferrocene in mice bearing Lewis lung carcinoma. Ferrocene was administered intraperitoneally 3 times (Ti, T8 and T15), each at a dose of 0.025, 0.05 and 0.2 mg/kg body weight. Mice were sacrificed 28 days post tumor inoculation and tumor load was assessed by weighing the lungs. Four mice were included in each group and the results are expressed as the means of lungs' weight ±SD.
FIG. 13 demonstrates anti-tumor effect of combined treatment with ferrocene and IL-2 in B16 melanoma bearing mice. Ferrocene was administered intraperitoneally at a single dose on day T[. IL-2 was administered intraperitoneally 3 times on days Tb T2 and T2 each, at a dose of 25 x 104 U/kg body weight). 5-6 mice were included in each group. Mice were sacrificed 25 days post tumor inoculation and tumor load was assessed by weighing the lungs. Results are expressed as the means of lungs' weight ±SD. FIGs. 14A-B demonstrate an adoptive transfer experiments: Anti-tumor effect of immunocytes from ferrocene-treated mice. 14A - Donor cells were obtained from B-16 melanoma bearing mice which were treated on day T0 with a single injection of ferrocene (0.2 mg/kg body weight). Cells were collected from donor mice 25 days post tumor inoculation. Splenocytes (10 cells/mouse)
/: and peritoneal mononuclear cells ("macrophages") (5 x 10 cells/mouse) were administered intravenously to B16 melanoma inoculated recipient mice on day T4. Recipient mice were sacrificed 25 day post tumor inoculation and tumor load was assessed by weighing the lungs. 2-4 recipient mice were included in each experiment and results are expressed as means ±SD. 14B - Photographs of lungs from recipient mice treated with donor ("macrophages") peritoneal mononuclear cells .
FIG. 15 demonstrates an adoptive transfer experiments: Anti-tumor effect of immunocytes from ferrocene-treated mice. Donor cells were obtained from B-16 melanoma bearing mice which were treated on day T0 with a single injection of ferrocene (0.2 mg/kg body weight). Cells were collected from donor mice 25 days post tumor inoculation. Splenocytes (108 cells/mouse) and peritoneal mononuclear cells ("macrophages") (5 x 106 cells/mouse) were administered intravenously to B16 melanoma inoculated recipient mice on day T . Recipient mice were sacrificed 25 day post tumor inoculation and tumor load was assessed by weighing the lungs. 2-4 recipient mice were included in each experiment. Photographs of lungs from recipient mice treated with donor ("macrophages") peritoneal mononuclear cells.
FIG. 16 demonstrate production of H202 in ferrocene solution. H202 production was measured by the aminotriazole-catalase inhibition test (see Examples section that follows). FIG. 17 demonstrates the effect of N-acetyl cysteine (NAC) on the anti-tumor effect of ferrocene in B-16 melanoma bearing mice. Ferrocene was injected intraperitoneally at a single dose of 0.25 mg/kg body weight (optimal) or 2.5mg/kg body weight (supraoptimal), on day T N-acetylcysteine was injected at a dose of 500 mg/kg body weight, twice a day, for 3 days (Ti, T2, T3). Mice were sacrificed 25 days post tumor inoculation and the tumor load was assessed by weighing the lungs. Five mice were included in each group and the results are expressed as means of lungs' weight ±SD.
FIG. 18 demonstrates that ferrocene induces phosphorylation of ERK1/2 in wild type Jurkat cells and not in C118S Jurkat cells. Jurkat cells (wild type and C118S) were incubated (2 x 105 cells/ml) for 24 hours in RPMI 1640 containing 0.5 % FCS. Ferrocene was added at a final concentration of 0.05, 0.2, 1 and 5 μM. PHA was added at a final concentration of 20 μg/ml. After incubation for 5 minutes at 37 °C, cells were centrifuged and suspended in lysis buffer. Western blot analysis for the detection of activated (phosphorylated) ERK1/2 was done as described in the Examples section that follows.
FIG. 19 demonstrates the effects of ferrocene on NF-κB activity. Cells were treated with the indicated concentrations of ferrocene for 4 hours prior to isolation of nuclei and assay for NF-κB binding activity as described in the Examples section that follows. Arrows indicate the protein/DNA complex.
FIG. 20 demonstrates the anti-tumor effect of H202 administered intraperitoneally on established lung metastases in B-16 melanoma-bearing mice. H202 was administered intraperitoneally, twice a day, each, at a dose of 5 μmoles/kg body weight, on day T1? T2, and T3 (1 cycle), or on days T T3, T8-Tιo and T15-T]7 (3 cycles). Mice were sacrificed 25 days post tumor inoculation and tumor load was assessed by lungs' weight. Four mice were included in each group. Results are expressed as means of lungs' weight ± SD.
FIGs. 21A-B demonstrate the anti-tumor effect of H202 intraperitoneally. The experimental design was similar to that depicted in Figure 20, except that 6 mice were included in each group. Scoring of tumor load was done by counting and measuring the size of lungs' metastases as follows: Lesion Size Score
Conglomerate 20
Large 3-5 mm 3
Medium 2-3 mm 2
Small < 2 mm 1
FIG. 22 shows photographs of lungs from mice inoculated with B-16 melanoma. Experimental data is outlined in Figures 21A-B.
DESCRIPTION OF THE PREFERRED EMBODIMENTS
The present invention is of methods and pharmaceutical compositions which can be used for inducing immune stimulation and/or treating cancer.
More specifically, the present invention is of methods and pharmaceutical compositions for inducing immune stimulation and/or for treating cancer, which comprise H202 and/or an H202 generating agent, such as metallocenes (e.g., ferrocene) and their derivatives.
The principles and operation of the present invention may be better understood with reference to the drawings and accompanying descriptions. Before explaining at least one embodiment of the invention in detail, it is to be understood that the invention is not limited in its application to the details set forth in the following description or exemplified by the Examples. The invention is capable of other embodiments or of being practiced or carried out in various ways. Also, it is to be understood that the phraseology and terminology employed herein is for the purpose of description and should not be regarded as limiting.
Oxidizing agents have been shown to regulate protein function by modification of cysteine residues. Their protein targets include Ras, calcium dependent potassium channels, N-methyl-D-aspartate and caspases (28). Ras is a key element of various signaling pathways and is implicated in the regulation of proliferation and differentiation by tyrosine kinase and G protein-coupled receptors (29). It was previously shown that p21ras is a common signaling target of reactive free radicals and cellular redox stress (21) and the immune-stimulatory properties of agents that generate free radicals, including iron-containing compounds such as hemin was described (9-13). Hemin, by itself, does not exhibit anti-tumor activity but shows this effect in combination with IL-2 (14).
While conceiving the present invention, it was hypothesized that metallocenes, stable metal-containing compounds, and metallocene derivatives could stimulate the immune system and/or provide for anti-cancer therapy. While reducing the present invention to practice this hypothesis was tested in various systems. Hence, it is demonstrated herein that ferrocene, induces in vitro and in vivo activation of mouse splenocytes and peritoneal macrophages. Ferrocene also has a marked anti-tumor effect in mice bearing B-16 melanoma and Lewis lung carcinoma.
The immune stimulatory and anti-tumor effects of ferrocene follow a bell shaped curve, namely ferrocene at high, supraoptimal concentrations, is far less effective. The inhibiting effects of ferrocene at high concentrations may be due to oxidation of thiol moieties and other susceptible molecules that are not relevant to the stimulatory process. Reactive oxygen species have been implicated in damaging biomolecular components including lipids, proteins and DNA.
Previous reports indicated that ferrocene at doses of 20-500 mg/kg body weight did not inhibit Ehrlich ascites tumor in mice (30), whereas other metallocene derivatives at high doses (180-300 mg/kg body weight) exhibited anti-tumor activity. The LD5o dose of these compounds ranged between 240-400 mg/kg body weight. It should be noted that, as reported herein, the effective dose of ferrocene that elicit an anti-tumor effect is approximately 2000 fold less than that reported for the anti-tumor effect of metallocene derivatives. The LD50 of ferrocene is 440 mg/kg body weight (30). The inhibitory effects of ferrocene derivatives, at high doses, on solid tumors in mice and two cell lines (31, 32) were demonstrated under conditions exhibiting immunotoxic activities (33).
It is plausible that the anti-tumor activity of metallocene derivatives, at high dose, resulted from a direct effect on the tumor itself.
It is postulated that the anti-tumor activity of ferrocene at low doses, 0.05-0.2 mg/kg bodyweight (administered intraperitoneally) and 0.5-2 μg/ml in drinking water, is mediated by immune-stimulation. This is based on the findings that ferrocene, at low doses, activates p21ras and ERK1/2, stimulates lymphocyte proliferation and activates peritoneal macrophages. Most importantly, adoptive transfer experiment demonstrated that mouse peritoneal monocuclear cells from ferrocene-treated mice have an anti-tumor effect in mice that were otherwise not treated with this compound.
Anti-tumor effects of agents known to stimulate the immune-system have been previously reported (1-5). They include IL-2, cytokines and agonistic antibodies such as anti CD3 (6,7) and anti BAT monoclonal antibody (34). Of interest are the findings described herein that H202 administered intraperitoneally, at a low dose, has an anti-tumor effect. H202-induced activation of the MAPK and NFKB pathways has been previously reported (35-37). Hydrogen peroxide was also reported to activate T lymphocytes (38). In several independent assays treatment with ferrocene showed a bell-shaped (optimum) response curve. Hence, methods of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy constitute an embodiment of the present invention. Hence, according to one aspect of the present invention there is provided a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy. The method according to this aspect of the present invention comprises determining an immune stimulation/cancer therapy dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to immune stimulation/cancer therapy within a predetermined range of concentrations. As is further exemplified in the Examples section, immune stimulation can be determined in vivo or in vitro (e.g., ex vivo). As is still further exemplified in the Examples section the immune stimulation can be determined with respect to macrophages and/or lymphocytes, by a cell proliferation assay, such as a thymidine incorporation assay or by a cellular response assay, such as an NO production assay, a TNF-α production assay and/or an oxygen burst assay.
