EP1827445A2 - Verbesserte behandlung von krebs und hif-1-vermittelten störungen mit adenosin-a3-rezeptorantagonisten - Google Patents

Verbesserte behandlung von krebs und hif-1-vermittelten störungen mit adenosin-a3-rezeptorantagonisten

Info

Publication number
EP1827445A2
EP1827445A2 EP05858564A EP05858564A EP1827445A2 EP 1827445 A2 EP1827445 A2 EP 1827445A2 EP 05858564 A EP05858564 A EP 05858564A EP 05858564 A EP05858564 A EP 05858564A EP 1827445 A2 EP1827445 A2 EP 1827445A2
Authority
EP
European Patent Office
Prior art keywords
substituted
cancer
hif
adenosine
alkyl
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Withdrawn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
EP05858564A
Other languages
English (en)
French (fr)
Inventor
Pier Andrea Borea
Pier Giovani Baraldi
Stefania Merighi
Stephen Maclennan
Edward Leung
Allan Moorman
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.)
King Pharmaceuticals Research and Development Inc
Original Assignee
King Pharmaceuticals Research and Development 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 King Pharmaceuticals Research and Development Inc filed Critical King Pharmaceuticals Research and Development Inc
Publication of EP1827445A2 publication Critical patent/EP1827445A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic 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
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to the use of adenosine receptor antagonists, preferably A 3 receptor antagonists, either alone or in combination with other agents for the treatment, prevention and/or management of diseases or disorders associated with overexpression of HIF-I ⁇ and/or increased HIF-I a activity (e.g., cancer, respiratory disease).
  • adenosine receptor antagonists preferably A 3 receptor antagonists
  • the methods and compositions of the invention are particularly useful for preventing, treating, or ameliorating symptoms associated with a cancer, disease or disorder associated with hypoxia-inducible factor 1- ⁇ (HIF- l ⁇ ) using the A 3 receptor antagonists of the invention.
  • the present invention provides methods to inhibit the growth of tumors, particularly solid tumors, and more particularly hypoxic tumors.
  • Adenosine recently called a "primordial signalling molecule" (Linden, 2001, Annu. Rev. Pharmacol. Toxicol, Unital 41: 775-87), has the potential of influencing development, is present in and modulates physiological responses in all mammalian tissues.
  • the actions of adenosine are most prominent in tissues where oxygen demand is high and there is reduction in oxygen tension, i.e., within solid tumors, where cell proliferation is greater than the rate of blood vessel formation (Sitkovsky, 2004 Annu. Rev. Immunol. 22, 657-82; Fredholm, 2001, Pharmacol. Rev. 53, 527-552).
  • the tumor has local areas of hypoxia and adenosine accumulates to high levels (Hockel, 2001, J. Natl. Cancer Inst. 93, 266-76).
  • adenosine accumulates to high levels (Hockel, 2001, J. Natl. Cancer Inst. 93, 266-76).
  • significant levels of adenosine are present in the extracellular fluid of solid tumors (Blay, 1997, Cancer Res., 57, 2602-5), suggesting a role for this nucleoside in tumor growth.
  • Adenosine has been linked to tumor development. Increased adenosine concentration has been reported inside tumoral masses. It has been speculated that it represents the anti-tumor agent that prevents tumor growth in muscle tissue in vivo and that impairs malignant cell growth and survival in vitro. However, it is known that adenosine acts as cyto-protective agent during ischemic damage in brain and heart. Adenosine is known to be released in hypoxia. Numerous studies have shown adenosine to protect cells in the heart from ischemic damage. Adenosine has been shown to have protective roles in numerous animal models and in man (Am. J. Cardiol. 79(12A):44-48 (1997).
  • both the A 1 and A 3 receptors offer protection against ischemia (Am. J. Physiol., 273(42)H501- 505 (1997)). However, it is the A 3 receptor that offers sustained protection against ischemia (PNAS 95:6995-6999 (1998)).
  • the ability of adenosine to protect tumor cells against hypoxia has not been recognized by others prior to the instant invention.
  • Adenosine interacts with cell surface receptors that are glycoproteins coupled to different members of G protein family.
  • a 1 , A 2A , A 2B and A 3 adenosine receptors
  • Selective antagonists for the A 3 receptor have been proposed for use as anti-inflammatory and antiischemic agents in the brain.
  • a 3 antagonists have been under development as antiasthmatic, antidepressant, anti-arrhythmic, renal protective, antiparkinson and cognitive enhancing drugs.
  • U.S. Pat. No. 5,646,156 to Marlene Jacobson et al. describes the inhibition of eosinophil activation by using selected A 3 antagonists.
  • adenosine A 3 receptors to be responsible for long-term protection against ischemia (Liang and Jacobson, PNAS, 1998, 95:6995-6999). While the present inventors have hypothesized that adenosine plays a protective role in other cell types, including tumor cells, in addition to myocytes, no efforts have been made to limit the protective effect of adenosine on tumor cells.
  • Hypoxia-inducible factor (HIF)-I is a transcription factor that functions as a master regulator of oxygen homeostasis (Semenza, 2001, Trends MoI. Med. 7, 345-350.
  • HIF-I is aheterodimer composed of an inducibly-expressed HIF- l ⁇ subunit and a constitutively-expressed HIF-I ⁇ subunit (Epstein, 2001,Ce//, 107, 43-54). HIF-I ⁇ and HIF-I ⁇ mRNAs are constantly expressed under normoxic and hypoxic conditions (Wiener, 1996 Biochem. Biophys. Res. Commun. 225, 485-488). The unique feature of HIF-I is the regulation of HIF-I ⁇ expression: it increases as the cellular O 2 concentration is decreased (Cramer, 2003, Cell, 112, 645-657, Pugh, 2003, Nat. Med. 9, 677-84).
  • HIF-I ⁇ is rapidly degraded by the ubiquitin proteasome system, whereas exposure to hypoxic conditions prevents its degradation (Minchenko, 2002 J Biol. Chem., 277, 6183-6187; Semenza, 2000, J. Appl Physiol, 88, 1474-1480).
  • HIF-I contributes to tumor progression and metastasis
  • Immunohistochemical analyses have shown that HIF- l ⁇ is present in higher levels in human tumors than in normal tissues (Zhong, 2000, Cancer Res. 60, 1541-5). Tumor progression is associated with adaptation to hypoxia, and there is an inverse correlation between tumor oxygenation and clinical outcome (Pugh, 2003, Ann. Med. 35, 380-90.; Semenza, 2000 J. Appl Physiol, 88, 1474-1480).
  • HIF-I activity in cells are correlated with tumorigenicity and angiogenesis in nude mice (Chen, 2003, Am. J. Pathol. 162,1283-91).
  • Tumor cells lacking HIF-I expression are markedly impaired in their growth and vascularization (Carmeliet, 1998, Nature 394, 485-90; Jiang, 1997, Cancer Res., 57, 5328- 5335 ; Maxwell, 1997, Proc. Natl. Acad. Sci. U.S.A., 94, 8104-8109; Ryan, 1998 EMBOJ. 17, 3005-3015). Therefore, since HIF-l ⁇ expression and activity appear central to tumor growth and progression, HIF-I inhibition has become an appropriate anticancer target (Kung, 2000, Nat. Med. 6, 1335-40).
  • a neoplasm, or tumor is a neoplastic mass resulting from abnormal uncontrolled cell growth which can be benign or malignant. Benign tumors generally remain localized. Malignant tumors are collectively termed cancers.
  • malignant generally means that the tumor can invade and destroy neighboring body structures and spread to distant sites to cause death (for review, see Robbins and Angell, 1976, Basic Pathology, 2d Ed., W.B. Saunders Co., Philadelphia, pp. 68-122). Cancer can arise in many sites of the body and behave differently depending upon its origin. Cancerous cells destroy the part of the body in which they originate and then spread to other part(s) of the body where they start new growth and cause more destruction.
  • Lung and prostate cancer are the top cancer killers for men in the United States.
  • Lung and breast cancer are the top cancer killers' for women in the United States.
  • One in two men in the United States will be diagnosed with cancer at some time during his lifetime.
  • One in three women in the United States will be diagnosed with cancer at some time during her lifetime.
  • cancer therapy may involve surgery, chemotherapy, hormonal therapy and/or radiation treatment to eradicate neoplastic cells in a patient ⁇ See, for example, Stockdale, 1998, "Principles of Cancer Patient Management", in Scientific American: Medicine, vol. 3, Rubenstein and Federman, eds., Chapter 12, Section IV). Recently, cancer therapy could also involve biological therapy or immunotherapy. All of these approaches pose significant drawbacks for the patient. Surgery, for example, may be contraindicated due to the health of the patient or may be unacceptable to the patient. Additionally, surgery may not completely remove the neoplastic tissue. Radiation therapy is only effective when the neoplastic tissue exhibits a higher sensitivity to radiation than normal tissue, and radiation therapy can also often elicit serious side effects.
  • Hormonal therapy is rarely given as a single agent and although can be effective, is often used to prevent or delay recurrence of cancer after other treatments have removed the majority of the cancer cells.
  • Biological therapies/immunotherapies are limited in number and may produce side effects such as rashes or swellings, flu-like symptoms, including fever, chills and fatigue, digestive tract problems or allergic reactions.
  • chemotherapeutic agents available for treatment of cancer.
  • a significant majority of cancer chemotherapeutics act by inhibiting DNA synthesis, either directly, or indirectly by inhibiting the biosynthesis of the deoxyribonucleotide triphosphate precursors, to prevent DNA replication and concomitant cell division ⁇ See, for example, Gilmmx et al, Goodman and Gilman's: The
  • agents which include alkylating agents, such as nitrosourea, anti-metabolites, such as methotrexate and hydroxyurea, and other agents, such as etoposides, campathecins, bleomycin, doxorubicin, daunorubicin, etc., although not necessarily cell cycle specific, kill cells during S phase because of their effect on DNA replication.
  • alkylating agents such as nitrosourea
  • anti-metabolites such as methotrexate and hydroxyurea
  • agents such as etoposides, campathecins, bleomycin, doxorubicin, daunorubicin, etc.
  • Other agents specifically colchicine and the vinca alkaloids, such as vinblastine and vincristine, interfere with microtubule assembly resulting in mitotic arrest.
  • Chemotherapy protocols generally involve administration of a combination of chemotherapeutic agents to increase the efficacy of treatment.
  • chemotherapy has many drawbacks (See, for example, Stockdale, 1998, "Principles Of Cancer Patient Management” in Scientific American Medicine, vol. 3, Rubenstein and Federman, eds., ch. 12, sect. 10).
  • Almost all chemotherapeutic agents are toxic, and chemotherapy causes significant, and often dangerous, side effects, including severe nausea, bone marrow depression, immunosuppression, etc.
  • many tumor cells are resistant or develop resistance to the chemotherapeutic agents.
  • those cells resistant to the particular chemotherapeutic agents used in the treatment protocol often prove to be resistant to other drugs, even those agents that act by mechanisms different from the mechanisms of action of the drugs used in the specific treatment; this phenomenon is termed pleiotropic drug or multidrug resistance.
  • drug resistance many cancers prove refractory to standard chemotherapeutic treatment protocols.
  • paclitaxel or docetaxel both taxane family medicaments
  • docetaxel administered at 100 mg/m 2 causes acute hypersensitivity reaction in 13% of patients, and severe hypersensitivity reaction in 1.2%. Due to these reactions, patients are normally premedicated with oral corticosteroids.
  • vincristine has been reported to be dose limited due to neurotoxicity.
  • An enhancing agent providing a neuro-protective effect is therefore desirable.
  • Chemotherapeutic agents are also costly to produce and provide to patients. If the agents can be used at reduced dosages, both the cost and the extent of undesirable side effects can be similarly reduced.
  • Another often-encountered challenge with chemotherapeutic treatment is limitations in effectiveness due to the cancerous growth or cells developing multidrug-resistance (MDR). As most cancer cells are genetically unstable they are prone to mutations likely to produce drug resistant cells.
  • MDR multidrug-resistance
  • Multi-drug resistance is the name given to the circumstance when a disease does not respond to a treatment drug or drugs.
  • MDR can be either intrinsic, which means the disease has never been responsive to the drug or drugs, or it can be acquired, which means the disease ceases responding to a drug or drugs that the disease had previously been responsive to MDR is characterized by cross-resistance of a disease to more than one functionally and/or structurally unrelated drugs.
  • MDR in the field of cancer is discussed in greater detail in “Detoxification Mechanisms and Tumor Cell Resistance to Anticancer Drugs,” by Kuzmich and Tew, particularly section VII "The Multidrug-Resistant Phenotype (MDR),” Medical Research Reviews, Vol. 11, No. 2, 185-217, (Section VII is at pp. 208- 213) (1991); and in “Multidrug Resistance and Chemosensitization: Therapeutic
  • MDR multi-drug resistance
  • P-gp pleitotropic-glycoprotein
  • MDR-I gene Endicott JA, Annu Rev Biochem 1989.
  • P-glycoprotein has been shown to play a major role in the intrinsic and acquired resistance of a number of human tumors.
  • Drugs that act as substrates for and are consequently detoxified by P-gp include the vinca alkaloids (vincristine and vinblastine), anthracyclines (Adriamycin), and epipodophyllotoxins (etoposide).
  • MDR-I gene has been identified as an additional risk factor in advanced ovarian cancer.
  • patients with phase III ovarian cancer were screened for MDR-I .
  • Ovarian cancer patients with high levels of MDR-I survived an average of 9.8 months.
  • the patients having low or no MDR-I expression survived an average of 30 months or more.
  • MRP multidrug resistance-associated protein
  • MDR has been reported that is neither P-gp nor MRP related. (Kellen editor, Alternative Mechanisms of Multidrug Resistance in Cancer, Birkhauser, 1995). The results obtained by inventors are consistent for using adenosine A 3 antagonists for countering P-gp or MRP multi-drug resistance, but not other forms of MDR.
  • ⁇ -[3-[[2-(3,4-Dimethoxyphenyl)ethyl]methylamino]propyl]-3,4-dimethoxy- ⁇ -(l-methylethyl)benzeneacetonitrile (Verapamil) has been utilized as a medicament to counter the effect of P-gp associated MDR.
  • Verapamil blocks L-type calcium channels and is used as a potent vasodilator of coronary and peripheral vessels and decreases myocardial oxygen consumption. Due to the Verapamil physiological effects, MDR use of Verapamil must be limited to patients not having low blood pressure, congestive heart failure, sinoatrial (SA) or atrioventricular (AV) node conduction disturbances, digitalis toxicity, Wolff-Parkinson- White syndrome, and further not being medicated with beta-blockers or Quinidine.
  • SA sinoatrial
  • AV atrioventricular
  • U.S. Patent No. 6,210,917 to Carson et al. discloses the use of L-alanosine and other adenosine kinase inhibitors for countering MDR.
  • U.S. Patent No. 6,391,884 identifies ATP-depleting agents including 2-deoxyglucose, cyanine, oligomycin, valinomycin and azide, as well as salts and derivatives thereof. Approaches relying upon ATP depletion or inhibition in countering MDR have yet to receive clinical success.
  • chemotherapeutic agent enhancers and compounds that prevent or counter MDR either alone or with ATP depleting agents in cancer treatment but without the limitations imposed by Verapamil.
  • the use of a combination of therapeutic agents is common in the treatment of neoplastic diseases.
  • paclitaxel TaxolTM
  • U.S. Patent No. 5,908,835 to Bissery et al. claims synergy of using paclitaxel or docetaxel in combination with an anthracycline antibiotic such as daunorubicin or doxorubicin.
  • U.S. Patent No. 5,728,687 to Bissery et al. claims synergy of using paclitaxel or docetaxel in combination with an alkylating agent, epidophyllotoxin, antimetabolite, or vinca alkaloid.
  • adenosine receptor antagonists and in particular A 3 receptor antagonists. It is therefore an object of the present invention to provide compositions and methods of enhancing the treatment of neoplastic cells by minimizing or eliminating the protective effect of adenosine on cells with the use of adenosine A 3 receptor antagonists.
  • compositions and methods suitable for countering P-gp and/or MRP associated multi-drug resistance It is further an object of the present invention to provide compositions and methods that reduce side effects of other currently used anti-cancer agents and/or therapies, in particular taxane induced hypersensitivity.
  • the present invention is based, in part, on the surprising discovery by the inventors that adenosine, a purine nucleoside present within hypoxic regions of solid tumors, modulates hypoxia-inducible factor 1 (HIF-I) expression.
  • HIF-I hypoxia-inducible factor 1
  • adenosine a purine nucleoside present within hypoxic regions of solid tumors, modulates hypoxia-inducible factor 1 (HIF-I) expression.
  • HIF-I a heterodimeric transcription factor composed of HIF-I a and HIF- l ⁇ subunits
  • angiogenesis for reviews see, e.g., Semenza 2000, Nature, 3: 721-32
  • the inventors have found that in the human A375 melanoma cell line adenosine up-regulates HIF- l ⁇ protein expression in response to hypoxia in a dose- and time-dependent manner.
  • the response to adenosine was not blocked by A 1 , A 2A or A 2B receptor antagonists, while it was abolished by A 3 receptor antagonists.
  • the inventors have found that Cl-IB-MECA, an adenosine analogue binding with high affinity to A 3 receptors, mimicked adenosine effect in hypoxic cells. Furthermore, A 3 receptor antagonists prevented HIF- l ⁇ protein accumulation in response to A 3 receptor stimulation.
  • the response to adenosine is generated at the cell surface since the inhibition of A 3 receptor expression, by using small interfering RNA, abolished the nucleoside effects.
  • the main intracellular signaling pathways sustained by A 3 receptor stimulation in hypoxia involve p44/p42 mitogen-activated protein kinase (MAPK) and p38 MAPK activation.
  • MAPK mitogen-activated protein kinase
  • the inventors have thus found for the first time that adenosine plays a critical role in HIF- l ⁇ regulation in hypoxia.
  • the inventors have found that compounds which are antagonists of the adenosine receptors, preferably the A 3 receptor, inhibit the protective effect of adenosine on growing tumor cells when such cells are starved of oxygen (/. e. , before an adequate vasculature is developed, or when anti-angiogenesis agents are administered.
  • the antagonists of the invention can bind in the same site as adenosine, or can be allosteric antagonists (i.e., bind at a site different from where adenosine binds, but adversely affect the ability of adenosine to bind to the site or adversely affect the ability of adenosine, once bound to the adenosine receptors, particularly A 3 receptors, to protect growing tumor cells).
  • the present invention relates to methods for the treatment, prevention, and/or management of diseases or disorders associated with overexpression of HIF- l ⁇ and/or increased HIF- l ⁇ activity (e.g., cancer, respiratory disorders such as asthma and obstructive pulmonary disorders) by using adenosine receptor antagonists, particularly A 3 receptor antagonists alone or in combination with A 1 , A 2 A, A 2 B receptor antagonists.
  • adenosine receptor antagonists particularly A 3 receptor antagonists alone or in combination with A 1 , A 2 A, A 2 B receptor antagonists.
  • the A 1 , A 2A and A 2 B receptor antagonists may act through different pathways than the A 3 receptor antagonist, the combination of these antagonists with A 3 receptor antagonist may be beneficial in the treatment, prevention, and/or management of diseases or disorders, e.g., cancer.
  • the methods of the invention relate to treatment, prevention, and/or management of diseases or disorders associated with overexpression of HIF-I ⁇ and/or increased HIF-I ⁇ activity by using adenosine receptor antagonists, particularly A 3 receptor antagonists alone.
  • adenosine receptor antagonists particularly A 3 receptor antagonists alone.
  • administration of antagonists for the adenosine receptors, particularly A 3 receptor antagonists antagonizes the protective effects against hypoxia and renders those cells susceptible to destruction due to hypoxia. Since the adenosine receptors, in particular, the A 3 receptor, are responsible for sustained cellular protection against ischemia, antagonists for the adenosine receptors, particularly A 3 receptors are particularly effective in enhancing the activity of anti-tumor agents.
  • the methods and compositions of the invention comprising A 3 receptor antagonists are particularly useful when the levels of HIF-I ⁇ expression and/or activity are elevated above the standard or background level, as determined using methods known to those skilled in the art and disclosed herein.
  • "elevation" of a measured level of HIF-I ⁇ relative to a standard level means that the amount or concentration of HIF- l ⁇ in a sample or subject is greater in a subject or sample relative to the standard as detected by any method now known in the art or to be developed in the future for measuring HIF-l ⁇ levels.
  • elevation of the measured level relative to a standard level may be any statistically significant detectable elevation.
  • Such an elevation in HIF- 1 ⁇ expression and/or activity may include, but is not limited to about a 10%, about a 20%, about a 40%, about an 80%, about a 2-fold, about a 4-fold, about an 10-fold, about a 20- fold, about a 50-fold, about a 100-fold, about a 2 to 20 fold, 2 to 50 fold, 2 to 100 fold, 20 to 50 fold, 20 to 100 fold, elevation, relative to the standard.
  • the therapeutic methods of the invention comprising administering a therapeutically effective amount of an A 3 receptor antagonist of the invention improve the therapeutic efficacy of treatment for diseases or disorders associated with overexpression of HIF-l ⁇ and/or increased HIF-l ⁇ activity (e.g., cancer, respiratory disorders such as asthma and obstructive pulmonary disorders) relative to the traditional modes of such therapies.
  • the methods of the invention reduce the HIF-I ⁇ level to the background level within one day, one week, 1 month, or 2 months, of the commencement of the therapeutic regime.
  • the methods of the invention result in a reduction of HIF-l ⁇ level to the background level.
  • the invention encompasses reduction of the HIF- l ⁇ level to a level which is within about 10%, about 20%, about 30%, about 40%, about 50% of the background level; about a 2-fold, about a 4-fold, about an 10-fold, about a 20- fold, about a 50-fold, about a 100-fold, about a 2 to 20 fold, 2 to 50 fold, 2 to 100 fold, 20 to 50 fold, 20 to 100 fold of the background level.
  • the methods of the invention reduce the level of HIF- 1 a to a particular level, that level is maintained during the treatment regimen, such that the maintained level of HIF-l ⁇ is sufficient and effective to result in regression of the disease, e.g., cancer.
  • the invention encompasses a method for treatment, prevention and/or management of diseases or disorders associated with overexpression of HIF-l ⁇ and/or increased HIF-l ⁇ activity (e.g., cancer, respiratory disorders such as asthma and obstructive pulmonary disorders) comprising administering a therapeutically and/or prophylactically effective amount of an A 3 receptor antagonist compound as disclosed herein alone or in combinationo with other therapeutic or prophylatic agents.
  • the invention also encompasses a method for determining the prognosis of a a disease or disorder associated with overexpression of HIF-I ⁇ and/or increased HIF-I ⁇ activity in a subject.
  • the subject is human.
  • the subject has been previously treated with a therapy regimen.
  • the invention encompasses measuring a level of HIF-I ⁇ in a subject to determine if the subject is in need of the therapeutic and or prophylactic methods of the inventon.
  • the invention encompasses measuring a level of HIF-I ⁇ in a sample obtained from the subject and comparing the level measured to a standard level, wherein elevation of the measured level of at least one HIF- 1 a relative to the standard level indicates that the subject is at an increased risk for progression of the disease or disorder, e.g., metastasis of the cancer.
  • the HIF-l ⁇ level may be measured using at least one of the methods for measuring and detecting HIF-I ⁇ such as those known to those skilled in the art and exemplified herein.
  • the invention encompasses compounds which are A 3 or A 1 receptor antagonists for use in the methods of the invention. Examples of such compounds are disclosed in U.S. Patent Nos 6,326,390; 6,407,236; 6,448,253; 6,358,964; and U.S.
  • the A 3 adenosine receptor agonist 2-Cl-IB-MECA facilitates epileptiform discharges in the CA3 area of immature rat hippocampal slices. Laudadio, Mark Anthony; Psarropoulou, Caterina. Department of Paediatrics, Ste- Justine Hospital Research Centre, Universite de Montreal, Montreal, QC, Can. Epilepsy Research (2004), 59(2-3), 83-94.
  • WO 2004051264 Al l,2,4-Triazolo[4,3-a]quinoxalin-l-one Moiety as an Attractive Scaffold To Develop New Potent and Selective Human A3 Adenosine Receptor Antagonists Synthesis, Pharmacological, and Ligand-Receptor Modeling Studies. Colotta, Vittoria; Catarzi, Daniela; Varano, Flavia; Calabri, Francesca Romana; Lenzi, Ombretta; Filacchioni, Guido; Martini, Stephan; Trincavelli, Letizia; Deflorian, Francesca; Moro, Stefano. Dipartimento di Scienze Farmaceutiche, Universita degli Studi di Firenze, Sesto Fiorentino, Italy. Journal of Medicinal Chemistry (2004), 47(14), 3580-3590.
  • Structural determinants of efficacy at A 3 adenosine receptors modification of the ribose moiety. Gao, Zhan-Guo; Jeong, Lak Shin; Moon, Hyung Ryong; Kim, Hea Ok; Choi, Won Jun; Shin, Dae Hong; Elhalem, Eleonora; Comin, Maria J.; Melman, NeIi; Mamedova, Liaman; Gross, Ariel S.; Rodriguez, Juan B.; Jacobson, Kenneth A. National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institutes of Health, Bethesda, MD, USA. Biochemical Pharmacology (2004), 67(5).
  • Pyrazolo[4,3-e]-l,2,4-triazolo[l,5-c]pyrimidine Derivatives as Adenosine Receptor Antagonists. Influence of the N5 Substituent on the Affinity at the Human A 3 and A 2B Adenosine Receptor Subtypes: A Molecular Modeling Investigation. Pastorin, Giorgia; Da Ros, Tatiana; Spalluto, Giampiero; Deflorian, Francesca; Moro, Stefano; Cacciari, Barbara; Baraldi, Pier Giovanni; Gessi, Stefania; Varani, Katia; Borea, Pier Andrea. Dipartimento di Scienze Farmaceutiche, Universita degli Studi di Trieste, Trieste, Italy. Journal of Medicinal Chemistry (2003), 46(20), 4287-4296.
  • QSAR of adenosine receptor antagonists II Exploring physicochemical requirements for selective binding of 2-arylpyrazolo[3,4-c]quinoline derivatives with adenosine A 1 and A 3 receptor subtypes. Roy, Kunal. Drug Theoretics and Cheminformatics Lab, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Calcutta, India. QSAR & Combinatorial Science (2003), 22(6), 614-621. Functional expression of adenosine A 2A and A 3 receptors in the mouse dendritic cell line XS-106. Dickenson, John M.; Reeder, Steve; Rees, Bob; Alexander, Steve; Kendall, Dave. School of Science, Faculty of Science and Mathematics, Department of Life Sciences, Nottingham Trent University, Nottingham, UK. European Journal of Pharmacology (2003), 474(1), 43-51.
  • a 3 adenosine and CBl receptors activate a PKC-sensitive Cl- current in human nonpigmented ciliary epithelial cells via a G-coupled MAPK signaling pathway. Shi, Chanjuan; Szczesniak, Anna; Mao, Lucy; Jollimore, Christine; Coca-Prados, Miguel; Hung, Orlando; Kelly, Melanie E. M. Laboratory for Retina and Optic Nerve Research, Dalhousie University, Why, NS, 475-486.
  • QSAR of adenosine receptor antagonists I: Exploration of receptor interaction sites of l,2-dihydro-2-phenyl-l,2,4-triazolo[4,3-a]quinoxalin-l-one derivatives using AMI calculations. Roy, Kunal. Drug Theoretics and Cheminformatics Lab, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, India. Indian Journal of Chemistry, Section B: Organic Chemistry Including Medicinal Chemistry (2003), 42B(6), 1485-1496.
  • 9-Ethyladenine derivatives as adenosine receptor antagonists 2- and 8-substitution results in distinct selectivities.
  • Klotz Karl-Norbert; Kachler, Sonja; Lambertucci, Catia; Vittori, Sauro; Volpini, Rosaria; Cristalli, Gloria.
  • Adenosine Receptor Subtypes Mediating Coronary Vasodilation in Rat Hearts. Hinschen, Andrea K.; Rose'Meyer, Roselyn B.; Headrick, John P. Griffith University, Heart Foundation Research Center, School of Health Science, Gold Coast Campus, Southport, Australia. Journal of Cardiovascular Pharmacology (2003), 41(1), 73-80.
  • Imidazo[2,l-i]purin-5-ones and Related Tricyclic Water-Soluble Purine Derivatives Potent A 2A - and A 3 -Adenosine Receptor Antagonists. Mueller, Christa E.; Thorand, Mark; Qurishi, Ramatullah; Diekmann, Martina; Jacobson, Kenneth A.; Padgett, William L.; Daly, John W. Pharmaceutical Institute Poppelsdorf, University of Bonn, Bonn, Germany. Journal of Medicinal Chemistry (2002), 45(16), 3440-3450.
  • Adenosine acts through an A 3 receptor to prevent the induction of murine anti-CD3- activated killer T cells.
  • thermodynamics at the human A 3 adenosine receptor Merighi, Stefania; Varani, Katia; Gessi, Stefania; Klotz, Karl-Norbert; Leung, Edward; Baraldi, Pier Giovanni; Borea, Pier Andrea. Pharmacology Unit, Department of Clinical and Experimental Medicine, Centro Nazionale di Eccellenza per Io Sviluppo di Metodologie Innovative per Io Studio ed il Trattamento delle Patologie Infiammatorie, University of Ferrara, Ferrara, Italy. Biochemical Pharmacology (2002), 63(2), 157-161.
  • [3H]-8-Ethyl-4-methyl-2-phenyl-(8R)-4,5,7 5 8-tetrahydro-lH-imidazo[2,l-i]purin-5-one [3H]PSB-11), a Novel High- Affinity Antagonist Radioligand for Human A3 Adenosine Receptors. Muller, ChristaE.; Diekmann, Martina; Thorand, Mark; Ozola, Vita. University of Bonn, Pharmaceutical Institute, Bonn, Germany. Bioorganic & Medicinal Chemistry Letters (2002), 12(3), 501-503.
  • Fluorosulfonyl- and bis-(ct ⁇ oroethyl)arnino-phenylamino functionalized pyrazolo[4,3- e]l,2,4-triazolo[l,5-c]pyrimidine derivatives irreversible antagonists at the human A 3 adenosine receptor and molecular modeling studies. Baraldi, Pier Giovanni; Cacciari, Barbara; Moro, Stefano; Romagnoli, Romeo; Ji, Xiao-duo; Jacobson, Kenneth A.; Gessi, Stefania; Borea, Pier Andrea; Spalluto, Giampiero. Dipartimento di Scienze Farmaceutiche, Universita degli Studi di Ferrara, Ferrara, Italy. Journal of Medicinal Chemistry (2001), 44(17), 2735-2742.
  • Receptor subtypes mediating adenosine-induced dilation of cerebral arterioles.
  • Pyrazolo[4,3-e]l,2,4-triazolo[l,5-c]pyrimidine derivatives a new pharmacological tool for the characterization of the human A 3 adenosine receptor.
  • Baraldi Pier Giovanni; Cacciari, Barbara; Romagnoli, Romeo; Spalluto, Giampiero; Varani, Katia; Gessi, Stefania; Merighi, Stefania; Borea, Pier Andrea. Dipartimento di Scienze Farmaceutiche, Universita degli Studi di Ferrara, Ferrara, Italy. Drug Development Research (2001), 52(1/2), 406-415.
  • Adenosine-enhanced ischemic preconditioning adenosine receptor involvement during ischemia and reperfusion. McCully, James D.; Toyoda, Yoshiya; Uematsu, Masahisa; Stewart, Robert D.; Levitsky, Sidney. Division of Cardiothoracic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA. American Journal of Physiology (2001), 280(2, Pt. 2), H591-H602.
  • Thiazole and thiadiazole analogs as a novel class of adenosine receptor antagonists, van Muijlwijk-Koezen, Jacqueline E.; Timmerman, Hendrik; Vollinga, Roeland C; von Kuenzel, Jacobien Frijtag; de Groote, Miriam; Visser, Sven; IJzerman, Adriaan P. Department of Pharmacochemistry Division of Medicinal Chemistry Leiden/Amsterdam Center for Drug Research, Vrije Universiteit, Amsterdam, Neth. Journal of Medicinal Chemistry (2001), 44(5), 749-762.
  • [3H]-MRE 3008-F20 the first high affinity radioligand antagonist for the human A 3 adenosine receptors. Baraldi, Pier Giovanni; Cacciari, Barbara; Romagnoli, Romeo; Varani, Katia; Merighi, Stefania; Gessi, Stefania; Borea, Pier Andrea; Leung, Edward; Hickey, Sarah L.; Spalluto, Giampiero. Dipartimento di Scienze Farmaceutiche, Universita di Ferrara, Ferrara, Italy. Bioorganic & Medicinal Chemistry Letters (2000), 10(12), 1403.
  • Patent evaluation a novel adenosine receptor pharmacophore. Novartis AG, Expert Opinion on Therapeutic Patents (2000), 10(5), 729-732.
  • [3H]MRE 3008F20 a novel antagonist radioligand for the pharmacological and biochemical characterization of human A 3 adenosine receptors. Varani, Katia; Merighi, Stefania; Gessi, Stefania; Klotz, Karl-Norbert; Lfung, Edward; Baraldi, Pier Giovanni; Cacciari, Barbara; Romagnoli, Romeo; Spalluto, Giampiero; Borfa, Pier Andrea. Department of Clinical and Experimental Medicine, Pharmacology Unit, University of Ferrara, Italy. Molecular Pharmacology (2000), 57(5), 968-975.
  • A3 adenosine receptor ligands history and perspectives. Baraldi, Pier Giovanni; Cacciari, Barbara; Romagnoli, Romeo; Merighi, Stefania; Varani, Katia; Borea, Pier Andrea; Spalluto, Giampiero. Dipartimento di Scienze Farmaceutiche, Universita di Ferrara, Ferrara, Italy. Medicinal Research Reviews (2000), 20(2), 103-128.
  • [3H]-MRE 3008-F20 the first high affinity radioligand antagonist for the human A 3 adenosine receptors. Baraldi, Pier Giovanni; Cacciari, Barbara; Romagnoli, Romeo; Varani, Katia; Merighi, Stefania; Gessi, Stefania; Borea, Pier Andrea; Leung, Edward; Hickey, Sarah L.; Spalluto, Giampiero. Dipartimento di Scienze Farmaceutiche, Universita di Ferrara, Ferrara, Italy. Bioorganic & Medicinal Chemistry Letters (2000), 10(3), 209-211.
  • Selective A 3 Adenosine Receptor Antagonists Water-Soluble 3,5-Diacyl- 1,2,4- trialkylpyridinium Salts and Their Oxidative Generation from Dihydropyridine Precursors.
  • Xie Rongyuan; Li, An-Hu; Ji, Xiao-Duo; Melman, NeIi; Olah, Mark E.; Stiles, Gary L.; Jacobson, Kenneth A. Molecular Recognition Section Laboratory of Bioorganic Chemistry National Institute of Diabetes Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA. Journal of Medicinal Chemistry (1999), 42(20), 4232-4238.
  • a 3 adenosine receptors protective vs. damaging effects identified using novel agonists and antagonists. Jacobson, Kenneth A.; Moro, Stefano; Kim, Yong-Chul; Li, An-Hu. Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA. Drug Development Research (1998), 45(3/4), 113-124.
  • Triazoloquinazoline Adenosine Antagonist (CGS 15943) Having High Potency at the Human A 2 B and A 3 Receptor Subtypes.
  • Adenosine A 3 receptors novel ligands and paradoxical effects. Jacobson, Kenneth A. Molecular Recognition Section, Lab. Bioorganic Chem., National Inst. Diabetes, Digestive and Kidney Diseases, national Inst. Health, Bethesda, MD, USA. Trends in Pharmacological Sciences (1998), 19(5), 184-191.
