EP1879597A2 - Méthodes permettant de réduire la gravité de la mucosite orale et gastro-intestinale - Google Patents

Méthodes permettant de réduire la gravité de la mucosite orale et gastro-intestinale

Info

Publication number
EP1879597A2
EP1879597A2 EP06831669A EP06831669A EP1879597A2 EP 1879597 A2 EP1879597 A2 EP 1879597A2 EP 06831669 A EP06831669 A EP 06831669A EP 06831669 A EP06831669 A EP 06831669A EP 1879597 A2 EP1879597 A2 EP 1879597A2
Authority
EP
European Patent Office
Prior art keywords
day
mucositis
patient
group
treatment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP06831669A
Other languages
German (de)
English (en)
Inventor
Tetsu Kawano
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.)
Eisai R&D Management Co Ltd
Original Assignee
Eisai Co Ltd
Eisai R&D Management Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eisai Co Ltd, Eisai R&D Management Co Ltd filed Critical Eisai Co Ltd
Publication of EP1879597A2 publication Critical patent/EP1879597A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/665Phosphorus compounds having oxygen as a ring hetero atom, e.g. fosfomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/136Amines having aromatic rings, e.g. ketamine, nortriptyline having the amino group directly attached to the aromatic ring, e.g. benzeneamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/7036Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin having at least one amino group directly attached to the carbocyclic ring, e.g. streptomycin, gentamycin, amikacin, validamycin, fortimicins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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 invention relates to methods for reducing the severity of oral and gastrointestinal mucositis.
  • Mucositis is a condition characterized by swelling, irritation, and discomfort of mucosal linings such as those of the gastrointestinal tract and the oral and oralpharyngeal cavities, and can result in mouth and throat sores, diarrhea, abdominal cramping and tenderness, and rectal ulcerations.
  • This condition occurs in approximately half of all cancer patients, and is a common side effect of cancer treatments involving radiation and/or chemotherapy.
  • the goal of these approaches to cancer treatment is to kill rapidly dividing cancer cells but, unfortunately, other rapidly dividing cells are killed by the treatment as well, including cells that line regions such as the gastrointestinal tract, leading to mucositis.
  • Mucositis Symptoms of mucositis generally occur five to ten days after the start of cancer treatment, and can take two to four weeks after cessation of treatment to clear. The incidence of mucositis, as well as its severity, depends on factors such as the type and duration of the cancer treatment. Mucositis occurs, for example, in virtually all patients who are treated by irradiation of the head and neck. It is also highly prevalent in patients treated with high dose chemotherapy and/or irradiation for the purpose of myeloablation, in preparation for stem cell or bone marrow transplantation.
  • Mucositis adversely impacts the quality of life of cancer patients in several ways.
  • the mouth and throat sores of mucositis can cause significant pain and make it difficult to eat, drink, and even take oral medication.
  • Mucositis is also accompanied by a severe risk of infection, as it can lead to a breach in the otherwise protective linings of the oral mucosa and gastrointestinal tract, which are colonized by a vast array of microorganisms.
  • efforts to counter the discomforts of mucositis can lead to disruptions in cancer treatment, alterations in treatment dosages, or shifting to different modes of treatment. Severe mucositis can also lead to the need for parenteral nutrition or hospitalization.
  • the development of effective approaches to preventing and treating mucositis is therefore important for improving the care of cancer patients.
  • the invention provides methods of reducing the severity of oral or gastrointestinal mucositis in patients.
  • the methods include a step of administering to the patients a composition containing one or more compounds that block activation of toll-like receptor 4 (TLR4), such as a lipid A analog, which may be within the formula:
  • R.1 is selected from the group consisting of:
  • each J, K, and Q independently, is straight or branched Cl to Cl 5 alkyl;
  • L is O, NH, or CH 2 ;
  • M is O or NH;
  • G is NH, O 5 S, SO, or SO 2 ;
  • R2 is straight or branched C5 to C15 alkyl
  • Ry is selected from the group consisting of straight or branched C5 to Cl 8 alkyl
  • E is NH, O, S, SO, or SO 2 ; each A, B, and D, independently, is straight or branched Cl to C15 alkyl;
  • R4 is selected from the group consisting of straight or branched C4 to C20 alkyl
  • each U and V independently, is straight or branched C2 to Cl 5 alkyl and W is hydrogen or straight or branched Cl to C5 alkyl;
  • R A is R.5 or R ⁇ -O-CH 2 -, R ⁇ being selected from the group consisting of hydrogen, J', -J'-OH, -J'-O-K', -J'-O-K'-OH, and -J' -0-PO(OH) 2 , where each J' and K', independently, is straight or branched Cl to C5 alkyl;
  • R6 is selected from the group consisting of hydroxy, halogen, Cl to C5 alkoxy and Cl to C5 acyloxy;
  • Al and A ⁇ are selected from the group consisting of
  • Z is straight or branched Cl to ClO alkyl; or a pharmaceutically acceptable salt or phosphate ester thereof.
  • One aspect of the invention includes phosphate esters of the above-noted formula, wherein at least one of the hydroxyl groups of A 1 or A 2 can be substituted to form a phosphate ester.
  • Lipid A analog that can be included in the compositions of the invention is a compound having the following structure:
  • the compound is of the following structure:
  • Patients that can be treated according to the invention include those who have oral or gastrointestinal mucositis, hi addition, patients who do not have, but are at risk of developing, oral or gastrointestinal mucositis can be treated according to the invention, hi the latter group of patients, the treatment can inhibit or prevent the development of mucositis.
  • Treatments that may cause or place a patient at risk of developing oral or gastrointestinal mucositis are radiation therapy and chemotherapy, as described further elsewhere herein or in the background section.
  • Patients that can be treated according to the invention thus include, for example, cancer patients, as well as patients that have recently been, will shortly be, or are currently subject to treatment with head or neck irradiation, or stem cell or bone marrow transplantation.
  • compositions used in the invention can be administered to a patient prior to, concurrently with, or after a treatment that has induced or places the patient at risk of developing oral or gastrointestinal mucositis, or a combination of these approaches can be used, hi an example, the composition is administered at the same time as, within 1-4 hours of, or on the same day as the treatment, and then for 1-3 (e.g., 1-2) days thereafter (e.g., 1-2 times per day).
  • 1-3 e.g., 1-2 days thereafter
  • compositions can be administered to patients by any acceptable manner known in the art, including topically (e.g., by gel, rinse, lozenge, cream, ointment, or patch), by intravenous infusion, orally (e.g., by tablet, capsule, lozenge, cream, ointment, or patch), rectally (e.g., by suppository, ointment, or enema), or vaginally (e.g., by cream, ointment, gel, or suppository).
  • treatment according to the invention can be carried out in combination with other approaches to treating mucositis, including antimicrobial and palliative treatments, as is discussed further below.
  • the invention also includes use of the compositions and compounds described herein in the preparation of medicaments for reducing the severity of oral or gastrointestinal mucositis.
  • medicaments can be used to treat patients who already have mucositis, in an effort to reduce the symptoms of the condition (partially or fully), to prevent the condition from worsening, and/or to reduce the level of worsening of the condition.
  • the medicaments can also be used with patients who do not yet have, but are at risk of developing, mucositis. As is discussed elsewhere herein, such patients include cancer patients who are scheduled to receive, are currently receiving, or have previously received cancer treatment involving radiation and/or chemotherapy.
  • administration of the medicaments can be carried out to reduce the severity of mucositis, to inhibit its development, or to prevent mucositis from occurring.
  • the compounds included in such medicaments can be any of those made reference to elsewhere herein, as well as compounds falling within the formulae provided elsewhere herein. A specific example of such a compound is as follows:
  • compositions including the compounds described herein, formulated for administration for reducing the severity of mucositis as described herein.
  • these compositions can include the compounds in formulations such as gels for topical administration, rinses, tablets, capsules, chewing gum, lozenges, creams, ointments, enemas, suppositories, or patches.
  • the invention provides several advantages. For example, in providing approaches to reducing the severity of mucositis, an uncomfortable side effect of treatments such as radiation and chemotherapy, the methods of the invention can contribute to the well being of patients as they face the challenges of such treatments. Further, the methods of the invention can decrease the incidence of infection, which is a common consequence of mucositis, hi addition, in providing increased comfort to patients, the methods of the invention can lead to increased compliance of patients with their therapeutic regimens, and also can contribute to increasing the speed of their recovery. Other features and advantages of the invention will be apparent from the following detailed description, the drawings, and the claims.
  • Figure 1 is a graph showing the percent weight change of C3H/HeOuJ and C3H/HeJ mice after snout irradiation treatment. Animals were weighed daily, the percent weight change from day 0 was calculated, and group means and standard errors of the mean (SEM) calculated for each day.
  • Figure 3 is a graph showing the mean serum IL-6 concentration of snout irradiation-treated C3H/HeOuJ and C3H/HeJ mice measured by ELISA analysis at the indicated time points.
  • Figure 4 is a graph showing the mean serum TNF- ⁇ concentration of snout irradiation-treated C3H/HeOuJ and C3H/HeJ mice measured by ELISA analysis at the indicated time points.
  • Figure 5 is a graph of epithelial histology scores for snout irradiation-treated C3H/HeOuJ and C3H/HeJ mice. Each sample was scored on a scale of 0-3 for epithelial cell layer damage.
  • Figure 6 is a graph of connective tissue histology scores for snout irradiation- treated C3H/HeOuJ and C3H/HeJ mice. Each sample was scored on a scale of 0-3 for connective tissue damage.
  • Figure 7 is a graph showing the mean numbers of inflammatory cells of snout irradiation-treated C3H/HeOuJ and C3H/HeJ mice measured at the indicated time points.
  • Figure 8 is a graph showing the mean numbers of mitoses in the epithelial cell layer of snout irradiation-treated C3H/HeOuJ and C3H/HeJ mice measured at the indicated time points.
  • Figure 9 is a graph showing the mean numbers of blood vessels per 10 high power fields of snout irradiation-treated C3H/HeOuJ and C3H/HeJ mice measured at the indicated time points.
  • Figure 10 is a graph showing the mean number of large blood vessels as a percentage of total blood vessels per 10 high power fields for C3H/HeOuJ and
  • C3H/HeJ mice measured at the indicated time points. The values are expressed as a percentage of the total numbers of blood vessels observed in those fields.
  • Figure 11 is a graph showing percent weight change of C3H/HeOuJ mice treated with the indicated amounts of eritoran, after snout irradiation treatment. The animals were weighed daily, the percent weight change from day 0 was calculated, and group means and standard errors of the mean (SEM) calculated for each day.
  • Figure 13 is a graph showing the minimum number of epithelial cell layers on the dorsal surface of the tongue for snout irradiation-treated C3H/HeOuJ mice treated with the indicated amounts of eritoran, at the indicated timepoints.
  • Figure 14 is a graph showing the maximum number of epithelial cell layers on the dorsal surface of the tongue for snout irradiation-treated C3H/HeOuJ mice treated with the indicated amounts of eritoran, at the indicated timepoints.
  • Figure 15 is a graph showing the minimum number of epithelial cell layers on the ventral surface of the tongue for snout irradiation-treated C3H/HeOuJ mice treated with the indicated amounts of eritoran, at the indicated timepoints.
  • Figure 16 is a graph showing the maximum number of epithelial cell layers on the ventral surface of the tongue for snout irradiation-treated C3H/HeOuJ mice treated with the indicated amounts of eritoran, at the indicated timepoints.
  • Figure 17 is a graph showing the percent weight change of animals treated with eritoran (E5564) under the indicated regimens, as well as un-irradiated and placebo controls. Animals were weighed daily, the percent weight change from day 0 was calculated, and group means and standard errors of the mean (SEM) calculated for each day.
  • SEM standard errors of the mean
  • Figure 19 is a graph showing the percent weight change of animals treated according to the regimens indicated in the figure. Data are shown for animals surviving until the end of the study only. Animals were weighed daily, the percent ( weight change from day 0 was calculated, and group means and standard errors of the mean (SEM) calculated for each day.
  • Figure 21 is a graph showing mean epithelial scores and standard errors of the mean for each of the indicated groups.
  • Figure 22 is a graph showing mean connective tissue scores and standard errors of the mean for each of the indicated groups.
  • Figure 23 is a graph showing mean inflammation scores and standard errors of the mean for each of the indicated groups.
  • Figure 24 is a graph showing mean number of mitoses per 10 hpf and standard errors of the means for each of the indicated groups.
  • Figure 25 is a graph showing the mean percent ulceration and standard error of the mean for each of the indicated groups.
  • Figure 26 is a graph showing the mean number of inflammatory cells per 10 hpf and standard errors of the means for each of the indicated groups.
  • Figure 27 is a graph showing the percentage of the infiltrating inflammatory cells that were neutrophils for each sample and the mean and standard deviation for each of the indicated groups.
  • Figure 28 is a graph showing the percentage of the infiltrating inflammatory cells that were lymphocytes for each sample and the mean and standard deviation for each of the indicated groups.
  • Figure 29 is a graph showing the percentage of the infiltrating inflammatory cells that were monocytes or macrophage for each sample and the mean and standard deviation for each of the indicated groups.
  • Figure 30 is a graph showing the number of small blood vessels per 10 hpf and the mean and standard errors of the means for each of the indicated groups.
  • Figure 31 is a graph showing the number of medium blood vessels per 10 hpf and the means and standard errors of the means for each of the indicated groups.
  • Figure 32 is a graph showing the number of large blood vessels per 10 hpf and the means and standard errors of the means for each of the indicated groups.
  • Figure 33 is a graph showing the number of mast cells per 10 hpf and the means and standard errors of the means for each of the indicated groups.
  • Figure 34 is a graph showing serum TNF- ⁇ levels measured using an ELISA assay and the mean and standard error of the mean for each of the indicated groups.
  • Figure 35 is a graph showing serum IL-6 levels measured using an ELISA assay and the mean and standard error of the mean for each of the indicated groups.
  • Figure 36 is a graph showing serum SAA levels measured using an ELISA assay and the mean and standard error of the mean for each of the indicated groups.
  • the present invention provides methods of reducing the severity of oral or gastrointestinal mucositis.
  • the methods can be used to treat patients who already have mucositis.
  • the methods can also be carried out with patients who do not have, but are at risk of developing mucositis (e.g., cancer or other patients scheduled to receive, currently receiving, or previously treated with radiation and/or chemotherapy).
  • treatment according to the invention can reduce the severity of mucositis resulting from their cancer treatment, inhibit the development of mucositis, or prevent mucositis.
  • TLR4 toll-like receptor 4
  • LPS lipopolysaccharide
  • TLR4 receptor activation is blocked by administration of a TLR4 antagonist, leading to beneficial effects in the reduction of severity of mucositis.
  • treatment according to the invention may block the effects of heat shock proteins (HSP's) in mucositis.
  • HSP's heat shock proteins
  • HSP60, 70, or 90 may be endogenous ligands of TLR4, and thus may play a role in the mucositis induced by radiation therapy.
  • TLR4 antagonists used in the methods of the invention can be, for example, analogs of the lipid A region of LPS, such as lipid A analogs that are within the formula set forth above, in the Summary of the Invention.