According to another aspect of the present invention there is provided a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy. The method according to this aspect of the invention comprises determining a p21ras signal transduction pathway component activity dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to p21ras signal transduction pathway component activity within a predetermined range of concentrations. Preferably, the p21ras signal transduction pathway component is p21ras and/or NF-κB.
Hence, according to yet additional aspects of the present invention there is provided (i) method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising determining p21ras oxidation dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to p21ras oxidation within a predetermined range of concentrations; (ii) a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising determining a GTPase activity dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to the GTPase activity within a predetermined range of concentrations; and (iii) a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy, the method comprising determining an NF-κB activity dose-response curve for the metallocene derivative and determining whether the dose-response curve has an optimum with respect to the NF-κB activity within a predetermined range of concentrations. Without being bound by a theory, it is postulated that ferrocene and metallocene derivatives are effective immunostimulant as they catalyze the generation of H202 which reacts with susceptible molecules, or react directly with thiol moeties such as CI 18 in p21ras.
Ferrocene at 5-500 nM under the experimental system described in the Examples section that follows, generated 9-12 micromolar H202. It is hence assumed that metallocene derivatives with a similar capabilities of H202 generation are effective immunostimulants and/or effective in cancer therapy.
Thus, according to still another aspect of the present invention there is provided a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy. The method according to this aspect of the present invention comprises monitoring the rate of H202 generation catalyzed by the metallocene derivative, and determining whether the rate is within a predetermined range.
Kinetic parameters determination assay is a multi data points assay and hence less amenable for highthroughput screening. However, based on the preferred range of parameters a single data point assay (a highthroughput assay) for H202 generation catalyzed by metallocene derivatives can be developed by those of skills in the art so as to be used in determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy. Hence, according to an additional aspect of the present invention there is provided a method of determining whether a metallocene derivative is an effective immunostimulant and/or effective in cancer therapy. The method according to this aspect of the invention comprises monitoring H202 generation catalyzed by the metallocene derivative, and determining whether the generation is above a predetermined value. According to yet an additional aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer. The method according to this aspect of the present invention comprises administering to a subject in need thereof at least one agent in an amount so as to induce H202 generation at a concentration effective in inducing immune stimulation and or providing cancer therapy. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one agent in an amount so as to induce H202 generation at a concentration effective in inducing immune stimulation following administration thereof.
According to a further aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial. The method according to this aspect of the present invention comprises administering to a subject in need thereof at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight.
According to yet a further aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer. The method according to this aspect of the present invention comprises administering to a subject in need thereof at least one agent in an amount effective in activating a p21ras signal transduction pathway component. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one agent in an amount effective in activating a p21ras signal transduction pathway component following administration thereof.
According to still a further aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer. The method according to this aspect of the present invention comprises administering to a subject in need thereof at least one agent in an amount effective in inducing p21ras oxidation. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one agent in an amount effective in inducing p21ras oxidation following administration thereof.
According to still a further aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer. The method according to this aspect of the present invention comprises administering to a subject in need thereof at least one agent in an amount effective in inducing GTPase activity. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one agent in an amount effective in inducing GTPase activity following administration thereof.
According to another aspect of the present invention there is provided a method of treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer. The method according to this aspect of the present invention comprises administering to a subject in need thereof at least one agent in an amount effective in inducing NF-κB activity. Accordingly, there is also provided a pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or treating cancer, each dose-unit of the pharmaceutical composition comprises, as an active ingredient, at least one agent in an amount effective in inducing NF-κB activity following administration thereof.
According to a presently preferred embodiment of the present invention the agent(s) used in the methods and compositions described above comprises at least one ferrocene and/or metallocene derivative and the amount thereof in a dose-unit is 0.025 - 10 mg/kg body weight, which translates to 0.25 - 1000 mg per dose-unit for individuals weighting 10-100 kg. Preferably, the amount of the ferrocene and/or metallocene derivative in a dose-unit is 0.1 - 5 mg/kg body weight, which translates to 1 - 500 mg per dose-unit for individuals weighting 10-100 kg. Still preferably, the amount of the ferrocene and/or metallocene derivative in a dose-unit is 0.2 - 3 mg/kg body weight, which translates to 2 - 300 mg per dose-unit for individuals weighting 10-100 kg.
Preferably the agent(s) used according to the present invention for treating a disease, syndrome or condition, in which immune stimulation is beneficial and/or for treating cancer are administered orally or by injection. Hence, the pharmaceutical compositions described herein are preferably formulated for oral administration or administration by injection. Other modes of application are further described herein. The present invention offers treatment for any and all cancers. These include, for example, carcinomas (e.g., colon, lung, breast, prostate, ovary), sarcomas (e.g., soft tissue, bone), lymphomas (e.g., Hodgkin lymphoma, non-Hodgkin lymphoma), hematological malignancies (e.g., myelogenous leukemia, lymphocytic leukenia). Hence, the pharmaceutical composition is preferably packaged and identified for the treatment of carcinomas (e.g., colon, lung, breast, prostate, ovary), sarcomas (e.g., soft tissue, bone), lymphomas (e.g., Hodgkin , non-Hodgkin), hematological malignancies (e.g., myelogenous leukemia, lymphocytic leukenia).
The present invention further offers treatment for acquired immune deficiency, such as AIDS, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency (e.g., SKID). Hence, the pharmaceutical composition can be packaged and identified for the treatment of a disease, syndrome or condition, such as an acquired immune deficiency, a radiotherapy induced immune deficiency, a chemotherapy induced immune deficiency and a congenital immune deficiency. The metallocene or metallocene derivatives used in the methods and pharmaceutical compositions of the present invention has a a general formula:
Figure imgf000024_0001
wherein, M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and R]-R8 are each independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryl and aryloxy.
As used herein, the term "alkyl" refers to a saturated aliphatic hydrocarbon including straight chain and branched chain groups. Preferably, the alkyl group has 1 to 10 carbon atoms. Whenever a numerical range; e.g., "1-10", is stated herein, it means that the group, in this case the alkyl group, may contain 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. More preferably, it is a medium size alkyl having 1 to 6 carbon atoms. Most preferably, it is a lower alkyl having 1 to 4 carbon atoms. The alkyl group may be substituted or unsubstituted. When substituted, the substituent group can be, for example, cycloalkyl, aryl, hydroxy, alkoxy, aryloxy, cyano, halo, carbonyl, nitro, silyl, or amino.
A "cycloalkyl" group refers to an all-carbon monocyclic or fused ring (i.e., rings which share an adjacent pair of carbon atoms) group wherein one of more of the rings does not have a completely conjugated pi-electron system. Examples, without limitation, of cycloalkyl groups are cyclopropane, cyclobutane, cyclopentane, cyclopentene, cyclohexane, cyclohexadiene, cycloheptane, cycloheptatriene, and adamantane. A cycloalkyl group may be substituted or unsubstituted. When substituted, the substituent group can be, for example, cycloalkyl, aryl, hydroxy, alkoxy, aryloxy, cyano, halo, carbonyl, nitro, silyl, or amino.
An "alkenyl" group refers to an alkyl group which consists of at least two carbon atoms and at least one carbon-carbon double bond.
An "alkynyl" group refers to an alkyl group which consists of at least two carbon atoms and at least one carbon-carbon triple bond.
An "alkoxy" group refers to both an -O-alkyl and an -O-cycloalkyl group, as defined herein.
An "aryl" group refers to an all-carbon monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of carbon atoms) groups having a completely conjugated pi-electron system. Examples, without limitation, of aryl groups are phenyl, naphthalenyl and anthracenyl. The aryl group may be substituted or unsubstituted. When substituted, the substituent group can be, for example, halo, trihalomethyl, cycloalkyl, aryl, hydroxy, alkoxy, aryloxy, cyano, carbonyl, nitro, silyl, or amino. An "aryloxy" group refers to an -O-aryl as defined herein.
A "halo" group refers to flourine, chlorine, bromine or iodine. A "trihalomethyl" group refers to a -CX3 group wherein X is a halo group as defined herein.
A hydroxy group refers to a -OH group. An "Amino" group refers to an -NH2 group. A "nitro" group refers to an -N02 group.
A "cyano" group refers to a -C=N group.
A "carbonyl" group refers to a -C( =0)-R' group, where R' is hydrogen, alkyl, cycloalkyl or aryl, as defined herein. A "silyl" group refers to a -Si(R')3, where R' is hydrogen, alkyl, cycloalkyl or aryl, as defined herein.