  • Flavonoid Derivatives as Adenosine Receptor Antagonists A Comparison of the Hypothetical Receptor Binding Site Based on a Comparative Molecular Field Analysis Model. Moro, Stefano; van Rhee, A. Michiel; Sanders, Lawrence H.; Jacobson, Kenneth A. Molecular Recognition Section Laboratory of Bioorganic Chemistry National Institute of Diabetes Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA. Journal of Medicinal Chemistry (1998), 41(1), 46-52.
  • Adenosine A 3 receptor agonists inhibit murine macrophage tumor necrosis factor-d production in vitro and in vivo. Bowlin, Terry L.; Borcherding, David R.; Edwards, Carl K., Ill; McWhinney, Charlene D. Department of Immunology, Marion Merrell Dow Research Institute, Marion Merrell Dow Pharmaceuticals, Cincinnati, OH, USA. Cellular and Molecular Biology (Paris) (1997), 43(3), 345-349.
  • Adenosine receptor antagonists structures and potential therapeutic applications. Mueller, C. E.; Stein, B. Inst. Pharmazie Strukturchemie, Univ. Wuerzburg, Wuerzburg, Germany. Current Pharmaceutical Design (1996), 2(5), 501-530.
  • 6-Phenyl-l,4-Dihydropyridine Derivatives as Potent and Selective A 3 Adenosine Receptor Antagonists.
  • Triazoloquinazoline Adenosine Antagonist (CGS 15943) are Selective for the Human A 3 Receptor Subtype.
  • Activation of adenosine A 3 receptors on macrophages inhibits tumor necrosis factor-d McWhinney, Charlene D.; Dudley, Mark W.; Bowlin, Terry L.; Peet, Norton P.; Schook, Larry; Bradshaw, Marita; De, Mamata; Borcherding, David R.; Edwards, Carl K., III. Department of Immunology, Marion Merrell Dow Research Institute, Marion Merrell Dow Pharmaceuticals, Inc., 2110 East Galbraith Road, Cincinnati, USA. European Journal of Pharmacology (1996), 310(2/3), 209-216.
  • Novel adenosine antagonists A 3 receptor-selective flavonoid and dihydropyridine derivatives. Jacobson, Kenneth A.; Van Rhee, A. Michiel; Jiang, Ji-Long; Ji, Xiao-duo; Karton, Yishai; Von Lubitz, Dag K. J. E.; Olah, Mark E.; Stiles, Gary L. Laboratory Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA. Book of Abstracts, 212th ACS National Meeting, Orlando, FL, August 25-29 (1996), MEDI-099. Publisher: American Chemical Society, Washington, D.C.
  • the A 3 receptor antagonists of the invention are particularly useful for prevention, treatment, and/or management of cancer, for example, as a single agent therapy or in combination with other modes of therapy for cancer.
  • the invention encompasses methods of treatment, prevention, or management of cancers which are A 3 receptor expressing cancers including, but not limited to, human leukemia, melanoma, pancreatic carcinoma, ovarian carcinoma, breast carcinoma, prostrate carcinoma, colon carcinoma, lung carcinoma, malignant melanomas, histiocytic lymphoma, and some forms of astrocytoma cells.
  • the invention encompasses methods of treatment, prevention, or management which have high level of expression of HIF-I a including, but not limited to, cervical cancer (early stage), lung cancer (non-small cell lung carcinoma), breast cancer (including lymph node positive breast cancer and lymph node negative breast cancer), oligodendroglioma, orpharyngeal squamous cell carcinoma, ovarian cancer, oesophageal cancer, endometrial cancer, head and neck cancer, gastrointestinal stromal tumor of the stomach
  • the methods of the invention are particularly useful in cancer therapy for providing tissue selectivity, such that the biological effect is observed in tumoral hypoxic cells, where high adenosine concentrations has resulted in increased HIF-I ⁇ accumulation.
  • solid tumors refer to a locus of tumor cells where the majority of the cells are tumor cells or tumor-associated cells, including but not limited to laryngeal tumors, brain tumors, and other tumors of the head and neck; colon, rectal and prostate tumors; breast and thoracic solid tumors; ovarian and uterine tumors; tumors of the esophagus, stomach, pancreas and liver; bladder and gall bladder tumors; skin tumors such as melanomas.
  • the tumors encompassed within the invention can be either primary or a secondary tumor resulting from metastasis of cancer cells elsewhere in the body to the chest.
  • the invention encompasses use of the A 3 or A 1 receptor antagonist compounds of the invention in inhibiting tumor growth in a mammal, including humans.
  • the invention encompasses methods for administering an effective amount of adenosine A 1 or A 3 antagonists, preferably, A 3 antagonists sufficient to inhibit the ability of adenosine to protect tumor cells.
  • the present invention encompasses methods for treatment, prevention and/or management of cancer comprising administering a compound of the invention including A 3 and/or A 1 receptor antagonists used in combination with other therapeutic and/or prophylactic agents, e.g., cytotoxic agents.
  • adenosine A 3 receptor antagonists synergistically enhance cytotoxic treatment and counter some forms of multi-drug resistance.
  • the combination therapies of the invention will attack the tumor cell directly, inhibit growth of new blood vessels around the tumor cell, and, by virtue of the adenosine A 3 antagonists, inhibit the ability of the cell to survive without the growth of new blood vessels.
  • high affinity adenosine A 3 receptor antagonists are useful as enhancers for many chemotherapeutic treatments of adenosine A 3 receptor expressing cancers.
  • high affinity adenosine A 3 receptor antagonists also counter P-glycoprotein (P-gp) effuse pump multidrug resistance (MDR).
  • P-gp P-glycoprotein
  • MDR multidrug resistance
  • high affinity adenosine A 3 receptor antagonists are helpful in reducing or ameliorating side effects of cytoxic agents especially taxane induced hypersensitivity. For example, lower doses of taxane may be used once used in combination with an adenosine A 3 receptor antagonist of the invention.
  • the present invention encompasses the use of high affinity adenosine A 3 receptor antagonists for enhancing chemotherapeutic treatment of cancers expressing adenosine A 3 receptors and countering multi-drug resistance in cancers expressing P-glycoprotein or MRP.
  • adenosine A 3 receptor antagonists are administered before or during administration of a taxane family, vinca alkaloid, camptothecin or antibiotic compound.
  • Preferred high affinity A 3 receptor antagonists include compounds of the general formulas ILA, HB, HC and HD described herein, vide infra.
  • the present invention encompasses therapies which involve administering one or more of the compounds of the invention, to an animal, preferably a mammal, and most preferably a human, for preventing, treating, or ameliorating symptoms associated with a cancer, a disease or disorder associated with hypoxia-inducible factor 1- ⁇ (HIF- l ⁇ ).
  • the invention further provides a pharmaceutical composition comprising a therapeutically or prophylactically effective amount of a compound of the invention that specifically binds an A 1 or A 3 receptor and a pharmaceutically acceptable carrier
  • the pharmaceutical formulation includes an adenosine A 3 receptor antagonist and one or more excipients.
  • the formulations can also include other anti-tumor agents, including cytotoxic agents and other anti-angiogenesis agents, including adenosine A 2A antagonists, such as ZM241385 and SCH 5861, adenosine A 2B antagonists, such as MRE- 20290-F20 and AS- 16, and anti-VEGF antibodies, including humanized and chimeric antibodies.
  • the composition includes an effective amount to inhibit tumor growth of an adenosine A 3 receptor antagonist, a cytotoxic agent, and an anti- angiogenesis agent.
  • a compound is an "antagonist of an adenosine A 3 receptor" if it is able to prevent the action due to an agonist and is able to displace [ I2 I]-AB-MECA in a competitive binding assay.
  • a compound is selective for the A 3 receptor if its affinity at the A 3 receptor is greater than its affinity at the A 1 , A 2 A and A 2 B receptors.
  • the ratio OfA 1 /A 3 and A 2 /A 3 activity is greater than about 50, preferably between 50 and 100, and more preferably, greater than about 100. Since the pharmacology at the A 3 receptor varies between species, especially between rodent A 3 and human A 3 receptors, it is important to determine the selectivity of the A 3 compounds in human adenosine receptors. The same holds true for adenosine A 1 and A 2 A receptors in terms of whether they are selective.
  • Adenosine A 1 Antagonists carry their common ordinary meaning. Adenosine A 1 antagonists are well known to those of skill in the art. Some of the more well known compounds include CPX, CVT 124, FK 352, and ILF 117, the structures of which are well known.
  • Adenosine A 3 Antagonists carry their common ordinary meaning.
  • Adenosine A 3 antagonists are well known to those of skill in the art. Some of the more well known compounds include MRS 1097, MRS 1191 and MRS 1220 (commercially available from Research Biochemicals International, Natick Mass.). Other suitable antagonists include those disclosed in Jacobson et al., Neuropharmacology, 36: 1157-1165 (1997); Yao et al, Biochem. Biophys. Res. Commun., 232:317-322 (1997); Kim etal., J. Med. Chem., 39(21):4142-4148 (1996); van Rhee et al, DrugDevel.
  • elevation of a measured level of HIF-I ⁇ or A 3 receptor relative to a standard level means that the amount or concentration of HIF-I ⁇ or A 3 receptor in a sample or subject is sufficiently greater in a subject or sample relative to the standard as detected by any method now known in the art or to be developed in the future for measuring HIF- lot or A 3 receptor levels.
  • elevation of the measured level relative to a standard level may be any statistically significant elevation detectable.
  • Such an elevation in HIF-I ⁇ expression and/or activity may include, but is not limited to about a 10%, about a 20%, about a 40%, about an 80%, about a 2-fold, about a 4-fold, about an 10- fold, about a 20-fold, about a 50-fold, about a 100-fold, about a 2 to 20 fold, 2 to 50 fold, 2 to 100 fold, 20 to 50 fold, 20 to 100 fold, elevation, relative to the standard.
  • standard level refers to a baseline amount of an HIF- l ⁇ level as determined in one or more normal subjects, i.e., a subject with no known history of past or current diseases, disorders or cancer.
  • the measurement of HIF-l ⁇ level may be carried out using an HIF-l ⁇ probe or a HIF- l ⁇ activity assay (see Section 6).
  • enhancement refers to a synergistic effect as determined from measurement of the enhancement factor, as defined below.
  • an enhancement factor of greater than one is considered synergistic. For example, if one of the compounds has little individual chemotherapeutic effect, an enhancement factor greater than one indicates a synergistic effect is occurring.
  • cancer refers to a neoplasm or tumor resulting from abnormal uncontrolled growth of cells. As used herein, cancer explicitly includes, leukemias and lymphomas.
  • cancer refers to a disease involving cells that have the potential to metastasize to distal sites and exhibit phenotypic traits that differ from those of non-cancer cells, for example, formation of colonies in a three-dimensional substrate such as soft agar or the formation of tubular networks or weblike matrices in a three- dimensional basement membrane or extracellular matrix preparation.
  • Non-cancer cells do not form colonies in soft agar and form distinct sphere-like structures in three-dimensional basement membrane or extracellular matrix preparations. Cancer cells acquire a characteristic set of functional capabilities during their development, albeit through various mechanisms.
  • cancer cell is meant to encompass both pre-malignant and malignant cancer cells.
  • cancer refers to a benign tumor, which has remained localized.
  • cancer refers to a malignant tumor, which has invaded and destroyed neighboring body structures and spread to distant sites.
  • the cancer is associated with a specific cancer antigen.
  • a 3 expressing cancers refers to human cancers that express the adenosine A3 receptor or that otherwise comprise elevated concentrations of adenosine A 3 receptors. Elevated concentration is determined by comparison to normal, non-cancerous tissues of a similar cell type. Examples of A 3 expressing cancers include, without limitation, human leukemia, melanoma, pancreatic carcinoma, breast carcinoma, prostrate carcinoma, colon carcinoma, lung carcinomamalignant melanomas, histiocytic lymphoma, and some forms of astrocytoma cells.
  • HIF- 1 ⁇ expressing cancers refers to human cancer that express HIF- l ⁇ or that otherwise comprise elevated concentrations of HIF-I ⁇ .
  • Elevated concentration is determined by comparison to normal, non-cancerous tissues of a similar cell type.
  • HIF-I ⁇ expressing cancers include without limitation cervical cancer (early stage), melanoma, lung cancer (non-small cell lung carcinoma), breast cancer (including lymph node positive breast cancer and lymph node negative breast cancer), oligodendroglioma, orpharyngeal squamous cell carcinoma, ovarian cancer, oesophageal cancer, endometrial cancer, head and neck cancer, gastrointestinal stromal tumor of the stomach.
  • hypoxia refers to any condition whereby the physiology of the tissue is compromised and/or blood supply in one or more tissues is compromised. These two terms may be used interchangeably.
  • the condition also encompasses any reduction in partial pressure of O 2 in one or more tissues.
  • hypooxia or “ischemia” encompasses any condition in which a cell, tissue or organ experiences a lack of oxygen or reduced blood flow.
  • the terms “treat,” “treating” and “treatment” refer to the eradication, removal, modification, or control of primary, regional, or metastatic cancer tissue that results from the administration of one or more therapeutic agents.
  • such terms refer to the minimizing or delaying the spread of cancer resulting from the administration of one or more therapeutic agents to a subject with such a disease.
  • the term also refers to the eradication, reduction or amelioration of a symptom or disorder related to aberrant levels of HIF-I ⁇ level and/or activity.
  • a "therapeutically effective amount” refers to that amount of the therapeutic agent sufficient to delay or minimize the spread of cancer.
  • a therapeutically effective amount may also refer to the amount of the therapeutic agent that provides a therapeutic benefit in the treatment or management of a disease or disorder.
  • a therapeutically effective amount with respect to a therapeutic agent of the invention means that amount of therapeutic agent alone, or in combination with other therapies, that provides a therapeutic benefit in the treatment or management of a disease or disorder. Used in connection with an amount of a compound of the invention, the term can encompass an amount that improves overall therapy, reduces or avoids unwanted effects, or enhances the therapeutic efficacy of or synergizes with another therapeutic agent.
  • prophylactic agent and “prophylactic agents” refer to any agent(s) which can be used in the prevention of a disorder, or prevention of recurrence or spread of a disorder.
  • a prophylactically effective amount may refer to the amount of prophylactic agent sufficient to prevent the recurrence or spread of hyperproliferative disease, particularly cancer, or the occurrence of such in a patient, including but not limited to those predisposed to hyperproliferative disease, for example those genetically predisposed to cancer or previously exposed to carcinogens.
  • a prophylactically effective amount may also refer to the amount of the prophylactic agent that provides a prophylactic benefit in the prevention of disease.
  • a prophylactically effective amount with respect to a prophylactic agent of the invention means that amount of prophylactic agent alone, or in combination with other agents, that provides a prophylactic benefit in the prevention of disease.
  • the term can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of or synergizes with another prophylactic agent, such as but not limited to a therapeutic antibody.
  • the terms “manage,” “managing” and “management” refer to the beneficial effects that a subject derives from administration of a prophylactic or therapeutic agent, which does not result in a cure of the disease or diseases.
  • a subject is administered one or more prophylactic or therapeutic agents to "manage” a disease so as to prevent the progression or worsening of the disease or diseases.
  • the terms “prevent”, “preventing” and “prevention” refer to the methods to avert or avoid a disease or disorder or delay the recurrence or onset of one or more symptoms of a disorder in a subject resulting from the administration of a prophylactic agent.
  • the term “in combination” refers to the use of more than one prophylactic and/or therapeutic agents.
  • the use of the term “in combination” does not restrict the order in which prophylactic and/or therapeutic agents are administered to a subject with a disorder, e.g., hyperproliferative cell disorder, especially cancer.
  • a first prophylactic or therapeutic agent can be administered prior to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 1 minute, 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second prophylactic or therapeutic agent to a subject which had, has, or is susceptible to a disorder.
  • the prophylactic or therapeutic agents are administered to a subject in a sequence and within a time interval such that the agent of the invention can act together with the other agent to provide an increased benefit than if they were administered otherwise.
  • Any additional prophylactic or therapeutic agent can be administered in any order with the other additional prophylactic or therapeutic agents.
  • alkyl refers to a monovalent straight, branched or cyclic saturated hydrocarbon group preferably having from 1 to 20 carbon atoms, more preferably 1 to 10 carbon atoms (“lower alkyl”) and most preferably 1 to 6 carbon atoms.
  • alkylene and “lower alkylene” refer to divalent radicals of the corresponding alkane.
  • other moieties having names derived from alkanes such as alkoxyl, alkanoyl, alkenyl, cycloalkenyl, etc. when modified by "lower,” have carbon chains often or less carbon atoms.
  • substituted alkyl refers to an alkyl group, preferably of from 1 to 10 carbon atoms ("substituted lower alkyl”), having from 1 to 5 substituents, and preferably 1 to 3 substituents, selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, substituted amino aminoacyl, aminoacyloxy, oxyacylamino, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, hydroxyamino, alkoxyamino, nitro, — SO-alkyl, —
  • alkyl refers to an alkyl group with an aryl substituent. Binding is through the alkyl group.
  • Alkaryl refers to an aryl group with an alkyl substituent, where binding is through the aryl group.
  • alkoxy refers to the group “alkyl-0 — ", where alkyl is as defined above. Preferred alkoxy groups include, by way of example, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1,2-dimethylbutoxy, and the like.
  • alkenyl refers to an unsaturated, straight or branched hydrocarbon group preferably having from 2 to 10 carbon atoms and more preferably 2 to 6 carbon atoms and having at least 1 and preferably from 1-2 double bonds.
  • alkynyl refers to an unsaturated, straight or branched hydrocarbon group preferably having from 2 to 10 carbon atoms and more preferably 2 to 6 carbon atoms and having at least 1 and preferably from 1-2 triple bonds.
  • acyl refers to the groups alkyl-C(O) — , substituted alkyl-C(O)— , cycloalkyl-C(O)— , substituted cycloalkyl-C(O)— , aryl-C(O)— , substituted aryl-C(O) — , heteroaryl-C(O) — and heterocyclic-C(O) — where alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, heteroaryl and heterocyclic are as defined herein.
  • aminoacyl refers to the group — C(O)NR 33 R 33 where each R 33 is independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.
  • acylamino refers to the group R 34 — C(O) — N(R 33 ) — where each R 34 is independently alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, heteroaryl and heterocyclic are as defined herein.
  • R 33 is independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.
  • acyloxy refers to the group R 34 — C(O) — where each R 34 is independently alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, cycloalkyl, aryl, heteroaryl and heterocyclic are as defined herein.
  • amino acid means an alpha amino acid selected from those amino acids which naturally occur in proteins but without regard for specific stereochemical properties.
  • protected amino acid means an amino acid of which the alpha amino group has been converted to a less reactive moiety, but a moiety which can be converted back to the amino group with relative ease.
  • amino acid residue and “amino acid moiety” are used synonymously herein.
  • aryl refers to an unsaturated aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring ⁇ e.g., phenyl) or multiple condensed (fused) rings ⁇ e.g., naphthyl or anthryl).
  • Preferred aryls include phenyl, naphthyl and the like.
  • such aryl groups can optionally be substituted with from 1 to 5 substituents and preferably 1 to 3 substituents selected from the group consisting of hydroxy, acyl, alkyl, alkoxy, alkenyl, alkynyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, amino, substituted amino, aminoacyl, acyloxy, acylamino, alkaryl, aralkyl, aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halo, nitro, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy, alkylthio, — SO-alkyl, — SO-substitute
  • cycloalkyl refers to cyclic alkyl groups of from 3 to 12 carbon atoms having a single cyclic ring or multiple condensed rings.
  • Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, and the like, or multiple ring structures such as adamantyl, and the like.
  • halo or halogen refer to fluoro, chloro, bromo and iodo and preferably is either fluoro or chloro.
  • heteroaryl refers to an aromatic carbocyclic group of from 1 to 15 carbon atoms and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur within at least one ring (if there is more than one ring).
  • heteroaryl groups can be optionally substituted with from 1 to 5 substituents and preferably 1 to 3 substituents selected from the group consisting of hydroxy, acyl, alkyl, alkoxy, alkenyl, alkynyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, amino, substituted amino, aminoacyl, acyloxy, acylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halo, nitro, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy, tbioheteroaryloxy, — SO-alkyl, — SO-substituted alkyl, — SO-aryl,
  • Preferred substituents include alkyl, alkoxy, halo, cyano, nitro, trihalomethyl, and thioalkoxy.
  • Such heteroaryl groups can have a single ring (e.g., pyridyl or furyl) or multiple condensed rings (e.g., indolizinyl or benzothienyl).
  • Heterocycle or “heterocyclic” refers to a monovalent saturated or unsaturated carbocyclic group having a single ring or multiple condensed rings, from 1 to 15 carbon atoms and from 1 to 4 hetero atoms selected from the group consisting of nitrogen, sulfur and oxygen within the ring.
  • heterocyclic groups can be optionally substituted with 1 to 5 substituents selected from the group consisting of alkyl, substituted alkyl, alkoxy, substituted alkoxy, aryl, aryloxy, halo, nitro, heteroaryl, thiol, thioalkoxy, substituted thioalkoxy, thioaryloxy, trihalomethyl, and the like.
  • substituents selected from the group consisting of alkyl, substituted alkyl, alkoxy, substituted alkoxy, aryl, aryloxy, halo, nitro, heteroaryl, thiol, thioalkoxy, substituted thioalkoxy, thioaryloxy, trihalomethyl, and the like.
  • Such heterocyclic groups can have a single ring or multiple condensed rings.
  • any of the above groups that contain one or more substituents it is understood, of course, that such groups do not contain any substitution or substitution patterns which are sterically impractical and/
  • carboxylic acid derivatives and “sulfonic acid derivatives” refer to -C(X)R 31 , — C(X)- N(R 31 ) 2 , -C(X)OR 31 , -C(X)SR 31 , — SO b R 31 , — SO b OR 31 , — SO b SR 31 , or SO b - N(R 31 ) 2 , where X is 0, S or NR 31 , where R 31 is hydrogen, alkyl, substituted alkyl or aryl, and activated derivatives thereof, such as anhydrides, esters, and halides such as chlorides, bromides and iodides.
  • “Pharmaceutically acceptable salts” refers to pharmaceutically acceptable salts of a compound of Formulas HA, HB, HC and HD which salts are derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like can be used as the pharmaceutically acceptable salt.
  • FIGS. IA through IH illustrate the saturation of [ 3 H]-MRE 3008-F20 binding to A 3 adenosine receptors in human cancers.
  • K D and B max values are reported in Table 8. Values are the means and S. E. of the mean of three separate experiments performed in triplicate. In the inset the Scatchard plot of the same data is shown.
  • Figure 2 illustrates colony formation assay of A375 cells.
  • Cells were treated with different drugs and colonies were counted after 7 days. The values represent the mean ⁇ SEM of four independent experiments.
  • Figure 3 A through 3D illustrate typical dose response curves of A375 cells exposed to increasing concentrations of cytotoxic agents vindesine (Figure 3A, Figure 3B) and TaxolTM brand paclitaxel (Figure 3C, Figure 3D).
  • the curves with open symbols represent the cytotoxic agent alone.
  • the curves with closed symbols represent the cytotoxic agent in the presence of 10 ⁇ M MRE 3008F2010.
  • Figure 3A and Figure 3C illustrate G 2 /M phases arrest calculated as percentage of untreated cells (control).
  • Figure 3B and Figure 3D illustrate the accumulation (percentage of total living cells) of the SUb-G 1 (apoptosis) population. Cells were fixed in 70% ethanol, stained with PI, and analysed by flow cytometry.
  • Figure 4A through 4F illustrate induction of G 2 /M phases arrest by MRE 3008F20 on exponentially growing A375 cells treated with paclitaxel or with vindesine.
  • A375 cells are treated for 24 hours with drug- vehicle as control (Figure 4A, Figure 4D); with 1 nM vindesine (Figure 4B), with 25 ng/ml paclitaxel ( Figure 4E); with 1 nM vindesine plus 10 ⁇ M MRE 3008F20 ( Figure 4C); or with 25 ng/ml paclitaxel plus 10 ⁇ M MRE 3008F20 ( Figure 4F).
  • Cells were fixed in 70% ethanol, stained with PI, and analysed by flow cytometry. The percentage of cells at Gi, S and G 2 /M phases was quantified.
  • FIG. 5 A illustrates dose-response curve for G 2 /M phases arrest of A375 cells exposed to increasing concentrations OfA 3 adenosine receptor antagonists in the presence of 1 nM vindesine. Cells were fixed in 70% ethanol, stained with PI, and analysed by flow cytometry.
  • Figure 6 A through 6D illustrate flow chromatogram for Rhodamine 123 (Rh 123) accumulation by A375 cells and HeL023 cells.
  • Figure 6A illustrates the accumulation by A375 cells.
  • Figure 6B illustrates the accumulation by A375 cells in the presence of 10 ⁇ M MRE 3008F20.
  • Figure 6C illustrates the accumulation by HeL023 cells.
  • Figure 6D illustrates the accumulation by HeL023 cells in the presence of 10 ⁇ M MRE 3008F20.
  • F MAX represents the maximum load of Rh 123 (gray filled area).
  • F RES shows residual Rh 123 fluorescence after the P-gp mediated drug efflux was allowed for 3 hours (black filled area). Rh 123 unstained cell chromatogram is reported as unfilled area.
  • Figure 7A illustrates the effect of inhibitors of MEK- (PD98059), ERK 1/2- (UO 126) and p38 MAPK ⁇ (SB203580) -signalling on the G 2 M phases arrest induced by paclitaxel (filled bars) and by vindesine (empty bars).
  • the residual G 2 /M phases arrest is reported as the percentage of control cells (cells treated with paclitaxel plus MRE 3008F20 for TU, TS, TP or with vindesine plus MRE 3008F20, for VU, VS, VP.
  • Figure 7B illustrates the effect of inhibitors of MEK- (PD98059), ERK 1/2- (U0126) and p38MAPK- (SB203580) -signalling on apoptosis induced by paclitaxel.
  • T paclitaxel (25 ng/ml)
  • V vindesine 1 ⁇ M
  • U U0126 30 ⁇ M
  • P PD9805920 ⁇ M
  • S SB203580 1 ⁇ M
  • M MRE 3008F20 10 ⁇ M
  • C cells treated with metabolic inhibitor vehicle (DMSO) (control).
  • DMSO metabolic inhibitor vehicle
  • Apoptotic cells (Apo) with sub-diploid DNA content are reported.
  • FIG. 9 Induction of HIF-I expression by hypoxia. A375 cells were cultured in normoxia for 4 hours (lane normoxia) or under hypoxic conditions for 2, 3, 4, 8, 16 and 24 hours (lanes 2-7). Whole cellular protein extracts were prepared and subjected to immunoblot assay using an anti-HIF-l ⁇ monoclonal antibody. The blot was then stripped and used to determine HIF-l ⁇ expression using an anti-HIF-l ⁇ monoclonal antibody. Tubulin shows equal loading protein.
  • FIG. 10 Iinduction of HIF-I ⁇ expression by adenosine.
  • A A375 cells were cultured in normoxia for 4 hours (lane normoxia). A375 cells were treated without (lane 1) or with adenosine 10 nM (lane 2), 100 nM (lane 3), 1 ⁇ M (lane 4), 10 ⁇ M (lane 5) and 100 ⁇ M (lane 6) in hypoxia for 4 hours. Cellular extracts were prepared and subjected to immunoblot assay using an anti-HIF-l ⁇ monoclonal antibody. The blot was then stripped and used to determine HIF-I ⁇ expression using an anti-HIF-l ⁇ monoclonal antibody. Tubulin shows equal loading protein.
  • A375 cells were treated without (lane 1, control) or with adenosine 100 ⁇ M (lanes 2-6) and exposed to the A 1 receptor antagonist DPCPX 100 nM (lane 3), or A 2A receptor antagonist SCH 58261 100 nM (lane 4), or A 2B receptor antagonist MRE 2029F20 100 nM (lane 5), or A 3 receptor antagonist MRE 3008F20 100 nM (lane 6) in hypoxia for 4 hours.
  • Cellular extracts were prepared and subjected to immunoblot assay using an anti- HIF-l ⁇ monoclonal antibody. The blot was then stripped and used to determine HIF- l ⁇ expression using an anti-HIF-l ⁇ monoclonal antibody.
  • Tubulin shows equal loading protein.
  • FIG. 11 Induction of HIF-I expression by A 3 receptor stimulation: time course.
  • A A375 cells were cultured in normoxia for 4 hours (lane normoxia) or under hypoxic conditions in the absence (-) or in the presence (+) of the A 3 receptor agonist Cl-IB- MECA (100 nM) for 2, 4, 8, 16 and 24 hours.
  • Whole cellular protein extracts were prepared and subjected to immunoblot assay using an anti-HIF-l ⁇ monoclonal antibody. The blot was then stripped and used to determine HIF- l ⁇ expression using an anti-HIF- l ⁇ monoclonal antibody.
  • Tubulin shows equal loading protein.
  • FIG. 12 Induction of HIF-I ⁇ expression by A 3 receptor stimulation: dose response.
  • A A375 cells were treated without (lane 1) or with Cl-IB-MECA 0.1 nM (lane 2), 1 nM (lane 3), 10 nM (lane 4) 100 nM (lane 5) and 1 ⁇ M (lane 6) in normoxia and in hypoxia for 4 hours.
  • Cellular extracts were prepared and subjected to immunoblot assay using an anti-HIF-l ⁇ monoclonal antibody. The blot was then stripped and used to determine HIF- l ⁇ expression using an anti-HIF-l ⁇ monoclonal antibody.
  • B Typical dose response curve of A375 cells exposed to adenosine in hypoxia is shown.
  • the immunoblot signals were quantified using molecular analyst /PC densitometry software (Bio-Rad).
  • FIG. 13 Effect of A 3 receptor antagonist MRE 3008F20.
  • A375 cells were treated in hypoxia for 4 hours without (-) or with (+) Cl-IB-MECA 10 nM, MRE 3008F20 10 nM (lanes 3, 4) and MRE 3008F20 100 nM (lanes 5, 6).
  • Cellular extracts were prepared and subjected to immunoblot assay using an anti-HIF-l ⁇ monoclonal antibody. The blot was then stripped and used to determine HIF- l ⁇ expression using an anti-HIF- l ⁇ monoclonal antibody.
  • B The immunoblot signals were quantified using molecular analyst /PC densitometry software (Bio-Rad).
  • blot was then stripped and used to determine HIF-I ⁇ expression using an anti-HIF-l ⁇ monoclonal antibody.
  • D Typical dose response curve of A375 cells exposed to MRE 3008F20 in hypoxia is shown.
  • the immunoblot signals were quantified using molecular analyst /PC densitometry software (Bio-Rad).
  • FIG. 14 A 3 receptor expression silencing by siRNA transfection.
  • A Analysis of siRNA transfection efficiency in A375 cells. Representative flow chromatograms of siRN A-FITC accumulation (gray filled area) in A375 cells transfected with siRNA-FITC. Unfilled area shows A375 cells transfected with RNAiFectTM Transfection reagent without S ⁇ RNA A3 . Fluorescence was quantified by flow cytometry 5 hours post-transfection.
  • (B) Relative A 3 receptor mRNA quantification, related to ⁇ -actin mRNA, by Real-Time RT-PCR. A375 cells were transfected by RNAiFectTM Transfection reagent or S1RNA A3 and cultured for 24, 48 and 72 hours.
  • C Western blot analysis using an anti-A 3 receptor polyclonal antibody of protein extracts from A375 cells treated by RNAiFectTM Transfection reagent (control) or SiRNA A3 and cultured for 24, 48 and 72 hours in normoxia. Tubulin shows equal loading protein.
  • Cellular extracts were prepared and subjected to immunoblot assay using an anti-HIF- l ⁇ monoclonal antibody. The blot was then stripped and used to determine HIF- 1 ⁇ expression using an anti-HIF- 1 ⁇ monoclonal antibody.
  • FIG. 16 A 3 receptor stimulation induces HIF- lot accumulation through a transcription-independent pathway.
  • A375 cells were pretreated with actinoniycin D (10 ⁇ g/ml) for 30 minutes and then exposed to hypoxia.
  • HIF-I ⁇ accumulation was induced by the exposure of A375 cells to Cl-IB-MECA 100 nM (+) for 4 hours in hypoxia in the absence (lane 2) or in the presence (lane 4) of actinomycin D.
  • Cellular extracts were prepared and subjected to immunoblot assay using an anti-HIF-l ⁇ monoclonal antibody. Tubulin shows equal loading protein.
  • B The immunoblot signals were quantified using molecular analyst/PC densitometry software (Bio-Rad).
  • FIG. 1 Induction of HIF-I ⁇ accumulation by A 3 receptor stimulation in normoxia.
  • A375 cells were exposed to 100 ⁇ M CoCl 2 alone (lane 1) or in the presence of Cl-IB-MECA 1 nM (lane 2), 10 nM (lane 3), 100 nM (lane 4), 1 ⁇ M (lane 5) and 10 ⁇ M (lane 6) in normoxia for 4 hours.
  • Cellular extracts were prepared and subjected to immunoblot assay using an anti-HIF- l ⁇ monoclonal antibody. The blot was then stripped and used to determine HIF- l ⁇ expression using an anti-HIF-l ⁇ monoclonal antibody.
  • A375 cells were exposed to 100 ⁇ M CoCl 2 for 4 hours in normoxia.
  • HIF-l ⁇ accumulation was induced by the exposure of A375 cells to Cl-IB-MECA 100 nM (+) for 4 (lanes 1 and 4) or 6 hours (lanes 5 and 6) in the absence (lanes 1 and 2) or in the presence (lanes 3-6) of cycloheximide (1 ⁇ M).
  • Cellular extracts were prepared and subjected to immunoblot assay using an anti-HIF- l ⁇ monoclonal antibody. Tubulin shows equal loading protein.
  • Figure 18. A 3 receptor activation does not affect HIF- l ⁇ degradation in normoxia.
  • A375 cells were incubated in hypoxia in the absence (lanes 1 to 4) or in the presence of Cl-IB-MECA 100 nM (lanes 5 to 8). After 4 hours, melanoma cells were exposed to normoxia and a time-course of HIF-l ⁇ disappearance was performed at 0, 5, 10 and 15 minutes. Cellular extracts were prepared and subjected to immunoblot assay using an anti-HIF-l ⁇ monoclonal antibody. Tubulin shows equal loading protein.
  • B The immunoblot signals were quantified using molecular analyst /PC densitometry software (Bio-Rad).
  • the present invention is based, in part, on the surprising discovery by the inventors that adenosine, a purine nucleoside present within hypoxic regions of solid tumors, modulates hypoxia-inducible factor 1 (HIF-I) expression.
  • HIF-I hypoxia-inducible factor 1
