Title of Invention

"COMPOUNDS DERIVED FROM 4--ANILINEQUINAZOLINES WITH ADENOSINE-KINASE INHIBITOR PROPERTIES"

Abstract The present invention relates to the use of 4-anilinoquinazoline derivatives as adenosine-kinase inhibitors. The present invention also relates to a method for protecting tissues and organs like heart, brain and kidneys affected by ischemia, and for treating heart insufficiency, myocardium infarct, arrhythmia, arterial hypertension, atherosclerosis, coronary artery restenosis after angioplasty, chronic renal insufficiency, cerebral vascular accident, and chronic inflammatory diseases (e.g. rheumatoid arthritis). The present invention also relates to the compound 6,7-dimethoxy- 4- (3"-7V",A^"-dimethylaminoanilino)quinazoline, or a pharmaceutically acceptable salt thereof, pharmaceutical composition comprising it and use of such compound in the manufacture of a medicament for treating or preventing diseases or conditions which are benefited from the adenosine kinase inhibition.
Full Text -ANIMNOQUINAZOLINES DERIVATIVES WITH ADENOSINE-, f KINASE INHIBITORY PROPERTIES".
FIELD OF THE INVENTION
This invention relates to compounds derived from 4-anilinoquinazolines that present adenosine-kinase inhibitory activity and use thereof in diseases or conditions that are benefited from the adenosine kinase inhibition. More particularly, the present invention relates to the compound 6,7-dimethoxy-4-(3'-7v~',./v"-dimethylaminoanilino)quinazoline, or a pharmaceutically acceptable salt thereof, pharmaceutical composition comprising it and the use of the adenosine kinase inhibitory properties of said compound for protecting tissues and organs like heart, brain and kidneys affected by ischemia, and for treating heart insufficiency, myocardium infarct, hypertension, atherosclerosis, coronary restenosis after angioplasty, chronic kidney
s
insufficiency, cerebral vascular accident, chronic inflammatory diseases (e.g. rheumatoid arthritis).
BACKGROUND OF THE INVENTION
The compounds included in the present invention are quinazoline derivatives previously described as inhibitors of tyrosine kinase from the family of EOF receptors [Fry et al, Science 1994, 265, 1093 and Patents No. BR9708640 and EP 566226], now they are unexpectedly found as inhibitors of adenosine kinase.
For a better comprehension of the present invention, some known compounds and their properties are defined and described, together with specific terms related to this invention.
Adenosine. Adenosine is a purinic nucleoside that regulates multiple cell functions, and its effects are mediated by at least 4 kinds of PI purinergic receptors located in cell membranes of almost all kinds of cells, namely Al, A2a, A2b and A3

[Fredholm et al, Pharmacol Rev 2001, 53:527]. Despite acting in practically in all cells and organs of the organism, its main effects are observed in the heart, brain, kidneys and immune system cells. Because its action is restricted to the site where it is released, adenosine is considered an autacoid [form the Greek autos - self and akos - relief, cure]. In general, its complex effects result in reduction of metabolic activity and protection against physiological and pathological stimuli associated to sustained increases of cell activity. Its protective effects are well known for ischemia/reperfusion deleterious actions, pro-inflammatory substances, as well as its effects as analgesic, modulator of neuronal activity related to sleep, sympatholitic, inhibitor of thrombosis aggregation, inhibitor of neutrophilic adhesion, inhibitor of free radical production and vasodilatator.
Pharmacological inhibitors of the adenosine catabolism. Considering its wide range of effects, there is no surprise in the rising interests in the therapeutic usage of adenosine, its mimetics and in substances that interfere in its metabolism and bioavailability. The usage of adenosine itself as a therapeutic agent is restricted due to its short half-life (estimated in less than 1 second in circulation) and its undesirable hemodynamic effects. These arguments are also valid for agonists and antagonists of receptor that like adenosine may have undesirable systemic effects. However, approaches that interfere in local metabolism and bioavailability of adenosine are promising. In this case, it should be noticed that adenosine is formed mainly as a result of the action of intra and extra-cellular 5'-nucleotidases that dephosphorylate 5'-AMP, and of the SAH-hydrolase on the S-adenosyl-homocysteinee (SAH) [Headrick et al, Am J Physiol Heart Circ Physiol 2003; 285:H1797]. Otherwise, extra-cellular adenosine is quickly absorbed by cells through a specific membrane carrier. In the intracellular environment, adenosine is deaminated turning into inosine, by adenosine deaminase or

re-phosphorylated into 5'-AMP by adenosine kinase. The high catabolism, besides the high membrane transport speed, determines that adenosine has a short half-life and highly localized physiological functions. The importance of these mechanisms for local adenosine bioavailability is confirmed by the increase in tissue concentration caused by inhibitors of membrane carriers of adenosine, as well as by the action activity inhibitors of adenosine kinase or deaminase [Headrick et al, Am J Physiol Heart Circ Physiol 2003; 285:H1797, Kowaluk & Jarvis, Expert Opin Investig Drugs. 2000; 9:551]. In this case of enzyme inhibitors, available evidences suggest a potential utility for the therapy of clinical situations, where deleterious effects of ischemia/reperfusion, inflammation and pain are involved.
Adenosine Kinase. Adenosine kinase, also known as adenosine 5'-phosphotransferase, is the most abundant nucleoside kinase in mammals, and catalyzes the phosphorylation of the 5'-hydroxyl of the ribofuranosyl of nucleoside analogs, using ATP or GTP as phosphate donor. The structures of adenosine kinases from different species, including human, have been determined, obtained from the placenta. The enzyme is a monomer, whose structure consists of a large a/p dominion with nine P-bands and eight a-helixes and a smaller o/P dominion with five p-bands and two a-helixes [Mathews et al, Biochemistry 1998; 37:15607]. The active site is located along the edge of the p-band in the larger o/p dominion, and this is where adenosine attaches itself, while the smaller o/P dominion blocks the upper face of the active site, and another nearby site receives the ATP. A magnesium binding site is located between binding sites of adenosine and ATP, and this is an essential ion for the catalysis of adenosine kinase. The model proposed for the activity of this kinase suggests that the amino acid aspartate, located in the position 300, is an important catalytic remainder involved in the deprotonation of the 5'-hydroxyl group during phosphate transfer.

The pharmacological inhibition of adenosine kinase has been described with adenosine analogs (e.g. aminoadenosine and iodotubercidine), as well as with pyridopyrimidine derivatives [Kowart et al, Bioorg Med Chem Lett 2001, 11:83; Lee et al. J Med Chem 2001, 44:2133; Zheng et al, Bioorg Med Chem Lett. 2001, 11:2071; Gomitsian et al, J Med Chem. 2002, 45:3639; Gfesser et al, Eur J Med Chem 2003, 38:245; Zheng et al, Bioorg Med Chem Lett 2003, 13:3041; Perner et al, J Med Chem. 2003; 46:5249]. Studies of therapeutic application of these compounds have shown beneficial effects of inhibition of the adenosine kinase in situations of myocardium ischemia, pain and inflammatory processes [Jarvis et al, Pain 2002, 96:107; Suzuki et al, BrJPharmacol 2001, 132:1615; Boyle et al, JPharmacol Exp Ther 2001, 296:495. Kowaluk et al J Pharmacol Exp Ther 2000, 295:1165. Jarvis et al, J Pharmacol Exp TherlQQQ, 295:1156. Smolenski et al. Circulation 2001, 104(suppl I):I-246]..
Ischemia/Reperfusion. Ischemia, defined as improper blood supply to tissues and organs, is one of the main causes of death and disability in populations all over the world, and its main determinant is the atherosclerotic disease of arteries. Its effects in the heart, brain or kidneys are caused mainly by the lack of oxygen, which leads to, depending on intensity and duration, to death or cell degeneration, resulting in different clinical situations like myocardium infarct, chest angina, heart insufficiency, brain vascular accident and kidney insufficiency. Otherwise, an additional deleterious effect is caused after restoration of blood flow in the ischemic area, a condition that many times occurs spontaneously or by therapeutic interference (e.g. coronary thrombosis). Mechanisms that cause deleterious effects of reperfusion are poorly known. Nevertheless, important pathogenic agents considered are the massive oxy radicals generation and the overload of intracellular calcium due to reperfusion. Therefore, tissue lesions caused by ischemia are frequently a consequence of a

combination of deleterious effects of the ischemia per se and of reperfusion. We should include in the context lesions to organs .(e.g. heart, kidney and liver) used in transplantations.
Surely, the obvious solution to the morbid-mortality caused by conditions of ischemia in several organs is the prevention of atherosclerotic disease. However, the impact of available strategies for primary prevention is still very limited. Therefore, effective prevention ways are needed and, particularly, therapeutic ways to limit the extension of tissue lesions caused by ischemia, and the preservation of the viability of ischemic tissues is one of the most imperious present therapeutic objectives.
In this context, it is important to mention that cells from multicellular organisms have a self-protection mechanism for the lesion by ischemia/reperfusion, activated by repeated events of sub-lethal ischemia, known as ischemic pre-conditioning [Yellon & Downey Physiol. Rev 2003, 83: 1113]. This mechanism has two ways of protection: one known as "classical" that last about 2 hours after the conditioning ischemia, followed after about 24 hours by a second protection window that lasts 3 days, known as "late protection". The current model for explaining preconditioning states that conditioning ischemia causes the release of various autacoids that trigger the protection process through the activation of membrane receptors [Yellon & Downey Physiol. Rev 2003, 83: 1113]. This activation triggers the combination of complex cell signaling ways that during lethal ischemia converge to one or more effectors to mediate protection. The effectors of this response are still poorly known. Nevertheless, in the therapeutic point-of-view, it is important that pharmacological agents that activate signaling ways at different levels may mimic the conditioning stimulus, leading to the expectation that pharmacological agents may be produced in order to explore