Depending on the disease, syndrome or condition to be treated, according to a preferred embodiment of the present invention, the method further comprises co-administering at least one additional agent having immune stimulation and/or anti-cancer activity. Accordingly, the pharmaceutical composition, further comprises at least one additional agent having immune stimulation and/or anti-cancer activity. Agents having immune stimulation activity include, for example, cytokines (e.g., IL-2, TNF, interferon), chemokines, immune-stimulatory antibodies (e.g., anti-CD3, anti-CTLA-4) or an anti-tumor vaccine. Agents having anti-cancer activity include, for example, chemotherapeutic agents, anti-metabolites, signal transduction inhibitors (e.g., protein tyrosine kinase inhibitors), anti-angiogenesis compounds, antibodies (e.g., anti-receptors, anti-rumor determinants), hormones and hormone antagonists, retinoids, steroids, pro-apoptotic agents, differentiating agents or radiation.
It is shown herein that ferrocene activity with respect to immune stimulation and/or cancer therapy is mediated, at least in part, by H202 generation. Hence, according to another aspect of the present invention there is provided a method of inducing immune stimulation and/or treating cancer in a body cavity. The method according to this aspect of the present invention comprises administering into the body cavity of a subject in need thereof H202 in an amount effective in immune stimulation. Preferably, the amount is of 10-2000 ml of 50-2500 μM H202 in a single or multiple administrations. The body cavity can be, for example, the peritoneal cavity or the pleural cavity. It is further shown herein that cells derived from mice treated with ferrocene are effective in inducing immune stimulation and/or treating cancer in adoptive transfer experiments. Hence, according to still another aspect of the present invention there is provided a method of inducing immune stimulation and/or treating cancer. The method according to this aspect of the present invention comprises exposing immune cells ex vivo to at least one ferrocene and/or metallocene derivative, and administering the cells to a subject in need thereof. According to yet another aspect of the present invention there is provided a method of inducing immune stimulation and/or treating cancer. The method according to this aspect of the present invention comprises exposing immune cells ex vivo to H202 in a concentration effective in inducing immune stimulation and administering the cells to a subject in need of thereof.
As used herein a "pharmaceutical composition" refers to a preparation of one or more of metallocene or metallocene derivatives (including salts and acids thereof) described herein, with other chemical components such as physiologically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
Herein the term "active ingredient" includes compounds directly accountable for a biological effect.
Hereinafter, the terms "physiologically acceptable carrier" and "pharmaceutically acceptable carrier" which may be interchangeably used refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
Herein the term "excipient" refers to an inert substance added to a pharmaceutical composition to further facilitate administration of a compound. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols. Techniques for formulation and administration of drugs may be found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference.
Suitable routes of administration may, for example, include oral, rectal, transmucosal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intravenous, inrtaperitoneal, intrapleural, intranasal, or intraocular injections.
Alternately, one may administer a preparation in a local rather than systemic manner, for example, via injection of the preparation directly into a solid tumor often in a depot or slow release formulation, such as described below.
Furthermore, one may administer the drug in a targeted drag delivery system, for example, in a liposome coated with a tumor specific antibody. The liposomes will be targeted to and taken up selectively by the tumor.
Pharmaceutical compositions of the present invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes. Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compounds into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
For injection, the compounds of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets-, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient. Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses. Pharmaceutical compositions, which can be used orally, include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration. For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
The preparations described herein may be formulated for parenteral administration, e.g., by bolus injection or continuos infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative. The compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
Pharmaceutical compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use. The preparation of the present invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
In addition to the formulations described previously, a preparation of the present invention may also be formulated for local administration, such as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the preparation may be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives such as sparingly soluble salts. Formulations for topical administration may include, but are not limited to, lotions, suspensions, ointments gels, creams, drops, liquids, sprays emulsions and powders.
According to a preferred embodiment of the present invention, the pharmaceutical composition is designed for a slow release of the active ingredients. The composition includes particles including a slow release carrier (typically, a polymeric carrier), such as, for example, polylactic acid, and the active ingredients. Slow release biodegradable carriers are well known in the art. These are materials that may form particles that may capture therein an active compound(s) and slowly degrade/dissolve under a suitable environment (e.g., aqueous, acidic, basic, etc.) and thereby degrade/dissolve in body fluids and release the active compound(s) therein. The particles are preferably nanoparticles (i.e., in the nanometer range, e.g., in the range of about 1 to about 500 nm in diameter, preferably about 50-200 nm in diameter, most preferably about 100 nm in diameter). The pharmaceutical compositions herein described may also comprise suitable solid of gel phase carriers or excipients. Examples of such carriers or excipients include, but are not limited to, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin and polymers such as polyethylene glycols.
Many of the compounds in the claimed preparations of the present invention may be provided as physiologically acceptable salts wherein the compound may form the negatively or the positively charged species.
Pharmaceutical compositions suitable for use in context of the present invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an amount of preparation effective to prevent, alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
For any preparation used in the methods of the invention, the therapeutically effective amount or dose can be estimated initially from cell culture assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (i.e., the concentration of the test compound, which achieves a half-maximal activity). Such information can be used to more accurately determine useful doses in humans. Toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the IC50 and the LD50 (lethal dose causing death in 50 % of the tested animals) for a subject compound. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl, et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 p.l). Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the therapeutic effects, termed the minimal effective concentration (MEC). The MEC will vary for each preparation, but can be estimated from in vitro data; e.g., the concentration necessary to achieve 50-90 % activity may be ascertained using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. HPLC assays or bioassays can be used to determine plasma concentrations.
Dosage intervals can also be determined using the MEC value. Preparations should be administered using a regimen, which maintains plasma levels above the MEC for 10-90 % of the time, preferable between 30-90 % and most preferably 50-90 %.
It is noted that, in the case of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration. In such cases, other procedures known in the art can be employed to determine the effective local concentration.
Depending on the severity and responsiveness of the condition to be treated, dosing can also be a single administration of a slow release composition described hereinabove, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
The amount of a composition to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
Compositions of the present invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accompanied by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert. Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. Suitable conditions are listed hereinabove.
Additional objects, advantages, and novel features of the present invention will become apparent to one ordinarily skilled in the art upon examination of the following examples, which are not intended to be limiting. Additionally, each of the various embodiments and aspects of the present invention as delineated hereinabove and as claimed in the claims section below finds experimental support in the following examples.
EXAMPLES
Reference is now made to the following examples, which together with the above descriptions, illustrate the invention in a non limiting fashion.
MATERIALS AND EXPERIMENTAL METHODS Ferrocene:
Ferrocene (98 % pure ) was obtained from Sigma, Israel (Cat. No.
F40-8). Stock solutions (5 mg/ml) were made in absolute ethanol and dilutions were made in NaCl. 0.9 % Hydrogen peroxide (H202), ACS reagent, 30 % in water was obtained from FLUKA, Israel (Cat. No. 95300). Recombinant human IL-2 (2 x 10 IU/mg in NaCl 0.9 %) was obtained from ReproTech, Inc., NJ., USA.
Mice: C57BL female mice, 6-8 weeks of age, were obtained from the Animal
Breeding Center of the Tel Aviv University.
Tumor Models:
Two mouse tumors grown in C57BL mice were used: B-16 melanoma and 3LL (Lewis lung carcinoma). B-16 and 3LL were obtained from large stocks of frozen cells and were grown in cultures. Culture medium included RPMI 1640, supplemented with 5 % FCS. The B-16 and 3LL cell stocks were prepared from in vivo subcutaneous growth in C57BL mice. To generate the stocks, B-16 cells were injected intravenously at 5 x 104 cells/mouse, whereas 3LL cells were injected intravenously at 2 x 105 cells/mouse. Isolation of cells:
Mouse splenocytes: Spleens were removed aseptically and crushed with the blunt end of a syringe in PBS. The cells were then centrifuged, washed twice with PBS and resuspended in RPMI 1640, supplemented with 0.1 mM non-essential amino acids, 1 mM sodium pyruvate, 5 x 105 2-mercaptoethanol, 100 μg/ml streptomycin, 100 U/ml penicillin, 0.03 % fresh glutamine, and 5 % heat-inactivated fetal calf serum. Cells (2 x 106/ml) were distributed (0.2 ml aliquots) in flat bottomed microwells and incubated at 37 °C, 5 % C02 for 68 hours. 3H-Thymidine incorporation (2 μCi/well) into DNA during the final 20 hours of incubation was determined. The means ±SD of CPM of triplicate cultures were determined.
