US20110038858A1 - Methods of treating hematologic cancers using pnp inhibitors such as forodesine in combination with alkylating agents or anti-cd20 agents - Google Patents

Methods of treating hematologic cancers using pnp inhibitors such as forodesine in combination with alkylating agents or anti-cd20 agents Download PDF

Info

Publication number
US20110038858A1
US20110038858A1 US12/747,456 US74745608A US2011038858A1 US 20110038858 A1 US20110038858 A1 US 20110038858A1 US 74745608 A US74745608 A US 74745608A US 2011038858 A1 US2011038858 A1 US 2011038858A1
Authority
US
United States
Prior art keywords
forodesine
agent
alkylating agent
subject
pnp inhibitor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/747,456
Other languages
English (en)
Inventor
Shanta Bantia
Philip Breitfeld
Yarlagadda S. Babu
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.)
Biocryst Pharmaceuticals Inc
Original Assignee
Biocryst Pharmaceuticals Inc
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 Biocryst Pharmaceuticals Inc filed Critical Biocryst Pharmaceuticals Inc
Priority to US12/747,456 priority Critical patent/US20110038858A1/en
Assigned to BIOCRYST PHARMACEUTICALS, INC. reassignment BIOCRYST PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BABU, YARLAGADDA S., BANTIA, SHANTA, BREITFELD, PHILIP
Publication of US20110038858A1 publication Critical patent/US20110038858A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • 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
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines

Definitions

  • This application relates to the treatment of hematologic cancers, for example, cancers of the blood, by methods that include administration of a purine nucleoside phosphorylase (PNP) inhibitor.
  • PNP purine nucleoside phosphorylase
  • methods of treating chronic lymphocytic leukemia (CLL) and acute lymphocytic leukemia (ALL) are described.
  • Cancer is now the second leading cause of death in the United States and over 8,000,000 persons in the United States have been diagnosed with cancer. In 1995, cancer accounted for 23.3% of all deaths in the United States (see, e.g., U.S. Dept. of Health and Human Services, National Center for Health Statistics, Health United States 1996-97 and Injury Chartbook 117 (1997)).
  • Cancer is now primarily treated with one or a combination of three types of therapies: surgery; radiation; and chemotherapy.
  • Surgery involves the bulk removal of diseased tissue. While surgery is sometimes effective in removing tumors located at certain sites, for example, in the breast, colon, and skin, it cannot be used in the treatment of tumors located in other areas, such as the backbone, nor in the treatment of disseminated neoplastic conditions such as leukemia.
  • Radiation therapy involves the exposure of living tissue to ionizing radiation causing death or damage to the exposed cells. Side effects from radiation therapy may be acute and temporary, while others may be irreversible.
  • Chemotherapy involves the disruption of cell replication or cell metabolism. It is used most often in the treatment of breast, lung, and testicular cancer. One of the main causes of failure in this treatment of cancer is the development of drug resistance by the cancer cells, a serious problem that may lead to recurrence of disease or even death. Thus, more effective cancer treatments are needed.
  • a method of treating a hematologic cancer in a subject.
  • the method includes the steps of: (a) administering to the subject an effective amount of a purine nucleoside phosphorylase (PNP) inhibitor; and (b) administering to the subject an effective amount of an alkylating agent or an anti-CD20 agent.
  • the PNP inhibitor is Forodesine.
  • the alkylating agent is selected from a mustard derivative, a nitrosourea derivative, a platinum compound, and an imidazole carboxamide compound.
  • the alkylating agent is Bendamustine.
  • the anti-CD20 agent is Rituximab.
  • the PNP inhibitor and alkylating agent or anti-CD20 agent are administered concurrently, while in other embodiments, the PNP inhibitor and alkylating agent or anti-CD20 agent are administered sequentially. In the latter embodiment, the alkylating agent or anti-CD20 agent can be administered one or more times prior to administration of the PNP inhibitor.
  • a method of treating a hematologic cancer in a subject resistant to one or more chemotherapeutic agents can include the steps of: (a) identifying a subject resistant to one or more chemotherapeutic agents; and (b) administering to the subject a PNP inhibitor.
  • the PNP inhibitor is Forodesine.
  • a method of treating a subject with a hematologic cancer can include the steps of: (a) detecting a p53 deletion in one or more cancer cells in a sample from the subject; and (b) administering to the subject a PNP inhibitor.
  • the PNP inhibitor is Forodesine.
  • the method can further include detecting the presence of a 17p deletion, and/or determining if one or more cancer cells in the sample are resistant to one or more chemotherapeutic agents (e.g., an alkylating agent and a purine nucleoside analogue).
  • composition comprising a PNP inhibitor and an alkylating agent or an anti-CD20 agent.
  • kits comprising a PNP inhibitor and an alkylating agent or an anti-CD20 agent.
  • the kit can further include a delivery system for the PNP inhibitor, the alkylating agent, the anti-CD20 agent, or any combination thereof.
  • the kit can also include instructions for treating a subject.
  • a kit comprises a PNP inhibitor.
  • the kit further includes a label that indicates that the contents are to be administered to a subject that is resistant to an alkylating agent.
  • the kit also includes a label that indicates that the contents are to be administered to a subject with a p53 deletion.
  • the kit can include a label that indicates that the contents are to be administered with an alkylating agent or an anti-CD20 agent.
  • Certain embodiments provide the use of a purine nucleoside phosphorylase (PNP) inhibitor and an alkylating agent to prepare a medicament useful for treating a hematologic cancer in an animal.
  • PNP purine nucleoside phosphorylase
  • Certain embodiments provide the use of a purine nucleoside phosphorylase (PNP) inhibitor and an anti-CD20 agent to prepare a medicament useful for treating a hematologic cancer in an animal.
  • PNP purine nucleoside phosphorylase
  • Certain embodiments provide the use of a purine nucleoside phosphorylase (PNP) inhibitor and an alkylating agent for treating a hematologic cancer.
  • PNP purine nucleoside phosphorylase
  • Certain embodiments provide the use of a purine nucleoside phosphorylase (PNP) inhibitor and an anti-CD20 agent for treating a hematologic cancer.
  • PNP purine nucleoside phosphorylase
  • FIGS. 1A and 1B details the cytotoxicity effect of Forodesine in CLL cells exhibiting both high and low ZAP-70 levels.
  • FIG. 2 displays the correlation between an intracellular increase in dGTP levels after Forodesine treatment and amount of cell death induced.
  • FIG. 3 displays values detailing the p53 deleted CLL cases having a high response to Forodesine.
  • FIG. 4 displays the data for treatment with Forodesine of CLL cases with low or no sensitivity to Bendamustine or Fludarabine treatment.
  • FIGS. 5A and 5B displays the combination index data for Forodesine/Bendamustine and Forodesine/Fludarabine.
  • FIG. 6 details the combination index data for Forodesine/Rituximab.
  • forodesine represents a novel chemotherapeutic approach able to induce apoptosis of CLL cells bypassing the ATM/p53 pathway.
  • certain embodiments provide methods of treating a hematologic cancer in a subject comprising the steps of administering to the subject an effective amount of a purine nucleoside phosphorylase (PNP) inhibitor; and administering to the subject an effective amount of an alkylating agent or an anti-CD20 agent.
  • PNP purine nucleoside phosphorylase
  • the PNP inhibitor is Forodesine.
  • the alkylating agent is selected from a mustard derivative, a nitrosourea derivative, a platinum compound, and an imidazole carboxamide compound.
  • the alkylating agent is a mustard derivative.
  • the alkylating agent is Bendamustine.
  • the anti-CD20 agent is Rituximab.
  • the PNP inhibitor and alkylating agent or anti-CD20 agent are administered concurrently.
  • the PNP inhibitor and alkylating agent or anti-CD20 agent are administered sequentially.
  • the alkylating agent or anti-CD20 agent is administered one or more times prior to administration of the PNP inhibitor.
  • the hematologic cancer is selected from chronic lymphocytic leukemia and acute lymphoblastic leukemia.
  • the hematologic cancer is chronic lymphocytic leukemia.
  • the hematologic cancer is acute lymphoblastic leukemia.
  • an effective amount of an alkylating agent is administered to the subject.
  • an effective amount of an anti-CD20 agent is administered to the subject.
  • the methods comprise administering to the subject an effective amount of a PNP inhibitor, an effective amount of an alkylating agent, and an effective amount of an anti-CD20 agent.
  • the PNP inhibitor, alkylating agent and anti-CD20 agent are administered concurrently.
  • the PNP inhibitor, alkylating agent and anti-CD20 agent are administered sequentially.
  • the alkylating agent and anti-CD20 agents are administered one or more times prior to administration of the PNP inhibitor.
  • Certain embodiments provide methods of treating a hematologic cancer in a subject resistant to one or more chemotherapeutic agents comprising the steps of identifying a subject resistant to one or more chemotherapeutic agents; and administering to the subject a PNP inhibitor.
  • the subject is resistant to one or more chemotherapeutic agents selected from the group consisting of an alkylating agent and a purine nucleoside analogue.
  • the alkylating agent is Bendamustine.
  • the purine nucleoside analogue is Fluradadine.
  • Certain embodiments provide methods of treating a subject with a hematologic cancer comprising the steps of detecting a p53 deletion in one or more cancer cells in a sample from the subject; and administering to the subject a PNP inhibitor.
  • the methods can further comprise detecting the presence of a 17p deletion.
  • the methods can further comprise determining if one or more cancer cells in the sample are resistant to one or more chemotherapeutic agents.
  • the cancer cell or cells are resistant to one or more chemotherapeutic agents selected from the group consisting of an alkylating agent and a purine nucleoside analogue.
  • compositions comprising a PNP inhibitor and an alkylating agent or an anti-CD20 agent.
  • the composition comprises Forodesine and Bendamustine.
  • the composition comprises Forodesine and Rituximab.
  • the composition comprises a PNP inhibitor, an alkylating agent, and an anti-CD20 agent.
  • the composition comprises Forodesine, Bendamustine and Rituximab.
  • kits comprising a PNP inhibitor and an alkylating agent or an anti-CD20 agent.
  • kits can further comprise a delivery system for the PNP inhibitor, the alkylating agent, the anti-CD20 agent, or any combination thereof.
  • kits can further comprise instructions for treating a subject.
  • kits comprise a PNP inhibitor and an alkylating agent.
  • kits comprise a PNP inhibitor and an anti-CD20 agent.
  • kits comprise Forodesine and Bendamustine.
  • kits comprise Forodesine and Rituximab.