  • An example of a Lipid A analog that can be included in the compositions of the invention is a compound having the following structure: or a pharmaceutically acceptable salt or phosphate ester thereof.
  • the compound is of the following structure:
  • eritoran also known as compound E5564, compound 1287, and SGEA
  • U.S. Patent No. 5,935,938 is described in U.S. Patent No. 5,935,938.
  • TLR4 antagonists that can be used in the invention include, for example, compound B531 (U.S. Patent No. 5,530,113), as well as other compounds described in the following patents: U.S. Patent No. 5,935,938; U.S. Patent No.
  • a TLR4 antagonist is administered to a patient before, during, and/or after treatment with a therapy that causes oral or gastrointestinal mucositis or puts the patient at risk of developing such mucositis.
  • treatments include radiation and chemotherapy, which act by blocking the growth of rapidly dividing cells, such as cancer cells and epithelial cells that line the surfaces of the gastrointestinal, respiratory, and genitourinary tracts.
  • Treatments that can lead to mucositis include radiation treatment (e.g., head and/or neck, whole body, targeted, and/or hyperfractionated radiation), as well as chemotherapeutic regimens used in the treatment of, or as adjuvant treatments for, conditions such as breast cancer, colon cancer, gastric cancer, genitourinary (e.g., bladder, prostate, or testicular) cancer, gynecologic (e.g., cervical, endometrial, ovarian, or uterine) cancer, head and neck/esophageal cancer, leukemia, lung (small cell or non small-cell) cancer, lymphoma (Hodgkin's or non-Hodgkin's), melanoma, multiple myeloma, pancreatic cancer, and sarcoma.
  • radiation treatment e.g., head and/or neck, whole body, targeted, and/or hyperfractionated radiation
  • chemotherapeutic regimens used in the treatment of, or as adjuvant treatments for
  • cancers such as these can be treated using approaches involving immunotherapy by use of agents such as, for example, rituximab, cetuximab, or bevacizumab, alone or in combination with chemotherapy or radiation therapy.
  • chemotherapeutic approaches that may induce mucositis include those utilizing (either as single agents or in combinations) platinum derivatives such as carboplatin, cisplatin, and oxaplatin; mitosis inhibitors such as paclitaxel, docetaxel, vinorelbine, vincristine, and vinblastine; topoisomerase inhibitors such as etoposide, irinotecan, and topotecan; antimetabolites such as gemcitabine, capecitabine, fludarabine, methotrexate, 5-fluorouracil, cladribine, pentostatin, and cytarabine; DNA synthesis inhibitors such as doxorubicin, epirubicin, idar
  • TLR4 antagonists such as those noted above can be administered using standard methods including, for example, topical approaches and intravenous infusion.
  • the particular approach and dosage used for a particular patient depends on several factors including, for example, the type of cancer treatment, the location(s) of any discomfort, and the general health of patient. Based on factors such as these, a medical practitioner can select an appropriate approach.
  • Treatment according to the invention can begin prior to cancer treatment (e.g., 1-2 days or up to 1 week prior to cancer treatment), at or near the same time as cancer treatment (e.g., simultaneously with, within 1-4 hours of, or on the same day as cancer treatment), or shortly after the cessation of cancer treatment (e.g., within 1-4 days of cessation, and/or prior to or upon appearance of symptoms). Treatment can then be maintained, for example, until any symptoms of mucositis have substantially cleared or the risk of developing such symptoms has passed.
  • treatment started before or at or near the same time as cancer treatment can be maintained, e.g., for 1-3, e.g., 1-2 days, hi other examples, treatment is maintained for 1-4 or 2-3 weeks following the cessation of cancer treatment, as determined to be appropriate by one of skill in the art.
  • the treatment according to the present invention is carried out prior to cancer treatment only; prior to and concurrently with cancer treatment only; prior to, concurrently with, and after cessation of cancer treatment; concurrently with cancer treatment only; concurrently with and after cessation of cancer treatment only; after cessation of cancer treatment only; or prior to and after cessation of cancer treatment only.
  • treatment according to the methods of the invention can be altered, stopped, or re-initiated in a patient, depending on the status of any symptoms of mucositis. Treatment can be carried out at intervals determined to be appropriate by those of skill in the art. For example, the administration can be carried out 1, 2, 3, or 4 times/day.
  • a TLR4 antagonist in the case of patients having or at risk of developing mucositis in the oral cavity, can be administered to the oral cavity in the form of a gel, paste, spray, cream, ointment, or patch that is applied to affected or at risk areas.
  • Such patients can also be treated by the use of an oral rinse, chewing gum, or lozenge including the drug.
  • the drug can be administered to patients affected in rectal or vaginal areas by use of formulations in the form of gels, creams, ointments, suspensions, or suppositories.
  • administration can be by use of an enema, hi another example, in the case of patients affected in the nasal cavity, the drug can be administered by topical administration, as described herein, or by inhalation of the drug (see, e.g., U.S. Patent No. 6,683,063).
  • the drug can be administered by injection (e.g., local injection), or by infusion (intravenous or intra-arterial), as discussed further below.
  • This approach to administration may be particularly convenient in the case of patients who already have a catheter in place for the administration of chemotherapeutic or other drugs.
  • Examples of such approaches, in which the drug administered is eritoran (see above) and the indicated amounts of the drug are based on an assumed average weight of a subject of 70 kg, are as follows.
  • the drug can be administered at a low dosage by continuous intravenous infusion.
  • the drug can be administered continuously at a rate of 10-500 (e.g., 50-400 or 100-200) ⁇ g/hour over the course of the treatment.
  • the drug in which a patient requires longer-term care, can be administered intermittently (e.g., every 12-24 hours) at a dosage of, for example, 0.1-20 (e.g., 1-8, 2-7, 3-6, or 4-5) mg/hour for 2-6 (e.g., approximately 4) hours.
  • the initial or loading dose is followed by maintenance doses that are less than (e.g., half) the loading dose or by continuous infusion as described above in the first example.
  • TLR4 antagonists such as eritoran
  • US-2003-0105033-A1 bolus or intermittent infusion
  • WO 00/41703 continuous infusion
  • devices and equipment e.g., catheters, such as central or peripheral venous catheters, tubing, drip chambers, flashback bulbs, injection Y sites, stopcocks, and infusion bags
  • catheters including a chlorhexidine-based antimicrobial coating have been found to disrupt the size of the micelles of the drug that are formed during formulation, leading to inadequate concentrations in blood.
  • a non-chlorhexidine-based antimicrobial coating such as an antimicrobial coating that includes one or more other antibiotics, such as rifampin or minicyclin.
  • kits that include one or more TLR4 antagonists (e.g., a Lipid A analog as described above, e.g., the compound eritoran) and instructions to use the drug in the methods described herein.
  • TLR4 antagonists e.g., a Lipid A analog as described above, e.g., the compound eritoran
  • the kits can also optionally include devices or equipment used in administration (e.g., a catheter lacking a chlorhexidine coating) and/or a solution for administering the drug, such as a 5% dextrose (e.g., glucose) solution.
  • a 5% dextrose e.g., glucose
  • the methods of the invention can be carried out in combination with antimicrobial or antifungal therapies, e.g., therapies involving administration of antibiotics such as nystatin, amphotericin, acyclovir, valacyclovir, clotimazole, and fluconazole.
  • antimicrobial or antifungal therapies e.g., therapies involving administration of antibiotics such as nystatin, amphotericin, acyclovir, valacyclovir, clotimazole, and fluconazole.
  • antibiotics such as nystatin, amphotericin, acyclovir, valacyclovir, clotimazole, and fluconazole.
  • patients with head and neck cancer receiving radiotherapy have colonization of the oropharyngeal region with gram-negative bacteria.
  • Selective decontamination of the oral cavity with anti-microbial agents has the benefit of reducing oral mucositis associated with radiation therapy, but there may be limitations to the beneficial effects of such treatment