Mouse peritoneal macrophages: C57BL female mice were anesthetized by ether and their peritoneal cavity washed with 3-5 ml of Ca++ and Mg++ free PBS. The washing fluid was centrifuged and the cells in the pellet were suspended in RPMI 1640 containing 20 % FCS and distributed into microwells. After incubation for 2 hours at 37 °C, the non-adherent cells were removed by washing (3 times) with Ca""", Mg1-1" free PBS.
Assessment of oxygen burst in macrophages by determination ofH202 produced, using a sensitive assay: The assay was done essentially as described by Scanonne et al., (25) and is based on the findings that H202 irreversibly inactivates catalase in the presence of 3-amine-l,2,4-triazole (26). Sodium perborate served as a substrate for catalase and its amount was determined by titration with KMn0 . TNF- a determination: TNF-α was determined using a bioassay, as previously described (27).
A9 cells (a TNF-α sensitive cell line) were incubated for 24 hours in serum free DMEM medium containing cycloheximide (50 μg/ml) in the presence of experimental aliquots. Recombinant TNF-α was used to generate a standard curve. Cytotoxicity was assessed using the neutral red vital staining assay. Nitric oxide assay:
Nitric oxide levels in experimental aliquots was determined colorimetrically using a Griess reagent, essentially as described (27). Western blot analysis: Cells were solubilized with lysis buffer containing 50 mM Tris-HCl (pH 7.5), 0.5 % TRITON X-100, 3 mM EGTA, 12 mM β-glycerophosphate, 150 mM sodium chloride, 50 mM sodium fluoride, 1 mM sodium vanadate, 2 mM DTT, 1 mM phenylmethylsulfonyl fluoride, 1 mM aprotinin and 0.1 % 2-mercaptoethanol. The cell lysates were then applied to a 10 % polyacrylamide gel. The eletcrophoresed proteins were transblotted onto a nitrocellulose membrane. After blocking with 5 % (w/v) of non fat dry milk, the membranes were incubated with a mouse monoclonal anti-MAP kinase, activated (Diphosphorylated ERK1 and ERK2), clone MAPK-YT (Sigma M-8159). The membrane was then incubated with HRP-conjugated goat anti-mouse antibody (Jackson) and developed with the enhanced chemiluminescence mixture. GTPase assay:
The GTPase assay was carried out as follows: Pure recombinant P21ras (1 μM) was added to assay tubes on ice containing 50 μl of NaCl, 200 mM; Tris, 25 mM; MgCl2, 10 mM; and EGTA; 0.5 mM, pH 7.4. To this was added 10 μl of each of DTT, 20 mM; ATP, 10 mM; adenosine 5'-(β, γ-imido) triphosphate, 5 mM; creatine phosphate, 100 mM; and creatine kinase, 500 U/ml. The final reaction volume was 100 μl and therefore the final concentration of components was 10-fold less than that added. Then, 100,000 dpm of GTP-γ32P (5000 Ci/mmol) were added as well as any test reagent, and tubes transferred to a shaking 37 °C water bath for 10 minutes. At the end of the assay period, 0.8 ml of an acid charcoal solution was added (I N HC1 containing 10 % (v/v) Norit A) and samples were spun for 3 minutes at 14,000 x g. The supernatants (450 μl) were counted in a liquid scintillation counter. Data are expressed as fmol P04 " released per minute per mg. Electromobility shift assay:
PBMC cells (5 x 106/ml) in 1 ml were treated for 4 hours at 37 °C in RPMI 1640 containing 5 % heat inactivated FCS and the indicated treatments. Cells were pelleted, washed once with cold PBS and resuspended in 250 μl of 10 mM HEPES, pH 7.9, 10 mM KC1, 1.5 mM MgCl2, 1 mM EDTA, 0.5 mM DTT, 0.5 mM PMSF, and 10 % glycerol. Cells were left on ice for 10 minutes and Nonidet P-40 was added to a final concentration of 0.04 %. After 5 minutes on ice, nuclear extracts were pelleted at 10,000 x g for 5 minutes. Supernatants were removed and 100 μl of 20 mM HEPES, pH 7.9, 200 mM KC1, 20 % glycerol, 0.5 mM PMSF, and 1 mM EDTA was added to the pellets. After another hour on ice, pellets were spun at 100,000 x g for 20 minutes. Supernatants were stored at -70 °C until use. Protein concentrations were determined by the method of Lowry (Gaunce,AP and DTorio,A., Microdetermination of protein by a automated Lowry method. Anal Biochem,37:204-207,1970). The NF-KB DNA probe used contained the 206 to -195 region of the IL-2 promoter (5*-CCAAGAGGGATTTCACCTAAATCC-3', SEQ ID NO:l).
Approximately 104 cpm (0.2 ng) of 5'-end labeled DNA was added to 5 μg nuclear protein and 2 μg Poly (dl-dC) in 20 μl of 10 mM Tris, pH 7.5, 50 5 mM NaCl, 1 mM EDTA, 1 mM DTT, and 5 % glycerol at room temperature for 20 minutes. Protein-DNA complexes were resolved on 4 % poly aery lamide gels in 45 mM tris, 45 mM borate, and 1 mM EDTA pH 8.3 at 150 V for 1.5 hours at room temperature. Gels were dried and exposed to x-ray film. The nonspecific probe used contained the -93 to -69 region of the IL-2 promoter l o (5'-TTACAAAATGTATAATGTGTATAA-3', SEQ ID NO:2).
Jurkat T-cells stably expressing p2lras C118S (1-189):
Jurkat T-cells stably expressing p21ras C118S (1-189), in which cysteine at position 118 was replaced by serine, were obtained from Dr. Harry M. Lander, Department of Biochemistry, Cornell University College of Medicine, 15 New York. These cells did not respond to stimulation by nitric oxide (NO) as assessed by activation of ERK1/2 but did so in response to PHA. These transfected cells expressed 7- 10-fold more p21ras than the wild-type cells as determined by Western blotting with the anti-p21ras antibody, Y13-259. This antibody cannot distinguish between wild-type and p21ras C118S, suggesting 20 that although endogenous p21ras was not specifically inhibited, ectopic expression of high levels of mutant p21ras apparently prevented its signaling. This dominant negative activity of p21rasC118S toward NO action may be due to its high level of expression (22).
25 EXPERIMENTAL RESULTS
Immune-stimulation properties of ferrocene:
Ferrocene stimulated H-thymidine incorporation in mouse splenocytes upon incubation for 3 days, in vitro (Figure 1A). Maximal stimulation was obtained by ferrocene at a dose of 0.05-0.2 μM. Ferrocene further enhanced 30 stimulation of splenocytes that had been treated with interleukin-2 (Figure IB). Ferrocene also stimulates H-thymidine incorporation upon administration in vivo. Splenocytes that were isolated from mice that had been injected 3 days earlier with ferrocene (0.05-1 mg/kg body weight) showed increased H-thymidine incorporation above controls (Figure 2). Purified peritoneal macrophages were not stimulated by ferrocene in vitro. Ferrocene activated peritoneal macrophages in vitro upon their incubation in the presence of mouse lymphocytes (Figure 3). Macrophage activation was assessed by oxygen burst in the presence of phorbol myristate acetate (PMA). Ferrocene also stimulated mouse peritoneal macrophages upon administration in vivo. Peritoneal macrophages were isolated 3 days following a single intraperitoneal ferrocene injection and their activation was assessed by measuring oxygen burst in the presence of PMA (Figure 4).
Activation of mouse peritoneal macrophages in vivo was also assessed by. production of TNF-α, NO and eliciting an oxygen burst (H202) (Figures 5A-C). Mice were injected intraperitoneally with ferrocene (50 μg/kg body weight). Three days later, macrophages were isolated and TNF-α and NO levels were determined in the supernatants after incubation in vitro for 6 hours in the presence of LPS (10 μg/ml). Oxygen burst was assessed by determination of H2O2 production in the presence of PMA Anti-tumor properties of ferrocene:
The anti-tumor effect of a single intraperitoneal administration of ferrocene, at different doses, in B-16 melanoma bearing mice was investigated (5 mice were included in each group). Mice were sacrificed 25 days post tumor inoculation and the lungs were weighed. Weight of lungs in B16 inoculated mice above controls represents tumor load.
Results indicated that ferrocene at a single dose of 20 μg/kg body weight was most effective in eliciting an anti-tumor effect. Ferrocene at higher doses (1 and 5 mg/kg body weight) was significantly less effective (Figure 6). A similar pattern of results was obtained upon multiple administrations of ferrocene at different doses, each at T T8 and T15 (T0, the day of tumor inoculation) (Figure 7). However, ferrocene at a dose of 50 μg/kg body weight, was more effective upon administration at Ti, T8 and T15 compared to administration at Ti, alone (Figure 8).
Ferrocene also has an anti-tumor effect upon administration in drinking water. The effect of ferrocene in drinking water on established lung metastases in B-16 melanoma bearing mice is depicted in Table 1, Figure 9A and Figure 9B. Maximal anti-tumor effect was attained upon administration of ferrocene in drinking water (1 μg/ml) for 1 week. Administration for 2 weeks was less effective and for 3 weeks had no anti-tumor effect.
Table 1
Anti-tumor effect of ferrocene in drinking water on established lung metastases of B-16 melanoma
Figure imgf000040_0001
* Scoring index was determined by assessment of tumor load by counting and measuring lungs' metastases as follows: conglomerate - 20; large (> 5 mm) - 3; medium (2-5 mm) - 2; small (< 2 mm) -1. Mean ±SD of lungs' weight (mg) of normal mice (n=6) was 203 ±7.
The anti-tumor effect on B-16 melanoma of ferrocene in drinking water (at different concentrations) administered for one and two weeks was investigated. The most effective anti-tumor protocols were the administration of ferrocene in drinking water for 2 weeks at a concentration of 0.5 μg/ml and administration in drinking water for 1 week at a concentration of 2 μg/ml. Other protocols were also effective to different degrees (Table 1 and Figures lOA and lOB).
Ferrocene administered in drinking water (1 μg/ml) for one week prolonged the survival of B-16 melanoma bearing mice (Figure 11). Ferrocene also has an anti-tumor effect in Lewis lung carcinoma bearing mice.
Intraperitoneal administration of ferrocene at a dose of 50 μg/kg body weight, 3 times each, on days Ti T8 and T15 markedly reduced established lung metastases of Lewis lung carcinoma (Figure 12). Ferrocene at a higher dose (200 μg/kg body weight) was less effective.
The anti-tumor effect of combined treatment with ferrocene at a low dose (50 μg/kg body weight at T along with IL-2 (250 x 103 U/kg body weight on days TI, T2 and T3) against B-16 melanoma was more pronounced than by treatment with each of the agents alone (Figure 13). Adoptive transfer experiments
