US20050053664A1 - Co-administration of a polysaccharide with a chemotherapeutic agent for the treatment of cancer - Google Patents

Co-administration of a polysaccharide with a chemotherapeutic agent for the treatment of cancer Download PDF

Info

Publication number
US20050053664A1
US20050053664A1 US10/657,508 US65750803A US2005053664A1 US 20050053664 A1 US20050053664 A1 US 20050053664A1 US 65750803 A US65750803 A US 65750803A US 2005053664 A1 US2005053664 A1 US 2005053664A1
Authority
US
United States
Prior art keywords
linked
galactomannan
cancer
chemotherapeutic agent
dose
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/657,508
Other languages
English (en)
Inventor
Eliezer Zomer
David Platt
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pro Pharmaceuticals Inc
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US10/657,508 priority Critical patent/US20050053664A1/en
Assigned to PRO-PHARMACEUTICALS, INC. reassignment PRO-PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PLATT, DAVID, ZOMER, ELI
Priority to PCT/US2004/028883 priority patent/WO2005025501A2/fr
Priority to EP04783211A priority patent/EP1662874A4/fr
Priority to JP2006525488A priority patent/JP2007505041A/ja
Priority to CNA2004800300176A priority patent/CN1867252A/zh
Priority to AU2004272022A priority patent/AU2004272022B2/en
Priority to BRPI0413410-9A priority patent/BRPI0413410A/pt
Publication of US20050053664A1 publication Critical patent/US20050053664A1/en
Assigned to PRO-PHARMACEUTICALS, INC. reassignment PRO-PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KLYOSOV, ANATOLE
Priority to IL174121A priority patent/IL174121A/en
Priority to US12/036,608 priority patent/US7893252B2/en
Assigned to 10X FUND, LP reassignment 10X FUND, LP SECURITY AGREEMENT Assignors: PRO-PHARMACEUTICALS, INC.
Priority to US12/962,296 priority patent/US20110077217A1/en
Priority to AU2010257400A priority patent/AU2010257400B2/en
Assigned to GALECTIN THERAPEUTICS, INC. reassignment GALECTIN THERAPEUTICS, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: 10X FUND L.P.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/208IL-12
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to compositions and methods for treating disease. Specifically, the instant invention relates to compositions comprising a polysaccharide and pharmaceutical agent, wherein the polysaccharide lowers the toxicity profile of the drug as well as increase its efficacy.
  • chemotherapeutic agents have significant limitations relating to their toxic effect on the patient and the efficacy of a particular dosage to target and kill tumor cells.
  • chemotherapeutic agents kill cancer cells once they begin to undergo division and replication. Cells are killed by disrupting cell division. For example, a chemotherapeutic agent may prevent the formation of new DNA or block some other essential function within a cell. Some chemotherapeutic agents may work by inducing apoptosis, essentially causing the cells to commit suicide by triggering the cells' programmed death process. Although these agents are effective for treating cancer cells that generally grow rapidly through unregulated cell division, they also kill healthy non-cancerous cells as they undergo ordinary cell division. This toxic effect is particularly apparent in fast-growing normal cells, such as bone marrow cells, those in the digestive tract, hair follicles, and reproductive cells.
  • chemotherapy harms healthy tissue
  • the effectiveness of a drug is limited by its dosage levels and treatment frequency such that it should not exceed the tolerance levels for non-cancerous cells.
  • the chemotherapy regimen often dramatically diminishes the quality of a patient's life through its physical and emotional side effects.
  • chemotherapy dosages must be kept within a range (i.e., the therapeutic index) that healthy tissue can tolerate, thus often reducing the optimal effectiveness of chemotherapy on diseased tissue.
  • the present invention comprises compositions and methods for treating disease, such as cancer.
  • the compositions of the present invention comprise one or more polysaccharides together with one or more pharmaceutical agents.
  • the methods of the instant invention comprise the co-administration of one or more pharmacological agents and one or more polysaccharides to a subject in need thereof, wherein the pharmaceutical preparation including the pharmacological agent and polysaccharide has reduced toxicity.
  • the compositions of the present invention has both reduced toxicity and increase efficacy.
  • a method for treating cancer in a subject comprising administering to the subject a mixture of one or more polysaccharides and an effective dose of a chemotherapeutic agent in a pharmaceutically acceptable formulation, wherein the polysaccharide is selected from group consisting of galactomannans, which are available from a number of plant and microbial sources.
  • This pharmaceutical formulation is then administered to a patient in need thereof in an acceptable manner well known to those skilled in the art.
  • a mixture of one or more polysaccharides and one or more chemotherapeutic agents is administered to a subject in need thereof, wherein the mixture comprises a sufficient amount of polysaccharide and chemotherapeutic agent in a ratio suitable for reducing the toxic side-effects in a subject while being effective against a particular pathology being treated, wherein the polysaccharide is selected from group consisting of galactomannans (from Cyamopsis tetragonolobus ), Arabinogalactan (from Larix occidentalis ), Rhamnogalacturonan (from potato), Carrageenan (from Eucheuma Seaweed), and the Locust Bean Gum (from Ceratonia siliqua ).
  • the toxic side-effects being defined as those physiological effects (symptoms) realized by the subject resulting from the administration of the chemotherapeutic agent absent the polysaccharide.
  • a pharmaceutical formulation in another embodiment, includes a mixture of one or more polysaccharides and an effective dose of one or more chemotherapeutic agents in a pharmaceutically acceptable formulation, wherein the polysaccharide is selected from group consisting of galactomannans (from Cyamopsis tetragonolobus ), Arabinogalactan (from Larix occidentalis ), Rhamnogalacturonan (from potato), Carrageenan (from Eucheuma Seaweed), and the Locust Bean Gum (from Ceratonia siliqua ).
  • galactomannans from Cyamopsis tetragonolobus
  • Arabinogalactan from Larix occidentalis
  • Rhamnogalacturonan from potato
  • Carrageenan from Eucheuma Seaweed
  • Locust Bean Gum from Ceratonia siliqua
  • the mixture in the formulation contains an amount of one or more polysaccharides and one or more chemotherapeutic agents in a ratio suitable for reducing any toxic side-effect in the subject.
  • the polysaccharide to chemotherapy ratio could be in the range from 10:1 up to 1:10. With the 50,000 MW modified galactomannan the optimum ratio was in the range from 6:1 to 1:3.
  • the mixture contains an amount of one or more polysaccharides and one or more chemotherapeutic agents in a ratio suitable for enhancing efficacy of chemotherapeutic effect for treating the cancer.
  • the mixture contains an amount of one or more polysaccharides and one or more chemotherapeutic agents in a ratio suitable for effectively treating cancer as well as reducing any potential toxic side-effect(s).
  • a method for treating cancer in a subject in need thereof that includes administrating an a mixture of one or more polysaccharides and an effective dose of one or more chemotherapeutic agents formulated so that the chemotherapeutic agent has enhanced therapeutic efficacy in the presence of the polysaccharide component.
  • Subject refers to an animal including a mammal, such as human, dog, cat, pig, cow, sheep, goat, horse, rat, mouse, and alike.
  • Patient refers to a human subject who has presented in a clinical setting with a particular symptom or symptoms consistent with a pathophysiological process.
  • Polysaccharide refers to polymers comprised primarily of monomers of one or more sugars and substituted sugars.
  • the sugar monomers can be modified in ways well known to those in the art.
  • “Efficacy” for a toxic therapeutic agent refers to the relationship between a minimum effective dose and the accompanying toxic side-effects. Efficacy of an agent is increased if a therapeutic end point can be achieved by administration of a lower dose or a shorter dosage regimen. If toxicity can be decreased, a therapeutic agent can be administered on a longer dosage regimen or even chronically with greater patient compliance and improved quality of life. Further, decreased toxicity of an agent enables the practitioner to increase the dosage to achieve the therapeutic endpoint sooner, or to achieve a higher therapeutic endpoint. “Efficacy” for a non-toxic therapeutic agent relates to improved therapeutic effect for treating a condition.
  • “Pharmaceutically acceptable carrier” refers to any and all solvents, dispersion media, e.g., human albumin or cross-linked gelatin polypeptides, coatings, antibacterial and antifungal agents, isotonic, e.g. sodium chloride or sodium glutamate, and absorption delaying agents, and the like that are physiologically compatible. The use of such media and agents for pharmaceutically active substances is well known in the art.
  • the carrier is suitable for oral, intravenous, intramuscular, subcutaneous, parenteral, spinal or epidural administration, (e.g., by injection or infusion).
  • the active compound can be coated in a material to protect the compound from the action of acids and other natural conditions that can inactivate the compound.
  • Active Compound refers to any and all chemotherapeutic agents used herein for the treatment of cancer, and any and all added agents that increase efficacy of the chemotherapeutic agent and/or decrease toxicity of the chemotherapeutic agent.
  • Parenteral Administration includes administration by bolus injection or infusion, as well as administration by intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
  • Toxic refers to any adverse effect caused by an agent when administered to a subject.
  • Tumor Regression was scored (excluding nonspecific deaths) as “partial” (less than 50 percent of its size at the beginning of the treatment), or “complete” (tumor becomes unpalpable).
  • “Duration of Regression” refers to the interval during which a tumor classified as a partial or complete regression continues to be below 50 percent of its size at first treatment.
  • Evaluation Size refers to the tumor mass selected at one or two mass doubling versus beginning with the initial tumor size at the start of treatment.
  • Time Required for Tumor Mass Doubling is the time to reach the evaluation size; it is used in the calculations of the overall delay in the growth of the median tumor [(T-C)/C ⁇ 100%], where T-C (days) is the difference in the median of times postimplant for tumors of the treated (T) groups to attain an evaluation size compared to the median of the control (C) group.
  • the T-C value is measured excluding non-specific deaths, and any other animal that dies whose tumor failed to attain the evaluation size.
  • Effective Dose is that dose of chemotherapeutic agent required to achieve a predetermined physiological effect, such as tumor size reduction, while not exceeding a patient's tolerance for the agent.
  • Enhanced therapeutic efficacy means that the therapeutic index of chemotherapeutic agent (alone or as a composition comprising one or more polysaccharides and one or more therapeutic agents) exceeds the desired physiological effect, such as tumor size reduction, tumor growth delay, etc., compared to that of a reference chemotherapeutic agent, and/or improving a patient's tolerance for the agent.
  • FIG. 1 is the stereochemical configuration of galactomannan
  • FIG. 2 is a graphical representation of the results of 1H-NMR spectrum of the galactomannan of the invention (from guar gum) and galactomannan from carob (locust bean) gum;
  • FIG. 3 is the Fourier transform of the 1H-NMR results shown in FIG. 2 ;
  • FIG. 4 is a graphical representation of the results of 13C-NMR spectrum of the galactomannan of the invention (from guar gum) and galactomannan from carob gum;
  • FIG. 6 is a schematic representation of the manufacturing and purification process to produce the galactomannan of the present invention.
  • FIG. 7 is a graph summary of the results of the first part of the efficacy study described in Example 3.
  • FIG. 8 is a graph summary of the results of the second part of the efficacy study described in Example 3.
  • FIG. 9 is a graph summary of the results of the efficacy study described in Example 4.
  • FIG. 11 is a graph summary of the results of the three efficacy studies described in Examples 3, 4, and 5.
  • chemotherapeutic agents have significant limitations relating to their toxic effect on the patient and the efficacy of a particular dosage to target and kill tumor cells.
  • chemotherapy harms healthy tissue
  • the effectiveness of a drug is limited by its dosage levels and treatment frequency such that it should not exceed the tolerance levels for non-cancerous cells.
  • the chemotherapy regimen often dramatically diminishes the quality of a patient's life through its physical and emotional side effects.
  • chemotherapy dosages must be kept within a range (i.e., the therapeutic index) that healthy tissue can tolerate, thus reducing the optimal effectiveness of chemotherapy on diseased tissue. Therefore, if the toxicity of chemotherapeutic agents can be reduced, practitioners would be able to increase the dosage of drug without the resultant toxic side effects.