  • endotoxin is a potent mediator of inflammation, it may contribute to the aggravation of mucositis and, thus, co-treatment with an antiendotoxin compound (e.g., a Lipid A analog, such as eritoran) and antibiotics can be used as a more effective approach to treating oral mucositis in such patients, according to the invention.
  • an antiendotoxin compound e.g., a Lipid A analog, such as eritoran
  • antibiotics can be used as a more effective approach to treating oral mucositis in such patients, according to the invention.
  • the methods of the invention can also be used in conjunction with palliative therapies including the use of topical rinses, gels, or ointments that include lidocaine, articaine, and/or morphine, as well as other analgesic or anti-inflammatory agents.
  • agents and approaches that can be used in combination with TLR4 antagonists, according to the methods of the invention, include the following: palifermin (recombinant keratinocyte growth factor; rHuKGF; KepivanceTM; Amgen) and AES- 14 (uptake-enhanced L-glutamine suspension)(Peterson, J. Support Oncol. 4(2 Suppl.
  • flurbiprofen e.g., administered as a tooth patch; Stokman et al., Support Care Cancer 13(l):42-48, 2005); diphenhydramine, magnesium hydroxide/aluminum hydroxide, nystatin, and corticosteroids (Chan et al., J. Oncol. Pharm. Pract.
  • oral transmucosal fentanyl citrate e.g., administered in the form of a lozenge; Shaiova et al., Support Care Cancer 12(4):268-273, 2004
  • clonazepam e.g., in the form of a tablet
  • Gremeau-Richard et al. Pain 108(102):51-57, 2004
  • capsaicin e.g., in the form of a lozenge; Okuno et al., J. Cancer Integr. Med. 2(3):179-183, 2004
  • ketamine e.g., in the form of an oral rinse; Slatkin et al., Pain Med.
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • G-CSF granulocyte colony- stimulating factor
  • the present invention is based, in part, on the following experimental results.
  • C3H/HeJ and C3H/HeOuJ Two strains of C3H mice (C3H/HeJ and C3H/HeOuJ) differ from one another by the presence or absence of the LPS receptor TLR4 (present in the C3H/HeOuJ strain).
  • C3H/HeJ mice are more sensitive to the lethal effects of total body radiation, but do not develop oral mucositis to the same ex ⁇ ent as do the C3H/HeOuJ mice after a localized acute radiation to the snout. The mechanistic basis for these differences is not understood.
  • mice The acute mouse snout radiation model in mice has been used to determine the radioprotective properties of experimental compounds.
  • the course of oral mucositis in this model is well defined and results in peak mucositis 10-12 days following radiation.
  • the acute model has little systemic toxicity, resulting in few radiation- induced animal deaths.
  • mice were used. Fifty-six (56) mice (28 each).
  • mice 4 C3H/HeOuJ and 4 C3H/HeJ were used as the no radiation control animals. Animals were sacrificed and blood and tissue taken according to the schedule described in Table 1.
  • mice 32 C3H/HeOuJ and 32 C3H/HeJ were used.
  • the mice were randomized into four (4) groups of either 28 animals (groups 1 and 2), the radiated groups, or 4 animals (groups 3 and 4), the un-irradiated control groups, as described in Table 2.
  • each animal was weighed. Animals in groups 1 and 2 received a single dose of 30 Gy radiation focused on the snout on day 0. A lead shield protected the remainder of the animal body. At 2 hours, 6 hours, 24 hours (1 day), 3 days, 6 days, 10 days, and 14 days after radiation, 4 animals from groups 1 and 2 were sacrificed and blood and tissue were collected as described below. Animals in groups 3 and 4 were sacrificed, the tongues dissected, and blood collected on day 1. The tongues from each animal were dissected into 3 pieces (anterior, middle, and posterior) and each tongue was fixed in formalin.
  • Mucositis was assayed by histological analysis of hematoxylin and eosin (H&E) stained sections of the formalin fixed tongues. Mucositis scoring was done in a blinded manner according to a validated scale. Serum samples were assayed for the cytokines TNF- ⁇ and IL-6 using a standard ELISA assay.
  • C3H/HeOuJ and C3H/HeJ mice (Jackson Laboratories), aged 5 to 6 weeks with body weights of 22.3 g, were used. Animals were individually numbered using an ear punch and housed in small groups of approximately 5 animals per cage. Animals were acclimatized prior to study commencement. During this period of at least 2 days, the animals were observed daily in order to reject animals that presented in poor condition.
  • the study was performed in animal rooms provided with filtered air at a temperature of 7O 0 F +/-5 0 F and 50% +/-20% relative humidity.
  • Animal rooms were set to maintain a minimum of 12 to 15 air changes per hour.
  • the room was on an automatic timer for a light/dark cycle of 12 hours on and 12 hours off with no twilight.
  • Bed-O-Cobs® bedding was used, and was changed a minimum of once per week.
  • Cages, tops, bottles, etc. were washed with a commercial detergent and allowed to air dry. Prior to use, these items were wrapped and autoclaved.
  • a commercial disinfectant was used to disinfect surfaces and materials introduced into the hood.
  • Floors were swept daily and mopped a minimum of twice weekly with a commercial detergent.
  • Walls and cage racks were sponged a minimum of once per month with a dilute bleach solution.
  • a cage card or label with the appropriate information necessary to identify the study, dose, animal number, and treatment group was placed on all cages. The temperature and relative humidity was recorded during the study, and the records retained.
  • mice were randomly and prospectively divided into four (4) treatment groups prior to irradiation. Each animal was identified by an ear punch corresponding to an individual number. A cage card was used to identify each cage or label marked with the study number, treatment group number, and animal numbers.
  • Machine calibration was checked within two weeks of the onset of the study.
  • a single dose of radiation (30 Gy/dose) was administered to all animals in groups 1 and 2 on day 0.
  • Radiation was generated with a 160 kilo volt potential (15-ma) source at a focal distance of 50 cm, hardened with a 0.35 mm Cu filtration system.
  • Irradiation was done at a rate of 121.5 cGy/minute. Animals were anesthetized prior to radiation, and placed under lead shielding such that only the snout was exposed
  • Animals in groups 3 and 4 were the un-irradiated control animals. The measurements from these animals provided a baseline control for all the irradiated samples in this study.
  • the 4 animals in each of groups 3 and 4 were sacrificed on day 1. Animals from groups 1 and 2 were sacrificed at several time points during the course of the study. At each time point, 4 animals per group were sacrificed. The time points were 2 hours, 6 hours, 24 hours, 3 days, 6 days, 10 days, and 14 days after radiation.
  • each tongue was removed and dissected into three pieces.
  • the anterior third of each tongue was fixed in formalin for subsequent histological analysis.
  • the middle third of each tongue was extracted to provide mRNA for analysis of cytokine expression levels.
  • the posterior section of each tongue was flash frozen in liquid nitrogen and stored for future analysis.
  • Enzyme linked immunosorbent assays were performed for cytokines TNF- ⁇ and IL-6 using kits purchased from R and D systems. These kits were used in accordance with the manufacturer's instructions. AU determinations were made in duplicate on serum samples stored at -80°C. If insufficient serum had been collected to run both IL-6 and TNF- ⁇ , samples were diluted 1 :2 or 1 :4, and run in duplicate in both assays. All assays were performed using 50 ⁇ L of sample per well.
  • H&E hematoxylin and eosin
  • Serum levels of cytokines IL-6 and TNF- ⁇ were evaluated by ELISA.
  • the mean serum concentration of IL-6 was 1.0 pg/mL. This level increased to 88.8 pg/mL at 6 hours post radiation, before falling to 12.0 pg/mL on day 3 following radiation and increasing to a peak level of 122.7 pg/mL on day 6. Days 10 and 14 showed a gradual decline from the peak levels seen on day 6.
  • the mean serum concentration of IL-6 was 13.8 pg/mL. All other readings were between 25 and 42 pg/mL with the exception of the day 10 time-point, when serum JL-6 concentrations increased to 69.4 pg/mL. These data are shown in Figure 3.
  • H&E hematoxylin and eosin
  • the epithelium and connective tissue regions of each sample were each given separate scores.
  • the scores for the epithelium are shown in Figure 5.
  • the mean epithelial histological score for C3H/HeOuJ mice that had not been irradiated was 0, and this was also the case for all post-radiation time points except day 1, when the mean score was 0.25, and days 6 and 10, when the mean score was 2.
