Title of Invention

VITRONECTIN RECEPTOR ANTAGONIST PHARMACEUTICALS

Abstract A compound, comprising: a targeting moiety and a chelator, wherein the targeting moiety is bound to the chelator, is a quinolone nonpeptide, and binds to a receptor that is upregulated during angiogenesis and the compound has 0-1 linking groups between the targeting moiety and chelator, wherein the receptor is the integrin <xvP3 or ctvPs and tne compound is of the formula: (Q)d-Ln-Ch or (Q)d-Ln-(Ch)d. wherein, Q is a compound of Formula (II):
Full Text FORM 2
THE PATENTS ACT 1970
[39 OF 1970]
&
THE PATENTS RULES, 2003
COMPLETE SPECIFICATION
[See Section 10; rule 13]
"VITRONECTIN RECEPTOR ANTAGONIST PHARMACEUTICALS"
BRISTOL-MYERS SQUIBB PHARMA COMPANY, of P.O. Box 4000, Princeton, New Jersey 08543-4000, United States of America,
The following specification particularly describes the invention and the manner in which it is to be performed:


TITLE
VITRONECTIN RECEPTOR ANTAGONIST PHARMACEUTICALS
FIELD OF THE INVENTION
5 The present invention provides novel pharmaceuticals useful for the diagnosis and treatment of cancer, methods of imaging tumors in a patient, and methods of treating cancer in a patient. The pharmaceuticals are comprised of a targeting moiety that binds to the vitronectin 10 receptor that is expressed in tumor vasculature, an
optional linking group, and a therapeutically effective radioisotope or diagnostically effective imageable moiety. The therapeutically effective radioisotope emits a gamma ray or alpha particle sufficient to be cytotoxic. 15 The imageable moiety is a gamma ray or positron emitting radioisotope, a magnetic resonance imaging contrast agent, an X-ray contrast agent, or an ultrasound contrast agent.
20 -*
BACKGROUND (MtTHB INVENTION
Cancer ^is a major publij^health concern in the United States and around the world. It is estimated that over 1 million new cases of invasive cancer will be
25 diagnosed in the United States in 1998. The most
prevalent forms of the disease are solid tumors of the lung, breast, prostate, col oft"'and rectum. Cancer is typically diagnosed by a combination of in vitro tests and imaging procedures. The imaging procedures include
30 X-ray computed tomography, magnetic resonance imaging, ultrasound imaging and radionuclide scintigraphy. Frequently, a contrast agent is administered to the patient to enhance the image obtained by X-ray CT, MRI and ultrasound, and the administration of a
35 radiopharmaceutical that localizes in tumors is required for radionuclide scintigraphy.
Treatment of cancer typically involves the use of external beam radiation therapy and chemotherapy, either alone or in combination, depending on the type and extent

of the disease. A number of chemotherapeutic agents are available, but generally they all suffer from a lack of specificity for tumors versus normal tissues, resulting in considerable side-effects. The effectiveness of these 5 treatment modalities is also limited, as evidenced by the high mortality rates for a number of cancer types, especially the more prevalent solid tumor diseases. More effective and specific treatment means continue to be needed. 10 Despite the variety of imaging procedures available for the diagnosis of cancer, there remains a need for improved methods. In particular, methods that can better differentiate between cancer and other pathologic conditions or benign physiologic abnormalities are 15 needed. One means of achieving this desired improvement would be to administer to the patient a
metallopharmaceutical that localizes specifically in the tumor by binding to a receptor expressed only in tumors or expressed to a significantly greater extent in tumors 20 than in other tissue. The location of the
metallopharmaceutical could then be detected externally either by its imageable emission in the case of certain radiopharmaceuticals or by its effect on the relaxation rate of water in the immediate vicinity in the case of 25 magnetic resonance imaging contrast agents.
This tumor specific metallopharmaceutical approach can also be used for the treatment of cancer when the metallopharmaceutical is comprised of a particle emitting radioisotope. The radioactive decay of the isotope at 30 the site of the tumor results in sufficient ionizing radiation to be toxic to the tumor cells. The specificity of this approach for tumors minimizes the amount of normal tissue that is exposed to the cytotoxic agent and thus may provide more effective treatment with 35 fewer side-effects.
Previous efforts to achieve these desired improvements in cancer imaging and treatment have centered on the use of radionuclide labeled monoclonal antibodies, antibody fragments and other proteins or


polypeptides chat bind to tumor cell surface receptors. The spec:ficity of these radiopharmaceuticals is frequently very high, but they suffer from several disadvantages. First, because of their high molecular 5 weight, they are generally cleared from the blood stream very slowly, resulting in a prolonged blood background in the images. Also, due to their molecular weight they do not extravasate readily at the site of the tumor and then only slowly diffuse through the extravascular space to 10 the tumor cell surface. This results in a very limited amount of the radiopharmaceutical reaching the receptors and thus very low signal intensity in imaging and insufficient cytotoxic effect for treatment.
Alternative approaches to cancer imaging and therapy 15 have involved the use of smal"3r~molecules, such as
peptides, that bind to tumor cell surface receptors. An In-Ill labeled somatostatin receptor binding peptide, In-lll-DTPA-D-Phe1-ccteotide# is in clinical use in many countries for imaging t umors .^that express the 20 somatostatin receptor (BaTcerT-et al. Life Sci., 1991, 49, 1583-91 and Krenning, et 'al. pEur. J. Nucl. Med., 1993, 20, 716-31K Higher doses cff*thi7s* radiopharmaceutical have been investigated for potential treatment of these types of cancer (Krenning, et al., Digestion, 1996, 57, 25 57-61) . Several groups are investigating the use of Tc-99m labeled ananlogs of In-lll-DTPA-D-Phe1-octeotide for imaging and Re-186 labeled analogs for therapy (Flanagan, et al., U.S. 5,556,939, Lyle, et al., U.S. 5,382,654, and Albert et al.,U.S. 5,650,134). 30 Angiogenesis is the process by which new blood
vessels are formed from pre-existing capillaries or post capillary venules; it is an important component of a variety of physiological processes including ovulation, embryonic development, wound repair, and collateral 35 vascular generation in the myocardium. It is also
central to a number of pathological conditions such as tumor growth and metastasis, diabetic retinopathy, and macular degeneration. The process begins with the

activation of existing vascular endothelial cells in response to a variety of cytokines and growth factors. Tumor released cytokines or angiogenic factors stimulate vascular endothelial cells by interacting with specific 5 cell surface receptors for the factors. The activated endothelial cells secrete enzymes that degrade the basement membrane of the vessels. The endothelial cells then proliferate and invade into the tumor tissue. The endothelial cells differentiate to form lumens, making 10 new vessel offshoots of pre-existing vessels. The new blood vessels then provide nutrients to the tumor permitting further growth and a route for metastasis.
Under normal conditions, endothelial cell proliferation is a very slow process, but it increases 15 for a short period of time during embryOgenesis,
ovulation and wound healing. This temporary increase in cell turnover is governed by a combination of a number of growth stimulatory factors and"growth suppressing factors. In pathological angiogenesis, tljis normal 20 balance is disrupted resulting in continued increased endothelial cell proliferation/ Some of the proangiogen^e factors that have -been identified include basic fibroblast growth factor (bFGF), angiogenin, TGF-alpha, TGF-beta, and vascular endothelium growth factor 25 (VEGF) . While mterferon-alpha, interferon-beta and
thrombospondm are examples of angiogenesis suppressors.
The proliferation and migration of endothelial cells in the extracellular matrix is mediated by interaction with a variety of cell adhesion molecules (Folkman, J., 30 Nature Medicine , 1995, 1, 27-31). Integrins are a
diverse family of heterodimeric cell surface receptors by which endothelial cells attach to the extracellular matrix, each other and other cells. The integrin auP, is a receptor for a wide variety for a wide variety of 35 extracellular matrix proteins with an exposed tripeptide Arg-Gly-Asp moiety and mediates cellular adhesion to its ligand: vitronectin, fibronectin, and fibrinogen, among
others. The mtegrin ocvp\ is minimally expressed on normal
■&


blood vessels, but is significantly upregulated or; vascular cells within a variety of human tumors. The role of the avP, receptors is to mediate the interaction of the endothelial cells and the extracellular matrix ar.d 5 facilitate the migration of the cells in the direction of the angiogenic signal, the tumor cell population. Angiogenesis induced by bFGF or TNF-alpha depend cr. the agency of the integrm av(33, while angiogenesis induced by VEGF depends on the integrin av(33 (Cheresh et. al., 10 Science, 1955, 270, 1500-2). Induction of expression of the integrins a,3, and a2P. on the endothelial cell surface is another important mechanism by which VEGF promotes angiogenesis (Senger, et. al., Proc. Natl. Acad, £ci USA, 1997, 84, 13612-7) . 15 Angiogenic factors interact with endothelial cell surface receptors such as the receptor tyrosine kinases EGFR, FGFR, PDGFR, Flk-1/KDR, Flt-1, Tek, tie, neuropilin-1, endoglin, endosialin, and Axl. The receptors Flk-1/KDR, neuropilin-1, and FH*-1 recognize 20 VEGF and these interactions play key'roles in VEGF-induced angiogenesis. The Tie subfamily>of:receptor tyrosine kinases are also expressed prominently during blood vessel formation.
Because of the importance of angiogenesis to tumor 25 . growth and metastasis, a number of chemotherapeutic approaches are being developed to interfere with or prevent this process. One of these approaches, involves the use of anti-angiogenic proteins such as angiostatin and endostatin. Angiostatin is a 38 kDa fragment of 30 plasminogen that has been shown in animal models to be a potent inhibitor of endothelial cell proliferation. (O'Reilly et. al. , Cell, 1994, 79, 315-328) Endostatin is a 20 kDa C-terminal fragment of collagen XVIII that has also been shown to be a potent inhibitor. (O'Reilly 35 et. al., Cell, 1997, 88, 277-285) Systemic therapy with endostatin has been shown to result in strong anti-tumor activity in animal models. However, human clinical trials

of these two chemotherapeutic agents of biological origin have been hampered by lack of availability.
Another approach to anti-angiogenic therapy is to use 5 targeting moieties that interact with endothelial cell surface receptors expressed in the angiogenic vasculature to which are attached chemotherapeutic agents. Burrows and Thorpe (Proc. Nat. Acad. Sci, USA, 1993, 90, 8996-9000) described the use of an antibody-immunotoxin 10 conjugate to eradicate tumors in a mouse model by destroying the tumor vasculature. The antibody was raised against an endothelial cell class II antigen of the major histocompatibility complex and was then conjugated with the cytotoxic agent, deglycosylated ricm 15 A chain. The same group (Clin. Can..-Res., 1995, 1, 1623-1634) investigated the use of antibodies raised against the endothelial cell surface receptor, endoglin, conjugated to deglycosylated ricin- A- chain. Both of these conjugates exhibited potent anti-tumor activity in mouse 20 -models. However, both still suffer'drawbacks to routine human use. As with most antibodies .or other large, foreign proteins, there is considerable; risk of immunologic toxicity which could limit or preclude administration to humans. Also, whiTe the vasculature 25 targeting may improve the local concentration of the
attached chemotherapeutic agents, the agents still must be. cleaved from the antibody carrTe'r* arid be transported or diffuse into the cells to be cytotoxic.
Thus, it is desirable to provide anti-angiogenic 30 pharmaceuticals and tumor or new vasculature imaging agents which do not suffer from poor diffusion or transportation, possible immunologic toxicity, limited availability, and/or a lack of specificity.
Another application of anti-angiogenic therapy is in 35 treating rheumatoid arthritis (RA). In RA, the ingrowth of a highly vascularized pannus is caused by the excessive production of angiogenic factors by the infiltrating macrophages, immune cells, or inflammatory cells. Therefore, it is desirable to have new

pharmaceuticals to destroy the highly vascularized pannus that results and thus treat the disease.
There is also a growing interest in therapeutic angiogenesis to improve blood flow in regions of the body 5 that have become ischemic or poorly perfused. Several investigators are using growth factors administered locally to cause new vasculature to form either in the limbs or the hearr. The growth factors VEGF and bFGF are the most common for this application. Recent 10 publications include.- Takeshita, S., et. al., J. Clm. Invest., 1994, 92, 662-670; and Schaper, W. and Schaper, J., Collateral Circulation:Heart, Brain, Kidney, Limbs, Kluwer Academic Publishers, Boston, 1993. The main applications that are under investigation in a number of 15 laboratories are for improving cardiac blood flow and in improving peripheral vessal blood flow in the limbs. For example, Henry, T. et. al. (J. Amer. College Cardiology, 1998, 31, 65A) describe the use of recombinant human VEGF in patients for improving myocardial perfusion by 20 therapeutic angiogenesis. Patients received infusions of rhVEGF and were monitored by nuclear perfusion imaging 30 and 60 days^post treatment to determine, improvement in myocardial perfusion. About 50% of patients showed improvement by nuclear perfusion imaging whereas 5/7 25 showed new collatoralization by angiography. Thus, it is desirable to discover a method of monitoring improved cardiac blood flow which is targeted to new collatoral vessels themselves and not, as in nuclear perfusion imaging, a regional consequence of new collatoral 30 vessels.
The detection, imaging and diagnosis of a number cf cardiovascular diseases need to be improved, including restenosis, atherosclerosis, myocardial reperfusion injury, and myocardial ischemia, stunning or infarction. 35 It has recently been determined that in all of these
disease conditions, the integrin receptor ocvP3 plays an important role.


For example, in the restenosis complication that occurs in -30-50% of patients having undergone angioplasty or stent placement, neointimal hyperplasia and ultimate reocclusion is caused by aggressively 5 proliferating vascular smooth muscle cells that express av|}3. (Cardiovascular Res., 1997, 36, 408-428; DDT, 1997, 2, 187-199; Current Pharm. Design, 1997, 3, 545-584)
Atherosclerosis proceeds from an intial endothelial damage that results in the recruitment and subintimal 10 migration of monocytes at the site of the injury. Growth factors are released which induce medial smooth muscle cells to proliferate and migrate to the intimal layer. The migrating smooth muscle cells express avp3.
In reperfusion injury, neutrophil transmigration is 15 integrin dependent and the integrins moderate initial
infiltration into the viable border zone. The induction of a5pi, cc4p>l and avp"5 in infiltrating neutrophils occurs within 3 to 5 hours after reperfusion as neutrophils move from the border 2one to the area of necrosis. 20 (Circulation, 1999, 100, 1-275)
Acute or chronic occlusion of a coronary artery is known to result in angiogenesis in the heart as native collateral vessels are recruited to attempt to relieve the ischemia. However, even a gradual occlusion usually 25 results in areas of infarction as the resulting
angiogenesis is not sufficient to prevent damage. Cardiac angiogenesis has been associated with increased expression of the growth factors VEGF and FGF and the upregulation of the growth factor receptors flt-1 and 30 flk-1/KDR. (Drugs, 1999, 58, 391-396)

SUMMARY OF THE INVENTION It is one object of the present invention to provide improved anti-angiogenic pharmaceuticals, comprised of a targeting moiety that binds to the vitronectin receptor 5 that is expressed in tumor neovasculature, an optional linking group, and a radioisotope. The vitronectin receptor binding compounds target the radioisotope to the tumor neovasculature. The beta or alpha-particle emitting radioisotope emits a cytotoxic amount of 10 ionizing radiation which results in cell death. The
penetrating ability of radiation obviates the requirement that the cytotoxic agent diffuse or be transported into the cell to be cytotoxic.
It is another object of the present invention to 15 provide pharmaceuticals to treat rheumatoid arthritis. These pharmaceuticals comprise a targeting moiety that binds to a receptor that is upregulated during angiogenesis, an optional linking group, and a radioisotope that emits cytotoxic radiation (i.e., beta 20 particles, alpha particles and Auger or Coster-Kronig electrons). In rheumatoid arthritis, the ingrowth of a highly vascularized pannus is caused by the excessive production of angiogenic factors by the infiltrating macrophages, immune cells, or inflammatory cells. 25 Therefore, the radiopharmaceuticals of the present
invention that emit cytotoxic radiation could be used to destroy the new angiogenic vasculature that results and thus treat the disease.
It is another object of the present invention to 30 provide imaging agents, comprised of vitronectin receptor binding compounds conjugated to an imageable moiety, such as a gamma ray or positron emitting radioisotope, a magnetic resonance imaging contrast agent, an X-ray contrast agent, or an ultrasound contrast agent. These 35 imaging agents are useful for imaging tumor
neovasculature, therapeutic angiogenesis interventions in the heart, natural angiogenic processes in response to acute or chronic coronary vessel occlusion, restenosis


post-angioplasty, atherosclerosis and plaque formation, and reperfusion injury.
It is another object of the present invention to provide compounds useful for preparing the 5 pharmaceuticals of the present invention. These compounds are comprised of a non-peptide quinolone containing targeting moiety that binds to a receptor that is upregulated during angiogenesis or during cardiovascular diseases, Q, an optional linking group, 10 Ln, and a metal chelator or bonding moiety, Ch- The
compounds may have one or more protecting groups attached to the metal chelator or bonding moiety. The protecting groups provide improved stability to the reagents for long-term storage and are removed either immediately 15 prior to or concurrent with the synthesis of the
radiopharmaceuticals. Alternatively, the compounds of the present invention are comprised of a peptide or peptidomimetic targeting moiety that binds to a receptor that is upregulated during angiogenesis or during 20 cardiovascular diseases, Q, an optional linking group, Ln/ anda surfactant, Sf.
The. pharjnaceuticals of the present invention may be used for diagnostic and/or therapeutic purposes. Diagnostic radiopharmaceuticals of the present invention 25 are pharmaceuticals comprised of a diagnostically useful radionuclide (i.e., a radioactive metal ion that has imageable gamma ray or positron emissions). Therapeutic radiopharmaceuticals of the present invention are pharmaceuticals comprised of a therapeutically useful 30 radionuclide, a radioactive metal ion that emits ionizing radiation such as beta particles, alpha particles and Auger or Coster-Kronig electrons.
The pharmaceuticals comprising a gamma ray or positron emitting radioactive metal ion are useful for 35 imaging tumors and by gamma scintigraphy or positron emission tomography. The pharmaceuticals comprising a gamma ray or positron emitting radioactive metal ion are also useful for imaging therapeutic angiogenesis, natural

angiogenic processes in response to acute or chronic
coronary vessel occlusion, restenosis post-angioplasty,
atherosclerosis and plaque formation, and reperfusion
injury by gamma scintigraphy or positron emission
5 tomography. The pharmaceuticals comprising a particle
emitting radioactive metal ion are useful for treating
cancer by delivering a cytotoxic dose of radiation tc the
tumors. The pharmaceuticals comprising a particle
emitting radioactive metal ion are also useful for
10 treating rheumatoid arthritis by destroying the formation
of angiogenic vasculature. The pharmaceuticals
comprising a paramagnetic metal ion are useful as
magnetic resonance imaging contrast agents. The
pharmaceuticals comprising one or more X-ray absorbing or
15 "heavy" atoms of atomic number 20 or greater are useful
as X-ray contrast agents. The pharmaceuticals comprising
a microbubble of a biocompatible gas, a liquid carrier,
and a surfactant microsphere, are useful as ultrasound
contrast agents.
20 *
DETAILED DESCRIPTION OF THE INVENTION
__7 ___
[1] Thus, in a first embodiment, the present invention provides a novel compound, comprising: a targeting
25 moiety and a chelator, wherein the targeting moiety is bound to the chelator, is a quinolone nonpeptide, and binds to a receptor that is upregulated during angiogenesis and the compound has 0-1 linking groups between the targeting moiety and chelator.
30
[2] In a preferred embodiment, the receptor is the integrin av^3 or avps and the compound is of the formula:
(Q)d-Ln-Ch or {Q)d-Ln-(Ch)d*
35 wherein, Q is a compound of Formula (II):




:ID
5 including stereoisomeric forms thereof, or mixtures of stereoisomeric forms thereof, or pharmaceutically acceptable salt or prodrug forms thereof wherein:
Rle is selected from:


10
15

Ae is -CH2- or -N(Ri0e)-;
Ale and Ee are independently -CH2- or -N(R10e)-;
De is -N(R10e)- or -S-;
Ee_pe is _C(R2e)^C(R3e; . cr -C(R2e)2C(R3e)2" ;
Je is -C(R2e)- or -N-;
Ke, Le and Me are independently -C(R2e)- or -C(R3e)-;
R2e and R3e are independently selected from.-
H, C1-C4 alkoxy, NRlieR12e, halogen, N02, CN, CF3, C1-C6 alkyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl), aryl (C1-C6 alkyl)-, (Ci-Ce alkyl )carbonyl, (C1-C6 alkoxy)carbonyl, arylcarbonyl, and aryl substituted with 0-4 R7e,
alternatively, when R2e and R3e are substituents on adjaceart atoms, they can be taken together with the carbon atoms to which they are attached to form a 5 7 membered carbocyclic or 5-7 membered heterocyclic aromatic or nonaromatic ring system, said carbocyclic or heterocyclic ring being substituted with 0-2 groups selected from C1-C4 alkyl, C1-C4 alkoxy, halo, cyano, amino, CF3 and N02;
R2ae is selected from:
H, C1-C10 alkyl, C2-Ce alkenyl, C3-C11 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl), aryl, aryl(Ci-C4 alkyl)-, (C2-C7 alkyl)carbonyl, arylcarbonyl, (C2-C10 alkoxy)carbonyl, Cj-Ci cycloalkoxycarbonyl, C7-C11 bicycloalkoxycarbonyl, aryloxycarbonyl, aryl(Ci-Cio alkoxy)carbonyl,
C1-C6 alkylcarbonyloxy(C1-C4 alkoxy)carbonyl, arylcarbonyloxy (C1-C4 alkoxy) carbonyl, and


C3-C7 cycloalkylcarbonyicxy|Ci-C4 alkoxy)carbonyl;
R7e is selected from:
H, hydroxy, C1-C4 alkyi, C1-C4 alkoxy, aryl, aryl(Ci~ C4 alkyl)-, (C1-C4 alkyljcarbonyl, C02R18ae, S02Rlle, SO2NR10eRlle, OR10e, and Nt'Rlle)R12e;
Ue is selected from:
-(CH2)ne-, -(CH2)neO(CH2)me-, - (CH2 > n*K(R12) (CH2 ) me" --NH(CH2)ne-. -(CH2)neC(=0; (CH2)me-. -(CH2)neS(0)pe(CH2)me-- " (CH2)neNKNK{CH2)me-, -N(R10e)C(=O)-, -NHC(=0) (CH2)ne-, -C (=0) N(R10e) - , and -N(R10e)S(0)pe-;
Ge is N or CR19e;
We is-C(=O)-N(R10e)-(Ci-C3 alkylene)-, in which the
alkylene group is substituted by R8e and by R9e:
R8e and R9e are independently selected from:
H. C02R18be, C(=0)R18be, CONR17R18be,
C1-C10 alkyl substituted with 0-1 R6e,
C2-C10 a*lkenyl substituted with 0-1 R6e,
C2-C10 alkynyl substituted with 0-1 R6e,
C3-C8 cycloalkyl substituted with 0-1 R6e,
C5-C6 cycloalkenyl substituted with 0-1 R6e,
(C1-C10 alkyl)carbonyl,
C3-C10 cycloalkyl (C1-C4 alkyl)-,
phenyl substituted with 0-3 R6e,
naphthyl substituted with 0-3 R6e,
a 5-10 membered heterocyclic ring containing 1-3 N, 0, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e,
C1-C10 alkoxy substituted with 0-2 R7e,
hydroxy, nitro, -N(R10e)Rlle, -N(R16e) Ri7e, aryl(Co-C6 alkyl)carbonyl, aryl(C3-C6 alkyl),

heteroaryl (Ci-C6 alkyl i , CONRla*eR20e, S02R18ae,
and SO2NR18aeR20e,
providing that any of the above alkyl, cycloalkyl,
aryl or heteroaryl groups may be unsubstituted
5 or substituted independently with 1-2 R7e;
F.6e is selected from:
H, Ci-Cio alkyl, hydroxy, Ci-Cio alkoxy, nitro, Ci-Cj
alkylcarbonyl, -N[Rlle)R12e, cyano, halo, CF3,
10 CHO, C02R18be, C(=0)R18be, C0NR17eR18be,
OC(=O)R10e, OR10e, OC(=O)NR10eRile.
NR10eC(=O)R10e, NR10eC(=O)OR21e,
NR10eC(=O)NR10eRlle, NR10eSO2NR10eRlle,
NR10eSO2R21e, S(0)pRlle, SO2NR10eRlle,
15 aryl substituted with 0-3 groups selected from
halogen, C1-C6 alkoxy, Ci-Ce alkyl, CF3, S(0)meMe, and -NMe2, aryl(Ci-C4 alkyl)-, said aryl being substituted with
0-3 groups selected from halogen, C1-C6 alkoxy,
~ >
20 C1-C6 alkyl, CF3, S(0)peMe, and -NMe2, and
a 5-10 membered heterocyclic ring containing 1-3 N, 0, or
S heteroatoJfls, wherein said heterocyclic ring may be
saturated, partially saturated, or fully unsaturated,
said heterocyclic ring being substituted with 0-2 R7e;
25
R10e is selected from:
H, CF3, C3-C6 alkenyl, C3-C11 cycloalkyl, aryl, (C3-C11 cycloalkyl)methyl, aryl(Cx-C4 alkyl), and Cr C10 alkyl substituted with 0-2 R6e;
30
Rlle is selected from:
H, hydroxy, Ci-C8 alkyl, C3-C6 alkenyl, C3-C11
cycloalkyl, (C3-C11 cycloalkyl)methyl, C1-C6 alkoxy,
benzyloxy, aryl, heteroaryl, heteroaryl (C1-C4 alkyl)-
35 , aryl(C1-C4 alkyl), adamantylmethyl, and
Cj-Cio alkyl substituted with 0-2 R4e;
R4e is selected from:

H, C:-C6 alkyl, C3-C7 cycloalkyl, C2-C1 cycloalkyl (C^-C^ alkyl)-, (C1-C10 alkyl!carbonyl, aryl, heteroaryl, aryl(C1-C6 alkyl)-, and heteroaryl(C1-C6 alkyl)-, wherein said aryl or
5 heteroaryl groups are substituted with 0-2
substituents independently selected from the group consisting of Ci-C4 alkyl, Ci-C4 alkoxy, F, CI, Br, CF3, and N02,
10 alternatively, when R10e and Rlle are both substituents on the same nitrogen atom (as in -NR10eRlle; they may be taken together with the nitrogen atom to which they are attached to form a heterocycle selected from: 3-azabicyclononyl, 1,2,3,4-tetrahydro-1-quinolinyl,
15 1,2,3,4-tetrahydro-2-isoquinolinyl, 1-piperidinyl, 1-morpholinyl, 1-pyrrolidinyl, thiamorpholinyl, thiazolidinyl, and 1-piperazinyl,-
said heterocycle being substituted with 0-3 groups selected from: C1-C6 alkyl, aryl, heteroaryl, 20 aryl(Ci-C4 alkyl)-, (C1-C6 alkyl)carbonyl, (C3-C7
cycloalkyl)carbonyl, ~(Ci~C6 alkoxy)carbonyl, aryl(Ci-C4 alkoxy)carbonyl, CiLG6 alkylsulfonyl, and arylsulfonyl;
25 R12e is selected from:
H, Ci-Ce alkyl, triphenylmethyl, methoxymethyl,
methoxyphenyldiphenylmethyl,
trimethylsilylethoxymethyl, (Ci-Ce alkyl)carbonyl, (C1-C6 alkoxy)carbonyl, (Ci-Ce alkyl)aminocarbonyl,
30 C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl (Ci-C4 alkyl)-, aryl, heteroaryl(C1-C6 alkyl)carbonyl, heteroarylcarbonyl, aryl{Ci-C6 alkyl)-, (C1-C6 alkyl)carbonyl, arylcarbonyl, CI-CG alkylsulfonyl, arylsulfonyl, aryl (Ci-Ce
35 alkyl)sulfonyl, heteroarylsulfonyl, heteroaryl(C1-C6 alkyl)sulfonyl, aryloxycarbonyl, and aryl(Ci-C6 alkoxy)carbonyl, wherein said aryl groups are substituted with 0-2 substituents selected from the

group consisting of C1-C4 alkyl, C1-C4 alkoxy, halo, CF3, and nitro;
R16e is selected from:
-C(=0)OR18ae, -C(=0)R18be, -C(=0)N(R18be}2. -C(=0)NHSO2R18ae, -C(=0)NHC(=0)Riebe, -C(=0)NHC(=0)OR1Sae, -C(=0)NKS02NHR18be- -S02R18ae, -S02N(R18be)2, and -S02NHC(=0)OR18be;
R17e is selected from:
H, Ci-C6 alkyl, C3-C7 cycloalkyl, C3-C7
cycloalkyl (C1-C4 alkyl)-, aryl, aryKCi-Ce alkyl)-,
and heteroaryl (Ci-Ce alkyl);
R18ae is selected from:
Cj-Cg alkyl optionally substituted with a bond to Ln, C3-C11 cycloalkyl optionally substituted with a bond to Ln, aryl(Ci-Ce alkyl)- optionally substituted with a bond to Ln, heteroaryl {C1-C6 alkyl)- optionally substituted with a bond to Ln, (Ci-Ce alkyl)heteroaryl optionally substituted with a bond to Ln, biaryl(C1-C6 alkyl) optionally substituted with a bond to Ln/ heteroaryl optionally substituted with a bond to Ln, phenyl substituted with 3-4 R19e and optionally substituted with a bond to Ln,
naphthyl substituted with 0-4 R19e and optionally substituted with a bond to Ln, and a bond to Ln, wherein said aryl or heteroaryl groups are optionally substituted with 0-4 R19e;
R.'.8be 1S H or Rl8ae.
Ri9e is selected from:
H, halogen, CF3, C02H, CN, N02, -NRlleR12e, OCF3, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C11 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl)-, aryl(C1-C6 alkyl)-, Ci-Ce alkoxy, C1-C4

alkoxycarbonyl, aryl, aryl-O-, aryl-S02~,
heteroaryl, and heteroaryl-S02-, wherein said aryl
and heteroaryl groups are substituted with 0-4 groups selected from hydrogen, halogen, CF3, C1-C3
alkyl, and C1-C3 alkoxy;
is selected from:
hydroxy, C1-C10 alkyloxy, C3-C11 cycloalkyloxy,
aryloxy, aryl(Ci-C4 alkyl)oxy,
C2-C10 alkylcarbonyloxy(C1-C2 alkyl)oxy-,
C2-C10 alkoxycarbonyloxy(C1-C2 alkyl)oxy-,
C2-C10 alkoxycarbonyl(C1-C2 alkyl)oxy-,
C3-C10 cycloalkylcarbonyloxy(C1-C2 alkyl/oxy-,
C3-C10 cycloalkoxycarbonyloxy(Ci-C2 alkyl)oxy-,
C3-C10 cycloalkoxycarbonyl(C1-C2 alkyl)oxy~,
aryloxycarbonyl(Ci"C2 alkyl)oxy-,
aryloxycarbonyloxy(C1-C2 alkyl)oxy-,
arylcarbonyloxy(Ci-C2 alkyl)oxy-,
C1-C5 alkoxy{C1-C5 alkyl)carbonyloxy(^-C2 alkyl)oxy,
(5- (C1-C5 alkyl)-1,3-dioxa-cyclopenten-2-one-
yDmethyloxy,
(5-arylTl'3-dioxa-cyclopenten-2-one-yl)methyloxy,
and (RlOe, (Rlle)N_(Cl_Cl0 alkoxy)-;
is selected from.-
Ci-Cs alkyl, C2-C6 alkenyl, C3-C11 cycloalkyl, (C3-C11 cycloalkyl)methyl, aryl, aryl(Ci-C4 alkyl)-, and Ci-C10 alkyl substituted with 0-2 R7e;
is selected from:
-C(=0)-R18be, -C(=0)N(R18be)2- -C(=0)NHS02R18ae, -C(=0)NHC(=0)R18be, -C(=0)NHC(=0)OR18ae, and -C(=0)NHS02NHR18be;
selected from.-


-COR20e, -SO3H, -PO3H, -CONHNHSO2CF3, -CONKSG2H18ae, -CONHS02NHR18be, -NHCOCF3, -NHCONHS02R18ae. -NHS02R18ae, -OPO3K2, -OSO3H, -PO3H2, -S02NHCOR18ae, -S02NHC02R1Bae,



10
15
with che following proviso: ne and me are chosen such
that the number of atoms connecting H*e and Ye is in the range of 8-14;
20 d is selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
d' is 1-100;
Ln is a linking group having the formula:
25
((W)h-(CR6R7)g)x-(Z)k-( (CR6aR7a)g'-(W)h-)X';
W is independently selected at each occurrence from the
group: 0, S, NH, NHCUO), C(=0)NH, NR8C(=0), C(=0)N 30 R6, C(=Oi, C{=0)0, OC{=0), NHC(=S)NH, NHC(=0)NH, S02, S02NH, (OCH2CH2)s- (CH2CH20)s., (OCH2CH2CH2)S" -{CH2CH2CH2c;t. and (aa)t-;
aa is independently at each occurrence an amino acid; 35
- 2.0 —

Z is selected from the group: aryl substituted with 0-3
R10, C3.10 cycloalkyl substituted with 0-3 R10, and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R10;
R6. R6a. R7, R7a, and R8 are independently selected at each occurrence from the group: H, -0, COOH, SO3H, PO3H, C1-C5 alkyl substituted with 0-3 R10, ary]
substituted with 0-3 R10, benzyl substituted with 0-3 R10, and C1-C5 alkoxy substituted with 0-3 Ri0,
NHC(=0)Rn, C(=0)NHRal, NHC (=0) NHR11. NHR11, R11. and a bond to Ch;
R10 is independently selected at each occurrence from the group: a bond to Ch# COOR11, C(=0)NHR:1, NHC{=0)R11, OH, NHR11, SO3H, PO3H, -OFO3H2, -OSO3H, aryl substituted with 0-3 R11, C1-5 alkyl substituted with 0-1 R12, C1-5 alkoxy substituted with 0-1 R12, and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O and substituted with 0-3 Rn;
R11 is independently selected at each occurrence from the group: H, alkyl substituted with 0-1 R12, aryl substituted with 0-1 R12, a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O and substituted with 0-1 R12, C3-10 cycloalkyl substituted with 0-1 R12, polyalkylene glycol substituted with 0-1 R12, carbohydrate substituted with 0-1 R12, cyclodextrin substituted with 0-1 R12, amino acid substituted with 0-1 R12, polycarboxyalkyl substituted with 0-1 R12, polyazaalkyl substituted with 0-1 R12, peptide substituted with 0-1 R12, wherein the peptide is comprised of 2-10 amino acids, 3,6-O-disulfo-B-D-galactopyranosyl, bis(phosphonomethyl)glycine, and a bond to Ch;


R12 is a oond to Ch;
k is selected from 0, 1, and 2; 5 h is selected from 0, 1, and 2;
h' is selected from 0, 1, and 2;
g is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
g' is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
s is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; 10 s" is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
s" is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
t is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
t' is selected from 0, 1. 2, 3, 4, 5, 6, 7, 8, 9, and 10;
x is selected from 0, 1, 2, 3, 4, and 5; 15 x' is selected from 0, 1, 2, 3, 4, and "5;
Ch is a metal bonding unit having a formula selected from the group:

A1. A2, A3, A4, A5, A6, A7, and A8 are independently 25 selected at each occurrence from the group: NR13 NRi3R14, S, SH, S(Pg), 0, OH, PR13, PR13R14, P(0)R15R16, and a bond to Ln;

E is a bond, CH, or a spacer group independently selecte; at each occurrence from the group: C1-C10 alkyl substituted with 0-3 R17, aryl substituted with 0-3 R17, C3-io cycloalkyl substituted with 0-3 R17, 5 heterocyclo-Ci-io alkyl substituted with 0-3 R17, wherein the heterocyclc group is a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0, C6-10 aryl-Ci-io alkyl substituted with 0-3 R17, Ci-io 10 alkyl-C6-io aryl- substituted with 0-3 R17, and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R17;
15 R13 and R14 are each independently selected from the group: a bond to Ln, hydrogen, Ci-Cio alkyl
substituted with 0-3 R17, aryl substituted with 0-3 R17, Ci-io cycloalkyl substituted with 0-3 R17. heterocyclo-Ci-io alkyl substituted with 0-3 R17,
20 wherein the heterocyclo group is a 5-10 membered
heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O, C6-10 aryl-Ci-io alkyl substituted with 0-3 R17, Ci-io alkyl-Ce-io aryl- substituted with 0-3 R17, a 5-10
25 membered heterocyclic ring system containing 1-4
heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R17, and an electron, provided that when one of R13 or R14 is an electron, then the other is also an electron;
30
alternatively, R13 and R14 combine to form =C(R20)(R21;
R15 and R16 are each independently selected from the
group: a bond to Ln, -OH, Ci-Cio alkyl substituted 35 with 0-3 R17, Ci~Cio alkyl substituted with 0-3 R17, aryl substituted with 0-3 R17, C3-10 cycloalkyl substituted with 0-3 R17, heterocyclo-Ci-10 alkyl substituted with 0-3 R17, wherein the heterocyclo


group is a b-:0 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and C. C^-io aryi-Ci-ic alkyl substituted with C 3 R1 ' , C- .io alkyl-C£-io aryl - substituted wits. 0-3 P.'"", and a 5-10 membered heterocyclic ring systex. containing 1-4 heteroatoms independently selected from h\ S, and 0 and substituted with 0-3 Ri7;
F-^ is independently selected at each occurrence from the group: a bond to Ln, =0, F, Ci, Br. I, -CFj, -CN, -C02R18, -C(=0)R18, -C(=0)N(R18)2, -CHO, -CH2OR18. -OC(=0)R18, -0C(=O)OR18a, -OR18, -OC{=0)N(R18)2. -NR1SC(=0)R18, -N"R19C(=0)OR18a, -NR19C(=0)N{R18;2. -NR1SS02N(R18!2- -NR19S02R18a. -SO3H, -S02R18a, -SR18, -S(-0)R18a, -S02N(R18)2, -N(R18)2, -NHC(=S!NHR18, =NOR18, N02, -C(=0)NHOR18, -C(=0)NHNR18Rl8a, -OCH2CO2H, 2-{1-morpholino)ethoxy, C1-C5 alkyl, C2-C4 alkenyl, C3-C6 cycjoalkyl, C3-C6 cycloalkylmethyl, C2-C6 alkoxyalkyl, aryl substituted with 0-2 R18, and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0;
Ri8r Rl8a# ancj pl9 are independently selected at each
occurrence from the group: a bond to Ln, K, Ci-Cg alkyl, phenyl, benzyl, C1-C6 alkoxy, halide, nitro, cyanc, and trifluoromethyl;
?g is a thiol protecting group;
F20 and R2: are independently selected fron! the group: H, Cl-Cio alkyl, -CN, -CO2R25, -C(=0)R25, -C (=0)N(R25 ) 2 , C2-C1C 1-alkene substituted with 0-3 R23, C2-C;o 1-alkyne substituted with 0-3 R23, aryl substituted with 0-3 R21, unsaturated 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O and substituted with 0-3


R2i, and unsaturated C;. 10 rarbocycle subsr : t ur -with 0-3 R23;
alternat i veiy, R20 and P.21, taken together with tr.e
5 divalent carbon radical to which they are attached
form:

10 P.22 and R23 are independent: ly selected from the group: H, R24, C1-C10 alkyl substituted with 0-3 R24, G2~C:o alkenyl substituted with 0-3 R24, C2-C1Q alkynyl substituted with 0-3 R24, aryl substituted with 0-3 R24, a 5-10 membered heterocyclic ring system
15 containing 1-4 heteroatoms independently selected
from N, S, and 0 and substituted with 0-3 R24, and C3-10 carbocycle substituted with 0-3 R24;
alternatively, R22, R23 taken together form a fused
20 aromatic or a 5-10 membered heterocyclic ring system
containing 1-4 heteroatoms independently selected from N, S, and O;
a and b indicate the positions of optional double bonds
25 and n is 0 or 1 ;
R24 is independently selected at each occurrence fron. the
group: =0, F, CI, Br, I, -CF3, -CN, -CO2R25.
-C(=0)R25, -C(=0)N(R25!2. -N(R25)3\ -CH2OR2".
30 -OC;=0)R2£j. -OC!-0)OR25a, -OR2S, -OC(=0)N(R2b)2,
-NR26C (=0>R2-. -NR26C i^O)OR25a, -NR26C (=0) N (R25 i 2 . -NE26S02N(R25>2' -NR26S02R25a- -SO3H, -S02R25a, -S?2S, -S(=0!R2Sa, -SC2N(R25)2, -N(R25)2, =NOR25,



including stereoisomeric forms thereof, or mixtures of stereoisomeric forms thereof, or pharmaceutically acceptable salt or prodrug forms thereof wherein:
10
Rle is selected from:


15
20

25

R2e and R3e are independently selected from:
H, C1-C4 alkoxy, mlleR12e, halogen, N02, CN, CF3 C1-C6 alkyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl (C1-C4 alkyl), aryl(Ci-C6 alkyl)-

(Ci-Ce alkyl)carbonyl, (C\-Cs alkoxy)carbonyl, arylcarbonyl, and aryl substituted with 0-4 R7e,
alternatively, when R2e and R3e are substituents on adjacent atoms, they can be taken together with the carbon atoms to which they are attached to form a 5 7 membered carbocyclic or 5-7 membered heterocyclic aromatic or nonaromatic ring system, said carbocyclic or heterocyclic ring being substituted with 0-2 groups selected from C1-C4 alkyl, C1-C4 alkoxy, halo, cyano, amino, CF3 and N02;
R2ae is selected from:
H, C1-C10 alkyl, C2-C6 alkenyl, C3-C11 cycloalkyl,
C3-C7 cycloalkyl(C1-C4 alkyl). aryl, aryl(C1-C4
alkyl)-, {C2-C7 alkyl)carbonyl, arylcarbonyl,
(C2-C10 alkoxy)carbonyl, C3-C7 cycloalkoxycarbonyl,
C7-C11 bicycloalkoxycarbonyl, aryloxycarbonyl,
aryl(C1-C10 alkoxy)carbonyl,
Cj-Ce alkylcarbonyloxy(Ci-C4 alkoxy)"cafbpnyl,
arylcarbonyloxy(Ci-C4 alkoxy)carbonyl, and
C3-C7 cycloalkylcarbonyloxy(Ci-C4 alkoxy) carbonyl ;
R7e is selected from:
H, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, aryl, aryl(Ci-C4 alkyl)-, (C1-C4 alkyl)carbonyl, C02R18ae, S02Rlle, SO2NR10eRlle, OR10e, and N(Rlle)R12e;
Ue is selected from:
-(CH2)ne-, -(CH2)neO(CH2)me-, -NH(CH2)ne-,
-N(Rl°e)C(=0)-, -NHC(=0) (CH2)ne-, and -C (=0) N(R10e) - ;
Ge is N or CR19e;
R8e is selected from:
H, C02R18be, C(=0)R18be, CONR17eR18be, C1-C10 alkyl substituted with 0-1 R6e, C2-C10 alkenyl substituted with 0-1 R6e,


C2-C1o alkynyl substituted with 0-1 R6e,
C3-C8 cycloalkyl substituted with 0-1 R6e,
C5-C6 cycloalkenyl substituted with 0-1 R6e,
(C1-C10 alkyl)carbonyl,
C3-C10 cycloalkyl(C1-C4 alkyl)-,
phenyl substituted with 0-3 R6e,
naphthyl substituted with 0-3 R6e,
a 5-10 membered heterocyclic ring containing 1-3 N, 0, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e;
is selected from: C1-C10 alkyl substituted with 0-1 R6e, C1-C10 alkoxy substituted with 0-2 R7e, H, nitro, N(Rlle)R12e, 0C(=O)Ri0e/ OR10e,
OC(=O)NR10eRlle, NR10eC(=dVRf*) R^*, -N(R16*)R17e, aryl (C0-C6&M$$tobonyl, aryl (Ci--JL W^ alkyl), heteroaryl(C?£?^tafc£l), CONR18aeR20e,
S02R18ae, and S02NR18aeR20eV providing that any of the above" alkyl, cycloalkyl, aryl or heteroaryl groups may be unsubstituted or substituted independently with 1-2 R7e;
s selected from:
H, C1-C10 alkyl, hydroxy, C1-C10 alkoxy, nitro, Ci-C10 alkylcarbonyl, -N(Rlle)Rl2e/ Cyano, halo, CF3, CHO, C02R18be, C(=0)R18be, C0NR17eR18be, OC(=O)R10e, OR10e, OC(=O)NR10eRHe,
NR10eC(=O)R10e, NR10eC(=O)OR21e, NR10eC(=O)NR10eRlle, NR10eSO2NR10eRlle/
NR10eSO2R21e, S(0)peRlle, SO2NR10eRlle, aryl substituted with 0-3 groups selected from halogen, Ci-C6 alkoxy, Ci-C6 alkyl, CF3, S(0)meMe, and -NMe2,


aryl(Ci-C4 alkyl}-, said aryl being substituted with 0-3 groups selected from halogen, C1-C6 alkoxy, C1-C6 alkyl, CF3, S(0)peMe, and -NMe2, and a 5-10 membered heterocyclic ring containing 1-3 N, C, or 5 S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e;
R10e is selected from:
W H, CF3, C3-C6 alkenyl. C3-C11 cycloalkyl, aryl,
(C3-C11 cycloalkyl)methyl, aryl (C1-C4 alkyl), and Ci-Cio alkyl substituted with 0-2 R6e;
Rlle is selected from: 15 H, hydroxy, Ci-Cg alkyl, C3-C6 alkenyl, C3-C11
cycloalkyl, (C3-C11 cycloalkyl)methyl, Ci-Cg alkoxy, benzyloxy, aryl, heteroaryl, heteroaryl(C1-C4 alkyl)-, aryl (C1-C4 alkyl), adamantylmethyl, and C1-C10 alkyl substituted with 0-2 R4e;
20
R4e is selected from:
H, CiJG^alkyl, C3-C7 cycloalkyl, C3-C7' : ■' ^ -cycloalkyl (C1-C4 alkyl}-, aryl, heteroaryl, aryKCj-C6 alkyl)-, and heteroaryl(Cj-Cg alkyl)-, wherein
25 said aryl or heteroaryl groups are substituted with 0-2 substituents independently selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, F, CI, Br, CF3, and NO2,
30 R12e is selected from:
H, C1-C6 alkyl, triphenylmethyl, methoxymethyl,
methoxyphenyldiphenylmethy1,
trimethylsilylethoxymethyl, (Ci-Ce alkyl)carbonyl, (C1-C6 alkoxy)carbonyl, (Ci-Cg alkyl)aminocarbony1, 35 C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(Ci-C4 alkyl)-, aryl, heteroaryl(C1-C6 alkyl)carbonyl, heteroarylcarbonyl, aryHCi-Cg alkyl)-, {C1-C6 alkyl)carbonyl, arylcarbonyl, C1-C6


alkylsulfonyl, arylsulfonyl, aryl(Ci-C6 alkyl)sulfonyl, heteroarylsulfonyl, heteroaryl(C1-C6 alkyl)sulfonyl, aryloxycarbonyl, and aryl(Ci~Cs alkoxy)carbonyl, wherein said aryl groups are substituted with 0-2 substituents selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, halo, CF3, and r.itro,-
R16e is selected from:
-C(=0)OR18ae, -C(=0)R18be, -C(=0)N(R18be)2, -S02R18ae. and -S02N(R18be)2;
R17e is selected from:
H, Ci-Ce alkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(Ci-C4 alkyl)-, aryl, aryl(Ci-Ce alkyl)-, and heteroaryl (Ci-Ce alkyl);
R18ae is selected from:
C3.-C8 alkyl optionally substituted with a bond to Ln, C3-C11 cycloalkyl .'optionally substituted with a bond to Ln, aryl(Ci-x|6 alkyl)- optionally substituted with a:«^M-to I^#vheteroaryl(Ci-C6 alkyl)- optionally substituted with a bond to Ln, (C1-C6 alkyl)heteroaryl optionally substituted with a bond to Ln, biaryl(Ci-C6
alkyl) optionally substituted with a bond to Ln, heteroaryl optionally substituted with a bond to L^, phenyl substituted with 3-4 R19e and optionally substituted with a bond to Ln,
naphthyl substituted with 0-4 R19e and optionally substituted with a bond to Ln, and a bond to Ln, wherein said aryl or heteroaryl groups are optionally substituted with 0-4 R19e;
R18be is H or Rl8ae.
R19(S is selected from:

H, halogen, CF3, CO:H, CN, NO2, -NRileP.12e, OCF;. Ci-Cs alkyl, C2-Cs alkenyl, C2-C6 alkynyl, C3-Cii cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl;-, aryli'Cj-Co alkyl;-, C1-C6 alkoxy, C1-C4 alkoxycarbonyl, aryl, aryl-O, aryl-S02-, heteroaryl, and heteroaryl-S02-. wherein said aryl
and heteroaryl croups are substituted with 0-4 groups selected from hydrogen, halogen, CF3, C:-C3 alkyl, and C1-C2 alkoxy;
0e is selected from:
hydroxy, C1-C10 alkyloxy, C3-C11 cycloalkyloxy,
aryloxy, aryl (C1-C4 alkyDoxy,
C2-C10 alkylcarbonyioxy(C1-C2 alkyDoxy-,
C2-C10 alkoxycarbonyloxy(C1-C2 alkyDoxy-,
C2-C1C alkoxycarbonyl (C1-C2 alkyDoxy-,
C3-C10 cycloalkylcarbonyloxy(C1-C2 alkyDoxy-,
C3-C10 cycloalkoxycarbonyloxy(Ci-C2 alkyDoxy-,
C3-C10 cycloalkoxycarbonyl(C1-C2 alkyl^oxy-,
aryloxycarbonyl (C1-C2 alkyDoxy-,
aryloxycarbonyloxy(C1-C2 alkyDoxy-,
arylcaifbonyloxy(C1-C2 alkyDoxy-,
C1-C5. alkoxy (C1-C5 alkyl) carbonyloxy (C1-C2 alkyDoxy,
(5- (C1-C5 alkyl)-1,3-dioxa-cyclopencen-2-one-
ylImethyloxy, (5-aryl-l,3-dioxa-cyclopencen-2-one-yl)mechyloxy,
and (Rioej (Rlle)N. (Ci-Cio alkoxy)-;
G is selected from:
Ci-Ce alkyl, C2-C6 alkenyl, C3-C11 cycloalkyl, (Cj-Cn cycloalkyl)methyl, aryl, aryl(Ci-C4 alkyl)-, and C1-C10 alkyl substituted with 0-2 R7e;
e is selected from:
-C(=0)-R18be. ■C(=0)N(R18he)2, -C(=0)NHS02R18fle,
-C(=0)NHC(=0)R18be. -C(=0)NHC(=0)OR18ae, and -C

me is 0-2;
ne is 0-4; and 5
Pe IS 0-2;
with the following proviso: ne and me are chosen such
that the number of atoms connecting R1 and -COR20e in 10 Formula (IV! is in the range of 8-14;
d is selected from 1, 2, 3, 4, and 5;
d' is 1-50; 15
W is independently selected at each occurrence from the
group: 0, NH. NHC(=0). C(=0}NH/ NR8C(=0), C(=0)NR8, C(=0), C(=0)0, OC(=0), NHC(=S)NH, NHC{=0)NH, S02, (OCH2CH2)s, (CH2CH20)s-, (OCH2CH2CH2)sJ* (CH2CH2CH20)t, 20 and (aa)t-;
aa is independently at each occurrence an amino acid;
Z is selected from the group: aryl substituted with 0-1 25 R10, C3-10 cycioalkyl substituted with 0-1 R10, and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O and substituted with 0-1 R10;
30 R6, R6a, R7, R7a, and R8 are independently selected at
each occurrence from the group: H, =0, COCK, SO3H, C1-C5 alkyl substituted with 0-1 R10, aryl
substituted with 0-1 R10, benzyl substituted with 0-1 R10, and C1-C5 aikoxy substituted with 0-1 R10,
35 NHC^OJR11. C(=0)NHRn, NHC (=0) NHRn, NHR11, R11, and a bond to Ch;
k is 0 or 1;

s is selected from 0, 1, 2, 3, 4, and 5;
s" is selected from 0, 1, 2, 3, 4, and 5;
s" is selected from 0, 1, 2, 3, 4, and 5;
t is selected from 0, 1, 2, 3, 4, and 5; 5
A1, A2, A3, A4, A5, A6, A7, and A8 are independently
selected at each occurrence from the group: NR13, NR13R14, S, SH, S(Pg), OH, and a bond to Ln;
10 E is a bond, CH, or a spacer group independently selected at each occurrence from the group: Ci-Cio alkyl
substituted with 0-3 R17, aryl substituted with 0-3 R17, C3-10 cycloalkyl substituted with 0-3 R17, and a
5-10 membered heterocyclic ring system containing 15 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R17;
R13, and R14 are each independently selected from the group: a bond to Ln, hydrogen, C1-C10 alkyl
20 substituted with 0-3 R17, aryl substituted with 0-3 Rl7, a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R17, and an electron, provided that when one of R13 or R14 iS
25 an electron, then the other is also an electron;
alternatively, R13 and R14 combine to form =C(R20) (R21);
R^7 is independently selected at each occurrence from the 30 group: a bond to hn. =0, F, CI, Br, I, -CF3, -CN, -CO2R18, -C(=0)Rl8, _C(=0)N(R18)2/ _CH2OR18,
-OC(=0)R18, -0C(=O)OR18a, -ORIS, -OC(=0)N(R18)2,
-NR^C^OJR18, -NR19c(=0)OR18a, -NR19C (=0) N(R18) 2, -NR19s02N(Rl8)2/ _NR19S02Rl8ar -SO3H, -S02R18a, 35 -S(=0)R18a, -S02N(Rl8)2/ _N(R18)2# -NHC(=S)NHR18, =NOR18, -C(=0)NHNR18Rl8a; -OCH2CO2H, and 2-(1-morpholino)ethoxy;

R18, R18a, and R19 are independently selected at each occurrence from the group: a bond to Ln, H, and C1-C6 alkyl;
5 R20 and R21 are independently selected from the group.- H, C1-C5 alkyl, -CO2R25, C2-C5 1-alkene substituted with 0-3 R23, C2-C5 1-alkyne substituted with 0-3 R23, aryl substituted with 0-3 R23, and unsaturated 5-10 membered heterocyclic ring system containing 1-4 10 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R23,-
alternatively, R20 and R21, taken together with the
divalent carbon radical to which they are attached 15 form:
R22 and R23 are independently selected from the group: H, 20 and R24;
alternatively, R22, R23 taken together form a fused
aromatic or a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected 25 from N, S, and 0;
R24 is independently selected at each occurrence from the group: -C02R25, -C(=0)N(R25)2, -CH2OR25, -OC(=0)R25, -OR25, -SO3H, -N(R25)2, and -OCH2CO2H; and,
30
R25 is independently selected at each occurrence from the group: H and C1-C3 alkyl.


