Title of Invention

PHOSPHORAMIDATE ALKYLATOR PRODRUGS

Abstract Phosphoramidate alkylator prodrugs can be used to treat cancer when administered alone or in combination with one or more anti-neoplastic agents.
Full Text

PHOSPHORAMIDATE ALKYLATOR PRODRUGS
CROSS-REFERENCES TO RELATED APPLICATIONS
[0001] This application claims benefit of U.S. provisional patent application No.
60/695,755 filed 29 Jun 2005, which is incorporated herein by reference.
BACKGROUND OF THE INVENTION
Field of Invention
[0002] The present invention provides compositions and methods for treating cancer and
other hyperproliferative disease conditions with phosphoramidate alkylator prodrugs. The
present invention generally relates to the fields of chemistry, biology, molecular biology,
pharmacology, and medicine.
Description of Related Art
[0003] Alkylating agents ("alkylators" or "mustards") used in cancer chemotherapy
encompass a diverse group of chemicals that have the ability to alkylate biologically vital
macromolecules such as DNA under physiological conditions (see Hardman et al, The
Pharmacological Basis of Therapeutics, 2001,1389-1399, McGraw-Hill, New York, USA).
DNA alkylation is postulated to be an important mechanism in the antitumor activity of
alkylators. The chemotherapeutic alkylators act as strong electrophiles, for example, through
the formation of neighboring-heteroatom-stabilized onium intermediates such as an aziridine
or an aziridinium cation.
[0004] Phosphoramidate based alkylators used in cancer therapy, such as
Cyclophosphamide and Ifosfamide, are an important subclass of chemotherapeutic alkylators.
Cyclophosphamide and Ifosfamide are each activated in the liver and the active alkylator
released alkylates nucleopbilic moieties such as the DNA within the tumor cells to act as a
chemotherapeutic agent. If the active alkylators are released away from the tumor, DNA and
other nucleophilic moieties such as the phosphate, amino, sulfhydryl, hydroxyl, carboxyl and
imidazo groups of biomolecules of healthy non-cancerous cells, can get alkylated. Such
alkylation of healthy cells canand result in unwanted toxic events in patients (see Hardman et
al., supra).


[0005] There remains a need for new phosphoramidate based alkylators that can be used to
treat cancer or other hyperproliferative disease conditions, preferably compounds less toxic to
normal cells. The present invention meets these needs and provides novel phosphoramidate
alkylator prodrugs as well as methods of therapy employing them, as summarized in the
following section.
BRIEF SUMMARY OF THE INVENTION
[0006] In one aspect the present invention provides compounds which are hypoxia
activated phosphoramidate alkylator prodrugs and methods of their synthesis. The
phosphoramidate alkylator prodrugs of the present invention can have the formula Alk-X
wherein Alk is a phosphoramidate alkylator, T is L-Z3 wherein L is a linker Z3 is a
bioreductive group.
[0007] In one aspect, the present invention provides phosphoramidate alkylator prodrugs of
formula (I):

wherein
[0008] Y1 is O, S, NRe, or NSO2R6 wherein each R6 is independently C1-C6 alkyl, C1-C6
heteroalkyl, aryl, orheteroaryl;
[0009] Y2 is O, S, NRs, NCORe, or NSO2R6;
[0010] each of R1-R5 independently is hydrogen, hydroxy!, amino, C1-C6 alkyl, C1-C6
heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, C1-C6 alkoxy, C1-C6 alkylamino, C1-C6
dialkylamino, aryl, heteroaryl, C1-C6 acyl, C1-C6heteroacyl, aroyl, or heteroaroyl; or together

any two of R1-R5 form a C3-C10 heterocycle; or each of R1- R5 independently is a Trigger T
wherein T is L-Z3
[0011] L is selected from
-[C(Z1)2-Y3]v-[C(=O)-O]q-[C(Z1)2-Z2-Y4]u-[C(Z1)2]z-[-C(Z1)=C(Z1)]g-Z3 and
-[C(Z1)2-Y3]v-(S(=O)2)q-[C(Z1)2-Z2-Y4]u-[C(Z1)2]2-[C(Z1)=C(Z1)]g-Z3; wherein each
z, v, q, u, and g independently is 0 or 1;
[0012] Y3 is S, O, or NR7 wherein each R7 is independently hydrogen, hydroxyl, C1-C6
alkyl, C1-C6 heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, C1-C6 alkoxy, C1-C6 alkylamino, C1-
C6 dialkylamino, aryl, heteroaryl, C1-C6 acyl, C1-C6heteroacyl, aroyl, or heteroaroyl;
[0013] Y4 is O, S, or -NR7-C(=O)-0-;
[0014] each Z\ independently is hydrogen, halogen, C1-C6 alkyl, C1-C6heteroalkyl, aryl,
heteroaryl, C3-C8 cycloalkyl, heterocyclyl, C1-C6 acyl, C1-C6heteroacyl, aroyl, or
heteroaroyl;
[0015] Z2 is C1-C6 alkylene, C1-C6 heteroalkylene,

wherein each X1 is independently N or CR8, each R8 is independently hydrogen, halogen,
nitro, cyano, CO2H, C1-C6 alkyl, C1-C6heteroalkyl, C1-C6 cycloalkyl, C1-C6 alkoxy, C1-C6
alkylamino, C1-C6 dialkylamino, aryl, CON(R7)2, C1-C6 acyl, C1-C6heteroacyl, aroyl, or
heteroaroyl;
[0016] X2 is NR7,S,or O;and
[0017] Z3 is selected from the group consisting of:


[0018] with the proviso that in formula (I):
[0019] (i) at least two of R1-R5 are selected from the group consisting of 2-haloalkyl, 2-
alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl;
[0020] (ii) at least one of R1-R5 is selected from the group consisting of 2-haloalkyl, 2-C1
C6 alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl; and at least one of NR2R3 and NR4R5 is "~KI; or
[0021] (iii) NR2R3 and NR4R5 both together are~"N:3; and
[0022] an individual isomer or a racemic or non-racemic mixture of isomers, bioisosteres,
pharmacophores, a pharmaceutically acceptable salt, solvate, hydrate, or a prodrug thereof.
[0023] In one embodiment, Z3 is a bioreductive group that can accept one or more electrons
in an oxidation-reduction reaction.
[0024] In a related embodiment, the present invention provides
phosphoramidate alkylator prodrugs having IC50 or GI50, in cells under hypoxia, of 50 uM to
0.01 nm. In a related embodiment, the present invention provides phosphoramidate alkylator

prodrugs having hypoxic cytotoxicity which are up to a million fold, up to 10,000 fold, and
up to 1000 fold less toxic in corresponding normoxic cells. In a related embodiment, the
cellular cytotoxicity is measured by antiproliferation assays and using the relative IC50 value
of a compound in hypoxic and normoxic cells. In a related embodiment, the cellular
cytotoxicity is measured by clonogenic assays and using the relative C10, C50, or C90 values of
the compounds in hypoxic and normoxic cells.
[0025] In another related embodiment, the present invention provides phosphoramidate
alkylator prodrugs having IC50 values, in cells under hypoxia, of 50 uM to 0.01 nM. In
another related embodiment, the present invention provides phosphoramidate alkylator
prodrugs which are up to 5000 fold less toxic in corresponding normoxic cells as measured
by the relative IC50 values in hypoxic and normoxic cells. In another related embodiment, the
present invention provides phosphoramidate alkylator prodrugs having an IC50 in cells in
hypoxia of 50 uM to 0.01 nM and which is up to 1000 fold less toxic in corresponding
normoxic cells as measured by the relative IC50 values in hypoxic and normoxic cells.
[0026] In a related embodiment, phosphoramidate alkylator prodrug of the present
invention has a hypoxic cytotoxicity of 0.1 nM to 50 uM and a hypoxia cytotoxicity ratio,
HCR, measured by the ratio of normoxic and hypoxic cytotoxicities, and defined in greater
detail further later in the application, of 10 to 100,000. In a related embodiment, the
phosphoramidate alkylator prodrug of the present invention has a hypoxic cytotoxicity of 0.1
nM to 50 uM and a HCR of 25 to 100,000. In another related embodiment, phosphoramidate
alkylator prodrug of the present invention has a hypoxic cytotoxicity of 0.1 nM to 5 uM and a
HCR of 50 to 10,000.
[0027] In one aspect the present invention provides novel phosphoramidate alkylators for
treatement of cancer and other hyperprolifcrative diseases,
[0028] In one aspect, the present invention provides a pharmaceutical formulation
comprising the phosphoramidate alkylator prodrugs of the invention ancTa pliarmaceutically
acceptable excipient, carrier, or diluent.
[0029] In one aspect, the present invention provides a method of treating cancer and other
hyperproliferative diseases comprising administering a therapeutically effective amount of a
phosphoramidate alkylator prodrug of the invention or one that is known, to a patient in need
of such therapy. In one embodiment, the cancer treated is resistant to first line, second line,
or third line therapy, or is a relapsed cancer. In another embodiment, the cancer treated is a

metastatic cancer. In another embodiment, the phosphoramidate alkylator prodrug of the
invention, or one that is known, is administered in combination with at least another anti-
cancer agent.
BRIEF DESCRIPTION OF THE DRAWINGS
[0030] Figure 1 demonstrates the effect of Compound 25 (50 mg/kg) on tumor growth in
the H460 xenograft mouse model.
[0031] Figure 2 demonstrates the effect of Compound 25 (100 mg/kg ) on tumor growth in
the H460 xenograft mouse model.
[0032] Figure 3 demonstrates the effect of Compound 25 (150 mg/kg ) dosed in
combination with CDDP on tumor growth in the H460 xenograft mouse model.
[0033] Figure 4 demonstrates the effect of Compound 25 dosed in combination with CDDP
on tumor growth in the H460 xenograft mouse model.
[0034] Figures 5, 6 and 7 demonstrate the effect of Compound 25 in combination with
Gemcitabine on tumor growth in the H460 xenograft mouse model.
DETAILED DESCRIPTION OF THE INVENTION
[0035] The detailed description of the different aspects and embodiments of the present
invention is organized as follows: Section I provides useful definitions; Section II describes
the compounds of the invention and methods for making them; Section III describes methods
of treatment, therapies, administrations, and formulations, employing the compounds of the
invention alone or in combination; and Section IV provides examples of synthetic methods
and biological assays for the compounds of the invention. This detailed description is
organized into sections only for the convenience of the reader, and disclosure found in any
section is applicable to any aspect of the invention.
I. Definitions
[0036] The following definitions are provided to assist the reader. Unless otherwise
defined, all terms of art, notations and other scientific or medical terms or terminology used
herein are intended to have the meanings commonly understood by those of skill in the
chemical and medical arts. In some cases, terms with commonly understood meanings are

defined herein for clarity and/or for ready reference, and the inclusion of such definitions
herein should not necessarily be construed to represent a substantial difference over the
definition of the term as generally understood in the art.
[0037] As used herein, "a" or "an" means "at least one" or "one or more."
[0038] "Alkyl' means a linear saturated monovalent hydrocarbon radical or a branched
saturated monovalent hydrocarbon radical having the number of carbon atoms indicated in
the prefix. As used in this disclosure, the prefixes (C1-Cqq), C1-qq, or C1-Cqq, wherein qq is
an integer from 2-20, have the same meaning. For example, (C1-Cg) alkyl, C1-g alkyl, or C\-
Cg alkyl includes methyl, ethyl, n-propyL 2-propyl, n-butyl, 2-butyl, tert-butyl, pentyl, and
the like. For each of the definitions herein {e.g., alkyl, alkenyl, alkoxy, araalkyloxy), when a
prefix is not included to indicate the number of main chain carbon atoms in an alkyl portion,
the radical or portion thereof will have six or fewer main chain carbon atoms. (C1-C6) alkyl
can be further optionally be substituted with substituents, including for example, deuterium
("D"), hydroxyl, amino, mono or di(C1-C6) alkyl amino, halo, C2-C6 alkenyl ether, cyano^
nitro, ethenyl, ethynyl, C1-C6 alkoxy, C1-C6 alkylthio, -COOH, -CONH2, mono- or di(C1-C6)
alkylcarbox-amido, -SO2NH2, -OSO2-(C1-C6) alkyl, mono or di(C1-C6) alkylsulfonamido,
aryl, heteroaryl, alkylsulfonyloxy, heteroalkylsulfonyloxy, arylsulfonyloxy or
heteroarylsulfonyloxy.
[0039] "Alkenyl" means a linear monovalent hydrocarbon radical or a branched
monovalent hydrocarbon radical having the number of carbon atoms indicated in the prefix
and containing at least one double bond, but no more than three double bonds. For example,
(C2-C6) alkenyl includes, ethenyl, propenyl, 1,3-butadienyl and the like. Alkenyl can be
further optionally be substituted with substituents, including for example, deuterium ("D"),
hydroxyl, amino, mono or di(C1-C6) alkyl amino, halo, C2-C6 alkenyl ether, cyano, nitro,
ethenyl, ethynyl, C1-C6 alkoxy, C1-C6 alkylthio, -COOH, -CONH2, mono- or di(C1-C6) alkyl-
carboxamido, -SO2NH2, -OSO2-(C1-C6) alkyl, mono or di(C1-C6) alkylsulfonamido, aryl,
heteroaryl, alkyl or heteroalkylsulfonyloxy, and aryl or heteroarylsulfonyloxy.
[0040] " Alkylator" means a reactive moiety capable of forming a covalent alkyl linkage to
macromolecules via an electrophillic reaction with a nucleophile on the macromolecule.
"Phosphoramidate alkylator" means an alkylator for which an aziridine or aziridinium
electrophile is present or generated by intramolecular cyclization.

[0041] "Alkylene" means a linear saturated divalent hydrocarbon radical having from one
to twelve carbon atoms or a branched saturated divalent hydrocarbon radical having from one
to twelve carbon atoms optionally substituted with substituents including for example,
deuterium ("D"), hydroxyl, amino, mono or di(C1-C6)alkyI amino, halo, C2-C6 alkenyl ether,
cyano, nitro, ethenyl, ethynyl, C1-C6 alkoxy, C1-C6 alkylthio, -COOH, -CONH2, mono- or di-
(C1-C6)alkyl-carboxamido, -SO2NH2, -OSO2-(C1-C6) alkyl, mono or di(C1-C6)
alkylsulfonarnido, aryl, heteroaryl, alkyl or heteroalkylsulfonyloxy, and aryl or
heteroarylsulfonyloxy. For example alkylene includes methylene, ethylene, propylene, 2-
methyl-propylene, pentylene, hexylene, and the like.
[0042] "Heteroalkylene" has essentially the meaning given above foran alkylene except that
one or more heteroatoms (i.e. oxygen, sulfur, nitrogen and/or phosphorous) may be present in
the alkylene biradical. For example, heteroalkylene includes, -CH2OCH2O-,-
CH2CH2OCH2CH2-, -CH2CH2N(CH3)CH2CH2-, -CH2CH2SCH2CH2-, and the like.
[0043] "AryV refers to a monovalent monocyclic or bicyclic aromatic hydrocarbon radical
of 6 to 10 ring atoms which is substituted independently with one to eight substituents,
preferably one, two, three, four ot five substituents selected from deuterium ("D"), alkyl,
cycloalkyl, cycloalkylalkyl, halo, nitro, cyano, hydroxyl, alkoxy, amino, acylamino, mono-
alkylamino, di-alkylamino, haloalkyl, haloalkoxy, heteroalkyl, COR (where R is hydrogen,
alkyl, cycloalkyl, cycloalkyl-alkyl, phenyl or phenylalkyl), -(CR'R")n-COOR (where n is an
integer from 0 to 5, R' and R" are independently hydrogen or alkyl, and R is hydrogen, alkyl,
cycloalkyl, cycloalkylalkyl, phenyl or phenylalkyl) or -(CR'R")n-CONRxRy (where n is an
integer from 0 to 5, R' and R" are independently hydrogen or alkyl, and Rx and Ry are
independently selected from hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, phenyl or
PllCIlYlalKyI)' & one embodiment, Rx and Ry together is cycloalkyl or heterocyclyl. More
specifically the term aryl includes, but is not limited to, phenyl, biphenyl, 1-naphthyl, and 2-
naphthyl, and the substituted forms thereof.
[0044] "Cycloalkyl" refers to a monovalent cyclic hydrocarbon radical of three to seven
ring carbons. The cycloalkyl group can have one or more double bonds and can also be
optionally substituted independently with one, two, three or four substituents selected from
alkyl, optionally substituted phenyl, or -C(0)Rz (where RZ is hydrogen, alkyl, haloalkyl,
amino, mono-alkylamino, di-alkylamino, hydroxyl, alkoxy, or optionally substituted phenyl).
More specifically, the term cycloalkyl includes, for example, cyclopropyl, cyclohexyl,

cyclohexenyi, phenylcyclohexyl, 4-carboxycyclohexyl, 2-carboxamidocyclohexenyl, 2-
dimethylaminocarbonyl-cyclohexyl, and the like.
[0045] "Heteroalkyl" means an alkyl radical as defined herein with one, two or three
substituents independently selected from cyano, -ORw, -NRxRy, and -S(0)pRz (where p is an
integer from 0 to 2 ), with the understanding that the point of attachment of the heteroalkyl
radical is through a carbon atom of the heteroalkyl radical. Rw is hydrogen, alkyl, cycloalkyl,
cycloalkyl-alkyl, aryl, aralkyl, alkoxycarbonyl, aryloxycarbonyl, carboxamido, or mono- or
di-alkylcarbamoyl. R" is hydrogen, alkyl, cycloalkyl, cycloalkyl-alkyl, aryl or araalkyl. Ry is
hydrogen, alkyl, cycloalkyl, cycloalkyl-alkyl, aryl, araalkyl, alkoxycarbonyl,
aryloxycarbonyl, carboxamido, mono- or di-alkylcarbamoyl or alkylsulfonyl. Rz is hydrogen
(provided that n is 0), alkyl, cycloalkyl, cycloalkyl-alkyl, aryl, araalkyl, amino, mono-
dkylamino, di-alkylamino, or hydroxyalkyl. Representative examples include, for example,
2-hydroxyethyl, 2,3-dihydroxypropyl, 2-methoxyethyl, benzyloxymethyl, 2-cyanoethyl, and
2-methylsulfonyl-ethyl. For each of the above, Rw, Rx, Ry, and Rz can be further substituted
by amino, halo, fluoro, alkylamino, di-alkylamino, OH or alkoxy. 'Additionally, the prefix
indicating the number of carbon atoms (e.g., C1-C10) refers to the total number of carbon
atoms in the portion of the heteroalkyl group exclusive of the cyano, -ORw, -NRxRy, or
-S(0)pRz portions.
[0046J In one embodiment, Rx and Ry together is cycloalkyl or heterocyclyl.
[0047] "Heteroaryl" means a monovalent monocyclic, bicyclic or tricyclic radical of 5 to 12
ring atoms having at least one aromatic ring containing one, two, or three ring heteroatoms
selected from N, O, or S, the remaining ring atoms being C, with the understanding that the
attachment point of the heteroaryl radical will be on an aromatic ring. The heteroaryl ring is
GptiuAally substituted independently with w 10 *$& a*^ preferably one, two, three
or four substituents, selected from alkyl, cycloalkyl, cycloalkyl-alkyl, halo, nitro, cyano,
hydroxyl, alkoxy, amino, acylarnino, mono-alkylamino, di-alkylamino, haloalkyl,
haloalkoxy, heteroalkyl, -COR (where R is hydrogen, alkyl, phenyl or phenylalkyl, -
(CR'R")n-COOR (where n is an integer from 0 to 5, R' and R" are independently hydrogen or
alkyl, and R is hydrogen, alkyl, cycloalkyl, cycloalkyl-alkyl, phenyl or phenylalkyl), or -
(CR'R")n-CONRxRy (where n is an integer from 0 to 5, R' and R" are independently
hydrogen or alkyl, and Rx and Ry are, independently of each other, hydrogen, alkyl,
cycloalkyl, cycloalkyl-alkyl, phenyl or phenylalkyl). In one embodiment, Rx and Ry together

is cycloalkyl or heterocyclyl. More specifically the term heteroaryl includes, but is not
limited to, pyridyl, furanyl, thienyl, thiazolyl, isothiazolyl, triazolyl, imidazolyl, isoxazolyl,
pyrrolyl, pyrazolyl, pyridazinyl, pyrimidinyl, benzofuranyl, tetrahydrobenzofuranyl,
isobenzofuranyl, benzothiazolyl, benzoisothiazolyl, benzotriazolyl, indolyl, isoindolyl,
benzoxazolyl, quinolyl, tetrahydroquinolinyl, isoquinolyl, benzimidazolyl, benzisoxazolyl or
benzothienyl, indazolyl, pyrrolopyrymidinyl, indolizinyl, pyrazolopyridinyl,
triazolopyridinyl, pyrazolopyrimidinyl, triazolopyrimidinyl, pyrrolotriazinyl,
pyrazolotriazinyl, triazolotriazinyl, pyrazolotetrazinyl, hexaaza-indenly, andheptaaza-indenyl
and 1he derivatives thereof. Unless indicated otherwise, the arrangement of thehetero atoms
within the ring can be any arrangement allowed by the bonding characteristics of the
constituent ring atoms.
[0048] "Heterocyclyl" or "cycloheteroalkyl" means a saturated or unsaturated non-aromatic
cyclic radical of 3 to 8 ring atoms in which one to four ring atoms are heteroatoms selected
from O, NR (where R is hydrogen, alkyl, cycloalkyl, cycloalkyialkyl, phenyl or phenylalkyl),
P(=O)ORw, or S(0)p (where p is an integer from 0 to 2), the remaining ring atoms being C,
where one or two C atoms can optionally be replaced by a carbonyl group. The heterocyclyl
ring can be optionally substituted independently with one, two, three or four substituents
selected from alkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, cycloalkyl, cycloalkyialkyl,
halo, nitro, cyano, hydroxyl, alkoxy, amino, mono-alkylarnino, di-alkylamino, haloalkyl,
haloalkoxy, -COR (where R is hydrogen, alkyl, cycloalkyl, cycloalkyialkyl, phenyl or
phenylalkyl), -(CR'R")n-COOR (n is an integer from 0 to 5, R' and R" are independently
hydrogen or alkyl, and R is hydrogen, alkyl, cycloalkyl, cycloalkyialkyl, phenyl or
phenylalkyl), or -(CR'R")„-CONRxRy (where n is an integer from 0 to 5, R' and R" are
independently hydrogen or alkyl, Rx and Ry are, independently of each other, hydrogen, alkyl,
cycloalkyl, cycloalkyialkyl, phenyl or phenylalkyl). More specifically the term MerOCyClyl
includes, but is not limited to, pyridyl, tetrahydropyranyl, N-methyIpiperidin-3-yl, N-
methylpyrrolidin-3-yl, 2-pyrrolidon-l-yl, furyl, quinolyl, thienyl, benzothienyl, pyrrolidinyl,
piperidinyl, morpholinyl, pyrrolidinyl, tetrahydrofuranyl, tefrahydrothiofuranyl, 1,1-dioxo-
bexahydro-1 A6-thiopyran-4-yl, tetrahydroimidazo [4,5-c] pyridinyl, imidazolinyl,
piperazinyl, and piperidin-2-only and the derivatives thereof. The prefix indicating the
number of carbon atoms {e.g., C3-C10) refers to the total number of carbon atoms in the
portion of the cycloheteroalkyl or heterocyclyl group exclusive of the number of heteroatoms.
[0049] "C1-C6 Acyl" means -CO-(C1-C6 alkyl), wherein the term alkyl is as defined above.

[0050] "C1-C6 Heteroacyl" means -CO-(C1-C6 heteroalkyl), wherein the term heteroalkyl
is as defined above.
{0051] "Aroyl" means -CO-aryl, wherein the term aryl is as defined above.
[0052] "Heteroaroyl" means -CO-heteroayl, wherein the term heteroaryl is as defined
above.
[0053] "Rsuisulfonyloxy" means Rsui-S(=O)2-0- including alkylsulfonyloxy,
heteroakyisulfonyloxy, cycloalkylsulfonyloxy, heterocyclylsuifonyloxy, arylsulfonyloxy and
heteroarylsulfonyloxy wherein Rsui is alkyl, heteroakyl, cycloalkyl, heterocyclyl, aryl and
heteroaryl respectively, and wherein alkyl, heteroakyl, cycloalkyl, heterocyclyl, aryl and
heteroaryl are defined above. Examples of alkylsulfonyloxy include Me-S(=O)2-0-, Et-
S(=0>2-0-, CF3-S(=O)2-0- and the like, and examples of arylsulfonyloxy include

Alkylsulfonyloxy, heteroakyisulfonyloxy, cycloalkylsulfonyloxy, heterocyclylsuifonyloxy,
arylsulfonyloxy, and heteroarylsulfonyloxy groups can be leaving groups in phosphoramidate
alkylators and can be replaced in a cell by nucleic acids such as DNA or RNA, and
imidazoles, carboxylates, or thiols of proteins, causing alkylation and cell death. The rate of
reaction of various RsuiSulfonyloxy groups with nucleic acids, proteins or water can be
modulated depending on for example the electron withdrawing nature and the steric bulk of
the Rsui moiety and can provide phosphoramidate alkylators and prodrugs thereof which are
more toxic to tumors in general and hypoxic zones of tumor in particular over healthy cells.
[0054] "Substituents" mean, along with substituents particularly described in the definition
of each of the groups above, those selected from: deuterieum, -halogen, -OR, -NR R , -SR ,
-SiR'R "R"', -0C(0)R', -C(0)R', -CO2R', -CONR'R", -0C(0)NR'R", -NR"C(0)R\ -NR'-
C(0)m"R"', -NR"C(0)2R'; -NH-C(NH2)=NH, -NR'C(NH2)=NH, -NH-C(NH2)=NR\ -
S(0)R', -S(0)2R\ -S(O) 2NR'R", -NR'S(0)2R", -CN and -N02, -R\ -N3, perfmoro(C1-
C4) alkoxy, and perfluoro(C1-C4) alkyl, in a number ranging from zero to the total number
of open valences on the radical; and where R\ R" and R'" are independently selected from
hydrogen, C]_g alkyl, C3.6 cycloalkyl, C2.g alkenyl, C2.g alkynyl, unsubstituted aryl and
heteroaryl, (unsubstituted aryl)-C1-4 alkyl, and unsubstituted aryloxy-C1-4 alkyl, aryl

substituted with 1-3 halogens, unsubstituted C1-g alkyl, C1-g alkoxy or C1-g thioalkoxy
groups, or unsubstituted aryl-C1-4 alkyl groups. When R' and R" are attached to the same
nitrogen atom, they can be combined with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-
membered ring. For example, -NR'R" is meant to include 1-pyrrolidinyl and 4-morpholinyl.
Other suitable substituents include each of the above aryl substituents attached to a ring atom
by an alkylene tether of from 1-4 carbon atoms. Two of the substituents on adjacent atoms of
the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -T2-
C(0)-(CH2)q-U3-, wherein T2 and U3 are independently -NH-, -0-, -CH2- or a single bond,
and q is an integer of from 0 to 2. Alternatively, two of the substituents on adjacent atoms of
the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-
(CH2)r-B-, wherein A and B are independently -CH2-, -O-, -NH-, -S-, -S(0>-, -S(0>2-, -S(O)
2NR'- or a single bond, and r is an integer of from 1 to 3. One of the single bonds of the new
ring so formed may optionally be replaced with a double bond. Alternatively, two of the
substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with
a substituent of the formula -(CH2)s-X5-(CH2)t-3 where s and t are independently integers of
from 0 to 3, and Xs is -O-, -NR'-, -S-, -S(O)-, -S(0>2-, or -S(0)2NR'-. The substituent R' in
-NR'- and -S(0)2NR'- is selected from hydrogen or unsubstituted C1-6 alkyl.
[0055] Certain compounds of the present invention possess asymmetric carbon atoms
(optical centers) or double bonds; the racemates, diastereomers, geometric isomers,
regioisomers and individual isomers (e.g., separate enantiomers) are all intended to be
encompassed within the scope of the present invention. The compounds of the present
invention may also contain unnatural proportions of atomic isotopes at one or more of the
atoms that constitute such compounds. For example, the compounds may be radiolabeled
with radioactive isotopes, such as for example tritium (H), iodine-125 ( I) or carbon-14
(}4C). All isotopic variations of the compounds of the present invention, whether radioactive
or not, are intended to be encompassed within the scope of the present invention.
[0056] The term "pharmaceutically acceptable salts" is meant to include salts of the active
compounds which are prepared with relatively nontoxic acids or bases, depending on the
particular substituents found on the compounds described herein. When compounds of the
present invention contain relatively acidic functionalities, base addition salts can be obtained
by contacting the neutral form of such compounds with a sufficient amount of the desired
base, either neat or in a suitable inert solvent. Examples of salts derived from

pharmaceutically-acceptable inorganic bases include aluminum, ammonium, calcium, copper,
ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, zinc and the
like. Salts derived from pharmaceutically-acceptable organic bases include salts of primary,
secondary and tertiary amines, including substituted amines, cyclic amines, naturally-
occuring amines and the like, such as arginine, betaine, caffeine, choline, N,N'-
dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol,
ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine,
glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine,
piperazine, piperadine, polyamine resins, procaine, purines, theobromine, triethylamine,
trimethylamine, tripropylamine, tromethamine and the like. When compounds of the present
invention contain relatively basic functionalities, acid addition salts can be obtained by
contacting the neutral form of such compounds with a sufficient amount of the desired acid,
either neat or in a suitable inert solvent Examples of pharmaceutically acceptable acid
addition salts include those derived from inorganic acids like hydrochloric, hydrobromic,
nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphpric,
dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and
the like, as well as the salts derived from relatively nontoxic organic acids like acetic,
propionic, isobutyric, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic,
benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like. Also included
are salts of amino acids such as arginate and the like, and salts of organic acids like
glucuronic or galactunoric acids and the like (see, e.g., Berge, S.M., et al, "Pharmaceutical
Salts", Journal of Pharmaceutical Science, 1977,66,1-19). Certain specific compounds of
the present invention contain both basic and acidic functionalities that allow the compounds
to be converted into either base or acid addition salts.
[0057] The neutral forms of ike compounds may be regenerated by contacting the salt with
a base or acid and isolating the parent compound in the conventional manner. The parent
form of the compound differs from the various salt forms in certain physical properties, such
as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the
compound for the purposes of the present invention.
[0058] Certain compounds of the present invention can exist in unsolvated forms as well as
solvated forms, including hydrated forms. In general, the solvated forms are equivalent to
unsolvated forms and are intended to be encompassed within the scope of the present
invention. Certain compounds of the present invention may exist in multiple crystalline or

amorphous forms. In general, all physical forms are equivalent for the uses contemplated by
the present invention and are intended to be within the scope of the present invention.
[0059] As used herein, a "glucose analog" includes mono-, di- and tri-saccharides. The
glucose analog includes sacchrides comprising glucosamine, N-acetyl-glucosamine; fructose;
mannose and mannose derivatives; glucose and glucose derivatives, including but not limited
to 2-deoxyglucose (2-DG), N-acetyl-2-amino-2-deoxyglucose, 3-amino-3-deoxy-gmcose, 2-
amino-2-deoxy-glucose; and galactose and galactose derivatives including but not limited to
D-2-deoxy-D-galactose, D-4-amino-4-deoxy-galactose and D-2-arnino-2-deoxy-galactose.
Thus, the glucose analog can differ from glucose or a derivative such as DG and glucosamine
in that it is an epimer thereof. In addition, the glucose analog can be a fluorinated derivative
of any of the foregoing compounds. Moreover, the oxygen in the ring of any of the foregoing
compounds can be substituted with an isostere selected from the group consisting of S,
sulfone, and the like. For example, glucose analog can be 5-tbio-D-gIucose or a derivative
thereof.
[0060] A wavy line " JVV " means the point of attachment of one group or moiety to

thio group is the point of attachment to another group or moiety.
[0061] The terms CO, C(O), C(=O), -CO- are used interchangeably herein. The terms CO2
and COO are used interchangeably herein.. The terms; SO2, S(0)2 are used interchangeably
herein. The terms SO and S(=O) are used interchangeably herein. The terms PO and P(=O)
are used interchangeably herein.
[0062] As used herein, a "bioisostere" of a chemical moiety Such SS mOleCUlC, pup, W
atom means another chemical moiety having similar size and spatial disposition of electron
pair or pairs. BioisOSteres and bioisosterism are well-known tools for predicting the
biological activity of compounds, based upon the premise that compounds with similar size,
shape, and electron density can have similar biological activity. Known bioisosteric
replacements include, for example, the interchangeability of-F, -OH, -NH2, -CI, and -CH3;
the mterchangeability of-Br and -1-C3H7; the interchangeability of-I and -*-C4H?; the
interchangeability of-0-, -S-, -NH-, -CH2, and -Se-; the interchangeability of-N=, -CH=, and
-P= (in cyclic or noncyclic moieties); the interchangeability of phenyl and pyridyl groups; the