  • 5-FU has been the mainstay for therapy of cancers like colorectal cancer for the last two decades.
  • Colorectal adenocarcinoma is the second leading cause of cancer deaths accounting for 10-12 percent of the total number.
  • the mortality rate in 1990 was 24/100,000 for males and 16/100,000 for females. Although survival from this disease has improved by approximately 8% over the past two decades largely due to earlier detection.
  • Topoisomerase I inhibitors such as irinotecan have been used in patients with advanced colorectal cancer who are chemotherapy naive or who have failed 5-FU regimens.
  • chemotherapy naive patients a Phase II study by the Southern Italy Oncology Group showed an 18% response rate in patients receiving 5-FU plus leucovorin, and a 40% response rate in patients receiving irinotecan given just prior to 5-FU plus leucovorin. Grade 3-4 toxicity was uncommon in both treatment arms.
  • two randomized trials showed that irinotecan improved response rates and survival when compared to best supportive care or continuous infusion 5-FU. Irinotecan produces more severe life-threatening myelosuppression and diarrhea than 5-FU. The overlapping toxicities of these two agents also limits the use of both compounds together at maximal dose intensity.
  • Oxaliplatin is a diaminocyclohexane platinum complex which has been shown to have a single agent response rate of 10% in previously treated colorectal cancer patients. When combined with 5-FU infusions in patients receiving initial therapy, response rates as high as 58% have been observed. Complications of this drug include peripheral neuropathy and oral-pharyngeal dysesthesias. Clinical trials are currently underway to compare oxaliplatin combinations with 5-FU or irinotecan to 5-FU as initial treatments for advanced colorectal cancer.
  • cancer One disease targeted by the present invention is cancer.
  • the types of cancer envisaged to be within the scope of the present invention include, but not limited to, chronic leukemia, breast cancer, sarcoma, ovarian carcinoma, rectal cancer, throat cancer, melanoma, colon cancer, bladder cancer, lung cancer, mammary adenocarcinoma, gastrointestinal cancer, stomach cancer, prostate cancer, pancreatic cancer, and Kaposi's sarcoma.
  • treatment regimes are envisaged to be within the scope of this invention include, but not limited to, anti-depressants, anti-inflammatory agents, gastroenterology drugs (for treating ulcers and associated disorders), anti-psychotic drugs, anti-hyperlipidemic agents, etc., as many therapeutic agents must be administered as a chronic medicine, i.e., on a long-term basis, potential reduction in dosage and improvement in quality of life become significant factors in availability, cost of therapeutic agents, and patient compliance.
  • the effective dose of the polysaccharide component is that amount needed to minimize or completely mitigate some, if not all, of the toxic side-effects experienced by a subject receiving a chemotherapeutic agent. In one aspect, the effective dose is that amount needed to enhance the efficacy of the therapeutic agent. In another aspect, the effective dose is that amount need to minimize or eliminate the toxic sides effects of a drug as well as increase the drug's efficacy in treating a subject.
  • Enhanced therapeutic efficacy means that the therapeutic index of chemotherapeutic agent (as a composition comprising one or more polysaccharides and exceeds the desired physiological effect, such as tumor size reduction, tumor growth delay, etc., compared to that of a reference chemotherapeutic agent, or/and improving a patient's tolerance for the chemotherapeutic agent.
  • a composition comprising one or more polysaccharides and one or more therapeutic agents.
  • the polysaccharides are formed from monomeric units. Modified polysaccharides are also considered to be within the scope of the present invention and can include modification by, for example, limited controlled depolymerization, or, for example, having lipid, protein, or nucleic acid moieties affixed to the monomeric units of the polysaccharide.
  • the polysaccharide of the composition can be branched or unbranched.
  • the polysaccharides include, but not limited to, galactomannans available from a number of plant and microbial sources.
  • the polysaccharide can be ⁇ -1,4-D-galactomannan and include a ratio of mannose lose in the range of about 1.7.
  • the molecular weight of the galactomannan polysaccharide is in the range of about 4,000 to about 200,000 D. In a particular aspect, the galactomannan has an average weight of about 40,000 to 60,000 D.
  • the structure of the galactomannans is a poly- ⁇ -1,4 mannan backbone, with side substituents affixed via ⁇ -1-6-glycoside linkages. In one aspect, the galactomannan polysaccharide can be ⁇ -1,4-D-galactomannan.
  • the polysaccharide is (((1,4)-linked ⁇ -D-mannopyranose)17-((1,6)-linked- ⁇ -D-galactopyranose)10)12).
  • the galactomannan can be a derivative of Guar gum from seeds of Cyamopsis tetragonoloba.
  • the polysaccharides of the present invention can have side branches of target specific carbohydrates, such as, galactose, rhamnose, mannose, or arabinose which provides the surface of a polymer recognition capabilities in targeting specific lectin type receptors on the surface of cells, especially tumor cells.
  • Branches can be a single unit or two or more units of oligosaccharide.
  • compositions of the present invention can further include an enhancer.
  • an enhancer is leucovorin.
  • the therapeutic agent of the present composition includes all known pharmaceuticals listed in, for example, the Physicians Desk Reference, as well as experimental therapeutic agents.
  • the therapeutic agents are chemotherapeutic agents.
  • dosage intensity has been employed to define the amount of drug delivered per unit time, usually in milligrams per square meter per week.
  • dose intensity delivered directly relates to treatment outcome. In most tumors in which cure is possible, this issue becomes critical and less than optimal dosing may result in treatment failure.
  • Cycle-active agents are drugs that require a cell to be in cycle, i.e., actively going through the cell cycle preparatory to cell division to be cytotoxic. Some of these drugs are effective primarily against cells in one of the phases of the cell. The importance of this designation is that cell cycle-active agents are usually schedule-dependent, and that duration of exposure is as important and usually more important than dose. In contrast, noncell cycle-active agents are usually not schedule-dependent, and effects depend on the total dose administered, regardless of the schedule. Alkylating agents are generally considered to be noncycle active, whereas antimetabolites are prototypes of cycle-active compounds.
  • 5-FU 5-fluorouracil
  • 5-FUdR 5-fluorodeoxyuridine
  • 5-FU exerts its cytotoxic effects by inhibition of DNA synthesis, or by incorporation into RNA, thus inhibiting RNA processing and function.
  • the active metabolite of 5-FU that inhibits DNA synthesis through potent inhibition of thymidylate synthase is 5-fluorodeoxyuridylate (5-FdUMP).
  • 5-FU 5-fluorodeoxyuridylate
  • Incorporation of 5-FU into DNA can occur also and may contribute to 5-FU cytotoxicity.
  • the major limiting toxicities of 5-FU and 5-FUdR include marrow and GI toxicity. Stomatitis and diarrhea usually occur 4-7 days after treatment. Further treatment is usually withheld until recovery from the toxic side-effects occurs. The nadir of leukopenia and of thrombocytopenia usually occurs 7-10 days after a single dose of a 5-day course.
  • the dose-limiting toxicity to infusions of 5-FUdR through the hepatic artery is transient liver toxicity, occasionally resulting in biliary sclerosis. Less common toxicities noted with 5-FU after systemic administration are skin rash, cerebellar symptoms and conjunctivitis.
  • methotrexate Another example of a cell cycle-active agent is methotrexate.
  • This folate antagonist was one of the first antimetabolites shown to induce complete remission in children with ALL.
  • Methotrexate (amethopterin) and aminopterin are analogs of the vitamin folic acid.
  • Methotrexate, and similar compounds acts by inhibiting the enzyme dihydrofolate reductase. As a consequence of this inhibition, intracellular folate coenzymes are rapidly depleted. These coenzymes are required for thymidylate biosynthesis as well as purine biosynthesis, as such, DNA synthesis is blocked by the use of methotrexate and alike.
  • methotrexate There is considerable toxicity associated with the use of methotrexate such as myelosuppression and GI distress.
  • An early sign of methotrexate toxicity to the GI tract is mucositis. Severe toxicity can result in diarrhea that is due to small bowel damage that can progress to ulceration
  • Cytosine arabinoside is an antimetabolite analog of deoxycytidine. In the analog, the OH group is in the ⁇ configuration at the 2′ position. This compound was first isolated from the sponge Cryptothethya crypta.
  • Ara-C is the drug of choice for the treatment of acute myelocytic leukemia. Ara-C is converted intracellularly to the nucleotide of triphosphate (ara-CTP) that is both an inhibitor of DNA polymerase and incorporated into DNA. The latter event is considered to cause the lethal action of ara-C. Nausea and vomiting are observed with patients being treated with ara-C.
  • Alkylating agents such as mechlorethamine, phenylalanine mustard, chlorambucil, ethylenimines and methyl melamines, and alkylsulfonates are employed to treat various cancers.
  • Cisplatin (diamino-dichloro-platinum) is a platinum coordination complex that has a broad spectrum antitumor activity. Cisplatin is a reactive molecule and is able to form inter- and intrastrand links with DNA in order to cross-link proteins with the DNA. Carboplatin is another platinum based antitumor drug.
  • antibiotics that have antitumor activity such as anthracyclines, such as doxorubicin, daunorubicin, and mitoxantrone.
  • anthracyclines such as doxorubicin, daunorubicin, and mitoxantrone.
  • Other antitumor antibiotics include bleomycin, dactinomycin, mitomycin C, and plycamycin.
  • antitumor drugs like asparaginase, that are considered to be within the scope of this invention.
  • these and the other drugs mentioned above all have a toxicity profile that is well known to those skilled in the art.
  • therapeutic agents that may be administered with one or more polysaccharides to reduce their toxicity or enhance efficacy include the following: anti-infectives including antibiotics, anti-virals and vaccines, antineoplastics, cardiovascular drugs including antiarrythmics, antihypertensives, etc., central nervous system drugs including analgesics, anorectics, anticonvulsants, anti-inflammatories and tranquilizers, etc.
  • anti-infectives including antibiotics, anti-virals and vaccines, antineoplastics, cardiovascular drugs including antiarrythmics, antihypertensives, etc.
  • central nervous system drugs including analgesics, anorectics, anticonvulsants, anti-inflammatories and tranquilizers, etc.
  • OTICS opthalmics
  • gastrointestinal including anti-ulcer drugs, anticholinergic drugs etc.
  • hormones, respiratory drugs including allergy medications, bronchodilators and decongestants, topical drugs and vitamins and minerals.
  • a polysaccharide like galactomannan in a mixture with a therapeutic drug such as 5-FU.
  • a therapeutic drug such as 5-FU.
  • galactomannan may increase cancer cell membrane fluidity and permeability as a result of galactose-specific interactions at the surface of the target cell.
  • the polysaccharide can thus serve as an effective vehicle for delivery of the drug to the target.
  • galactomannan may act to inhibit aggregation of tumor cells and their adhesion to normal cells so that the cancer fails to metastasize.
  • the polysaccharide may release the anti-cancer drug.
  • the toxicity of a therapeutic agent may be reduced because the drug is inactive as long as it is bound to the polymer.
  • the polysaccharide may release therapeutic agent.
  • Another possible mode of action for the polysaccharide like galactomannan may involve its interaction with some regulatory sites in a biological system, for example, if those sites are governed by galactose-specific residues, such as galectins.
  • Yet another possible mode of action may involve an inhibitory effect of the polysaccharide having a certain chemical structure (a certain Man:Gal ratio) and a certain size (molecular weight) on enzymatic systems responsible for a rapid clearance of therapeutic agent from the body, and therefore may potentially increase the bioavailability and prolong the mean residence time of drug in the body, thus improving the therapeutic profile of a drug in cancer therapy.
  • Galactomannan can be obtained from a variety of natural sources such as plants and microbial sources. The polysaccharide can also be synthetically made. Galactomannan can be derived from carob gum ( Ceratonia siliqua ), guar gum ( Cyamopsis tetragonoloba ), and honey locust ( Gleditsia triacanthos ), are examples of commercial available galactomannans.
  • a method for treating a disease comprising administering to the subject a mixture of one or more polysaccharides and an effective dose of one or more chemotherapeutic agents in a pharmaceutically acceptable formulation, wherein the polysaccharide is selected from group consisting of galactomannans from various sources.
  • This pharmaceutical formulation is then administered to a patient in need thereof in any acceptable manner known in the art.
  • the components, i.e., the polysaccharide and pharmaceutical can be administered separately to a subject.