[4] In an even more preferred embodiment, the present invention provides a compound including stereoisomeric forms thereof, or mixtures of stereoisomeric forms thereof, or pharmaceutical^ acceptable salt or prodrug 5 forms thereof wherein:
Rle is selected from:


10

R2e and R3e are independently selected from:
H, C1-C4 alkoxy, NRlleR12e, halogen, NO2, CN, CF3,
15 Ci-Ce alkyl, C3-C6 alkenyl, C3-C7 cycloalkyl,
C3-C7 cycloalkyl (C1-C4 alkyl), aryKCi-Ce alkyl)-, (C1-C6 alkyl)carbonyl, (Ci-Cg alkoxy)carbonyl, arylcarbonyl, and aryl substituted with 0-4 R7e, 20 alternatively, when R2e and R3e are substituents on adjacent atoms, they can be taken together with the carbon atoms to which they are attached to form a 5-7 membered carbocyclic or 5-7 membered heterocyclic aromatic or nonaromatic ring system,
25 said carbocyclic or heterocyclic ring being
substituted with 0-2 groups selected from C1-C4 alkyl, C1-C4 alkoxy, halo, cyano, amino, CF3 and NO2;
R2ae is selected from:
30 H, C1-C10 alkyl, C2-Ce alkenyl, C3-C11 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl), aryl, aryl(C1-C4 alkyl)-, (C2-C7 alkyl)carbonyl, arylcarbonyl, (C2-C10 alkoxy)carbonyl, C3-C7 cycloalkoxycarbonyl, C7-C11 bicycloalkoxycarbonyl, aryloxycarbonyl,
35 aryl(C1-C10 alkoxy)carbonyl,

Ci-Ce alkylcarbonyloxy{C1-C4 alkoxy)carbonyl,
arylcarbonyloxy(Ci-C4 alkoxy) carbonyl, and
C3-C7 cycloalkylcarbonyloxy (C1-C4 alkoxy) carbonyl;
R7e is selected from:
H, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, aryl, aryKCi-C4 alkyl)-, (C1-C4 alkyl) carbonyl, C02R18ae, S02Rlle, SO2NR10eRlle, 0R10e, and N(Rlle)Rl2e;
Ue is selected from:
-(CH2)ne-, -NH(CH2)ne-. -N(R10e)C(=0)-, and -NHC(=0) (CH2)ne;
G« is N or CR19e;
R8e is H;
R9e is selected from:
H, nitro, N(Rne)Ri2e, OC(=O)R10e, OR10e,
OC(=O)NRl0eRlle. NR10eC(=O)R10e, NR10eC (=0) 0R21e, NR10eC(=O)NR10eRlle/ NR10eSO2NR10eRlle/
NR10eSO2R21e, hydroxy, OR22e, -N(R10e)Ri:ie, -N(Rl6e)R17e, aryl(Co-C4 alkyl) carbonyl, aryl(Ca-C4 alkyl), heteroaryl (C1-C4 alkyl) , CONR18aeR20e, S02R18ae, and SO2NR18aeR20e, providing that any of the above alkyl, cycloalkyl, aryl or heteroaryl groups may be unsubstituted or substituted independently with 1-2 R7e;
RiOe is selected from:
H, CF3, C3-C6 alkenyl, C3-C6 cycloalkyl, aryl, (C3-C6 cycloalkyl)methyl, aryl(C1-C4 alkyl), and C1-C4 alkyl substituted with 0-2 R6e;
R6e is selected from:
H, C1-C4 alkyl, hydroxy, C1-C4 alkoxy, nitro, C1-C4 alkylcarbonyl, -N(Rlle)Ri2e, cyano, halo, CF3, CHO, C02R18be, C(=0)R18be, CONRl7eR18be,

OC(=O)R10e, OR10e, OC(=O)NR10eRlle, NR10eC(=O)RlOe/ NR10eC(=O)OR21e, NR10eC(=O)NR10eRlle, NR10eSO2NR10eRlle, NR10eSO2R21e, S(0)pRlle, SO2NR10eRlle,
5 aryl substituted with 0-3 groups selected from
halogen, C1-C4 alkoxy, C1-C4 alkyl, CF3,
S(0)meMe, and -NMe2,
aryl(C1-C4 alkyl)-, said aryl being substituted with
0-3 groups selected from halogen, C1-C4 alkoxy,
10 C1-C4 alkyl, CF3, S(0)peMe, and -NMe2, and
a 5-10 membered heterocyclic ring containing 1-3 N, 0, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being 15 substituted with 0-2 R7e;
Rlle is selected from:
H, hydroxy. C1-C4 alkyl, C3-C6 alkenyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)methyl, C1-C4 alkoxy, 20 benzyloxy, aryl, heteroaryl, heteroaryl(C1-C4 alkyl)-, aryl (C1-C4 alkyl), adamantylmethyl, and Ci-C4*alkyl substituted with 0-2 R4e;
R4e is selected from: 25 H, Ca-C4 alkyl, C3-C7 cycloalkyl, C3-C7
cycloalkyl(Ca-C4 alkyl)-, aryl, heteroaryl, aryl(C1-
C4 alkyl)-, and heteroaryl(C^-C^ alkyl)-, wherein
said aryl or heteroaryl groups are substituted with 0-2 substituents independently selected from the 30 group consisting of C1-C4 alkyl, C1-C4 alkoxy, F, CI, Br, CF3, and N02,
R12e is selected from:
H, C1-C4 alkyl, (C1-C4 alkyl) car bony 1, (C1-C4 35 alkoxyJcarbonyl, phenyl(C1-C4 alkyl)-,
phenylsulfonyl, phenyloxycarbonyl, and phenyl(C1-C4 alkoxy)carbonyl, wherein said phenyl groups are substituted with 0-2 substituents selected from the

group consisting of C1-C4 alkyl, C1-C4 alkoxy, halo, CF3, and nitro;
R16e is selected from:
5 -C(=0)OR18ae -C(=0)R18be, -C(=0)N(R18be)2, -S02R18ae,
and -S02N(R18be)2;
RI7e is selected from:
H, C1-C4 alkyl, C3-C6 cycloalkyl, C3-C6
10 cycloalkyl(C1-C4 alkyl)-, aryl, aryl(Ci-C6 alkyl)-,
and heteroaryl (Ci-Ce alkyl);
R18ae is selected from:
Ci~C$ alkyl optionally substituted with a bond to Ln,
15 C3-C11 cycloalkyl optionally substituted with a
bond to Ln, aryl(Ci~C6 alkyl)- optionally substituted with a bond to Ln, heteroaryl (C1-C6 alkyl)- optionally substituted with a bond to Ln/ (Ci-Ce alkyl)heteroaryl optionally
20 substituted with a bond to Ln, biaryl(Ci-Ce
alkyl) optionally substituted with a bond to Ln, heteroaryl optionally substituted with a bond to Ln, phenyl substituted with 3-4 R19e and optionally substituted with a bond to Ln,
25 naphthyl substituted with 0-4 R19e and
optionally substituted with a bond to Ln, and a bond to Ln, wherein said aryl or heteroaryl groups are optionally substituted with 0-4 R19e;
30 R18be is H or R18*^.
R19e is selected from:
H, halogen, CF3, C02H, CN, N02/ -NRlleR12e, OCF3, C1-C6 alkyl, C2-C6 alkenyl, C2-Ce alkynyl, 35 C3-C6 cycloalkyl, C3-Ce cycloalkyl(C1-C4 alkyl)-, aryl(Ci-C4 alkyl)-, C1-C6 alkoxy, C1-C4 alkoxycarbonyl, aryl, aryl-O-, aryl-S02-, heteroaryl, and heteroaryl-S02-, wherein said aryl


and heteroaryl groups are substituted with 0-4 groups selected from hydrogen, halogen, CF3, C1-C3 alkyl, and C1-C3 alkoxy;
R20e j_s selected from:
hydroxy, C1-C6 alkyloxy, C3-C6 cycloalkyloxy,
aryloxy, aryl (Ci-C4 alkyDoxy,
C2-C10 alkylcarbonyloxy(C1-C2 alkyDoxy-,
C2-C10 alkoxycarbonyloxy(Ci-C2 alkyDoxy-,
C2-C10 alkoxycarbonyl (C1-C2 alkyDoxy-,
C3-C10 cycloalkylcarbonyloxy(C1-C2 alkyDoxy-,
C3-C10 cycloalkoxycarbonyloxy(Ci-C2 alkyDoxy-,
C3-C10 cycloalkoxycarbonyl (C1-C2 alkyDoxy-,
aryloxycarbonyl (C1-C2 alkyDoxy-,
aryloxycarbonyloxy (C1-C2 alkyDoxy-,
arylcarbonyloxy (C1-C2 alkyDoxy-,
C1-C5 alkoxy (C1-C5 alkyl) carbonyloxy (Ca-C2 alkyDoxy,
(5-(C1-C5 alkyl)-1,3-dioxa-cyclopenten-2-one-
yDinethyloxy, (5-aryl-1,3-dioxa-cyclopenten-2-one-yl)methyloxy,
and {R10e)/R1Je)N-(C1-Cio alkoxy)-;
R21e is selected from:
C1-C4 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)methyl, aryl, aryl(C1-C4 alkyl)-, and C1-C10 alkyl substituted with 0-2 R7e;
p22e iS selected from:
-C(=0)-R18be, -C(=0)N(R18be)2, -C(=0)NHS02R18ae, -C(=O)NHC(=0)R18be, -C(=0)NHC(=0)ORl8ae, and -C(=0)NHS02NHR18be;
me is 0-2;
ne is 0-4;
pe is 0-2;




5
A1 is selected from the group: OH, and a bond to Ln;
A2, A4, and A6 are each N; 10 A3, A5, and A8 are each OH;
A7 is a bond to Ln or NH-bond to Ln;
E is a C2 alkyl substituted with 0-1 R17;
15
R17 is =0;

alternatively, Ch is A5
A1 is selected from the group: OH, and a bond to Ln; 25 A2, A3 and A4 are each N; A5, A6 and A8 are each OH;
A7 is a bond to Ln; 30


E is a C2 alkyl substituted with 0-1 R

R17 is =0;
5 alternatively, Cj, is A
10
A1 is NK2 or N=C(R20) (R21); E is a bond; A2 is NHR13;

15

R13 is a heterocycle substituted with R17, the heterocycle being selected from pyridine and pyrimidine;
R17 is selected from a bond to Ln, C(=0)NHR18 and C(=0)R18;



20

R18 is a bond to Ln;
R24 is selected from the group: -CO2R25, -OR25, -SO3H, and -N(R25)2; and,

R25 is independently selected at each occurrence from the 25 group: hydrogen and methyl.
[5] In another even more preferred embodiment, the present invention provides a compound including enantiomeric or diastereomeric forms thereof, or mixtures 30 of enantiomeric or diastereomeric forms thereof, or pharmaceutically acceptable salt or prodrug forms thereof, wherein Q is selected from the group:
3-[7-[(imidazolin-2-ylamino)methyl]-l-methyl-6, 8-
35 difluoroquinoline-4-one-3-ylcarbonylamino]-2-
(3,5-dimethylisoxazol-4-ylsulfonylamino)propionic acid,


3- [7-[ (imidazolin-2-ylamino)methyl] -l-methyl-6, 8-difluoroquinoline-4-one-3-ylcarbonylamino] -2-(benzyloxycarbonylamino)propionic acid, 3- [7- [ (imidazolin-2-ylamino)methyl] -l-methyl-6, 8-difluoroquinoline-4-one-3-ylcarbonylamino] -2-(n-butyloxycarbonylamino) propionic acid, 3- [7- [ (imidazolin-2-ylamino)methyl] -l-methyl-6 , 8-dif luoroquinoline-4~one-3-ylcarbonylamino] -2-(n-butylsulfonylamino)propionic acid, 3- [7- [ (tetrahydropyrimid- 2 -ylamino) methyl] - l-methyl-6, 8-dif luoroquinoline-4-one-3-ylcarbonylamino] -2- (benzyl oxycarbonyl ami no) propionic acid, 3- [1- [ (tetrahydropyrimid-2-ylamino)methyl] -1-methyl-6, 8-dif luoroquinoline-4-one-3-ylcarbonylamino] -2-(n-butyloxycarbonylamino)propionic acid, 3- [7- [ (tetrahydropyrimid-2-ylamino)methyl] -1-methyl-6, 8-dif luoroquinoline-4-one-3-ylcarbonylamino] -2-(phenylsulfonylamino)propionic acid, 3- [7- [ (tetrahydropyrimid-2-ylamino)methyl] -1-methyl-6, 8-dif luoroquinoline-4-one-3-ylcarbonylamino] -2- (n-butylsulfonyl)aminopropionic acid, 3-[7- [ fr2-aminothiazol-4-yl)methyl]-l-methyl-6, 8-
difluoroquinoline-4-one-3-ylcarbonylamino] -2-(benzyloxycarbonylamino)propionic acid, 3- [7- [ (imidazolin-2-ylamino)methyl] -l-methyl-6, 8-dif luoroquinoline-4-one-3-ylcarbonylamino] -2-((2,4,6-trimethylphenyl)sulfonylamino)propionic acid, 3- [7- [ (tetrahydropyrimid-2-ylamino) methyl] -1-methyl-6, 8-difluoroquinoline-4-one-3-ylcarbonylamino] -2-((2,4,6-
trimethylphenyl) sulfonylamino) propionic acid, 3- [7- [ (imidazol-2-ylamino)methyl] -l-methyl-6, 8-
dif luoroquinoline-4-one-3-ylcarbonylamino] -2-(3,5-dimethylisoxazol-4-ylsulfonylamino)propionic acid,

3- [7- [ (imidazol-2-ylamino)methyl] -l-methyl-6, 8-
difluoroquinoline-4-one-3-ylcarbonylamino]-2-(benzyloxycarbonylamino) propionic acid, 3- [7- [ (imidazol-2-ylamino)methyl] -l-methyl-6, 8-
dif luoroquinoline-4-one-3-ylcarbonylamino] -2-((2,4,6-trimethylphenyl)sulfonylamino)propionic acid, 3- [7- [ (imidazol-2-ylamino)methyl] -l-methyl-6, 8-
dif luoroquinoline-4-one-3-ylcarbonylamino] -2-( (4-biphenyl) sulf onylamino) propionic acid, 3- [7- [ (imidazol-2-ylamino)methyl] -l-methyl-6, 8-
difluoroquinoline-4-one-3-ylcarbonylamino]-2-(1-naphthylsulfonylamino)propionic acid, 3- (7- [ (benzimidazol-2-ylamino)methyl] -l-methyl-6, 8~ difluoroquinoline-4-one-3-ylcarbonylamino] -2-((2,4, 6-trimethylphenyl) sulf onylamino) propionic acid, 3- [7- [ (4-methylimidazol-2-ylamino)methyl] -1-methyl-6, 8-difluoroquinoline-4~one-3-ylcarbonylamino] -2-((2,4,6-
trimethylphenyl) sulf onylamino) propionic acid, 3- [7- [4r4, 5 -dimethyl imi dazol- 2 -ylamino) methyl] -1-methyl-6, 8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-
trimethylphenyl) sulf onylamino) propionic acid, 3- [7- [ (4,5,6,7-tetrahydrobenzimidazol-2-
ylamino)methyl] -l-methyl-6, 8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-trimethylphen-yl) sulf onylamino) propionic acid, 3- [7- [ (pyridin-2-ylamino)methyl] -l-methyl-6, 8-
difluoroquinoline-4-one-3-ylcarbonylamino] -2-( (2,4, 6-trimethylphenyl) sulf onylamino) propionic acid, 3- [7- (2-aminopyridin-6-yl)-l-methyl-6,8-
difluoroquinoline-4-one-3-ylcarbonylamino] -2-( (2,4, 6-trimethylphenyl) sulf onylamino) propionic acid,

3-[7-[(7-azaben2imidazol-2-yl)methyl]-l-methyl-6, 8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-trimethylphenyl)sulfonylamino)propionic acid, 5 3-[7-[(benzimidazol-2-ylamino)methyl]-l-(2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-
ylcarbonylamino]pro-pionic acid,
3-[7-[(pyridin-2-ylamino)methyl]-1-(2-phenylethyl)-
6,8-difluoroquinoline-4-one-3-
10 ylcarbonylamino]propionic acid,
3-[7-[(imidazolin-2-ylamino)methylJ-1-(2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-
ylcarbonylamino]propionic acid,
3-[7-[(imidazol-2-ylamino)methyl]-1-(2-phenylethyl)-
15 6,8-difluoroquinoline-4-one-3-
ylcarbony1amino]propionic acid, 3-[7-f(imidazoline2-ylamino)methyl]-1-(2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-
ylcarbonylamino]-2-
20 (benzyloxycarbonylamino)propionic acid,
3-[7-[(imidazolin-2-ylamino)methyl]-1-(2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-(n-butyloxycarbonylamino)propionic acid, 25 3-[7-{(imidazolin-2-ylamino)methyl]-l-(2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-
ylcarbonylamino]-2-
(phenylsulfonylamino)propionic acid,
3-[7-[(imidazolin-2-ylamino)methyl]-1-(2-
30 phenylethyl)-6,8-difluoroquinoline-4-one-3-
ylcarbonylamino]-2-(n-
butylsulfonylamino)propionic acid,
3-[7-[(tetrahydropyrimid-2-ylamino)methyl]-1-(2-
phenylethyl)-6,8-difluoroguinoline-4-one-3-
35 ylcarbonylamino]-2-
(benzyloxycarbonylamino)propionic acid, 3-[7-[(tetrahydropyrimid-2-ylamino)methyl]-1- (2-phenylethyl)-6,8-difluoroquinoline-4-one-3-


ylcarbonylamino]-2-(n-
butyloxycarbonylamino)propionic acid,
3-[7-[(tetrahydropyrimid-2-ylamino)methyl]-1-(2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-
5 ylcarbonylamino]-2-
(phenylsulfonylamino)propionic acid,
3-[7-[(tetrahydropyrimid-2-ylamino)methyl]-1-(2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-
ylcarbonylamino]-2-(n-
10 butylsulfonyDaminopropionic acid,
3-[7-[(2-ammothiazol-4-yl)methyl]-l-(2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-(phenylsulfonylamino)propionic acid, 15 3-[7-[(2-aminothiazol-4-yl)methyl]-1-(2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-
(benzyloxycarbonylamino)propionic acid,
3-[7-[(imidazolin-2-ylamino)methyl]-1-(2-
20 phenylethyl)-6,8-difluoroquinoline-4-one-3-
ylcarbonylamino]-2-((2,4,6-
trimethylpheny1)sulfonylamino)propionic acid,
3-[7-[(tetrahydropyrimid-2-ylamino)methyl]-1-(2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-
25 ylcarbonylamino]-2-((2,4, 6-
trimethylphenyl)sulfon-ylamino)propionic acid,
3-[7-[(imidazol-2-ylamino)methyl]-l-(2-phenylethyl)
6,8-difluoroquinoline-4-one-3-ylcarbonylamino]
2~(benzyloxycarbonylamino)propionic acid,
30 3-[7-[(imidazol-2-ylamino)methyl]-l-(2-phenylethyl)-
6,8-difluoroquinoline-4-one-3-ylcarbonylamino]
2-(phenylsulfonylamino)propionic acid,
3-[7-[(imidazol-2-ylamino)methyl]-1-(2-phenylethyl)
6, 8-difluoroquinoline-4-one-3-ylcarbonylamino]
35 2- ((2, 6, dichlorophenyDsulfonylamino)propionic
acid, 3-[7-[(imidazol-2-ylamino)methyl]-1-(2-phenylethyl) -6, 8-difluoroquinoline-4-one-3-ylcarbonylamino] -

2-((2,4,6-
trimethylphenyl) sulfonylamino)propionic acid,
3- [7- [ (imidazol-2-ylamino)methyl] -1- (2-phenylethyl) -
6, 8-dif luoroquinoline-4-one-3-ylcarbonylamino] -
5 2-((4-biphenyl)sulfonylamino)propionic acid,
3-[1-[(benzimidazol-2-ylamino)methyl]-1- (2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-{(2,4,6-
trimethylphenyl) sulf onylamino) propionic acid, 10 3- [7- [ (4-methylimidazol-2-ylamino)methyl] -1- (2-phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-{(2,4,6-
trimethylphenyl)sulfonylamino)propionic acid,
3- [7- [ (4, 5-dimethylimidazol-2-ylamino)methyl] -1- (2-
15 phenylethyl) -6, 8-dif luoroquinoline-4—one-3-
ylcarbonylamino]-2-((2,4,6-
trimethylphenyl)sulfonylamino)propionic acid, 3-[7-[(4,5,6,7-tetrahydrobenzimidazol-2-
ylamino)methyl]-1-(2-phenylethyl)-6,8-
20 difluoroquinoline-4-one-3-ylcarbonylamino] -2-
((2,4, 6-trimethylphenyl) sulf onylamino) propionic
acfid,
3-[7- [ (pyridin-2-ylamino)methyl]-l- (2-phenylethyl) -
6,8-difluoroquinoline-4-one-3-ylcarbonylamino] -
25 2-((2,4,6-
trimethylphenyl)sulfonylamino)propionic acid,
3-[7-(2-aminopyridin-6-yl)-l-(2-phenylethyl)-6,8-
dif luoroquinoline-4-one-3-ylcarbonylamino] -2-
((2,4, 6-trimethylphenyl) sulf onylamino) propionic
30 acid, and
3-[7- [(7-azabenzimidazol-2-yl)methyl] -1- (2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-
ylcarbonylamino]-2-((2,4, 6-
trimethylphenyl)sulfonylamino)propionic acid.
35
[6] In another'even more preferred embodiment, the present invention provides a compound selected from the group:

2-(((4-(4~(((3-(2-(2-(3- sulfophenyl) vinyl) amino) (3-pyridyl) )carbonylamino)-propoxy)ethoxy)-5 ethoxy) propyl) amino) sul f ony 1) phenyl) phenyl) -
sulf onyl) amino) -3- ( (7- ( (imidazol-2-ylamino)methyl) -
l-methyl-4-oxo(3-
hydroquinolyl))carbonylamino)propanoic acid;
10 3- ( (7- { (imidazol-2-ylamino)methyl) -l-methyl-4-oxo(3-
hydroquinolyl) ) carbonylamino) -2-(((4-(4-(((3-(2-(2-(3-(2-(1.4,7,10-tetraaza-4,7,10-tris (carboxylmethyDcyclododecyl)acetylamino) -propoxy) ethoxy) ethoxy) propyl) amino) sulf onyl) -
15 phenyl)phenyl)sulfonyl)amino)propanoic acid;
2-{((4-(3-(N-(3-(2-(2-{3-({6-((l-aza-2-(2-
sulfophenyl) vinyl) amino) (3-pyridyl) ) carbonylamino) -propoxy) ethoxy) ethoxy) propyl) carbamoyl)propoxy) -2,6-20 dimethylphenyl) sul fonyl) amino) -3- ((7- ((imidazol-2-ylamino)methyl) -l-methyl-4-oxo(3-hydroquinolyl) ) -carbonylamino)propanoic;
3-( (l-(3-( (6-( (l-aza-2-(2-sulfophenyl)vinyl)amino) (3-25 pyridyl) ) carbonylamino)propyl)-7-((imidazole-2-ylamino)methyl)-4-oxc(3-
hydroquinolyl))carbonylamino)-2-(((2,4,6-trimethylphenyl) sulf onyl) amino) propanoic acid;
30 3-((l-(3-( (6-( (l-aza-2-(2-sulfophenyl)vinyl)amino) (3-
pyridyl))carbonylamino)propyl)-7-(((1-
hydroxyimidazole-2-yl)amino)methyl)-4-oxo(3-
hydroquinolyl))carbonylamino)-2-(((2,4,6-
trimethylphenyl) sulf onyl) amino) propanoic acid; 35
3-((l-(3-(3-(N-(3-(2-(2-(3-((6-((l-aza-2-(2-
sulfophenyl)vinyl)amino)(3-
pyridyl) ) carbonylamino)propoxy) ethoxy) -
ethoxy) propyl) carbamoyl) propanoylamino) propyl) -7-
40 ( (imidazole-2-ylamino)methyl) -4-oxo(3-



10
15
20

hydroquinolyl))carbonylamino)-2-(((2,4,6-trimethylphenyl)sulfonyl)amino)propanoic acid;
2-(2-aza-2-(5-(N-(l,3-bis(3-(2-(2-(3-(3-



10


2-(((4-(3-(N-(3-(2-(2-(3-(2-(l,4,7,10-tetraaza-4,7,10-5 tris(carboxymethyl)cyclododecylacetylamino)-6-aminohexanoylamino)propoxy)ethoxy)ethoxy)-propyl)carbamoyl)propoxy)-2,6-
dimethylphenyDsulfonyl) amino) -3- ((7- ((imidazol-2-ylamino)methyl)-l-methyl-4-oxo(3-hydroquinolyl))-10 carbonylamino)propanoic acid ;
2-(((4-(3-(N-(3-(2-(2-(3-(2-(l,4,7,10-tetraaza-4,7,10-tris(carboxymethyl)cyclododecylacetylamino)-6- (2-(bis (phosphonomethyl) amino) acetyl amino) hexanoyl ami no 15 ) propoxy) ethoxy) ethoxy) propyl) carbamoyl) propoxy) -
2, 6-diHiethylphenyl)sulfonyl)amino)-3- ((7- ( (imidazol-2-ylamino)methyl)-l-methyl-4-oxo(3-hydroquinolyl))-carbonylamino)propanoic acid conjugate; and
20 2-(((4-(3-(N-(3-(2-(2- (3- (2-(2-((2-((2-
(bis(carboxymethyl)-
amino)ethyl) (carboxymethyl)amino) ethyl) (carboxymethy
1) amino) acetylamino) -3-sulfopropyl)propoxy) ethoxy) -
ethoxy)propyl)carbamoyl)propoxy) -2,6-25 dimethylphenyl)sulfonyl)amino) -3-((7-((imidazol-2-
ylamino)methyl)-l-methyl-4-oxo(3-hydroquinolyl))-
carbonylamino)propanoic acid;








10

or a pharmaceutical^ acceptable salt form thereof.
[7] In a further preferred embodiment, the present invention provides a kit comprising a compound of Claim 2, or a pharmaceutical^ acceptable salt form thereof and a pharmaceutical^ acceptable carrier.
[8] In an even further preferred embodiment, the kit further comprises one or more ancillary iigands and a reducing agent.

15 [9] In a still further preferred embodiment, the ancillary Iigands are tricine and TPPTS.
[10] In another still further preferred embodiment, the reducing agent is tin(II) .
20
[11] In a second embodiment, the present invention provides a novel diagnostic or therapeutic metallopharmaceutical composition, comprising: a metal, a chelator capable of chelating the metal and a targeting 25 moiety, wherein the targeting moiety is bound to the chelator, is a quinolone non-peptide and binds to a
-5if _

receptor that is upregulated during angiogenesis and the compound has 0-1 linking groups between the targeting moiety and chelator.
5 [12] In a preferred embodiment, wherein the metallopharmaceutical is a diagnostic
radiopharmaceutical, the metal is a radioisotope selected from the group: 99mTc, 95Tc, mIn, 62Cu, 64Cu, 67Ga, and 68Ga, and the linking group is present between the non-10 peptide targeting moiety and chelator.
[13] In another preferred embodiment, the targeting moiety is a quinolone non-peptide and the receptor is avp3 or avPs-15
[14] In another preferred embodiment, the radioisotope is 99mrpc or 95TC/ ^Q radiopharmaceutical further comprises a first ancillary ligand and a second ancillary ligand capable of stabilizing the radiopharmaceutical. 20
[15] In another preferred embodiment, the radioisotope is 99mTc. r
[16] In another preferred embodiment, the 25 radiopharmaceutical is selected from the group:
99mTc(2-(((4-(4-(((3-{2-(2-(3-((6-(diazenido)(3-pyridyl))carbonylamino)propoxy)ethoxy)-ethoxy > propyl)amino)sulfonyl)phenyl)phenyl)-30 sulfonyl)amino)-3-((7-((imidazol-2-ylamino)methyl)-1-methyl-4-oxo(3-
hydroquinolyl))carbonylamino)propanoic acid)(tricine)(TPPTS);
35 99mTc(2-{{(4-(3-(N-(3- (2- (2- (3- {(6-(diazenido) (3-pyridyl) ) carbonylamino)propoxy) ethoxy) -ethoxy)propyl)carbamoyl)propoxy)-2,6-dimethylphenyl)sulfonyl)amino)-3-((7-((imidazol-2-

ylamino)methyl)-l-methyl-4-oxo(3-hydroquinolyl))-carbonylamino)propanoic acid)(tricine)(TPPDS);
s^TcO- ( (l-(3-( (6-(diazenido) (3-5 pyridyl))carbonylamino)propyl)-7-((imidazole-2-ylamino)methyl)-4-oxo(3-
hydroquinolyl))carbonylamino)-2-(((2,4,6-trimethylphenyl)sulfonyl)amino)propanoic acid)(tricine)(TPPTS); 10
9^Tc{3- ( (1- (3- ( (6- (diazenido) (3-
pyridyl))carbonylamino)propyl)-7- (( (1-hydroxyimidazole-2-yl)amino)metttyl)-4-oxo(3-hydroquinolyl))carbonylamino)-2-(((2,4,6-15 trimethylphenyl)sulfonyl)amino)propanoic acid)(tricine)(TPPTS);
9SnTc(3-( (l-(3-(3-(N-(3-(2-(2-(3-((6- (diazenido) (3-pyridyl))carbonylamino)propoxy)ethoxy)-
20 ethoxy)propyl)carbamoyl)propanoylamino)propyl) - 7 -((imidazole-2-ylamino)methyl)-4-oxo(3-hydrocjuinolyl) ) carbonylamino) -2- (((2,4,6-trimethylphenyl)sulfonyl)amino)propanoic acid)(tricine)(TPPTS);
25
99mTc(2-(2-(5-(N-(l,3-bis(3-(2-(2-(3-(3-(N-(3-(3-(N-(3-carboxy-2-(((2,4,6-trimethylphenyl)sulfonyl)amino)-ethyl)carbamoyl)-7-((imidazole-2-ylamino)methyl)4-oxohydroquinolyl)propyl)carbamoyl)propanoylamino)
30 propoxy)ethoxy)ethoxy)propyl)carbamoyl)(2-pyridyl)diazenido)\ (.tricine\ CTPPTS);
99mrC(3-{tl_(3-{2-[(6-(diazenido)(3-35 pyridyl))carbonylamino](2R)- 3-sulfopropyl}propyl)-7-[(imidazol-2-ylamino)methyl]-4-oxo(3-hydroquinolyl)]carbonylamino}(2S)-2-U(2,4,6-

trimethylphenyl) sulfonyl] amino} propanoic acid) (tricine)(TPPTS).
[17] In another preferred embodiment, the radioisotope is 5 i"ln.
[18] In another preferred embodiment, the radiopharmaceutical is selected from the group:





[19] In another preferred embodiment wherein the metallopharmaceutical is a therapeutic
radiopharmaceutical, the metal is a radioisotope selected 10 from the group: 186Re, 188Re, 153Sm, 166Ho, 177Lu, 149Pm,
90Y, 212Bi/ 103pd# 109pd/ 159Gd/ 140La, 198Au# 199Au, 169Yb,
i75Yb/ i65py# i66Dy/ 67CU/ i05Rh, iiiAg, and 192Ir, and the


linking group is present between the non-peptide targeting moiety and chelator.
[20] In another preferred embodiment, the targeting moiety 5 is a quinolone non-peptide and the receptor is av^3 or
[21] In another preferred embodiment, the radioisotope is
10
[22] In another preferred embodiment, the radioisotope is "7Lu.
[23] In another preferred embodiment, the 15 radiopharmaceutical is selected from the group:





10

[24] In another preferred embodiment, the radioisotope is
90y.
[25] In another preferred embodiment, the radiopharmaceutical is selected from the group;




-*s _




10

[26] In another preferred embodiment wherein the metallopharmaceutical is a MRI contrast agent, the metal is a paramagnetic metal ion selected from the group: Gd(III), Dy(III), Fe(III), and Mn(II), the targeting moiety is a quinolone nonpeptide and the linking group is present between the targeting moiety and chelator.
[27] In another preferred embodiment, the targeting moiety is quinolone non-peptide and the receptor is ocvfb or avps.

[28] In another preferred embodiment, the metal ion is 15 Gd(III).
[29] In another preferred embodiment, the contrast agent is




[30] In another preferred embodiment wherein the metallopharmaceutical is a X-ray contrast agent, the metal is selected from the group: Re, Sm, Ho, Lu, Pm, Y, Bi, Pd, Gd, La, Au, Au, Yb, Dy, Cu, Rh, Ag, and Ir, the targeting moiety is a quinolone non-peptide, the receptor is av^3 or avPs, and the linking group is present between the targeting moiety and chelator.
10
[31] In another preferred embodiment, the present invention provides a novel method of treating rheumatoid arthritis in a patient comprising: administering a therapeutic radiopharmaceutical of Claim 19 capable of 15 localizing in new angiogenic vasculature to a patient by injection or infusion.
[32] In another preferred embodiment, the present invention provides a novel method of treating cancer in a 20 patient comprising: administering to a patient in need thereof a therapeutic radiopharmaceutical of Claim 19 by injection or infusion.
[33] In another preferred embodiment, the present 25 invention provides a novel method of imaging therapeutic angiogenesis in a patient comprising: (1) administering a diagnostic radiopharmaceutical, a MRI contrast agent,

or a X-ray contrast agent of Claim 11 to a patient by injection or infusion. (2) imaging the area of the patient wherein the desired formation of new blood vessels is located. 5
[34) In another preferred embodiment, the present invention provides a novel method of imaging cancer in a patient comprising.- (1) administering a diagnostic radiopharmaceutical of Claim 12 to a patient by injection 10 or infusion; (2) imaging the patient using planar or SPECT gamma scintigraphy, or positron emission tomography.
(35] In another preferred embodiment, the present 15 invention provides a novel method of imaging cancer in a patient comprising: (1) administering a MRI contrast agent of Claim 26,- and (2) imaging the patient using magnetic resonance imaging.
20
25
[36] In another preferred embodiment, the present invention provides a novel method of imaging cancer in a patient comprising: (1) administering a X-ray. contrast agent of Claim 30; and (2) imaging the patient using X-ray computed tomography.
30
[37] in a third embodiment, the present invention provides a novel compound, comprising: a targeting moiety and a surfactant, wherein the targeting moiety is bound to the surfactant, is a nonpeptide, and binds to a receptor that is upregulated during angiogenesis and the compound has 0-1 linking groups between the targeting moiety and surfactant.
[38] In a preferred embodiment, the targeting moiety 35 comprises a guinolone non-peptide and the linking group is present between the targeting moiety and surfactant.


5

[39] In another preferred embodiment, the receptor is the integrin av^3 or avp5 and the compound is of the formula:
(Q)d-Ln-Sf wherein, 0 is a compound of Formula (II) :


10

including stereoisomeric forms thereof, or mixtures of stereoisomeric forms thereof, or pharmaceutically 15 acceptable salt or prodrug forms thereof wherein:
Rle is selected from:


Ae is -CH2- or -N(R10e)-;
Ale and Be are independently -CH2- or -N(R10e)-; 10
De is -N(RlOe)_ or _s_.
Ee-Fe is -C(R2e)=C(R3e)- or -C (R2e) 2C (R3e) 2-; 15 Je is -C(R2e)- or -N-;
Ke, Le and M*5 are independently -C(R2e)- or -C(R3e)-; R2e and R3e are independently selected from:


H, C1-C4 alkoxy, NRlleR12e, halogen, NO2, CN, CF3, C1-C6 alkyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl (C1-C4 alkyl), aryl (Ci-Ce alkyl)-, (C1-C6 alkyl)carbonyl, (Ci-Ce alkoxy)carbonyl, 5 arylcarbonyl, and aryl substituted with 0-4 R7e,
10
alternatively, when R2e and R3e are substituents on adjacent atoms, they can be taken together with the carbon atoms to which they are attached to form a 5-7 membered carbocyclic or 5-7 membered heterocyclic aromatic or nonaromatic ring system, said carbocyclic or heterocyclic ring being substituted with 0-2 groups selected from C1-C4 alkyl, C1-C4 alkoxy, halo, cyano, amino, CF3 and NO2;
15
R2ae is selected from:
H, C1-C10 alkyl, C2-C6 alkenyl, C3-C11 cycloalkyl,
C3-C7 cycloalkyl (C1-C4 alkyl), aryly aryl(C1-C4
20
alkyl)-, (C2-C7 alkyl)carbonyl, arylcarbonyl,
(C2-C10 alkoxy)carbonyl, C3-C7 cycloalkoxycarbonyl,
C7-C11 bicycloalkoxycarbonyl, aryloxycarbonyl,
aryl(Ci^Cio alkoxy)carbonyl,
Ci-Ce alkylcarbonyloxy(C1-C4 alkoxy)carbonyl,
25
arylcarbonyloxy(C1-C4 alkoxy)carbonyl, and
C3-C7 cycloalkyl carbonyloxy (C1-C4 alkoxy) carbonyl ,-
R7e is selected from:
H, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, aryl, aryKCi-C4 alkyl)-, (C1-C4 alkyl)carbonyl, C02R18ae, S02Rlle, 30 SO2NR10eRlle, OR10e, and N(Rlle)R12e;
Ue is selected from:
-(CH2)ne-/ -(CH2)neO(CH2),ne-' - (CH2 ) neN(R12) (CH2)me- , -NH(CH2)ne-, -(CH2)neC(=0)(CH2)me-, 35 -(CH2)neS(0)pe(CH2)me-, -(CH2)n*NHNH(CH2)me~,
-N(R10e)C(=O)-, -NHC(=0) (CH2)ne-, -C (=O)N(R10e) - , and -N(R10e)S(O)pe-;

aryl substituted with 0-3 groups selected from
halogen, C1-C6 alkoxy, Ci-Cg alkyl, CF3,
S(0)meMe, and -NMe2,
aryl(C1-C4 alkyl)-, said aryl being substituted with
5 0-3 groups selected from halogen, C1-C6 alkoxy,
Ci-Ce alkyl, CF3, S(0)peMe, and -NMe2, and a 5-10 membered heterocyclic ring containing 1-3 N, C, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, 10 said heterocyclic ring being substituted with 0-2 R7e;
R10e is selected from:
H, CF3, C3-C6 alkenyl, C3-C11 cycloalkyl, aryl, (C3-C11 cycloalkyl)methyl, aryl(C1-C4 alkyl), and Ci-15 C10 alkyl substituted with 0-2 R6e;
Rlle is selected from:
H, hydroxy, Ci-Cg alkyl, C3-C6 alkenyl, C3-C11 cycloalkyl, (C3-C11 cycloalkyl)methyl, Ci-Ce alkoxy, 20 benzyloxy, aryl, heteroaryl, heteroaryl(C1-C4 alkyl)-, aryl(Ci-C4 alkyl), adamantylmethyl, and C1-C10 ^Ikyl substituted with 0-2 R4e;
25
R4e is selected from:
H, Cx-C€ alkyl, C3-C7 cycloalkyl, C3~C7
cycloalkyl (Ca-C4 alkyl)-, (C1-C3.0 alkylJcarbonyl,
aryl, heteroaryl, aryl(C1-C6 alkyl)-, and
heteroaryl (C-pCg alkyl)-, wherein said aryl or
30
heteroaryl groups are substituted with 0-2 substituents independently selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, F, CI, Br, CF3, and N02,
alternatively, when R10e and Rlle are both substituents on 35 the same nitrogen atom (as in -NR10eRlle) they may be taken together with the nitrogen atom to which they are attached to form a heterocycle selected from: 3-azabicyclononyl, 1,2,3,4-tetrahydro-l-quinolinyl,

1,2,3, 4-tetrahydro-2-isoquinolinyl, 1-piperidinyl, 1-morpholinyl, 1-pyrrolidinyl, thiamorpholinyl, thiazolidinyl, and 1-piperazinyl;
said heterocycle being substituted with 0-3 croups
5 selected from: C1-C6 alkyl, aryl, heteroaryl,
aryl(Ci-C4 alkyl)-, (C1-C6 alkyl) carbonyl, (C3-C7 cycloalkyDcarbonyl, (Ci-Ce alkoxy) carbonyl, aryl (Ci-C4 alkoxy)carbonyl, Ci~Cs alkylsulfonyl, and arylsulfonyl;
10
R12e is selected from:
H, C1-C6 alkyl, triphenylmethyl, methoxymethyl, methoxyphenyldiphenylmethyl, trimethylsilylethoxymethyl, (C1-C6 alkyl) carbonyl,
15 (C1-C6 alkoxy) carbonyl, (Ci-Ce alkyl)aminocarbonyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl (Ci-C4 alkyl)-, aryl, heteroaryl(Ci-Ce alkyl)carbonyl, heteroarylcarbonyl, aryl (C1-C6 alkyl)-, (C1-C6 alkyl)carbonyl, arylcarbonyl, C1-C6 20 alkylsulfonyl, arylsulfonyl, aryl (C1-C6
alkyDsulfonyl, heteroarylsulfonyl, heteroaryl (C1-C6 alkyDsulfonyl, aryloxycarbonyl, and aryl (C1-C6 alkoxy)carbonyl, wherein said aryl groups are substituted with 0-2 substituents selected from the 25 group consisting of C1-C4 alkyl, C1-C4 alkoxy, halo, CF3, and nitro;
R16e is selected from:
-C(=0)OR18ae, -C(=0)R18be, -C(=0)N(R18be)2, 30 -C(=0)NHS02R18ae, -C(=0)NHC(=0)R18be,
-C(=0)NHC(=0)0R18ae, -C(=0)NHS02NHR18be, -S02R18ae, -S02N(R18be)2, and -S02NHC R17e is selected from: 35 H, Ci-Ce alkyl, C3-C7 cycloalkyl, C3-C7
cycloalkyl(C1-C4 alkyl)-, aryl, aryl(Ci-C6 alkyl)-, and heteroaryl(C1-C6 alkyl);

Ri8ae iS selected from:
C1-C8 alkyl optionally substituted with a bond to Ln,
C3-C11 cycloalkyl optionally substituted with a
bond to Ln, aryl(Ci-C6 alkyl)- optionally
5 substituted with a bond to Ln, heteroaryl(C1-C6
alkyl)- optionally substituted with a bond to Ln. (Ci-Ce alkyl)heteroaryl optionally substituted with a bond to Ln, biaryl {C1-C6 alkyl) optionally substituted with a bond to
10 Ln, heteroaryl optionally substituted with a
bond to Wj, phenyl substituted with 3-4 R19e and optionally substituted with a bond to Ln, naphthyl substituted with 0-4 R19e and optionally substituted with a bond to Ln, and a
15 bond to Ln, wherein said aryl or heteroaryl
groups are optionally substituted with 0-4 R19e;
R18be is H or Rl8ae.
20 R19e is selected from:
H, halogen, CF3, C02H, CN. N02, -NRlleR12e, OCF3, Ci-Ce a^kyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C11 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl)-, aryl(Ci-Ce alkyl)-, C1-C6 alkoxy, C1-C4
25 alkoxycarbonyl, aryl, aryl-O-, aryl-S02~,
heteroaryl, and heteroaryl-SC>2-, wherein said aryl
and heteroaryl groups are substituted with 0-4 groups selected from hydrogen, halogen, CF3, C1-C3 alkyl, and C1-C3 alkoxy;
30
R20e is selected from:
hydroxy, C1-C10 alkyloxy, C3-C11 cycloalkyloxy,
aryloxy, aryl(C1-C4 alkyl)oxy,
C2-C10 alkylcarbonyloxy(Ci-C2 alkyl)oxy-,
35 C2-C10 alkoxyc^rbonyloxy(Ci-C2 alkyl )oxy-, C2-C10 alkoxycarbonyl (C1-C2 alkyl )oxy-, C3-C10 cycloalKylcarbonyloxy(Ci-C2 alkyl )oxy-, C3-C10 cycloalXoxycarbonyloxytCi-C2 alkyl)oxy-,


C3-C10 cycloalkoxycarbonyl (C1-C2 alkyDoxy-, aryloxycarbonyl (C1-C2 alkyDoxy-, aryloxycarbonyloxy(C1-C2 alkyDoxy-, arylcarbonyloxy(C1-C2 alkyDoxy-,
C1-C5 alkoxy(C1-C5 alkyl)carbonyloxy(C1-C2 alkyDoxy, (5- (C1-C5 alkyl)-1,3-dioxa-cyclopenten-2-one-
ylImethyloxy, (5-aryl-l, 3-dioxa-cyclopenten-2-one-yl)methyloxy,
and (R10e) (Rlle)N-(Ci-Cio alkoxy)-;
R21e is selected from:
C1-C8 alkyl, C2-C6 alkenyl, C3-C11 cycloalkyl, (C3-C11 cycloalkyl)methyl, aryl, aryl(C1-C4 alkyl)-, and Ci-C10 alkyl substituted with 0-2 R7e;
R22e is selected from:
-C(=0)-R18be, ~C(=0)N(R18be)2, -C(=0)NHS02R18ae. -C(=0)NHC(=0)Rl8be, -C(=0)NHC(=0)OR18ae, and -C(=0)NHS02NHRl8be;
Ye is selected from:
-COR20e, -SO3H, -PO3H, -CONHNHSO2CF3, -CONHS02R18ae, -CONHS02NHR18be, -NHCOCF3, -NHCONHS02R18ae, -NHS02R18ae, -OPO3H2, -OSO3H, -PO3H2, -S02NHCOR18ae, -S02NHC02R18ae,
H , H , and HO N0;
me is 0-2; ne is 0-4; pe is 0-2; re is 0-2;

with the following proviso: ne and ine are chosen such
that the number of atoms connecting Rle and Ye is in the range of 8-14; 5
d is selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
Ln is a linking group having the formula:
10 ( W is independently selected at each occurrence from the
group: 0, S, NH, NHC(=0), C(=0)NH, NR8C(=0), C(=0)N
R8, C(=0), C(*0)0, OC(=0), NHC( = S)NH, NHC(=0)NH# S02,
15 S02NH, (OCH2CH2) 20-200. (CH2CH20) 20-200- (OCH2CH2CH2)20-
200/ (CH2CH2CH20)20-200- and (aa)t-; aa is independently at each occurrence an amino acid;
20 Z is selected from the group: aryl substituted with 0-3 R10- C3-10 cycloalkyl substituted with 0-3 R10, and a 5-10 mombered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O and substituted with 0-3 R10;
25
R6, R6a, R7, R7a, and R8 are independently selected at each occurrence from the group: H, =0, COOH, SO3H, PO3H, C1-C5 alkyl substituted with 0-3 R10, aryl
substituted with 0-3 R10, benzyl substituted with 0-3 30 R10, and C1-C5 alkoxy substituted with 0-3 R10,
NHC(=0)Ri;L, CUOJNHR11, NHC(=0)NHRn, NHR1*, R", and a bond to Sf;
R10 is independently selected at each occurrence from the 35 group: a bond to Sf, COOR11, C(=0)NHRn, NHC(=0)Rn, OH, NHR11, SO3H, PO3H, -OPO3H2, -OSO3H, aryl substituted with 0-3 R11, C1-5 alkyl substituted with 0-1 R12, C1-5 alkoxy substituted with 0-1 R", and a


5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R11;
5 R11 is independently selected at each occurrence from the group-. H, alkyl substituted with 0-1 R12, aryl substituted with 0-1 R12, a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and
10 substituted with 0-1 R12, C3-10 cycloalkyl
substituted with 0-1 R12, polyalkylene glycol substituted with 0-1 R12, carbohydrate substituted with 0-1 R12, cyclodextrin substituted with 0-1 R12, amino acid substituted with 0-1 R12, polycarboxyalkyl
15 substituted with 0-1 R12, polyazaalkyl substituted with 0-1 R12, peptide substituted with 0-1 R12, wherein the peptide is comprised of 2-10 amino acids, 3, 6-O-d.isulio-B-E>-galactopyranosyl, bis(phosphonomethyl)glycine, and a bond to Sf;
20
R12 is a bond to Sf;

1, and 2;
1, and 2;
1, , and 2 /
1, 2, 3, ■ 4, 5, 6, 7, 8, 9, and : 10;
1, . 2, 3, 4, 5, 6, 7, 8, 9, and 10;
1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
k is selected from 0,
h is selected from 0, 25 h' is selected from 0,
g is selected from 0,
g' is selected from 0,
t' is selected from 0,
x is selected from 0, 1, 2, 3, 4, and 5; 30 x' is selected from 0, 1, 2, 3, 4, and 5;
Sr is a surfactant which is a lipid or a compound of the

35

formula: A
A9 is selected from the group: OH and OR27;
A10 is OR27;

R27 is C(=0)Ci-2o alkyl;
E1 is Ci-io alkylene substituted with 1-3 R28; 5
R28 is independently selected at each occurrence from the group: R30, -PO3H-R30, =0, -C02R29, -C(=0)R29, -C(=0)N(R29)2, _CH2OR29, -OR29, -N(R29)2, C1-C5 alkyl, and C2-C4 alkenyl;
10
R2^ is independently selected at each occurrence from the group: R30, H, C1-C6 alkyl, phenyl, benzyl, and
trifluoromethyl; 15 R30 is a bond to Ln;
and a pharmaceutical^ acceptable salt thereof.
[40] In another preferred embodiment, the compound is of 20 the formula:

25

r Q-Ln-Sf
wherein, Q is a compound of Formula (IV)
R19e 0 0 R8e



19e
COR
R
Rle-Ue Ge' "N

20e



R

18ae

(IV)
30 including stereoisomeric forms thereof, or mixtures of stereoisomeric forms thereof, or pharmaceutically acceptable salt or prodrug forms thereof wherein:



Rle is selected from:
R2e and R3e are independently selected from:
H, C1-C4 alkoxy, NRlleR12e, halogen, N02, CN, CF3, C1-C6 alkyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 alternatively, when R2e and R3e are substituents on adjacent atoms, they can be taken together with the carbon atoms to which they are attached to form a 5-7 membered carbocyclic or 5-7 membered heterocyclic aromatic or nonaromatic ring system, said carbocyclic or heterocyclic ring being substituted with 0-2 grouos selected from C1-C4 alkyl, C1-C4 alkoxy, halo, cyano, amino, CF3 and NO2;
R2ae j_s selected from:
H, C1-C10 alkyl, C2-C6 alkenyl, C3-CH cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl), aryl,

aryl(Ci-C4 alkyl)-, (C2-C7 alkyl)carbonyl,
arylcarbonyl,
(C2-C10 alkoxy)carbonyl, C3-C7 cycloalkoxycarbonyl,
C7-C11 bicycloalkoxycarbonyl, aryloxycarbonyl,
aryl(Ci-Cio alkoxy)carbonyl,
C1-C6 alkylcarbonyloxy (C1-C4 alkoxy)carbonyl,
arylcarbonyloxy(C1-C4 alkoxy)carbonyl, and
C3-C7 cycloalkylcarbonyloxy(C1-C4 alkoxy)carbonyl;
is selected from: H, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, aryl, aryl(Cj-C4 alkyl)-, (C1-C4 alkyl)carbonyl, C02R18ae, S02Rlle, SO2NR10eRlle, OR10e, and N (Rlle) R12e;
is selected from:
-(CH2)ne-, -(CH2)neO(CH2)me-, -NH(CH2)ne-, -N(R10e)C(=O)-, -NHC(=0) (CH2)ne-, and -C ( =0) N(R10e) -;
is N or CR19e;
is selected from: H, C02»18be. C(=0)R18be, CONR17eR18be, C1-C10 alkyl substituted with 0-1 R6e, C2-Cio alkenyl substituted with 0-1 R6e, C2-C10 alkynyl substituted with 0-1 R6e, C3-C8 cycloalkyl substituted with 0-1 R6e, C5-C6 cycloalkenyl substituted with 0-1 R6e, (C1-C10 alkyl)carbonyl, C3-C10 cycloalkyl(C1-C4 alkyl)-, phenyl substituted with 0-3 R6e, naphthyl substituted with 0-3 R6e,
a 5-10 membered heterocyclic ring containing 1-3 N, O, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e;
is selected from:


Ci-Cio alkyl substituted with 0-1 R6e, C1-C10 alkoxy substituted with 0-2 R7e, H, nitro, N(Rlle)Rl2e, OC(=O)R10e, OR10e,
OC(=O)NR10eRlle, NR10eC(=O)R10e, NR10eC (=0) OR21e,
5 NR10eC(=O)NR10eRlle, NR10eSO2NR10eRlle,
NR10eSO2R21e, hydroxy, OR22e, -N(R10e) Rlle, -
N(R16e)R17e, aryl(C0-C6 alkyl) carbonyl, aryl (Ci-
C6 alkyl), heteroaryl(Ci-C6 alkyl), CONR18aeR20e,
S02R18ae, and SO2NR18aeR20e,
10 providing that any of the above alkyl, cycloalkyl,
aryl or heteroaryl groups may be unsubstituted or substituted independently with 1-2 R7e;
R6e is selected from:
15 H, Ci-Cio alkyl, hydroxy, Ci-Cio alkoxy, nitro, Ci-Cio
alkylcarbonyl, -N(Rlle)R12e, cyano, halo, CF3/ CHO, C02R18be, c(=0)R18be, CONR17eR18be, OC(=0)Rl°e, OR10e, OC(=O)NR10eRlle, NR10eC(=O)R10e, NR10eC(=O)OR21e,
20 NRl0eC(=O)NR10eRlle/ NR10eSO2NR10eRlle,
NR10esO2R21e, S(0)peRlle, SO2NR10eRlle, aryl substituted with 0-3 groups selected from halogen, Ci-Ce alkoxy, Ci-Ce alkyl, CF3, S(0)meMe, and -NMe2, 25 aryl(C1-C4 alkyl)-, said aryl being substituted with 0-3 groups selected from halogen, Ci-Ce alkoxy, Ci-Ce alkyl, CF3, S(0)peMe, and -NMe2, and a 5-10 membered heterocyclic ring containing 1-3 N, 0, or S heteroatoms, wherein said heterocyclic ring may be 30 saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e;
R10e is selected from:
H, CF3, C3-C6 alkenyl, C3-C11 cycloalkyl, aryl, 35 (C3-C11 cycloalkyl)methyl, aryl(Ci-C4 alkyl), and Ci-C10 alkyl substituted with 0-2 R6e;
Rlle is selected from:

H, hydroxy, CI-CB alkyl, C3-C6 alkenyl, C3-C11 cycloalkyl, (C3-C11 cycloalkyl)methyl, Ci-Ce alkoxy, benzyloxy, aryl, heteroaryl, heteroaryl(C1-C4 alkyl)-, aryl(Ci-C4 alkyl), adamantylmethyl, and C1-C10 alkyl substituted with 0-2 R4e;
R4e is selected from:
H, C1-Ce alkyl, C^-Cj cycloalkyl, C^-Cj
cycloalkyl(C2-C4 alkyl)-, aryl, heteroaryl, arylfCj-
C6 alkyl)-, and heteroaryl (C^Cg alkyl)-, wherein
said aryl or heteroaryl groups are substituted with 0-2 substituents independently selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, F, CI, Br, CF3, and NO2,
R12e is selected from:
H, Ci-Ce alkyl, triphenylmethyl, methoxymethyl, methoxyphenyldiphenylmethyl,
trimethylsilylethoxymethyl, (Ci-Ce alkyl)carbonyl, (C1-C6 alkoxy)carbonyl, (C1-C6 alkyl)aminocarbonyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(Ci-C4 alkyjr)-, aryl, heteroaryl(Ci-Cs alkyl)carbonyl, heteroarylcarbonyl, aryl (C1-C6 alkyl)-, (Ci-Ce alkyl)carbonyl, arylcarbonyl, C1-C6 alkylsulfonyl, arylsulfonyl, aryl(C1-C6 alkyl)sulfonyl, heteroarylsulfonyl, heteroaryl (C1-C6 alkyl)sulfonyl, aryloxycarbonyl, and aryl(C1-C6 alkoxy)carbonyl, wherein said aryl groups are substituted with 0-2 substituents selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, halo, CF3, and nitro;
R16e is selected from:
-C(=0)OR18ae, -C(=0)R18be, -C(=0)N(R18be)2, -S02R28ae, and -S02N(R18b*)2;
R17e is selected from:


H, Ci-Ce alkyl, C3-C7 cycloalkyl, C3-C7
cycloalkyl(C1-C4 alkyl)-, aryl, aryl(Ci-C6 alkyl)-,
and heteroaryl(C1-C6 alkyl);
5 Risae iS selected from:
C1-C8 alkyl optionally substituted with a bond to Ln, C3-C21 cycloalkyl optionally substituted with a bond to Ln, aryl(Ci-C6 alkyl)- optionally substituted with a bond to Ln, heteroaryl(Ci-Cg
10 alkyl)- optionally substituted with a bond to
Ln, (Ci-Ce alkyl)heteroaryl optionally substituted with a bond to Ln, biaryl (Ci-Ce alkyl) optionally substituted with a bond to Ln, heteroaryl optionally substituted with a
15 bond to Ln, phenyl substituted with 3-4 R19e and
optionally substituted with a bond to Ln,
naphthyl substituted with 0-4 R19e and
optionally substituted with a bond to Ln, and a
bond to Ln, wherein said aryl or heteroaryl
20 groups are optionally substituted with 0-4 R19e;
Rl8be is H of R18ae;
R19e is selected from:
25 H, halogen, CF3, C02H, CN, N02, -NRlleR12e, OCF3, Ci-Cg alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C11 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl)-, aryl(Ci-C6 alkyl)-, C1-C6 alkoxy, C1-C4 alkoxycarbonyl, aryl, aryl-O-, aryl-S02-/
30 heteroaryl, and heteroaryl-SC>2-, wherein said aryl and heteroaryl groups are substituted with 0-4 groups selected from hydrogen, halogen, CF3, C1-C3 alkyl, and C1-C3 alkoxy;
35 R20e is selected from:
hydroxy, C1-C10 alkyloxy, C3-C11 cycloalkyloxy,
aryloxy, aryl (C1-C4 alkyDoxy,
C2-C10 alkylcarbonyloxy(Ci-C2 alkyDoxy-,


contain all or part of the formulation can independently be in the form of a sterile solution or a lyophilized solid.
Another aspect of the present invention contemplates 5 a method of imaging cancer in a patient involving: (1) synthesizing a diagnostic radiopharmaceutical of the present invention, using a reagent of the present invention, capable of localizing in tumors; (2) administering said radiopharmaceutical to a patient by 10 injection or infusion; (3) imaging the patient using
planar or SPECT gamma scintigraphy, or positron emission tomography.
Another aspect of the present invention contemplates a method of imaging cancer in a patient involving: (1) 15 administering a paramagnetic metallopharmaceutical of the present invention capable of localizing in tumors to a patient by injection or infusion,- and (2) imaging the patient using magnetic resonance imaging.
Another aspect of the present invention contemplates 20 a method of imaging cancer in a patient involving: (1) administering a X-ray contrast agent of the present invention capable of localizing in tumors to a patient by injection or infusion; and (2) imaging the patient using X-ray computed tomography. 25 Another aspect of the present invention contemplates a method of imaging cancer in a patient involving: (1) administering a ultrasound contrast agent of the present invention capable of localizing in tumors to a patient by injection or infusion; and (2) imaging the patient using 30 sonography.
Another aspect of the present invention contemplates a method of treating cancer in a patient involving: (1) administering a therapeutic radiopharmaceutical of the present invention capable of localizing in tumors to a 35 patient by injection or infusion.


DEFINITIONS
The compounds herein described may have asymmetric centers. Unless otherwise indicated, all chiral, 5 diastereomeric and racemic forms are included in the present invention. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. It will be 10 appreciated that compounds of the present invention
contain asymmetrically substituted carbon atoms, and may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms or by 15 synthesis from optically active starting materials. Two distinct isomers (cis and trans) of the peptide bond are known to occur; both can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. The D and 20 L-isomers of a particular amino acid are designated herein using the conventional 3-letter abbreviation of the amino acid, as indicated by the following examples: D-Leu, or L-Leu.
When any variable occurs more than one time in any 25 substituent or in any formula, its definition on each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0-2 R52, then said group may optionally be substituted with up to two R52, and R52 at 30 each occurrence is selected independently from the
defined list of possible R52. Also, by way of example, for the group -N(R53)2, each of the two R53 substituents on N is independently selected from the defined list of possible R53. Combinations of substituents and/or 35 variables are permissible only if such combinations
result in stable compounds. When a bond to a substituent is shown to cross the bond connecting two atoms in a

ring, then such substituent may be bonded to any atom on the ring.
The term "nonpeptide" means preferably less than three amide bonds in the backbone core of the targeting 5 moiety or preferably less than three amino acids or amino acid mimetics in the targeting moiety.
The term "metallopharmaceutical" means a pharmaceutical comprising a metal. The metal is the cause of the imageable signal in diagnostic applications and 10 the source of the cytotoxic radiation in radiotherapeutic applications. Radiopharmaceuticals are metallopharmaceuticals in which the metal is a radioisotope.
By "reagent" is meant a compound of this invention 15 capable of direct transformation into a
metallopharmaceutical of this invention. Reagents may be utilized directly for the preparation of the metallopharmaceuticals of this invention or may be a component in a kit of this invention. 20 The term "binding agent" means a
metallopharmaceutical of this invention having affinity for and capable of binding to the vitronectin receptor. The binding agents of this invention have Ki By "stable compound" or "stable structure" is meant 25 herein a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious pharmaceutical agent.
The term "substituted", as used herein, means that 30 one or more hydrogens on the designated atom or group is replaced with a selection from the indicated group, provided that the designated atom's or group's normal valency is not exceeded, and that the substitution results in a stable compound. When a substituent is keto 35 (i.e., =0), then 2 hydrogens on the atom are replaced. The term "bond", as used herein, means either a single or double bond.
The term "salt", as used herein, is used as defined in the CRC Handbook of Chemistry and Physics, 65th

Edition, CRC Press, Boca Raton, Fla, 1984, as any substance which yields ions, other than hydrogen or hydroxyl ions. As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed 5 compounds modified by making acid or base salts.
Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. 10 The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals 15 without excessive toxicity, irritation, allergic
response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein 20 the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic 25 acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include 30 those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, tartaric, citric, ascorbic, pamoic, maleic, 35 hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.

The pharmaceutical^ acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared 5 by reacting the free acid or base forms of these
compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or 10 acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Eastcn, PA, 1985, p. 1418, the disclosure of which is hereby incorporated by reference. As used herein, "alkyl" is intended to include both 15 branched and straight-chain saturated aliphatic
hydrocarbon groups having the specified number of carbon atoms, examples of which include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, sec-butyl, t-butyl, pentyl, hexyl, heptyl, octyl, nonyl, 20 and decyl; "cycloalkyl* or "carbocycle" is intended to include saturated and partially unsaturated ring groups, including mono-, bi- or poly-cyclic ring systems, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl and adamantyl; "bicycloalkyl" or 25 "bicyclic" is intended to include saturated bicyclic ring groups such as [3.3.0]bicyclooctane, [4.3.0]bicyclononane, [4.4.O]bicyclodecane (decalin), [2.2.2]bicyclooctane, and so forth.
As used herein, the term "alkene" or "alkenyl" is 30 intended to include hydrocarbon chains having the
specified number of carbon atoms of either a straight or branched configuration and one or more unsaturated carbon-carbon bonds which may occur in any stable point along the chain, such as ethenyl, propenyl, and the like. 35 As used herein, the term "alkyne" or "alkynyl" is intended to include hydrocarbon chains having the specified number of carbon atoms of either a straight or branched configuration and one or more unsaturated


carbon-carbon triple bonds which may occur in any stable point along the chain, such as propargyl, and the like.
As used herein, "aryl" or "aromatic residue" is intended to mean phenyl or naphthyl, which when 5 substituted, the substitution can be at any position. As used herein, the term "heterocycle" or "heterocyclic system" is intended to mean a stable 5- to 7- membered monocyclic or bicyclic or 7- to 10-membered bicyclic heterocyclic ring which is saturated partially 10 unsaturated or unsaturated (aromatic), and which consists of carbon atoms and from 1 to 4 heteroatoms independently selected from the group consisting of N, 0 and S and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene 15 ring. The nitrogen and sulfur heteroatoms may optionally be oxidized. The heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom which results in a stable structure. The heterocyclic rings described herein may be substituted on carbon or on a 20 nitrogen atom if the resulting compound is stable. If specifically noted, a nitrogen in the heterocycle may optionally be quaternized. It is preferred that when the total number of S and 0 atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one 25 another. It is preferred that the total number of S and 0 atoms in the heterocycle is not more than 1. As used herein, the term "aromatic heterocyclic system" is intended to mean a stable 5- to 7- membered monocyclic or bicyclic or 7- to 10-membered bicyclic heterocyclic 30 aromatic ring which consists of carbon atoms and from 1 to 4 heteroatoms independently selected from the group consisting of N, 0 and S. It is preferred that the total number of S and 0 atoms in the aromatic heterocycle is not more than 1. 35 Examples of heterocycles include, but are not limited to, lH-indazole, 2-pyrrolidonyl, 2H,6H-l,5,2-dithiazinyl, 2H-pyrrolyl, 3H-indolyl, 4-piperidonyl, 4aH-carbazole, 4H-quinolizinyl, 6H-1,2,5-thiadiazinyl, acridinyl, azocinyl,

benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazalonyl, carbazolyl, 5 4aif-carbazolyl, --carbolinyl, chromanyl, chromenyl,
cinnolinyl, decahydroquinolinyl, 2H. SH-1,5, 2-dithiazinyl, dihydrofuro(2,3-jb] tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, lH-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 10 isobenzofuranyl, isochromanyl, isoindazolyl,
isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1, 2, 4-oxadiazolyl, 1, 2,5-oxadiazolyl, 1,3,4-oxadiazolyl, 15 oxazolidinyl., oxazolyl, oxazolidinylperimidinyl, phenanthridinyl, phenanthrolinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, pteridinyl, piperidonyl, 4-piperidonyl, pteridinyl, purinyl, pyranyl, 20 pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, 25 carbolinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, 6#-l,2, 5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3, 4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, 30 thienoimidazolyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, xanthenyl. Preferred heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, indolyl, benzimidazolyl, 35 lH-indazolyl, oxazolidinyl, benzotriazolyl,
benzisoxazolyl, oxindolyl, benzoxazolinyl, or isatinoyl. Also included are fused ring and spiro compounds containing, for example, the above heterocycles.

As used herein, the term "alkaryl" means an aryl group bearing an alkyl group of 1-10 carbon atoms; the term "aralkyl" means an alkyl group of 1-10 carbon atoms bearing an aryl group; the term "arylalkaryl" means an 5 aryl group bearing an alkyl group of 1-10 carbon atoms bearing an aryl group; and the term "heterocycloalkyl" means an alkyl group of 1-10 carbon atoms bearing a heterocycle.
A "polyalkylene glycol" is a polyethylene glycol, 10 polypropylene glycol or polybutylene glycol having a
molecular weight of less than about 5000, terminating in either a hydroxy or alkyl ether moiety.
A "carbohydrate" is a polyhydroxy aldehyde, ketone, alcohol or acid, or derivatives thereof, including 15 polymers thereof having polymeric linkages of the acetal type.
A "cyclodextrin" is a cyclic oligosaccharide. Examples of cyclodextrins include, but are not limited to, oc-cyclodextrin, hydroxyethyl-a-cyclodextrin, 20 hydroxypropyl-a-cyclodextrin, (3-cyclodextrin, hydroxypropyl-p-cyclodextrin, carboxymethyl-p-cyclodextrin, dihydroxypropyl-p-cyclodextrin, hydroxyethyl-p-cyclodextrin, 2,6 25 di-O-methyl-p-cyclodextrin, sulfated-p-cyclodextrin, y-cyclodextrin, hydroxypropyl-y-cyclodextrin, di hydroxypropyl-y- cyclodextrin,
hydroxyethyl-y-cyclodextrin, and sulfated y-cyclodextrin. As used herein, the term * poly carboxya Iky 1" means an 30 alkyl group having between two and about 100 carbon atoms and a plurality of carboxyl substituents; and the term "polyazaalkyl" means a linear or branched alkyl group having between two and about 100 carbon atoms, interrupted by or substituted with a plurality of amine 35 groups.


A "reducing agent" is a compound that reacts with a radionuclide, which is typically obtained as a relatively unreactive, high oxidation state compound, to lower its oxidation state by transferring electron (s) to the 5 radionuclide, thereby making it more reactive. Reducing agents useful in the preparation of radiopharmaceuticals and in diagnostic kits useful for the preparation of said radiopharmaceuticals include but are not limited to stannous chloride, stannous fluoride, formamidine 10 sulfinic acid, ascorbic acid, cysteine, phosphines, and cuprous or ferrous salts. Other reducing agents are described in Brodack et. al., PCT Application 94/22496, which is incorporated herein by reference.
A "transfer ligand" is a ligand that forms an 15 intermediate complex with a metal ion that is stable enough to prevent unwanted side-reactions but labile enough to be converted to a metallopharmaceutical. The formation of the intermediate complex is kinetically favored while the formation of the metallopharmaceutical 20 is thermodynamically favored. Transfer ligands useful in the preparation of metallopharmaceuticals and in diagnostic kits useful for the preparation of diagnostic radiopharmaceuticals include but are not limited to gluconate, glucoheptonate, mannitol, glucarate, 25 N, N,N' ,N'-ethylenediaminetetraacetic acid, pyrophosphate and methylenediphosphonate. In general, transfer ligands are comprised of oxygen or nitrogen donor atoms.
The term "donor atom" refers to the atom directly attached to a metal by a chemical bond. 30 "Ancillary" or "co-ligands" are ligands that are incorporated into a radiopharmaceutical during its synthesis. They serve to complete the coordination sphere of the radionuclide together with the chelator or radionuclide bonding unit of the reagent. For 35 radiopharmaceuticals comprised of a binary ligand system, the radionuclide coordination sphere is composed of one or more chelators or bonding units from one or more reagents and one or more ancillary or co-ligands, provided that there are a total of two types of ligands,


chelators or bonding units. For example, a radiopharmaceutical comprised of one chelator or bonding unit from one reagent and two of the same ancillary or co-ligands and a radiopharmaceutical comprised of two 5 chelators or bonding units from one or two reagents and one ancillary or cc?-ligand are both considered to be comprised of binary ligand systems. For radiopharmaceuticals comprised of a ternary ligand system, the radionuclide coordination sphere is composed 10 of one or more chelators or bonding units from one or more reagents and one or more of two different types of ancillary or co-ligands, provided that there are a total of three types of ligands, chelators or bonding units. For example, a radiopharmaceutical comprised of one 15 chelator or bonding unit from one reagent and two
different ancillary or co-ligands is considered to be comprised of a ternary ligand system.
Ancillary or co-ligands useful in the preparation of radiopharmaceuticals and in diagnostic kits useful for 20 the preparation of said radiopharmaceuticals are comprised of one or more oxygen, nitrogen, carbon, sulfur, phosphorus, arsenic, selenium, and tellurium donor atoms. A ligand can be a transfer ligand in the synthesis of a radiopharmaceutical and also serve as an 25 ancillary or co-lig^nd in another radiopharmaceutical. Whether a ligand is termed a transfer or ancillary or co-ligand depends on whether the ligand remains in the radionuclide coordination sphere in the radiopharmaceutical, which is determined by the 30 coordination chemistry of the radionuclide and the chelator or bonding unit of the reagent or reagents. A •chelator" or "bonding unit" is the moiety or group on a reagent that binds to a metal ion through the formation of chemical bonds with one or more donor atoms. 35 The term "binding site" means the site in vivo or in vitro that binds a biologically active molecule.
A "diagnostic kit" or "kit" comprises a collection of components, termed the formulation, in one or more vials which are used by the practicing end user in a

clinical or pharmacy setting to synthesize diagnostic radiopharmaceuticals. The kit provides all the requisite components to synthesize and use the diagnostic radiopharmaceutical except those that are commonly 5 available to the practicing end user, such as water or saline for injection, a solution of the radionuclide, equipment for heating the kit during the synthesis of the radiopharmaceutical, if required, equipment necessary for administering the radiopharmaceutical to the patient such 10 as syringes and shielding, and imaging equipment.
Therapeutic radiopharmaceuticals, X-ray contrast agent pharmaceuticals, ultrasound contrast agent pharmaceuticals and metallopharmaceuticals for magnetic resonance imaging contrast are provided to the end user 15 in their final form in a formulation contained typically in one vial, as either a lyophilized solid or an aqueous solution. The end user reconstitutes the lyophilized with water or saline and withdraws the patient dose or just withdraws the dose from the aqueous solution 20 formulation as provided.
A "lyophilization aid" is a component that has favorable physical properties for lyophilization, such as the glass transition temperature, and is added to the formulation to improve the physical properties of the 25 combination of all the components of the formulation for lyophi1i zat ion.
A "stabilization aid" is a component that is added to the metallopharmaceutical or to the diagnostic kit either to stabilize the metallopharmaceutical or to 30 prolong the shelf-life of the kit before it must be used. Stabilization aids can be antioxidants, reducing agents or radical scavengers and can provide improved stability by reacting preferentially with species that degrade other components or the metallopharmaceutical. 35 A "solubilization aid" is a component that improves the solubility of one or more other components in the medium required for the formulation.
A "bacteriostat" is a component that inhibits the growth of bacteria in a formulation either during its

storage before use of after a diagnostic Kit is used to synthesize a radiopharmaceutical.
The following abbreviations are used herein:
5 Acm acetamidomethyl
b-Ala, beta-Ala
or bAla 3-aminopropionic acid
ATA 2-ammothiazole-5-acetic acid or 2-
aminothiazole-5-acetyl group
JO Boc t-butyloxycarbonyl
CBZ, Cbz or Z Carbobenzyloxy
Cit citrulline
Dap 2,3-diaminopropionic acid
DCC dicyclohexylcarbodiimide
]5 DIEA diisopropylethylamine
DMAP 4-dimethylaminopyridine
EOE ethoxyethyl
HBTU 2-(lH-Benzotriazol-1-yD-1.1,3,3-
tetramethyluronium
20 hexafluorophosphate
hynic boc-hydrazinonicotinyl group or 2-
[[[5- r [carbonyl]-2-
pyridinyl]hydrazono]methyl] -benzenesulfonic acid, 25 NMeArg or MeArga-N-methyl arginine
NMeAsp a-N-methyl aspartic acid
NMM N-methylmorpholine
OcHex O-cyclohexyl
OBxl 0-benzyl
30 oSu O-succinimidyl
TBTU 2-QH-Benzotriazol-l-yl)-l,l/3,3-
tetrajnethyluronium tetrafluoroborate
THF tetrahydrofuranyl
THP tetrahydropyranyl
35 Tos tosyl
Tr trityl


The following conventional three-letter amino acid abbreviations are used herein; the conventional one-letter amino acid abbreviations are NQT used herein:

Ala = alanine
Arg - arginine
Asn = asparagine
Asp = aspartic acid
Cys = cysteine
Gin = glutamine
Glu = glutamic acid
Gly = glycine
His = histidine
He = isoleucine
Leu = leucine
Lys = lysine
Met = methionine
Nle = norleucine
Orn = ornithine
Phe = phenylalanine
Phg = phenylglycine
Pro = proline
Sar = sarcosine
Ser = serine
Thr = threonine
Trp = tryptophan
Tyr = tyrosine
Val - valine
30 As used herein, the term "bubbles", as used herein, refers to vesicles which are generally characterized by the presence of one or more membranes or walls surrounding an internal void that is filled with a gas or precursor thereto. Exemplary bubbles include, for
35 example, liposomes, micelles and the like.
As used herein, the term "lipid" refers to a synthetic or naturally-occurring amphipathic compound which comprises a hydrophilic component and a hydrophobic component. Lipids include, for example, fatty acids,



neutral fats, phosphatides, glycolipids, aliphatic alchols and waxes, terpenes and steroids.
As used herein, the term "lipid composition" refers to a composition which comprises a lipid compound. 5 Exemplary lipid compositions include suspensions, emulsions and vesicular compositions.
As used herein, the term "lipid formulation" refers to a composition which comprises a lipid compound and a bioactive agent. 10 As used herein, the term "vesicle" refers to a
spherical entity which is characterized by the presence of an internal void. Preferred vesicles are formulated from lipids, including the various lipids described herein. In any given vesicle, the lipids may be in the 15 form of a monolayer or bi layer, and the mono- or bi layer lipids may be used to form one of more mono- or bilayers. In the case of more than one mono- or bilayer, the mono-or bilayers are generally concentric. The lipid vesicles described herein include such entities commonly referred 20 to as liposomes, micelles, bubbles, microbubbles,
microspheres and the like. Thus, the lipids may be used to form a unilamellar vesicle (comprised of one monolayer or bilayer), an oligolamellar vesicle (comprised of about two or about three monolayers or bilayers) or a 25 multilamellar vesicle (comprised of more than about three monolayers or bilayers) . The internal void of the vesicles may be filled with a liquid, including, for example, an aqueous liquid, a gas, a gaseous precursor, and/or a solid or solute material, including, for 30 example, a bioactive agent, as desired.
As used herein, the term "vesicular composition" refers to a composition which is formulate from lipids and which comprises vesicles.
As used herein, the term "vesicle formulation" 35 refers to a composition which comprises vesicles and a bioactive agent.
As used herein, the term "lipsomes" refers to a generally spherical cluster or aggregate of amphipathic compounds, including lipid compounds, typically in the


form of one or more concentric layers, for example, bilayers. They may also be referred to herein as lipid vesicles.
Angiogenesis is the process of formation of new 5 capillary blood vessels from existing vasculature. It is an important component of a variety of physiological processes including ovulation, embryonic development, wound repair, and collateral vascular generation in the myocardium. It is also central to a number of 10 pathological conditions such as tumor growth and metastasis, diabetic retinopathy, and macular degeneration. The process begins with the activation of existing vascular endothelial cells in response to a variety of cytokines and growth factors. The activated 15 endothelial cells secrete enzymes that degrade the
basement membrane of the vessels. The endothelial cells then proliferate and migrate into the extracellular matrix first forming tubules and subsequently new blood vessels. 20 Under normal conditions, endothelial cell
proliferation is a very slow process, but it increases for a short^period of time during embryogenesis, ovulation and wound healing. This temporary increase in cell turnover is governed by a combination of a number of 25 growth stimulatory factors and growth suppressing factors. In pathological angiogenesis, this normal balance is disrupted resulting in continued increased endothelial cell proliferation. Some of the pro-angiogenic factors that have been identified include 30 basic fibroblast growth factor (bFGF), angiogenin, TGF-alpha, TGF-beta, and vascular endothelium growth factor (VEGF), while interferon-alpha, interferon-beta and thrombospondin are examples of angiogenesis suppressors. Angiogenic factors interact with endothelial cell 35 surface receptors such as the receptor tyrosine kinases EGFR, FGFR, PDGFR, Flk-1/KDR, Flt-1, Tek, Tie, neuropilin-1, endoglin, endosialin, and Axl. The receptors Flk-1/KDR, neuropilin-1, and Flt-1 recognize VEGF and these interactions play key roles in VEGF-


induced angiogenesis. The Tie subfamily of receptor tyrosine kinases are also expressed prominently during blood vessel formation.
The proliferation and migration of endothelial cells 5 in the extracellular matrix is mediated by interaction with a variety of cell adhesion molecules. Integrins are a diverse family of heterodimeric cell surface receptors by which endothelial cells attach to the extracellular matrix, each other and other cells. Angiogenesis induced
10 by bFGF or TNF-alpha depend on the agency of the integrin avb3, while angiogenesis induced by VEGF depends on the integrin avb5 (Cheresh et. al., Science, 1995, 270, 1500-2). Induction of expression of the integrins albl and a2bl on the endothelial cell surface is another important
15 mechanism by which VEGF promotes angiogenesis (Senger, et. al.. Proc. Natl. Acad, Sci USA, 1997, 94, 13612-7).
The pharmaceuticals of the present invention are comprised of a non-peptide targeting moiety for the vitronectin receptor that is expressed or upregulated in
20 angiogenic tumor vasculature.
The ultrasound contrast agents of the present invention comprise a plurality of vitronectin receptor targeting moieties attached to or incorporated into a
25 microbubble of a biocompatible gas, a liquid carrier, and a surfactant microsphere, further comprising an optional linking moiety, Ln, between the targeting moieties and the microbubble. In this context, the term liquid carrier means aqueous solution and the term surfactant
30 means any amphiphilic material which produces a reduction in interfacial tension in a solution. A list of suitable surfactants for forming surfactant microspheres is disclosed in EP0727225A2, herein incorporated by reference. The term surfactant microsphere includes
35 nanospheres, liposomes, vesicles and the like. The
biocompatible gas can be air, or a fluorocarbon, such as a C3-C5 perfluoroalkane, which provides the difference in echogenicity and thus the contrast in ultrasound imaging.

The gas is encapsulated or contained in the microsphere to which is attached the biodirecting group, optionally via a linking group. The attachment can be covalent, ionic or by van der Waals forces. Specific examples of 5 such contrast agents include lipid encapsulated
perfluorocarbons with a plurality of tumor neovasculature receptor binding peptides, polypeptides or peptidomimetics.
X-ray contrast agents of the present invention are 10 comprised of one or more vitronectin receptor targeting moieties attached to one or more X-ray absorbing or "heavy" atoms of atomic number 20 or greater, further comprising an optional linking moiety, Ln, between the targeting moieties and the X-ray absorbing atoms. The 15 frequently used heavy atom in X-ray contrast agents is iodine. Recently, X-ray contrast agents comprised of metal chelates (Wallace, R., U.S. 5,417,959) and polychelates comprised of a plurality of metal ions (Love, D., U.S. 5,679,810) have been disclosed. More 20 recently, multinuclear cluster complexes have been
disclosed as X-ray contrast agents (U.S. 5,804,161, PCT WO91/14460, *and PCT WO 92/17215).
MRI contrast agents of the present invention are comprised of one or more vitronectin receptor targeting 25 moieties..attachedjto one or more paramagnetic metal ions, further comprising an optional linking moiety, Ln, between the targeting moieties and the paramagnetic metal ions. The paramagnetic metal ions are present in the form of metal complexes or metal oxide particles. U.S. 30 5,412,148, and 5,760,191, describe examples of chelators for paramagnetic metal ions for use in MRI contrast agents. US. 5,801,228, U.S. 5,567,411, and U.S. 5,281,704, describe examples of polychelants useful for complexing more than one paramagnetic metal ion for use 35 in MRI contrast agents. U.S. 5,520,904, describes
particulate compositions comprised of paramagnetic metal ions for use as MRI contrast agents.


The pharmaceuticals of the present invention have the formulae, (Q) The pharmaceuticals of the present invention can be synthesized by several approaches. One approach involves the synthesis of the targeting non-peptide moiety, Q, and 20 direct attachment of one or more moieties, Q, to one or more metal chelators or bonding moieties, Ch, or to a paramagnetic metal ion or heavy atom containing solid particle, or to an echogenic gas microbubble. Another approach involves the attachment of one-er more moi-et-ies, 25 Q, to the linking group, Ln, which is then attached to one or more metal chelators or bonding moieties, Ch, or to a paramagnetic metal ion or heavy atom containing solid particle, or to an echogenic gas microbubble. Another approach involves the synthesis of a non-peptide, 30 Q, bearing a fragment of the linking group, Ln, one or more of which are then attached to the remainder of the linking group and then to one or more metal chelators or bonding moieties, C^, or to a paramagnetic metal ion or heavy atom containing solid particle, or to an echogenic 35 gas microbubble.
The non-peptide vitronectin binding moieties, Q, optionally bearing a linking group, Ln, or a fragment of

the linking group, can be synthesized using standard synthetic methods known to those skilled in the art. Preferred methods include but are not limited to those methods described below. 5 The attachment of linking groups, Ln, to the non-peptides, Q; chelators or bonding units, Ch, to the non-peptides, Q, or to the linking groups, Ln; and non-peptides, bearing a fragment of the linking group to the remainder of the linking group, in combination forming 10 the moiety, (O)d-Ln' and then to the moiety Ch; can all be performed by standard techniques. These include, but are not limited to, amidation, esterification, alkylation, and the formation of ureas or thioureas. Procedures for performing these attachments can be found in Brinkley, 15 M., Bioconjugate Chemistry 1992, 3(1), which is incorporated herein by reference.
A number of methods can be used to attach the non-peptides, Q, to paramagnetic metal ion or heavy atom containing solid particles, X2, by one of skill in the 20 art of the surface modification of solid particles. In general, the targeting moiety Q or the combination (Q)dLn is attached ^o a coupling group that react with a constituent of the surface of the solid particle. The coupling groups can be any of a number of silanes which 25 react" with "surface"" hydroxy1 groups "on" the" solid particle surface, as described in co-pending United States Patent Application Serial No. 09/356,178, and can also include polyphosphonates, polycarboxylates, polyphosphates or mixtures thereof which couple with the surface of the 30 solid particles, as described in U.S. 5,520,904.
A number of reaction schemes can be used to attach the non-peptides, Q, to the surfactant microsphere, X3. These are illustrated in following reaction schemes where Sf represents a surfactant moiety that forms the 35 surfactant microsphere.
Acylation Reaction:


Sf-C(=0)-Y+ Q-NH2 or > Sf-C(=0)-NH-Q
Q-OH or Sf-C(=0)-0-Q
Y is a leaving group or active ester
5 Disulfide Coupling:
Sf-SH + Q-SH > Sf-S-S-Q
Sulfonamide Coupling: 10
Sf-S{=0)2-Y + Q-NH2 > Sf-S(=0)2-NH-
0
Reductive Amidation: 15
Sf-CHO + Q-NH2 > Sf-NH-Q
In these reaction schemes, the substituents Sf and 0 can
20 be reversed as well.
The linking group Ln can serve several roles. First it provides a spacing group between the metal chelator or bonding moiety, C^, the paramagnetic metal ion or heavy atom containing solid particle, X2, and the surfactant
25 microsphere, X3, and the one or more of the non-peptides, Q, so as to minimize the possibility that the moieties Ch-X, Ch-X1, X2, and X3, will interfere with the interaction of the recognition sequences of Q with angiogenic tumor vasculature receptors. The necessity of
30 incorporating a linking group in a reagent is dependent on the identity of Q, Ch-X, Ch-X1, X2, and X3. If Ch-X, Ch-X1, X2, and X3, cannot be attached to Q without substantially diminishing its affinity for the receptors, then a linking group is used. A linking group also
35 provides a means of independently attaching multiple non-peptides, Q, to one group that is attached to Ch-X, Ch-X1, X2, or X3.


The linking group also provides a means of incorporating a pharmacokinetic modifier into the pharmaceuticals of the present invention. The pharmacokinetic modifier serves to direct the 5 biodistibution of the injected pharmaceutical other than by the interaction of the targeting moieties, Q, with the vitronectin receptors expressed in the tumor neovasculature. A wide variety of functional groups can serve as pharmacokinetic modifiers, including, but not 10 limited to, carbohydrates, polyalkylene glycols, peptides or other polyamino acids, and cyclodextrins. The modifiers can be used to enhance or decrease hydrophilicity and to enhance or decrease the rate of blood clearance. The modifiers can also be used to 15 direct the route of elimination of the pharmaceuticals. Preferred pharmacokinetic modifiers are those that result in moderate to fast blood clearance and enhanced renal excretion.
The metal chelator or bonding moiety, Ch, is 20 selected to form stable complexes with the metal ion chosen for the particular application. Chelators or bonding moieties for diagnostic radiopharmaceuticals are selected to form stable complexes with the radioisotopes that have imageable gamma ray or positron emissions, such 25 as 99mIc, 9STc^_iiixa/_62.Cu, ^i°.Cu, 64Cu, 67£a,- 8Ga_A6Y_ Chelators for technetium, copper and gallium isotopes are selected from diaminedithiols, monoamine-monoamidedithiols, triamide-monothiols, monoamine-diamide-monothiols, diaminedioximes, and 30 hydrazines. The chelators are generally tetradentate with donor atoms selected from nitrogen, oxygen and sulfur. Preferred reagents are comprised of chelators having amine nitrogen and thiol sulfur donor atoms and hydrazine bonding units. The thiol sulfur atoms and the 35 hydrazines may bear a protecting group which can be
displaced either prior to using the reagent to synthesize a radiopharmaceutical or preferably in situ during the synthesis of the radiopharmaceutical.

Exemplary thiol protecting groups include those listed in Greene and Wuts, "Protective: Groups in Organic Synthesis" John Wiley & Sons, New York (1991), the disclosure of which is hereby incorporated by reference. 5 Any thiol protecting group known in the art can be used. Examples of thiol protecting groups include, but are not limited to, the following: acetamidomethyl. benzamidomethyl, 1-ethoxyethyl, benzoyl, and triphenylmethyl. 10 Exemplary protecting groups for hydrazine bonding units are hydrazones which can be aldehyde or ketone hydrazones having substituents selected from hydrogen, alkyl, aryl and heterocycle. Particularly preferred hydrazones are described in co-pending U.S.S.N. 15 08/476,296 the disclosure of which is herein incorporated by reference in its entirety.
The hydrazine bonding unit when bound to a metal radionuclide is termed a hydrazido, or diazenido group and serves as the point of attachment of the radionuclide 20 to the remainder of the radiopharmaceutical. A diazenido group can be either terminal (only one atom of the group is bound to^the radionuclide) or chelating. In order to have a chelating diazenido group at least one other atom of the group must also be bound to the radionuclide. The 25 atoms bound to the metal are termed donor atoms.
Chelators for inIn and 86Y are selected from cyclic and acyclic polyaminocarboxylates such as DTPA, DOTA, D03A, 2-benzyl-DOTA, alpha-(2-phenethyl)1,4,7,10-tetraazazcyclododecane-l-acetic-4,7,10-30 tris(methylacetic)acid, 2-benzyl-
cyclohexyldiethylenetriaminepentaacetic acid, 2-benzyl-6-methyl-DTPA, and 6,6"-bis[N,N,N*,N"-tetra(carboxymethyl)aminomethyl)-4'-(3-amino-4-methoxyphenyl)-2,2':6',2"-terpyridine. Procedures for 35 synthesizing these chelators that are not commercially available can be found in Brechbiel, M. and Gansow, 0., J. Chem. Soc. Perkin Trans. 1992, 1, 1175; Brechbiel, M. and Gansow, O., Bioconjugate Chem. 1991, 2, 187; Deshpande, S., et. al. , J. Nucl. Med. 1990, 31, 473;


Kruper, J., U.S. Patent 5,064,956, and Toner, J., U.S. Patent 4,859,777, the disclosures of which are hereby incorporated by reference in their entirety.
The coordination sphere of metal ion includes all 5 the ligands or groups bound to the metal. For a
transition metal radionuclide to be stable it typically has a coordination number (number of donor atoms) comprised of an integer greater than or equal to 4 and less than or equal to 8; that is there are 4 to 8 atoms 10 bound to the metal and it is said to have a complete
coordination sphere. The requisite coordination number for a stable radionuclide complex is determined by the identity of the radionuclide, its oxidation state, and the type of donor atoms. If the chelator or bonding unit 15 does not provide all of the atoms necessary to stabilize the metal radionuclide by completing its coordination sphere, the coordination sphere is completed by donor atoms from other ligands, termed ancillary or co-ligands, which can also be either terminal or chelating. 20 A large number of ligands can serve as ancillary or co-ligands, the choice of which is determined by a variety of considerations such as the ease of synthesis of the radiopharmaceutical, the chemical and physical properties of the ancillary ligand, the rate of
2^ formation, the yield, and the number of_isomeric..Jroxms of
the resulting radiopharmaceuticals, the ability to administer said ancillary or co-ligand to a patient without adverse physiological consequences to said patient, and the compatibility of the ligand in a 30 lyophil-ized kit formulation. The charge and
lipophilicity of the ancillary ligand will effect the charge and lipophilicity of the radiopharmaceuticals. For example, the use of 4,5-dihydroxy-l,3-benzene disulfonate results in radiopharmaceuticals with an 35 additional two anionic groups because the sulfonate groups will be anionic under physiological conditions. The use of N-alkyl substituted 3,4-hydroxypyridinones results in radiopharmaceuticals with varying degrees of


lipophilicity depending on the size of the alkyl substituents.
Preferred technetium radiopharmaceuticals of the present invention are comprised of a hydrazido or 5 diazenido bonding unit and an ancillary ligand, ALI, or a bonding unit and two types of ancillary ALI and AL2> or a tetradentate chelator comprised of two nitrogen and two sulfur atoms. Ancillary ligands ALI are comprised of two or more hard donor atoms such as oxygen and amine 10 nitrogen (sp3 hybridized). The donor atoms occupy at
least two of the sites in the coordination sphere of the radionuclide metal; the ancillary ligand ALI serves as one of the three ligands in the ternary ligand system. Examples of ancillary ligands ALI include but are not 15 limited to dioxygen ligands and functionalized
aminocarboxylates. A large number of such ligands are available from commercial sources.
Ancillary dioxygen ligands include ligands that coordinate to the metal ion through at least two oxygen 20 donor atoms. Examples include but are not limited to:
glucoheptonate, gluconate, 2-hydroxyisobutyrate, lactate, tartrate, mannitol, glucarate, maltol, Kojic acid, 2, 2-bis (hydroxymethyl) propionic acid,
4,5-dihydroxy-l,3-benzene disulfonate, or substituted or 25 uri sub'stitutelTl,~2 or 3,T fiydroxypyrfclin'onesT "The names for the ligands in these examples refer to either the protonated or non-protonated forms of the ligands.)
Functionalized aminocarboxylates include ligands that have a combination of amine nitrogen and oxygen 30 donor atoms. Examples include but are not limited to: iminodiacetic acid, 2,3-diaminopropionic acid, nitrilotriacetic acid, N,N'-ethylenediamine diacetic acid, N,N,N'-ethylenediamine triacetic acid, hydroxyethylethylenediamine triacetic acid, and 35 N,N"-ethylenediamine bis-hydroxyphenylglycine. (The names for the ligands in these examples refer to either the protonated or non-protonated forms of the ligands.)

A series of functionalized aminocarboxylates are disclosed by Bridger et. al. in U.S. Patent 5,350,837, herein incorporated by reference, that result in improved rates of formation of technetium labeled hydrazino 5 modified proteins. We have determined that certain of these aminocarboxylates result in improved yields of the radiopharmaceuticals of the present invention. The preferred ancillary ligands ALI functionalized aminocarboxylates that are derivatives of glycine; the 10 most preferred is tricine
(tris(hydroxymethylImethylglycine).
The most preferred technetium radiopharmaceuticals of the present invention are comprised of a hydrazido or diazenido bonding unit and two types of ancillary 15 designated ALI and AL2- or a diaminedithiol chelator. The second type of ancillary ligands AL2 are comprised of one or more soft donor atoms selected from the group: phosphine phosphorus, arsine arsenic, imine nitrogen (sp2 hybridized), sulfur (sp2 hybridized) and carbon (sp 20 hybridized); atoms which have p-acid character. Ligands AL2 can be monodentate, bidentate or tridentate, the denticity is defined by the number of donor atoms in the ligand.- One of the two donor atoms in a bidentate ligand and one of the three donor atoms in a tridentate ligand 25 must be a soft-donor atomT~ We"ha~ve aisc"lose'a~irT
co-pending U.S.S.N. 08/415,908, and U.S.S.N. 60/013360 and 08/646,886, the disclosures of which are herein incorporated by reference in their entirety, that radiopharmaceuticals comprised of one or more ancillary 30 or co-ligands AL2 are more stable compared to
radiopharmaceuticals that are not comprised of one or more ancillary ligands, AL2,* that is, they have a minimal number of isomeric forms, the relative ratios of which do not change significantly with time, and that remain 35 substantially intact upon dilution.
The ligands AL2 that are comprised of phosphine or arsine donor atoms are trisubstituted phosphines, trisubstituted arsines, tetrasubstituted diphosphines and


tetrasubstituted diarsines. The ligands AL2 that are comprised of imine nitrogen are unsaturated or aromatic nitrogen-containind, 5 or 6-membered heterocycles. The ligands that are comprised of sulfur (sp2 hybridized) 5 donor atoms are thiocarbonyls, comprised of the moiety C=S. The ligands comprised of carbon (sp hybridized) donor atoms are isonitriles, comprised of the moiety CNR, where R is an organic radical. A large number of such ligands are available from commercial sources.
10 Isonitriles can be synthesized as described in European ?^evx Ql Preferred ancillary ligands AL2 are trisubstituted phosphines and unsaturated or aromatic 5 or 6 membered
15 heterocycles. The most preferred ancillary ligands AL2
are trisubstituted phosphines and unsaturated 5 membered
heterocycles.
The ancillary ligands AL2 may be substituted with
alkyl, aryl, alkoxy, heterocycle, aralkyl, alkaryl and
20 arylalkaryl groups and may or may not bear functional
groups comprised of heteroatoms such as oxygen, nitrogen, phosphorus or sulfur- Examples of such functional groups include but are not limited to: hydroxyl, carboxyl, carboxamide, nitro, ether, ketone, amino, ammonium,
25 sulfonate,- sul-fonamide, phosphonate, and—phosphonamide. The functional groups may be chosen to alter the lipophilicity and water solubility of the ligands which may affect the biological properties of the radiopharmaceuticals. such as altering the distribution
30 into non-target tissues, cells or fluids, and the mechanism and rate Chelators or bending moieties for therapeutic radiopharmaceuticals are selected to form stable complexes with the radioisotopes that have alpha
35 particle, beta particle, Auger or Coster-Kronig electron emissions, such as l86Re, a88Re, 153Sm, 166Ho, 177Lu, 149Pm, 90Y, 212Bi# 103Pd, 109pd, l59Gd, 140La, 198Au, 199Au, 169Yb, 175YD/ 165^, 166Dy( 67CU( 105Rh/ lllAg, and 192Ir.