  • kits comprise a PNP inhibitor, an alkylating agent, and an anti-CD20 agent.
  • kits comprise Forodesine, Bendamustine and Rituximab.
  • kits that comprise a PNP inhibitor.
  • kits comprise a label that indicates that the contents are to be administered to a subject that is resistant to an alkylating agent.
  • kits further comprise a label that indicates that the contents are to be administered to a subject with a p53 deletion.
  • kits further comprise a label that indicates that the contents are to be administered with an alkylating agent or an anti-CD20 agent.
  • forodesine is not incorporated to DNA.
  • Forodesine treatment leads to a dGTP increase in CLL cells, and this increase correlated with cell cytotoxicity, indicating that the dGTP levels reached after forodesine treatment would be a surrogate marker indicative of the cytotoxic response.
  • the susceptibility of CLL to forodesine may be due to the high dCK activity observed in this cells, activity positively regulated by the phosphorylation of dCK on Ser-74. A significant positive correlation was observed between phospho-dCK/dCK ratio and forodesine-induced apoptosis.
  • dCK also catalyzes the phosphorylation required for the activation of several anti-leukemic nucleoside analogues, such as fludarabine, gemcitabine or cladribine.
  • the antagonic effect observed between forodesine and fludarabine can be explained by the reduction on dGTP levels observed after combination of fludarabine with forodesine.
  • dGTP-mediated cell death Several mechanisms for dGTP-mediated cell death have been proposed. For example, accumulated deoxynucleosides can be phosphorylated in the mitochondria by deoxyguanosin kinase and thymidine kinase, leading to abnormal accumulation of dNTPs that might interfere with mitochondrial DNA synthesis and repair, giving to increased sensitivity to mitochondrial damage, p53 activation and apoptosis.
  • the imbalance in mitochondrial dGTP could also affect mitochondrial ATP synthesis and/or inactivation of anti-oxidants enzymes of the mitochondrial electron transport chain, leading to ROS production. Mitochondrial genome is highly sensitive to oxidative stress damage that may rapidly initiate apoptosis.
  • forodesine activated the mitochondrial apoptotic pathway by production of ROS and ⁇ m loss, leading to caspase-dependent and independent apoptosis.
  • ROS are generated by the mitochondrial electron transport, and under severe oxidative stress the increase in O 2 ⁇ , OH ⁇ or H 2 O 2 levels provoke loss of ⁇ m and cell death.
  • These events induced by forodesine were reverted by pre-incubation of CLL cells with Tiron and NAC, specific scavengers of O 2 ⁇ , which supports a role of oxidative stress preceding the activation of mitochondrial apoptotic pathway.
  • Phosphorylation and activation of p53 is induced by the DNA damage response, but also by several stress signals.
  • E2F-1 increases p53 phosphorylation at residues that are also phosphorylated in response to DNA damage, but also is able to induce cell death by activation of the p53 homologue p73 via a p53-independent apoptotic pathway. How ROS generation can regulate E2F-1, p53 and/or p73 activation and cell death remains poorly understood.
  • An early event of the mitochondrial apoptotic pathway is the formation of the apoptosome and activation of caspase-9, which cleaves and activates caspase-3 as well caspase-8.
  • Forodesine induced a time-related activation of caspase-9 and -3, as well pro-caspase-8 simultaneously to caspase-9 activation.
  • caspase-8 induced the cleavage of BID protein to its pro-apoptotic truncated form, that activates mitochondrial apoptotic pathway.
  • caspase-8 reduced ⁇ m loss induced by forodesine, but the effect on cell death at later times was moderated, suggesting that caspase-8/BID lead to the amplification loop of the mitochondrial apoptotic pathway.
  • Caspase-9 activation could be not enough per sec to induce apoptosis, so the activation of caspase-8/BID together with the decrease in the inhibitos of apoptosis XIAP and survivin would increase caspase-9 and -3 activities, and therefore potentiates apoptosis induced by forodesine.
  • the BCL-2 family of proteins controls the commitment to apoptotic cell death.
  • a balance between pro-survival such as BCL-2 and MCL-1 and pro-apoptotic BCL-2 members (BAX, BAK and the BH3-only proteins BIM, PUMA, NOXA, BAD, BID, BMF, BIK and HRK) controls the outcome of many death signalling pathways.
  • pro-survival such as BCL-2 and MCL-1 and pro-apoptotic BCL-2 members
  • BIM pro-apoptotic BCL-2 members
  • BIM is associated with MCL-1, so the decrease in MCL-1 levels would make CLL cells susceptible to this BH3-only protein.
  • BIM as well truncated Bid, have dual functions as both inhibitors of anti-apoptotic BCL-2 members and direct activators of pro-apoptotic BAX and BAK. In this sense, the increase in BIM levels, together with Bid activation would lead to BAX and BAK activation, and together with MCL-1 decrease, the remaining anti-apoptotic capacity of BCL-2 would be overwhelmed.
  • ROS scavengers block the induction of FOXO3a and BIM. It has been described that the expression of both FOXO1a and BIM and subsequent apoptosis is regulated by p73 in tumoral cells defective of p53. After forodesine treatment, a protein and mRNA marked up-regulation of p73 and BIM independent of p53 status was shown, as well an increase in FOXO1 and FOXO3a levels, increased that was detected at early times. The expression of FOXO-regulated target genes can be controlled by any of the FOXO members, suggesting a redundant mechanism of action, as shown by the transcriptional regulation of BIM exerted by both FOXO1A and FOXO3A.
  • results described herein provide evidence of a mechanism involved in forodesine induced cell death in CLL cells independent of p53 status, revealing that different programmed cell death pathways can coexists in the same cell and can be selectively induced by diverse stimuli.
  • forodesine as a single agent, or in combination with bendamustine or rituximab, to be highly effective in the treatment of CLL. Therefore, forodesine, available, e.g., in oral and intra-venous formulation with low toxicity profiles, is a treatment option for patients with poop prognosis (e.g., 17p-), patients with refractory disease and/or treatment option for elderly patients.
  • poop prognosis e.g., 17p-
  • a multi-centre, open-label phase I clinical trial of forodesine in relapsed CLL patients has been initiated.
  • hematologic cancers for example, cancers of the blood, in a subject.
  • these types of cancers include, e.g., acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), myeloproliferative diseases, multiple myeloma, and myelodysplastic syndrome, Hodgkin's lymphoma, non Hodgkin's lymphoma (malignant lymphoma) and Waldenström's macroglobulinemia.
  • the hematologic cancer is CLL.
  • the hematologic cancer is ALL.
  • a subject can include both mammals and non-mammals.
  • Mammals include, for example, humans; nonhuman primates, e.g. apes and monkeys; cattle; horses; sheep; rats; mice; pigs; and goats.
  • Non mammals include, for example, fish and birds.
  • the method includes administering a purine nucleoside phosphorylase (PNP) inhibitor and an alkylating agent or an anti-CD20 agent to the subject.
  • PNP purine nucleoside phosphorylase
  • a PNP inhibitor and an alkylating agent are administered.
  • a PNP inhibitor and an anti-CD20 agent are administered.
  • a PNP inhibitor is administered after identifying a subject resistant to one or more chemotherapeutic agents (e.g., Bendamustine or Fluarabine).
  • a PNP inhibitor is administered after detecting a p53 deletion in a subject.
  • a PNP inhibitor can induce an increase of plasma 2′-deoxyguanosine (dGuo) and the accumulation of intracellular deoxyguanosine triphosphate (dGTP) leading to cell death induction.
  • PNP inhibitors can include those disclosed in U.S. Pat. Nos. 4,985,433; 4,985,434, 5,008,265; 5,008,270; 5,565,463 and 5,721,240 assigned to BioCryst Pharmaceuticals, Inc., disclosures of which are incorporated herein by reference.
  • the PNP inhibitor is Forodesine or a salt thereof, including the HCl salt:
  • an alkylating agent refers to a chemotherapeutic compound that chemically modifies DNA and disrupts its function. Some alkylating agents cause formation of cross links between nucleotides on the same strand, or the complementary strand, of a double-stranded DNA molecule, while still others cause base-pair mismatching between DNA strands.
  • An alkylating agent can be a mustard derivative, a nitrosourea derivative, a platinum compound, or an imidazole carboxamide compound.
  • alkylating agents include Bendamustine, Busulfan, Carboplatin, Carmustine, Cisplatin, Chlorambucil, Cyclophosphamide, dacarbazine, Hexamethylmelamine, Ifosphamide, Lomustine, Mechlorethamine, Melphalan, Mitotane, Mytomycin, Pipobroman, Procarbazine, Streptozocin, Thiotepa, and Triethylenemelamine.
  • the alkylating agent can be Bendamustine, or a salt thereof, including the HCl salt:
  • An anti-CD20 agent can be any agent that targets (e.g., selectively binds to) the B-cell cells surface protein CD20.
  • the anti-CD20 agent is an antibody specific for CD20. Without being bound by theory, it is thought that these agents can operate by one of three mechanisms: (1) complement-mediated cytotoxicity; (2) antibody-dependent cell-mediated cytotoxicity; and (3) induction of apoptosis.
  • anti-CD20 agents include Rituximab, Ibritumomab, Trastuzumab, Gemtuzumab, and Alemtuzumab. In some embodiments, the anti-CD20 agent is Rituximab.
  • the PNP inhibitor, alkylating agent, and/or anti-CD20 agent can be administered by any route, e.g., intra-operative, intrathecal, intradiskal, peridiskal, epidural (including periradicular and transforaminal), any combination of intradiskal, epidural, and peridural, perispinal, IV, intramuscular, SC, oral, intranasal, inhalation, transdermal, and parenteral.