interchangeability of -C=C- and -S- (for example, benzene and thiophene); the
interchangeability of an aromatic nitrogen (^^(R^-Ra,.) for an unsaturated carbon (Rar-
C(=Rar)-Rar); and the interchangeability of-CO-, -SO-, and -SO2-. These examples are not
limiting on the range of bioisosteric equivalents and one of skill in the art will be able to
identify other bioisosteric replacements known in the art. See, for example, Patani et ah,
1996, Chem. Rev. 96:3147-16; and Burger, 1991, A. Prog. Drug Res. 37:287-371.
[0063] A reasonable quantitative prediction of the binding ability or the function of a
known molecule can be made based on the spatial arrangement of a small number of atoms or
functional groups in the molecule. As used herein, such an arrangement is called a
"pharmacophore", and once the pharmacophore or pharmacophores in a molecule have been
identified, this information can be used to identify other molecules containing the same or
similar pharmacophores. Such methods are well known to persons of ordinary skill in the art
of medicinal chemistry, and as the structural information described in this application
identifies the pharmacophore of phosphoramidate alkylator prodrugs and phosphoramidate
alkylators. An example of programs available to perform pharmacophore -related searches is
the program 3D Pharmacophore search from the Chemical Computing Group (see
http://www.chemcomp.com/fdept/prodinfo.htm).
[0064] "Optional" or "optionally" means that the subsequently described event or
circumstance can, but need not, occur, and that the description includes instances where the
event or circumstance occurs and instances in which it does not. For example, "heterocyclo
group optionally mono- or di- substituted with an alkyl group" means that the alkyl can, but
need not be, present, and the description includes situations where the heterocyclo group is
mono- or disubstituted with an alkyl group and situations where the heterocyclo group is not
substituted with an alkyl group.
|0065] A combination of substituents or variables is permissible only if such a combination
results in a stable or chemically feasible compound. A stable compound or chemically
feasible compound is one in which the chemical structure is not substantially altered when
kept at a temperature of 4 °C or less, in the absence of moisture or other chemically reactive
conditions, for at least a week.
[0066] As used herein, a "prodrug" means a compound that, after administration, is
metabolized or otherwise converted to an active or more active form with respect to at least
one biological property, relative to itself. To produce a prodrug, a pharmaceutically active

compound (or a suitable precursor thereof) is modified chemically such that the modified
form is less active or inactive, but the chemical modification is effectively reversible under
certain biological conditions such that a pharmaceutically active form of the compound is
generated by metabolic or other biological processes. A prodrug can have, relative to the
drug, altered metabolic stability or transport characteristics, fewer side effects or lower
toxicity, or improved flavor, for example {see the reference Nogrady, 1985, Medicinal
Chemistry A Biochemical Approach, Oxford University Press, New York, pages 388-392).
Prodrugs can also be prepared using compounds that are not drugs but which upon activation
under certain biological conditions generate a pharmaceutically active compound. As used
herein a phosphoramidate alkylator prodrug is a prodrug that upon activation releases the
active phosphoramidate alkylator.
[0067] As used herein, a "cytotoxic agent" is an agent or compound that produces a toxic
effect on cells. As used herein, a "cytostatic agent" is an agent that inhibits or suppresses
cellular growth and multiplication.
[0068] As used herein "hypoxic cells" are cells residing in a hypoxic environment in vivo
such as, for example, in a hypoxic tumor zone, or in vitro. As used herein "normoxic cells"
are cells residing in a normoxic environment in vivo or in vitro. As used herein "hypoxic
cytotoxicity" of a compound or agent is its cytotoxicity on hypoxic cells. As used herein
"normoxic cytotoxicity" of a compound or agent is its cytotoxicity on normoxic cells.
[00691 As used herein, a "bioreductive group" refers to a group that accepts electrons in an
oxidation-reduction reaction. The bioreductive group is a group (1) that can be reduced, i.e.,
a group that can accept electrons, hydrogen, and/or or an hydride ion; (2) that can be reduced
111 VIVO (HluVOI ill YlfTv'i P) mat can ^e reduced in vivo and/or in vitro under hypoxia; (4) that
can be reduced m VW 9* VI W&0 ty Dittos*, thiols, or by photochemical or
eleCtrOChemiCal means; Or (5) that Can be eliminated and/or cleaved by abiological process,
such as by enzymatic hydrolysis, metabolism etc.
[0070] F°r example, and as described in more detail below, one bioreductive group is a
nitroimidazole that may be substituted with a variety of groups. Other examples of
bioreductive groups include, but are not limited to, groups based on electron deficient
nitrobenzenes, electron deficient nitrobenzoic acid amides, nitroazoles, nitroimidazoles,
mtrothiophenes, nitrOthiaZOleS, nitTOOXazoleS, nitrofurans, and nitropyrroles, where each of
these classes of moieties may be optionally substituted, such that the redox potential for the

bioreductive group lies within a range where the group can undergo reduction in the hypoxic
conditions of a tumor, by DT-diaphorase, and/or by a thiol. One of skill in the art will
understand, in view of the disclosure herein, how to substitute these and other bioreductive
groups to provide a a bioreductive group having a redox potential that lies within said range.
[0071] Generally, one of skill in the art can "tune" the redox potential of a bioreductive
group by modifying that group to contain electron withdrawing groups, electron donating
groups, or some combination of such groups. For example, nitrothiophene, nitrofbran, and
nitrothiazole groups may be substituted with one or more electron donating groups, including
but not limited to methyl, methoxy, or amine groups, to achieve the desired redox potential.
In another example, the nitropyrrole moiety can be substituted with an electron withdrawing
group, including but not limited to cyano, carboxamide, -CF3, and sulfonamide groups, to
achieve the desired redox potential. For this purpose, strong electron withdrawing groups
such as cyano, sulfone, sulfonamide, carboxamide, or-CF3, and milder electron withdrawing
groups such as -CKb-halogen where halogen is -F, -CI, or -Br, can be used.
[0072] As used herein, an "anti-neoplastic agent", "anti-tumor agent", or "anti-cancer
agent", refers to any agent used in the treatment of cancer. Such agents can be used alone or
in combination with other compounds and can alleviate, reduce, ameliorate, prevent, or place
or maintain in a state of remission of clinical symptoms or diagnostic markers associated with
neoplasm, tumor or cancer. Anti-neoplastic agents include, but are not limited to, anti-
angiogenic agents, alkylating agents or alkylators, antimetabolite, certain natural products,
platinum coordination complexes, anthracenediones, substituted ureas, methylhydrazine
derivatives, adrenocortical suppressants, certain hormones and antagonists, anti-cancer
polysaccharides, chemoprotectants, and certain herb or other plant extracts.
[0073] As used herein, "cancer" refers to one of a pup of more than 100 diseases caused
by the uncontrolled growth and spread of abnormal cells that can take the form of solid
tumors, lymphomas, and non-solid cancers such as leukemia.
[0074] As used herein, "malignant cancer" refers to cancer cells or cancers that have the
capacity of metastasis, with loss of both growth and positional control.
[0075] As used herein, "neoplasm" (neoplasia) or "tumor" refers to abnormal new cell or
tissue growth, which can be benign or malignant.

[0076] As used herein, "treating" a condition or patient refers to taking steps to obtain
beneficial or desired results, including clinical results. For purposes of this invention,
beneficial or desired clinical results include, but are not limited to, alleviation or amelioration
of one or more symptoms of cancer or other hyperproliferative disease conditions,
diminishment of extent of disease, delay or slowing of disease progression, amelioration,
palliation or stabilization of the disease state, and other beneficial results described below.
[0077] As used herein, "reduction" of a symptom or symptoms (and grammatical
equivalents of this phrase) means decreasing of the severity or frequency of the symptom(s),
or elimination of the symptom(s).
[0078] As used herein, "administering" or "administration of a drug to a subject (and
grammatical equivalents of this phrase) includes both direct administration, including self-
administration, and indirect administration, including the act of prescribing a drug. For
example, as used herein, a physician who instructs a patient to self-administer a drug.and/or
provides a patient with a prescription for a drug is administering the drug to the patient.
[0079] As used herein, a "therapeutically effective amount" of a drug is an amount of a
drug that, when administered to a subject with cancer, will have the intended therapeutic
effect, e.g., alleviation, amelioration, palliation or elimination of one or more manifestations
of cancer in the subject. The full therapeutic effect does not necessarily occur by
administration of one dose, and can occur only after administration of a series of doses.
Thus, a therapeutically effective amount can be administered in one or more administrations.
[0080] As used herein, a "prophylactically effective amount" of a drug is an amount of a
drug that, when administered to a subject, will have the intended prophylactic effect, e.g.,
preventing or delaying the onset (or reoccurrence) of disease or symptoms, or reducing the
likelihood of the onset (or reoccurrence) of disease or symptoms. Tke full prophylactic effect
does not necessarily occur by administration of one dose, and can occur only after
administration of a series of doses. Thus, a prophylactically effective amount can be
adrninistered in one or more administrations.
[0081] AS USed herein, a "Second line" therapy refers to therapy that is given for the
treatment of a cancer which has failed to respond to a first chemotherapy regimen or "first
line" chemotherapy. "Third line" therapy refers to therapy that is given for the treatment of a
cancer when both initial treatment, first-line therapy, and subsequent treatment, second-line
therapy, don't work, or stop working is called.

[0082] As used herein "LogP" means a measure of lipophilicity of a substance determined
based on the partitioning of the substance betwen octanol and water.
IIa. Compounds
[0083] Most drug-mediated cancer therapies, including phosphoramidate alkylator-based
therapies, rely on poisons, called cytotoxic agents, selective for dividing cells targeting, for
example, their replicating DNA, microtutbule, and various growth factors and growth factor
receptors. These drugs are effective, because cancer cells generally divide more frequently
than normal cells. However, such drugs almost inevitably do not kill all of the cancer cells in
the patient One reason is that cancer cells can mutate and develop drug resistance. Another
is that not all cancer cells divide more frequently than normal cells and slowly-dividing
cancer cells can be as, or even more, insensitive to such cytotoxic agents as normal cells.
[0084] Some cancer cells reside in a poorly vascularized solid tumor, are unable to generate
the energy required for cell division and divide slowly. As a tumor grows, it requires a blood
supply and, consequently, growth of new vasculature. The new vasculature that supports
tumor growth is often disordered; leaving significant regions of the tumor under-vascularized
and even the vascularized regions subject to intermittent blockage. These under-vascularized
and blocked regions of the tumor become hypoxic - they have a lower oxygen concentration
or a lower oxygen partial pressure than the corresponding normal tissue, and the cells in them
exhibit slower rates of division. Thus, the median oxygen concentration of only ten percent
of solid tumors falls in the normal range of 40 to 60 mm Hg, and fifty percent of solid tumors
exhibit median oxygen concentrations of less than 10 mm Hg.
[(11)891 Tn6 hVDOXlC STOilS Of &e ^mor rePresent a signifi03111 source of metastases and
cancer cells resistant to therapy (see for example, De Jaeger et aL, Br J Cancer. 2001,
84(9):1280-5 and Rofstad et aL, Br J Cancer. 1999,80(11):1697-707). Not surprisingly,
then, low tumor oxygen levels are associated with a poor response to therapy, increased
metastases and poor Survival. The mechanisms of activation and action of
Cyclophosphamide and Ifosfamide can exemplify how these agents can not specifically target
the difficult to kill hypoxic zone of a tumor.
[0086J Both Cyclophosphamide and Ifosfamide are prodrugs and can be oxidatively
activated in the liver via intermediates to yield active phosphoramidate alkylators, Alkylators

1 (cylophosphamide mustard) and 2 (ifosfamide mustard), respectively (see below). The
charge neutral Hemiacetals 1 and 2, can have a half life of many minutes and can permeate in
and out of the cell. In contrast, the anionic Alkylators 1 and 2 are much less cell membrane
permeable and once formed extracellularly inefficiently kills the cell by alkylating cellular
DNA.
[0087] When the phosphoramidate alkylators reach the tumor, they generally kill cells in
the fast growing, well vascularized, normoxic, outer zone of the tumor. However, these
phosphoramidate alkylators are not as effective in permeating into the less vascularized,
slower growing, progressively hypoxic inner tumor zones and in killing tumor cells therein.
Before any of these active alkylators reach the tumor, they can react with healthy cells and
result in toxicity and/or cell death.

10088] While te hypoxic tumor ie fiffi«*to freat? ** hvrxic tumor zone can generate
reduced derivatives of a variety of chemical groups (see the reference Workman et al, 1993,
Cancer andMetOSt. ifev. 12; 73-82), and prodrugs of cytotoxins can be developed to exploit
such bioreductive environments (PCT Application Nos. US 04/009667 and US 05/08161;
PCT/US2005/041959 and PCT/US2005/042095, all Matteucci et al). Such a hypoxia
reducable (or hypoxia activated) prodrug can be constructed by employing a bioreductive
group (Z3) together with an alkylator. The bioreductive group is employed as part of a
Trigger moiety covalently bonded or attached to the phosphoramidate alkylator.

[0089] The compounds of the invention can generally be described as phosphoramidate
alkylator prodrugs. In general, the phosphoramidate alkylator prodrugs of the invention have
the following structure
Alk-Trigger
wherein Alk is a phosphoramidate alkylator and Trigger T has a structure L-Z3, wherein the
linker L is bonded to a bioreductive group Z3. La one embodiment, the Trigger T is a hypoxia
activated trigger.
[0090] Phosphoramidate alkylator derivatives are reported in the references, Borch et al, J.
Med. Chem. 2000,43: 2258-65; 2001,44:69-73; 2001,44:74-7; Hernick et al. J. Med.
Chern. 2002,45: 3540-8; Hernick et al, J. Med. Chem. 2003,46:148-54; US Patent Nos.
4,908,356; 5,306,727; 5,403,932; 5,190,929; 5,472,956; and 6,656,926; US Patent
Application Publication No.US 2003/0008850; and Papot et al, Curr. Med. Chem., 2002, 2,
155-85, isolated compounds of which are disclosed therein, and are not the subject of the
present invention. In some embodiments, the phosphoramidate alkylator prodrugs of the
present invention have one or more of the following characteristics: (i) a higher hypoxic
toxicity or lower value of IC50 or IC90, in hypoxic tissue, (ii) lower normoxic cytoxicity, and
(iii) less toxic side effect profile or some combination of these attributes. In some
embodiments, the phosphoramidate alkylator prodrugs of the present invention differ from
known phosphoramidate alkylator derivatives by: (i) the nature of the phosphoramidate
alkylator released, (ii) the nature of the linker (L) and/or the bioreductive group Z3, (iii) the
presence of more than one bioreductive group moiety, or some combination of these
attributes (iv) increased hypoxia selective cytoxicity measured by larger HCR values (v)
increased aqueous solubility (vi) increased stability to liver microsomal degradation and/or
(vii) providing effective phospnoramide alkylator pfodfllgS that 9T6 acMral and aVOld
enantiomer specific in vivo metabolism.
[0091] To understand why the prodrug compounds of the present invention represent a
significant advance over known anti-cancer phosphoramidate alkylator derivatives, an
understanding of tumor biology particularly under hypoxia and pharmacokinetics, and
pharmacodynamics of prodrugs provided herein in particular is helpful.
[00921 For effective tumor therapy, a hypoxia activated prodrug should be much less toxic
t0 healthy normoxic cells compared to hypoxic tumor cells. In some embodiments, the
hypoxia activated prodrugs of the invention are less active and less toxic to normoxic cells

than hypoxic cells. When such a prodrug of the invention encounters the hypoxic, reducing
environment within solid tumor tissue, reduction of the bioreductive group causes
dissociation of the the phosphoramidate alkylator or the active cytotoxin. The
phosphoramidate alkylator is released within the tumor zone and can more easily penetrate
the hypoxic region of the solid tumor. These phosphoramidate alkylators can kill cells in the
difficult to reach hypoxic region of the solid tumor while minimizing death of non-cancerous
healthy cells and toxic side effects to the patient. Thus the present invention provides
hypoxia activated prodrugs that are much less toxic to healthy, normoxic cells compared to
hypoxic, tumor cells.
[0093] In certain embodiments, the phosphoramidate alkylator prodrugs of the present
invention employ nitro containing aromatic or indole quinone moieties as bioreductive
groups in the Trigger T. In the hypoxic tumor, the nitro group is reduced to an
hydroxylarnino or an amino group and flow of an electron pair from the amino or
hydroxylamino group through the conjugated % electron system of the Trigger T releases the
phosphoramidate alkylator. In another embodiment, in a hypoxic tumor, an indole quinone is
reduced to an indole hydroquinone and flow of an electron pair from the hydroquinone
through the Trigger T releases the phosphoramidate alkylator. The released phosphoramidate
alkylator kills cells in and/or near the hypoxic tumor.
[0094] A number of enzymes can be responsible for the reduction of the bioreductive group
Z3 in the Trigger. For example, cytochrome P450 reductase enzymes can reduce the nitro or
a quinone moiety in a bioreductive group in a first step respectively to a N02('-) or a
serniquinone radical anion. The hypoxic tumor zone can have a higher concentration of the
reductase enzyme compared to normoxic tissue. Under normoxia, as in well vascularized
Mi assiirx ii in pro ftp * T) OI to "***"" "**mion
formed can react with oxygen to revert back to the bioreductive group and not ultimately
generate or release the phosphoramidate alkylator. The aryl or heteroaryl moiety covalently
bonded to the NO2C-) or the serniquinone radical anion modulates the oxygen sensitivity of
the radical anion.
[0095] The oxygen sensitivity of the bioreductive group varies depending partly on the
reduction potential of the bioreductive group. Thus, for example, one bioreductive group can
get reduced in a hypoxic tumor zone having 1 % oxygen, another in a zone having 0.1%
oxygen, and yet another in a zone having 0.01 % oxygen.

[0096] A bioreductive group loses some or all of its hypoxic specificity when it is so easily
reduced that the cytochrome P450 reductase enzyme or other reducing agents ("reducing
agents") in healthy normoxic tissue can reduce it in the presence of oxygen. If a NO2C-) or a
semiquinone radical anion in a bioreductive group does not react or reacts slowly with
oxygen, the radical anion itself can release the phosphoramidate alkylator, or can be further
reduced and release (he phosphoramidate alkylator, causing toxicity to healthy normoxic cells
and tissue. The novel phosphoramidate alkylator prodrugs of the present invention are more
toxic to the hypoxic cancer cells and tissue compared to the healthy normoxic cells and
tissue.
[0097] The ease or difficulty of reducing the bioreductive group Z3 can be measured by the
reduction potential of the bioreductive group and is influenced by the linker (L), and the
phosphoramidate alkylator (Alk-H). For example, attachment of the bioreductive group to an
electron withdrawing linker or an electron withdrawing phosphoramidate alkylator can make
the bioreductive group easier to reduce compared to when it is covalently bonded to an
electron rich linker or'an electron rich phosphoramidate alkylator.
[0098] The Trigger T can be oxidized, hydrolyzed, or thiolyzed and can release the
phosphoramidate alkylator in a hypoxia non-senselective manner. Telcyta™, a
phosphoramidate alkyltor prodrug that is in the clinic, can release an active toxin in absence
of hypoxia by the action of glutathione transferase (see, e.g., phophorarnidate alkylator If in
the "Methods of Treatment" section). The chemical nature of the linker and/or the
phosphoramidate alkylator can influence the oxidative, hydrolytic, or thiolytic stability of the
prodrug with respect to phosphoramidate alkylator release. In one embodiment of the present
invention a hypoxia activated phosphoramidate alkylator prodrug, does not release the
phosphoramidate alkylator in a hypoxia non-specific, oxidation, hydrolysis, or thiolysis.
[0099] According to the present invention, a properly employed Trigger in a
phosphoramidate alkylator prodrug can be used to "tune" the pharmacokinetic property of the
prodrug without altering its cytotoxic properties. For example, a high volume of distribution
of an anticancer agent ensures that the prodrug is absorbed in the tissue quickly. According
to the present invention, in one embodiment, the volume of distribution of a phosphoramidate
alkylator prodrug can be modulated by employing a Trigger T containing an amino group
capable of forming an anunamum cation under physiological conditions, in one
embodiment, a Trigger T containing a quaternary ammonium group can yield a prodrug

compound of the invention having a high volume of distribution while avoiding possible
endosomal trapping. In another emdodiment, a Trigger T comprising a carboxyl functionality
will exist as the anionic carboxylate anion form. CC^Oin the extracellular space outside of
normal healthy tissue and not pass easily through the normal cell membrane. The lower pH
in tumor extracellular space can convert the CO20 to the uncharged "CO2H" form allowing
the prodrug to pass through tumor cell membrane.
[0100] A phosphoramidate alkylator containing a hydroxyl, amino, mercapto, and/or a
carboxyl group can be transformed into a prodrug by covalently attaching a Trigger T to one
or more of these functional groups. During the transformation from a phosphoramidate
alkylator to a prodrug, a hydroxyl group in the phosphoramidate alkylator can be
transformed, for example, to an ether or an acetal; an amino to an allcylarnino, a carbamate, or
an amide; a carboxyl group to an ester; and a mercapto group to a thioether or a tbioacyl, as
described in greater detail in the Method of Synthesis and the Experimental sections below.
These transformations can yield a prodrug which is less polar or more lipophilic than the
corresponding phosphoramidate alkylator. Non polar phosphoramidale alkylator prodrugs
may not be readily soluble in aqueous pharmaceutical carriers or diluents. Solubility
enhancer groups like CO2H, amino, alkylamino, dialylamino, and hydroxyl can be employed
in the Trigger T to modulate the solubility of the prodrug and overcome any problems
encountered in preparing aqueous formulations of the phosphoramidate alkylator prodrugs.
[0101] Phosphoramidate alkylators of the present invention can have one or more N-(2-
haloalkyl) or N-(2-haloethyl) and/or one or more aziridine moiety covalently bonded
to a P=0 moiety as shown below. Upon release of the anionic phosphoramidate alkylator
moiety an aziridine or aziridium species forms which can alkylate DNA (See EXAMPLE
wSnm*% Deplngpnfc«W*«BWta|ffltUKOfll!l(lIli
substituents, the aziridinium formation kinetics can vary. For example, as shown in the
reaction sequence below, the rate of alkylation can increase when tne W2% moiety is
changed from NH2 to ^see Engle et al, J. Med. Chan., 1987,25:1347-57).
Substituents on the nitrogen atoms can alter the geometry of the phosphoramidate alkylator,
the delOCalizatiOIl Of the lOIie electron pair on this nitrogen atom in the P=0 moiety, the
availability of the nitrogen lone electron pairs for aziridinium or subsequent aziridine
formation, and the aqueOUS SOluMity Of the phosphoramidate alkylator prodrug and the
phosphoramidate alkylator.


[0102] The present invention arises in part out of the discovery that phosphoramidate
aikylator prodrugs employing 2-rdtroiinidazole-bioreductive group show unexpectedly high
hypoxic cytotoxicity, low normoxic toxicity and high HCR and improved solubility. For
example, Compounds 24 and 25 were respectively, 400 to 1000 fold more toxic in hypoxic
cells than in normoxic cells in an anti-proliferation cytotoxicity assay with a IC5o of 0.05 uM
in cells under hypoxia. (See EXAMPLE section). Phosphoramidate aikylator prodrugs
containing ifosfamide mustard or ifosfamide mustard analogs and having formulas:
Z3-CH2-0-P(=O)(NHCH2CH2X4)2, Z3-CH2-0-P(=O)(NHCH(R9)CH2X4)2, and
Z3-CH(Z2)-0-P(=O)(NHCH(R9)CH2X4)2;
wherein Z2 is methyl; R9 is hydrogen, methyl, or isopropyl; Z3 is l-N-methyl-2-
nitroimidazol-5-yl), 2-nitrothiophen-5-yl, or 2-nitrofuran-5-yl; and each X4 is CI or Br were
unexpectedly more toxic in hypoxic cells compared to normoxic cells, and/or possessed
unexpectedly high HCR values, in anti-proliferation cell cytotoxicity assays, in contrast to the
HCR values of known phosphoramidate aikylator derivatives having 2-nitrotliiophene-5-yl,
2-nitrofuran-5-yl, or 5-nitro imidazolyl, bioreductive groups (Z3), and N,N'(tetrakis-2-
choloroethyl) phosphoramidate mustard or cyclofosfamide mustard; or an indole quinonyl
group as Z3 and ifosfamide mustard (see e.g., compounds P4, P14-17, P19, and P21-22, in
BorCh It a/., J. Afei Cftffll.3 andUS Patent No. 6,656,926 both supra).
[0103] In one aspect, the present invention provides phosphoramidate alkylators prodrugs
onormula(l):


wherein
[0104] Y, is O, S, NRe or NSO2R6 wherein each R6 is independently C1-C6 alkyl, C1-C6
heteroalkyl, aryl, or heteroaryl;
[0105] Y2 is O, S, NRe, NCOR6, or NSO2R6;
[0106] each of R1-R5 independently is hydrogen, hydroxyl, amino, C1-C6 alkyl,C1-C6
heteroalkyl, C3-C8 cycloalkyl,heterocyclyl, C1-C6alkoxy, CpCg alkylamino, C1-C6
dialkylamino, aryl, heteroaryl, C1-C6 acyl, C1-C6heteroacyl, aroyl, or heteroaroyl; or together
any two of R1-R5 form a C3-C10 heterocycle; or each of R1-R5 independently is a Trigger T
wherein Trigger is L-Z3;
[0107] L is selected from
-[CCZi^-YjktCCOy-Olq-tqZOz^-Y^u^CCZ.W^KlZO^ZOlgSand
-[C(Z1)2-Y3]v-(S(=O)2V[C(Z1)2-Z2-Y4]u-[C(Z1)2]z-[C(Z1)=C(Z1)]g-;
wherein each z, v, q, u, and g independently is 0 or 1;

[0108] Y3 is S, O, or NR7 wherein each R7 is independently hydrogen, hydroxyl, C1-C6
alkyl, C1-C6heteroalkyl, C3-Cg cycloalkyl, heterocyclyl, C1-C6 alkoxy, C1-C6 alkylamino, C1-
C6 dialkylamino, aryl, heteroaryl, C1-Cg acyl, C1-C6heteroacyl, aroyl, or heteroaroyl;
[0109] Y4 is O, S, or -NR7-C(=O)-0-;
[0110] each Z\ independently is hydrogen, halogen, C1-C6 alkyl, C1-C6 heteroalkyl, aryl,
heteroaryl, C3-CS cycloalkyl, heterocyclyl, C1-C6 acyl, C1-Cgheteroacyl, aroyl, or heteroaroyl;
[0111] Z2 is C1-C6 alkylene, C1-C6heteroalkylene,

[0112] eaeh X1 is independently N or CR8 wherein R8 is independently hydrogen, halogen,
OH, OP(=O)(OH)2, nitro, cyano, CO2H, C1-C6 alkyl, C,-C6 heteroalkyl, C,-C6 cycloalkyl, Cr
C6 alkoxy, C1-C6 alkylamino, C1-C6 dialkylamino, aryl, CON(R7)2, C1-C6 acyl, Cr
Ceheteroacyl, aroyl, or heteroaroyl;
[0113] X2isNR7,S,orO;and

[0114] Z3 is a bioreductive group selected from the group consisting of:

with the proviso that in formula (I) :
[0115] (i) at least two of RrR5 are selected from the group consisting of 2-haloalkyl, 2-
alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-aryisulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl;
[0116] (ii) at least one of RrR5 is selected from the group consisting of 2-haloalkyl, 2-
alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl; and at least oneofNRaRj andl^R-sis
[0117] (iii)?a9ii?Wj^d^re
[0118] an individual isomer or a racemic or non-racemic mixture of isomers, bioisosteres,
pharmacophores, a pharmaceutically acceptable salt, solvate, hydrate, Of a flf6dfUg thereof.
[0119] In one embodiment, z is 1.
[0120] In one embodiment, R2-Rs are not the same.
[0121) In one embodiment, any one Of R"-R IS


[0122] In one embodiment, the present invention provides hypoxia activated
phosphoramidate alkylator prodrugs each employing two phosphoramidate alkylators. In one
embodiment, the phosphoramidate alkylator prodrug of the present invention employs an 1-
N-alkyl-2-nitroimidazol-5-yl moiety or a l-N-methyl-2-nitroimidazol-5-yl moiety as a
bioreductive group or Z3. In one embodiment, the phosphoramidate alkylator prodrug of the
present invention employs a 2-nitrofuran moiety as a bioreductive group or Z3.
[0123] In one embodiment, the present invention excludes the compounds:





5 wherein Ra is H, Br (P14), NMe2 (P15), CN (P16), or CONH2 (PI7),




wherein R is H, Me or allyl;
[0124] 3-(5-Methoxy-l -methyl-4,7-indolequinonyl)-methyl bis[N-methyl-N-(2-
bromoethyl)] phosphorodiamidate (P27),
[0125] 3-(5-Methoxy-l -methyl-4,7-indolequinonyl)methyl N,N-bis(2-bromoethyl)-
phosphorodiamidate (P28),
[0126] ^(s-Methoxy^-metliyW^WolequifionyllnieW bis[N-mcthyl-N-(2-
bromoethyl)]phosphorodiami(iate{P29),
[0127] 2-(5-Methoxy-l-methyl-4,7-indolequinonyl)methylN,N-bis(2-chloroethyl)
phosphorodiamidate (P30),
[0128] 2-(5-Methoxy4-methyl-4,7-indoleqmnonyl)methylN,N-bis(2-bromoethyl)-
phosphorodiamidate (P31),
[0129] 3-(5-Methoxy-l-methyl-4,7-indolequin-onyl)methylN,N-bis(2-bromoethyl)-
phosphorodiamidate (P32),

[0130] 2-(5-Methoxy-l -memyl-4,7-indolequinonyl)methyl bis[N-methyl-N-(2-
bromoethyl)]phosphorodiamidate (P3 3),
[0131] 2-(5-Methoxy-l -methyl-4,7-indolequinonyl)methyl N,N-bis(2-chloroethyl)-
phosphorodiamidate (P34), and
[0132] 2-(5-Methoxy-l -methyl-4,7-indolequinonyi)methyl N,N-bis(2-bromoethyl)-
phosphorodiamidate (P35)
[0133] In a related embodiment, the present invention provides a compound of formula (I)
with the proviso that
[0134] (i) at least one of R1-R.5 are selected from the group consisting of 2-
alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl and
[0135] at least one of R1-R5 are selected from the group consisting of 2-haloalkyl, 2-
alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl; or
[0136] (ii) at least one of R1-R5 is selected from the group consisting of 2-haloalkyl, 2-C1-
C6alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl; and at least one of NR2R3 and NR4R5 is or
[0137] (iii) each NR2R3 and NR4R5 are
[0138] In another related embodiment, the present invention provides a compound of
formula (I) with the proviso that the formula (I) excludes R2 and R3 together forming a
m&fpMffie ring or h and Rs together forming a morpholine ring,
[0139] In one embodiment, the present invention excludes a compound of the following
structure:


wherein Zj is hydrogen or C1-C6 alkyl.
[0140] In one embodiment, the present invention provides compounds wherein the Trigger
Tis:
[C(Z1)2-Y33-(C(=O)-OHC(Z1)2-Z2-Y4]-[C(Z1)2]2-[C(Z1)-C(Z])3-Z3;
[CCZ,)2-Y3HC(Z1)r2i-Y4]-[C(Z1)2k[C(Z1)=C(Z1)]-Z3;
[C(Z1)2-Y3]-[C(Z1)2]2-[C(Z1)=C(Z1)]-Z3;
[C(Z1)2-Y3-[C(Z1)2]2-Z3;
[C(Z1)2-Y3]-(C(=O)-O)-[C(Z1)2]r[C(Z1)-C(Z0]-Z3;
[C(Z1)2-Y3]-(C(=0>0)-[C(Z1)2]^[C(Z1)=C(Z1)]-Z3;
[C(Z1)2-Z2-Y4]-[C(Z1)2V[C(Z1)=C(Z1)]-Z3;
-taz^jz-tqz^cxzoi-Zj;

[0141] In an additional embodiment, Z3 is:


[0142] In one embodiment, each -C(Z1)r is: -CH2-, -CHMe-, -CH(CN)-, -CH(CO2H)-, -
CH(CONH2>, -CH(CF3)-, -CH(CHF2)-, -C(Me)2-, -C(Et)2-, -CH(CH2NMe2>, -
CH(CH2NMe2>, -C(CH2NMe2)2-, or -C(CH2CO2H)2-.
[0143] In one embodiment,-C(Z1)2-Y3- is: -CH2-0-, -CH2-S-, -CH2-NMe, -CH2-NH-,
CH(Me)-0-, CH(Me)-S-; -CH(Me)-NMe-, -CH(Me)-NH-; -CMe2-NMe-, -CM^-NMe-, or-
CMe2-NMe-.
[0144] In one embodiment -Z2-Y4- together is:


[0145] In one embodiment, -[C(Z0=C{Zi)]- is:-CH=CH-, -C(CN)=CH-, -CH=C(CN)-, -
C(Ar)=CH-, -CH=CAr~, -C(COAr)=CH-,-CH=C(COAr)-, -C(COR,2)=CH- or-
CH=C(CORi2)-, wherein Ar is aryl optionally substituted with up to five substituents selected
from the group consisting of OH, OMe, CF3,0-CHF2, OCF3, N02, CN, halo, halomethyl,
dihalomethyl, trihalomethyl, hydroxymethyl, CO2H, CONH2, COMMe2, and CONHMe; and
Ri2 is is independently hydrogen, C1-C6 alkyl, C1-Cgheteroalkyl, C3-Cg cycloalkyl, or
heterocyclyl.
[0146] In another embodiment, Trigger is:

hi another embodiment, Trigger is

wherein each Z] independently is H or C1-C6 alkyl.
[0147] In another embodiment, Trigger is:

wherein each Zi is hydrogen or C1-C6 alkyl and Rs is H, OH, or -OP(=O)(OH)2-
(0148] In one embodiment, the present invention provides compounds of formulas (II) and
(III):


wherein each R2-R5 independentiy is selected from the group consisting of hydrogen,
hydroxyl, C1-Q alkyl, Q-Qheteroalkyl, C3-C8 cycloalkyl, heterocyclyl, C1-C6alkoxy, C1-Cg
alkylamino, C1-C6dialkylamino, aryl and heteroaryl; or together any two of R2-R5 form a C3-
C10 heterocycle; each Yi independently is S or O; and each Trigger T is defined as in formula
(D;
[0149] with the proviso that in formulas (H) or (III):
[0150] (i) at least two of R1-R5 are selected from the group consisting of 2-haloalkyl, 2-
alkylsulfonyloxyalkyl, 2JieteroaIkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2
heteroalkylsulfonyloxyalkyl; or
[0151] (ii) at least one of R1-R5 is selected from the group consisting of 2-haloalkyl, 2-Cr
C6 alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl; and at least one of NR2R3 and NR4R5 is ; or
[0152] (iii) each NR2R3 and NR4R5 are and
[0153] an individual isomer or a racemic or non-racemic mixture of isomers, bioisosteres,
pharmacophores, a pharmaceutically acceptable salt, solvate, hydrate, or a prodrug mereOT.
[0154] In one embodiment, the present invention provides a compound of formula (II)
wherein Trigger T is -CH2-Z3, -CH(Z1)-Z3, or -C{Z\h-li wherein Z, is C,-alkyl and Z3 is:

with the proviso that in formula (II):

[0155] (i) one of R2 and R3 is H and one of R4 and R5 is H;
[0156] (ii) one of R2 and R3 is C1-alkyl and one of R4 and R5 is C1 -alkyl; or
[0157] (iii) at least one of R2-R5 is hydroxy!, amino, C3-Cg cycloalkyl, heterocyclyl, C1-C6
alkoxy, C1-C6 alkylamino, Q-Cgdiallcylarnino, aryl, heteroaryl, C1-C6 acyl, C1-C6heteroacyl,
or aroyl or heteroaroyl.
[0158] In one embodiment, the present invention provides a compound of formula (II)
wherein Z3 is a bioreductive group selected from:

with the proviso that in formula (I):
[0159] (i) at least one of R1-R5 are selected from the group consisting of 2-
alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl and
[0160] at least one of R1-R5 are selected from the group consisting of 2-haloalkyl, 2-
alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl; or
[0161] (ii) at least one of R1-R5 is selected from the group consisting of 2-haloalkyl, 2-Cr
C6 aIkylsu]fonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
Keteroalkylsulfonykyalkyl; and ai least one Of NR1R3 ^ W^Jls or
[0162] (iii) each NR2R3 and NR4R5 are
[0163] In one aspect, the present invention provides phosphoramidate alkylator prodrugs of
formula (I):


wherein
[0164] R, is a -[CCZiVYjlv-lCC^-Olq-^Z^-Zz-Y^u-Zj or
-[CiZ^-Yi^SfrOy^q-lCiZiyz-Tji-Y^-Z^, wherein each v, q, and u independently is 0 or 1;
and Z3 is a glucose or an analog thereof with the proviso that it excludes glucose conjugates
of phosphoramidate alkylators described in the reference Wiessler et ah, US Patent No.
5,622,936;
[0165] each of R2-R5 independently is hydrogen, hydroxyl, amino, C1-C6~alkyl, C1-C6
heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, C1-C6alkoxy, C1-C6alkylamino, C1-C6
dialkylamino, aryl and heteroaryl, C1-Cg acyl, C1-C6heteroacyl, aroyl, or heteroaroyl; or
together any two of R1-R5 form a C1-Cjoheterocycle;
[0166] with the proviso that in formula (I):
[0167] (i) at least two of R2-R5 are selected from the group consisting of 2-haloalkyl, 2-
alkylsulfonyloxyalkyl, 2_~heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl;
[0168] (ii) at least one of R2-R5 is selected from the group consisting of 2-haloalkyl, 2-Cr
C6 alkylsulfonyloxyaIkyI, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl; and at least one of NR2R3 and NR4R5 is or
>
[0169] (iii) each NR2R3 and NR4R5 are and
[0170] an individual isomer or a racemic or non-racemic mixture of isomers, bioisosteres,
pharmacophores, a pharmaceutically acceptable salt, solvate, hydrate, or a prodrug thereof.
[0171] In another embodiment, the present invention provides the compounds:


[0172] In another embodiment, the present invention provides the compounds:

wherein X4 and Z3 are defined as above.
[0173] In another embodiment, the present invention provides the compounds:


In one embodiment, R6 is -(N-CHbCEfeXOa.
[0174] In another embodiment, the present invention provides the compounds:

wherein R2 - R5 are defined as in formula (H).
[0175] The following scheme exemplifies hypoxic reduction of the phosphoramidate
alkylator prodrug to yield the corresponding pWspktamid&te altyMoi.