  • the mean epithelial histological score was 0 at all time points except day 6, when the score was 0.75.
  • the data for the mean connective tissue histological scores is shown in Figure 6.
  • the mean connective tissue histological score for C3H/HeOuJ mice that had not been irradiated was 2.
  • the mean number of inflammatory cells per ten high powered fields for each strain of mouse at each time point was calculated and the results are shown in Figure 7.
  • the numbers of cells seen in the connective tissue of un-irradiated animals was higher than expected in both strains of mice, and was lower at all post irradiation time-points in the C3H7HeJ mice, hi the C3H/HeOuJ mice, the numbers of inflammatory cells seen at most time points was also lower than those observed in unirradiated controls, except for day 10, when the number of cells was approximately 2 times higher than the un-irradiated controls (and about 10 times higher than the 2 hour and 6 hour time-points) and on day 14, when the numbers observed were about 50% higher than the un-irradiated controls.
  • the number of mitotic figures seen in the epithelial cell layer was counted and the mean number of mitoses per ten high power fields for each strain at each time point is shown in Figure 8.
  • the number of mitotic figures counted in the epithelial cell layer of C3H/HeOuJ mouse tongues was generally low, with a mean of 0.4 in unirradiated mice and numbers lower than this at all time points except day 6, when a mean of 2.75 was observed.
  • the numbers of mitoses seen in unirradiated mice was lower than in C3H/HeOuJ mice with a mean of 0.1. However, this increased to 0.4 by day 10 post radiation.
  • the number of blood vessels per ten high power fields was counted for each sample and the mean number for each strain of mouse at each time point calculated. These data are shown in Figure 9.
  • the number of blood vessels per 10 high power fields was 26.6 in un-irradiated C3H/HeOuJ mice and 27.2 in un-irradiated C3H/HeJ mice.
  • the number of blood vessels had apparently dropped to 4.5 by 2 hours post radiation, rising to 20.6 on day 1, before falling to 8.5 on day 3, and increasing on days 6 and 10, before reaching a peak of 33.7 on day 14. This represents an increase of ' 27% relative to the un-irradiated controls and 648% relative to the 2 hour time point.
  • Analysis of serum cytokine levels showed that the un-irradiated C3 HTHeJ control mice had higher levels than their C3H/HeOuJ counterparts, but that the C3 HTHeOuJ mice showed greater increases in serum cytokines following radiation than C3 HTHeJ, with peak levels of both IL-6 and TNF- ⁇ being seen on day 6 post irradiation. 3. Histologically, very little change was seen in C3HTHeJ mice.
  • C3H/HeOuJ mice showed a significant disturbance of the epithelium on days 6 and 10 following radiation. Histological scores for the connective tissue were high in the un-irradiated control C3H/HeOuJ mice and declined at 2 hours to 6 days post radiation, returning to near control levels at days 10 and 14 post radiation. 4. The numbers of inflammatory cells present showed little change in the
  • C3H/HeJ mice but increased to a peak on day 10 post radiation in C3H/HeOuJ mice, coinciding with the tissue peak tissue cytokine levels in these animals. Infiltrates were predominantly lymphocytic in nature.
  • Example I 5 two strains of C3H mice (C3H/HeJ and C3H/HeOuJ) differ from one another by the presence or absence of the LPS receptor TLR4 (present in the C3H/HeOuJ strain), hi the experiments described above, it is established that the C3H/HeOuJ strain is susceptible to oral mucositis induced by focal radiation to the snout, while the C3H/HeJ strain is relatively resistant to radiation induced mucositis. Evaluation of the pro-inflammatory cytokines in these animals showed that the induction of these cytokines via the LPS receptor TLR4 in the C3H/HeOuJ mice may play a role in the development of oral mucositis. The purpose of the study described below was to evaluate a compound that blocks stimulation of TLR4 (eritoran) in the murine model of oral mucositis.
  • mice The acute mouse snout radiation model in mice has been used to determine the radio-protective properties of experimental compounds.
  • the course of oral mucositis in this model is well defined and results in peak mucositis 10-12 days following radiation.
  • the acute model has little systemic toxicity, resulting in relatively few radiation induced animal deaths.
  • a dose of 30 Gy was used to induce oral mucositis.
  • mice A total of fifty-four (54) animals were used in this study. Forty-eight (48) C3H/HeOuJ mice were divided into 3 groups of 16 animals per group (groups 1-3). An additional 6 animals were put into a separate control group (group 4) as described in Table 2.
  • mice Fifty-four (54) male C3H/HeOuJ mice aged 6-7 weeks and weighing approximately 22 g were used. There were three (3) treatment groups of sixteen (16) animals each, and a control group of six (6) animals that received no radiation. All animals had a jugular cannula inserted into the left jugular vein on day -3. Beginning on day 0, animals in groups 1 and 4 were dosed twice a day by injection via cannula with placebo. Animals in group 2 were dosed with 2 FV injections of eritoran at 1 mg/kg daily, starting 2 hours or less before radiation on day 0 and continuing until day 10.
  • Animals in group 3 were dosed with 2 FV injections of eritoran at 10 mg/kg daily, starting 2 hours or less before radiation on day 0 and continuing until day 10. Animals in groups 1, 2, and 3 were given a single dose of 30 Gy radiation directed to the snout on day 0. The 6 animals in group 4 were used as the no radiation control animals (see Table 2). Eight animals in each of groups 1, 2, and 3 were sacrificed and blood and tissue taken according to the schedule described in Table 2.
  • C3H/HeOuJ mice (Jackson Laboratories), aged 5 to 6 weeks with body weight of 21.3 g, were used. Animals were individually numbered using an ear punch and individually housed. Animals were acclimatized prior to study commencement. During this period of at least 2 days, the animals were observed daily in order to reject animals that presented in poor condition.
  • Example II The study was performed in animal rooms as described above in section 4.2 of Example I.
  • mice were randomly and prospectively divided into three (3) treatment groups prior to irradiation. Each animal was identified by an ear punch corresponding to an individual number. A cage card was used to identify each cage or label marked with the study number, treatment group number, and animal numbers. 4.5 Radiation
  • Machine calibration was checked within two weeks of the onset of this study.
  • a single dose of radiation (30 Gy/dose) was administered to all animals in groups 1 and 2 on day 0. Radiation was generated with a 160 kilovolt potential (15-ma) source at a focal distance of 50 cm, hardened with a 0.35 mm Cu filtration system.
  • Irradiation was done at a rate of 121.5 cGy/minute. Animals were anesthetized prior to radiation, and placed under lead shielding such that only the snout is exposed
  • H&E hematoxylin and eosin
  • the mean percentage weight gain for each group for each day of the study is shown in Figure 11.
  • the un-irradiated control group gained an average of 8.1% during the study, as compared with a mean loss of 0.8% in the placebo group.
  • a mean weight gain of 4.0% was seen in the group receiving
  • Each tongue was processed for routine hematoxylin and eosin histology. Because of several technical reasons, a total of 44 samples were evaluated. Of these 44 samples, 6 were in the un-irradiated control group, 12 were in the placebo group (6 each on days 6 and 10), 14 were in the eritoran 1 mg/kg treated group (7 each on days 6 and 10), and 12 were in the eritoran 10 mg/kg treated group (7 on day 6, and 5 on day 10).
  • Hyperkeratosis was also seen in 14 of the samples, none of which were in the unirradiated control group. Hyperkeratosis was most commonly seen in samples from the eritoran 10 mg/kg treated group, where it was applied to 7 of the 12 samples (3/7 at day 6 and 4/5 at day 10). Hyperkeratosis was seen in 5 of the 14 samples in the eritoran 1 mg/kg treated group (1/7 at day 6 and 4/7 on day 10). Only 2 samples in the placebo group were seen with hyperkeratosis, one at each time-point.
  • Epithelial hyperplasia was seen in only 5 samples, however 4 of these samples were in the eritoran 10 mg/kg treatment group (2 at each time-point) and the fifth was in the placebo group (day 6). These observations seem to indicate a substantial improvement in both eritoran treatment groups relative to the placebo controls, with the high dose treatment group (10 mg/kg) showing a tendency to hyperplasia and hyperkeratosis.
