EP1901740A2 - Unit dosage forms of temozolomide - Google Patents

Unit dosage forms of temozolomide

Info

Publication number
EP1901740A2
EP1901740A2 EP07796436A EP07796436A EP1901740A2 EP 1901740 A2 EP1901740 A2 EP 1901740A2 EP 07796436 A EP07796436 A EP 07796436A EP 07796436 A EP07796436 A EP 07796436A EP 1901740 A2 EP1901740 A2 EP 1901740A2
Authority
EP
European Patent Office
Prior art keywords
days
patient
per day
day
day cycle
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Withdrawn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
EP07796436A
Other languages
German (de)
English (en)
French (fr)
Inventor
Jacqueline Rose Bersch
Mark Manzo
Sumant Ramachandra
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.)
Merck Sharp and Dohme Corp
Original Assignee
Schering Corp
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 Schering Corp filed Critical Schering Corp
Publication of EP1901740A2 publication Critical patent/EP1901740A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • 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
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4883Capsule finishing, e.g. dyeing, aromatising, polishing

Definitions

  • This invention relates to unit dosage forms of temozolomide. These unit dosage forms are particularly well-suited for decreasing the pill burden and increasing patient compliance.
  • the invention also relates to methods of treating proliferative disorders in a patient with these unit dosage forms.
  • the invention additionally relates to kits comprising these unit dosage forms.
  • Brain tumors comprise approximately 2% of all malignant diseases. Stupp et al., J. Clin. One, 20(5):1375-1382 (2002). More than 17,000 cases are diagnosed every year in the United States, with approximately 13,000 associated deaths.
  • the standard protocol for treating a malignant glioma involves cytoreduction through surgical resection, when feasible, followed by radiotherapy (RT) with or without adjuvant chemotherapy. Stupp et al., supra.
  • temozolomide Schering Corp. under the trade name of Temodar® in the United States and Temodal® in Europe.
  • the chemical name for temozolomide is 3,4- dihydro-3-methyl-4-oxoimidazo[5,l-d]-as-tetrazine-8-carboxamide (see U.S. Pat. No. 5,260,291).
  • MTIC methyl-(triazen-l-yl)irnidazole-4-carboxamide
  • Temodar® capsules are currently indicated in the United States for the treatment of adult patients with newly diagnosed gliobastoma multiforme as well as refractory anaplastic astrocytoma, i.e., patients at first relapse who have experienced disease progression on a drug regimen containing a nitrosourea and procarbazine.
  • Temodal® is currently approved in Europe for the treatment of patients with malignant glioma, such as glioblastoma multiforme or anaplastic astrocytoma for newly diagnosed patients as well as those showing recurrence or progression after standard therapy.
  • a typical regimen for patients with glioma taking temolozomide consists of two phases, a concomitant phase, followed by a maintenance phase.
  • the patient receives an oral administration of temozolomide at 75 mg/m 2 (approximately 140 mg for a patient having a Body Surface Area (BSA) between 1.8 and 1.9 m 2 ) for 42 days concomitant with RT. See Table 1.
  • BSA Body Surface Area
  • temozolomide is administered at 150 mg/m 2 (approximately 280 mg for a patient having a BSA between 1.8 and 1.9 m 2 ) once daily for five days followed by 23 days without treatment.
  • the dosage may be escalated to 200 mg/m 2 (approximately 360 mg for a patient having a BSA between 1.8 and 1.9 m 2 ) for the first 5 days of each subsequent cycle.
  • the capsule formulations of temozolomide contain 5, 20, 100 or
  • temozolomide 250 mg.
  • a patient having a BSA of between 1.8 and 1.9 would need to consume 4 capsules per day to receive the approximately 280 mg of temozolomide dosage (1 x 250 mg, 1 x 20 mg and 2 x 5 mg). See Table 2.
  • Such a high pill burden would likely result in poor patient compliance with therapies that require self-administration of temozolomide, thus producing sub-optimal therapeutic benefit of the drug.
  • Other factors that may affect the patient's compliance with self-administering the appropriate number of pills is the intensity of the treatment regimens for gliomas, including brain surgery and RT.
  • the number of different medications that the patient may have to take to alleviate the side effects of the surgery or RT or to remedy or alleviate other unrelated conditions further exacerbate the already high pill burden.