  • the polysaccharide component of this embodiment can be prepared using methods articulated herein. If for example galactomannan is one of the components for the present embodiment, it can be extracted from Guar gum which itself is obtained from seeds of the Cyamopsis tetragonoloba.
  • the polysaccharide can be stored as a powder or as an aqueous solution, for example, in physiological saline. Other acceptable physiological solutions can be used as well.
  • a pharmaceutical preparation can be formed using the prepared polysaccharide and one or more pharmaceutical agents.
  • the polysaccharide theraputic' IV dose can be in the range of 10 to 200 mg/kg and is usually optimized to the optimal therapeutic dose of the chemotherapeutic agent for best therapeutic performance. For 5-FU the optimum dose (in mice) has been established at 30 to 150 mg/kg with best results at about 120 mg/kg. However, it could vary with other chemotherapy.
  • the pharmaceutical carriers that can be used for the administration of the present composition are well known to those skilled in the art.
  • the routes of administration include oral, intravenous, subcutaneous, intraperitoneal, intramuscular, and alike.
  • the route of administration can be any route sufficient to introduce the composition into a subject in a manner consistent with good medical practice. These various routes of administration are well known to those skilled in the art.
  • a mixture of one or more polysaccharides and chemotherapeutic agent is administered to a subject in need thereof, wherein the mixture comprises a sufficient amount of polysaccharide and chemotherapeutic agent in a ratio suitable for reducing the toxic side-effects in a subject while being effective against the particular pathology being addressed, wherein the polysaccharide is selected from group consisting of galactomannans from different plant sources.
  • Arabinogalactan from Larix occidentalis
  • Rhamnogalacturonan from potato
  • Carrageenan from Eucheuma Seaweed
  • Locust Bean Gum from Ceratonia siliqua .
  • the toxic side-effects being defined as those physiological effects (symptoms) realized by the subject resulting from the administration of the chemotherapeutic agent absent the polysaccharide.
  • a sufficient amount of polysaccharide is then understood herein to mean that amount required to minimize or mitigate toxic side-effects resulting from the administration of a pharmaceutical agent.
  • a pharmaceutical formulation in another embodiment, includes a mixture of one or more polysaccharides and an effective dose of a chemotherapeutic agent in a pharmaceutically acceptable formulation, wherein the polysaccharide is selected from group consisting of galactomannans from plant sources.
  • the mixture in the formulation contains an amount of one or more polysaccharides and a chemotherapeutic agent in a ratio suitable for effectively treating cancer as well as for reducing any toxic side-effect in the subject.
  • the mixture contains an amount of one or more polysaccharides and a chemotherapeutic agent in a ratio suitable for enhancing efficacy of chemotherapeutic effect for treating the cancer.
  • a method for treating cancer in a subject in need thereof that includes administrating an a mixture of one or more polysaccharides and an effective dose of a chemotherapeutic agent formulated so that the chemotherapeutic agent has enhanced therapeutic efficacy and reduced toxic effect upon the subject.
  • Any of the identified compounds of the present invention can be administered to a subject, including a human, by itself, or in pharmaceutical compositions where it is mixed with suitable carriers or excipients at doses therapeutically effective to prevent, treat or ameliorate a variety of disorders, including those characterized by that outlined herein.
  • a therapeutically effective dose further refers to that amount of the compound sufficient result in the prevention or amelioration of symptoms associated with such disorders.
  • the compounds of the present invention can be targeted to specific sites by direct injection into those sites.
  • Compounds designed for use in the central nervous system should be able to cross the blood-brain barrier or be suitable for administration by localized injection.
  • compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or alleviate the existing symptoms and underlying pathology of the subject being treating. Determination of the effective amounts is well within the capability of those skilled in the art.
  • the therapeutically effective 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 IC50 (the dose where 50% of the cells show the desired effects) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • a therapeutically effective dose refers to that amount of the compound that results in the attenuation of symptoms or a prolongation of survival in a subject. Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of a given population) and the ED50 (the dose therapeutically effective in 50% of a given population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50. Compounds which exhibit high therapeutic indices are preferred. 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 of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage can vary within this range 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 a patient's condition. Dosage amount and interval can be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the desired effects.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • compositions of the present invention can be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for use in accordance with the present invention thus can be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries that facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the agents of the invention can 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 barriers 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 subject to be treated.
  • Pharmaceutical preparations for oral use can be obtained solid excipient, optionally grinding a 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, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • PVP polyvinylpyrrolidone
  • disintegrating agents can be added, such as the 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 can be used, which can optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions that 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 can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration.
  • compositions can 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 pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoromethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoromethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoromethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoromethane, carbon dioxide or other suitable gas.
  • the compounds can be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection can be presented in unit dosage for, e.g., in ampoules or in multidose containers, with an added preservatives.
  • the compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds can be prepared as appropriate oily injection suspension. Suitable lipohilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions can contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension can also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient can 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 compounds can also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds can also be formulated as a depot preparation.
  • Such long acting formulations can be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds can be formulated with suitable polymeric or hydrophobic materials (e.g., as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, e.g., as a sparingly soluble salt.
  • a pharmaceutical carrier for the hydrophobic compounds of the invention is a co-solvent system comprising benzyl alcohol, a non-polar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • benzyl alcohol a non-polar surfactant
  • a water-miscible organic polymer a water-miscible organic polymer
  • an aqueous phase a co-solvent system
  • the proportions of a co-solvent system can be varied considerably without destroying its solubility and toxicity characteristics.
  • identity of the co-solvent components can be varied.
  • hydrophobic pharmaceutical compounds can be employed.
  • Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs.
  • Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds can be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known to those skilled in the art. Sustained-release capsules can, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein stabilization can be employed.
  • compositions also can comprise suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase 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.
  • salts can be provided as salts with pharmaceutically compatible counterions.
  • Pharmaceutically compatible salts can be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms.
  • Suitable routes of administration can, e.g., include oral, rectal, transmucosal, transdermal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • one can administer the compound in a targeted drug delivery system e.g., in a liposome coated with an antibody specific for affected cells.
  • the liposomes will be targeted to and taken up selectively by the cells.
  • compositions can, if desired, be presented in a pack or dispenser device which can contain one or more unit dosage forms containing the active ingredient.
  • the pack can, e.g., comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device can be accompanied by instruction for administration.
  • Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier can also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. Suitable conditions indicated on the label can include treatment of a disease such as described herein.
  • Pharmaceutically acceptable carriers are commonly added in typical drug formulations.
  • galactomannan has been used as a carrier for oral delivery of agents, which are in a non-liquid form. See U.S. Pat. Nos. 4,447,337; 5,128,143; and 6,063,402, the entire teaching of which is incorporated herein by reference.
  • the galactomannan oligomer of the present invention is a polysaccharide. In one aspect it has an average molecular weight of about 48,000 D. Shown below is the acceptable chemical nomenclature and structural formula for the galactomann of the present invention. Also shown is the stereochemical configuration.
  • FIG. 2 shows the structure of the galactomannan polysaccharide of the present invention as determined by Nuclear Magnetic Resonance (NMR).
  • the galactomannan of the present invention was compared to galactomannan from Carob (locust bean) gum.
  • An easy identification of the two principal sugar residues, that is mannose (Man) and galactose (Gal) comes from two peaks, at 4.8 p.p.m. (a doublet), respectively, since the ratio of Man/Gal in the galactomannan from guar gum (the galactomannan of the present invention) is 1.7:1 and that in the galactomannan from carob gum (Ceratonia siliqua) is 4:1.
  • FIG. 3 shows the Fourier Transform 1H-NMR spectrum of the guar gum galactomannan of the present invention.
  • the signal for the galactose anomeric protons appears at approximately 4.9 ppm (doublet).
  • the signal for the mannose anomeric protons appears at approximately 4.6 ppm (broad signal).
  • These signals are completely completely separated from those of the free monosachharides; the galactose a proton at 5.1 ppm and ⁇ at 4.5 ppm, the mannose ⁇ at 4.8 ppm and ⁇ at 5.0 ppm.
  • the ratio of mannose to galactose units can be easily calculated for this working standard at 1.7.
  • FIG. 4 shows the 13C-NMR spectrum of the galactomannan of the present invention, showing detailed positions of the chemical shifts and their intensities. This study confirms the above chemical structure for the galactomannan.
  • FIG. 4 illustrate the following:
  • Mannose residues are attached to each other “head-to-tail”, forming a (1 ⁇ 4) backbone chain.
  • the ratio of mannose to galactose in the galactomannan of the invention was 1.7 (that was the integral intensity ratio of C-1 signal of mannose and galactose, and also calculated from dual frequencies for unsubstituted and substituted Man residues, measured from C-4 (Man) resonance.
  • FIG. 5 shows the quantitation and molecular weight of the galactomannan of the present invention by HPLC/RI-MALLS (high performance liquid chromatography/refractive index-multi-angle laser light scattering).
  • High Performance Liquid Chromatography using Gel Permeation Chromatography (GPC) separation technology (also known as Size Exclusion Chromatograpy—SEC) is a well-established technique for the characterization of polymers.
  • GPC Gel Permeation Chromatography
  • MALLS Multi-Angle Laser Light Scattering
  • RI Refractive Index
  • Another advantage of GPC-MALLS over classical GPC is that besides molecular weights, additional information concerning radii and conformation in solution can be followed.
  • the principle of the GPC-MALLS method is based on the fact that light is more strongly scattered by large molecules than by small molecules.
  • the MALLS detector measures the degree of light scattering of a laser beam with detectors placed at fifteen different angles.
  • the output of the light scattering detector is proportional to the multiplication of the concentration and the molecular weight of macromolecules. Therefore, the shape of the light scattering peak is asymmetric. Further, it does not coincide with the RI peak, because the RI detector signal is proportional to the concentration only (see FIG. 5 ).