  • Increased cellularity was seen in 10 samples, 2 in the placebo group (one each on day 6 and day 10), 5 in the eritoran 1 mg/kg treated group (one on day 6 and 4 on day 10), and 3 in the eritoran 10 mg/kg treated group (one on day 6 and 2 on day 10).
  • Two types of infiltrate were observed, round cell or lymphocytic infiltrates were noted in 8 samples, were evenly distributed among the groups and time-points, and were seen in one of the 6 un-irradiated controls.
  • the mean minimum number of cell layers was 2.8 (day 6) and 3.8 (day 10) in the 1 mg/kg group and 3.8 (day 6) and 3.3 (day 10) in the 10 mg/kg group. These data are shown in Figure 13.
  • the mean maximum number of epithelial cell layers on the dorsal surface in the un-irradiated controls was 8.
  • the mean number of cell layers was 4.8 on day 6 and 3.5 on day 10.
  • Li the eritoran treated groups the mean minimum number of cell layers was 5.3 (day 6) and 6.4 (day 10) in the 1 mg/kg group, and 6.2 (day 6) and 6.1 (day 10) in the 10 mg/kg group. These data are shown in Figure 14.
  • the mean minimum number of cell layers in the un-irradiated controls was 4 cell layers.
  • the mean number of cell layers was 1.7 on day 6 and 0.9 on day 10.
  • the mean minimum number of cell layers was 2.0 (day 6) and 2.8 (day 10) in the 1 mg/kg group, and 3.0 (day 6 and day 10) in the 10 mg/kg group.
  • C3H mice C3H/HeJ and C3H/HeOuJ
  • the C3H/HeOuJ strain are susceptible to oral mucositis induced by focal radiation to the snout, while the C3H/HeJ strain are relatively resistant to radiation induced mucositis.
  • evaluation of the pro-inflammatory cytokines in these animals shows that the induction of these cytokines via the LPS receptor (TLR4) in the C3H/HeOuJ mice may play a role in the development of oral mucositis.
  • TLR4 LPS receptor
  • the acute mouse snout radiation model has been used to determine the radioprotective properties of experimental compounds.
  • the course of oral mucositis in this model is well defined and results in peak mucositis 10-12 days following radiation.
  • the acute model has little systemic toxicity, resulting in relatively few radiation induced animal deaths.
  • a dose of 30 Gy was used to induce oral mucositis.
  • the objective of this study was to examine the effect of scheduling of eritoran, administered intraveneously, on the severity and duration of oral mucositis induced by radiation. Mucositis was induced using an acute radiation dose of 30 Gy directed to the mouse snout. At 10 days after radiation, groups of four mice from each treatment group were sacrificed. At the time of sacrifice, the tongues were removed and fixed in formalin for subsequent histological analysis. At the time of sacrifice, blood was taken from each animal and serum was prepared for subsequent cytokine analysis. These samples were used for the measurement of serum Tumor Necrosis Factor (TNF- ⁇ ), Interleukin-6 (IL-6), and Serum Amyloid A (SAA) levels.
  • TNF- ⁇ Tumor Necrosis Factor
  • IL-6 Interleukin-6
  • SAA Serum Amyloid A
  • mice were obtained from Jackson Laboratories. These animals were shipped with jugular cannulae already implanted. The animals were randomly divided into 6 groups of 10 animals per group as described in Table 4.
  • Animals in group 4 received eritoran at 10 mg/kg twice daily from day 0 until day 3, and then placebo twice daily until the end of the dosing period.
  • Animals in group 5 received placebo twice daily from day 0 until day 2, then eritoran 10 mg/kg twice daily from day 3 until day 6, and then placebo twice daily until the end of the dosing period.
  • Animals in group 6 received placebo twice daily from day 0 until day 5, then eritoran 10 mg/kg twice daily until the end of the dosing period. All drug and placebo administration was via intravenous via jugular cannula.
  • each animal was weighed to an accuracy of 0.1 g.
  • all animals were sacrificed and the tongues taken for histological analysis. Blood was taken at the time of sacrifice and serum was stored at -8O 0 C.
  • mice C3H/HeOuJ mice (Jackson Laboratories), aged 5 to 6 weeks with body weights of 23.2 g, were used. Animals had jugular cannulas installed by Jackson Laboratories prior to shipment, and were individually numbered using an ear punch and individually housed. Animals were acclimatized prior to study commencement. During this period of at least 2 days, the animals were observed daily in order to reject animals that presented in poor condition.
  • mice were randomly and prospectively divided into three (3) treatment groups prior to irradiation. Each animal was identified by an ear punch corresponding to an individual number. A cage card was used to identify each cage or label marked with the study number, treatment group number, and animal numbers.
  • Machine calibration was checked within two weeks of the onset of this study.
  • a single dose of radiation (30 Gy/dose) was administered to all animals in groups 1 and 2 on day 0. Radiation was generated with a 160 kilo volt potential (15-ma) source at a focal distance of 50 cm, hardened with a 0.35 mm Cu filtration system.
  • Irradiation was done at a rate of 121.5 cGy/minute. Animals were anesthetized prior to radiation, and placed under lead shielding such that only the snout is exposed
  • H&E hematoxylin and eosin
  • Enzyme linked immunosorbent assays were performed for cytokines TNF- ⁇ and IL-6 using kits purchased from R and D systems. Determination of serum amyloid A was performed using an ELISA kit from Biosource International. These kits were used in accordance with the manufacturer's instructions. All determinations were made in duplicate on serum samples stored at -80°C. Samples were run in duplicate in all three assays, and if insufficient serum had been collected to run IL-6, SAA 5 and TNF- ⁇ assays, samples were diluted 1 :4. All assays were performed using 50 ⁇ L of sample per well.
  • Body weights are evaluated for differences between the treatment groups.
  • mice A total of 108 cannulated animals were used in this study. Due to the limited availability of the C3H/HeOuJ mice, these animals were processed in 3 groups over a period of 6 weeks. 57 of these mice survived until day 10. Of the 51 mice that did not survive until day 10, 21 died or were euthanized on day O 5 11 due to anesthesia and radiation related issues, and 10 due to problems with the cannula (died after initial injection due to presumed clot, cannula not patent, or cannula pulled out). Of the remaining 30 animals that died or were enthanized during the study, 2 died on day 1, 5 on day 2, 4 on day 3, 7 on day 4, 3 each on days 5 and 6, 1 each on days 7 and 8, and 2 each on days 9 and 10.
  • the distribution of deaths by group was relatively equal.
  • the mean percentage weight gain for each group for each day of the study is shown in Figure 17.
  • the un-irradiated control group gained and average of 3.2% during the study, as compared with a mean loss of 12.1% in the placebo group, hi the groups receiving eritoran at 10 mg/kg, a mean weight loss of 7.8% was seen in the group treated on days 0-10 as compared with a net loss of 2.2% in the group treated on days 0 to 3, a net loss of 7.3% in the group treated on days 3 to 6, and a net loss of 8.9% for the group treated on days 6 to 9.
  • AUC Area Under the Curve
  • Each tongue was processed for routine hematoxylin and eosin histology and slides were reviewed in a blinded manner.
  • a total of 57 samples were evaluated, and of these, 9 were in the un-irradiated control group, 9 were in the placebo group, 11 were in the group treated with eritoran at 10 mg/kg from day 0 to day 9, 11 were in the group treated with eritoran at 10 mg/kg from day 0 to day 3, 9 were in the group treated with eritoran at 10 mg/kg from day 3 to day 6, and 9 were in the group treated with eritoran at 10 mg/kg from day 6 to day 9.
  • epitheial score Three sections from each sample were evaluated for the following parameters: epitheial score, connective tissue score, inflammation score, mitoses per 10 high power fields (hpf), percent ulceration, number of inflammatory cells per 10 hpf (percent neutrophils, lymphocytes, and monocytes/macrophage), the number of small, medium, and large blood vessels per 10 hpf, and the number of mast cells per 10 hpf. 5.3.1 Epithelial Score
  • Epithelial histology was scored on a 4 point 0-3 scale as outlined in section 4.7.1. These scores are shown in Figure 21.
  • the un-irradiated animals all had scores of 0.
  • the placebo control group had a mean score of 1.1 , as did the group treated with eritoran at 10 mg/kg from day 0 to day 9.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had a mean score of 0.45.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had a mean score of 0.89.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had a mean score of 0.75.