  • the patient is at risk for cognitive deficits and compromised neurological status as a result of the intense treatment regimens.
  • the present invention provides an improved unit dosage form of temozolomide comprising about 140 mg temozolomide.
  • the present invention also provides an improved unit dosage form of temozolomide comprising about 180 mg of temozolomide. These unit dosage forms reduce the pill burden and increase patient compliance.
  • the unit dosage forms are in the form of a capsule.
  • the capsules are color-coded.
  • the 140 mg temozolomide capsule differs in color from the 180 mg temozolomide capsule.
  • the present invention also provides methods of increasing patient compliance with a regimen by administering these unit dosage forms.
  • the present invention also provides methods of treating proliferative disorders with these unit dosage forms.
  • the proliferative disorder is selected from a glioma, melanoma, a lung cancer, a lymphoma, a head and neck cancer, ovarian cancer, colorectal and/or colon cancer or esophageal cancer, or other solid tumor or hematologic malignancy.
  • the methods of treating proliferative disorders comprise administering the unit dosage forms according to a dosing regimen.
  • the regimen is based upon the methylation state of the promoter region of the O 6 -methylguanine-DNA transferase (MGMT) gene in a sample obtained from the patient.
  • the regimen is based upon the presence or absence of the MGMT protein in a sample obtained from the patient.
  • the regimen is based upon the level or enzymatic activity of the MGMT protein detected in a sample obtained from the patient.
  • the regimen is based upon the presence (or level) of messenger RNA of MGMT in a sample obtained from the patient.
  • kits comprising these unit dosage forms.
  • the kits comprise both unit dosage forms.
  • the kits further comprise one or more unit dosage forms comprising about 5, 20 or 100 mg of temozolomide.
  • the kit is a blister pack.
  • pharmaceutically acceptable carrier or adjuvant refers to a nontoxic carrier or adjuvant that may be administered to a patient, together with temolozimide, and that does not destroy the pharmacological activity thereof.
  • color-coded capsule refers to a capsule in which any portion or all of the capsule is colored and the color represents a particular meaning, e.g. a given capsule color represents a specific dosage amount of temozolomide.
  • treating or “treatment” is intended to mean mitigating or alleviating the symptoms a cell proliferative disorder in a mammal such as a human or the improvement of an ascertainable measurement associated with a cell proliferative disorder.
  • neoplasm refers to a new, abnormal growth of cells or a growth of abnormal cells that reproduce faster than normal.
  • a neoplasm creates an unstructured mass (a tumor) which can be either benign or malignant.
  • benign refers to a tumor that is noncancerous, e.g., its cells do not invade surrounding tissues or metastasize to distant sites.
  • malignant refers to a tumor that is cancerous, metastastic, invades contiguous tissue or is no longer under normal cellular growth control.
  • brain tumor includes glioma, glioblastoma multiforme, ependymoma; astrocytoma, medulloblastoma, neuroglioma, oligodendroglioma and meningioma.
  • sample refers to a specimen that can obtained as or isolated from normal or tumor tissue, brain tissue, cerebrospinal fluid, blood, plasma, serum, urine, stool, saliva lymph, lymph nodes, spleen, liver, bone marrow, or any other biological specimen containing either MGMT protein or nucleic acid of the MGMT gene.
  • MGMT refers to O 6 -methylguanine-DNA methyltransferase. MGMT is also known as an O 6 -alkylguanine-DNA-alkyltransferase (AGAT).
  • GM-CSF means a protein which (a) has an amino acid sequence that is substantially identical to the sequence of mature (i.e., lacking a signal peptide) human GM-CSF described by Lee et al., Proc. Natl. Acad. Sci. U.S.A., 82:4360 (1985) and (b) has biological activity that is common to native GM-CSF.
  • substantially impair biological activity means that the sequences are identical or differ by one or more amino acid alterations (deletions, additions, substitutions) that do not substantially impair biological activity.
  • kit refers to one or more containers for containing the unit dosage forms of the present invention.
  • the present invention provides a unit dosage form of temozolomide comprising about 140 mg temozolomide and a pharmaceutically acceptable carrier.