  • the molecular weight of the polymer eluting from the column can be calculated from the quotient of MALLS and RI signals.
  • a graph of the molecular weight versus the elution volume is obtained and (average) molecular weights and molecular weight distributions can be calculated.
  • the present investigators have adapted the GPC/IR-MALLS technique to quantitate the drug substance and characterize the molecular weight average and distribution throughout the R&D and scale up phases for the invention.
  • the use of the MALLS analysis removes many factors interfering with MW estimation by the “Classical GPC”.
  • GPC separations are based on differences in hydrodynamic volume instead of differences in molecular weight. Differences in molecular conformation, e.g. branching in dextrans, can strongly influence the hydrodynamic volume.
  • GPC elution of positively or negatively charged polymers can be non-ideal because of repulsion or attraction by the stationary phase.
  • the GPC-MALLS results are not affected by these chromatographical drawbacks, and absolute molecular weights are obtained.
  • the use of dual monitoring of the HPLC elution profile of the galactomannan of the invention provides two important chemical specifications, e.g., quantitative measurement by the Refractive Index signal and the absolute molecular weight by the Multi Angle Laser Light Scattering (MALLS) detector. Furthermore, these chromatograms can provide data on molecular stability and breakdown derivatives of the galactomannan of the invention.
  • MALLS Multi Angle Laser Light Scattering
  • FIG. 5 Shown in FIG. 5 is a flow chart of an example for a purification and manufacturing process for a galactomannan of the present invention.
  • High grade Guar gum is dissolved in warm water at 1% at 45° C. for 2 hr.
  • the pH is reduced to 2.2 with 1 M HCl and solution is heated to 95° C. for ⁇ 2 hours.
  • pH is adjust to 5.8 with 1 M NaOH.
  • the solution is then cool to 20° C. and filter with glass filter.
  • CuSO4/Na-K tartrate is added and the precipitate is collected on 200 mesh filter, wash with water solution and than washed in 5% HCl in 96% EtOH. Than washed with 75% EtOH and twice with 96% EtOH. And finally vacuum freeze-dried as white solids.
  • Galactomannan from a readily available source (e.g., Guar gum), was selected for process optimization and manufacturing.
  • the soluble galactomannan oligomer was tested in-vivo (in animals) for both efficacy and overall reduction of toxicity.
  • the manufacturing process described above produces a product in the form of a highly soluble oligomer of Galactomannan (GM) from certified premium Guar Gum powder (from seeds of Cyamopsis tetragonoloba ).
  • the process is designed to generate a highly pure soluble and homogeneous oligomer with an average molecular weight in the range of about 48,000 daltons, and mannose to galactose ratio in the range of about 1:7.
  • the process incorporates four major phases; controlled depolymerization to produce the desired galactomannan oligomer and three purification steps, removal of insoluble impurities, removal of water soluble impurities, removal of organic soluble impurities, and finally freeze drying to generate a pure and stable form of galactomannan powder.
  • Galactomannan can be packaged and delivered as a sterile concentrated solution in a single use vial, while bulk galactomannan can be produced and stored as powder.
  • the process described herein is for both bulk drug and final drug product.
  • the galactomannan drug product can be combined and administered together with a pharmaceutical like 5-fluorouracil to form the active ingredients of a pharmaceutical preparation.
  • the drug product contains normal saline for infusion (about 0.9 M sodium chloride in water) and has a pH of about 6.5.
  • the galactomannan of the present invention is a galactomannan derivative comprising exposed galactose moieties attached to a mannose backbone.
  • the compound is thought to interact with galactose-binding lectins or galectins that are generally located on cell surfaces.
  • Lectins are carbohydrate-binding proteins, typically located on the cell surface, which mediate various types of cellular interactions. It is generally accepted that lectins mediate many biological recognition events in plants and in animal tissues, and in tumor cell lines. Lectins play a role in cell-cell adhesion, and in the organization of the extracellular matrix.
  • lectins can act as receptors involved in selective intercellular adhesion and cell migration, recognition of circulating glycoproteins, and modulation of cell-cell and cell-matrix interactions.
  • Galectins are members of a family of ⁇ -galactoside-binding lectins with related amino acid sequences. Galectins and lectins have been the target of many experimental agents (monoclonal antibodies, simple sugars, and some polysaccharides, such as pectins) which allegedly interact with them on the cancer cell surface. The use of some of these agents has been shown to result in inhibition of tumor cell colony development.
  • the galactomannan of the invention can be distinctly identified and characterized, unlike many known polysaccharides. Its exposed galactose residues can readily interact with biological targets, such as lectins and galectins, thereby modulating signal transduction, cell-cell interactions or other functions. It thus can block the actions of galectins (or other receptors), thereby competing with their specific (or non-specific) ligands.
  • the galactomannan of the invention enhances the antineoplastic effects of 5-FU in animal models of colorectal cancer, although the precise mechanism has yet to be defined.
  • 5-fluorouracil has been the standard first-line agent used either alone, or in combination with, other agents in the treatment of metastatic colorectal cancer.
  • Preliminary animal studies with a variety of soluble galactomannan oligomers have shown promising response to the combination therapy of 5-FU and galactomannan with mannose to galactose ratio of 1:7.
  • the galactomannan from Cyamopsis tetragonoloba was administered intravenously (i.v.) once every four days for a total of three injections (q4d ⁇ 3) at a doses of 120 mg/kg/dose (360 mg/m2/dose), or was co-administered as one injection with 5-FU on the same q4d ⁇ 3 treatment schedule at doses of 120 mg/kg/dose of GM and 75 mg/kg/dose (225 mg/m2/dose) of 5-FU.
  • 5-FU alone was administered i.v. on the same q4d ⁇ 3 treatment schedule at doses of 75 mg/kg/dose (225 mg/m2/dose).
  • 5-FU was formulated in saline fresh on each day of treatment at a concentration of 3.75 mg/mL, at pH 9.2.
  • GM powder was dissolved in the 5-FU solution to yield the GM concentration of 6 mg/mL and 5-FU concentration of 3.75 mg/mL. Both individual compounds and their mixture were administered according to exact body weight with injection volume being 0.2 mL/10 g body weight.
  • a dosage of 75 mg/kg/dose of 5-FU (i.e., 225 mg/kg total dose over 8 days) was in excess of the maximum tolerated dosage and produced treatment-related deaths for three of ten mice within two weeks.
  • the treatment caused a delay in a median to quadrupling of tumor volume from 12.5 to 23.7 days. Again, there was no tumor regression after 56 days of the study; however, two relatively small tumors were observed that grew from 75 mm3 each at initiation of treatment to 126 mm3 and 567 mm3 by the end of the study. Median tumor volume increased from 101 mm3 at treatment initiation to 2254 mm3 after 56 days of the study. Mean survival time shifted from 14.2 days (control, untreated animals) to 23.7 days.
  • GM at a dosage of 120 mg/kg/dose administered alone on a q4d ⁇ 3 schedule, was well tolerated. No deaths or body weight loss was observed.
  • the median to quadrupling of tumor volume equaled 15.5 days, that is slightly longer than the value for untreated animals (12.5 days). There was no tumor regression after 56 days of study, however, two relatively small tumors (compared to median tumor volume) were observed that grew from 100 mm3 and 126 mm3, at initiation of treatment, to 270 mm3 and 729 m3, respectively, by the end of the study.
  • Median tumor volume increased from 100 mm3 at treatment initiation to 1813 mm3 after 56 days of the study, that is noticeably less compared to 2058 mm3 for untreated animals, and 2254 mm3 for 5-FU (75 mg/kg/dose)-treated animals.
  • Mean survival time was prolonged from 14.2 days (control, untreated animals) to 19.2 days.
  • a second study using COLO 205 tumors in mice evaluated the compatibility of the investigator's galactomannan/5-FU with Leucovorin (given orally, 25 mg/kg/dose) and dose escalation of galactomanna from 6 to 600 mg/kg/day (or 18 to 1800 mg/m2).
  • the combination of 5-FU+galactomannan (at 48 mg/kg and 120 mg/kg, respectively) had the best anti-tumor response for both mean tumor size and time required to quadruple tumor weight, being superior to the 5-FU alone or 5-FU+Leucovorin combination.
  • tetragonoloba was administered intravenously (i.v.) once every four days for a total of four injections (q4d ⁇ 4) at a dosage of 120 mg/kg/dose (360 mg/m2/dose) or was co-administered as one injection with 5-FU on the same q4d ⁇ 4 treatment schedule at a dosage of 6, 30, 120, and 600 mg/kg/dose (18, 90, 360, and 1800 mg/m2/dose, respectively) of GM and 48 mg/kg/dose (144 mg/m2/dose) dose of 5-FU.
  • 5-FU alone was administered i.v. on the same q4d ⁇ 4 treatment schedule at dosages of 48 mg/kg/dose.
  • 5-FU was formulated in saline fresh on each day of treatment at a concentration of 4.8 mg/mL, at pH 9.2.
  • GM powder was dissolved in the 5-FU solution to yield the GM concentration of 0.6, 3.0, 12, and 60 mg/mL and 5-FU concentration of 4.8 mg/mL.
  • Leucovorin powder (clinical formulation, Leucovorin calcium for injection) was reconstituted with water for injection to yield a concentration of 10 mg/mL. On each day of treatment the stock solution was diluted with water for injection to yield a concentration of 2.5 mg/mL.
  • 5-FU and GM and their mixture with each other and leucovorin were administered by exact body weight with injection or p.o. volume being 0.1 mL/10 g body weight.
  • mice Two more combination-treatment groups were also included in the study.
  • One group of mice was treated with 5-FU (48 mg/kg/dose, i.v., q4d ⁇ 4), followed by oral gavage (p.o.)with leucovorin, administered two hours after 5-FU at a dosage of 25 mg/kg/dose.
  • Another two groups of mice were treated with 5-FU in a combination with the galactomannan (48 mg/kg/dose and 120 mg/kg/dose, respectively, i.v., q4d ⁇ 4), followed by p.o. leucovorin treatment, administered at a dosage of 25 mg/kg/dose two hours after 5-FU plus the galactomannan.
  • a dosage of 48 mg/kg/dose of 5-FU (that is, 192 mg/kg total dose over 12 days) was well tolerated and produced some growth delay in the median to quadrupling of tumor volume, increasing it from 7.2 to 8.7 days.
  • Two tumors in the group of 10 mice were significantly (three times or more) smaller, compared with the median tumor size, after 13 days of treatment, growing from 100 and 163 mm3 at initiation of treatment to 270 mm3 and 138 mm3, respectively, by the end of the study.
  • Median tumor volume increased from 172 mm3 at treatment initiation to 800 mm3 after 13 days of the study, less than the control value 1288 mm3.
  • GM at a dosage of 120 mg/kg/dose administered alone on a q4d ⁇ 4 schedule did not delay growth (the median to quadrupling of tumor volume equaled 6.9 days, compared to that of 7.2 days in the control group). No tumor regression occurred after 13 days of study, and no relatively small tumors (compared to median tumor volume) were observed. Median tumor volume increased from 157 mm3 at treatment initiation to 1152 mm3, a value essentially equal to that of the untreated animals (1288 mm3).
  • Treatment with 5-FU in the presence of leucovorin did not add anything to the efficacy of the drug.
  • Median days to quadrupling of tumor weight even decreased from 8.7 days to 8.2 days, and tumor weight at Day 13 from treatment initiation slightly decreased as median (from 800 mg to 775 mg), but noticeably increased as mean (from 734 mg to 969 mg).
  • a second human colon tumor, HT-29, in NU/NU-nuBR nude mice was studied to evaluate the anti-tumor efficacy of the combinations of 5-FU at 48 mg/kg/dose, galactomannan at 30 and 120 mg/kg/dose given I.V. every 4 days for a total of four injections. All groups received oral leucovorin, 25 mg/kg/dose. At Day 33, the mean tumor volume was lowest for 5-FU+leucovorin+galactomannan 120 mg/kg.
  • the GM was co-administered i.v. via tail vein injection once every four days for a total of four injections (q4d ⁇ 4) at a doses of 30 and 120 mg/kg/dose as one injection with 5-FU (48 mg/kg/dose), followed by oral gavage (p.o.) of leucovorin, administered two hours after the injection, at a dose of 25 mg/kg/dose on the same q4d ⁇ 4 schedule.
  • GM or 5-FU were administered also on the same q4d ⁇ 4 treatment schedule at doses of 120 mg/kg/dose (GM) or 48 mg/kg/dose (5-FU), followed by 25 mg/kg of leucovorin, administered two hours later.
  • GM was formulated in 0.9% sterile saline fresh on each day of treatment at a concentration of 12 mg/mL.
  • Leucovorin powder (clinical formulation, Leucovorin calcium for injection) was reconstituted with 0.9% sterile saline to yield a concentration of 2.5 mg/mL.
  • 5-FU was formulated in 0.9% sterile saline fresh on each day of treatment at a concentration of 4.80 mg/mL, at pH 9.2.