  • the inventors have found that in the human A375 melanoma cell line adenosine up-regulates HIF- l ⁇ protein expression in response to hypoxia in a dose- and time-dependent manner.
  • the response to adenosine was not blocked by A 1 , A 2A or A 2 B receptor antagonists, while it was abolished by A 3 receptor antagonists.
  • the inventors have found that Cl-IB-MECA, an adenosine analogue binding with high affinity to A 3 receptors, mimicked adenosine effect in hypoxic cells. Furthermore, A 3 receptor antagonists prevented HIF- l ⁇ protein accumulation in response to A 3 receptor stimulation.
  • the response to adenosine is generated at the cell surface since the inhibition of A 3 receptor expression, by using small interfering RNA, abolished the nucleoside effects.
  • the main intracellular signaling pathways sustained by A 3 receptor stimulation in hypoxia involve p44/p42 mitogen- activated protein kinase (MAPK) and p38 MAPK activation.
  • MAPK mitogen- activated protein kinase
  • the inventors have thus found for the first time that adenosine plays a critical role in HIF- l ⁇ regulation in hypoxia.
  • the inventors have found that compounds which are antagonists of the adenosine receptors, preferably the A 3 receptor, inhibit the protective effect of adenosine on growing tumor cells when such cells are starved of oxygen (/. e. , before an adequate vasculature is developed, or when anti-angiogenesis agents are administered.
  • the antagonists of the invention can bind in the same site as adenosine, or can be allosteric antagonists (i.e., bind at a site different from where adenosine binds, but adversely affect the ability of adenosine to bind to the site or adversely affect the ability of adenosine, once bound to the adenosine receptors particularly A 3 receptors, to protect growing tumor cells).
  • the present invention relates to methods for the treatment, prevention, and/or management of diseases or disorders associated with overexpression of HIF-I ⁇ and/or increased HIF- l ⁇ activity (e.g., cancer, respiratory disorders such as asthma and obstructive pulmonary disorders) by using adenosine receptor antagonists, particularly A 3 receptor antagonists, alone or in combination with A 1 receptor antagonists.
  • adenosine receptor antagonists particularly A 3 receptor antagonists
  • administration of antagonists for the adenosine receptors antagonizes the protective effects against hypoxia and renders those cells susceptible to destruction due to hypoxia.
  • adenosine receptors in particular, the A 3 receptors
  • antagonists for the adenosine receptors, particularly A 3 receptors are particularly effective in enhancing the activity of anti-tumor agents.
  • the methods and compositions of the invention comprising A 3 receptor antagonists are particularly useful when the levels of HIF-I ⁇ expression and/or activity are elevated above the standard or background level, as determined using methods known to those skilled in the art and dislosed herein.
  • "elevation" of a measured level of HIF-I ⁇ relative to a standard level means that the amount or concentration of HIF-I ⁇ in a sample or subject is sufficiently greater in a subject or sample relative to the standard as detected by any method now known in the art or to be developed in the future for measuring HIF- l ⁇ levels.
  • elevation of the measured level relative to a standard level may be any statistically significant elevation detectable.
  • Such an elevation in HIF-I ⁇ expression and/or activity may include, but is not limited to about a 10%, about a 20%, about a 40%, about an 80%, about a 2-fold, about a 4-fold, about an 10-fold, about a 20- fold, about a 50-fold, about a 100-fold, about a 2 to 20 fold, 2 to 50 fold, 2 to 100 fold, 20 to 50 fold, 20 to 100 fold, elevation, relative to the standard.
  • standard level refers to a baseline amount of HIF-I ⁇ level as determined in one or more normal subjects, i.e., a subject with no known history of past or current diseases, disorders or cancer.
  • the measurement of HIF-I ⁇ level may be carried out using an HIF-l ⁇ probe or a HIF-l ⁇ activity assay (see Section 5.4.4).
  • HIF-l ⁇ activity assay see Section 5.4.4.
  • reference to measuring a level of HIF-l ⁇ in a method of the invention relates to any proxy for HIF-l ⁇ levels.
  • levels may include, but are not limited to, the abundance of HIF-l ⁇ nucleic acid or amino acid sequences in a sample from a subject.
  • a level of HIF-I ⁇ may correspond to the abundance of full-length HIF- l ⁇ protein.
  • a level of HIF-I ⁇ may correspond to abundance of a fragment, analog or derivative of HIF-I ⁇ protein.
  • a level of HIF-I ⁇ can be determined by measuring the abundance of nucleic acids (or sequences complementary thereto) that corresponds to all or fragments of HIF-I ⁇ .
  • the abundance of niRNA encoding HIF- l ⁇ is measured.
  • a probe with which the amount or concentration of HIF- l ⁇ can be determined includes but is not limited to an antibody, an antigen, a nucleic acid, a protein, or a small molecule.
  • the probe is the HIF-l ⁇ protein or a fragment thereof .
  • the probe is an antibody that immunospecifically binds to HIF-l ⁇ , such as e.g., a monoclonal antibody or a binding fragment thereof.
  • measuring a level of HIF-I ⁇ comprises testing at least one aliquot of the sample, said step of testing comprising: (a) contacting the aliquot with an antibody or a fragment thereof that is immunospecific for HIF-I ⁇ , and (b) detecting whether and how much binding has occurred between the antibody or a fragment thereof and at least one species of HIF-I ⁇ in the aliquot.
  • measuring a level of HIF-I ⁇ comprises testing at least one aliquot, said step of testing comprising: (a) contacting the aliquot with a nucleic acid probe that is hybridizable to HIF- l ⁇ mRNA, and (b) detecting whether and how much hybridization has occurred between the nucleic acid probe and at least one species of HIF-I ⁇ mRNA in the aliquot.
  • measuring a level of HIF-I ⁇ involves quantitating the amount of complex formation. For example the amount of complex formation between the antibody or a fragment thereof and at least one species of HIF- 1 ⁇ in the aliquot would correlate with the amount of at least one species of HIF-I ⁇ in the aliquot of the sample.
  • the antibody or other probe is labeled with a detectable marker.
  • the detectable marker is a chemiluminescent, enzymatic, fluorescent, or radioactive label.
  • the therapeutic methods of the invention comprising administering a therapeutically effective amount of an A 3 receptor antagonist of the invention improve the therapeutic efficacy of treatment for diseases or disorders associated with overexpression of HIF-l ⁇ and/or increased HIF-l ⁇ activity (e.g., cancer, respiratory disorders such as asthma and obstructive pulmonary disorders) relative to the traditional modes of such therapies.
  • the methods of the invention reduce the HIF-l ⁇ level to the background level within within one day, one week, 1 month, or 2 months of the commencement of the therapeutic regime.
  • the methods of the invention reduce the level of HIF- l ⁇ to a particular level, that level is maintained during the treatment regimen, such that the maintained level of HIF-I ⁇ is sufficient and effective to result in regression of the disease, e.g., cancer.
  • the methods of the invention result in a reduction of HIF-I ⁇ level to the background level.
  • the invention encompasses reduction of the HIF-I ⁇ level to a level which is within about 10%, about 20%, about 30%, about 40%, about 50% of the background level.
  • the invention encompasses a method for treatment, prevention and/or management of diseases or disorders associated with overexpression of HIF-l ⁇ and/or increased HIF-l ⁇ activity (e.g., cancer, respiratory disorders such as asthma and obstructive pulmonary disorders) comprising administering a therapeutically and/or prophylactically effective amount of an A 3 receptor antagonist compound as disclosed herein.
  • the invention also encompasses a method for determining the prognosis of a a disease or disorder associated with overexpression of HIF-I ⁇ and/or increased HIF- l ⁇ activity in a subject.
  • the subject is human, hi some embodiments, the subject has been previously treated with a therapy regimen.
  • the invention encompasses measuring at least a level of HIF-I ⁇ in a subject to determine if the subject is in need of the therapeutic and or prophylcatic methods of the inventon.
  • the invention encompasses measuring a level of at least one HIF-I ⁇ in a sample obtained from the subject and comparing the level measured to a standard level, wherein elevation of the measured level of at least one HIF-I ⁇ relative to the standard level indicates that the subject is at an increased risk for progression of the disease or disorder, e.g., metastasis of the cancer.
  • the invention encompasses compounds which are adenosine receptor antagonists particularly A 3 antagonists for use in the methods of the invention. Examples of such compounds are disclosed in U.S. Patent Nos 6,326,390; 6,407,236; 6,448,253;
  • a 3 receptor antagonists of the invention are particularly useful for prevention, treatment, and/or management of cancer, for example, as a single agent therapy or in combination with other modes of therapy for cancer.
  • the invention encompasses methods of treatment, prevention, or management of cancers which are A 3 expressing cancers including, but not limited to, human leukemia, melanoma, pancreatic carcinoma, ovarian carcinoma, breast carcinoma, prostrate carcinoma, colon carcinoma, lung carcinoma, malignant melanomas, histiocytic lymphoma, and some forms of astrocytoma cells.
  • cancers which are A 3 expressing cancers including, but not limited to, human leukemia, melanoma, pancreatic carcinoma, ovarian carcinoma, breast carcinoma, prostrate carcinoma, colon carcinoma, lung carcinoma, malignant melanomas, histiocytic lymphoma, and some forms of astrocytoma cells.
  • the invention encompasses methods of treatment, prevention, or management which have high level of expression of HIF-I ⁇ including, but not limited to, cervical cancer (early stage), lung cancer (non-small cell lung carcinoma), breast cancer (including lymph node positive breast cancer and lymph node negative breast cancer), oligodendroglioma, orpharyngeal squamous cell carcinoma, ovarian cancer, oesophageal cancer, endometrial cancer, head and neck cancer, gastrointestinal stromal tumor of the stomach
  • the methods of the invention are particularly useful in cancer therapy for providing tissue selectivity, such that the biological effect is observed in tumoral hypoxic cells, where high adenosine concentrations has resulted in increased HIF- l ⁇ accumulation.
  • solid tumors refer to a locus of tumor cells where the majority of the cells are tumor cells or tumor-associated cells, including but not limited to laryngeal tumors, brain tumors, and other tumors of the head and neck; colon, rectal and prostate tumors; breast and thoracic solid tumors; ovarian and uterine tumors; tumors of the esophagus, stomach, pancreas and liver; bladder and gall bladder tumors; testicular cancer; skin tumors such as melanomas.
  • the tumors encompassed within the invention can be either primary or a secondary tumor resulting from metastasis of cancer cells elsewhere in the body to the chest.
  • the invention encompasses use of the adenosine receptor antagonists of the invention, particulalry A 3 receptor antagonists in inhibiting tumor growth in a mammal, including humans.
  • the invention encompasses methods for administering an effective amount of an adenosine receptor antagonists, preferably A3 receptor antagonists to inhibit the ability of adenosine to protect tumor cells.
  • the present invention encompasses methods for treatment, prevention and/or management of cancer comprising administering an adenosine receptor antagonist, particularly an A 3 receptor antagonist combination with other therapeutic and/or prophylactic agents, e.g., cytotoxic agents.
  • an adenosine receptor antagonist particularly an A 3 receptor antagonist combination with other therapeutic and/or prophylactic agents, e.g., cytotoxic agents.
  • cytotoxic agents e.g., cytotoxic agents.
  • the inventors have surprisingly found that adenosine A 3 receptor antagonists synergistically enhance cytotoxic treatment and counter some forms of multi-drug resistance.
  • the combination therapies of the invention will attack the tumor cell directly, inhibit growth of new blood vessels around the tumor cell, and, by virtue of the adenosine A 3 antagonists, inhibit the ability of the cell to survive without the growth of new blood vessels.
  • high affinity adenosine A 3 receptor antagonists are useful as enhancers for many chemotherapeutic treatments of adenosine A 3 receptor expressing cancers.
  • high affinity adenosine A 3 receptor antagonists also counter P-glycoprotein (P-gp) effuse pump multidrug resistance (MDR).
  • P-gp P-glycoprotein
  • MDR multidrug resistance
  • the present invention encompasses the use of high affinity adenosine A 3 receptor antagonists for enhancing chemotherapeutic treatment of cancers expressing adenosine A 3 receptors and countering multi-drug resistance in cancers expressing P-glycoprotein or MRP.
  • adenosine A 3 receptor antagonists are administered before or during administration of a taxane family, vinca alkaloid, camptothecin or antibiotic compound.
  • Preferred high affinity A 3 receptor antagonists include compounds of the general formulas HA, HB, HC and IID described herein, vide infra.
  • the present invention encompasses therapies which involve administering one or more of the compounds of the invention, to an animal, preferably a mammal, and most preferably a human, for preventing, treating, or ameliorating symptoms associated with a cancer, a disease or disorder associated with hypoxia-inducible factor 1 -a (HIF- 1 ⁇ ).
  • the invention further provides a pharmaceutical composition comprising a therapeutically or prophylactically effective amount of a compound of the invention that specifically binds an A 1 or A 3 receptor and a pharmaceutically acceptable carrier
  • the pharmaceutical formulation includes an A 3 receptor antagonist and one or more excipients.
  • the formulations can also include other anti-tumor agents, including cytotoxic agents and other anti-angiogenesis agents, including adenosine A 2A antagonists, such as ZM241385 and SCH 5861, adenosine A 2B antagonists, such as MRE-20290-F20 and AS-16, and anti-VEGF antibodies, including humanized and chimeric antibodies.
  • the composition includes an effective amount to inhibit tumor growth of an adenosine A 3 receptor antagonist, a cytotoxic agent, and an anti-angiogenesis agent.
  • the invention provides methods for treating a disease or disorder associated with HIF- l ⁇ in a patient, comprising administering to a patient in need thereof an effective amount of a compound or a pharmaceutically acceptable salt of the compound having the following general formulas HA and HB:
  • A is imidazole, pyrazole, or triazole
  • R is -C(X)R 1 , -C(X)-N(R 1 );., -C(X)OR 1 , -C(X)SR 1 , -SO b R 1 , — SO b OR 1 , — SO b SR 1 , or SO b - N(R ⁇ 2 ;
  • R 1 is hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, lower alkenyl, lower alkanoyl, or, if linked to a nitrogen atom, then taken together with the nitrogen atom, forms an azetidine ring or a 5-6 membered heterocyclic ring containing one or more heteroatoms such as N, O, S;
  • R 2 is hydrogen, alkyl, alkenyl, alkynyl, substituted alkyl, substituted alkenyl, substituted alkynyl, aralkyl, substituted aralkyl, aryl, substituted aryl, heteroaryl, or substituted heteroaryl;
  • R 3 is furan, pyrrole, thiophene, benzofuran, indole, benzothiophene, optionally substituted with one or more substituents as described herein for substituted heteroaryl rings;
  • X is O, S, or NR 1 ; b is 1 or 2.
  • R 1 is hydrogen; C 1 to C 8 alkyl; C 2 to C 7 alkenyl, C 2 to C 7 alkynyl; C 3 to C 7 cycloalkyl; C 1 to C 5 alkyl substituted with one or more halogen atoms, hydroxy groups, C 1 to C 4 alkoxy, C 3 to C 7 cycloalkyl or groups of formula — NR 32 R 32 , — C(O)NR 32 R 32 ; aryl, substituted aryl wherein the substitution is selected from the group consisting of C 1 to C 4 alkoxy, C 1 to C 4 alkyl, nitro, amino, cyano, C 1 to C 4 haloalkyl, Ci to C 4 haloalkoxy, carboxy, carboxyamido; C 7 to C 10 aralkyl in which the aryl moiety can be substituted with one or more of the substituents indicated above for the aryl group; a group of formula — (CH 2 ) m
  • C 1 to C 8 alkyl groups are methyl, ethyl, propyl, butyl and isopentyl.
  • Examples of C 3 to C 7 cycloalkyl groups include cyclopropyl, cyclopentyl, and cyclohexyl.
  • Examples of C 1 to C 5 alkyl groups substituted with C 3 to C 7 cycloalkyl groups include cyclohexylmethyl, cyclopentylmethyl, and 2-cyclopentylethyl.
  • substituted C 1 to C 5 alkyl groups include 2-hydroxyethyl, 2-methoxyethyl, trifluoromethyl, 2-fluoroethyl, 2-chloroethyl, 3-aminopropyl, 2-(4-methyl-l-piperazine)ethyl, 2-(4-morpholinyl)ethyl, 2- aminocarbonylethyl, 2-dimethylaminoethyl, 3-dimethylaminopropyl.
  • Aryl is preferably phenyl, optionally substituted with Br, Cl, F, methoxy, nitro, cyano, methyl, trifluoromethyl, difluoromethoxy groups.
  • Examples of 5 to 6 membered ring heterocyclic groups containing N, O and/or S include piperazinyl, morpholinyl, thiazolyl, pyrazolyl, pyridyl, furyl, thienyl, pyrrolyl, triazolyl, tetrazolyl.
  • Examples of C 7 to C 1O aralkyl groups comprise benzyl or phenethyl optionally substituted by one or more substituents selected from Cl, F, methoxy, nitro, cyano, methyl, trifluoromethyl, and difluoromethoxy.
  • R 1 is hydrogen, C 1 to C 8 alkyl, aryl or C 7 to C 10 aralkyl, optionally substituted, preferably with halogen atoms.
  • X is O
  • R 2 is C 2 -C 3 alkyl or substituted alkyl and R 3 is furan. In a specific embodiment R 2 is alkyl.
  • R is a phenethyl group in which the phenyl ring is substituted with one or more substituents selected from the group consisting of chlorine, fluorine atoms, methoxy, nitro, cyano, methyl, trifluoromethyl, and difluoromethoxy groups.
  • substituents selected from the group consisting of chlorine, fluorine atoms, methoxy, nitro, cyano, methyl, trifluoromethyl, and difluoromethoxy groups.
  • the invention provides methods for treating a disease or disorder associated with HIF- l ⁇ in a patient, comprising administering to a patient in need thereof an effective amount of a compound or a pharmaceutically acceptable salt of the compound having the general formula HA: wherein:
  • A is imidazole, pyrazole, or triazole;
  • R is -C(X)R 1 , -C(X)-N(R 1 );,, -C(X)OR 1 , -C(X)SR 1 , -SO n R 1 , -SO n SR 1 , or — SO n N(R 1 );,;
  • R 1 is hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, heteroaryl, heterocyclic, lower alkenyl, lower alkanoyl, or, if linked to a nitrogen atom, then taken together with the nitrogen atom, forms an azetidine ring or a 5-6 membered heterocyclic ring containing one or more heteroatoms;
  • R 2 is hydrogen, alkyl, substituted alkyl, alkenyl, aralkyl, substituted aralkyl, heteroaryl, substituted heteroaryl or aryl;
  • R 3 is furan, pyrrole, thiophene, benzofuran, benzypyrrole, benzothiophene, optionally substituted with one or more substituents selected from the group consisting of hydroxy, acyl, alkyl, alkoxy, alkenyl, alkynyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, amino, substituted amino, aminoacyl, acyloxy, acylamino, alkaryl, aryl, substituted aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halo, nitro, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, aminoacyloxy, thioalkoxy, substituted thioalkoxy, -SO-alkyl, -SO-substituted alkyl, -SO-aryl, -SO- heteroaryl, -SO 2 -
  • the method is conducted using the compound of the general formula IIA, wherein A is a triazolo ring. In another specific embodiment of the invention the method is conducted using the compound of the general formula IIA, wherein A is a pyrazolo ring.
  • the method uses as high affinity adenosine A3 receptor antagonist the "phenyl-carbamoyl-amino" compounds of the general formula HC and pharmaceutical salts thereof:
  • A is imidazole, pyrazole, or triazole
  • R 2 is hydrogen, alkyl, substituted alkyl, alkenyl, aralkyl, substituted aralkyl, heteroaryl, substituted heteroaryl or aryl;
  • R 3 is furan; and R 6 is aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle or substituted heterocycle.
  • Non-limiting examples of the compounds of the general formula HA and HC include compounds listed below in Tables 2 and 3.
  • N 1 - ⁇ 4-[(4-methylpiperazin-l -yl)sulfonyl] -phenyl ⁇ urea 106d N-[2-(2-furyl)-8-methyl-8H-pyrazolo[4,3-e][l,2,4]triazolo[l,5-c]pyrimidin-5-yl]-N'- pyridin-4-yl urea 107d N-[2-(2-furyl)-8-methyl-8H-pyrazolo[4,3-e][l,2,4]triazolo[l,5-c]pyrimidin-5-yl]-N'- pyridin-4-ylurea hydrochloride
  • the invention provides methods for treating a disease or disorder associated with HIF-I ⁇ in a patient, comprising administering to a patient in need thereof an effective amount of a compound or a pharmaceutically acceptable salt of the compound having the following general formula HD:
  • R is -C(X)R 1 , -C(X)-N(R 1 K -C(X)OR 1 , -C(X)SR 1 , -SO b R 1 , — SO b OR 1 , —
  • R 1 is hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, heterocyclic, lower alkenyl, lower alkanoyl, or, if linked to a nitrogen atom, then taken together with the nitrogen atom, forms an azetidine ring or a 5-6 membered heterocyclic ring containing one or more heteroatoms such as N, O, S;
  • R 2 is hydrogen, halogen, preferably chloro, alkyl, alkenyl, alkynyl, substituted alkyl, substituted alkenyl, substituted alkynyl, aralkyl, substituted aralkyl, aryl, substituted aryl, heteroaryl or substituted heteroaryl;
  • R 3 is furan, pyrrole, thiophene, benzofuran, indole, benzothiophene, optionally substituted with one or more substituents as described herein for substituted heteroaryl rings;
  • X iS O 5 S 5 Or NR 1 ; and b is 1 or 2.
  • the compound is of the formula IID as described above, with the proviso that R 2 is not halogen when R is — C(X)R 1 .
  • a preferred R group is — C(X)- N(R 1 ⁇ 5 where X is O.
  • R 1 is hydrogen; C 1-8 alkyl; C 2-7 alkenyl, C 2-7 alkynyl; C 3-7 cycloalkyl; Ci .5 alkyl substituted with one or more halogen atoms, hydroxy groups, C 1-4 alkoxy, C 3-7 cycloalkyl or groups of formula — NR 32 , — C(O)NR 32 ; aryl, substituted aryl wherein the substitution is selected from the group consisting of C M alkoxy, C 1-4 alkyl, nitro, amino, cyano, C 1-4 haloalkyl, C 1-4 haloalkoxy, carboxy, carboxyamido; C 7-10 aralkyl in which the aryl moiety can be substituted with one or more of the substituents indicated above for the aryl group; a group of formula —
  • Het is a 5-6 membered aromatic or non aromatic heterocyclic ring containing one or more heteroatoms selected from the group consisting of N, O, and S and m is an integer from 1 to 5;
  • Ci -8 alkyl groups are methyl, ethyl, propyl, butyl and isopentyl.
  • Examples OfC 3-7 cycloalkyl groups include cyclopropyl, cyclopentyl, and cyclohexyl.
  • Examples Of Ci -5 alkyl groups substituted with C 3-7 cycloalkyl groups include cyclohexylmethyl, cyclopentylmethyl, and 2- cyclopentylethyl.
  • substituted C 1-5 alkyl groups include 2-hydroxyethyl, 2- methoxyethyl, trifluoromethyl, 2-fluoroethyl, 2-chloroethyl, 3-aminopropyl, 2-(4methyl-l- piperazine)ethyl, 2-(4-morpholinyl)ethyl, 2-aminocarbonyIethyl, 2-dimethylaminoethyl, 3- dimethylaminopropyl.
  • Aryl is preferably phenyl, optionally substituted with Cl, F, methoxy, nitro, cyano, methyl, trifluoromethyl, difluoromethoxy groups.
  • Examples of 5 to 6 membered ring heterocyclic groups containing N, O and/or S include piperazinyl, morpholinyl, thiazolyl, pyrazolyl, pyridyl, furyl, thienyl, pyrrolyl, triazolyl, tetrazolyl.
  • Examples of C 7- io aralkyl groups comprise benzyl or phenethyl optionally substituted by one or more substituents selected from Cl, F, methoxy, nitro, cyano, methyl, trifluoromethyl, and difluoromethoxy.
  • R 1 is hydrogen, Ci -8 alkyl, aryl or C 7- io aralkyl, optionally substituted, preferably with halogen atoms.
  • X is O
  • R 2 is chloro
  • Particularly preferred compounds are those in which R is a phenethyl group in which the phenyl ring is substituted with one or more substituents selected from the group consisting of chlorine, fluorine atoms, methoxy, nitro, cyano, methyl, trifluoromethyl, and difluoromethoxy groups.
  • Non-limiting examples of compounds of the formula HD include compounds listed below in Table.
  • Compounds of the general formulas HA, IIB and HC can be prepared, for example, as described in U.S. Patent No.'s 6,407,236; 6,448,253 and U.S. Patent Application Serial No. 10/134,219, filed April 26, 2002; the disclosures of which are hereby incorporated by reference in their entireties.
  • Compounds of the general formula HD can be prepared, for example, as described in U.S. Patent No. 6,358,964; the disclosure of which is hereby incorporated by reference in its entirety.
  • certain synthetic intermediates used to prepare compounds used in the invention may contain reactive moeities.
  • reactive and fragile functional groups often must be protected prior to a particular reaction, or sequence of reactions, and then restored to their original forms after the last reaction is completed.
  • groups are protected by converting them to a relatively stable derivative. For example, a hydroxyl group may be converted to an ether group and an amine group converted to an amide or carbamate.
  • the present invention relates to methods for the treatment, prevention, and/or management of diseases or disorders associated with overexpression of HIF-I ⁇ and/or increased HIF-I ⁇ activity (e.g., cancer, respiratory disorders such as asthma and obstructive pulmonary disorders) by using A 3 receptor antagonists alone or in combination with Ai receptor antagonists.
  • diseases or disorders associated with overexpression of HIF-I ⁇ and/or increased HIF-I ⁇ activity e.g., cancer, respiratory disorders such as asthma and obstructive pulmonary disorders
  • compositions of the invention comprising A 3 receptor antagonists are particularly useful when the levels of HIF-I ⁇ expression and/or activity are elevated above the standard or background level, as determined using methods known to those skilled in the art and dislosed herein.
  • elevation of the measured level relative to a standard level may be any statistically significant detectable elevation.
  • Such an elevation of HIF-I ⁇ expression and/or activity may include, but is not limited to about a 10%, about a 20%, about a 40%, about an 80%, about a 2-fold, about a 4-fold, about a 10- fold, about a 20-fold, about a 50-fold, about a 100-fold, about a 2 to 20 fold, 2 to 50 fold, 2 to 100 fold, 20 to 50 fold, 20 to 100 fold, elevation, relative to the standard.
  • the term "about” as used herein, refers to levels of elevation of the standard numerical value plus or minus 10% of the numerical value.
  • the therapeutic methods of the invention comprising administering a therapeutically effective amount of an A 3 receptor antagonist of the invention improve the therapeutic efficacy of treatment for diseases or disorders associated with overexpression of HIF- l ⁇ and/or increased HIF- l ⁇ activity (e.g., cancer, respiratory disorders such as asthma and obstructive pulmonary disorders) relative to the traditional modes of such therapies.
  • the methods of the invention reduce the HIF- l ⁇ level to the background level within 1 day, 1 week, 1 month, at least 2 months of the commencement of the therapeutic regime.
  • the methods of the invention result in a reduction of an HIF-I ⁇ level to the background level.
  • the invention encompasses reduction of the HIF- l ⁇ level to a level which is within about 10%, about 20%, about 30%, about 40%, about 50% of the background level.
  • the invention encompasses a method for treatment, prevention and/or management of diseases or disorders associated with overexpression of HIF-I ⁇ and/or increased HIF- l ⁇ activity (e.g., cancer, respiratory disorders such as asthma and obstructive pulmonary disorders) comprising administering a therapeutically and/or prophylactically effective amount of an A3 receptor antagonist compound as disclosed herein.
  • Compounds of the present invention that function as prophylactic and/or therapeutic agents of a disease or disorder can be administered to an animal, preferably a mammal, and most preferably a human, to treat, prevent or ameliorate one or more symptoms associated with the disease or disorder.
  • the subject is preferably a mammal such as non-primate (e.g., cows, pigs, horses, cats, dogs, rats, etc.) and a primate (e.g., monkey, such as a cynomolgous monkey and a human).
  • the subject is a human.
  • Compounds of the invention can be administered in combination with one or more other prophylactic and/or therapeutic agents useful in the treatment, prevention or management of a HIF-I ⁇ -mediated disorders, which are improved or ameliorated by modulation of HIF-I expression or activities.
  • the amount of the compounds of the invention (including adenosine A 1 and/or A 3 antagonists) required to be effective will, of course, vary with the individual mammal being treated and is ultimately at the discretion of the medical or veterinary practitioner.
  • the factors to be considered include the condition being treated, the route of administration, the nature of the formulation, the mammal's body weight, surface area, age and general condition, and the particular compound to be administered.
  • a suitable effective dose is in the range of about 0.1 ⁇ g/kg to about 100 mg/kg, about 0.1 ⁇ g/kg to about 500 mg/kg, about 0.1 ⁇ g/kg to about lg/kg, about 100 ug/kg to about 500 mg/kg, about 100 ug/kg to aboutlg/kg, about 1 mg/kg to about 100 mg/kg, about 1 mg/kg to about 500 mg/kg, about 1 mg/kg to about lg/kg of the patient's body weight.
  • the total daily dose may be given as a single dose, multiple doses, e.g., two to six times per day, or by intravenous infusion for a selected duration. Dosages above or below the range cited above are within the scope of the present invention and may be administered to the individual patient if desired and necessary.
  • compositions of the invention comprise the administration of one or more compounds of the invention to subjects/patients suffering from or expected to suffer from a disease or disorder.
  • the invention encompasses combination therapies by administration of one or more compounds of the invention in combination with administration of one or more other therapies that are traditionally used for the treatment and/or prevention of the particular disease or disorder being treated or prevented.
  • the A 3 and/or A 1 receptor antagonists of the invention may be administered in combination with one or more cancer therapies such as, but not limited to, chemotherapies, radiation therapies, hormonal therapies, and/or biological therapies/immunotherapies.
  • Prophylactic and therapeutic compounds that may be used in the methods and compositions of the invention include, but are not limited to, proteinaceous molecules, including, but not limited to, peptides, polypeptides, proteins, including post-translationally modified proteins, antibodies, etc.; small molecules (less than 1000 daltons), inorganic or organic compounds; nucleic acid molecules including, but not limited to, double-stranded or single-stranded DNA, double-stranded or single-stranded RNA, as well as triple helix nucleic acid molecules.
  • Prophylactic and therapeutic compounds can be derived from any known organism (including, but not limited to, animals, plants, bacteria, fungi, and protista, or viruses) or from a library of synthetic molecules, hi certain embodiments, one or more compounds of the invention are administered to a mammal, preferably a human, concurrently with one or more other therapeutic agents useful for the treatment of cancer or a disorder.
  • the term "concurrently” is not limited to the administration of prophylactic or therapeutic agents at exactly the same time, but rather it is meant that compounds of the invention and the other agent are administered to a subject in a sequence and within a time interval such that the compounds of the invention can act together with the other agent to provide an increased benefit than if they were administered otherwise.
  • each prophylactic or therapeutic agent may be administered at the same time or sequentially in any order at different points in time; however, if not administered at the same time, they should be administered sufficiently close in time so as to provide the desired therapeutic or prophylactic effect.
  • Each therapeutic agent can be administered separately, in any appropriate form and by any suitable route.
  • the prophylactic or therapeutic agents are administered less than 1 hour apart, at about 1 hour apart, at about 1 hour to about 2 hours apart, at about 2 hours to about 3 hours apart, at about 3 hours to about 4 hours apart, at about 4 hours to about 5 hours apart, at about 5 hours to about 6 hours apart, at about 6 hours to about 7 hours apart, at about 7 hours to about 8 hours apart, at about 8 hours to about 9 hours apart, at about 9 hours to about 10 hours apart, at about 10 hours to about 11 hours apart, at about 11 hours to about 12 hours apart, no more than 24 hours apart or no more than 48 hours apart, hi preferred embodiments, two or more components are administered within the same patient visit.
  • the dosage amounts and frequencies of administration provided herein are encompassed by the terms therapeutically effective and prophylactically effective.
  • the dosage and frequency further will typically vary according to factors specific for each patient depending on the specific therapeutic or prophylactic agents administered, the severity and type of cancer, the route of administration, as well as age, body weight, response, and the past medical history of the patient. Suitable regimens can be selected by one skilled in the art by considering such factors and by following, for example, dosages reported in the literature and recommended in the Physician 's Desk Reference (58 th ed.,
  • Compounds of the invention can be used alone or in combination with other anti-cancer agents, e.g., therapeutic antibodies known in the art to prevent, inhibit or reduce the growth of primary tumors or metastasis of cancerous cells.
  • a compound of the invention when administered alone or in combination with an anti-cancer agent, inhibits or reduces the growth of primary tumor or metastasis of cancerous cells by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10% relative to the growth of primary tumor or metastasis in absence of said compound of the invention.
  • compounds of the invention particularly the A 1 and A 3 receptor antagonists in combination with an anti-cancer agent inhibit or reduce the growth of primary tumor or metastasis of cancer by at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, at least 50%, at least 45%, at least 40%, at least 45%, at least 35%, at least 30%, at least 25%, at least 20%, or at least 10% relative to the growth or metastasis in absence of said compounds.
  • the methods of the invention are particularly useful for the treatment of solid tumors, hypoxic tumors, A 3 expressing tumors and HIF- l ⁇ expressing tumors.
  • the compounds of the invention particularly the A 1 and A 3 receptor antagonists can be used to treat solid tumors.