  • diluents include calcium carbonate, potato starch, alginic acid, and lactose.
  • inert diluents include lubricating agents, such as magnesium stearate.
  • the pharmaceutically acceptable carriers are lactate anhydrous, colloidal silicon dioxide, sodium starch glycolate, tartartic acid and stearic acid.
  • the unit dosage forms according to the present invention may be prepared in the form of tablets, pills, granules, dispersible powders or capsules.
  • the unit dosage forms are in the form of capsules.
  • the capsules are color-coded for easy identification.
  • the 140 mg temozolomide capsule differs in color from the 180 mg temozolomide capsule.
  • each capsule strength has a different color, thus indicating dosage of temozolomide.
  • the unit dosage forms of the present invention may be used to increase patient compliance by reducing the pill burden.
  • the present invention provides methods of increasing patient compliance by administering to the patient one or more unit dosage forms containing about 140 mg or 180 mg of temozolomide.
  • the methods further comprise administering one or more unit dosage forms containing about 5 mg, 20 mg or 100 mg of temozolomide.
  • the unit dosage forms may be administered to a patient according to a dosing regimen and/or dosing schedule.
  • dosing regimens and schedules are illustrated in Table 3.
  • the dosing regimens of Table 3 may be simplified by using the unit dosage forms of the present invention. Suggested capsule combinations using the unit dosage forms of the present invention are illustrated in Table 4. In 18 of 29 daily doses listed in Table 4, the suggested capsule combinations of the present invention are lower than the currently available capsule combinations. See Table 5. Importantly, the suggested capsule combinations of the present invention increase in only 2 daily doses (see, 255 mg and 370 mg in Table 2 compared to Table 4). By simplifying the dosing regimens, the pill burden on the patient is significantly reduced.
  • a patient on a 195 mg of temozolomide per day regimen would need to consume 8 of the current capsule formulations to achieve the therapeutic dosage (i.e., 1 x 100 mg, 4 x 20 mg and 3 x 5 mg). See Tables 2 and 5.
  • that patient would need to consume only 4 units dosage forms (i.e., 1 x 180 mg and 3 x 5 mg). See Tables 4 and 5.
  • a patient on a 280 mg of temozolomide per day regimen would need to consume 4 of the current capsule formulations to achieve the therapeutic dosage (i.e., 1 x 250 mg, 1 x 20 mg and 2 x 5 mg). See Tables 2 and 5.
  • Table 6 further illustrates how the unit dosage forms of the present invention decrease the pill burden on the patient. As shown in Table 6, the number of capsules is lower in 28 out of 45 regimens when the patient receives the unit dosage forms of the present invention. See Table 6. Importantly, the number increases in only 8 of the remaining 17 regimens. Table 4.
  • the unit dosage forms of the present invention may be used to treat a proliferative disorder in a patient.
  • Proliferative disorders include, but are not 5 limited to, benign/malignant tumors such as brain tumors, prostate, lung cancer, breast cancer, ovarian, testicular cancer, liver, kidney, spleen, bladder, colorectal and/or colon cancer, head and neck, melanoma, carcinoma, sarcoma, lymphoma, mycosis fungoides as well as leukemia or other hematologic malignancies.
  • the methods are used to treat glioma, melanoma, lung cancer, 10 lymphoma, colorectal and/or colon cancer, head and neck or ovarian cancer.
  • the methods are used to treat glioma.
  • the unit dosage forms of the present invention are administered according to one of the dosing regimens presented in Tables 3 and 4.
  • the dosing regimen is 150-200 mg/m 2 per day for 5 days in 15 a 28 day cycle. In other embodiments, the dosing regimen is 100 mg/m 2 per day for 14 days in a 21 day cycle. In other embodiments, the dosing regimen is
  • the unit dosage forms of the present invention are used to treat glioma.
  • the unit dosage forms are 20 administered to a patient having a glioma according to one of the dosing regimens presented in Tables 3 and 4.
  • the dosing regimen is 150-200 mg/m 2 per day for 5 days in a 28 day cycle.
  • the dosing regimen is 100 mg/m 2 per day for 14 days in a 21 day cycle.
  • the dosing regimen is 150 mg/m 2 per day for 7 days in a 14 day cycle.
  • the unit dosage forms are administered in combination with a growth factor.