Based on the assumption that ferrocene mediates its effect by immune stimulation, whether immunocytes from ferrocene treated mice could elicit an anti-tumor effect in otherwise untreated tumor-inoculated mice was investigated. Donor mice were inoculated with B-16 melanoma and treated at day T2 with ferrocene (200 μg/kg body weight). Mice were sacrificed at T25 splenocytes and peritoneal cells (containing 50 % lymphocytes and 50 % macrophages) were isolated and injected intravenously into B-16 inoculated mice (TI). As depicted in Figures 14A and 14B, the peritoneal mononuclear cells from ferrocene-treated mice were most effective in eliciting an anti-tumor effect. Figure 15 represents an additional experiment illustrating the anti-tumor effect of peritoneal mononuclear cells from ferrocene-treated mice. Mechanisms of action of ferrocene: It is postulated that the primary molecular target of ferrocene is p21ras which has been identified as a key molecule in T lymphocyte activation (15-19). p21ras has previously been shown to be activated by cellular redox stress (21, 22).
The following experiments support this postulation.
1. Auto-oxidation of ferrocene in PBS generates H202 (Figure 16). H202 was measured using the aminotriazole-catalase inhibition assay (see
Materials and Experimental Methods section above). The effect of
N-acetylcysteine (NAC), a free radical scavenger, on ferrocene-induced anti-tumor effect (B-16 melanoma) was investigated.
Ferrocene at an optimal dose of 0.25 mg/kg (250 μg/kg) body weight or at a supraoptimal dose of 2.5 mg/kg body weight) was injected intraperitoneally to C57BL mice which had been inoculated with B16-melanoma on day Ti.
NAC was injected intraperitoneally at a dose of 500 mg/kg body weight twice a day for 3 consecutive days (Tl3 T2, T3)
The anti-tumor effect of ferrocene at an optimal dose of 250 μg/kg body weight was markedly inhibited by N-acetylcysteine. In contrast, NAC .enhanced the anti-tumor effect in mice that were treated with ferrocene, at a single-supraoptimal dose of 2.5 mg/kg body weight which, by itself, had a very little anti-tumor effect (Figure 17). These results are in accordance with the findings presented herein that the anti-tumor effect of ferrocene follows a "bell shape" curve, namely it is most effective at an optimal dose and is less active at higher doses.
3. To establish that p21ras cysl lδ is the primary target site for oxidation by ferrocene, the stimulatory effect of ferrocene on ERK1/2 (which is mediated by ras) was investigated in wild type Jurkat cells and in Jurkat T cells in which p21ras cysl lδ was replaced by a serine residue (referred to as p21ras C118S, see Materials and Experimental Methods), which is not susceptible to redox-stress modification.
As depicted in Figure 18, ferrocene activated ERK1/2 in wild type Jurkat cells but failed to do so in Jurkat CI 18S. On the other hand, PHA which does not activate p21ras via redox stress, phosphorylated ERK1/2 in both cells. Levels of activated ERK1/2 in the cell lysates were determined by Western blot analysis using anti-phosphorylated ERK1/2.
4. Ferrocene stimulates GTPase activity catalyzed by pure recombinant p21ras in a bell-shaped pattern (Table 2). Ferrocene, at a high concentration (50 nM) was inhibitory.
Table 2
Figure imgf000043_0001
* The effect of ferrocene on GTPase activity of p21 was determined as outlined in Materials and Experimental Methods.
5. Ferrocene also activates and translocates NFKB in human PBM cells (Figure 19), a pathway that is mediated by ras. Binding of NFKB to a DNA fragment was demonstrated in an electromobility shift assay as described in Materials and Experimental Methods. Anti-tumor effect ofH202, administered intraperitoneally
Ferrocene generates hydrogen peroxide (H202) upon auto-oxidation (see Figure 16). Following the observation that ferrocene elicits an anti-tumor effect, whether H202 itself will share this property was investigated. H202 was injected intraperitoneally, twice a day at TI, T2 and T3 (1 cycle) or at day TrT3, T8-Tιo and T15-T17 (3 cycles), each time at a dose of 5 μM/kg body weight. Mice were sacrificed 25 days post tumor inoculation and tumor load was assessed.
Four mice were included in each experiment and the results are expressed as means of lungs' weight ±SD (Figure 20). An additional experiment depicted in Figures 21 A, 2 IB and 22 also demonstrated the anti-tumor effect of H202. The experimental protocol in this experiment was similar to that outlined in Figure 20. Six mice were included in each group and the results are expressed as the means of lungs' weight ±SD (Figures 21A-B). Photographs of the lungs are shown in Figure 22.
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims. All publications, patents and patent applications mentioned in this specification are herein incorporated in their entirety by reference into the specification, to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated herein by reference. In addition, citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the present invention. REFERENCES CITED (Additional references are cited in the text)
1. Margolin KA. Interleukin-2 in the treatment of renal cancer. Semin Oncol 27:194-203, 2000.
2. Porter AG. The prospects for therapy with tumour necrosis factors and their antagonists. Trends Biotechnol 9:158-62, 1991.
3. Queirolo P, Ponte M, Gipponi M, et al. Adoptive immunotherapy with tumor-infiltrating lymphocytes and subcutaneous recombinant interleukin-2 plus interferon alfa-2a for melanoma patients with nonresectable distant disease: a phase I/II pilot trial. Melanoma Istituto Scientifico Tumori Group. Ann Surg Oncol 6:272-8, 1999.
4. Keilholz U, Stoter G, Punt CJ, et al. Recombinant interluekin-2 -based treatments for advanced melanoma: the experience of the European Organization for Research and Treatment of Cancer Melanoma Cooperative Group. Cancer J Sci Am., Suppl 1 S22-8, 1997.
5. Dutcher JP, Atkins M, Fisher R, et al. Interleukin-2-based therapy for metastatic renal cell cancer: the Cytokine Working Group experience, 1989-1997. Cancer J Sci Am., Suppl 1 S73-8, 1997.
6. Hank JA, Albertini M, Wesly OH, et al. Clinical and immunological effects of treatment with murine anti-CD3 monoclonal antibody along with interieukin 2 in patients with cancer. Clin Cancer Res 1: 481-91, 1995.
7. Glennie MJ, Johnson PW. Clinical trials of antibody therapy. Immunol Today 21 :403-10, 2000.
8. Mungan NA, Witjes JA. Bacille Calmette-Guerin in superficial transitional cell carcinoma. Br J Urol 82:213-23, 1998.
9. Novogrodsky A, Suthanthiran M, Stenzel KH. Immune stimulatory properties of metalloporphyrins. J Immunol 143: 3981-7, 1989. 10. Novogrodsky A, Suthanthiran M, Stenzel KH. Ferro-mitogens: iron-containing compounds with lymphocyte-stimulatory properties. Cell Immunol 133(2) 295-305, 1991.
11. Lander HM, Levine DM, Novogrodsky A. Haemin enhancement of glucose transport in human lymphocytes: stimulation of protein tyrosine phosphatase and activation of p561ck tyrosine kinase. Biochem J 291 : 281-7, 1993.
12. Lander HM, Sehajpal P, Levine DM, Novogrodsky A. Activation of human peripheral blood mononuclear cell by nitric oxide-generating compounds. J Immunol 150:1509-16, 1993.
13. Lander HM Sehajpal PK, Novogrodsky A. Nitric oxide signaling : a possible role for G proteins. J Immunol 151 :7182-7, 1993.
14. Tsuji A, Wang J, Stenzel KH, Novogrodsky A. Immune stimulatory and anti-tumour properties of haemin. Clin Exp Immunol. 93:308-12, 1993.
15. Downward J, Graves JD, Warne PH, et al. Stimulation of p2 Iras upon T-cell activation. Nature 346:719-23, 1990.
16. Izquierdo M, Leevers SJ, Marshall CJ, Cantrell D. P21ras couples the T cell antigen receptor to extracellular signal-regulated kinase 2 in T lymphocytes. J Exp Med 178: 1199-208, 1993.
17. Graves JD, Downward J, Izquierdo-Pastor M, et al. The growth factor IL-2 activates p21ras proteins in normal human T lymphocytes. J Immunol 148:2417-22, 1992.
18. Schneider H, Cai YC, Prasad KV, et al. T cell antigen CD28 binds to the GRB-2/SOS complex, regulators of p21ras. Eur J Immunol 25:1044-50, 1995.
19. Galandrini R, Palmieri G, Piccoli M, et al. CD16-mediated p21ras activation is associated with She and p36 tyrosine phosphorylation and their binding with Grb2 in human natural killer cells. J Exp Med 183:179-86, 1996. 20. Lander HM, Ogiste JS, Pearce SF, Levi R, Novogrodsky A. Nitric oxide-stimulated guanine nucleotide exchange on p21ras. J Biol Chem 270:7017-20, 1995.
21. Lander HM, Ogiste JS, Teng KK, Novogrodsky A. p21ras as a common signaling target of reactive free radical and cellular redox stress. J Biol Chem. 210: 21195-8, 1995.
22. Lander HM, Hajjar DP, Hempstead BL, et al. A molecular redox switch on p21(ras). Structural basis for the nitric oxide-p21(ras) interaction. J Biol Chem 272: 4323-6, 1997.
23. Deora AA, Win T, Vanhaesebroeck B, Lander HM. A redox-triggered ras-effector interaction. Recruitment of phosphatidylinositol 3 '-kinase to Ras by redox stress. JBiol Chem 273: 29923-8, 1998.
24. Deora AA, Hajjar DP, Lander HM. Recruitment and activation of Raf-1 kinase by nitric oxide-activated Ras. Biochemistry 39: 9901-8, 2000.
25. Scannone J, Wellner D, Novogrodsky A. A study of amino acid oxidase specificity using a new assay . Biochemistry 3:1742-5, 1964.
26. Margoliash E, Novogrodsky A. A study of the inhibition of catalase by 3-amino-l,2,4-triazole. Biochem J 68:468-7 '5, 1958.
27. Novogordsky A, Vanichkin A, Patya M, Gazit A, Osherov N, Levitzki A. Prevention of lipopolysaccharide-induced lethal toxicity by tyrosine kinase blockers. Science 264:1319-22, 1994.
28. Lander HM. An essential role for free radicals and derived species in signal transduction. FASEB 11:118-24, 1997.
29. Marshal CJ. Current Opin Cell Biol 8:197-204, 1996.
30. Kopf-Maier P, Kopf H, Neuse EW. Ferrocenium Salts - The first antineoplastic iron compounds. Angew Chem Int Ed Engl 23:456-7, 1984.
31. Kopf-Maier P, Klapotke T. Tumor inhibition by ferricenium complexes. Activity against some solid experimental tumors. Arzneimittelforschung 39:369-371, 1989. 32. Hall IH, Warrant AE, Lee CC, Wasczcak MD, Sneddon LG. Cytotoxicity of ferratricarbadecaboranyl complexes in murine and human tissue cultured cell lines. Anticancer Res 18:951-62, 1998.
33. Gasiorowski K, Brokos JB, Szyba K, Wozniak D, Fraser DM, Zakeeruddin SM, Graetzel M. Evaluation of genotoxic and immunotoxic activities of potential glucose biosensor components: ferrocenes. Biometals 12:19-26, 1999.
34. Hardy B, Yampolski I, Kovjazin R, Galli M, Novogrodsky A. A monoclonal antibody against a human B lymhoblastoid cell line induces tumor regression in mice. Cancer Res 54:5796-6, 1994.
35. Wang X, Martindale JL, Liu Y, Holbrook NJ. The cellular response to oxidative stress: influences of mitogen-activated protein kinase pathways on cell survival. Biochem J. 333:291-300, 1998.
36. Preston TJ, Muller WJ, Singh G. Scavenging of extracellular H202 by catalase inhibits the proliferation of HER-2/Neu-transformed rat-1 fibroblasts though the induction of a stress response. J Biol Chem 276: 9558-64, 2001.
37. Lakshminarayanan, V, Drab- Weiss EA, Roebuck KA. H202 and tumor necrosis-alpha induce differential binding of redox-responsive transcription factors AP-1 and NF-kappa B to the interieukin- 8 promotor in endothelial and epithelial cells. JBiol Chem 273:32670-8, 1998.
38. Hehner SP, Brietkreutz R, Shubinsky G, Unsoeld H, Schulze-Osthoff K, Schmitz ML, Droge W. Enhancement of T cell receptor signaling by a mild oxidative shift in the intracellular thiol pool. J Immunol 165:4319-28, 2000.
39. Tsunawaki S, Nathan CF. Enzymatic basis of macrophage activation. J Biol Chem 259:4305-12, 1984.