  • GM powder and 5-FU were dissolved in 0.9% fresh saline to yield the GM concentration of 3.0 mg/mL or 12 mg/mL, and 5-FU concentration of 4.80 mg/mL. Both individual compounds and their mixture were administered according to exact body weight with injection volume being 0.1 mL/10 g body weight.
  • control (untreated) tumors grew well in all mice, with a median of 13.3 days for quadrupling of tumor volume.
  • a dosage of 48 mg/kg/dose of 5-FU (192 mg/kg total dose over 12 days of the treatment) along with an oral administration of leucovorin as described above was within the maximum tolerated dosage, producing no treatment-related deaths in the group of ten mice within three weeks.
  • the treatment caused a delay of two days for the quadrupling of tumor volume (from 13.3 to 15.3 days). Median tumor volume increased from 179 mm3 at treatment initiation to 1120 mm3 on Study Day 26.
  • 3H-galactomannan was independent of the injected dose and did not change its pattern when 5-FU (342 mg/m2) was combined with 3H-galactomannan.
  • 3H-galactomannan elimination from plasma, kidneys, lungs and tumor in the various groups was rapid, an average of approximately 50% of the one-hour radioactivity was detected at six hours except in tumor-bearing mice, where the radioactivity in tumor samples from mice treated with 6 or 60 mg/kg of 3H-galactomannan with or without 5-FU averaged approximately 72% remaining after six hours. Elimination of 3H-galactomannan from the liver was more gradual than in other tissues, and on average, more than 50% of the radioactivity detected at one hour after injection was still present at 24 hours.
  • mice Male NCr-nu athymic nude mice (Charles Rivers Laboratories, Raleigh, N.C.) were acclimated and housed as described above. The first set of animals (a total of 18 mice) were non-tumored mice. The second set of 18 mice were tumored as follows. Thirty to forty mg fragments from an in vivo passage of COLO 205 human colon tumor were implanted subcutaneously (s.c.) in mice as described above, and allowed to grow. Tumors were allowed to reach 245-392 mg in weight before the start of treatment. A sufficient number of mice were implanted so that tumors in a weight range as narrow as possible were selected for the trial on the day of treatment initiation. Those animals selected with tumors in the proper size range were divided into the various treatment groups.
  • Tritiation of GM from G. triacanthos was performed as follows. 12.8 mg of GM was dissolved in 2.0 mL of water and exposed to 25 Curies of tritium gas in the presence of Pd/BaSO4 catalyst (120 mg, totally insoluble in water). After one hour the gas supply was removed, the catalyst was filtered away, and the aqueous solution of GM was evaporated to dryness repeatedly (four-fold, adding water), until no labile tritium was found. Total yield of the labeled GM was 3.8 mCi, specific radioactivity was 300 ⁇ Ci/mg.
  • mice All 36 mice, divided into 18 groups, were given a single intravenous injection of cold or tritiated GM (either 6 or 60 mg/kg) or of a combination of GM (60 mg/kg, cold or tritiated) and 5-FU (114 mg/kg) on the same day.
  • Non-labeled GM was formulated in saline, and tritiated GM was added to the solution so that each animal received 10 ⁇ Ci of radioactivity.
  • 5-FU was dissolved in the solution containing GM (at a concentration of 6 mg/mL). All dosing solutions (100 ⁇ L each) were counted in duplicate.
  • mice per group Two mice per group were bled at 1, 6, and 24 hrs after injection, and plasma was prepared. Animals were then sacrificed; livers, kidneys, lungs, and tumors (from tumored animals) were collected, weighed and flash-frozen for further analysis.
  • livers were dissolved in 10 mL of Soluene 350 (Packard Instruments, Downers Grove, Ill.) and incubated first for 4 hrs at 500C, and at room temperature, until tissues were solubilized. One millilter of the resulting solution was counted in a scintillation counter as a single sample. Based on tissue weight and the sample volume, the number of ⁇ Ci of tritiated GM per gram of tissue was calculated.
  • Soluene 350 Packard Instruments, Downers Grove, Ill.
  • Kidneys were treated in the same manner, but dissolved in 2 mL of Soluene. After the tissue was solubilized at room temperature, 15 mL of Safety Solve scintillation fluid (Research Products International, Mount Prospect, Ill.) was added and samples were incubated overnight. Five mL of the resulting solution were diluted in 15 mL of Safety Solve and counted in a scintillation counter as a single sample. Lungs were treated in the same manner but dissolved in 1 mL of Soluene. Plasma samples (50 ⁇ L each) were placed direct into Safety Solve and counted as a single sample.
  • Safety Solve scintillation fluid Research Products International, Mount Prospect, Ill.
  • tumors were dissolved in one or two milliliters of Soluene and incubated for three days at 500C to solubilize. Fifteen milliliters of Safety Solve were then added and samples were incubated overnight at room temperature. Two milliliters of water were then added and samples were counted in a scintillation counter as a single sample.
  • mice Male NCr-nu athymic nude mice were divided into two principal sets, 18 animals in each. The first set of animals were non-tumored mice. The second set were tumored as follows. Thirty- to -forty mg fragments from an in vivo passage of COLO 205 human colon tumor were implanted subcutaneously (s.c.) in mice as described above, and allowed to grow. Tumors were allowed to reach 245-392 mg in weight before the start of treatment. A sufficient number of mice were implanted so that tumors in a weight range as narrow as possible were selected for the trial on the day of treatment initiation. Those animals selected with tumors in the proper size range were divided into the various treatment groups.
  • mice All 36 mice, divided into 18 groups, were given a single intravenous injection of cold or tritiated GM (either 6 or 60 mg/kg) or of a combination of GM (60 mg/kg, cold or tritiated) and 5-FU (114 mg/kg) on the same day. Tritiation of GM (with the resulting specific activity of 300 ⁇ Ci/mg) is described above. Cold GM was formulated in saline, and tritiated GM was added to the solution so that each animal received 10 ⁇ Ci of radioactivity. 5-FU was dissolved in the solution containing GM (at a concentration of 6 mg/mL).
  • mice per group Two mice per group were bled at 1, 6, and 24 hrs after injection, and plasma was prepared. Animals were then sacrificed; livers, kidneys, lungs, and tumors (from tumored animals) were collected, weighed and flash-frozen for further analysis, as described above.
  • the galactomannan freely binds to liver, kidney, lung, tumor, and plasma, and did not reach limits of the binding, e.g., did not reach saturation of the binding between the 6 mg/kg and 60 mg/kg doses.
  • 6 mg/kg (with a relative radioactivity of 1.0) and 60 mg/kg (with a relative radioactivity of 0.1) doses of the galactomannan were administered, the amount of bound radioactive galactomannan was the same; that is, the amount of bound galactomannan increased 10-fold for the 10-times higher dose.
  • the distribution of radioactivity in whole tissues as well as per weight or volume (in plasma) was practically identical for 6 and 60 mg/kg of GM, hence, the respective data were pooled. Also, the distribution of radioactivity in whole tissues as well as per weight or volume (in plasma) was practically identical for tumored and non-tumored animals (except in tumors, that obviously were present only in tumored animals), hence, the respective data were pooled. Overall, the data were averaged for eight animals, except the data for tumors, that are average for four animals.
  • 5-FU and GM work in a synergism when delivered into the tumor. This might explain why GM in a combination with 5-FU increases efficacy of the drug against COLO 205 human colon tumor, bearing by mice (see above).
  • 5-FU and GM work as antagonists (apparently, compete with each other for the same binding sites in the liver) when delivered into the liver.
  • Acute toxicology studies of the galactomannan of the present invention were performed in mice, rats and dogs and subchronic toxicology studies were performed in rats and dogs. The following is a summary of the findings of these studies:
  • Alopecia severe transient decreases in body weight and feed consumption, and transient depression of erythrocyte parameters and platelets were observed in all groups receiving 5-FU.
  • Treatment-related histopathological lesions were resolved in animals sacrificed at study day 29 (only one female survived in Group 2 and Group 6).
  • the galactomannan of the invention has minimal histological effects with only 1/30 rats injected with 48 mg/kg dose of the GMP galactomannan clinical solution having granulomatous inflammation. None of the 30 rats injected with the 96 mg/kg dose of the GMP galactomannan clinical solution were affected. It was concluded that galactomannan clinical solutions, up to a dose of 15 mg/kg (555 mg/m2; 9 mg/mL), is safe for human use and poses no undue risk to health.
  • Adverse effects in 5-FU groups included ataxia, prostration, vocalization, convulsions, tremors, hypersensitivity to touch, aggressive behavior (resulting in only three doses being given to the 6/3.2 5-FU/galactomanna group), emesis, salivation, soft stools and decreased red cell parameters and platelet counts.
  • Gross/microscopic changes in unscheduled-death animals included congestion of one or more organs (suggesting cardiovascular dysfunction) and atrophy of the mucosa of the GI tract. At the day 5 necropsy, treatment-related histological changes were largely limited to the 5-FU females. At day 29 necropsy, there were no remarkable changes in the survivors.
  • Galactomannan at 500 mg/m2 produced no observed adverse effects.
  • Mutagenicity studies were conducted and included two Ames bacterial reverse mutation assays in which the galactomannan of the invention was evaluated by itself in the first study and combined with 5-FU in the second study.
  • the test articles in both studies were evaluated in bacterial assays using Salmonella typhimurium strains TA97a, TA98, TA100, TA1535 and Escherichia coli strain WP2 uvrA (pKM101), both in the presence and absence of an exogenous metabolic activation system. No evidence of mutagenic activity was detected in either of the two studies.
  • Cytokines and chemokines have been shown, in in vitro studies, to promote cancer cells susceptibility to destruction by the immune response. (Cytokines and chemokines are well known to those skilled in the art and a list of such can be found in any modern biology/medicine text). In few cases it has been shown that cytokines directly inhibit tumor cell growth. Cytokines function as messengers of the immune system by regulating the intensity and duration of the immune response by exerting a variety of effects on lymphocytes and other immune cells. Cytokines also control cellular proliferation and differentiation. In the USA, the Food and Drug Administration (FDA) has already approved the use of two cytokines-IL-2 and ⁇ -interferon for treatment of cancer.
  • FDA Food and Drug Administration
  • IL-2 has biological activity against renal cell disease, melanoma, lymphoma, and leukemia. Interferon has been effective against these cancers as well as against Kaposi's sarcoma, chronic myelogenous leukemia, and hairy cell leukemia.
  • cytokine The Effect of galactomannan on chemotherapy drugs has been also shown in with these proteinous chemotherapeutics, e.g., cytokine.
  • cytokine a combination of IL-12 (10 ⁇ g/kg) and IL-12 (40 mg/kg) alone or in combination with galactomannan (120 mg/kg) were used to treat a mice model implanted with human colon tumor HT-29.
  • the IV injections started once tumor size reached 110 mg and were repeated 4 times at 4 days interval, the result one week after the last injection (day 26) already showed a significant improvement in inhibition of tumor growth was achieved.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Inorganic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Dermatology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Polysaccharides And Polysaccharide Derivatives (AREA)
US10/657,508 2003-09-08 2003-09-08 Co-administration of a polysaccharide with a chemotherapeutic agent for the treatment of cancer Abandoned US20050053664A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
US10/657,508 US20050053664A1 (en) 2003-09-08 2003-09-08 Co-administration of a polysaccharide with a chemotherapeutic agent for the treatment of cancer
PCT/US2004/028883 WO2005025501A2 (fr) 2003-09-08 2004-09-07 Co-administration de polysaccharide avec un agent chimiotherapeutique pour le traitement du cancer
EP04783211A EP1662874A4 (fr) 2003-09-08 2004-09-07 Co-administration de polysaccharide avec un agent chimiotherapeutique pour le traitement du cancer
JP2006525488A JP2007505041A (ja) 2003-09-08 2004-09-07 癌治療のための化学療法薬とポリサッカライドの同時投与
CNA2004800300176A CN1867252A (zh) 2003-09-08 2004-09-07 多糖与用于癌症治疗的化疗剂的共同给药
AU2004272022A AU2004272022B2 (en) 2003-09-08 2004-09-07 Co-administration of polysaccharide with a chemotherapeutic agent for the treatment of cancer
BRPI0413410-9A BRPI0413410A (pt) 2003-09-08 2004-09-07 co-administração de um polissacarìdeo com um agente quimioterapêutico para o tratamento de cáncer
IL174121A IL174121A (en) 2003-09-08 2006-03-05 A mixture containing non-intestinal and oligosaccharide galactomanan
US12/036,608 US7893252B2 (en) 2003-09-08 2008-02-25 Selectively depolymerized galactomannan polysaccharide
US12/962,296 US20110077217A1 (en) 2003-09-08 2010-12-07 Selectively depolymerized galactomannan polysaccharide
AU2010257400A AU2010257400B2 (en) 2003-09-08 2010-12-23 Co-administration of polysaccharide with a chemotherapeutic agent for the treatment of cancer