  • any route e.g., intra-operative, intrathecal, intradiskal, peridiskal, epidural (including periradicular and transforaminal), any combination of intradiskal, epidural, and peridural, perispinal, IV, intramuscular, SC, oral, intranasal, inhalation, transdermal, and parenteral.
  • the PNP inhibitor, alkylating agent, and/or anti-CD20 agent can be formulated with a pharmaceutically acceptable carrier selected on the basis of the selected route of administration and standard pharmaceutical practice.
  • the PNP inhibitor, alkylating agent, and/or anti-CD20 agent may be formulated into dosage forms according to standard practices in the field of pharmaceutical preparations. See Alphonso Gennaro, ed., Remington's Pharmaceutical Sciences, 18th Edition (1990), Mack Publishing Co., Easton, Pa.
  • Suitable dosage forms may comprise, for example, tablets, capsules, solutions, parenteral solutions, troches, suppositories, or suspensions.
  • the PNP inhibitor, alkylating agent, and/or anti-CD20 agent may be mixed with a suitable carrier or diluent such as water, an oil (particularly a vegetable oil), ethanol, saline solution, aqueous dextrose (glucose) and related sugar solutions, glycerol, or a glycol such as propylene glycol or polyethylene glycol.
  • a suitable carrier or diluent such as water, an oil (particularly a vegetable oil), ethanol, saline solution, aqueous dextrose (glucose) and related sugar solutions, glycerol, or a glycol such as propylene glycol or polyethylene glycol.
  • Solutions for parenteral administration preferably contain a water soluble salt of the PNP inhibitor, alkylating agent, and/or anti-CD20 agent.
  • Stabilizing agents, antioxidant agents and preservatives may also be added. Suitable antioxidant agents include sulfite, ascorbic acid, citric acid and its salt
  • Suitable preservatives include benzalkonium chloride, methyl- or propyl-paraben, and chlorbutanol.
  • the composition for parenteral administration may take the form of an aqueous or non-aqueous solution, dispersion, suspension or emulsion.
  • the PNP inhibitor, alkylating agent, and/or anti-CD20 agent may be combined with one or more solid inactive ingredients for the preparation of tablets, capsules, pills, powders, granules or other suitable oral age forms.
  • the PNP inhibitor, alkylating agent, and/or anti-CD20 agent may be combined with at least one excipient such as fillers, binders, humectants, disintegrating agents, solution retarders, absorption accelerators, wetting agents absorbents or lubricating agents.
  • a PNP inhibitor, alkylating agent, and/or anti-CD20 agent will, of course, be determined by the particular circumstances of the individual patient including the size, weight, age and sex of the patient, the nature and stage of the disease being treated, the aggressiveness of the disease disorder, and the route of administration of the compound.
  • Doses and schedules of the compounds e.g., forodesine, bendamustine and rituximab, may be administered singly or in combination according to, e.g., doses and schedules indicated in FDA approved labels.
  • the PNP inhibitor and alkylating agent or anti-CD20 agent are administered concurrently, while in other embodiments the PNP inhibitor and alkylating agent or anti-CD20 agent are administered sequentially.
  • the alkylating agent or anti-CD20 agent can be administered one or more times prior to administration of the PNP inhibitor (e.g., two times, three times, four times, five times, 10 times, or 20 times).
  • the PNP inhibitor may be administered one or more times prior to the administration of the alkylating agent or anti-CD20 agent (e.g., two times, three times, four times, five times, 10 times, or 20 times).
  • treatment of a hematologic cancer in a subject may include identifying a subject who is resistant to one or more chemotherapeutic agents.
  • One of the main causes of failure in the treatment of cancer is the development of drug resistance by the cancer cells. This is a very serious problem that may lead to recurrence of disease or even death.
  • the subject resistant to one or more chemotherapeutic agents can be identified by means known in the art.
  • the subject may be resistant to any known chemotherapeutic agent.
  • the subject can be resistant to an alkylating agent (e.g., Bendamustine) and/or purine nucleoside analogue (e.g., Fluradadine).
  • the subject can be administered a PNP inhibitor.
  • the PNP inhibitor is Forodesine HCl.
  • treatment of a hematologic cancer in a subject can include the detection of the presence of a p53 deletion, e.g. a 17p deletion, in one or more cancer cells in a sample from the subject.
  • a p53 deletion e.g. a 17p deletion
  • the 17p deletion is a marker that can identify a hematologic cancer subject that may exhibit a different biological and clinical behavior.
  • p53 alterations can convey drug resistance and shorter survival periods.
  • a subject that presents a p53 deletion can be administered a PNP inhibitor.
  • the PNP inhibitor is Forodesine HCl.
  • chemotherapeutic agents in particular, a PNP inhibitor and an alkylating agent or a PNP inhibitor and an anti-CD20 agent.
  • chemotherapeutic agents in particular, a PNP inhibitor and an alkylating agent or a PNP inhibitor and an anti-CD20 agent.
  • a PNP inhibitor and an alkylating agent or anti-CD20 agent can produce a synergistic effect.
  • This effect can be demonstrated through the development of a combination index (CI).
  • the index can be calculated as a function of the fraction of cells affected according to the procedure of Chou et al., Advance Enz. Regul., 22, 27-55 (1985). This is a well-known test that evaluates coefficient interactions against a range of cell death proportions. For example, if treatment with drug A results in 30% cell death and treatment with drug B results in 50% cell death, than it would be expected that the combination of the two drugs would result in 65% cell death.
  • the ratio of the predicted cell death to that actually measured upon combination of the drugs is less than one, then a synergistic effect is observed. If, however, the ratio is greater than one, then an antagonistic effect is observed.
  • the combination of Forodesine and Bendamustine shows a synergistic effect
  • the combination of Forodesine and Fludarabine shows an antagnonistic effect (see Example 5).
  • compositions comprising a PNP-inhibitor and an alkylating agent or a PNP-inhibitor and an anti-CD20 agent.
  • a PNP-inhibitor can include Forodesine (BCX-1777).
  • an alkylating agent can include Bendamustine.
  • an anti-CD20 agent is Rituximab.
  • compositions provided herein contain a PNP inhibitor and an alkylating agent or a PNP-inhibitor and an anti-CD20 agent in amounts that are useful in the treatment of hematologic cancers, and a pharmaceutically acceptable carrier.
  • Pharmaceutical carriers suitable for administration of the compounds provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration.
  • compositions can be, in one embodiment, formulated into suitable pharmaceutical preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as transdermal patch preparation and dry powder inhalers (see, e.g., Ansel Introduction to Pharmaceutical Dosage Forms, Fourth Edition 1985, 126).
  • concentration of the PNP inhibitor and the alkylating agent or a PNP-inhibitor and an anti-CD20 agent in the pharmaceutical composition will depend on absorption, inactivation and excretion rates of the compounds, the physicochemical characteristics of the compounds, the dosage schedule, and amount administered as well as other factors known to those of skill in the art. For example, the amount that is delivered is sufficient to treat chronic lymphocyte leukemia, as described herein.
  • the pharmaceutical composition may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment is a function of the disease being treated and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.
  • the pharmaceutical compositions are provided for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water emulsions containing suitable quantities of the compounds or pharmaceutically acceptable derivatives thereof.
  • the pharmaceutically therapeutically active compounds and derivatives thereof are, in one embodiment, formulated and administered in unit-dosage forms or multiple-dosage forms.
  • Unit-dose forms as used herein refers to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the therapeutically active compound sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent.
  • unit-dose forms include ampoules and syringes and individually packaged tablets or capsules. Unit-dose forms may be administered in fractions or multiples thereof.
  • a multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form. Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pints or gallons. Hence, multiple dose form is a multiple of unit-doses which are not segregated in packaging.
  • Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.
  • a carrier such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension.
  • the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • nontoxic auxiliary substances such as wetting agents, emulsifying agents, solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents.
  • compositions containing a PNP inhibitor and an alkylating agent or a PNP-inhibitor and an anti-CD20 agent in the range of 0.005% to 100% with the balance made up from non-toxic carrier may be prepared. Methods for preparation of these compositions are known to those skilled in the art.
  • the contemplated compositions may contain 0.001%-100% active ingredient, in one embodiment 0.1-95%, in another embodiment 75-85%.
  • kits typically include a PNP inhibitor and an alkylating agent or a PNP-inhibitor and an anti-CD20 agent.
  • a kit can include one or more delivery systems, e.g., for the PNP inhibitor, the alkylating agent, the anti-CD20, or any combination thereof, and directions for use of the kit (e.g., instructions for treating a subject).