Claims

WHAT IS CLAIMED IS:
1. A method of determining whether a metallocene derivative is an effective immunostimulant, the method comprising determining an immune stimulation dose-response curve for the metallocene derivative and determining whether said dose-response curve has an optimum with respect to immune stimulation within a predetermined range of concentrations.
2. The method of claim 1, wherein said immune stimulation is determined in vivo.
3. The method of claim 1, wherein said immune stimulation is determined in vitro.
4. The method of claim 1, wherein said immune stimulation is determined with respect to macrophages.
5. The method of claim 1, wherein said immune stimulation is determined with respect to lymphocytes.
6. The method of claim 1, wherein said immune stimulation is determined by a cell proliferation assay.
7. The method of claim 6, wherein said cell proliferation assay includes a thymidine incorporation assay.
8. The method of claim 1, wherein said immune stimulation is determined by a cellular response assay.
9. The method of claim 8, wherein said cell response assay is selected from the group consisting of an NO production assay, a TNF-α production assay and an oxygen burst assay.
10. A method of determining whether a metallocene derivative is an effective immunostimulant, the method comprising determining a p21ras signal transduction pathway component activity dose-response curve for the metallocene derivative and determining whether said dose-response curve has an optimum with respect to p21ras signal transduction pathway component activity within a predetermined range of concentrations.
11. The method of claim 10, wherein said p21ras signal transduction pathway component is selected from the group consisting of p21ras and F-κB.
12. A method of determining whether a metallocene derivative is an effective immunostimulant, the method comprising determining p21ras oxidation dose-response curve for the metallocene derivative and determining whether said dose-response curve has an optimum with respect to p21ras oxidation within a predetermined range of concentrations.
13. A method of determining whether a metallocene derivative is an effective immunostimulant, the method comprising determining a GTPase activity dose-response curve for the metallocene derivative and determining whether said dose-response curve has an optimum with respect to said GTPase activity within a predetermined range of concentrations.
14. A method of determining whether a metallocene derivative is an effective immunostimulant, the method comprising determining an NF-κB activity dose-response curve for the metallocene derivative and determining whether said dose-response curve has an optimum with respect to said NF-κB activity within a predetermined range of concentrations.
15. A method of determining whether a metallocene derivative is an effective immunostimulant, the method comprising determining H202 generation catalyzed by the metallocene derivative, and determining whether the rate is within a predetermined range.
16. A method of determining whether a metallocene derivative is an effective immunostimulant, the method comprising monitoring H20 generation catalyzed by the metallocene derivative, and determining whether said generation is above a predetermined value.
17. A method of determining whether a metallocene derivative is a candidate for cancer therapy, the method comprising determining a cancer therapy dose-response curve for the metallocene derivative and determining whether said dose-response curve has an optimum with respect to said cancer therapy within a predetermined range of concentrations.
18. The method of claim 17, wherein said cancer therapy is determined in vivo.
19. The method of claim 17, wherein said cancer therapy is determined in vitro.
20. The method of claim 17, wherein said cancer therapy is determined in a test animal inoculated with cancer cells.
21. A method of determining whether a metallocene derivative is a candidate for cancer therapy, the method comprising determining a p21ras signal transduction pathway component activity dose-response curve for the metallocene derivative and determining whether said dose-response curve has an optimum with respect to p21ras signal transduction pathway component activity within a predetermined range of concentrations.
22. The method of claim 21, wherein said p21ras signal transduction pathway component is selected from the group consisting of p21ras and NF-κB.
23. A method of determining whether a metallocene derivative is a candidate for cancer therapy, the method comprising determining p21ras oxidation dose-response curve for the metallocene derivative and determining whether said dose-response curve has an optimum with respect to p21ras oxidation within a predetermined range of concentrations.
24. A method of determining whether a metallocene derivative is a candidate for cancer therapy, the method comprising determining a GTPase activity dose-response curve for the metallocene derivative and determining whether said dose-response curve has an optimum with respect to said GTPase activity within a predetermined range of concentrations.
25. A method of determining whether a metallocene derivative is a candidate for cancer therapy, the method comprising determining an NF-κB activity dose-response curve for the metallocene derivative and determining whether said dose-response curve has an optimum with respect to said NF-κB activity within a predetermined range of concentrations.
26. A method of determining whether a metallocene derivative is a candidate for cancer therapy, the method comprising determining determining H202 generation catalyzed by the metallocene derivative, and determining whether the rate is within a predetermined range.
27. A method of determining whether a metallocene derivative is a candidate for cancer therapy, the method comprising monitoring H202 generation catalyzed by the metallocene derivative, and determining whether said generation is above a predetermined value under predetermined experimental conditions.
28. A method of treating a disease, syndrome or condition, in which immune stimulation is beneficial, the method comprising administering to a subject in need thereof at least one agent in an amount so as to induce H202 generation at a concentration effective in inducing immune stimulation.
29. The method of claim 28, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
30. The method of claim 29, wherein said amount is between 0.025 - 10 mg/kg body weight.
31. The method of claim 29, wherein said amount is between 0.1 - 5 mg/kg body weight.
32. The method of claim 29, wherein said amount is between 0.2 - 3 mg/kg body weight.
33. The method of claim 29, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
34. The method of claim 29, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
35. The method of claim 29, wherein said disease, syndrome or condition is selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency.
36. The method of claim 29, wherein said metallocene or metallocene derivative has a general formula: wherein,
M is a metal selected from the group consisting of Fe (iron), Cr
Figure imgf000054_0001
(chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
Ri-Rg are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
37. The method of claim 29, further comprising co-administering at least one additional agent having immune stimulation activity.
38. The method of claim 37, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
39. A method of treating a disease, syndrome or condition, in which immune stimulation is beneficial, the method comprising administering to a subject in need thereof at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight.
40. The method of claim 39, wherein said amount is between 0.1 - 5 mg/kg body weight.
41. The method of claim 39, wherein said amount is between 0.2 - 3 mg/kg body weight.
42. The method of claim 39, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
43. The method of claim 39, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
44. The method of claim 39, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
45. The method of claim 39, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000055_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
Ri-Rg are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
46. The method of claim 39, further comprising co-administering at least one additional agent having immune stimulation activity.
47. The method of claim 46, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
48. A method of treating a disease, syndrome or condition, in which immune stimulation is beneficial, the method comprising administering to a subject in need thereof at least one agent in an amount effective in activating a p21ras signal transduction pathway component.
49. The method of claim 48, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
50. The method of claim 49, wherein said p21ras signal transduction pathway component is selected from the group consisting of p21ras andNF-κB.
51. The method of claim 49, wherein said amount is between 0.1 - 5 mg/kg body weight.
52. The method of claim 49, wherein said amount is between 0.2 - 3 mg/kg body weight.
53. The method of claim 49, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
54. The method of claim 49, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
55. The method of claim 49, wherein said disease, syndrome or condition is selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency.
56. The method of claim 49, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000057_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); andR R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
57. The method of claim 49, further comprising co-administering at least one additional agent having immune stimulation activity.
58. The method of claim 57, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
59. A method of treating a disease, syndrome or condition, in which immune stimulation is beneficial, the method comprising administering to a subject in need thereof at least one agent in an amount effective in inducing p21ras oxidation.
60. The method of claim 59, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
61. The method of claim 60, wherein said amount is between 0.1 - 5 mg/kg body weight.
62. The method of claim 60, wherein said amount is between 0.2 - 3 mg/kg body weight.
63. The method of claim 60, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
64. The method of claim 60, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
65. The method of claim 60, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
66. The method of claim 60, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000059_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); andand Ri-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
67. The method of claim 60, further comprising co-administering at least one additional agent having immune stimulation activity.
68. The method of claim 67, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
69. A method of treating a disease, syndrome or condition, in which immune stimulation is beneficial, the method comprising administering to a subject in need thereof at least one agent in an amount effective in inducing GTPase activity.
70. The method of claim 69, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
71. The method of claim 70, wherein said amount is between 0.1 - 5 mg/kg body weight.
72. The method of claim 70, wherein said amount is between 0.2 - 3 mg/kg body weight.
73. The method of claim 70, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
74. The method of claim 70, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
75. The method of claim 70, wherein said disease, syndrome or condition is selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency.
76. The method of claim 70, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000061_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
Rι-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
77. The method of claim 70, further comprising co-administering at least one additional agent having immune stimulation activity.
78. The method of claim 77, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
79. A method of treating a disease, syndrome or condition, in which immune stimulation is beneficial, the method comprising administering to a subject in need thereof at least one agent in an amount effective in inducing NF-κB activity.
80. The method of claim 79, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
81. The method of claim 80, wherein said amount is between 0.1 - 5 mg/kg body weight.
82. The method of claim 80, wherein said amount is between 0.2 - 3 mg/kg body weight.
83. The method of claim 80, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
84. The method of claim 80, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
85. The method of claim 80, wherein said disease, syndrome or condition is selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency.
86. The method of claim 80, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000062_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) andNb ( niobium); and
Rι-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
87. The method of claim 80, further comprising co-administering at least one additional agent having immune stimulation activity.
88. The method of claim 87, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
89. A method of treating a cancer, the method comprising administering to a subject in need thereof at least one agent in an amount so as to induce H202 generation at a concentration effective in inducing immune stimulation.
90. The method of claim 89, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
91. The method of claim 90, wherein said amount is between 0.025 - 10 mg/kg body weight.
92. The method of claim 90, wherein said amount is between 0.1 - 5 mg/kg body weight.
93. The method of claim 90, wherein said amount is between 0.2 - 3 mg/kg body weight.
94. The method of claim 90, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
95. The method of claim 90, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
96. The method of claim 90, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
97. The method of claim 90, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000064_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); andand, R R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
98. The method of claim 90, further comprising co-administering at least one additional agent having anti-cancer activity.
99. The method of claim 98, wherein said at least one additional agent is an immunostimulant.
100. A method of treating a cancer, the method comprising administering to a subject in need thereof at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight.
101. The method of claim 100, wherein said amount is between 0.1 - 5 mg/kg body weight.
102. The method of claim 100, wherein said amount is between 0.2 - 3 mg/kg body weight.
103. The method of claim 100, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
104. The method of claim 100, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
105. The method of claim 100, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
106. The method of claim 100, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000066_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
Rι.-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
107. The method of claim 100, further comprising co-administering at least one additional agent having anti-cancer activity.
108. The method of claim 107, wherein said at least one additional agent is an immunostimulant.
109. A method of treating a cancer, the method comprising administering to a subject in need thereof at least one agent in an amount effective in activating a p21ras signal transduction pathway component.
110. The method of claim 109, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
111. The method of claim 110, wherein said p21ras signal transduction pathway component is selected from the group consisting of p21ras and NF-κB.
112. The method of claim 110, wherein said amount is between 0.1 - 5 mg/kg body weight.
113. The method of claim 110, wherein said amount is between 0.2 - 3 mg/kg body weight.
114. The method of claim 110, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
115. The method of claim 110, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
116. The method of claim 110, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
117. The method of claim 110, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000067_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and Rι-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
118. The method of claim 110, further comprising co-administering at least one additional agent having anti-cancer activity.
119. The method of claim 118, wherein said at least one additional agent is an immunostimulant.
120. A method of treating a cancer, the method comprising administering to a subject in need thereof at least one agent in an amount effective in inducing p21ras oxidation.
121. The method of claim 120, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
122. The method of claim 121, wherein said amount is between 0.1 - 5 mg/kg body weight.
123. The method of claim 121, wherein said amount is between 0.2 - 3 mg/kg body weight.
124. The method of claim 121, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
125. The method of claim 121, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
126. The method of claim 121, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
127. The method of claim 121, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000069_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
R]-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
128. The method of claim 121, further comprising co-administering at least one additional agent having anti-cancer activity.
129. The method of claim 128, wherein said at least one additional agent is an immunostimulant.
130. A method of treating a cancer, the method comprising administering to a subject in need thereof at least one agent in an amount effective in inducing GTPase activity.
131. The method of claim 130, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
132. The method of claim 131, wherein said amount is between 0.1 - 5 mg/kg body weight.
133. The method of claim 131, wherein said amount is between 0.2 - 3 mg/kg body weight.
134. The method of claim 131, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
135. The method of claim 131, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
136. The method of claim 131, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
137. The method of claim 131, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000070_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
R R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
138. The method of claim 131, further comprising co-administering at least one additional agent having anti-cancer activity.
139. The method of claim 138, wherein said at least one additional agent is an immunostimulant.
140. A method of treating a cancer, the method comprising administering to a subject in need thereof at least one agent in an amount effective in inducing NF-κB activity.
141. The method of claim 140, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
142. The method of claim 141, wherein said amount is between 0.1 - 5 mg/kg body weight.
143. The method of claim 141, wherein said amount is between 0.2 - 3 mg/kg body weight.
144. The method of claim 141, wherein administering said at least one ferrocene and/or metallocene derivative is by oral administration.