Chelators for rhenium, copper, palladium, platinum, iridium, rhodium, silver and gold isotopes are selected from diaminedithiols, monoamine-monoamidedithiols, triamide-monothiols, monoamine-diamide-monothiols, 5 diaminedioximes, and hydrazines. Chelators for yttrium, bismuth, and the lanthanide isotopes are selected from cyclic and acyclic polyaminocarboxylates such as DTPA, DOTA, D03A, 2-benzyl-DOTA, alpha-(2-phenethyl)1,4,7,10-tetraazacyclododecane-l-acetic-4, 7,10-10 tris(methylacetic)acid, 2-benzyl-
cyclohexyldiethylenetriaminepentaacetic acid, 2-benzyl-6-methyl-DTPA, and 6,6"-bis(N,N,N",N"-tetra(carboxymethyl)aminomethyl)-4'-(3-amino-4-methoxyphenyl)-2,2■:6',2"-terpyridine. 15 Chelators for magnetic resonance imaging contrast agents are selected to form stable complexes with paramagnetic metal ions, such as Gd(III), Dy(III), Fe(III), and Mn(II), are selected from cyclic and acyclic polyaminocarboxylates such as DTPA, DOTA, D03A, 20 2-benzyl-DOTA, alpha-(2-phenethyl)1,4,7,10-tetraazacyclododecane-l-acetic-4,7,10-tris(methylaeetic)acid, 2-benzyl-
cyclohexyldiethylenetriaminepentaacetic acid, 2-benzyl-6-methyl-DTPA, and 6,6"-bis[N,N,N",NB-25 tetra(carboxymethyl)aminomethyl)-4'-(3-amino-4-methoxyphenyl)-2,2':6',2"-terpyridine.
The technetium and rhenium radiopharmaceuticals of the present invention comprised of a hydrazido or diazenido bonding unit can be easily prepared by admixing 30 a salt of a radionuclide, a reagent of the present
invention, an ancillary ligand A^i, an ancillary ligand AL2» and a reducing agent, in an aqueous solution at temperatures from 0 to 100 °C. The technetium and rhenium radiopharmaceuticals of the present invention 35 comprised of a tetradentate chelator having two nitrogen and two sulfur atoms can be easily prepared by admixing a salt of a radionuclide, a reagent of the present

invention, and a reducing agent, in an aqueous solution at temperatures from 0 to 100 °C.
When the bonding unit in the reagent of the present invention is present as a hydrazone group, then it must 5 first be converted to a hydrazine, which may or may not be protonated, prior to complexation with the metal radionuclide. The conversion of the hydrazone group to the hydrazine can occur either prior to reaction with the radionuclide, in which case the radionuclide and the 10 ancillary or cc-ligand or ligands are combined not with the reagent but with a hydrolyzed form of the reagent bearing the chelator or bonding unit, or in the presence of the radionuclide in which case the reagent itself is combined with the radionuclide and the ancillary or 15 co-ligand or ligands. In the latter case, the pH of the reaction mixture must be neutral or acidic.
Alternatively, the radiopharmaceuticals of the present invention comprised of a hydrazido or diazenido bonding unit can be prepared by first admixing a salt of 20 a radionuclide, an ancillary ligand ALI, and a reducing agent in an aqueous solution at temperatures from 0 to 100 °C to form an intermediate radionuclide complex with the ancillary ligand ALI then adding a reagent of the present invention and an ancillary ligand AL2 and reacting 25 further at temperatures from 0 to 100 °C.
Alternatively, the radiopharmaceuticals of the present invention comprised of a hydrazido or diazenido bonding unit can be prepared by first admixing a salt of a radionuclide, an ancillary ligand ALI, a reagent of the 30 present invention, and a reducing agent in an aqueous solution at temperatures from 0 to 100 °C to form an intermediate radionuclide complex, and then adding an ancillary ligand AL2 and reacting further at temperatures from 0 to 100 °C. 35 The technetium and rhenium radionuclides are preferably in the chemical form of pertechnetate or perrhenate and a pharmaceutically acceptable cation. The pertechnetate salt form is preferably sodium

pertechnetate such as obtained from commercial Tc-99m generators. The amount of pertechnetate used to prepare the radiopharmaceuticals of the present invention can range from 0.1 mCi to 1 Ci, or more preferably from 1 to 5 200 mCi.
The amount of the reagent of the present invention used to prepare the technetium and rhenium radiopharmaceuticals of the present invention can range from 0.01 ug to 10 mg, or more preferably from 0.5 ug to 10 200 ug. The amount used will be dictated by the amounts of the other reactants and the identity of the radiopharmaceuticals of the present invention to be prepared.
The amounts of the ancillary ligands ALI used can 15 range from 0.1 mg to 1 g, or more preferably from 1 mg to 100 mg. The exact amount for a particular radiopharmaceutical is a function of identity of the radiopharmaceuticals of the present invention to be prepared, the procedure used and the amounts and 20 identities of the other reactants. Too large an amount of ALI will result in the formation of by-products comprised o^technetium labeled ALI without a biologically active molecule or by-products comprised of technetium labeled biologically active molecules with the ancillary 25 ligand ALI but without the ancillary ligand AL2 • Too small an amount of ALI will result in other by-products such as technetium labeled biologically active molecules with the ancillary ligand AL2 but without the ancillary ligand ALi, or reduced hydrolyzed technetium, or 30 technetium colloid.
The amounts of the ancillary ligands AL2 used can range from 0.001 mg to 1 g, or more preferably from 0.01 mg to 10 mg. The exact amount for a particular radiopharmaceutical is a function of the identity of the 35 radiopharmaceuticals of the present invention to be prepared, the procedure used and the amounts and identities of the other reactants. Too large an amount of AL2 will result in the formation of by-products


comprised of technetium labeled AL2 without a biologically active molecule or by-products comprised of technetium labeled biologically active molecules with the ancillary ligand AL2 but without the ancillary ligand ALI . If the 5 reagent bears one or more substituents that are comprised of a soft donor atom, as defined above, at least a ten-fold molar excess of the ancillary ligand AL2 to the reagent of formula 2 is required to prevent the substituent from interfering with the coordination of the 10 ancillary ligand AL2 to the metal radionuclide.
Suitable reducing agents for the synthesis of the radiopharmaceuticals of the present invention include stannous salts, dithionite or bisulfite salts, borohydride salts, and formamidinesulfinic acid, wherein 15 the salts are of any pharmaceutical^ acceptable form. The preferred reducing agent is a stannous salt- The amount of a reducing agent used can range from 0.001 mg to 10 mg, or more preferably from 0.005 mg to 1 mg.
The specific structure of a radiopharmaceutical of 20 the present invention comprised of a hydrazido or
diazenido bonding unit will depend on the identity of the reagent of tne present invention used, the identity of any ancillary ligand ALI, the identity of any ancillary ligand AL2- and the identity of the radionuclide. 25 Radiopharmaceuticals comprised of a hydrazido or
diazenido bonding unit synthesized using concentrations of reagents of 1 mg/mL concentrations will be comprised of two hydrazido 30 or diazenido groups from two reagent molecules. For most applications, only a limited amount of the biologically active molecule can be injected and not result in undesired side-effects, such as chemical toxicity, interference with a biological process or an altered 35 biodistribution of the radiopharmaceutical. Therefore, the radiopharmaceuticals which require higher concentrations of the reagents comprised in part of the


biologically active molecule, will have to be diluted or purified after synthesis to avoid such side-effects.
The identities and amounts used of the ancillary ligands ALI and AL2 will determine the values of the 5 variables y and z. The values of y and z can
independently be an integer from 1 to 2 . In combination, the values of y and z will result in a technetium coordination sphere that is made up of at least five and no more than seven donor atoms. For monodentate 10 ancillary ligands hu2- 2 can be an integer from 1 to 2;
for bidentate or tridentate ancillary ligands &U2, z is 1. The preferred combination for monodentate ligands is y equal to 1 or 2 and z equal to 1. The preferred combination for bidentate or tridentate ligands is y 15 equal to 1 and z equal to 1.
The indium, copper, gallium, silver, palladium, rhodium, gold, platinum, bismuth, yttrium and lanthanide radiopharmaceuticals of the present invention can be easily prepared by admixing a salt of a radionuclide and 20 a reagent of the present invention, in an aqueous solution at temperatures from 0 to 100 °C. These radionuclide's are typically obtained as a dilute aqueous solution in a mineral acid, such as hydrochloric, nitric or sulfuric acid. The radionuclides are combined with 25 from one to about one thousand equivalents of the
reagents of the present invention dissolved in aqueous solution. A buffer is typically used to maintain the pH of the reaction mixture between 3 and 10.
The gadolinium, dysprosium, iron and manganese 30 metallopharmaceuticals of the present invention can be easily prepared by admixing a salt of the paramagnetic metal ion and a reagent of the present invention, in an aqueous solution at temperatures from 0 to 100 °C. These paramagnetic metal ions are typically obtained as a 35 dilute aqueous solution in a mineral acid, such as
hydrochloric, nitric or sulfuric acid. The paramagnetic metal ions are combined with from one to about one thousand equivalents of the reagents of the present

invention dissolved in aqueous solution. A buffer is typically used to maintain the pH of the reaction mixture between 3 and 10.
The total time of preparation will vary depending on 5 the identity of the metal ion, the identities and amounts of the reactants and the procedure used for the preparation. The preparations may be complete, resulting in > 80% yield of the radiopharmaceutical, in 1 minute or may require more time. If higher purity 10 metallopharmaceuticals are needed or desired, the
products can be purified by any of a number of techniques well known to those skilled in the art such as liquid chromatography, solid phase extraction, solvent extraction, dialysis or ultrafiltration. 15 Buffers useful in the preparation of
metallopharmaceuticals and in diagnostic kits useful for the preparation of said radiopharmaceuticals include but are not limited to phosphate, citrate, sulfosalicylate, and acetate. A more complete list can be found in the 20 United States Pharmacopeia.
Lyophilization aids useful in the preparation of diagnostic Kits useful for the preparation of radiopharmaceuticals include but are not limited to mannitol, lactose, sorbitol, dextran, Ficoll, and 25 polyvinylpyrrolidine(PVP).
Stabilization aids useful in the preparation of metallopharmaceuticals and in diagnostic kits useful for the preparation of radiopharmaceuticals include but are not limited to ascorbic acid, cysteine, monothioglycerol, 30 sodium bisulfite, sodium metabisulfite, gentisic acid, and inositol.
Solubilization aids useful in the preparation of metallopharmaceuticals and in diagnostic kits useful for the preparation of radiopharmaceuticals include but are 35 not limited to ethanol, glycerin, polyethylene glycol, propylene glycol, polyoxyethylene sorbitan monooleate, sorbitan monoloeate, polysorbates, poly(oxyethylene)poly(oxypropylene)poly(oxyethylene)


block copolymers (Pluronics) and lecithin. Preferred solubilizing aids are polyethylene glycol, and Pluronics.
Bacteriostats useful in the preparation of metallopharmaceuticals and in diagnostic kits useful for 5 the preparation of radiopharmaceuticals include but are not limited to benzyl alcohol, benzalkonium chloride, chlorbutanol, and methyl, propyl or butyl paraben.
A component in a diagnostic kit can also serve more than one function. A reducing agent can also serve as a 10 stabilization aid, a buffer can also serve as a transfer ligand, a lyophilization aid can also serve as a transfer, ancillary or co-ligand and so forth.
The diagnostic radiopharmaceuticals are administered by intravenous injection, usually in saline solution, at 15 a dose of 1 to 100 mCi per 70 kg body weight, or preferably at a dose of 5 to 50 mCi. Imaging is performed using known procedures.
The therapeutic radiopharmaceuticals are administered by intravenous injection, usually in saline 20 solution, at a dose of 0.1 to 100 mCi per 70 kg body
weight, or preferably at a dose of 0.5 to 5 mCi per 70 kg body weight.^
The magnetic resonance imaging contrast agents of the present invention may be used in a similar manner as 25 other MRI agents as described in U.S. Patent 5,155,215; U.S. Patent 5,087,440; Margerstadt et al., Magn. Reson. Med., 1986, 3, 808; Runge et al., Radiology, 1988, 166, 835; and Bousquet et al., Radiology, 1988, 166, 693. Generally, sterile aqueous solutions of the contrast 30 agents are administered to a patient intravenously in dosages ranging from 0.01 to 1.0 mmoles per kg body weight.
For use as X-ray contrast agents, the compositions of the present invention should generally have a heavy 35 atom concentration of 1 mM to 5 M, preferably 0.1 M to 2 M. Dosages, administered by intravenous injection, will typically range from 0.5 mmol/kg to 1.5 mmol/kg, preferably 0.8 mmol/kg to 1.2 mmol/kg. Imaging is


performed using known techniques, preferably X-ray computed tomography.
The ultrasound contrast agents of the present invention are administered by intravenous injection in an 5 amount of 10 to 3 0 uL of the echogenic gas per kg body weight or by infusion at a rate of approximately 3 pL/kg/min. Imaging is performed using known techniques of sonography.
Other features of the invention will become apparent 10 in the course of the following descriptions of exemplary embodiments which are given for illustration of the invention and are not intended to be limiting thereof.
EXAMPLES
15 Representative materials and methods that may be
used in preparing the compounds of the invention are described further below.
l-methyl-4-oxo-7- ( ( (i- (triphenylmethyl) imidazol-2-yl)amino)methyl)hydroquinoline-3-carboxylic acid, ethyl
20 7-bromo-4-oxohydroquinoline-3-carboxylate, 1-
(triphenylmethyl)imidazole-2-ylamine, and methyl 3-amino-2- ( ( (2, 4, 6-trimethylphenyl)sulfonyl)amino)propanoate hydrochloride were prepared as described in PCT WO 98/23608. Boc-L-cysteic acid, Boc-L-cysteic acid N-
25 hydroxyphenyl ester, and Boc-L-cysteic acid p-nitrophenyl ester were prepared as described in Liebigs Ann. Chem. 1979, 776-783. Benzotriazole-1-yloxy-tris-pyrrolidinophosphonium hexafluorophosphate (PyBOP) was purchased from Novabiochem.
30 (tert-butoxy)-N-(3-bromopropyl) formamide and 2-(2-aza-2-((5-((2,5-dioxopyrrolidinyl)carbonyl) (2-pyridyl) )-amino)vinyl)benzenesulfonic acid were prepared as described in PCT WO 96/40637. All other chemicals and solvents (reagent grade) were used as supplied from the
35 vendors cited without further purification. t-
Butyloxycarbonyl (Boc) amino acids and other starting amino acids may be obtained commercially from Bachem Inc., Bachem Biosciences Inc. (Philadelphia, PA), Advanced ChemTech (Louisville, KY) , Peninsula


Laboratories (Belmont, CA), or Sigma (St. Louis, MO). 2-(lH-Benzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HBTU) and TBTU were purchased from Advanced ChemTech. N-methylmorpholine (NMM), m-cresol, 5 D-2-aminobutyric acid (Abu), trimethylacetylchloride, diisopropylethylamine (DIEA), 1,2,4-triazole, stannous chloride dihydrate, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDO, triethylsilane (Et3SiH) and tris(3~sulfonatophenyl)phosphine trisodium
10 salt (TPPTS) were purchased from Aldrich Chemical
Company. Bis(3-sulfonatophenyl)phenylphosphine disodium salt (TPPDS) was prepared by the published procedure (Kuntz, £., U.S. Patent 4,248,802). (3-SulfonatophenyUdiphenylphosphine monosodium salt
15 (TPPMS)was purchased from TCI America, Inc. Tricine was obtained from Research Organics, Inc. Technetium-99m-pertechnetate (99mTcC>4~) was obtained from a DuPont Pharma
"Mo/99mTc Technelite® generator. In-111-chloride (Indichlor®) was obtained from Amersham Medi-Physics,
20 Inc. Sm-153-chloride and Lutetium-177-chloride were
obtained from the University of Missouri Research Reactor (MURR). Yttr^um-90 chloride was obtained from the Pacific Northwest Research Laboratories. Dimethylformamide (DMF) , ethyl acetate, chloroform (CHCI3), methanol
25 (MeOH), pyridine and hydrochloric acid (HC1) were obtained from Baker. Acetonitrile, dichloromethane (DCM), acetic acid (HOAc), trifluoroacetic acid (TFA), ethyl ether, triethylamine, acetone, and magnesium sulfate were commercially obtained. Absolute ethanol was
30 obtained from Quantum Chemical Corporation.
Synthesis of Boc-Glu-(OTFP)-OTFP 35

To a solution of Boc-Glu-OH (28.9 g, 117 mmol) in DMF (500 mL) at room temperature, and under nitrogen, was 5 added a solution of ,3,5,6-tetrafluorophenol (48.2 g,
290 mmol) in DMF (50 mL). After stirring for 10 min. EDC (55.6 g, 290 mmol) was added and the reaction mixture was stirred for about 96 h. The volatiles were removed in vacuo and the residue was triturated in 0.1 N HC1 (750 10 mL). To this mixture was added ethyl acetate (600 mL), the layers separated. The aqueous layer was extracted with ethyl acetate (3 x -500 mL), and all the ethyl acetate fractions were combined, washed with water (300
mL) and brine (300 mL) , dried (MgS04), and concentrated to
15 give a tan solid (62 g). The tan solid was washed with acetonitrile to give the title compound (45.5 g, 73%) in purified form.
ESMS: Calculated for C22H17FsN06, 543.09; found, 566.0
[M+Na]*1. 20
*" Example 1
2-(((4-(4-(((3-(2-(2-(3-((6-((l-Aza-2-(2-sulfophenyl)vinyl)amino)(3-pyridyl))carbonylamino)-
propoxy)et hoxy)-
25 ethoxy)propyl)amino)sulfonyl)phenyl)phenyl)sulfonyl)-
amino)-3-((7-((imidazol-2-ylamino)methyl)-l-methyl-4-
oxo(3-hydroguinolyl))carbonylamino)propanoic Acid
Trifluoroacetate Salt
30 Part A - N-(3-(2-(2-(3-aminopropoxy)ethoxy)ethoxy)-propyl)(phenylmethoxy)formamide
A solution of 4,7,10-trioxa-l,13-tridecanediamine (158 mL, 0.72 mol), TEA (16.7 mL, 0.12 mol), and MeOH (3 00 mL) in peroxide-free THF (1,000 mL) was placed in a
35 3 liter 3-neck flask fitted with a mechanical stirrer, a thermometer, and an addition funnel with nitrogen line. The addition funnel was charged with a solution of benzyl chloroformate (17.1 mL, 0.12 mol) in peroxide-free THF

(1,000 mL). The contents of the flask were cooled below 5 °C. The contents of the addition funnel were added to the flask with rapid stirring over 4 h while keeping the temperature below 5 °C. The solution was stirred an 5 additional 3 0 min and concentrated to give a thick syrup. This syrup was taken up in saturated NaCl (1800 mL) and 10% Na2C03 (200 mL) and extracted with ether (3 x 1,000 mL). The combined ether extracts were washed with saturated NaCl (500 mL) , dried (MgSC>4), and concentrated
10 to give a pale yellow oil (36.74 g). Flash
chromatography on a 7 x 29 cm silica gel column (DCM/MeOH/TEA, 20/15/0.5) gave the title compound as a colorless syrup (19.14 g, 45%). !H NMR (CDCI3): 7.33-7.25 (m, 5H), 5.59 (s, 1H), 5.06 (s, 2H) , 3.62-3.45 (m,
15 12H), 3.32-3.25 (m, 2H), 2.74 (t, J = 6.7 Hz, 2H), 1.75 (pentet, J = 6.0 Hz, 2H), 1.67 (pentet, J = 6.4 Hz, 2H) , 1.33 (s, 2H); MS: m/e 355.4 [M+H]; High Resolution MS: Calcd for C18H31N2O5 [M+H]: 355.2233, Found: 355.2222.
20 K O
Part B - Methyl 3- ((tert-Butoxy)carbonylamino) -2- (( (4- (4-
( ( (3-(2-(2-(3-
( (phenylmethoxy) carbonylamino) propoxy) ethoxy) -
25 ethoxy)propyl)amino)sulfonyl)phenyl)phenyl)sulfonyl)-amino)propanoate
Biphenyl-4,4'-disulfonyl chloride (2.64 g, 7.5 mmol, freshly recrystallized from CHCI3) and DCM (200 mL) were placed in a 500 mL 3-neck flask fitted with a
30 thermometer, an addition funnel, and a nitrogen line.
The addition funnel was charged with a solution of N-(3-(2- (2- (3-aminopropoxy) ethoxy) ethoxy)propyl) -(phenylmethoxy) formamide (1.77 g, 5.0 mmol) and DIEA (0.87 mL, 5.0 mmol) in DCM (40 mL) . The contents of the
35 flask were cooled below 5 °C. The contents of the addition funnel were added to the flask with rapid

stirring over 3 h while keeping the temperature of the flask below 5 °C. The addition funnel was charged with a solution of N-(3-Boc-L-cc,p, -diaminopropionic acid methyl ester hydrochloride (2.55 g, 10 mmol) and DIEA (3.8 mL, 5 22 mmol) in DCM (25 mL). This solution was added to the flask with stirring at 5 °C over 15 min, and stirred at ambient temperatures for an additional 20 h. The reaction solution was washed consecutively with 0.1 N HCl (100 mL) and water (2 x 100 mL) , dried (MgSCM), and
10 concentrated to give a viscous oil (5.79 g) . Flash chromatography on a 5 x 21 cm silica gel column (85/15 EtOAc/hexanes, followed by 100% EtOAc) gave a colorless amorphous solid. Recrystallization from toluene (85 mL) gave the title compound as a colorless solid (2.52 g,
15 59%). MP: 104.5-106.5 °C; 2H NMR (CDCI3): 8.00-7.90 (m, 4H), 7.72-7.64 (m, 4H) , 7.46-7.24 (m, 5H), 5.96-5.88 (m, 1H), 5.86-5.73 (m, 1H), 5.41 (s, 1H) , 5.16-5.00 (in, 3H) , 4.15-4.02 (m, 1H), 3.68-3.39 (m, 17H), 3.34-3.22 (m, 2H) , 3.13-3.03 (m, 2H), 1.80-1.62 (m, 4H), 1.39 (s, 9H) ; 13C
20 NMR (CDCI3): 170.2, 156.5, 156.1, 143.9, 143.0, 140.4, 139.4, 136.^, 128.4, 128.1, 128.0, 127.9, 127.9, 127.8, 127.3, 80.1, 70.6, 70.5, 70.2, 70.1, 70.0, 69.6, 66.5, 56.1, 52.9, 43.2, 42.4, 39.3, 29.4, 28.5, 28.2; MS: m/e 868.3 [M+NH4]; High Resolution MS: Calcd for C39H55N4O13S2
25 [M+H]: 851.3207, Found: 851.3226.
Part C - Methyl 3-((l-Methyl-4-oxo-7-(((1-30 (triphenylmethyl) imi dazol - 2-yl) amino) methyl) (3-
hydroquinolyl))carbonylamino)-2-(((4-(4-(((3-(2-(2-(3-
((phenylmethoxy)carbonylamino)-
propoxy) ethoxy) ethoxy) propyl) amino) sulfonyl) phenyl) phenyl
)sulfonyl)amino)propanoate 35 The product of Part B, above (748 mg, 0.88 mmol) was
dissolved in 25/75 TFA/DCM (15 mL) and allowed to stand


at ambient temperatures under nitrogen for 15 min. The TFA was removed under vacuum and the resulting amber oil was taken up in 50/50 ACN/water (50 mL), and treated portion wise with Bio-Rad AG-3-X4A resin, hydroxide form, 5 to raise the pH from 2 to 6. The resin was removed by filtration and the filtrate was lyophilized to give a sticky pale yellow foam.
In a separate flask, l-methyl-4-oxo-7-(((1-(triphenylmethyl)imidazol-2-
10 yl )amino)methyl)hydroquinoline-3-carboxylic acid (432 mg, 0.80 iranol), TEA (0.33 mL), and HBTU (3 64 mg, 0.96 mmol) were dissolved in anhydrous DMF (25 mL). The resulting solution was stirred at ambient temperatures under a nitrogen atmosphere for 10 min and combined with a
15 solution of the yellow foam in anhydrous DMF (15 mL). The DMF was removed under vacuum after 18 h to give a viscous yellow oil. This oil was taken up in EtOAc (175 mL), washed consecutively with water (25 mL), saturated NaHC03 (50 mL), and saturated NaCl (25 mL), dried (MgS04) ,
20 and concentrated to give a viscous yellow oil.
Purification by flash chromatography on a 7 x 25 cm silica gel column using a CHCl3/EtOAc/MeOH step gradient (47/47/6, 46/46/8, 60/30/10) gave the title compound as a pale yellow solid (510 mg, 50%). MP: 136-140 °C; MS: m/e
25 1273.4 [M+H]; High Resolution MS: Calcd for C68H73N8O13S2 [M+H]: 1273.4738, Found: 1273.4730.


30

Part D - 3-( (l-Methyl~4-oxo-7- (((1-(triphenylmethyU-imidazol^-yDaminoJmethyl) (3-
hydroquinolyl)Jcarbonylamino)-2-(((4-(4- ( ( (3-(2-(2-(3-( (phenylmethoxy)carbonylamino)propoxy) ethoxy) -

ethoxy) propyl)amino)sulfonyl)phenyl)phenyl)sulfonyl)-amino)propanoic Acid
The product form Part C, above (295 mg, 0.232 mmol) was dissolved in a mixture of peroxide-free THF (12 mL), 5 water (1.8 mL) , and 3 N LiOH (1.2 mL), and stirred at ambient temperatures under a nitrogen atmosphere for 3 0 min. The THF was removed under vacuum and the resulting mixture was dissolved in CHCI3 (75 mL) and water (50 mL). The aqueous layer was adjusted to pH 3 with 0.5 N HC1 and
10 the layers were thoroughly mixed. The aqueous layer was extracted with additional CHCI3 (2 x 25 mL). The combined CHCI3 extracts were washed with saturated NaCl (50 mL), dried (MgS04), and concentrated to give the title compound as a pale yellow solid (291 mg, 100%) .
15 MS: m/e 1259.3 [M+H]; High Resolution MS: Calcd for C67H71N8O13S2 [M+H]: 1259.4582, Found: 1259.4610.

20 Part E - 2-(((4-(4-(((3-(2-(2-(3~Aminopropoxy)ethoxy)-ethoxy) propyl) amino) sulf onyl) phenyl) phenyl) sulf onyl) -amino)-3-((7-((imidazol-2-yl)methyl)-l-methyl-4-oxo(3~ hydroquinolyl) )carbonylamino) propanoic Acid
The product from Part D, above (279 mg, 0.222 mmol)
25 was dissolved in degassed TFA (30 mL) and treated with Et3SiH (0.424 mL, 2.66 mmol). The solution was heated at 70 °C under a nitrogen atmosphere for 1 h and concentrated to a viscous oil. This oil was dissolved in water (20 mL) and washed with ether (2 x 20 mL). The
30 combined ether washings were back-extracted with water (10 mL) . The combined water extracts were diluted with an equal volume of ACN and treated with Bio-Rad AG-3-X4A resin, hydroxide form to raise the pH from 4 to 6. The resin was removed by filtration and the filtrate was
35 lyophilized to give the title compound as a colorless
solid (220 mg). MS: m/e 883.4 [M+H], 442.5 [M+2H]; High


Resolution MS: Calcd for C40H51N8O11S2 [M+H]: 833.3118, Found: 833.3118.
Part F - 2-(((4-(4-(((3-(2-(2-(3-((6-((l-Aza-2-(2-sulfophenyl)vinyl)amino)(3-pyridyl))carbonylamino)-propoxy)ethoxy)-ethoxy)propyl)amino)sulfonyl)phenyl)phenyl)sulfonyl)-
10 amino)-3-((7-((imidazol-2-ylamino)methyl)-l-methyl-4-oxo(3-hydroquinolyl))carbonylamino)propanoic Acid Trifluoroacetate Salt
A solution of the product from Part F, above (15 mg, 0.0135 mmol), TEA (0.007 mL), and 2-(2-aza-2-((5-((2,5-
15 dioxopyrrolidinyDcarbonyl) (2-pyridyl))amino) vinyl) -
benzenesulfonic acid (9.0 mg, 0.0204 mmol) in anhydrous DMF (2.5 mL) was allowed to stand at ambient temperatures under a nitrogen atmosphere for 22 h. The DMF was removed under vacuum and the glassy solid was dissolved
20 in 20% ACN and purified by preparative HPLC on a Vydac C-18 column (22 x 250 mm) using 0.1% TFA in water for 5 min followed by a 2.52%/min gradient of 0 to 63% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 21.2 min was collected and
~±*8-
25 lyophilized to give the title compound as a colorless powder (3.5 mg, 20%). MS: m/e 1186.7 [M+H]; High Resolution MS: Calcd for C53H60N11O15S3 [M+H]: 1186.3432, Found: 1186.3410.


3-((7-((Imidazol-2-ylamino)methyl)-1-methyl-4-oxo (3-hydroquinolyl) )carbonylammo) -2- ( ( (4- (4- (((3- (2- (2- (3- (2-
(1,4,7,10-tetraaza-4,7,10-
tris (carboxylmethyl)cyclododecyl)acetylamino) -
5 propoxy)ethoxy)ethoxy)propyl(amino)sulfonyl)-
phenyl)phenyl)sulfonyl)amino)propanoic Acid
Bis(trifluoroacetate) Salt
Part A - Phenylmethyl 2-(1,4,7,10-Tetraaza-4,7,10-10 tris(((tert-butyl)oxycarbonyl)methyl)cyclododecyl)acetate A solution of tert-butyl (1,4,7,lO-tetraaza-4,7-
bis(((tert-butyl)oxycarbonyl)methyl)cyclododecyl)acetate (0.922 g, 1.79 mmol), TEA (1.8 mL) and benzyl
bromoacetate (0.86 mL, 5.37 mmol) in anhydrous DMF (24 15 mL) was stirred at ambient temperatures under a nitrogen
atmosphere for 24 h. The DMF was removed under vacuum
and the resulting oil was dissolved in EtOAc (300 mL) .
This solution was washed consecutively with water (2 x 50
mL) and saturated NaCl (50 mL), dried (MgS04) , and 20 concentrated to give the title compound as an amorphous
solid (1.26 g). MS: m/e 663.5 [M+H].

25 Part B - 2-(1,4,7,10-tetraaza-4,7,10-tris(((tert-butyl )oxycarbonyl)methyl)cyclododecyl)acetic acid
The product from Part A, above (165 mg, .0.25 mmol) was hydrogenolyzed over 10% Pd on carbon (50 mg) in EtOH (15 mL) at 60 psi for 24 h. The catalyst was removed by
30 filtration through filter aid and washed with EtOH. The filtrates were concentrated to give the title compound as an amorphous solid (134 mg, 94%). MS: m/e 573.5 [M+H].
~129-- 1.2 *> '



Part C - Methyl 3-((7-((Imidazol-2-ylamino)methyl)-1-methyl-4-oxo(3-hydroquinolyl) ) carbonylamino) ~2- (((4- (4-5 ( ( (3-(2-(2-(3-(2-(1.4,7,10-tetraaza-4,7,10-tris(((tert-butyl) oxycarbonyl) methyl) cyclododecyl) acetylamino) -propoxy)ethoxy)-
ethoxy)propyl)amino) sulfonyl) phenyl5phenyl) sulfonyl) -amino)propanoate Pentakis(trifluoroacetate) Salt 10 A solution of the product of Example 1, Part C (68 mg, 0.0534 mmol) and Et3SiH (0.051 mL, 0.32 mmol) in degassed TFA (5.0 mL) was stirred at 70 °C under a nitrogen atmosphere for 1 h and concentrated to dryness. The resulting amber oil was dissolved in anhydrous DMF (2 15 mL) and treated with TEA until basic to pH paper. A solution of the product of Part B, above (46 mg, 0.080 mmol) in anhydrous DMF (1.0 mL) was added, followed by HBTU (24 mg, 0.064 mmol), and the solution was stirred at ambient temperatures under a nitrogen atmosphere for 3 h. 20 The DMF was removed under vacuum and the residue was
dissolved in 50% ACN and purified by preparative HPLC on a Vydac C-18 column (22 x 250 mm) using a 2.1%/min gradient of 0 to 63% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 23.8 25 min was collected and lyophilized to give the title compound as a colorless powder (16 mg, 15%) . MS: m/e 1451.7 [M+H]; High Resolution MS: Calcd for C69H103N12O18S2 [M+H]: 1451.6954, Found: 1451.698.


30

Part D - 3- ((7-((Imidazol-2-ylamino)methyl)-l-methyl-4-oxo(3-hydroquinolyl))carbonylamino) -2- ( ((4- (4- ( ((3- (2- (2-

-t^d—

(3-(2-(l,4,7,10-tetraaza-4,7,10-tris (carboxylmethyl) cyclododecyl) acetylamino) -propoxy)ethoxy) ethoxy) propyl) amino)sulfonyl) -phenyl) phenyl) sulfonyl) amino) propanoic Acid 5 Bis (trifluoroacetate) Salt
The product of Part C, above (16 mg, 0.0102 mmol) was dissolved in a mixture of peroxide-free THF (1 mL), water (0.115 mL), and 3 N LiOH (0.075 mL), and stirred at ambient temperatures under a nitrogen atmosphere for 24 10 h. The reaction was concentrated to give an oily solid. This solid was dissolved in 50% ACN and purified by preparative HPLC on a Vydac C-18 column (22 x 250 mm) using a 2.52%/min gradient of 0 to 63% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product 15 peak eluting at 24.0 min was collected and lyophilized to give a colorless powder (6.0 mg) . This solid was dissolved in degassed TFA (2.0 mL) and Et3SiH (0.050 mL) , stirred at 70 °C under a nitrogen atmosphere for 4.5 h, and concentrated to dryness. The resulting oil was 20 dissolved in 25% ACN and purified by preparative HPLC on a Vydac C-18 column (22 x 250 mm) using a 1.5%/min gradient of 1269.5 [M+H], 635.5 [M+2H], 424.3 [M+3H]; High Resolution MS: Calcd for C56H77Ni20i8S2 [M+H]: 1269.4920, Found: 1269.4950.




30

•2 TFA
Example 3
2-{{(4-(3-{N-(3-(2-(2-(3-( ( 6- ( (l-Aza-2- (2-
sulfophenyl) vinyl) amino) (3-pyridyl)) carbonylamino) -

propoxy)ethoxy)ethoxy)propyl)carbamoylJpropoxy)-2,6-
dimethylphenyDsulfonyl) amino) -3- ( (7- ( (imidazol-2-
ylamino)methyl)-l-methyl-4-oxo(3-hydroquinolyl))-
carbony1amino)propanoic Acid Trifluoroacetate Salt
5
Part A - Ethyl 4-(3,5-Dimethylphenoxy)butanoate
Sodium metal (17.12 g, 0.744 mol) was added to anhydrous EtOH (350 mL) and stirred until dissolved. 3, 5-Dimethylphenol was added and the solution was stirred 10 15 min at ambient temperatures. Ethyl 4-bromoacetate (58.7 mL, 0.41 mol) was added and the solution was stirred at ambient temperatures under a nitrogen atmosphere for 28 h. The EtOH was removed under vacuum and the oily solid was partitioned between water (1 L) 15 and EtOAc (500 mL). The aqueous layer was extracted with additional EtOAc (500 mL). The combined EtOAc extracts were washed consecutively with saturated NaHC03 (300 mL) and saturated NaCl (300 mL), dried (MgSC>4), and concentrated to give an amber liquid. This liquid was 20 vacuum fractional distilled through a 15 cm Vigreux
column. The main fraction was collected from 91-117 °C/6 mm Hg to gave the title compound as a colorless liquid (77.77 g, 89%). *H NMR (CDC13): 6.59 (s, 1H), 6.52 (s, 2H) , 4.16 (q, J - 7.16 Hz, 2H), 3.98 (t, J = 6.14 Hz, 25 2H), 2.49 (t, J = 7.34 Hz, 2H), 2.28 (s, 6H), 2.11-2.07 (m, 2H), 1.26 (t, J = 7.16 Hz, 3H); Anal, calcd for C14H20O3: C71.16; H, 8.53, Found: C71.35; H, 8.59.

30
Part B - 4-(3,5-Dimethylphenoxy)butanoic Acid
The product of part A, above (75.52 g, 0.320 mol) and KOH pellets (38.5 g, 0.584 mol) were dissolved in absolute EtOH (1.50 L) and heated at reflux for 3 h. The 35 solution was concentrated to a colorless solid, which was taken up in water (2.0 L) and washed with ether (2 x 750

mL). The aqueous layer was adjusted to pH 1 with coned HC1 (55 mL) and the resulting oily ppt was extracted into EtOAc (2 x 500 mL). The combined EtOAc extracts were washed consecutively with water (300 mL) and saturated 5 NaCl, dried (MgSO^), and concentrated to give a colorless solid (64.13 g). Recrystallization from hexanes (500 mL) gave the title compound as a colorless solid (59.51 g, 89%). MP: 66-68.5 °C; 2H NMR (CDCI3): 11.70 (bs, 1H) , 6.59 (s, 1H), 6.52 (s, 2H), 3.99 (t, J = 6.06 Hz, 2H), 10 2.57 (t, J = 7.29 Hz, 2H), 2.28 (s, 6H), 2.12-2.08 (m, 2H); Anal, calcd for Ci2Hi603: C, 69.21; H, 7.74, Found: C, 69.23; H, 7.40.

15
Part C - 4-(4-(Chlorosulfonyl)-3,5-dimethylphenoxy)butanoic Acid
A solution of the product of Part B, above (20.8 g, 0.100 mol) in CHC13 (100 mL) was cooled to 0 °C and
20 treated with chlorosulfonic acid (36 mL, 0.54 mol) dropwise and with rapid stirring while keeping the temperature of the reaction at 0 °C. The resulting gelatinous mixture was stirred an additional 10 min and poured onto an ice/water mixture (600 mL). The resulting
25 solid ppt was collected by filtration, washed with water (3 x 75 mL), and dried under vacuum to give a colorless solid (12.52 g). MP: 114-115 °C (with decomp); *H NMR (CDCI3): 13.84 (bs, 1H), 6.50 (s, 2H), 3.91 (t, J = 6.48 Hz, 2H), 2.48 (s, 6H), 2.32 (t, J = 7.32 Hz, 2H), 1.89-
30 1.84 (m, 2H); IR (KBr cm'1): 1705 (s), 1370 (s), 1175 (s); MS: m/e 305.1 [M-H].


Part D - 4- (4- ( ( (2- ( (tert-Butoxy) carbonylamino) -1-(methoxycarbonyl) ethyl)amino)sulfonyl) -3,5-dimethylphenoxyjbutanoic Acid
A solution of N-($-Boc-L-a,fJ,-diaminopropionic acid 5 methyl ester hydrochloride (568 mg, 2.10 mmol) and DIEA (0.73 mL, 4.2 iranol) in DCM (5 mL) was cooled to 0 °C and treated with a suspension of the product of Part C, above (656 mg, 2.10 mmol) in DCM (20 mL) in small portions over a 15 min period. The reaction was stirred at ambient 10 temperatures under a nitrogen atmosphere for 18 h. The reaction was diluted with DCM (100 mL) and washed with water (3 x 75 mL) . The organic phase was dried (MgSC>4), and concentrated to give crude product (698 mg), which was purified by preparative HPLC on a Vydac C-18 column 15 (50 x 250 mm) using a 0.96%/min gradient of 18 to 58.5% ACN containing 0.1% TFA at a flow rate of 80 mL/min. The main product fraction eluting at 23.8 min was collected adjusted to pH 3, partially concentrated to remove ACN, and extracted with DCM (2 x 100 mL) . The DCM extracts 20 were dried (MgS04) and concentrated to give the title
compound as a colorless solid (297 mg, 29%) . *H NMR (CDCI3): 5 6^61 (s, 2H), 5.66 (d, J = 7.2 Hz, 1H) , 4.90 (s, 1H), 4.03 (bs, 2H), 3.86 (bs, 1H), 3.59 (s, 3H), 3.49 (bs, 2H), 2.62 (s, 6H), 2.58-2.51 (m, 2H) , 2.18-2.07 (m, 25 2H), 1.41 (s, 9H); MS: m/e 489.4 [M+H]; High Resolution
MS: Calcd for C21H33N2O9S [M+Na] : 511.1726, Found:
511.1747; Anal, calcd for C21H32N2O9S: C, 51.62; H, 6.61;
N, 5.74, Found: C, 51.47; H, 6.27; N, 5.48.

H f>iH
30
Part E - Methyl 3-((tert-Butoxy)carbonylamino)-2-(((2,6-
dimethyl-4-(3-(N-(3-(2-(2-(3-
( (phenylmethoxy) carbonylamino) propoxy) ethoxy) -

ethoxy) propyl) carbamoyl) propoxy) phenyl) -sulfonyl)amino)propanoate
A solution of the product from Part D, above (233 mg, 0.477 mmol), the product of Example 1, Part A (190 5 mg, 0.536 mmol), TEA (0.2 mL, 1.43 mmol), and HBTU (226 mg, 0.701 mmol) in anhydrous DMF (8 mL) was stirred at ambient temperatures under a nitrogen atmosphere for 1 h. The DMF was removed under vacuum and the oily residue was taken up in EtOAc (50 mL) and washed consecutively with
10 0.1 N HC1 (35 mL), water (35 mL) , and saturated NaCl (35 mL) , dried (MgSCu), and concentrated to give crude product as a yellow viscous oil. Flash chromatography on a 3 x 18 cm silica gel column (EtOAc/MeOH. 95/5) gave the title compound as a colorless viscous oil (393 mg, 100%).
15 iH NMR (CDCI3): 8 7.34-7.28 (m, 5H) , 6.60 (s, 2H) , 6.26
(bs, 1H), 5.67 (bs, 1H) , 5.29 (bs, 1H) , 5.08 (s, 2H) , 4.88 (bs, 1H) , 3.99 (t, J = 6.1 Hz, 2H), 3.88-3.84 (m, 1H), 3.62-3.40 (m, 17H) , 3.37-3.26 (m, 4H) , 2.62 (s, 6H), 2.32 (t, J = 7.2 Hz, 2H), 2.08 (t, J = 6.3 Hz, 2H), 1.79-20 1.70 (m, 4H), 1.41 (s, 9H); MS: m/e 825.5 [M+H]; High Resolution MS: Calcd for C39H61N4O13S [M+H): 825.3955, Found: 825.39*40.

Part F - Methyl 3-Amino-2-(((2, 6-dimethyl~4-(3-(N-(3-(2-(2- (3- ((phenylmethoxy) carbonylamino)propoxy)ethoxy) -ethoxy) propyl) carbamoyl) propoxy) phenyl) -s u1f ony1)amino)propanoate 30 The product of Part E, above (750 mg, 0.91 mmol) was dissolved in 4 M HCl/dioxane (25 mL) and stirred at ambient temperatures for 1 h. The solution was diluted with ether (500 mL) and the resulting gummy ppt was

triturated with fresh ether (2 x 250 mL). The gummy solid was dissolved in water (100 mL) and adjusted to pH 9 with NaHCCb, causing an oily ppt to form. This ppt was extracted into DCM (2 x 75 mL). The DCM extracts were 5 dried (MgSC>4) and concentrated to give the title compound as a colorless oil (386 mg, 56%). MS: m/e 725.5 [M+H].

10 Part G - Methyl 2- (((2,6-Dimethyl-4-(3-(N-(3-(2-(2-(3-((phenylmethoxy)carbonylamino)propoxy)ethoxy)ethoxy) -propyl)carbamoyl)propoxy)phenyl)sulfonyl)amino)-3-((1-methyl-4-oxo-7-(((1-(triphenylmethyl)imidazol-2-yl)amino)methyl)(3-
15 hydroquinolyl) ) carbonylamino) propanoate
A solution of l-methyl-4-oxo-7-(((1-(triphenylmethyl)imidazol-2-
yl)amino)methyl)hydroguinoline-3-carboxylic acid (274 mg, 0.51 mmol), TEA (0.22 mL, 1.52 mmol), and HBTU (192 mg,
20 0.51 mmol) in anhydrous DMF (3 mL) was stirred at ambient temperatures for 5 man. A solution of the product of Part F, above (367 mg, (0.51 mmol) in anhydrous DMF (7 mL) was added and the resulting solution was stirred at ambient temperatures under a nitrogen atmosphere for 2 h.
25 The DMF was removed under vacuum and the resulting oily solid was dissolved in EtOAc (150 mL). The EtOAc solution was washed consecutively with water (50 mL), saturated NaHC03 (25 mL), and saturated NaCl (25 mL), dried (MgS04), and concentrated to give a yellow solid.
30 Purification by flash chromatography on a silica gel
column using a EtOAc/MeOH step gradient (95/5, 92.5/7.5) gave the title compound as a pale yellow solid (254 mg, 43%). MS: m/e 1247.7 [M+H], 624.6 [M+2H].




Part K - 2-(((2,6-Dimethyl-4-(3-(N-(3-(2-(2-(3-5 ((phenylmethoxy)carbonylamino)propoxy)ethoxy)ethoxy)-propyl)carbamoyl)propoxy)phenyl)sulfonyl)amino)-3-((1-methyl-4-oxo-7-(((1-(triphenylmethyl)imidazol-2-yl)amino)methyl)(3-hydroquinolyl))carbonylamino)propanoic Acid
10 The product of Part G, above (60.0 mg, 0.048 nunol) was dissolved in a mixture of peroxide-free THF (2.5 mL), water (0.37 mL), and 3 N LiOH (0.244 mL) , and stirred at ambient temperatures under a nitrogen atmosphere for 30 min. The THF was removed under vacuum and the resulting
15 mixture was dissolved in CHCI3 (25 mL) and water (20 mL). The aqueoua- layer was adjusted to pH 3 with 0.1 N HCl and the layers were thoroughly mixed. The aqueous layer was extracted with additional CHCI3 (2 x 20 mL). The combined CHCI3 extracts were washed with saturated NaCl
20 (30 mL), dried (MgSC>4), and concentrated to give the title compound as a pale yellow solid (44.0 mg, 74%). MS: m/e 1233.7 [M+H]; High Resolution MS: Calcd for C67H77N8O13S [M+H]: 1233.5330, Found: 1233.5330.

QO Q
OH
25

DMF was removed under vacuum and the amber oil was dissolved in 50% ACN and purified by preparative HPLC on a Zorbax C-18 RX column (21.2 x 250 mm) using a 1.5%/min gradient of 0 to 45% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 21.0 min was collected and lyophilized to give the title compound as a colorless powder (8.9 mg, 20%). MS: m/e 1160.6 [M+H], 581.0 [M+2H].


10

Example 4
3-( (l-(3-((6-((l-Aza-2-(2-sulfophenyl)vinyl)amino)(3-
15 pyridyl) )carbonylamino)propyl)-7- ((imidazole-2-ylamino)-
methyl)-4-oxo(3-hydroquinolyl))carbonylamino)-2-(((2,4,6-
trimethylphenyl)sulfonyl)amino)propanoic Acid
Trifluoroacetate Salt
20 Part A - Ethyl 1-(3-((tert-Butoxy)carbonylamino)propyl) -7-bromo-4-oxohydroquinoline-3-carboxylate
A mixture of ethyl 7-bromo-4-oxohydroquinoline~3-carboxylate (6.28 g, 0.0212 mol), (tert-butoxy)-N-(3-bromopropyl)formamide (30.3 g, 0.127 mol), and anhydrous
25 K2CO3 (12.5 g, 0.904 mol) in anhydrous DMF (200 mL) was stirred at 60 °C under a nitrogen atmosphere for 4 h, and then at ambient temperatures for 72 h. The DMF was removed under vacuum and the resulting oily solid was dissolved in EtOAc (500 mL). The EtOAc solution was
30 washed consecutively with water (500 mL), saturated NaHC03 (500 mL), and saturated NaCl (500 mL), dried (MgSCj), and concentrated to give a red oil. This oil


was taken up in EtOAc (250 mL) and cooled, causing a solid ppt to form. This ppt was collected by filtration, washed with cold EtOAc, and dried to give the title compound as a colorless solid (6.25 g, 65%). MP: 140-142 5 °C; *H NMR (CDC13): 8.49 (s, IH), 8.39 (d, J = 8.6 Hz, IH), 7.58 (S, IH), 7.53 (d, J = 8.6 Hz, IH), 4.72 (bs, IH), 4.39 (q, J = 7.1 Hz, 2H), 4.20 (t, J = 7.6 Hz, 2H), 3.28-3.24 (m. 2H) , 2.10-2.06 (m, 2H) , 1.46 (s, 9H), 1.40 (t, J = 7.1 Hz, 3H); MS: m/e 455.2. [M+H]; High 10 Resolution MS: Calcd for C2oH26BrN205 [M+H]: 453.1025, Found: 453.1028.



Part B - Ethyl 1-(3-( (tert-Butoxy)carbonylamino)propyl)-
15 4-oxo-7-vinylhydroquinoline-3-carboxylate
The product from Part A, above (2.98 g, 6.60 mmol). was dissolved in toluene (50 mL) at a temperature of 100 °C and treated with tetrakis(triphenylphosphine)palladium(0) (152 mg, 0.132
20 mmol). After 5 min the mixture was treated with
tributyl(vinyl)tin (1.93 mL, 6.60 mmol) and stirred 4.5 h at 100 °C under a nitrogen atmosphere, and 18 h at ambient temperatures. Additional tributyl(vinyl)tin (0.386 mL) and tetrakis(triphenylphosphine)palladium(O) ^
25 (152 mg) were added and the mixture was heated at 100 °C%
for an additional 17 h. The toluene was removed under I
f vacuum and the solid residue was triturated with ether tf
give the title compound as a pale green solid (1.67 g,
63%). MP: 133-135 °C; XH NMR (CDCI3) : 8.52 (d, J = 8
30 Hz, IH), 8.51 (s, IH), 7.55 (d, J = 8.4 Hz, IH), 7.?/
IH), 6.88-6.82 (m, IH) , 5.97 (d, J = 17.4 Hz, IH),
0
(d, J = 10.8 Hz, IH), 4.75 (bs, IH), 4.42 (q, J
2H), 4.27 (t, J = 7.8 Hz, 2H) , 3.6-3.25 (m, 2H) ,'
2.11 (m, 2H), 1.49 (s, 9H), 1.45 (t, J = 7.2 Hz, 3R>

m/e 401.3 [M+HJ; High Resolution MS: Calcd for C22H29N2O5 [M+H]: 401.2076, Found: 401.2075.

Part C - Ethyl 1-(3-((tert-Butoxy)carbonylamino)propyl)-7-formyl-4-oxohydroquinoline-3-carboxylate
A solution of the product of Part B, above {1.50 g, 3.75 mmol) in dioxane (119 mL) and water (39 mL) was
10 treated with a solution of osmium tetroxide (19.6 mg,
0.077 mmol) in dioxane (0.600 mL) and stirred at ambient temperatures under a nitrogen atmosphere for 5 min. Sodium periodate (2.40 g, 11.2 mmol) was added and the stirred at ambient temperatures for 2 h. The dioxane was
15 removed under vacuum and the residue was taken up in DCM (500 mL). The DCM solution was washed consecutively with water (500 mL) and saturated NaCl (500 mL), dried (MgSC>4), and concentrated to give the title compound as
an orange oily solid (1.52 g, 100%). XH NMR (CDCI3): 20 10.17 (s, 1H), 8.68 (d, J = 8.2 Hz, 1H), 8.64 (s, 1H) , 8.01 (s, 1H), 7.88 (d, J = 8.2 Hz, 1H) , 4.82 (bs, 1H) , 4.41-4.35 (m, 4H), 3.28 (s, 2H) , 2.15-2.07 (m, 2H) , 1.45 (S, 9H), 1.41 (t, J = 7.1 Hz, 3H); MS: m/e 403.3 [M+H]; High Resolution MS: Calcd for C21H27N2Q6 [M+H]: 403.1870, 25 Found: 403.1875.

Part D - Ethyl 1-(3-((tert-Butoxy)carbonylamino)propyl)-4-0x0-7-(((1-{triphenylmethyl)imidazole-2-30 yl)amino)methyl)hydroquinoline-3-carboxylate


A solution of the product of Part C, above (544 mg, 1.35 mmol) and 1- (triphenylmethyl) imidazole-2-ylamine (456 mg, 1.35 mmol) in toluene (60 mL) was heated at reflux under a nitrogen atmosphere with removal of water 5 for 5 h. The solution was cooled, treated with
Na(OAc)3BH (1.14 g, 5.3 8 mmol) and stirred at ambient temperatures for 18 h. The mixture was diluted with EtOAc (400 mL), washed consecutively with water (500 mL) and saturated NaCl (500 mL) , dried (MgSC>4), and
10 concentrated to give an orange solid. This solid was
dissolved in 50% ACN and purified by preparative HPLC on a Vydac C-18 column (50 x 250 mm) using a 0.60%/min gradient of 18 to 52% ACN containing 0.1% TFA at a flow rate of 49 mL/min. The main product peak eluting at 3 0.8
15 min was collected and lyophilized to give the title
compound as a pale yellow solid (407 mg, 60%) . MS: m/e 712.4 [M+H]; High Resolution MS: Calcd for C43H46N5O5 [M+H}: 712.3499, Found: 712.3485.
20
Part E - 1-(3-((tert-Butoxy)carbonylamino)propyl)-4-oxo-7- (((1-(triphenylmethyl)imidazole-2-yl)amino)methyl)-hydroquinoline-3-carboxylic Acid
25 A mixture of the product of Part D, above (997 mg, 1.40 mmol), water (7.3 mL), 3 N LiOH (3.5 mL), and THF (50 mL) was stirred at ambient temperatures under a nitrogen atmosphere for 3 h. The THF was removed under vacuum and the resulting mixture was dissolved in CHCI3
30 (500 mL) and water (100 mL). The aqueous layer was adjusted to pH 3 with 1.0 N HC1 and the layers were thoroughly mixed. The organic layer was washed consecutively with water (500 mL) and saturated NaCl (500 mL), dried (MgS04) , and concentrated to give the title
'-=142 J-

10

compound as a pale yellow solid (998 mg). MP: 153-160 °C; XH NMR (CDC13): 8 14.83 (s, IH) , 8.76 (s, IH) , 8.68 (s, IH) , 8.24 (d, J = 6 Hz, IH), 7.49-7.35 (m, 9H), 7.12-7.10 (m, 6H), 6.82 (s, IH) , 6.52 (s, IH) , 6.24 (d, J = 6 Hz, IH), 5.75 (bs, IH), 4.87-4.83 (m, 2H), 4.77 (bs, IH), 4.51 (t, J = 9 Hz, 2H), 3.38 (s, 2H), 2.23 (s, 2H), 1.42 (s, 9H); MS: m/e 684.3 [M+H]; High Resolution MS: Calcd for C41H42N5O5 [M+H]: 684.3186, Found: 684.3181.

O 0
OH

Part F - Methyl 3-((1-(3-((tert-
Butoxy)carbonylamino)propyl)-4-oxo-7-(((1-
(triphenylmethyl)imidazole-2-yl)amino)methyl) (3 -
15 hydroquinolyl))carbonylamino)-2-(((2,4,6-trimethylphenyDsulfonyl) amino)propanoate
A solution of the product of Part E, above (300 mg, 0.437 mmol), TEA (0.243 mL, 1.75 mmol), and HBTU (230 mg, 0.606 mmol) in anhydrous DMF (4 mL) was stirred at
20 ambient temperatures for 5 min. A solution of methyl 3-amino-2-(((2,4,6-
trimethylphenyl)sulfonyl)amino)propanoate hydrochloride (184 mg, 0.637 mmol) in anhydrous DMF (3 mL) was added and the solution was stirred at ambient temperatures
25 under a nitrogen atmosphere for 2 h. The solution was
diluted with EtOAc (200 mL) and washed consecutively with water (2 x 50 mL), saturated NaHC03 (50 mL), and saturated NaCl (50 mL) , dried (MgSC>4), and concentrated to give a viscous amber oil. Purification by flash
30 chromatography on a 2.5 x 24 cm silica gel column using a EtOAc/MeOH step gradient (98/2, 95/5, 75/25) gave the title compound as a pale yellow oil (330 mg, 78%). MS: m/e 966.6 [M+H]; High Resolution MS: Calcd for C54H60N7O8S [M+H]: 966.4224, Found: 966.4224.