  • a kit can include the PNP inhibitor and/or the alkylating agent.
  • a kit can include the PNP inhibitor and/or the anti-CD20 agent.
  • the kit can include a PNP inhibitor and a label that indicates that the contents are to be administered to a subject resistant to alkylating agents, such as Bendamustine.
  • the kit can include a PNP inhibitor and a label that indicates that the contents are to be administered to a subject with a p53 deletion (e.g., a 17p deletion).
  • a kit can include a PNP inhibitor and a label that indicates that the contents are to be administered with an alkylating agent or an anti-CD20 agent.
  • an amount of compound in a method refers to the amount of a compound that achieves the desired pharmacological effect or other effect, for example an amount that inhibits the abnormal growth or proliferation, or induces apoptosis of cancer cells, resulting in a useful effect.
  • treating and “treatment” mean causing a therapeutically beneficial effect, such as ameliorating existing symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, postponing or preventing the further development of a disorder and/or reducing the severity of symptoms that will or are expected to develop.
  • the purine nucleoside phosphorylase inhibitor forodesine induces p53-independent mitochondrial apoptosis in chronic lymphocytic leukemia cells through Mcl-1 downregulation and induction of p73 and Bim.
  • Chronic lymphocytic leukemia is a clinical heterogeneous entity derived from the monoclonal expansion of long-life CD5 + B-lymphocytes with abnormal regulation of apoptosis and low proliferation rates.
  • CLL Chronic lymphocytic leukemia
  • An unmutated profile of immunoglobulin genes, high expression of ZAP-70 protein, high CD38 expression and the presence of certain cytogenetic abnormalities are associated with poorer overall survival and a shorter time to disease progression of CLL patients.
  • Chemotherapeutic anti-cancer drugs induce apoptosis usually either via DNA damage response and p53 activation, and/or directly via perturbation of mitochondrial function.
  • the DNA-damage response pathway appears to play a key role in CLL drug-induced apoptosis, as cells from CLL patients with TP53 abnormalities show resistance to conventional chemotherapy and short survival.
  • the lack of p53 function by deletion of p53 and mutation of the remaining allele increases in CLL patient's refractory to chemotherapy, being “double-hits” during the acquisition of drug resistance that abrogates the transcriptional and mitochondrial apoptotic activity of p53. Therefore, new effective approaches should induce apoptosis through DNA-damage-independent pathways and/or direct activation of apoptotic pathways independent of p53.
  • CLL cells A hallmark of CLL cells is their resistance to apoptosis induction, being Bcl-2 family proteins critical regulators of apoptosis and survival in CLL cells. High levels of anti-apoptotic Bcl-2 and Mcl-1 proteins are associated with aggressive disease and resistance to chemotherapy in CLL. MCL-1 has also an important role in prolonging the CLL cell survival, as MCL-1 levels inversely correlated with in vitro and clinical response to several drugs.
  • Purine nucleoside phosphorylase is an enzyme in the purine salvage pathway that phosphorylates purine analogues to their respective bases and deoxyribose phosphate.
  • Forodesine or immucillin H (BCX-1777) is a potent transition-state analogue inhibitor of PNP that has demonstrated inhibition of T-cell proliferation in vitro and in vivo Bantia et al., Int Immunopharmacol, 1(6), 1199-210 (2001); Kicska et al., PNAS, 98(8), 4593-4598 (2001); Bantia et al., Int Immunopharmacol, 3(6), 879-887 (2003); and Vogel et al., Semin Oncol, 34(6 Suppl 5), S8-12 (2007)).
  • Forodesine has shown a low toxicity profile (Gandhi et al., Blood, 106(13), 4253-4260 (2005); and Korycka et al., Mini Rev Med Chem, 7(9), 976-983 (2007)) and is being studied in clinical trials for patients with T-cell prolymphocytic leukemia, cutaneous T-cell lymphoma and B-cell acute lymphoblastic leukemia (Galmarini et al., IDrugs, 9(10), 712-722 (2006)). Also, forodesine appears to exert an ex vivo cytotoxic effect in CLL cells (Balakrishnan et al., Blood, 108(7), 2392-2398 (2006)).
  • CLL cells have high (dCK) activity, which is a primary enzyme for the conversion of dGuo to dGMP, which is then converted to dGTP, being CLL cells susceptible to PNP inhibition.
  • purine nucleoside analogs such as fludarabine or bendamustine, forodesine in not incorporated into DNA and represents a new class of selective anti-tumour agent with a novel and not fully understood mechanism of action.
  • forodesine appears to be a highly effective therapy in the treatment of CLL patients independent of their ZAP-70, CD38, p53 status or cytogenetic abnormalities. Further, forodesine appears to induce activation of the mitochondrial apoptotic pathway by decreasing the levels of Mcl-1 protein and induction of p73 and pro-apoptotic Bim protein. Interestingly, no significative differences in these apoptotic markers were observed based on p53 status, suggesting a common apoptotic pathway independent of p53-mediated cell death.
  • a dose escalation study was conducted to evaluate the in vitro forodesine cytotoxicity in primary leukemic lymphocytes from CLL patients.
  • an external source of dGuo was added together with forodesine.
  • No differences on cell death were observed using higher doses of forodesine (even up to 15 ⁇ M).
  • increasing the dose of dGuo resulted in a higher induction of cell death, so the subsequent studies were performed using a single dose of forodesine (2 ⁇ M) together with 10 or 20 ⁇ M of dGuo.
  • cytotoxic effect of forodesine and dGuo were analyzed in primary leukemic cells from 43 patients with CLL.
  • a mean cytotoxicity with respect to control of 44.2% ⁇ 11.4 and 57.4% ⁇ 13.1 at 24 h and 48 h respectively was observed.
  • the cytotoxic effect was higher than 60% in 21 cases (48.8% of total), between 40-60% in 18 cases (41.8% of total) and lower than 40% in only 4 cases (9.4% of total).
  • Cytotoxicity of forodesine 2 ⁇ M and dGuo (10-20 ⁇ M) in PBMCs from healthy donors was relatively lower compared to CLL cells, both in T-lymphocytes (CD3 + cells) and B-lymphocytes (CD19 + cells).
  • the mean cytotoxicity in CLL with no 17p deletion was 55.3 ⁇ 8, 50.6 ⁇ 11 and 49.2 ⁇ 18 for forodesine, fludarabine and bendamustine, respectively.
  • the acquisition of 17p deletion, leading to p53 alterations, is associated with in vitro and in vivo fludarabine resistance in CLL patients (Dohner et al., Blood, 85(6), 1580-1589 (1995); and Turgut et al., Leuk Lymphoma, 48(2), 311-320 (2007)).
  • the majority CLL patients with 17p deletion showed a high response to forodesine, with a mean cytotoxicity at 48 hours of 60.1% ⁇ 21 (forodesine 2 ⁇ M and dGuo 20 ⁇ M).
  • the combination index or CI value was calculated using Chou and Talalay's algorithm (Chou et al., Adv Enzyme Regul, 22, 27-55 (1984)) as an indicative marker for the antagonic or synergistic effect for the combination of two different drugs. Briefly, a CI value higher than 1 is indicative of an antagonistic effect, whereas a CI value lower than 1 is indicative of a synergistic effect.
  • the combination studies performed with fludarabine and forodesine showed CI values higher than 1 in all CLL cases analyzed (48 hours), indicating an antagonic effect between both drugs. The mean cytotoxicity achieved with the forodesine and fludarabine combination was lower than caused by both drugs.
  • Forodesine was also effective in those CLL cases with low response to bendamustine, but in contrast to fludarabine, a high increase on cell death was observed with the combination of bendamustine and forodesine and a potent synergistic effect (CI ⁇ 1) was observed. Frodesine clearly enhance the cytotoxic response of a low dose of bendamustine (10 ⁇ M, mean cytotoxicity of 32.95%), achieving a mean cytotoxic effect for the combination of both drugs of 70.5%.
  • the intracellular levels of dGTP in primary cells from 26 CLL patients treated with forodesine 2 ⁇ M and dGuo 10 ⁇ M was alanyzed.
  • a significative direct correlation (p ⁇ 0.05) between the fold increase in the dGTP levels analyzed at 18 hours and forodesine-cytotoxicity at 48 hours was observed.
  • Forodesine induced a high increase in intracellular dGTP levels (up to 96 times fold increase with respect to control basal levels, reaching values between 6 and 129 pmoles of dGTP/10 6 millions of cells).
  • Four CLL cases showed no or low increase in dGTP levels, and a low cytotoxic response to forodesine was observed in these cases.
  • deoxycytidine is the primary substrate of dCK, it should inhibit the phosphorylation of dGuo by dCK, affecting the intracellular increase in dGTP and subsequent apoptosis induced by forodesine.
  • Pre-incubation of cells with deoxycytidine (5-10 ⁇ M) inhibited forodesine-induced cell death.
  • nucleoside analogues used in anti-cancer chemotherapy are phosphorylated by dCK in order to be active.
  • Deoxycytidine also reverted the loss of cell viability induced by fludarabine, whereas cell death induced by bendamustine (drug that may acts independent of dCK) was not reverted.
  • dCK activity is positively regulated by phosphorylation at Ser-74 (Smal et al., J Biol Chem, 281(8), 4887-4893 (2006); Smal et al., Nucleosides Nucleotides Nucleic Acids, 25(9-11), 1141-1146 (2006); Smal et al., Cancer Lett, 253(1), 68-73 (2007)).
  • An increase in the phosphorylated form (at Ser-74) of dCK in CLL cells was observed, an increase not exerted by bendamustine.
  • Forodesine induced loss of mitochondrial transmembrane potential ( ⁇ m) at early times. Reactive-oxygen species (ROS) can be generated after mitochondrial damage and may subsequently mediate apoptosis (Villamor et al., Curr Pharm Des, 10(8), 841-853 (2004)).
  • ROS Reactive-oxygen species