therapeutically the powerful tissue protection activated by endogenous mechanisms responsible to ischemic pre-conditioning.
Thus, it is known that adenosine is the main triggering agent in the activation of cell ways involved in the classical or late pre-conditioning [Headrick et al, Am J Physiol Heart Circ Physiol 2003; 285:H1797]. Consistent results from clinical studies point out benefits of the use of adenosine for the preservation of the ischemic myocardium, but clinical evidences are still scarce for its therapeutic effect in brain and kidney ischemic lesions, yet it does not mean that it is not effective. It has been demonstrated, for instance, that its action restores ATP stocks in endothelium cells and myocytes, inhibits the formation of free radicals, inhibits the accumulation and the activity of neutrophils, and improves microcirculation [Mahaffey et al, J Am Col
Cardiol 1999, 34:1711]. Additionally, because adenosine is the main endogenous agent
x
that activates ischemic pre-conditioning, its effect is particularly important in acute coronary syndromes, for they are usually caused by dynamic coronary occlusion with intermittent blood flow periods, having a potential deleterious effect due to the ischemia/reperfusion mechanism. In models of acute coronary syndrome in experimental animals, adenosine reduces consistently the size of the infarct, improves ventricular function and improves coronary flow [Yellon & Downey Physiol. Rev 2003, 83: 1113; Headrick et al, Am J Physiol Heart Circ Physiol 2003; 285:H1797]. Clinical studies demonstrated that adenosine administration reduces the extension of myocardium infarcts, improves the conditions of myocardium flow, reduces the incidence of heart insufficiency and of myocardium infarct with Q wave in patients submitted to primary angioplasty, also reduces variation of the segment S-T, lactate production and ischemic symptoms in patients submitted to elective angioplasty [Mahaffey et al, J Am Col Cardiol 1999, 34: 1711]. Recently, results from the study

AMISTAD (Acute Myocardial Infarction Study of Adenosine), planned to test the hypothesis that adenosine reduces the size of myocardium infarct in patients submitted to thrombolysis, demonstrated reduction in sizes of previous infarcts in patients treated with adenosine [Mahaffey et al, J Am Col Cardiol 1999, 34:1711]. However, no differences between the clinical evolution of treated and non-treated patients was observed. The absence of measurable clinical benefits with adenosine in this study reflects problems with biases in choice of patient groups, but also pharmacokinetic and pharmacodynamic problems of adenosine, as well as its short half-life and undesirable hemodynamic effects.
Therefore, it is possible that pharmacological agents that modify local adenosine bioavailability show to be effective for the protection of the myocardium and other tissues submitted to ischemia/reperfusion.
Inflammation. Chronic inflammatory diseases represent a wide range of diseases that attack organs and tissues in different ways and extensions. In this group one may include, among others, asthma, rheumatoid arthritis, inflammatory diseases of the intestine, psoriasis and atherosclerosis [Barnes & Karim, N Engl J Med 1997; 336:1066; Ross, N Engl J Med 1999; 340:115]. In spite of representing different physiopathological situations, all inflammatory diseases present the activation and collapse of the immune system responsible for the amplification and support of the inflammatory process. Causes of these diseases remain unknown, but there is little doubt that the pathological process results from the interaction between genetic and environmental factors. Genes, like those in asthma atopy, HLA antigens in rheumatoid arthritis and intestine inflammatory diseases, may determine the susceptibility of patients to the disease, but frequently unknown environmental factors may determine clinical presentation and course. Once established, the chronic inflammatory process

develops itself alone. Anti-inflammatory agents and immunosuppressors may suppress the vicious circle, but there is still no healing treatment for any chronic inflammatory diseases.
Deleterious effects of chronic inflammatory processes occur through several mechanisms, but main determinants are local production of pro-inflammatory cytokines and transformation of tissue inflammatory cells in autonomous lineages. These transformations and cytokine production are processes regulated by complex signaling ways that involve many transduction elements and transcription factors. Nevertheless, one transcription factor, NF-fcB, seems to be a key element for the activation and transformation of tissue inflammatory cells [Barnes & Karim, N Engl J Med 1997; 336:1066; Lawrence et al, Nat Afet/2001, 7:1291]. This factor is related to the expression of genes responsible for adhesion and recruitment of circulation inflammatory cells (e.g. neutrophils, eosinophils and T lymphocytes) in inflammatory sites, as well as for cytokine and enzyme production in chronic inflammatory diseases. One of these genes is the inducible NOS, whose expression in increased in the epithelium of aerial ways cells and macrophages of asthma patients, in colon epithelium of ulcerative colitis patients and in synovial cells of inflamed joints. The cyclooxygenase-2, another inducible enzyme regulated by NF-»cB, is responsible by the increase in prostaglandin and tromboxane production in inflammatory diseases. On the other hand, the production of interleukin-lp\ TNF-a, interleukin-6, stimulant factor of granulocyte/macrophage colonies, and many chemotactic cytokines is increased in patients of asthma, rheumatoid arthritis, psoriasis and intestine inflammatory disease. All these cytokines have an important role in these inflammatory processes. Interleukin-ip and TNF-a may influence the severity of these diseases, possibly by permanently

activating NF-»*B. The treatment of rheumatoid arthritis patients with drugs that block the action of TNF-a may control the disease.
Adenosine is an endogenous immunomodulator with anti-inflammatory and immunosupressor properties, which acts through multiple mechanisms still not completely established. Some evidences point out that adenosine inhibits the activation of NF-reB that is induced by TNF, what may contribute for its role in the suppression of inflammations and immunomodulation [Kowaluk et al J Pharmacol Exp Ther 2000, 295:1165. Jarvis et al, J Pharmacol Exp Ther 2000, 295:1156]. Therefore, the use of adenosine kinase inhibitors may present therapeutic benefits to a wide range of clinical situations directly or indirectly dependent of inflammatory and immunological processes. Among conditions that could benefit from the use of adenosine kinase are chronic degenerative inflammatory diseases (e.g. rheumatoid arthritis, systemic erythematous lupus etc.), asthma, atherosclerosis, ulcerative colitis, and Chron disease.
Pain. Chronic or acute pain are among most frequent clinical conditions. Mechanisms involved in its beginning and sustenance are multiple and comprise from neuronal degeneration to inflammation. Pain initiator stimuli are transmitted to the central nervous system by the activation of non-myelinized (C fibers) and myelinized (Aft fibers) afferents. Cell bodies of these fibers are located in the dorsal root, trigeminal ganglion, and nodous ganglion. These fibers establish multiple connections with the spinal medulla or cerebral trunk, and with specific areas of the prosencephalon, where the stimulus is integrated. Following the tissue lesion or the inflammation, a large number of endogenous substances is released, and these substances may activate or sensibilize nociceptor afferents. These substances comprise H+, ATP, bradikinine, 5-HT, histamine, prostaglandins, P substance and adenosine [Bevan, 1999. In: Watt, P.D., Melzack, R. (Eds.), Textbook of Pain, fourth ed. Churchill Livingstone, Edinburgh, pp.

85-103].. Some of these mediators act through binders associated to cationic channels
(e.g. H+, ATP, 5-HT3), while others act through G-protein-coupled receptors (GPCRs) (e.g. prostaglandins, bradikinines, 5-HT). Changes in the excitability of nociceptor afferents may result from the activation of multiple intracellular signaling ways mediated by kinase proteins with subsequent phosphorylation of specific sodium channels of sensorial neurons. There are three basic therapeutic approaches for controlling pain: (1) suppression of the source, (2) change in central perception, and (3) transmission blocking for the central nervous system.
Adenosine and its analogs have analgesic effect. Their actions are complex and multiple, including action in central and peripheral mechanisms. Thus, spinal administration of adenosine or its analogs [eg. 5'-Af-ethyl-carboxamidoadenosine (NECA)] produces analgesia through an effect mediated by Al receptors, whose activation produces liberation inhibition of nociceptive afferents CGRP [Mauborgne et al, Eur. J. Pharmacol 2002, 441:47]. Likewise, the same effect has been demonstrated for adenosine metabolism inhibitors [Sawynok, Curr. Opin. Cent. Periph. Nerv. Syst. Invest. Drugs 1999, 1:27; Kowaluk et al, Exp. Opin. Invest. Drugs 2000, 9:551]. The inhibition of adenosine kinase with 5'-amino-5'-deoxyadenosine or iodotubercidine increase the bioavailability of adenosine in the spinal medulla [Golembiowska et al, Brain Res 1995, 699:315].
Adenosine acts also directly on peripheral nerves by interfering in the process of nociceptor activation, through complex mechanisms. Its actions may result in inhibition or increase of pain through the action on nociceptor afferents via Al and A2A receptors, and it results from the reduction or increase of cAMP, respectively [Khasar et al, Neuroscience 1995, 67:189]. However, its central actions are more powerful and result in analgesic effect.