  • Connective histology was scored on a 4 point 0-3 scale as outlined in section 4.7.1. These scores are shown in Figure 22.
  • the un-irradiated animals all had scores of 0.
  • the placebo control group had a mean score of 0.4
  • the group treated with eritoran at 10 mg/kg from day 0 to day 9 had a mean score of 0.6.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had a mean score of 0.4.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had a mean score of 0.6.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had a mean score of 0.8.
  • the un-irradiated animals all had scores of 0.
  • the placebo control group had a mean score of 0.4
  • the group treated with eritoran at 10 mg/kg from day 0 to day 9 had a mean score of 0.5.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had a mean score of 0.4.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had a mean score of 0.6.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had a mean score of 0.8. 5.3.4 Number of Mitoses
  • the number of mitoses was counted in 10 high power fields (hpf). These data are shown in Figure 24.
  • the un-irradiated animals had an average of 1.2 mitoses per 10 hpf.
  • the placebo control group had an average of 3.9 mitoses per 10 hpf.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 9 had an average of 2.3 mitoses per 10 hpf.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had an average of 1.5 mitoses per 10 hpf.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had an average of 1.6 mitoses per 10 hpf.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had an average of 1.5 mitoses per 10 hpf.
  • the percentage ulceration was estimated for each sample. These data are shown in Figure 25.
  • the un-irradiated animals had no ulceration.
  • the placebo control group had mean ulceration of 13.3%.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 9 had mean ulceration of 13.2%.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had mean ulceration of ' 2.7%.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had mean ulceration of 16.7%.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had mean ulceration of 10.0%.
  • the inflammatory cell infiltrate present in each sample was enumerated by counting the total number of inflammatory cells per 10 hpf, and evaluated for cell type by estimating the percentage of cells within the infiltrate that were neutrophils, lymphocytes, or monocytes/macrophage.
  • the numbers of inflammatory cell data are shown in Figure 26, the percent neutrophils in Figure 27, the percent lymphocytes in Figure 28, and the percent monocytes/macrophage in Figure 29.
  • the un-irradiated animals had an average of 9.3 cells per 10 hpf, with an average composition of 98.9% lymphocytes and 1.1% monocytes/macrophage, with no neutrophils seen.
  • the placebo control group had an average of 44.9 cells per 10 hpf, with an average composition of 10.6% neutrophils, 86.7% lymphocytes, and 2.8% monocytes/macrophage.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 9 had an average of 43.6 cells per 10 hpf, with an average composition of 13.6% neutrophils, 82.7% lymphocytes, and 4.5% monocytes/macrophage.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had an average of 33.3 cells per 10 hpf, with an average composition of 6.4% neutrophils, 93.2% lymphocytes, and 0.5% monocytes/macrophage.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had an average of 31.5 cells per 10 hpf, with an average composition of 7.2% neutrophils, 91.1% lymphocytes, and 1.1% monocytes/macrophage.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had an average of 52.1 cells per 10 hpf, with an average composition of 8% neutrophils, 91.0% lymphocytes, and 1.0% monocytes/macrophage.
  • the number of blood vessels present in each sample was quantified by counting the total number of blood vessels in 10 hpf, and evaluated for vessel size by counting the number of small, medium, and large vessels in this sample. These data are shown in Figures 30-32.
  • the un-irradiated animals had an average of 5.6 blood vessels per 10 hpf, with an average composition of 63.3% small, 20.7% medium, and 16.0% large vessels seen.
  • the placebo control group had an average of 8.8 blood vessels per 10 hpf, with an average composition of 63.9% small, 22.3% medium, and 13.9% large vessels.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 9 had an average of 9.4 blood vessels per 10 hpf, with an average composition of 74.6% small, 16.1% medium, and 9.3% large vessels seen.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had an average of 8.2 blood vessels per 10 hpf, with an average composition of 72.5% small, 14.9% medium, and 12.6% large vessels.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had an average of 7.0 blood vessels per 10 hpf, with an average composition of 67.9% small, 20.5% medium, and 11.6% large vessels.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had an average of 7.5 blood vessels per 10 hpf, with an average composition of 72.1% small, 17.3% medium, and 10.6% large vessels. 5.3.8. Mast Cells
  • the number of mast cells present in each sample was determined by counting the number cells per 10 hpf. These data are shown in Figure 33.
  • the un-irradiated animals had 23.7 mast cells per 10 hpf.
  • the placebo control group had 26 mast cells per 10 hpf.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 9 had 18.4 mast cells per 10 hpf.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had 24.4 mast cells per 10 hpf.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had 24.2 mast cells per 10 hpf.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had 24.5 mast cells per 10 hpf.
  • Serum levels of TNF- ⁇ , IL-6, and SAA were measured using commercially available ELISA kits.
  • the un-irradiated animals had serum TNF- ⁇ levels of 43.0 pg/mL.
  • the placebo control group had mean serum TNF- ⁇ levels of 63.8 pg/mL.
  • the group ' treated with eritoran at 10 mg/kg from day 0 to day 9 had mean serum TNF- ⁇ levels of
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had mean serum TNF- ⁇ levels of 20.3 pg/mL.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had mean serum TNF- ⁇ levels of 40.2 pg/mL.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had mean serum TNF- ⁇ levels of
  • the un-irradiated animals had serum IL-6 levels of 48.7 pg/mL.
  • the placebo control group had mean serum IL-6 levels of 154.2 pg/mL.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 9 had mean serum IL-6 levels of 85.6 pg/mL.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had mean serum IL-6 levels of 19.7 pg/mL.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had mean serum IL-6 levels of 50.4 pg/mL.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had mean serum IL-6 levels of 119.3 pg/mL.
  • the un-irradiated animals had serum SAA levels of 597 ⁇ g/mL.
  • the placebo control group had mean serum SAA levels of 427 ⁇ g/mL.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 9 had mean serum SAA levels of 344 ⁇ g/mL.
  • the group treated with eritoran at 10 mg/kg from day 0 to day 3 had mean serum SAA levels of 279 ⁇ g/mL.
  • the group treated with eritoran at 10 mg/kg from day 3 to day 6 had mean serum SAA levels of 475 ⁇ g/mL.
  • the group treated with eritoran at 10 mg/kg from day 6 to day 9 had mean serum SAA levels of 652 ⁇ g/mL.
  • mice treated with eritoran on days 0-3 showed a significant improvement in weight loss relative to the placebo treated control group.
  • the group treated with eritoran on days 0-3 had the lowest epithelial score, connective tissue score, and percent ulceration, indicating that it had suffered less damage than other groups. This group also had the lowest inflammation score and was second lowest in the number of inflammatory cells and mitoses. 6. Among the groups receiving radiation, the group treated with eritoran on days 0-3 had the lowest serum levels of TNF- ⁇ , JL-6, and SAA, showing the efficacy of this regimen in reducing inflammatory responses.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention se rapporte à des méthodes permettant de réduire la gravité de la mucosite orale et gastro-intestinale, qui consistent à administrer un antagoniste du récepteur Toll 4.
EP06831669A 2005-05-13 2006-05-15 Méthodes permettant de réduire la gravité de la mucosite orale et gastro-intestinale Withdrawn EP1879597A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US68073305P 2005-05-13 2005-05-13
PCT/IB2006/003538 WO2007031879A2 (fr) 2005-05-13 2006-05-15 Méthodes permettant de réduire la gravité de la mucosite orale et gastro-intestinale