Part G - 3-((1-(3-((tert-Butoxy)carbonylamino)propyl)-4-5 oxo-7-(((1-(triphenylmethyl)imidazole-2-
yl)amino)methyl)(3-hydroquinolyl))carbonylamino)-2-(((2,4, 6-trimethylphenyl)sulfonyl)amino)propanoic Acid A solution of the product of Part F, above (51 mg, 0.052 mmol), water (0.27 mL) , and 3 N LiOH (0.13 mL) in
10 MeOH (2 mL) was allowed to stand at ambient temperatures for 3.5 h and concentrated under vacuum. The resulting solid was dissolved in water (10 mL) and adjusted to pH 3 with 1.0 N HC1. The aqueous mixture was extracted with DCM (2 x 3 0 mL). The combined DCM extracts were washed
15 with saturated NaCl (30 mL), dried (MgS04> , and
concentrated to give the title compound as a colorless solid (72 mg). MS: m/e 952.5 [M+H]; High Resolution MS: Calcd for C53H58N7O8S [M+H]-. 952.4067, Found: 952.4056.

20
Part H - 3-((1-(3-Aminopropyl)-7-((imidazole-2-ylamino)-methyl)-4-oxo(3-hydroquinolyl))carbonylamino)-2-(((2,4,6-trimethylphenyl)sulfonyl)amino)propanoic Acid Bis(trifluoroacetate) Salt
25 The product of Part I, above (0.052 mmol) and Et3SiH (0.042 mL, 0.26 mmol) were dissolved in degassed TFA (2 mL), heated at 70 °C for 2.5 h, and concentrated to give an amber oil. This oil was dissolved in water (25 mL) and washed with ether (2 x 15 mL). The combined ether
30 washings were back-extracted with water (15 mL) . The

combined water extracts were lyophilized to give the title compound as a colorless powder (34 mg, 78%). MS: m/e 610.4 [M+H]; High Resolution MS: Calcd for C29H36N7O6S [M+H]: 610.2448, Found: 610.2462.

Part I - 3-((l-(3-((6-((l-Aza-2-(2-sulfophenyl)vinyl)amino)(3-10 pyridyl))carbonylamino)propyl)-7-((imidazole-2-ylamino)-methyl)-4-oxo(3-hydroquinolyl))carbonylamino)-2-(((2,4,6-trimethylphenyDsulfonyl) amino) propanoic Acid Trifluoroacetate Salt
A solution of the product of Part H, above (13.7 mg,
15 0.0163 mmol), TEA (0.015 mL, 0.108 mmol), and 2-(2-aza-2-((5-((2,5-dioxopyrrolidinyl)carbonyl)(2-pyridyl))-amino)vinylJbenzenesulfonic acid (8.2 mg, 0.0186 mmol) in anhydrous DMF (2.0 mL) was allowed to stand at ambient temperatures under a nitrogen atmosphere for 24 h. The
20 DMF was removed under reduced pressure and the amber oil was dissolved in 50% ACN and purified by preparative HPLC on a Vydac C-18 column (22 x 250 mm) using 0.1% TFA in water for 5 min followed by a 2.52%/min gradient of 0 to 63% ACN containing 0.1% TFA at a flow rate of 20 mL/min.
25 The main product peak eluting at 21.4 min was collected and lyophilized to give the title compound as a colorless powder (12.5 mg, 75%). MS: m/e 913.3 [M+H]; High Resolution MS: Calcd for C42H45N10OioS2 [M+H]: 913.2761, Found: 913.2751.
30



Example 5
3- ((1- (3-((6-((l-Aza-2-(2-sulfophenyl)vinyl)amino) (3-
pyridyl) ) carbonylamino)propyl) -7- (( (l-hydroxyimidazole-2-
yl)amino)methyl) -4-oxo(3-hydroquinolyl) ) carbonylamino) -2-
(((2,4, 6-trimethylphenyl) sulfonyl) amino) propanoic Acid
Trifluoroacetate Salt

10
Part A - Methyl 3- ((1-(3-Aminopropyl)-7-((imidazole-2-ylamino)methyl) -4-oxo (3-hydroquinolyl) ) carbonylamino) -2-( ( (2,4, 6-trimethylphenyl)sulfonyl)amino)propanoate Bis(trifluoroacetate) Salt
15 A solution of the product of Example 4, Part F (12 0 mg, 0.124 mmol) and Et3SiH (0.99 mL, 6.20 mmol) in TFA (10 mL) was heated at 70 °C for 1 h, and concentrated to give an amber oil. This oil was dissolved in water (50 mL) and washed with ether (2 x 30 mL) . The combined
20 ether washings were back-extracted with water (20 mL) . The combined water extracts were lyophilized to give the title compound as a colorless powder (105 mg, 100%). MS: m/e 624.4 [M+H]; High Resolution MS: Calcd for C30H38N7O6S [M+H]: 624.2604, Found: 624.2608.

25
Part B - 3-((1-(3-Aminopropyl)-7-(((l-hydroxyimidazol-2-yl) amino)methyl) -4-oxo (3-hydroquinolyl) ) carbonylamino) -2-


(((2,4,6-trimethylphenyl)sulfonyl)amino)propanoic Acid Trifluoroacetste Salt
A mixture of the product of Part A, above (105 mg, 0.126 mmol), water (3.0 mL), and 3 N LiOH (1.82 mL) in 5 peroxide-containing THF (4 mL) was allowed to stand at ambient temperatures for 1 h and concentrated under vacuum. The resulting solid was dissolved in water (10 mL) and adjusted to pH 5 with 1.0 N HC1. Insoluble impurities were removed by filtration and the filtrate
10 was lyophilized to give a colorless solid. This solid was dissolved in water and purified by preparative HPLC on a Vydac C-18 column (22 x 250 mm) using 0.1% TFA in water for 5 min followed by a 2.52%/min gradient of 0 to 63% ACN containing 0.1% TFA at a flow rate of 20 mL/min.
15 The main product peak eluting at 19.5 min was collected and lyophilized to give the title compound as a colorless powder (10.0 mg, 11%). MS: m/e 314.0 [M+2H]



20
Part C - 3-( (1-(3-((6-((l-Aza-2-(2-
sulfophenyl)vinyl)amino)(3-
pyridyl) ) carbonylamino)propyl) -7- ( ( (l-hydroxyimidazole-2-
yl) amino)methyl)-4-oxo(3-hydroquinolyl))carbonylamino)-2-
25 (((2,4,6-trimethylphenyl)sulfonyl)amino)propanoic Acid Trifluoroacetate Salt
A solution of the product of Part E, above (10.0 mg, 0.0135 mmol), TEA (0.018 mL, 0.129 mmol), and 2-(2-aza-2-((5- ( (2,5-dioxopyrrolidinyl)carbonyl) (2-pyridyl))-
30 amino)vinyl)benzenesulfonic acid (7.2 mg, 0.0163 mmol) in anhydrous DMF (4 mL) was allowed to stand at ambient temperatures under a nitrogen atmosphere for 20 h. The DMF was removed under vacuum and the amber oil was dissolved in 30% ACN and purified by preparative HPLC on

a Vydac C-18 column (22 x 250 mm) using 0.1% TFA in water for 5 min followed by a 2.52%/min gradient of 0 to 63% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 21.5 min was collected and lyophilized to give the title compound as a colorless powder (3.5 mg, 25%). MS: m/e 929.4 [M+H3,- High Resolution MS: Calcd for C42H45N10O11S2 [M+H]: 929.2710, Found: 929.2698.

10
Example §
3-({l-(3~(3-(N-(3-(2-(2-(3-( (6-( (l-Aza-2-(2-
sulfophenyl)vinyl)amino)(3-
15 pyridyl))carbonylamino)propoxy)ethoxy)-
ethoxy)propyl)carbamoyl)propanoylamino)propyl) -7-
((imidazole-2-ylamino)methyl)-4-oxo(3-hydroquinclyl) ) -
carbonylamino)-2-{((2,4,6-trimethylphenyl)sulfonyl) -
amino)propanoic Acid Trifluoroacetate Salt
20
Part A - 3-(N-(3- (2-(2-(3-((tert-Butoxy)carbonylamino)-propoxy) ethoxy) ethoxy) propyl) carbamoyl) propanoic Acid
A solution of N-(3-(2-(2-(3-aminopropoxy) ethoxy) ethoxy) propyl) (tert-butoxy) formamide 25 (as described by D. S. Wilbur et al. in Bioconjugate
Chem. 1998, 9, 322-330) (2.00 g, 6.24 mmol), TEA (1.0 mL, 7.49 mmol), and succinic anhydride (624 mg, 6.24 mmol) in anhydrous DMF (5 mL) was stirred at ambient temperatures under a nitrogen atmosphere for 4 h. The DMF was removed 30 under reduced pressure to give the title compound as a pale yellow oil (2.80 g) . MS: m/e 839.5 [2M-H], 419.4 [M-H].



Part B - Methyl 3-((1-(3-(3-(N-(3-(2-(2-(3-((tert-Butoxy) carbonylamino)propoxy) ethoxy) ethoxy)propyl) carbamo 5 yl)propanoylamino)propyl-4-oxo-7-( ( (1-(triphenylmethyl)-imidazole-2-yl)amino)methyl)(3-hydroquinolyl))carbonylamino)-2-(((2,4,6-trimethylphenyl)sulfony1)amino)propanoate
The product of Example 4, Part F (46.1 mg, 0.477
10 mmol) was dissolved in 50% TFA/DCM (2.0 mL) for 15 min at ambient temperatures and concentrated to give a yellow oil. This oil was dissolved in anhydrous DMF (1.0 mL) and made basic to pH paper with TEA. In a separate flask, the product of Part A, above (26.1 mg, 0.062
15 mmol), TEA (0.014 mL, 0.099 mmol), and HBTU (27.7 mg, 0.074 mmol) were dissolved in anhydrous DMF (1.0 mL). The resulting solution was allowed to react for 5 min and combined with the DMF solution from the TFA deprotection reaction. The combined solutions were allowed to stand
20 at ambient temperatures under a nitrogen atmosphere for 20 min and concentrated under vacuum. The resulting oil was dissolved in 50% ACN and purified by preparative HPLC on a Vydac C-18 column (22 x 250 mm) using a 1.8%/min gradient of 18 to 72% ACN containing 0.1% TFA at a flow
25 rate of 20 mL/min. The main product peak eluting at 26.8 min was collected and lyophilized to give the title compound as a colorless powder (44.5 mg, 68%). MS: m/e 1268.6 [M+H]; High Resolution MS: Calcd for C68H86N9O13S [M+H]: 1268.6065, Found: 1268.6070.


30

Part C - 3-((l-(3-(3-(N-(3-(2-(2-(3-( (tert-Butoxy) carbonylamino) propoxy) ethoxy) ethoxy) propyl) carbamo yl)propanoylamino)propyl-4-oxo-7-(((1- (triphenylmethyl) -imidazole-2-yl)amino)methyl)(3-5 hydroquinolyl))carbonylamino)-2-(((2,4,6-
trimethylphenyl)sulfonyl)amino)propanoic Acid
A solution of the product of Part B, above (31.1 mg, 0.0227 iranol), 3 n LiOH (0.091 mL) , and water (0.117 mL) in MeOH (1.30 mL) was stirred at ambient temperatures for
10 8.5 h. The MeOH was removed under vacuum and the aqueous mixture was diluted with water (30 mL) and adjusted to pH 4 with 1.0 N HC1. The resulting aqueous mixture was extracted with DCM (2 x 50 mL) . The combined DCM extracts were washed with saturated NaCl (50 mL) , dried
15 (MgSCj), and concentrated to give the title compound as a colorless solid (24.6 mg, 86%).

0 0 0
OH
20 Part D - 3-( (l-(3- (3-(N- (3- (2- (2-(3-aminopropoxy)ethoxy)-ethoxy) propyl) carbamoyl) propanoylamino) propyl) -7-((imidazole-2-ylamino)methyl)-4-oxo(3-hydroquinolyl))carbonylamino)-2-(((2,4,6-trimethylphenyl)sulfonyl)amino)propanoic Acid
25 Bis(trifluoroacetate) Salt
A solution of the product of Part C, above (24.6 mg, 0.0194 mmol) and Et^SiH (0.016 mL, 0.097 mmol) in TFA (2.0 mL) was heated at 70 °C under a nitrogen atmosphere for 3 h, and concentrated to give a yellow solid. This
30 solid was dissolved in water (50 mL) and washed with
ether (2 x 25 mL) . The aqueous layer was lyophilized to

give the title compound as a pale yellow solid (20.7 mg, 93%). MS: m/e 912.5 [M+H].

Part E - 3-( (l-(3-(3-(N-(3-(2-(2-(3-((6- A solution of the product of Part D, above (15.5 mg, 0.0136 mmol), TEA (0.010 mL, 0.0746 mmol), and 2-(2-aza-2- ((5-((2,5-dioxopyrrolidinyl)carbonyl) (2-pyridyl))-amino)vinyl)benzenesulfonic acid (8.0 mg, 0.0182 mmol) in anhydrous DMF (2.0 mL) was allowed to stand at ambient temperatures under a nitrogen atmosphere for 24 h. The DMF was removed under vacuum and the resulting yellow oil was dissolved in 50% ACN and purified by preparative HPLC on a Vydac C-18 column (22 x 250 mm) using 0.1% TFA in water for 5 min followed by a 2.52%/min gradient of 0 to 63% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 21.7 min was collected and lyophilized to give the title compound as a colorless powder (7.2 mg, 40%). MS: m/e 1215.5 [M+H]; High Resolution MS: Calcd for C56H71N12O15S2 [M+H]: 1215.4603, Found: 1215.4580.


Example 7 5 2-(2-Aza-2-(5-(N-(1,3-bis (3-(2-(2-(3-(3-(N-(3-(3-(N-(3-carboxy-2- (((2,4, 6-trimethylphenyDsulfonyl)amino)ethyl) -
carbamoyl)-7-((imidazole-2-ylamino)methyl)4-oxohydroquinolyl)propyl)carbamoyl)propanoylamino)propoxy) ethoxy)ethoxy)propyl)carbamoyl) (2-pyridyl))amino)vinyl) -10 benzenesulfonic Acid Bis(trifluoroacetate) Salt
Part A - N,N,-Bis(3-(2-(2-(3-(3-(N-(3-(3-(N-(3-carbomethoxy-2-(((2,4,6-trimethylphenyl) -sulfonyl)amino)ethyl)carbamoyl)-4-oxo-7-( ((1-
15 (triphenylmethyl)imidazole-2-yl)amino)methyl)-
hydroguinolyl) propyl) carbamoyl) propanoylamino) propoxy) eth oxy)ethoxy)propyl-2-((tert-butoxy)carbonyiamino)pentane-1,5-diamide
A solution of the product of Example 6, Part B (50.5
20 mg, 0.0398 mmol) in 50/50 TFA/DCM (2 mL) was allowed to react for 20 min at ambient temperatures and concentrated to a viscous oil. This oil was taken up in anhydrous DMF and made basic to pH paper with TEA. This solution was treated with Boc-L-Glu-OH (4.5 mg, 0.0181 mmol) and HBTU
25 (16.6 mg, 0.0438 mmol), and allowed to stand at ambient temperatures for 2 h. The DMF was removed under vacuum and the resulting oil was dissolved in 60% ACN and purified by preparative HPLC on a Vydac C-18 column (22 x 250 mm) using a 1.8%/min gradient of 18 to 72% ACN
30 containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 21.5 min was collected and lyophilized to give the title compound as a colorless

propanoy 1 amino)propoxy) ethoxy) ethoxy) propyl )pentane-l, 5-diamide Tris(trifluoroacetate) Salt Conjugate
The product of Part B, above, is dissolved in degassed TFA, treated with triethylsilane, and heated at 50 °C under nitrogen for 1 h. The solution is concentrated under vacuum and the resulting residue is purified by preparative HPLC on a C18 column using a water:ACN:0.1% TFA gradient. The product fraction is lyophilized to give the title compound.
10

Example 10
15 DOTA/2- ( ( (4-(3-(N-(3-(2-(2- (3- (2-Amino-3-sulfopropyi) -
propoxy) ethoxy) ethoxy) propyl) carbamoyl )propoxy) -2,6-
dimethylphenyDsulfonyl) amino) -3- ( (7- ( (imidazol-2-
ylamino)methyl) -l-methyl-4-oxo(3-hydroquinolyl) ) -
carbonyl ami no) propanoic Acid Trifluoroacetate Salt
20 Conjugate
Part A - 2-( ( (4-(3-(N-(3-(2-(2-(3-(2-((tert-Butoxy)-
carbonylamino) -3-sulfopropyi) propoxy) ethoxy! ethoxy) -
propyl)carbamoyl)propoxy)-2,6-25 dimethylphenyDsulfonyl) amino) -3- ((7- ( (imidazol-2-
ylamino)methyl) -l-methyl-4-oxo(3-hydroQuinolyl)) -
carbonylamino)propanoic Acid
The product of Example 3, Part I is dissolved in
anhydrous DMF and treated with the N-hydroxysuccinimide 30 ester of Boc-cysteic acid (as described in Liebigs Ann.


Chem. 1919, 776-783) and DIEA. The solution is stirred at ambient temperatures under nitrogen for 18 h, and the DMF is removed under vacuum. The resulting residue is purified by preparative HPLC on a C18 column using a 5 water:ACN:0.1% TFA gradient. The product fraction is lyophilized to give the title compound.
Part B - DOTA-tri-t-butyl Ester/2-(((4-(3-(N-(3-(2-(2-(3-(2-Amino-3-sulfopropyl)propoxy)ethoxy)ethoxy)propyl)-10 carbamoyl)propoxy)-2,6-dimethylphenyl)sulfonyl)amino)-3-((7-((imidazol-2-ylamino)methyl)-1-methyl-4-oxo(3-hydroquinolyl))carbonylamino)propanoic Acid TetraJcis (trifluoroacetate) Salt Conjugate
The product of Part A, above, is dissolved in 15 degassed TFA and stirred at ambient temperatures for 15 min. The solution is concentrated under vacuum, and the resulting residue is dissolved in 50% ACN and lyophilized to remove the last traces of TFA.
In a separate flask, a solution of the product of 20 Example 2, Part B and DIEA in anhydrous DMF are treated with HBTU and allowed to react 15 min at ambient temperatures under nitrogen. The deprotected product from above is added to this solution and stirring is continued at ambient temperatures under nitrogen for 18 25 h. The DMF is removed under vacuum and the resulting residue is purified by preparative HPLC on a C18 column using a water:ACN:0.1% TFA gradient. The product fraction is lyophilized to give the title compound.
30 Part C - DOTA/2-(((4-(3-(N-(3-(2-(2-(3-(2-Amino-3-sulfopropyl)-
propoxy) ethoxy) ethoxy) propyl) carbamoyl)propoxy) -2,6-dimethylphenyl)sulfonyl)amino)-3-((7-((imidazol-2-ylamino)methyl) -1-methyl-4-oxo(3-hydroquinolyl))-
35 carbonylamino)propanoic Acid Trifluoroacetate Salt Conjugate
The product of Part B, above, and Et3SiH are dissolved in degassed TFA and heated at 50 °C under nitrogen for 1 h. The solution is concentrated and the


resulting residue is purified by preparative HPLC on a C18 column using a water:ACN:0.1% TFA gradient. The product fraction is lyophilized to give the title compound.



Example 11 10 DOTA/2-(( (4-(3-(N- (3-(2-(2- (3-(2-Amino-3-(4-
(phosphonooxy)-phenyl) propanoylamino) propoxy) ethoxy) ethoxy) propyl) carbarn oyl)propoxy)-2,6-dimethylphenyl)sulfonyl)amino)-3-((7-((imidazol-2-ylamino)methyl)-1-methyl-4-oxo(3-15 hydroquinolyl))carbonylamino)propanoic Acid
Trifluoroacetate Salt Conjugate
The title compound is prepared by the same procedure described for Example 10 by substituting Boc-Tyr(PO3H2) -20 OSu for Boc-Cys(03H)-OSu.

25 Example 12

D0TA/2-( ((4-(3-(N-(3-(2-{2-(3-(2-Amino-3-(4-(sulfooxy)-
phenyl) propanoylamino) propoxy) ethoxy) ethoxy) propyl) carbarn
oyl) propoxy) -2, 6-dimethylphenyl) sulfonyl) amino) -3- ((7-
( (imidazol-2-ylamino)methyl) -l-methyl-4-oxo(3-
5 hydroquinolyl))carbonylamino)propanoic Acid
Trifluoroacetate Salt Conjugate

The title compound is prepared by the same procedure described for Example 10 by substituting Boc-Tyr(SO3H)-10 OSu for Boc-Cys(03H)-OSu.


Example 13
D0TA/2-( ( (4-(3-(N-(3-(2-{2-(3-(2-Amino-4-(N-(ethyl-3,6-0-
disulf o-(5-D-galactopyranosyl) carbamoyl) butanoylamino) -
propoxy) ethoxy)ethoxy)propyl) carbamoyl)propoxy) -2,6-
5 dimethylphenyDsulfonyl) amino) -3- ((7- ((imidazol-2-
ylamino)methyl) -l-methyl-4-oxo(3-hydroquinolyl) ) -
carbonylamino)propanoic Acid Conjugate
Part A - Preparation of Boc-Glu(aminoethyl-3, 6-0-disulfo-
10 P-D-galactopyranosyl)-OSu
A solution of Boc-Glu-OMe, aminoethyl-3,6-O-disulfo-P-D-galactopyranoside (as described in Tet. Lett. 1997, 53, 11937-11952), DIEA, and HBTU in anhydrous DMF is stirred at ambient temperatures under nitrogen for 18 h.
15 The DMF is removed under vacuum and the resulting residue is hydrolyzed using aqueous NaOH. The reaction solution is adjusted to pH 7 and purified by preparative anion exchange chromatography using a resin such as DEAE Cellulose and a Et3NH2CQ;j gradient. The product fraction
20 is treated with a cation exchange resin, sodium form, to give the intermediate carboxylic acid as the sodium salt.
The above compound, N-hydroxysuccinimide, and DCC are dissolved in anhydrous DMF and stirred at ambient temperatures under nitrogen for 18 h. The DMF is removed
25 under vacuum and the resulting residue is purified by preparative anion exchange chromatography as above to give the title compound as the triethylammonium salt.
Part B - DOTA/2-( ( (4- 30 (ethyl-3,6-0-disulfo-P-D-
galactopyranosyl)carbamoyl)butanoylamino)-propoxy) ethoxy) ethoxy)propyl)carbamoyl)propoxy) -2,6-dimethylphenyl)sulfonyl)amino)-3-((7-((imidazol-2-ylamino)methyl)-1-methyl-4-oxo(3-hydroquinolyl))-
35 carbonylamino)propanoic Acid Conjugate
The title compound is prepared by the same procedure described for Example 10 by substituting Boc-


Glu(aminoethyl-3, 6-0-disulfo-fJ-D-galactopyranosyl)-OSu for Boc-Cys(03H)-OSu.
Example 14
D0TA/2-(( (4-(3-(N-(3-(2-(2-(3-(2-Amino-4-(N-(6-deoxy-(5-
cyclodextryl)carbamoyl)butanoylamino)-
propoxy)ethoxy)ethoxy)propyl)carbamoyl)propoxy)-2, 6-
10 dimethylphenyl)sulfonyl)amino)-3-((7-((imidazol-2-
ylamino)methyl)-1-methyl-4-oxo(3-hydroquinolyl))-
carbonylamino)propanoic Acid Bis(trifluoroacetate) Salt
Conjugate
15 Part A - Preparation of Boc-Glu(6-amino-6-deoxy-[i-cyclodextryl)-OMe
A solution of Boc-Glu-OMe, 6-amino-6-deoxy-p-cyclodextrin (as described in J. Org. Chem. 1996, 61, 903-908), DIEA, and HBTU in anhydrous DMF is stirred at
20 ambient temperatures under nitrogen for 18 h. The DMF is removed under vacuum and the resulting residue is purified by preparative HPLC on a C18 column using a water:ACN:0.1% TFA gradient. The product fraction is lyophilized to give the title compound.
25
Part B - Preparation of Boc-Glu(6-amino-6-deoxy-p-cyclodextryl)-OSu
The product of Part A, above, is hydrolyzed by stirring in a mixture of LiOH, THF, and water at ambient 30 temperatures under nitrogen for 4 h. The THF is removed

under vacuum and the resulting mixture is diluted with water and adjusted to pH 3 using 0.1 N HC1. The mixture is extracted with EtOAc, and the combined extracts are dried (MgS04) and concentrated. The resulting material 5 is dissolved in anhydrous DMF along with N-
hydroxysuccinimide, and DCC, and stirred at ambient temperatures under nitrogen for 18 h. The DMF is removed under vacuum and the resulting residue is purified by preparative HPLC on a CI8 column using a water:ACN:0.1% 10 TFA gradient. The product fraction is lyophilized to give the title compound.
Part C - DOTA/2-(((4-(3-(N-(3-(2-(2-(3-(2-Amino-4-(N-(6-deoxy-p-cyclodextryl)carbamoyl)butanoylamino)-
15 propoxy)ethoxy)ethoxy)propyl)carbamoyl)propoxy) - 2, 6 -dimethylphenyl)sulfonyl)amino)-3-((7-((imidazol-2-ylamino)methyl)-l-methyl-4-oxo(3-hydroquinolyl))-carbonylamino)propanoic Acid Bis(trifluoroacetate) Salt Conjugate
20 The title compound is prepared by the same procedure described for Example 10 by substituting Boc-Glu(6-amino-6-deoxy-P-cyclodextryl)-OSu for Boc-Cys(O3H)-OSu.

25
Example 15 DOTA/2- ({ (4- (3- (N- (3- (2- (2- (3- (2-Amino-4- (N- (co¬met hoxypolyethylene (5,000)glycoxyethyl)carbamoyl)-30 butanoylamino) propoxy) ethoxy) ethoxy) propyl) carbamoyl) -propoxy)-2, 6-dimethylphenyl)sulfonyl)amino)-3-((7-


((imidazol-2-ylamino)methyl)-l-methyl-4-oxo(3-
hydroquinolyl))carbonylamino)propanoic Acid
Bis(trifluoroacetate) Salt Conjugate
5 Part A - Preparation of Boc-Glu(amino-(i>-methoxypolyethylene glycol)-OMe
A solution of Boc-Glu-OMe, amino-co-methoxypolyethylene glycol, (MW = 5,000), DIEA, and HBTU in anhydrous DMF is stirred at ambient temperatures under 10 nitrogen for 18 h. The DMF is removed under vacuum and the resulting residue is purified by preparative HPLC on a C18 column using a water:ACN:0.1% TFA gradient. The product fraction is lyophilized to give the title compound.
15
Part B - Preparation of Boc-Glu(amino-o>-methoxypolyethylene glycol)-OSu
The product of Part A, above, is hydrolyzed by stirring in a mixture of LiOH, THF, and water at ambient
20 temperatures under nitrogen for 4 h. The THF is removed under vacuum and the resulting solution is adjusted to pH 7 using 0.1 N HC1. The solution is desalted using a Sephadex PD-10 desalting column and the product eluant is lyophilized. The resulting material is dissolved in
25 anhydrous DMF along with N-hydroxysuccinimide, and DCC, and stirred at ambient temperatures under nitrogen for 18 h. The DMF is removed under vacuum and the resulting residue is purified by preparative HPLC on a C18 column using a water:ACN:0.1% TFA gradient. The product
30 fraction is lyophilized to give the title compound.
Part C - DOTA/2-U (4-(3-(N-(3-(2-(2-(3-(2-Amino-4-(N- (co-methoxypolyethylene (5, 000)glycoxyethyl) carbamoyl) -butanoylamino) propoxy) ethoxy) ethoxy) propyl) carbamoyl) -35 propoxy) -2, 6-dimethylphenyl) sulfonyl) amino) -3- ((7-((imidazol-2-ylamino)methyl) -l-methyl-4-oxo{3-hydroguinolyl) )carbonylamino)propanoic Acid Bis(trifluoroacetate) Salt Conjugate


The title compound is prepared by the same procedure described for Example 10 by substituting Boc-Glu(amino-(i>-methoxypolyethylene glycol)-OSu for Boc-Cys(O3H)-OSu.



Example 16
2-{((4-(3-(N-(3-(2-(2-(3-(2-(l,4,7,10-Tetraaza-4,7,10-
10 tris(carboxymethyl)cyclododecylacetylamino)-6-
aminohexanoylaminojpropoxy)ethoxy)ethoxy)-
propyl)carbamoyl)propoxy) - 2, 6 -
dimethylphenyl)sulfonyl)amino)-3-((7-((imidazol-2-
ylamino)methyl)-l-methyl-4-oxo(3-hydroquinolyl) ) -
15 carbonylamino)propanoic Acid Tris(trifluoroacetate) Salt
The title compound is prepared by the same procedure described for Example 10 by substituting Boc-Lys(Cbz)-OSu for Boc-Cys(O3H)-OSu.


20

Example 17 ~±49—

2-(((4-(3-(N-(3-(2-(2-(3-(2-(l,4,7,i0-Tetraaza-4,7,10-
tris (carboxymethyl) cyclododecylacetylamino ) -6- (2-
(bis (phosphonomethyl) amino) acety 1 amino)hexanoylamino) -
propoxy) ethoxy) ethoxy) propyl) carbamoyl) propoxy) -2,6-
5 dimethylphenyDsulfonyl) amino) -3- ( (7- ( (imidazol-2-
ylamino) methyl) -1-methyl-4-oxo (3-hydroquinolyl) ) -
carbonylamino)propanoic Acid Conjugate
A solution of bis(phosphonomethyl)glycine, DIEA, and 10 HBTU in anhydrous DMF is stirred at ambient temperatures under nitrogen for 15 min, and treated with the product of Example 16. Stirring is continued for 18 h and the DMF is removed under vacuum. The resulting residue is purified by ion exchange chromatography.
15

Example 18
20 2-(((4-(3-(N-(3-(2-(2-(3-(2-(2-((2-((2-
(bis(carboxymethyl)-
amino) ethyl) (carboxymethyl) amino) ethyl) (carboxymethyl) ami
no)acetylamino)-3-sulfopropyl)propoxy)ethoxy) -
ethoxy) propyl) carbamoyl) propoxy) -2, 6-dimethylphenyl) -
25 sulfonyl)amino)-3-((7-((imidazol-2-ylamino) methyl)-1-
methyl-4-oxo(3-hydroquinolyl)) carbonylamino) propanoic
Acid

The product of Example 10, Part A is dissolved in degassed TFA and stirred at ambient temperatures for 15 min. The solution is concentrated under vacuum, and the resulting residue is dissolved in 50% ACN and lyophilized 5 to remove the last traces of TFA. The material is dissolved in anhydrous DMF along with DIEA and diethylenetriaminepentaacetic dianhydride. The resulting solution is stirred at ambient temperatures under nitrogen for 18 h. The DMF is removed under vacuum and 10 the resulting residue is purified by preparative HPLC on a C18 column using a water:ACN:0.1% TFA gradient. The product fraction is lyophilized to give the title compound.

V|J 7 fQ2 HOOC^N^COOH
Ns^COOH
15 HO3S' ° COOH
The following procedure describe the synthesis of radiopharmaceuticals of the present invention of the
20 formula 99mTc(VnA)(tricine)(phosphine), in which (VnA) represents a vitronectin receptor antagonist compound of the present invention bonded to the Tc through a diazenido (-N=N-) or hydrazido (=N-NH-) moiety. The diazenido or hydrazido moiety results from the reaction
25 of the hydrazinonicotinamido group, present either as the free hydrazine or protected as a bydrazone, with the Tc-99m. The other two ligands in the Tc coordination sphere are tricine and a phosphine.
30 Examples 19-23
Synthesis of Complexes [99mTc(HYNlc-VnA)(tricine)(TPPTS)].
To a lyophilized vial containing 4.84 mg TPPTS, 6.3 mg tricine, 40 mg mannitol, succinic acid buffer, pH 4.8,


10

and 0.1% Pluronic F-64 surfactant, was added 1.1 mL sterile water for injection, 0.2 mL (20 ug.) of the appropriate HYNIC-conjugated vitronectin antagonist (VnA) in deionized water or 50% aqueous ethanol, and 0.2 mL of 99mrco4- (50±5 mCi) in saline. The reconstituted kit was
heated in a
100°C water bath for 15 minutes, and was allowed to cool 10 minutes at room temperature. A sample of the reaction mixture was analyzed by HPLC. The RCP results are listed in the Table 1.



15

HPLC Method
Column: Zorbax CI8, 25 cm x 4.6 mm
Flow rate : 1.0 mL/min
Solvent A: 10 mM sodium phosphate buffer, pH 6.0
Solvent B : 100 % CH3CN



20


Gradient A (Exs. 19 , 20, 21)
t (min) 0 20 21 30 31 40
% Solvent B 0 25 75 75 0 0
Gradient B (Ex. 22)
t (min) 0 20 30 31 40
% Solvent B 0 50 50 0 0

25 Gradient C (Ex. 23)
t (min) 0 20 21 30 31 40
% Solvent B 10 30 75 75 0 0
Table 1. Analytical and Yield Data for 30 99mTc(VnA)(tricine)(TPPTS) Complexes

Example No. Reagent No. Ret. Time (min) % Yield
19 1 8.8 73
20 3 17.2 81
21 4 17.6 68
22 6 11.7 79
23 7 16.4 52


Example 24 Synthesis of the In-Ill Complex of 3-((7-((Imidazol-2-ylamino)methyl)-l-methyl-4-oxo(3-5 hydroquinolyl))carbonylamino)-2-( ( (4-(4-(((3-(2-(2-(3-(2-(l,4,7,10-tetraaza-4,7,10-
tris(carboxylmethyl)cyclododecyl)acetylamino)-propoxy)ethoxy)ethoxy)propyl)amino)sulfonyl)-phenyl)phenyl)sulfonyl)amino)propanoic Acid
10 To a lead shielded and crimped autosampler vial was added 35 \ig of the conjugate of Example 2 and 1.0 mg gentisic acid, sodium salt dissolved in 70 p.L ammonium acetate buffer (0.4 M, pH 4.7) followed by the addition of 2 mCi , 20 uL In-Ill in 0.05 N HC1 (specific activity:
15 17 ug/mCi) . The reaction mixture was heated at 70 - 80 °C for 60 min and analyzed by HPLC and ITLC. The complex was formed in 93% yield and had a retention time of 19.6 min.
20 HPLC Method
Column: Zorbax Rx C18, 2 5 cm x 4.6 mm
Column Temperature: Ambient
Flow: 1.0 mL/min
Solvent A: 10 % Acetonitrile/0.1%TFA/H20 25 Solvent B: Acetonitrile
Detector: Sodium iodide (Nal) radiometric probe
Gradient
t (min) 0 25 26 35 36 45
%B 10 20 60 60 10 10 30
Examples 25-26
Synthesis of 177Lu and 90Y Complexes of 3-((7-((Imidazol-
2-ylamino)methyl)-l-methyl-4-oxo(3-
hydroquinolyl))carbonylamino)-2-( ( (4- (4-(((3-(2-(2-(3-(2-35 (1,4,7,10-tetraaza-4,7,10-
tris (carboxylmethyl) cyclododecyl) acetylamino) -
propoxy) ethoxy) ethoxy) propyl) amino) sulfonyl) -
phenyl) phenyl) sulfonyl) amino) propanoic Acid.


To a clean sealed 5 mL vial was added 0.3 mL of a solution of the comjugate of Example 2 (200 ug/mL in 0.5 M ammonium acetate buffer, pH 6.9), followed by 0.05 mL of gentisic acid (sodium salt, 10 mg/mL in 0.5 M ammonium 5 acetate buffer, pH 6.9) solution, 0.3 mL of 0.5 M
ammonium acetate buffer (pH 6.9), and 0.010 mL of l LuCl3 or 90YCl3 solution (1000 mCi/mL for 177LuCl3 and 500 mCi/mL for 90YCl3) in 0.05 N HC1. The resulting mixture was heated at 100 °C for 30 min. After cooling to room
10 temperature, a sample of the resulting solution was analyzed by radio-HPLC and ITLC. The radiolabeling yields were = 90% (after correction for small amount of colloid) for both complex, and the retention time was 19.2 min.
15
HPLC Method
Column: Zorbax C18 , 25 cm x 4.6 mm Flow rate : 1.0 mL/min Solvent A: 0.1% TFA aqueous solution 20 Solvent B : 100 % CH3CN
t (min) 0 20 25 30 31 40
% Solvent B 10 25 60 60 10 10
The instant thin layer chromatography (ITLC) method used Gelman Sciences silica-gel strips and a 1:1 mixture of 25 acetone and saline as eluant.
Example 27 Synthesis of 177Lu Complex of the DOTA Conjugate of 3-((l-(3-(3-(N-(3-(2-(2-(N-(L-Asp-L-Asp)3-aminopropoxy)-ethoxy)ethoxy)propyl)carbamoyl)propanoylamino)propyl-7 -
30 ((imidazole-2-ylamino)methyl)-4-oxo(3-
hydroquinolyl))carbony1amino)-2-({(2, 4, 6-trimethylphenyDsulfonyl) amino) propanoic Acid.
To a clean sealed 5 mL vial was added 0.5 mL of a solution of the conjugate of Example 8 (200 ug/mL in 0.5
35 M ammonium acetate buffer, pH 6.9), followed by 0.05 mL of gentisic acid (sodium salt, 10 mg/mL in 0.5 M ammonium


acetate buffer, pH 6.9) solution, 0.25 mL of 0.5 M ammonium acetate buffer (pH 6.9), and 0.05 mL of * LuCl3 solution (200 mCi/mL) in 0.05 N HC1. The resulting mixture was heated at 100 °C for 30 min. After cooling to 5 room temperature, a sample of the resulting solution was analyzed by radio-HPLC and ITLC. The radiolabeling yield was 75% (after correction for colloid), and the retention time was 20 min.
10 HPLC Method
Column: Zorbax CI8 , 25 cm x 4.6 mm
Flow rate : 1.0 mL/min
Solvent A: 10 mM phosphate buffer, pH = 6
Solvent B : 100 % CH3CN
15 t (min) .0 20 25 30 31 40
% Solvent B 0 20 50 50 0 0
Example 28 Synthesis of the Gadolinium Complex of 2-(((4-(3-(N-(3-
20 (2-(2-(3-(2-(2-((2-( (2-(bis(carboxymethyD-
amino) ethyl) (carboxymethyl)amino) ethyl) (carboxymethyl) ami no)acetylamino)-3-sulfopropyl)propoxy)ethoxy)-ethoxy) propyl) carbamoyl) propoxy) -2 , 6-dimethylphenyl) -sulfonyl)amino)-3-((7-((imidazol- 2-ylamino)methyl)-1-
25 methyl-4-oxo(3-hydroquinclyl) ) carbonylamino)propanoic Acid
The gadolinium complex of the conjugate of Example 18 is prepared according to the following procedure. 3-3.5 mg of the conjugate is dissolved in 2 mL 1 M ammonium
30 acetate buffer at pH 7.0 , and one equivalent Gd(N03)3 solution (0.02 M in water) is added to it. The reaction mixture is allowed to stay at room temperature for 3-5 hours and the product is isolated by HPLC. The fraction containing the complex is lyophilized and dissolved in 1
35 mL H2O. The identity of the complex is confirmed by mass spectroscopy.

Example 29 Synthesis of (2S)-2-[({2,6-Dimethyl-4- [3-(N-{2-[3-sulfo-2- (3-sulfo-2-{2-[l,4,7,10-tetraaza-4,7,10-tris(carboxymethyl)
cyclododecyllacetylamino)propyl)propyl]ethyl)carbamoyl)-propoxy]phenylJsulfonyl)amino)-3-({7- [ (imidazol-2-ylamino)methyl]~l-methyl-4-oxo(3-hydroquinolyl)}-carbonylamino)propanoic Acid Trifluoroacetate Salt


10

Part A - Preparation of Methyl (2S)-3-[ (tert-Butoxy)-15 carbonylamino]-2-[({2, 6-dimethyl-4-[3- (N-{2-
[ (phenylmethoxy) carbonylamino) ethyl} carbamoyl) propoxy] -phenyl}sulfonyl)amino]propanoate

20
A solution of the product of Example 3, Part D (369 mg, 0.756 irnnol), DIEA (0.52 mL, 3.0 mmol), and HBTU (315 mg, 0.832 mmol) in anhydrous DMF (14 mL) was stirred at ambient temperatures under nitrogen for 5 min, and 25 treated with benzyl N-(2-aminoethyl)carbamate
hydrochloride (192 mg, 0.832 mmol), and stirred an additional 1 h. The DMF was removed under vacuum, and

acid (425 mg, 1.03 mmol), and DIEA (0.435 mL, 2.55 mmol) in anhydrous DMF (20 mL) was stirred at ambient temperatures under nitrogen for 3 h. The DMF was removed under vacuum and the resulting oil was purified by HPLC 5 on a Vydac C-18 column (50 x 250 mm) using a 1.12%/min gradient of 9 to 54% ACN containing 0.1% TFA at a flow rate of 80 mL/min. The main product peak eluting at 37.3 min was lyophilized to give the title compound as a colorless solid (410 mg, 80.2%). MS: m/e 1204.4 [M+H], 10 962.3 [M+H-Trt].
Part F - Preparation of (2R) -N- {2- [4- (4-{ [ ((IS) -1-(Methoxycarbonyl)-2-{ [l-methyl-4-oxo-7- ({[1-(triphenylmethyl) imidazol-2-yl]amino}methyl) (3-15 hydroguinolyl) ] carbonylamino}ethyl) amino] sulfonyl) -3", 5-dimethylphenoxy)butanoylamino]ethyl}-2-aminopropanesulfonic Acid

20
The product of Part E, above (410 mg, 0.341 mmol) was dissolved in 50/50 TFA/DCM (20 mL) and allowed to react at ambient temperatures for 10 min. The solution was concentrated and the resulting amber oil was 25 dissolved in 50% ACN (50 mL) and lyophilized to give the title compound as a colorless solid (371 mg, 98.6%). MS: m/e 1104.4 [M+H], 862.3 [M+H-Trt]; High Resolution MS: Calcd for C55H62N9O12S2 [M+H]: 1104.3959; Found: 1104.393.
30 Part G - Preparation of (2R) -N- [ (1R) -1- (N-{2- [4- (4-
{[ ((lS)-l-(Methoxycarbonyl)-2-{ [l-methyl-4-oxo-7- ({[1-
hydroguinolyl) ] carbonylamino) ethyl) amino] sulfonyl} -3, 5-


dimethylphenoxy) butanoylamino] ethyl} carbamoyl) -2-sulfoethyl] -2- [ (tert-butoxy) carbonylamino] propanesulfonic Acid

A solution of the product of Part F, above (110 mg, 0.100 mmol), the p-nitrophenyl ester of Boc-L-cysteic acid (82.4 mg, 0.200 mmol), and DIEA (0.104 mL, 0.600 10 mmol) in anhydrous DMF (5.0 mL) was stirred at ambient temperatures under nitrogen for 48 h. The DMF was removed under vacuum and the resulting amber oil was purified by HPLC on a Vydac C-18 column (50 x 250 mm) using a 1.12%/min gradient of 9 to 54% ACN containing
15 0.1% TFA at a flow rate of 80 mL/min. The main product peak eluting at 37.0 min was lyophilized to give the title compound as a colorless solid (96.0 mg, 70.9%). MS: m/e 1355.3 [M+H], 1113.3 [M-Trt+H]. 1013.2 [M-Trt-BOC+H].
20
Part H - Preparation of (2R)-N-[(1R)-1-(N-[2-[4-(4-{[((lS)-l-(Methoxycarbonyl)-2-{ [l-methyl-4-oxo-7- ({[1-(triphenylmethyl) imidazol-2-yl] amino)methyl) (3-hydroquinolyl) ] carbonylamino)ethyl) amino] sulfonyl)-3, 5-
25 dimethylphenoxy) butanoylamino] ethyl) carbamoyl) -2-sulfoethyl] -2-aminopropanesulfonic Acid



The product of Part G, above (21 mg, 0.0155 mmol) was dissolved in 50/50 TFA/DCM (5.0 mL) and allowed to 5 react at ambient temperatures for 10 min. The solution was concentrated and the residue was taken up in 50% ACN (15 mL) and lyophilized to give the title compound as a colorless solid (18.7 mg, 96.2%). MS: m/e 1255.3 [M+H], 1013.2 [M+H-Trityl]; High Resolution MS: Calcd for 10 C58H67N10O16S3 [M+H]: 1255.3899; Found: 1255.391.
Part I - Preparation of (2R)-N-[(1R)-1-(N-{2-[4-(4-{[((lS)-l-(Methoxycarbonyl)-2-{[l-methyl-4-oxo-7-({[1-(triphenylmethyl)imi dazol-2-yl]amino}methyl)(3-15 hydroquinolyl) ] carbonylamino}ethyl) amino] sulfonyl}-3, 5-dimethylphenoxy)butanoylamino]ethyl}carbamoyl)-2-sulfoethyl]-2-(2-(1,4,7,10-tetraaza-4,7,10-tris[(tert-butoxycarbony1)methyl]cyclododecyl}acetylamino) propanesulfonic Acid
20

A solution of 2- (1,4,7,10-tetraaza-4,7,10-tris (((tert-butyl)oxycarbonyl) methyl) cyclododecyl) acetic 25 acid (30.0 mg, 0.0327 mmol) (as described in DM-7003), DIEA (0.034 mL, 0.196 mmol), and HBTU (9.3 mg, 0.0245

mmol) in anhydrous DMF (1.5 mL) was stirred under nitrogen at ambient temperatures for 15 min and treated with the product of Part H, above (18.7 mg, 0.0137 mmol). The DMF was removed under vacuum after 75 min and the 5 resulting amber oil was purified by HPLC on a Vydac C-18 column (22 x 250 mm) using a 0.9%/min gradient of 22.5 to 58.5% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 26.1 min was lyophilized to give the title compound as acolorless 10 fluffy solid (7.5 mg, 53%). MS: m/e 1809.7 [M+H].
Part J - Preparation of (2S)-2-[({2,6-Dimethyl-4-[3-(N-{2-[3-sulfo-2-(3-sulfo-2-{2-[1,4,7,lO-tetraaza-4,7,10-tris (carboxymethyl) cyclododecyl ] acetylamino)propyl) -15 propyl] ethyl} carbamoyl )propoxy] phenyl Jsulfonyl) amino] -3-({7-[(imidazol-2-ylamino)methyl]-l-methyl-4-oxo(3-hydroquinolyl)}carbonylamino)propanoic Acid Trifluoroacetate Salt
20 The product of Step I, above (7.5 mg, 0.0039 mmol) was dissolved in a solution of peroxide-free THF (1.40 mL) and water (0.21 mL), and treated with 3 N LiOH (0.14 mL). The mixture was stirred at ambient temperatures under nitrogen for 1 h, and concentrated to dryness under
25 vacuum. The resulting solid residue was dissolved in 95/5 TFA/Et3SiH (2.0 mL) and heated at 70 °C under nitrogen for 1 h. The solution was concentrated under vacuum and the resulting solid residue was purified by HPLC on a Vydac C-18 column (22 x 250 mm) using a
30 0.90%/min gradient of 0 to 27% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 20.5 min was lyophilized to give the title compound as a colorless fluffy solid (4.2 mg, 71.9%). MS: m/e 1385.3 [M+H]; High Resolution MS: Calcd for C54H77N14O23S3 [M+H]:
35 1385.4448; found: 1385.446.

Example 30
Synthesis of DOTA/(2S) -2-{ t(4-{3- [N-(2-{2-[(4S)-4-(N-{1-
[N- (2-{4- [4- ({[ (IS) -l-Carboxy-2- ({7- [ (imidazol-2-
ylamino)methyl] -l-methyl-4-oxo (3-hydroquinolyl) }-
5 carbonylamino)ethyl]aminoisulfonyl) -3, 5-dimethylphenoxy] -
butanoylamino)ethyl) carbamoyl] -2-sulfoethyl}carbamoyl) -4-
aminobutanoylamino]-3-
sulfopropyl)ethyl) carbamoyl]propoxy} -2 , 6-
dimethylphenyl)sulfonyl]amino)-3- ({7- [ (imidazol-2-
10 ylamino)methyl] -l-methyl-4-oxo(3-hydroquinolyl) }-
carbonylamino)propanoic Acid Conjugate
Bis(trifluoroacetate) Salt


15

Part A - Preparation of Di-2, 3, 5, 6-tetrafluorophenyl (2S) -2- [ (tert-Butoxy)carbonylamino]pentane-1, 5-dioate

20
To a solution of Boc-L-Glu-OH (28.9 g, 117 mmol) in DMF (500 mL) at ambient temperatures and under nitrogen, was added a solution of 2, 3, 5, 6-tetrafluorophenol (48.2


g, 290 mmol) in DMF (50 mL). After stirring for 10 min, EDC (55.6 g, 290 mmol) was added and the mixture was stirred for 96 h. The volatiles were removed under vacuum and the residue was triturated with 0.1 N HCl (750 5 mL) . To this mixture was added EtOAc (600 mL) and the layers were separated. The aqueous layer was extracted with EtOAc (3 x 500 mL), and all EtOAc extracts were combined, washed consecutively with water (300 mL) and saturated NaCl (300 mL), dried (MgSC>3) , and concentrated 10 to give a tan solid (62 g) . The tan solid was washed with ACN to give the title compound (45.5 g, 73.0%) in purified form. MS: m/e 566.0 [M+Na].
Part B - Preparation of (2R)-2-[4-(N-{(1R)-1-[N-(2-{4-[4-15 ({ [ (lS)-2-({7-[([l-(Triphenylmethyl)imidazol-2-
yl]amino)methyl]-1-methyl-4-oxo(3-hydroquinolyl)}-carbonylamino)- 1-(methoxycarbonyl)ethyl]amino}sulfonyl) -3 , 5-dimethylphenoxy]butanoylamino}ethyl)carbamoyl]-2-sulfoethyl)carbamoyl)(4S)-4-[(tert-butoxy)carbonylamino] -20 butanoylamino]-N-(2-{4-[4-({[(IS)-2-({7-[(imidazol-2-ylamino)methyl]-1-methyl-4-oxo(3-hydroquinolyl)}-carbonylamino) -1- (methoxycarbonyl) ethyl] amino}sulfonyl) -3, 5-dimethylphenoxy]butanoylamino}ethyl)propanesulfonic Acid
25


A solution of the product of Example 29, Part F (130 mg, 0.118 mmol), the product of Part A, above Part C - Preparation of (2R) -2- [4- (N- { (1R) -1- [N- (2- (4- [4-15 ({ [ (IS) -2- ({7- [ ([1- (Triphenylmethyl) imidazol-2-
yl]amino)methyl]-l-methyl-4-oxo(3-hydroquinolyl) }-carbonylamino) - 1 - (methoxycarbonyl) ethyl ] amino} sulf ony 1) -3, 5-dimethylphenoxy]butanoylamino}ethyl) carbamoyl] -2-sulfoethyl}carbamoyl) (4S) -4-aminobutanoylamino] -N- (2-{4-20 [4-({[ (lS)-2-({7-[ (imidazol-2-ylamino)methyl]-l-methyl-4-oxo(3-hydroquinolyl)}carbonylamino)-1-(methoxycarbonyl)ethyl]amino}sulfonyl) -3 , 5-dimethylphenoxy] butanoylamino} ethyl) propanesulf onic Acid


f*l 25 O O O

The product of Part B, above (107 mg, 0.0442 mmol) was dissolved in 50/50 TFA/DCM (5.0 mL) and allowed to react at ambient temperatures under nitrogen for 10 min. The solution was concentrated and the resulting amber oil 5 was dissolved in 50% ACN (25 mL) and lyophilized to give the title compound as a pale yellow solid (105 mg, 98.0%). MS: m/e 1159.9 [M+2H], 1039.4 [M+2H-Trt].
Part D - Preparation of DOTA tri-t-Butyl Ester/(2R)-2-[4-10 (N-{(lR)-l-[N-(2-{4-[4-({[(lS)-2-({7-[([l-
(Triphenylmethyl! imidazol-2-yl] amino)methyl] -l-methyl-4-oxo(3~hydroquinolyl)}carbonylamino)-1-(methoxycarbonyl)ethyl]amino}sulfonyl)-3,5-dimethylphenoxy]butanoylamino}ethyl) carbamoyl] -2-15 sulfoethyl)carbamoyl) (4S) -4-aminobutanoylamino] -N- (2- (4-[4- ({[ (IS) -2- ({7- [ (imidazol-2-ylamino)methyl] -l-methyl-4-oxo(3-hydroquinolyl))carbonylamino)-1-(methoxycarbonyl)ethyl]amino}sulfonyl)-3,5-dimethylphenoxy]butanoylamino}ethyl) propanesulfonic Acid 20 Conjugate

A solution of 2-(1,4,7,10-tetraaza-4,7,10-25 tris (((tert-butyl) oxycarbonyl) methyl) cyclododecyl) acetic acid (31.6 mg, 0.0346 mmol) (as described in DM-7003), DIEA (0.072 mL, 0.416 mmol), and HBTU (9.8 mg, 0.026

mmol) in anhydrous DMF (1.8 mL) was stirred under nitrogen at ambient temperatures for 15 min and treated with the product of Part C, above (40.0 mg, 0.0173 mmol). The DMF was removed under vacuum after 90 min and the 5 resulting pale yellow oil was purified by HPLC on a Vydac C-18 column (50 x 250 mm) using a 1.01%/min gradient of 22.5 to 63.0% ACN containing 0.1% TFA at a flow rate of 80 mL/min. The main product peak eluting at 27.6 min was lyophilized to give the title compound as a colorless 10* solid (29.0 mg, 62.4%). MS: m/e 1437.6 [M+2H], 1316.6 [M+2H-Trt].
Part E - Preparation of DOTA/ 15 [(imidazol-2-ylamino)methyl]-l-methyl-4~oxo(3-
hydroquinolyl)Jcarbonylamino)ethyl]amino}sulfonyl)-3,5-dimethylphenoxy]butanoylamino}ethyl)carbamoyl ] -2-sulfoethyl}carbamoyl)-4-aminobutanoylamino]-3-sulfopropyl}ethyl)carbamoyl]propoxy)-2,6-
20 dimethylphenyl)sulfonyl]amino)-3-({7-[(imidazol-2~ ylamino)methyl]-1-methyl-4-oxo(3-hydroquinoly 1)}-carbonylamino)propanoic Acid Conjugate Bis(trifluoroacetate) Salt
25 A mixture of the product of Part D, above (30.0 mg, 0.0104 mmol), peroxide-free THF (3.2 mL), water (0.485 mL), and 3 N LiOH (0.320 mL, 0.96 mmol) was stirred at ambient temperatures under nitrogen for 2 h. The solution was concentrated under vacuum and the resulting
30 solid residue was dissolved in 95/5 TFA/Et3SiH (5.0 mL) . The solution was heated at 70 °C under nitrogen for 1 h and concentrated under vacuum. The resulting oily solid was purified by HPLC on a Vydac C-18 column (22 x 250 mm) using a 0.90%/min gradient of 0 to 27% ACN containing
35 0,1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 27.8 min was lyophilized to give the title compound as a cololess fluffy solid (12.8 mg, 48.5%). MS: m/e 1096.8 [M+2H], 731.8 [M+3H]; High


Resolution MS: Calcd for C91H122N23O33S4 [M+H] : 2192.7458; Found: 2192.741.
5 Example 31
Synthesis of 2-[({4-[3-(N-{2-[(2R)-2-((2R)-3-Sulfo-2-{2-
(1,4,7,10-tetraaza-4,7,10-
tris (carboxymethyl)cyclododecyl] acetylamino}propyl)-3 -
sulfopropyl]ethyl}carbamoyl)propoxy]-2,6-
10 dimethylphenyl}sulfonyl)amino](2S)-3-({7-[(imidazol-2-
ylamino)methyl]-l-methyl-4-oxo(3-hydroquinolyl)}-
carbonylamino)propanoic Acid Trifluoroacetate Salt

15
Part A - Preparation of 2-({ [4-(3-{N-[2-((2R)-2-Amino-3-sulfopropyl)ethyl]carbamoylJpropoxy)-2,6-dimethylphenyl]-sulfonyl}amino)(2S)-3-{[l-methyl-4-oxo-7-({[1-(triphenylmethyl)imidazol-2-yl]aminojmethyl)(3-20 hydroguinolyl)]carbonylamino}propanoic Acid

A mixture of the product of Example 29, Part F (12 5 25 mg, 0.113 mmol), peroxide-free THF (3.8 mL), water (0.57 mL), and 3 N LiOH (0.38 mL, 1.13 mmol) was stirred at ambient temperatures under nitrogen for 1 h. The mixture

was adjusted to pH 1 using 1 N HC1 (0.70 mL) and concentrated to dryness under vacuum. The resulting solid was purified by HPLC on a Vydac C-18 column (50 x 250 mm) using a 0.90%/min gradient of 18 to 54% ACN 5 containing 0.1% TFA at a flow rate of 80 mL/min. The main product peak eluting at 21.0 min was lyophilized to give the title compound as a colorless solid (96.0 mg, 77.9%). MS: m/e 1090.3 [M+H], 848.2 [M+H-Trt]; High Resolution MS: Calcd for C54H60N9O12S2 [M+H]: 1090.3808; 10 Found: 1090.381.
Part B - Preparation of 2-({[4-(3-{N-[2-((2R)-2-{(2R)-2-[(tert-Butoxy)carbonylamino]-3-sulfopropyl}-3-sulfopropy1)ethyl]carbamoyl}propoxy) - 2, 6 -15 dimethylphenyl] sulfonyl}amino) (2S) -3- { [l-methyl-4-oxo-7-({[1-(triphenylmethyl)imidazol-2-yl3amino)methyl) (3-hydroquinolyl)]carbonylamino}propanoic Acid

20
A solution of Boc-L-cysteic acid (37.0 mg, 0.128 mmol), DIEA (0.040 mL, 0.228 mmol), and PyBOP (53.0 mg, 0.102 mmol) in anhydrous DMF (1.0 mL) was stirred at ambient temperatures under nitrogen for 15 min, and added
25 to a solution of the product of Part A, above (93.0 mg, 0.0854 mmol) and DIEA (0.045 mL, 0.256 mmol) in anhydrous DMF (3.0 mL) . The resulting solution was stirred at ambient temperatures under nitrogen for 1.5 h and concentrated to a viscous amber oil. Purification by
30 HPLC on a Vydac C-18 column (50 x 250 mm) using a
0.68%/min gradient of 18 to 45% ACN containing 0.1% TFA at a flow rate of 80 mL/min. The main product peak

eluting at 36.4 min was lyophilized to give the title compound as a colorless solid (94.0 mg, 82.1%) . MS: m/e 1341.2 [M+H] , 1099.1 [M+H-Trt], 999.1 [M+H-Trt-Boc].
5 Part C - Preparation of 2- {[ (4- {3- [N- (2- {(2R) -2- [ (2R) -3-Sulfo-2-(2-{l,4,7,10-tetraaza-4,7,10-tris[ (tert-butoxycarbonyl} methyl ] cyclododecyl} acetylamino) propyl ] - 3 -sulf opropy 1} ethyl) carbamoyl] propoxy} -2, 6-dimethyIpheny 1) -sulfonyl]amino) (2S)-3-{ [l-methyl-4-oxo-7- ({[1-10 (triphenylmethyl) imidazol-2-yl]amino}methyl) (3-hydroquinolyl) jcarbonylamino}propanoic Acid

15 A solution of the product of Part B, above (90.0 mg, 0.0672 mmol) in 50/50 TFA/DCM (10.0 mL) was allowed to react at ambient temperatures under nitrogen for 10 min and concentrated under vacuum to give the intermediate amine as an amber oil. MS: m/e 1241.3 [M+H], 999.3 [M+H-
20 Trt]; High Resolution MS: Calcd for C57H65N10O16S3 [M+H]: 1241.3742; Found: 1241.375.
A solution of 2-(l,4,7,10-tetraaza-4,7,10-tris ( ((tert-butyl) oxycarbonyl)methyl) cyclododecyl) acetic acid (123 mg, 0.134 mmol) (as described in DM-7003), DIEA
25 (0.092 mL, 0.538 mmol), and PyBOP (52.4 mg, 0.101 mmol) in anhydrous DMF (1.5 mL) was stirred under nitrogen at ambient temperatures for 15 min, and added to a solution of the free amine produced above (90.0 mg, 0.0672 mmol) and DIEA (0.046 mL, 0.269 mmol) in anhydrous DMF (1.5
30 mL) . The DMF was removed under vacuum after 1 h and the resulting amber oil was purified by HPLC on a Vydac C-18 column (50 x 250 mm) using a 0.288%/min gradient of 30.6

to 45% ACN containing 0.1% TFA at a flow rate of 80 mL/min. The main product peak eluting at 25.8 min was lyophilized to give the title compound as a colorless solid (92.0 mg, 76.3%). MS: m/e 1795.6 [M+H], 1553.5 5 [M+H-Trt]; High Resolution MS: Calcd for C85H115N14O23S3 [M+H]: 1795.7422; Found: 1795.744.
Part D - Preparation of 2-[({4-[3- (N- {2-[(2R)~2~ ((2R)-3-Sulfo-2-{2-[l,4,7,10-tetraaza-4,7,10-
10 tris(carboxymethyl)cyclododecyl] acetylamino}propyl)-3-sulfopropyl]ethyl}carbamoyl)propoxy]~2, 6-dimethylphenyljsulfonyl)amino](2S)-3- ({7-[(imidazol-2-ylamino)methyl]-1-methyl-4-oxo(3-hydroquinolyl)}-carbonylamino)propanoic Acid Trifluoroacetate Salt
15
A solution of the product of Part C, above (89.0 mg, 0.0496 mmol) in 97/3 TFA/Et3SiH (10.0 mL) was heated at
70 °C under nitrogen for 3 0 min and concentrated under vacuum. The resulting oily solid was purified by HPLC on
20 a Vydac C-18 column (50 x 250 mm) using a 0.45%/min gradient of 4.5 to 22.5% ACN containing 0.1% TFA at a flow rate of 80 mL/min. The main product peak eluting at 19.5 min was lyophilized to give steochemically pure title compound as a colorless fluffy solid (65.0 mg,
25 87.5%). MS: m/e 1385.4 [M+H].
Example 32
Alternative Synthesis of Intermediate 2-({ [4-(3-{N-[2-
((2R) -2-Amino-3-sulfopropyl) ethyl] carbamoyl}propoxy) -2,6-
30 dimethylphenyl]sulfonyl]amino) (2S)-3-{ [l-methyl-4-oxo-7-
({[1- (triphenylmethyl) imidazol-2-yl]amino)methyl) (3-
hydroquinolyl)]carbonylamino}propanoic Acid



Part A - Preparation of (2S)-2-{[(4-{3-[N-(2-Aminoethyl)carbamoyl]propoxy}-2, 6-dimethylphenyi; 5 sulfonyl]amino}-3-{[l-methyl-4-oxo-7-({[1-
(triphenylmethyl)imidazol-2-yl]amino}methyl)(3-hydroquinolyl)]carbonylamino}propanoic Acid

10
A mixture of the product of Example 29, Part D (956 mg, 1.004 mmol), peroxide-free THF (35 mL), water (5.3 mL), and 3 N LiOH (3.53 mL, 10.6 mmol) was stirred at ambient temperatures under nitrogen for 1 h, and adjusted
15 to pH 5-6 using 1 N HC1 (10 mL) . The THF was removed
under vacuum causing a gummy yellow solid to precipitate. The water layer was removed by decantation and the solid was washed with water (15 mL). The solid was dried under vacuum to give the title compound as a dry yellow solid
20
Part B - Preparation of 2-{[(4-{3-[N-(2-{(2R)-2-[(tert-Butoxy) carbonylamino] -3-sulfopropyl}ethyl) carbamoyl] -propoxy} -2, 6-dimethylphenyl) sulfonyl] amino} (2S)-3-{ [1-methyl-4-oxo-7-({[1- (triphenylmethyl)imidazol-2-
25 yl]amino}methyl)(3-hydroquinolyl)]carbonylamino}propanoic Acid


A solution of Boc-L-cysteic acid (175 rng, 0.60 mmol), DIEA (0.208 mL, 1.20 mmol), and PyBOP (250 mg, 5 0.480 mmol) in anhydrous DMF (5.0 mL) was stirred at
ambient temperatures under nitrogen for 17 min, and added to a solution of the product of Part A, above (375 mg, 0.400 mmol) and DIEA (0.070 mL, 0.400 mmol) in anhydrous DMF (4.0 mL) . The resulting solution was stirred at
10 ambient temperatures under nitrogen for 45 min and concentrated under vacuum to give an amber oil. Purification by PLC on a Vydac C-18 column (50 x 250 mm) using a 0.292%/min gradient of 31.5 to 43.2% ACN containing 0.1% TFA at a flow rate of 80 mL/min. The
15 main product peak eluting at 22.0 min was lyophilized to give the title compound as a colorless solid (430 mg, 90.4%). MS: m/e 1190.3 [M+H], 948.3 [M+H-Trt] .
Part C - Preparation of 2-({[4-(3-{N-[2-((2R)-2-Amino-3-20 sulfopropyl) ethyl] carbamoyl Jpropoxy) -2 , 6-dimethylphenyl] -sulfonyl)amino)(2S)-3-{[l-methyl-4-oxo-7-({[1-(triphenylmethyl)imidazol-2-yl]amino)methyl)(3-hydroquinolyl)]carbonylamino}propanoic Acid
25 A solution of the product of Part B, above (430 mg, 0.3'62 mmol) in 50/50 TFA/DCM (15 mL) was allowed to react at ambient temperatures under nitrogen for 10 min and concentrated under vacuum. The resulting amber oil was taken up in 50% ACN (50 mL) and lyophilized to give the
30 title compound as a pale yellow solid (398 mg, 100%) . MS: m/e 1090.3 [M+H], 848.2 [M+H-Trt).


Example 3 3
Synthesis of D0TA/2-{[(4-{3-[N-(2-{(2R)-2-I(2R)-2-(4-{N-
[ (lR)-l-(N-{ (1R)-1-[N-(2-{4-[4-({[(IS)-l-Carboxy-2-({7-
[ (imidazol-2-ylamino)methyl] -1-methyl-4-oxo(3-
hydroquinolyl) }carbonylamino) -1-carboxyethyl]amino}-
sulfonyl) -3, 5-dimethylphenoxy] butanoylamino) ethyl) -
carbamoyl]-2-sulfoethyl}carbamoyl)-2-
sulfoethyl]carbamoyl)(2S) -2-aminobutanoylamino)-3-
sulfopropyl] -3-sulfopropyl)ethyl) carbamoyl]propoxy) -2, 6-
dimethylphenyl)sulfonyl]amino} (2S)-3-({7-[(imidazol-2-
ylamino)methyl]-l-methyl-4-oxo(3-hydroquinolyl)}-
carbonylamino)propanoic Acid Conjugate

0 0 O
15
Part A - Preparation of 2-{[(4-{3-[N-(2-{(2R)-2- [ (2R) -2-(4-{N-[(lR)-l-(N-{(lR)-l-[N-(2-{4-[4-({[(IS)-l-Carboxy-2-({7- [ ({1-(triphenylmethyl)imidazol-2-yl}amino)methyl]-1-methyl-4-oxo(3-hydroquinolyl)}carbonylamino)-1-
20 carboxyethyl]amino}sulfonyl)-3,5-
dimethylphenoxy]butanoylamino}ethyl) carbamoyl] -2-sulfoethyl}carbamoyl)-2-sulfoethyl]carbamoyl} (2S)-2-[ (tert-butoxy)carbonylamino]butanoylamino)-3-sulfopropyl] -3-sulfopropyl)ethyl) carbamoyl]propoxy} -2,6-
25 dimethylphenyl)sulfonyl]amino}(2S)-3-({7-[ ({1-


(triphenylmethyl)imidazol-2-yl}amino)methyl]-1-methyl-4 ■ oxo(3-hydroguinolyl)}carbonylamino)propanoic Acid

A solution of the product of the first half of Example 31, Part C (136 mg, 0.110 mmol), DIEA (0.076 mL, 0.44 mmol), and the product of Example 30, Part A (26.2 mg, 0.050 mmol) in anhydrous DMF (3.0 mL) was stirred at
10 ambient temperatures under nitrogen for 7 h. The DMF was removed under vacuum and the viscous amber oil was purified by HPLC on a Vydac C-18 column (50 x 250 mm) using a 0.45%/min gradient of 27 to 45% ACN, followed by a 0.72% gradient of 45-63% ACN containing 0.1% TFA at a
15 flow rate of 80 mL/min. The main product peak eluting at 75.2 min was lyophilized to give the title compound as a colorless solid (129 mg, 47.9%). MS: m/e 1347.3 [M+2H].
Part B - Preparation of DOTA tri-t-Butyl Ester Conjugate 20 Of 2-{[(4-{3-[N-(2-{(2R)-2-[(2R)-2-(4-{N-[(lR)-l-(N-
{{lR)-l-[N-(2-{4-[4-({[(lS)-l-Carboxy-2-({7~[({l-
(triphenylmethyl)imidazol-2-yl)amino)methyl]-1-methyl-4-
oxo'(3-hydroquinolyl) }carbonylamino) -1-
carboxyethyl] amino}sulfonyl)-3,5-25 dimethylphenoxy]butanoylamino}ethyl)carbamoyl]-2~


sulfoethyl}carbamoyl)-2-sulfoethyl]carbamoyl}(2S)-2-aminobutanoylamino)-3-sulfopropyl]-3-sulfopropyl}-ethy1)carbamoyl]propoxy}- 2,6-dimethylpheny1)sulfony1] amino}(2S)-3-({7-[({1-(triphenylmethyl)imidazol-2-yl}amino)methyl]-1-methyl-4-oxo(3-hydroquinolyl)}-carbonylamino)propanoic Acid

10 The product of Part A, above (34.0 mg, 0.0126 mmol) was dissolved in 50/50 TFA/DCK (12 mL) and allowed to react at ambient temperatures under nitrogen for 10 min. The solution was concentrated and the resulting amber oil was dried under vacuum.
15 A solution of 2-(1,4,7,10-tetraaza-4,7,10-
tris(((tert-butyl)oxycarbonyl)methyl)cyclododecyl)acetic acid (23.1 mg, 0.0253 mmol), DIEA (0.020 mL, 0.115 mmol), and PyBOP (9.8 mg, 0.019 mmol) in anhydrous DMF (2.0 mL) was stirred under nitrogen at ambient temperatures for 15
20 min, and added to a solution of the product from the deprotection reaction, above and DIEA (0.020 mL, 0.115 mmol) in anhydrous DMF (2.0 mL) . The DMF was removed under vacuum after 2 h, and the resulting residue was purified by HPLC on a Vydac C-18 column (50 x 2 50 mm)
25 using a 0.45%/min gradient of 27 to 49.5% ACN containing

0.1% TFA at a flow rate of 80 mL/min. The main product peak eluting at 43.8 min was lyophilized to give the title compound as a colorless solid (16.0 mg, 40.4%). MS: m/e 1574.8 [M+2H], 1453.7 [M+2H-Trt], 1332.2 [M+2H-5 2Trt].
Part C - Preparation of DOTA/2-{[(4-{3-[N-(2-{(2R)-2-[(2R)-2-(4-{N-[(lR)-l-(N-{(lR)-l-[N-(2-{4-[4-({[(lS)-l-Carboxy-2-({7- [ (imida2ol-2-ylamino)methyl]-1-methyl-4-
10 oxo(3-hydroquinolyl)}carbonylamino)-1-carboxyethyl]amino}sulfonyl)-3, 5-dimethylphenoxy]butanoylamino}ethyl)carbamoyl]-2-sulfoethyl}carbamoyl)-2-sulfoethyl]carbamoyl}(2S)-2-aminobutanoylamino)-3-sulfopropyl]-3-sulfopropyl}-
15 ethyl)carbamoylIpropoxy}-2,6-dimethylphenyl)sulfonyl] -
amino} (2S)-3-({7-[ (imidazol- 2 -ylamino) methyl] -l-methyl-4-oxo(3-hydroquinolyl)}carbonylamino)propanoic Acid Conjugate
20 The product of Part B, above (14.0 mg, 0.00445 mmol) was dissolved in 95/5 TFA/Et3SiH (8.0 mL) and heated at 70 °C under nitrogen for 1 h. The solution was concentrated under vacuum and the resulting yellow solid was purified by HPLC on a Vydac C-18 column (22 x 250 mm) 25 using a 0.9%/min gradient of 0 to 27% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 24.5 min was lyophilized to give the title compound as a colorless solid (8.2 mg, 73.9%). MS: m/e 1247.7 [M+2H]. 30
Example 34 Synthesis of (2S)-3-{[7-[(Imidazol-2-ylamino)methyl]-4-
oxo-l-(3-{2-[l,4,7,10-tetraaza-4,7,10-
tris(carboxymethyl)cyclododecyl]acetylamino}propyl) (3-
35 hydroquinolyl)]carbonylamino}-2-{[(2,4,6-
trimethylphenyl)sulfonyl]amino}propanoic Acid Tris(trifluoroacetate) Salt



Part A - Preparation of (2S)-3-({7-[(lmidazol-2-ylamino)methyl]-4-oxo-l-[3-(2-{l,4,7, iO-tetraaza-4,7,10-5 tris[(tert-
butoxycarbonyl) methyl ] cyclododecyl} acetylamino) propyl ] (3 ■ hydroquinolyl)}carbonylamino)-2-{[ (2,4,6-trimethylphenyl)sulfonyl]amino}propanoic Acid Tris(trifluoroacetate) Salt
10


o
0 O

o u
F3C OH
O
A,

t-Bu-02C—/ \ / v-C0rt-Bu A
F3C OH
A solution of 2-(1,4,7,10-tetraaza-4,7,10-tris(((tert-butyl)oxycarbonyl) methyl)cyclododecyl)acetic
15 acid (89 mg, 0.0974 mmol) (as described in DM-7003), DIEA (0.103 mL, 0.607 mmol), and HBTU (28.0 mg, 0.0735 mmol) in anhydrous DMF (1.0 mL) was stirred under nitrogen at ambient temperatures for 15 min and treated with a solution of the product of Example 4, Part H (30.0 mg,
20 0.049 mmol) in anhydrous DMF (1.0 mL) . The DMF was removed under vacuum after 3 h and the residue was purified by HPLC on a Vydac C-18 column (22 x 250 mm) using a 1.08%/min gradient of 18 to 72% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product

peak eluting at 17.5 min was lyophilized to give the title compound as a colorless solid (48.0 mg, 65.0%). MS: m/e 1164.7 [M+H].
5 Part B - Preparation of (2S)-3-{[7-[(lmidazol-2-
ylamino)methyl]-4-oxo-1-(3-{2-[1,4,7,10-tetraaza-4,7,10-tris(carboxymethyl)cyclododecyl]acetylamino}propyl)(3-hydroquinolyl)]carbonylamino} - 2 - {[(2,4,6-trimethylphenyl)sulfonyl]amino}propanoic Acid 10 Tris(trifluoroacetate) Salt
A solution of the product of Part A, above (48.0 mg, 0.0375 mmol) in 95/5 TFA/Et3SiH (2.1 mL) was stirred at
50 °C under nitrogen for 2 h. The solution was 15 concentrated under vacuum and the oily residue was purified by HPLC on a Vydac C-18 column (22 x 250 mm) using a 1.2%/min gradient of 0 to 36% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 18.6 min was lyophilized to give the title 20 compound as a colorless solid (25.7 mg, 51.2%). MS: m/e 996.5 [M+H]; High Resolution MS: Calcd for C45H62N11O13S [M+H]: 996.4249; Found: 996.4278.
Example 3 5
25 Synthesis of 3-({1-[3-((2R)-3-Sulfo-2-{2-[1,4,7,10-
tetraaza-4, 7,10-tris(carboxymethyl)cyclododecyl]-
acetylamino}propyl)propyl]-7-[(imidazol-2-
ylamino)methyl]-4-oxo(3-
hydroquinolyl)}carbonylamino)(2S)-2-{[(2,4,6-
30 trimethylphenyl)sulfonyl]amino}propanoic Acid
Bis(trifluoroacetate) Salt


Part A - Preparation of 3-{[1-(3-{(2R)-2-[ (tert-Butoxy)carbonylamino] -3-sulfopropyl}propyl) -7- [ (imidazol-5 2-ylamino)methyl]-4-oxo(3-
hydroquinolyl)]carbonylamino}(2S)-2-{[(2,4,6-trimethylphenyl)sulfonyl]amino}propanoic Acid

10
A solution of the product of Example 4, Part H (105 mg, 0.125 mmol), the N-hydroxysuccinimide ester of Boc-Cysteic acid (as described in Liebigs Ann. Chem. 1979, 776-783) (146 mg, 0.467 mmol), and DIEA (0.120 mL, 0.69
15 mmol) in anhydrous DMF (1.5 mL) was stirred at ambient temperatures under nitrogen for 24 h. The DMF was removed under vacuum and the resulting solid residue was purified by HPLC on a Vydac C-18 column (22 x 250 mm) using a 0.68%/min gradient of 9 to 36% ACN containing
20 0.1% TFA at a flow rate of 2 0 mL/min. The main product peak eluting at 30.3 min was lyophilized to give the title compound as a colorless solid (73.0 mg, 67.9%). MS: m/e 861.3 [M+H].

Part B - Preparation of 3-({1-[3-((2R)-2-Amino-3-sulfopropyl)propyl]-7-[(imidazol-2-ylamino)methyl]-4-oxo(3-hydroquinolyl)}carbonylamino) (2S)-2-{[(2,4,6-trimethylphenyl)sulfonyl]amino)propanoic Acid 5 Trifluoroacetate Salt

The product of Part B, above £70.0 mg, 0.0814 mmol) 10 was dissolved in 2:1 DCM/TFA (1.5 mL) and allowed to
react at ambient temperatures under nitrogen for 30 min. The solution was concentrated under vacuum and the amber oil was dissolved in 50% ACN (25 mL) and lyophilized to give the title compound as a colorless solid (70.8 mg, 15 99.5%). MS: m/e 761.2 [M+H]; High Resolution MS: Calcd for C32H41N8O10S2 [M+H]: 761.2387; Found: 761.2393.
Part C - Preparation of 3-[(1-{3-[ (2R)-3-Sulfo-2-(2-{1,4,7,10-tetraaza-4,7,10-trisl(tert-butoxycarbonyl)-
20 methyl]cyclododecyl}acetylamino)propyl]propyl}-7-[ (imidazol-2-ylamino)methyl]-4-oxo(3-hydroquinolyl))carbonylamino] (2S)-2-{[(2,4,6-trimethylphenyl)sulfonyl]amino]propanoic Acid Bis(trifluoroacetate) Salt
25


A solution of 2-(1,4,7,10-tetraaza-4,7,10-tris(((tert-butyl)oxycarbonyl)methyl)cyclododecyl)acetic 5 acid (20.8 mg, 0.0228 mmol) (as described in DM-7003), DIEA (0.006 mL, 0.034 mmol), and HBTU (6.5 mg, 0.0171 mmol) in anhydrous DMF (0.5 mL) was stirred under nitrogen at ambient temperatures for 5 min and treated with a solution of the product of Part B, above (10.0 mg,
10 0.0114 mmol) and DIEA (0.006 mL, 0.034 mmol) in anhydrous DMF (0.5 mL). Stirring was continued at ambient temperatures for 24 h, and the reaction was diluted with water (3.0 mL), treated with concentrated ammonium hydroxide (0.003 mL), and stirred an additional 10 min.
15 The solution was adjusted to pH 3 using 0.1 N HCl (6.0 mL) and diluted further with 10% ACN (5.5 mL). This solution was purified directly by KPLC on a Vydac C-18 column (22 x 250 mm) using a 0.68%/min gradient of 9 to 36% ACN containing 0.1% TFA at a flow rate of 20 mL/min.
20 The main product peak eluting at 3 6.0 min was lyophilized to give the title compound as a colorless solid (12.0 mg, 68.3%). MS: m/e 1315.6 [M+H].
Part D - Preparation of 3-({1-[3- ( (2R)-3-Sulfo-2-{2-25 [l,4,7,10-tetraaza-4,7,10-
tris(carboxymethyl)cyclododecyl]-
acetylamino}propyl)propyl]-7-[(imidazol-2-
ylamino)methyl]-4-oxo(3-
hydroquinolyl)Jcarbonylamino)(2S)-2-{[(2,4,6-30 trimethylphenyDsulfonyl] amino)propanoic Acid
Bis(trifluoroacetate) Salt


A solution of the product of Part C, above (12.0 mg, 0.00778 mmol) in 95/5 TFA/Et3SiH (1.0 mL) was stirred at ambient temperatures under nitrogen for 18 h. The 5 solution was concentrated under vacuum and the oily
residue was purified by HPLC on a Vydac C-18 column (22 x 250 mm) using a 1.2%/min gradient of 0 to 36% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 21.1 min was lyophilized to 10 give the title compound as a colorless solid (8.1 mg,
75.7%). MS: m/e 1147.3 [M+H]; High Resolution MS: Calcd for C48H67N12O17S2 [M+H]: 1147.4189; Found: 1147.418.
Example 3 6 15 Synthesis of 3-{[1- (3-{2-[(6-{[(IE)-l-Aza-2-(2-
sulfophenyl)vinyl]amino}(3-pyridyl))carbonylamino](2R)-3-
sulfopropyl}propyl)-7- [ (imidazol-2-ylamino)methyl)-4-
oxo(3-hydroquinolyl)]carbonylamino)(2S)-2-{[{2,4,6-
trimethylphenyl)sulfonyl]amino)propanoic Acid
20

A solution of the product of Example 35, Part B (10.0 mg, 0.0101 mmol), DIEA (0.007 mL, 0.040 mmol), and
25 2-{2-aza-2-((5-((2,5-dioxopyrrolidinyl)carbonyl)(2-
pyridyl))amino)vinyl)benzenesulfonic acid (5.3 mg, 0.0120 mmol) in anhydrous DMF (0.5 mL) was allowed to stand at ambient temperatures under a nitrogen atmosphere for 48 h. Additional 2-(2-aza-2- ((5-((2, 5-dioxopyrrolidinyl)-
30 carbonyl)(2-pyridyl))amino)vinyl)benzenesulfonic acid (2.0 mg, 0.00455 mmol) was added and stirring was continued an additional 48 h. The DMF was removed under vacuum and the residue was purified by HPLC on a Vydac C-

18 column (22 x 250 mm) using a 0.9%/min gradient of 0 to 36% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 30.0 min was lyophilized to give the title compound as a colorless solid (2.5 mg, 5 23.3%). MS: m/e 1064.3 [M+H]; High Resolution MS: Calcd for C45H50N11O14S3 [M+H]: 1064.27005; Found: 1064.272.
Example 37 Synthesis of 3-U1-(3-{ (2R)-2-[4-(N-{ (1R)-1-[N-(3-{3-[N-10 ( (2S)-2-Carboxy-2-{ [ (2, 4 , 6-trimethylphenyl) sulfonyl] -aminojethyl) carbamoyl] -7- [ (imidazol-2-ylarnino) methyl] -4-oxohydroquinolyl)propyl) carbamoyl] -2-sulfoethyl] -carbamoyl) (2S)-2-{2-[1,4,7,10-tetraaza-4, 7 ,10-tris (carboxymethyl) cyclododecyl] acetylamino)butanoylamino 15 ] -3-sulfopropyl}propyl) -7- [ (imidazol-2-ylamino ) methyl] -4-oxo(3-hydroquinolyl)]carbonylamino}(2S)-2- { [(2,4,6-trimethylphenyl)sulfonyl]amino}propanoic Acid Bis(trifluoroacetate) Salt

Part A - Preparation of 3-{[1-(3-{(2R)-2-[4-(N-{ (1R)-1-[N-(3-{3-[N-( (2S)-2-Carboxy-2-{[(2,4,6-trimethylphenyl)-sulfonyl] amino)ethyl) carbamoyl] -7- [ (imidazol-2-25 ylamino)methyl J -4-oxohydroguinolyl}propyl) carbamoyl] -2-sulfoethyl}carbamoyl) (2S)-2-[(tert-butoxy)carbonylamino]


5

butanoylamino]-3-sulfopropyl}propyl)-7-[(imidazol-2-ylamino)methyl]-4-oxo(3-
hydroquinolyl)]carbonylamino}(2S)-2-{((2,4,6-trimethylphenyDsulfonyl] amino}propanoic Acid
0 0 O


A solution of the product of Example 35, Part B (38.0 mg, 0.0434 mmol), DIEA (0.015 mL, 0.0869 iranol), and 10 the product of Example 30, Part A (10.9 mg, 0.0202 mmol) in anhydrous DMF (1.0 mL) was stirred at ambient temperatures under nitrogen for 48 h. The DMF was removed under vacuum and the amber oil was purified by HPLC on a Vydac C-18 column (22 x 250 mm) using a 15 0.68%/min gradient of 9 to 36% ACN containing 0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 36.1 min was lyophilized to give the title compound as a colorless solid (13.5 mg, 38.6%). MS: m/e 1732.4 [M+H], 1632.2 [M+H-Boc]. 20
Part B - Preparation of 3-{[1-(3-{(2R)-2-[A-(N-{(1R)-1-[N- (3-{3-[N-((2S)-2-Carboxy-2-{[(2,4,6-trimethylphenyl)-sulfonyl]amino)ethyl! carbamoyl] -7- [ (imidazol-2-ylamino)methyl] -4-oxohydroquinolyl}propyl) carbamoyl]-2-25 sulfoethyl)carbamoyl)(2S)-2-aminobutanoylamino]-3-
sulfopropyl}propyl)-7-[(imidazol-2-ylamino)methyl]-4-


oxo(3-hydroquinolyl)Jcarbonylamino}(25)-2-{[(2,4,6-trirnethylphenyl)sulfonyl] amino} propanoic Acid Trifluoroacetate Salt

The product of Part A, above (13.5 mg, 0.00779 mmol was dissolved in 50/50 TFA/DCM (1.0 mL) and allowed to react at ambient temperatures under nitrogen for 4 5 min. 10 The solution was concentrated under vacuum to give the title compound as a pale amber oil. MS: m/e 1633.3 [M+K].
Part C - Preparation of 3-{[1-(3-{(2R)-2-[4-(N-{(1R)-1-15 [N-(3-{3-[N-((2S)-2-Carboxy-2-{[(2,4,6-trimethylphenyl)-sulfonyl]amino}ethyl)carbamoyl]-7-[(imidazol-2-ylamino)methyl]-4-oxohydroquinolyl}propyl)carbamoyl]-2-sulfoethyl}carbamoyl)(2S)-2-{2-[1,4,7,10-tetraaza-4,7,10-tris[(tert-20 butoxycarbonyl)methyl]cyclododecyl]acetylamino}-
butanoylamino]-3-sulfopropyl}propyl)-7-[(imidazol-2-ylamino)methyl]-4-oxo(3-hydroquinolyl)]carbonylamino}-(2S)-2-{[(2,4, 6-trimethylphenyl)sulfonyl]amino}propanoic Acid Bis(trifluoroacetate) Salt 25
-207-


A solution of 2-(1,4,7,10-tetraaza-4,7,10-tris(((tert-butyl)oxycarbonyl)methyl)cyclododecyl)acetic 5 acid (15.0 mg, 0.0164 mmol) (as described in DM-7003), DIEA (0.004 mL), and HBTU (4.7 mg, 0.0124 mmol) in anhydrous DMF (0.5 mL) was stirred under nitrogen at ambient temperatures for 8 min and treated with a solution of the product of Part B, above (0.00779 mmol)
10 and DIEA (0.004 mL) in anhydrous DMF (0.5 mL). The solution was stirred at ambient temperatures for 2 4 h, treated with 0.1 N NaOH (0.33 mL) , stirred an additional 5 min, and adjusted to pH 3 with 0.1 N HC1 (0.60 mL). This solution was diluted with water (4.5 mL) and
15 purified directly by HPLC on a Vydac C-18 column (22 x 250 mm) using a 1.01%/min gradient of 9 to 49.5% ACN containing-0.1% TFA at a flow rate of 20 mL/min. The main product peak eluting at 26.7 min was lyophilized to give the title compound as a colorless solid (7.0 mg,
20 37.2%). MS: m/e 1094.4 [M+2H]; High Resolution MS: Calcd for C97H136N21O29S4 [M+H] : 2186.8696; Found: 2186.867.
Part D - Preparation of 3- {[1-(3-{(2R)-2-[4-(N-{(1R)-1-[N-(3-{3-[N-((2S)-2-Carboxy-2-{[(2,4, 6-trimethylphenyl)-25 sulfonyl]amino}ethyl) carbamoyl] -7- [ (imidazol-2-
ylamino)methyl] - 4-oxohydroquinolyl)propyl) carbamoyl] -2-


sulfoethyl} carbamoyl) (2S)-2-{2-[l,4,7,10-tetraaza-4, 7,10-tris (carboxymethyl)cyclododecyl]acetylamino}butanoylamino ] -3-sulf opropyl)propyl) -7- [ (imidazol-2-ylamino ) methyl] -4-oxo (3-hydroquinolyl) ] carbonylamino) (2S) -2- {[(2,4,6-5 trimethylphenyl) sulf onyl] amino)propanoic Acid Bis(trifluoroacetate) Salt
A solution of the product of Step C, above (7.0 mg, 0.00290 mmol) in 95/5 TFA/Et3SiH (1.0 mL) was heated to
10 reflux under nitrogen for 3 h. The solution was concentrated under vacuum and the oily residue was purified by HPLC on a Vydac C-18 column (22 x 250 mm) using a 1.2%/min gradient of 0 to 36% ACN containing 0.1% TFA at a flow rate of 2 0 mL/min. The main product peak
15 eluting at 26.5 min was lyophilized to give the title compound as a colorless solid (4.5 mg, 66.1%). High
Resolution MS: Calcd for C85H112N2102gS4 [M+H] : 2018.6818;
Found: 2018.683.
20 Example 3 8
Synthesis of the In-Ill Complex of the Conjugate Example 29
To a shielded and crimped 2 cc autosampler vial was
added 70 ug of the conjugate of Example 29 dissolved in 25 140 |il 0.5 M ammonium acetate buffer (pH 4.8) followed by the addition of 2 mg gentisic acid sodium salt and 2.6 mCi (7 p.1) In-Ill in 0.05M HC1. The reaction mixture (specific activity was heated at 85°C for 20 minutes and analyzed by HPLC. Yield: 87.9% (total for the two 30 isomers); Ret. Time: 12.5, 13.1 min.
HPLC Method
Column: Zorbax Rx C18, 25 cm x 4.6 mm Column Temperature: Ambient 35 Flow: 1. 0 ml/min
Solvent A: 10 mM ammonium acetate Solvent B: Acetonitrile


Detector: IN-US p-ram, and UV at 220 nm wavelength.
Gradient
t (min) 0 25 26 35 36 45 %B 7 7 60 60 7 7 5
Example 3 9
Synthesis of the In-Ill Complex of the Conjugate Example
30
To a lead shielded and crimped 2 cc autosampler vial
10 was added 120 jig of the conjugate of Example 30 dissolved in 240 jiL ammonium acetate buffer (0.5 M, pH 4.7) followed by the addition of 2 mg of gentisic acid (sodium salt) dissolved in 20 (1L of H.O, and 2.3 mCi, (10 \ih) In-Ill (NEN) in 0.05 N HC1 (specific activity: 52 jig/mCi).
15 The reaction mixture was heated at 100 °C for 2 0 min and analyzed by HPLC. Yield: 34.7% (total for the two isomers), Ret. Time: 16.6 and 17.3 min.
HPLC Method 20 Column-. Zorbax Rx CI8, 2 5 cm x 4.6 mm
Column Temperature: Ambient
Flow: 1.0 ml/min
Solvent A: 10 mM ammonium acetate
Solvent B: Acetonitrile 25 Detector: IN-US P~ram, and UV at 220 nm wavelength.
Gradient
t (min) 0 25 26 35 36 45
%B 10 15 60 60 10 10
30 Example 4 0
Synthesis of the In-Ill Complex of the Conjugate Example
31
To a shielded and crimped 2 cc autosampler vial was
added 70 p.g of the conjugate of Example 31 dissolved in 35 140 fil 0.5 M ammonium acetate buffer (pH 4.8) followed by
the addition of 2 mg gentisic acid sodium salt and 2.6


mCi (7 jil) In-Ill in 0.05M HC1. The reaction mixture (specific activity was heated at 85°C for 20 minutes and analyzed by HPLC. Yield: 92.2%; Ret. Time: 12.9 min.
5 HPLC Method
Column: Zorbax Rx CI8, 2 5 cm x 4.6 mm Column Temperature: Ambient Flow. 1.0 ml/min
Solvent A: 10 mM ammonium acetate 10 Solvent B: Acetonitrile
Detector: IN-US p-ram, and UV at 220 nm wavelength.
Gradient
t (min) 0 25 26 35 36 45
%B 7 7 60 60 7 7
Example 41 Synthesis of the In-Ill Complex of the Conjugate Example 33
20 To a shielded and crimped 2 cc autosampler vial was added 107 jig of the conjugate of Example 33 dissolved in 140 jil 0.5 M ammonium acetate buffer (pH 4.8) followed by the addition of 2 mg gentisic acid sodium salt and 2.6 mCi (7 jil) in-Ill in 0.05M HC1. The reaction mixture
25 (specific activity was heated at 85°C for 20 minutes and analyzed by HPLC. Yield: 77.9%; Ret. Time-. 17.8 min.
HPLC Method
Column: Zorbax Rx C18, 25 cm x 4.6 mm 30 Column Temperature: Ambient Flow: 1.0 ml/min
Solvent A: 10 mM ammonium acetate Solvent B: Acetonitrile Detector: IN-US (3-ram, and UV at 220 nm wavelength.
35 Gradient
t (min) 0 25 26 35 36 45
%B 9 11 60 60 9 9


Example 42 Synthesis of the In-Ill Complex of the Conjugate Example 5 34
To a lead shielded and crimped autosampler vial was
added 25 jig of the conjugate of Example 34 and 1.0 mg gentisic acid, sodium salt dissolved in 50 \iL ammonium acetate buffer (0.4 M, pH 4.7) followed by the addition 10 of 1.2 mCi, (5 jiL) In-Ill in 0.05 N HC1 (specific
activity: 21 ug/mCi) . The reaction mixture was heated at 80 °C for 45 min and analyzed by HPLC and ITLC. 93.5% yield by HPLC, Ret. Time: 16.7 min.
15 HPLC Method
Column: Zorbax Rx C18, 2 5 cm x 4.6 mm
Column Temperature: Ambient
Flow: 1.0 ml/min
Solvent A: 25 mM sodium phosphate buffer at pH 6 20 Solvent B: Acetonitrile
Detector: Sodium iodide (Nal) radiometric probe, and UV
at 220 nm wavelength.
Gradient
t (min) 0 25 26 35 36 45 25 %B 10 20 60 60 10 10
Example 43
Synthesis of the In-111 Complex of the Conjugate Example 35
30 To a lead shielded and crimped 1 cc autosampler vial was added 40-50 jig of the conjugate of Example 35 dissolved in 100 jiL ammonium citrate buffer (0.4 M, pH 4.7) followed by the addition of 2 mCi, (5 JiL) In-Ill in 0.05 N HC1 (specific activity: 25 jig/mCi). The reaction
35 mixture was heated at 90-100 C for 3 0 min and analyzed by HPLC. Yield: 95%; Ret. Time 12.5 min.

HPLC Method
Column: Zorbax Rx C18, 25 cm x 4.6 mm Column Temperature: Ambient Flow: 1.0 ml/min 5 Solvent A: 25 mM sodium phosphate buffer at pH 6 Solvent B: Acetonitrile
Detector: Sodium iodide (Nal) radiometric probe, and UV at 220 nm wavelength. Gradient 10 t (min) 0 25 26 35 36 45 %B 10 20 60 60 10 10
Example 44 Synthesis of the In-111 Complex of the Conjugate Example
15 37
To a lead shielded and crimped 2 cc autosampler vial was added 150 jig of the conjugate of Example 37 dissolved in 300 uL ammonium citrate buffer (0.3 M, pH 4.8) followed by the addition of 4.5 mCi, (25 UL) In-111 (NEN) in 0.05
20 N HC1 (specific activity: 33 ng/mCi). The reaction
mixture was heated at 100 °C for 20 min and analyzed by HPLC. RCP: 80%, Ret. Time: 21 min.
HPLC Method 25 Column: Zorbax Rx C18, 2 5 cm x 4.6 mm
Column Temperature: Ambient
Flow: 1.0 ml/min
Solvent A: 25 mM sodium phosphate buffer at pH 6
Solvent B: Acetonitrile 30 Detector: Sodiuin iodide (Nal) radiometric probe, and UV
at 220 nm wavelength.
Gradient
t (min) 0 25 26 35 36 45
%B 17 19 60 60 17 17


Examples 45 - 51 Synthesis of Y-90 and Lu-177 Complexes of the Conjugates of Examples 30, 31, 34, 35 and 37
To a clean sealed 5 mL vial was added 0.5 -1.0 mL of 5 the appropriate conjugate solution (200 ug/mL in 0.5 M ammonium acetate buffer, pH 7.0-8.0), followed by 0.05 mL of sodium gentisate (10 mg/mL in 0.5 M ammonium acetate buffer, pH 7.0-8.0) solution, and 10 - 40 uL of ,0YC13 or 177LuCl3 solution (10 - 20 mCi) in 0.05 N HC1. The 10 reaction mixture was heated at 100 °C for 5-10 min. After cooling to room temperature, a sample of the resulting solution was analyzed by HPLC and by ITLC.

Complex Ex # Isotope Conjugate Ex/# Ret. Time (min) % Yield HPLC Method
45 Y-90 30 14.0, 16.0 90 D
46 Y-90 31 14.0 90.5 F
47 Lu-177 31 13.0 85 D
48 Y-90 34 8.0 81.9 A
49 Y-90 35 16.0 89 B
50 Y-90 37 8.2 83.5 B
51 LU-177 37 14.0 70 G
15 HPLC Method A-. The HPLC method using a reverse phase Cis Zorbax column (4.6 mm x 25 cm, 80 A pore size) at a flow rate of 1.0 mL/min with a gradient mobile phase from 85% A (25 mM pH 6.0 phosphate buffer) and 15% B (acetonitrile) to 75% A and 25% B at 20 min.
20
HPLC Method B: The HPLC method using a reverse phase Cis Zorbax column (4.6 mm x 25 cm, 80 A pore size) at a flow rate of 1.0 mL/min with a gradient mobile phase from 90% A (25 mM pH 6.0 phosphate buffer) and 10% B
25 (acetonitrile) to 80% A and 20% B at 20 min.
HPLC Method D: The HPLC method using a reverse phase CIB Zorbax column (4.6 mm x 25 cm, 80 A pore size) at a flow rate of 1.0 mL/min with a gradient mobile phase from 87% 30 A (25 mM pH 6.0 phosphate buffer) and 13% B (acetonitrile) to 86% A and 14% B at 20 min.

HPLC Method F*. The HPLC method using a reverse phase Cis Zorbax column (4.6 mm x 25 cm, 80 A pore size) at a flow rate of 1.0 mL/min with a gradient mobile phase from 92% 5 A (25 mM ammonium acetate buffer, pH = 6.8) and 8% B (acetonitrile) to 90% A and 10% B at 20 min.
HPLC Method G: The HPLC method using a reverse phase Ci8 Zorbax column (4.6 mm x 25 cm, 80 A pore size) at a flow 10 rate of 1.0 mL/min with an isocratic mobile phase of 87% A (25 mM ammonium acetate buffer, pH = 6.8) and 13% B (acetonitrile) from 0 to 20 min.
Example 52
15 Synthesis of 99mTc(3-{[1-(3-{2-[(6-(diazenido)(3-
pyridyl) ) carbonylamino] (2R) -3-sulf opropyl}propyl) -7-[ (imidazol-2-ylarnino)methyl] -4-oxo (3-hydroquinolyl)]carbonylamino}(2S) -2- { [ (2 , 4, 6-trimethylphenyDsulfonyl] amino}propanoic acid)
20 (tricine)(TPPTS)
To a lyophilized vial containing 4.84 mg TPPTS, 6.3 mg tricine, 40 mg mannitol, succinic acid buffer, pH 4.8, and 0.1% Pluronic F-64 surfactant, was added 1.1 mL sterile water for injection, 0.2 mL (20 pg) of the the
25 conjugate of Example 36 in deionized water or 50% aqueous ethanol, and 0.2 mL of 99mTcO,r (50±5 mCi) in saline. The reconstituted kit was heated in a 100 °C water bath for 15 minutes, and was allowed to cool 10 minutes at room temperature. A sample of the reaction mixture was
30 analyzed by HPLC. The yield was 89.0% and the retention time 12.8, 13.2 min (2 isomers). HPLC Method
Column: Zorbax C18 , 25 cm x 4.6 mm Flow rate : 1.0 mL/min
35 Solvent A: 10 mM sodium phosphate buffer, pH 6.0 Solvent B : 100 % CH3CN Gradient 0 - 25% B over 20 min.

Utility
The pharmaceuticals of the present invention are useful for imaging angiogenic tumor vasculature, therapeutic cardiovascular angiogenesis, and cardiac 5 pathologies associated with the expression of vitronectin receptors in a patient or for treating cancer in a patient. The radiopharmaceuticals of the present invention comprised of a gamma ray or positron emitting isotope are useful for imaging of pathological processes 10 involving angiogenic neovasculature, including cancer,
diabetic retinopathy, macular degeneration, restenosis of blood vessels after angioplasty, and wound healing, as well as atherosclerotic plague, myocardial reperfusion injury, and myocardial ischemia, stunning or infarction. 15 The radiopharmaceuticals of the present invention
comprised of a beta, alpha or Auger electron emitting isotope are useful for treatment of pathological processes involving angiogenic neovasculature, by delivering a cytotoxic dose of radiation to the locus of 20 the angiogenic neovasculature. The treatment of cancer is affected by the systemic administration of the radiopharmaceuticals resulting in a cytotoxic radiation dose to tumors.
The compounds of the present invention comprised of 25 one or more paramagnetic metal ions selected from
gadolinium, dysprosium, iron, and manganese, are useful as contrast agents for magnetic resonance imaging (MRI) of pathological processes involving angiogenic neovasculature, as well as atherosclerotic plaque, 30 myocardial reperfusion injury, and myocardial ischemia, stunning or infarction.
The compounds of the present invention comprised of one or more heavy atoms with atomic number of 20 or greater are useful as X-ray contrast agents for X-ray 35 imaging of pathological processes involving angiogenic neovasculature, as well as atherosclerotic plaque, myocardial reperfusion injury, and myocardial ischemia, stunning or infarction.

The compounds of the present invention comprised of an echogenic gas containing surfactant microsphere are useful as ultrasound contrast agents for sonography of pathological processes involving angiogenic 5 neovasculature, as well as atherosclerotic plaque,
myocardial reperfusion injury, and myocardial ischemia, stunning or infarction.
Representative compounds of the present invention 10 were tested in the following in vitro assays and in vivo models and were found to be active.
Immobilized Human Placental avb3 Receptor Assay
The assay conditions were developed and validated
15 using [1-125]vitronectin. Assay validation included Scatchard format analysis (n=3) where receptor number (Bmax) and Kd (affinity) were determined. Assay format is such that compounds are preliminarily screened at 10 and 100 nM final concentrations prior to IC50
20 determination. Three standards (vitronectin, anti-avB3 antibody, LM609, and anti-avB5, P1F6) and five reference peptides have been evaluated for IC50 determination. Briefly, the method involves immobilizing previously isolated receptors in 9 6 well plates and incubating
25 overnight. The receptors were isolated from normal,
fresh, non-infectious (HIV, hepatitis B and C, syphilis, and HTLV free) human placenta. The tissue was lysed and tissue debris removed via centrifugation. The lysate was filtered. The receptors were isolated by affinity
30 chromatography using the immobilized avb3 antibody. The plates are then washed 3x with wash buffer. Blocking buffer is added and plates incubated for 120 minutes at room temperature. During this time compounds to be tested and [1-125]vitronectin are premixed in a reservoir
35 plate. Blocking buffer is removed and compound mixture pipetted. Competition is carried out for 60 minutes at room temperature. Unbound material is then removed and wells are separated and counted via gamma scintillation.


ray contrast agents. After administration of the appropriate amount of the X-ray absorbing compounds, the whole animal can be placed in a commercially available X-ray imager to image the tumors. The effectiveness of the 5 contrast agents can be readily seen by comparison to the images obtain from animals that are not administered a contrast agent.
This model can also be used to assess the compounds of the present invention comprised of an echogenic gas
10 containing surfactant microsphere as ultrasound contrast agents. kitex adrriiTiist.rat.iov, ot ^VYS appropriate S^CAITA: of the echogenic compounds, the tumors in the animal can be imaging using an ultrasound probe held proximate to the tumors. The effectiveness of the contrast agents can
15 be readily seen by comparison to the images obtain from animals that are not administered a contrast agent.
Rabbit Matrigel Model
This model was adapted from a matrigel model
20 intended for the study of angiogenesis in mice. Matrigel (Becton & Dickinson, USA) is a basement membrane rich in laminin, collagen IV, entactin, HSPG and other growth factors. When combined with growth factors such as bFGF [500 ng/ml] or VEGF [2 ug/ml] and injected subcutaneously
25 into the mid-abdominal region of the mice, it solidifies into a gel and stimulates angiogenesis at the site of injection within 4-8 days. In the rabbit model, New Zealand White rabbits (2.5-3.0 kg) are injected with 2.0 ml of matrigel, plus 1 ug bFGF and 4 ug VEGF. The
30 radiopharmaceutical is then injected 7 days later and the images obtained.
This model can also be used to assess the effectiveness of the radiopharmaceuticals of the present invention comprised of a beta, alpha or Auger electron
35 emitting isotope. The radiopharmaceuticals are
administered in appropriate amounts and the uptake at the angiogenic sites can be quantified either non-invasively by iwagiivg tor those isotopes ^itft a coincident, imageable gamma emission, or by excision of the angiogenic sites


and counting the amount of radioactivity present by standard techniques. The therapeutic effect of the radiopharmaceuticals can be assessed by monitoring the rate of growth of the angiogenic sites in control rabbits 5 versus those in the rabbits administered the radiopharmaceuticals of the present invention.
This model can also be used to assess the compounds of the present invention comprised of paramagnetic metals as MRI contrast agents. After administration of the 10 appropriate amount of the paramagnetic compounds, the whole animal can be placed in a commercially available magnetic resonance imager to image the angiogenic sites. The effectiveness of the contrast agents can be readily seen by comparison to the images obtain from animals that 15 are not administered a contrast agent.
This model can also be used to assess the compounds of the present invention comprised of heavy atoms as X-ray contrast agents. After administration of the appropriate amount of the X-ray absorbing compounds, the 20 whole animal can be placed in a commercially available X-ray imager to image the angiogenic sites. The effectiveness of the contrast agents can be readily seen by comparison to the images obtain from animals that are not administered a contrast agent. 25 This model can also be used to assess the compounds of the present invention comprised of an echogenic gas containing surfactant microsphere as ultrasound contrast agents. After administration of the appropriate amount of the echogenic compounds, the angiogenic sites in the 30 animal can be imaging using an ultrasound probe held proximate to the tumors. The effectiveness of the contrast agents can be readily seen by comparison to the images obtain from animals that are not administered a contrast agent. 35
Canine Spontaneous Tumor Model
Adult dogs with spontaneous mammary tumors were sedated with xylazine (20 mg/kg)/atropine (1 ml/kg) . Upon sedation the animals were intubated using ketamine


(5 mg/kg)/diazepam (0.25 mg/kg) for full anethesia. Chemical restraint was continued with ketamine (3 mg/kg)/xylazine (6 mg/kg) titrating as necessary. If required the animals were ventilated with room air via an 5 endotrachael tube (12 strokes/min, 25 ml/kg) during the study. Peripheral veins were catheterized using 20G I.V. catheters, one to serve as an infusion port for compound while the other for exfusion of blood samples. Heart rate and EKG were monitored using a cardiotachometer 10 (Biotech, Grass Quincy, MA) triggered from a lead II
electrocardiogram generated by limb leads. Blood samples are generally taken at -10 minutes (control), end of infusion, (1 minute), 15 min, 30 min, 60 min, 90 min, and 120 min for whole blood cell number and counting. 15 Radiopharmaceutical dose was 300 uCi/kg adminitered as an i.v. bolus with saline flush. Parameters were monitored continuously on a polygraph recorder (Model 7E Grass) at a paper speed of 10 mm/min or 10 mm/sec.
Imaging of the laterals were for 2 hours with a 20 2 56x256 matrix, no zoom, 5 minute dynamic images. A
known source is placed in the image field (20-90 uCi) to evaluate region of interest (ROD uptake. Images were also acquired 24 hours post injection to determine retention of the compound in the tumor. The uptake is 25 determined by taking the fraction of the total counts in an inscribed area for ROI/source and multiplying the known uCi. The result is uCi for the ROI.
This model can also be used to assess the effectiveness of the radiopharmaceuticals of the present 30 invention comprised of a beta, alpha or Auger electron emitting isotope. The radiopharmaceuticals are administered in appropriate amounts and the uptake in the tumors can be quantified either non-invasively by imaging for those isotopes with a coincident imageable gamma 35 emission, or by excision of the tumors and counting the amount of radioactivity present by standard techniques. The therapeutic effect of the radiopharmaceuticals can be assessed by monitoring the size of the tumors over time. This model can also be used to assess the compounds

of the present invention comprised of paramagnetic metals as MRI contrast agents. After administration of the appropriate amount of the paramagnetic compounds, the whole animal can be placed in a commercially available 5 magnetic resonance imager to image the tumors. The
effectiveness of the contrast agents can be readily seen by comparison to the images obtain from animals that are not administered a contrast agent.
This model can also be used to assess the compounds
10 of the present invention comprised of heavy atoms as X-ray contrast agents. After administration of the appropriate amount of the X-ray absorbing compounds, the whole animal can be placed in a commercially available X-ray imager to image the tumors. The effectiveness of the
15 contrast agents can be readily seen by comparison to the images obtain from animals that are not administered a contrast agent.
This model can also be used to assess the compounds of the present invention comprised of an echogenic gas
20 containing surfactant microsphere as ultrasound contrast agents. After administration of the appropriate amount of the echogenic compounds, the tumors in the animal can be imaging using an ultrasound probe held proximate to the tumors. The effectiveness of the contrast agents can
25 be readily seen by comparison to the images obtain from animals that are not administered a contrast agent.
Cardiovascular disease models that can be used to assess the diagnostic radiopharmaceuticals, magnetic resonance, X-ray and ultrasound contrast agents of the
30 present invention are reviewed in J. Nucl. Cardiol., 1998, 5, 167-83. There are several well established rabbit models of atherosclerosis; one model produces predominantly proliferating smooth muscle cells by balloon deendothelialization of infradiaphragmatic
35 abdominal aorta to simulate restenotic lesions; another model that produces simulated advanced human atherosclerotic plaque by balloon deendothelialization followed by a high cholesterol diet.

WE CLAIM:
1. A compound, comprising: a targeting moiety and a chelator, wherein the targeting moiety is bound to the chelator, is a quinolone nonpeptide, and binds to a receptor that is upregulated during angiogenesis and the compound has 0-1 linking groups between the targeting moiety and chelator,
wherein the receptor is the integrin (Q)d-Ln-Ch or (Q)d-Ln-(Ch)d. wherein, Q is a compound of Formula (II):

(II)
including stereoisomeric forms thereof, or mixtures of stereoisomeric forms thereof, or pharmaceutical^ acceptable salt or prodrug forms thereof wherein:
225


Rle is selected from:
Ae is -CH2- or -N(R10e)-;
Ale and Be are independently -CH2- or -N(R10e)-;
De is -N(RlOe)- or -S-;
Ee_Fe is _C(R2e)=c{R3e)_ or _c (R2e)2C (R3e) 2_.
Je is -C(R2e)- or -N-;
Ke, Le and Me are independently -C(R2e)- or -C(R3e)~; R2e and R3e are independently selected from:
226

K, C1-C4 alkoxy, ijRUsRusi halogen, N02, CM, CF3, Ci-Ce alkyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl (d-C4 alkyl), aryl (Ci-Ce alkyl) -, (Ci-Ce alkyl)carbonyl, (C1-C6 alkoxy)carbonyl, arylcarbonyl, and aryl substituted with 0-4 R7e,
alternatively, when R2e and R3e are substituents on adjacent atoms, they can be taken together with the carbon atoms to which they are attached to form a 5-7 membered carbocyclic or 5-7 membered heterocyclic aromatic or nonaromatic ring system, said carbocyclic or heterocyclic ring being substituted with 0-2 groups selected from C1-C4 alkyl, C1-C4 alkoxy, halo, cyano, amino, CF3 and N02;
R2ae is selected from:
H, C1-C10 alkyl, C2-C6 alkenyl, C3-C11 cycloalkyl, C3-C7 cycloalkyl (C1-C4 alkyl), aryl, aryl(Ci-C4 alkyl)-, (C2-C7 alkyl)carbonyl, arylcarbonyl, {C2-C10 alko*y)carbonyl, C3-C7 cycloalkoxycarbonyl, C7-CXI bicycloalkoxycarbonyl, aryloxycarbonyl, aryl (C1-C10 alkoxy) carbonyl,
Ci-Ce alkylc^rbonyloxy(Ci-C4 alkoxy) carbonyl, arylcarbonyl0xy(C1-C4 alkoxy)carbonyl, and C3-C7 cycloalkylcarbonyloxy(Ci-C4 alkoxy)carbonyl;
R7e is selected from:
H, hydroxy, d-c4 alkyl, Ci~C4 alkoxy, aryl, aryl (Ci-C4 alkyl)-, (C1-C4 alkyl)carbonyl, C02R18ae, S02Rlle, SO2NR10eRlle, ORl0e, and NiR^^R1^;
Ue is selected froin:
-(CH2)ne-» -(CH2)neO(CH2)I„e-, - (CH2)neN(Rl2) (CH2)ra*-, -NH(CH2)n«-, -(CH2)neC(=0) (CH2 )„,-, -(CH2)neS(0)J3e{CH^jJ5e-, - {CE2)^NHm(CH2)me-/
-N(RlO«)C{=0)-# -NHC(=0)(CH2)n*-, -C (=O)N(Rl0e) _, and
-N(RlOe)S(0}pe..;

G* is N or CR1***;
We is-C(=O)-N(R10e)-(Ci-C3 alkylene)-, in which the alkylene group is substituted by R8e and by R9e:
R8e and R9e are independently selected from: H, C02R18be, C(=0)R18be, CONR17R18be, Ci-Cio alkyl substituted with 0-1 R6e, C2-C10 alkenyl substituted with 0-1 R6e, C2-C10 alkynyl substituted with 0-1 R6e, C3-C8 cycloalkyl substituted with 0-1 R6e, C5-C6 cycloalkenyl substituted with 0-1 R6e, (C1-C10 alkyl)carbonyl, C3-C10 cycloalkyl(C1-C4 alkyl)-, phenyl substituted with 0-3 R6e, naphthyl substituted with 0-3 R6e,
a 5-10 membered heterocyclic ring containing 1-3 N, O, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e, C1-C10 alkoxy substituted with 0-2 R7e, hydroxy, nitro, -N(R10e)Rlle, -N(R16e)R17e, aryl (C0-C6 alkyl)carbonyl, aryl(C3-C6 alkyl), heteroaryl(Ci-C6 alkyl), CONRi8aeR20e, S02Ra8ae, and SO2NR18aeR20e, providing that any of the above alkyl, cycloalkyl, aryl or heteroaryl groups may be unsubstituted or substituted independently with 1-2 R7e'"
R6e is selected from:
H, C1-C10 alkyl, hydroxy, C1-C10 alkoxy, nitro, C1-C10 alkyl carbonyl, -N(Rlle)Ri2e, cyano, halo, CF3, CHO, C02R18be, C(=0)R18be, CONR17^^**, OC(=O)R10e, OR10e, OC(=O)NR10eRHe, NR10eC(=O)R10e, NR10eC(=O)OR21e, NRl°eC (=0) NR10eRlle, NRl0«SO2NR10eRlle/
NR10eSO2R21e, S(0)pRlle, SO2NR10eRlle,

aryl substituted with 0-3 groups selected from halogen, Ci-Ce alkoxy, Ci-Cs alkyl, CF3, S(0)meMe, and -NMe2, aryl(Ci-C4 alkyl)-, said aryl being substituted with 0-3 groups selected from halogen, C1-C6 alkoxy, C1-C6 alkyl, CF3, S(0)PeMe, and -NMe2, and a 5-10 membered heterocyclic ring containing 1-3 N, 0, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e;
R10* is selected from:
H, CF3, C3-C6 alkenyl, C3-G11 cycloalkyl, aryl, (C3-Cia cycloalkyl)methyl, aryl(C1-C4 alkyl), and Ci-C10 alkyl substituted with 0-2 R6e;
Rile iS selected from.-
H, hydroxy, Ci-Cs alkyl, C3-C6 alkenyl, C3-C11 cycloalkyl, (C3-C11 cycloalkyl)methyl, Ci-Cg alkoxy, ben2yloxy, aryl, heteroaryl, heteroaryl(C1-C4 alkyl)-, aryl (C1-C4 alkyl), adamantylmethyl, and C1-C10 alkyl substituted with 0-2 R4e;
R4e is selected from:
H, C1-C6 alkyl, C3-C7 cycloalkyl, C3-C7
cycloalkyl(C1-Ci alkyl)-, (Ci-Cj,o alkyl)carbonyl, -
aryl, heteroaryl, aryKC^-Cg alkyl)-, and
heteroaryl(Ca-C6 alkyl)-, wherein said aryl or
heteroaryl groups are substituted with 0-2 substituents independently selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, F, CI, Br, CF3, and NO2,
alternatively, when R10e and Rlle are both substituents on the same nitrogen atom (as in -NR10eR11(S) they may be taken together with the nitrogen atom to which they are attached to form a heterocycle selected from: 3-azabicyclononyl, 1,2,3,4-tetrahydro-l-quinolinyl,

1,2, 3,4-tetrahydro-2-isoquinolinyl, 1-piperidinyl, 1-morpholinyl, 1-pyrrolidinyl, thiamorpholinyl, thiazolidinyl, and 1-piperazinyl; said heterocycle being substituted with 0-3 groups selected from: C1-C6 alkyl, aryl, heteroaryl, aryl(Ci-C4 alkyl)-, (Ci-Ce alkyl)carbonyl, (C3-C7 cycloalkyl) carbonyl, (C1-C6 alkoxy) carbohyl, aryl (Ci-C4 alkoxy)carbonyl, Ci-Cg alkylsulfonyl, and arylsulfonyl;
Ri2e ±s selected from:
H, Ci-Ce alkyl, triphenylmethyl, methoxymethyl, methoxyphenyldiphenylmethyl,
trimethylsilylethoxymethyl, (Ci-Ce alkyl) carbonyl, (Ci-C6 alkoxy) carbonyl, (C1-C6 alkyl) amino carbonyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C;-C4 alkyl)-, aryl, heteroaryl(C1-C6 alkyl)carbonyl, heteroaryl carbonyl, aryl (Ci-Ce alkyl) -, (Ci-Ce alkyl)carbonyl, arylcarbonyl, C1-C6 alkylsulfonyl, arylsulfonyl, aryl(Ci-C6 alkyDsulfonyl, heteroarylsulfonyl, heteroaryl (Cj-Ce alkyDsulfonyl, aryl oxy car bony 1, and aryl (C2-C6 alkoxy) carbonyl, wherein said aryl groups are substituted with 0-2 substituents selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, halo, CF3, and nitro;
R16e is selected from.-
-C(=O)0R18ae, -C{=0)R18be, -C(=0)N(R18be)2, -C(=0) NHS02R18ae, -C(=0)NHC(=0)R18be, -C(=0)NHC(=0)OR18ae, -C(=0)NHS02NHR18be, -S02R18ae, -S02N(R18be)2, and -S02NHC(=0)OR18be;
R17e is selected from:
H, Ca-Ce alkyl, C3-C7 cycloalkyl, C3-C7
cycloalkyl(C1-C4 alkyl)-, aryl, aryl(Ci-C6 alkyl)-,
and heteroaryl (Ci-Cg alkyl);

Ri8ae iS selected from:
Ci-Cs alkyl optionally substituted with a bond to Ln, C3-C11 cycloalkyl optionally substituted with a bond to Ln, aryKCi-Cs alkyl)- optionally substituted with a bond to LB, heteroaryl(C1-C6 alkyl)- optionally substituted with a bond to Ln, (Ci-Cs alkyl)heteroaryl optionally substituted with a bond to Ln, biaryl (C1-C6 alkyl) optionally substituted with a bond to Ln, heteroaryl optionally substituted with a bond to Ln, phenyl substituted with 3-4 R19e and optionally substituted with a bond to L^,
naphthyl substituted with 0-4 R19e and optionally substituted with a bond to Ln/ and a bond to Ln/ wherein said aryl or heteroaryl groups are optionally substituted with 0-4 R19e;
R18be is H or RiS";
R19e is selected from;
H, halogen, CF3, C02H, CN, NO2, -NRlleR12e, OCF3, Ci-Cs alkyl, C2-C6 alkenyl, C2-C6 alkynyl, . C3-CH cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl)-, aryKCi-Ce alkyl)-, Ci-Ce alkoxy, C1-C4 alkoxycarbonyl, aryl, aryl-0-, aryl~S02-, heteroaryl, and heteroaryl-SC>2-, wherein said aryl
and heteroaryl groups are substituted with 0-4 groups selected from hydrogen, halogen, CF3, C1-C3 alkyl, and C1-C3 alkoxy;
K20e is selected from:
hydroxy, C1-C10 alkyloxy, C3-C11 cycloalkyloxy, aryloxy, aryl(Ci-C4 alkyl )oxy, C2-C10 alkylcarbonyloxy(Ci-C2 alkyl)oxy-, C2-C10 alkoxycarbonyloxy (C1-C2 alkyl )oxy-, C2-C10 alkoxycarlxmyl (C1-C2 alkyl>oxy-, C3-Cio cycloalkylcarbonyloxy(Ci-C2 alkyl)oxy-, C3-C10 cycloalkoxycarbonyloxy (Ci-c2"- alkyl) oxy-,

C3-C10 cycloalkoxycarbonyl(Ci-C2 alkyDoxy-, ary!oxycarbonyl(Ci-C2 alkyl)oxy-,
arylcarbonyloxy(Ci-C2 alkyDoxy-/
C1-C5 alkoxy{Ci-C5 alkyl)carbonyloxy(C1-C2 alkyDoxy,
(5-(Ci-C5 alkyl)-1, 3-dioxa-cyclopenten-2-one-
yDmethyloxy, (5-aryl-l,3-dioxa-cyclopenten-2-one-yl)methyloxy,
and (R10e) (Rlle)N-(Ci-Cio alkoxy)-;
R21e is selected from:
Ci-Ce alkyl, C2-Ce alkenyl, C3-C11 cycloalkyl, (C3-C11 cycloalkyl)methyl, aryl, aryl(Ci-C4 alkyl)-, and Ci~ C10 alkyl substituted with 0-2 R7e;
-C(=0)-R18be, -C(=0)N{R18be)2, ~C(=0)NHSO2R18ae, -C(=0)NHC(=0)R18be, -C(=O)NHC(=0)OR18ae, and -C(=0)NHS02NHR18be;
Ye is selected from:
-COR20e, -SO3H, -PO3H, -CONHNHSO2CF3, -CONHS02R18ae, -CONHS02NHR18be, -NHCOCF3, -NHC0NHSO2R18a«, -NHS02R18ae, -OPQ3H2, -OSO3H, -PD3H2/ -S02NHCOR18ae, -SO2NHC02R18ae,


with the following proviso: ne and me are chosen such
that the number of atoms connecting Rle and Ye is in the range of 8-14;
d is selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
d' is 1-100;
Ln is a linking group having the formula:
((W)h-(CR6R7)g)x-(Z)k-{(CR6aR7a)g'-(W)h.}X';
W is independently selected at each occurrence from the
group: 0, S, NH, NHC{=0), C(=0)NH, NR8C(=0), C(=0)N R8, C(=0), C(=0)0, OC(=0), NHC(=S)NH, NHC(=0)NH, S02, S02NH, (OCH2CH2)s, (CH2CH20)s-, aa is independently at each occurrence an amino acid;
Z is selected from the group: aryl substituted with 0-3 R10. C3-10 cycloalkyl substituted with 0-3 R10, and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R10;
R6, R6a, R7, R7a, and R8 are independently selected at each occurrence from the group: H, =0, COOH, SO3H, PO3H, C1-C5 alkyl substituted with 0-3 R10, aryl substituted with 0-3 R10, benzyl substituted with 0-3 R10, and C1-C5 alkoxy substituted with 0-3 R10, NHC(=0)Ri:i, C(=0)NHR1:L, NHC(=0)NHR11, NHR*1, R", and a bond to Ch;
R10 is independently selected at each occurrence from the group: a bond to Ch, COOR11, C(=0)NHRn, NHC^OR11, OHr mm11, S03H, P03H, -OP03H2, -OSO3H, aryl

substituted with 0-3 R11, C1-5 alkyl substituted with 0-1 R12, C1-5 alkoxy substituted with 0-1 R12, and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R11;
R11 is independently selected at each occurrence from the group: H, alkyl substituted with 0-1 R12, aryl substituted with 0-1 R12, a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-1 R12, C3-10 cycloalkyl substituted with 0-1 R12, polyalkylene glycol substituted with 0-1 R12, carbohydrate substituted with 0-1 R12, cyclodextrin substituted with 0-1 R12, amino acid substituted with 0-1 R12, polycarboxyalkyl substituted with 0-1 R12, polyazaalkyl substituted with 0-1 R12, peptide substituted with 0-1 R12, wherein the peptide is comprised of 2-10 amino acids, 3, 6-O-disulfo-B-D-galactopyranosyl, bis(phosphonomethyl)glycine, and a bond to Ch;
R12 is a bond to Ch;
k is selected from 0, 1, and 2; h is selected from 0, 1, and 2; h" is selected from 0, 1, and 2;
g is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; g' is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; s is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; s' is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; s" is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; t is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; t' is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10; x is selected from 0, 1, 2, 3, 4, and 5; x' is selected from 0, 1, 2, 3, 4, and 5;

s a metal bonding unit having a formula selected from group:

A1, A2, A3, A4, A5, A6, A7, and A8 are independently
selected at each occurrence from the group: NR13, NR13R14, S, SH, S(Pg), 0, OH, PR*3, PR13R14, P(0)R15R16, and a bond to Ln;
E is a bond, CH, or a spacer group independently selected at each occurrence from the group: Ci-Cio alkyl
substituted with 0-3 R17, aryl substituted with 0-3 R17, C3-10 cycloalkyl substituted with 0-3 R17, heterocyclo-Ci-io alkyl substituted with 0-3 R17, wherein the heterocyclo group is a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0, Cg-io aryl-Ci-10 alkyl substituted with 0-3 R17, C1-10 alkyl-C6^io aryl- substituted with 0-3 R17, and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, s> and O and substituted with 0-3 R17;
R13 and R14 are each independently selected from the group: a bond to Ln, hydrogen, C1-C10 alkyl substituted with 0-3 R17, aryl substituted with 0-3

R17 alternatively, R13 and R14 combine to form =C(R20) (R21) ;
R15 and R16 are each independently selected from the
group: a bond to Lj,, -OH, Ci-Cio alkyl substituted with 0-3 R^7, Ci-Cio alkyl substituted with 0-3 R17, aryl substituted with 0-3 R17, C3-10 cycloalkyl substituted with 0-3 R17, heterocyclo-Ci-10 alkyl substituted with 0-3 R17, wherein the heterocyclo group is a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O, C6-10 aryl-Ci-10 alkyl substituted with 0-3 R17, Ci-10 alkyl-Cg-io aryl- substituted with 0-3 R17, and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R";
R17 is independently selected at each occurrence from the group: a bond to L„, =0, F, CI, Br, I, -CF3, -CN, -CO2R18, -C(=0!R18, -C(=0)N(R18]2, -CHO, -CH2OR18, -OC(=0)R18, -OC(=0)OR18a, -OR18, -OC (=0)N{R18)2, -NRl9c(=0)R18, _NRl9C(=o)OR18a, -NRl^C(=0)N(R18)2, -NR19S02N(R18)2, -NRl9S02R18a, -SO3K, -S02R18a, -SR18, -S(=0)Rl8a, .S02N(R18)2, -N(R18)2, -NHC(=S)NHR18, =NOR18, N02, -C(=0)NHOR18,

-C(=0)NHNR18R18a, -OCH2CO2H, 2-(l-morpholano)ethoxy, C1-C5 alkyl, C2-C4 alkenyl, C3-C6 cycloalkyl, C3-C6 cycloalkylmethyl, C2-C6 alkoxyalkyl, aryl substituted with 0-2 R18, and a 5-10 membered heterocyclic ring system containing 1-T4 heteroatoms independently selected from N, S, and 0;
R18, R18a, and R19 are independently selected at each occurrence from the group: a bond to Ln, H, Ci-Cs alkyl, phenyl, benzyl, Ci-Ce alkdxy, halide, nitro, cyano, and trifluoromethyl;
Pg is a thiol protecting group;
R20 and R21 are independently selected from the group: H, C1-C10 alkyl, -CN, -C02R25, -C(*0)R25, -C (=0)N(R25)2, C2-C10 1-alkene substituted with 0-3 R23, C2-C10 1-alkyne substituted with 0-3 R23, aryl substituted with 0-3 R23, unsaturated 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R23, and unsaturated C3-10 carbocycle substituted
with 0-3 R23;
alternatively, R20 and R21, taken together with the
divalent carbon radical to which they are attached form:

R22 and R23 are independently selected from the group: H, R24, C1-C10 alkyl substituted with 0-3 R24, C2-C10 ' alkenyl substituted with 0-3 R24, C2-C10 alkynyl substituted with 0-3 R24, aryl substituted with 0-3

R24, a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R24, and C3-10 carbocycle substituted with 0-3 R24;
alternatively, R22, R23 taken together form a fused
aromatic or a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0;
a and b indicate the positions of optional double bonds and n is 0 or 1;
R24 is independently selected at each occurrence from the group: =0, F, Cl, Br, I, -CF3, -CN, -C02R25, -C(=0)R25, -C(=0)N(R25)2, -N(R25)3+, ~CH2OR2S, -0C(=0)R25, -0C(=O)OR25a, -OR25, -0C(=O)N(R25) 2, -NR26C(=0)R25, -NR26C(=0)OR2Sa, -NR26C (=0)N(R25) 2, -NR26S02N(R25)2, -NR26S02R25*, -S03H, -iS02R25a, -SR25, -S(=0)R25a, -S02N(R25)2, -N(R25)2, =NOR25, -C(=0)NHOR25, -OCH2CO2H, and 2- (l-morpholino)ethoxy; and,
R25, R25a, and R26 are each independently selected at each occurrence from the group: hydrogen and C1-C6 alkyl;
and a pharmaceutical^ acceptable salt thereof.
(IV)
238
2. A compound as claimed in claim 1, wherein Q is a compound of Formula (IV):


including stereoisomer^ forms thereof, or mixtures of stereoisomer^ forms thereof, or pharmaceutical^ acceptable salt or prodrug forms thereof wherein:
Rle is selected from:

R2e and R3e are independently selected from:
H, C1-C4 alkoxy, NRlleR12e, halogen, N02/ CN, CF3, Ci-Ce alkyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl (C1-C4 alkyl), aryl (Ci-C6 alkyl)-, (Ci-06 alkyl)carbonyl, (Ci-Cg alkoxy) carbonyl, arylcarbonyl, and aryl substituted wit-h 0^4 R7e,

alternatively, when R2e and R3e are substituents on adjacent atoms, they can ba taken together with the carbon atoms to which they are attached to form a 5-7 membered carbocyclic or 5-7 membered heterocyclic aromatic or nonaromatic ring system, said carbocyclic or heterocyclic ring being substituted with 0-2 groups selected from C1-C4 alkyl, C1-C4 alkoxy, halo, cyano, amino, CF3 and NO2;
R2ae ^s selected from:
H, C1-C10 alkyl, C2-C6 alkenyl, C3-C11 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl), aryl, aryl(Ci-C4 alkyl)-, (C2-C7 alkyl}carbonyl,
arylcarbonyl,
(C2-C10 alkoxy)carbonyl, C3-C7 cycloalkoxycarbonyl, C7-C11 bicycloalkoxycarbonyl, aryloxycarbonyl, aryl (C1-C10 alkoxy)carbonyl, C1-C6 alkylcarbonyloxy(Ci-C4 alkoxy)carbonyl, arylcarbonyloxy(C1-C4 alkoxy)carbonyl, and C3-C7 cycloalkylcarbonyloxy(Ci-C4 alkoxy)carbonyl;
R7e is selected from:
H, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, aryl, aryKCi-Ct alkyl)-, (C1-C4 alkyl)carbonyl, C02R18ae, S02Rlle, SO2NR10«Rile, OR10e, and N(Rlle)Rl2e;
U* is selected from:
-(CH2)ne-, -(CH2)neO(CH2)n.e-, -NH (CH2)ne-^ -N(R10e)C(=O)-, -NHC(=0) (CH2)ne-, and -C (=0) N(Rl°e)-;
G« is N or CR19e;
R8e is selected from:
H, C02R18be, C(=0)Rl8be, CONRl'eRlSbe, C1-C10 alkyl substituted with 0-1 R6e, C2-Cio alkenyl substituted with 0-1 R6e,
C2-C10 alkynyl substituted with 0-1 R6e,
C3-Cg cycloalkyl substituted with 0-1 R^e,

Cs-Cg cycloalkenyl substituted with 0-1 R6e,
(Ci-Cio alkyl)carbonyl,
C3-C10 cycloalkyl(C1-C4 alkyl)-,
phenyl substituted with 0-3 R6e,
naphthyl substituted with 0-3 R6e,
a 5-10 membered heterocyclic ring containing 1-3 N, O, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e;
e is selected from:
C1-C10 alkyl substituted with 0-1 R6e,
C1-C10 alkoxy substituted with 0-2 R7e,
H, nitro, N{Rlle)R12e, OC(=O)R10e, OR10e,
OC (sCONR!0^1*, NR10*C(=.O)R10e, NR^c (=0)OR21e, NR1OeC(=O)NR10eRUe, NR10eSO2NR10eRlle/ NR10eS02R21e, hydroxy, ORa3e. ~N(R10e)Rlle, -N{Ri6«)Ri7*, aryl(Co-C6 alkyl)carbonyl, aryKCi-C6 alkyl), heteroaryl (Ci-Ce alkyl), CONR18aeR20e,
S02Ra8ae/ and SQ2NR18aeR20e,
providing that any of the above alKyl, cycloalkyl, aryl or heteroaryl groups may be unsubstituted or substituted independently with 1-2 R7e;
is selected from: H, C1-C10 alkyl, hydroxy, C1-C20 alkoxy, nitro, C1-C10 alkyl carbonyl, -N(Rlle)R12e, cyano, halo, CF3, CHO, C02R18be, C(=0)R18b«, "CONR17eR18be, OC(=O)R10e, OR10*, OC(=0)NRl°«Rlle, NR10eC(=O)Rle, NRlOeCfsOJOR216, NRlOteC (sso)NRl°"Rll», NRl0eSO2NR10eRlle. NRl0eSO2R21e, S(0)peRlle, SO2NR10eR1:Le, aryl substituted with 0-3 groups selected from halogen, Ci-Cg alkoxy, C1-C6 alkyl, CF3, - S(0)m«Me, and -NMe2,

aryl(Ci-C4 alkyl)-, said aryl being substituted with 0-3 groups selected from halogen, C1-C6 alkoxy, Ci-Cs alkyl, CF3, S(0)p*Me, and -NMe2, and a 5-10 membered heterocyclic ring containing 1-3 N, 0, or S heteroatoms, wherein said heterocyclic ring .may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e;
Rioe iS selected from:
H, CF3, C3-C6 alkenyl, C3-C11 cycloalkyl, aryl, (C3-C11 cycloalkyl) methyl, aryl (C1-C4 alkyl), and Ci-C10 alkyl substituted with 0-2 R6e;
Rile is selected from:
H, hydroxy, Ci-Cs alkyl, C3-C6 alkenyl, C3-C11 cycloalkyl, (C3-C11 cycloalkyl)methyl, C1-C6 alkoxy, benzyloxy, aryl, heteroaryl, heteroaryl(Ci~C4 alkyl)-, aryl(Ci-C4 alkyl), adamantylmethyl, and C1-C10 alkyl substituted with 0-2 R4«;
R4e is selected from:
H, Ci-Cg alkyl, C3-C7 cycloalkyl, C3~C7
cycloalkyl(Cx-C4 alkyl)-, aryl, heteroaryl, aryl(C1-
C6 alkyl)-, and heteroaryl(C1~C6 alkyl)-, wherein
said aryl or heteroaryl groups are substituted with 0-2 substituents independently selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, F, CI, Br, CF3, and NO2,
Ri2e iS selected from:
H, Ci-Ce alkyl, triphenylmethyl, methoxymethyl, methoxyphenyldiphenylmethyl,
trimethylsilylethoxymethyl, (C1-C6 alkyl) carbonyl, (Ci-Ce alkoxy)carbonyl, (C1-C6 alkyl)aminocarbonyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(Ci-C4 alkyl)-, aryl, heteroaryl(Ci-Cg alkyl)carbonyl, heteroaryl carbonyl, aryl(Ci-C6 alkyl)-, (C2.-C6 alkyl)carbonyl, arylcarbonyl, Ci-Cg

alkylsulfonyl, arylsulfonyl, aryl(C1-C6 alkylJsulfonyl, heteroarylsulfonyl, heteroaryl(Ci-Cg alkyl)sulfonyl, aryloxycarbonyl, and aryl (Ci-Cg alkoxy)carbonyl, wherein said aryl groups are substituted with 0-2 substituents selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, halo, CF3, and nitro;
R16e is selected from:
-C(=0)0R18ae, -C(=0)R18be, -C(=0)N(R18be)2, -S02R18ae, and -S02N(Rl8J:>e)2;
Ri7e iS selected from:
H, Ci-Cg alkyl, C3-C7 cycloalkyl, C3-C7
cycloalkyl(C1-C4 alkyl)-, aryl, aryl (C1-C6 alkyl)-,
and heteroaryl (Ci-Ce alkyl);
RiSae is selected from:
C1-C8 alkyl optionally substituted with a bond to Ln, C3-C11 cycloalkyl optionally substituted with a bond to Ln. aryl(Ci-Cg alkyl)- optionally substituted with a bond to Ln, heteroaryl(C1-C6 alkyl)- optionally substituted with a bond to Ln. (Ci-Cg alkyl)heteroaryl optionally substituted with a bond to Ln, biaryl(Ci-Ce alkyl) optionally substituted with a bond to Ln, heteroaryl optionally substituted with a 'bond to Ln, phenyl substituted with 3-4 R19e and optionally substituted with a bond to Ln,
naphthyl substituted with 0-4 R19e and optionally substituted with a bond to Ln, and a bond to Ln, wherein said aryl or heteroaryl groups are optionally substituted with 0-4 R19e;
R18be is H or R18ae. R19e is selected from:

H, halogen, CF3/ C02H, CN, N02, -NRlleRl2e/ OCF3^ Ci-Ca alkyl, C2-Ce alkenyl, C2-Ce alkyr.yl, C3-C11 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl)-, aryl(Ca-C6 alkyl)-, Cj-Cs alkoxy, C1-C4 alkoxycarbonyl, aryl, aryl-O-, aryl-S02-, heteroaryl, and heterqaryl-S02-, wherein said aryl and heteroaryl groups are substituted with 0-4 groups selected from hydrogen, halogen, CF3, C1-C3 alkyl, and C1-C3 alkoxy;
R20e j_s selected from:
hydroxy, Cj-Cio alkyloxy, C3-C11 cycloalkyloxy, aryloxy, aryl(C1-C4 alkyl)oxy, C2-C10 alkylcarbonyloxy(Ci-C2 alkyl)oxy-, C2-C10 alkoxycarbonyloxy(Ci-C2 alkyl)oxy^, C2-C10 alkoxycarbonyl(C1-C2 alkyDoxy-, C3-C10 cycloalkylcarbonyloxy(C1-C2 alkyDoxy-, C3-C10 cycloalkoxycarbonyloxy(Cj;-C2 alkyDoxy-, C3-C10 cycloalkoxycarbonyl {C1-C2 alkyDoxy-, aryloxycarbonyl (C1-C2 alkyl) oxy-, aryloxycarbonyloxy(Ci-C2 alkyDoxy-, arylcarbonyloxy C1-C5 alkoxy (C1-C5 alkyl) carbonyloxy (Ci~C2 alkyDoxy, (5-{Ci-C5 alkyl)-l, 3-dioxa-cyclopenten-2-one-yl)methyloxy,
(5-aryl-l,3-dioxa-cyclopenten-2-one-yl)methyloxy,
and (RlOe, {Riip)N_(Cl„Cl0 aik0xy)_;
R21e is selected from:
CI-CB alkyl, C2-C6 alkenyl, C3-C11 cycloalkyl, (C3-C11 cycloalkyl)methyl, aryl, aryl(C1-C4 alkyl)-, and C1-C10 alkyl substituted with 0-2 R7e;
R22e is selected from:
-C(=0)-Ri8be/ -C(=0)N(R18be)2, -C (=0) NHS02R18*e, -C(=0)NHC(=0)R18be, -C(=0)NHC.(=0)0Rl8ae^ ^3 -C (=0) NHS02NHR18be;

me is 0-2;
n* is 0-4; and
pe is 0-2;
with the following proviso: ne and- me are chosen such
that the number of atoms connecting R1 and -COR20e in Formula (IV) is in the range of 8-14;
d is selected from 1, 2, 3, 4, and 5;
d' is 1-50;
W is independently selected at each occurrence from the
group: 0, NH, NHC(=0), C(=0)NH, NR8C(=0), C(=0)NR8, C(=0), C(=0)0, OC(=0), NHC(=S)NH, NHC(=0)NH, S02, (OCH2CH2)s, (CH2CH20}s., (OCH2CH2CH2)s«, (CH2CH2CH20)t, and (aa)c;
aa is independently at each occurrence an amino acid;
Z is selected from the group: aryl substituted with 0-1 R10, C3-.10 cycloalkyl substituted with 0-1 R10, and a
5-10 membered heterocyclic ring system containing
1-4 heteroatoms independently selected from N, S,
and O and substituted with 0-1 R10; 1
R6, R6a, R7, R7a, and R8 are independently selected at each occurrence from the group: H, =0, COOH, SO3H, C1-C5 alkyl substituted with 0-1 R10, aryl substituted with 0-1 R10, benzyl substituted with 0-1 R10, and C1-C5 alkoxy substituted with 0-1 R10, NHC^OJR11, CUOJNHR11, NHC(=0)NHR", NHR^, R", and a bond to Ch;
k is 0 or 1;

s is selected from 0, 1, 2, 3, 4, and 5; s" is selected from 0, 1, 2, 3, 4, and 5; s" is selected from 0, 1, 2, 3, 4, and 5; t is selected from 0, 1, 2, 3, 4, and 5;
A1, A2, A3, A4, A5, A6, A7, and A8 are independently
selected at each occurrence from the group: NR13, NR13R14, S, SH, S(Pg), OH, and a bond to Ln;
E is a bond, CH, or a spacer group independently selected at each occurrence from the group: Ci-Cio alkyl substituted with 0-3 R17, aryl substituted with 0-3 R17, C3_io cycloalkyl substituted with 0-3 and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R17;
R13, and R14 are each independently selected from the group: a bond to Ln, hydrogen, Ci-Cio alkyl substituted with 0-3 R17, aryl substituted with 0-3 R*7, a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R17, and an electron, provided that when one of R13 or R14 is an electron, then the other is also an electron;
alternatively, R13 and R14 combine to form =C{R20) (R21) ;
R17 is independently selected at each occurrence from the group: a bond to Ln, =0, F, CI, Br, I, -CF3, -CN, -C02R18, -C(=0)R18, -C(=0)N -NR19S02N{Rl8)2, -NR19S02R18a, -SO3H, -S02R18a, -S(=0)R18a, -S02N(R18)2. -N(R18)2, -NHC(=S)NHRl8, =NOR18, -C(=0)NHNRl8Rl8a, -OCH2CO2H, and 2- (1-morpholino) ethoxy;

R1?}, ^lSa, an(j ^19 ^re independently selected at each occurrence from the group: a bond to Ln, H, and C1-C6 alkyl;
R20 and R21 are independently selected from the group: H, C1-C5 alkyl, -CO2R25, C2-C5 1-alkene substituted with 0-3 R23, C2-C5 1-alkyne substituted with 0-3 R23, aryl substituted with 0-3 R23, and unsaturated 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0 and substituted with 0-3 R23;
alternatively, R20 and R21, taken together with the
divalent carbon radical to which they are attached form:

R22 and R23 are independently selected from the group: H, and R24;
alternatively, R22, R23 taken together form a fused
aromatic or a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and 0;
R24 is independently selected at each occurrence from the group: -CO2R25, -C(=0)N(R25)2, -CH2OR25, -OC(=0)R25, -OR25, -SO3H, -N(R25)2, and -OCH2CO2H; and,
R25 is independently selected at each occurrence from the -group: H and C1-C3 alkyl.

3. A compound as claimed in claim 2, including stereoisomeric forms thereof, or mixtures of stereoisomeric forms thereof, or pharmaceutically acceptable salt or prodrug forms thereof wherein:
Rle is selected from:

R2e and R3e are independently selected from:
H, C1-C4 alkoxy, NRlleR12e, halogen, NO2, CN, CF3, C1-C6 alkyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl (C1-C4 alkyl), aryl(Ci-C6 alkyl)-, (C1-C6 alkyl)carbonyl, (Ci-Ce alkoxy)carbonyl, arylcarbonyl, and aryl substituted with 0-4 R7e,
alternatively, when R2e and R3e are substituents on adjacent atoms, they can be taken together with the carbon atoms to which they are attached to form a 5-7 membered carbocyclic or 5-7 membered heterocyclic aromatic or nonaromatic ring system, said carbocyclic or heterocyclic ring being substituted with 0-2 groups selected from C1-C4 alkyl, C1-C4 alkoxy, halo, cyano, amino, CF3 and NO2;

R2a* is selected from:
H. Ca-Cio alkyl, C2-Cs alkenyl, C3-C11 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl), aryl, aryl(Ci-C4 alkyl)-, (C2-C7 alkyl)carbonyl,
arylcarbonyl,
(C2-C10 alkoxy)carbonyl, C3-C7 cycloalkoxycarbonyl,
C7-C11 bicycloalkoxycarbonyl, aryloxycarbonyl,
aryl (C1-C10 alkoxy) carbonyl, Ci-Cfi alkylcarbonyloxy(Ci-C4 alkoxy)carbonyl, aryl carbonyl oxy(Ci-C4 alkoxy) carbonyl, and C3-C7 cycloalkylcarbonyloxy(Ci-C4 alkoxy)carbony 1;
R7e is selected from:
H, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, aryl, aryl (Cj.-C4 alkyl)-, (C1-C4 alkyl)carbonyl, C02R18ae, S02Rlle, SO2NR10eRlle, OR10e, and N(Rlle)R12e;
Ue is selected from:
-(CH2)ne-, -NH(CH2)ne-, -N(R10e)C(=0)-, and -NHC(=0) (CH2)ne;
Ge is N or CR19e;
R8« is H;
R9e is selected from:
H, nitro, N(RIle)R12e, OC(=O)R10e, OR10',
OC(=0)NR10eRlle, NR10eC(=O)R10e, NR10eC (=0)0R21e, NRa0eC(=O)NR10eR1:le, NR10eSO2NR;i0eR;L:Le, NR10eSO2R21e, hydroxy, OR22*5, -N(R10e)Rlle, -N(R16e)R17e, aryl(C0-C4 alkyl) carbonyl, aryl (Cj-C4 alkyl), heteroaryl(Ci-C4 alkyl), CONR18aeR20e, S02Ra8*e, and S02NR18a«R20e, providing that any of the above alkyl, cycloalkyl, aryl or heteroaryl groups may be unsubstituted or substituted independently with 1-2 R7e;

R10e is selected from:
H, CF3, C3-C6 alkenyl, C3-C6 cycloalkyl, aryl, (C3-C6 cycloalkyl)methyl, aryl(C1-C4 alkyl), and C1-C4 alkyl substituted with 0-2 R6e;
R6e is selected from:
H, C1-C4 alkyl, hydroxy, C1-C4 alkoxy, nitro, C1-C4 alkylcarbonyl, -N{Rlle)R12e/ cyano, halo, CF3, CHO, C02R18be, C(=0)R18be, CONR17eR18be,
OC(=O)R10e, OR10e, OC(=O)NR10eRlle,
NR10eC(=O)R10e, NR10eC(=O)OR2Ie.
HR10eC (=0) NR10eRlle, NR^SC^NR10*^11*,
NR10eS02R21e, S(0)pRlle, S02NR10eRlle, aryl substituted with 0-3 groups selected from
halogen, C1-C4 alkoxy, C1-C4 alkyl, CF3,
S(0)meMe, and -NMe2, aryl (C1-C4 alkyl)-, said aryl being substituted with
0-3 groups selected from halogen, C1-C4 alkoxy, C1-C4 alkyl, CF3, S(0)peMe, and -NMe2, and a 5-10 membered heterocyclic ring containing 1-3 N, O, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e;
Riie iS selected from:
H, hydroxy, C1-C4 alkyl, C3-C6 alkenyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)methyl, C1-C4 alkoxy,
benzyloxy, aryl, heteroaryl,
heteroaryl (C1-C4 alkyl)-, aryl(Ci-C4 alkyl),
adamantylmethyl, and
C1-C4 alkyl substituted with 0-2 R4e;

R4e is selected from;
H, Cx-C4 alkyl, C3-C7 cycloalkyl, C3-C7
cycloalkyl (C^-^ alkyl)-, aryl, heteroaryl, aryKC^-
C4 alkyl)-, and heteroaryl(Cj-C^ alkyl)-, wherein
said aryl or heteroaryl groups are substituted with 0-2 substituents independently selected from the group consisting of C3.-C4 alkyl, C1-C4 alkoxy, F, CI, Br, CF3, and NO2,
Ri2e iS selected from:
H. C1-C4 alkyl, (C1-C4 alkyl)carbonyl, (C1-C4" alkoxy) carbonyl, phenyl (C1-C4 alkyl) -, phenylsulfonyl, phenyloxycarbonyl, and phenyl(C1-C4 alkoxy)carbonyl, wherein said phenyl groups are
substituted with 0-2 substituents selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, halo, CF3, and nitro;
Ri6e iS selected from:
-C(=0)OR18a« -C(*0)R18be, -C(=0)N(R18be)2/ -S02R18a"e, and -S02N(R18be)2;
R17e is selected from:
H,. C1-C4 alkyl, C3-C6 cycloalkyl, C3-C6
cycloalkyl (C1-C4 alkyl)-, aryl, aryl (Ci-Ce alkyl)-,
and heteroaryl(C1-C6 alkyl);

Ri8ae is selected from:
Ci-Cs alkyl optionally substituted with a bond to Ln, C3-C11 cycloalkyl optionally substituted with a bond to Ln, aryKCi-Cg alkyl)- optionally substituted with a bond to Ln, heteroaryl (C1-C6 alkyl}- optionally substituted with a bond to in- (Ci-Cg alkyl)heteroaryl optionally substituted with a bond to Ln, biaryl (Ci-Ce alkyl) optionally substituted with a bond to Ln, heteroaryl optionally substituted with a bond to Ln R18be is H or Rl8ae.
Rl9e iS selected from:
H, halogen, CF3, CO2H, CN, N02, -NRlleR12e, OCF3, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C3-C6 cycloalkyl(C1-C4 alkyl)-, aryl(Ci-C4 alkyl)-, C1-C6 alkoxy, C1-C4 alkoxycarbonyl, aryl, aryl-O-, aryl-S02-,
heteroaryl, and heteroaryl-SO2--, wherein said aryl and heteroaryl groups are substituted with 0-4 groups selected from hydrogen, halogen, CF3, C1-C3 alkyl, and C1-C3 alkoxy;

p20e is selected from:
hydroxy, Ci-Cg alkyloxy, C3-C6 cycloalkyloxy,
aryloxy, aryl(C1-C4 alkyDoxy,
C2-C10 alkylcarbonyloxy(Ci-C2 alkyDoxy-,
C2-C10 alkoxycarbonyloxy(Ci-C2., alkyDoxy-,
C2~Cio alkoxycarbonyl (C1-C2 alkyDoxy-,
C3-C10 cycloalkylcarbonyloxy(Ci-C2 alkyDoxy-,
C3-C10 cycloalkoxycarbonyloxy(Ci-C2 alkyDoxy-,
C3-C10 cycloalkoxycarbonyl (C1-C2 alkyDoxy-,
aryloxycarbonyl (C1-C2 alkyDoxy-,
aryloxycarbonyloxy (C1-C2 alkyl)oxy-,
arylcarbonyloxy(C1-C2 alkyl)oxy-,
C1-C5 alkoxy(Ci-Cs alkyl)carbonyloxy(C1-C2 alkyDoxy,
(5- {C1-C5 alkyl) -1,3-dioxa-cyclopenten-2-one-
yl) methyl oxy, (5-aryl-l,3-dioxa-cyclopenten-2-one-yl)methyloxy,
and tR10e) (Rlle)N-(Ci-Cio alkoxy)-;
R21e is selected from:
C3.-C4 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)methyl, aryl, aryl (C1-C4 alkyl)-, and C1-C10 alkyl substituted with 0-2 R7e;
R22e iS selected from:
-C(=0)-R18be, -C(=0)N(R18be)2, -C{=0)NHS02R18ae, -C(=0)NHC(=0)R18^, -C(=0)NHC(=0)OR18ae, and -C (=0) NHS02NHRl8be;
me is 0-2;
ne is 0-4;

e is 0-1; h is

A1 is selected from the group: OH, and a bond to Ln;
A2, A4, and A6 are each N;
A3, A5, and A8 are each OH;
A7 is a bond to Ln or NH-bond to Ln;
E is a C2 alkyl substituted with 0-1
R17 is =0; alternatively, CQ is

A1 is selected from the group: OH, and a bond to Ln;
A2, A3 and A4 are each N; A5, A6 and A8 are each OH; A7 is a bond to Ln,-


E is a C2 alkyl substituted with 0-1 R17; R17 is =0;
alternatively,
A1 is NH2 or N=C(R3°](R21);
E is a bond;
A2 is NHR13;
R13 is a heterocycle substituted with R17, the heterocycle being selected from pyridine and pyrimidine;
R17 is selected from a bond to Ln, C(=0) NHR18 and C(=0)R18;
R18 is a bond to LR;
R24 is selected from the group: -CO2R25, -OR25, -SO3H, and -N(R25)2; and,
R25 is independently selected at each occurrence from the group: hydrogen and methyl.
4. A compound as claimed in claim 1, including enantiomeric or diastereomeric forms thereof, or mixtures of enantiomeric or diastereomeric forms thereof, or pharmaceutical^ acceptable salt or prodrug forms thereof, wherein Q is selected from the group:
3-[7-r(imida2olin-2-ylamino)methyl]-l-methyl-6,8-dif luoroguinoline-4-one-3 -yl car bony lamino] -2-(3,5-dimethylisoxazol-4-ylsulfonylamino)propionic acid,

3- [7-( (inridazolin-2-ylainino)methyl]-l-meth.yl-6,8-di f luoroguinoline-4 -one-3 -ylcarbonylamino] -2 -(benzyloxycarbonylamino)propionic acid, 3- [7-[ (imidazolin-2-ylamino)methyl]-l-methyl-6,8-difluoroguinoline-4-one-3-ylcarbonylamino]-2-(n-butyloxycarbonylamino) propionic acid, 3- [7-[ (imidazolin-2-ylamino) methyl]-l-methyl-6,8-dif luoroguinoline~4-one-3 -ylcarbonylamino] -2-(n-butylsulfonylamino)propionic acid, 3- [7- [ (tetrahydropyrimid-2-ylamino)methyl] -l-methyl-6,8-dif luoroguinoline-4-one-3-ylcarbonylaitu.no] -2- (benzyloxycarbonylamino)propionic acid, 3- 17- [ (tetrahydropyrimid-2-ylamino)methyl] -1-methyl-6,8-dif luoroguinoline-4-one-3-ylcarbonylamino] -2- (n-butyloxycarbonylamino) propionic acid, 3- [7- [ (tetrahydropyrimid-2-ylamino) methyl] -1-methyl-6,8-dif luoroguinoline-4-one-3-ylcarbonylamino] -2- (phenylsulfonylamino)propionic acid, 3- (7- C (tetrahydropyrimid-2-ylamino)methyl] -l-methyl-6,8-difluoroguinoline-4-one-3-ylcarbonylamino]-2-(n-butylsulfonyl)aminopropionic acid, 3- [7-[ (2-aminothiazol-4-yl)methyl]~l-methyl-6,8~ difluoroquinoline-4-rone-3-ylcarbonylamino]-2-(benzyloxycarbonylamino)propionic acid, 3- [7-[(iroidazolin-2-ylamino)methyl]-l-methyl-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4, 6-trimethylphenyl)sulfonylamino)propionic acid, 3- [7- [ (tetrahydropyrimid-2-ylamino)methyl] -1-methyl-6,8-dif luoroguinoline-4-one-3-ylcarbonylamino] -2-((2,4,6-
trimethylphenyDsulfonylamino)propionic acid, 3- [7-[(imidazol-2-ylamino)methyl]-l-methyl-6,8-
dif luoroguinoline-4-one-3-ylcarbonylamino] -2-(3,5-dimethylisoxazol-4-ylsulfonylamino)propionic acid,

3-[7-[(imidazol-2-ylamino)methyl]-l-methyl-6, 8-
difluoroquinoline-4-one-3-ylcarbonylamino]-2-(benzyloxycarbonylamino) propionic acid, 3- [7- [ (imidazol-2-ylamino)methyl] -l-methyl-6, 8-
difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4, 6-trimethylphenyl)sulfonylamino)propionic acid, 3- [7- ((imidazol-2-ylamino) methyl]-l-methyl-6, 8-
difluoroquinoline-4-one-3-ylcarbonylamino]-2-((4-biphenyl) sulfonylamino) propionic acid, 3- [7- [ (imidazol-2-ylamino)methyl] -l-methyl-6,8-
difluoroguinoline-4-one-3-ylcarbonylamino]-2-(1-naphthylsulfonylamino 5 propionic acid, 3-[7-[ (benzimidazol-2-ylamino) methyl] -l-methyl-6,8-difluoroguinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-trimethylphenyl) sulf onylamino)propionic acid, 3- [7- [ (4-methylimidazol-2-ylamino)methyl] -1-methyl-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-
trimethylphenyl) sulfonyl amino) propionic acid, 3-[7-[(4,5-dimethylimidazol-2-ylamino)methyl] -1-methyl-6,8-dif luoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-trimethylphenyl) sulfonylamino) propionic acid, 3- [7-[ (4,5, 6,7-tetrahydrobenzimidazol-2-
ylamino)methyl] -l-methyl-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4, 6-trimethylphen-yl) sulf onylamino) propionic acid, 3- f7-f (pyridin-2-ylamino)methyl] -l-methyl-6,8-
difluoroquinoline-4-one-3-ylcarbonylamino]-2-
((2,4,6-trimethylphenyl) sulf onylamino) propionic
acid, '■•■
3-(7-(2-aminopyridin-6-yl)-l-methyl-6,8-
difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4, 6-trimethylphenyl) sulf onylamino) propionic acid,

3-[7-[ (7-azabenzimidazol-2~yl)methyl]-l-methyl-6,8-difluoroguinoline-4-one-3-ylcarbonylainino] -2-{(2,4,6-trimethylphenyl)sulfonylamino)propionic acid, 3-[7- [(benzimidazol-2-ylamino)methyl]-l-(2-
phenylethyl) -6,8-difluoroquinoline-4-one-3-ylcarbonylamino]pro-pionic acid, 3- [7- [ (pyridin-2-ylamino)methyl] -1- (2-phenylethyl) -6,8-difluoroguinoline-4-one-3~ ylcarbonylamino]propionic acid, 3-[7-[ {imidazolin-2-ylamino)methyl]-1- (2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]propionic acid, 3-£7-[ (imidazol-2-ylamino)methyl]-l- (2-phenylethyl) -6,8-difluoroquinoline-4-one-3-ylcarbonylamino]propionic acid, 3-[7-£ (imidazolin-2-ylamino)methyl]-l- phenylethyl) -6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-
(benzyloxycarbonylamino)propionic acid, 3-I7-r(imidazolin-2-ylainino)methyl]-l-(2-
phenylethyl)-6, 8-dif luoroquinoline-4-one-3-ylcarbonylamino]-2-(n-butyloxycarbonylamino)propionic acid, 3-[7-[(imidazolin-2-ylamino)methylJ-1-(2-
phenylethyl)-6, 8-difluoroguinoline~4-one-3-ylcarbonylamino]-2-(phenylsulfonylamino)propionic acid, 3-[7-[(imidazolin-2-ylamino)methyl]-l- (2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-(n-butylsulfonylamino) propionic acid, 3- [7- [ (tetrahydropyrimid-2-ylamino) methyl J -1- (2--phenylethyl) -6, 8-difluoroquinoline-4-one-3~ ylcarbonylamino]-2-
(benzyloxycarbonyl amino) propionic acid, 3- [7- [ (tetrahydropyrimid-2-ylamino)methyl] -1- (2-phenylethyl) -6,8-dif luoroquinoline-4-one-3-

ylcarbonylamino]-2-(n-butyloxycarbonylamino) propioni c acid, 3- [7- [ (tetrahydropyriMd-2-ylamino) methyl] -1- (2-phenylethyl) -6, 8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-(phenylsulfonylamino) propionic acid, 3-[7-[(tetrahydropyrimid-2-ylamino)methyl]-l- (2-phenylethyl) -6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-(n-butylsulfonyUaminopropionic acid, 3-[7-[(2-aminothiazol-4-yl)methyl]-1- (2-
phenylethyl) -6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-(phenylsulfonylamino)propionic acid, 3-[7-[(2-aminothiazol-4-yl)methyl]-1- (2-
phenylethyl) ~6,8-dif luoroquinoline-4:-one-3-ylcarbonylamino] -2-
(benzyloxycarbonylamino) propionic acid, 3- [1- [ {imidazolin-2-ylamino)methyl] -1- (2-
phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-
trimethylphenyl) sulf onylamino) propionic acid, 3- [l-[ (tetrahydropyrimid-2-ylamino)methyl]-l- (2-phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-
trimethylphenyl) sulfon-ylamino)propionic acid, 3- [7- [ (imidazol-2-ylamino)methyl] -1- (2-phenylethyl) -6,8-dif luoroquinoline-4-one-3-ylcarbonylamino] -2- (benzyloxycarbonylamino) propionic acid, 3- [7- [ (imi dazol-2-ylamino) methyl] -1-(2-phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino] -2- (phenylsulf onylamino) propionic acid, 3- [7- { (imidazol-2-ylamino)methyl] -1- (2-phenylethyl) -6,8-dif luoroquinoline-4-one-3-ylcarbonylamino] -2- ((2, 6, dichlorophenyl) sulf onylamino) propionic acid, 3- [7- t (imidazol-2-ylamino)methyl] -1- (2-phenylethyl)-6,8-dif luoroquinoline-4-one-3-ylcarbonylamino] -

2-((2,4,6r
trimethylphenyl) sulfonylamino) propionic acid, 3- [7- [ (imidazol-2-ylaroino)methyl] -1- (2-phenylethyl) -6, 8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((4-biphenyl)sulfonylamino)propionic acid, 3- [7- [ (benzimidazol-2-ylamino)methyl] -1- (2-
phenylethyl)-6,8-difluoroguinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-trimethylphenyl) sulfonylamino)propionic acid, 3- [7-1 (4-methyliinidazol-2-ylamino)methyl] -1- (2-phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-tritnethylphenyl) sulfonylamino) propionic acid, 3-[7-[(4, 5-dimethylimidazol-2-ylamino)methyl]-l- (2-phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-trimethylphenyl) sulf onylamino) propionic acid, 3-17-f(4,5,6,7-tetrahydrobenzimidazol-2-
ylamino) methyl] -1- (2-phenylethyl)-6,8-difluoroquinoline-4-one-3-ylcarbonylamino3-2-((2,4, 6-trimethylphenyl) sulf onylamino) propionic acid, 3-[7-[ (pyridin-2-ylamino)methyl] -1- (2-phenylethyl) -6,8-difluoroguinoline-4-one-3-ylcarbonylamino]-2-((2>4,6-
t rime thy lphenyl) sulf onylamino) propionic acid, 3-[7-(2-aminopyridin-6-yl)-l-(2-phenylethyl)-6,8-difluoroguinoline-4-one-3-ylcarbonylamino]-2-((2,4,6-trimethylphenyl)sulfonylamino)propionic acid, and 3- [7- [ (7-azabenzimidazol-2-yl)methyl] -1- (2-
phenylethyl) -6, 8-difluoroquinoline-4-one-3-
yl car bony 1 amino] -2- ((2, 4, 6-
trimethylphenyl) sulf onylamino) propionic acid.
A compound as claimed in claim 1, wherein the compound selected from the group:

2-(((4-(4-(((3-(2-(2-(3-((6-((l-aza-2-(2-
sulf©phenyl) vinyl) amino) (3-pyridyl)) carbonylamino) -
propoxy)ethoxy)-
ethoxy) propyl) amino) s ul f onyl) phenyl) phenyl) -
sulfonyl)amino)-3-( (7- ((imidazol-2-ylamino) methyl)-
l-methyl-4-oxo(3-
hydroquinolyl)) carbonylamino)propanoic acid;
3-( (7- ((imidazol-2-ylamino) methyl)-l-methyl-4-oxo(3-
hydroquinolyl)) carbonylamino) -2- {((4-(4-( ((3-(2-(2-
(3-(2-(l,4,7,10-tetraaza-4,7,10-
tris (carboxylmethyl) cyclododecyl) acetylamino) -
propoxy) ethoxy) ethoxy) propyl) amino) sulfonyl) -
phenyl) phenyl) sulf onyl) amino) propanoic acid;
2-(({4-(3-(N-(3-(2-(2-(3-((6-((l-aza-2-(2-
sulfophenyl)vinyl)amino) (3-pyridyl))carbonylamino) -propoxy) ethoxy) ethoxy) propyl) carbamoyl)propoxy) -2, 6j dimethylphenyl)sulfonyl)amino)-3-( (7- ((imidazol-2-ylamino) methyl) -1-methyl-4-oxo (3-hydroquinolyl)) -carbonylamino)propanoi c;
3-((l-(3-((6-((l-aza-2-(2-sulfophenyl)vinyl)amino)(3-pyridyl)) carbonylamino)propyl) -7- ((imidazole-2-ylamino)methyl)-4-oxo(3-hydroguinolyl))carbonylamino)-2-(((2,4,6-trimethylphenyl)sulfonyl)amino)propanoic acid;
3-((l-(3-((6-((l-aza-2-(2-sulfophenyl)vinyl)amino)(3-pyridyl))carbonylamino)propyl)-7-(((1-hydroxyimidazole-2-yl) amino)methyl) -4-oxo(3-hydroguinolyl))carbonylamino)-2-(((2,4,6-tr imethylphenyl) sulf onyl) amino) propanoic acid;
3-((1-(3-(3-(N-(3-(2-(2-(3-((6-((l-aza-2-(2-s ul f ophenyl) vinyl) amino) (3 -pyridyl)) carbonylamino)propoxy) ethoxy)-ethoxy) propyl) carbamoyl) propanoylamino) propyl) -7-((imidazole-2-ylamino)methyl)-4-oxo{3-

hydroquinolyl))carbonylamino) -2- (({2,4,6-trimethyl phenyl)sulfonyl)amino)propanoic acid;
262
2-(2-aza-2-(5- (N- (1,3-bis(3-(2-(2-(3-(3-(N-(3-{3- (N- (3-carboxy-2- (((2,4,6-trimethylphenyl)sulfonyl)amino) -ethyl) carbamoyl)-7- ((imidazole-2-ylamino)methyl) 4-oxohydroquinolyl) propyl) carbamoyl) propanoyl amino) pro poxy) ethoxy) ethoxy)propyl) carbamoyl) (2-pyridyl)) amino) vinyl)benzenesulfonic acid;



?/rc


2-( dimethylphenyl)sulfonyl)amino)-3-((7-((imidazol-2-ylamino)methyl)-l-methyl-4-oxo(3-hydroquinolyl))-carbonylamino)propanoic acid ;
2-(((4-(3-(N~(3-(2-(2-(3-(2-(l,4,7,10-tetraaza-4,7,l0-tris (carboxymethyl) cyclododecylacetylamino) -6- (2-(bis (phosphonomethyl) amino) acetylamino)hexanoylamino ) propoxy) ethoxy) ethoxy) propyl) carbamoyl) propoxy) -2, 6-dimethylphenyl) sulf onyl) amino) -3- ((7- ((imidazol-2-ylamino)methyl)-l-methyl-4~oxo(3-hydroctuinolyl)) -carbonylamino)propanoic acid conjugate; and
2- {((4-(3-(N-(3-(2-(2-(3-(2-(2-((2-((2-(bis(carboxymethyl)-
amino)ethyl) (carboxymethyl)amino)ethyl) (carboxymethy 1) amino) acetylamino) -3-sulfopropyl) propoxy) ethoxy) -ethoxy) propyl) carbamoyl) propoxy) -2,6-

265
dimethyl phenyl )sulfonyl) amino)-3-((7-((imldazol-2-ylamino) methyl) -1-methyl-4-oxo (3-hydroquinolyl)) -carbonyl amino) propanoic acid;




C02H

»

266


0 ^0
N N HHN^S03H
H02C^ v_y ^COzH *i
H H UI^N^YOH
0 0 O .;
or a pharmaceutically acceptable salt form thereof.
6. A kit comprising a compound as claimed in claim 1, or pharmaceutically acceptable salt form thereof and a pharmaceutically acceptable carrier.
7. A kit comprising as claimed in claim 6, wherein the kit further comprises one or more ancillary ligands and a reducing agent.
8. A kit as claimed in claim 7, wherein the ancillary ligands are tricine and TPPTS.
9. A kit as claimed in claim 7, wherein the reducing agent, is tin (II) .
10. A diagnostic or therapeutic metallopharmaceutical composition, comprising a compound as claimed in claim 1 and further comprising a metal.

11. A composition as claimed in claim 10, wherein the metallopharmaceutical is a diagnostic radiopharmaceutical, the metal is a radioisotope selected from the group: 99mTc, 95Tc, U1ln, 62Cu, 64Cu, 67Ga, and 68Ga, and the linking group is present between the non-peptide targeting moiety and chelator.
12. A composition as claimed in claim 11, wherein the targeting moiety is a quinolone non-peptide and the receptor is 13. A composition as claimed in claim 12, wherein the radioisotope is 99mTc or 95Tc, the radiopharmaceutical further comprises a first ancillary ligand and a second ancillary ligand capable of stabilizing the radiopharmaceutical.
14. A composition as claimed in claim 13, wherein the radioisotope is 99mTc.
15. A composition as claimed in claim 14, wherein the radiopharmaceutical is selected from the group:
99nvrc (2T {((4- (4- (((3~ (2- (2- (3- ((6- (diazenido) (3-pyridyl)) carbonylamino) propoxy) ethoxy) -ethoxy) propyl) amino) sulf onyl) phenyl) phenyl) -sulfonyl)amino)-3- {(7- ((imidazol-2-ylamino)niethyl) -l-methyl-4-oxo{3-
hydroquinolyl)) carbonylamino) propanoic acid)(tricine)(TPPTS);
99nTc (2- (((4- (3- (N- (3- (2- (2- (3- ((6- (diazenido) (3-pyridyl)) carbonylamino) propoxy) ethoxy) -ethoxy) propyl) carbamoyl) propoxy) -2,6-dimethylphenyl)sulfonyl)amino)-3- ((7- ((imidazol-2-

ylamino) methyl) -l-methyl-4-oxo (3-hydroquinolyl)) -carbonylamino)propanoic acid) (tricine) (TPPDS);
99nj;c o- ((i- (3- ((6- (diazenido) (3-
pyridyl)) carbonylamino)propyl) -7- ((imidazole-2-ylamino) methyl)-4-oxo (3-
hydroquinolyl))carbonylamino)-2- (((2,4,6-trimethylphenyl) sulf onyl) amino) propanoic acid)(tricine)(TPPTS);
"«Tc(3-((1-(3-((6-(diazenido)(3-
pyridyl)) carbonylamino) propyl)-7-(((1-hydroxyimidazole-2-yl)amino)methyl) -4-oxo (3-hydroquinolyl)) carbonylamino) -2-(((2,4,6-trimethylphenyl) sulfonyl) amino) propanoic acid)(tricine)(TPPTS);
99nTc (3- ((1- (3- (3- (N- (3- (2- (2- (3- ((6- (diazenido) (3-pyridyl)) carbonylamino) propoxy) ethoxy) -ethoxy) propyl) carbamoyl) propanoylamino) propyl) -7-((imidazole-2-ylamino)methyl) -4-oxo(3-hydroquinolyl) ) carbonylamino)-2- (((2,4,6-trimethylphenyl) sulfonyl) amino) propanoic acid)(tricine)(TPPTS);
99nTc(2-(2-(5-(N-(l,3-bis(3-(2-(2-(3-(3-(N-(3-(3-(N-(3-carboxy-2- (((2,4,6-trimethylphenyl) sulfonyl)amino) -ethyl)carbamoyl)-7- ((imidazole-2-ylamino)methyl)4-oxohydroquinolyl) propyl) carbamoyl) propanoylamino) propoxy) ethoxy) ethoxy) propyl) carbamoyl) (2 -pyridyl)diazenido))(tricine)(TPPTS);
99wTc(3-{[1-(3-{2-[(6-(diazenido)(3-
pyridyl)) carbonylamino] (2R) -3-sulfopropyl}propyl) -7-[ (imidazol-2-ylamino)methyl]-4-oxo(3-hydroguinolyl) ]carbonylamino} (2S)-2~{ [ (2,4,6-

trimethylphenyl) sulfonyl] eunino}propanoic acid) (tricine)(TPPTS).
16. A composition as claimed in claim 12, wherein the radioisotope is 111In.
17. A composition as claimed in claim 16, wherein the radiopharmaceutical is selected from the group:



271



18. A composition as claimed in claim 10, wherein the
metallopharmaceutical is a therapeutic
radiopharmaceutical, the metal is a radioisotope selected from the group: ^6Re, «8Re, ^3Smt igsHO/ 177LU/ i49Em/
*°Y, 212Bi/ 103pd; 109pd, 159Gd/ 140La/ J98Au, W9Au# 169Yb, 175Yb( ISScy, 166ny, 67CU/ 105Rh, 111^, ^ W2Ir, and che
linking group is present between the non-peptide targeting moiety and chelator.
19. A composition as claimed in claim 18, wherein the
targeting moiety is a quinolone non-peptide and the
receptor is avP3 or oivpV
20. A composition as claimed in claim 19, wherein the radioisotope is 153Sm.
21. A composition as claimed in claim 19, wherein the radioisotope is 177Lu.
22. A composition as claimed in claim 21, wherein the radiopharmaceutical is selected from the group:
o o o



and

23. A composition as claimed in claim 19, wherein the radioisotope is 90Y.
24. A composition as claimed in claim 23, wherein the radiopharmaceutical is selected from the group;




276


25. A composition as claimed in claim 10, wherein the metallopharmaceutical is a MRI contrast agent, the metal is a paramagnetic metal ion selected from the group: Gd(III), Dy(III), Fe(III), and Mn(II), the targeting moiety is a quinolone nonpeptide and the linking group is present between the targeting moiety and chelator.
26. A composition as claimed in claim 25, wherein the targeting moiety is quinolone non-peptide and the receptor
is avp3 or avps.
27. A composition as claimed in claim 26, wherein the metal ion is Gd(III).
28. A composition as claimed in claim 27, wherein the contrast agent is
277


29. A composition as claimed in claim 10, wherein the metallopharmaceutical is a X-ray contrast agent, the metal is selected from the group: Re, Sm, Ho, Lu, Pm, Y, Bi, Pd, Gd, La, Au, Au, Yb, Dy, Cu, Rh, Ag, and Ir, the targeting moiety is a quinolone non-peptide, the receptor is avp3 or avP5, and the linking group is present between the targeting moiety and chelator.
30. A compound, comprising: a targeting moiety and a surfactant, wherein the targeting moiety is bound to the surfactant, is a nonpeptide, and binds to a receptor that is upregulated during angiogenesis and the compound has 0-1 linking groups between the targeting moiety and surfactant.
31. A compound as claimed in claim 30, wherein the targeting moiety comprises a quinolone non-peptide and linking group is present between the targeting moiety and surfactant.

32. A compound as claimed in claim 31, wherein the receptor is the integrin avp3 or ctvps and the compound is of the formula:
(Q)d-Ln-Sf wherein, Q is a compound of Formula (II):

including stereoisomeric forms thereof, or mixtures of stereoisomeric forms thereof, or pharmaceutically acceptable salt or prodrug forms thereof wherein:
Rle is selected from:


Ae is -CH2- Or -N(R10«)-;
Ale and Be are independently -CH2- or -N(R10e)-/
D« is -N(R10e)- or -S-;
E*-Fe is -C(R2e)=C(R3e)- or -C(R2e)2C(R3e)2-;
Je is -C(R2e)- or -N-;
K«, Le and M* are independently -C(R2«)- or -C(R3e)-;
R2e and R3e are independently selected from:
H, C1-C4 alkoxy, NR"«R"ef halogen, NO2, CN, CF3, Ci-Ce alkyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl {C1-C4 alkyl), aryl(Ci-C6 alkyl)-, (C1-C6 alkyl)carbonyl, (Ci-C$ alkoxy)carbonyl, arylcarbonyl, and aryl substituted with 0-4 R7e,
alternatively, when R2e and R3e are substituents on adjacent atoms, they can be taken together with the carbon atoms to which they are attached to form a 5^ 7 membered carbocyclic or 5-7 membered heterocyclic aromatic or nonaromatic ring system, said carbocyclic or heterocyclic ring being substituted with 0-2 groups selected from C1-C4 alkyl, C1-C4 alkoxy, halo, cyano, amino, CF3 and NO2;
R2«e is selected from:
H, C1-C10 alkyl, C2-C6 alkenyl, C3-CU cycloalkyl, C3-C7 cycloalkyl (C1-C4 alkyl), aryl, aryl(Ci-C4 alkyl)-, (C2-C7 alkyl)carbonyl, arylcarbonyl, (C2-C10 alkoxy)carbonyl, C3-C7 cycloalkoxycarbonyl, C7-C11 bicycloalkoxycarbonyl, aryloxycarbonyl, aryl {C1-C10 alkoxy)carbonyl, Ci-Cg alkylearbonyloxy(Ci-C4 alkoxy)carbonyl, arylcarbonyloxy(Ci-C4 alkoxy)carbonyl, and

C3-C7 cycloalkylcarbonyloxy(Ci-C4 alkoxy)carbonyl;
R7e is selected from:
H, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, aryl, aryKCi-C4 alkyl)-, (C1-C4 alkyl)carbonyl, C02R1Bae, S02Rlle* SO2NR10*Rlle, OR10e, and N(Rlle)Ri2e;
Ue is selected from:
- (CH2)ne~, - (CH2)neO (CK2) me-, - (CH2) neN(R12) (CH2) ae-. -NH(CH2)ne-, -(CH2)neC(=0) (CH2)me-,
- (CH2) neS (0) pe {CH2) me-, - (CH2) neNHNH (CH2) me-, -N(R10e)C(=O)-, -NHC(=0) (CH2)ne-, -C(=O)N(R10e) -, and
G« is N or CR19e;
We is-C(=O)-N R8e and R9e are independently selected from: H, C02R18be, C(=0)R18be, CONR17R18be, C1-C10 alkyl substituted with 0-1 R6e, C2-Cio alkenyl substituted with 0-1 R6e, C2-Cio alkynyl substituted with 0-1 R6e, C3-C8 cycloalkyl substituted with 0-1 R6«, C5-C6 cycloalkenyl substituted with 0-1 R6e, (C1-C10 alkyl)carbonyl, C3-C10 cycloalkyl (C1-C4 alkyl)-, phenyl substituted with 0-3 R6e, naphthyl substituted with 0-3 R6e, a 5-10 membered heterocyclic ring containing 1-3 N, 0, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e, C1-C10 alkoxy substituted with 0-2 R7e, hydroxy, nitro, -N(R*°«)R"-e, -NfR16*^7*, aryl(Co~C6 alkyl)carbonyl, aryl(C3-C6 alkyl),

heteroaryl (Ci-C6 alkyl), CONR18aeR20e, S02R18ae, and SO2NR18aaR20«, providing that any of the above alkyl, cycloalkyl, aryl or heteroaryl groups may be unsubstituted or substituted independently with 1-2 R7e;
R6e is selected from:
H, C1-C10 alkyl, hydroxy, C1-C10 alkoxy, nitro, C1-C10 alkylcarbonyl, -N(Rlle)R12e, cyano, halo, CF3, CHO, C02R18be, C(=0)R18be, CONR17eRi8be, OC(=O)R10e, OR10«, OC(=O)NR10eRlle, NR1OeC(=O)R10e, NR10eC(=O)OR21e, NRl0eC(=O)NR10eRlle, NR1OeSO2NRl0eRlle, NR10eSO2R21e, S(0)pRlle, S02NR1 aryl substituted with 0-3 groups selected from halogen, C1-C6 alkoxy, C1-C6 alkyl, CF3, SlO)meHe, and -NMe2. aryl(Ci-C4 alkyl)-, said aryl being substituted with 0-3 groups selected from halogen, Ci-Ce alkoxy, Ci-Ce alkyl, CF3, SfOJp^Me, and -NMe2, and a 5-10 membered heterocyclic ring containing 1-3 N, 0, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e;
plOe is selected front:
H, CF3, C3-C6 alkenyl, C3-C11 cycloalkyl, aryl, (C3-C11 cycloalkyl) methyl, aryl (C1-C4 alkyl), and Cj-C10 alkyl substituted with 0-2 R6«;
Rlle is selected from:
H, hydroxy, Ci-Cg alkyl, C3-C6 alkenyl, C3-C11 cycloalkyl, {C3-C11 cycloalkyl)methyl, C1-C6 alkoxy, benzyloxy, aryl, heteroaryl, heteroaryl (C3.-C4 alkyl)-, aryl(Gi-C4 alkyl), adamantylmethyl, and C1-C10 alkyl substituted with 0-2 R4e;
R4e is selected from:

H, C^-Cg alkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl (C^-C^ alkyl)-, (C1-C10 alkyl)carbonyl, aryl, heteroaryl, aryl ^-Cg alkyl)-, and heteroaryl (C^-Cg alkyl)-, wherein said aryl or heterparyl "groups are substituted with 0-2 substipuents independently selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, F, CI, Br, CF3, and NO2,
alternatively, when R10e and Rlle are both substituents on the same nitrogen atom (as in -NR10eRlle) they may be taken together with the nitrogen atom to which they are attached to form a heterocycle selected from: 3-azabicyclononyl, 1,2,3,4-tetrahydro-lrquinolinyl, 1,2,3,4-tetrahydro-2-isoguinolinyl, 1-piperidinyl, 1-morpholinyl, 1-pyrrolidinyl, thiamorpholinyl, thiazolidinyl, and 1-piperazinyl; said heterocycle being substituted with 0-3 groups selected from: C1-C6 alkyl, aryl, heteroaryl, aryl(Ci-C4 alkyl)-, (Ci-Ce alkyl)carbonyl, (C3-C7 cycloalkyl) carbonyl, (C1-C6 alkoxy)carbonyl, aryl(Ci-C4 alkoxy)carbonyl, C1-C6 alkylsulfonyl, and arylsulfonyl,-
Ri2e iS selected from:
H, Ci-Cg alkyl, triphenylmethyl, methoxymethyl, methoxyphenyldiphenylmethyl,
trimethylsilylethoxymethyl, (Ci-C6 alkyl) carbonyl, (C1-C6 alkoxy)carbonyl, (C1-C6 alkyl)aminocarbonyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(Ci-C4.alkyl)-/ aryl, heteroaryl (Ci-Cg alkyl) carbonyl, heteroarylcarbonyl, aryl(Ci-C6 alkyl)-, (Ca-C6 alkyl) carbonyl, aryl carbonyl, Ci-Cg alkylsulfonyl, arylsulfonyl, aryl(Ci-C6 alkyDsulfonyl, heteroarylsulfonyl, heteroaryl (Ci-Cg alkyDsulfonyl, aryloxycarbonyl, and arylfCi-Ce alkoxy) carbonyl, wherein said aryl groups are substituted with 0-2 substituents selected from the

group consisting of C1-C4 alkyl, C1-C4 alkoxy, halo, CF3, and nitro;
R16e is selected from:
-C(=0)qR18ae, -C(*0)R18be, -C(=0)N(R18be)2, -C(=0)JpiS02R18ae, -C(=0)NHC(=0)R18b«, -C(=O)NHC(=O)0R1Ba*, -C(=0)NHS02NHR18ba, -S02R18ae, -S02N(RJf&be)2, and -S02NHC(=0)OR18be;
R17e is selected from:
H, C1-C6 alkyl, C3-C7 cycloalkyl, C3-C7
cycloalkyl(Ci-C4 alkyl)-, aryl, aryl(Ci-Cs alkyl)-,
and heteroaryl(Ci-C6 alkyl);
Ri8ae ±s selected from:
C1-C8 alkyl ;optionally substituted with a bond to Ln, C3-C1X cycloalkyl optionally substituted with a bond to Ln, aryl(Ci-C6 alkyl)- optionally substituted with a bond to Ln, heteroaryl {Ci-Cg
alkyl)- optionally substituted with a bond to Ln, (Ci-Ce alkyl)heteroaryl optionally substituted with a bond to Ln, biaryl(Ci-Cg alkyl) optionally substituted with a bond to Ln, heteroaryl optionally substituted with a bond to Ln, phenyl substituted with 3-4 R19e and optionally substituted with a bond to Ln, naphthyl substituted with 0-4 R19e and optionally substituted with a bond to Ln, and a bond to Ln, wherein said aryl or heteroaryl groups are optionally substituted with 0-4 R19e;
R:8be is H or R18ae;
Ri9e is selected from:
H, halogen, CF3, C02H, CN, N02, -NRlleR12e, OCF3, C1-C8 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C11 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl)-, aryl(Ci-C6 alkyl)-, C1-C6 alkoxy, C1-C4

alkoxycarbonyl, aryl, aryl-O-, aryl-S02-, heteroaryl, and heteroaryl-SO2-, wherein said aryl
and heteroaryl groups are substituted with 0-4 groups selected from hydrogen, halogen, CF3, C1-C3 alkyl, and C1-C3 alkoxy;
R20e is selected from:
hydroxy, C1-C10 alkyloxy, C3-C11 cycloalkyloxy,
aryloxy, aryl(Ci-C4 alkyDoxy,
C2-C10 alJcylcarbonylojsytCi-C^ alkyDoxy-,
C2-C10 alkoxycarbonyl oxy {C1-C2 alkyDoxy-,
C2-C10 alkoxycarbonyl(C1-C2 alkyDoxy-,
C3-C10 cycloalkyicarbonyloxy(Ca-C2 alkyDoxy-,
C3-C10 cycloalkoxycarbonyloxy(Ci-C2 alkyDoxy-,
C3-C10 cycloalkoxycarbonyl (C1-C2 alkyDoxy-,
aryloxycarbonyl{C1-C2 alkyl)oxy-,
aryloxycarbonyloxy {C1-C2 alkyl)oxy-,
arylcarbonyloxy(Ci-C2 alkyDoxy-,
C1-C5 alkoxy(Ci-Cs alkyl)carbonyloxy(Cj-C2 alkyDoxy,
(5-(C1-C5 alkyl)-1,3-dioxa-cyclopenten-2-one-
yDmethyloxy, (5-aryl-l,3-dioxa-cyclopenten-2-one-yl)methyloxy,
and (R10e) (Rlle)N-(Ca-Cio alkoxy)-;
R21e is selected from:
C1-C8 alkyl, C2-C6 alkenyl, C3-C11 cycloalkyi, (C3-C11 cycloalkyi)methyl, aryl, aryl(Ci~C4 alkyl)-, and Ci-C10 alkyl substituted with 0-2 R7e;
R22e iS selected from:
-C(»0)-Rl8be# _C(_0)N(R18be)2f -C(=0)NHS02R18*e, -
C(=0)NHC{=0)R18be, -C(=0)NHC(=0)OR18ae, and -C (»OJ NHS02NHR18be;
Ye is selected from:

-COR20V -SO3H, -PO3H, -CONHNHSO2CF3, -CONHSQ2R18ae, -CONHS02NHRl8b«, -NHCOCF3, -NHCONHS02R18ae, -NHS02R18ae, -OPO3H2, -OSO3H, -PO3H2, -S02NHCOR18ae, -S02NHC02R18ae,

me is 0-2;
ne is 0-4; pe is 0-2; re is 0-2;
with the following proviso: ne and me are chosen such
that the number of atoms connecting Rle and Ye is in the range of 8-14;
d is selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
Ln is a linking group having the formula:
( (W)h- (CR6R7)g)x- (Z)k- ((CR6*R7«)g.- (W)h. )x'/
W is independently selected at each occurrence from the
group: 0, S, NH, NHC(=0), C(=0)NH, NR8C(=0), C(=0)N R8, C(=0), C(=6)0, OC(=0), NHC(=S)NH, NHC(=0)NH, S02, S02NH, (OCH2CH2) 20-200/ {CH2CH2O) 20-200. (0CH2CH2CH2)20-
200* (CH2CH2CH2O)20-200/ and (aa)t-;
aa is independently at each occurrence an amino acid;
Z is selected from the group: aryl substituted with 0-3 R10, C3-10 cycloalkyl substituted with 0-3 R10, and a 5-10 membered heterocyclic ring system containing
286

1-4 heteroatoms .independently selected from N, S, and 0 and substituted with 0-3 R10;
R6, R6a, R7, R7a, and R8 are independently selected at • each occurrence from the group: H, =0, COOH, S03H, PO3H, C1-C5 alkyl substituted with 0-3 R10, aryl substituted with 0-3 R10, benzyl substituted with 0-3 R10, and C1-C5 alkoxy substituted with 0-3 R10, NHC(=0)R", C(=0)NHRlx, NHC(=0)NHRi:i, NHRU, R1*, and a bond to Sf;
R10 is independently selected at each occurrence from the group: a bond to Sf, COOR11, C{=0)NHRia, NHC R11 is independently selected at each occurrence from the group: H, alkyl substituted with 0-1 R12, aryl substituted with 0-1 R12, a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O and substituted with 0-1 R12, C3-10 cycloalkyl
substituted with 0-1 R12, polyalkylene glycol substituted with 0-1 R12, carbohydrate substituted with 0-1 R12, cyclodextrin substituted with 0-1 R12, amino acid substituted with 0-1 R12, polycarboxyalkyl substituted with 0-1 R12, polyazaalkyl substituted with 0-1 R12, peptide substituted with 0-1 R12, wherein the peptide is comprised of 2-10 amino acids, 3,6-O-disulfo-B-D-galactqpyranosyl, bis (phosphonomethyl) glycine, and a bond to Sf;
R12 is a bond to Sf;

k is selected from 0, l, and 2;
h is selected from 0, 1, and 2;
h' is selected from 0, 1, and 2;
g is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, s, and 10;
g' is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
t' is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10;
x is selected from 0, 1, 2, 3, 4, and 5;
x' is selected from 0, 1, 2, 3, 4, and 5;
Sf is a surfactant which is a lipid or a compound of the formula: " ;
A9 is selected from the group: OH and OR27;
A10 is OR27;
R27 is C(=0)Ci-2o alkyl;
Ea is Ci-io alkylene substituted with 1-3 R28;
R28 is independently selected at each occurrence from the group: R30, -PO3H-R30, =0, -CO2R29, ~C(=0)R29, -C(=0)N{R29}2, -CH2OR29, -OR29, -N(R29)2, C1-C5 alkyl, and C2-C4 alkenyl;
R29 is independently selected at each occurrence from the group: R3", H, Ci-Cg alkyl, phenyl, benzyl, and
trif luoiromethyl ; R30 is a bond to Ln;
and a pharmaceutical^ acceptable salt thereof.

33. A compound as claimed in claim 32, wherein the compound is of the formula:

wherein, Q |s a compound of Formula (IV):

(IV)
including stereoisomeric forms thereof, or mixtures of stereoisomeric forms thereof, or pharmaceutically acceptable salt or prodrug forms thereof wherein: Rle is selected from:


R2e and R3e are independently selected from:
H, C1-C4 alkoxy, NRlleR12e, halogen, N02, CN, Cf3, C1-C6 alkyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl), aryl(Ci-C6 alkyl)-, (Ci-Cfi alkyl)carbonyl, (C1-C6 alkoxy)carbonyl, arylcarbonyl, and aryl substituted with 0-4 R7e,
alternatively, when R2e and R3e are substituents on adjacent atoms, they can be taken together with the carbon atoms to which they are attached to form a 5-7 membered carbocyclic or 5-7 membered heterocyclic aromatic or nonaromatic ring system, said carbocyclic or heterocyclic ring being substituted with 0-2 groups selected from C1-C4 alkyl, C1-C4 alkoxy, halo, cyano, amino, CF3 and N02;
R2ae is selected from:
H, C1-C10 alkyl, C2-C6 alkenyl, C3-C11 cycloalkyl, C3-C7 cycloalkyl(C1-C4 alkyl), aryl, aryl(Ci-C4 alkyl)-, (C2-C7 alkyl)carbonyl, arylcarbonyl, (C2-C10 alkoxy)carbonyl, C3-C7 cycloalkoxycarbonyl, C7-C11 bicycloalkoxycarbonyl, aryloxycarbonyl, aryl (C1-C10 alkoxy) carbonyl, C1-C6 al kyl car bony loxy{Ci-C4 alkoxy) carbonyl, arylcarbonyloxy(Ci-C4 alkoxy)carbonyl, and C3-C7 cycloalkylcarbonyloxy(Ci-C4 alkoxy)carbonyl;
R7e is selected from:
H, hydroxy, C1-C4 alkyl, C1-C4 alkoxy, aryl, aryKCi-C4 alkyl)-, (C1-C4 alkyl)carbonyl, C02R18ae, S02Rlle, SO2NR10eRlle/ ORl°e, and NtR11*^12'?
U* is selected from:
-(CH2)n«-, -(CH2)neO(CH2)me-/ -NH(CH2)n*-, -N(Rl°«)C(=0)-, -NHC(=0)(CH2)ne-, and -C (=0)N(R*°e)-;
Ge is N or CR19e;
290

R8e is selected' from:
H, C02R18be, C R9e is selected from:
C1-C10 alkyl substituted with 0-1 R6e, Ci-Cio alkoxy substituted with 0-2 R7e, H, nitro, N(R^«)Ri2e, OC(=O)R10«, OR10e,
OC(=O)NR10eRlle, NR10eC(=O)R10e, NR10eC (=0)OR21e, NR10eC(:sO)NR10eRlle, NRl°eSO2NR10*R1:ie, NR10eSO2R21?, hydroxy, OR22e, -NtR10*^11*, -N(R16*)R17e, aryl(Co-Cfi alkyl} carbonyl, aryl (Ci-Ce alkyl), heteroaryl (Ci-Cg alkyl), CONR18aeR2°e, S02R1Bae, and SO2NR18aeR20«, providing that any of the above alkyl, cycloalkyl, aryl or heteroaryl groups may be unsubstituted or substituted independently with 1-2 R7e;
R6e is selected from:
H, C1-C10 alkyl, hydroxy, C1-C10 alkoxy, nitro, C1-C16. alkylcarbonyl, -N(R1Ie)R12e, cyano, halo, CF3, CHO, C02Rl8be, C(=0)Rl8i«, CONR*7^81**, OC(=O)R10e, QR10e, OC{=0)NRl°eRlle, NR10eC(=O)R10e, NR10eC(=O)OR21e,

NRlO«c (=0) NR10eRUe, NR10eSO2NR10eRi:Le/ NR10eSO2R21e/ S(0)peRlle, S02NR10eRlle,
aryl substituted with 0-3 groups selected from halogen, Ci-Cg alkoxy, C1-C6 alkyl, CF3/ S(0)meMe, and -NMe2, aryl(Ci-C4 alkyl)-, said aryl being substituted with 0-3 groups selected from halogen, Ci-Cg alkoxy, Cx-Cs alkyl, CF3, S(0)peMe, and -NMe2, and a 5-10 membered heterocyclic ring containing 1-3 N, 0, or S heteroatoms, wherein said heterocyclic ring may be saturated, partially saturated, or fully unsaturated, said heterocyclic ring being substituted with 0-2 R7e;
R10« J[S selected from:
H, CF3, C3-C6 alkenyl, C3-C11 cycloalkyl, aryl, (C3-C11 cycloalkyl)methyl, aryl(C1-C4 alkyl), and Cj-C10 alkyl substituted with 0-2 R6e;
Rile iS selected from:
H, hydroxy, Ci-Cs alkyl, C3-C6 alkenyl, C3-C11 cycloalkyl, (C3-C11 cycloalkyl)methyl, Cx-Ce alkoxy, benzyloxy, aryl, heteroaryl, heteroaryl(C1-C4 alkyl)-, aryl(Ci-C4 alkyl), adamantylmethyl, and C1-C10 alkyl substituted with 0-2 R4e;
R4e is selected from:
H, Cj-Cg alkyl, C3-C7 cycloalkyl, C3-C7
cycloalkyl(Cj-C^ alkyl)-, aryl, heteroaryl, aryl(Ca-
C6 alkyl)-, and heteroaryl(C^-Cg alkyl)-, wherein
said aryl or heteroaryl groups are substituted with 0-2 substituents independently selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, F, Cl, Br, CF3, and NO2,
R!2« is selected from:
H, C1-C6 alkyl, triphenylmethyl, methoxymethyl, methoxyphenyldiphenylmethyl, trimethylsilylethoxymethyl, (C1-C6 alkyl Jcarbonyl,

(Ci-Ce alkoxy)carbonyl, (C1-C6 alkyl)aminocarbonyl, C3-C6 alkenyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl(Ci-C4 alkyl)-, aryl, heteroaryl(Ci-Cg alkyl)carbonyl, heteroarylcarbonyl, aryl(Ci-C6 alkyl)-, (Ca-Cg alkyl)carbonyl, arylcarbony1, d-C6 alkylsulfonyl, arylsulfonyl, aryKCi-Ce alkyl}sulfonyl, heteroarylsulfonyl, heteroaryl(C1-C6 alkyl)sulfonyl, aryloxycarbonyl, and aryl(Ci-Ce alkoxy)carbonyl, wherein said aryl groups are substituted with 0-2 substituents selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, halo, CF3, and nitro;
Rl6e iS selected from:
-C(=0)OR18ae, -C(=0)Rlflbe, -C(=0)N(R18fae)2, -S02R18ae, and -S02N(R18be)2;
R17e is selected from:
H, C1-C6 alkyl, C3-C7 cycloalkyl, C3-C7 cycloalkyl (C1-C4 alkyl)-, aryl, aryl (C1-C6 alkyl)-, and heteroaryl(Ca-C6 alkyl); ,
Ri8ae is selected from:
C1-C8 alkyl optionally substituted with a bond to Ln, C3-C11 cycloalkyl optionally substituted with a bond to Ln, aryl(Ci-Ce alkyl)- optionally substituted with a bond to Ln, heteroaryl (Ci-Ce alkyl)- optionally substituted with a bond to Ln# (C1-C6 alkyl)heteroaryl optionally substituted with a bond to Ln. biaryKCi-Cg alkyl) optionally substituted with a bond to Ln, heteroaryl optionally substituted with a bond to Ln, phenyl substituted with 3-4 R19e and optionally substituted with a bond to Ln, naphthyl substituted with 0-4 R19* and optionally substituted with a bond to Ln, and a bond to LQ, wherein said aryl or heteroaryl groups are optionally substituted with 0-4 R19e;

Rl8be is H or RlB&e.
Rl9« is selected from:
ff, halogen, CF3, CO2H.CN, ti02, ~NRlleR12e, OCF2. C1-C8 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C11 cycloalkyl/ C3-C7 cycloalkyl{C1-C4 alkyl)-, aryl(Ci-C6 alkyl)-, Ci-Ce alkoxy, C1-C4 alkoxycarbonyl, aryl, aryl-O-, aryl-S02-, heteroaryl, and heteroaryl-SC>2-, wherein said aryl and heteroaryl groups are substituted with 0-4 groups selected from hydrogen, halogen, CF3, C1-C3 alkyl, and C1-C3 alkoxy;
R20e is selected from:
hydroxy, C1-C10 alkyloxy, C3-C11 cycloaikyloxy,
aryloxy, aryl(Ci-C4 alkyDoxy,
C2-Cio alkyl carbohyl oxy {C1-C2 alkyDoxy-,
C2-C10 alkoxycarbonyloxy(Ci-C2 alkyDoxy-,
C2-C10 alkoxycarbonyl(C1-C2 alkyDoxy-,
C3-C10 cycloalkylcarbonyloxy(ei-C2 alkyDoxy-,
C3-C10 cycloalkoxycarbonyloxy(Ci-C2' alkyDoxy-,
C3-C10 cycloalkoxycarbonyl(C1-C2 alkyDoxy-,
aryloxycarbonyl(C1-C2 alkyl)oxy-,
aryloxycarbonyloxy{C1-C2 alkyl)oxy-,
arylcarbonylpxy(Ci-C2 alkyDoxy-,
Ci-Cs alkoxy{C1-C5 alkyl)carbonyloxy(Ci-C2 alkyDoxy,
(5-(C1-C5 alkyl)-1,3-dioxa-cyclopenten-2-one-
yUmethyloxy, (5-aryl-l, 3-dioxa-cyclopenten-2-one-yl) methyloxy,
and (R10«) (Rlle) N-{C1-C10 alkoxy)-;
R21e is selected from:
Ci-C8 alkyl, C2-C6 alkenyl, C3-C11 cycloalkyl, (C3-C11 cycloalkyl) methyl, aryl, aryl (C1-C4 alkyl) -, and C1-C10 alkyl substituted with 0-2 R7e;

R22e is selected from:
-C(=0)-R18be, -C(=0}N(R18be)2. -C(*0}NHS02R18ae, -C(=0)NHC(=0)R18be, -C(=0)NHC(=0)OR18ae, and -C (=0)NHS02NHR18be;
me is 0-2;
ne is 0-4; and
pe is 0-2;
with the following proviso: ne and me are chosen such
that the number of atoms connecting R1 and -COR20e in Formula (IV) is in the range of 8-14;
w is independently selected at each occurrence from the group: O, S, NH, NHC(=0), C(=0)NH, NR8C{=0), C(=0)NR8, C(=0), C(=0)0, OC(=0), NHC(=S)NH, NHC(=0)NH, S02, S02NH,
(OCH2CH2) 20-200/ (CH2CH20) 20-200. (OCH2CH2CH2)2o-200/ (CH2CH2CH20)20-200/ and (aa)t-;
aa is independently at each occurrence an amino acid;
Z is selected from the group: aryl substituted with 0-1 Rl°, C3-10 cycloalkyl substituted with 0-1 R10, and a 5-10 membered heterocyclic ring system containing 1-4 heteroatoms independently selected from N, S, and O and substituted with 0-1 R10;
R6, R6a, R7, R7a, and R8 are independently selected at
each occurrence from the group: H, =0, COOH, SO3H, . C1-C5 alkyl substituted with 0-1 R10, aryl
substituted with 0-1 R10t benzyl substituted with 0-1 R10, and C1-C5 alkoxy substituted with 0-1 R10,
mc(=o)R11, c(eO)NHRn, mc(=o)mm^1, NHR", R", snd a bond to Sf;
k is 0 or 1; '

Sf is a surfactant which is a lipid or a compound of the
formula: A .
A9 is OR27;
A10 is OR27;
R27 is C(=0)Ci-i5 alkyl;
E1 is C1-4 alkylene substituted with 1-3 R28;
R28 is independently selected at each occurrence from the group: R30, -PO3H-R30, =0, -CO2R29, -C{=0)R29, -CH20R29, -OR29, and C1-C5 alkyl;
R29 is independently selected at each occurrence from the group: R30, H, C1-C6 alkyl, phenyl, and benzyl;
R30 is a jjond t0 Ln.
and a pharmaceutical^ acceptable salt thereof.
34. An ultrasound contrast agent composition, comprising:
(a) a compound as claimed in claim 32, comprising: an quinolone that binds to the integrin avp3, a surfactant and a linking group between the quinolone and the surfactant;
(b) a parenterally acceptable carrier; and,
(c) an echogenic gas.
35. An ultrasound contrast agent composition as claimed in
claim 34, further comprising: 1,2-dipalmitoyl-sn-glycero-3-
phosphotidic acid, 1,2-dipalmitoyl-sn-glycero-3-
phosphatidylcholine, and N-(methoxypolyethylene glycol
5000 carbamoyl)-1,2-dipalmitoyl-sn-glycero-3-
phosphatidylethanolamine.

36. An ultrasound contrast agent composition as claimed in claim 35, wherein the echogenic gas is a C2-5 perfluorocarbon.
37. A therapeutic radiopharmaceutical composition, comprising:

(a) a therapeutic radiopharmaceutical as claimed in claim 18; and,
(b) a parenterally acceptable carrier.
38. A diagnostic pharmaceutical composition, comprising:
(a) a diagnostic radiopharmaceutical, a MRI contrast agent, or a X-ray contrast agent as claimed in claim 10; and,
(b) a parenterally acceptable carrier.
Dated this 22nd day of May, 2001.
OF REMFRY & SAGAR ATTORNEY FOR THE APPLICANTS

Documents:

in-pct-2001-00576-mum-cancelled pages(8-8-2007).pdf

in-pct-2001-00576-mum-claims(granted)-(8-8-2007).doc

in-pct-2001-00576-mum-claims(granted)-(8-8-2007).pdf

in-pct-2001-00576-mum-correspondence(9-8-2007).pdf

in-pct-2001-00576-mum-correspondence(ipo)-(30-1-2008).pdf

in-pct-2001-00576-mum-form 1(19-4-2001).pdf

in-pct-2001-00576-mum-form 1(22-5-2001).pdf

in-pct-2001-00576-mum-form 1a(5-3-2007).pdf

in-pct-2001-00576-mum-form 2(granted)-(8-8-2007).doc

in-pct-2001-00576-mum-form 2(granted)-(8-8-2007).pdf

in-pct-2001-00576-mum-form 3(19-7-2007).pdf

in-pct-2001-00576-mum-form 3(22-5-2001).pdf

in-pct-2001-00576-mum-form 5(5-3-2007).pdf

in-pct-2001-00576-mum-form 6(26-4-2002).pdf

in-pct-2001-00576-mum-form-pct-isa-210(22-5-2001).pdf

in-pct-2001-00576-mum-petition under rule 137(27-7-2007).pdf

in-pct-2001-00576-mum-power of authority(5-3-2007).pdf


Patent Number 214104
Indian Patent Application Number IN/PCT/2001/00576/MUM
PG Journal Number 13/2008
Publication Date 28-Mar-2008
Grant Date 30-Jan-2008
Date of Filing 22-May-2001
Name of Patentee BRISTOL-MYERS SQUIBB PHARMA COMPANY
Applicant Address P.O.BOX 4000, PRINCETON, NEW JERSEY 08543-4000,
Inventors:
# Inventor's Name Inventor's Address
1 THOMAS DAVID HARRIS 56 ZION HILL ROAD, SALEM, NEW HAMSHIRE 03079,
2 MILIND RAJOPADHYE 21 HONEYSUCKLE ROAD, WESTFORD, MASSACHUSETTS 01886,
PCT International Classification Number A61K 51/04
PCT International Application Number PCT/US99/30315
PCT International Filing date 1999-12-17
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/112,732 1998-12-18 U.S.A.