Anilinoquinazolines: Derivatives of 4-anilinoquinazolines are widely described in the literature as powerful and selective inhibitors of the activity of tyrosine kinases from the family of EGF receptors [Fry et al, Science 1994, 265, 1093; Fry et al, Pharmacol. Ther. 1999, 82, 207 and Levitzki et al, Pharmacol. Ther. 1999, 82, 231]. Furthermore, knowledge of the inhibition process of these enzymes seems to be the way for the therapy of many diseases, like cancer, psoriasis, diabetes, cardiovascular diseases etc [Fry et al, Science 1994, 265, 1093]. Based on this evidence, many detailed studies arose on the biological function of many derivatives from this structure class [Rewcastle et al, J. "Med. Chem. 1995, 38, 3482 and Bridges et al, J. Med. Chem. 1996, 39, 267].
Many studies on the structure-activity relationship (SAR) involving many series of quinazoline derivatives lead to advances in power, specificity and pharmacokinetic properties of these inhibitors [Fry et al, Pharmacol. Ther. 1999, 82, 207 and Rewcastle et al, Curr. Org. Chem. 2000, 4, 679]. Three quinazoline compounds are under clinical investigation in cancer patients: ZD1839 (Iressa) [Rewcastle et al, Curr. Org. Chem. 2000, 4, 679], CP358774 [Rewcastle et al, Curr. Org. Chem. 2000, 4, 679 e Moyer et al, Cancer Res. 1997, 57, 4838] and CI1033 [Tsou et al, J. Med. Chem. 2001, 44, 2719]. Pre-clinical data (IC5o in the order of pmol.L"1) support the possibility of using these compounds in conventional chemotherapy with potential anti-tumoral agents [Ciardiello et al, Drugs 2000, 60 (supl.l), 25].
Inhibition power, in all series of evaluated and synthesized compounds, seems to be associated to electron donor substitute groups in positions 6 and/or 7 of quinazoline (OMe, OEt e NHb), and to halogens (mainly Br and Cl), like substitutes in the meta position of the aniline ring. The meta-substituted aniline group showed to be

the best substitute for position 4 of the quinazoline system [Bridges etal, J. Med. Chem. 1996, 39, 267].
Studies with quinazoline derivatives have not been limited only to the investigation of the activity of tyrosine kinase from the family of EGF receptor [Rewcastle et al, Curr. Org. Chem. 2000, 4, 679]. Prazosin is a quinazoline with antagonistic properties of a-adrenergic receptors. This compound has a vasodilatator effect, and is used in the anti-hypertensive therapy, as well as some of its structural derivatives like ciclazosin, which has a stringer affinity to al-adrenergic receptors, and may be applied in'the treatment of benign prostate hyperplasia [Melchiorre et. al., Bioorganic &Medicinal Chemistry Letters 1998, 8, 1353-1358]. Another good example is PD15303 5, which entered the stage of clinical triage by Sugen (like SU5271) for use in the treatment of skin diseases, like psoriasis and skin cancer [McMahon et al, WO9810767; Chem. Abstr. 1998, 128, 261949]. Other examples of biologically active quinazolines are those presented as powerful and specific inhibitors of type 5 phosphodiesterase (PDE5) [Ukita et al, J. Med. Chem. 2001, 44, 2204]. This enzyme is highly specific in the hydrolysis of the cyclic nucleotide cGMP (guanosine 3',5'- cyclic monophosphate), which controls vascular functions [Gorbin et al, J. Biol. Chem. 1999, 274, 13729]. Thus, an inhibitor that increases the cGMP level inside cells is considered a potential pharmaceutical for the treatment of cardiovascular diseases, such as hypertension, angina, and heart insufficiency [Ukita et al, J. Med. Chem. 2001, 44, 2204].
SUMARY OF THE INVENTION
The present invention relates to the use of anilinoquinazoline derivatives as adenosine kinase inhibitors. The 4-anilinoquinazoline compounds referred-hereinafter have the molecular formula I, as shown in Figure 1, wherein R] and R2are alkoxy group

as methoxy (-OCH3), and R3 is hydrogen (-H), halogen (F, Cl, Br or I), methoxy (-OCH3), methyl (-CH3), acetyl [-C(O)CH3], N,N- dimethylamino [-N(CH3)2] or nitro (-NO2). The substituent R3 may occupy positions 3' or 4' of the jV-phenyl group, generating meta and para-substituted 4-anilinoquinazoline compounds.
The present invention also relates to the use 4-anilinoquinazoline derivatives as adenosine kinase inhibitors in the manufacture of a medicament for treating or preventing diseases which are benefited from the adenosine kinase inhibition.
The present invention also relates to the preferred compound 6,7-dimethoxy-4-(3'-A^"AP-dimethylaminoanilino)quinazoline, or a pharmaceutical ly acceptable salt thereof, particularly a hydrochloride salt.
BRIEF DESCRIPTION OF THE FIGURES
We refer to the following figures that accompany this descriptive report, in order to allow a better understanding and illustration of the present invention:
Figure 1 presents Molecular Formula I, related to the 4-anilinoquinazoline derivatives, that is included in the scope of the present invention, wherein R! and R2 are alkoxy group as methoxy (-OCH3), and R3 is hydrogen (-H), halogen (F, Cl, Br and I), methoxy (-OCH3), methyl (-CH3), acetyl [-C(O)CH3], N,N-dimethylamino [-N(CH3)2] or nitro (-NO2). The substituent R3 may occupy positions 3' or 4' of group JV-phenyl, generating meta and para-substituted 4-anilinoquinazoline compounds.

Figure 2 presents different benzodiazine cores. Structure (1) represents a quinazoline, (2) represents a quinoline and (3) a quinoxaline. The name quinazoline (German: Chinazoline) was proposed, because these compounds are isomers of quinolines (2) and quinoxalines (3).
Figure 3 presents Scheme 1, which presents stages for preparing 4-anilinoquinazolines (9) included in the present invention.
Figure 4 presents experimental stages for preparing 6,7-dimethoxyquinazolin-4(3/#-one (2a), and experimental stages for preparing 4-chloro-6,7-dimethoxyquinazoline (3a).
Figure 5 presents the General Procedure for substituting chlorine, in 4-chloro-6,7-dimethoxyquinazoline (3a).
Figure 6 presents Chromatograms obtained through HPLC experiments of rat myocardium extracts for dosage of tissue adenosine and AMP.
Figure 7 shows representative Examples of systolic pressure records of the left ventricle (LVP) from concentration-response experiments of quinazoline compounds in isolated rat hearts.
Figure 8 presents concentration-response curves for compounds 4d (Example 7), 3a (Example 1) and 4i (Example 17) on the systolic pressure of the left ventricle in isolated rat hearts (expressed as fractional values).
Figure 9 presents concentration-response curves for 4i (Example 17), 3a (Example 1) and 4d (Example 7) on the heart rate of isolated rat hearts (expressed as absolute values).
Figure 10 presents the correlation between ventricular pressure and heart rate, pointing out the existence of an inverse relationship between ventricular pressure and heart rate in the presence of growing doses of compound 4i (Example 17).

Figure 11 presents correlation Diagrams between the increase of systolic pressure and decrease in heart rate in isolated hearts submitted to injections (bolus) of 4i (Example 17) perfused with propanolol, diltiazem and with reduction of calcium concentration in the perfusion buffer.
Figure 12 presents the concentration-response relationship for 4i (Example 17) in an isolated heart submitted to electrical stimulus.
Figure 13 presents experiments of the concentration-response kind with adenosine and the specific inhibitor of its receptor (8-phenylteophylin- 8-FT).
Figure 14 presents representative heart rate diagrams of isolated rat hearts perfused with pure HEPES buffer (control) or HEPES buffer with InM of 4i (Example 17) plus IjaM of an specific inhibitor of adenosine receptors, 8-PT (4i + 8-PT).
DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to the use of 4-anilinoquinazoline derivatives as adenosine kinase inhibitors having the Molecular Formula I (Fig.l), wherein RI and R2 are alkoxy group such as methoxy (-OCH3), and R3 is hydrogen (-H), halogen (F, Cl, Br and I), methoxy (-OCH3), methyl (-CH3), acetyl [-C(O)CH3], N,N-dimethylamino [-N(CH3)2] and nitro (-NO2) or a pharmaceutically acceptable salt thereof. The substituent R3 may occupy positions 3' or 4' of the N-phenyl group, generating meta and para-substituted 4-anilinoquinazoline compounds.
The way of choosing 4-anilinoquinazoline substituents was established by structure-activity studies, in an attempt of allowing compounds with higher power and specificity to inhibit adenosine kinases. Thus, it was established that compounds of Formula I comprise all proper elements in a 4-anilinoquinazoline that possibly allow high power and/or efficiency for the inhibition of the enzyme. Such elements are

electron donor substituents in positions 6 and 7 of the quinazoline, one small or medium-sized lipophilic substituent in positions meta or para in the group W-phenyl (preferably in position meta), one free NH in position 4, and on free CH in positions 2, 5 and 8 of the quinazoline.
Therefore, the compounds included in the scope of the present invention are the following:
6,7-Dimethoxy-4-anilinoquinazoline
6,7-Dimethoxy-4-(3'-fluoroanilino)quinazoline
6,7-Qimethoxy-4-(4'-fluoroanilino)quinazoline
6,7-Dimethoxy-4-(3'-chloroanilino)quinazoline
6,7-Dimethoxy-4-(4'-chloroanilino)quinazoline
6,7-Dimethoxy-4-(3'-bromoanilino)quinazoline
6,7-Dimethoxy-4-(4'-bromoanilino)quinazoline
6,7-Dimethoxy-4-(3' -iodine)phenylaminequinazoline
6,7-Dimethoxy-4-(4'-iodoanilino) quinazoline
6,7-Dimethoxy-4-(3'-methoxyanilino)quinazoline
6,7-Dimethoxy-4-(4'-methoxyanilino)quinazoline
6,7-Dimethoxy-4-(3'-methylanilino)quinazoline
6,7-Dimethoxy-4-(4'-methylanilino)quinazoline
6,7-Dimethoxy-4-(3'-acetylanilino)quinazoline
6,7-Dimethoxy-4-(4' -acetylanilino)quinazol ine
6,7-Dimethoxy-4-(3'-Af',Af'-dimethylaminoanilino)quinazoline
6,7-Dimethoxy-4-(4'-A^,Af'-dimethylaminoanilino)quinazoline
6,7-Dimethoxy-4-(3'-nitroanilino)quinazoline
6,7-Dimethoxy-4-(4'-nitroanilino)quinazoline

or their pharmaceutically acceptable salts.
The more preferred compound of the present invention is 6,7-dimethoxy-4-(3'-N',A^-dimethylaminoanilino)quinazoline.
The following considerations upon the substituents of compounds of Formula I are important:
- the term "alkoxy" means an alkyl group attached to an oxygen atom.
Representative examples of "alkoxy" groups comprise methoxy, ethoxy, terc-butoxy,
propoxy and isobutoxy;
- the term "halogen" comprises fluorine, chlorine, bromine and iodine;
- the term "alkyl" means a straight or ramified chain of hydrocarbon.
Representative examples of alkyl groups are methyl, ethyl, propyl, isopropyl, isobutyl,
butyl, terc-butyl, sec-butyl, pentyl and hexyl;
- the term "acetyl" means a methyl attached to a carbonylic carbon atom;
- the term 'W,A^-dimethylamino" means two methyls attached to a
nitrogen atom.
Compounds of the present invention may exist in forms that are non-solvated, as well as solvated, with pharmacologically acceptable solvents like water, DMSO, ethanol and similar. Generally, solvated forms are considered equivalent to non-solvated forms for the purposed of the present invention.
In the experimental stages, situations related to processing of the compounds are considered, which are described in Examples from 2 to 20, and the experimental conditions for obtaining them are the following:
(i) Solvents were evaporated in a rotating evaporator [Asten (250 rpm) and Wheaton (200 rpm)] after removing solid remains, such as drying agents, by filtering;