  • Solid tumors which can be treated in accordance with the methods of the invention include without limitation fibrodysplasia ossificans progressiva, prostate cancer, benign prostatic hyperplasia, recessive dystrophic epidermolysis bullosa (RDEB), Lewis lung carcinoma, breast cancer, and brain tumors.
  • RDEB recessive dystrophic epidermolysis bullosa
  • a 3 expressing cancers refers to human cancers that express the adenosine A 3 receptor or that otherwise comprise elevated concentrations of adenosine A 3 receptors. Elevated concentration is determined by comparison to normal, non-cancerous tissues of a similar cell type.
  • a 3 expressing cancers include, without limitation, human leukemia, melanoma, pancreatic carcinoma, breast carcinoma, prostrate carcinoma, colon carcinoma, lung carcinomamalignant melanomas, histiocytic lymphoma, and some forms of astrocytoma cells.
  • HIF- l ⁇ expressing cancers refers to human cancer that express HIF-I ⁇ or that otherwise comprise elevated concentrations of HIF-I ⁇ . Elevated concentration is determined by comparison to normal, non-cancerous tissues of a similar cell type.
  • HIF-I ⁇ expressing cancers include without limitation cervical cancer (early stage), lung cancer (non-small cell lung carcinoma), breast cancer (including lymph node positive breast cancer and lymph node negative breast cancer), oligodendroglioma, orpharyngeal squamous cell carcinoma, ovarian cancer, oesophageal cancer, endometrial cancer, head and neck cancer, gastrointestinal stromal tumor of the stomach.
  • the methods and compositions of the invention comprise the administration of one or more A 3 receptor antagonist compounds of the invention (alone or in combination with other anti-cancer agents) to subjects/patients suffering from or expected to suffer from cancer, e.g., have a genetic predisposition for a particular type of cancer, have been exposed to a carcinogen, or are in remission from a particular cancer.
  • the methods and compositions of the invention may be used as a first line or second line cancer treatment. Included in the invention is also the treatment of patients undergoing other cancer therapies and the methods and compositions of the invention can be used before any adverse effects or intolerance of these other cancer therapies occurs.
  • the invention also encompasses methods for administering one or more A 3 receptor antagonist compounds of the invention to treat or ameliorate symptoms in refractory patients.
  • that a cancer is refractory to a therapy means that at least some significant portion of the cancer cells are not killed or their cell division arrested.
  • the determination of whether the cancer cells are refractory can be made either in vivo or in vitro by any method known in the art for assaying the effectiveness of treatment on cancer cells, using the art-accepted meanings of "refractory" in such a context.
  • a cancer is refractory where the number of cancer cells has not been significantly reduced, or has increased.
  • the invention also encompasses methods for administering one or more A 3 receptor antagonist compounds of the invention to prevent the onset or recurrence of cancer in patients predisposed to having cancer.
  • the A 3 receptor antagonists of the invention are administered to reverse resistance or reduced sensitivity of cancer cells to certain hormonal, radiation and chemotherapeutic agents thereby resensitizing the cancer cells to one or more of these agents, which can then be administered (or continue to be administered) to treat or manage cancer, including to prevent metastasis.
  • the invention provides methods for treating cancer in a subject by administering one or more A 3 receptor antagonists of the invention in combination with any other treatment or to patients who have proven refractory to other treatments but are no longer on these treatments.
  • the patients being treated by the methods of the invention are patients already being treated with chemotherapy, radiation therapy, hormonal therapy, or biological therapy/immunotherapy.
  • these patients are refractory patients and those with cancer despite treatment with existing cancer therapies, hi other embodiments, the patients have been treated and have no disease activity and one or more A 3 receptor antagonist compounds of the invention are administered to prevent the recurrence of cancer.
  • the invention also provides methods of treatment of cancer as an alternative to chemotherapy, radiation therapy, hormonal therapy, and/or biological therapy/immunotherapy where the therapy has proven or may prove too toxic, i.e., results in unacceptable or unbearable side effects, for the subject being treated.
  • the subject being treated with the methods of the invention may, optionally, be treated with other cancer treatments such as surgery, chemotherapy, radiation therapy, hormonal therapy or biological therapy, depending on which treatment was found to be unacceptable or unbearable.
  • the invention provides administration of one or more A 3 receptor antagonist compounds of the invention without any other cancer therapies for the treatment of cancer, but who have proved refractory to such treatments.
  • patients refractory to other cancer therapies are administered one or more A 3 receptor antagonist compounds in the absence of cancer therapies.
  • a high affinity adenosine A 3 receptor antagonist and a chemotherapeutic cancer agent are administered to the patient.
  • the combination therapy enhances the effect of the chemotherapeutic cancer agent and prevents multi-drug resistance from developing.
  • the chemotherapeutic cancer agents showing desirable response to the present invention are typical of agents noted for developing P-gp or MRP class multi-drug resistance, including, without limitation, taxane compounds, vinca alkaloids, camptothecins and antibiotics useful as chemotherapeutic agents.
  • the invention encompasses combination therapies for treating cancers that have already developed multi-drug resistance.
  • the high affinity adenosine A 3 receptor antagonist counters the existing MDR while further enhancing the effect of the chemotherapeutic cancer agent.
  • the present invention is not effective for all forms of MDR cancers.
  • the MDR cancers showing desirable response to the present invention are in the P-gp and MRP classes.
  • the high affinity adenosine A 3 receptor antagonist is preferably administered either before or during administration of the taxane compound.
  • this is done to reduce the incidence of hypersensitivity to the taxane agent.
  • Commercially available compounds of the taxane family are paclitaxel (commercially available under the tradename TAXOL from Bristol-Myers Squibb Company, Princeton, NY and as a generic drug from IVAX Corp, Miami, FL) and docetaxel (commercially available under the tradename TAXOTERE from Aventis Pharmaceuticals, Collegeville, PA), which are encompassed in the instant invention.
  • the composition comprises an effective amount of an adenosine A 3 receptor antagonist and a chemotherapeutic agent that is a taxane family compound, for example paclitaxel or docetaxel to inhibit tumor growth.
  • a chemotherapeutic agent that is a taxane family compound, for example paclitaxel or docetaxel to inhibit tumor growth.
  • Paclitaxel is commercially available under the tradename TAXOL from Bristol-Myers Squibb Company, Princeton, NY and as a generic drug from IVAX Corp, Miami, FL.
  • Docetaxel is commercially available under the tradename TAXOTERE from Aventis Pharmaceuticals, Collegeville, PA.
  • the A 3 antagonist may reduce the growth rate of cancerous cells, interfere with adenosine protective effects against hypoxia, enhance the chemotherapeutic effect of the taxane, reduce hypersensitivity reactions in the patient and counter development of multi- drug resistance.
  • Cancers and related disorders that can be treated or prevented by methods and compositions comprising A 1 and/or A 3 receptor antagonists of the present invention include, but are not limited to, the following: Leukemias including, but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblasts, promyelocyte, myelomonocytic, monocytic, erythroleukemia leukemias and myelodysplastic syndrome, chronic leukemias such as but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, hairy cell leukemia; polycythemia vera; lymphomas such as but not limited to Hodgkin's disease, non-Hodgkin's disease; multiple myelomas such as but not limited to smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myelom
  • cancers include myxosarcoma, osteogenic sarcoma, endotheliosarcoma, lymphangioendotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma, cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma and papillary adenocarcinomas (for a review of such disorders, see Fishman et ah, 1985, Medicine, 2d Ed., J.B. Lippincott Co., Philadelphia and Murphy et al, 1997, Informed Decisions: The Complete Book of Cancer Diagnosis, Treatment, and Recovery, Viking Penguin, Penguin Books U.S.A., Inc., United States of America).
  • carcinoma including that of the bladder, breast, colon, kidney, liver, lung, ovary, pancreas, stomach, cervix, thyroid and skin; including squamous cell carcinoma; hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Berketts lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyoscarcoma; other tumors, including melanoma, seminoma, tetratocarcinoma, neuroblastom
  • cancers caused by aberrations in apoptosis would also be treated by the methods and compositions of the invention.
  • Such cancers may include but not be limited to follicular lymphomas, carcinomas with p53 mutations, hormone dependent tumors of the breast, prostate and ovary, and precancerous lesions such as familial adenomatous polyposis, and myelodysplastic syndromes.
  • malignancy or dysproliferative changes (such as metaplasias and dysplasias), or hyperproliferative disorders, are treated or prevented by the methods and compositions of the invention in the ovary, bladder, breast, colon, lung, skin, pancreas, or uterus.
  • sarcoma, melanoma, or leukemia is treated or prevented by the methods and compositions of the invention.
  • Cancers associated with the cancer antigens may be treated or prevented by administration of the A 3 receptor antagonists of the invention in combination with an antibody that binds the cancer antigen and is cytotoxic.
  • the A 3 receptor antagonists of the invention enhance the antibody mediated cytotoxic effect of the antibody directed at the particular cancer antigen.
  • cancers associated with the following cancer antigen may be treated or prevented by the methods and compositions of the invention.
  • KS 1/4 pan-carcinoma antigen Perez and Walker, 1990, J. Immunol. 142:32-37; Bumal, 1988, Hybridoma 7(4):407-415
  • ovarian carcinoma antigen CA125
  • prostatic acid phosphate (Tailor et al , 1990, Nucl. Acids Res. 18(1):4928), prostate specific antigen (Henttu and Vihko, 1989, Biochem. Biophys. Res. Comm. 10(2):903-910; Israeli et al, 1993, Cancer Res. 53:227-230), melanoma-associated antigen p97 (Estin et al, 1989, J Natl. Cancer Instil 81(6):445-44), melanoma antigen gp75 (Vijayasardahl et al, 1990, J. Exp. Med.
  • HMW-MAA high molecular weight melanoma antigen
  • CEA carcinoembryonic antigen
  • CEA polymorphic epithelial mucin antigen
  • human milk fat globule antigen Colorectal tumor-associated antigens such as: CEA, TAG-72 (Yokata et al, 1992, Cancer Res.
  • melanoma specific antigens such as ganglioside GD2 (Saleh et al, 1993, J.Immunol., 151, 3390-3398), ganglioside GD3 (Shitara et al,
  • ganglioside GM3 Hoon et ⁇ /., 1993, Cancer Res. 53:5244-5250
  • tumor-specific transplantation type of cell-surface antigen TSTA
  • virally-induced tumor antigens including T-antigen DNA tumor viruses and envelope antigens of RNA tumor viruses
  • oncofetal antigen-alpha-fetoprotein such as CEA of colon
  • bladder tumor oncofetal antigen Hellstrom et al, 1985, Cancer. Res. 45:2210-2188
  • differentiation antigen such as human lung carcinoma antigen L6, L20 (Hellstrom et al, 1986, Cancer Res.
  • antigens of fibrosarcoma human leukemia T cell antigen-Gp37 (Bhattacharya-Chatterjee et ⁇ /., 1988, J oflmmun. 141:1398-1403), neoglycoprotein, sphingolipids, breast cancer antigen such as EGFR (Epidermal growth factor receptor), HER2 antigen (pi 85 ⁇ 112 ), polymorphic epithelial mucin (PEM) (Hilkens et al, 1992, Trends in Bio. Chem. Sci.
  • PEM polymorphic epithelial mucin
  • malignant human lymphocyte antigen-APO-1 (Bernhard et al, 1989, Science 245:301-304), differentiation antigen (Feizi, 1985, Nature 314:53-57) such as I antigen found in fetal erthrocytes and primary endoderm, I(Ma) found in gastric adencarcinomas, M 18 and M39 found in breast epithelium, SSEA-I found in myeloid cells, VEP8, VEP9, MyI, VIM-D5,and D 2 56-22 found in colorectal cancer, TRA-I- 85 (blood group H), C14 found in colonic adenocarcinoma, F3 found in lung adenocarcinoma, AH6 found in gastric cancer, Y hapten, Le y found in embryonal carcinoma cells, TL5 (blood group A), EGF receptor found in A431 cells , E 1 series (blood group B) found in pancreatic cancer, FCl 0.2 found in embryonal carcinoma cells
  • the high affinity adenosine A 3 receptor antagonists of the invention can be administered alone or in combination with other therapeutic and/or prophylactic agents.
  • the methods of the invention encompass the administration of one or more chemotherapeutic cancer agents.
  • Chemotherapeutic cancer agents are defined as agents that attack and kill cancer cells.
  • Chemotherapeutic agents that may be used inthe methods of the invention include taxane compounds and derivatives thereof, paclitaxel and its derivatives or docetaxel and its derivatives. Additional taxane derivatives and methods of synthesis are disclosed in U.S. Patent No. 6,191,287 to Holton et al, U.S. Patent No. 5,705,508 to Ojima et al, U.S. Patent Nos. 5,688,977 and 5,750,737 to Sisti et al, U.S. Patent No. 5,248,796 to Chen et al, U.S. Patent No. 6,020,507 to Gibson et al., U.S. Patent No. 5,908,835 to Bissery, all of which are incorporated herein by reference in their entireties.
  • chemotherapeutic cancer agents include topoisomerase I inhibitors, such as camptothecin compounds.
  • camptothecin compounds include CamptosarTM (irinotecan HCL), HycamtinTM (topotecan HCL) and other compounds derived from camptothecin and its analogues.
  • chemotherapeutic cancer agents that may be used in the methods and compositions of the invention are podophyllotoxin derivatives, such as etoposide, teniposide and mitopodozide.
  • the invention further encompasses other chemotherapeutic cancer agents known as alkylating agents, which alkylate the genetic material in tumor cells. These include without limitation cisplatin, cyclophosphamide, nitrogen mustard, trimethylene thiophosphoramide, carmustine, busulfan, chlorambucil, belustine, uracil mustard, chlornaphazin, and dacarbazine.
  • the invention encompasses antimetabolites as chemotherapeutic agents.
  • chemotherapeutic cancer agents examples include cytosine arabinoside, fluorouracil, methotrexate, mercaptopurine, azathioprime, and procarbazine.
  • An additional category of chemotherapeutic cancer agents that may be used in the methods and compositions of the invention include antibiotics. Examples include without limitation doxorubicin, bleomycin, dactinomycin, daunorubicin, mithrarnycin, mitomycin, mytomycin C, and daunomycin. There are numerous liposomal formulations commercially available for these compounds.
  • the invention further encompasses other chemotherapeutic cancer agents including without limitation anti-tumor antibodies, dacarbazine, azacytidine, amsacrine, melphalan, ifosfamide and mitoxantrone.
  • chemotherapeutic cancer agents including without limitation anti-tumor antibodies, dacarbazine, azacytidine, amsacrine, melphalan, ifosfamide and mitoxantrone.
  • the A 3 antagonists can be administered alone or in combination with other anti-tumor agents, including cytotoxic/antineoplastic agents and anti-angiogenic agents. Cytotoxic/anti-neoplastic agents are defined as agents which attack and kill cancer cells.
  • cytotoxic/anti-neoplastic agents are alkylating agents, which alkylate the genetic material in tumor cells, e.g., cis-platin, cyclophosphamide, nitrogen mustard, trimethylene thiophosphoramide, carmustine, busulfan, chlorambucil, belustine, uracil mustard, chlornaphazin, and dacabazine.
  • Other cytotoxic/anti-neoplastic agents are antimetabolites for tumor cells, e.g., cytosine arabinoside, fluorouracil, methotrexate, mercaptopuirine, azathioprime, and procarbazine.
  • cytotoxic/anti-neoplastic agents are antibiotics, e.g., doxorubicin, bleomycin, dactinomycin, daunorubicin, mithramycin, mitomycin, mytomycin C, and daunomycin. There are numerous liposomal formulations commercially available for these compounds. Still other cytotoxic/anti-neoplastic agents are mitotic inhibitors (vinca alkaloids). These include vincristine, vinblastine and etoposide.
  • Miscellaneous cytotoxic/anti-neoplastic agents include taxol and its derivatives, L-asparaginase, anti-tumor antibodies, dacarbazine, azacytidine, amsacrine, melphalan, VM-26, ifosfamide, mitoxantrone, and vindesine.
  • Anti-angiogenic agents are well known to those of skill in the art. Suitable anti-angiogenic agents for use in the methods and compositions of the invention include anti-VEGF antibodies, including humanized and chimeric antibodies, anti-VEGF aptamers and antisense oligonucleotides. Other known inhibitors of angiogenesis include angiostatin, endostatin, interferons, interleukin 1 (including ⁇ and ⁇ ) interleukin 12, retinoic acid, and tissue inhibitors of metalloproteinase-1 and -2. (TIMP-I and -T). Small molecules, including topoisomerases such as razoxane, a topoisomerase II inhibitor with anti- angiogenic activity, can also be used.
  • the anti-angiogenesis compound is an adenosine A 2a antagonist.
  • a 2a antagonists are well known to those of skill in the art. Examples of these compounds include SCH 58261 from Schering Plough, ZM241385 (Palmer et al., MoI. Pharmacol., 48:970-974 (1995), the tricyclic non-xanthine antagonists and the triazoloquinazolines, including CP 66,713, disclosed inNikodijevic et al., J. Pharmacol. Exp. Ther., 259:286-294 (1991), as well as the compounds disclosed in von Lubitz et al., Eur. J. Pharmacol, 287:295-302 (1995). The contents of each of the references above are hereby incorporated by reference.
  • Anti-cancer agents that can be used in combination with A 3 receptor antagonists of the invention in the various embodiments of the invention, including pharmaceutical compositions and dosage forms and kits of the invention, include, but are not limited to: acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer
  • anti-cancer drugs include, but are not limited to: 20-epi-l,25 dihydroxyvitamin D3; 5-etbynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein- 1 ; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-
  • B betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; canarypox IL-2; capecitabine; carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorlns; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A;
  • Preferred additional anti-cancer drugs are 5-fluorouracil and leucovorin.
  • therapeutic antibodies that can be used in methods of the invention include but are not limited to HERCEPTIN® (Trastuzumab) (Genentech, CA) which is a humanized anti-HER2 monoclonal antibody for the treatment of patients with metastatic breast cancer; REOPRO® (abciximab) (Centocor) which is an anti-glycoprotein Ilb/IIIa receptor on the platelets for the prevention of clot formation; ZENAP AX® (daclizumab) (Roche Pharmaceuticals, Switzerland) which is an immunosuppressive, humanized anti-CD25 monoclonal antibody for the prevention of acute renal allograft rejection; PANOREXTM which is a murine anti-17-IA cell surface antigen IgG2a antibody (Glaxo Wellcome/Centocor); BEC2 which is a murine anti-idiotype (GD3 epitope) IgG antibody (I
  • MAb-VEGF NSCLC metastatic VEGF
  • IDEC Zevahn (Rituxan + yttrium-90) low grade of follicular, CD 20 relapsed or refractory, CD20-positive, B-cell NHL and Rituximab- refractory NHL
  • Cetuximab + cisplatin head & neck cancer EGF receptor extendensive incurable local-regional disease & distant metasteses
  • IMC-ICl 1 colorectal cancer with VEGF-receptor liver metasteses
  • CEA-Cide metastatic solid tumors CEA
  • CEA-Scan Tc-99m-labeled colorectal cancer CEA arcitumomab (radioimaging)
  • CEA-Scan Tc-99m-labeled Breast cancer CEA arcitumomab (radioimaging)
  • CEA-Scan Tc-99m-labeled lung cancer CEA arcitumomab (radioimaging)
  • CEA-Scan Tc-99m-labeled intraoperative tumors CEA arcitumomab (radio imaging) Company Product Disease Target
  • LymphoScan Tc-99m-labeled lymphomas CD22 (radioimaging)
  • CTLA-4 Medarex MDX-101 (CTLA-4) Prostate and other CTLA-4 cancers
  • NeoRx CD20-streptavidin (+ biotin-yttrium Non-Hodgkins CD20 90) lymphoma
  • MAb lung/kidney cancer lung & kidney cancer NA nacolomab tafenatox (C242 + colon & pancreatic NA staphylococcal enterotoxin) cancer
  • the A 3 receptor antagonists of the invention may be used in combination with one or more ATP depleting agents for enhancing the therapeutic efficacy of treating a disease or disorder as disclosed herein, i.e., cancer.
  • the A 3 receptor antagonists of the invention may be used with verapamil for enhancing the therapeutic efficacy of conventional modes of treating a disease or disorder as discussed herein, i.e., cancer.
  • Compounds of the invention can be used alone or in combination with other therapeutic and/or prophylactic agents to prevent, treat or manage respiratory disorders such as asthma and obstructive pulmonary disorders (COPD).
  • COPD obstructive pulmonary disorders
  • the methods and compositions comprising A 1 and/or A 3 receptor antagonists of the invention are effective for treatment, prevention, and/or management of asthma.
  • the therapeutic and prophylactic methods of the invention for asthma may be used in combination with other methods known in the art for the treatment, prevention and/or management of asthma including but not limited to inhaled beta 2 agonists, inhaled corticosteroids, retinoic acid, anti-IgE antibodies, phosphodiesterase inhibitors, leukotriene antagonists, anti IL-9 antibody, and/or anti-mucin therapies (e.g., anti hCLCAl therapy such as LomucinTM).
  • ⁇ -adrenergic drugs e.g.
  • epinephrine and isoproterenol theophylline, anticholinergic drugs (e.g., atropine and ipratorpium bromide), and corticosteroids, adrenergic stimulants (e.g., catecholamines (e.g., epinephrine, isoproterenol, and isoetharine), resorcinols (e.g., metaproterenol, terbutaline, and fenoterol), and saligenins (e.g., salbutamol), other steroids, immunosuppressant agents (e.g., methotrexate and gold salts), mast cell modulators (e.g., cromolyn sodium (INTALTM) and nedocromil sodium (TILADETM)), and mucolytic agents (e.g., acetylcysteine)).
  • adrenergic stimulants e.g., catecholamines
  • the therapeutic and prophylactic methods of the invention may be used in combination with other therapeutic methods known in the art for the treatment, prevention and/or management of COPD including but not limited tiotropium and/or ipratropium
  • respiratory disorders that may be treated, prevented or managed using the methods and compositions of the invention include without limitation lung fibrosis, bronchial hyper responsiveness. 6.4 COMPOSITIONS AND METHODS OF ADMINISTERING
  • the invention provides methods and pharmaceutical compositions comprising compounds of the invention.
  • the invention also provides methods of treatment, prophylaxis, and amelioration of one or more symptoms associated with a disease, disorder or cancer by administering to a subject an effective amount of compound of the invention.
  • the subject is an animal, preferably a mammal such as non- primate (e.g., cows, pigs, horses, cats, dogs, rats etc.) and a primate (e.g., monkey such as, a cynomolgous monkey and a human).
  • the subject is a human.
  • compositions of the invention include bulk drug compositions useful in the manufacture of pharmaceutical compositions (e.g., impure or non-sterile compositions) and pharmaceutical compositions (i.e., compositions that are suitable for administration to a subject or patient) which can be used in the preparation of unit dosage forms.
  • Such compositions comprise a prophylactically or therapeutically effective amount of a prophylactic and/or therapeutic agent disclosed herein or a combination of those agents and a pharmaceutically acceptable carrier.
  • compositions of the invention comprise a prophylactically or therapeutically effective amount of compounds of the invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises a therapeutically or prophylactically effective amount of an A 3 or A 1 receptor antagonist and a pharmaceutically acceptable carrier.
  • said pharmaceutical composition further comprises one or more anti-cancer agents.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete), excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • compositions can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the ingredients of compositions of the invention are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • compositions of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include, but are not limited to those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with captions such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the compounds described above are preferably administered in formulations including an active compound, i.e., an adenosine Aj and/or A 3 receptor antagonist, together with an acceptable carrier for the mode of administration.
  • suitable pharmaceutically acceptable carriers are known to those of skill in the art.
  • the compositions can optionally include other therapeutically active agents, including anti-angiogenic and anti-cancer agents.
  • Other optional ingredients include antivirals, antibacterials, antiinflammatories, analgesics, and immunosuppresants.
  • the carrier must be pharmaceutically acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • the compounds of the invention can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol ⁇ e.g., ethanol, isopropanol, or hexadecyl alcohol), glycols ⁇ e.g., propylene glycol or polyethylene glycol), glycerol ketals, ⁇ e.g., 2,2- dimethyll,3-dioxolane-4-methanol), ethers, ⁇ e.g., poly(ethyleneglycol) 400), an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant (e.g., a soap or a detergent),
  • a pharmaceutically acceptable surfactant e.g., a soap or
  • Formulations suitable for parenteral administration conveniently include sterile aqueous preparation of the active compound which is preferably isotonic with the blood of the recipient.
  • Such formulations may conveniently contain distilled water, 5% dextrose in distilled water or saline.
  • Useful formulations also include concentrated solutions or solids containing the compounds which upon dilution with an appropriate solvent give a solution suitable for parental administration above.
  • Formulations suitable for parenteral administration include but are not limited to aqueous and non-aqueous solutions, isotonic sterile injection solutions, which may comprise anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that may comprise suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the parenteral formulations will typically contain from about 0.5 to about
  • compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17.
  • HLB hydrophile-lipophile balance
  • Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water, for injections, immediately prior to use.
  • sterile liquid carrier for example, water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • the compounds of the inventione, e.g., high affinity adenosine A 3 receptor antagonists may be made into injectable formulations.
  • the requirements for effective pharmaceutical carriers for injectable compositions are well known to those of ordinary skill in the art. See Pharmaceutics and Pharmacy Practice, J. B.
  • the compound can be incorporated into an inert carrier in discrete units such as capsules, cachets, tablets or lozenges, each containing a predetermined amount of the active compound; as a powder or granules; or a suspension or solution in an aqueous liquid or non-aqueous liquid, e.g., a. syrup, an elixir, an emulsion or a draught.
  • Suitable carriers may be starches or sugars and include lubricants, flavorings, binders, and other materials of the same nature.
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions.
  • Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and cornstarch.
  • Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers.
  • Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable in a machine the active compound in a free-flowing form, e.g., a powder or granules, optionally mixed with accessory ingredients, e.g., binders, lubricants, inert diluents, surface active or dispersing agents. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered active compound with any suitable carrier.
  • a syrup or suspension may be made by adding the active compound to a concentrated, aqueous solution of a sugar, e.g., sucrose, to which may also be added any accessory ingredients.
  • Such accessory ingredients may include flavoring, an agent to retard crystallization of the sugar or an agent to increase the solubility of any other ingredient, e.g., as a polyhydric alcohol, for example, glycerol or sorbitol.
  • the compounds can also be administered locally by topical application of a solution, ointment, cream, gel, lotion or polymeric material (for example, a PluronicTM, BASF), which may be prepared by conventional methods known in the art of pharmacy.
  • a solution, ointment, cream, gel, lotion or polymeric material for example, a PluronicTM, BASF
  • topical formulations may also contain preservatives, perfumes, and additional active pharmaceutical agents.
  • Topical formulations for high affinity adenosine A 3 receptor antagonists include ointments, creams, gels and lotions that may be prepared by conventional methods known in the art of pharmacy.
  • Such topical formulation may also furhter comprise preservatives, perfumes, and additional active pharmaceutical agents.
  • Preferred additional pharmaceutical agents include the chemotherapeutic agents for cancer treatments noted to be enhanced or benefited by the A 3 receptor antagonist (for example by preventing MDR).
  • An example of a preferred topical formulation includes a high affinity adenosine A 3 receptor antagonist and a taxane family compound.
  • Oils, which can be used in parenteral formulations include petroleum, animal, vegetable, and synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral.
  • Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts
  • suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (e) mixtures thereof.
  • the compounds of the invention may be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases.
  • bases such as emulsifying bases or water-soluble bases.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
  • Formulations for rectal administration may be presented as a suppository with a conventional carrier, e.g., cocoa butter or Witepsol S55 (trademark of Dynamite Nobel Chemical, Germany), for a suppository base.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active compound into association with a carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier or a finely divided solid carrier and then, if necessary, shaping the product into desired unit dosage form.
  • the formulations of this invention may further include one or more cytotoxic agent as well as one or more optional accessory ingredient(s) utilized in the art of pharmaceutical formulations, e.g., diluents, buffers, flavoring agents, binders, surface active agents, thickeners, lubricants, suspending agents, preservatives (including antioxidants) and the like.
  • cytotoxic agent e.g., diluents, buffers, flavoring agents, binders, surface active agents, thickeners, lubricants, suspending agents, preservatives (including antioxidants) and the like.
  • Various delivery systems are known and can be used to administer a composition comprising compounds of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules.
  • the compounds of the invention are formulated in liposomes for targeted delivery of the compounds of the invention.
  • Liposomes are vesicles comprised of concentrically ordered phopsholipid bilayers which encapsulate an aqueous phase. Liposomes typically comprise various types of lipids, phospholipids, and/or surfactants. The components of liposomes are arranged in a bilayer configuration, similar to the lipid arrangement of biological membranes. Liposomes are particularly preferred delivery vehicles due, in part, to their biocompatibility, low immunogenicity, and low toxicity. Methods for preparation of liposomes are known in the art and are encompassed within the invention, see, e.g., Epstein et al, 1985, Proc.
  • the invention also encompasses methods of preparing liposomes with a prolonged serum half-life, i.e., enhanced circulation time, such as those disclosed in U.S. Patent No. 5,013,556.
  • Preferred liposomes used in the methods of the invention are not rapidly cleared from circulation, i.e., are not taken up into the mononuclear phagocyte system (MPS).
  • MPS mononuclear phagocyte system
  • sterically stabilized liposomes which are prepared using common methods known to one skilled in the art.
  • sterically stabilized liposomes contain lipid components with bulky and highly flexible hydrophilic moieties, which reduces the unwanted reaction of liposomes with serum proteins, reduces oposonization with serum components and reduces recognition by MPS.
  • Sterically stabilized liposomes are preferably prepared using polyethylene glycol.
  • the invention also encompasses liposomes that are adapted for specific organ targeting, see, e.g., U.S. Patent No. 4,544,545.
  • Particularly useful liposomes for use in the compositions and methods of the invention can be generated by reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • a fragment of an antibody of the invention may be conjugated to the liposomes using previously described methods, see, e.g., Martin et al, 1982, J Biol. Chem. 257: 286-288, which is incorporated herein by reference in its entirety.
  • Microspheres formed of polymers or proteins are well known to those skilled in the art, and can be tailored for passage through the gastrointestinal tract directly into the blood stream. Alternatively, the compound can be incorporated and the microspheres, or composite of microspheres, implanted for slow release over a period of time ranging from days to months. See, for example, U.S. Pat. Nos. 4,906,474, 4,925,673 and 3,625,214, the contents of which are hereby incorporated by reference.
  • Preferred microparticles are those prepared from biodegradable polymers, such as polyglycolide, polylactide and copolymers thereof.
  • biodegradable polymers such as polyglycolide, polylactide and copolymers thereof.
  • the compositions can be delivered in a vesicle, in particular a liposome (See Langer, Science 249:1527-1533 (1990); Treat et al, in Liposomes in the Therapy of Infectious Disease and Cancer. Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 3 17-327; see generally ibid.).
  • compositions can be delivered in a controlled release or sustained release system. Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more A3 receptor antagonists of the invention. See, e.g., U.S. Patent No.
  • a pump may be used in a controlled release system (See Langer, supra; Sefton, 1987, CRC CrU. Ref. Biomed. Eng. 14:20; Buchwald et al, 1980, Surgery 88:507; and Saudek etal, 1989, N. Engl. J. Med. 321:574).