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10/657,508 US20050053664A1 (en) 2003-09-08 2003-09-08 Co-administration of a polysaccharide with a chemotherapeutic agent for the treatment of cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/036,608 Division US7893252B2 (en) 2003-09-08 2008-02-25 Selectively depolymerized galactomannan polysaccharide

Publications (1)

Publication Number Publication Date
US20050053664A1 true US20050053664A1 (en) 2005-03-10

Family

ID=34226574

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/657,508 Abandoned US20050053664A1 (en) 2003-09-08 2003-09-08 Co-administration of a polysaccharide with a chemotherapeutic agent for the treatment of cancer
US12/036,608 Expired - Lifetime US7893252B2 (en) 2003-09-08 2008-02-25 Selectively depolymerized galactomannan polysaccharide
US12/962,296 Abandoned US20110077217A1 (en) 2003-09-08 2010-12-07 Selectively depolymerized galactomannan polysaccharide

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/036,608 Expired - Lifetime US7893252B2 (en) 2003-09-08 2008-02-25 Selectively depolymerized galactomannan polysaccharide
US12/962,296 Abandoned US20110077217A1 (en) 2003-09-08 2010-12-07 Selectively depolymerized galactomannan polysaccharide

Country Status (8)

Country Link
US (3) US20050053664A1 (fr)
EP (1) EP1662874A4 (fr)
JP (1) JP2007505041A (fr)
CN (1) CN1867252A (fr)
AU (2) AU2004272022B2 (fr)
BR (1) BRPI0413410A (fr)
IL (1) IL174121A (fr)
WO (1) WO2005025501A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023250141A3 (fr) * 2022-06-24 2024-04-11 Enalare Therapeutics Inc. Méthodes de traitement d'insuffisance ventilatoire neurologique

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050053664A1 (en) * 2003-09-08 2005-03-10 Eliezer Zomer Co-administration of a polysaccharide with a chemotherapeutic agent for the treatment of cancer
WO2008011216A2 (fr) 2006-05-16 2008-01-24 Pro-Pharmaceuticals, Inc. Polysaccharides à dents de galactose dans une formulation pour des thérapies antifibrotiques
BR112014015940B1 (pt) 2011-12-28 2022-08-09 Galectin Therapeutics, Inc Polissacarídeo de grau farmacêutico, composição, e seus usos
BR112014030534A2 (pt) 2012-06-06 2017-06-27 Galectin Therapeutics Inc composições de galacto-ramnogalacturonato para o tratamento de doenças associadas com óxido nítrico sintase induzível elevada
WO2014043708A1 (fr) 2012-09-17 2014-03-20 Traber Peter G Procédé pour l'amélioration d'immunothérapies spécifiques dans le traitement du cancer
US8828971B2 (en) 2012-10-10 2014-09-09 Galectin Therapeutics, Inc. Galactose-pronged carbohydrate compounds for the treatment of diabetic nephropathy and associated disorders
KR101989972B1 (ko) * 2012-11-05 2019-06-18 (주)나디안바이오 약물 유발성 신장질환 예방 또는 치료용 약제 조성물
WO2014130648A1 (fr) * 2013-02-20 2014-08-28 Galectin Therapeutics, Inc. Méthode pour le traitement de la fibrose pulmonaire
JP6535013B2 (ja) 2013-09-19 2019-06-26 ザ・リサーチ・ファウンデーション・フォー・ザ・ステイト・ユニヴァーシティ・オブ・ニューヨーク 糖尿病または肝臓脂肪症を処置する方法および物質
WO2016160590A1 (fr) * 2015-03-27 2016-10-06 The Research Foundation For The State University Of New York Procédés et matériaux pour traiter le cancer
AU2016243516A1 (en) 2015-03-27 2017-11-09 The Research Foundation For The State University Of New York Methods and materials for reducing amyloid beta levels within a mammal
US10676723B2 (en) 2015-05-11 2020-06-09 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
CN105412155B (zh) * 2015-12-02 2019-03-12 皖南医学院 拟康氏木霉胞外多糖治疗和预防结肠癌的应用、及其联用化疗药物治疗结肠癌的应用
EP3813883A1 (fr) 2018-06-29 2021-05-05 Glykos Biomedical Oy Conjugués
WO2021123506A1 (fr) 2019-12-18 2021-06-24 Glykos Biomedical Oy Conjugué stable