(ii) Melting points were determined in a MOAPF-301 equipment and are incorrect;
(iii) Structures of compounds of Formula 1 and its intermediates were characterized by their infrared spectra, mass and 'll RMN and by elementary analysis. Infrared spectra were obtained in a Perkin••Winer II IR 1600 or 1-T1R 1605 equipment. Yet mass spectra were obtained in a VG Auto Spec (Varian) spectrometer. Data from the elementary analysis were obtained in Perkin-I liner (2400) analy/er.'H RMN spectra were acquired in a INOVA-500 (Varian) spectrometer, operating at 500 MHz. All *H RMN spectra were'obtained at 21°C in (CDibSO, and referenced with Me4Si. Values of 'H RMN signals were determined in delta (f>) scale, and multiplicities are presented as following; d, dublet; dd, double dublet; ddd, double double dublcts; t, triplet; dt, dublet of triplets; tdd, triplet of dublet of dublets.
(iv) The following abbreviations were used:
(€03)280 - deuteron dimelhs isulfo.vuk
Me4Si - tetramethylsilane
DMF - A'.A'-dimethylforniamule
CH2Cl2 -dichloromethane
Na2CO3 -sodium carbonate
NaOH - sodium hydroxide
A general review on qnuuiy.oline synthesis processes may be found in textbooks and in a recent thesis. Anolhei review emphasi/es that the most used start material has been anthranilic acid (4), according to the general method presented in Scheme 1, represented in Figure 3.
In this procedure, the first step involves addition of a carbon unit to an anthranilic acid derivative (4), which lead1- to in situ cycli/ation to quinazoline (5)

(Scheme 1). This transformation may be carried out using reagents like formic acid, formamide, and amidins. The synthesis of this precursor was described in 1895 by Niementowski, who reacted anthranilic acid with formamide. However, superior results are frequently obtained with reagents such as formamidine acetate. The second step in derivatization involves the conversion of intermediate 5 to 4-chloroquinazoline 6, through reaction with phosphoryl chloride (POCb) or with thionyl chloride, containing a catalytic amount of DMF. An alternative procedure, which is better for low soluble quinazolines, involves conversion to analog thione 7 followed by alkylation in sulfur, in order to provide an'alkylthio derivative 8. Finally, the reaction of derivative 4- chloro (6) or of 4- alkylthio (8) with an aniline derivative provides the final product (9) (Scheme 1, Fig. 3).
In this way, synthetic stages explored for preparation of Formula 1 compounds (Fig.l) and its intermediates are illustrated in Example 1. The synthetic method, which is already well described in the literature, and physic-chemical and spectroscopical data are described in Examples 1 to 20, as follows:
In Example 1, we describe experimental stages for preparation of 6-,-?-6,7-dimethoxyquinazolin-4(3/#-one (2a) and 4-chloro-6,7-dimethoxyquinazoline (3a), which are precursors of 4-anilinoquinazoline derivatives from Formula I. Moreover, it presents the general substitution procedure of the chlorine atom of intermediate (3a), in order to synthesize all target-compounds described in Examples 2 to 20.
EXAMPLE 1
Obtainment of 6,7-dimethoxyquinazoHn-4(3//)-one (2a)
A mixture of 2-amino-4,5-dimethoxybenzoic acid (1.0 g, 5.08 mmol) and formamidine acetate (4.50 g, 43.3 mmol) was disposed in a 50 mL ball. The solid mixture was heated at 140°C in a silicone bath for 8 h. During heating, the fusion of

solids happened, and then the resolidification of the reaction environment. I he mixture was left cooling, and then a NaOH solution was added to it (0.33 mol.l. ') until pi I \\as adjusted to 8. The grayish pink solid was collected through filtering in a liuelmer funnel, washed with water (3 x 10 mL), and dried in vacuum to provide the desired compound (0.79 g, 3.83 mmol, 76%), which was used without purification in the next stage: m.p. 296-298°C (Lit. [Bridges et al, J. Med Chem. 1996, 39, 267| m.p. ")v 298°C).
'H RMN [500 MHz, (CD3)2SO, ppm] 5: 12.07 (1H, s, 11-3) 8.00 (111. s. H-2), 7.45 (1H, s, H-5), 7.14 (1H, s, H-8), 3.91 (3H, s, H-6a), 3.87 (311, s. 11- /a).
Obtainment of 4-chIoro-6,7-dimethoxyquinazoline (3a)
A 6,7-dimethoxyquinazolin-4(3/^)-one suspension (2a) (0 71' - vM i mmol) in thionyl chloride (7.0 mL) containing 10 drops of A'.A'-dimeiln IIunnainiile (DMF) was agitated and heated under reflux for 3 h, until a solution was ohiumed I he reaction mixture was left cooling at room temperature. The reaction emii'mmem uas diluted in dichloromethane and water (160 mL) and left in ice bath. The maieiial uas treated under agitation with 30 mL of saturated Na2CO3 solution. Na2COi uas earelnlK added until pH was adjusted to 7-8 range. Then, the aqueous phase was e\ira<..ud chjcb x ml and the organic phase compound was washed with sail sulmion dried over magnesium sulfate filtered solvent esapoiaied in a rotatory evaporator order to provide as yellow solid g mmol which used without purification ihe ne stage: melting point> RMN of 'H [500 MHz, (CD3)2SO, ppm] 6: 8.90 (1 H, s. 11 : i H-5), 7.34 (1H, s, H-8), 4.08 (6H, s, H-6a and H-7a).

IV (KBr/ cm"') v: 2975 (C-1I), 1619 (C-N of an aromatic), 1511 (C-C of an aromatic), 1233 (C-O-C), 789 (C-H), 872 (C-C1).
Obtainment of 6,7-I)imethoxy-4-[3'- or 4'-(R3)-anilino]quinazoline (4a-4j and 4b'-4j'). With substituents R= H (a), F (b), CI (c), Br (d), I (e), OCH3 (f), CH3 (g), C(0)CH3 (h), N(CH3)2 (i) and NO2 (j)
General chlorine substitution procedure: a mixture of 4-chloro-6,7-dimethoxyquinazoline (3a) (0.10 g, 0.445 mmol) and of the correspondent aniline (5.50 mmol) in isopropanol (20 inL) was mechanically agitated and heated to reflux temperature for 2 Hours. We observed that when the heating of the reaction mixture reached the range of 70-90°C, the solid dissolved completely, and then began the precipitation of the desired compound, showing that the nucleophilic aromatic substitution reaction was happening. The yellow solids were filtered, washed with isopropanol (2 x 50 mL), and after vacuum drying, the desired compounds were obtained. Products were isolated as hydrochlorides through direct filtering of the reaction mixture.
According to this experimental procedure, the following compounds of Formula I (Fig.l) were synthesized.
EXAMPLE 2
Obtainment of 6,7-Dimethoxy-4-(anilino)quinazoline hydrochloride (4a): yield of 0.110 g (0.346 mmol, 77.0%), melting point 268-270°C (Lit.[ Bridges et al,J. Meet. Chem. 1996, 39, 267J melting point >250°C).
RMN of 'H [500 MHz, (CD3)2SO,p/wj] 8: 11.46 (IH, s, NH), 8.80 (IH, s, H-2), 8.35 (IH, s, H-5), 7.70 (2H, d, 3.l- 8.0 Hz, H-2' e H-6'), 7.50 (2H, t, 3J= 8.0 Hz, H-3' e H-5'), 7.38 (IH, s, H-8), 7.32 (111, t, 3J= 8,0 Hz, H-4'), 4.04 (3H, s, H-6a), 4.01 (3H, s, H-7a).

IV (KBr/cm ') v: 3418 (N-H), 3062 (( II ot an aromatic). 1635-1459 (C-N o! an aromatic). 1459 (C-C olan aromatic), 1279 (C-O-C), 867-748 (C-H).
MS(l::i). in// (%); 281.1 |M' ] (84,25), 280.1 (M-H'f (100).
Llemenlarv analysis lor Ci6Hr,NtO2.HCI (%) - calculated: C, 60.48; H, 5.07; N, 13.22. Dclermincd: C', 60.48; H, 4.92; N, 13 16,
KXAMPLK3
Obtain menl of 6,7-l)tmethoxy-4-(3'-fluoroaniIino)quinazoline hydrochloride (4b); yield of 0.100 g (0.298 rnmol, 67%), melting point 219-22TC (Lit. | bridges KMN ol'll |500 Mil/, (CDO.'SO, ppm\ '6: 11.51 (1H, s, NH), 8.86 (1H, s, 11-2). 8.41 (111. s, 11-5). 7.74 (111, dt, -'j,,.[.-- 1 1.0 Hz and 4J,,_H= 2,2 H/,, H-2'), 7.63 (III, ddd. '.1 8.3 11/ and '.I,,,,,,,- 2.2 11/, 4J,m,m 1.0 Hz, H-6'), 7.52 (1H, dt, 3JH.n= 8,3 11/and '.I,, i 6.7 11/. 11-5'), 7.39 (111, s, 11-8). 7.15 (IH, tdd, 3J,,.H = 3Jn-F= 8.3 Hz, 4JH i, 2.2 11/ aiul '.In.,, 1.0 11/. 11-4'), 4.04 (311. s, H-6a), 4.00 (311, s, H-7a).
IV (KBr/cm ') v: 3412 (N-ll), 3062 (C'-H of an aromatic), 1635 (C-N of an aromatic), 1490 (( --(' ol an aromatic), 1279 (C-O-C), 985 (C-F), 872-774 (C.'-H).
MS(LI), m// (%): 299.0 |M' J (91.4), 298,0 [M-H]4 (100).
Llernenlary analysis ior C,,,! 1|4N3O71 ,11C! (%) - calculated: C, 57.24; H, 4.50; N, 12.51. Dclermincd: C. 57.14: 11, 4,38; N, 12,34,
FXAMl'LI 4
Oblaininent of 6,7-Dimethoxy-4-(4'-fluoroanilino)quinazoline hydrochloride (41)'): yield of 0.098 g (0.2,92 mmol, 65%). melting point 269-272°C (I it. | Barker. I'alenl LJ' 566226A 1 | melting point 227-2JO°C),