Publications (1)

Publication Number Publication Date
EP1879597A2 true EP1879597A2 (fr) 2008-01-23

Family

ID=37772612

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06831669A Withdrawn EP1879597A2 (fr) 2005-05-13 2006-05-15 Méthodes permettant de réduire la gravité de la mucosite orale et gastro-intestinale

Country Status (7)

Country Link
EP (1) EP1879597A2 (fr)
JP (1) JP2008540510A (fr)
KR (1) KR20080038085A (fr)
CN (1) CN101262870A (fr)
AU (1) AU2006290437A1 (fr)
CA (1) CA2606727A1 (fr)
WO (1) WO2007031879A2 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT2185581E (pt) * 2007-07-06 2015-12-09 Promedior Inc Processos e composições úteis no tratamento de mucosite
WO2009152517A1 (fr) * 2008-06-13 2009-12-17 Case Western Reserve University Compositions et procédés pour le traitement d’une inflammation de la cornée
WO2010127367A2 (fr) * 2009-05-01 2010-11-04 Alfagene Bioscience, Inc. Système de cellules épithéliales intestinales primaires dérivées de cellules souches gastro-intestinales humaines et ses procédés d'utilisation
BR112014024139A8 (pt) * 2012-03-28 2018-01-16 Univ Maryland administração de eritoran ou sais farmaceuticamente aceitáveis do mesmo para tratar infecções de ortomixovírus
CN107669692B (zh) * 2017-09-07 2020-09-29 中国人民解放军第二军医大学 Mpla在制备电离辐射致肠道损伤防治药物中的应用

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030130212A1 (en) * 1999-01-14 2003-07-10 Rossignol Daniel P. Administration of an anti-endotoxin drug by intravenous infusion
WO2003013440A2 (fr) * 2001-08-10 2003-02-20 Eisai Co., Ltd. Traitement et prevention de maladies et de conditions associees a la proteine de choc thermique

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2007031879A2 *

Also Published As

Publication number Publication date
CA2606727A1 (fr) 2007-03-22
KR20080038085A (ko) 2008-05-02
JP2008540510A (ja) 2008-11-20
CN101262870A (zh) 2008-09-10
WO2007031879A2 (fr) 2007-03-22
AU2006290437A1 (en) 2007-03-22
WO2007031879A3 (fr) 2007-10-04

Similar Documents

Publication Publication Date Title
US7727974B2 (en) Methods of reducing the severity of mucositis
RU2767664C2 (ru) Комбинированная терапия противоопухолевым алкалоидом
Coleman et al. A randomised phase II study of two different schedules of pegylated liposomal doxorubicin in metastatic breast cancer (EORTC-10993)
JP4391083B2 (ja) 効果的な抗腫瘍治療
AU758191B2 (en) Antitumor composition containing a synergistic combination of an anthracycline derivative with a camptothecin derivate
PT1385498E (pt) Ácidos gordos como factores de sobrevivência e activação de neutrófilos
JP2020055884A (ja) がんのためのグルフォスファミド組み合わせ療法
EP1879597A2 (fr) Méthodes permettant de réduire la gravité de la mucosite orale et gastro-intestinale
EP2107906B1 (fr) Nouvelle utilisation therapeutique pour le traitement des leucemies
WO2017132445A1 (fr) Régime clincal pour le traitement du syndrome myélodysplasique avec un inhibiteur de phosphatase
CN109528731B (zh) 具有协同作用治疗多发性骨髓瘤的药物组合物及其应用
TW201904579A (zh) Mcl-1抑制劑與血液癌症標準療法之組合,其用途及醫藥組合物
TWI454280B (zh) Pharmaceutical composition or combination
CN108126201B (zh) 肠道菌抗生素在制备防治化疗并发症药物中的应用
CN112915086B (zh) 一种含有Akt靶向激酶抑制剂的药物组合物
KR101969451B1 (ko) 만성 호흡기 질환 치료제 및 만성 호흡기 질환의 예방 또는 개선용 식품 조성물.
NZ768469B2 (en) Fractionated dosing of a phospholipid ether analog for the treatment of cancer
CN115461058A (zh) 用于治疗血液癌的方法和方案
TW201513871A (zh) 用於癌症治療之抗群集素(clusterin)單一療法
KR20210054544A (ko) 포유류 체내의 myc-발현 종양에 대한 타우로리딘 치료
CN116327771A (zh) 一种用于治疗炎症及相关疾病的联合用药物
TWI434829B (zh) 以密環菌乙素來治療癌症之新穎用途

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20071128

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: EISAI R&D MANAGEMENT CO., LTD.

17Q First examination report despatched

Effective date: 20080811

RIN1 Information on inventor provided before grant (corrected)

Inventor name: SHIROTA, HIROSHI

Inventor name: KOBAYASHI, SEIICHI

Inventor name: KAWANO, TETSU

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

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20111201