  • polymeric materials can be used to achieve controlled release of A3 receptor antagonists (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, 1983, J., Macromol. ScL Rev. Macromol. Chem. 23:61; See also Levy et al, 1985, Science 228:190; During et al, 1989, Ann. Neurol. 25:351; Howard et al, 1989, J. Neurosurg. 1 1:105); U.S. Patent No.
  • polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters.
  • a controlled release system can be placed in proximity of the therapeutic target (e.g., the lungs), thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • polymeric compositions useful as controlled release implants are used according to Dunn et al (See U.S. 5,945,155). This particular method is based upon the therapeutic effect of the in situ controlled release of the bioactive material from the polymer system.
  • the implantation can generally occur anywhere within the body of the patient in need of therapeutic treatment, hi another embodiment, a non-polymeric sustained delivery system is used, whereby a non-polymeric implant in the body of the subject is used as a drug delivery system.
  • the organic solvent of the implant Upon implantation in the body, the organic solvent of the implant will dissipate, disperse, or leach from the composition into surrounding tissue fluid, and the non-polymeric material will gradually coagulate or precipitate to form a solid, microporous matrix (See U.S. 5,888,533). Controlled release systems are discussed in the review by Langer (1990, Science 249:1527-1533). Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more therapeutic agents of the invention.
  • Methods of administering a compound of the invention include, but are not limited to, parenteral administration (e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous), epidural, and mucosal (e.g., intranasal and oral routes).
  • parenteral administration e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous
  • epidural e.g., intranasal and oral routes
  • mucosal e.g., intranasal and oral routes
  • the compounds of the invention are administered intramuscularly, intravenously, or subcutaneously.
  • the compositions may be administered by any convenient route, for example, by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • an inhaler or nebulizer e.g., a pressurized device, a pressurized device, and a pressurized device.
  • the compounds are administered intravenously in a liposome or microparticle with a size such that the particle can be delivered intraveneously, but gets trapped in a capillary bed around a growing tumor.
  • Suitable particle sizes for this embodiment are those currently used, for example, in liposomes sold under the name DaunoXome.TM., which are believed to be between about 200 and 500 um.
  • the compounds are then released locally, over time, at the location of the tumor. This can be effective in limiting systemic effects of the administration of cytotoxic agents, and also targets the adenosine antagonist and any other anti-angio genie agents directly to the site at which blood vessel growth is to be inhibited.
  • the compounds are administered in a tissue coating, preferably a polymeric tissue coating, more preferably, a biodegradable tissue coating, which is applied to the site at which a tumor is surgically removed.
  • a tissue coating preferably a polymeric tissue coating, more preferably, a biodegradable tissue coating, which is applied to the site at which a tumor is surgically removed.
  • Suitable polymeric materials are disclosed, for example, in U.S. Pat. No. 5,410,016 to Hubbell et al., the contents of which are hereby incorporated by reference.
  • the polymeric barrier in combination with the adenosine A3 antagonists, and optionally in combination with other anti-angiogenic agents and/or cytotoxic agents, provides a physical barrier as well as a chemical barrier to further tumor growth and vasculation around the tumor.
  • the two embodiments described above can be particularly useful when cytotoxic agents with pronounced systemic side effects are used. By targeting the administration of these compounds to a specific site, and locally delivering the compounds, systemic effects can be minimized.
  • anti-VEGF antibodies can be covalently coupled to microparticles or liposomes including the compounds. The antibodies can be used to not only target the delivery of the compounds, but also to assist in inhibiting tumor growth by inhibiting angiogenesis.
  • the amount of the composition of the invention which will be effective in the treatment, prevention or amelioration of one or more symptoms associated with a disorder can be determined by standard clinical techniques.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the condition, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • compositions of the invention may be desirable to administer locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • Treatment of a subject with a therapeutically or prophylactically effective amount of compounds of the invention can include a single treatment or, preferably, can include a series of treatments.
  • a subject is treated with compounds of the invention in the range of between about 0.1 ⁇ g/kg to about 100 mg/kg, about 0.1 ⁇ g/kg to about 500 mg/kg, about 0.1 ⁇ g/kg to about lg/kg, about 100 ug/kg to about 500 mg/kg, about 100 ug/kg to aboutlg/kg, about 1 mg/kg to about 100 mg/kg, about 1 mg/kg to about 500 mg/kg, about 1 mg/kg to about lg/kg of the patient's body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks.
  • the pharmaceutical compositions of the invention are administered once a day, twice a day, or three times a day. In other embodiments, the pharmaceutical compositions are administered once a week, twice a week, once every two weeks, once a month, once every six weeks, once every two months, twice a year or once per year. It will also be appreciated that the effective dosage of the compounds used for treatment may increase or decrease over the course of a particular treatment.
  • the amount of a compound required to be effective as an antagonist of adenosine A 3 receptors will, of course, vary with the active moiety selected, the individual mammal being treated and is ultimately at the discretion of the medical or veterinary practitioner. The factors to be considered include the binding affinity of the active, the route of administration, the nature of the formulation, the mammal's body weight, surface area, age and general condition, and the particular compound to be administered.
  • the total daily dose may be given as a single dose, multiple doses, e.g., two to six times per day, or by intravenous infusion for a selected duration. Dosages above or below the range cited above are within the scope of the present invention and may be administered to the individual patient if desired and necessary. For example, for a 75 kg mammal, a dose range would be about 75 ug to about 50 mg per day, and a typical dose would be about 50 mg per day. If discrete multiple doses are indicated, treatment might typically be 50 mg of a compound of the present invention given 3 times per day.
  • the invention also provides that the compounds of the invention are packaged in a hermetically sealed container such as an ampoule or sachette indicating the quantity of antibody.
  • the compounds of the invention are supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted, e.g., with water or saline to the appropriate concentration for administration to a subject.
  • the compounds of the invention are supplied as a dry sterile lyophilized powder in a hermetically sealed container at a unit dosage of at least 5 mg, more preferably at least 10 mg, at least 15 mg, at least 25 mg, at least 35 mg, at least 45 mg, at least 50 mg, at least 75 mg, at least 100 mg, at least 200 mg, at least 500 mg or at least 1 gram..
  • the lyophilized compounds of the invention should be stored at between 2 and 8°C in their original container and the compounds should be administered within 12 hours, preferably within 6 hours, within 5 hours, within 3 hours, or within 1 hour after being reconstituted.
  • compounds of the invention are supplied in liquid form in a hermetically sealed container indicating the quantity and concentration of the compound.
  • the liquid form of the compounds are supplied in a hermetically sealed container at least 1 mg/ml, more preferably at least 2.5 mg/ml, at least 5 mg/ml, at least 8 mg/ml, at least 10 mg/ml, at least 15 mg/kg, at least 25 mg/ml, at least 50 mg/ml, at least 100 mg/ml, at least 150 mg/ml, at least 200 mg/ml of the compounds.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active compound into association with a carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier or a finely divided solid carrier and then, if necessary, shaping the product into desired unit dosage form.
  • the formulations may further include one or more optional accessory ingredient(s) utilized in the art of pharmaceutical formulations, e.g., diluents, buffers, flavoring agents, binders, surface active agents, thickeners, lubricants, suspending agents, preservatives (including antioxidants) and the like.
  • optional accessory ingredient(s) utilized in the art of pharmaceutical formulations, e.g., diluents, buffers, flavoring agents, binders, surface active agents, thickeners, lubricants, suspending agents, preservatives (including antioxidants) and the like.
  • the formulations include, but are not limited to, those suitable for oral, rectal, topical or parenteral (including subcutaneous, intramuscular and intravenous) administration. Preferred are those suitable for oral or parenteral administration.
  • pharmaceutically acceptable carriers described herein for example, vehicles, adjuvants, excipients, or diluents, are well known to those who are skilled in the art and are readily available to the public. It is preferred that the pharmaceutically acceptable carrier be one which is chemically inert to the active compounds and one which has no detrimental side effects or toxicity under the conditions of use.
  • compositions of the present invention there are a wide variety of suitable formulations of the pharmaceutical composition of the present invention.
  • suitable formulations for oral, aerosol, parenteral, subcutaneous, intravenous, intraarterial, intramuscular, interperitoneal, intrathecal, rectal, and vaginal administration are merely exemplary and are in no way limiting.
  • the compounds of the invention e.g., high affinity adenosine A 3 receptor antagonist, alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation.
  • aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer.
  • compositions, prophylactic or therapeutic agents of the invention are preferably tested in vitro, e.g., in a cell culture system, and then in vivo, e.g., in an animal model organism, such as a rodent animal model system, for the desired therapeutic activity prior to use in humans.
  • assays which can be used to determine whether administration of a specific pharmaceutical composition is indicated include cell culture assays in which a patient tissue sample is grown in culture, and exposed to or otherwise contacted with a pharmaceutical composition, and the effect of such composition upon the tissue sample is observed, e.g., inhibition of or decrease in growth and/or colony formation in soft agar or tubular network formation in three-dimensional basement membrane or extracellular matrix preparation.
  • the tissue sample can be obtained by biopsy from the patient. This test allows the identification of the therapeutically most effective prophylactic or therapeutic molecule(s) for each individual patient.
  • therapeutic agents and methods may be screened using cells of a tumor or malignant cell line.
  • in vitro assays can be carried out with representative cells of cell types involved in an autoimmune or inflammatory disorder (e.g., T cells), to determine if a pharmaceutical composition of the invention has a desired effect upon such cell types.
  • cell proliferation can be assayed by measuring 3 H-thymidine incorporation, by direct cell count, by detecting changes in transcriptional activity of known genes such as proto-oncogenes (e.g., fos, myc) or cell cycle markers; cell viability can be assessed by trypan blue staining, differentiation can be assessed visually based on changes in morphology, decreased growth and/or colony formation in soft agar or tubular network formation in three-dimensional basement membrane or extracellular matrix preparation. Combinations of prophylactic and/or therapeutic agents can be tested in suitable animal model systems prior to use in humans.
  • Such animal model systems include, but are not limited to, rats, mice, chicken, cows, monkeys, pigs, dogs, rabbits, etc. Any animal system well-known in the art may be used. In a specific embodiment of the invention, combinations of prophylactic and/or therapeutic agents are tested in a mouse model system. Such model systems are widely used and well-known to the skilled artisan. - Prophylactic and/or therapeutic agents can be administered repeatedly. Several aspects of the procedure may vary such as the temporal regime of administering the prophylactic and/or therapeutic agents, and whether such agents are administered separately or as an admixture.
  • the prophylactic and/or therapeutic agents of the invention can be tested in clinical trials to establish their efficacy. Establishing clinical trials will be done in accordance with common methodologies known to one skilled in the art, and the optimal dosages and routes of administration as well as toxicity profiles of the compositions of the invention can be established using routine experimentation. Toxicity and efficacy of the prophylactic and/or therapeutic protocols of the instant invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
  • Prophylactic and/or therapeutic agents that exhibit large therapeutic indices are preferred. While prophylactic and/or therapeutic agents that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such agents to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage of the prophylactic and/or therapeutic agents for use in humans.
  • the dosage of such agents lies preferably within a range of circulating concentrations that include the ED5 0 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC 50 i.e., the concentration of the test compound that achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • the anti-cancer activity of the therapies used in accordance with the present invention also can be determined by using various experimental animal models for the study of cancer such as the SCID mouse model or transgenic mice or nude mice with human xenografts, animal models, such as hamsters, rabbits, etc. known in the art and described in Relevance of Tumor Models for Anticancer Drug Development (1999, eds. Fiebig and Burger); Contributions to Oncology (1999, Karger); The Nude Mouse in Oncology Research (1991, eds. Boven and Winograd); and Anticancer Drug Development Guide (1997 ed. Teicher), herein incorporated by reference in their entireties.
  • SCID mouse model or transgenic mice or nude mice with human xenografts animal models, such as hamsters, rabbits, etc. known in the art and described in Relevance of Tumor Models for Anticancer Drug Development (1999, eds. Fiebig and Burger); Contributions to Oncology (1999, Karger);
  • the protocols and compositions of the invention are preferably tested in vitro, and then in vivo, for the desired therapeutic or prophylactic activity, prior to use in humans.
  • Therapeutic agents and methods may be screened using cells of a tumor or malignant cell line. Many assays standard in the art can be used to assess such survival and/or growth; for example, cell proliferation can be assayed by measuring 3 H-thymidine incorporation, by direct cell count, by detecting changes in transcriptional activity of known genes such as proto-oncogenes (e.g., fos, myc) or cell cycle markers; cell viability can be assessed by trypan blue staining, differentiation can be assessed visually based on changes in morphology, decreased growth and/or colony formation in soft agar or tubular network formation in three-dimensional basement membrane or extracellular matrix preparation, etc.
  • proto-oncogenes e.g., fos, myc
  • cell cycle markers e.g., cell cycle markers
  • cell viability can be assessed by trypan blue stain
  • Compounds for use in therapy can be tested in suitable animal model systems prior to testing in humans, including but not limited to in rats, mice, chicken, cows, monkeys, rabbits, hamsters, etc., for example, the animal models described above. The compounds can then be used in the appropriate clinical trials. 6.5.1 ANTI-PROLIFERATIVE ASSAYS
  • the antiproliferative activity of compounds of the invention can be readily determined using no more than routine experimentation using cell growth inhibitory assays. Selection of cell lines for such assays is based upon desired future pharmaceutical use. Numerous cell lines are available for study from American Type Culture Collection, Manassas, VA. Cell lines suitable for assays include, but are not limited to, HL-60 human leukemia, A375 human melanoma, SKMES human lung carcinoma, HT29 human colon carcinoma and Panc-1 human pancreatic carcinoma.
  • Cell lines can be maintained in RPMI- 1640 medium supplemented with 100 units/ml penicillin G sodium, 100 ⁇ g/ml streptomycin sulfate, 0.25 ⁇ g/ml amphotericin B (fungizone), 2 mM glutamine, 10 mM HEPES, 25 ⁇ g/ml gentamycin, and 10% heat-inactivated fetal bovine serum.
  • Such maintained cells can be cultured in a T25 Falcon Tissue Culture flask in a humidified incubator at 37°C with 5% CO 2 -95% air.
  • such maintained cells can be sub-cultured twice a week with approximate doubling times of 22 hours for A375 human melanoma, 46 hours for SKMES human lung carcinoma, 48 hours for HT29 human colon carcinoma and 60 hours for Panc-1 human pancreatic carcinoma.
  • MTT assay One of the growth inhibitory assays that can be used in accordance with the methods of the invention is the MTT assay.
  • An exemplary assay may comprise the following steps: Cells (1000-1500 cells/well) are seeded in a 96-well micro culture plate in a total volume of 100 ⁇ l/well. After overnight incubation in a humidified incubator at 37°C with 5% CO 2 - 95% air, chemotherapeutic drug solutions diluted with culture medium at various concentrations are added in the amount of 100 ⁇ l to each well. The plates are placed in a humidified incubator at 37 0 C with 5% CO 2 - 95% air for 7-10 days. The plates are then centrifuged briefly and 100 ⁇ l of the growth medium is removed.
  • MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide] reagent (1 mg/ml in Dulbecco's phosphate-buffered saline) for 4 hr at 37°C.
  • the resultant purple formazan precipitate is solubilized with 200 ⁇ l of 0.04 N HCl in isopropanol.
  • Absorbance is measured at a wavelength of 595 nm and at a reference wavelength of 655 nm using a Bio-Rad Model 3550 Microplate Reader. Preferably, all tests are run in duplicate for each dose level.
  • Bio-Rad brands Microplate Readers when properly equipped, transmit measured test results to a personal computer for interpretation via computer programs such as the EZED50 program.
  • the EZED50 computer program estimates the concentration of agent that inhibits cell growth by 50% as compared to the control cells. This is termed the IC 50 and is determined by curve fitting test data using the following four logistic equation.
  • a max is the absorbance of the control cells
  • a m j n is the absorbance of the cells in the presence of the highest agent concentration
  • Y is the observed absorbance
  • X is the agent concentration
  • IC 50 is the concentration of agent that inhibits the cell growth by 50% compared to the control cells
  • n is the slope of the curve. If testing concentrations are properly selected (i.e., no or little growth inhibition at low concentrations and complete inhibition at high concentrations), EZED50 program fits the data extremely well and estimates the IC 50 value accurately.
  • EZED50 cannot estimate the IC50 value accurately (as indicated by an erroneously fitted Amax value). In these instances, the data could be re-analyzed by fixing the Amax value.
  • EZED50 will overestimate the potency of the testing agent (the fitted ICsovalue is lower than the "actual" IC 50 value). In this case, the IC 5O could be estimated more accurately if both the A max and A m j n values are fixed.
  • the best way to determine the IC50 accurately is to decrease or increase the concentrations of the test agent and repeat the test.
  • Inhibitory assay testing is also used to determine enhanced therapeutic effects from combining A 3 receptor antagonists with other tumor inhibiting agents.
  • the inhibitory cell growth assays are run with and without selected concentrations of A 3 receptor antagonists.
  • An enhanced therapeutic effect is present when the IC 50 of the tumor inhibiting agent is lower with the A 3 receptor antagonist.
  • the enhancement factor (EF) is calculated by dividing the IC5 0 value of the tumor inhibiting agent for a tumor cell line by the IC 50 value of the agent with A 3 receptor antagonist for the same cell line:
  • EF Enhanced Factor
  • any EF above 1.0 is synergistic if the A 3 receptor antagonist concentration is below the threshold of cell growth inhibition.
  • the activity and selectivity of the compounds of the invention as adenosine A 3 antagonists can be readily determined using no more than routine experimentation using any of the assays disclosed herein or known to one skilled in the art. Since the A 1 and A 2a receptors express similar pharmacology between humans and rodents, endogenous receptors from the rat can be used for the A 1 and A 2a binding assays.
  • An exemplary rat A 1 and A 2 A Adenosine receptor binding assay may comprise the following steps.
  • Membrane preparations Male Wistar rats (200-250 g) can be decapitated and the whole brain (minug brainstem, striatum and cerebellum) dissected on ice. The brain tissues can be disrupted in a Polytron (setting 5) in 20 vols of 50 mM Tris HCl, pH 7.4. The homogenate can then be centrifuged at 48,000 g for 10 min and the pellet resuspended in Tris-HCL containing 2 IU/ml adenosine deaminase, type VI (Sigma Chemical Company, St. Louis, Mo., USA).
  • the membranes can be centrifuged and the pellets stored at -70 0 C.
  • Striatal tissues can be homogenized with a Polytron in 25 vol of 50 mM Tris HCl buffer containing 10 mM MgCl 2 pH 7.4. The homogenate can then be centrifuged at 48,000 g for 10 min at 4 0 C. and resuspended in Tris HCl buffer containing 2 IU/ml adenosine deaminase. After 30 min incubation at 37 0 C 5 membranes can be centrifuged and the pellet stored at -70 0 C.
  • the radioligand binding assays may comprise the following: Binding of [ 3 H]-DPCPX (1,3- dipropyl-8-cyclopentylxanthine) to rat brain membranes can be performed essentially according to the method previously described by Bruns et ah, 1980, Proc. Natl. Acad. Set. 11, 5547-5551, which is incorporated herein by reference in its entirety. Displacement experiments can be performed in 0.25 ml of buffer containing 1 nM [ 3 H]-DPCPX, 100 ul of diluted membranes of rat brain (100 ⁇ g of protein/assay) and at least 6-8 different concentrations of examined compounds. Non specific binding can be determined in the presence of 10 uM of CHA (N6 cyclohexyladenosine) and this is always ⁇ 10% of the total binding. Incubation times are typically 120 min at 25 0 C.
  • Radioligand binding assays may comprise the following: Binding of [ 3 H]- SCH 58261 (5-amino-7-(2- ⁇ henylethyl)-2-(2-furyl)-pyrazolo[4,3-e]-l,2,4-triazolo[l,5- c]pyrimidine) to rat striatal membranes (100 ug of protein/assay) can be performed according to methods described in Zocchi et ah, 1996, J. Pharm. andExper. Ther. 276:398- 404, which is incorporated herein by reference in its entirety. In competition studies, at least 6-8 different concentrations of examined compounds should be used.
  • Non specific binding can be determined in the presence of 50 uM of NECA (5'-(N- ethylcarboxamido)adenosine). Incubation time is typically 60 min at 25 0 C. Bound and free radioactivity can be separated by filtering the assay mixture through Whatman GF/B glass-fiber filters using a Brandel cell harvester (Gaithersburg, Md., USA). The incubation mixture can be diluted with 3 ml of ice-cold incubation buffer, rapidly vacuum filtered and the filter can be washed three times with 3 ml of incubation buffer. The filter bound radioactivity can be measured, for example, by liquid scintillation spectrometry. The protein concentration can be determined, for example, according to a Bio-Rad method (Bradford, 1976, Anal. Biochem. 72:248, which is incorporated herein by reference in its entirety) with bovine albumin as reference standard.
  • NECA 5'-(N- ethylcarboxa
  • An exemplary assay for human cloned A 3 Adenosine Receptor Binding Assay may comprise the following: Binding assays can be carried out according to methods described in Salvatore et al, 1993, Proc. Natl. Acad. Sd. 90:10365-10369; which is incorporated herein by reference in its entirety. In saturation studies, an aliquot of membranes (8 mg protein/ml) from HEK-293 cells transfected with the human recombinant A 3 adenosine receptor (Research Biochemical International, Natick, Mass., USA) can be incubated with 10-12 different concentrations of [ 125 1]AB-MECA ranging from 0.1 to 5 nM.
  • Competition experiments can be carried out in duplicate in a final volume of 100 ul in test tubes containing 0.3 nM [ 125 1]AB-MECA, 50 mM Tris HCl buffer, 10 mM MgCl 2 , pH 7.4 and 20 ul of diluted membranes (12.4 mg protein/ml) and at least 6-8 different concentrations of examined ligands. Incubation time was 60 min at 37 0 C, according to the results of previous time-course experiments. Bound and free radioactivity were separated by filtering the assay mixture through Whatman GF/B glass-fiber filters using a Brandel cell harvester. Non-specific binding was defined as binding in the presence of 50 uM R-PIA and was about 30% of total binding.
  • the incubation mixture was diluted with 3 ml of ice- cold incubation buffer, rapidly vacuum filtered and the filter was washed three times with 3 ml of incubation buffer.
  • the filter bound radioactivity was counted in a Beckman gamma 5500B gamma counter.
  • the protein concentration can be determined according to a Bio- Rad method with bovine albumin as reference standard.
  • Ki Inhibitory binding constant
  • the efficacy of the compounds can be tested in animal models.
  • the efficacy of the adenosine A 3 antagonists alone or in combination with conventional anti-tumor agents such as cytotoxic/anti-neoplastic agents and anti- angiogenic agents can be compared to the conventional agents alone.
  • conventional anti-tumor agents such as cytotoxic/anti-neoplastic agents and anti- angiogenic agents
  • a tumor of a given size is present in a rat or mouse.
  • the mouse is treated with the agent and the size of the tumor is measured over time.
  • the mean survival time of the animals can also be measured.
  • the adenosine A 3 antagonists which are used must actively bind the adenosine A 3 receptor in the animal which is to be tested.
  • Xenografts can be implanted into the animal to test the ability of species specific cytotoxic compounds. However, the effect of the anti- angiogenic therapy depends on how well the anti-angiogenetic compounds work on inhibiting the specific animal's vasculation.
  • Assay methods that have demonstrated the efficacy of endostatin and angiostatin in suppressing tumor growth can be applied to A 1 , A 2a , and A 3 antagonists.
  • Lewis lung carcinoma, T241 fibrosarcoma or Bl 6F10 melanoma can be grafted to mice (Boehm et al, 1977, Nature, 390:404-407; which is incorporated herein by reference in its entirety).
  • tumors such as PC-3 human prostate carcinoma, the CCLl 88 human colon carcinoma and the UBC urinary bladder carcinoma have been shown to release angiogenic agents (Chen et al, 1996, Cancer Res., 55:4230-4322; which is incorporated herein by reference in its entirety), these tumors can also be grafted in mice for the evaluation of anti-tumor activities of adenosine antagonists.
  • Adenosine antagonists can be administered to mice by the methods disclosed herein. This therapy can be used alone or in conjunction with anti-angiogenic therapy or conventional chemotherapy.
  • the invention encompasses methods to assess quantitative and qualitative aspects of HIF-I ⁇ expression.
  • the increased expression of an HIF- lot gene or gene product indicates a predisposition for the development of cancer or a disease or disorder associated with enhanced HIF- l ⁇ expression.
  • enhanced expression levels of an HIF-I ⁇ gene or gene product can indicate the presence of cancer in a subject or the risk of metastasis of said cancer in said subject.
  • Techniques well known in the art e.g., quantitative or semi-quantitative RT PCR or Northern blot, can be used to measure expression levels of HIF-I ⁇ . Methods that describe both qualitative and quantitative aspects of HIF-I ⁇ gene or gene product expression are described in detail in the examples infra.
  • the measurement of HIF-l ⁇ gene expression levels can include measuring naturally occurring HIF- l ⁇ transcripts and variants thereof as well as non-naturally occurring variants thereof, however for the diagnosis and/or prognosis of diseases or disorders in a subject the HIF-l ⁇ gene product is preferably a naturally occurring HIF-l ⁇ gene product or variant thereof.
  • the invention relates to methods of measuring the expression of the HIF- 1 ⁇ gene or gene product in a subj ect.
  • any method known in the art for detecting and/or quantitating an HIF- l ⁇ level may be used in the methods and kits of the invention, a number of which are exemplified herein. Particularly preferred are methods known in the art for detecting and/or quantitating an HIF-l ⁇ activity or an HIF-l ⁇ related activity, e.g., phosphorylation of downstream effector molecules in the HIF-l ⁇ pathway.
  • the invention encompasses measuring an HIF- l ⁇ activity or an HIF- l ⁇ related activity including but not limited to, measuring an activity of one or more downstream effectors of an HIF-l ⁇ signaling cascade. Measuring an HIF-l ⁇ activity or an HIF-l ⁇ related activity can be done using any of the methods disclosed herein or any standard method known to one skilled in the art.
  • the invention encompasses quantitation of a nucleic acid encoding HIF- lot in a sample obtained from a subject using methods disclosed herein or any standard method known in the art.
  • the invention encompasses quantitation of HIF-I ⁇ protein in a sample obtained from a subject with a disease or disorder, e.g., cancer. Any method known in the art for the detection and quantitation of a HIF- l ⁇ protein is encompassed within the present invention.
  • the methods and kits of the invention encompass detection and/or quantitation of a nucleic acid sequence encoding HIF- l ⁇ in a sample obtained from a subject.
  • the invention provides methods for amplifying a specific HIF- l ⁇ nucleic acid sequence in a sample obtained from a subject with cancer, and detecting and/or quantitating the same.
  • Nucleic acids encoding HIF- l ⁇ are well known in the art. See, for example, Wang et al, 1995, Proc. Natl. Acad. ScL USA, 92: 5510-4; and WO 96/39426 each of which is incorporated herein by reference in their entireties.
  • kits of the invention may use any nucleic acid amplification or detection method known to one skilled in the art, such as those described in U.S. Patent No.'s 5,525,462; 6,528,632; 6,344,317; 6,114,117; 6,127,120; 6,448,001; all of which are incorporated herein by reference in their entirety.
  • the nucleic acid encoding an HIF-I ⁇ is amplified by PCR amplification using methodologies known to one skilled in the art.
  • amplification of target sequences i.e., nucleic acid sequences encoding HIF- l ⁇
  • LCR ligase chain reaction
  • QP-replicase amplification amplification of target sequences in a sample obtained from a subject with cancer
  • transcription amplification amplification of transcription amplification
  • self-sustained sequence replication each of which provides sufficient amplification.
  • the PCR process is well known in the art and is thus not described in detail herein.
  • PCR methods and protocols see, e.g., Innis et ah, eds., PCR Protocols, A Guide to Methods and Application. Academic Press,
  • the invention encompasses methods to determine quantitative and/or qualitative levels of expression of HIF-I ⁇ . Any technique known in the art for measuring the expression of an HIF-I ⁇ is within the scope of the invention, including but not limited, to quantitative and/or semi-quantitative RT PCR and Northern blot analysis.
  • the invention encompasses detecting and/or quantitating an HIF- l ⁇ nucleic acid using fluorescence in situ hybridization (FISH) in a sample, preferably a tissue sample, obtained from a subject with cancer in accordance with the methods of the invention.
  • FISH fluorescence in situ hybridization
  • a sample preferably a tissue sample
  • FISH fluorescence in situ hybridization
  • the invention encompasses measuring naturally occurring HIF- 1 ⁇ transcripts and variants thereof as well as non-naturally occurring variants thereof.
  • the HIF-I ⁇ transcript is preferably a naturally occurring HIF- l ⁇ transcript.
  • the invention relates to methods of prognosis of a cancer in a subject by measuring the expression of an HIF- 1 ⁇ transcript in a subject.
  • the increased level of mRNA encoding an HIF- l ⁇ as compared to a standard, e.g., a non-cancerous sample, would indicate the increased risk of developing cancer in said subject.
  • the increased level of mRNA encoding an HIF- l ⁇ as compared to a standard would indicate the risk of metastasis of the cancer in said subject or the likelihood of a poor prognosis in said subject.
  • the invention encompasses isolating RNA from a sample obtained from a subject with cancer, and testing the RNA utilizing hybridization or PCR techniques as described above for determining the level of an HIF-I ⁇ .
  • the invention encompasses synthesizing cDNA from the isolated RNA by reverse transcription. All or part of the resulting cDNA is then used as the template for a nucleic acid amplification reaction, such as a PCR or the like.
  • the nucleic acid reagents used as synthesis initiation reagents ⁇ e.g., primers) in the reverse transcription and nucleic acid amplification steps of this method are chosen from among the HIF- l ⁇ nucleic acid reagents described below.
  • the preferred lengths of such nucleic acid reagents are at least 9- 30 nucleotides.
  • the nucleic acid amplification may be performed using radioactively or non-radioactively labeled nucleotides.
  • enough amplified product may be made such that the product may be visualized by standard ethidium bromide staining or by utilizing any other suitable nucleic acid staining method.
  • standard Northern analysis techniques known to one skilled in the art can be performed on a sample obtained from a subject with cancer.
  • the preferred length of a probe used in Northern analysis is 9-50 nucleotides. Utilizing such techniques, quantitative as well as size related differences among HIF- l ⁇ transcripts can also be detected.