RMN of'H [500 MHz, (CD3)2SO,ppm] 5: 11.56 (IH, s, NH), 8.79 (IH, s, H-2), 8.40 (IH, s, H-5), 7.75 (2H, dd, 3J= 9:0 Hz and 4JH-F= 5.0 Hz, H-2' e H-6'), 7.38 (IH, s, H-8), 7.32 (2H, t, 3J= 9.0 Hz, H-3' e H-5'), 4.02 (3H, s, H-6a), 3.98 (3H, s, H-7a).
IV (KBr/cnT1) v: 3418 (N-H), 3031 (C-H of an aromatic), 1635 (C-N of an aromatic), 1511 (C-N of an aromatic), 1284 (C-O-C), 826 (C-F), 774 (C-H).
MS(EI), m/z (%): 299.1 [M+] (92), 298.1 [M-H]+ (100).
Elementary analysis for QeHnNsChF.HCl (%) - calculated: C, 57.24; H, 4.50; N, 12.51. Determined: C, 57.22; H, 4.41; N, 12.38.
EXAMPLE 5
Obtainment of 6,7-Dimethoxy-4-(3'-chloroanilino)quinazoline hydrochloride (4c): yield of 0.113 g (0.321 mmol, 72%), melting point 226-228°C (Lit. [Bridges et al, J. Med. Chem. 1996, 39, 267], melting point 261-262°C).
RMN of'H [500 MHz, (CD3)2SO, ppm] 5: 11.60 (IH, s, NH), 8.88 (IH, s, H-2), 8.45 (IH, s, H-5), 7.93 (IH, t, 3J= 2.0 Hz, H-2'), 7.77 (IH, ddd, 3J= 8.0 Hz, 4J= 2.0 Hz and 4J~ 1.0 Hz, H-4'), 7.51 (IH, t, 3J= 8.0 Hz, H-5'), 7.40 (IH, s, H-8), 7.37 (IH, ddd, 3J= 8.0 Hz, 4J= 2.0 Hz and 4J~ 1.0 Hz, H-6'), 4.04 (3H, s, H-6a), 4.00 (3H, s, H-7a).
IV (KBr/cm-1) v: 3428 (N-H), 3041 (C-H of an aromatic), 1640 (C-N of an aromatic), 1521 (C-C of an aromatic), 1284 (C-Q-C), 991 (C-C1), 877-774 (C-H).
MS(EI), m/z (%): 315.0 [M+] (71.3), 314.0 [M-H]+ (100).
Elementary analysis for C^Hn^C^O-HCl (%) - calculated: C, 54.56; H, 4.29; N, 11.93. Determined: C, 54.43; H, 4.17; N, 11.27.

EXAMPLE 6
Obtainment of 6,7-Dimethoxy-4-(4'-chloroanilino)quinazoline hydrochloride (4c') [Hennequin et al, /. Med. Chem. 1999, 42, 5369]: yield of 0.105 g (0.298 mmol, 67%), melting point 282-284°C.
RMN of'H [500 MHz, (CD3)2SO,ppm] 5: 11.16 (IH, s, NH); 8.74 (IH, s, H-2); 8.28 (IH, s, H-5), 7.79 (2H, d, 3J= 8.5 Hz, H-2' and H-6'), 7.50 (2H, d, 3J= 8.5 Hz, H-3' and H-5'), 7.33 (IH, s, H-8), 4.00 (3H, s, H-6a); 3.97 (3H, s, H-7a).
IV (KBr/cm'1) v: 3397 (N-H), 3041 (C-H of an aromatic), 1635 (C-N of an aromatic), 1516 (C-C of an aromatic), 1243 (C-O-C), 985 (C-C1), 857-774 (C-H).
MS(EI), m/z (%): 315.0 [M+] (82.8), 314.0 [M-Hf (100).
Elementary analysis for Ci6H,4N3O2Cl.HCl (%) - calculated: C, 54.56; H, 4.29; N, 11.93. Determined: C, 54.77; H, 4.49; N, 11.27.
EXAMPLE 7
Obtainment of 6,7-Dimethoxy-4-(3'-bromoanilino)quinazoline hydrochloride (4d): yield of 0.165 g (0.416 mmol, 93%), melting point 263-265°C (Lit. [Bridges et al, J. Med. Chem. 1996, 39, 267], melting point 264-266°C).
RMN of'H [500 MHz, (CD3)2SO, ppm] 6: 11.70 (IH, s, NH), 8.88 (IH, s, H-2), 8.45 (IH, s H-5), 8.04 (IH, t, 4J= 2.0 Hz, H-2'), 7.80 (IH, ddd, 3J= 8.0 Hz, 4J= 2,0 Hz and 4J ~ 1.0 Hz, H-4'), 7.49 (IH, ddd, 3J= 8.0 Hz, 4J= 2.0 Hz and 4J ~ 1.0 Hz, H-6'), 7.43 (IH, t, 3J= 8.0 Hz, H-5'), 7.39 (IH, s, H-8), 4.03 (3H, s, H-6a), 4.00 (3H, s, H-7a).
IV (KBr/cnf1) v: 3418 (N-H), 3031 (C-H of an aromatic), 1640 (C-N of an aromatic), 1521 (C-C of an aromatic), 1279 (C-O-C), 872-779 (C-H), 600 ( C-Br).
MS(EI), m/z (%): 359.0 [M+] (77.5), 360.0 [M-H]+ (100).

Elementary analysis for C|6H,4N3O2Br.HCl (%) - calculated: C, 48.45; H, 3.81; N, 10.59. Determined: C, 48.85; H, 3.54; N, 10.64.
EXAMPLE 8
Obtainment of 6,7-Dimethoxy-4-(4'-bromoanilino)quinazoline hydrochloride (4d'): yield of 0.126 g (0.318 mmol, 71%), melting point 277-279°C.
RMN of'H [500 MHz, (CD3)2SO, ppm] 8: 11.19 (IH, s, NH), 8.88 (IH, s, H-2), 8.22 (IH, s, H-5), 7.70 (4H, high singlet, H-2', H-3', H-5' and H-6'), 7.32 (IH, s, H-8), 4.04 (3H, s, H-6a), 4.02 (3H, s, H-7a).
»
IV (KBr/cm-1) v: 3449 (N-H), 3144 (C-H of an aromatic), 1629 (C-N of an aromatic), 1516 (C-C of an aromatic), 1284 (C-O-C), 867-774 (C-H), 501 (C-Br).
MS(EI), m/z (%): 358.9 [M+] (81.1), 358.9 [M-H]+ (100).
Elementary analysis for QeHnNsCbBr.Ha (%) - calculated: C, 48.45; H, 3.81; N, 10.59. Determined: C, 48.38; H, 3.61; N, 10.54.
EXAMPLE 9
Obtainment of 6,7-Dimethoxy-4-(3'-iodoanilino)quinazoline hydrochloride (4e): yield of 0.119 g (0.268 mmol, 60%), melting point 218-220°C (Lit. [Bridges et al, J. Med Chem. 1996, 39, 267], melting point 273°C).
RMN of 'H [500 MHz, (CD3)2SO, ppm] 5: 11.50 (IH, s, NH), 8.85 (IH, s, H-2), 8.39 (IH, s H-5), 8.15 (IH, t, 4J= 1.5 Hz, H-2'), 7.81 (IH, ddd, 3J= 8.0 Hz, 4J= 1.5 Hz and 4J ~ 1.0 Hz, H-4'), 7.70 (IH, ddd, 3J= 8.0 Hz, 4J= 1.5 Hz and 4J ~ 1.0 Hz, H-6'), 7.39 (IH, s, H-8), 7.27 (IH, t, 3J= 8.0 Hz, H-5'), 4.03 (3H, s, H-6a), 4.00 (3H, s, H-7a).
IV (KBr/cm'1) v: 3418 (N-H), 3026 (C-H of an aromatic), 1629 (C-N of an aromatic), 1516 (C-C of an aromatic), 1279 (C-O-C), 877-779 (C-H), 600 ( C-I).