  • suitable growth factors include, but are not limited to, GM-CSF, G-CSF, IL-I, IL-3, IL-6, or erythropoietin.
  • Non-limiting growth factors include Epogen® (epoetin alfa), Procrit®.
  • the growth factor is G-CSF.
  • the unit dosage forms are administered in a combination therapy. In some embodiments, the unit dosage forms are administered in combination with a poly(ADP-ribose) polymerase(s) (PARP) inhibitor.
  • PARP poly(ADP-ribose) polymerase(s)
  • the PARP inhibitor may be administered either prior to, concomitantly with or after administration of the unit dosage forms of the present invention.
  • Suitable PARP inhibitors include but are not limited to CEP-6800 (Cephalon; described in Miknyoczki et al., MoI Cancer Ther, 2(4):371-382 (2003)); 3- aminobenzamide (also known as 3-AB; Inotek; described in Liaudet et al., Br J Pharmacol, 133(8):1424-1430 (2001)); PJ34 (Inotek; described in Abdelkarim et al., Int J MoI Med, 7(3):255-260 (2001)); 5-iodo-6-amino-l,2-benzopyrone (also known as INH(2)BP; Inotek; described in Mabley et al., Br J Pharmacol, 133(6):909-919 (2001), GPI 15427 (described in Tentori et al., Int J Oncol, 26(2):415-422 (2005)); 1,5-dihydroxyisoquinoline (also known as DIQ; described in Walisser and Thies, Exp Cell Res,
  • the unit dosage forms are administered in combination with an O 6 -alkylguanine-DNA-alkyltransferase (Atase) inhibitor such as O 6 -benzylguanine (O 6 BG) or Lomeguatrib [6-(4-bromo-2-thienyl) methoxy] purin-2-amine] (a.k.a. PatrinTM or Patrin-2) described in U.S. Pat. No. 6,043,228 (Cancer Research UK) to accentuate hematopoietic toxicity.
  • the Atase inhibitor such as O 6 BG can be administered either prior to, concomitantly with or after administration of the unit dosage forms of the present invention.
  • the unit dosage forms are administered in combination with an anti-emetic agent.
  • Suitable anti-emetic agents include, but are not limited to, Palonosetron, Tropisetron, Ondansetron, Granisetron, Bemesetron or a combination of at least two of the foregoing.
  • the amount of active anti-emetic substance in one dosage unit amounts to 2 to 10 mg, an amount of 5 to 8 mg active substance in one dosage unit being especially preferred.
  • a daily dosage comprises generally an amount of active substance of 2 to 20 mg, particularly preferred is an amount of active substance of 5 to 16 mg.
  • a neurokinin- 1 antagonist such as aprepitant may be administered either alone or in combination with a steroid such as dexamethasone in conjunction with an anti-emetic agent.
  • the unit dosage forms are administered in combination with an NK-I agonist alone or with a steroid.
  • the NK-I antagonist is one or more of the NK-I antagonists described in U.S. Pat. No. 7,049,320 alone or in combination with one or more of an 5HT3 inhibitor and a steroid.
  • the unit dosage forms are administered in combination with a farnesyl protein transferase inhibitor.
  • the unit dosage forms are administered with another antineoplastic agent.
  • Suitable antineoplastic agents include, but are not limited to, Uracil Mustard, Chlormethine, Cyclophosphamide, Ifosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylenemelamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, dacarbazine, Methotrexate, 5- Fluorouracil, Floxuridine, Cytarabine, 6-Mercaptopurine, 6-Thioguanine, Fludarabine phosphate, Pentostatine, Gemcitabine, Vinblastine, Vincristine, Vindesine, Bleomycin, Dactinomycin, Daunorubicin, Doxorubicin, Epirubicin, Idarubicin, Paclitaxel, Mithramycin, Deoxycoformycin, Mitomycin-C, L- Asparaginase, Interferons, E
  • Medroxyprogesteroneacetate Leuprolide, Flutamide, Toremifene, Goserelin, Cisplatin, Carboplatin, Hydroxyurea, Amsacrine, Procarbazine, Mitotane, Mitoxantrone, Levamisole, Navelbene, Anastrazole, Letrazole, Capecitabine, Reloxafine, Droloxafine, Hexamethylmelamine, Oxaliplatin (Eloxatin®), Iressa (gefinitib, ZdI 839), XELOD A® (capecitabine), Tarceva® (erlotinib), Azacitidine (5-Azacytidine; 5-AzaC), and mixtures thereof.
  • the unit dosage forms may be administered with other anti-cancer agents such as the ones disclosed in U.S. Pat. Nos. 5,824,346, 5,939,098, 5,942,247, 6,096,757, 6,251,886, 6,316,462, 6,333,333, 6,346,524, and 6,703,400, all of which are incorporated by reference.
  • Temozolomide as an alkylating agent, causes cell death by binding to DNA which structurally distorts the DNA helical structure preventing DNA transcription and translation.