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5733572A (en) * 1989-12-22 1998-03-31 Imarx Pharmaceutical Corp. Gas and gaseous precursor filled microspheres as topical and subcutaneous delivery vehicles
US20020068077A1 (en) * 2000-09-25 2002-06-06 Anatole Klyosov Methods and compositions for reducing side effects in chemotherapeutic treatments
US6413494B1 (en) * 1998-07-23 2002-07-02 Samyang Corporation Composition and pharmaceutical dosage form for colonic drug delivery using polysaccharides
US6645946B1 (en) * 2001-03-27 2003-11-11 Pro-Pharmaceuticals, Inc. Delivery of a therapeutic agent in a formulation for reduced toxicity
US6982255B2 (en) * 2001-03-27 2006-01-03 Anatole Klyosov Delivery of a therapeutic agent in a formulation for reduced toxicity
US7012068B2 (en) * 2001-03-27 2006-03-14 Pro-Pharmaceuticals, Inc. Co-administration of a polysaccharide with a chemotherapeutic agent for the treatment of cancer
US20060057131A1 (en) * 2001-12-20 2006-03-16 Eric Simard Malleable protein matrix and uses thereof
US7166299B2 (en) * 1998-07-24 2007-01-23 Seo Hong Yoo Preparation of aqueous clear solution dosage forms with bile acids

Family Cites Families (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5612114B2 (fr) 1974-06-07 1981-03-18
JPS5535049A (en) 1978-09-04 1980-03-11 Otsuka Pharmaceut Factory Inc Amino acid transfusion for cancerous patient
US4231938A (en) 1979-06-15 1980-11-04 Merck & Co., Inc. Hypocholesteremic fermentation products and process of preparation
US4444784A (en) 1980-08-05 1984-04-24 Merck & Co., Inc. Antihypercholesterolemic compounds
MX7065E (es) 1980-06-06 1987-04-10 Sankyo Co Un procedimiento microbiologico para preparar derivados de ml-236b
US4450171A (en) 1980-08-05 1984-05-22 Merck & Co., Inc. Antihypercholesterolemic compounds
US5308838A (en) 1982-05-07 1994-05-03 Carrington Laboratories, Inc. Uses of aloe products
US5118673A (en) 1982-05-07 1992-06-02 Carrington Laboratories, Inc. Uses of aloe products
US4739073A (en) 1983-11-04 1988-04-19 Sandoz Pharmaceuticals Corp. Intermediates in the synthesis of indole analogs of mevalonolactone and derivatives thereof
WO1984004041A1 (fr) 1983-04-14 1984-10-25 Univ California Systeme d'administration de medicaments specifiques du colon
ES2058070T3 (es) 1986-05-09 1994-11-01 Pulverer Gerhard Utilizacion de monosacaridos especificos para la preparacion de un medicamento para impedir metastasis de tumores malignos.
US4681893A (en) 1986-05-30 1987-07-21 Warner-Lambert Company Trans-6-[2-(3- or 4-carboxamido-substituted pyrrol-1-yl)alkyl]-4-hydroxypyran-2-one inhibitors of cholesterol synthesis
US5490991A (en) 1986-07-03 1996-02-13 Advanced Magnetics, Inc. Directed delivery of radioprotectants using a receptor specific carrier
JP2569746B2 (ja) 1987-08-20 1997-01-08 日産化学工業株式会社 キノリン系メバロノラクトン類
US4804770A (en) 1988-04-29 1989-02-14 E. R. Squibb & Sons, Inc. Process for preparing a keto-phosphonate intermediate useful in preparing HMG-CoA reductase inhibitors
JPH0220292A (ja) 1988-07-06 1990-01-23 Agency Of Ind Science & Technol 低分子化キトサンの製造方法
FR2642065B1 (fr) 1989-01-24 1991-05-24 Lipha Derives d'acides benzocycloalcenyl dihydroxy alcanoiques, procede de preparation et medicaments les contenant
JP2798287B2 (ja) 1989-02-08 1998-09-17 ユニチカ株式会社 抗炎症剤
US5569483A (en) 1989-02-10 1996-10-29 Alko Group Ltd. Degraded polysaccharide derivatives
FI94339C (fi) 1989-07-21 1995-08-25 Warner Lambert Co Menetelmä farmaseuttisesti käyttökelpoisen /R-(R*,R*)/-2-(4-fluorifenyyli)- , -dihydroksi-5-(1-metyylietyyli)-3-fenyyli-4-/(fenyyliamino)karbonyyli/-1H-pyrroli-1-heptaanihapon ja sen farmaseuttisesti hyväksyttävien suolojen valmistamiseksi
US5015632A (en) 1990-01-08 1991-05-14 Olin Corporation Chitosan pyrithione as an antimicrobial agent useful in personal care products
JP3253982B2 (ja) 1991-03-13 2002-02-04 ピアス株式会社 にきび用化粧料
US5605938A (en) 1991-05-31 1997-02-25 Gliatech, Inc. Methods and compositions for inhibition of cell invasion and fibrosis using dextran sulfate
JP2648897B2 (ja) 1991-07-01 1997-09-03 塩野義製薬株式会社 ピリミジン誘導体
JP3164854B2 (ja) 1991-10-31 2001-05-14 雪印乳業株式会社 皮膚衛生用シート
US5639737A (en) 1991-11-04 1997-06-17 Co Enzyme Technology Ltd. Method and compositions for treating malignant tumors and inhibiting growth and metastases of malignant tumors
WO1993008810A1 (fr) 1991-11-05 1993-05-13 Carrington Laboratories, Inc. Utilisations de produits issus de l'aloes, tel que de l'acemannan, dans le traitement des maladies necessitant l'intervention du systeme immunitaire pour la guerison
DE4236237A1 (de) 1992-10-27 1994-04-28 Behringwerke Ag Prodrugs, ihre Herstellung und Verwendung als Arzneimittel
US20020107222A1 (en) 1993-03-01 2002-08-08 David Platt Modified polysaccharides for treatment of cancer
DE4309553A1 (de) 1993-03-24 1994-09-29 Bayer Ag Verfahren zur Herstellung von 3R,5S-(+)-Natrium-erythro-(E)-7-(4-(4-flurophenyl)-2,6-diisopropyl-5-methoxymethyl-pyrid-3-yl)-3,5-dihydroxy-hept-6-enoat
FI943242A (fi) 1993-07-15 1995-01-16 Jcr Pharmaceutical Co Ltd Lääke munuais- ja maksatauteihin
US5908761A (en) 1993-12-05 1999-06-01 Yeda Research And Development Co. Ltd. Galectin-8 and galectin-8-like proteins and DNA molecules coding therefor
US5498702A (en) 1993-12-16 1996-03-12 California Natural Products Treated pectinic acid process and product
US5620961A (en) 1994-04-25 1997-04-15 Markovic; Nenad S. Fructose ester-β-cyclodextrin complexes and processes for making and using same
GB9412394D0 (en) 1994-06-21 1994-08-10 Danbiosyst Uk Colonic drug delivery composition
US5834442A (en) 1994-07-07 1998-11-10 Barbara Ann Karmanos Cancer Institute Method for inhibiting cancer metastasis by oral administration of soluble modified citrus pectin
EP0721784A4 (fr) 1994-07-29 1997-09-17 Kanegafuchi Chemical Ind Preparation de medicament a liberation lente
US6190657B1 (en) 1995-06-07 2001-02-20 Yale University Vectors for the diagnosis and treatment of solid tumors including melanoma
US5626856A (en) 1995-06-30 1997-05-06 Safe & Dry Company, Inc. Cosmetic delivery vehicles and related compositions
DE19524515A1 (de) 1995-07-05 1997-01-09 Deutsches Krebsforsch Saccharid-Konjugate
US5681923A (en) 1995-10-06 1997-10-28 Platt; David Tumor derived carbohydrate binding protein
EP0888776A4 (fr) 1996-03-15 2005-01-26 Takara Bio Inc Produit obtenu par traitement thermique d'acide uronique, et aliments, boissons ou medicaments contenant ce produit
US5861142A (en) 1996-03-25 1999-01-19 Schick; Mary Pichler Method for promoting hair, nail, and skin keratinization
WO1998008528A1 (fr) 1996-08-30 1998-03-05 Biomeasure Incorporated Procede d'inhibition de la fibrose avec un agoniste de la somatostatine
US5869289A (en) 1996-10-09 1999-02-09 Incyte Pharmaceuticals, Inc. Human galectin homolog
US5891861A (en) 1996-10-15 1999-04-06 Platt; David Composition and method for treating fungal disease in animals
US5995100A (en) * 1997-01-07 1999-11-30 Ncr Corporation Method of automatically generating custom controls for selected modules in a library
AU7201998A (en) * 1997-05-06 1998-11-27 Zhi Liu Drug delivery system
US5831052A (en) 1997-05-07 1998-11-03 Incyte Pharmaceuticals, Inc. New human translocation associated protein
US6110492A (en) * 1997-05-28 2000-08-29 Jenner Biotherapies, Inc. Immunogenic compositions
AU751932B2 (en) 1997-10-15 2002-08-29 Polarx Biopharmaceuticals, Inc. Compositions and methods for the treatment of primary and metastatic neoplastic diseases using arsenic compounds
US5972995A (en) 1997-10-16 1999-10-26 Children's Hospital Medical Center Of Northern California Compositions and methods for cystic fibrosis therapy
SI1064942T1 (en) 1998-03-26 2004-12-31 Fujisawa Pharmaceutical Co., Ltd. Sustained release preparation of a macrolide
JP2002515457A (ja) * 1998-05-15 2002-05-28 アフトン コーポレーション 腫瘍治療のための併用療法
JP2002522398A (ja) 1998-08-06 2002-07-23 帝人株式会社 細胞外基質の過剰産生と蓄積に対し薬効を示す医薬組成物
CA2279791C (fr) 1998-08-14 2011-11-08 Marcus B. Gohlke Supplement alimentaire combinant du colostrum et de la lactoferrine dans une formule posologique absorbable par les muqueuses
US6500807B1 (en) 1999-02-02 2002-12-31 Safescience, Inc. Modified pectin and nucleic acid composition
US6912230B1 (en) 1999-02-05 2005-06-28 Tecore Multi-protocol wireless communication apparatus and method
US6274566B1 (en) 1999-02-23 2001-08-14 Econugenics, Inc. Methods for treating mammals with modified alginates and pectins
DE60036540T2 (de) 1999-03-25 2008-07-03 Mitsubishi Tanabe Pharma Corp. Rho-kinase-inhibitoren für die vorbeugung oder behandlung von interstitieller pneumonie und pulmonaler fibrose
WO2000059481A1 (fr) 1999-04-06 2000-10-12 Pharmaquest Ltd. Forme posologique pharmaceutique destinee a l'administration intermittente de d-threo-methylphenidate et d'un second stimulant du systeme nerveux central
AU4055000A (en) 1999-04-13 2000-11-14 Daniel K. Hsu Galectin expression is induced in cirrhotic liver and hepatocellular carcinoma
CN1191860C (zh) 1999-04-22 2005-03-09 比奥根公司 利用整联蛋白α4亚单位的拮抗剂治疗纤维变性的方法
GB9909135D0 (en) 1999-04-22 1999-06-16 Univ Wales Medicine Cystic fibrosis medicaments
US6893637B1 (en) 1999-10-21 2005-05-17 Zymogenetics, Inc. Method of treating fibrosis
US6562369B2 (en) * 1999-12-16 2003-05-13 Dermatrends, Inc. Transdermal administration of androgenic drugs hydroxide-releasing agents as permeation enhancers
DE60030164T2 (de) * 1999-12-30 2007-08-30 Sugen, Inc., San Francisco 3-Heteroarylidenyl-2-Indolinon Derivate für die Modulierung der Aktivität einer Proteinkinase und für die Verwendung bei der Chemotherapie von Krebs
CO5251465A1 (es) 2000-01-26 2003-02-28 Pfizer Prod Inc Composiciones y procedimientos para tratar la osteoporosis y reducir el colesterol
US7283844B2 (en) 2000-04-04 2007-10-16 Thompson Scott D Multi-beam antenna wireless network system
US6550807B1 (en) * 2000-11-21 2003-04-22 Trw Vehicle Safety Systems Inc. Air bag module with electronically modulated vent
SE0100172D0 (sv) 2001-01-22 2001-01-22 Ulf Nilsson New inhibitors against galectins
US6914055B2 (en) * 2001-03-27 2005-07-05 Anatole Klyosov Delivery of a therapeutic agent in a formulation for reduced toxicity
US6680306B2 (en) 2001-06-21 2004-01-20 Glycogenesys, Inc. Method for enhancing the effectiveness of cancer therapies
US6964761B1 (en) 2001-12-12 2005-11-15 New York University Method of treating idiopathic pulmonary fibrosis with aerosolized IFN-γ
US6986995B2 (en) 2002-02-28 2006-01-17 Prometheus Laboratories, Inc. Methods of diagnosing liver fibrosis
US20040091503A1 (en) 2002-08-20 2004-05-13 Genitrix, Llc Lectin compositions and methods for modulating an immune response to an antigen
CA2498739A1 (fr) 2002-09-12 2004-03-25 Pharmexa A/S Immunisation contre la ghreline autologue
US20040254419A1 (en) 2003-04-08 2004-12-16 Xingwu Wang Therapeutic assembly
US20050053664A1 (en) * 2003-09-08 2005-03-10 Eliezer Zomer Co-administration of a polysaccharide with a chemotherapeutic agent for the treatment of cancer
US20060024357A1 (en) 2004-05-12 2006-02-02 Medivas, Llc Wound healing polymer compositions and methods for use thereof
CA2547573A1 (fr) 2003-12-05 2005-06-23 Warner-Lambert Company Llc Pyrroles de n-alkyl comme inhibiteurs de l'hmg-coa-reductase
US20050282773A1 (en) * 2004-01-14 2005-12-22 Pro-Pharmaceuticals, Inc. Modified polysaccharides in combination with anti-cancer drugs for enhanced treatment of cancer
WO2005079314A2 (fr) * 2004-02-13 2005-09-01 Pro-Pharmaceuticals, Inc. Compositions et procedes utilises pour traiter l'acne et la candida
US20070258969A1 (en) * 2004-03-19 2007-11-08 Eliezer Zomer Compositions and methods for targeting metastatic tumors using multivalent ligand-linked carbohydrate polymers
US8529889B2 (en) 2004-06-29 2013-09-10 Massachusetts Institute Of Technology Methods and compositions related to the modulation of intercellular junctions
WO2006017417A2 (fr) 2004-08-02 2006-02-16 Pro-Pharmaceuticals, Inc. Compositions et procedes permettant d'ameliorer l'efficacite d'une chimiotherapie a l'aide de cytotoxines microbiennes
WO2008011216A2 (fr) * 2006-05-16 2008-01-24 Pro-Pharmaceuticals, Inc. Polysaccharides à dents de galactose dans une formulation pour des thérapies antifibrotiques