MS(EI), m/z (%): 406.9 [M+] (95), 405.9 [M-H]+ (100).
Elementary analysis for CieHuNsCfel.HCl (%) - calculated: C, 43.31; H, 3.41 ;N, 9.47. Determined: C, 43.26; H, 3.35; N, 9.26.
EXAMPLE 10
Obtainment of 6,7-Dimethoxy-4-(4'-iodoaniIino)quinazoline hydrochloride (4e'): yield of 0.121 g (0.273 mmol, 61%), melting point 266-269°C.
RMN of 'H [500 MHz, (CD3)2SO, ppm] 8: 11.50 (IH, s, NH), 8.83 (IH, s, H-2), 8.38 (IH, s, H-5), 7.82 (2H, d, 3J= 8,5 Hz, H-3' and H-5'), 7.58 (2H, d, 3J= 8.5 Hz, H-2' and H-6')/7.37 (IH, s, H-8), 4.02 (3H, s, H-6a), 4.00 (3H, s, H-7a).
IV (KBr/cnT1) v: 3397 (N-H), 3031 (C-H of an aromatic), 1635 (C-N of an aromatic), 1516 (C-C of an aromatic), 1290 (C-O-C), 872-779 (C-H), 501 (C-I).
MS(EI), m/z (%): 407.0 [M+] (100), 406.0 [M-H]+ (93.2).
Elementary analysis for CieHnNsChl.HCl (%) - calculated: C, 43.31; H, 3.41; N, 9.47. Determined: C, 43.44; H, 3.42; N, 9.28.
EXAMPLE 11
Obtainment of 6,7-Dimethoxy-4-(3'-methoxyanilino)quinazoline hydrochloride (4f): yield of 0.094 g (0.270 mmol, 61%), melting point 216-218°C.
RMN of'H [500 MHz, (CD3)2SO, ppm] 5: 11.39 (IH, s, NH), 8.81 (IH, s, H-2), 8.37 (IH, s, H-5), 7.39 (IH, s, H-8), 7.39 (IH, t, 3J= 8.0 Hz; H-5'), 7.35 (IH, t, 4J= 2.0 Hz, H-2'), 7.31 (IH, ddd, 3J= 8.0 Hz, 4J= 2.0 Hz e 4J ~ 1.0 Hz, H-6'), 6.90 (IH, ddd, 3J= 8.0 Hz, 4J= 2.5 Hz and 4J ~ 1.0 Hz, H-4'), 4.03 (3H, s, H-6a), 4.00 (3H, s, H-7a), 3.80 (3H, s, H-7').
IV (KBr/cm'1) v: 3438 (N-H), 3005 (C-H of an aromatic), 1635 (C-N of an aromatic), 1496 (C-C of an aromatic), 1279 (C-O-C), 872-774 (C-H).

MS(EI), m/z (%): 311.0 [M+] (79.3), 310.0 [M-H]+ (100).
Elementary analysis for CnHnNsOs.HCl (%) - calculated: C, 58.71; H, 5.22; N, 12.08. Determined: C, 58.52; H, 5.00; N, 12.17.
EXAMPLE 12
Obtainment of 6,7-Dimethoxy-4-(4'-methoxyaniIino)quinazoline hydrochloride (4f): yield of 0.101 g (0.291 mmol, 65%), melting point 205-207°C.
RMN of 'H [500 MHz, (CD3)2SO, ppm] 6: 11.52 (IH, s, NH), 8.76 (IH, s, H-2), 8.38 (IH, s, H-5), 7.59 (2H, d, 3J= 9.0 Hz; H-2' and H-6'), 7.38 (IH, s, H-8), 7.02 (2H, d, 3J= 9.6 Hz, H-3' and H-5'), 4.01 (3H, s, H-6a), 3.97 (3H, s, H-7a), 3.80 (3H, s, H-7').
IV (KBr/cnT1) v: 3403 (N-H), 2949 (C-H of an aromatic), 1635 (C-N of an aromatic), 1516 (C-C of an aromatic), 1243 (C-O-C), 862-774 (C-H).
MS(EI), m/z (%): 311.1 [M+] (100), 310.1 [M-Hf (64.9).
Elementary analysis for Ci7H17N3O3.HCl (%) - calculated: C, 58.71; H, 5.22; N, 12.08. Determined: C, 58.68; H, 5.03; N, 12.10.
EXAMPLE 13
Obtainment of 6,7-Dimethoxy-4-(3'-methylanilino)quinazoIine hydrochloride (4g) [Fry et al, Annu. Rep. Med Chem. 1996, 31, 151]: yield of 0.075 g (0.226 mmol, 51%), melting point 221-223°C.
RMN of'H [500 MHz, (CD3)2SO, ppm] 5: 11.24 (IH, s, NH), 8.78 (IH, s, H-2), 8.29 (IH, s, H-5), 7.50 (2H, signal overlap, H-2' and H-5'), 7.36 (2H, signal overlap, H-8 and H-6'), 7.14 (IH, d, 3J= 8.0 Hz, H-4'), 4.02 (3H, s, H-6a), 3.99 (3H, s, H-7a), 2.37 (3H, s, H-7').

IV (KBr/cm'1) v: 3418 (N-H), 3008 (C-H of an aromatic), 1635 (C-N of an aromatic), 1511 (C-C of an aromatic), 1279 (C-O-C), 775 (C-H).
MS(EI), m/z (%): 295.0 [M+] (87.4), 294.0 [M-H]+ (100).
Elementary analysis for CiyHnNaCfe.HCl (%) - calculated: C, 61.54; H, 5.47; N, 12.66. Determined: C, 61.96; H, 5.55; N, 12.96.
EXAMPLE 14
Obtainment of 6,7-Dimethoxy-4-(4'-methylanilino)quinazoline hydrochloride (4g'): yield of 0.096 g (0.290 mmol, 65%), melting point 227-229°C.
RMKof'H [500 MHz, (CD3)2SO,ppm] 5: 11.26 (IH, s, NH), 8.75 (IH, s, H-2), 8.30 (IH, s, H-5), 7.57 (2H, d, 3J= 8.3 Hz, H-2' and H-6'), 7.36 (IH, s, H-8), 7.28 (2H, d, 3J= 8.3 Hz, H-3' and H-5'), 4.01 (3H, s, H-6a), 3.98 (3H, s, H-7a), 2.35 (3H, s, H-7').
IV (KBr/cm-1) v: 3419 (N-H), 2949 (C-H), 1635 (C-N of an aromatic), 1506 (C-C of an aromatic), 1279 (C-O-C), 867-779 (C-H).
MS(EI), m/z (%): 295.1 [M+] (85.1), 294.1 [M-H]+ (100).
Elementary analysis for CnHnNaOa.HCl (%) - calculated: C, 61.54; H, 5.47; N, 12.66. Determined: C, 61.27; H, 5.53; N, 12.42.
EXAMPLE 15
Obtainment of 6,7-Dimethoxy-4-(3'-acetylanilino)quinazoline hydrochloride (4h): yield of 0.097 g (0.270 mmol, 61%), melting point 219-221°C (Lit. [Barker, Patent No. 566226A1] melting point > 240°C).
RMN of'H [500 MHz, (CD3)2SO, ppm] 6: 11.49 (IH, s, NH), 8.79 (IH, s, H-2), 8.45 (IH, s, H-5), 8.34 (IH, t, 4J= 2.0 Hz, H-2'), 8.10 (IH, ddd, 3J= 8.1 Hz, 4J= 2.1 Hz, 4J= 1.1 Hz, H-6'), 7.87 (IH, ddd, 3J= 8.0 Hz, 4J= 1.7 Hz, 4J= 1.1 Hz,H-4'), 7.62

(IH, t, 3J= 8.0 Hz, H-5'), 7.42 (IH, s, H-8), 4.05 (3H, s, H-6a), 3.99 (3H, s, H-7a), 2.63 (3H, s, H-8').
IV (KBr/cm'1) v: 3428 (N-H), 3036 (C-H of an aromatic), 1681 (C=O), 1635 (C-N of an aromatic), 1516 (C-C of an aromatic), 1279 (C-O-C), 882-779 (C-H).
MS(EI), m/z (%): 323.0 [M+] (83.9), 322.0 [M-H]+ (100).
Elementary analysis for CigHnNaOs.HCl (%) - calculated: C, 60.09; H, 5.04; N, 11.68. Determined: C, 59.07; H, 4.69; N, 11.72.
EXAMPLE 16
Obtafnment of 6,7-Dimethoxy-4-(4'-acetylaniIino)quinazoIine hydrochloride (4h'): yield of 0.110 g (0.306 mmol, 69%), melting point 218-220°C.
RMN of 'H [500 MHz, (CD3)2SO, ppm] 5: 11.33 (IH, s, NH), 8.84 (IH, s, H-2), 8.35 (IH, s, H-5), 8.03 (2H, d, 3J= 9.0 Hz, H-3' and H-5'), 7.98 (2H, d, 3J= 9.0 Hz, H-2' and H-6'), 7.36 (IH, s, H-8), 4.03 (3H, s, H-6a), 3.98 (3H, s, H-7a), 2.60 (3H, s, H-8').
IV (KBr/cm'1) v: 3412 (N-H), 2995 (C-H of an aromatic), 1671 (CO), 1635 (C-N of an aromatic), 1516 (C-C of an aromatic), 1279 (C-O-C), 872-779 (C-H).
MS(EI), m/z (%): 323.1 [M+] (73), 322.1 [M-Hf (100).
/
Elementary analysis for CigHnNsOa.HCI (%) - calculated: C, 60.09; H, 5.04; N, 11.68. Determined: C, 59.07; H, 4.67; N, 11.73.
EXAMPLE 17
Obtainment of 6,7-Dimethoxy-4-(3'-7V',Ar'-
dimethylaminoanilino)quinazoline hydrochloride (4i): yield of 0.128 g (0.355 mmol; 80%), melting point 198-200°C.