  • Forodesine also induced the production of ROS in CLL primary cells. Mitochondrial depolarization and ROS production was observed as early events, as they were evident after 10 hours of treatment with forodesine.
  • GSH glutathione-reduced ethyl ester
  • NAC N-acetyl cysteine
  • Tiron reduced ⁇ m loss and ROS production induced by forodesine.
  • caspase-8 In correlation with activation of caspase-8, forodesine also induced a decreased of BH3-only protein BID, a main substrate of caspase-8, to give its truncated pro-apoptotic form, that also activates the mitochondrial apoptotic pathway.
  • a decrease of the inhibitors of apoptosis XIAP and survivin and the proteolityic cleavage of PARP, a caspase-3 substrate was also observed, in correlation with apoptosis induction.
  • caspase activation in forodesine-induced apoptosis was investigated, as activation of the mitochondrial apoptotic pathway leads to caspase-dependent but also independent cell death.
  • Treatment of cells with the broad range caspase inhibitor z-VAD.fmk partially reduced phospatidylserine exposure after forodesine treatment for 24 hours, suggesting the implication of both, caspase-dependent as well independent mechanisms of action.
  • caspase-8/BID Activation of caspase-8/BID might play a key role during apoptosis induction exerted by forodesine, or be a secondary side-event due to caspase-9 activation and therefore downstream of mitochondria.
  • apoptosis induction analyzed by Annexin V staining, was partially blocked by the specific caspase-8 inhibitor z-IETD.fmk, whereas the reversion on ⁇ m loss observed was more pronounced.
  • the mitochondrial apoptotic pathway is regulated by a tight balance between pro- and anti-apoptotic members that belong to the BCL-2 family proteins, such as anti-apoptotic BCL-2 and MCL-1 proteins and pro-apoptotic BAX, BAK, BID and BIM proteins among others.
  • CLL cells express high levels of the anti-apoptotic proteins MCL-1 and BCL-2 that inversely correlates with in vitro and clinical response to chemotherapy. The effect of forodesine incubation on the levels of these anti-apoptotic proteins was investigated. Upon forodesine treatment, the levels of the anti-apoptotic MCL-1 protein considerably decreased, whereas BCL-2 protein levels were not affected.
  • the BH3-only protein BIM interacts with BCL-2 and others anti-apoptotic BCL-2 family members to induce apoptosis.
  • Forodesine induced an increase in the protein levels of the pro-apoptotic BIM protein levels.
  • the changes in BIM and MCL-1 protein levels was confirmed by densitometric analysis in seven CLL cases treated with forodesine.
  • BCL-2 and MCL-1 act as guardians of the mitochondrial membrane, preserving its integrity from the action of effector pro-apoptotic proteins, such as BAX and BAK.
  • BIM and truncated BID are the only BH3-only proteins that have the capacity to act as direct activators of BAX and BAK, and the activation of BAX and BAK by flow cytometry after incubation of CLL cells with forodesine was analyzed.
  • Forodesine induced the conformational change of BAX and BAK that allow these proteins to insert into the outer mitochondrial membrane, its oligomerization, the subsequent induction of mitochondrial membrane permeabilization and activation of the cell death machinery.
  • Forodesine Treatment Triggered an Increase of p73 at mRNA and Protein Levels and the Induction of FOXO1 and FOXO3A.
  • forodesine exerted a cytotoxic effect in all CLL cases irrespective of the p53 status
  • forodesine was able to induce stabilization of p53 protein in CLL cases with no 17p deletion.
  • Induction of the TAp73 protein, a p53 related protein needed for p53-mediated apoptosis, is able to overcome the resistance to apoptosis of CLL cells lacking functional p53 (Dicker et al., Blood, 108(10), 3450-3457 (2006)).
  • Forodesine treatment induces a clear up-regulation of p73 mRNA and TAp73 protein levels in all CLL cases analyzed.
  • p73 regulates the induction of the pro-apoptotic protein BIM through upregulation of the transcription factors FOXO1 and FOXO3a in a way p53-dependent but also p53-independent in tumoral cells (Amin et al., Cancer Res, 67(12), 5617-5621 (2007)), although the mechanism of FOXO induction is unknown.
  • the levels of FOXO1 and FOXO3a in CLL cells treated with forodesine was investigated. Forodesine induced an increase in both, FOXO1 and FOXO3a, in correlation with the increase in p73 and BIM protein levels observed.
  • Forodesine BCX-1777/immucillin H for laboratory use was provided by BioCryst Pharmaceuticals Inc. (Birmingham, USA) and deoxyguanosine (dGuo) was purchased from Sigma.
  • dGTP deoxyguanosine triphosphate
  • dNTPs dNTPs
  • [ 3 H]dATP were obtained from Amersham Biosciences. Fludarabine (Shering, Berlin, Germany), bendamustine hydrochloride (TreandaTM, provided by Cephalon, Inc., Frazer, Pa.), rituximab (Roche, Basel, Switzerland) and 2′-deoxycytidine (Sigma) were used for cytotoxicity assays.
  • the present in vitro study was carried out in primary leukemic lymphocytes from 43 patients diagnosed with CLL according to the World Health Organization classification. Informed consent was obtained from each patient.
  • PBMCs Peripheral blood mononuclear cells
  • FBS heat-inactivated fetal bovine serum
  • mononuclear cells from CLL patients (2 ⁇ 10 6 cells/mL) were cultured in RPMI 1640 culture medium (Gibco), supplemented with 10% FBS, 2 mM glutamine and 50 ⁇ g/mL penicillin-streptomycin, in a humidified atmosphere at 37° C. containing 5% carbon dioxide.
  • the percentage of tumoral cells (CD19 + , CD5 + ), ZAP-70 and CD38 expression levels were analyzed by flow cytometry and quantified as previously described (Crespo et al., N Engl J Med, 348(18), 1764-1775 (2003)).
  • the cut-off point for high expression levels of ZAP-70 was ⁇ 20% and ⁇ 30% for CD38 (Hus et al., Ann Oncol, 17(4), 683-690 (2006)). All the CLL samples used in this study carried more than 95% of tumoral cells.
  • Cytogenetic alterations were assessed by fluorescence in situ hybridization (FISH) using the multiprobe commercial kit from Vysis (Downers Grove, Ill.), that contains locus-specific probes to determine the deletions of 17p13.1 (p53), 11q22.3 (ATM) and 13q14.3 (D13S319) and a centromeric probe to detect trisomy 12.
  • FISH fluorescence in situ hybridization
  • the mutations of p53 gene are usually missense mutations, and the mutant protein has a prolonged half-life enabling its detection by Western Blot.
  • p53 mutations were confirmed by direct sequencing according to the IARC TP53 consortium.
  • cells were pre-treated for 4, 12 or 24 hours with fludarabine or bendamustine, or 1 hour with the monoclonal antibody anti-CD20 rituximab before adding forodesine (2 ⁇ M) plus dGuo (10-20 ⁇ M).
  • forodesine 2 ⁇ M
  • dGuo 10-20 ⁇ M
  • Cell viability and apoptosis induction was analyzed by changes in cell complexity by means of FSC/SSC, quantification of phosphatidyl serine (PS) exposure by double staining with Annexin-V conjugated to fluorescein isothiocyanate (FITC) and propidium iodide (PI) (BenderMedsystems, Vienna, Austria).
  • PS phosphatidyl serine
  • FITC fluorescein isothiocyanate
  • PI propidium iodide
  • PBMCs were labeled simultaneously with anti-CD3-FITC (Immunotech, Marseille, France), anti-CD19-PE (Becton Dickinson) and Annexin-V-APC.
  • Cell viability and cytotoxicity were plotted as percentage with respect to control cells.
  • Loss of mitochondrial transmembrane potential ( ⁇ m) were evaluated by staining cells with 20 nM of DiOC 6 (3,3-diexyloxacarbocyanine iodide, Molecular Probes) and reactive oxygen species (ROS) production was determined by staining cells with 2 ⁇ M dihydroethidine (DHE; Molecular Probes) and flow cytometry analysis.
  • DiOC 6 3,3-diexyloxacarbocyanine iodide
  • ROS reactive oxygen species
  • cells were washed once in PBS and fixed in paraformaldehyde 4%, permeabilized with 0.1% saponin and 0.5% bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • Cells were stained with 1 ⁇ g/ml of primary antibodies against conformational active Bak (Oncogene Research), or Bax (clone 6A7, BD Pharmingen) antibodies at room temperature.
  • a secondary goat anti-mouse FITC DAKO
  • goat anti-rabbit FITC Supertechs
  • Cells were lysed for 15 min in RIPA buffer supplemented with protease and phosphatase inhibitors (leupeptine 10 ⁇ g/ml, apoprotinine 10 ⁇ g/ml, 1 ⁇ M PMSF, 1 ⁇ M sodium ortovanadate, 1 ⁇ M NaF, 2 ⁇ M sodium pyrophosphate decahydrate (Sigma).
  • protease and phosphatase inhibitors leupeptine 10 ⁇ g/ml, apoprotinine 10 ⁇ g/ml, 1 ⁇ M PMSF, 1 ⁇ M sodium ortovanadate, 1 ⁇ M NaF, 2 ⁇ M sodium pyrophosphate decahydrate (Sigma).
  • protease and phosphatase inhibitors leupeptine 10 ⁇ g/ml, apoprotinine 10 ⁇ g/ml, 1 ⁇ M PMSF, 1 ⁇ M sodium ortovanadate, 1 ⁇ M NaF, 2 ⁇ M sodium pyrophosphat
  • membranes were probed with the following primary antibodies: anti-Bim, anti-Bak (Ab1) anti-Caspase-8 (Ab-3), anti-p53 (Ab-2) (Calbiochem); anti-Bid, anti-Caspase-9, anti-FoxO1 and anti-XIAP (Cell Signaling Technologies); anti-Bcl-2, anti-dCK, anti-Mcl-1 (S-19) and anti-p73 (clone 5B429) (Santa Cruz Biotechnology); anti-PARP (Roche); anti-Caspase-3 and anti-Bax (clone 6A7) (BD-Pharmingen); anti-survivin (Abcam); anti- ⁇ -actin and anti- ⁇ -tubulin (Sigma) and anti-FoxO3A (Upstate).
  • Rabbit Anti-phospho dCK (Ser79) and rabbit anti-dCK were a kindly provided by Caroline Smal and Institute Bontemps (Universite Catholique de Louvain, Belgium). After the incubation with the appropriate primary antibody, the blots were developed with horseradish peroxidase (HRP)-labeled anti-mouse (Sigma), anti-rabbit (Sigma) or anti-goat (Dako) antibodies by using enhanced chemiluminiscence (ECL) reagents (Pierce). Equal protein loading was confirmed with ⁇ -actin or ⁇ -tubulin expression and relative protein quantification was done with Image Gauge Fujifilm software (Fuji).
  • HRP horseradish peroxidase
  • ECL enhanced chemiluminiscence
  • Ct comparative cycle threshold