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5733572A (en) * 1989-12-22 1998-03-31 Imarx Pharmaceutical Corp. Gas and gaseous precursor filled microspheres as topical and subcutaneous delivery vehicles
US6413494B1 (en) * 1998-07-23 2002-07-02 Samyang Corporation Composition and pharmaceutical dosage form for colonic drug delivery using polysaccharides
US7166299B2 (en) * 1998-07-24 2007-01-23 Seo Hong Yoo Preparation of aqueous clear solution dosage forms with bile acids
US20020068077A1 (en) * 2000-09-25 2002-06-06 Anatole Klyosov Methods and compositions for reducing side effects in chemotherapeutic treatments
US6642205B2 (en) * 2000-09-25 2003-11-04 Pro-Pharmaceuticals, Inc. Methods and compositions for reducing side effects in chemotherapeutic treatments
US6645946B1 (en) * 2001-03-27 2003-11-11 Pro-Pharmaceuticals, Inc. Delivery of a therapeutic agent in a formulation for reduced toxicity
US6982255B2 (en) * 2001-03-27 2006-01-03 Anatole Klyosov Delivery of a therapeutic agent in a formulation for reduced toxicity
US7012068B2 (en) * 2001-03-27 2006-03-14 Pro-Pharmaceuticals, Inc. Co-administration of a polysaccharide with a chemotherapeutic agent for the treatment of cancer
US20060057131A1 (en) * 2001-12-20 2006-03-16 Eric Simard Malleable protein matrix and uses thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023250141A3 (fr) * 2022-06-24 2024-04-11 Enalare Therapeutics Inc. Méthodes de traitement d'insuffisance ventilatoire neurologique

Also Published As

Publication number Publication date
AU2004272022B2 (en) 2010-10-07
CN1867252A (zh) 2006-11-22
WO2005025501A3 (fr) 2005-05-19
AU2004272022A1 (en) 2005-03-24
BRPI0413410A (pt) 2006-10-10
US20110077217A1 (en) 2011-03-31
AU2010257400B2 (en) 2012-01-12
JP2007505041A (ja) 2007-03-08
US7893252B2 (en) 2011-02-22
US20080207516A1 (en) 2008-08-28
IL174121A (en) 2014-11-30
IL174121A0 (en) 2006-08-01
WO2005025501A2 (fr) 2005-03-24
EP1662874A4 (fr) 2012-08-01
AU2010257400A1 (en) 2011-01-20
EP1662874A2 (fr) 2006-06-07

Similar Documents

Publication Publication Date Title
US7893252B2 (en) Selectively depolymerized galactomannan polysaccharide
Zhao et al. Challenges and potential for improving the druggability of podophyllotoxin-derived drugs in cancer chemotherapy
EP2127652B1 (fr) Procédé de traitement d'un cancer utilisant un agent anti-cancer en combinaison
MXPA06013559A (es) Tratamiento de linfoma de celula t usando 10-propargil-10-deazaaminopterina.
WO2019217164A1 (fr) Compositions et méthodes de traitement du cancer et d'autres maladies
JP5916040B2 (ja) メトトレキサートをオロチン酸誘導体として投与することでその副作用と毒性を下げる組成物及び方法
WO2008134528A1 (fr) Compositions de conjugués d'agent anticancéreux-acide hyaluronique et procédés
EP1383516B1 (fr) Co-administration d'un polysaccharide avec un agent chimiotherapeutique permettant le traitement du cancer
US20130252904A1 (en) Compositions and methods for treating cancer
WO2015125641A1 (fr) Composition pharmaceutique contenant un dérivé de camptothécine polymérisé, et dérivé inhibiteur de hsp90 polymérisé
WO2019072014A1 (fr) FORMULATION CONTENANT UN COMPOSÉ 5α-ANDROSTANE A-DÉCARBONISÉ POUR AUGMENTER LE NOMBRE DES GLOBULES BLANCS, ET SON UTILISATION
WO2008109349A1 (fr) Schéma posologique amélioré servant au traitement du cancer avec du 5-fluorouracile, du 5,10-méthylènetétrahydrofolate et de la capécitabine
EP2301556A1 (fr) Co-administration d'un polysaccharide avec un agent chimiotherapeutique dans le traitement du cancer
KR101560339B1 (ko) 의약 조성물 또는 조합제
Di Lauro et al. Epirubicin, cisplatin and docetaxel combination therapy for metastatic gastric cancer
KR102265488B1 (ko) 암 화학 요법시의 부작용 경감제
JP2007531697A (ja) 疎水性薬剤のデリバリーのための組成物と方法
WO2015125640A1 (fr) Composé polymère auquel sont liés un dérivé de camptothécine et un inhibiteur de hsp90, et son utilisation
EP3965734A1 (fr) Méthode destinée à augmenter le taux d'hémoglobine d'un patient atteint de cancer
WO2011016811A1 (fr) Procédés pour réduire l'incidence d'une mucosité induite par chimiothérapie
CN111728960B (zh) 富马酸比索洛尔联合多西他赛在制备抗肿瘤药物中的应用
CN114712353A (zh) 烟酰胺及含有其的组合物的制药用途
CN110314159A (zh) 酪氨酸激酶抑制剂联合治疗肿瘤疾病的用途
JP2006518355A (ja) インドロピロロカルバゾール誘導体及び他の抗腫瘍薬を含む併用療法
WO2010019396A1 (fr) Administration de glufosfamide pour le traitement du cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: PRO-PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZOMER, ELI;PLATT, DAVID;REEL/FRAME:014947/0718

Effective date: 20031222

AS Assignment

Owner name: PRO-PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KLYOSOV, ANATOLE;REEL/FRAME:016811/0436

Effective date: 20050912

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: 10X FUND, LP, FLORIDA

Free format text: SECURITY AGREEMENT;ASSIGNOR:PRO-PHARMACEUTICALS, INC.;REEL/FRAME:023134/0968

Effective date: 20090212

AS Assignment

Owner name: GALECTIN THERAPEUTICS, INC., GEORGIA

Free format text: RELEASE BY SECURED PARTY;ASSIGNOR:10X FUND L.P.;REEL/FRAME:050399/0679

Effective date: 20190729