  • the invention encompasses gene expression assays in situ, i.e., directly upon tissue sections (fixed and/or frozen) of patient tissue obtained from biopsies or resections, such that no nucleic acid purification is necessary.
  • Nucleic acid reagents such as those described below may be used as probes and/or primers for such in situ procedures (see, e.g., Nuovo, GJ., 1992, PCR In Situ Hybridization: Protocols And Applications. Raven Press, NY, which is incorporated herein by reference in its entirety).
  • the target HIF- l ⁇ nucleic acids of the invention can also be detected using other standard techniques well known to those of skill in the art. Although the detection step is typically preceded by an amplification step, amplification is not necessarily required in the methods of the invention.
  • the HIF-I ⁇ nucleic acids can be identified by size fractionation (e.g., gel electrophoresis). The presence of different or additional bands in the sample as compared to the control is an indication of the presence of target nucleic acids of the invention.
  • the target HIF-I ⁇ nucleic acids can be identified by sequencing according to well known techniques.
  • oligonucleotide probes specific to the target HIF- l ⁇ nucleic acids can be used to detect the presence of specific fragments.
  • Sequence-specific probe hybridization is a well known method of detecting desired nucleic acids in a sample comprising a biological fluid or tissue sample and is within the scope of the present invention. Briefly, under sufficiently stringent hybridization conditions, the probes hybridize specifically only to substantially complementary sequences. The stringency of the hybridization conditions can be relaxed to tolerate varying amounts of sequence mismatch. If the target is first amplified, detection of the amplified product utilizes this sequence-specific hybridization to insure detection of only the correct amplified target, thereby decreasing the chance of a false positive caused by the presence of homologous sequences from related organisms or other contaminating sequences.
  • hybridization formats well known in the art including but not limited to solution phase, solid phase, mixed phase, or in situ hybridization assays are encompassed within the nucleic acid detection methods of the invention.
  • solution phase hybridizations both the target nucleic acid and the probe or primer are free to interact in the reaction mixture.
  • solid phase hybridization assays either the target or probes are linked to a solid support where they are available for hybridization with complementary nucleic acids in solution.
  • Exemplary solid phase formats include Southern hybridizations, dot blots, and the like.
  • the invention encompasses homogenous based hybridization assays as well as heterogeneous based assays for detection and/or quantitation of HIF-I ⁇ nucleic acid sequences in accordance with the methods of the invention.
  • Heterogeneous based assays depend on the ability to separate hybridized from non-hybridized nucleic acids.
  • One such assay involves immobilization of either the target or probe nucleic acid on a solid support so that non-hybridized nucleic acids which remain in the liquid phase can be easily separated after completion of the hybridization reaction ⁇ see, e.g., Southern, 1975, J. MoI Biol. 98: 503-517; which is incorporated herein by reference in its entirety).
  • homogeneous assays depend on other means for distinguishing between hybridized and non-hybridized nucleic acids. Because homogeneous assays do not require a separation step, they are generally considered to be more desirable.
  • One such homogeneous assay relies on the use of a label attached to a probe nucleic acid that is only capable of generating a signal when the target is hybridized to the probe ⁇ see, e.g., Nelson, et al, 1992, Nonisotopic DNA Probe Techniques. Academic Press, New York, N.Y., pages 274-310; which is incorporated herein by reference in its entirety).
  • the invention encompasses any method known in the art for enhancing the sensitivity of the detectable signal in such assays, including but not limited to the use of cyclic probe technology (Bakkaoui et al, 1996, BioTeckniques 20: 240-8, which is incorporated herein by reference in its entirety); and the use of branched probes (Urdea et al, 1993, Clin. Chem. 39: 725-6; which is incorporated herein by reference in its entirety).
  • nucleic acid probes capable of specifically hybridizing to a target can be labeled by any one of several methods typically used to detect the presence of hybridized nucleic acids.
  • One common method of detection is the use of autoradiography, using probes labeled with 3 H, 125 1, 35 S, 14 C, or 32 P, or the like.
  • the choice of radioactive isotope depends on research preferences due to ease of synthesis, stability, and half lives of the selected isotopes.
  • Other labels include compounds ⁇ e.g., biotin and digoxigenin), that bind to anti- ligands or antibodies labeled with fluorophores, chemiluminescent agents, or enzymes.
  • probes can be conjugated directly to labels such as fluorophores, chemiluminescent agents or enzymes.
  • labels such as fluorophores, chemiluminescent agents or enzymes.
  • the choice of label depends on sensitivity required, ease of conjugation with the probe, stability requirements, and available instrumentation.
  • the probes and primers of the invention can be synthesized and labeled using techniques known to one skilled in the art. Oligonucleotides for use as probes and primers may be chemically synthesized according to the solid phase phosphoramidite triester method described by Beaucage, S. L. and Caruthers, M. H., 1981, Tetrahedron Lett. 22(20): 1859-1862, using an automated synthesizer, as described inNeedham-VanDevanter, D. R., et al.
  • oligonucleotides can be by either native acrylamide gel electrophoresis or by anion-exchange HPLC, as described in Pearson, J. D. and Regnier, F. E., 1983, J. Chrom. 255:137-149. All of the references cited supra are incorporated herein by reference in their entirety.
  • the methods and kits of the invention encompass detection and/or quantitation of HIF-I ⁇ in a sample obtained from a subject. Any method known to one skilled in the art for the detection and quantitation of an HIF-I ⁇ protein is encompassed within the present invention.
  • HIF- l ⁇ protein sequences useful in the methods and kits of the invention are well known in the art. See, for example, Wang et al, 1995, Proc. Natl. Acad. ScL USA, 92: 5510-4; Merighi et al, HIF-l ⁇ Expression in Cancer Cells in up- regulated by adeerrozine A 3 receptors, paper submitted to Nature, 2004; and WO 96/39426 each of which is incorporated herein by reference in its entireties.
  • HIF- l ⁇ proteins and anti- HIF- l ⁇ antibodies and immunospecific fragments thereof are suitable in the assays of the invention.
  • Detection and quantitation of an HIF- l ⁇ gene product encompasses the detection of proteins exemplified herein.
  • Detection of elevated levels of an HIF-l ⁇ gene product in a sample obtained from a subject in accordance with the methods of the invention is generally compared to a standard sample.
  • antibodies directed against naturally occurring HIF- l ⁇ proteins may be used in the methods of the invention.
  • the invention encompasses the use of any standard immunoassay method known to one skilled in the art, including but not limited to Western blot, ELISA, and FACS .
  • the invention encompasses use of an immunoassay comprising contacting a sample from a subject with an anti- HIF- l ⁇ antibody or an immunospecific fragment thereof under conditions such that immunospecific binding to the HIF- l ⁇ receptor in the sample can occur, thereby forming an immune complex, and detecting and/or measuring the amount of complex formed.
  • an antibody to an HIF-l ⁇ is used to assay a sample for the presence of the HIF-l ⁇ , wherein an increased level of the HIF-l ⁇ is detected relative to a standard sample.
  • the biological sample may be brought in contact with and immobilized onto a solid phase support or a carrier such as nitrocellulose or other solid support capable of immobilizing cells, cell particles or soluble proteins.
  • a solid phase support or a carrier such as nitrocellulose or other solid support capable of immobilizing cells, cell particles or soluble proteins.
  • the support can be washed with suitable buffers followed by treatment with the antibody that selectively or specifically binds to an HIF- l ⁇ protein.
  • the solid phase support can then be washed with buffer to remove unbound antibody.
  • the amount of antibody bound to the solid support can then be detected by conventional means.
  • Solid phase support or carrier refers to any support capable of binding an antigen or an antibody.
  • Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
  • the nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention.
  • the support material may have virtually any possible structural configuration so long as the coupled molecule is capable of binding to an antigen or antibody.
  • the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tube, or the external surface of a rod.
  • the surface may be flat such as a sheet, test strip, etc.
  • Preferred supports include polystyrene beads. Those skilled in the art will know many other suitable carriers for binding antibody or antigen, or will be able to ascertain the same by use of routine experimentation.
  • the anti- HIF-I ⁇ antibody or an immunospecific fragment thereof can be detectably labeled by linking the same to an enzyme and using the labeled antibody in an enzyme immunoassay (EIA) (Voller, A., "The 1 Enzyme Linked Immunosorbent Assay (ELISA)", 1978, Diagnostic Horizons 2:1, Microbiological Associates Quarterly Publication, Walkersville, MD; Voller, A. et al, 1978, J Clin. Pathol. 31:507-520; Butler, J.E., 1981, Meth. Enzymol. 73:482; Maggio, E. ed., 1980, Enzyme Immunoassay, CRC Press, Boca Raton, FL,; Ishikawa, E. et ah, eds., 1981, Enzyme
  • the enzyme bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorimetric or visual means.
  • Enzymes that can be used to detectably label the antibody include but are not limited to malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta- galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase, among others.
  • the detection can be accomplished by colorimetric methods that employ a chromogenic substrate for the enzyme. Detection can also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.
  • Detection can also be accomplished using any other method known to one skilled in the art.
  • a radioimmunoassay RIA
  • the radioactive isotope can be detected by such means as the use of a gamma counter or a scintillation counter, or by autoradiography.
  • the invention encompasses labeling the antibody with a fluorescent compound.
  • fluorescent labeling compounds include fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine.
  • the antibody can also be detectably labeled using fluorescence emitting metals such as 152 Eu, or others of the lanthanide series.
  • metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).
  • DTPA diethylenetriaminepentacetic acid
  • EDTA ethylenediaminetetraacetic acid
  • the invention further encompasses detectably labeling the antibody by coupling it to a cheniiluminescent compound.
  • the presence of the chemiluminescent- tagged antibody is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction.
  • particularly useful chemiluminescent labeling compounds are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.
  • a bioluminescent compound can be used to label the antibody of the present invention.
  • Bioluminescence is a type of chemiluminescence found in biological systems in which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence.
  • Bioluminescent compounds for purposes of labeling include, e.g., luciferin, luciferase and aequorin.
  • the invention also encompasses methods for indirect detection of HIF-I ⁇ .
  • the invention encompasses use of an immunoassay comprising contacting a sample derived from a subject with cancer with an anti- HIF- l ⁇ antibody (primary antibody) or an immunospecific fragment thereof under conditions such that immunospecific binding to the HIF- l ⁇ protein in the sample can occur, thereby forming an immune complex, adding a secondary antibody that is labeled under conditions such that immunospecific binding to the primary antibody occurs and detecting and/or quantitating the amount of complex formed indirectly.
  • Anti- HIF- l ⁇ antibodies or immunospecific fragments thereof may be used quantitatively or qualitatively to detect an HIF- l ⁇ in a sample.
  • the anti- HIF- l ⁇ antibodies or immunospecific fragments thereof may be used histologically, e.g., immunofluorescence or microscopic studies, using common techniques known to one skilled in the art, for in situ detection of an HIF-I ⁇ receptor.
  • In situ detection may be accomplished by preparing a histological specimen from a subject, such as a paraffin embedded section of tissue, e.g., breast tissues, and applying thereto a labeled antibody of the present invention.
  • the antibody (or fragment) is preferably applied by overlaying the labeled antibody (or fragment) onto the biological sample.
  • the methods of the invention may use any nucleic acid encoding HIF- l ⁇ , an analog, fragment or derivative thereof, as a proxy for determining a HIF-I a level.
  • a nucleic acid is intended to include DNA molecules (e.g., cDNA, genomic DNA), RNA molecules (e.g., hnRNA, pre-mRNA, mRNA) and DNA or RNA analogs (e.g., peptide nucleic acids) generated using techniques known to one skilled in the art.
  • the nucleic acid measured as a proxy for a HIF-I ⁇ level can be single-stranded or double stranded.
  • nucleotide sequences for use in the methods and kits of the invention may include all or a portion of any of the following: nucleotide sequences disclosed in U.S. Patent Nos. 6,455,674 (issued to Einat et al); 6,652,799 (issued to Semenza); 6,222,018(issued to Semenza); Wang et al. , 1995 PNAS USA, 92: 5510-4; and WO 96/39426.
  • the invention encompasses all or a portion of the nucleotide sequence of human HIF-l ⁇ with GENBANK Accession Nos. NM_OO153O and NM_181054.
  • any HIF-I ⁇ nucleic acid known in the art may be useful in the methods and kits of the invention.
  • Such nucleic acids generally encode at least a portion of HIF-I ⁇ , or have a sequence that hybridizes to a HIF- l ⁇ , -encoding nucleic acid under hybridizing conditions, as described herein.
  • the methods of the invention may use a coding sequence or a 5' or 3' untranslated region of a nucleic acid encoding HIF- l ⁇ or a fragment thereof as a probe, including naturally occurring and non-naturally occurring variants.
  • a non- naturally occurring variant is one that is engineered by man (e.g., a peptide nucleic acid probe).
  • HIF-l ⁇ , or an mRNA encoding HIF- l ⁇ in a sample from a subject is detected or measured, naturally occurring gene products are detected, including but not limited to wild-type gene products as well as mutants, allelic variants, splice variants, polymorphic variants, etc.
  • variants will be highly homologous to the wild-type gene product encoding HIF-I ⁇ , e.g., having at least 90%, 95%, 98% or 99% amino acid sequence identity (as determined by standard algorithms known in the art, see, e.g., Altschul, 1990 Proc. Natl. Acad. Sci. U.S.A. 87: 2264-2268; Altschul, 1993, Proc. Natl. Acad. ScL U.S.A. 90: 5873-5877; Altschul et al, 1990 J. MoI. Biol. 215: 403-410).
  • HIF-I ⁇ variants to be used as probes may be encoded by a nucleic acid which is hybridizable under stringent conditions to a nucleic acid encoding HIF-l ⁇ .
  • Nucleic acid hybridization methods are well known in the art (see, e.g., Sambrook et al, 2001 Molecular Cloning. A Laboratory Manual. 3 rd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York; Ausubel et al, eds., 1994-1997, in the Current Protocols in Molecular Biology: Series of laboratory technique manuals, John Wiley and Sons, Inc.; Shilo and Weinberg, 1981, Proc. Natl. Acad. Sci. U.S.A.
  • stringent conditions refers to the ability of a first polynucleotide molecule to hybridize, and remain bound to a second filter-bound polynucleotide molecule in 0.5 M NaHPO 4 , 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65°C, followed by washing in 0.2X SSC/0.1% SDS at 42°C ⁇ see Ausubel et al. (eds.), 1989, Current Protocols in Molecular Biology, Vol.
  • the variants being detected or measured comprise (or, if nucleic acids, encode) not more than 1, 2, 3, 4, 5, 10, 15 or 20 point mutations (substitutions) relative the wild-type sequence.
  • An isolated nucleic acid probe encoding HIF- 1 a or a portion thereof can be obtained by any method known in the art, e.g., from a deposited plasmid, by PCR amplification using synthetic primers hybridizable to the 3' and 5' ends of the sequence, and/or by cloning from a cDNA or genomic library using standard screening techniques, or by polynucleotide synthesis.
  • Use of such probes for detection and quantitation of specific sequences is well known in the art. See e.g., Erlich, e.d, 1989, PCR Technology Principles and Applications for DNA Amplification. Macmillan Publishers Ltd., England; Sambrook et al, Molecular Cloning: A Laboratory Manual, 3 rd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2001.
  • the methods of the invention may use a gene coding sequence, e.g., cDNA, of HIF-I ⁇ , which preferably hybridizes under stringent conditions as described above to at least about 6, preferably about 12, most preferably about 18 or more consecutive nucleotides of the gene coding sequence of HIF-I ⁇ protein, useful for the detection of an HIF-I ⁇ protein
  • nucleic acid sequence encoding HIF- l ⁇ protein such as those exemplified herein as a hybridization probe
  • full length nucleic acid molecules encoding HIF-I ⁇ protein can be quantitated using standard hybridization techniques (see, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual. 3 rd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY 5 2001) for use in the methods of the invention, i.e., as a proxy for HIF- l ⁇ level.
  • the HIF-l ⁇ sequences used in the methods of the invention are preferably human sequences.
  • homologs of the human HIF-I a isolated from other animals can also be used in the methods of the invention as a proxy for HIF- l ⁇ level, particularly where the subject is a non-human animal.
  • the invention also includes the use of HIF- l ⁇ homologs identified from non-human animals such as non-human primates, rats, mice, farm animals including but not limited to cattle, horses, goats, sheep, pigs, etc., household pets including but not limited to cats, dogs, etc. , in the methods of the invention.
  • the methods of the invention may use fragments of any of the nucleic acids disclosed herein in any of the methods of the invention.
  • a fragment preferably comprises at least 10, 20, 50, 100, or 200 contiguous nucleotides of a sequence described herein.
  • Standard recombinant DNA techniques known in the art may be used to provide a HIF- l ⁇ protein or a nucleic acid encoding an HIF-I ⁇ protein, or a fragment thereof, for use in the methods and kits of the invention.
  • the corresponding nucleotide sequence encoding HIF- l ⁇ protein of interest can be cloned.
  • HIF-I ⁇ proteins or fragments thereof, for the generation of antibodies for methods of the invention.
  • HIF- l ⁇ polypeptides and fragments can also be used as protein abundance or activity standards in the methods of the invention.
  • the invention encompasses amino acid sequences of HIF-l ⁇ as dislcosed in U.S. Patent Nos. 6,455,674 (issued to Einat et al); 6,652,799 (issued to Semenza); 6,222,018(issued to Semenza); Wang et al, 1995 PNAS USA, 92: 5510-4; 6,436,654 (issued to Berkenstam); WO 96/39426.
  • the amino acid sequences cited in the above-identified references are incorporated herein by reference in their entirety.
  • the invention encompasses the amino acid sequences of human HIF-Ia with GENBANK Accession No.s NP_001521 andNP_851397 each of which is incorporated herein by reference in its entirety.
  • the HIF-I ⁇ protein comprises an amino acid sequence that exhibits at least about 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% sequence similarity to the amino acid sequence of any of HIF-I ⁇ proteins known in the art.
  • proteins are provided consisting of or comprising a fragment of HIF-I ⁇ protein consisting of at least ten contiguous amino acids.
  • the fragment consists of or comprises at least 20, 30, 40, or 50 contiguous amino acids from a HIF- l ⁇ protein for use, for example, in raising antibodies.
  • Such fragments can also be useful, for example, as standards or controls in the methods and kits of the invention.
  • host-expression vector systems may be utilized to express HIF- l ⁇ proteins or fragments for use in the methods of the invention. Such host-expression systems are well known and provide the necessary means by which a protein of interest may be produced and subsequently purified. Examples of host-expression vector systems that may be used in accordance with the invention are: bacterial cells ⁇ e.g., E. coli, B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing a HIF-l ⁇ nucleic acid coding sequence, yeast cells ⁇ e.g., Saccharomyces, Pichia) transformed with a recombinant yeast expression vector containing the HIF-l ⁇ coding sequence; insect cells infected with a recombinant virus expression vector ⁇ e.g., baculovirus) containing the HIF-l ⁇ coding sequence; plant cells infected with a recombinant virus expression vector ⁇ e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with a recombinant plasmid expression vector ⁇ e.g., Ti plasmid) containing the HIF- l ⁇ coding sequence; or mammalian cells ⁇ e.g., COS, CHO, BHK, 293, 3T3) harboring recombinant expression constructs containing promoters derived from
  • a number of expression vectors may be advantageously selected depending upon the use intended for the HIF-I ⁇ being expressed.
  • vectors that direct the expression of high levels of protein products that are readily purified may be desirable.
  • Such vectors include but are not limited to the E. coli expression vector pUR278 (Ruther et al, 1983, EMBO J. 2:1791), in which the HIF-l ⁇ coding sequence can be ligated into the vector in-frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, 1985, Nucleic Acids Res.
  • pGEX vectors can also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to a column comprising of glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include, e.g., thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) can be used as a vector to express foreign genes.
  • the virus grows in Spodoptera frugiper da cells.
  • the HIF- l ⁇ coding sequence can be cloned individually into nonessential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • Successful insertion of a HIF- l ⁇ coding sequence will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the polyhedrin gene).
  • These recombinant viruses can be used to infect Spodoptera frugiper da cells in which the inserted gene is expressed (e.g., see Smith et at, 1983, J. Virol. 46:584; Smith, U.S. Patent No. 4,215,051).
  • the HIF- l ⁇ coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination.
  • Insertion in a non-essential region of the viral genome will result in a recombinant virus that is viable and capable of expressing HIF- l ⁇ in infected hosts (see, e.g., Logan & Shenk, 1984, Proc. Natl. Acad. ScL USA 81:3655).
  • Specific initiation signals may also be required for efficient translation of inserted HIF- l ⁇ coding sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where an entire HIF-I ⁇ gene, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed.
  • exogenous translational control signals including, if necessary, the ATG initiation codon
  • exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic.
  • the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure correct translation of the entire insert.
  • the efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. ⁇ see Bittner ⁇ t al, 1987. Methods in Enzvmol. 153: 516).
  • a host cell strain may be chosen that modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications ⁇ e.g., glycosylation) and processing ⁇ e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • eukaryotic host cells that possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product can be used.
  • Such mammalian host cells include but are not limited to CHO 5 VERO, BHK, HeLa, COS, MDCK, 293, 3T3, WI38, and in particular, breast cancer cell lines such as, for example, BT483, Hs578T, HTB26, BT20 and T47D, and normal mammary gland cell lines such as, for example, CRL7030 and Hs578Bst.
  • HIF- l ⁇ gene product For long-term, high-yield production of recombinant proteins, stable expression is preferred.
  • cell lines that stably express the HIF- l ⁇ gene product can be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements ⁇ e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • engineered cells can be allowed to grow for 1-2 days in an enriched media, and then can be switched to a selective media.
  • a selectable marker in a recombinant construct such as a plasmid, can confer resistance to the selective media, allow cells to stably integrate the plasmid into their chromosomes, and grow to form foci which, in turn, can be cloned and expanded into cell lines.
  • This method can advantageously be used to engineer cell lines that stably express the HIF- l ⁇ gene product.
  • Such engineered cell lines can be particularly useful in screening and evaluating compounds that affect the endogenous activity of the HIF- 1 ⁇ gene product.
  • a number of selection systems including but not limited to the herpes simplex virus thymidine kinase (Wigler etal, 1977, Cell 11: 223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, 1962, Proc. Natl. Acad. ScL USA 48: 2026), and adenine phosphoribosyltransferase (Lowy et ah, 1980, Cell 22: 817) genes can be employed in tk " , hgprf or aprt " cells, respectively.
  • anti-metabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al, 1980, Proc Natl. Acad. ScL USA 77: 3567; O'Hare et al, 1981, Proc. Natl. Acad. ScL USA 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, 1981, Proc. Natl. Acad. ScL USA 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al, 1981, J. MoI. Biol. 150: 1); and hygro, which confers resistance to hygromycin (Santerre et al., 1984, Gene 30: 147).
  • the methods and kits of the invention encompass use of anti- HIF- l ⁇ antibodies or fragments thereof that specifically recognize one or more epitopes of a HIF- l ⁇ protein. Accordingly, any HIF-I ⁇ protein, derivative, or fragment can be used as an immunogen to generate antibodies that immunospecifally bind a HIF- l ⁇ protein. Such antibodies and fragments can be used in the detection and quantitation of a HIF- l ⁇ in a sample to carry out any of the methods of the invention as disclosed herein.
  • Such antibodies can include but are not limited to polyclonal antibodies, monoclonal antibodies (mAbs), humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab') 2 fragments, Fv fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • mAbs monoclonal antibodies
  • humanized or chimeric antibodies single chain antibodies
  • Fab fragments F(ab') 2 fragments, Fv fragments, fragments produced by a Fab expression library
  • anti-idiotypic (anti-Id) antibodies anti-idiotypic antibodies
  • epitope-binding fragments of any of the above In a specific embodiment, antibodies to human HIF- l ⁇ protein are used. Described herein are general methods for the production of antibodies or immunospecific fragments thereof. Any of such antibodies or fragments can be produced by standard immunological methods or by recombinant expression of nucleic acid molecules
  • any of various host animals can be immunized by injection with a HIF- l ⁇ gene product, or a portion thereof.
  • host animals can include but are not limited to rabbits, mice, and rats.
  • Various adjuvants can be used to increase the immunological response depending on the host species, including but not limited to Freund's (complete or incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, dinitrophenol or potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum.
  • BCG Bacille Calmette-Guerin
  • Anti- HIF- l ⁇ monoclonal antibodies are preferred for use in the methods and kits of the invention.
  • Monoclonal antibodies can be obtained by any technique that provides for the production of antibody molecules by continuous cell lines in culture. These include but are not limited to the hybridoma technique of Kohler and Milstein, (1975, Nature 256: 495; and U.S. Patent No. 4,376,110), the human B-cell hybridoma technique (Kosbor et al, 1983, Immunology Today 4: 72; Cole et al, 1983, Proa Natl. Acad. Sci. USA 80: 2026), and the EBV-hybridoma technique (Cole et al. , 1985, Monoclonal Antibodies And Cancer Therapy, Alan R.
  • Such antibodies can be of any immunoglobulin class, including IgG, IgM, IgE, IgA, IgD, and any subclass thereof.
  • the hybridoma producing the mAb of this invention can be cultivated in vitro or in vivo.
  • chimeric antibodies By splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used in preparing antibodies useful in the present invention.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region.
  • the invention thus contemplates chimeric antibodies that are specific or selective for a HIF- l ⁇ protein. While often designed to be therapeutic, such chimeric antibodies can be useful to quantitate a HIF- l ⁇ level according to the methods of the invention.
  • humanized antibodies can be used in the methods and kits of the invention.
  • humanized antibodies are antibody molecules from non-human species having one or more hypervariable regions or complementarity determining regions (CDRs) from the non-human species and framework regions from a human immunoglobulin molecule.
  • CDRs complementarity determining regions
  • the extent of the framework region and Cars have been precisely defined (see, "Sequences of Proteins of Immunological Interest", Kabat, E. et al, U.S. Department of Health and Human Services (1983)).
  • Examples of techniques that have been developed for the production of humanized antibodies are known in the art and useful within the scope of the present invention. (See, e.g., Queen, U.S. Patent No. 5,585,089 and Winter, U.S. Patent No.
  • Humanized antibodies are typically developed as therapeutic agents. However, such antibodies can also be used in the methods and kits of the present invention, as they can be used to quantitate a HIF- l ⁇ level in accordance with the instant invention.
  • Phage display technology can be used to increase the affinity of an antibody to a HIF- l ⁇ gene product. This technique can be useful in obtaining higher affinity antibodies to an HIF- l ⁇ gene product, which could be used for the diagnosis and prognosis of a subject with cancer according to the present invention.
  • affinity maturation employs mutagenesis or CDR walking and re-selection using the HIF- l ⁇ gene product antigen to identify antibodies that bind with higher affinity to the antigen when compared with the initial or parental antibody (see, e.g.,G ⁇ ase ⁇ et ⁇ l., 1992, J. Immunology 149:3903). Mutagenizing entire codons rather than single nucleotides results in a semi-randomized repertoire of amino acid mutations. Libraries can be constructed consisting of a pool of variant clones, each of which differs by a single amino acid alteration in a single CDR, and contain variants representing each possible amino acid substitution for each CDR residue.
  • Mutants with increased binding affinity for the antigen can be screened by contacting the immobilized mutants with labeled antigen. Any screening method known in the art can be used to identify mutant antibodies having increased avidity to the antigen (e.g., ELISA) (see ' ⁇ u et ⁇ l, 1998, Proc N ⁇ tl. Ac ⁇ d ScL USA 95:6037; Y ⁇ Xon et ⁇ l, 1995, J Immunology 155:1994). CDR walking that randomizes the light chain may also be useful (see Schier et ⁇ l, 1996, J. MoI Bio. 263:551).
  • ELISA ELISA
  • single chain antibodies can be adapted to produce single chain antibodies against HIF- l ⁇ gene products.
  • Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.
  • Techniques for the assembly of functional Fv fragments in E. coli can also be used (Skerra et al, 1988, Science 242: 1038).
  • Antibody fragments that recognize specific epitopes can be generated by known techniques. Such fragments can be used for quantitating a HIF- l ⁇ gene product according to any available method known in the art.
  • fragments include but are not limited to: F(ab') 2 fragments, which can be produced by pepsin digestion of the antibody molecule; and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab') 2 fragments; Fab fragments, which can be generated by treating the antibody molecule with papain and a reducing agent; and Fv fragments.
  • Fab expression libraries can be constructed (Huse et al, 1989, Science 246:1275-1281) to allow rapid and easy identification of monoclonal Fab fragments having the desired specificity.
  • a molecular clone of an antibody to an antigen of interest can be prepared by techniques known to one skilled in the art. Recombinant DNA methodology ⁇ see e.g., Maniatis et al., 1982, Molecular Cloning. A Laboratory Manual. Cold Spring Harbor Laboratory, Cold Spring Harbor, New York) can be used to construct nucleic acid sequences that encode a monoclonal antibody molecule, or an immunospecific fragment thereof.
  • Antibody molecules can be purified by well-known techniques, e.g., immunoabsorption or immunoaffinity chromatography, chromatographic methods such as HPLC (high performance liquid chromatography), or a combination thereof.
  • screening for the desired antibody can be accomplished by techniques known in the art, e.g. , ELISA (enzyme-linked immunosorbent assay).
  • ELISA enzyme-linked immunosorbent assay
  • generated hybridomas can be assayed for a product that binds to a HIF- l ⁇ fragment containing such domain.
  • the foregoing antibodies can be used to quantify a HIF- l ⁇ protein, e.g., to measure levels thereof in appropriate samples, in the methods and kits of the invention.
  • HIF-I ⁇ Any antibody directed to one or more epitopes of an HIF-I ⁇ can be used in the methods and kits of the invention.
  • Commercially available HIF-l ⁇ antibodies can be used in accordance with the instant invention, for example those available from Novus Biologicals, Inc. (Littleton, CO); Affinity BioReagents, (Golden, CO). 6.9 KITS
  • the invention provides a pharmaceutical pack or kit comprising one or more containers filled with compounds of the invention. Additionally, one or more other prophylactic or therapeutic agents useful for the treatment of a disease can also be included in the pharmaceutical pack or kit.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the present invention provides kits that can be used in the above methods. In one embodiment, a kit comprises one or more compounds of the invention.
  • kits further comprises one or more other prophylactic or therapeutic agents useful for the treatment of cancer, in one or more containers.
  • a kit further comprises one or more anti-cancer agents that bind one or more cancer antigens associated with cancer.
  • the other prophylactic or therapeutic agent is a chemotherapeutic.
  • the prophylactic or therapeutic agent is a biological or hormonal therapeutic.