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Oncology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Endocrinology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US12/747,456 2007-12-10 2008-12-10 Methods of treating hematologic cancers using pnp inhibitors such as forodesine in combination with alkylating agents or anti-cd20 agents Abandoned US20110038858A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/747,456 US20110038858A1 (en) 2007-12-10 2008-12-10 Methods of treating hematologic cancers using pnp inhibitors such as forodesine in combination with alkylating agents or anti-cd20 agents

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US1276207P 2007-12-10 2007-12-10
PCT/US2008/086256 WO2009076455A2 (en) 2007-12-10 2008-12-10 Methods of treating hematologic cancers using pnp inhibitors such as forodesine in combination with alkylating agents or anti-cd20 agents
US12/747,456 US20110038858A1 (en) 2007-12-10 2008-12-10 Methods of treating hematologic cancers using pnp inhibitors such as forodesine in combination with alkylating agents or anti-cd20 agents

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/086256 A-371-Of-International WO2009076455A2 (en) 2007-12-10 2008-12-10 Methods of treating hematologic cancers using pnp inhibitors such as forodesine in combination with alkylating agents or anti-cd20 agents

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/190,987 Continuation US20140178372A1 (en) 2007-12-10 2014-02-26 Methods of treating hematologic cancers

Publications (1)

Publication Number Publication Date
US20110038858A1 true US20110038858A1 (en) 2011-02-17

Family

ID=40431122

Family Applications (5)

Application Number Title Priority Date Filing Date
US12/747,456 Abandoned US20110038858A1 (en) 2007-12-10 2008-12-10 Methods of treating hematologic cancers using pnp inhibitors such as forodesine in combination with alkylating agents or anti-cd20 agents
US14/190,987 Abandoned US20140178372A1 (en) 2007-12-10 2014-02-26 Methods of treating hematologic cancers
US16/400,414 Active 2029-01-05 US11110092B2 (en) 2007-12-10 2019-05-01 Methods of treating hematologic cancers
US17/393,897 Abandoned US20220040188A1 (en) 2007-12-10 2021-08-04 Methods of treating hematologic cancers
US18/386,050 Pending US20240100055A1 (en) 2007-12-10 2023-11-01 Methods of treating hematologic cancers

Family Applications After (4)

Application Number Title Priority Date Filing Date
US14/190,987 Abandoned US20140178372A1 (en) 2007-12-10 2014-02-26 Methods of treating hematologic cancers
US16/400,414 Active 2029-01-05 US11110092B2 (en) 2007-12-10 2019-05-01 Methods of treating hematologic cancers
US17/393,897 Abandoned US20220040188A1 (en) 2007-12-10 2021-08-04 Methods of treating hematologic cancers
US18/386,050 Pending US20240100055A1 (en) 2007-12-10 2023-11-01 Methods of treating hematologic cancers

Country Status (26)

Country Link
US (5) US20110038858A1 (no)
EP (3) EP2237778A2 (no)
JP (2) JP5543362B2 (no)
KR (1) KR101545367B1 (no)
CN (2) CN103736091B (no)
AU (1) AU2008335167B2 (no)
CA (2) CA2881035C (no)
CY (1) CY1120203T1 (no)
DK (1) DK2564846T3 (no)
EA (2) EA018415B1 (no)
ES (1) ES2670714T3 (no)
GE (2) GEP20156288B (no)
HR (1) HRP20180712T1 (no)
HU (1) HUE037342T2 (no)
IL (1) IL206264A (no)
LT (1) LT2564846T (no)
MY (2) MY163023A (no)
NO (1) NO2564846T3 (no)
NZ (3) NZ601072A (no)
PH (1) PH12014501454A1 (no)
PL (1) PL2564846T3 (no)
PT (1) PT2564846T (no)
SG (1) SG188870A1 (no)
SI (1) SI2564846T1 (no)
UA (2) UA104579C2 (no)
WO (1) WO2009076455A2 (no)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014205194A1 (en) * 2013-06-22 2014-12-24 Nitor Therapeutics Compositions and methods for potentiating immune response for the treatment of infectious diseases and cancer
WO2016022166A1 (en) * 2014-08-07 2016-02-11 Nitor Therapeutics Use of pnp inhibitor to treat relapse of malignancy after hematopoietic stem cell transplant