  • the damaging action of alkylating agents can be repaired by cellular DNA repair enzymes, in particular MGMT.
  • the level of MGMT varies in tumor cells, even among tumors of the same type.
  • the gene encoding MGMT is not commonly mutated or deleted. Rather, low levels of MGMT in tumor cells are due to an epigenetic modification; the MGMT promoter region is methylated, thus inhibiting transcription of the MGMT gene and preventing expression of MGMT.
  • U.S. Publication 20060100188 discloses methods for treating cancer in a patient comprising administering temozolomide according to improved dosing regimen and/or schedules based on the patient's MGMT level.
  • the dosing regimen is based upon the methylation state of the MGMT gene in a sample obtained from the patient.
  • the methylation state is assessed by a determination of whether the MGMT gene is methylated.
  • the methylation state is assessed by a quantitative determination of the level of methylation of the MGMT gene.
  • the methylation state is assessed by determination of whether MGMT protein is expressed.
  • the methylation states is assessed by a determination of the level of MGMT protein expressed or measurement of the enzymatic activity of MGMT in the patient sample.
  • Assessing whether the MGMT gene is methylated may be performed using any method known to one skilled in the art.
  • Techniques useful for detecting methylation of a gene or nucleic acid include, but are not limited to, those described by Ahrendt et al., J. Natl. Cancer Inst., 91 :332-339 (1999); Belsinky et al., Proc. Natl. Acad. Sci.
  • Methylation-specific PCR can rapidly assess the methylation status of virtually any group of CpG sites within a CpG island, independent of the use of methylation-sensitive restriction enzymes. See, MSP; Herman et al., Proc. Natl. Acad Sci. USA, 93(18):9821-9826 (1996); Esteller et al., Cancer Res., 59:793-797 (1999)) see also U.S. Pat. No. 5,786,146, issued JuI. 28, 1998; U.S. Pat. No.
  • the MSP assay entails initial modification of DNA by sodium bisulfite, converting all unmethylated, but not methylated, cytosines to uracil, and subsequent amplification with primers specific for methylated versus unmethylated DNA.
  • MSP requires only small quantities of DNA, is sensitive to 0.1% methylated alleles of a given CpG island locus, and may be performed on DNA extracted from paraffin-embedded samples. MSP eliminates the false positive results inherent to previous PCR-based approaches that relied on differential restriction enzyme cleavage to distinguish methylated from unmethylated DNA. This method is very simple and can be used on small amounts of tissue or a few cells. As would be understood by those skilled in the art, if the gene encoding MGMT is not methylated, the MGMT protein is expressed and can be detected (e.g., by Western blot, immuno-histochemical techniques or enzymatic assays for MGMT activity, etc.) as detailed below herein.
  • the level of MGMT protein expressed in a sample obtained from a patient may be assessed by measuring MGMT protein, e.g., by Western blot using an antibody specific to MGMT, see for example, U.S. Pat. No. 5,817,514.
  • the level of MGMT expressed in a sample may also be assessed by measuring the MGMT protein using an immunohistochernistry technique on a defined number of patient cells, e.g., employing a labeled antibody specific for MGMT and comparing the level with that expressed by the same defined number of normal lymphocytes known to express MGMT (see, for example, U.S. Pat. No. 5,407,804 by Yarosh for a description of useful quantitative immunohistochemical assays).
  • the level of MGMT may be assessed by enzymatic assay, i.e., the ability to methylate the O 6 or N 7 guanine position of DNA.
  • the measured level of MGMT protein expressed is compared to that expressed by normal lymphocytes known to express MGMT.
  • MGMT specific activity of MGMT may be assessed and based on a comparison with cell lines known to express MGMT classified as follows:
  • the level of methylation of MGMT may be assessed by quantitative determination of the methylation of the gene encoding MGMT.
  • the quantitative technique called COBRA (Xiong et al., Nuc. Acids Res., 25:2532-2534 (1997)) may be used in this determination.
  • the "methyl light” technique of Eads et al., Nuc. Acids Res., 28(8):e32 (2000); U.S. Pat. No. 6,331,393 may also be used.
  • the level of methylation of gene encoding MGMT in cells of the patient is compared to that of an equivalent number of cells of normal lymphocytes known to express