  • MRE 3046F20 5-N-(4-methylphenyl- carbamoyl)amino-8-methyl-2(2-furyl)-pyrazolo-[4,3e]-l,2,4-triazolo[l,5-c]pyrimidine; IL-10 salt, 5-N-(4-diethylamino-phenyl-carbamoyl)amino-8-methyl-(2(2furyl)-pyrazolo- ⁇ ejl ⁇ -triazolotl ⁇ -clpyrimidine); MRE 3008F20, 5-N-(4-methoxyphenyl- carbamoyl)amino-8-propyl-2(2furyl)-pyrazolo-[4,3e] 1 ,2,4-triazolo[ 1 ,5-c]pyrirnidine, MRE 3055F20, 5-N- (4-phenylcarbamoyl)amino- 8-propyl- 2 (2furyl) -pyr
  • RNAse was purchased from Boehringer.
  • Cell Culture For chemotherapeutic enhancement testing, the human melanoma A375, human lung carcinoma SKMES, colon carcinoma HT29, and pancreatic cancer Panc-1 cell lines were originally obtained from the ATCC (American Type Culture Collection) and have been maintained at PRC.
  • Cell lines are maintained in RPMI-1640 medium supplemented with 100 units/ml penicillin G sodium, 100 ⁇ g/ml streptomycin sulfate, 0.25 ⁇ g/ml amphotericin B (fungizone), 2 mM glutamine, 10 mM HEPES, 25 ⁇ g/ml gentamycin, and 10% heat-inactivated fetal bovine serum.
  • the cells are cultured in a T25 Falcon Tissue Culture flask in a humidified incubator at 37°C with 5% CO 2 -95% air. The cells are sub-cultured twice a week.
  • the doubling times of A375, SKMES 3 HT29, and Panc-1 cultures are approximately 22, 45, 48, and 60 hr, respectively.
  • A375 andNCTC2544 cells are grown adherently and maintained in DMEM and EMEM medium, respectively, containing 10% fetal calf serum, penicillin (100 U/ml), streptomycin (100 ⁇ g/ml), L-glutamine (2 mM) at 37°C in 5% CO 2 /95% air.
  • HeL023 and Jurkat were grown in RPMI-1640 medium, containing 10% fetal calf serum, penicillin (100 U/ml), streptomycin (100 ⁇ g/ml), L- glutamine (2 mM) at 37°C in 5% CO 2 /95% air.
  • Cells are passaged two or three times weekly at a ratio between 1 :5 and 1:10.
  • Lymphocytes were isolated from buffy coats kindly provided by the Blood Bank of the University Hospital of Ferrara. Blood donated by healthy volunteers, after informed consent for research was obtained. Lymphocytes were isolated by density gradient centrifugation (Ficoll/Histopaque 1.077 g/ml). The cells are stimulated with purified phytohemoagglutinin (1 ⁇ g/ml) and expanded in RPMI medium added of interleukin 2 (20 Units/ml) and 10% fetal calf serum.
  • Colony Formation Assay Exponentially growing A375 cells were seeded at 300 cells per well in six- well plates with 2 ml of fresh medium and treated with paclitaxel, vindesine and adenosine receptor agonists and antagonists dissolved in DMSO solution. Control plates received the same volume of DMSO alone. After 7 days of growth at 37°C in humidified atmosphere containing 5% CO 2, the cells were fixed with absolute methanol for 5' and stained with 1/10 Giemsa/phosphate-buffered saline (PBS) staining solution for 10 minutes. Staining solution was removed and colonies of greater than 30 cells were scored as survivors. For each treatment, six individual wells were scored.
  • PBS Giemsa/phosphate-buffered saline
  • A375 and NCTC2544 adherent cells were trypsinized, mixed with floating cells, washed with PBS and permeabilised in 70% (vol/vol) ethanol/PBS solution at 4 0 C for at least 24 hours.
  • Jurkat, HeL023 and PBMC cells were centrifuged for 10 minutes at 1000 x g. The cell pellet was then resuspended and permeabllised in 70% (vol/vol) ethanol/PBS solution at 4°C for at least 24 hours.
  • the cells were washed with PBS and the DNA was stained with a PBS solution, containing 20 ⁇ g/ml of propidium iodide and 100 ⁇ g/ml of RNAse, at room temperature for 30 minutes.
  • Cells were analysed by FACScan (Becton-Dickinson) and the content of DNA was evaluated by the CeIl-LISYS program (Becton-Dickinson). Cell distribution among cell cycle phases and apoptotic cells was evaluated as previously described (Secchiero et al., 2001). Briefly, the cell cycle distribution is shown as the percentage of cells containing 2n (G 1 phase), 4n (G 2 and M phases), 4n>x>2n. DNA amount (S phase) judged by propidium iodide staining. The apoptotic population is the percentage of cells with DNA content lower than 2n.
  • Rhodamine 123 Efflux Assay 5x10 s cells from each subline were loaded with 50 ng/ml of rhodamine 123 for 30 minutes at 37C in medium. The cells were washed and resuspended in dye-staining agent free medium for 3 hours at 37 C. to allow rhodamine 123 efflux. The cells were then washed twice and the fluorescence of intracellular rhodamine 123 were analysed with Flow cytometer (residual fluorescence FRES). The fluorescence was compared with loaded cells maintained at 4 C. to prevent drug export (maximal fluorescence FMAX).
  • the cells were treated with adenosine receptor antagonists to evaluate the ability of adenosine receptors to interfere with P-gp drug efflux activity. Cells from each subline that were not being exposed to rhodamine 123 were used as negative controls.
  • Metabolic Inhibitors Cells were treated for 30 minutes with metabolic inhibitors or with drug vehicle (DMSO) prior to being challenged with adenosine receptor antagonists and paclitaxel or vindesine. After 24 hours, cells were collected for flow cytometry analysis.
  • PD 98059 was used at 20 uM as an inhibitor of MEK to prevent MEK- 1 activation.
  • UOl 26 was used at 10 uM as inhibitor of MEK-I and MEK-2 to prevent extracellular signal-regulated kinase ERK-I and ERK-2 activation.
  • SB 203580 was used at 1 uM as an inhibitor of p38MAP kinase (p38MAPK).
  • Inventors have verified the concentration of adenosine A 3 receptors in human cancer cells that is elevated in the cancerous tumors compared to non-cancerous tumors or normal tissue.
  • Table 8 and FIG. 1 indicate elevated levels of adenosine A 3 receptors found by the inventors in human A375 (human melanoma), Panc-1 (human pancreatic carcinoma), MX-I (human breast carcinoma), PC-I (human prostate carcinoma), HT29 (human colon carcinoma), and SKMES (human lung carcinoma) and A2780AD10 (human ovarian carcinoma).
  • a 3 receptor antagonists Three A 3 receptor antagonists have been tested for their Degree of Growth Inhibitory activity and enhancement of inhibitory growth function of known anti-neoplastic agents.
  • Table 6 indicates receptor binding assay results for three A 3 receptor antagonist compounds. Structures of the three compounds are as follow:
  • MRE3008F20 5-[[(4-Methoxyphenyl)amino]carbonyl]amino-8-propyl-2-(2-furyl)- pyrazolo[4,3-e] 1 ,2,4-triazolo[l,5-c]pyrimidine
  • IL- 10 N- l-(4-diethylamino-phenyl)-N'-S-[8-methyl-2-(2-furyl)-pyrazolo[4,3-e] l,2,4-triazolo[ l,5-c]pyrimidine]-urea H
  • IL- 11 N-l-(4-dimethylamino-phenyl)-N'-5-[8-methyl-2-(2-&ryl)-pyrazolo[4,3-e] ,2,4 -triazolofl ,5-c]pyrimidine]-urea
  • Table 6 shows MRE3008F20, IL-IO and IL-11 to be potent, selective antagonists for the human adenosine A 3 receptor.
  • Irinotecan HCl (Camptosar®; 20 mg/ml) was obtained from Pharmacia & Upjohn Co.
  • Paclitaxel (Taxol®, 6 mg/ml) was obtained from Bristol-Myers Squibb, Co.
  • Docetaxel (Taxotere®) was obtained from Rhone-Poulenc Rorer.
  • Vinblastine sulfate salt was obtained from Sigma Chemical Co., (V1377).
  • Irinotecan was diluted with culture medium. All other agents were dissolved in 100% DMSO at appropriate concentrations.
  • DMSO stock solutions were diluted 100-fold with growth medium so that the final DMSO concentration was 1%.
  • MTT (3 -[4,5- Dimethylthiazol-2-yl]2,5-diphenyltetrazolium bromide) was obtained from Sigma Chemical Co.
  • RPMI-1640 medium antibiotic antimycotic 100 X consisting of 10,000 units/ml penicillin G sodium, 10,000 ⁇ g/ml streptomycin sulfate, 25 ⁇ g/ml Amphotericin B (fungizone), glutamine (20OmM), HEPES buffer (1 M), gentamicin (50 mg/ml), sodium bicarbonate (7.5%), and fetal bovine serum were obtained from GibcoBRL. The complement in fetal bovine serum was inactivated at 56°C for 30 min.
  • the human melanoma A375, human lung carcinoma SKMES, colon carcinoma HT29, and pancreatic cancer Panc-1 cell lines were originally obtained from the ATCC (American Type Culture Collection) and have been maintained in RPMI-1640 medium supplemented with 100 units/ml penicillin G sodium, 100 ⁇ g/ml streptomycin sulfate, 0.25 ⁇ g/ml amphotericin B (fungizone), 2 mM glutamine, 10 mM HEPES, 25 ⁇ g/ml gentamycin, and 10% heat-inactivated fetal bovine serum.
  • HT29 is approximately 30-fold more refractory than the A375 cell line to doxorubicin (IC 50 value of 0.211 vs. 0.0064 ⁇ g/ml) and mitoxantrone (IC 50 value of 0.1 vs. 0.0032 ⁇ g/ml), respectively.
  • the HT29 cell line has an altered form of topoisomerase II, and is therefore more refractory to doxorubicin and mitoxantrone, which mediate their growth inhibitory activity by trapping topoisomerase II, DNA, and drug in ternary complexes.
  • Inventors verified the concentration of adenosine A 3 receptors in human cancer cells that is elevated in the cancerous tumors compared to non-cancerous tumors or normal tissue.
  • Table 8 and Figure 1 indicate elevated levels of adenosine A 3 receptors found by the inventors in human A375 (human melanoma), Panc-1 (human pancreatic carcinoma), MX-I (human breast carcinoma), PC-I (human prostate carcinoma), HT29 (human colon carcinoma), and SKMES (human lung carcinoma).
  • the growth inhibitory activity of paclitaxel against the human melanoma A375 cell line was determined in the absence or in the presence of 10 ⁇ g/ml of MRE3008F20 or 5 ⁇ g/ml each of IL-10 and IL-Il (Table 9). At these sub-cytotoxic concentrations, MRE3008F20, IL-10, and IL-Il (approximately 30-45% growth inhibition in the presence of A 3 antagonists alone), enhanced the growth inhibitory activity of paclitaxel by 8-12-fold; IC 50 values decreased from 0.0046 ⁇ g/ml (paclitaxel alone) to 0.0004-0.00054 ⁇ g/ml (paclitaxel plus MRE3008F20, IL-10, and IL-11).
  • IL-10 and IL-11 enhance the growth inhibitory activity of paclitaxel in a concentration-dependent manner.
  • Results of further testing of the enhancement effects with taxane family compounds is given in Table 10. Both of the common taxane compounds paclitaxel (taxolTM) and docetaxel (taxotereTM) are included.
  • Table 10 illustrates concentration-dependencies for concentrations of 1, 3, and 10 ⁇ g/ml for MRE3008F20 and for concentrations of 0.5, 1.5, and 5 ⁇ g/ml for compounds IL- 10 and IL- 11.
  • IL-10 and IL-11 enhanced the growth inhibitory activity of paclitaxel in a concentration-dependent manner. This testing also indicated that compounds MRE3008F20, IL-10 and IL-Il have enhancement factors with docetaxel that are indicative of a synergistic effect.
  • Table 11 Growth Inhibitory Activity of A 3 Antagonists and Anti-Neoplastic Agents used Jointly with SKMES Cells
  • the growth inhibitory activity of paclitaxel and docetaxel against the human colon carcinoma HT29 cell line was determined in the absence or in the presence of 1 ⁇ g/ml of MRE3008F20 or 0.5 ⁇ g/ml each of IL-10 and IL-Il (Table 12). It was found that A 3 antagonists enhance the growth inhibitory activity of paclitaxel, docetaxel, doxorubicin and irinotecan.
  • Table 12 Growth Inhibitory Activity of A 3 Antagonists and Taxan Compounds Used Jointly With HT29 Cells
  • chemotherapeutic cancer agents can be selected for A 3 antagonist enhancement from the form of MDR they are associated with, the inventors have further established that high affinity A 3 adenosine receptor antagonists can be used to counter P-gp and MRP associated multi-drug resistance.
  • inventors evaluated whether A 3 adenosine receptor could protect the cell by the toxic effect of conventional chemotherapeutic drug. Colony formation assay experiments were performed on A375 melanoma cells treated with increasing concentration of paclitaxel (0.25-75 ⁇ g/ml). A 3 stimulation is achieved with the selective agonist CI-IB- MECA while A 3 blockade is obtained with the selective antagonist MRE 3008F20.
  • Colony formation of A375 cells is increased of about 30% when the adenosine A 3 agonist CI-IB-MECA (10 ⁇ M) is applied. This is seen in the "C" bar of Figure 2 being 130% of the DMSO control bar "D" Figure 2 further shows that increased colony formation occurs when CI-IB-MECA is combined with the taxane family compound paclitaxel (0.75 ⁇ g/ml). When paclitaxel alone is added ("T" bar of Figure 2), colony formation is 64% of the control. Colony formation is increased 32% to 85% of control when the CI-IB-MECA is combined ("TC" bar of Figure 2).
  • a 3 receptors trigger a pro-survival signal, not able to restore colony formation ability of A375 cells treated with the taxane paclitaxel, while the blockage of As increased paclitaxel mediated deleterious effects (p ⁇ 0.01, "TM" bar versus "T” bar of Figure 2).
  • the inventors also analysed the ability of A 3 adenosine receptor antagonists to enhance chemotherapeutic effects by performing an acute treatment of A375 cells with the taxane family compound paclitaxel and the vinca alkaloid vindesine.
  • Cell proliferation and apoptosis are quantified by flow cytometer analysis after propidium-iodide (PI) DNA staining.
  • PI propidium-iodide
  • EC 50 concentration exerting the 50% of the G 2 /M accumulation (EC 50 ).
  • EC 50 concentration exerting the 50% of the G 2 /M accumulation (EC 50 ).
  • EC 50 are 16.60+2.00 ng/ml and 1.90+0.20 nM for paclitaxel and vindesine, respectively (means of four experiments).
  • Analyses further show that EC50 of paclitaxel and vindesine for decrease of Gi population are not significantly changed respect to EC 50 of G 2 M. arrest.
  • the S-phase population, representative of replicating DNA, is not appreciably changed.
  • FIG. 3A and 3C show that MRE 3008F20 (10 ⁇ M) improved vindesine and paclitaxel ability to alter cell proliferation: MRE 3008F20 reduced paclitaxel and vindesine EC 50 of 1.9 and 4.0 fold, respectively. Similar results were obtained analysing EC 50 values calculated for the G 1 population.
  • MRE 3008F20 (10 ⁇ M) reduced EC MAX of 2.0 and 2.1 fold, for paclitaxel and vindesine, respectively (Figure 3B and 3D).
  • Representative flow cytometry profiles of DNA content in A375 cells (Apo (sub diploid cells), G 1 , S and G 2 /M phases) are shown in Figure 4.
  • Figure 4C shows that, under both treatments (A 3 antagonist MRE3008F20 plus vinca alkaloid vindesine), vindesine response increased as cells progressed from G 1 to G 2 /M phases respect to vindesine-treated cells alone (Figure 4B). Similar results were obtained with the taxane compound paclitaxel (Figure 4E-F).
  • A375 cells were treated with vindesine (1 nM) with increasing concentrations of adenosine receptor antagonists (MRE3055F20, MRE3062F20, MRE3046F20, MRE3008F20, IL-IO, CGS 15943, ZM 241385).
  • MRE3055F20 highest
  • MRE3062F20 MRE 3046F20
  • MRE3008F20 MRE3008F20
  • IL-10 adenosine receptor antagonists
  • Table 14 Adenosine receptor antagonist parameters of enhancing activity to vindesine and binding affinity to A 3 adenosine receptor in A375 cells.
  • SEC 50 adenosine receptor antagonist dose that induces 50% of the enhancing activity to vindesine (1 nM), calculated on G 2 /M accumulation dose response curve. Ki equilibrium constant of binding affinity at human A 3 adenosine receptor. Data represents the mean of four independent experiments.
  • HeLa23 cell line has the lowest sensitivity to paclitaxel and vindesine alone. Interestingly, HeLa23 cells also show the greatest amount of enhancement by MRE3008F20 co-treatment.
  • MRE3008F20 is 10 ⁇ M.
  • EC 50 values were obtained by analysing G 2 /M accumulation dose response curve.
  • EC MAX values were obtained by analysing SUb-G 1 accumulation dose response curve.
  • Paclitaxel values have units of ng/ml.
  • Vindesine values have units of nM. nd: not done.
  • the greater relative enhancement in the HeL023 cell line may be related to a factor absent in other cell lines or related to the intracellular concentration of actives that is modulated by P-glycoprotein drug expulsion activity.
  • Rhodamine 123 (Rhl23) retention a P-gp functional assay, was studied in all cell lines. This assay was performed by incubating cells with RhI 23 and determining Rhl23 accumulation by measuring its fluorescence. The maximum load of Rhl23 (F MA X) was quantified at the end of treatment harvesting the cells and storing them at 4°C to prevent any active Rhl23 efflux. The P-gp drug efflux was allowed incubating Rhl23- loaded cells in fresh new medium Rhl23-free for 3 hours at 37°C. After this incubation, residual fluorescence (F R E S ) was measured and compared to FM AX -
  • Figure 6A shows the flow chromatogram for RhI 23 accumulation by A375 cells when the adenosine A 3 antagonist is not present.
  • Two cell populations are found, characterised by an F RES having a mean fluorescence intensity lower than F MAX .
  • Figure 6B shows the A375 cellular accumulation of Rhl23 in the presence of MRE3008F20 (10 ⁇ M).
  • the response yielded a F RES chromatogram comparable to FMAX, consistent with a completely blockade of P-gp mediated Rh 123 transport.
  • the Jurkat and NCTC2544 cells studied did not appear to have a significant change of Rhl23 fluorescence after 3 hours of incubation at 37°C (F RES was similar to F MAX ) consistent with the absence or not detectable P-pg drug pumping activity in these cells.
  • the high affinity adenosine A 3 antagonists IL-IO, MRE3008F20, MRE3055F20, MRE3062F20 and MRE3046F20 are strong inhibitors of P-gp activity.
  • the low affinity antagonists, ZM241385 and CGS15943 had much lower effect on P-gp activity. This may be due to the higher A 3 affinity or due to the existence of a structure-activity relationship for the inhibition of P-gp drug expulsion activity mediated by adenosine receptor antagonists.
  • Table 16 data represents the mean of four independent experiments.
  • MAPK mitogen-activated protein kinase
  • PD98059 a selective inhibitor of MEK1/2 (dudley dt, PNAS 92:7686, 1995), was used to inhibit the MEK pathways.
  • UO 126 an agent approximately 100-fold more potent than PD98059, was used as inhibitor of ERK activation.
  • SB203580 selectively inhibits p38 MAPK activity (Young PR JBC 272:12116 1997).
  • A375 cells were pretreated with PD98059 (20 ⁇ M), U0126 (30 ⁇ M), SB203580 (1 ⁇ M] or with DMSO (as control) and 20 then challenged with vindesine (1 nM) or with paclitaxel (15 ng/ml). At 24 hours post treatment, cells were harvested and apoptosis and cell cycle were analysed.
  • U0126 prevented MRE3008F20-induced accumulation in G 2 /M by 23 ⁇ 5% and by 48 ⁇ 5% in presence of paclitaxel (bar "TU” of Figure 7A) and vindesine (bar “VU” 30 of Figure 7A), respectively. This infers that the molecular mediator of enhancement activity was ERK.
  • the A 3 adenosine receptor antagonists reduce EC5 0 doses of chemotherapeutic drugs (quantified by analysing G 2 /M accumulation rate) to 12.3, 1.9, 1.2-fold for paclitaxel and 36.3, 4.0, 2.5-fold for vindesine when challenged on HeL023, A375 and Jurkat cell lines, respectively.
  • This enhancement activity was confirmed also when the apoptotic degree was evaluated: EC MAX dose of chemotherapeutic drugs are reduced to 10.5, 2.0, 1.5-fold for paclitaxel on HeL023, A375 and Jurkat cells, respectively, and 31.5, 2.0-fold for vindesine on HeL023 and A375 cells, respectively.
  • variable degree of EC50 (and ECMA X ) value observed in different cell types suggest a cell type specific participation in drug-induced enhancement.
  • the prerequisite of the drug activity is its delivery to the target site.
  • efficiency of drug is limited by appearing resistance, i.e., lack of cell sensitivity to the administered drug.
  • the tumor cells with multidrug resistance (MDR) phenotype are characterised by lowered intracellular accumulation of the compounds they are resistant to.
  • P-gp membrane associated P-glycoprotein
  • MRP and not P-gp transporter is expressed in Jurkat leukemia cells (T lymphocytic cell line). This is consistent with the results as HeL023 and A375 cells produced a P-gp efflux-activity whereas in Jurkat cells had low rhodamine 123 efflux. It is shown that adenosine A 3 antagonists are useful for enhancement in both P-gp expressing and MRP expressing cell lines.
  • CGS 15943 and ZM241385 correlates with the lower adenosine A 3 affinity of these compounds.
  • the high affinity A 3 antagonist compounds tested have greater potency in both enhancement and inhibiting P- gp and MRP drug resistance. This may be due to the higher affinity or to the molecular structure of such compounds.
  • the tested high affinity compounds have a phenyl-carbarnoyl-amino derivative in the N5 position of a 2-furylpyrazolo-triazolo- pyrimidine structure.
  • CGS 15943 and ZM241385 in addition to having much lower A 3 affinity also have very different molecular structures.
  • the silicone fluid and active compound are mixed together and the colloidal silicone dioxide is added to increase viscosity.
  • the material is then dosed into a subsequent heat sealed polymeric laminate comprised of the following: polyester release liner, skin contact adhesive composed of silicone or acrylic polymers, a control membrane which is a polyolefin, and an impermeable backing membrane made of a polyester multilaminate.
  • the resulting laminated sheet is than cut into 10 sq. cm patches.
  • the active compound and the starch are granulated with water and dried. Magnesium stearate is added to the dried granules and the mixture is thoroughly blended. The blended mixture is compressed into tablets.
  • the active compound and buffering agents are dissolved in the propylene glycol at about 50°)C.
  • the water for injection is then added with stirring and the resulting solution is filtered, filled into ampules, sealed and sterilized by autoclaving.
  • Active compound 1Og Buffering Agents q.s. Water for injection q.s. 100OmL
  • U2OS osteosarcoma U87MG glioblastoma human cells were obtained from American Tissue Culture Collection (ATCC). Tissue culture media and growth supplements were obtained from BioWhittaker. GasPak PouchTM System was obtained from Becton Dickinson. Unless otherwise noted, all other chemicals were purchased from Sigma.
  • Anti- HIF- 1 ⁇ and anti-HIF 1 ⁇ antibodies mAb were obtained from Transduction Laboratories (BD, Milano, Italy).
  • U0126 inhibitor of MEK-I and MEK-2
  • SB202190 inhibitor of p38 MAP kinase
  • Anti-ACTIVE ® MAPK and anti-ERK 1/2 pAb
  • Phospho-p38 and p38 MAP Kinase antibodies were from Cell Signaling Technology.
  • Anti- Adenosine A 3 receptor (polyAb) was from Aviva Antibody Corporation.
  • Cell culture and hypoxia treatment Cells were maintained in DMEM
  • [ 3 Hl -Thymidine incorporation cell proliferation test: Cells were seeded in fresh medium with 1 uCi/ml of [ 3 H]-Thymidine in DMEM containing 10% fetal calf serum, penicillin (100 U/ml), streptomycin (100 ug/ml), L-glutamine (2 mM). After 24 hours of labelling, cells were trypsinised, dispensed in 4 wells of a 96 well plate, and filtered through Whatman GF/C glass-fiber filters using a Micro-Mate 196 cell harvester (Packard Instrument Company). The filter bound radioactivity was counted on Top Count Microplate Scintillation Counter (efficiency 57%) with Micro-Scint 20.
  • A375 adherent cells were trypsinized, mixed with floating cells, washed with PBS and permeabilized in 70% (vol/vol) ethanol/PBS solution at 4 0 C for at least 24 hours. The cells were washed with PBS and the DNA was stained with a PBS solution, containing 20 ug/ml of propidium iodide and 100 ug/ml of RNAse, at room temperature for 30 minutes. Cells were analysed with an EPICS XL flow cytometer
  • the apoptotic population is the percentage of cells with DNA content lower than 2n.
  • Small interfering RNA (siRNA) design To generate a small interfering RNA that targets A 3 receptor mRNA (S ⁇ RNAA3), eight oligonucleotides consisting of ribonucleosides, except for the presence of 2'-deoxyribonucleosides at the 3' end, were synthesized and annealed, according to the recommendations of Elbashir (ref), to the manufacturer's instructions (SilencerTM siRNA Construction Kit, Ambion) and as previously described (Mirandola, 2004).
  • SiRNA Small interfering RNA
  • oligo-1 sense sequence: 5'-GCU UAC CGU CAG AUA CAA GUU-3' (SEQ ID NO:1) and antisense 5'-CUU GUA UCU GAC GGU AAG CUU-3' (SEQ ID NO:2).
  • sense sequence 5'-GAC GGC UAA GUC CUU GUU UUU-3' (SEQ ID NO:3) and antisense 5'-AAA CAA GGA CUU AGC CGU CUU-3' (SEQ ID NO:4).
  • oligo-3 sense sequence: 5'-ACA CUU GAG GGC CUG UAU GUU-3' (SEQ ID NO:5) and antisense 5'-CAU ACA GGC CCU CAA GUG UUU-3' (SEQ ID NO:6).
  • sense sequence 5'-CCU GCU CUC GGA GGA UGC CUU- 3' SEQ ID NO:7
  • antisense 5'-GGC AUC CUC CGA GAG CAG GUU-3' SEQ ID NO:8.
  • Target sequences were aligned to the human genome database in a BLAST search to ensure sequences without significant homology to other genes.
  • the target sequences for oligo-1, oligo-2, oligo-3 and oligo-4 are localized at position 337, 679, 1009 and 1356 bases downstream of the start codon of A 3 receptor mRNA sequence (L20463), respectively.
  • A375 cells were plated in six- well plates and grown to 50-70% confluence before transfection. Transfection of siRNA was performed at a concentration of 100 nM using RNAiFectTM Transfection Kit (Qiagen). Cells were cultured in complete media and at 24, 48 and 72 hours total RNA was isolated for Real- Time RT-PCR analysis of A 3 receptor mRNA and for Western blot analysis of A 3 receptor protein. At 48 hours from the transfection, A375 cells were serum starved for another 24 hours and then exposed to increasing concentrations of the A 3 adenosine receptor agonist Cl-IB-MECA for 4 hours in hypoxia. Total protein were then harvested for Western blot analysis.
  • RNAiFectTM Transfection reagent without SiRNAA 3 .
  • siRNA-FITC labelled Qiagen. After 24 hours of transfection, cells were tripsinized and resuspended in PBS for flow cytometry analysis. Fluorescence obtained from FITC-siRNA transfected cells was compared to autofluorescence generated by untransfected control.
  • primer and probe sequences were used for real-time RT-PCR: A 3 forward primer, 5'-ATG CCT TTG GCC ATT GTT G-3' (SEQ ID NO:9); A 3 reverse primer, 5'-ACA ATC CAC TTC TAC AGC TGC CT-3' (SEQ ID NO: 10); A 3 MGB probe, 5'-FAM-TCA GCC TGG GCA TC-TAMRA-3' (SEQ ID NO:11); for the real-time RT-PCR of the HIF-l ⁇ gene the assays-on-demandTM Gene expression Product Accession No. NM 019058 was used (Applied Biosystems, Monza, Italy).
  • the fluorescent reporter FAM and the quencher TAMRA are 6-carboxy fluorescein and 6-carboxy-N,N,N',N'-tetramethylrhodamme, respectively.
  • the endogenous control human ⁇ -actin kit was used, and the probe was fluorescent-labeled with VICTM (Applied Biosystems, Monza, Italy).
  • Metabolic inhibitors Cells were treated for 30 minutes with metabolic inhibitors or with drug vehicle (DMSO) prior to being challenged with adenosine or adenosine analogues.
  • U0126 was used at 10 and 30 uM as inhibitor of MEK-I and MEK-2 to prevent p44 and p42 MAPK activation.
  • SB202190 was used at 1 and 10 uM as inhibitor of p38 MAPK.
  • Adenosine induces HIF- l ⁇ protein accumulation in hypoxia.
  • adenosine receptors The family of adenosine receptors consists of four subtypes of G protein- coupled receptors, designed A 1 , A 2A , A 2B and A 3 . We have previously demonstrated that all four adenosine receptors are expressed in human melanoma A375 cells. To evaluate the functional role of adenosine receptor subtypes on HIF- 1 ⁇ protein expression under hypoxic conditions, we tested the effect of adenosine in combination with DPCPX (an Ai receptor antagonist), SCH 58261 (a selective A 2A receptor antagonist), MRE 2029F20 (a selective A 2B receptor antagonist), and MRE 3008F20 (a selective A 3 receptor antagonist) (Baraldi 2004; Merighi 2001; Varani 2000).
  • DPCPX an Ai receptor antagonist
  • SCH 58261 a selective A 2A receptor antagonist
  • MRE 2029F20 a selective A 2B receptor antagonist
  • MRE 3008F20 a selective A 3 receptor antagonist
  • a 3 receptor antagonist MRE 3008F20 abrogated the adenosine-induced increase of HIF-l ⁇ protein expression (Fig. lOC-D). Furthermore, HIF-I ⁇ expression was unaffected by adenosine or by synthetic adenosine receptor antagonists. These results indicate that adenosine may increase HIF-l ⁇ protein expression via A 3 receptors. A 3 adenosine receptor induces HIF-l ⁇ protein accumulation in hypoxia.
  • a 3 receptor stimulation did not affect the expression of HIF-I ⁇ protein, either in normoxia or in hypoxia.
  • HIF- l ⁇ protein increase induced by 10 nM Cl-IB-MECA
  • MRE 3008F20 0.3-30 nM
  • IC 50 0.90 ⁇ 0.08 nM
  • A375 cells were mock transfected or transfected with small interfering RNAs that target A 3 receptor mRNA (siRNA A3 ) for degradation.
  • A375 cells were also transfected with a siRNA control labeled with fluorescein. By flow cytometry we devisved a transfection efficiency of 85 ⁇ 5% (Fig. 7A). After transfection, the cells were cultured in complete media and at 24, 48 and 72 hours total RNA was isolated for Real-Time RT-PCR analysis ofA 3 receptor mRNA and for Western blot analysis of A 3 receptor protein. As expected, A 3 receptor mRNA levels were significantly reduced in cells transfected with SIRNA A3 (Fig. 14B). Furthermore, A 3 receptor protein expression was strongly reduced in siRNA ⁇ -treated cells (Fig. 14C-D). Neither mock transfection nor transfection with an siRNA targeted to an irrelevant mRNA inhibited A 3 receptor mRNA or protein expression.
  • A375 cells were exposed to increasing concentrations of the A 3 adenosine receptor agonist Cl-IB-MECA (1-100 nM) for 4 hours in hypoxia. Total protein were then harvested for Western blot analysis. As control, A375 cells were exposed to RNAiFectTM Transfection reagent without SIRNA A3 . We found that the inhibition of A 3 receptor expression is sufficient to block Cl-IB-MECA-induced HIF-l ⁇ accumulation (Fig. 14E).
  • A375 cells were pretreated with 10 ⁇ g/ml actinomycin D (Act-D) to inhibit de novo gene transcription. Then, A375 cells were cultured for 4 hours in hypoxia in the presence of increasing concentrations of Cl-IB-MECA (100 nM). We found that A 3 receptor stimulation was able to increase HIF-l ⁇ protein expression also in the presence of Act-D (Fig. 16). HIF- l ⁇ has been shown to be degraded through the proteasome pathway during normoxia. The enzymatic hydroxylation of proline 564 of HIF-I ⁇ controls the turnover of the protein by tagging it for interaction with the von Hippel Lindau protein (Ivan 2001; Jaakkola, 2001; Yu 2001; Maxwell 1999).
  • A375 cells were cultured in normoxia for 4 hours in the presence of 100 ⁇ M CoCl 2 , preventing oxygen-dependent HIF-I ⁇ protein degradation, and then A375 cells were treated with 100 nM Cl-IB-MECA in the presence or absence of CHX (1 ⁇ M). In cells exposed to CHX 5 Cl-IB-MECA failed to increase HIF-I ⁇ levels within 6 hours, as observed in the absence of CHX (Fig. 17B). Together, these results suggest that A 3 receptor activation increases HIF- l ⁇ protein levels through a translation-dependent pathway. After return of hypoxic A375 cultures to normoxia the levels of HIF-l ⁇ protein decreased very rapidly and were abrogated after 15 minutes (Fig. 18A).
  • phosphorylation of p44 and p42 was induced in response to nanomolar concentrations of Cl-IB-MECA and the induction of p44/p42 kinases phosphorylation status was maximum by the treatment of A 3 receptor agonist 100 nM (Fig. 19C). Furthermore, we have monitored the activation levels of p38 MAPK upon A 3 receptor stimulation by the detection of its phosphorylated form on Western blot. As can be seen in Fig. 19D, a strong increase in the phosphorylation of p38 MAPK was observed after 4 hours of A 3 receptor stimulation in hypoxia.
  • Hypoxia represents one of the first events in the growth of the cancer; this process creates conditions that, on one hand, are conducive to the accumulation of extracellular adenosine and, on the other hand, stabilize hypoxia-inducible factors, such as HIF-l ⁇ (Winn, 1981; Decking 1997; Ledoux 2003).
  • results of the present study indicate a new way by which hypoxia may contribute to cancer development, based on the natural pathways of adenosine receptor- mediated signaling.
  • adenosine is able to increase HIF- l ⁇ protein expression in response to hypoxia in a dose- and time-dependent manner in A375 human melanoma cells, whereas HIF-I ⁇ protein levels are not affected.
  • a 3 receptor inhibition at the mRNA and protein level is sufficient to block A 3 receptor-induced HIF- loc protein accumulation. Therefore, our results indicate that the cell surface A 3 adenosine receptor transduces extracellular hypoxic signals into the cell interior.
  • a 3 receptors are present in melanoma cells and their expression appears to be a bridge between the hypoxic insult and HIF- l ⁇ accumulation, regulating the cellular response to hypoxia, like an oxygen-sensing receptor. The extent to which A 3 receptor influences the ability of tumor cells to respond to hypoxia will require further investigation.
  • a 3 adenosine receptor stimulation had no effect on HIF- l ⁇ mRNA accumulation, as observed by Real-Time RT-PCR experiments. Accordingly, Act-D experiments indicate that A 3 receptor does not regulate HIF- l ⁇ protein expression through a transcription-dependent mechanism.
  • the lack of adenosine effect on HIF-I ⁇ at transcriptional level is not surprising in view of the fact that hypoxic regulation of HIF-I ⁇ is primarily determined by stabilization of HIF-l ⁇ protein (Huang, 1998).
  • a 3 adenosine receptors does not increase the half-life of HIF-l ⁇ protein while may increase the rate of HIF-l ⁇ protein synthesis, in a manner similar to the effect of various growth factors (Zhong 2000; Fukuda, 2002). Nevertheless, we cannot exclude the possibility that A 3 adenosine receptor regulates the translation of a protein, which inhibits HIF- l ⁇ degradation. Phosphorylation and dephosphorylation activities have been suggested to be critical in the signaling pathway leading to HIF-I activation.
  • hypoxia induces the phosphorylation of HIF-I ⁇ by p44/p42 and p38 MAPKs, which increases both HIF- l ⁇ nuclear localization and transcriptional activity
  • adenosine has been shown to directly enhance MAPK activity in A375 human melanoma cells (Merighi et al., 2002) but also in non human cell lines stably transfected with the human A 3 receptor (Hammarberg 2004; Schulte 2000-2002-2003).
  • HIF- l ⁇ is overexpressed in tumors as a result of hypoxia and is involved in key aspects of tumor biology, such as angiogenesis, invasion and altered energy metabolism (Ratcliffe 2000). It is recognized that the inhibition of HIF-I activity represents a novel therapeutic approach to cancer therapy, especially in combination with angiogenesis inhibitors, which would further increase intratumoral hypoxia and thus provide an even greater therapeutic window for use of an HIF-inhibitor. Recent studies indicate that pharmacologic inhibition of HIF-l ⁇ and particularly of HIF -regulated genes that are important for cancer cell survival may be more advantageous than HIF-gene inactivation therapeutic approaches (Mabjeesh et al., 2003). Many normal tissues function at p ⁇ 2 values sufficient to activate HIF, and the system has important functions under normal physiological conditions (Hopfl, 2004). This will need to be considered in the development of pharmacological inhibitors for clinical use.
  • a 3 adenosine receptor antagonists may be useful in cancer therapy.
  • a 3 receptor antagonists can block the survival of hypoxic solid tumors.
EP05858564A 2004-11-22 2005-11-22 Verbesserte behandlung von krebs und hif-1-vermittelten störungen mit adenosin-a3-rezeptorantagonisten Withdrawn EP1827445A2 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US63055704P 2004-11-22 2004-11-22
PCT/US2005/042551 WO2007040565A2 (en) 2004-11-22 2005-11-22 Enhancing treatment of cancer and hif-1 mediated disoders with adenosine a3 receptor antagonists