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6090365A (en) * 1993-09-16 2000-07-18 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20 antibodies
US8206711B2 (en) * 1998-11-09 2012-06-26 Biogen Idec Inc. Treatment of chronic lymphocytic leukemia using anti-CD20 antibodies

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US199697A (en) 1878-01-29 Improvement in butter-sharers
ES2106732T3 (es) 1989-02-27 1997-11-16 Biocryst Pharm Inc Desazaguaninas substituidas en 9 y no substituidas en 8.
US4985433A (en) 1989-10-31 1991-01-15 Biocryst, Inc. 2-amino-7-(pyridinylmethyl)-3H,5H-pyrrolo[3,2-d]pyrimidin-4-ones and pharmaceutical uses and compositions containing the same
US5008270A (en) 1989-10-31 1991-04-16 Biocryst, Inc. 2-amino-7-(heterocyclomethyl)-3H,5H-pyrrolo[3,2-d]pyrimidin-4-ones and pharmaceutical uses and compositions containing the same
US4985434A (en) 1989-10-31 1991-01-15 Biocryst, Inc. 7-substituted derivatives of 2-amino-3H,5H-pyrrolo(3,2-d)pyrimidin-4-ones and pharamceutical uses and compositions containing the same
US5008265A (en) 1989-10-31 1991-04-16 Biocryst, Inc. 2-amino-7-(alicyclomethyl)-3H,5H,-pyrrolo[3,2-d]pyrimidin-4-ones and pharmaceutical uses and compositions containing the same
PL319485A1 (en) * 1994-10-05 1997-08-04 Chiroscience Ltd Purinic and guanic compounds as pnp inhibitors
US6387923B2 (en) * 2000-03-22 2002-05-14 Biocryst Pharmacueticals, Inc. Imminoribitol PNP inhibitors, preparation thereof and use thereof
DE10306724A1 (de) * 2002-02-28 2003-09-18 G O T Therapeutics Gmbh Vesikuläre Verkapselung von Bendamustin
CN103393681B (zh) * 2002-05-17 2017-04-12 细胞基因公司 用于治疗和控制多发性骨髓瘤的方法及组合物
US8003625B2 (en) * 2005-06-29 2011-08-23 Threshold Pharmaceuticals, Inc. Phosphoramidate alkylator prodrugs
EP2007423A2 (en) 2006-04-05 2008-12-31 Pfizer Products Incorporated Ctla4 antibody combination therapy

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6090365A (en) * 1993-09-16 2000-07-18 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20 antibodies
US8206711B2 (en) * 1998-11-09 2012-06-26 Biogen Idec Inc. Treatment of chronic lymphocytic leukemia using anti-CD20 antibodies

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Balakrishnan et al.,"Forodesine, an inhibitor of purine nucleoside phosphorylase, induces apoptosis in chronic lymphocytic leukemia cells", Blood , Vol 108, No 7 (October 1), 2006: pages 2392-2398. *
Merup et al., " 6q Deletions in Acute Lymphoblastic Leukemia and Non-Hodgkin's Lymphomas", Blood, Vol 91, No 9 (May 1), 1998: pp 3397-3400. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014205194A1 (en) * 2013-06-22 2014-12-24 Nitor Therapeutics Compositions and methods for potentiating immune response for the treatment of infectious diseases and cancer
US9452217B2 (en) 2013-06-22 2016-09-27 Nitor Therapeutics Methods for potentiating immune response for the treatment of infectious diseases and cancer
US9616129B2 (en) 2013-06-22 2017-04-11 Nitor Therapeutics Compositions and methods for potentiating immune response, enhancing immunotherapy, and increasing vaccine potency
WO2016022166A1 (en) * 2014-08-07 2016-02-11 Nitor Therapeutics Use of pnp inhibitor to treat relapse of malignancy after hematopoietic stem cell transplant
EP3177294A4 (en) * 2014-08-07 2017-12-27 Nitor Therapeutics Use of pnp inhibitor to treat relapse of malignancy after hematopoietic stem cell transplant

Also Published As

Publication number Publication date
PH12014501454B1 (en) 2016-01-18
GEP20135855B (en) 2013-06-25
EA201370055A1 (ru) 2013-11-29
IL206264A (en) 2017-04-30
JP2014094962A (ja) 2014-05-22
SI2564846T1 (en) 2018-06-29
PT2564846T (pt) 2018-05-23
KR20100101139A (ko) 2010-09-16
GEP20156288B (en) 2015-05-25
US11110092B2 (en) 2021-09-07
US20140178372A1 (en) 2014-06-26
CA2881035C (en) 2019-04-16
IL206264A0 (en) 2010-12-30
EP2564846B1 (en) 2018-02-28
CN103736091A (zh) 2014-04-23
PH12014501454A1 (en) 2016-01-18
NO2564846T3 (no) 2018-07-28
HUE037342T2 (hu) 2018-08-28
NZ601072A (en) 2014-01-31
CN101969947A (zh) 2011-02-09
HRP20180712T1 (hr) 2018-06-15
JP5543362B2 (ja) 2014-07-09
US20190255058A1 (en) 2019-08-22
UA109571C2 (uk) 2015-09-10
AU2008335167A1 (en) 2009-06-18
CY1120203T1 (el) 2018-12-12
DK2564846T3 (en) 2018-05-22
CN103736091B (zh) 2016-09-28
NZ586416A (en) 2012-07-27
EP2237778A2 (en) 2010-10-13
EA025340B1 (ru) 2016-12-30
ES2670714T3 (es) 2018-05-31
US20240100055A1 (en) 2024-03-28
MY163023A (en) 2017-07-31
WO2009076455A2 (en) 2009-06-18
WO2009076455A3 (en) 2009-08-27
UA104579C2 (uk) 2014-02-25
CA2708606A1 (en) 2009-06-18
SG188870A1 (en) 2013-04-30
EA201070669A1 (ru) 2010-12-30
CN101969947B (zh) 2014-10-22
LT2564846T (lt) 2018-05-10
US20220040188A1 (en) 2022-02-10
PL2564846T3 (pl) 2018-08-31
EP2564846A1 (en) 2013-03-06
EA018415B1 (ru) 2013-07-30
CA2881035A1 (en) 2009-06-18
AU2008335167B2 (en) 2014-02-13
NZ617989A (en) 2015-05-29
KR101545367B1 (ko) 2015-08-18
MY193789A (en) 2022-10-27
JP2011506345A (ja) 2011-03-03
CA2708606C (en) 2015-04-28
EP2581083A1 (en) 2013-04-17

Similar Documents

Publication Publication Date Title
US20240100055A1 (en) Methods of treating hematologic cancers
Saha et al. MDM2 antagonist nutlin plus proteasome inhibitor velcade combination displays a synergistic anti-myeloma activity
EP3835323A1 (en) Combination therapy for cancer
US11513122B2 (en) Predicting response to PD-1 axis inhibitors
RU2765997C2 (ru) Комбинация анти-pd-l1 антитела и ингибитора днк-пк для лечения злокачественного новообразования
US20170306050A1 (en) Compositions and methods for treating cancer, inflammatory diseases and autoimmune diseases
JP2023182572A (ja) がんの診断及び治療方法
US11124571B2 (en) Methods of sensitizing cancer to immunotherapy
AU2013201347B2 (en) Methods of treating hematologic cancers
US20230192863A1 (en) Methods of sensitizing cancer to immunotherapy

Legal Events

Date Code Title Description
AS Assignment

Owner name: BIOCRYST PHARMACEUTICALS, INC., ALABAMA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BANTIA, SHANTA;BREITFELD, PHILIP;BABU, YARLAGADDA S.;SIGNING DATES FROM 20100513 TO 20100604;REEL/FRAME:024485/0657

STCB Information on status: application discontinuation

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