{0176) In another embodiment, the present invention provides the compounds:

wherein R2 - Rs are defined as in formula (II).
[0177] The following scheme exemplifies hypoxic reduction of the phosphoramidate
alkylator prodrug to yield the corresponding phosphoramidate alkylator.

[0178] In one embodiment, the present invention provides the compounds of the formulas
(IV)-(VII)



wherein each R9 independently is hydrogen, deuterium, aryl, heteroaryl, C1-Q alkyl, Q-Q
heteroalkyL, C3-C& cycloalkyl, heterocyclyl, C1-C6 acyl, C1-C6heteroacyl, aroyl, heteroaroyl,
d-C6 alkoxycarbonyl, C1-C6 alkylaminocarbonyl, di C1-C6 alkylaminocarbonyl, or C1-C6
alkoxy; or together two R9 groups form a heterocycle; each Rio is hydrogen, C1-Q alkyl, C1-
C6 heteroalkyL C3-C8 cycloalkyl, heterocyclyl aroyl or heteroaroyl, or together two RJ0
groups form a heterocycle;
[0179] R11 is independently is hydrogen, deuterium, aryl, heteroaryl, C1-C6 alkyl, C1-C6
heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, C1-C6 acyl, C1-C6heteroacyl, aroyl, heteroaroyl,
C1-C6 alkoxycarbonyl, C1-C6 alkylaminocarbonyl, di Q-Q alkylaminocarbonyl, or C1-C6
alkoxy; or together two R9 groups form a heterocycle with the proviso that when Rn is C1-C6
alkyl or C1-C6 heteroalkyl then Rn excludesor together two Rj 1 groups form
a heterocycle;
[0180] X4 is CI, Br, alkylsulfonyloxy, heteroalkylsulfonyloxy, arylsulfonyloxy, or
heteroalkylsulfonyloxy; and
[0181] Trigger T is [C(Z02-Y3MCH))-O)q-[C(^^
[0182] In a related embodiment, in formulas (IV) - (VII), each R9 is independently
hydrogen, deuterium, C,-C3 alkyl, C1-C6heteroalkyl, C3-C6 cycloalkyl, heterocyclyl, aryl or
heteroaryl. In another embodiment, each R9 is independently hydrogen, deuterium, or C,-C6
alkyl. In another related embodiment, each R9 is independently methyl, ethyl, propyl,
isopropyl, isoktyl, tertiary butyl, or cyclopropyl.

[0183] In one embodiment, the present invention provides a compound of formula (IV)
wherein one of Rio is -(GftOe-Intercalator wherein an Intercalator is an aromatic or
heteroaromatic moiety capable of intercalating between a nucleic acid base pair.
[0184] In another embodiment, the present invention provides the compound:

wherein X4 and R10 is defined as in formula (IV).
[0185] In another embodiment, the present invention provides the compound:

[0186] In one embodiment, the present invention provides the compound of formula (VIII):


wherein each R9 is independently hydrogen, methyl, ethyl, propyl, isopropyl, or cyclopropyl;
and N(Rio)2 is selected from NH2, NHMe, NMe2, NEt;
NHOMe,andNHOH.
[0187] In one embodiment, the present invention provides the compound of the formula
(IX):

wherein each R9 independently is hydrogen, methyl, ethyl, propyl, isopropyl, or cyclopropyl.
[0188] In one embodiment, the present invention provides the compound of the formula
(X):

wherein each R9 independently is hydrogen, methyl, ethyl, propyl, isopropyl, or cyclopropyl;
and each Rn is independently hydrogen, methyl, ethyl, propyl, isopropyl, benzyl, substituted
methyl, cyclopropyl, methoxy, and hydroxyl; or together two Rn form a heterocycle.
[0189] In one embodiment, the present invention provides the compounds of the formulas
(X-A),(X-B)and(X-C):


wherein X2 and X4, are defined as in formula (I), and Rio, and Rn are defined as in formulas
(IV), (VI) and (VII).
10190] taeemMimeflt.ftoiJ^
(XI)-(XV):


wherein each Rn independently is hydrogen, methyl or substituted methyl, benzyl, isopropyl,
propyl, cyclopropyl, methoxy, and hydroxyl; and Xi, X2, and Zj are defined as above; and X4
is CI, Br, alkylsulfonyloxy, heteroalkylsulfonyloxy, cycloalkylsulfonyloxy,
MerOCyClOalWSUlfOnylOXy, arylsulfonyloxy, or heteroarylsulfonyloxy. In one embodiment,
111 COmpOUndS Of formulas (XII), (XIV), and (XV), when X, is CI or Br then Rn excludes
isopropyl. In one embodiment, a compound of formula (X) excludes a compound wherein Z3
is


[0191] In one embodiment, the present invention provides a compound of formula (XII),
(XIV), or (XV) wherein each Ru is hydrogen. Examples of compounds of formula XII, XIV,
or XV include compounds 5,7, 8,9,10,13,14,15,19,23,24,25,26, 32, 34, and 36. In one
embodiment, the present invention provides phosphoramidate alkylator prodrugs of formulas
XII, XTV, or XV wherein Rn excludes propyl or isopropyl. In another embodiment, the
present invention excludes the compound:

[0192] In one embodiment the present invention provides a phopsphoramidate alkylator
prodrug wherein Rn is C3-C8 cycloalkyl. In another embodiment, the cycloalkyl is
cyclopropyl. In general, a cyclopropyl group can be more stable than an alkyl group to
oxidatively metabolizing proteins in the cell, particularly in the liver the prodrug compounds
of the invention provide a pharmacokinetically improved phopsphoramidate alkylator
prodrug compared to known phosphoramidate alkylator prodrugs.
[0193] In one embodiment, the present invention provides the compounds of the formula
(XVI)

wherein K is C1-C6alkylene or C1-C6 heteroalkylene. In one embodiment K is (C(Ri2)2)ej
CH2CH2(-X6-CH2CH2)f, or CH2(-X6-CH2)f wherein e is 1-10, f is 0-3, and X6 is 0, S, or
NR12 wherein each Ri2 is independently defined as above.

[0194] In one embodiment, the present invention provides the compounds of the formula
(XVII -(XVIII)

wherein e is 0-4, X4 is CI or Br, alkylsulfonyloxy, heteroalkylsulfonyloxy, arylsulfonyloxy,
or heteroarylsulfonyloxy; Xe is O, S, or NR12 wherein R12 is defined as above.
[0195] In one embodiment, the present invention provides the compound of formula (XIX):
1
wherein e is 0-4, and X4 is CI, Br, alkylsulfonyloxy, heteroalkylsulfonyloxy, arylsulfonyloxy,
or heteroarylsulfonyloxy. In a related embodiment, the present invention provides a
compound of formula (XIX) wherein e is 1. See EXAMPLE section for examples of
compounds of formulas described herein.
[0196] In one embodiment, the present invention provides the compound of formula (XX):


(XX)
wherein R8 is glucose or a glucose analog; e is 0-4, and X4 is CI, Br, alkylsulfonyloxy,
heteroalkylsulfonyloxy, arylsulfonyloxy, or heteroarylsulfonyloxy. As used herein, a glucose
analog includes mono, di and tri saccharides. In a related embodiment, the present invention
provides a compound of formula XX wherein e is 1.
[0197] In one embodiment, the present invention provides the compounds:

wherein X4 is CI, Br; or alkylsulfonyloxy,
[0198] In one embodiment, the present invention provides the compounds;


wherein R9 and X4 are defined as in formula VI.
[0199] In one embodiment, the present invention provides the compound of the formula
(XXI)

wherein Yj is S or O; and Trigger T is defined as in formula (I).
[0200] In another embodiment, the present invention provides the oxime-phosphoramidate
alkylator conjugate: —

[0201] In one embodiment, such an oxime-phosphoramidate alkylator conjugate can be
hydrolyzed enzymatically to produce


[0202] In another aspect, the present invention provides the compounds of the formula
(XXII):

wherein
[0203] Rj - R5, Yi, and Y2 are defined as in formula (I);
[0204] each R1-R5 and Rt *-R5* independently is selected from the group consisting of
hydrogen, hydroxyl, C1-C6 alkyl, C1-C6alkoxy, C1-C6alkylamino, C1-C6dialkylamino, aryl,
heteroaryl; or together R2 and R2* form a heterocycle; or each R1-R5 and Ri*-R5*
independently is a Trigger T selected from the group consisting of
-[C(Z1)2-Y3]v-[C(=O)-O]q-[C(Z1)2-Z2-Y4]„-[C(Z1)2]z-[-C(Z1>=C(Z1)]g-Z3and
-[C(Z1)2-Y3]v-(S(=O)2)q-[C(Z1)2-Z2-Y4]u-[C(Z1)2]2-[C(Z1)=C(Z1)]g-Z3-;
with the proviso that in formula (XXII):
[0205] (i) at least two of R,-R5 and R,*-R5* are 2-haloalkyl, 2-alkylsulfonyloxyalkyl, 2.
heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, or 2 heteroalkylsulfonyloxyalkyl; or
[0206] ^ at least one of RrR5 and Rt*-R5* is 2-haloalkyl, 2-C1-C6 alkylsulfonyloxyalkyl,
at least one of NR2R3 and NR2*R3* is or
[0207] (iii) each NR2R3 and NR2*R3* both and
an individual isomer or a racemic or non-racemic mixture of isomers, bioisosteres,
pharmacophores, apharmaceutically acceptable salt, solvate, hydrate, or a prodrug thereof.
[0208] each Z independently is C, S, or P;
[0209] each t independently is 1 or 2;

[0210] each r independently is 0 or 1;
[0211] K is selected from the group consisting of Q-Csalkylene, C1-C6heteroalkylene,
arylene, or heteroarylene, (C(R9)2)n; and (Y5-(C(R9)2)m-Y4-(C(R9)2)m-Y6)n wherein n is 1-8;
[0212] each m independently is 1-4;
[0213] each R9 is independently C1-C6 alkyl or heteroalkyl, or together when covalently
bonded to the same carbon atom or adjacent carbon atoms are cycloalkyl or heterocyclyl; and
[0214] each Y4,Y5, and Y6 independently is O, S, NR7, or a bond; with the proviso that one
of Y4,Y5, and Y6 has to be O, S, or NR7.
[0215] In another aspect, the present invention provides the compounds of the formula
(XXHI):

wherein
[02161 RJ - Rs, Yi, and Y2 are defined as in formula (I);
[0217] each fy-Rj and Rf-Rf mdepfcfltly 15 ftlftttfi ftQUUuG gTOUp CQM5till£0f
hydrogen, hydroxyl, C1-C6 alkyl, C1-C6alkoxy, C1-C6alkylamino, C1-C6dialkylamino, aryl,
heteroaryl; or together R2 and R2* form a heterocycle; or each R1-R5* independently is a
Trigger T selected from the group consisting of
-[C(Z1)2-Y3]v-[C(=O)-O]q-[C(Z1)2-Z2-Y4]u-[C(Z1)2]z-[-C(Z1)=C(Z1)]g-Z3and
-[C(Z1)rY3]v-(S(=O)^^^
with the proviso that in formula (XXIII)
[0218] (i) at least two of R2-R5 and R2*-R5* are 2-haloalkyl, 2-alkylsulfonyloxyalkyl, 2-
heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, or 2 heteroalkyl-sulfonyloxyalkyl;

[0219] (ii) at least one of R2-R5 and R2*-R5* is 2-haIoaIkyl, 2-C1-C6 alkylsulfonyloxyalkyl,
2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, or 2-heteroalkylsulfonyloxyalkyl; and
one of NR2R3 and NR2*R3* is or
[0220] (iii) NR2R3 and NR2*R3* together are both or NR4R5 and NR4*R5*
together are both and
[0221] an individual isomer or a racemic or non-racemic mixture of isomers, bioisosteres,
pharmacophores, a pharmaceutically acceptable salt, solvate, hydrate, or a prodrug thereof.
[0222] L2is

wherein X is defined as above.
[0223] In another embodiment, the present invention provides the compound of the formula
(XXIV):

wherein R2, R3, JU, R2*, R3*, &»*, Z, K and Trigger are as defined in Formula (XXII).
[0224] In another embodiment, the present invention provides the compounds of formula
(XXIV) having the structure of formula (XXV) or (XXVI):


[0225] In another embodiment, the present invention provides the compound of the formula
(XXVI):
5
wherein Xi, X2, X4, and e are defined as in formula (XXV).
[0226] In another aspect, the present invention provides the compound of the formula
(XXVn):
[0
wherein R2-R5, r, k, Yi, and Trigger T are defined as in formula (XXIV).
[0227] In one embodiment, the the present invention provides a compound of Formula:


wherein
[0228] In another embodiment, the present invention provides a Z3 selected from the group
consisting of:


[0229J In another embodiment, the present invention provides a moiety having the formula:
selected from the group consisting of:




[0230] In another embodiment, the present invention provides a T selected from the group
consisting of:

wherein each Zi, R7, and R8 is defined as above. Within this embodiment, L\ is hydrogen,
methyl, or ethyl; R7 is methyl, trifluoroethyl, ethyl, propyl, and cyclohexyl; and R8 is OH or
0P(=O)(0H)2. Within this embodiment,


wherein each R9 is hydrogen or C1-C6 alkyl and each X4 is halo or RsuiS(=O)20-. In another
embodiment, R9 is hydrogen, methyl, ethyl, isopropyl, or isobutyl; and X4 is chloro, bromo,
or methanesulfonyloxy.
[0231J In another embodiment, the present invention provides a compound of formula:

wherein T is defined as above or more particularly T is L-Z3 wherein L CH2, CHMe, CMez,



[0232] In one aspect the present invention provides deuterated phosphoramidate alkylators
and deuterated phosphoramidate alkylator prodrugs of formula

wherein X4 is halo or R^S^O^O. In another embodiment, X4 is CI or Br. Such deuterated
phosphoramidate alkylators and their prodrugs are equally cytotoxic with respect to hypoxic
tumor tissue as their non-deuterated or hydrogenated analogs, such as compounds 25, 36 and
the likes,. However, the presence of such deuterated analogs in vivo, for example in blood
plasma, can be determined more efficiently compared to their corresponding
phosphoramidate alkylators and/or phosphoramidate alkylator prodrugs by nuclear magnetic
resonance methods and such deuterated analogs can be useful in determining
pharmacokinetic or pharmacodynamic properties of the phosphoramidate alkylators and/or
phosphoramidate alkylator prodrugs. Pharmacokinetic and or pharmacodynamic information
of phosphoramidate alkylators and/or phosphoramidate alkylator prodrugs is used in
determining dosage, frequency of dosing, and similar administration related parameters. The
synthesis of a octadeuterated-compound 25 and octadeuterated isofosfamide alkylator is
described in the EXAMPLE section.

[0233] In another group of embodiments, the present invention provides the individual and
selective groupings of the compounds of the EXAMPLES. Examples of compounds of the
invention include:













154 155
[0234] In One embodiment, the phQSphoraimdate alkylator prodrug contains

as Z3 and shows hypoxic tumor specific toxicity while being much less toxic to healthy,
normoxictisue.

[0235] In one embodiment, the present invention provides a novel phosphoramidate
alkylator prodrug which upon bioreduction releases the corresponding novel or known
phosphoramidate alkylator

wherein X4 is defined as in formula (T), and R9, Rio, and Rn, are defined as in formulas (IV)-
(VII), and ionized forms thereof. In a related embodiment, X4 is CI, Br, methanesulfonyloxy,
benzenesulfonyioxy, or para-toluenesulfonyloxy.
#
[0236] In one embodiment, the present invention provides a novel phosphoramidate
alkylator prodrug which upon bioreduction releases the corresponding novel or known
phosphoramidate alkylators


and ionized forms thereof;
wherein N(Rio)2 is selected from the group consisting of NH2, NHMe, NMe2, NEt2,
^JHOMeandNHOH;

each Rn is independently hydrogen, Me, ethyl, cyclopropyl, isopropyl, propyl, benzyl,
substituted methyl, cyclopropyl, methoxy, and hydroxyl; or together two Rn form a
heterocycle.
[0237] The anti-cancer agent Cyclophosphamide metabolizes to Id (Rio is hydrogen) and
Ifosfamide metabolizes to le (each Rn is hydrogen), when used in cancer treatment.
Glufosfamide, which is being evaluated in the clinic for cancer treatment, releases an
alkylator of formula le (each Rn is hydrogen, see Wiessler et al, US Pat. No. 5,622,936;
PCT application No. USO5/03370 entitled "Anti Cancer Therapies", US Pat. Appl. No.
60/638995 entitled "Glufosfamide Combination Therapy" and Attorney docket No. 021305-
005900US filed on 11 May 2005 entitled "Glufosfamide Combination Therapy"). Telcyta™
which is being evaluated in the clinic for cancer treatment, releases If (Rosen et ah, Clin
Cancer Res. 2004,10(ll):3689-98).
[0238] Known phosphoramidate alkylator prodrugs such as ifosfamide and
cyclophosphamide metabolize to produce cytotoxic by products such as acrolein and
chloroacetaldehyde which cause undesirable patient side-effects such as hemorrhagic cystitis,
coma or death. In one embodiment, the present invention provides a phosphoramidate
alkylator prodrug which upon metabolism produces less toxic by products per treatment as
compared to those produced by the metabolism of ifosfamide and/or cyclophosphamide. In
one embodiment, the phosplioromidate alkylator prodrugs of the present invention do not
produce acrolein by in vivo metabolism. Examples of toxic by products resulting from
metabolism of the prodrugs of the invention include chloro, bromo, alkylsulfonyloxy,
heteroalkylsulfonyloxy, arylsulfonyloxy, orheteroarylsulfonyloxy-acetaldehyde, (for
metabolic production of chloroacetaldehyde from ifosfamide see the reference Hardman et
al, supra, page 1396). In another embodiment, the present invention provides a
phosphoramidate alkylator prodrug which upon oxidative metabolism produces 5-95% as
much chloroacetaldehyde or an equivalent as defined above, per treatment, as produced by
ifosfamide metabolism.

[0239] The phosphoramidate alkylator derivative formed upon the reduction of Z3 can be
different from the phosphoramidate alkylator being protected and the phosphoramidate
alkylator prodrug and is termed a modified phosphoramidate alkylator prodrug. For example,
a phosphoramidate alkylator prodrug can yield a modified phosphoramidate alkylator prodrug
Alk-Triggermod upon reduction of the bioreductive group (Z3). When reduction of the
bioreductive group forms a modified phosphoramidate alkylator prodrug, the linker (L)
bonded to the phosphoramidate alkylator can undergo degradation to yield either the
phosphoramidate alkylator or some other modified phosphoramidate alkylator prodrug.
[02401 hi one embodiment, the present invention provides a compound which demonstrates
a bystander effect upon activation in hypoxic tissue by incorporating a linker (L) as described
above. In one embodiment, the bystander effect allows a modified phosphoramidate
alkylator of the present invention to diffuse or penetrate into tumor zones which are not
hypoxic enough to activate the prodrug compounds of the invention but reside nearby the
hypoxic tumor zone which can activate these prodrugs.

[0241] Upon reduction of the bioreductive group (Z3) within the Trigger T is modified to
Zs-mod, to yield a modified phosphoramidate alkylator prodrug such as phosphoramidate
alkylator- TM or Alk- TM conjugate. In one embodiment the TM is selected from:
[C(Z1)2-y3]-(C(=O)-o)-[C(Z1)2-Z2-Y4]-[C(z1)2]z-[C(z,>=c(z1)]-Z3.mod;
[C(Z1)2-Y3]-[C(Z])2-Z2-Y4]-[C(Z1)2]z-[C(Z1)=C(Z1)]-Z3.mod;
[C(Z])2-Y3]-[C(Z1)2]z-[C(Z1)=C(Z1)]-Z3.mod;
[C(Z1)2-Y3]-[C(Z1)2]z-Z3-mod;
[C(Z1)2-Y3]-(C(=O)-0)-[C(Zl)2]z-[C(Z1)=C(Z1)]-Z3.mod;
[ClZ^-Ysl-CCeO^-lCCZOzlz-ZB-nKx.;
[C(Z1)2-Y3]-(C(=O)-0)-[C(Z1)2MC(Z1K(Z1)]-Z3.nwd;
[qzOrZrY^ HTO^qz^,)]-^ and
-[qZi)2]z-[C(Z1)=C(Z1)]-Z3.mo modified Z3.
[0242] In another embodiment, the TM is selected from:
[qZ1)2-Y3]-(CeO)-0)-[C(Z1)2-Z2-Y4]-H; [C(Z1)2-Y3]-[qZ,)2-Z2-Y4]-H; and
[C(Z!)rY3l-H;
[0243] In One embodiment, Trigger T includes the following linkers (L) having the
formula:

-[C(Z1)2-Y3]-(C(=O)-0)-[C(Z1)2-Z2-Y4]-[C(Z1)2]z-[C(Z1)=C(Z!)]-;
-[C(Z,>.-Y3]-[C(Z1)2-Z2-Y4]-[C(Z1)2]2-[C(Z1)=C(Z1)]-;
-[C(Z1)2-Y3]-[C(Z1)2]2-[C(Z1)=C(Z1)]-;-[C(Z1)2-Y3]-[C(Z1)2]z-;
-[C(Z1)2-Y3]-(C(=O)-0)-[C(Z1)2]z-[C(Z1)=C(Z1)]-;-[C(Z1)2-Y3]-(C(=O)-0)-C(Z1)2-;
-[C(Z1)2-Y3]-(C(=O)-0) -[qzOzMCxzo^xz,)]-; md
-[C(Z!)2-Z2-Y4] -[C(Z1)2]Z-[C(Z1)=C(Z1)]-; -[C(Z1)2]2-[C(Z1)=C(Z1)]- and -[C(Z1)2]Z-.
[0244] In one embodiment, the present invention provides a Trigger T which upon
bioreduction is modified to TriggerMod or TM and the phosphoramidate alleviator is separated
from TM in less than 0.1 second. In another embodiment, the phosphoramidate alkylator is
separated from TM in between 0.01 to 0.10 second. In another embodiment, the
phosphoramidate alkylator is separated from TM in between 0.1 to 1.0 second. In another
embodiment, the active phosphoramidate is separated from TM in between 1.0 to 10.0
seconds. In another embodiment, the phosphoramidate alkylator is separated from TM in
between 10.0 to 100.0 seconds.
[0245] In a related embodiment, upon activation or reduction, a phosphoramidate alkylator
prodrug yields a prodrug with a modified Trigger T (TM) which subsequently releases the
phosphoramidate alkylator 20 to 500 um from the site of activation or reduction; or 20 to 100
um from the site of activation or reduction. Bystander effect of a phosphoramidate alkylator
prodrug of the present invention can be measured using cellular spheroids and multilayer
cellular assay (for example of such assays see Kyle et al, Cancer Res. 2004,64(17):6304-9
and West et al, Cancer Chemother. Pharmacol., 1987, 20(2): 109-14); and as described in
greater detail in Examples 35 and 37. Tumor cells can be grown in culture as multicellular
spheroids to create an in vitro model of the tumor microenvironment in solid tumors
containing a hypoxic region and a quiescent cell population responding to the environmental
stresses of limited nutrients and increased waste production. These spheroids have the unique
property of developing gradients of oxygen and nutrients as the aggregate of cells continue to
divide and grow outward. After the viable rim reaches approximately 150 um in size, a
hypoxic region develops, that drives the cells in this region into a quiescent state and
eventually to cell death. A necrotic core develops as a result of the dying cells. The spheroid
can be divided into 4 distinct compartments for modeling the effectiveness of a hypoxic
activated prodrug: 1) the outer aerobic and actively dividing region; 2) a region of
intermediate hypoxia; 3) a region of hypoxia where cells are not cycling; 4) and a necrotic
core containing dead cells and cellular debris. The response of a drug will depend on a

number of factors; the ability of compound to penetrate into the deepest regions of the
spheroid. The activation of hypoxic activated prodrug (HAP) by nitroreductases; the
reactivity of the activated drug in the cell in which it was activated; and the ability of the
activated drug to leave the site from where it was activated and kill nearby cells (bystander
effect). The assessment of the effectiveness of a compound can therefore be evaluated on a
number of different levels. The effect of the compound alone can be compared to cells in
monolayer culture versus intact spheroids. The HAP can used as a monotherapy. The
hypoxic fraction of the spheroid can be modulated by varying the concentration of O2 of the
equilibrating gas and therefore change the ratio of the aerobic and hypoxic compartments.
HAP's can be combined with other chemotherapeutic agents that either target only the outer
aerobic cells or are able to target the entire spheroid. The expected cell kill can be predicted
by knowing the hypoxic fraction and the expected cell kill of each of the monotherapies.
[0246] In one embodiment, the present invention provides a phosphoramidate alkylator
prodrug which upon activation such as bioreduction releases the phosphoramidate alkylator
with ahalf life of less than 0.1 second; between 0.01 Jo 0.10 second, between 0.1 to 1.0
second, between 1.0 to 10.0 seconds, and between 10.0 to 100.0 seconds.
[0247] Anti cancers drugs can bind to tissue surrounding the vasculature and/or have high
molecular weights that impede diffusion and not reach in therapeutically effective
concentrations hypoxic tumor zones that can be up to 150 - 200 uM away from the
vasculature. In one embodiment, the present invention provides phosphoramidate alkylator
prodrugs that can reach hypoxic cancer cells away from the vasculature. Some methods for
determining the bystander effect are described in greater detail in Examples 35 and 37. The
phosphoramidate alkylator used in a hypoxia activated prodrug plays an important role to
efficiently kill tumor cells. For example, for a hypoxia activated phosphorarnidate alkylator
prodrug, the cytoxicity of the phosphoramidate alkylator and its rate of cellular alkylation,
and the cell membrane permeability of the prodrug and the phosphoramidate alkylator impact
the hypoxic selectivity and hypoxic cytotoxicity of the phosphoramidate alkylator prodrug.
[02481 In one embodiment, the present invention provides phosphoramidate alkylator
prodrugs that are safer than the corresponding phosphoramidate alkylators formed in vivo (at
least ten and up to one million-fold safer.. In one embodiment, the increased safety results
from a modification at the site of attachment of the Trigger T (activation of the
phosphoramidate alkylator prodrug releases the alkylator/cytotoxic agent). In either event,
the phosphoramidate alkylator prodrugs are converted into the corresponding alkylator in

hypoxic tissues by virtue of the activation or reduction of the bioreductive group (Z3),
resulting in its removal and the concomitant or subsequent release or generation of the
phosphoramidate alkylator.
[0249] In one embodiment, the Trigger T is covaleotly bonded to the phosphoramidate
alkylator, in a manner that masks or reduces the cytotoxic activity of the phosphoramidate
alkylator. This masking effect can vary and can depend on the cytotoxic activity of the
phosphoramidate alkylator. Typically, the phosphoramidate alkylator prodrug will show at
least about 10 fold less cytotoxic activity than the corresponding phosphoramidate alkylator,
and can show up to about a million fold or less cytotoxic activity. In one version, the
cytotoxic activity of the phosphoramidate alkylator prodrug is about 100 fold to about 10,000
fold less than the cytotoxic activity of the corresponding phosphoramidate alkylator. As one
example, for a phosphoramidate alkylator with an IC50, IC90, or LC50 of 1 nM, the IC50, IC90,
or LC50 of the corresponding phosphoramidate alkylator prodrug can be 1 uM or greater.
[0250] In one version, compounds provided herein include as phosphoramidate alkylator
prodrug, any phosphoramidate alkylator that can be linked to a Trigger T in a manner that
yields a phosphoramidate alkylator prodrug that is at least about 10-fold to about 1,000,000-
fold, and typically about 100 to about 10,000-fold, less active as a cytotoxic agent than the
corresponding phosphoramidate alkylator or modified phosphoramidate alkylator that is
released from the compounds under hypoxic conditions.
[0251] To determine if a phosphoramidate alkylator prodrug is selectively active under
anoxic or hypoxic conditions, cells are exposed to the drug either with air (normoxic) or
without oxygen (anoxia) or with very little oxygen (hypoxia). One of skill in the art will
recognize that cytotoxicity of a phosphoramidate alkylator prodrug as measured in an anti-
piouferation assay is expressed by the IC50; and Hie cytotoxicity of a phosphoramidate
alkylator prodrug as measured in a clonogenic survival experiment is expressed as IC10 or
LCio, IC90 or LC90, or IC99 or LC99. The ratio of cytotoxicity as measured for example by
IC50, IC90, LC50, LCoo, or LC99 determined in normoxia and hypoxia is called hypoxia
cytotoxicity ratio (HCR) and can be a measure of the hypoxia selective cytotoxicity of the
prodrugs of the present invention. The larger the HCR of the phosphoramidate alkylator
prodrug the higher is its hypoxic cell selective toxicity and greater the hypoxic tumor killing
ability of the prodrug relative to healthy normoxic cells. The HCR determined based on IC99
or LC99 is larger than; that determined based on IC90 or LC90.

[0252] In a related embodiment, the phosphoramidate alkylator prodrug of the present
invention has a hypoxic cytotoxicity of 0.1 nM to 50 uM and a HCR of 10 to 100,000. In a
related embodiment, the phosphoramidate alkylator prodrug of the present invention has a
hypoxic cytotoxicity of 0.1 nM to 50 uM and a HCR of 25 to 100,000 (see EXAMPLE
section). In another related embodiment, the phosphoramidate alkylator prodrug of the
present invention has a hypoxic cytotoxicity of 0.1 nM to 5 uM and a HCR of 50 to 10,000
such as, for example, the compounds as described in Examples 29,30 and 31.
[0253] In one embodiment, the present invention provides a phosphoramidate alkylator
prodrug having hypoxic toxicity which is 5 to 1,000,000 folds more man the corresponding
normoxic toxicity. In another embodiment, me present invention provides a
phosphoramidate alkylator prodrug having hypoxic toxicity which is 10 to 10,000 folds more
than the corresponding normoxic toxicity. In another embodiment, the present invention
provides a phosphoramidate alkylator prodrug having hypoxic toxicity which is 25 to 5,000
folds more than the corresponding normoxic toxicity.
[0254] Tumors have a gradient of oxygen concentration that can vary from 10%, in tissues
adjacent to the vasculature, to 0.5% in tissues about 150 uM away, and lower in tissues
further away from the vasculature and near the necrotic core. In one embodiment, the present
invention provides phosphoramidate alkylator prodrugs that can generate phosphoramidate
alkylators, 5-1,000,00; 10-10,00; and 25-5,000 folds more toxic than the corresponding
prodrug, under a variety of oxygen concentrations. In one embodiment, the present invention
provides phosphoramidate alkylator prodrugs generate phosphoramidate alkylators, 5-
1,000,00; 10-10,00; and 25-5,000 folds more toxic than the corresponding prodrug, under
about 0.5-0.6% oxygen concentrations.
[025S] The logP of a phopsphramidate alkylator prodrug of the present invention can
measure the lipophilicity or the hydrophilicity of the prodrug. In one embodiment, the
present invention provides a phosphoramidate alkylator prodrug having a logP less than 0.
Such phosphoramidate alkylator prodrugs can be hydrophilic, such as a prodrug having
formula XV wherein each Ru is H and can be easily formulated as an aqueous formulation
for i.v. or i.p. injection. Another example of such prodrugs are compounds 24,25 and 36.
[0256] In one embodiment, the present invention provides a phosphoramidate alkylator
prodrug having a logP greater than 0. In one embodiment, the present invention provides a
phosphoramidate alkylator prodrug having a logP between 0 and 4 such as those exemplified

by formulas XIV; XX and XV wherein each Rn is methyl or, cyclopropyl, and administered
in a patient can pass the cell membrane to penetrate inside cancer cells. Another example a
prodrug having a logP between 0 and is 5,6,7, or 16. (for measured logP of phopsphramidate
alkylator prodrugs of the present invention see EXAMPLES section).
lib. Method of synthesis
[0257] The present invention arises in part out of the discovery that compound 36, which
could not be isolated by reacting

l-N-methyl-2-nitroimidazole-5-methanol, and n-butyl lithium in a suitable solvent, was
readily synthesized by employing a Mitsunobu-type reaction wherein l-N-methyl-2-
nitroimidazole-5-methanol was activated by the addition of triphenylphosphine and
diisopropyl azodicarboxylate, and reacted with to yield compound 36.
[0258] Thus, in one aspect the present invention provides a method of synthesizing a
phosphoramidate compound comprising reacting a phosphoramidic or a phosphordiamidic
acid and an alcohol to yield a phosphoramidate. In another aspect, the present invention
provides methods of synthesizing the novel phosphoramidate alkylator prodrug compounds
of the invention or those that are known. In one embodiment, the present invention provides
a method of synthesizing a phosphoramidate alkylator prodrug comprising reacting, a novel
or known phosphoramidate alkylator, a Trigger-OH, a trisubstituted phosphine, and a dialkyl
azodicarboxylate to yield a novel or known phosphoramidate alkylator prodrug. In one
embodiment of the method, in a first step the Trigger-OH is reacted with the trisubstituted
phosphine and the dialkyl azodicarboxylate to yield an intermediate, and in a second step, the
phosphoramidate alkylator is added to the intermediate obtained from the first step to yield

the product. Such a Mitsunobu type reaction is particularly suitable for synthesis of novel or
known phosphoramidate alkylator prodrugs or derivatives, Alk-Trigger, wherein Trigger is
L-Z3, wherein Z3 is:

wherein R9 is as defined above.
[0259] In one embodiment, the present invention provides a method of synthesizing a
phosphoramidate alkylator prodrug comprising reacting each of novel or known
phosphoramidate alkylators:


with a Trigger-OH, a trisubstituted phosphide, and a dialkyi azodicarboxylate to yield
respectively,

wherein X4, R5, R7, and R8 are as defined as in formula (I).
[0260] In one embodiment, the present invention provides a method to synthesize a
compound of formula:

comprising reacting (a) a novel or known phosphoramidate alkylator of formula:

wherein R2-R5 are defined as in formula (T) with the proviso that
(i) at least two of Ri-R5 are selected from (he group consisting of 2-haloalkyl, 2-
alkylsulfonyloxyalkyl, 2.-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyloxyalkyl;

(ii) at least one of R1-R5 is selected from the group consisting of 2-haloalkyl, 2-C1-C$
alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-
heteroalkylsulfonyioxyalkyl; and at least one of NR2R3 and NR4R5 is or
(iii) NR2R3 and NR4R5 both together are'
(b) a Trigger-OH wherein Trigger is defined as in Formula (I), a trisubstituted phosphine, and
(c) a dialkyl azodicarboxylate to yield the compound of formula:

10261] In one embodiment, the compound of formula:

is selected from the group consisting of:

$262} In another embodiment, the group of formula:

is selected from the group consisting of:




[0263] In another embodiment, the reaction includes a solvent such as THF, dioxane, a Cr
Ce alkyl acetate, chloroform, dichloromethane, acetonitrile and the like. In another
embodiment, each substituent in the trisubstituted phosphine is independently selected from a
C1-C6 alkyl, C1-Csheteroalkyl, C3-Cg cycloalkyl, heterocyclyl, aryl, heteroaryl, and C1-C6
alkoxy substituent. In another embodiment, Trigger T- is

wherein Xi, X2, Z\, and Z2 are defined as in formula (I).
[0264] In another embodiment, the present invention provides a method to synthesize a
phosphoramidate alkylator prodrug comprising

[0265] (i) reacting in a solvent selected from THF, dioxane, dichloromethane, chloroform,
ethyl acetate, propyl acetate, butyl acetate, or acetonitrile a compoumd of formula:

wherein each Rj 1 is independently hydrogen, cycloprpyl, methyl, ethyl, benzyl, or methoxy;
each R9 is independently hydrogen, methyl, ethyl, propyl, or cyclopropyl; and X4 is halo,
methylsulfonyloxy, phenylsulfonyloxy, 4-methylphenylsulfonyloxy, and 4-
halophenylsulfonyloxy;
(ii) a trisubstituted phosphine selected from triphenylphosphine, tributylphosphine,
tributylphosphite; and
(iii) diethyl or diisopropyl azodicarboxylate;
to yield a product of formula;

[0266] In another embodiment, the present invention provides a method of synthesizing a
compound of formula:

comprising the steps of:

(i) reacting in an aprotic solvent, a Trigger-OH, wherein Trigger is defined as in
Formula (I); a trisubstituted phosphine; and a dialkyl azodicarboxylate to yield an
Intermediate (i);
(ii) reacting the Intermediate (i) obtained from step (i) with a compound of formula

wherein each R9, Rn, and X4 is defined as in Formula (I), to yield the compound of formula:

[0267] In another embodiment, the trisubstituted phosphine is P(Ri2)3 wherein each R12 is
H, C1-C6 alkyl, C1-C6 heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, aryl, or heteroaryl. In
another embodiment, the trisubstituted phosphine is a polymer supported trisubstituted
phosphine. In another embodiment, the trisubstituted phosphine is triphenylphosphine,
tributylphosphine, tripropylphosphine, triethylphosphine, or trimethylphosphine. In another
embodiment, the trisubstituted phosphine is a polymer supported triphenyl phosphine.
Polymer supported uisubsututed phosphines are commercially available, for example, from
Varian Inc. of Palo Alto, California. In another embodiment, the present invention provides a
method of synthesizing the compounds wherein each Rn is hydrogen. In another
embodiment, the present invention provides a method of synthesizing the compounds


[0268] In another embodiment, the present invention provides the method of making a
compound wherein the Trigger selected from the group consisting of:

and Z3 is

[0269] In one embodiment, the present invention provides a method to synthesize a
phosphoramidate alkylator prodrug comprising the steps of:
[0270] (a) remixing POCI3 with a N-2-haloethyl-N-(Ri3)ammonium salt, wherein RJ3 is
hydrogen, C1-C6 alkyl, C1-C6heteroalkyl, C3-Cg cycloalkyl,heterocyclyl, aryl, heteroaryl, to
yield a dichlorophosphoramidate intermediate;
[0271] (b) reacting the dichlorophosphoramidate intermediate in step (a) with a N-2-
haloethyl-N-(Ri3)ammonram salt, wherein R13 is hydrogen, C1-C6 alkyl, C1-C6heteroalkyl,

C3-C8 cycloalkyl, heterocyclyl, aryl, heteroaryl, and a base in a solvent to yield a
monochlorophosphoramidate intermediate; and
[0272] (c) reacting the monochlorophosphoramidate intermediate obtained in step (b) with
Trigger-OH and a base in a solvent to yield the phosphoramidate alkylator prodrug.
[0273] In one embodiment, the dichlorophosphoramidate intermediate of step (a) is
separated from the rest of the reaction mixture before subjecting it to the reaction in step (b).
In another embodiment, the separation is performed by first removing excess POCI3 in vacuo
and then distilling the dichlorophosphoramidate under reduced pressure.
[0274] In one embodiment, the phosphoramidate alkylator prodrug of step (c) is separated
from the rest of the reaction mixture by flash column chromatography on silica gel. In one
embodiment, the base employed in step (b) is a tertiary amine. Suitable tertiary amines
employed in step (b) include trialkyl amines, such as, triethyl amine or
diisopropylemylamine. In one embodiment, the solvent employed in step (b) is
tetrahydrofuran (THF) or dioxane.
[0275] In one embodiment, the monochlorophosphoramidate intermediate of step (b) is
separated from the rest of the reaction mixture by flash column chromatography on silica gel
before subjecting it to the reaction in step (c). In one embodiment, the base useful in step (c)
is lithium, sodium, or potassium hexaalkyldisilazide; sodium or potassium hydride; or lithium
diisopropylamide. In one embodiment, the solvent employed in step (c) is dimethoxyethane,
diglyme, diethylether, or THF.
[0276] In one embodiment, the present invention provides a method to synthesize a
phosphoramidate alkylator prodrug comprising the steps of:
[0277] (a) reacting in a solvent about 1 equivalent each of POCl3, a Trigger-OH, and a base
to yield a dichlorophosphate intermediate; and
[0278] (b) reacting the dichlorophosphate intermediate in step (a) with a N-2-haloethyl-N-
(Ri3)ammomum salt, wherein R13 is hydrogen, C1-C6 alkyl, C1-C6heteroalkyl, C3-Cg
cycloalkyl, heterocyclyl, aryl, heteroaryl, and a base in a solvent to yield the
phosphoramidate alkylator prodrug.

[0279J In one embodiment, steps (a) and (b) are performed at temperatures below 0°C. In
another embodiment, step (b) is performed at a temperature between 20-100°C higher than
the temperature of step (a).
[0280] In another embodiment, the present invention provides a method for synthesizing
heterocyclic phopsphoramidate ankylator prodrugs of the present invention as shown below:

wherein X4 = Br or CI; e = 1-3
[0281] In one embodiment, the present invention provides a method to synthesize a
phosphoramidate alkylator prodrug comprising the steps of:
[0282] (a) reacting PCI3 with a N,N-di(2-haloethyl)ammonium salt and a base in a solvent
to yield a monochlorophosphamide derivative;
[0283] (b) reacting the monochlorophosphamide derivative with Trigger-OH to yield an
intermediate; and
[0284] (c) oxidizing the intermediate in step (b) to yield the phosphoramidate alkylator
prodrug.
[0285] In one embodiment, the base used in step (b) is triethylainine. In another
embodiment, the solventused in step (c) is dimethoxyethane, diglyme, or a C1-C6 alkyl
acetate. In another embodiment, Trigger-OH is step (c) is

[0286] Various l-N-alkyI-2-aminoimidazole-5-carboxylate can be synthesized as described
Qr*hpmfltir.fl11v below:


[0287] The 1 -N-alkyl-2-aminoimidazole-5-carboxylates can be reduced to yield various 1 -
N-alkyl-2-amino-5-hydroxymethylimidazole derivatives employed in the present invention as
bioreductive group Z3.
[0288] The synthetic methods are provided in further detail in the EXAMPLES section
below.
[0289] Synthesis of bioreductive groups and phosphoramidate alkylator prodrugs, and
methods of the present invention can be adapted from the references Matteucci et ah, PCT
Appl. Pub. No. WO 04/009667, and Hypoxia activated produgs US Pat. AppL entitled.
"Hypoxia Activated anti-Cancer Agents"; deGroot et al, 2001, Current Med. Chem. 5:1093-
1122; Denny et al, US Pat. Nos. 5,750,782; 5,780,585; 5,872,129; and 6,251,933; Davis et
al., PCT AppL Pub. Nos. WO 04/85421 and WO 04/85361;'and Lin et al, US Pat. Appl. Pub.
Nos. 2004/254103 and 2005/043244, and Borch et al, {supra).
[0290] Examples of methods to synthesize phosphoramidate alkylator prodrugs of the
present invention are provided in further detail in the "EXAMPLES" section below.
Hla. Methods of Treatment
[0291] In one embodiment, the present invention provides a method of treating cancer in a
patient in need of therapy thereof by administering to the patient a phosphoramidate alkylator
prodrug of the present invention or one that is Known, Known phosphoramidate aikyiators
are provided by the references Borch et al, supra. In one embodiment, the phosphoramidate
alkylator prodrug employed in treating cancer according to the methods provided by the
present invention has the formula selected from (I) - (XXVII). In one embodiment, the
phosphoramidate alkylator prodrug employed in treating cancer according to the methods
provided by the present invention is selected from the compounds exemplified in the
EXAMPLE section.
[0292] Cancer therapy with alkylating agents can lead to development of cancers that are
resistant to these alkylating agents. Alkylating agents can kill cancer cells in the more rapidly

dividing or higher oxygen containing cancer region as compared to the cancer cells in the
slower growing hypoxic cancer region. The latter cells survive the treatment by alkylators
and can produce cells resistant to such alkylators. Increased activity of guanine-O6-
Alkyltransferase, glutathione, glutathione transferases, the nucleotide excision repair
pathway, and/or the mismatch repair proteins, and decreased permeation of actively
transported drugs such as mechlorethamine and melphalan, are postulated to be responsible
for cancer resistance to alkylators (for example, see, Hardman et al, pages 1393 and 1433,
supra).
[0293] The prodrugs of the present invention are effective in treating cancers resistant to
other therapies. Slowly dividing cancer cells in the hypoxic cancer zone act as a source of
resistant cancer cells and strains and are killed by the prodrugs of the present invention. In
one embodiment, the present invention provides a method of treating a cancer resistant to
treatment by one or more alkylators by administering the compounds of the present invention
alone or in combination with another anticancer agent. In one embodiment, a
phosphoramidate alkylator prodrug of the invention is administered in combination with a
drug having substantially no nephrotoxicity. In one embodiment the phosphoramidate
alkylators prodrug is administered with carboplatin.
[0294] In one embodiment, the present invention provides phosphoramidate alkylators
prodrugs which are not cross-resistant with known alkylators. In another embodiment,
present invention provides phosphoramidate alkylators prodrugs which are not cross-resistant
with the alkylators cyclophosphamide, ifosfamide, glufosfamide, mechlorethamine,
melphalan, chlorambucil, dacarbazine, temozolomide, carmustine, streptozocin, bendamustin,
busulfan, thiotepa, cisplatin, carboplatin, and oxaliplatin.
[0295] In one embodiment, the present invention provides a method of treating cancer by
administering as a first line therapy the compounds of the present invention alone or in
combination with other anti-cancer agents. In another embodiment, the present invention
provides a method of treating a metastatic cancer by administering as a first line therapy the
compounds of the present invention alone or in combination with other anti-cancer agents. In
one embodiment, the present invention provides a method of treating cancer by administering
as a second line therapy the compounds of the present invention alone or in combination with
other anti-cancer agents. In one embodiment, the present invention provides a method of
treating cancer by administering as a third line therapy the compounds of the present

invention alone or in combination with other anti-cancer agents. In one embodiment, the
present invention provides a method of treating cancer by administering after a prior
treatment with surgery and/or radiation therapy the compounds of the present invention alone
or in combination with other anti-cancer agents. In one embodiment, the present invention
provides a method of treating cancer, the cancer having relapsed after prior chemotherapy,
sugery, radiation or any combination of them, by administering the compounds of the present
invention alone or in combination with other anti-cancer agents.
[0296] In methods for treating cancer provided by the present invention, an effective
amount of phosphoramidate alkylator prodrugs is administered to the subject. Generally, the
subject can be any human or non-human mammal. The preferred subject is a human subject.
Other particular subjects include but are not limited to non-human primates, dogs, cats, farm
animals and horses, hi one version, the phosphoramidate alkylator prodrug is administered
alone. In one version the phosphoramidate alkylator prodrug is administered in combination
with one or more additional anti-cancer agents. In one version the phosphoramidate alkylator
prodrug is administered in conjunction with a therapeutic cancer treatment, including but not •
limited to surgery and radiation. The phosphoramidate alkylator prodrug will typically be
administered in a pharmaceutical composition. Various pharmaceutical compositions that
can be used are described in the Formulations section infra.
[0297] The phosphoramidate alkylator prodrug and their pharmaceutical compositions can
be used to treat any type of cancer in a subject, particularly in a human subject. Cancers that
can be treated include but are not limited to leukemia, breast cancer, skin cancer, bone
cancer, liver cancer, brain cancer, cancer of the larynx, gallbladder, pancreas, rectum,
parathyroid, thyroid, adrenal, neural tissue, head and neck, stomach, bronchi, kidneys, basal
COll CarCillOina, squamous cell carcinoma of both ulcerating and papillary type, metastatic
skin carcinoma, osteosarcoma, Ewing's sarcoma, veticulum cell sarcoma, myeloma, giant cell
tumor, small-C6ll lung tumor, gallstones, islet cell tumor, primary brain tumor, acute and
chronic lymphocytic and granulocytic tumors, hairy-C6ll tumor, adenoma, hyperplasia,
medullary carcinoma, pheochromocytoma, mucosal neuronals, intestinal ganglioneuromas,
hyperplastic corneal nerve tumor, marfanoid habitus tumor, Wilm's tumor, seminoma,
leiomyomater tumor, cervical dysplasia and in situ carcinoma, neuroblastoma,
retinoblastoma, soft tissue sarcoma, malignant carcinoid, topical skin lesion, mycosis
fungoide, rhabdomyosarcoma, Kaposi's sarcoma, osteogenic and other sarcoma, malignant

hypercalcemia, renal cell tumor, polycythermia vera, adenocarcinoma, glioblastoma
multiforma, leukemias, lymphomas, malignant melanomas, and epidermoid carcinomas.
[0298] The phosphoramidate alkylator prodrug can particularly be used in the treatment of
cancers containing significant areas of hypoxic tissue. Such cancers include but are not
limited to lung cancer, especially non-small cell lung cancer, breast cancer, colon cancer,
head and neck cancer, ovarian cancer, pancreatic cancer, and prostate cancer. Examples of
types of cancers that can be treated with the phosphoramidate alkylator prodrugs of the
invention are provided in the following references, each of which is incorporated in its
entirety herin by reference Tidmarsh et ah, PCT Pat. Appl. No. PCT/US2005/047314 filed on
22 December 2005, and PCT Pat. Appl. entitled "Glufosfamide combination therapy",
Attorney Docket No. 021305-005900PC; and US Pat App. No. 60/760,599 and 60/719,787
and PCT Pat. Pub. No. WO 2005/076888. Several of these cancers are discussed for
illustrative purposes below. Those of skill in the art will appreciate that cancer chemotherapy
often involves the simultaneous or successive administration of a variety of anti-cancer
agents, and as discussed further below, a phosphoramidate alkylator prodrug can bemsed in
combination therapies as provided by the methods described herein. Thus, in the description
of illustrative cancers containing hypoxic regions amenable to treatment with a
phosphoramidate alkylator prodrug, examples of combination therapies are also described.
[0299] Lung cancer affects more than 100,000 males and 50,000 females in the United
States, most of who die within 1 year of diagnosis, making it the leading cause of cancer
death. Current protocols for the treatment of lung cancer involve the integration of
chemotherapy with or without radiotherapy or surgery. A phosphoramidate alkylator prodrug
can be used as a single agent or in combination with existing combination therapies. A
variety of combination chemotherapy regimens have been reported for small cell lung cancer,
including the combinations consisting of cyclophosphamide, doxorubicin and vincristine
(CAV); etoposide and cisplatin (VP-16); and cyclophosphamide, doxorubicin and VP-16
(CAVP-16). Modest survival benefits from combination chemotherapy (etoposide plus
cisplatin) treatment have been reported for non-small cell lung cancer.
[0300] Likewise, several different cytotoxic drugs have produced at least temporary
regression of ovarian cancer. The most active drugs in the treatment of ovarian cancer have
been alkylating agents, including cyclophosphamide, ifosfamide, melphalan, chlorambucil,
tbiotepa, cisplatin, and carboplatin. Current combination therapies for ovarian cancer include

cisplatin or carboplatin in combination with cyclophosphamide at 3- to 4-week intervals for
six to eight cycles. The compounds and methods described herein provide prodrug forms and
methods for treating ovarian cancer in which a phosphoramidate alkylator prodrug as
described herein is used as a single agent or in existing such combination therapy, either to
replace an agent or in addition to the agent(s) currently used.
[0301] Cancer of the prostate is the most common malignancy in men in the United States
and is the second most common cause of cancer death in men above age 55, and this cancer
has been reported to consist primarily of hypoxic tissue. Several chemotherapy protocols
have been reported for use in late stage disease following relapse after hormonal treatment.
Agents for the treatment of prostate cancer include the alkylators estramustine phosphate,
prednimustine, and cisplatin. Combination chemotherapy is also used to treat prostate
cancer, including treatment with estramustine phosphate plus prednimustine and cisplatin,
and 5-fluorouracil, melphalan, and hydroxyurea. The present invention provides methods for
treating prostate cancer in which a phosphoramidate alkylator prodrug of the present
*
invention is used in such combinations, either to replace an agent or in addition to the
agent(s) currently used.
[0302] Cancer of the large bowel is the second most common cause of cancer death in the
United States and is likewise a cancer characterized by hypoxic regions. While
chemotherapy in patients with advanced colorectal cancer has proven to be of only marginal
benefit, 5-fluorouracil is the most effective treatment for this disease. 5-Fluorouracil is useful
alone or in combination with other drugs, but is associated with only a 15 to 20 percent
likelihood of reducing measurable tumor masses by 50 percent or more. Using 5-FU in
combination with the compounds and methods described herein, and the methods for treating
colon cancer using a prodrug can offer significant topftlfa tofit and pvMM M
meeting the unmet need for better treatment methods for this disease.
[0303] In one version of the treatment methods, a phosphoramidate alkylator prodrug can
be used in various known approaches to cancer therapy including but not limited to "anti-
body-directed enzyme prodrug therapy" (ADEPT), "virus-directed enzyme prodrug therapy
(VDEPT), "gene-directed enzyme prodrug therapy" (GDEPT), and "bacteria-directed enzyme
prodrug therapy" (BDEPT). The general uses of a phosphoramidate alkylator prodrug are not
limited to the foregoing treatment methods.

[0304] n another aspect, the present invention provides a method of treatment of non-
cancer hyperproliferative diseases characterized by cellular hyperproliferation (e.g., an
abnormally increased rate or amount of cellular proliferation). In one embodiment, the
hyperproliferative disease treated according to the present method is selected from the group
consisting of allergic angiitis and granulomatosis (Churg-Strauss disease), asbestosis, asthma,
atrophic gastritis, benign prostatic hyperplasia, bullous pemphigoid, coeliac disease, chronic
bronchitis and chronic obstructive airway disease, chronic sinusitis, Crohn's disease,
demyelinating neuropathies, dermatomyositis, eczema including atopic dermatitis, eustachean
tube diseases, giant cell arteritis, graft rejection, hypersensitivity pneumonitis,
hypersensitivity vasculitis (Henoch-Schonlein purpura), irritant dermatitis, inflammatory
hemolytic anemia, inflammatory neutropenia, inflammatory bowel disease, Kawasaki's
disease, multiple sclerosis, myocarditis, myositis, nasal polyps, nasolacrimal duct diseases,
neoplastic vasculitis, pancreatitis, pemphigus vulgaris, primary glomerulonephritis, psoriasis,
periodontal disease, polycystic kidney disease, polyarteritis nodosa, polyangitis overlap
syndrome, primary sclerosing cholangitis, rheumatoid arthritis, serum sickness, surgical
adhesions, stenosis or restenosis, scleritis, scleroderma, strictures of bile ducts, strictures (of
duodenum, small bowel, and colon), silicosis and other forms of pneumoconiosis, type I
diabetes, ulcerative colitis, ulcerative proctitis, vasculitis associated with connective tissue
disorders, vasculitis associated with congenital deficiencies of the complement system,
vasculitis of the central nervous system, and Wegener's granulomatosis.
[0305] In some embodiments of the invention, a compound of the present invention is
administered to treat a hyperproliferative disease selected from the group consisting of
psoriasis, multiple sclerosis, rheumatoid arthritis, restenosis, and benign prostatic hyperplasia.
In one embodiment, the hyperpriliferative disease treated is psoriasis, a disease characterized
by the cellular hyperproliferation of keratinocytes which builds up on the skin to form
elevated, scaly lesions. In another embodiment, the hyperproliferative disease treated is
multiple sclerosis, a disease characterized by progressive demyelination in the brain. In
another embodiment, the hyperproliferative diseases treated is rheumatoid arthritis, a
multisystem chronic, relapsing, inflammatory disease that can lead to destruction and
ankylosis of joints affected. In another embodiment, the compounds of the present invention
are administered to prevent a hyperproliferative disease resulting from cellular proliferation
on a prosthesis implanted in a subject by coating the prosthesis with a composition containing
a compound of the present invention. In another embodiment, the hyperproliferative disease

treated is benign prostatic hyperplasia, a disease in which prostate epithelial cells grow
abnormally and thereby block urine flow.
Hlb. Formulations, modes of administration, dosages
[0306] A phosphoramidate alkylator prodrug will typically be formulated as
pharmaceutical formulations for administration to a subject. Described in this section are
modes of administration, formulations, and dosages that can be used when treating cancers
using a phosphoramidate alkylator prodrug described herein.
[0307] Administration of a phosphoramidate alkylator prodrug for the treatment of cancer
can be effected by any method that enables delivery of the prodrugs to the site of action, the
hypoxic region of a tumor. Many cancer drugs are administered by intravenous injection, and
a phosphoramidate alkylator prodrug can be formulated for such administration, including not
only ready-for-injection formulations but also lyophilized or concentrated formulations that
*
must be rehydrated or diluted, respectively, prior to injection. In addition to these
formulations, a phosphoramidate alkylator prodrug can be formulated for administration by
oral routes, intraduodenal routes, parenteral injection (including intravenous, subcutaneous,
intramuscular, intravascular or infusion), topical, and rectal routes. Those of skill in the art
will recognize that a phosphoramidate alkylator prodrug may be activated by bacteria in the
gut. If such activation is not desired, then the practitioner can employ a route of
administration or a formulation that results in absorption of a phosphoramidate alkylator
prodrug prior to its entry into the large intestine or colon. The actual route of administration
and corresponding formulation of the phosphoramidate alkylator prodrug will depend on the
type of cancer being treated, the phosphoramidate alkylator prodrug selected for
administration, the severity of the cancer, and the age, weight, and condition of the patient,
among other factors.
[0308] The amount of a phosphoramidate alkylator prodrug administered, and thus the
amount of the phosphoramidate alkylator prodrug contained in the dose administered and the
product comprising that dose, will be dependent on the subject being treated, the severity of
the cancer, localization of the cancer, the rate of administration, the disposition of the prodrug
(e.g., molecular weight, solubility and hypoxic and normoxic cytotoxicity), the cytotoxic
agent released by a phosphoramidate alkylator prodrug, and the discretion of the prescribing
physician.

[0309] In one embodiment, the present invention provides a method of cancer treatment in
a patient wherein an effective dosage is typically in the range of about 0.001 to about O.lg per
kg body weight, or about 0.1 to about 35 mg/kg/day in single or divided doses. For a 70 kg
human, this would amount to about 0.05 to about 7 g/day, about 0.2 to about 2.5 g/day. In
some instances, dosage levels below the lower limit of the aforesaid range can be more than
adequate, while in other cases still larger doses can be employed without causing any harmful
side effect; larger doses can also be divided into several small doses for administration
throughout the day by infusion for an hour or continuously using a peripherally inserted
central catheters (PICC line) and portable intravenous bag and pump.
[0310] In one embodiment, the effective dose of a compound of the present invention for
treatement of cancer and other hyperproliferative diseases is in the range of about 0.1 to about
35 mg/kg/day, about 0.5 to about 20 mg/kg/day, about 0.5 to about 15 mg/kg/day; about 0.5
to about 10 mg/kg/day; about 0.5 to about 8 mg/kg/day; and about 1 to about 5 mg/kg/day in
single or divided doses. In one embodiment, the effective dose of a compound of the present
invention for treatement of cancer and other hyperproliferative diseases is in the range of
about 2 to about 8 mg/kg/day; about 2 to about 4 mg/kg/day; and about 2 mg/kg/day in single
or divided doses. In one embodiment, the effective dose of a compound of the present
invention for treatement of cancer and other hyperproliferative diseases is in the range of
about 0.25 to about 2.5 mg/kg/day; about 0.25 to about 1 mg/kg/day; and about 0.25 to about
0.5 mg/kg/day in single or divided doses. In one embodiment, the dose is administered i.v.
daily, either as a monotherapy (compound of the present invention alone) or in conjunction
(combination) with standard of care therapies. In one embodiment, the effective dose for
treatement of cancer and other hyperproliferative diseases is in the range as described earlier
administered once a week.
[0311] In one embodiment, a larger dose is administered intermittently (less frequently); a
dose in the range of about 3 to about 20 mg/kg; about 6 to about 10 mg/kg; or 8 mg/kg is
administered on once every three days for two weeks. In another embodiment, a dose in the
range of about 5 to about 30 mg/kg; about 10 to about 15 mg/kg; or 12.5 mg/kg of the
phosphoramidate alkylator prodrug is administered once a week for four weeks. In one
embodiment, a dose in the range of about 0.5 to about 8 mg/kg/day is administered for 5 days
over two weekly cycles.
[0312] In another embodiment, for treatment of human patients, the maximum daily dose
of a phosphoramidate alkylator prodrug is not greater than 500 mg/kg patient weight and,

accordingly, a phosphoramidate alkylator prodrug is administered in a daily dose in the range
of about 1 mg of a phosphoramidate alkylator prodrug/kg of patient weight to about 500 mg
of a phosphoramidate alkylator prodrug/kg of patient weight. In one embodiment, a
phosphoramidate alkylator prodrug is administered in a daily dose in the range of about 5
mg/kg to about 500 mg/kg of the body weight of the patient to be treated. In another
embodiment, the therapeutically effective dose is a daily dose of a phosphoramidate alkylator
prodrug is about 10 mg/kg to about 250 mg/kg of the body weight of the patient to be treated.
In another embodiment, the therapeutically effective dose of a phosphoramidate alkylator
prodrug is about 25 mg/kg to about 150 mg/kg of the body weight of the patient to be treated.
In another embodiment, the therapeutically effective dose of a phosphoramidate alkylator
prodrug is about 25 mg/kg to about 50 mg/kg of body weight of the patient to be treated. In
another embodiment, the therapeutically effective dose of a phosphoramidate alkylator
prodrug is about 1.25 mg/kg to about 12.5 mg/kg of body weight of the patient to be treated.
[0313] Guidance concerning administration can also be provided by and from studies in
humans and other mammalian animals. A therapeutically effective dose determined for an
animal can be converted to the corresponding human equivalent dose (HED) as described in
the table below:

b For example, cynomolgus, rhesus, or stumptail.

[0314] To achieve therapeutic effectiveness, the therapeutically effective daily dose of a
phosphoramidate alkylator prodrug is usually administered multiple times to the patient. In
one embodiment, a phosphoramidate alkylator prodrug is administered daily for a period of
time. Typically, daily administration for at least 3 consecutive days will be employed. In
related embodiments, administration is for at least 5 consecutive days, at least 7 consecutive
days, or at least 10 consecutive days. Depending on the dose, formulation, and route of
adiainistration selected by the practitioner and the convenience of the patient, the entire daily
dose can be administered once daily, or the daily dose can be administered in multiple
smaller doses through the course of a day (including by infusion with a pump or intravenous
administration). For example, the dose can be divided into two smaller doses and
administered twice daily, or divided into three smaller doses and administered thrice daily. It
will be apparent to one of skill in the art of cancer treatment that, as used herein, "daily"
administration is not limited to one administration per day but can include multiple
administrations.
[0315J Adniinistration schedules other than consecutive daily administration can also be
used. Administration once every other day (qod) is particularly convenient, and
administration once every third day, or once a week can be appropiate in some instances, but
in any event, a phosphoramidate alkylator prodrug is repeatedly administered over a period of
time. For example, whether administration is daily (including, as noted, a divided daily
dose), every other day, or less frequently, in one embodiment a phosphoramidate alkylator
prodrug is administered at least 2 days per week for at least two, three, four, five or at least
six consecutive weeks, or, alternatively, for at least two, three, four, five or at least six weeks
within a six-month period, or, alternatively, for at least two, three, four, five or at least six
weeks within a twelve-month period. In one embodiment, a phosphoramidate alkylator
prodrug is administered at least 3 days per week for at least two, three, four, five or at least
six consecutive weeks, or, alternatively, for at least two, three, four, five or at least six weeks
within a six-month period, or, alternatively, for at least two, three, four, five or at least six
weeks within a twelve-month period. In one embodiment a phosphoramidate alkylator
prodrug is administered at least 10 days per month,- optionally at least 20 days per month, for
at least one month or at least two, three, four, five or at least six consecutive months, or,
alternatively, at least one, two, three, four, five or at least six months in a 6-month period.
[0316] In one embodiment, the administration of the therapeutically effective dose is
continued for multiple days, typically for at least three consecutive days, and often for at least

five to ten consecutive days, or for a week, or for several weeks or more. Thus, a patient can
be administered a phosphoramidate alkylator prodrug in accordance with the present methods
for several days, a week, a month, two months, three months, six months, or a year or longer.
[0317] Consistent with administration regimens of other anticancer agents, a
phosphoramidate alkylator prodrug can be administered in multiple "rounds" of
administration. For example, in some embodiments, a phosphoramidate alkylator prodrug
can be administered once daily for at least three to ten, or at least five to ten consecutive days,
and such three to ten or five to ten day treatments can be repeated once, twice, or three or
more times, sometimes with a no-treatment (with a phosphoramidate alkylator prodrug)
period ranging from one to several weeks between each multiple-day treatment Similarly, in
some embodiments, a phosphoramidate alkylator prodrug is adrninistered every other day for
two to ten administrations, more often three to ten administrations, or five to ten
administrations, and such two, three or five to ten administrations qod can be repeated once,
twice, or three or more times with a no-treatment (with a phosphoramidate alkylator prodrug)
period ranging from one to several weeks between each* multiple-day treatment. Other
multiple-round schedules for administration will be apparent to the skilled practicioner
quided by this disclosure.
[0318] In one aspect, "administering a therapeutically effective dose or regimen of a
phosphoramidate alkylator prodrug" refers to (i) administering a phosphoramidate alkylator
prodrug in the ranges stated (e.g., 1 mg to 1 g of a phosphoramidate alkylator prodrug per kg
of patient weight, typically 25 to 150 mg of a phosphoramidate alkylator prodrug per kg of
patient weight) for a specified minimum number of days within a specified time period,
wherein the administration of a phosphoramidate alkylator prodrug has a therapeutic effect on
the cancer in the patient. Illustrative therapeutically effective dose regimens for a
phosphoramidate alkylator prodrug include those described herein, such as administration of
a phosphoramidate alkylator prodrug for 3 consecutive days, 5 consecutive days, 7
consecutive days, 10 consecutive days, at least 3 days per week, at least 3 days per week for
one month, at least 10 days per month, and at least 20 days per month.
[0319] In optimizing a phosphoramidate alkylator prodrug treatment regimen according to
the present invention, the dose and frequency of a phosphoramidate alkylator prodrug
administration can be selected to achieve a maximal sustained area under the plasma
concentration curve (AUC) over the course of treatment. The theoretically optimal dosing

regimen will result in a maximal exposure of the tumor cells to a phosphoramidate alkylator
prodrug, as measured by AUC, while minimizing the maximal plasma concentration (Cmax)
for any single administration. A higher Cmax will contribute to toxicity while the AUC will
determine efficacy. As is understood in the art for other cancer therapeutic drugs, treatment
with a phosphoramidate alkylator prodrug can be suspended temporarily if toxicity is
observed, or for the convenience of the patient, without departing from the scope of the
invention, and then resumed.
[0320] In one embodiment, the pharmacokinetics of the phosphoramidate alkylator prodrug
of the present invention employed for the treatment of cancer can determine the dose, the
method of administration, and the kind of cancer that is treated with the phosphoramidate
alkylator prodrug. In one embodiment, the phosphoramidate alkylator prodrug of the present
invention can have a in vivo half life of between 1 to 300 minutes. In one embodiment, the
compounds of the present invention can have a in vivo half life of between 3 to 10 minutes.
In one embodiment, the compounds of the present invention can have a in vivo half life of
between 10 to 30 minutes. A short half life of the phosphoramidate alkylator prodrug can
require an infusion time in treatment that is longer than that required for a phosphoramidate
alkylator prodrug having a longer half life. A short half life of the phosphoramidate alkylator
prodrug can increase the maximum tolerated dose (MID) for that prodrug.
[0321] In another embodiment, the present invention provides phosphoramidate alkylator
prodrugs that remain up to 20% unchanged when incubated with mouse liver microsomal
(update with human example and data if available) protein for 30 minutes. In another
embodiment, the present invention provides phosphoramidate alkylator prodrugs that remain
20-80% unchanged when incubated with mouse liver microsomal protein for 30 minutes. In
another embodiment, the present invention provides phosphoramidate alkylator prodrugs that
remain greater than 80% unchanged when incubated with mouse liver microsomal protein for
30 minutes. In another embodiment, examples of phosphoramidate alkylator prodrugs of the
present invention which when incubated with mouse liver microsomal protein for 30 minutes
remain greater than 80% unchanged include 1,25, and 36. The higher the MLM stability of a
prodrug of the invention, its therapeutically effective dose and undesirable patient side effects
will be lower.
[0322] In a related embodiment, the bioreductive group of the phosphoramidate alkylator
prodrugs of the present invention upon reduction/activation in a hypoxic tumor zone form a

phosphoramidate alkylator- TM conjugate. The phosphoramidate alkylator- TM conjugate can
diffuse and reach other parts of the tumor or other tumors in the case of a metastatic disease.
Various pharmacokinetic parameters such as volume of distribution under steady state (Vss),
clearance (CL), area under curve (AUC), mouse liver microsomal stability (MLM stability),
plasma stability, and Cmax of phosphoramidate alkylator prodrugs of the present invention
were measured and listed in the EXAMPLES section (see also Hardman et al., supra).
[0323] In re-treatment regimens, the dose can be adjusted to reflect patient tolerance of the
prior treatment. In any event, as toxicity is observed during repeat administration, dosing can
be temporarily stopped as severe symptoms are observed. The period of temporary halting of
administration (drug holiday) can be ended at the time when the first organ of toxicity no
longer contains significant concentrations of a phosphoramidate alkylator prodrug or a
phosphoramidate alkylator released therefrom (which can be measured or determined
indirectly by cessation of symptoms). Therefore, an intermittent dosing period can be
defined not only by specific days but individualized by drug holidays that are based on
symptoms and normal organ clearance of a phosphoramidate alkylator prodrug or a
phosphoramidate alkylators released therefrom.
[0324] A formulation of a phosphoramidate alkylator prodrug can, for example, be in a
form suitable for oral administration as a tablet, capsule, pill powder, sustained release
formulation, solution, and suspension; for parenteral injection as a sterile solution, suspension
or emulsion; for topical administration as an ointment or cream; and for rectal administration
as a suppository. A formulation of a phosphoramidate alkylator prodrug can be in unit
dosage forms suitable for single administration of precise dosages and will typically include a
conventional pharmaceutical carrier or excipient.
[0325] Suitable pnarmaceutlcal carriers include inert diluents or fillers, water and various
organic solvents. The pharmaceutical compositions can, if desired, contain additional
ingredients such as flavorings, binders, excipients, and the like. Thus for oral administration,
tablets containing various excipients, such as citric acid can be employed together with
various disintegrants, such as starch, alginic acid, and certain complex silicates, and with
binding agents such as sucrose, gelatin and acacia. Additionally, lubricating agents such as
magnesium stearate, sodium lauryl sulfate, and talc can be used to prepare the tablet forms of
formulations of a phosphoramidate alkylator prodrug described herein. Solid compositions of
a similar type can be employed in soft and hard filled gelatin capsules. Preferred materials,

therefore, include lactose or milk sugar and high molecular weight polyethylene glycols.
When aqueous suspensions or elixirs are desired for oral administration, the prodrug therein
can be combined with various sweetening or flavoring agents, coloring matters or dyes and, if
desired, emulsifying agents or suspending agents, together with diluents such as water,
ethanol, propylene glycol, glycerin, or combinations thereof.
[0326] Exemplary parenteral administration forms include solutions or suspensions of a
phosphoramidate alkylator prodrug in sterile aqueous solutions, for example, aqueous
polyethylene glycols, propylene glycol or dextrose solutions. Such dosage forms can be
suitably buffered, if desired.
[03271 Methods of preparing various pharmaceutical compositions with a specific amount
of active drug are known, or will be apparent, to those skilled in this art in view of this
disclosure. For examples, see Remington's Pharmaceutical Sciences, Mack Publishing
Company, Philadelphia, Pa., 17th Edition (1984).
[0328] The methods of cancer treatment employing a phosphoramidate alkylator prodrug of
the present invention are effective in killing the most difficult to kill cancer cells growing in
the hypoxic region of a tumor. Once released in the hypoxic region a phosphoramidate
prodrug can diffuse from the hypoxic cells and kill the cancer cells in adjacent regions
containing increasing populations of rapidly dividing cells. The hypoxic region acts as a
drug-factory to produce within a tumor an alkylator for killing adjacent normoxic cancer cells
leading to a higher concentration of the phosphoramidate alkylator within the tumor, relative
to normal tissues. The use of the prodrug to generate the phosphoramidate alkylator within
the tumor can reduce toxic side-effects arising due to normal cell toxicity. After cancer cell
in the normoxic region of the tumor are destroyed, a hypoxic region can become normoxic
and start to divide. At this point, such cells can be killed by the phosphoramidate alkylators
generated from a phosphoramidate alkylator prodrug of this invention or those known, or by
other anticancer agents or cytoxins administered in combination with the phosphoramidate
alkylator prodrug, as described in the following section.
IHc. Combination therapies
[0329] In accordance with the methods of the invention, a phosphoramidate alkylator
prodrug can be co-administered in combination with other anti-cancer agents ("anticancer

agent"). Without intending to be bound by any particular mechanism or effect, such co-
administration can in some cases provide one or more of several advantages over known
cancer therapies, such as, for example co-administration of a phosphoramidate alkylator
prodrug and the anticancer agent has a synergistic effect on induction of cancer cell death.
Co-administration provides a better therapeutic result than administration of the anticancer
agent alone, e.g., greater alleviation or amelioration of one or more symptoms of the cancer,
dinxinishment of extent of disease, delay or slowing of disease progression, amelioration,
palliation or stabilization of the disease state, partial or complete remission, prolonged
survival or other beneficial therapeutic results.
[0330] The co-administration of a phosphoramidate alkylator prodrug increases the
sensitivity of cancer cells to the anticancer agent, allowing lower doses of the anticancer
agent to be adrninstered to the patient or allowing an anticancer agent to be used for treatment
of cells otherwise resistent to the anticancer agent or otherwise refractory to treatment While
the known anti-cancer agents in general targets the rapidly dividing cells in the normoxic
region, the phosphoramidate alkylator prodrugs of the invention target the hypoxic cells in
the regions of tumors that are not efficiently killed by the anticancer agent alone.
[0331] As used herein, a phosphoramidate alkylator prodrug is "co-administered" with
another anticancer agent (also referred to herein as, "Agent") when a phosphoramidate
alkylator prodrug and Agent are administered as part of the same course of therapy. In one
embodiment, a phosphoramidate alkylator prodrug is first administered prior to
administration of the Agent, (i.e., the initiation of the other cancer therapy), and treatment
with a phosphoramidate alkylator prodrug is continued throughout the course of
administration of the Agent (i.e., the course of the other therapy). In another embodiment, a
phosphoramidate alkylator prodrug is administered after the initiation or completion of the
other cancer therapy. In other embodiments, a phosphoramidate alkylator prodrug is first
administered contemporaneously with the initiation of the other cancer therapy. See for
example combination therapies as described in EXAMPLE section.
[0332] In one embodiment, a phosphoramidate alkylator prodrug is first administered prior
to administration of the Agent, and treatment with a phosphoramidate alkylator prodrug is
continued after the cessation of administration of the Agent. In one embodiment, a
phosphoramidate alkylator prodrug is first administered prior to administration of the Agent,
and treatment with a phosphoramidate alkylator prodrug is continued during part of the

period of administration of the Agent. For certain drugs, such as certain topoisomerase
inhibitors, a phosphoramidate alkylator prodrug administration can be initiated and
completed prior to the administration of the second drug.
(0333] In the presence of oxygen, the radical anion formed upon the reduction of Z3 reacts
with oxygen to yield superoxide and Z3. Superoxide is a cytotoxin and the production of
superoxide in normoxic tissues can lead to unwanted side effects. In one embodiment, the
present invention provides a phosphoramidate alkylator prodrug administered in combination
with a chemoprotective agent or a chemoprotectant Chemoprotective agents protect healthy
tissue from the toxic effects of anticancer drugs. In one embodiment, the chemoprotective
agent is a thiol or a disulfide. In one embodiment, the chemoprotectant can reduce
superoxide, m another embodiment, the the chemoprotectant can react with the "Michael-
receptor" generated from a phosphoramidate alkylator prodrug and prevent "Michael-
receptor" from reacting with proteins and nucleic acid (see below).
«
[0334] Anticancer drug therapy today typically involves multiple rounds, or "cycles," of
administration of the anti-cancer agent(s). In the context of administering a phosphoramidate
alkylator prodrug, each cycle of administration (as well as a complete set of cycles) can be
viewed as administration of a second drug. A phosphoramidate alkylator prodrug can be
administered in any or all of the multiple cycles of treatment with the other Agent; in general,
a phosphoramidate alkylator prodrug is administered on a daily basis for at least two or more
days during each cycle. In one aspect of the invention, a phosphoramidate alkylator prodrug
is co-administered with the Agent according to a schedule repeated at each round.
[0335] In one version of the method of treating cancer using the a phosphoramidate
alkylator prodrug, a phosphoramidate alkylator prodrug is administered in combination with
an effective amount of one or more chemotherapeutic agents, an effective amount of
radiotherapy, an appropriate surgery procedure, or any combination of such additional
therapies.

[0336] When a phosphoramidate alkylator prodrug is used in combination with one or more
of the additional therapies, a phosphoramidate alkylator prodrug and additional therapy can
be administered at the same time or can be administered separately. For example, if a
phosphoramidate alkylator prodrug is administered with an additional chemotherapeutic
agent, the two agents can be administered simultaneously or can be administered sequentially
with some time between administrations. One of skill in the art will understand methods of
administering the agents simultaneously and sequentially and possible time periods between
administration. See for example combination therapies as described in the EXAMPLE
section.
[0337] The Agents can be administered as the same or different formulations and can be
administered via the same or different routes.
[0338] Chemotherapeutic agents mat can be used in combination with the a
phosphoramidate alkylator prodrug of the invention include, but are not limited to, busulfan,
improsulfan, piposulfan, benzodepa, carboquone, 2-deoxy-D-glucose, lonidamine and
analogs thereof (refrence apps), glufosfamide, gemcitibine, erlotinib, meturedepa, uredepa,
altretamine, imatinib, triethylenemelamine, triemylenephosphoramide,
triethylenethiophosphoramide, trimethylolomelamine, chlorambucil, chlornaphazine,
estramustine, ifosfamide, gefitinib, mechlorethamine, mechlorethamine oxide hydrochloride,
melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard,
carmustine, chlorozotocin, fotemustine, nimustine, ranimustine, dacarbazine, mannomustine,
mitobronitol, mitolactol, pipobroman, aclacinomycins, actinomycin F(l), anthramycin,
azaserine, bleomycin, cactinomycin, carubicin, carzinophilin, chromomycin, dactinomycin,
daunorubicin, daunomycin, 6-diazo-5-oxo-l-norleucine, mycophenolic acid, nogalamycin,
olivomycin, peplomycin, plicamycin, porfiromycin, puromycin, streptonigrin, streptozocin,
tubercidin, ubenimex, zinostatin, zorubicin, denopterin, pteropterin, trimetrexate, fludarabine,
6-mercaptopurine, thiamiprine, tbioguanine, ancitabine, azacitidine, 6-azauridine, carmofur,
cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, 5-fmorouracil, tegafur, L-
asparaginase, pulmozyme, aceglatone, aldophosphamide glycoside, aminolevulinic acid,
amsacrine, bestrabucil, bisantrene, carboplatin, defofamide, demecolcine, diaziquone,
elfornitbine, elliptinium acetate, etoglucid, flutamide, gallium nitrate, hydroxyurea,
interferon-alpha, interferon-beta, interferon-gamma, interleukin-2, lentinan, mitoguazone,
mitoxantrone, mopidamol, nitracrine, pentostatin, phenamet, pirarubicin, podophyllinic acid,
2-ethylhydrazide, procarbazine, razoxane, sizofiran, spirogermanium, paclitaxel, tamoxifen,

erlotonib, teniposide, tenuazonic acid, triaziquone, 2,2,,2"-trichlorotriethylamine, urethan,
vinblastine, cyclophosphamide, and vincristine. Other chemotherapeutic agents that can be
used include platinum derivatives, including but not limited to cis platinum, carboplatin, and
oxoplatin.
[0339] In one version, a phosphoramidate alkylator prodrug described herein can be used in
combination with an antiangeogenisis inhibitor including but not limited to Avastin and
similar therapeutics. In one version of the combination treatment methods, a subject is
treated with an antiangeogenisis inhibitor and subsequently treated with a phosphoramidate
alkylator prodrug. In one version of the combination treatment methods, a subject is treated
with an antiangeogenisis inhibitor and subsequently treated with a phosphoramidate alkylator
prodrug with another chemotherapeutic agent, including but not limited to Cisplatin, and
carboplatin. In one version of these combination methods of treatment using an
antiangeogenisis inhibitor, the method is used to treat breast cancer.
[0340J In another embodiment, a phosphoramidate alkylator prodrug is administered with
an anti-cancer agent that acts, either directly or indirectly, to inhibit the epidermal growth
factor or EGFR receptor. EGFR inhibitors suitable for coadministration with a
phosphoramidate alkylator prodrug of the invention include gefitinib and erlotonib.
[0341] In another version, a phosphoramidate alkylator prodrug is administered with an
anti-cancer agent that acts, either directly or indirectly, to inhibit hypoxia-inducible factor 1
alpha (HIF1 a) or to inhibit a protein or enzyme, such as a glucose transporter or VEGF,
whose expression or activity is increased upon increased HIFla levels. HIFla inhibitors
suitable for use in this version of the methods and compositions described herein include PI 3
kinase inhibitors; LY294002; rapamycin; histone deacetylase inhibitors such as [(E)-
(lS,4S)10S,21R)-7-[(Z)-ethylidene]-4,21-diisopropyl-2-oxa-12,13-dithia-5,8,20,23-
tetraazabicyclo-[8,7,6]-tricos-16-ene-3,6s9,19,22-pentanone (FR901228, depsipeptide); heat
shock protein 90 (Hsp90) inhibitors such as geldanamycin, 17-aIlylarnino-geldanamycin (17-
AAG), and other geldanamycin analogs, and radicicol and radicicol derivatives such as
KF58333; genistein; indanone; staurosporin; protein kinase-1 (MEK-1) inhibitors such as
PD98059 (2*-ammo-3'-methoxyflavone); PX-12 (1-methylpropyl 2-imidazolyl disulfide);
pleurotin PX-478; quinoxaline 1,4-dioxides; sodium butyrate (NaB); sodium nitropurruside
(SNP) and other NO donors; microtubule inhibitors such as novobiocin, panzem (2-
methoxyestradiol or 2-ME2), vincristines, taxanes, epothilones, discodermolide, and

derivatives of any of the foregoing; coumarins; barbituric and thiobarbituric acid analogs;
camptothecins; and YC-1, a compound described in Biochem. Pharmacol., 15 Apr 2001,
67(8):947-954, incorporated herein by reference, and its derivatives.
[0342] In another version, a phosphoramidate alkylator prodrug is administered with an
anti-angiogenic agent, including but not limited to anti-angiogenic agents selected from the
group consisting of angiostatin, an agent that inhibits or otherwise antagonizes the action of
VEGF, batimastat, captopril, cartilage derived inhibitor, genistein, endostatin, interleukin,
lavendustin A, medroxypregesterone acetate, recombinant human platelet factor 4, Taxol,
tecogalan, thalidomide, thrombospondin, TNP-470, and Avastin. Other useful angiogenesis
inhibitors for purposes of the combination therapies provided by the present methods and
compositions described herein include Cox-2 inhibitors like celecoxib (Celebrex), diclofenac
(Voltaren), etodolac (Lodine), fenoprofen (Nalfon), indomethacin (Indocin), ketoprofen
(Orudis, Oruvail), ketoralac (Toradol), oxaprozin (Daypro), nabumetone (Relafen), sulindac
(Clinoril), tolmetin (Tolectin), rofecoxib (Vioxx), ibuprofen (Advil), naproxen (Aleve,
Naprosyn), aspirin, and acetaminophen (Tylenol).
[0343] In addition, because pyruvic acid plays an important role in angiogenesis, pyruvate
mimics and glycolytic inhibitors like halopyruvates, including bromopyruvate, can be used in
combination with an anti-angiogemc compound and a phosphoramidate alkylator prodrug to
treat cancer. In another version, a phosphoramidate alkylator prodrug is administered with an
anti-angiogenic agent and another anti-cancer agent, including but not limited to a cytotoxic
agent selected from the group consisting of alkylators, Cisplatin, Carboplatin, and inhibitors
of microtubule assembly, to treat cancer.
[0344] In addition to the combination of a phosphoramidate alkylator prodrug with the
Agents described above, the present methods and compositions described herein provides a
variety of synergistic combinations of a phosphoramidate alkylator prodrug and other anti-
cancer drugs. Those of skill in the art can readily determine the anti-cancer drugs that act
"synergistically" with a phosphoramidate alkylator prodrug as described herein. For
example, the reference Vendetti, "Relevance of Transplantable Animal-Tumor Systems to the
Selection of New Agents for Clinical Trial," Pharmacological Basis of Cancer
Chemotherapy, Williams and Wilkins, Baltimore, 1975, and Simpson Herren et al., 1985,
"Evaluation of In Vivo Tumor Models for Predicting Clinical Activity for Anticancer Drugs,"

Proc. Am. Assoc. Cancer Res. 26: 330, each of which is incorporated herein by reference,
describe methods to aid in the determination of whether two drugs act synergjstically.
[0345] While synergy is not required for therapeutic benefit in accordance with the
methods of described herein, in one embodiment, the present invention provides a method of
cancer treatment, wherein there is synergy between a phosphoramidate alkylator prodrug and
another anticancer agent. Two drugs can be said to possess therapeutic synergy if a
combination dose regimen of the two drugs produces a significantly better tumor cell kill than
the sum of the single Agents at optimal or maximum tolerated doses. The "degree of
synergy" can be defined as net log of tumor cell kill by the optimum combination regimen
minus net log of tumor cell kill by the optimal dose of the most active single Agent.
Differences in cell kill of greater than ten-fold (one log) are considered conclusively
indicative of therapeutic synergy.
[0346] When a phosphoramidate alkylator prodrug is used with another anti-cancer agent, a
phosphoramidate alkylator prodrug will, at least in some embodiments, be administered prior
to the initiation of therapy with the other drug or drugs and administration will typically be
continued throughout the course of treatment with the other drug or drugs. In some
embodiments, the drug co-administered with a phosphoramidate alkylator prodrug will be
delivered at a lower dose, and optionally for longer periods, than would be the case in the
absence of a phosphoramidate alkylator prodrug administration. Such "low dose" therapies
can involve, for example, administering an anti-cancer drug, including but not limited to
paclitaxel, docetaxel, doxorubicin, cisplatin, or carboplatin, at a lower than approved dose
and for a longer period of time together with a phosphoramidate alkylator prodrug
administered in accordance with the methods described herein.
[0347] These methods can be used to improve patient outcomes over currently practiced
therapies by more effectively killing cancer cells or stopping growth of cancer cell as well as
diminishing unwanted side effects of the other therapy. When employed in combination with
a phosphoramidate alkylator prodrug, the additional anti-cancer agent(s) is dosed using either
the standard dosages employed for those Agents (i.e., when used without a phosphoramidate
alkylator prodrug) or are less than those standard dosages.
{0348] The administration of a phosphoramidate alkylator prodrug in accordance with the
methods described herein can therefore allow the physician to treat cancer with existing (or
later approved) drugs at lower doses (than currently used), thus ameliorating some or all of

the toxic side effects of such drugs. The exact dosage for a given patient varies from patient
to patient, depending on a number of factors including the drug combination employed, the
particular disease being treated, and the condition and prior history of the patient, but can be
determined using only the skill of the ordinarily skilled artisan in view of the teachings
herein.
[0349] Specific dose regimens for known and approved chemotherapeutic agents or
antineoplastic agents (i.e., the recommended effective dose) are known to physicians and are
given, for example, in the product descriptions found in the Physician's Desk Reference 2003,
(Physicians' Desk Reference, 57th Ed) Medical Economics Company, Inc., Oradell, NJ
and/or are available from the Federal Drug Administration. Illustrative dosage regimens for
certain anti-cancer drugs are also provided below.
[0350] Cancer drugs can be classified generally as alkylators, anthracyclines, antibiotics,
aromatase inhibitors, bisphosphonates, cyclo-oxygenase inhibitors, estrogen receptor
modulators, folate antagonists, inorganic aresenates, microtubule inhibitors, modifiers,
nitrosoureas, nucleoside analogs, osteoclast inhibitors, platinum containing compounds,
retinoids, topoisomerase 1 inhibitors, topoisomerase 2 inhibitors, and tyrosine kinase
inhibitors. In accordance with the methods described herein, a phosphorarnidate alkylator
prodrug can be co-administered with any anti-cancer drug from any of these classes or can be
administered prior to or after treatment with any such drug or combination of such drugs. In
addition, a phosphorarnidate alkylator prodrug can be administered in combination with a
biologic therapy (e.g., treatment with interferons, interleukins, colony stimulating factors and
monoclonal antibodies). Biologies used for treatment of cancer are known in the art and
include, for example, trastuzumab (Herceptin), tosirumomab and 131I Tositumomab (Bexxar),
rituximab (Rituxan).
[0351] Alkylators useful in the practice of the methods described herein include but are not
limited to busulfan (Myleran, Busulfex), chlorambucil (Leukeran), ifosfamide (with or
without MESNA), cyclophosphamide (Cytoxan, Neosar), ghrfosfarnide, melphalan, L-PAM
(Alkeran), dacarbazine (DTIC-Dome), and temozolamide (Temodar). In accordance with the
methods described herein a phosphorarnidate alkylator prodrug is co-administered with an
alkylator to treat cancer. In one version, the cancer is chronic myelogenous leukemia,
multiple myeloma, or anaplastic astrocytoma.

[0352] In one embodiment, the present invention provides a method of treating cancer
treatable by administering an alkylator by administering the phosphoramidate alkylator
prodrugs of the present invention or those alone or in combination with at least another
alkylator or a prodrug thereof. Alkylators, such as, for example, cyclophosphamide,
ifosfamide, glufosfamide, mechlorethamine, melphalan, chlorambucil, dacarbazine,
temozolomide, carmustine, streptozocin, bendamustin, busulfan, thiotepa, cisplatin,
carboplatin, and oxaliplatin, and types of cancers treated using any one of such alkylators
alone or in combination with other anti cancer or chemoprotective agents are described for
example in the reference Hardman et ah, (supra).
[0353] In one embodiment, the present invention provides a method of treating cancer by
coadministeTing a phosphoramidate alkylator prodrug with at least the alkylator
Cyclophosphamide, in the treatment of Stages III and IV malignant lymphomas, multiple
myeloma, leukemia, mycosis fungoides, neuroblastoma, ovarian adenocarcinoma,
retinoblastoma, and carcinoma of the breast. Cyclophosphamide is administered for
induction therapy in doses of 1500-1800 mg/m2 that are administered intravenously in
divided doses over a period of three to five days; for maintenance therapy, 350-550 mg/m2
are administered every 7-10 days, or 110-185 mg/m2 are administered intravenously twice
weekly. In accordance with the methods described herein, a phosphoramidate alkylator
prodrug is co-administered with cyclosphosphamide administered at such doses or at lower
doses and/or for a longer duration than normal for administration of Cyclosphosphamide
alone.
[0354] In one embodiment, the present invention provides a method of treating cancer by
administering a phosphoramidate alkylator produg of the invention together with a cancer
tre&toeut regimen using at least the alkylator Mechlorethamine. For example,
Mechlorethamine is used in the combination chemotherapy regimen MOPP
(mecMorethamine, Oncovin (vincristine), procarbazine, and prednisone) in patients with
Hodgkin's disease and administered by intravenous bolus actministration is doses 6mg/m on
days 1 and 8 of the 28 day cycles of each course of treatment.
[0355] In one embodiment, the present invention provides a method of treating cancer by
administering a phosphoramidate alkylator produg of the invention with a cancer treatment
regimen using at least the alkylator Ifosfamide. Ifosfamide is used to treat pediatric and adult
sarcomas, carcinomas of cervix and lung, and in combination with other drugs for germ cell

testicular cancer. Ifosfamide is used as part of the ICE (Ifosfamide, Carboplatin, and
Etoposide) ans RICE (Rituxan and ICE) regimens for treating lymphomas (see Hardman et
ah, supra).
[0356] In one embodiment, the present invention provides a method of treating cancer by
administering a phosphoramidate alkylator produg of the invention with a cancer treatment
regimen using at least the alkylator Glufosfamide. Glufosfamide is in the clinic for the
treatment of pancreatic cancer or Gemzar resistant pancreatic cancer. Glufosfamide can be
used for treating breast cancer, Morbus Hodgkin, gastrointestinal tract cancer, or as part of
the GCE (Glufosfamide, Carboplatin, and Etoposide) or RGCE (Rituxan and GCE) regimen,
for treating lymphomas. (Tidmarsh et al., PCT Pat. Appl. No. PCT/US2005/047314 filed on
22 December 2005, and PCT Pat Appl. entitled "Glufosfamide combination therapy",
Attorney Docket No. 021305-005900PC; and US Pat App. No. 60/760,599 and 60/719,787
and PCT Pat. Pub. No. WO 2005/076888, incorporated in their entirety herin by reference).
[0357] In one embodiment, the present invention provides a method of treating cancer by
administering a phosphoramidate alkylator produg of the invention with a cancer treatment
regimen using at least an alkylator selected from the group consisting of ethylenimines and
methylmelamines. In another embodiment the ethyleriimine is Triethylenemelamine or
Thiotepa.
[0358] Thiotepa can be used to treat adenocarcinomas of the breast, ovary, and bladder,
malignant lymphomas, bronchiogenic carcinomas, and Wilms' tumor. Thiotepa was used at
high doses in combination chemotherapy with cyclophosphamide in patients with refractory
malignancies treated with autologous bone transplantation and to treat a variety of cancers
including bladder, ovarian, breast, lung, brain, and lymphomas (see, International Agency for
Research on Cancer Monograph on tk MlmM o/CMiopic M ofCkmwls to
Humans, 1975,9:286, Lyon, France; International Agency for Research on Cancer
Monographs on the Evaluation of Carcinogenic Risk of Chemicals to Humans, 1990,50:
415, Lyon, France; and MEDLINEplus, 2003, Drug Information: Thiotepa, National Library
of Medicine). The methylmelamine Altretamine is used to treat advanced ovarian cancer
after failure of first round therapies.
[0359] In one embodiment, the present invention provides a method of treating cancer by
administering a phosphoramidate alkylator produg of the invention with a cancer treatment
regimen using at least the alkylator Melphalan, Chlorambucil, or Bendamustine. Melphalan

is used to treat multiple myolema and can be administered orally. Chlorambucil is used to
treat chronic lyphocytic leukemia and primary macroblobulinemia. Bendamustine,
developed by Salmedix Inc. can be used to treat hematological malignancies, such as, for
example, non-Hodgkin's lymphoma, chronis lymphocytic leukemia, and multiple myeloma.
[0360] In one embodiment, the present invention provides a method of treating cancer by
administering a phosphoramidate alkylator produg of the invention with a cancer treatment
regimen using at least the alkylator Busulfan. Busulfan is used to treat chronic granulocytic
leukemia and chronic myelogenous leukemia. High doses of busulfan can be used in
combination with Cyclophosphamide to treat patients with acute myelogenous leukemia
before bone marrow transplantation.
[0361] m one embodiment, the present invention provides a method of treating cancer by
administering a phosphoramidate alkylator produg of the invention with a cancer treatment
regimen using at least a nitrosourea alkylator. In another embodiment, the nitrosourea
alkylator is Carmustine. Carmustine can be used to treat Hodgkin's disease, lymphomas,
myelomas, malignant astrocytomas, metastatic tumors of the brain, melanoma, and
gastrointestinal tumors. In another embodiment, the nitrosourea is Streptozocin which is used
to treat pancreatic islet cell carcinoma.
[0362] In one embodiment, the present invention provides a method of treating cancer by
administering a phosphoramidate alkylator produg of the invention with a cancer treatment
regimen using at least a triazene alkylator. In one embodiment, the triazene alkylator is
Dacarbazine. Dacarbazine is used to treat malignant melanoma, Hodgkin's disease, and adult
sarcoma. In another embodiment, the triazene alkylator is Temozolomide. Temozolomide
can be used to treat malignant gliomas.
[0363] In one embodiment, the present invention provides a method of treating CdflC6r Ity
administering a phosphoramidate alkylator produg Of the invention with a cancer treatment
regimen using at least a platinum coordination complex alkylator. In one embodiment, the
platinum coordination complex alkylator is Cisplatin. Cisplatin can be used to treat cancer of
bladder, head and neck, endometrium, small cell carcinoma of the lung, and some neoplasms
of childhood. Cisplatin alone or with cyclophosphamide is used to treat advanced ovarian
cancer. Combination chemotherapy of Cisplatin with Bleomycin, Etoposide, and Vinblastine
is USed to treat advanced testicular cancer; and with one of Paclitaxel, Cyclophosphamide, or
Doxorubicin to treat ovarian carcinoma.

[0364] Anthracyclines useful in the practice of the methods described herein include but
are not limited to, doxorubicin (Adriamycin, Doxil, Rubex), mitoxantrone (Novantrone),
idarubicin (Idamycin), valrubicin (Valstar), and epirubicin (Ellence). In accordance with the
methods described herein a phosphoramidate alkylator prodrug is co-administered with an
anthracycline to treat cancer. In one version, the cancer is acute nonlymphocytic leukemia,
Kaposi's sarcoma, prostate cancer, bladder cancer, metastatic carcinoma of the ovary, and
breast cancer.
[0365] As one example the compound (8S,10S)-10-[(3-Animo-2,3,6-trideoxy-alpha.-L-
lyxo-hexopyim>syl)oxy]-8-glycoloyl-7,^
naphthacenedione, more commonly known as doxorubicin, is a cytotoxic anthracycline
antibiotic isolated from cultures of Streptomycespeucetius var. caesius. Doxorubicin has
been used successfully to produce regression in disseminated neoplastic conditions such as
acute lymphoblastic leukemia, acute myeloblastic leukemia, Wilm's tumor, neuroblastoma,
soft tissue and bone sarcomas, breast carcinoma, ovarian carcinoma, transitional cell bladder
carcinoma, thyroid carcinoma, lymphomas of both Hodgkin and non-Hodgkin types,
bronchogenic carcinoma, and gastric carcinoma. Doxorubicin is typically administered in a
dose in the range of 30-75 mg/m2 as a single intravenous injection administered at 21-day
intervals; weekly intravenous injection at doses of 20 mg/m2; or 30 mg/m2 doses on each of
three successive days repeated every four weeks. In accordance with the methods of the
methods described herein, a phosphoramidate alkylator prodrug is co-administered starting
prior to and continuing after the administration of doxorubicin at such doses (or at lower
doses). Cyclic Anthracycline cytotoxin prodrugs useful in the practice of the methods
described herein are provided by the reference Matteuci et ah, PCT Patent Aplication No.
USO5/008161.
[0366] Antibiotics useful in the practice of the methods described herein include but are not
limited to dactmornycin, actinomycin D (Cosmegen), bleomycin (Blenoxane), daunorubicin,
and daunomycin (Cerubidine, DanuoXome). In accordance with the methods described
herein a phosphoramidate alkylator prodrug is co-administered with an antibiotic to treat
cancer. In one version, the cancer is a cancer selected from the group consisting of acute
lymphocytic leukemia, other leukemias, and Kaposi's sarcoma.
[0367] Aromatase inhibitors useful in the practice of the methods described herein include
but are not limited to anastrozole (Arimidex) and letroazole (Femara). In accordance with the

methods described herein a phosphoramidate alkylator prodrug is co-administered with an
aromatase inhibitor to treat cancer. In one version, the cancer is breast cancer.
[0368] Bisphosphonate inhibitors useful in the practice of the methods described herein
include but are not limited to zoledronate (Zometa). In accordance with the methods
described herein a phosphoramidate alkylator prodrug is co-administered with a
biphosphonate inhibitor to treat cancer. In one version, the cancer is a cancer selected from
the group consisting of multiple myeloma, bone metastases from solid tumors, or prostate
cancer.
[03691 Cyclo-oxygenase inhibitors useful in the practice of the methods described herein
include but are not limited to celecoxib (Celebrex). In accordance with the methods
described herein a phosphoramidate alkylator prodrug is co-administered with a cyclo-
oxygenase inhibitor to treat cancer. In one version, the cancer is colon cancer or a pre-
cancerous condition known as familial adenomatous polyposis.
[0370] Estrogen receptor modulators useful in the practice of the methods described herein
include but are not limited to tamoxifen (Nolvadex) and fulvestrant (Faslodex). In
accordance with the methods described herein a phosphoramidate alkylator prodrug is co-
administered with an estrogen receptor modulator to treat cancer. In one version, the cancer
is breast cancer or the treatment is administered to prevent the occurrence or reoccurrence of
breast cancer.
[0371] Folate antagonists useful in the practice of the methods described herein include but
are not limited to methotrexate and tremetrexate. In accordance with the methods described
herein a phosphoramidate alkylator prodrug is coadministered with a folate antagonist to
treat cancer. In one version, the cancer is osteosarcoma.
[0372] As one example, the compound N-[4-[[(2,4-diamino-6-pteridinyl)methyl
memylamho]benzoyl]-L-glutamic acid, commonly known as methotrexate, is an antifolate
drug that has been used in the treatment of gestational choriocarcinoma and in the treatment
of patients with chorioadenoma destruens and hydatiform mole. It is also useful in the
treatment of advanced stages of malignant lymphoma and in the treatment of advanced cases
of mycosis fungoides. Methotrexate is administered as follows. For choriocarcinoma,
intramuscular injections of doses of 15 to 30 mg are administered daily for a five-day course,
such courses repeated as needed with rest period of one or more weeks interposed between
courses of therapy. For leukemias, twice weekly intramuscular injections are administered in

doses of 30 mg/m2. For mycosis fungoides, weekly intramuscular injections of doses of 50
mg or, alternatively, of 25 mg are administered twice weekly. In accordance with the
methods described herein, a phosphoramidate alkylator prodrug is co-administered with
methotrexate administered at such doses (or at lower doses). 5-Methyl-6-[[(3,4,5-
trimemoxyphenyl)-ainino]methyl]-2,4-quinazolinediamine (commonly known as
trimetrexate) is another antifolate drug that can be co-administered with a phosphoramidate
alkylator prodrug.
[0373] Inorganic arsenates useful in the practice of the methods described herein include
but are not limited to arsenic trioxide (Trisenox). In accordance with the methods described
herein a phosphoramidate alkylator prodrug is co-administered with an inorganic arsenate to
treat cancer. In one version, the cancer is refractory acute promyelocytic leukemia (APL).
[0374] Microtubule inhibitors (as used herein, a "microtubule inhibitor" is any agent that
interferes with the assembly or disassembly of microtubules) useful in the practice of the
methods described herein include but are not limited to vincristine (Oncovin), .vinblastine
(Velban), paclitaxel (Taxol, Paxene), vinorelbine (Navelbine), docetaxel (Taxotere),
epothilone B or D or a derivative of either, and discodermolide or its derivatives. Tubulin
binding anticancer drugs and prodrugs thereof which can be used in the practice of the
methods of the present invention are provided in the reference Matteucci et al., PCT Patent
Application No. PCT/US2005/042095; US Patent Applications entitled "Tubulin Binding
Anti Cancer Agents and Prodrugs Thereof (Attorney RefNos. 021305-008500US, 021305-
008400US and 021305-004520US). In accordance with the methods described herein a
phosphoramidate alkylator prodrug is co-administered with a microtubule inhibitor to treat
cancer. In one version, the cancer is ovarian cancer, breast cancer, non-small cell lung
CailC^ Kaposi's sarcomaj and metastatic cancer of breast or ovary origin. As one example,
the compound 22-oxo-vincaleukoblastine, also commonly known as vincristine, is an alkaloid
obtained from the common periwinkle plant (Yinoa rosea, Linn,) and is useful in the
treatment of acute leukemia. It has also been shown to be useful in combination with other
oncolytic agents in the treatment of Hodgkin's disease, lymphosarcoma, reticulum-C6ll
sarcoma, rhabdomyosarcoma, neuroblastoma, and Wilm's tumor. Vincristine is administered
in weekly intravenous doses of 2 mg/m2 for children and 1.4 mg/m2 for adults. In accordance
with the methods described herein, a phosphoramidate alkylator prodrug is co-administered
with vincristine administered at such doses. In one version, a phosphoramidate alkylator
prodrug is not administered prior to treatment with a microtubule inhibitor, such as a taxane,

but rather, administration of a phosphoramidate alkylator prodrug is administered
simultaneously with or within a few days to a week after initiation of treatment with a
microtubule inhibitor.
[03751 Modifiers useful in the practice of the methods described herein include but are not
limited to Leucovorin (Wellcovorin), which is used with other drugs such as 5-fluorouracil to
treat colorectal cancer. In accordance with the methods described herein a phosphoramidate
alkylator prodrug is co-administered with a modifier and another anti-cancer agent to treat
cancer. In one version, the cancer is colon cancer. In one version, the modifier is a
compound that increases the ability of a cell to take up glucose, including but not limited to
the compound N-hydroxyurea. N-hydroxyurea has been reported to enhance the ability of a
cell to take up 2-deoxyglucose (see the reference Smith et al., 1999, Cancer Letters 141:85,
incorporated herein by reference), and administration of N-hydroxyurea at levels reported to
increase 2-deoxyglucose uptake or to treat leukemia together with administration of 2-
deoxyglucose and a phosphoramidate alkylator prodrug as described herein is one version of
*
the therapeutic methods provided herein. In another such version, a phosphoramidate
alkylator prodrug is co-administered with nitric oxide or a nitric oxide precursor, such as an
organic nitrite or a spermineNONOate, to treat cancer, as the latter compounds stimulate the
uptake of glucose.
[0376] Nitrosoureas useful in the practice of the methods described herein include but are
not limited to procarbazine (Matulane), lomustine, CCNU (CeeBU), carmustine (BCNU,
BiCNU, Gliadel Wafer), and estramustine (Emcyt). In accordance with the methods
described herein a phosphoramidate alkylator prodrug is co-administered with a nitrosourea
to treat cancer. In one version, the cancer is prostate cancer or glioblastoma, including
recurrent glioblastoma multiforme.
[0377] Nucleoside analogs useful in the practice of the methods described herein include
but are not limited to mercaptopurine, 6-MP (Purinethol), fluorouracil, 5-FU (Adrucil),
thioguanine, 6-TG (Thioguanine), hydroxyurea (Hydrea), cytarabine (Cytosar-U, DepoCyt),
floxuridine (FUDR), fludafabilie (Fludara), azacytidine (Vidaza), pentostatin (Nipent),
cladribine (Leustatin, 2-CdA), gemcitabine (Gemzar), and capecitabine (Xeloda). In
accordance with the methods described herein a phosphoramidate alkylator prodrug is co-
administered with a nucleoside analog to treat cancer. In one version, the cancer is B-C6ll
lymphocytic leukemia (CLL), hairy cell leukemia, adenocarcinoma of the pancreas,

metastatic breast cancer, non-small cell lung cancer, or metastatic colorectal carcinoma. As
one example, the compound 5-fluoro-2,4(lH,3H)-pyrimidinedione, also commonly known as
5-fluorouracil, is an antimetabolite nucleoside analog effective in the palliative management
of carcinoma of the colon, rectum, breast, stomach, and pancreas in patients who are
considered incurable by surgical or other means. 5-Fluorouracil is administered in initial
therapy in doses of 12 mg/m2 given intravenously once daily for 4 successive days with the
daily dose not exceeding 800 mg. If no toxicity is observed at any time during the course of
the therapy, 6 mg/kg are given intravenously on the 6th, 8th, 10th, and 12th days. No therapy
is given on the 5th, 7th, 9th, or 11th days. In poor risk patients or those who are not in an
adequate nutritional state, a daily dose of 6 mg/kg is administered for three days, with the
daily dose not exceeding 400 mg. If no toxicity is observed at any time during the treatment,
3 mg/kg.can be given on the 5th, 7th, and 9th days. No therapy is given on the 4th, 6th, or
8th days. A sequence of injections on either schedule constitutes a course of therapy. In
accordance with the methods described herein, a phosphorarnidate alkylator prodrug is co-
administered with 5-FU administered at such doses or with the prodrug form Xeloda with
correspondingly adjusted doses. As another example, the compound 2-amino-l,7-dihydro-
6H-purine-6-thione, also commonly known as 6-thioguanine, is a nucleoside analog effective
in the therapy of acute non-pymphocytic leukemias. 6-Thioguanine is orally administered in
doses of about 2 mg/kg of body weight per day. The total daily dose can be given at one
time. If after four weeks of dosage at this level there is no improvement, the dosage can be
cautiously increased to 3 mg/kg/day. In accordance with the methods described herein, a
phosphorarnidate alkylator prodrug is co-administered with 6-TG administered at such doses
(or at lower doses).
[0378] Osteoclast inhibitors useful in the practice of the methods described herein include
but are not limited* b pamldWate (AflA). Ill JITCOte Wl U methods described herein
a phosphorarnidate alkylator prodrug is co-administered with an osteoclast inhibitor to treat
cancer. In one version, the cancer is osteolytic bone metastases of breast cancer, and one or
IflOrC additional anti-cancer agents are also co-administered with a phosphorarnidate alkylator
prodrug.
[0350] Platinum compounds useful in the practice of the methods described herein include
but are not limited to cisplatin (Platinol) and carboplatin (Paraplatin). In accordance with the
methods described herein a phosphorarnidate alkylator prodrug is co-administered with a
platinum compound to treat cancer. In one version, the cancer is metastatic testicular cancer,

metastatic ovarian cancer, ovarian carcinoma, and transitional cell bladder cancer. As one
example, the compound cis-Diaminedichloroplatinum (II), commonly known as cisplatin, is
useful in the palliative treatment of metastatic testicular and ovarian tumors, and for the
treatment of transitional cell bladder cancer which is not amenable to surgery or radiotherapy.
Cisplatin, when used for advanced bladder cancer, is administered in intravenous injections
of doses of 50-70 mg/m2 once every three to four weeks. In accordance with the methods
described herein, a phosphoramidate alkylator prodrug is co-administered with cisplatin
administered at these doses (or at lower doses). One or more additional anti-cancer agents
can be co-administered with the platinum compound and a phosphoramidate alkylator
prodrug. As one example, Platinol, Blenoxane, and Velbam can be co-administered with a
phosphoramidate alkylator prodrug. As another example, Platinol and Adriamycin can be co-
administered with a phosphoramidate alkylator prodrug.
[0351] Retinoids useful in the practice of the methods described herein include but are not
limited to tretinoin, ATRA (Vesanoid), alitretinoin (Panretin), and bexarotene (Targretin). In
accordance with the methods described herein a phosphoramidate alkylator prodrug is co-
administered with a retinoid to treat cancer. In one version, the cancer is a cancer selected
from the group consisting of APL, Kaposi's sarcoma, and T-C6ll lymphoma.
{0352] Topoisomerase 1 inhibitors useful in the practice of the methods described herein
include but are not limited to topotecan (Hycamtin) and irinotecan (Camptostar). In
accordance with the methods described herein a phosphoramidate alkylator prodrug is co-
administered with a topoisomerase 1 inhibitor to treat cancer. Topoisomerase inhibitors and
prodrugs thereof useful in the practice of the methods of the present invention are provided in
the reference Matteucci et al., PCT Patent Application No. PCT/US2005/041959. In one
version, the cancer is metastatic carcinoma of the ovary, colon, or rectum, or small cell lung
cancer. As noted above, however, in one version of the methods described herein,
administration of a phosphoramidate alkylator prodrug either precedes or follows, or both,
adrnrnistrab'on of a topoisomerase 1 inhibitor but i5 not administered concurrently therewith.
[0353] Topoisomerase 2 inhibitors useful in the practice of the methods described herein
include but are not limited to etoposide, VP-16 (Vepesid), teniposide, VM-26 (Yumon), and
etoposide phosphate (Etopophos). In accordance with fee methods described herein a
phosphoramidate alkylator prodrug is co-administered with a topoisomerase 2 inhibitor to
treat cancer. In one version, the cancer is a cancer selected from the group consisting of

refractory testicular tumors, refractory acute lymphoblastic leukemia (ALL), and small cell
lung cancer. As noted above, however, in one version of the methods described herein,
administration of a phosphoramidate alkylator prodrug either precedes or follows, or both,
administration of a topoisomerase 2 inhibitor but is not administered concurrently therewith.
[0354] Tyrosine kinase inhibitors useful in the practice of the methods described herein
include but are not limited to imatiriib (Gleevec). In accordance with the methods described
herein a phosphoramidate alkylator prodrug is (X)-ad*rninistered with a tyrosine kinase
inhibitor to treat cancer. In one version, the cancer is CML or a metastatic or unresectable
malignant gastrointestinal stromal tumor.
[0355] Lonidamine analogs useful in the practice of the present invention are provided in the
Matteucci et al. U.S. Pat. Appl. Nos. 11/346632; 60/764,427; 60/764,438; and applications
entitled "Heterocyclic Lonidarnine Analogs" (Attorney Docket No. 021305-007220US;
021305-007900US) and PCT Publication Nos. WO 2006/015191, WO 2006/015263 and WO
2006/01007 A2..
[0356] Thus, described herein are methods of treating cancer in which a phosphoramidate
alkylator prodrug or a pharmaceutically acceptable salt thereof and one or more additional
anti-cancer agents are administered to a patient. Specific versions of such other anti-cancer
agents include without limitation 5-memyl-6-[[(3,4,5-trimethoxyphenyl)amino]-methyl]-2,4-
quinazolinediamine or a pharmaceutically acceptable salt thereof, (8S,10S)-10-(3-amino-
2,3,6-trideoxy-alpha-L-lyxo-hexopyranosyl)oxy]-8-glycoloyl-7,8,9,10-tetrahydro-6,8,ll-
trihydroxy-l-methoxy-5,12-naphthacenedione or a pharmaceutically acceptable salt thereof;
5-fluoro-2,4(lH,3H)-pyrimidinedione or a pharmaceutically acceptable salt thereof; 2-amino-
l,7-dihydro-6H-purine-6-thione or a pharmaceutically acceptable salt thereof; 22-oxo-
YinCaleukoWastine or a pharmaceutically acceptable salt thereof; 2-bis[(2-
cUoroe%l)aniino]tetrahydVo-2H-l,3,2-oxazapliosphorine, 2-oxide, or a pharmaceutically
acceptable salt thereof; N44-[[(2,4-diamino-6-pteridinyl)me%l]-methylamino]benzoyl]-L-
ghitamic acid, or a pharmaceutically acceptable salt thereof; or dsdiamminedichloro-
platinum (II).
IV. EXAMPLES

[0357] In the following examples, any reference to a compound designated by a letter is a
reference to the structure shown next to or above that letter in the corresponding reaction
schemes.
Synthesis
[0358] Methods to synthesize the phosphoramidate alkylator prodrugs of the present
invention are provided in section lib. Starting materials used in the synthesis of the
phosphoramidate alkylator prodrugs of the present invention were bought, when available,
from commercial manufacturers, such as, for example, the Sigma-Aldrich Co. 1-N-Methyl-
2-mtroimidazole-5-methanol was purchased from Syngene, India. Non-commercially
available starting materials can be synthesized in via standard literature procedures. Such
procedures can be identified via literature search tools such as SciFinder available from the
American Chemical Society or Beilstein, available from MDL Software.
[0359] Reactions with moisture sensitive compounds, such as, for example, POCl3 and
PCI3, and their mono and dichloro derivatives were performed employing anhydrous solvents
and under nitrogen or argon. Separation of a product from the reaction mixture was
performed employing a work-up where necessary, followed by vacuum distillation,
crystallization, column chromatography, or preparative thick layer chromatography. A
suitable eluent for the column chromatography of a compound can be determined by reading
this disclosure and/or by determining the Rf of the compound by thin layer chromatography
and choosing a solvent which allows separation of the desired compound from undesired
compounds. The choice of a particular eluent can depend, among other factors, on the polar
nature of the compound, existence of other closely eluting compounds, type of stationary
phase such as silica gel or alumina used, and the amount of pressure used to elute the solvent
through the stationary phase. In practice, different compositions of solvents can be used to
separate the same compound.
[0360] Separated compounds were analyzed for their purity by Standard analytical
techniques, such as, TLC, NMR spectroscopy, and LC-MS, and stored in a freezer or a
fridge, avoiding moisture, light, or air. Stock solutions of phosphoramidate alkylator prodrug
compounds were prepared in DMSO and stored in a freezer.
Example 1
Synthesis of Compound 23


[0361] To a solution of 5-nitrofurfuryl alcohol (200 mg, 1.4 mmol) in THF (10 ml) at -
78°C POCI3 was added in one portion followed by a dropwise addition of triethylamine
(TEA, 0.22 ml, 1.54 mmol). Temperature was increased to -30°C in one hour and then 2-
cUoroemylarnine hydrochloride was added followed by TEA (1 ml, 7 mmol). After the
temperature was raised to room temperature (rt), the reaction was continued for one more
hour, the reaction mixture was quenched with water and the organic layer was separated. The
aqueous layer was extracted with DCM and the combined organic solution was dried and
concentrated. Compound 23 was separated by flash column chromatography and analyzed
by LC/MS and NMR spectroscopy to be pure.

[0362] A suspension of ^Memyl-2-chloroeihylammonium chloride (10 gm) in POCI3 (40
ml) was refluxed (135°C) over night. After removing excess POCI3 under vacuum product 5i
was distilled out under vacuum as light yellow oil and analyzed by H and P NMR
spectroscopy to be pure.

[0363] To solution of 5i (1 gm, 4.75 mmol) and AT-Methyl-2-chloroethylammonium
chloride (0.62 gm, 4.75 mmol) in THF at -78°C diisopropylethylamine (DIEA, 1.65 ml, 9.5
mmol) was slowly added and the reaction mixture was warmed to rt. After stirring at rt for
one hour, the reaction mixture was diluted with ethyl acetate and washed with brine. The
organic layer was dried over MgSC>4 and concentrated to yield a residue which was separated
by flash chromatography yielding compound 5ii as oil.
[0364] To a solution of //-methyl 2-nitroimidazole-5-methanol (0.5 g, 3.2 mmol) in
dimethoxyethane (DME) lithium bis(trimemyMyl)amide (3.2 mmol, 3.2 ml, 1 M in THF)
was added at -78°C. After 5 min, 5ii (2.9 mmol, 770 mg) was added and the reaction mixture
was warmed up to -20°C, diluted with ethyl acetate and washed with brine. The organic layer
was dried over MgSC>4 and concentrated. Purification by flash chromatography with 6-12%
methanol in DCM yielded 5.
[0365] Compounds of 8 and 16 were synthesized employing the procedure used for the
preparation of compound_5.



[0366] To a solution of ethanolamine (6.03 mL, 100 mmol) and K2CO3 (13.8 g, 100 mmol)
in DMF (38 mL) a solution of p-toluenesulfonyl chloride (19 g, 100 mmol) was added drop
wise at rt and the reaction mixture was heated up to 120°C (bath temperature). K2CO3 (27.6
g, 200 mmol) was added to the reaction mixture, followed by dropwise addition of 1,3-
dibromopropane (10 g, 50 mmol). After heating for two more hours, the reaction was cooled
to rt, poured into water (250 mL), and extracted with ethyl acetate. The organic layer was
dried with Na2SC>4 and concentrated to yield compound_35a as yellow oil which was
employed in the next reaction.
[0367] A solution of compound_35a (5 g) in aqueous HBr (48%, 50 ml) was distilled to
remove the aqueous portion (about 20 ml), and the reaction mixture was refluxed for 40 h.
Additional aqueous portion (5 ml) was removed by distillation and the reaction mixture was
refluxed (4 h). The reaction mixture was cooled to rt, diluted with water (20 mL), and
filtered through a celite pad. The filtrate was concentrated to dryness to yield a residue which
was coevaporated with ethanol thrice, and following addition of a large volume of acetone a
White Solid product 35b was filtered and washed with acetone twice and employed in the
phosphorylation provided below.

[0368] A suspension of compound_35b (1 g) in POCl3 (14 mL) was heated at 130°C for
about 14 h and excess POCI3 removed under vacuum at 130°C (bath temperature). The
residue was purified by column chromatography on silica gel employing 10-80%
ETOAc/hexane to yield product 35c which was convereted to compound 35 of the present
invention employing the same procedure as provided in Example 2 employing for column
chromatographic separation silica gel andlO-80% acetone/toluene as the eluent.
Example 4
Synthesis of Compound 7
[0369] Compound 7 was prepared by employing N-cyclopropyl-2-chloroethylammonium
chloride as provided below

[0370] To a solution of cyclopropylamine (25g) in dry THF (30 ml) a solution of 2-
bromoethanol (17.6g, 0.141 mol) in 30 ml THF was added dropwise over 35 minutes. The
reaction mixture was stirred for 1 hour at rt, and heated at 50°C for 75 minutes. After
cooling, the reaction mixture was concentrated to yield an orange oil to which was added a

solution of sodium hydroxide (7g) in water (50 ml). The reaction mixture was stirred for 10
minutes, and extracted 4 times with ethyl acetate (75 ml). The combined organic layer was
dried (MgSO^ and evaporated to give an orange oily residue. The residue was distilled in
vacuo at 53-56°C (1 mm Hg) to yield an intermediate alcohol (5.94 g, 42% yield) as a clear,
colorless liquid which was analyzed by LC/MS and 1H NMR to be pure.
[0371] To a solution of the intermediate alcohol (3.7 g, 36.6 mmol) in dry THF (30 ml) a
solution of HC1 in dioxane (4.0M, 18.3 ml, 73.2 mmol) was added. The reaction mixture was
cooled to 0°C and SOClj (6.50g, 54.9 mmol) was added by syringe. The reaction mixture
was refluxed (6 h), cooled, and concentrated to yield a residue. The residue was triturated
with dry ether (100 ml), filtered, and residual volatiles removed in vacuo to yield 7i (5.42g,
95% yield) which was analyzed by 'H NMR to be pure.
[0372] 7i (3.00g, 192 mmol) was added to POCb (15 ml) and refluxed under nitrogen for
7.5 hours. The reaction mixture was concentrated and the resulting oil distilled in vacuo
through a short path distillation apparatus to yield 7ii as a clear, pale yellow oil (3.6g, 79%
yield) which was analyzed by 'H NMR to be pure.
[0373] 7ii (0.50 g, 2.11 mmol) and N-cyclopropyl-2-cMoroefoylamine hydrochloride (0.33
g, 2.11 mmol) were combined in dry THF under argon. The reaction mixture was cooled to -
78°C and DIEA (0.545 g, 4.22 mmol) added slowly by syringe, wanned to rt slowly, stirred
for 1.5 hours and concentrated to give an orange oily residue. The residue was separated by
flash chromatography over silica using 0-50% of hexane in ethyl acetate to give 315 mg
(47% of theoretical) of pale yellow oil which was analyzed to be 7iii by MS.
[0374] N-methyl-2-nitroimidazole-5-methanol (76.8 mg, 0.489 mmol) was partially
dissolved in dry THF (2 ml) under argon. The reaction mixture was cooled to -78°C and a
solution of lithium bis(trimethylsilyl)am; added. After 15 minutes, a solution of 7iii (172 mg, 0.538 mmol) in 2 ml THF was added.
After 15 minutes the reaction mixture Was Slowly Waimed to rt, stirred for 2 hours, poured
into 25 ml water and extracted 3 times with ethyl acetate (30 ml). The combined organic
layers Were dried OVer MgSO4 and concentrated to give a yellow oily residue. The residue
was separsted by flash chromatography in 0-10% methanol in DCM to yield compound^
(110 mg, 51% yield) as a yellow oil which was analyzed by LC-MS and 'H NMR to be pure.


[0375J To a suspension of bis(2-cMoroemyl)ammonium chloride (1.43 g, 8.01 mmol) in
dichloromethane (DCM) phosphorus trichloride (0.32 ml, 3.64 mmol) was added at rt
followed by addition of TEA (3.05 ml, 21.84 mmol). The reaction mixture was stirred at rt
for 30 minutes and then 2V-methyl 2-nitroimidazolyl methanol (0.474 g, 3.31 mmol) in DME
was added. After stirring for 0.5 hour, the reaction mixture was cooled to -20°C and tert-
butyl hydroperoxide (0.7 ml, 3.82 mmol, 5.5 M in Decane) was added. The reaction mixture
was warmed to rt over a period of one hour, and poured into 10% aqueous HC1. The organic
layer was separated and the aqueous layer was extracted with DCM. The combined organic
solution was dried with MgSCU and concentrated to yield a residue which was purified by
flash chromatography with 6-12% methanol in DCM yielding 6.
[0376] Compound 15 was synthesized using the method described for the synthesis of
Compound 6 above.


[0377] To a solution of 7/-methyl-2-nitroimidazole-5-methanol (180 mg, 1.14 mmol),
triphenylphosphine (300 mg, 1.14 mmol), and isophosphoramide mustard (lc, 127 mg, 0.57
mmol) in THF (10 ml) diisopropyl azodicarboxylate (DIAD, 0.22ml, 1.14 mmol) was added
dropwise at rt. After two hours reaction mixture was concentrated and the residue separated
by flash chromatography with 30-100% acetone in toluene yielding compound.36.
[0378] Compounds 23 and 26 were synthesized employing the procedure of Example 6.

[0379] N-methyl-2-nitroimidazole-5-methanol (50 mg, 0.318 mmol) was dissolved in dry
THF (2 ml) under nitrogen. The solution was cooled to -78°C and a solution of lithium
bis(trimethylsilyl)amide (1M in toluene, 0.35 ml, 0.35 mmol) was added by syringe. After 5
minutes a solution of bis(chloroethyl) phosphoramidic dichloride (91 mg, 0.35 mmol) in THF
(2 ml) was added. After stirring at 78°C for 30 minutes, the temperature was reduced to -
20°C employing a NaCl/ice bath and anhydrous ammonia was bubbled through the reaction
mixture for 5 minutes. The reaction mixture was purged with nitrogen, warmed to rt, poured
into 25 ml water and extracted with ethyl acetate (4 x 25 ml). The combined organic layers
were dried (MgSO4) and concentrated to give pale yellow oil which was separated by flash
chromatography over silica gel using 0-10% methanol in dichloromethane yielding
compound 1 (32 mg, 28 % yield) of an oil which soldified on standing and was analyzed by
LC/MS and 1H NMR to be pure.
Example 8
Synthesis of Compounds 25,26
[0380] To a solution of 2-bromoethylarnmmoiiuiin bromide (19.4 g) in DCM (90 mL) at -
10°C was added a solution of POCl3 (2.3 mL) in DCM (4 mL) followed by addition of a
solution of TEA (14.1 mL) in DCM (25 mL). The reaction mixture was filtered, the filtrate
concentrated to ca. 30% of the original volume and filtered. The residue was washed with
DCM (3x25 mL) and the combined DCM portions concentrated to yield a solid to which a

mixture of THF (6 mL) and water (8 mL) was added. THF was removed in a rotary
evaporator, the resulting solution chilled overnight in a fridge. The precipitate obtained was
filtered, washed with water (10 mL) and ether (30 mL), and dryed in vacuo to yield 2.1 g of:

[0381] Isophosphoramide mustard

can be synthesized employing the method provided in Example 8, substituting 2-
bromoemylammmonium bromide with 2-chloroethylammmonium chloride. Synthesis of
Isophosphoramide mustard has been described (see for example Wiessler et al., supra).
[0382] The phosphoramidate alkylator toxin:

was transformed into compounds 24 and 25, employing the method provided in Example 6
and the appropriate Trigger-OH.
Example 9*
Synthesis of Compounds 37-105
[0383] The following compounds 37-105 were synthesized employing the Mitsunobu type
Coupling described for the synthesis of 25 or 36 above, and upon appropriate substitution of
the Trigger-OH and the ifosfamide mustard analog employed. For example, for the synthesis
of compounds 40,81,83,87,89,95,96,100, and 104, the ifosfamide mustard analog
employed was H0P(=O)(NHCH2CH2C1)2; in compounds 50,53,55,56,58 - 65,68 - 71,73
- 75,77 - 80,82„ 84 - 86,88,90 - 92,94,97 - 99,101 -103, and 105, the ifosfamide
mustard analog employed is HOP(=O)(NHCH2CH2Br)2; in compounds 37,39,52,54, and

93, the ifosfamide mustard analog employed is the R enantiomer of
HOP(=O)(NHCHMeCH2Cl)2; in compounds 38,41, 51, and 57 the ifosfamide mustard
analog employed is the S enantiomer of HOP(=O)(NHCHMeCH2Cl)2; in compounds 43 - 45
and 49 the ifosfamide mustard analog employed was the R enantiomer of
HOP(=O)(NHCH(CHMe2)CH2Cl)2; and in compounds 46 - 48, the ifosfamide mustard
analog employed was the S enantiomer of HOP(=O)(NHCH(CHMe2)CH2Cl)2.
[0384] The various Trigger-OH compounds employed in the synthesis of Compounds 37 -
105, included the following Trigger-OH compounds: l-N-methyl-2-nitroimidazole-5-
methanol, l-N-memyl-5-nitroimidazole-2-methanol, 5-nitrofuran-2-methanol, 5-
nitrothiophene-2-methanol;



[0385] The following compounds were according to the method described in Example 6.






Examples 10-26 describes the synthesis of various Trigger-OH compounds employed in the
synthesis of phosphoramidate alkylator prodrugs of the invention.
Example 10
Synthesis of Compound 52i


[0386] A solution of compound 52ii (100 mg, 0.48 nunol), 52iii (73 mg, 0.48 mmol), and
KOAc (190 mg, 1.92 mmol) in DMF (5 ml) was degassed thrice and PdCl2(dppf) (36 mg,
0.048 mmol) added to it at rt under an argon atmosphere. The reaction mixture was heated at
> 60°C for two hours, diluted with ethyl acetate (EA) and washed with brine. The organic layer
was dried, concentrated, and the residue separated by column chromatography on silica gel
employing as eluent EA/Hex (0 - 80%) to yield 52i.
[0387] Compounds 55i, 63i, 59i, 65i, and 68i were prepared in a similar manner as described
schematically below:
i

[0388] To a solution of compound 68ii (100 mg, 0.31 mmol) and 3-amino-l-propanol
(0.047 ml, 0.62 mmol) in THF (2.5 ml), DIEA (0.162 ml, 0.93 mmol) was added at rt. The
reaction mixture was stirred overnight and concentrated to yield a residue which was
separated by column chromatography on silica gel employing as eluent EA/Hex (0-80%) to
yield compound 68i.
[0389] Compound 69i was made similarly as depicted in the scheme below.

[0390] To a solution of compound 70ii (100 mg, 0.87 mmol) and compound 70iii (112 mg,
0.87 mmol) in acetone (8 ml), was added K2CO3 (78.6 mg, 0.87 mmol) at rt. The reaction
mixture was heated at 60°C with stirring for 1 h, filtered, and concentrated to yield a residue
which was separated by column chromatography on silica gel employin (EA\Hex) 0-60% to
yield compound 70i.
[0391] Compound 51i was made similarly as depicted in the scheme below.

[0392] A solution of compound 59ii (200 mg, 0.96 mmol) and 59iii (127 mg, 0.96 mmol) in
DMF (3 ml) was degassed thrice and PdCl2(dppf) (50 mg, 0.07 mmol) was added to it,

followed by Cul (8.5 mg, 0.043 mmol) and TEA (0.27 ml, 1.92 mmol), at rt, under argon
atmosphere and the reaction mixture was heated at 60 °C for two hours. The reaction mixture
was diluted with EA, washed with brine, the organic layer separated, dried, and concentrated
to yield a residue which was separated by column chromatography on silica gel employing as
eluent EA\Hex (0-70%) to yield compound 58i.

[0393] To a suspension of 67i (472 mg, 2.69 mmol) in DCM (20 ml) was added
phenyldichlorophosphate (0.2 ml, 1.34 mmol) at -20°C, followed by the dropwise addition of
TEA (0.75 ml, 5.38 mmol) and stirring. The reaction mixture was warmed up to rt, stirred at
rt for 1 h, poured into brine, the organic layer separated, and the aqueous layer extracted with
DCM. The combined organic layers were dried with MgSC>4 and concentrated. The residue
was separated by column chromatography on silica gel employingas eluent EA/hexane (10-
100%) to yield compound 67ii. To a solution of compound 67ii (42 mg) in EtOH (5 ml) was
added platinum(IV)oxide (20 mg), the reaction mixture degassed, and vigorously stirred
under hydrogen for 0.5 h. The reaction mixture was diluted with MeOH, filtered through a
syringe filter, the filtrate concentrated under vacuum and coevaperated with toluene to yield
compound 67iii. Compound 67iii was reacted with l-N-methyl2-Ditroimidazole-5-m6thanol
employing a Mitsunobu type reaction as described for the synthesis of Compound 36.
Example 15
Synthesis of Compoundsl06 and 107


[0394] To a solution of 5-nitrofiirfuryl alcohol (200 mg, 1.4 mmol) in THF (10 ml) was
added POCl3 (0.13 ml, 1.4 mmol) at -78°C, followed by the dropwise addition of TEA (0.216
ml, 1.54 mmol). The reaction temperature was wanned up to -10°C in 1 h, 2-
(phenylsulfonyl)ethylamine hydrochloride (832 mg, 3.5 mmol) added to it, followed by the
addition of TEA (1 ml, 7 mmol). The reaction was warmed to rt, stirred for 1 h, quenched
with water and the organic layer separated. The aqueous layer was extracted with DCM
twice, the combined organic layers were dried, concentrated to yield a residue which was
separated by column chromatography on silica gel employing as eluent acetone\toluene (30
to 100%) to yield product 106. Compound 107:

was synthesized using a similar method.
[0395] Compounds 108-112, shown below:



were synthesized employing the procedure described for the synthesis of compound 35 in

[0396] Compound 113 was synthesized following a procedure described in Example 7 as
described here. To a solution of 113ii (181 mg, 1.16 mmol) in THF (8 mL) was added
dropwise LiN(TMS)2 (1.2 mL, 1 M THF solution, 1.2 mmol) at -78 °C, followed by the
addition of li. The reaction mixture was wanned up to -20°C and NH3 bubbled through the
reaction mixture for 5 minutes. Water (20 mL) was added to the reaction mixture and the
reaction mixture extracted thrice with EA (30mL). The combined organic layers were dried
and concentrated to yield a residue which was separated by column chromatography on silica
gel employing acetone\toluene (30-100%) to yield compound 113.
[0397] Compounds 114-117 were synthesized according to the method described for
Compound 13 and substituting with the appropriate Trigger-OH as
starting material.




[0398] 48% HBr (60 mL) was added dropwise to rf4-ethanolarnine at 0°C. The reaction
roixture was stirred for lhr at rt and then gently refluxed and slowly distilled, 16 mL liquid
being collected in 2 hrs until 155°C (oil bath). This was replaced twice with 60 mL of 48%
HBr and the distillation continued for an additional 5 hr. 90 mL liquid was collected. The
resultant solution was heated at 165°C for 2hr and evaporated under vacuum. The residue
was recrystalled from an absolute ethol (10 mL)-ethyl acetate (30 mL) to 11.3g of J4-2-
bromoemarnine hydrobromide (compound 64i). Compound 64i (19.5 mmol, 1.0 eq.) was
added dropwise to a suspension of d4-2-bromoethamine hydrobromide (40.0 mmol, 2.05 eq.)
in dry DCM (100 mL) under argon,at -20°C, followed by the dropwise addition of TEA (81.9
mmol, 4.2 eq.) at -20°C. The reaction mixture was stirred at -20°C for 0.5 h, and at rt for 2 h,
poured into water, and extracted twice with DCM (30 mL). The combined organic layers
were washed with brine, dried over ^SO^ and concentrated under reduced pressure to yield
a residue which was separated by column chromatography on silica gel employing as eluent
Hexane/EA (100:70(v/v)) to yield 7.0 g of compound 64ii. Pt02 (0.7 g) was added to a
solution of compound 64ii (7.0 g) in MeOH (160 mL), the reaction mixture degassed and
exchanged with H2 thrice, stirred under H2 for 3 h at rt, and diluted with MeOH until the

white solid in the reaction mixture dissolved. The diluteed reaction mixture was
filtered, the filtrate concentrated under reduced pressure to yield a residue ehich was
washed with anhydrous ether twice to yield 2.9 g of compound 64iii. To a suspension
of compounds 64iii (1.92 g 1.0 eq.), l-N-methyl-2-nitroimidazolemethanol (l.Olg, 1.1
eq.), and PPh3 (2.39 g, 1.5 eq.) in THF (20 ml) was added DIAD (1.76 ml, 1.5 eq.),
under argon, at 0°C. The reaction mixture was stirred for 2 hours while being warmed
up from 0°C to rt, following which volatiles were removed under vacuum to yield a
residue. The residue was separated by flash chromatography on silica gel employing
as eluent Acetone/Toluene (100:70(v/v)) to yield 1.35 g of compound 64.

[0399] The vinyl derivative, 2iii, was synthesized according to the reference Cavalleri
et

al. J. Het. Chem., 1972, 9: 979, and oxymercurated as follows. Hg(OAc)2 (208 mg,
0.653 mmol) was dissolved in water (0.7 mL) and THF (0.7mL), followed by the
addition of compound 2iii (lOOmg, 0.653 mmol). The reaction mixture was stirred at rt
for 1.5 h, NaBH4 (25 mg) added to it in portions, and after stirring for 15 min the
reaction poured into water, extracted with EA, the EA layer dried and concentrated to
yield a residue which was separated by silica gel column chromatography employing
as eluent EA/Hexane (0-100%) to yield compound 2i (16 mg).

[0400] A solution of 94iii (7.1 g) in AC2O (9.7 mL) was added dropwise into a solution
of Fuming nitric acid (1.5 mL) was added into AcOH (12 mL) at 0°c. The reaction
mixture was warmed up to rt, stirred 1 hr, fuming nitric acid (1 mL) was added
dropwise into it and stirred for 1.5 h. The reaction mixture was poured into water,
extracted with EA, the EA layer dried and concentrated to yield a residue which was
separated by silica gel column

chromatography employing as eluent EA/Hexane (0-100%) to yield compound 94ii.
Compound 94ii (600 mg, 1.77 mmol) was suspended in methanol (10 mL) at 0°C
followed by the addition of NaBH4 (141 mg) in portions into the reaction mixture over
5 min. NaBH4 (100 mg) was added once every hour thrice, the reaction mixture was
stirred for 3.5 h, poured into water, extracted with EA, the EA layer dried and
concentrated to yield a residue which was separated by silica gel column
chromatography employing as eluent EA/Hexane (0-100%) to yield compound 94i
(289 mg) as yellow solid.

[0401] A mixture of A (1.4 g), CuCN (0.56g) and DMF (25 mL) was stirred at 140°C
for 35 min and on 300 mL of crushed ice and stirred for 10 min, The reaction mixture
was then filtered and the residue was separated by column chromatography
employing as eluent Hexane:EA (1:0 to 2:3) to yield compound 96iii as yellow oil (617
mg). Compound 96iii.

was converted to alcohol 96i and separated by column chromatography, following a
similar method as employed for compound 94iii in and using THF instead of MeOH as
solvent in the reaction.

[0402] A mixture of 99ii (500 mg), PdCI2(PPh3)2 (208 mg), and Cul (56.4 mg) was
suspended in TEA (15 mL), the reaction mixture was degassed and flushed with Ar 6
times each. Propyne was bubbled through the reaction mixture for 15 min, and the
reaction continued under a propyne atmosphere at 50°C bath for 2 h. The reaction
mixture was poured into EA, filtered, the filtrate concentrated to yield a residue which
was separated by

silica gel column chromatography employing as eluent EA/Hexane (0-100%) to yield
compound 99i (286 mg).

[0403] Ethyl formate (500 mL) was added to sarcosin methyl ester hydrochloride (82 g,
585.7 mmol, grounded into powder prior to reaction) contained in a 1-L round-bottomed
flask. The reaction mixture was cooled in an ice-water bath, stirred, a gas outlet connected
with the flask, NaH (60% oil suspension, 54 g, 1.35 mol) added slowly during a period of 2 h,
and stirred at rt for about 14 h. Volatiles were removed using a rotary evaporator to yield a
residue which was triturated twice with hexane (500 mL) to yield a sticky light brown paste
which was dissolved in ethanol (400 mL) and cone HCI (50 mL) and stirred at 110°C for 1.5
h. After the reaction mixture cooled down, the white precipitate was filtered off and the
residue washed with 2 x 25 mL of ethanol. The filtrate was evaporated to yield a thick brown
oil to which was added 10% aqueous HO Ac, H2NCN (45 g, 1.07 mol), and sodium acetate
(88 g, 1.07 mol). The reaction mixture was stirred at 90-100°C for 1.5 h to yield a clear
solution which was cooled, its pH adjusted to 1 using concentrated HCI and the resulting
solution concentrated to 1/5 its original volume using a rotary evaporator at a temperature not
more than 45°C. The concentrated reaction mixture was carefully neutralized by addition of
K2CO3 to a pH of 8-9 and extracted with EA (5 x 200 mL followed by 3 x 50 mL). The
combined ethyl acetate layers were dried over MgSO4, filtered, and volatiles removed to
yield 48 g of 1 -N-methyl-2~amino imidazole-5-carboxylic acid ethyl ester.
Example 23
Synthesis of l-N-methvl-2-amino imidazole-5-carboxvlic acid ethyl ester


[0404] Ethyl formate (850 mL) was added to sarcosine methyl ester HC1 salt (205 g, 1.46
mol, grounded into powder prior to use), potassium carbonate (205 g, 1.48 mol), and EtOH
(800 mL,), stirred overnight at rt, and filtered. The filtrate was concentrated in a rotary
evaporator during which the residue separated into two layers. The upper layer was separated
and the lower layer was extracted with EA. Combined EA layers and the upper layer was
dried over MgSC>4, filtered, and concentrated to yield 185 g (81%) of N-formyl sarcosine
methyl ester which was used for the following reaction. NaH (60% oil suspension, 16.0 g,' 0.4
mol) was carefully added in several portions in 1 h to a mixture of N-formyl sarcosine methyl
ester (50 g, 0.34 mol) and ethyl formate (160 mL) cooled in an ice-water bath. The reaction
mixture was stirred, the temperature raised to rt, and the stirring continued overnight. The
reaction mixture was triturated twice with hexane (100 mL each time), the residue dissolved
in EtOH (100 mL) and concentrated HC1 (60 mL), and the reaction mixture stirred at 110°C.
After 1 h, the reaction mixture was cooled down, filtered, the residue washed with EtOH and
the filtrate concentrated to yield a thick brown oil. The oil was added to 10% HOAc in water
(200 mL), NH2CN (35 g) and sodium acetate (90 g), stirred at 95°C. After lh the reaction
mixture was concentrated to 1/3 its original volume in a rotary evaporator and its pH adjusted
to about 9 by addition of sodium carbonate. The reaction mixture was then extracted with
EA (8 x 100 mL), the combined EA layers dried, filtered, and concentrated to yield a residue
which was purified by recrystallization to yield l-N-methyl-2-amino imidazole-5-carboxylic
acid ethyl ester ("amino ester").
Example 24
Synthesis of l-N-methvl-2-nitroimidazole-5-carboxvlis acid ethyl ester


[0405] A solution of the amino ester (36.94 g, 0.218 mol) in 200 ml of acetic acid was
added drop wise to a solution of sodium nitrite (100 g, 1.449 mol) and water (300 ml) cooled
in an ice-water bath, and stirred. The temperature of the reaction mixture, which was
measured to be around -5 - 10°C was raised to rt and and the reaction mixture stirred
overnight. The reaction mixture was extracted with DCM (3 x 150 mL). The combined
DCM layers were dried and evaporated to yield a reddish residue which was separated by
column chromatography on silica gel employing as ehjent EA/hexane (30%) to yield 1-N-
methyl-2-nitroimidazole-5-carboxylic acid ethyl ester ("nitro ester") as a light brown solid
(27 g, yield 62%).
[0406] This method described in Example 24 and employing aqueous acetic acid is an
improvement of the method using about 7% sulfuric acid (v/v) for the diazonium ion
formation from the amino ester. Using aqueous sulfuric acid, the reaction volume becomes
large causing difficulty in stirring the reaction mixture effectively. For example, a reaction
involving 150 g of the amino ester required a reaction mixture volume of about 12 L. The
sticky nitro ester formed as product in aqueous sulfuric acid and disrupted the stirring of the
reaction mixture.

[0407] A suspension of the nitro ester (39.2 g, 196.9 mmol) in IN NaOH (600 mL) and
water (200 mL) was stirred at rt for about 20 h to give a clear light brown solution. The pH
of the reaction mixture was adjusted to about 1 by addition of cone. HC1 and the reaction
mixture extracted with EA (5 x 150 mL). The combined ethyl acetate layers were dried over
MgSO4 and concentrated to yield l-N-methyl-2-nitroimidazole-5-carboxylis acid ("nitro
acid") as a light brown solid (32.2 g, 95%).


[0408] A mixture of the nitro acid (30.82 g, 180.23 mmol) and triethylamine (140 mL, 285
mmol) in anhydrous THF (360 mL) was stirred while the reaction mixture was cooled in a
dry ice-acetonitrile bath (temperature mmol) was added drop wise to this cooled reaction mixture during a period of 10 min and
stirred for 1 h followed by the addition of sodium borohydride (36 g, 947 mmol) and
dropwise addition of water during a period of 1 h while maintaining a temperature around or
less than 0°C. The reaction mixture was warmed up to 0°C. The solid was filtered off and
washed with THF. The combined THF portions were evaporated to yield l-N-methyl-2-
nitroimidazole-5-methanol as an orange solid (25 g) which was recrystallized from ethyl
acetate.
Example 27
Synthesis of Compound 119
[0409] To a suspension of l-N-methyl-2-nitroimidazole-5-methanol (50 mg, 0.32 mmol) in
DME, LiN(TMS)2 was added at -78°C with vigorous stirring. After 10 min, compound 119i
(67 mg, 0.32 mmol) was added and the reaction mixture was warmed to if. After 1 h, the
reaction mixture was concentrated and the residue was separated by chromatography on silica
gel (0 -100% acetone\toluene) to yield Compound 119.

Examples 28A-28V

[0410] Compounds 134 to 155 were synthesized by employing the corresponding
substituted phosphormarmdate and hydroxy substituted Trigger (Trigger-OH), according to
the procedures described in Examples 1-27 above.

Example 29B
Antiproliferation Assay
[0412] To determine the effect of phosphoramidate alkylator prodrugs on cell proliferation,
the antiproliferative activity of these compounds was tested in a multi-well Alamar Blue-
based assay. Cell growth in the presence and absence of the test compound was compared, as
measured by a fluorescence plate reader at excitation 550nm and emission 590nm (see
Biosource International Inc., Tech Application Notes, Use of Alamar Blue in the
measurement of Cell Viability and Toxicity, Determining IC50). The following cell lines were
tested with 20,000 cells/well/500uL medium: NC1-H460 cells (ATCC HTB-177, RPMI
medium (Gibco Products, Invitrogen Corporation, Carlsbad, CA)), HT29 cells (ATCC HTB-
38, RPMI medium (Gibco)), MES-SA cells (ATCC CRL-1976, McCoy's 5a medium
(ATCC)), MES-SA/Dx5 cells ((ATCC CRL-1977), McCoy's 5a medium (ATCC)), ACHN
cells (ATCC CRL-1611, Minimum essential medium, Eagle (ATCC)), PC3 cells (ATCC
CRL-1435, Ham's F12K medium (ATCC)). The cells were seeded in glass inserts placed in
each well of a 24-well plate in the density and medium as specified above one day prior to
compound testing. After 24 hours, these plates were divided into two groups - anoxia group
and air group. A test compound was added to each well (200uL volume) in the treatment
groups at concentrations varying from 100,30,10,3,1,0.3,0.1,0.03, to 0.01 uM. All test

compounds were serially diluted in complete medium with final DMSO concentrations less
than or equal to 1% in each well. The cells in the anoxia treatment group were incubated for
2 hours in a Bactron II anaerobic chamber. The cells in the air treatment group were
incubated for 2 hours in standard tissue-culture incubators. Following the 2 hour treatment
with a test compound, the test compound was removed from each well, cells were washed
with 500uL medium, and incubated for 3 days in 500uL fresh medium. After 3 days, cells
were stained with 10% Alamar Blue for 2 hours after which the capacity of cells to proliferate
was measured (as mentioned above), and the 50% growth inhibitory concentration (GIso (also
referred to IC5o herein)) of test compounds was calculated and tabulated in Table X below.







Example 30
Antiproliferation Assay - Oxygen Dependence
[0413] To determine the oxygen dependence of phosphoramidate alkylator prodrugs, the
antiproliferative activity of these compounds was tested in a multi-well Alamar Blue-based
assay as previously described (see Example 29). NC1-H460 cells (ATCC HTB-177, RPMI
medium (Gibco)) or HT29 (ATCC HTB-38, RPMI medium (Gibco)) were seeded at 20,000
cells/well/500uL medium in glass inserts in 24-well plates one day prior to testing. The cells
were incubated for 2 hours in a Bactron II anaerobic chamber flushed with gasses of the
desired oxygen concentrations varying from anoxia, 0.1%, 0.3%, 0.6%, 1%, 10% oxygen,
and air. The calculated IC50 values (uM) are tabulated in Table Yl (H460 cells) or Table Y2
(HT29 cells) below.





Example 31
Clonogenic Assay - Oxygen Dependence
[0414] To determine the oxygen dependence of phosphoramidate alkylator prodrugs, a
clonogenic survival assay was performed. Cells were plated in 60 mm glass dishes (5x105
cells per dish in 5mL of medium) 2 days prior to compound testing. The following cell lines
were tested: NC1-H460 cells (ATCC HTB-177, RPMI medium (Gibco)), HT29 cells (ATCC
HTB-38, RPMI medium (Gibco)), PC3 cells (ATCC CRL-1435, Ham's F12K medium
(ATCC)). A solution of the test compound was made in complete medium immediately
before the test and added directly to cells (2mL volume). Anoxia or hypoxia (less than
200ppm O2) was achieved by exposing the glass dishes in a Bactron II anaerobic chamber or
in aluminum vessels (see Example 33) for 2 hours. For the anaerobic chamber, desired levels
of oxygenation between 200ppm and air were achieved by flushing the anaerobic chamber
with pre-calibrated gasses prior to experimentation. For the aluminum vessels, anoxia or

hypoxia was achieved by exposing the glass dishes in pre-warmed, air tight aluminum jigs to
a series of five rapid evacuations and flushings with 95% nitrogen plus 5% carbon dioxide in
a 37°C water bath on a shaking platform (controls are flushed as well). After the fifth
evacuation and flushing, the platform (with water bath and jigs) was shaken for 5 minutes,
after which one more evacuation and flushing were performed, and the jigs were transferred
to a shaker in a 37 degree C incubator for the remainder of the 1 to 2 hour drug exposure.
Levels of oxygenation between 200 ppm and air were achieved by varying the degree and
number of evacuations. The oxygen concentrations in the medium and gas phases were
checked using an oxygen electrode (Annua, Phoenixville, PA) in a specially modified
aluminum jig that allowed for monitoring of both gas and liquid phases. Following the
exposure to drug, the glass dishes were removed from the chamber or aluminum vessels and
the drug was washed off the cells by rinsing with medium. The cells were then trypsinized
and plated for clonogenic survival in plastic Petri dishes. Ten to 14 days later, the dishes
were stained with crystal violet (0.25% in 95% ethanol), and colonies containing-more than
50 cells were counted (see Example 33). The P0% growth inhibitory concentration (IC90,
90% killing, 10% survival) of test compounds was calculated and tabulated in Table Y3
below.


[0415] To determine the electrochemical properties and reduction potentials of
phosphoramidate alkylator prodrugs, cyclic voltammograms of these compounds were
generated by Bioanalytical Systems, Inc. All experiments were conducted with glassy carbon
(3.0mm diameter) working electrodes, Ag/AgCl reference electrodes, and platinum wire
auxiliary electrodes. Compounds were dissolved in lmL methanol to make final drug
concentrations between 0.5 and 1.5mM after the addition of 9mL Phosphate Buffered Saline
(PBS). The solution was added to an electrochemical cell vial and sparged with Argon for 5
minutes to remove most of the oxygen. Cyclic voltammetry was performed at 100 mV/sec
and at 10,000 mV/sec scan rates at a glassy carbon working electrode. One test run was
performed at a CGME mercury electrode (CGME in SMDE mode, 150um bore capillary,
size 8 drop), but little difference was observed between mercury and glassy carbon
voltammograms, so the mercury electrode was not used further. The single electron or
multiple electron reduction potentials of compounds were generated at each scan rate and are
tabulated in the table below.


Clonogenic Survival Assay
[0416] The phosphoramidate alkylator prodrugs of the invention were tested in the assay as
follows. Exponentially growing human H460 cells (obtained from the ATCC) were seeded
into 60mm notched glass plates at a density of between 2.5 and 5 xlO5 cells per plate and
grown in RPMI medium supplemented with 10 % fetal bovine serum for 2 days prior to
initiating drug treatment. On the day of the test, drug stocks of known concentrations were
prepared in complete medium, and 2 ml of the desired stock added to each plate. To achieve
complete equilibration between the surrounding gas phase and the liquid phase, the lid of the
glass plate was removed and the plate shaken for 5 minutes on an orbital shaker. The plates
were recovered and stored inside a glove-box. The glove-box was evacuated and gassed with
either a certified anoxic gas mixture (95% nitrogen and 5% carbon dioxide) or with an
aerobic (normoxic) gas mixture (95% air and 5% carbon dioxide). Cells were then incubated
with the drug for 2 hours at 37°C.
[0417J At the end of prodrug treatment, plates were removed from each vessel, and the
prodrug was promptly removed from the cells. Plates were washed with phosphate buffered
saline and a solution of trypsin-EDTA and then trypsinized for 5 minutes at 37°C. Detached
cells were neutralized with medium plus serum and collected by centrifugation for 5 min at
lOOxg. Cells were resuspended at approximately lxlO6 cells/ml and diluted 10 fold to yield
stock concentrations for plating. The concentration of each stock was determined by
counting with a Coulter Z2 particle counter. Known numbers of cells were plated, and the
plates were placed in an incubator for between 7 and 10 days. Colonies were fixed and
stained with a solution of 95% ethanol and 0.25% crystal violet. Colonies of greater than 50
cells were counted, and the surviving fraction was determined.
[0418] lOmdeeDtaritWiic
assays were performed in the same way as
described above and in Example 31.
[0419] Cytotoxicity of compounds (Tables 1A and IB) were determined in hypoxia and in
normoxia by clonogenic assay employing H460 and HT29 cell lines as provided in Examples
31 and 33 and expressed as IC90 in uM, and by anti-proliferation assay performed by
modifying a multi-well assay described by Hay et ah, I Med. Client., 2003,46:169-82
employing H460, HT29, HCT116, and DX-5 cell lines and expressed as IC50 in uM (see
Example 29). The ratio of IC50 or IC90 determined in normoxia and hypoxia is called hypoxia

cytotoxicity ration (HCR) and can be a measure of the hypoxia selective cytotoxicity of the
prodrugs of the present invention.




Effect of Compound 25 on Cell Cycle Distribution
[0420] Cells (H60, PC3 and HT29) were seeded at a density of 1.0 x 106 cells/3ml medium
per 60mm dish. After 24 h attachment, cells were exposed to Compound 25 at the indicated
concentrations for 2 h under either normoxia (air) or anoxia (nitrogen). Cells were washed
twice, and incubated for additional 22 h in fresh medium. Cells were trypsinized,
centrifuged, and fixed in 75% ethanol at least for 24 h at -20°C. Cell cycle distribution was
determined using Guava Cell Cycle reagent (Guava, Hayward, CA) by flow cytometry
(Guava, Hayward, CA). The data demonstrate that Compound 25 induces cell cycle arrest in
oxygen- and concentration-dependent manner in multiple human cancer cell lines.



Example 35
Spheroid Model
[0421] Two human cancer cell lines were used in these spheroid studies to determine the
efficacy of the hypoxic activated phophoramidate alkylator prodrugs. HT29 colorectal
adenocarcinoma (colon carcinoma) cells were seeded directly into a 125 ml spinner flask at
10,000 cells/ mL and grown in RPMI medium supplemented with 10% FBS and antibiotics.
As these cells divided, they adhered to each other and formed spheroids. H460 lung
carcinoma cells were seeded into a flask coated with a non-adherent surface to form small
balls of cells that can be seeded into a spinner flask. To initiate H460 cell seeds, 150 cm2
tissue culture flasks were coated with 1% agarose and then 10,000 cells per flask were added
and allowed to grow in RPMI medium supplemented with 10% FBS and antibiotics for 3 to 5
days before seeding into spinner cultures. For both cell lines, growth medium was changed
every day after the spheroids became visible to the eye.
[0422] In order to determine the morphology and the location of hypoxic regions within an
intact spheroid, whole spheroids were prepared for histology. For frozen sections, intact
spheroids were washed in phosphate buffered saline (PBS) and embedded in OCT and
rapidly frozen in a dry ice/2-methylbutane solution before being stored at -80°C. For paraffin
embedded sections, intact spheroids were fixed in a freshly prepared solution of 4%
paraformaldehyde in PBS and subsequently embedded and sectioned.
[0423] To assess the ability of a phosphoramidate alkylator prodrug to penetrate to the
inner hypoxic cancer cells, become activated, release the phosphoramidate alkylator, and kill
those inner cancer cells, the clonogenic survival of spheroids exposed to drug for 2 h was
measured.

[0424] Spheroids were placed in a new growth medium and incubated for at least 1 h
before beginning experiments. Spheroids between 500 and 600 urn were isolated by filtering
the spheroid culture through a series of sterile mesh filters of defined size. Between 10 and
20 spheroids were placed on a siliconized notched 60 mm Pyrex dish in 3 mL of medium
with the desired concentration of the test compound. The dishes were placed in sealed
aluminum vessels and exposed to a series of evacuations and gassings with certified gases
containing 5% CO2 and a defined amount of 02 (0% O2, 3% O2,10% 02 or air). Spheroids
were incubated in a shaking water bath to ensure both the equilibrium of the dissolved O2 in
solution and the integrity of the spheroids in solution for 2 h. The test compound was
removed and the spheroids were washed before being completely digested with trypsin.
Since the necrotic core contains cellular debris a treatment with DNase I was required to
yield a uniform single cell-suspension. Cells were resuspended at 106/mL and plated for
clonogenic survival.
[0425] Initial dose response experiments were performed in monolayer cells under nitrogen,
0.6% O2, or air to establish the appropriate dose range and the oxygen dependence of
phosphoramidate alkylator release from a phosphoramidate alkylator prodrug. Clonogenic
survival was the end point and the data are summarized by the IC90 or C90 values (the
inhibitory concentration required to kill 90% of the cells and yield 10% survival).
Daunorubicin and cisplatin, each of which penetrates into speroids to a different extent, were
employed to kill the outer aerobic cancer cells of the spheroid. Daunorubicin was used to
penetrate the outer layers of a multicellular spheroid due to its high affinity toward cells and
cisplatin was used at doses appropriate kill only the outer aerobic cancer cells. As a control
for a bioreductive drug that killed cells under hypoxia in monolayer cultures, but not in
multicellular cell culture due to its high reactivity and poor penetration, Tirapazamine was
used both in monolayer based experments and in spheroids as tabulated below for H460 cells
exposed for 2 h.


[0426] A series of phosphoramidate alkylator prodrugs were tested in spheroids to
determine their ability to penetrate into the inner lying hypoxic cancer cells, become
activated, and kill the hypoxic cells. The results are tabulated below.

[0427] Similar results for the efficacy of Compound 25 were demonstrated in the HT29
spheroids as tabulated below:

[0428] The phosphoramidate alkylator prodrug was combined simultaneously with
cisplatin or daunorubicin and the spreroids exposed for 2 h to the combination, followed by
measurement of clonogenic survival. The results are tabulated below:

[0429] Phosphoramidate alkylator prodrugs demonstrate the ability to penetrate into the inner
lying cells in the spheroid and kill hypoxic cancer cells alone and in combination with
another agent that targets aerobic cancer cells.
Example 36
Antiproliferation Assay - DNA Mutant Repair Cells

[0430] Chinese hamster ovary cells mutant to specific DNA repair pathways were obtained
from ATCC. The following cell lines were tested with 2,500 or 3,000 cells/well/500uL
Dulbecco's Modified Eagle Medium (Gibco) supplemented with 10% fetal bovine serum and
antibiotics: AA8 cells (ATCC CRL-1859), EM9 cells (ATCC CRL-1861), UV41 cells
(ATCC CRH860), UV135 cells (ATCC CRW867), ERS1SF cells. All cell lines were
initially screened with an anti-proliferation assay and those demonstrating sensitivity were
retested with the clonogenic assay (as previously described) to confirm the proliferation
results. Cells were exposed to selected doses of phosphoramidate alkylator prodrugs of the
present invention for 2 h under hypoxic or aerobic conditions, the test compound was
removed, and the cells assayed. The following table lists the cell lines, the pathway mutated,
and the specific gene defect:

[0431] The following table lists the effect of exposure of various cell lines to Compounds 25
and 36 under anoxic or aerobic conditions and assayed by proliferation as measured by IC50.

[0432] The following table lists the IC90 values for clonogenic survival for selected cells
exposed to Comopund 25 under anoxic or aerobic conditions.

[0433] Only cell lines defective in homologous recombination were sensitive to Compound
25 under hypoxia. Since UV41 participates in both the nucleotide excision repair pathway as
well as with the homologous recombination repair pathway, Compound 25 possibly also

produced a significant amount of monoadducts. However, UV135 which is also involved in
nucleotide excision repair was not sensitive to Compound 25. The predominant lesions
produced by Compound 25 were DNA interstrand crosslinks. These results were confirmed
in UV41 and irslSF cells with the clonogenic assay. The exposure under aerobic conditions
produced the same spectrum of sensitivities as seen under hypoxia, indicating that the aerobic
toxicity was also caused by DNA interstrand crosslink formation. Compound 36 exhibited a
similar pattern of sensitivity in the mutant cell lines, indicating that Compound 36 also
produced DNA interstrand crosslinks.
Example 37
Multilayered Cell Culture Assay
[0434] This example demonstrates the effect of Compound 25 on tissue penetration using
multilayered cell culture (MCC) and to assess any bystander effect. MCCs were incubated
with oxygenated media (20% O2 & 5% O2) or hypoxic media (approximately 0% O2) and the
test compound was exposed from one side (exposed,surface, normoxic side) while the other
side was temporarily closed off (far side, hypoxic side). When MCCs are incubated in
media at 20% O2 or 5% O2 a gradient in oxygen develops from the surface exposed to the
media towards the far surface of the culture. The furthest 50 um of tissue becomes depleted
of oxygen. The extent of O2 depletion is greater with 5% than the 20% Oz gassed media;
incubation with 5% O2 reflects the in vivo situation most closely. Incubating MCCs with
media at 0% O? models perfusion limited hypoxia, where tumor blood vessels become
completely depleted of oxygen and test compound must penetrate extensive distances to
reach all cells. This situation therefore poses a greater barrier to drug penetration, if binding
of activated drug acts to limit its penetration.
[0435] MCC based experiments were carried out with media gassed with 0, 5 or 20% 02
for 45 minutes prior to and during incubation with the test compound. HCT116 cells were
grown to a thickness of 150pm on a solid support and one side of the culture was clamped off
to develop diffusion limited hypoxia. Cultures were exposed to test compound for 1 hr under
0% O2,5% O2 or 20% O2 and efficacy assessed by measuring the inhibition of BrdU
incorporation. The cultures were incubated for a second hour in fresh media at 20% 02 and
removed from the apparatus and returned to a normal growth chamber, where media flows
over both sides of the MCC. Cultures were incubated for 24 hours prior to BrdUrd labeling
and subsequent cryosectioning. BrdUrd labeling on the exposed and far sides of the MCC

were analyzed using immunohistochemical staining, microscope imaging and computer
image analysis to assess the effect of Compound 25 on cell proliferation.
[0436] When cultures were exposed to graded doses of Compound 25 under 20% O2, 5 fold
less compound was required on the far (hypoxic) side compared to the exposed (normoxic)
side to produce comparable results, demonstrating penetration and hypoxic activation of
Compound 25. When MCC's were exposed to test compound under a more physiologically
relevant condition of 5% O2, Compound 25 was 10 fold more effective at inhibiting BrdU
incorporation on the hypoxic side as compared to the normoxic side. Normoxic sides of
cultures at 5% & 20% O2 were equally affected by exposure to Compound 25.
[0437] Compound 25 is more effective on the hypoxic side of cultures under 5% O2 than
with 0% O2. Comparison of normoxic versus hypoxic sides of cultures under 5% O2
demonstrated that Compound 25 penetrates effectively through relatively well oxygenated
tissue. Compound 25 is capable of killing hypoxic cells located about 150um from functional
blood vessels. Approximately 3-fold reduction in exposure to Compound 25 to the hypoxic
side was observed under 0% O2 relative to the exposure under 5% O9 conditions. Bystander
effect was observed only at the highest concentration.
[0438] The following table lists the effect of e exposed to graded doses of Compound 25 as
measured by IC50 (concentration to inhibit BrdU incorporation by 50%).

Example 38
Metabolism of Compound 25
By Human and Mouse Microsomal Protein
[0439] An in vifro assessment of metabolic stability of a phosphoramidate alkylator
prodrug (Compound 25) was performed using human (HLM), rat (RLM) and mouse (MLM)
liver microsomal proteins containing cytochrome P450 enzymes. A solution of Compound
25 (500uL, 5uM) was prepared by diluting a DMSO stock solution 100 fold in a
watermethanol bridge solution, adding microsomal protein (1 mg/mL) in PBS/MgCl2, and
enzymatic reactions initiated by adding an NADPH solution. 50 ul of the reaction mixture
was withdrawn at 0,10,20, and 30 minutes after addition of the NADPH solution, the

proteins were precipitated with acetonitrile and the clear supernatant was analyzed for the
amount of Compound 25 by reversed phase LC-MS/MS. Nifedipine and testosterone were
used as positive controls. The first study compared RLM to MLM (Table 1) and the second
study compared HLM to RLM (Tables 2A and 2B)



Example 39
Iv Vivo Pharmacokinetics of Phosphoramidate Alkylator Prodrugs
[0440] Various plasma pharmacokinetic parameters of phosphoramidate alkylator prodrugs
were determined in CD-I mice except where noted as listed below in Table 3.



Example 41
Cytochrome P450 Inhibition of the Metabolism of Compound 25
[0442] Eight reaction wells with 100 /J.L of a solution containing 50 mM potassium
phosphate, pH 7.4,2.6 mM NADP+, 6.6 mM glucose-6-phosphate, 0.8 U/mL of glucose-6-
phosphate dehydrogenase, andl :3 serial dilutions of the test compound (such as Compound
25) were prepared along with eight wells of 1:3 serial dilutions of a suitable positive control
inhibitor (such as furafylline for CYP1A2, sulfaphenazole for CYP2C9,' N-benzymirvanol for
CYPC219, quinidine for CYP2D6 and ketoconazole for CYP3A4). The concentrations of
test compound ranges from 0.0229 fiM to 200 fiM. The reactions were initiated by adding
100 (iL of a pre-warmed enzyme/substrate solution. A zero time-point control reaction was
prepared by adding 50 mL of 10% formic acid (400 mL of acetonitrile for 2C19) in water to
100 mL of cofactor solution to inactivate the enzymes, then adding 100 mL of
enzyme/substrate solution. A control reaction with no inhibitor was also prepared. After a
suitable incubation at 37 °C, the reactions were terminated by the addition of 50 mL of 10%
formic acid in water (400 mL of acetonitrile for 2C19). The reactions were prepared and
analyzed for the metabolite forms of the probe substrate (phenacetin for CYP1A2, diclofenac
for CYP2C9, (S)-mephenytoin for CYPC219, dextromethorphan for CYP2D6 and
midazolam, testosterone and nifedipine for CYP3A4) using HPLC/MS/MS. Each assay was
performed in duplicate. A summary of the IC50 values are listed below.



Example 42
Determination of the Potential Metabolites of Compound 25
Formed in Mouse, Rat, Dog and Human Hepatocytes
[0443] Compound 25 is incubated with mouse, rat, dog, monkey and human cryopreserved
hepatocytes at a concentration of 10 uM. The reactions are stopped at 0 (pre-incubation), 30,
60 and 120 minutes by quenching with acetonitrile prior to centrirugation and analysis by
high-performance liquid chromatography (HPLC) in conjunction with tandem mass
spectrometry (LC/MS/MS). Potential metabolites are identified by performing full scans
from 100 to 520 amu. The product ion spectra of the potential metabolites are subsequently
collected and compared to the product ion spectrum of the parent compound to determine
whether each potential metabolite is related to Compound 25. The disappearance of the
parent compound (Compound 25) and the appearance of potential metabolites over time are
monitored by comparing the peak heights at each time point acquired.
Example 43
Determination of the In Vivo Pharmacokinetics of Compound 25 and its Metabolite (s) in
Rat, Dog and Monkey
[0444] Pharmacokinetic parameters of Compound 25 and its metabolite(s) in Sprague
Dawley rats are determined following single intravenous administration of 5,20, 50 and 100
mg/kg Compound 25. The pharmacokinetics of Compound 25 and its metabolite(s) will also
be determined in beagle dogs and cynomologus monkeys following single intravenous
administration of 20 mg/kg Compound 25. Concentrations of Compound 25 and its
metabolite(s) in plasma are determined by a LC/MS/MS method and mean pharmacokinetic
parameters are computed.
Example 44
Mass Balance Study in Rats

[0445] Normal and bile-cannulated Sprague-Dawley rats are administered 14C-Compound
25 as a single intravenous dose. Blood plasma, urine, feces and are collected at specified
times and concentrations of total radioactivity are determined by liquid scintillation counting
(LSC).
Example 45
Quantitative Whole Body Autoradiography
[0446] Sprague-Dawley rats are administered a single intravenous dose of 14C-Compound
25. At specified times, one rat per time point is euthanized. Blood is centrifuged to obtain
plasma, and the blood and plasma are analyzed for concentration of radioactivity. Frozen rat
carcasses are embedded in 2% CMC, frozen into a block and sectioned at 40 um in a Leica
CM 3600 cryomicrotome. Collected sections are freeze-dried, mounted and exposed on
phosphorimaging plates along with 14C autoradiographic standards for subsequent calibration
of the image analysis software. Exposed screens are scanned using a Molecular Dynamics
Storm 820 or 860. The concentration of radidactivity in select tissues including adipose
(brown and white), adrenal gland, blood, brain (cerebrum, cerebellum, medulla) bone, bone
marrow, cecum and contents, epididymis, esophagus, eyeball (Uveal tract, aqueous humor,
lens), Harderian gland, heart, kidney (cortex, medulla, papilla and entire section), large
intestine and contents, liver, lung, lymph node submaxillary), pancreas, pituitary gland,
prostate gland, salivary gland, seminal vesicles, skeletal muscle, skin, stomach (and
contents), small intestine (and contents), spleen, spinal cord, trachea, thyroid and urinary
bladder (and contents) are measured by image analysis. Autoradioluminographs and digital
images are produced for each animal.
Example 46
Plasma Protein Binding of Compound 25
[0447] The protein binding in mouse, rat, dog, monkey and human plasma of Compound 25
is determined using ultrafiltration. Ultrafiltration is performed by aliquoting plasma spiked at
three concentrations with Compound 25 into a Centrifree® device in triplicate. All plasma
samples are then equilibrated to 37°C. The Centrifree® apparatus is centrifuged at 37°C for
30 minutes at 2500 x g. A 75 uL aliquot of the ultrafilitrate is spiked with the I.S. (deuterated
Compound 25) and analysed using LC/MS/MS. The ultrafiltrates are analyzed and quantified
using human ultrafiltrate standards for the calibration curve.

Example 47
[0448] Example 47 demonstrates the usefulness of a compound of this invention in treating
cancer employing a HT-29 human colon carcinoma xenograft mouse model.
[0449] Female CB17/SCE) mice (purchased from Charles River, Cambridge, MA), 7-8
weeks of age, were allowed to acclimatize for at least three days, and handled under
pathogen-free conditions. Human colon carcinoma cell line HT-29 was obtained from the
American Type Culture Collection. The cell lines were cultured in RPMI1640 media
supplemented with 10% fetal bovine serum. Cells were maintained in a 37°C incubator with
5% CO2. The HT-29 cells were harvested from culture and inoculated at 3 x 106 cells/ animal
in the peritoneal subcutaneous space. When the tumors grew to an average volume of 100
mm3 (day 8), each group of 10 mice was administered for three weeks, vehicle alone (saline
and PEG (lOmL/kg each), Group 1), Compound 36 alone (dissolved in 30% cyclodextrin in
PBS) at a daily dose of 20,60, or 200 mg/kg (Groups 2,3 and 4, respectively), and
Compound 36 at a daily dose of 20,60, and 200 mg/kg given 2-3 hours after a dose of 10
mg/kg of 5FU (in saline) (Group 5,6 and 7, respectively) and compared to a group receiving
only 5FU at 10 mg/kg (Group 8) as tabulated below.
[0450] The body weight of each mouse was recorded twice per week. Growth of each
xenograft was monitored by externally measuring tumors in two dimensions using a digital
caliper twice per week. Tumor volume (V) was determined by the following equation: V =
(L x W2) /2, where L is the length and W is the width of a xenograft. Tumor volumes were
measured twice weekly.
[0451] Administration of Compound 36 at 20,60, and 200 mg/kg/day each reduced tumor
growth compared to administration of vehicle alone. Administration of a combination of
Compound 36 and 5FU resulted in greater and dose related inhibition of tumor growth
compared to vehicle. In addition combinations of 60 and 200 mg/kg of Compound 36
reduced tumor growth to a greater degree than 5FU alone.



[0452] Associated with these anti-tumor effects, there was some degree of weight loss and
occasional mortality, particularly in the group treated with the high dose of Compound 36 but
in other groups as well. Overall, Compound 36 showed varying rates of tumor growth
inhibition.
Example 48
[0453] Example 48 demonstrates the usefulness of a compound of this invention in treating
cancer employing a NCI H460, human colon carcinoma xenograft mouse model.
{0454] Female CB17/SCID mice (purchased from Charles River, Cambridge, MA), 7-8
weeks of age, were allowed to acclimatize for at least three days, and handled under
pathogen-free conditions. Human colon carcinoma cell line NCI H460 was obtained from the
American Type Culture Collection. The cell lines were cultured and harvested according to
the procedure described in Example 47 and inoculated at 1 x 106 cells/ animal in the
peritoneal subcutaneous space. When the tumors grew to an average volume of 100 mm3
(day 8), each group of mice was administered for three weeks, as tabulated in the table below:
Compound 25 (2.5 mg/ml in 10% PEG; administration route - i.p.) and Taxol (1 mg/ml in
5% EtOH, 5% Cremophor and 90% saline; administration - i. v. 2 h after administration of
Compound 25). The body weight and tumor volumes were measured as described in
Example 47 above.



qld/qd = every day; q2d = every second day; q7d = every seventh day.
[0455] Results are presented in Table X2 based on tumor volume measurement on day 29
when vehicle treated mice had reached a volume of 946 mm3. Groups of 5 mice receiving
saline or no treatment were added in order to indicate any vehicle effects but are not used for
comparisons in this analysis.

[0456] The results demonstrate that all three regimens for dosing Compound 25 provided
similar degrees of tumor growth inhibition and that combination therapy, particularly with
every day dosing provided additional benefit Each combination therapy was associated with
some degree of weight loss but not large enough to cause any mortality. Overall the results
indicate that Compound 25 is efficacious in this model of lung cancer and provides additional
benefit to that provided by the standard chemotherapeutic agent, taxol.
[0457] Using the mouse to HED conversion, Compound 25 can be administered at a
therapeutically effective dose of about 2 to about 8 mg/kg/day, for the treatment of cancer,
particularly lung cancer, alone or in combination with Taxol™, wherein the daily dose can be
administered with a decreasing frequency of dosing for higher doses compared to lower
doses.
Example 49
[0458] Example 49 describes the usefulness of a compound of this invention in treating
cancer as demonstrated employing a H460, non-small lung carcinoma xenograft mouse
model. Female CB17/SCID mice (purchased from Charles River, Cambridge, MA), 7-8
weeks of age, were allowed to acclimatize for at least three days, and handled under

pathogen-free conditions. Human non-small lung carcinoma cell line NCI H460 was
obtained from the American Type Culture Collection. The cell lines were cultured and
harvested as described in Example 47 above, and inoculated at 3 x 106 cells/ animal in the
peritoneal subcutaneous space. When the tumors grew to an average volume of 100 mm3
(day 8), each group of mice (ten per group) was administered for three weeks, as tabulated in
the table below: Compound 25 (2.5mg/ml in 10% PEG; administration route - i.p.);
Compound 24 (0.3,0.lmg/ml in 10%PEG, administration route - i.p.) and Taxol (1 mg/ml in
5% EtOH, 5% Cremophor and 90% saline; administration - i.v. 2 h after administration of the
test compound).

[0459] The body weight and tumor volume was determined as described in Example 47
above. Results for tumor growth inhibition measured on day 27 are as tabulated below.
Comparisons were made on day 27 because that was the last day of measurements for the
vehicle group and those animals were sacrificed.

[0460] These results demonstrate that daily doses of 3 mg/kg of Compound 24 and 25
mg/kg of Compound 25 inhibited tumor growth and that Compound 25 had a slightly greater
benefit both as monotherapy and in combination with taxol. These effects were accompanied
by mild weight reductions, particularly in the Compound 25 + taxol group.

[0461] Using the mouse to HED conversion, Compound 25 can be administered at a
therapeutically effective dose of 2 mg/kg/day, for the treatment of cancer, particularly lung
cancer, alone or in combination with Taxol™, and Compound 24 can be administered at a
therapeutically effective dose of 0.25 mg/kg/day, for the treatment of cancer, particularly
lung cancer, alone or in combination with Taxol™.
Example 50
[0462] Example 50 describes the usefulness of a compound of this invention in treating
cancer as demonstrated employing a HT-29, human colon carcinoma xenograft mouse model.
Female CB17/SCID mice (purchased from Charles River, Cambridge, MA), 7-8 weeks of
age, were allowed to acclimatize for at least three days, and handled under pathogen-free
conditions. Human colon carcinoma cell line HT29 was obtained from the American Type
Culture Collection. The cell lines were cultured and harvested as described in Example 47
above, and inoculated at 3 x 106 cells/ animal in the peritoneal subcutaneous space. When the
tumors grew to an average volume of 100 mm3 (day 8), each group of mice (ten per group)
was administered for three weeks, as tabulated in the table below: Compound 24 (in
10%PEG), administration route - i.p., administrated 2 h before 5-FU or cisplatin (CDDP; in
saline) on the days the combination therapy was scheduled; 5FU alone (in saline), or CDDP
alone.


[0463] In control groups, tumors were implanted in two locations as part of separate study
of effect of location on control group tumor growth. These results had no impact on the
interpretation of the study and all treatments were compared to the vehicle group with tumors
on the same are of the body. The body weight and tumor volume were measured as described
in Example 47. Tumor growth inhibition measured on day 25 when vehicle tumors had
reached the maximal size and animals in that group were sacrificed is tabulated below

[0464J The results demonstrate that Compound 24 as monotherapy resulted in tumor
growth inhibition of slightly more than 40% whereas combining Compound 24 administered
in combination with CDDP or 5FU provided about 50-70% growth inhibition. According to
this Example, the most therapeutically effective combination was that of Compound 24 and
5FU. The effects on tumor growth were associated with minor decreases in weights of the
mice during treatment; however the mice recovered the lost weight after the end of treatment.
{0465] Using the mouse to HED conversion, Compound 24 can be administered at a
therapeutically effective dose of about 0.25 to about 0.50 mg/kg/day, for the treatment of
cancer, particularly colon cancer, alone or in combination with 5FU or CDDP.
Example 51
[0466] Example 51 describes the usefulness of a compound of this invention in treating
cancer as demonstrated employing a H460, non-small lung carcinoma xenograft mouse
model. Female CB17/SCID mice (purchased from Charles River, Cambridge, MA), 7-8
weeks of age, were allowed to acclimatize for at least three days, and handled under
pathogen-free conditions. Human non-small cell lung carcinoma cell line NCI H460 was
obtained from the American Type Culture Collection. The cell lines were cultured and
harvested as described in Example 47 above, and inoculated at 3 x 10 cells/ animal in the

peritoneal subcutaneous space. When the tumors grew to an average volume of 100 nun',
treatment was initiated in which groups of 10 mice received vehicle (Group 1), CDDP at 3 or
6 mg/kg (Groups 2 and 3, respectively, IVone time), Compound 25 at 50 mg/kg in saline 5
times per week for two weeks (Group 4), Compound 25 at 100 mg/kg every three days for 5
times (Group 5) or the combination of each dose of Compound 25 with either 3 or 6 mg/kg of
CDDP (Groups 6 and 7, respectively). Results for groups receiving 50 mg/kg of Compound
25 are illustrated in the Figure 1. Figure 2 shows similar results for 100 mg/kg of Compound
25.
[0467] These results performed with a saline formulated version of Compound 25
demonstrate significant dose related decrease in tumor volume and increase in tumor growth
delay with a daily dose of 50 mg/kg, and 100 mg/kg with less frequent dosing compared to
that employed for the 50 mg/kg daily dose. These data also demonstrate that both dosing
regimens add to the effects of CDDP in this model.
[0468] Using the mouse to HED conversion, Compound 25 can be administered at a
therapeutically effective doses of about 4 to about 8 mg/kg/day, for the treatment of cancer,
particularly lung cancer, alone or in combination with 5FU or CDDP, wherein the daily dose
can be administered with a decreasing frequency of dosing for higher doses compared to
lower doses.
Example 52
[0469] Example 52 describes the usefulness of a compound of this invention in treating
cancer as demonstrated employing a HT-29, human colon carcinoma xenograft mouse model.
Female CB17/SCID mice (purchased from Charles River, Cambridge, MA), 7-8 weeks of
age, were allowed to acclimatize for at least three days, and handled under pathogen-free
conditions. Human colon carcinoma cell line HT29 was obtained from the American Type
Culture Collection. The cell lines were cultured and harvested as described in Example 47
and inoculated at 3 x 106 cells/ animal in the peritoneal subcutaneous space. When the
tumors grew to an average volume of 100 mm (day 8), each group of mice (ten per group)
was administered for three weeks, as tabulated in the table below: Compound 25 in saline,
administration route - i.p., administrated 2 h before CDDP on the days the combination
therapy is scheduled and CDDP (in saline, IV).
Treatment protocol


[0470] The body weight and tumor volume was determined as described in Example 47.
Data are based on tumor volumes at day 25 when tumors in the vehicle group had reached
sufficient size to require that the mice be sacrificed. The results of inhibition of tumor
growth are tabulated below.

[04711 Tb-e results demonstrate that monotherapy administering Compound 25, formulated
in saline, at 50mg/kg/day and 100 mg/kg/day with a variety of dose regimens results in
inhibition of tumor growth in this model of colon cancer, and that treatment combination of
Compound 25 and CDDP enhanced the effectiveness of Compound 25 for treatment of colon
cancer in this model. These effects were accompanied by modest body weight loss, more so
in the combination groups; the mice recovered the lost body weights after the treatment
ended.

[0472] Using the mouse to HED conversion, Compound 25 can be administered at a
therapeutically effective doses of about 4 to about 8 mg/kg/day, for the treatment of cancer,
particularly non-small cell lung cancer, alone or in combination with CDDP, wherein the
daily dose can be administered with a decreasing frequency of dosing for higher doses
compared to lower doses.
Example 53
[0473J Example 53 describes the usefulness of a compound of this invention in treating
cancer as demonstrated employing a H460, non-small lung carcinoma xenograft mouse
model. Female CB17/SCID mice (purchased from Charles River, Cambridge, MA), 7-8
weeks of age, were allowed to acclimatize for at least three days, and handled under
pathogen-free conditions. Human colon carcinoma cell line NCI H460 was obtained from the
American Type Culture Collection. The cell lines were cultured and harvested as described
in Example 47 and inoculated at 3 x 106 cells/ animal in the peritoneal subcutaneous space.
When the tumors grew to an average volume of 100 mm , treatment was initiated in which
groups of 10 mice received vehicle (Group 1), CDDP at 6 mg/kg (IV one time, Group 2),
Compound 25 at 150 mg/kg in saline, once a week for two weeks (i.p., Group 3), or the
combination of the two agents (Group 4).
[0474] The results shown in Figure 3 demonstrate that 150 mg/kg per week of Compound
25 provided greater reduction in tumor growth than CDDP alone and that the combination of
the two agents resulted in added benefit. These results also indicate that during the two week
period of dosing mean tumor volume did not change indicating complete inhibition of tumor
growth. These data indicate that Compound 25 administered at 150 mg/kg/day as
monotherapy, once a week, is the most effective all the dosing regimens described in the
preceding examples (Examples 47-52). Little change in body weight was observed
suggesting reduced toxicity with this dosing regimen.
[0475] Using the mouse to HED conversion, Compound 25 can be administered at a
therapeutically effective dose of about 12 mg/kg/day, for the treatment of cancer, optionally
administered at a frequency of once every week, particularly non-small cell lung cancer,
alone or in combination with CDDP.
Example 54

[0476] Example 54 describes efficacy of Compound 25 via an ip bolus injection or ip
infusion alone or in combination with Cisplatin in H460 xenografts mouse model. Female
Nu-Foxnlnu homozygous nu/nu mice (purchased from Charles River, Cambridge, MA), 6
weeks of age, were allowed to acclimatize for at least three days, and handled under
pathogen-free conditions. Human colon carcinoma cell line HT29 was obtained from the
American Type Culture Collection. The cell lines were cultured and harvested as described
in Example 47 and inoculated at 3 x 106 cells/ animal in the peritoneal subcutaneous space.
When the tumors grew to an average volume of 100 mm3 (day 8), each group of mice (ten per
group) was administered for three weeks, as tabulated in the table below: compound
Compound 25 (formulated as a 15 mg/ml saline solution, administration route - i.p.,
adrninistrated 2 h before CDDP on the days the combination therapy is scheduled and CDDP
in saline, IV.

[0477] The body weight and tumor volume was determined as described in Example 47.
The results are indicated in Figure 4. The data indicate that, while continuous application of
Compound 25 alone or in combination with CDDP is efficacious, intermittent, such as once a
week dosing can provide greater therapeutic benefit in the treatment of certain cancers such
as non-small cell lung cancer.
Example 55
Compound 25 And Gemcitabine Combination Therapy
[0478] A combination of Compound 25 and gemcitabine was administered to nude mice
that were carrying tumors derived from type MiPaca2 human pancreatic cancer cells.
MiaPaca-2 tumor is a highly invasive, rapidly growing tumor that results in death within 20-

30 days in untreated animals. The tumor cells had been transfected with the gene for red
fluorescent protein. Mice were administered doses of vehicle control, gemcitabine,
Compound 25, Compound 24, or gemcitabine/ Compound 25 combinations or gemcitabine/
Compound 24 were administered i.p., as tabulated below (8 mice/group). Compounds 24 and
25 were formulated in saline and provided by Threshold Pharmaceuticals, Inc. as a dry
powder. Gemcitabine was obtained commercially and prepared freshly according to
manufacturer's instructions.

[0479] Tumors were imaged once weekly until the end of the study at which time open
body images were obtained to confirm effects. In Group 1, the tumors grew rapidly (Figure
5) and resulted in 100% lethality by day 30 (Figure 6).
[0480] Groups 3 and 4 resulted in minor effects on tumor volume and had little effect on
survival. Group 2 significantly reduced tumor volume and prolonged survival. Group 6
provided modest reduction in tumor size but no additional effects on survival. In contrast,
Group 5 demonstrated significantly reduced tumor growth and significantly prolonged
survival compared to Group 2. Five out of 8 tumors in Group 5 regressed rapidly after
treatment and within a short period failed to emit fluorescence (Figure 7).
[0481] Four of these tumors remained at zero fluorescence until the end of the experiment
and the tumors were considered to be cured. No tumors in Group 2 were considered to be
cured. These results demonstrate that combination treatment with Compound 25 and
gemcitabine is of greater benefit in this model of cancer compared to monotherapy with the
stand of care, gemcitabine. These results demonstrate that tumor reduction in animals
administered with a combination of Compound 25 at 30 mg/mg/day and gemcitabine is
significantly greater than that in animals treated with gemcitabine as a single agent.

[0482] Using the mouse to HED conversion, Compound 25 can be administered at a
therapeutically effective doses of about 2.5 mg/kg/day, for the treatment of cancer,
particularly pancreatic cancer, in combination with gemcitabine.
Example 56
[0483] It is recognized that efficacious molecules for treatment of human diseases
including cancer maybe toxic at doses near or sometime much greater than doses necessary to
achieve beneficial effects. To determine appropriate dose and route of administering such a
compound, it is necessary to understand its toxicity. Routinely, initial approaches to
determining the toxic dose involve the use of rodents such as mice to provide preliminary
data that might support the design of similar studies in larger animals and humans. Test
compounds (Compounds 24,25 and 36) were tested in mice as preliminary experiments for
determining doses to be used in larger animals. Compound 25_was tested at doses as high as
300 mg/kg as a single dose and found to cause renal toxicities such as tubular necrosis and
protein spillage into the urine. Transient reductions in white blood cells were also observed.
However, little toxicity was noticed at lower doses (100 and 200 mg/kg). These doses
selected represent an approximation of doses that might be used in larger animals such as rats
and dogs for the purpose of confirming that such toxicities exist and for predicting if renal
function should be measured in humans.
[0484] Although the present invention has been described in detail with reference to
specific embodiments, those of skill in the art will recognize that modifications and
improvements are within the scope and spirit of the invention, as set forth in the claims which
follow. All publications and patent documents (patents, published patent applications, and
unpublished patent applications) cited herein are incorporated herein by reference as if each
such publication or document was specifically and individually indicated to be incorporated
herein by reference. Citation of publications and patent documents is not intended as an
admission that any such document is pertinent prior art, nor does it constitute any admission
as to the contents or date of the same. The invention having now been described by way of
written description and example, those of skill in the art will recognize that the invention can
be practiced in a variety of embodiments and that the foregoing description and examples are
for purposes of illustration and not limitation of the following claims.

WE CLAIM:
1. A. compound of the formula :

wherein each X4 are both CI or Br.
2. A compound as claimed in claim 1 having the formula:

3. A compound as claimed in claim 1 having the formula:

4. A pharmaceutical formulation comprising the compound as claimed in claim
1 and a pharmaceutically acceptable excipient, carrier, or diluent.

5. A composition comprising a compound as claimed in claim 1 for treating
cancer.
6. A pharmaceutical formulation comprising the compound as claimed in claim
2 and a pharmaceutically acceptable excipient, carrier, or diluent,
7. A pharmaceutical formulation comprising the compound as claimed in claim
3 and a pharmaceutically acceptable excipient, carrier, or diluent.
8. A composition comprising a compound as claimed in claim 2 for treating
cancer.
9. A composition comprising a compound as claimed in claim 3 for treating
cancer.



ABSTRACT


Title : PHOSPHORAMIDATE ALKYLATOR PRODRUGS
Phosphoramidate alkylator prodrugs can be used to treat cancer when administered
alone or in combination with one or more anti-neoplastic agents.

Documents:

00257-kolnp-2008-abstract.pdf

00257-kolnp-2008-claims.pdf

00257-kolnp-2008-correspondence others 1.1.pdf

00257-kolnp-2008-correspondence others.pdf

00257-kolnp-2008-description complete.pdf

00257-kolnp-2008-drawings.pdf

00257-kolnp-2008-form 1.pdf

00257-kolnp-2008-form 13.pdf

00257-kolnp-2008-form 2.pdf

00257-kolnp-2008-form 3.pdf

00257-kolnp-2008-form 5.pdf

00257-kolnp-2008-international publication.pdf

00257-kolnp-2008-others.pdf

257-KOLNP-2008-(16-10-2012)-ABSTRACT.pdf

257-KOLNP-2008-(16-10-2012)-ANNEXURE TO FORM 3.pdf

257-KOLNP-2008-(16-10-2012)-CLAIMS.pdf

257-KOLNP-2008-(16-10-2012)-CORRESPONDENCE.pdf

257-KOLNP-2008-(16-10-2012)-DRAWINGS.pdf

257-KOLNP-2008-(16-10-2012)-FORM-1.pdf

257-KOLNP-2008-(16-10-2012)-FORM-2.pdf

257-KOLNP-2008-(16-10-2012)-OTHERS.pdf

257-KOLNP-2008-(16-10-2012)-PETITION UNDER RULE 137.pdf

257-KOLNP-2008-CANCELLED PAGES.pdf

257-KOLNP-2008-CORRESPONDENCE OTHERS 1.2.pdf

257-KOLNP-2008-CORRESPONDENCE OTHERS 1.3.pdf

257-KOLNP-2008-CORRESPONDENCE OTHERS 1.4.pdf

257-KOLNP-2008-CORRESPONDENCE OTHERS 1.5.pdf

257-KOLNP-2008-CORRESPONDENCE-1.6.pdf

257-KOLNP-2008-CORRESPONDENCE-1.7.pdf

257-KOLNP-2008-CORRESPONDENCE.pdf

257-KOLNP-2008-EXAMINATION REPORT.pdf

257-KOLNP-2008-FORM 1-1.1.pdf

257-KOLNP-2008-FORM 13.pdf

257-KOLNP-2008-FORM 18-1.1.pdf

257-kolnp-2008-form 18.pdf

257-KOLNP-2008-FORM 26-1.1.pdf

257-KOLNP-2008-FORM 26.pdf

257-KOLNP-2008-GRANTED-ABSTRACT.pdf

257-KOLNP-2008-GRANTED-CLAIMS.pdf

257-KOLNP-2008-GRANTED-DESCRIPTION (COMPLETE).pdf

257-KOLNP-2008-GRANTED-DRAWINGS.pdf

257-KOLNP-2008-GRANTED-FORM 1.pdf

257-KOLNP-2008-GRANTED-FORM 2.pdf

257-KOLNP-2008-GRANTED-FORM 3.pdf

257-KOLNP-2008-GRANTED-FORM 5.pdf

257-KOLNP-2008-GRANTED-SPECIFICATION-COMPLETE.pdf

257-KOLNP-2008-INTERNATIONAL PUBLICATION.pdf

257-KOLNP-2008-INTERNATIONAL SEARCH AUTHORITY REPORT.pdf

257-KOLNP-2008-INTERNATIONAL SEARCH REPORT & OTHERS.pdf

257-KOLNP-2008-OTHERS 1.1.pdf

257-KOLNP-2008-OTHERS-1.1.pdf

257-KOLNP-2008-PCT REQUEST.pdf

257-KOLNP-2008-PETITION UNDER RULE 137.pdf

257-KOLNP-2008-REPLY TO EXAMINATION REPORT.pdf

abstract-00257-kolnp-2008.jpg


Patent Number 258585
Indian Patent Application Number 257/KOLNP/2008
PG Journal Number 04/2014
Publication Date 24-Jan-2014
Grant Date 22-Jan-2014
Date of Filing 17-Jan-2008
Name of Patentee THRESHOLD PHARMACEUTICALS, INC.
Applicant Address 1300 SEAPORT BOULEVARD, 5TH FLOOR REDWOOD CITY, CALIFORNIA
Inventors:
# Inventor's Name Inventor's Address
1 MATTEUCCI, MARK 135 FAWN LANE PORTOLA VALLEY, CALIFORNIA 94028
2 JIAO, HAILONG 861 COMET DRIVE, FOSTER CITY, CALIFORNIA 94404
3 KAIZERMAN, JACOB P.O. BOX 2498, MENLO PARK, CALIFORNIA 94026
4 AMMONS, STEVE 1568 HUDSON STREET, REDWOOD CITY CALIFORNIA 94061
5 DUAN, JIAN-XIN 628 MACIE CIRCLE, SO. SAN FRANCISCO, CALIFORNIA 94080
PCT International Classification Number A61K 31/685
PCT International Application Number PCT/US2006/025881
PCT International Filing date 2006-06-29
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/695,755 2005-06-29 U.S.A.