Publications (1)

Publication Number Publication Date
EP1827445A2 true EP1827445A2 (de) 2007-09-05

Family

ID=37906593

Family Applications (1)

Application Number Title Priority Date Filing Date
EP05858564A Withdrawn EP1827445A2 (de) 2004-11-22 2005-11-22 Verbesserte behandlung von krebs und hif-1-vermittelten störungen mit adenosin-a3-rezeptorantagonisten

Country Status (6)

Country Link
EP (1) EP1827445A2 (de)
JP (1) JP2008520746A (de)
AU (1) AU2005336924A1 (de)
CA (1) CA2586420A1 (de)
IL (1) IL183307A0 (de)
WO (1) WO2007040565A2 (de)

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2318649T3 (es) 2000-10-20 2009-05-01 EISAI R&D MANAGEMENT CO., LTD. Procedimiento de preparacion de derivados de 4-fenoxi quinolinas.
US7994159B2 (en) 2003-03-10 2011-08-09 Eisai R&D Management Co., Ltd. c-Kit kinase inhibitor
CN101337930B (zh) 2003-11-11 2010-09-08 卫材R&D管理有限公司 脲衍生物的制备方法
US8969379B2 (en) 2004-09-17 2015-03-03 Eisai R&D Management Co., Ltd. Pharmaceutical compositions of 4-(3-chloro-4-(cyclopropylaminocarbonyl)aminophenoxy)-7=methoxy-6-quinolinecarboxide
WO2007015578A1 (ja) 2005-08-02 2007-02-08 Eisai R & D Management Co., Ltd. 血管新生阻害物質の効果を検定する方法
CA2652442C (en) 2006-05-18 2014-12-09 Eisai R & D Management Co., Ltd. Antitumor agent for thyroid cancer
KR101472600B1 (ko) 2006-08-28 2014-12-15 에자이 알앤드디 매니지먼트 가부시키가이샤 미분화형 위암에 대한 항종양제
CA2676796C (en) 2007-01-29 2016-02-23 Eisai R & D Management Co., Ltd. Composition for treatment of undifferentiated gastric cancer
WO2009060945A1 (ja) 2007-11-09 2009-05-14 Eisai R & D Management Co., Ltd. 血管新生阻害物質と抗腫瘍性白金錯体との併用
AU2009210098B2 (en) * 2008-01-29 2013-06-13 Eisai R & D Management Co., Ltd. Combined use of angiogenesis inhibitor and taxane
WO2010054763A1 (de) * 2008-11-14 2010-05-20 Bayer Schering Pharma Aktiengesellschaft Heterocyclisch substituierte aryl-verbindungen als hif-inhibitoren
EP2586443B1 (de) 2010-06-25 2016-03-16 Eisai R&D Management Co., Ltd. Antitumorwirkstoff mit verbindungen mit kinasehemmungswirkung in kombination
CA2828946C (en) 2011-04-18 2016-06-21 Eisai R&D Management Co., Ltd. Therapeutic agent for tumor
ES2705950T3 (es) 2011-06-03 2019-03-27 Eisai R&D Man Co Ltd Biomarcadores para predecir y valorar la capacidad de respuesta de sujetos con cáncer de tiroides y de riñón a compuestos de lenvatinib
CN104755463A (zh) 2012-12-21 2015-07-01 卫材R&D管理有限公司 非晶态形式的喹啉衍生物及其生产方法
WO2014153363A1 (en) * 2013-03-18 2014-09-25 Northeastern University Method for generation of broadly neutralizing anti-pathogen antibodies
MX368099B (es) 2013-05-14 2019-09-19 Eisai R&D Man Co Ltd Biomarcadores para predecir y evaluar el grado de respuesta de sujetos con cancer de endometrio a compuestos de tipo lenvatinib.
JO3783B1 (ar) 2014-08-28 2021-01-31 Eisai R&D Man Co Ltd مشتق كوينولين عالي النقاء وطريقة لإنتاجه
DK3263106T3 (da) 2015-02-25 2024-01-08 Eisai R&D Man Co Ltd Fremgangsmåde til undertrykkelse af bitterhed af quinolinderivat
CA2978226A1 (en) 2015-03-04 2016-09-09 Merck Sharpe & Dohme Corp. Combination of a pd-1 antagonist and a vegfr/fgfr/ret tyrosine kinase inhibitor for treating cancer
CN107801379B (zh) 2015-06-16 2021-05-25 卫材R&D管理有限公司 抗癌剂
WO2021028382A1 (en) * 2019-08-12 2021-02-18 Bayer Aktiengesellschaft [1,2,4]triazolo[1,5-c]quinazolin-5-amines
WO2022029063A1 (en) 2020-08-04 2022-02-10 Bayer Aktiengesellschaft Pyrido[1,2,4]triazolo[1,5-c]pyrimidin-5-amines
CN114574580B (zh) * 2022-02-14 2022-11-29 山东大学 靶向a2br联合化疗在三阴性乳腺癌治疗中的应用

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6326390B1 (en) * 1998-08-25 2001-12-04 King Pharmaceuticals Reseach And Development, Inc. Use of adenosine A3 receptor antagonists to inhibit tumor growth
US6921825B2 (en) * 1998-09-16 2005-07-26 King Pharmaceuticuals Research & Development, Inc. Adenosine A3 receptor modulators
US6448253B1 (en) * 1998-09-16 2002-09-10 King Pharmaceuticals Research And Development, Inc. Adenosine A3 receptor modulators
IL133680A0 (en) * 1999-09-10 2001-04-30 Can Fite Technologies Ltd Pharmaceutical compositions comprising an adenosine receptor agonist or antagonist

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2007040565A2 *

Also Published As

Publication number Publication date
CA2586420A1 (en) 2007-04-12
WO2007040565A3 (en) 2009-04-16
JP2008520746A (ja) 2008-06-19
IL183307A0 (en) 2008-04-13
AU2005336924A1 (en) 2007-04-12
WO2007040565A9 (en) 2007-07-05
WO2007040565A2 (en) 2007-04-12

Similar Documents

Publication Publication Date Title
EP1827445A2 (de) Verbesserte behandlung von krebs und hif-1-vermittelten störungen mit adenosin-a3-rezeptorantagonisten
US20060204502A1 (en) Enhancing treatment of cancer and HIF-1 mediated disorders with adenosine A3 receptor antagonists
KR102356433B1 (ko) 치환된 아미노퓨린 화합물, 이의 조성물, 및 그것에 의한 치료 방법
JP7019422B2 (ja) 癌を治療するための方法
JP6957650B2 (ja) 血小板由来成長因子受容体アルファの遺伝的異常に関連する癌の治療のための、1−[4−ブロモ−5−[1−エチル−7−(メチルアミノ)−2−オキソ−1,2−ジヒドロ−1,6−ナフチリジン−3−イル]−2−フルオロフェニル]−3−フェニルウレアおよびアナログの使用
US20180360852A1 (en) System and method for diagnosis and treatment
TW201200149A (en) New uses of an immunomodulatory protein (GMI) from Ganoderma microsporum
TW201242601A (en) Methods of treating cancer using 3-(5-amino-2-methyl-4-oxo-4H-quinazolin-3-yl)-piperidine-2,6-dione
AU2013202305A1 (en) Methods for treating cancer using TOR kinase inhibitor combination therapy
Lee et al. Suppression of inflammation response by a novel A3 adenosine receptor agonist thio-Cl-IB-MECA through inhibition of Akt and NF-κB signaling
WO2009011897A1 (en) Combinations for the treatment of b-cell proliferative disorders
CN113194752A (zh) Pi3k相关疾病或病症的组合疗法
Visco et al. The cytotoxic effects of bendamustine in combination with cytarabine in mantle cell lymphoma cell lines
CN112512527A (zh) 恩扎妥林和btk抑制剂的组合及其用途
CN114727996A (zh) 用于治疗白血病或骨髓增生异常综合征的与维奈托克、吉瑞替尼、米哚妥林或其他化合物组合的阿扎胞苷
JP2023080357A (ja) リンパ球悪性疾患を治療するための方法
JP2018518524A (ja) 物質使用障害、依存症及び精神障害の治療のための組成物及び方法
US20110151020A1 (en) Active substance combination with gemcitabine for the treatment of epithelial cancer
CN113164415A (zh) 依拉司群和阿贝西利在患乳腺癌女性中的联合应用
CN110996955A (zh) 以乙型肝炎病毒感染为特征的肝细胞癌的治疗
Pasquini et al. Anxiety levels and related pharmacological drug treatment: a memorandum for the third millennium
JP2022524047A (ja) 効力を判定するための方法

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: 20070530

AK Designated contracting states

Kind code of ref document: A2

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

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

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: 20071219

R17D Deferred search report published (corrected)

Effective date: 20090416