RMN of 1H [500 MHz, (CD3)2SO,ppm] 8: 10.39 (IH, s, NH), 8.60 (IH, s, H-2), 8.11 (IH, s, H-5), 7.27 (IH, s, H-8), 7.22 (IH, t, 3J= 8.0 Hz, H-5'), 7.10 (2H, overlap of H-2' and H-6'), 6.59 (IH, ddd, 3J= 8.2 Hz, 4J= 2.4.Hz and 4J ~ 1.0 Hz, H-4'), 3.99 (3H, s, H-6a), 3.95 (3H, s, H-7a), 2.93 (6H, s, H-7' and H-8').
IV (KBr/cmf1) v: 3418 (N-H), 3119 (C-H of an aromatic), 1624 (C-N of an aromatic), 1511 (C-C of an aromatic), 1228 (C-O-C), 846-764 (5 =C-H).
MS(EI), m/z (%): 324.1 [M+] (100), 323.1 [M-H]+ (75.5).
Elementary analysis for C18H2oN4O2.HCl (%) - calculated: C, 59.91; H, 5.87;N, 15.53. Determined: C, 60.01; H, 5.66; N, 15.62.
EXAMPLE 18
Obtainment of 6,7-Dimethoxy-4-(4'-Ar',Ar>-
dimethylaminoanilino)quinazoline hydrochloride (4i'): yield of 0.110 g (0.305 mmol, 69%), melting point 204-206°C.
RMN of *H [500 MHz, (CD3)2SO, ppm] 5: 11.83 (IH, s, NH), 8.80 (IH, s, H-2), 8.51 (IH, s, H-5), 7.80 (2H, d, 3J= 8.50 Hz, H-2' and H-6'), 7.59 (2H, d, 3J= 8.50 Hz, H-3' and H-5'), 7.42 (IH, s, H-8), 4.03 (3H, s, H-6a), 3.98 (3H, s, H-7a), 3.11 (6H,s,H-7').
MS(EI), m/z (%): 324.1 [M+] (100), 323.1 [M-H]+ (23.2).
Elementary analysis for CisHioN^.HCl (%) - calculated: C, 59.91; H, 5.87; N, 15.53. Determined: C, 59.34; H, 5.60; N, 15.29.
EXAMPLE 19
Obtainment of 6,7-Dimethoxy-4-(3'-nitroanilino)quinazoline hydrochloride (4j): yield of 0.093 g (0.256 mmol, 58%), melting point 279-281°C (Lit. [Barker, Patent No. 566226A1] melting point > 240°C).

RMN of'H [500 MHz, (CD3)2SO, ppm] 6: 11.10 (1H, s, NH), 8.82 (1H, s H-2), 8.76 (1H, t, 4J= 2.0 Hz, H-2'), 8.34 (1H, s, H-5), 8.33 (1H, ddd, 3J= 8.0 Hz, 4J= 2.2 Hz, 4J ~ 1.0 Hz, H-4'), 8.07 (1H, ddd, 3J= 8.2 Hz, 4J= 2.2 Hz and 4J ~ 1.0 Hz, H-6'), 7.74 (1H, t, 3J= 8.5 Hz, H-5'), 7.31 (1H, s, H-8), 4.04 (3H, s, H-6a), 4.00 (3H, s, H-7a).
IV (KBr/cm'1) v: 3443 (N-H), 3026 (C-H of an aromatic), 1635 (C-N of an aromatic), 1511 (C-C of an aromatic), 1532 (NO2), 1284 (C-O-C), 872-733 (C-H).
MS(EI), m/z (%): 326.0 [M+] (100), 325.0 [M-H]+ (83.5).
Elementary analysis for QeHnNAt.HCl (%) - calculated: C, 52.97; H, 4.17; N, 15.44. Determined: C, 52.68; H, 4.04; N, 15.04.
EXAMPLE 20
Obtainment of 6,7-Dimethoxy-4-(4'-nitroanilino)quinazoline hydrochloride (4j'): yield of 0.121 g (0.334 mmol, 75%), melting point 228-230°C.
RMN of'H [500 MHz, (CD3)2SO, p/wj] 8: 10.70 (1H, s, NH), 8.78 (1H, s, H-2), 8.32 (2H, d, 3J= 9.0 Hz, H-3' and H-5'), 8.18 (2H, d, 3J= 9.0 Hz, H-2' and H-6'), 8.11 (1H, s, H-5), 7.31 (1H, s, H-8), 4.02 (3H, s, H-6a), 4.00 (3H, s, H-7a).
IV (KBr/crrf1) v: 3428 (N-H), 3119 (C-H of an aromatic), 1635 (C-N of an aromatic), 1511 (C-C of an aromatic), 1573 (NO2), 1279 (C-O-C), 867-779 (C-H).
MS(EI), m/z (%): 326.1 [M+] (86.9), 325.1 [M-H]+ (100).
Elementary analysis for C^HnNAt-HCl (%) - calculated: C, 52.97; H, 20 4.17;N, 15.44. Determined: C, 52.76; H, 4.10; N, 14.98.
Example 21 - BIOLOGICAL METHODS
PHARMACOLOGICAL ACTION MECHANISMS
We present results from experiments that support the present claim.
I. Effect on adenosine bioavailability and on cardiac adenosine

Physiological and pharmacological data of experiments carried out with quinazoline compounds point out that their cardiovascular effects are mediated by adenosine or by the activation of its receptors. We conducted adenosine dosages in the myocardium of isolated rat hearts with compounds 4d (Example 7) and 4i (Example 17). Chromatograms obtained through HPLC experiments in rat myocardium extracts for dosage of tissue adenosine and AMP are presented in Figure 6. As pointed out in the chromatograms (1st peak = adenosine; 2nd peak = AMP), the treatment with the compound 4d increased substantially the amount of myocardial adenosine. Average results point out a basal adenosine value of 0.48 nmol/mg of protein, and in hearts treated with 4d values were 0.75 nmol/mg of protein.
Experiments conducted in HPLC with adenosine derivatized using 2-chloroacetaldehyde for fluorescence detection used as a substrate in myocardium extracts, were able to confirm our hypothesis that compounds 4d (Example 7) and 4i (Example 17) are adenosine kinase inhibitors.
II. Effect of quinazoline compounds on systolic pressure of the left ventricle and heart rate of isolated rat hearts.
Next, typical examples of pressure records are represented, which were conducted to evaluate the effect of injections of increased concentrations of quinazoline compounds, as well, as the vehicle (DMSO), on the function of isolated rat hearts. We evaluated the effects of three different quinazoline compounds [4d (Example 7), 3a (Example I) and 4i (Example 17)]. All compounds caused increase in the systolic pressure of the left ventricle (LVP), depending on the injected concentration, an effect that was not observed when the vehicle DMSO was separately administered. As demonstrated in the following examples, the compounds tested presented different potencies of their pressor effects in isolated hearts. 4i (Example 17) was the compound

that produced a higher pressor response, when infused in concentrations between 30pM - 2}iM (maximum pressor response = 21 ± 3 mmHg), while 4d (Example 7) presented the lowest response (maximum pressor response = 8 ± 4 mmHg). Representative examples of systolic pressure records of the left ventricle (LVP) from concentration-response experiments with quinazoline compounds in isolated rat hearts are presented in Figure 7.
Next, we present concentration-response relationships of arterial pressure and heart frequency, respectively, of the three quinazoline compounds mentioned before. Systolic pressure responses of the left ventricle were expressed as fractional values, while heart rate responses were expressed as absolute values. In Table 1, we present values of Emax, ECso ( nM ) and LogECso in the fractional pressor response to infusion of increased concentrations of compounds like 4d (Example 7), 3a (Example 1) and 4i (Example 17) in isolated rat hearts ( mean ± S.E.M.). In Figure 8, we present concentration-response curves to compounds 4d (Example 7), 3a (Example 1) and 4i (Example 17) on systolic pressure of the left ventricle in isolated rat hearts (expressed as fractional values). Data are presented as mean ± S.E.M. * p TABLE 1

4i (Example 17) 3a (Example 1) 4d (Example 7)
Emax 0.9 ±0.2 0.6 ± 0.2 0.3 ±0.1
ECSO 0.1±0.03 0.6 ± 0.5 6.0 ±1.2
LogECso -9.9 ± 0.3 -9.2 ± 0.5 -8.2 ± 0.2

The increase in systolic pressure was approximately 35%, 30% and 14% for 4i (Example 17), 3a (Example 1) and 4d (Example 7), respectively, according to basal absolute values of systolic pressure. Emax values calculated for the respective curves were 0.9 ± 0.2 (4i, Example 17); 0.6 ± 0.2 (3a, Example 1); 0.3 ± 0.1 (4d, Example 7), in fractional values (Table 1), but the statistical test did not demonstrate significant difference between groups. EC50 values for pressure curves of 4i (Example 17) and of 3a (Example 1) were statistically higher than those of 4d (Example 7). However, there was no difference between ECso of 4i (Example 17) and 3a (Example 1). Responding to the three compounds mentioned, there a decrease in the heart rate dependent concentration. The bradycardia responses were different from each other, and were approximately 24%, 29% and 25% for 4i (Example 17), 3a (Example 1) and 4d (Example 7), respectively, if compared to basal values. In Table 2, we present initial and final values of heart rate (bpm), responding to infusions of increased concentrations of 4i (Example 17), 3a (Example 1) and 4d (Example 7) in isolated rat hearts (mean + S.E.M).
TABLE 2
4i (Example 17) 3a (Example 1) 4d (Example 7)
Initial heart rate 228 ± 7 242 ± 5 219±12
Final heart rate' 173 ± 8 173 ± 1 164 + 3
In Figure 9, we present concentration-response curves to 4i (Example 17), 3a (Example 1) and 4d (Example 7) on the heart rate of isolated rat hearts (expressed as absolute values). Data are presented as mean ± S.E.M.
In all concentration-response experiments of compounds 4i (Example 17), 3a (Example 1) and 4d (Example 7) in isolated hearts, we observed an increase in

svslolic pressure ol ihc left ventricle with simultaneous decrease in hcail MK. both dependent on compound concentration on perlusiuu hutlcr. .As in hcails isolaied and perfused \\illi crystalloid, solutions, variations in heart rale rna\ modih oxygen inllo\\ lo llie imocardium. and consequently its function, il is possible lhal heart rale - d \\ith increased doses of 4i (Fxample 17). In figure id \\e represent the correlatinu lieisve-en ventricular pressure and heart rate, suHL'.esini;j an inverse relationship U.-iueen \'enlncnl.ir pressure aiul heart rate in the presence ol increased doses ul cunip»mul 4i (1 ..sample 1 /)
1 here was also a negative currelalion heluecn levels ol \-...>-,n i>. ulai pressure and heart iale in isolated hearts heated -Aith increased (.loses ol i.onip-iund 4i ((•.sample 17) in the presence ol Mockers like propanolol and dillia/em. and reduced concentrations ol calcium in the perlusion butler In Figure I I. \\e presenied diagrams ol the correlation belueen increase in systolic pressure and decrease in he.ni iale in isolated hearts submitted to injections (bolus) ul 4i ([-..sample 17) peiln-.ed sMth, propanolol. diltia/em and \vith reduction ol the eaiemm concenlralion in the peclusiuu bnlier 1 )ata are presented as mean i S.K.M
In order to confirm our hypolhesis I ha I (iressor response o| (.umpnund 4i depends on bradxcardia ami not on a direct action ol 4i in the inotropisin ol i--..-lated rai hearts. \\e coiklncted experiments where heart rale \\,i-, held constant dm in:.1 inlusiuii o! increased concenii,ili(»ns of 4i (l;.xamp!e 17). through the action of ..MI electiical laloi. In Figure 12 \ve represent the coiicenlration-response relaliou--.liip lo 4i

(I Aampic I/) in isolated heal 1 submitted to clec li ical si muilus ahiiosi annulled ilk" pressor response lo 4i (lixamplc I /).
Kesiills nblamed \\ ilii 4i (kxainple 17) and otlici (.|inna/oliMc i omptmink, point on) Ilia! ihe main lunctional ellecl ol these compounds on isolated lai hcaiis r-niaihcardia aiul tlie prcssor ellecl depends on bnuKcardia, and llin-. is a con-,ci.|ueiK.c iii pai in. ulai ities ol ihe preparation used (i.e isolated heart) and noi ,i dneu elleel. Bci..air-.c llie iirad\ eardia response \\'as not allei'.'d hy propanolol 01 dihia/em hloekiii!1. 01 even by decrease in calcium concentration in ihe perlusion butlei. i-nr h\p.iilie-as i •• dial this elleel ol •. in isolated heads, similai to those observed with qmna/oline componinl--, (i.e. hiad\ >. aidi.i and increase ot \\stolic (iressure ol the lell ventiicle). In ordei i.> o.nhim um h\ pi'ihesis, \se conducted concentration-eflcct e.xperinu.'iit.s \\il\\ a.dennane and ihe nnspccilic inlnbilor ol its receptor (S-phenyllheopIn Iline S-l'l ). \\hkh denninsiiaird as indicated m I ii'iire I s, that adenosine produced an increase ol Ih1.' mini I;.' in ihe s'eiiti k.'ular picssnie ol isolated hearts, and reduction ol approximated IV",, m heaii iaie i om pa red lo ahsoiule basal \ allies, in the eoncen Ira lion ol I jiM of adeno -.me ( ma Mininn
kt ,,, value lor adenosine pressor response uas 7.0 ' I.I n.\l. and loi bradxcardia response' uas I.HI 1,4 nM. In Table x \\e present \alues "i I , I ( , mM) and l.oul'l'.,, in (lie Iraelional pressor response to inlusion ol increased ^viiiv.cntiaiion-, D| ade'nosine in isolated hearts perfused uith pure IlkPkS bnll'er. or \viih 111 I'! S bnllei \\ Mh N -/"/ ( mean i S k.M ).

Hoili ellects \\crc altered by the action ol die mispci. iii-, a icccplor inhibitor K-i'l (IfiM), added lo tlic pcrlusion hnllci. A(.coidin:.' l
presented in I ii'iiic l.i. the increase in svstolk pressure in icspoir-c ts. \\hen the inliihiini \sas piesenl. lliere \\as a shili oflhe liC'^o value In liu: lell. |iresentiii!.! the ne\\ \alue ul •«(H|() nM ('lahle M- il i.umpand to basal values ol I able -4, \\here \vc present initial and linal heart iate s alues tbpni) in die re/spouse lo inlusion of increased coneenlrations of adeiiosme in isolated he.ui-, perfused uiih pine lll-l'l^S bidTerand IIHI'r.S buftet \\ith \-/'/ ( mean • S.I Al.i.




0.43 i-() 0s 7.0-M.I -8.2M).?.
'I ABI I -Atlenoaint-
Initial Ih'tin Rule 2281:2
Until Henri Rate 185±2

In l'i;j,ure 14. u'e present didLiianis dial leprcscni lieaii i ih
isolated lal heart', ]>'.'rinsed \sith pure llb'.PI-'.S bulk'i dondol) or III |'| s huik-r \s iii.Mol 4id sample 1 7). or 1 IklM'S buffer \vilh I nM ol 4i i \ \arnp e I'.') pin- in\l

uiispecihe inhibitor ol adenosine receptors, 8-P'l (4i > X-PI). Data are pie-denied as mean ' S.I1 .M. lor 4 experiments. * p •- 0.05 compared to the v ontrol. and demounti-aled thai perliision of hearts \vilh 4i (I'.xample 17) reduces hear! rate in approximated l'/"u. li compared lo hearl rate values ol control hearts, Hoxvever. pcrliiMon i>l In aits \\iih 4i (I xample 17) and K-PI annulled bradycardia caused In 4i (Pxainplc I /). lonlumm:1 om h\ polhesis that ibe quina/oline compound could be aelum in a direct »i indirect \\a\ on adenosine receptors. In Table 5, we present lieait rale values ihpm) I rum individual i.-.\periments ol isolated hearts perfused with pure III PI S butler (conirnl). Ill PI s bnfler \\ith InM of 4i (I'.xample 17), or 1-I1{PI;S bnflei with InM of 4i (1 xample l/i and !(iM ol'\- ITjmcan i S.l-.M.).
'I'ABLf, 5
•// (!'\isniph' I '} Control 4il4i (Example -7/ 7) (InM)
(InM) i .V- /'/

CLAIM:-
1 - COMPOUNDS, characterized by Molecular Formula I, according to figure
1, where RI and R2 correspond to the alcoxy group like methoxyl; R3 corresponds to
substitutes like hydrogen (H), halogen (F, Cl, Br and I), methoxyl (OCH3), methyl (CH3),
acetyl [C(O)CH3], 7V,7V-dimethylamine [N(CH3)2] and nitro (NO2), where each substitute may
occupy positions 3' or 4' of the jV-phenyl group, generating meta and />ara-substituted
quinazolinic compounds.
2 - compound according to claim 1, wherein R3 is Af.TV-dimethylamine
[N(CH3)a], and being located in position 3' of the JV-phenyl group.
3 - compound according to claim 1, wherein R3 is N,N- dimethylamine
[N(CH3)i], and being located in position 4' of the jV-phenyl group.
4 compounds according to claim 1, wherein R3 may be methyl, chlorine,
bromine, iodine, acetyl, nitro and methoxyl. Substitutes are located in position 4' of the N-
phenyl ring.
5 - compounds according to claims 1, 2, 3 and 4, characterized by having a
pharmacologically acceptable formula (substitutes in positions 3' or 4') for usages stated in
claims 8 to 19.
6 - A pharmacologically acceptable composition comprising the compound as
claimed in claim 1 .
1 - COMPOUNDS, comprising an adenosine-kinase inhibitor effect of quinazoline derived compounds that were previously known as reversible inhibitors of tyrosine-kinases, and producing protection for the myocardium, brain, kidneys, and liver to the effects of ischemia/reperfusion.
8 - COMPOUNDS, and PROCESS of inhibiting adenosine-kinases, both characterized as a process that comprises administering an amount of a compound according to claims 1 to 7 to a patient who needs adenosine-kinase inhibition.

9- A compound having the formula:
(FIGURE REMOVE)
or a pharmaceutically acceptable salt thereof, which inhibits the enzyme adenosine kinase.
10- The compound according to claim 19 which is 6,7-dimethoxy-4-(3'-
dimetylaminoanilino)quinazoline.
11- The compound according to claim 19 which is 6,7-dimemoxy-4-(4'-
dimetylaminoanilino)quinazoline.
12- The compound according to claims 20 or 21 which is in the form of
hydrochloride salts.
13- A pharmaceutical composition comprising a therapeutically effective
amount of a compound according to claims 20 or 21 in combination'with a pharmaceutically
acceptable carrier.
14. Compounds, pharmacologically acceptable composition, compounds comprising an adenosine-kinase inhibitor effect, a compound, a pharmaceutical composition substantially as illustrated in the foregoing examples.

Documents:

http://ipindiaonline.gov.in/patentsearch/GrantedSearch/viewdoc.aspx?id=xG9z3BvYfKEqgQz+levM2w==&amp;loc=+mN2fYxnTC4l0fUd8W4CAA==


Patent Number 268634
Indian Patent Application Number 5554/DELNP/2006
PG Journal Number 37/2015
Publication Date 11-Sep-2015
Grant Date 09-Sep-2015
Date of Filing 25-Sep-2006
Name of Patentee UNIVERSIDADE ESTADUAL DE CAMPINAS-UNICAMP
Applicant Address CIDADE UNIVERSITARIA "ZEFERINO VAZ" DISTRITO DE BARAO GERALDO, 13084-971 CAMPINAS -SP, BRAZIL.
Inventors:
# Inventor's Name Inventor's Address
1 GOMES FRANCHINI, KLEBER FAULDADE DE CIENCIAS MEDICA-UNICAMP,DISTRITO DE BARAO GERALDO, 13084-971 CAMPINAS -SP, BRAZIL.
2 ABDALLA SAAD, MARIO, JOSE RUA JOSE DA SILVA MARTINHO, 136,13094-530 CAMPINAS-SP, BRAZIL
3 RITTNER NETO, ROBERTO RUA DR ANTONIO HOSSRI 45, 13083-370 CAMPINAS -SP, BRAZIL.
4 MARIN, RODRIGO, MIGUEL RUA ARTUR RAMOS, 381, 13041-302- CAMPINAS -SP, BRAZIL
5 ROCCO APARECIDA, SILVANA RUA CAPITAO JOSE DE SOUZA, 56-AP.12, 13020-470 CAMPINAS-SP, BRAZIL
PCT International Classification Number A61K 31/517
PCT International Application Number PCT/BR2004/000196
PCT International Filing date 2004-10-07
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 PI 0400869-3 2004-03-02 Brazil