145. The method of claim 141, wherein administering said at least one ferrocene and/or metallocene derivative is by injection.
146. The method of claim 141, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
147. The method of claim 141, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000072_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) andNb ( niobium); and
Rι-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
148. The method of claim 141, further comprising co-administering at least one additional agent having anti-cancer activity.
149. The method of claim 148, wherein said at least one additional agent is an immunostimulant.
150. A method of inducing immune stimulation in a body cavity, the method comprising administering into the body cavity H202 in an amount effective in immune stimulation.
151. The method of claim 150, wherein said amount is of 10-2000 ml of 50-2500 μM H202.
152. The method of claim 150, wherein said body cavity is selected from the group consisting of peritoneal and pleural cavities.
153. A method of treating cancer in a body cavity, the method comprising administering into the body cavity H202 in an amount effective in immune stimulation.
154. The method of claim 153, wherein said amount is of 10-2000 ml of 50-2500 μM H202.
155. The method of claim 153, wherein said body cavity is selected from the group consisting of peritoneal and pleural cavities.
156. A method of inducing immune stimulation, the method comprising exposing immune cells ex vivo to at least one ferrocene and/or metallocene derivative, and administering said cells to a subject in need of immune stimulation.
157. A method of treating cancer, the method comprising exposing immune cells ex vivo to at least one ferrocene and/or metallocene derivative, and administering said cells to a subject in need of such treatment.
158. A method of inducing immune stimulation, the method comprising exposing immune cells ex vivo to H202 in a concentration effective in inducing immune stimulation and administering said cells to a subject in need of immune stimulation.
159. A method of treating cancer, the method comprising exposing immune cells ex vivo to H2θ2 in a concentration effective in inducing immune stimulation and administering said cells to a subject in need of such treatment.
160. A pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount so as to induce H202 generation at a concentration effective in inducing immune stimulation following administration thereof.
161. The pharmaceutical composition of claim 160, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
162. The pharmaceutical composition of claim 161, wherein said amount is between 0.025 - 10 mg/kg body weight.
163. The pharmaceutical composition of claim 161, wherein said amount is between 0.1 - 5 mg/kg body weight.
164. The pharmaceutical composition of claim 161, wherein said amount is between 0.2 - 3 mg/kg body weight.
165. The pharmaceutical composition of claim 161, wherein said pharmaceutical composition is formulated for oral administration.
166. The pharmaceutical composition of claim 161, wherein said pharmaceutical composition is formulated for administration by injection
167. The pharmaceutical composition of claim 161, packaged and identified for treatment of a disease, syndrome or condition selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency.
168. The pharmaceutical composition of claim 161, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000075_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
Rj-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
169. The pharmaceutical composition of claim 161, further comprising at least one additional agent having immune stimulation activity.
170. The pharmaceutical composition of claim 169, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
171. A pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight.
172. The pharmaceutical composition of claim 171, wherein said amount is between 0.1 - 5 mg/kg body weight.
173. The pharmaceutical composition of claim 171, wherein said amount is between 0.2 - 3 mg/kg body weight.
174. The pharmaceutical composition of claim 171, wherein said pharmaceutical composition is formulated for oral administration.
175. The pharmaceutical composition of claim 171, wherein said pharmaceutical composition is formulated for administration by injection
176. The pharmaceutical composition of claim 171, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
177. The pharmaceutical composition of claim 171, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000076_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and b ( niobium); and
Ri-Rg are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
178. The pharmaceutical composition of claim 171, further comprising at least one additional agent having immune stimulation activity.
179. The pharmaceutical composition of claim 178, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
180. A pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in activating a p21ras signal transduction pathway component following administration thereof.
181. The pharmaceutical composition of claim 180, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
182. The pharmaceutical composition of claim 181, wherein said p21ras signal transduction pathway component is selected from the group consisting of p21ras and NF-KB.
183. The pharmaceutical composition of claim 181, wherein said amount is between 0.1 - 5 mg/kg body weight.
184. The pharmaceutical composition of claim 181, wherein said amount is between 0.2 - 3 mg/kg body weight.
185. The pharmaceutical composition of claim 181, wherein said pharmaceutical composition is formulated for oral administration.
186. The pharmaceutical composition of claim 181, wherein said pharmaceutical composition is formulated for administration by injection
187. The pharmaceutical composition of claim 181, packaged and identified for treatment of a disease, syndrome or condition selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency.
188. The pharmaceutical composition of claim 181, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000078_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and Rι-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
189. The pharmaceutical composition of claim 181, further comprising at least one additional agent having immune stimulation activity.
190. The pharmaceutical composition of claim 189, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
191. A pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing p21ras oxidation following administration thereof.
192. The pharmaceutical composition of claim 191, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
193. The pharmaceutical composition of claim 192, wherein said amount is between 0.1 - 5 mg/kg body weight.
194. The pharmaceutical composition of claim 192, wherein said amount is between 0.2 - 3 mg/kg body weight.
195. The pharmaceutical composition of claim 192, wherein said pharmaceutical composition is formulated for oral administration.
196. The pharmaceutical composition of claim 192, wherein said pharmaceutical composition is formulated for administration by injection
197. The pharmaceutical composition of claim 192, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
198. The pharmaceutical composition of claim 192, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000080_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
R R are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
199. The pharmaceutical composition of claim 192, further comprising at least one additional agent having immune stimulation activity.
200. The pharmaceutical composition of claim 199, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
201. A pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing GTPase activity following administration thereof.
202. The pharmaceutical composition of claim 201, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
203. The pharmaceutical composition of claim 202, wherein said amount is between 0.1 - 5 mg/kg body weight.
204. The pharmaceutical composition of claim 202, wherein said amount is between 0.2 - 3 mg/kg body weight.
205. The pharmaceutical composition of claim 202, wherein said pharmaceutical composition is formulated for oral administration.
206. The pharmaceutical composition of claim 202, wherein said pharmaceutical composition is formulated for administration by injection
207. The pharmaceutical composition of claim 202, packaged and identified for treatment of a disease, syndrome or condition selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency.
208. The pharmaceutical composition of claim 202, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000082_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
RrR8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
209. The pharmaceutical composition of claim 202, further comprising at least one additional agent having immune stimulation activity.
210. The pharmaceutical composition of claim 209, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
211. A pharmaceutical composition for treating a disease, syndrome or condition, in which immune stimulation is beneficial, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing NF-κB activity following administration thereof.
212. The pharmaceutical composition of claim 211, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
213. The pharmaceutical composition of claim 212, wherein said amount is between 0.1 - 5 mg/kg body weight.
214. The pharmaceutical composition of claim 212, wherein said amount is between 0.2 - 3 mg/kg body weight.
215. The pharmaceutical composition of claim 212, wherein said pharmaceutical composition is formulated for oral administration.
216. The pharmaceutical composition of claim 212, wherein said pharmaceutical composition is formulated for administration by injection
217. The pharmaceutical composition of claim 212, packaged and identified for treatment of a disease, syndrome or condition selected from the group consisting of cancer, acquired immune deficiency, radiotherapy induced immune deficiency, chemotherapy induced immune deficiency and congenital immune deficiency.
218. The pharmaceutical composition of claim 212, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000083_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb j( niobium); and
Ri-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
219. The pharmaceutical composition of claim 212, further comprising at least one additional agent having immune stimulation activity.
220. The pharmaceutical composition of claim 219, wherein said at least one additional agent is selected from the group consisting of a cytokine, a chemokine, an immune-stimulatory antibody and an anti-timor vaccine.
221. A pharmaceutical composition for treating a cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount so as to induce H202 generation at a concentration effective in inducing immune stimulation following administration thereof.
222. The pharmaceutical composition of claim 221, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
223. The pharmaceutical composition of claim 222, wherein said amount is between 0.025 - 10 mg/kg body weight.
224. The pharmaceutical composition of claim 222, wherein said amount is between 0.1 - 5 mg/kg body weight.
225. The pharmaceutical composition of claim 222, wherein said amount is between 0.2 - 3 mg/kg body weight.
226. The pharmaceutical composition of claim 222, wherein said pharmaceutical composition is formulated for oral administration.
227. The pharmaceutical composition of claim 222, wherein said pharmaceutical composition is formulated for administration by injection
228. The pharmaceutical composition of claim 222, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
229. The pharmaceutical composition of claim 222, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000085_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
Rι.-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
230. The pharmaceutical composition of claim 222, further comprising co-administering at least one additional agent having anti-cancer activity.
231. The pharmaceutical composition of claim 230, wherein said at least one additional agent is an immunostimulant.
232. A pharmaceutical composition for treating a cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one ferrocene and/or metallocene derivative, in an amount of 0.025 - 10 mg/kg body weight.
233. The pharmaceutical composition of claim 232, wherein said amount is between 0.1 - 5 mg/kg body weight.
234. The pharmaceutical composition of claim 232, wherein said amount is between 0.2 - 3 mg/kg body weight.
235. The pharmaceutical composition of claim 232, wherein said pharmaceutical composition is formulated for oral administration.
236. The pharmaceutical composition of claim 232, wherein said pharmaceutical composition is formulated for administration by injection
237. The pharmaceutical composition of claim 232, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
238. The pharmaceutical composition of claim 232, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000087_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
R R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
239. The pharmaceutical composition of claim 232, further comprising co-administering at least one additional agent having anti-cancer activity.
240. The pharmaceutical composition of claim 239, wherein said at least one additional agent is an immunostimulant.
241. A pharmaceutical composition for treating a cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in activating a p21ras signal transduction pathway component following administration thereof.
242. The pharmaceutical composition of claim 241, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
243. The pharmaceutical composition of claim 242, wherein said p21ras signal transduction pathway component is selected from the group consisting of p21ras and NF-KB.
244. The pharmaceutical composition of claim 242, wherein said amount is between 0.1 - 5 mg/kg body weight.
245. The pharmaceutical composition of claim 242, wherein said amount is between 0.2 - 3 mg/kg body weight.
246. The pharmaceutical composition of claim 242, wherein said pharmaceutical composition is formulated for oral administration.
247. The pharmaceutical composition of claim 242, wherein said pharmaceutical composition is formulated for administration by injection
248. The pharmaceutical composition of claim 242, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
249. The pharmaceutical composition of claim 242, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000089_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) andNb ( niobium); and
RrR8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
250. The pharmaceutical composition of claim 242, further comprising co-administering at least one additional agent having anti-cancer activity.
251. The pharmaceutical composition of claim 250, wherein said at least one additional agent is an immunostimulant.
252. A pharmaceutical composition for treating a cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing p21ras oxidation following administration thereof.
253. The pharmaceutical composition of claim 252, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
254. The pharmaceutical composition of claim 253, wherein said amount is between 0.1 - 5 mg/kg body weight.
255. The pharmaceutical composition of claim 253, wherein said amount is between 0.2 - 3 mg/kg body weight.
256. The pharmaceutical composition of claim 253, wherein said pharmaceutical composition is formulated for oral administration.
257. The pharmaceutical composition of claim 253, wherein said pharmaceutical composition is formulated for administration by injection
258. The pharmaceutical composition of claim 253, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
259. The pharmaceutical composition of claim 253, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000090_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
R R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
260. The pharmaceutical composition of claim 253, further comprising co-administering at least one additional agent having anti-cancer activity.
261. The pharmaceutical composition of claim 260, wherein said at least one additional agent is an immunostimulant.
262. A pharmaceutical composition for treating a cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing GTPase activity following administration thereof.
263. The pharmaceutical composition of claim 262, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
264. The pharmaceutical composition of claim 263, wherein said amount is between 0.1 - 5 mg/kg body weight.
265. The pharmaceutical composition of claim 263, wherein said amount is between 0.2 - 3 mg/kg body weight.
266. The pharmaceutical composition of claim 263, wherein said pharmaceutical composition is formulated for oral administration.
267. The pharmaceutical composition of claim 263, wherein said pharmaceutical composition is formulated for administration by injection
268. The pharmaceutical composition of claim 263, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
269. The pharmaceutical composition of claim 263, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000092_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
Rι-R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
270. The pharmaceutical composition of claim 263, further comprising co-administering at least one additional agent having anti-cancer activity.
271. The pharmaceutical composition of claim 270, wherein said at least one additional agent is an immunostimulant.
272. A pharmaceutical composition for treating a cancer, each dose-unit of the pharmaceutical composition comprising, as an active ingredient, at least one agent in an amount effective in inducing NF-κB activity following administration thereof.
273. The pharmaceutical composition of claim 272, wherein said at least one agent comprises at least one ferrocene and/or metallocene derivative.
274. The pharmaceutical composition of claim 273, wherein said amount is between 0.1 - 5 mg/kg body weight.
275. The pharmaceutical composition of claim 273, wherein said amount is between 0.2 - 3 mg/kg body weight.
276. The pharmaceutical composition of claim 273, wherein said pharmaceutical composition is formulated for oral administration.
277. The pharmaceutical composition of claim 273, wherein said pharmaceutical composition is formulated for administration by injection
278. The pharmaceutical composition of claim 273, wherein said cancer is selected from the group consisting of a carcinoma, a sarcoma, a lymphoma and a leukemia.
279. The pharmaceutical composition of claim 273, wherein said metallocene or metallocene derivative has a general formula:
Figure imgf000093_0001
wherein,
M is a metal selected from the group consisting of Fe (iron), Cr (chromium), V (vanadium), Mn (manganese), Co (cobalt), Zn (zinc), Ni (nickel), Ti (titanium), Zr (zirconium), Hf (hafnium) and Nb ( niobium); and
R R8 are each independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, alkoxy, aryl and aryloxy.
280. The pharmaceutical composition of claim 273, further comprising co-administering at least one additional agent having anti-cancer activity.
281. The pharmaceutical composition of claim 280, wherein said at least one additional agent is an immunostimulant.
PCT/IL2002/000803 2001-10-17 2002-10-02 Methods and pharmaceutical compositions for stimulating the immune system and/or treating cancer WO2003032895A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32954801P 2001-10-17 2001-10-17
US60/329,548 2001-10-17