  • MGMT normal lymphocytes expressing MGMT have a low level of methylation of the MGMT gene; conversely, cells with high levels of methylation of the MGMT gene express low levels of the MGMT protein (see for example, Costello et al., J. Biol. Chem., 269(25): 17228-17237 (1994); Qian et al., Carcinogen, 16(6):1385-1390 (1995)).
  • COBRA may be used to determine quantitatively DNA methylation levels at specific gene loci in small amounts of genomic DNA. Restriction enzyme digestion is used to reveal methylation-dependent sequence differences in PCR products of sodium bisulfite-treated DNA. (Tano et al., Proc. Natl. Acad. Sci. USA, 87:686-690 (1990) describe isolation and sequence of the human MGMT gene). Methylation levels in original DNA sample are represented by the relative amounts of digested and undigested PCR product in a linearly quantitative fashion across a wide spectrum of DNA methylation levels. This technique may be reliably applied to DNA obtained from microdissected paraffin-embedded tissue samples. COBRA thus combines the powerful features of ease of use, quantitative accuracy, and compatibility with paraffin sections. An illustrative example of a RT-PCR assay useful for assessing the level of
  • MGMT mRNA is described in Watts et al., MoI. Cell. Biol., 17(9):5612-5619 (1997). In brief, total cellular RNA is isolated by guanidium isothiocyanate cell lysis followed by centrifugation through a 5.7 M CsCl gradient for 2.5 hr at 2O5,OOOxg. RNA is quantitated in a Beckman TL-100 spectrophotometer by measurements of absorbance at 260 nm.
  • Total cellular RNA is reverse transcribed by incubating a 40 ⁇ l reaction mixture composed of 200 ng of RNA; IxPCR buffer (10 mM Tris [pH 8.3], 50 mM KCl, 1.5 mM MgCl 2 ); 1 mM each dATP, dCTP, dGTP, and dTTP; 200 pmol of random hexamer, 40 U of RNasin, and 24 U of avian myeloblastosis virus reverse transcriptase (Boehringer Mannheim, Indianapolis, Ind.) at 42° C for 60 min. The reaction is then stopped by incubation at 99° C for 10 min.
  • MGMT-specific PCR is performed by adding 80 ⁇ l of amplification reaction buffer (Ix PCR buffer, 25 pmol of MGMT-specific primers and/or a control sequence, and 2 U of Taq DNA polymerase) to 20 ⁇ l of the reverse transcription reaction mixture followed by incubation at 94° C for 5 min; 30 cycles of 94° C. for 1 min, 60° C for 15 s, and 72° C. for 1 min; a final extension at 72° C. for 5 min; and a quick chill to 4° C.
  • the upstream primer sequence from exon 4 (nt 665 to 684) of the MGMT gene can be used.
  • Nucleotide positions can be derived from the cDNA sequence (Tano et al., Proc. Natl. Acad. Sci. USA, 87:686-690 (1990)).
  • a control primer sequence can be employed in the same cDNA reaction (e.g., primers for the histone 3.3 gene).
  • 10% of the respective PCR products are separated through a 3% agarose gel and visualized by ethidium bromide staining.
  • the unit dosage forms of the present invention are used to treat a patient having a glioma.
  • the dosing regimen is based upon the detection of methylated MGMT gene in a sample.
  • the dosing regimen is 150-200 mg/m 2 per day for 5 days in a 28 day cycle.
  • the dosing regimen is (i) 100 mg/m 2 per day for 14 days in a 21 day cycle; (ii) 150 mg/m 2 per day for 7 days in a 14 day cycle; or (iii) 100 mg/m 2 per day for 21 days in a 28 day cycle.
  • the sample is a tumor biopsy sample.
  • the MGMT gene is detected using MSP.
  • the dosing regimen is based upon the presence or absence of MGMT protein. In those embodiments basedupon the presence or absence of the MGMT protein, when the MGMT protein is not detected in a sample obtained from a patient having a glioma, the dosing regimen is 150-200 mg/m 2 per day for 5 days in a 28 day cycle; when the MGMT protein is detected, the dosing regimen is selected from (i) 100 mg/m 2 per day for 14 days in a 21 day cycle; 150 mg/m 2 per day for 7 days in a 14 day cycle; or (iii) 100 mg/m 2 per day for 21 days in a 28 day cycle.
  • the sample is a tumor biopsy sample.
  • the MGMT protein is detected using Western blot immunoassay, an immunohistochemical technique, or an enzymatic assay for MGMT protein.
  • the dosing regimen is based upon the level or activity of the MGMT protein detected in a sample. When the level or enzymatic activity of the MGMT protein detected in a sample obtained from the patient is Low, compared to that of normal lymphocytes, the dosing regimen is (i) 150-200 mg/m 2 per day for 5 days in a 28 day cycle; or (ii) 250 mg/m 2 per day for 5 days in a 28 day cycle in combination with a growth factor.
  • the dosing regimen is (i) 100 mg/m 2 per day for 14 days in a 28 day cycle; (ii) 300 mg/m 2 per day for 5 days in a 28 day cycle in combination with a growth factor; (iii) 75 mg/m 2 per day for 21 days in a 28 day cycle; or (iv) 75 mg/m 2 per 42 days in a 56 day cycle.
  • the dosing regimen is selected from (i) 100 mg/m 2 per day for 14 days in a 21 day cycle; (ii) 150 mg/m 2 per day for 7 days in a 14 day cycle; or (iii) 100 mg/m 2 per day for 21 days in a 28 day cycle.
  • the sample is a tumor biopsy sample.
  • the unit dosage forms are used to treat patients having a proliferative disorder selected from melanoma, lung cancer, lymphoma, head cancer, neck cancer, ovarian cancer, colorectal cancer, colon cancer and esophogeal cancer.
  • the dosing regimen is based upon the detection of methylated MGMT gene in a sample. When methylation of the MGMT gene is not detected in a sample obtained from a patient, the dosing regimen is (i) 100 mg/m 2 per day for 14 days in a 21 day cycle; (ii) 150 mg/m 2 per day for 7 days in a 14 day cycle; or (iii) 100 mg/m 2 per day for 21 days in a 28 day cycle.
  • the sample is a tumor biopsy sample.
  • the MGMT gene is detected using MSP.
  • the dosing regimen is based upon the detection of MGMT protein in a sample.
  • the dosing regimen is selected from (i) 100 mg/m 2 per day for 14 days in a 21 day cycle; 150 mg/m 2 per day for 7 days in a 14 day cycle; or (iii) 100 mg/m 2 per day for 21 days in a 28 day cycle.
  • the sample is a tumor biopsy sample.
  • the MGMT protein is detected using Western blot immunoassay, an immunohistochemical technique, or an enzymatic assay for MGMT protein.
  • the dosing regimen is based upon the level or activity of the MGMT protein detected in a sample.
  • the dosing regimen is (i) 100 mg/m 2 per day for 14 days in a 21 day cycle; (ii) 150 mg/m 2 per day for 7 days in a 14 day cycle; or (iii) 100 mg/m 2 per day for 21 days in a 28 day cycle.
  • the sample is a tumor biopsy sample. Kits Comprising The Unit Dosage Forms Of The Present Invention
  • the unit dosage forms of the present invention may be included in a kit.
  • the kit may be in any conventional shape or form as known in the art which is made of a pharmaceutically acceptable material, for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a "refill" of tablets for placement into a different container), or metal or plastic foil, such as a blister pack with individual doses for pressing out of the pack or a sachet or envelope-like container that may be ripped open to release the unit dosage form according to a therapeutic schedule.
  • the kit is a blister pack.
  • the kit may include more than one container to market a single dosage form.
  • capsules or tablets may be contained in a bottle, which is in turn contained within a box.
  • the kit is air-tight.
  • the kit is made from a light resistant material.
  • the kit comprise one or more unit dosage forms comprising about 5, 20, 100 140 or 180 mg of temozolomide.
  • the kit is a blister pack.
  • the different dosage forms may be arranged in the form of capsules, tablets or pills in rows lying next to one another in the blister pack. At the time indicated by the physician, the patient would in each case successively take a capsule, tablet or pill from each row within a short length of time (in particular within 5 minutes).
  • the kit may be an FDA approved kit.
  • the kit is accompanied by instructions for administration.
  • the kit may also be accompanied by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or of human or veterinary administration.
  • Such notice for example, may be of the labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert.
  • the unit dosage forms may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • the pill burden would be decreased for patients receiving the unit dosage forms of the present invention in 16 of the 24 possible dosing regimens.
  • the decreased pill burden would be especially significant in patients having a BSA between 1.8-2.0.
  • the pill burden would be decreased in 6 of the possible 8 regimens.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurosurgery (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
EP07796436A 2006-06-26 2007-06-25 Unit dosage forms of temozolomide Withdrawn EP1901740A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US81662306P 2006-06-26 2006-06-26
PCT/US2007/014761 WO2008002544A2 (en) 2006-06-26 2007-06-25 Unit dosage forms of temozolomide

Publications (1)

Publication Number Publication Date
EP1901740A2 true EP1901740A2 (en) 2008-03-26

Family

ID=38704663

Family Applications (1)

Application Number Title Priority Date Filing Date
EP07796436A Withdrawn EP1901740A2 (en) 2006-06-26 2007-06-25 Unit dosage forms of temozolomide

Country Status (13)

Country Link
EP (1) EP1901740A2 (es)
JP (1) JP2008534692A (es)
KR (2) KR20080015777A (es)
CN (1) CN101309686A (es)
AR (1) AR061618A1 (es)
AU (1) AU2007221979A1 (es)
BR (1) BRPI0702847A (es)
CA (1) CA2610439A1 (es)
CL (1) CL2007001864A1 (es)
NO (1) NO20074913L (es)
TW (1) TWI326598B (es)
WO (1) WO2008002544A2 (es)
ZA (1) ZA200708280B (es)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102526038B (zh) * 2011-01-12 2013-09-11 北京人福军威医药技术开发有限公司 替莫唑胺的脑靶向药物组合物及其应用
US10744171B2 (en) * 2016-03-21 2020-08-18 Duke University Sequential anti-cancer treatment
US11147810B2 (en) 2017-03-13 2021-10-19 Ftf Pharma Private Limited Pharmaceutical composition of oral suspension of anti-neoplastic alkylating agents

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102160864A (zh) * 1999-03-30 2011-08-24 先灵公司 用替莫唑胺改善癌症治疗
CN101098696A (zh) * 2004-11-09 2008-01-02 先灵公司 基于患者mgmt水平来治疗癌症的替莫唑胺的改进给药方案

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2008002544A2 *

Also Published As

Publication number Publication date
NO20074913L (no) 2009-03-26
TWI326598B (en) 2010-07-01
ZA200708280B (en) 2009-03-25
TW200808803A (en) 2008-02-16
AU2007221979A9 (en) 2008-01-10
WO2008002544A3 (en) 2008-02-07
JP2008534692A (ja) 2008-08-28
CA2610439A1 (en) 2007-12-26
BRPI0702847A (pt) 2008-04-01
KR20100055543A (ko) 2010-05-26
CN101309686A (zh) 2008-11-19
KR20080015777A (ko) 2008-02-20
WO2008002544A2 (en) 2008-01-03
AU2007221979A1 (en) 2008-01-10
AR061618A1 (es) 2008-09-10
CL2007001864A1 (es) 2008-02-08

Similar Documents

Publication Publication Date Title
AU2005304672B2 (en) Improved dosing regimen of temozolomide for treating cancer based on the patient's MGMT level
Plummer et al. Phase I study of the poly (ADP-ribose) polymerase inhibitor, AG014699, in combination with temozolomide in patients with advanced solid tumors
US20100210700A1 (en) Methods of treatment using intravenous formulations comprising temozolomide
MXPA05007207A (es) Metodos para evaluar y tratar cancer.
US20080090242A1 (en) Panel of biomarkers for prediction of fti efficacy
US20080319039A1 (en) Unit dosage forms of temozolomide
WO2008002544A2 (en) Unit dosage forms of temozolomide
US20060111365A1 (en) Combinations comprising N- {5-[4- [4-methy L-piperazino-methyl) -benzoylamido] -2-methylphenyl) -4- (3-pyridyl) -2-pyrimidine-amine and at least one telomerase inhibitor
US20100240723A1 (en) Methods of treating cell proliferative disorders using a compressed temozolomide dosing schedule
MX2007012093A (es) Formas de dosificacion unitaria de temozolomida
Yung Future directions for temozolomide therapy
CA2472846A1 (en) Methods for assessing and treating leukemia
US20220211710A1 (en) Compositions and methods for treating cancer with nucleoside-metabolism modulators
WO2021074338A1 (en) Methods of determining whether patients suffering from acute myeloid leukemia will achieve a response to an myc-targeting therapy
KR20240026704A (ko) Ginsenoside Rk1을 포함하는 전립선암 예방 또는 치료용 약학적 조성물
Srivastava et al. CANCER CHEMOTHERAPYSONAL SRIVASTAVA, SAKSHI MISHRA, JAYANT DEWANGAN, AMAN DIVAKAR, PRABHASH KUMAR PANDEY, ANDSRIKANTA KUMAR RATH

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: 20070928

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 MT NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

RAX Requested extension states of the european patent have changed

Extension state: RS

Payment date: 20070928

Extension state: MK

Payment date: 20070928

Extension state: HR

Payment date: 20070928

Extension state: BA

Payment date: 20070928

Extension state: AL

Payment date: 20070928

17Q First examination report despatched

Effective date: 20080509

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1109746

Country of ref document: HK

TPAC Observations by third parties

Free format text: ORIGINAL CODE: EPIDOSNTIPA

RBV Designated contracting states (corrected)

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 MT NL PL PT RO SE SI SK TR

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

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20120201

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1109746

Country of ref document: HK