Publications (2)

Publication Number Publication Date
WO2003032895A2 true WO2003032895A2 (en) 2003-04-24
WO2003032895A3 WO2003032895A3 (en) 2005-12-08

Family

ID=23285920

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2002/000803 WO2003032895A2 (en) 2001-10-17 2002-10-02 Methods and pharmaceutical compositions for stimulating the immune system and/or treating cancer

Country Status (1)

Country Link
WO (1) WO2003032895A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102008004100A1 (en) * 2008-01-11 2009-07-16 Westfälische Wilhelms-Universität Münster Körperschaft des öffentlichen Rechts New metallocene compounds are estrogen receptor modulators useful e.g. to treat preferably cancer, manic disorder, gynecomastia and/or female infertility, and to prevent mamma carcinoma

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2839552A (en) * 1955-08-08 1958-06-17 Ethyl Corp Cyclomatic manganese compounds
US3960911A (en) * 1973-11-21 1976-06-01 Frisons Limited Ferrocene compounds and preparation
US5167948A (en) * 1987-08-07 1992-12-01 Mallinckrodt Medical, Inc. Diagnostic or radiotherapeutic composition comprising a hydrogen containing compound

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2839552A (en) * 1955-08-08 1958-06-17 Ethyl Corp Cyclomatic manganese compounds
US3960911A (en) * 1973-11-21 1976-06-01 Frisons Limited Ferrocene compounds and preparation
US5167948A (en) * 1987-08-07 1992-12-01 Mallinckrodt Medical, Inc. Diagnostic or radiotherapeutic composition comprising a hydrogen containing compound

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102008004100A1 (en) * 2008-01-11 2009-07-16 Westfälische Wilhelms-Universität Münster Körperschaft des öffentlichen Rechts New metallocene compounds are estrogen receptor modulators useful e.g. to treat preferably cancer, manic disorder, gynecomastia and/or female infertility, and to prevent mamma carcinoma

Also Published As

Publication number Publication date
WO2003032895A3 (en) 2005-12-08

Similar Documents

Publication Publication Date Title
Clark et al. The clinical pharmacology of etoposide and teniposide
JP4881536B2 (en) Cytotoxic factors that regulate cell death
Adamson Antitumor activity of tilorone hydrochloride against some rodent tumors: preliminary report
Kovjazin et al. Ferrocene‐induced lymphocyte activation and antitumor activity is mediated by redox‐sensitive signaling
JP2002511878A (en) Enhanced treatment of hyperproliferative conditions with antioxidants
JPH02108636A (en) Anti-tumor combination agent
US9226946B2 (en) Pharmaceutical combination for the treatment and/or chemosensibilization of refractory tumors to anticancer drugs
US10729669B2 (en) Compositions and methods for treating cancer
P Popolin et al. A review of ruthenium complexes activities on breast cancer cells
WO1995003051A1 (en) Use of quinoline-3-carboxamide compounds for inhibiting the production of tumour necrosis factor (tnf) and/or for the treatment of septic shock
EP2474528B1 (en) Antineoplasic compounds and pharmaceutical compositions thereof
US9399644B2 (en) [1,3] dioxolo [4,5-G] quinoline-6(5H)thione derivatives as inhibitors of the late SV40 factor (LSF) for use in treating cancer
US6020365A (en) Arglabin compounds and therapeutic uses thereof
WO2003032895A2 (en) Methods and pharmaceutical compositions for stimulating the immune system and/or treating cancer
AU7141898A (en) Pharmaceutical compositions of arglabin and arglabin derivatives
JP3002144B2 (en) Pharmaceutical composition for promoting cancer cell differentiation and method for producing the same
US9657044B2 (en) Method for antagonizing STAT3 dimerization and compounds for use therein
US20050004091A1 (en) Biologically active complex
CN110693903B (en) Medicine for treating acute monocytic leukemia and application of arsenic trioxide and dihydroartemisinin
CN104606189B (en) A kind of application of compound in mTOR inhibitors are prepared
WO2024060520A1 (en) Medicament for preventing and treating tumor and use thereof
FI91753C (en) Process for the preparation of a therapeutically useful complex containing tellurium
US6693127B1 (en) Pharmaceutical compositions of arglabin and arglabin derivatives
CN107929717B (en) Pharmaceutical composition of siramesine and snake venom cytotoxin-CTX 1
US20040131701A1 (en) Method to treat infectious diseases and/or to enhance antimicrobial efficacy of drugs

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GM HR HU ID IL IN IS JP KE KG KP KZ LC LK LR LS LT LU LV MA MD MK MN MW MX MZ NO NZ OM PH PT RO RU SD SE SG SI SK SL TJ TM TN TR TZ UA UG US UZ VC VN YU ZA ZM

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ UG ZM ZW AM AZ BY KG KZ RU TJ TM AT BE BG CH CY CZ DK EE ES FI FR GB GR IE IT LU MC PT SE SK TR BF BJ CF CG CI GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP