(12) STANDARD PATENT (11) Application No. AU 2011202075 B8 (19) AUSTRALIAN PATENT OFFICE

(54) Title Phosphoramidate alkylator prodrugs

(51) International Patent Classification(s) C07H 19/10 (2006.01) C07H 19/00 (2006.01)

(21) Application No: 2011202075 (22) Date of Filing: 2011.05.05

(43) Publication Date: 2011.05.26 (43) Publication Journal Date: 2011.05.26 (44) Accepted Journal Date: 2013.01.17 (48) Corrigenda Journal Date: 2013.05.30

(62) Divisional of: 2006263433

(71) Applicant(s) Threshold Pharmaceuticals, Inc.

(72) Inventor(s) Jiao, Hailong;Kaizerman, Jacob;Matteucci, Mark;Duan, Jian-Xin

(74) Agent / Attorney Spruson & Ferguson, L 35 St Martins Tower 31 Market St, Sydney, NSW, 2000

(56) Related Art US5472956 A MISIURA K. et al: Acta Biochim. Polonica, 2002, 49(1)169-176. 2011202075 05 May 2011 5

AH21 alone

(4064035

Phosphoramidate or

IkRTK in

combination PHOSPHORAMIDA

alkylator with

one

prodrugs or

more

TE Abstract

anti-neoplastic

ALKYLA can

be

used

TOR

to

agents.

treat

PRODRUGS

cancer

when

administered 2011202075 05 May 2011 The performing 5845c(4166805_1) Actual of Address Name Invention

following

Applicant

and Inventor(s):

it for

Title:

known Address statement

Service: :

to

me/us:

is

a

full

description COMPLETE FOR Threshold Jian-Xin Jacob (CCN Sydney 31 St Spruson Mark Hailong United Boulevard, Phosphoramidate

Martins Market PATENTS

A

Matteucci

3710000177)

Kaizerman States STANDARD

AUSTRALIA

Jiao NSW Duan &

of

Pharmaceuticals, Street Tower Ferguson 5th

SPECIFICATION this

2000 of

ACT

Floor,

America invention,

Level alkylator

1990 PATENT

Redwood

35

including prodrugs

Inc.,

City,

of

1300 the California,

best

S&FRef: Seaport

method

94063,

838876D1

of

2011202075 05 May 2011 30 25 20 15 10 5

.

the al., imidazo alkylation other released chemotherapeutic Pharmacological Cyclophosphamide or DNA macromolecules alkylators. pharmacology, encompass Description present Field 60/695,755 [0004] other [0003] [0001] [0002]

an

formation supra}.

nucleophilic aziridinium hyperproliferative of alkylation

invention

groups Invention alkylates Phosphoramidate This The Alkylating

of

The a

filed of

healthy of diverse

PHOSPHORAMIDArE present application

Related

and

of

neighboring-heteroatom-stabilized chemotherapeutic

is such

29

cation. CROSS-REFERENCES Basis

generally nucleophilic agent

biomolecules moieties postulated

and and medicine.

Jun

agents cells

group

invention as

Art

Ifosfamide , of

2005, disease DNA If

claims canand BACKGROUND

Therapeutics,

based

such of ( the relates “

alkylators to

chemicals

which under moieties

active

provides

conditions be of

as

benefit alkylators result

alkylators

to healthy an are the are

physiological is the

alkylators important

phosphate,

each

incorporated an in

such

2001, that of compositions or fields

unwanted important

TO non-cancerous

with

U.S. used “

activated

ALKYLATOR OF

mustards act have as

RELATED

1389-1399,

of

the phosphoramidate THE are

mechanism as provisional in

chemistry,

amino,

the onium

strong conditions

DNA cancer released

herein toxic subclass

in INVENTION ” ability

and )

the used

within

sulfhydryl, intermediates electrophiles, cells,

APPLICATIONS events therapy, McGraw-Hill, methods

by

liver

patent in

away

biology, to

in of

(see

reference.

the

PRODRUGS

alkylate can cancer chemotherapeutic the

and

in alkylator

from antitumor Hardman application such

patients get for tumor

hydroxyl, the molecular

treating alkylated. such for the biologically

as active

New

cells

prodrugs. example,

tumor,

(see

et activity as

No.

York, al.,

carboxyl alkylator an cancer

to biology,

Hardman

act Such DNA alkylators. The

vital

through of USA).

The as

and

a and and

et

2 2012

Dec

Cyclophosphamide ^nfj Ifosfamide 06

[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.

2011202075 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-T wherein Aik is a

phosphoramidate alkylator, T is L-Z3 wherein L is a linker Z3 is a bioreductive group.

[0006a] In another aspect there is provided a compound having the formula:

wherein

L is selected from the group consisting of CH2, CHMe, CMe2,

1

and Z3 is selected from the group consisting of

6554197_ I.RTK 2a 2012

Dec

HO 06

2011202075

each R.g is independently selected from the group consisting of hydrogen, aryl, heteroaryl, C1-C4

alkyl, C1-C6 heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, C1-C6 acyl, Ci-C6heteroacyl, aroyl,

heteroaroyl, C1-C6 alkoxycarbonyl, Cj-Cg alkylaminocarbonyl, di C1-C6 alkylaminocarbonyl and

C1-C6 alkoxy;

each X4 is selected from the group consisting of halo, alkylsulfonyloxy, heteroalkylsulfonyloxy,

cycloalkylsulfonyloxy, heterocyciylsulfonyloxy, arylsulfonyloxy and heteroarylsulfonyloxy;

or a pharmaceutically acceptable salt thereof.

[006b] In another aspect there is provided a method to synthesize a compound of the first

aspect comprising reacting a compound having the formula Z3-L-OH, a trisubstituted phosphine,

a compound of the formula:

6554I97_| RTK 2b 2012

Dec

06

and a dialkyl azodicarboxylate to yield the compound of claim 1.

[0007] In one aspect, the present invention provides phosphoramidate alkylator prodrugs of formula (I):

2011202075 Rs

(0

wherein

[0008] Y| is O, S, NR^. or NSO2R.6 wherein each R.6 is independently C1-C6 alkyl, C1-C6 heteroalkyl, aryl, or heteroaryl;

[0009] Y2 is O, S, NR^, NCOR6, or NSO2R6; [0010] each of R1-R5 independently is hydrogen, hydroxyl, amino, Ci-Ce alkyl, C1-C6 heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, C1-C6 alkoxy, C1-C6 alkylamino, C1-C6

dialkylamino, aryl, heteroaryl, C1-C6 acyl, C1-C6 heteroacyl, aroyl, or heteroaroyl; or together

6554197 I RTK any two of R1-R5 form a C3-C10 heterocycle; or each of Ri — Rs independently is a Trigger T wherein T is L-Z3

[0011] L is selected from

-[C(Zl)2-Y3]v-[C(=O)-O]q-[C(Z1)2-Z2-Y4]u-[C(Zi)2]z-[-C(Z1)=C(ZI)]g-Z3and 5 -[C(Z1)2-Y3]v-(S(=O)2)q-[C(Zl)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, CpCe heteroalkyl, C3-Cg cycloalkyl, heterocyclyl, Ci-Cg alkoxy, Ci-C6alkylamino, Cr Ce dialkylamino, aryl, heteroaryl, Ci-Cg acyl, Ci-Ccheteroacyl, aroyl, or heteroaroyl;

10 [0013] Y4 is O, S, or -NR7-C(=O)-O-;

[0014] each Zj independently is hydrogen, halogen, Cj-C6 alkyl, C]-C6 heteroalkyl, aryl, heteroaryl, C3-C8 cycloalkyl, heterocyclyl, Ci-C6 acyl, CrC6 heteroacyl, aroyl, or hetero aroyl;

[0015] Z2 is C1-C6 alkylene, C]-C6 heteroalkylene,

15

wherein each Xi is independently N or CR8, each R8 is independently hydrogen, halogen, nitro, cyano, CO2H, C)-C6 alkyl, C]-C6heteroalkyl, Ci-C6 cycloalkyl, CrCe alkoxy, Cj-Ce alkylamino, CrC6 dialkylamino, aryl, CONCR-^, CrC6 acyl, Ci-C6heteroacyl, aroyl, or heteroaroyl;

20 [0016] X2 is NR7, S, or O; and

[0017] Z3 is selected from the group consisting of:

3 2011202075 05 May 2011 15 10 5

J

phosphoramidate ------pharmacophores, in 0.01 heteroalkylsulfonyloxyalkyl; heteroalkylsulfonyloxyalkyl; [0023] [0022] Ce alkylsulfonyloxyalkyl, [0021] [0020] [00191 [0018]

an

alkylsulfonyloxyalkyl,

nm.

oxidation-reduction

[0024] In an (iii) (ii) (i) with In

one individual at a at

NR2R3

related the least

least

embodiment,

alkylator a proviso

pharmaceutically

two

one

and embodiment, In

isomer 2-heteroalkylsulfonyloxyalkyl,

of

a of NR4R5

2-heteroalkylsulfonyloxyalkyl, reaction. that related

prodrugs R1-R5

R1-R5

and Z3

or in

both

is

a formula

are at embodiment, is

racemic a the

least

selected

having

bioreductive selected acceptable together

present

one (I):

or

IC50

from

of

from non-racemic

4

are"^; invention

the

NR2R3

salt, or

group the

the present GI50,

solvate,

group

2-arylsulfonyloxyalkyl,

group

and and

that

provides in 2-arylsulfonyloxyalkyl,

mixture invention

cells NR4R5

consisting

can hydrate, consisting

under accept

phosphoramidate

is of

'" provides

or isomers,

hypoxia, NC1;

of

one of

a

2-haloalkyl, prodrug

2-haloalkyl, or

or

and more bioisosteres,

no of

and

2

2- thereof. 50

alkylator electrons

2-

μΜ 2-Cj-

2-

to

) 2011202075 05 May 2011 30 25 20 15 10 5

phosphoramidate hyperproliferative or of acceptable comprising phosphoramidate [0028] [0029] treatement HCR nM alkylator HCR, detail hypoxia [0027] invention normoxic present by prodrugs up prodrugs [0026] alkylator tide another cytotoxicity of cellular [0025]

third such

a the to

compounds to compound

of

1000

further

measured

50 relative

therapy.

fine invention

related cytotoxicity 50 of

In

In In prodrug In In prodrugs which having

μΜ has

cells

excipient, of

to fold

50 the one one one a therapy, another is

later cancer related and a 10,000.

μΜ

measured IC50

phosphoramidate

as

embodiment, hypoxic in in aspect, less aspect, aspect are by In hypoxic

alkylator

alkylator of provides

measured a hypoxic diseases hypoxic

in to

having

one the values

HCR up

related

toxic

the

or is and the embodiment, carrier, 0.01

the measured to

ratio is the the

embodiment, present

cytotoxicity

application, by

other

of cytotoxicity 5000 a

in

nM

IC50

present

in prodrug present and present comprising phosphoramidate prodrug and relapsed embodiment,

by 25 clonogenic of corresponding

or hypoxic the

and

hyperproliferative

normoxic normoxic values, fold to normoxic the

diluent. invention

present alkylator by

100,000.

invention

which phosphoramidate invention invention relative

of

of

less antiproliferation cancer.

of of

the

and

the which the in administering

0.1 assays 10

toxic

invention

cancer is the cells. and cells

cells. invention

normoxic has present

prodrugs

to IC50 nM normoxic

up In In

provides

present are provides provides alkylator

hypoxic 100,000. in 5

a

under another to and

another

to hypoxic In

values treated corresponding up

diseases. 1000

invention 50

a provides using

to

or of

related

cells.

alkylator hypoxia, invention

assays μΜ a

cells. novel

a a a

prodrugs cytotoxicities,

one the related in fold therapeutically embodiment,

In is

pharmaceutical million method

cytotoxicity

the and

hypoxic

resistant a invention

In that

less embodiment, phosphoramidate related has phosphoramidate In and relative

another a

prodrug

embodiment, of

provides a

is hypoxia normoxic

having fold, toxic a of using related

50

known,

hypoxic and

treating to embodiment,

μΜ

and the Ci

up of and related first in

the normoxic effective of

0 an

0.1

embodiment, cytotoxicity formulation phosphoramidate

, to corresponding to cancer

a

defined the the C cells to

relative

line, IC50 cytotoxicity pharmaceutically

0.01 10,000 5 cancer

nM phosphoramidate o, a

cellular present embodiment,

alkylators patient alkylator or

as

in second

treated to nM. amount

cells.

C90

the measured in

cells IC$o and 5 fold,

greater

μΜ

ratio, values

In in the

other line,

of in value is

and

need of for and

a

0.1

the a

of

a

2011202075 05 May 2011 30 25 20 15 10 5

the herein chemical defined, invention, metastatic I. section invention organized and invention of the cancer on the [0036] [0035] combination [0032] [0030] [0034] [0031] [0033] .

Definitions

treatment,

compounds tumor

H460 H460 biological

are

agent. is

all

The

The Figure Figure Figures Figure Figure and

growth xenograft xenograft applicable

is alone

intended into or cancer.

terms

organized

with

one therapies, on medical following detailed

assays sections of

or tumor 3 4 2 1

in

5, that

of the

CDDP demonstrates demonstrates demonstrates demonstrates in

In

to the 6 mouse mouse

DETAILED art,

to

combination;

invention

for another and is have

arts. description

as

growth BRIEF any

only administrations, H460

definitions known, notations

the

on follows: 7

model. model.

the aspect Tn demonstrate

for tumor compounds

xenograft embodiment,

some

in DESCRIPTION

meanings

and the the the the the is

the DESCRIPTION

of and of Section administered

and

growth are

methods effect convenience effect effect effect cases,

the

H460 the

other

provided

Section

mouse

and different

invention. the of commonly

of of of of

I

terms in

xenograft

the

provides the scientific

effect

formulations, Compound Compound Compound Compound for the

model. phosphoramidate

invention. IV OF

in

making

to of with

H460

aspects

OF

combination provides of

assist

THE the

understood

useful

mouse

Compound or THE commonly

reader, xenograft

25 25

them; 25 25 medical

DRAWINGS the

and

employing This

INVENTION (150 (50 dosed (100 definitions;

examples

model. reader.

embodiments

and Section

mg/kg with

detailed

by

mg/kg

mg/kg

alkylator 25 terms understood mouse

in

disclosure those

in combination at Unless

the of

)

combination

III least

or )

) Section

on

description model.

synthetic

dosed on

compounds of

describes terminology prodrug

tumor

of

skill otherwise another tumor

meanings

found

the

in II

in

with growth present describes methods

of growth

the

with methods is in anti

of the

any CDDP used are

­ the

in

in

2011202075 05 May 2011 30 25 20 15 10 5

electrophile macromolecules heteroaryl, hydroxyl, "Phosphoramidate carboxamido, ethenyl, [0040] further (C2-C6) monovalent heteroarylsulfonyloxy. and aryl, nitro, the herein alkylcarbox-amido, ( the prefix saturated definition [0039] can the Cg defined an [0038] [0037] “ D

integer

alkyl

prefix. radical ” like. be containing

), heteroaryl,

ethenyl,

is hydroxyl, should further optionally

alkenyl herein

ethynyl, includes not

"Alkylator" For "Alkenyl" As "Alkyl" monovalent

amino,

of from or alkyl

As

is

hydrocarbon included used

the

each

portion

-SO2NH2, not

present optionally ethynyl,

used for

at

includes, 2-20,

alkylsulfonyloxy,

via or term

Ci-C amino,

mono methyl, means least be herein, necessarily

of clarity alkylator"

heteroalkylsulfonyloxy,

means -SO2NH2, in

an

substituted

means the

thereof have to as hydrocarbon 6 or

this

one

CpCi electrophillic alkoxy,

or

indicate -OSO mono generally a

generated definitions

be radical ethenyl, and/or "a"

ethyl,

linear

di(C]-C

disclosure, a double the

substituted a

linear

means

will or

be alkoxy, reactive

2 -OSO same or -(Ci-C6)

C

n-propyl,

"an"

with for construed saturated having the

r

di(Cj-C6) have

propenyl, 6 understood bond, heteroalkylsulfonyloxy,

C6 monovalent ) by radical

ready

an herein 2 meaning.

alkyl number means

reaction -(Ci-C6)

substituents,

Ci-Ce

alkylthio, the intramolecular moiety

six alkylator

with

alkyl, the but

prefixes 2-propyl,

reference, amino, monovalent

or having to

(e.g.,

alkyl and

number "at alkylthio,

1,3-butadienyl no substituents, of

fewer represent

with 7 capable in alkyl, mono For

hydrocarbon

least

main

more

aryl

-COOH,

for the alkyl,

amino,

halo,

the

example, including (Ci-C a

main

n-butyl,

which of mono art. one" nucleophile or

and or

chain than cyclization. number

of

-COOH,

hydrocarbon

carbon alkenyl, heteroarylsulfonyloxy. C a di(Ci-Ce)

qq

halo, the 2 substantial

forming

chain -CONH2, including

-C or ),

three

or an arylsulfonyloxy carbon and

2-butyl, Ci-qq (Ci-Cg) 6 for radical inclusion "one

di(C|-Cg)

alkenyl aziridine of C2-C6 atoms

carbon

-CONH2,

the alkoxy, double example,

carbon on

alkylsulfonamido, a or ,

atoms

or

like. mono-

covalent

for radical

the or

difference

alkyl, more." tert-butyl, alkenyl indicated

ether, Ci-C

of

atoms. a

or example, bonds.

alkylsulfonamido,

macromolecule. araalkyloxy),

atoms branched

Alkenyl such

in mono- deuterium

aziridinium or Cj-s qq

or or an cyano,

, alkyl

ether,

di(C[-C6)

wherein

a

definitions

(Ci-Ce)

indicated in alkyl over pentyl,

For alkyl, branched

or

deuterium the

can linkage

cyano, nitro, example, difCj-Ce)

the portion, aryl,

(“

prefix

when

or be

qq D

alkyl and

alkyl-

C ” in

), is

r to

a

2011202075 05 May 2011 30 25 20 15 10 5

More amino, alkyl, optionally ring naphthyl, specifically phenylalkyl), independently integer integer [0044] cycloalkyl, alkylamino, cycloalkyl, preferably heteroarylsulfonyloxy. alkyl, methyl-propylene, to of CH2CH2OCH2CH2-, cyano, deuterium to the one alkylsulfonamido, (Ci-C6)alkyl-carboxamido, [0041] [0042] [0043]

twelve twelve 6

alkylene

or carbons. to

specifically, optionally cycloalkyl,

more nitro, mono-alkylamino, 10 from from

"Cycloalkyl"

"Heteroalkylene" "Alkylene" "Aryl"

and carbon carbon

ring substituted ( one,

cycloalkylalkyl, cycloalkylalkyl, “

the

di-alkylamino,

heteroatoms biradical. D 0 0 ethenyl,

The the

hi ” selected to to atoms

), term two,

refers

substituted one

atoms atoms 5, 5, hydroxyl, substituted

cycloalkyl-alkyl,

the cycloalkyl aryl, pentylene,

R R

three, aryl

-CH2CH means

embodiment, which

’ ’ ethynyl,

independently

term

refers and to and

from For heteroaryl, or For optionally

includes,

a (i.e.

di-alkylamino,

a

four R R monovalent has cycloalkyl

example,

haloalkyl, -SO2NH2, amino, phenyl, phenyl example halo, branched a is

hydrogen, ”

” to forms

group 2 linear hexylene,

oxygen, N(CH3)CH

substituted C1-C6 are are

essentially a ot

monovalent

nitro,

independently independently five

substituted

but

R phenyl alkyl mono

thereof. or

can

or saturated

with x alkoxy, heteroalkylene alkylene

saturated haloalkoxy, and phenylalkyl) -OSO includes, sulfur, is substituents -C(O)R

alkyl,

monocyclic cyano, have

and

or not

hydroxyl,

2 or one,

independently

the R CH2-, or

heteroalkylsulfonyloxy,

2 y

the di(Ci-C6)alkyl limited -(Ci-C6)

C1-C6

phenylalkyl),

one together

nitrogen cycloalkyl, cyclic divalent

meaning with 8 includes

Z two, hydroxyl,

divalent (where for like.

hydrogen -CH hydrogen

or

heteroalkyl,

substituents selected alkylthio, example, or three alkoxy,

or hydrocarbon more includes, to,

2

alkyl,

CH -(CR is hydrocarbon

bicyclic and/or

methylene, given R phenyl,

hydrocarbon

cycloalkyl

with z or alkoxy, cycloalkylalkyl, 2

double is SCH2CH

or or

-(CR

amino, or from four mono R

hydrogen,

cyclopropyl,

above phosphorous) -COOH, ” alkyl, one alkyl,

-CH

optionally

)

COR aromatic including n biphenyl,

’ -CONR

substituents

R amino, deuterium

bonds radical

to

2 or ” ethylene,

halo,

OCH

2 or radical )

and and foran and

-,

eight n (where

di(Ci-Ce) radical -COOR

heterocyclyl. and -CONH2,

alkyl,

aryl R x R and hydrocarbon 2 C

acylamino,

of R

substituted O-,-

for

x alkylene phenyl 1 2 substituents,

cyclohexyl, is the

y having -C6 -naphthyl,

and

three

may

(

(where R propylene,

can

or having hydrogen, “ example,

selected

haloalkyl,

(where D

like. is

alkenyl

R ”

mono- also be

hydrogen, ), y to or

from are except alkyl,

present

n seven

mono from More

phenyl).

be

n is radical

and from

ether,

is

alkyl, 2- or an one

an one

2- ­ that

di-

in

2011202075 05 May 2011 30 25 20 15 10 5

hydrogen haloalkoxy, hydroxyl, cycloalkyl, (CR alkyl, or (CR optionally ring attachment selected -S(O) indicating by 2-hydroxyethyl, hydrogen, radical atoms 2-methylsulfonyl-ethyl. alkylamino, (provided di-alkylcarbamoyl. cycloalkyl-alkyl, integer [0047] [0046] aryloxycarbonyl, substituents dimethylaminocarbonyl-cyclohexyl, cyclohexenyl, [0045]

four amino,

’ ’ atoms R R

P

and

” R ” in

is substituents, )n-CONR ) from

Z n

from

the

-COOR "Heteroaryl" In

portions. "Heteroalkyl"

through or

alkoxy, R that halo, substituted the alkyl,

having

point cycloalkyl-alkyl,

one heteroalkyl,

di-alkylamino, independently is alkyl, portion 0

N,

number

phenylcyclohexyl, n hydrogen, to

fluoro, embodiment,

is

2,3-dihydroxypropyl,

cycloalkyl,

O, of x aryl, 2 carboxamido, (where

a amino, R at

and 0),

),

carbon the y

or R selected of

least with (where x

independently alkyl,

means of

aralkyl,

S, R

is the

heteroaryl alkylamino, means

x -COR

For

n carbon the

hydrogen, alkyl, acylamino,

and one

the

heteroalkyl

is selected atom

or

cycloalkyl, cycloalkyl-alkyl,

phenyl n each from remaining

an a

R

understanding

R hydroxyalkyl. aromatic

is alkoxycarbonyl, an

monovalent (where x cycloalkyl, mono-

y

integer

atoms

of

an and 4-carboxycyclohexyl, are,

alkyl

of radical alkyl,

the

from integer di-alkylamino,

alkyl, or

with the and

mono-alkylamino, R independently

2-methoxyethyl, R group phenylalkyl). y or heteroalkyl

(e.g., radical

ring ring from cycloalkyl-alkyl,

together

above,

cycloalkyl, is the

cyano, will di-alkylcarbamoyl one cycloalkyl,

hydrogen, cycloalkyl-alkyl, from monocyclic,

containing atoms

like.

that Ci-Cio)

0 exclusive

Representative

aryl,

9 to be

as

to aryloxycarbonyl, R

-OR

eight

0

the on is w defined 5,

,

radical. to being

araalkyl, OH cycloalkyl

R

R an cycloalkyl-alkyl, of W point

refers

x In 5,

, alkyl, cycloalkyl-alkyl, ’ substituents, ,

-NR each aromatic of or and benzyloxymethyl, one, R

2-carboxamidocyclohexenyl,

R one di-alkylamino, bicyclic

y C, herein aryl,

the , alkoxy.

of

and to and x

R R

phenyl with R alkoxycarbonyl, other, two, embodiment, or phenyl examples

w attachment ” cyano, the

y

or araalkyl,

,

is are alkylsulfonyl.

R R

and with

ring.

carboxamido, ” or heterocyclyl. z the hydrogen,

or total

can

independently Additionally, hydrogen, are

preferably

or tricyclic three

-S(O)pR or

understanding -OR

one, halo,

phenylalkyl,

independently The number be

phenylalkyl), include,

haloalkyl, aryl amino,

of

W

further ring 2-cyanoethyl, R two ,

heteroaryl

nitro, the -NR x

or alkyl, z radical

and alkyl,

(where

one,

R heteroatoms or

araalkyl. of

heteroalkyl mono or for z x

the

R

substituted

three is

hydrogen cyano,

carbon R

mono- cycloalkyl,

y

two,

example, y , - hydrogen that

of

prefix or

together

or ­

p 2- ring

5

is

-

three the

and

to R

or an

y

is

12 or is

) 2011202075 05 May 2011 30 25 20 15 10 5

within portion piperazinyl, number hexahydro-lA piperidinyl, methylpyrrolidin-3-yl, includes, phenylalkyl), hydrogen phenylalkyl), [0049] cycloalkyl, independently haloalkoxy, halo, ring selected where P(=O)OR pyrazolotriazinyl, benzothienyl, benzoxazolyl, from cyclic triazolopyridinyl, pyrrolyl, limited is and isobenzofuranyl, constituent [0048]

cycloalkyl

the can

nitro, O,

the

radical one

of of to,

derivatives

NR from be

"Cj-Ce but pyrazolyl,

"Heterocyclyl" W or ring the

carbon

pyridyl, or ,

cycloalkylalkyl, cyano,

optionally ring morpholinyl,

and or -COR

alkyl, (where

is

or

or two -(CR cycloheteroalkyl 6 of

alkyl, indazolyl,

hydrogen quinolyl, -thiopyran-4-yl, can

S(O) not

heterocyclyl.

piperidin-2-only atoms. -(CR

Acyl" 3

benzothiazolyl,

pyrazolopyrimidinyl, atoms C

triazolotriazinyl, hydroxyl, to be

and ’ (where limited

furanyl, thereof. R

P pyridazinyl, R aryl, atoms

” (where 8 ’

any

R )

substituted is

2-pyrroIidon-l-yl, ring R n means

-COOR

tetrahydroquinolinyl,

(e.g., or hydrogen, pyrrolopyrymidinyl,

) or is

pyrrolidinyl, arylalkyl, n arrangement

R -CONR

to, can

alkyl, hydrogen, atoms "cycloheteroalkyl" thienyl, phenyl

Unless is p alkoxy,

pyridyl, -CO-(Ci-C« More C3-C10)

is hydrogen, optionally or tetrahydroimidazo

(n pyrimidinyl,

an benzoisothiazolyl, and R*

heterocyclyl independently in x

is R

heteroaryl, pyrazolotetrazinyl, or alkyl,

integer indicated thiazolyl,

specifically

and which y amino,

an the

phenylalkyl). refers (where alkyl,

tetrahydropyranyl, tetrahydrofuranyl, allowed

triazolopyrimidinyl,

integer

R

derivatives furyl, be alkyl, cycloalkyl,

y

alkyl), from one

are,

mono-alkylamino, cycloalkyl, replaced to

benzofuranyl, 10 otherwise, indolizinyl, isoquinolyl, n isothiazolyl,

group heteroarylalkyl, means

the

by quinolyl,

to is

from cycloalkyl,

independently the with 0

an

wherein [4,5-c] four

the to total

term benzotriazolyl,

More

thereof. integer

exclusive cycloalkylalkyl, 2), 0

by one, a

bonding

hexaaza-indenly, ring

to

saturated

number cycloalkylalkyl,

the

the

a

thienyl,

heteroaryl tetrahydrothiofuranyl, pyridinyl, pyrazolopyridinyl, N-methylpiperidin-3-yl,

5,

benzimidazolyl, two, carbonyl triazolyl, specifically

the

cycloalkylalkyl,

atoms

tetrahydrobenzofuranyl,

arrangement remaining R pyrrolotriazinyl, from The

term

characteristics of cycloalkyl, of di-alkylamino, and three

of

benzothienyl, or

each the 0 prefix are

carbon

indolyl, R

alkyl unsaturated imidazolinyl, to includes, group. imidazolyl,

or

” number

phenyl heteroatoms

5, the other, ring are

four

and

phenyl indicating R of is

atoms term independently ’ benzisoxazolyl cycloalkylalkyl,

The

as atoms isoindolyl, the

and phenyl

heptaaza-indenyl substituents

hydrogen, or

but

of of defined haloalkyl,

pyrrolidinyl,

hetero heterocyclyl phenylalkyl), non-aromatic heterocyclyl 1,1-dioxo- or R isoxazolyl,

heteroatoms. the

in is

being

N-

selected

the the or not are

atoms above.

alkyl,

C,

or

[0050] "C|-C6 Heteroacyl" means -CO-(Ci-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 5 above.

[0053] "Rsuisulfonyloxy" means Rsui-S(=O)2-O- including alkylsulfonyloxy, heteroakylsulfonyloxy, cycloalkylsulfonyloxy, heterocyclylsulfonyloxy, arylsulfonyloxy and heteroarylsulfonyloxy wherein Rsui is alkyl, heteroakyl, cycloalkyl, heterocyclyl, aryl and heteroaryl respectively, and wherein alkyl, heteroakyl, cycloalkyl, heterocyclyl, aryl and 10 heteroaryl are defined above. Examples of alkylsulfonyloxy include Me-S(=O)2-O-, Et- S(=O)2-O-, CF3-S(=O)2-O- and the like, and examples of arylsulfonyloxy include

'----- ' · '----- ' · '----- ' and the like. Alkylsulfonyloxy, heteroakylsulfonyloxy, cycloalkylsulfonyloxy, heterocyclylsulfonyloxy, arylsulfonyloxy, and heteroarylsulfonyloxy groups can be leaving groups in phosphoramidate 15 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 Rjui moiety and can provide phosphoramidate alkylators and prodrugs thereof which are 20 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’”, -OC(O)R’, -C(O)R’, -CO2R’, -CONR’R”, -OC(O)NR’R”, -NR”C(O)R’, -NR’-

C(O)NR”R’”, -NR”C(O)2R’, -NH-C(NH2)=NH, -NR’C(NH2)=NH, -NH-CtTJH^NR’, -

25 S(O)R’, -S(O)2R’, -S(O) 2NR’R”, -NR’SCO^R”, -CN and -NO2, -R’, -N3, perfluoro(C]-

C4) alkoxy, and perfluoro(C]-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_g cycloalkyl, C2_8 alkenyl, C2_g alkynyl, unsubstituted aryl and

heteroaryl, (unsubstituted aryl)-C]-4 alkyl, and unsubstituted aryloxy-Cj-4 alkyl, aryl

11 2011202075 05 May 2011 30 25 20 15 10 5

base, by present particular compounds or ( [0056] with atoms regioisomers invention encompassed (optical -NR from a substituents ring the the by membered nitrogen groups, substituted [0055] 2 C(O)-(CH (CH and Other 14 NR substituent

C). ηοζ

contacting

an

aryl aryl

q 2 ’

so radioactive

’ -

either 0 )r-B-,

-

is alkylene

that suitable All and

are or to invention

formed

or centers) or or

an The atom,

Certain a

substituents may 3,

2

isotopic intended

unsubstituted constitute -S(O) heteroaryl heteroaryl

ring.

integer single ) neat with

which wherein and on q

of

and term within -U the

substituents also

may tether they adjacent

the

3 or 2 isotopes, X or

1-3

-, For compounds neutral

contain individual NR

bond,

5

are

of "pharmaceutically variations in

wherein double contain formula

to is

optionally can A halogens, the

such example, ’

of ring a ring from - prepared -O-, found be

and suitable is

be from

and scope atoms

aryl-Cj-4

form

encompassed relatively

selected

such

may may compounds.

bonds;

-NR B 0 include combined

unnatural

T

-(CH

isomers

r on

to

2 of of

are

1-4 is

of

unsubstituted of

and be -NR of

as optionally

optionally ’ with inert 2.

the

-, the the an

independently

such 2

the

carbon the for replaced -S-, the )

from

Alternatively,

U alkyl

s integer each ’ compounds acidic present compounds

R -X

relatively (e.g.,

3 solvent.

present

example proportions aryl acceptable racemates, with

compounds -S(O)-, are

5 within

is For

-(CH hydrogen

of

groups. atoms.

meant

be be or independently

functionalities,

separate

of the

with the

invention example,

heteroaryl C]-g

2 replaced replaced invention.

from 12

)t-,

-S(O) Examples

the nitrogen tritium nontoxic above

described -CH2-, of

a

Two

to salts" diastereomers,

two

When

of where

double scope or alkyl,

with

the include 1 enantiomers) 2

atomic unsubstituted

to

-,

aryl

of of the possess

with ( with

present

3

is

or

ring atom 3. -Ο-,

a H),

R

acids the the The

of C] s

of

-NH-,

meant compounds bond.

sufficient

base herein. ’ substituents -S(O) and

One

the and a 1-pyrrolidinyl a

salts -g

iodine-125 isotopes substituents substituents may -NH-,

substituent substituent compounds to

or asymmetric

alkoxy invention,

t present

addition

R -O-,

of

form 2 are to Alternatively,

geometric

derived bases,

NR are optionally ”

When

the include Cj-g -S-, are

independently

amount

-CH2-

’ all at

a -.

may or

single

attached

attached

invention.

3-, ( -S(O)-, one depending

salts intended

The

l25 C[-g

of on on alkyl. of

from compounds whether

of

and

4-, carbon be

I) salts

isomers,

the the

or

or

adjacent adjacent of the be

bonds substituent or

can

radiolabeled 5-,

a thioalkoxy 4-morpholinyl.

more

the

two replaced formula -S(O)2-, formula

to to carbon-14 present single of

6-, be

to

radioactive atoms a

the desired integers

on the

of of

ring be

obtained of

or

atoms atoms of

the

same the the

bond, active

the

7-

-A- -T

the

with -S(O) R atom

new

2 ’

-

of

in of of

i

2011202075 05 May 2011 30 25 20 15 10 5

are unsolvated invention. solvated compound as form [0058] a benzenesulfonic, propionic, [0057] to Salts glucuronic the the dihydrogenphosphoric, nitric, trimethylamine, piperazine, glucosamine, pharmaceutically-acceptable addition either invention ethanolamine, dibenzylethylenediamine, occuring like. contacting secondary ferric,

base

be solubility

salts present like,

” of converted Salts ,

neat

or carbonic, ferrous, Journal

the

of

forms,

as salts

Certain The acid

amines

contain

isobutyric, Certain for amino and forms

the or or derived invention well compound piperadine,

in

neutral

galactunoric in include histidine, and

the

neutral ethylenediamine,

polar of lithium, tertiary

including

into

as

monohydrogencarbonic, a tripropylamine,

compounds

and p-tolylsulfonic,

and

Pharmaceutical acids purposes relatively isolating suitable

compounds the from

either

solvents,

forms

contain the

are malonic, those

form

salts differs

amines,

polyamine such magnesium, sulfuric, hydrabamine,

pharmaceutically-acceptable like, intended hydrated

diethylamine, base inert

acids

the of of

derived derived of basic

as

of

both inorganic

from

but such

the the

such

of parent

benzoic, including arginate

or the solvent.

N-ethylmorpholine, monohydrogensulfuric, and tromethamine

citric,

the present Science,

functionalities, compounds otherwise

acid basic to resins,

forms.

as the present

compounds from from

manganic,

the

be present

compound arginine, isopropylamine,

various addition bases

tartaric,

and encompassed and succinic, like

2-diethyl Examples

relatively phosphoric, inorganic substituted

procaine, invention.

In

1977, invention

the the acidic

(see, invention include 13 general, may

salt manganous, and betaine,

with like, methanesulfonic, salts. salts

66, in

acid

suberic, aminoethanol, e.g.,

be

functionalities forms of nontoxic the acids purines, the

amines,

N-ethylpiperidine, a and 1-19). are

aluminum, can

monohydrogenphosphoric,

pharmaceutically regenerated organic within the

addition

sufficient

lysine,

Berge, like.

may conventional caffeine,

hydriodic,

equivalent

like

salts

exist in solvated fumaric,

potassium, Certain theobromine, certain exist

When organic

cyclic the

hydrochloric,

methylglucamine, bases S.M., of

in salts

2-dimethylaminoethanol,

amount ammonium, scope

organic

choline,

unsolvated in

and that

by

forms

mandelic,

compounds or

physical

amines, to specific can include multiple

et

acids manner.

contacting

phosphorous the

allow the

al,

of sodium,

acceptable glucamine, be

of

acids are

N,N like. “ the

parent triethylamine,

like

Pharmaceutical obtained

the hydrobromic,

naturally- salts

compounds properties, the

equivalent forms

crystalline calcium, present phthalic,

The

- like acetic,

desired Also of

zinc

compounds

the

morpholine, form of

the

parent

acid as

acids

primary,

salt by and included

present

well

of copper,

acid,

such to

or

with of the

the and

as

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 " analog" includes mono-, di- and tri-saccharides. The glucose analog includes sacchrides comprising glucosamine, N-acetyl-glucosamine; fructose; 5 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-glucose, 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-amino-2-deoxy-galactose. Thus, the glucose analog can differ from glucose or a derivative such as DG and glucosamine 10 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-thio-D-glucose or a derivative thereof.

15 [0060] A wavy line " " means the point of attachment of one group or moiety to

another. For example, both indicate that the 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(O)2 are used interchangeably 20 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 as molecule, group, or 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 25 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, -Cl, and -CH3; the interchangeability of -Br and -1-C3H7; the interchangeability of-I and -Z-C4H9; the interchangeability of -O-, -S-, -NH-, -CH2, and -Se-; the interchangeability of -N=, -CH=, and 30 -P= (in cyclic or noncyclic moieties); the interchangeability of phenyl and pyridyl groups; the

14 ) 2011202075 05 May 2011 30 25 20 15 10 5

one metabolized kept conditions, feasible results [0066] mono- need substituted [0065] group http://www.chemcomp.com/ known event circumstance the alkylators. identifies of identified, interchangeability [0064] similar functional identify limiting C(=R interchangeability "pharmacophore", [0063] 1996,

medicinal

program biological

at not

ar

or

optionally

Chem.

)-Rar); in or

a molecule pharmacophores.

compound

other on circumstance temperature

As be, A

A "Optional" a disubstituted the

groups

this

stable with for combination An reasonable the

used

or present, 3D

chemistry, Rev. pharmacophore

can, an<

bioisosteric

property,

at information otherwise range example

i an Pharmacophore

mono-

can

least herein,

or

the

in but 96:3147-76;

of of and is alkyl

chemically

or the

and be of an -C=C- one of interchangeability

need

occurs a

with once quantitative

"optionally"

4 relative

week. bioisosteric or and made aromatic molecule. of

of

group. a

the converted

in °C Such

replacements “

di-

programs substituents

can not, prodrug

an which

the and as

description

or fdept/prodinfo.htm). and

of based substituted

alkyl and the

be methods

less,

feasible occur, pharmacophore

to phosphoramidate -S- search

nitrogen instances

used

itself. structural

the

Burger, As prediction

” to

means

(for equivalents on group in available means

an chemical

used and

the

to the from

or

known

of-CO-, compound. are example, includes

active

To with

identify

variables

(Rar-NiR^-Rar)

that that and absence spatial

in 1991,

herein,

well

a information produce the

15

which

of

compound to an

situations the in the

or or

and structure

Chemical the

known -SO-,

perform

A.

situations alkyl

the

alkylator benzene

arrangement other more pharmacophores description subsequently

such

of

is

Prog. binding one A it

art.

a

moisture permissible

does

stable and prodrug, group

molecules to active

an of

described

that, where is

pharmacophore

See,

Drug for Computing persons and

skill

-SO prodrugs arrangement where not not.

ability

” compound

an after

includes means for

form

thiophene); 2 of

or substantially

the in -. Res. a

described For unsaturated

a pharmaceutically

other the only example,

the These

containing

of

in administration, or heterocyclo in small

with

example, and

37:287-311.

that this ordinary heterocyclo

the a art Group

if

instances

molecule

chemically is

or

phosphoramidate

examples respect such will number -related function application the

event called the

chemically Patani

altered

carbon

(see alkyl the

be

a "heterocyclo skill

group

combination

or

able to

where a

same have

of

group

searches

et

of

is

can,

are reactive

at in

active when

(R

atoms

al.,

a

to least is the

been not ar

or the but

- is not

art

or

is

2011202075 05 May 2011 25 30 20 10 15 5

bioreductive nitroimidazole these nitrothiophenes, nitrobenzenes, Prodrugs such electrochemical can herein in a oxidation-reduction “ under [0070] cytotoxicity are such cellular effect toxicity, drug, generated [0069] active certain form compound [0068] [0067] Chemistry normoxic

group

vivo

cells be

as

as, is classes

altered

on certain

phosphoramidate

reduced a

and/or biological

by less

that

growth

for phosphoramidate

residing or

For As As As cells. can

by

A enzymatic

(or cytotoxicity

improved ”

example,

active

can

used used used

Biochemical groups

of metabolic example, metabolic of biological

also

in

a

electron that

in

As moieties

and

means; a suitable nitrothiazoles, accept vitro·,

in

herein, herein herein, vivo conditions compound be

or

used may

multiplication. a reaction. include,

prepared inactive, hydrolysis, normoxic in flavor,

and/or (3)

and

deficient electrons,

stability

or or

be conditions a herein, alkylator.

“ precursor may of a a

hypoxic Approach, hypoxic

that

alkylator

other (5)

“ “ substituted as

a bioreductive

cytotoxic

but for such or

compound

be in described

The

that but using

can

nitrooxazoles,

agent a environment

biological

vitro or example optionally are nitrobenzoic

metabolism

“ hydrogen,

that the

can bioreductive cells tumor be cytostatic thereof) transport

generate

prodrug

not

compounds

Oxford

by reduced is

chemical

agent be with a ”

in

limited its pharmaceutically

or

DT-diaphorase, are

group zone, (see eliminated

more processes.

agent cytotoxicity is ” substituted,

a

is a and/or cells characteristics,

agent University

is in modified variety

16 etc. pharmaceutically

nitrofurans, acid the in

a

or

modification an group vivo

to,

prodrug detail

refers that vivo

is

residing reference

in

agent amides,

groups

or its

is

or vitro.

and/or

of are

and/or

an is A an

below, cytotoxicity

to chemically

in

such groups. on

Press,

a or prodrug

not that

thiols, hydride agent

a

vitro.

active group and in based

hypoxic nitroazoles, As

compound

group Nogrady, cleaved fewer

drugs in

that is a upon

one

used nitropyrroles, hypoxic New

vitro that effectively or active

As Other

(1) form on the

can ion;

that

bioreductive

side by but such

activation on

cells. herein inhibits used York, electron by that

under redox

have,

photochemical

(2)

1985,

of normoxic

accepts

that compound. which

examples effects a nitroimidazoles, environment

that

can

the biological herein that

As

pages

reversible “

produces hypoxia; potential

relative normoxic

or Medicinal

the compound be deficient where

used releases upon

can

electrons or suppresses

group

reduced,

“ modified

cells. 388-392). of hypoxic lower

be

herein

As activation

to each process,

(4)

a for in reduced under or

is the cells the toxic used

vivo

is that in

the a of i.e.,

” an

2011202075 05 May 2011 30 25 20 15 10 5

tissue capacity by tumors, polysaccharides, [0075] [0074] platinum derivatives, neoplasm, [0073] angiogenic or in agent but groups such nitrothiazole groups, [0072] achieve group, In group groups understand, bioreductive conditions [0071]

combination maintain the another

not

as

” growth,

uncontrolled , by

including

such to refers

limited lymphomas,

cyano, or the of

As As As

As coordination Generally,

modifying

provide

tumor

some example,

metastasis, of

agents,

desired

adrenocortical in used used in used used as

groups group

a to which

a

view

-CH2-haIogen sulfone, to tumor, with

state any

combination

chemoprotectants, but

or herein, herein, herein, herein, methyl,

a

alkylating

lies redox growth

cancer.

a

of one the may other that and agent not can

of bioreductive

complexes, by with

the

within sulfonamide,

remission nitropyrrole

of limited be non-solid “ group “ “

an

be

DT-diaphorase, potential. neoplasm methoxy, malignant

cancer compounds used disclosure suppressants,

skill and

benign loss

Anti-neoplastic “ substituted

anti-neoplastic

agents of

where a

to

spread

in

in of to range such ”

contain

refers

anthracenediones, of the

the group both cyano, cancers

or

” and

or

or For moiety cancer halogen

clinical carboxamide, herein, (neoplasia)

groups.

malignant.

treatment art and where of amine

alkylators,

certain with growth

this certain to

having

abnormal

can electron

and/or carboxamide,

can such ” one

can agents

agent

how purpose, refers

one symptoms

the 17 is groups, “

For alleviate,

tune

herb of

-F, and hormones

of be as

a by group or

or

to

” a

redox example, ,

withdrawing

cancer. leukemia. , -Cl, ”

to include, substituted

or

cells

“ positional more group a

substitute or “

tumor the substituted

to anti-tumor thiol. cancer -CF strong

other

can or

or

achieve -CF reduce, redox potential that

electron

-Br, 3 of and diagnostic

, Such

undergo

but

nitrothiophene, 3

One

and refers plant electron cells , can more

and

control. these with antagonists, can potential

are groups,

the

ureas, ameliorate,

milder agent agents of take

certain that or

donating sulfonamide extracts.

to than be

not

skill an desired reduction and

cancers

abnormal markers withdrawing

used. the ”

lies

electron methylhydrazine limited

,

electron electron can

100 of or other in

natural form

within

a “

the anti-cancer

redox groups, nitrofuran, be prevent,

diseases anti-cancer

bioreductive that

bioreductive associated in

to, art used of withdrawing groups, new

withdrawing donating

the products, have

said

solid potential. anti- will

groups

including

alone cell hypoxic or caused

range.

the

and to place

or with

or

2011202075 05 May 2011 30 25 20 15 10 5

diminishment treatment therapy, cancer line administered does preventing [0081] administration likelihood Thus, drug provides administration of effect, drug palliation example, administration, beneficial beneficial [0080] grammatical equivalents [0078] of [0079] or [0077] [0076]

cancer one elimination ”

not that, chemotherapy. thaf

a

when e.g., or

therapeutically

don

necessarily

As

As As a As

As

As

more of in as

when when or

of or or patient

or

alleviation,

of

’ the used used a used

used used both used stabilization used t

equivalents in the desired desired

of

delaying cancer work,

this of of

of

symptoms

and one administered subject. administered extent onset

the a herein, herein, one herein, initial herein, herein, herein, herein, with

phrase) series

indirect

or occur

or

results, clinical symptom(s). “ which

dose, Third

more

(or the of effective stop a amelioration, treatment,

The prescription a of a a “ a

“ “ of of disease,

reoccurrence) of

administering by reduction treating onset physician “ means “

and

this has second

prophylactically working

therapeutically administration, doses.

the line administrations.

including results

cancer

full to to administration

failed can phrase) disease

a ” a (or amount

therapeutic subject, therapy decreasing subject delay ”

first-line

line Thus, occur a reoccurrence) ” or include,

who is

palliation

for of condition to

other clinical

” called.

state, includes

” or a

of respond can therapy

a

with instructs a or refers will only symptom

slowing drug

disease prophylactically

effective including

therapy,

hyperproliferative “

but be of

effective of effect 18 and administration

have results. cancer,

after or one

or

the administered

is

to are

both refers

to

other elimination patient

of administering

therapy

a

or

of

does severity

dose,

a or the

not

administration patient

and amount

disease

first the symptoms. direct disease

amount will symptoms

For

to beneficial intended limited

not

subsequent

act and

refers therapy

chemotherapy

have that purposes

or to

administration,

effective ” necessarily

of or of

in

” can progression,

of of

self-administer frequency

disease

is ’

of prescribing symptoms, to,

one to

the

a

the one prophylactic a

(and given

occur

results The

that

drug a taking drug

alleviation

of

drug intended

or

drug

of treatment, or

amount

a

is full grammatical more

conditions,

this

to

more

is series for only occur

given

described regimen is of

steps to

a an prophylactic amelioration,

or the an a invention,

subject including

the the

administrations.

after therapeutic amount drug.

manifestations

reducing or can effect, of a by

amount

to

for

treatment

symptom(s), second-line patient. drug

amelioration

doses.

obtain

be or

the

below. (and For

“ e.g., of and/or

first self

of

the

effect a

of a ­

a

1 i ■ i

2011202075 05 May 2011 30 25 20 15 10 5

the activated metastases, supply exhibit Cyclophosphamide then, [0086] 84(9):1280-5 cancer of exhibit or [0085] and and tumor the the receptors. based cancer is than Ila. example, therapies, [0084] [0083] [0082]

that solid a

difficult

patient energy blocked Compounds even lower

normal low

on

growth

not

cells and, slower cells median tumors

Both

The

in Most Some As the tumor the

their

rely

oxygen all

These required

and the consequently, to

resistant

can One

regions cells.

used

vascularized partitioning and hypoxic cancer

is

Cyclophosphamide rates

kill

on

replicating

drug-mediated oxygen liver falls cancer

poor

. oxygen be

often

reason

drugs Rofstad

poisons,

herein hypoxic partial

However,

as, and of

for

in

via of cells to survival.

disordered; areas

division.

or cells

the

concentrations the

therapy cell

Ifosfamide are levels intermediates is

pressure even of

divide LogP etal.,

growth regions DNA,

that normal called

tumor zone

effective, division

reside of the

such

cancer

are the more, The cancer

(see substance

Br Thus, of

more

microtutbule,

means

cytotoxic become

of

leaving subject associated range and tumor than

in drugs

can a J mechanisms

for

new and

tumor. Cancer. a insensitive because therapies,

cells

of to frequently Ifosfamide

the poorly exemplify

the

example,

a

of divide

almost represent yield less vasculature.

measure hypoxic to significant betwen median

corresponding 40 can agents,

intermittent

with

19 1999, than

vascularized cancer

to active

and

mutate slowly.

including

inevitably

to of

60

than

De

octanol are how

a oxygen

- 10 of a selective

such various activation

80(1 poor

mm they

significant

cells Jaeger phosphoramidate

regions lipophilicity prodrugs mm

normal

The

and these

As

cytotoxic 1):1697-707). Hg,

blockage.

response

have

phosphoramidate

and normal Hg.

concentration generally

do

new develop growth solid a

et

for

agents and

tumor

of

and not

cells water.

al., a

and

source dividing vasculature

the lower tumor,

fifty

kill

tissue, action

agents of Br to

factors

and can drug These tumor

can grows, divide

a therapy, J all

percent

substance oxygen

of alkylators, Not Cancer. be slowly-dividing not

are

of of resistance. and cells of as

metastases

under-vascularized under-vascularized and

oxidatively

- more

it only

unable the alkylator-based surprisingly, specifically normal that

the requires

increased targeting, of

growth

concentration cancer 2001,

supports ten

cells solid frequently determined

Alkylators to

cells. percent

Another

and

generate

in a factor tumors cells

blood

target for them

in

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 5 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. 10 Before any of these active alkylators reach the tumor, they can react with healthy cells and result in toxicity and/or cell death.

Cyclophosphamide Hemiacetal 1 Alkylator 1

Liver oxidation

Ifosfamide Hemiacetal 2 Alkylator 2

[0088] While the hypoxic tumor is difficult to treat, the hypoxic tumor zone can generate 15 reduced derivatives of a variety of chemical groups (see the reference Workman et al., 1993, Cancer and Metast. Rev. 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 20 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.

20 ) 2011202075 05 May 2011 30 25 20 15 10 5

hypoxia pharmacodynamics to understanding [0092] significant enantiomer increased (vii) presence attributes [0091] alkylator known present Med. embodiments, (iii) toxicity present Application 4,908,356; the activated linker wherein alkylator 155-85, Chem. [0090] [0089]

healthy

following

less providing

Chem.

L

2002,45: phosphoramidate

invention invention.

or activated isolated

toxic is Aik

For To

of released, Phosphoramidate The prodrugs.

trigger. (iv) aqueous normoxic

advance lower bonded

5,306,727;

specific

more

understand

Publication

2000, effective is

increased

compounds structure side

of the

effective a

compounds

phosphoramidate tumor value 3540-8; than

prodrugs

have

phosphoramidate

solubility (ii)

effect to

43·. In

of over

in cells In

a

some

prodrugs tumor

the vivo one

5,403,932; 2258-65; general, one bioreductive of

biology

hypoxia why

phosphoramide No.US

known

alkylator profile

compared

Hemick

IC50 nature of

bioreductive

of or alkylator metabolism. embodiments,

(vi) the therapy, of the

the more

or

which the provided particularly

2003/0008850;

anti-cancer

invention prodrug or 2001, selective increased

of

IC90, invention

5,190,929; et

derivatives

alkylator

some phosphoramidate

the

of to alkylator group

al., derivatives

a

are hypoxic the

44:

in

hypoxia linker

group J. alkylator

Alk-Trigger combination conjpounds herein

hypoxic

disclosed

cytoxicity

following the 69-73; Med. can Z3. stability

phosphoramidate are

under and

21

5,472,956; prodrugs

(L)

by: phosphoramidate moiety,

generally

In tumor less

and

activated

in are Chem. Trigger

and/or one prodrugs (i) tissue,

hypoxia 2001,44: particular

to active therein, Papot

reported

measured

characteristics: the

alkylator of

cells.

embodimenk liver of

or

of 2003,46:148-54;

the

be nature and the T these (ii)

some

prodrug

the et and

has

and

that microsomal present described

and

bioreductive In

74-7;

al., lower 6,656,926;

is

in present alkylator

prodrugs less

by attributes.

a some

helpful. pharmacokinetics, combination

of

the

are alkylator

are Curr. structure

larger

the should Hemick

toxic normoxic

references,

invention achiral the not (i)

embodiments,

invention

as phosphoramidate Med.

derivatives,

a Trigger

the

degradation US

of

HCR phosphoramidate

to higher

prodrugs be group In L-Z3, US

the

et and

normoxic

subject

Patent

of Chem., much some

cytoxicity, al. represent

Patent

values

invention

Borch

these avoid

differ T Z3,

hypoxic wherein J.

is

and

less Med.

of of

an (iii)

the 2002, a

and/or

Nos.

(v)

cells et hypoxia from the

the

toxic

a

the

al.,

and have the

2,

J.

2011202075 05 May 2011 30 25 20 15 10 5

(0093 oxygen, reduction bonded get the generate formed healthy reductase [0095] Z3 semiquinone a alkylator through reduced phosphoramidate hydroxylamino hydroxylamino invention hypoxic, hypoxia groups healthy phosphoramidate [0094] difficult the dissociation than environment

quinone

in

reduced radical hypoxic

hypoxic J the

in

can to tissue,

cells

and the to

to activated or In

The Trigger.

A

kills tumor

potential

enzyme the

employ the moiety

anion.

an

reach

release certain react number

in

Trigger

region yet

of

radical and

within oxygen Trigger

cells. NO2C-)

indole cells a in

the

group or cells. hypoxic another

with the hypoxic

toxic

in alkylator. alkylator

nitro compared prodrugs

For

an

embodiments,

of the the

of in

of T When

a solid anion. presence hydroquinone

sensitivity

amino

the T.

or

bioreductive the

oxygen and/or releases through example, phosphoramidate enzymes phosphoramidate

side containing

in the tumor

In bioreductive

solid tumor region

such a

is

effects the that

The

semiquinone zone group In to near

released

to of

the the

normoxic another tumor. zone hypoxic

a

can

cytoclirome are of

hypoxic tissue, revert

oxygen, of

prodrug the

having the

conjugated phosphoramidate

aromatic

and to the group

and

the much be

having

phosphoramidate the hypoxic

within group.

bioreductive

responsible These flow back reduction embodiment, solid

flow

tumor, alkylator alkylator.

patient. tumor 0.01

tissue.

the radical in less of

or

a of 1

to P450

of the tumor

N

22

% the phosphoramidate

π first Thus,

% tumor. toxic

indole an the ¢)2(-)

the

an

zone electron

oxygen,

invention

oxygen. Under anion tumor of

Thus

electron or

reductase

electron step bioreductive

for nitro The while

group to the

alkylator.

for

the

in can or quinone

healthy, the

alkylator

modulates respectively

the

bioreductive a zone

normoxia, aryl the example, active group system

another

have hypoxic minimizing

reduction varies

pair encounters

pair present

semiquinone

or enzymes

and

moieties

a

is cytotoxin. heteroaryl The

from normoxic

alkylators from group

of higher

prodrugs

in depending

reduced can

one tumor, the

the as

invention

a released

to

of

the

group zone the

more

in death

oxygen bioreductive can the and Trigger a

the concentration as

well NO2C-)

amino

hydroquinone

radical

an cells

hypoxic, to moiety can reduce bioreductive

of

The having

not bioreductive easily

causes

of partly indole an

phosphoramidate

the vascularized provides

sensitivity

kill

ultimately non-cancerous T

compared or

or present anion releases

the

covalently penetrate

0.1 cells

on

reducing a group quinone

nitro

% the of

in

group

the

of

to

the

can the or

is

2011202075 05 May 2011 30 25 20 15 10 5

alkylator embodiment, capable of prodrug phosphoramidate to phosphoramidate prodrug [0099] invention phosphoramidate phosphoramidate the of phosphoramidate electron the phosphoramidate reduction tissue. toxic reduced [0098] electron and reduced [0097] oxygen, semiquinone agents [0096]

the

an hypoxia

“ bioreductive

tissue. Methods

anticancer to present

)

of

without

the with rich and that prodrug withdrawing the According in The The A a potential

forming

by

hypoxia healthy

The bioreductive hypoxic

radical

release

the

linker

a respect radical invention, Trigger

ease of the Trigger

agent novel

altering Treatment cytochrome

group

can

alkylator alkylator alkylator alkyltor alkylator alkylator action

or

an of normoxic or anion

to activated the

cancer

anion to be

difficulty

T linker

the ammonium ensures an phosphoramidate

the

T easier

phosphoramidate

phosphoramidate

can modulated

in

its of group

containing electron

bioreductive

itself prodrug

present

one prodrug ” in

in

can in (Alk-H). glutathione cells cytotoxic

be or

section).

P450

to a a phosphoramidate tissue a that

an oxidized, βπώοίΐϊπιβηζ influence bioreductive hypoxia loses of hypoxia

can reduce

and

rich reducing electron

cation invention, that reductase the

by

can release

can

a some

tissue For properties.

quaternary prodrug

phosphoramidate The

employing

group is transferase compared

be

non-specific, reduce non-senselective

alkylator

under hydrolyzed, the in

example, alkylator alkylator,

used

withdrawing

or chemical the

the compared

the

group

oxidative, enzyme the a

23 and all

properly

phosphoramidate

is

bioreductive

physiological it clinic, alkylator to

volume

of

absorbed

in ammonium For is

prodrugs a to

“ (see, does release.

attachment

its Trigger tune influenced

the causing when nature

or or example, oxidation,

to hypoxic can

hydrolytic,

presence

” employed phosphoramidate other not e.g., thiolyzed

alkylator. the

of

prodrug, manner. the

release it

in

of distribution of

T

In react

healthy

group toxicity is

phophoramidate

pharmacokinetic

conditions. the group reducing

containing

the the

one

of covalently by specificity a

hydrolysis,

alkylator, high

of tissue

the or

present

linker

an

the and Trigger does Z3 embodiment Telcyta™, or

oxygen. can

reacts normoxic

to bioreductive

active

thiolytic

can linker volume can

healthy agents quickly.

not of

yield

and/or

an

invention be In alkylator bonded

when a in release slowly or

release

toxin

phosphoramidate or amino

(L), one measured

a If

can

a alkylator

(“ a of cells

property stability thiolysis.

normoxic

a prodrug the

of

reducing it

and

According NO

distribution

in

be

with to the group is

the

group the

can

and are

absence

so an 2 further the

(

present

-)

by

more of make easily

If of

to

or cells

the the in

the

an

a

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. CChOin the extracellular space outside of normal healthy tissue and not pass easily through the normal cell membrane. The lower pH 5 in tumor extracellular space can convert the CO2C) 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 10 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 alkylamino, a carbamate, or an amide; a carboxyl group to an ester, and a mercapto group to a thioether or a thioacyl, 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 15 corresponding phosphoramidate alkylator. Non polar phosphoramidate 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.

20 [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 (OTN\I) moiety covalently bonded

to a P=O moiety as shown below. Upon release of the anionic phosphoramidate alkylator moiety an aziridine or aziridium species forms which can alkylate DNA (See EXAMPLE section, Example 36). Depending upon the electron witlidrawing nature of R2 and R3 25 substituents, the aziridinium formation kinetics can vary. For example, as shown in the reaction sequence below, the rate of alkylation can increase when the NR2R3 moiety is

changed from NH2 to (see Engle et al., J. Med. Chem., 1987, 25:1347-57). Substituents on the nitrogen atoms can alter the geometry of the phosphoramidate alkylator, the delocalization of the lone electron pair on this nitrogen atom in the P=O moiety, the 30 availability of the nitrogen lone electron pairs for aziridinium or subsequent aziridine formation, and the aqueous solubility of the phosphoramidate alkylator prodrug and the phosphoramidate alkylator.

24 2011202075 05 May 2011 20 15 10 5

HCR Borch of unexpectedly unexpectedly Z3-CH(Z2)-O-P(=O)(NHCH(R group 2-nitrofuran-5-yl, nitroimidazol-5-yl), wherein choloroethyl) Z3-CH2-O-P(=O)(NHCH hypoxic [0103] containing in cells example, alkylator [0102]

formula cells

Phosphoramidate of values than

as

et

the under

Z2 cytotoxicity, al., Z3

In The prodrugs

Compounds in

present

(I): ifosfamide

is one of and

J. normoxic

high more methyl; present phosphoramidate hypoxia.

known Med.

aspect, ifosfamide

or invention

HCR

employing toxic

2-nitrothiophen-5-yl, 5-nitro Chem.,

invention

low Alkylator

phosphoramidate R

mustard 24

cells lone the 9 (See

values,

2 in is and

CH normoxic

present

mustard hydrogen, hypoxic pair

in imidazolyl,

andUS EXAMPLE

2 25

X4)2, 2-nitroimidazole-bioreductive

an 9

or

)CH Prodrug

mustard arises

on in were

anti-proliferation ifosfamide

anti-proliferation invention N

Z Patent 2

(see toxicity cells X4) 3

in methyl, -CH respectively,

alkylator

bioreductive 2

or part

e.g.,

;

section). R4 compared or 2 alkylator Phosphoramidate

No. cyclofosfamide -O-P(=O)(NHCH(R

and 2-nitrofuran-5-yl; (I) mustard

out provides 25

compounds or

6,656,926

high of isopropyl; derivatives

R3

Phosphoramidate cytotoxicity

400 the

to cell

groups

analogs HCR

phosphoramidate

normoxic discovery

to

cytotoxicity

both P4,

1000

mustard; and

Z3 (Z group having

Pl and

supra). and 3

9 is

), assay improved )CH fold

4-17, cells,

that

and l-N-methyl-2- having

%*?(+) Aziridinium each show

2-nitrothiophene-5-yl, 2

or more

alkylator 0(-)

assays, X4)

with phosphoramidate

N,N Pl

and/or an

X4 alkylators

2 9, unexpectedly

formulas: ,

solubility. indole

toxic ’

a and (tetrakis-2- and is

IC50 in

possessed Cl

prodrugs

dna P21-22, contrast

in

or quinonyl of

prodrugs hypoxic

Br

0.05 For

were high

to in

μΜ

the

2011202075 05 May 2011 25 20 15 10 5

Ceheteroacyl, Ce OH, heteroaryl, [0113] [0112] -[C(Z,) -[C(Z [0111] Cg wherein wherein dialkylamino, heteroalkyl, alkyl, any heteroalkyl, wherein [0110] [0109] [0108] [0107] [0106] [0105] [0104]

alkoxy, dialkylamino,

two OP(=O)(OH)

Cj-Cgheteroalkyl, I

) 2 2

Trigger of -Y3]v-(S(=O) -Y each X each Z each Y Y Yi Y3 L each

2 2 Ci-Ce

4 2 C3-C8 3 R1-R5 is

is ]v-[C(=O)-O] is C3-C8 aryl, is is is is

selected

z,

aroyl,

Xi

Zi Ci-Ce aryl, NR of O, O, S, O,

v,

is

Ri-Rj alkylamino,

aryl, independently O, or is cycloalkyl, 2 form S, S, S, 7

q,

, , cycloalkyl, L-Z3;

heteroaryl,

independently

nitro, or heteroaryl; S,

or NR NRg or alkylene, u, 2

heteroaryl,

) from

or heteroaroyl;

-NR NR CI a and independently 6 -[C(Z

C3-C8 , C3-C10

q

O; or cyano, NC0R -[C(Z 7

'

7 g NSO wherein

and -C(=O)-O-;

heterocyclyl, Ci-C

independently

1

Ci-Ce heterocyclyl, )

Ci-Ce 2 cycloalkyl,

heterocycle; 1 is -Z

)

s 2 CO 2 Ci-Ce

, R6

hydrogen, -Z

N 6 2 or

-Y4] dialkylamino,

2 heteroalkylene, 2

each or wherein

acyl, H, -Y4]u-[C(Z NSO

is

u CRs

acyl, hydrogen, C -[C(Z

R

r Ci-Ceheteroacyl, 2 CpCe heterocyclyl,

R 7

C6 Ci-Ce

is wherein or

halogen, is 6 26 Ci-Ceheteroacyl, each

;

1

alkyl, 0

) independently each 2

1 or

] ) acyl,

z 2

-[C(Z aryl, alkoxy,

]

Rg hydroxyl, 1 z

-[-C(Z

of ; Ci-C Re

is

C1-C6

Ci-Ceheteroacyl,

R1-R5 CON(R

1 independently is Ci-Cg )=C(Z

6

Ci-Ce independently

1 heteroalkyl, )=C(Z

aroyl, alkyl,

amino, independently hydrogen,

alkoxy, aroyl, 7 ! )] )

alkylamino, 2 g I ,

-;

)] or Ci-C« Ci-C

g

Cj-Ce

-;and or heteroaroyl;

C|-Ce Ci-Ce

Ci-C« heteroaroyl;

aroyl, 6 hydroxyl,

heteroalkyl, hydrogen, acyl,

alkyl,Ci-C

is

Ci-Ce alkylamino, alkyl,

cycloalkyl, or a Ci-

Trigger

heteroaroyl;

or

CpCe

Ci-C halogen,

together

aryl, 6

T 6

Cr Ci-

2011202075 05 May 2011 15 10 5

pharmacophores, heteroalkylsulfonyloxyalkyl; [0119] heteroalkylsulfonyloxyalkyl [0121] [0120] alkylsulfonyloxyalkyl, alkylsulfonyloxyalkyl, with [0118] [0117] [0116] [0115] [0114]

the

proviso In In In an (iii) (i) Z3 (ii)

one one one individual at is

at each

a least least

embodiment, embodiment, embodiment, bioreductive that

a

of pharmaceutically

two

one in NR2R3

isomer 2-heteroalkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl,

formula

of of

Ri-Rs

R1-R5 ;

and

group and R any z

or

2 is

(I) -R

NR4R5 a

are

at is one 1

racemic 5 .

:

selected least selected are

selected acceptable

of

not are

R one 2

or -R

the

^ from

from

27 of from s non-racemic

N is same.

\); NR2R3 salt,

the the

the and

solvate,

group

group

2-arylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, group

and

mixture

NR4R5

consisting consisting

hydrate, consisting

is of

or

isomers,

of: of

of

a

2-haloalkyl, prodrug 2-haloalkyl, or

and and bioisosteres, NO

2- 2- thereof. 2

2

2- ­

[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 5 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:

O (CIH2CH2C)2N II N(CH2CH2CI)2

0 I (CIH2CH2C)2N^J 1^, NH2 °\ t____ ^NO: Me no2 h2c------

h A-J Me 2 3

10 Pl P2

O 0 (CIH2CH2C)2N II nh2

no2

>

P3 P4

28 2011202075 05 May 20 P9 PIO 29 h 3 ch 2 co Pll N(CH 2 CH 2 CI) 2 I 2011202075 05 May 2011 5 wherein

R a

is

H,

Br

(Pl P12 4),

NMe2

(Pl 5),

CN

(Pl 30 6),

or

CONH 2

(Pl 7), P13 N(CH NO NfCHzCHjCIfe 2 CH 2 2 CI) 2 2011202075 05 May 2011 o 2 n Br P20 31 Cl ο CM Cl 2011202075

P36 P37 wherein R is H, Me or allyl;

5 {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] 2-(5-Methoxy-l -methyl-4,7-indolequinonyl)methyl bis[N-methyl-N-(2- 10 bromoethyl)]phosphorodiamidate (P29),

[0127] 2-(5-Methoxy-l -methyl-4,7-indolequinonyl)methyl N,N-bis(2-chloroethyl) phosphorodiamidate (P30),

[0128] 2-(5-Methoxy-l -methyl-4,7-indolequinonyl)methyl N,N-bis(2-bromoethyl)- phosphorodiamidate (P31),

15 [0129] 3-(5-Methoxy-l-methyl-4,7-indolequin-onyl)methyl N,N-bis(2-bromoethyl)- phosphorodiamidate (P32),

32 [0130] 2-(5-Methoxy-1 -methyl-4,7-indolequinonyl)methyl bis[N-methyl-N-(2- bromoethyl)]phosphorodiamidate (P33),

[0131] 2-(5-Methoxy-l -methyl-4,7-indolequinonyl)methyl N,N-bis(2-chloroethyl)- phosphorodiamidate (P34), and

5 [0132] 2-(5-Methoxy-1 -methyl-4,7-indolequinonyl)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-R5 are selected from the group consisting of 2- 10 alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyIoxyalkyl, 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

15 [0136] (ii) at least one of R1-R5 is selected from the group consisting of 2-haloalkyl, 2-Ci- Cj alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2- heteroalkylsulfonyloxyalkyl; and at least one of NR2R3 and NR4R5 is VWN\}; or

[0137] (iii) each NR2R3 and NR4R5 are

[0138] In another related embodiment, the present invention provides a compound of 20 formula (I) with the proviso that the formula (I) excludes R2 and R3 together forming a morpholine ring or R4 and R5 together forming a morpholine ring.

[0139] In one embodiment, the present invention excludes a compound of the following structure:

O .Cl

Cl

33 wherein Z| is hydrogen or Ci-Ce alkyl.

[0140] In one embodiment, the present invention provides compounds wherein the Trigger T is:

5 [CCZjh-YaJ-ICiZjh-Zz-Y^ -[C(Z1)2]z-[C(Z1)=C(Zi)]-Z3; [C(Z1)2-Y3]-[C(Z1)2]z-[C(Z1)=C(ZI)]-Z3; [C(Z1)2-Y3 -[C(Z1)2]z-Z3; [C(Z1)2-Y3]-(C(=O)-O)-[C(Z1)2]z-[C(Z1)=C(Z1)]-Z3; [C(Z1)2-Y3]-(C(=O)-O)-[C(Z1)2]z-Z3; 10 [CiZjh-YaHC^OJ-O) -[CiZOJz-fCiZjhCiZOl-Zj; [C(Zi)2-Z2-Y4]-(C(Z1)2]z-[C(Z1)=C(ZI)]-Z3; -[C(Zi)2]z-[C(Z1)=C(Z1)]-Z3;

15 [0141] In an additional embodiment, Z3 is:

34 2011202075 05 May 20 10 5

CMe CH(Me)-O-, [0144] CH(CONH [0143] CH(CH 2 -NMe-.

2 NMe In In

2 one one )->

CH(Me)-S-; 2

)-, -CH(CF

embodiment embodiment,-C(Zj)

-C(CH 3 )-, 2 NMe

-CH(Me)-NMe-,

-CH(CHF

-Z 2 ) 2 2 -Y4-together -,

or 2 -Y 2 -C(CH )-, 3

- -C(Me)2-,

is:

-CH(Me)-NH-;

2 -CH CO 35

is: 2 2 H) -O-,

-C(Et) 2 -.

-CH 2 -, 2 -CMe -S-,

-CH(CH

-CH 2 -NMe-, 2 2 -NMe, NMe

-CMez-NMe-, 2

)-, -CH

-

2 -NH->

or-

j 2011202075 05 May 2011 10 5

(ΠΙ): wherein [0148] wherein [0147] heterocyclyl. In R12 dihalomethyl, from CH=C(CORi2)-, C(Ar)=CH-, [0146] [0145]

another

is

the

is

each each independently In In group In In

embodiment;

one another another one

-CH=CAr-, Ζ,γ Zi

trihalomethyl,

consisting embodiment, is independently embodiment,

wherein

hydrogen

embodiment, embodime^

hydrogen,

Trigger -C(COAr)=CH-,-CH=C(COAr)-,

Ar of

or

is OH, hydroxymethyl,

the -[C(Zj)=C(Zi)J-

is aryl Ci-Ce

is

present

Trigger H Trigger OMe, C^-Ce

optionally or

alkyl

Ci-Q

CF3,

allcyl, invention

is: is:

and

36 alkyl. O-CHF2, CO2H,

substituted Ci-C is:-CH=CH-,

R 8

is provides 6

heteroalkyl, CONH H,

OCF3,

OH,

with

-C(COR

2 compounds

-C(CN)=CH-, , or

NO CONMe2,

up

-OP(=O)(OH)

Ο 2

to ,

3 12 CN, -Ο

five )=CH- 8

cycloalkyl,

halo, of

and substituents

-CH=C(CN)-,

formulas

or

CONHMe;

halomethyl,

2 - .

or

(II)

selected

and and

-

Trigger and

(Π) (ΙΠ)

wherein each R2-R5 independently is selected from the group consisting of hydrogen, hydroxyl, Ci-Ce alkyl, Ci-C6heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, Ci-Ce alkoxy, Ci-Ce 5 alkylamino, Ci-Ce dialkylamino, aryl and heteroaryl; or together any two of R2-R5 form a C3- C10 heterocycle; each Y1 independently is S or O; and each Trigger T is defined as in formula

(i);

[0149] with the proviso that in formulas (II) or (ΠΙ):

[0150] (i) at least two of R1-R5 are selected from the group consisting of 2-haloalkyl, 2- 10 alkylsulfonyloxyalkyl, 2.heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2 heteroalkylsulfonyloxyalkyl; or

[0151] (ii) at least one of R1-R5 is selected from the group consisting of 2-haloalkyl, 2-Ci- Ce alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2- '0; or heteroalkylsulfonyloxyalkyl; and at least one ofNR2R3 and NR4R5 is

ΧΛ/νΉ 15 [0152] (iii) each NR2R3 and NR4R5 are \l; 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 thereof.

[0154] In one embodiment, the present invention provides a compound of formula (II)

wherein Trigger T is -CH2-Z3, -CH(Zj)-Z3 , or-C(Zi)2-Z3 wherein Zi is Cj-alkyl and Z3 is:

20

with the proviso that in formula (U):

37 [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 Ci-alkyl and one of R4 and R5 is C|-alkyl; or

[0157] (iii) at least one of R2-R5 is hydroxyl, amino, C3-C8 cycloalkyl, heterocyclyl, Ci-C6 alkoxy, C1-C6 alkylamino, C1-C6 dialkylamino, aryl, heteroaryl, C1-C6 acyl, C]-C6heteroacyl, 5 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):

10 [0159] (i) at least one of Rj-Rs are selected from the group consisting of 2- alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2- heteroalkylsulfonyloxyalkyl and

[0160] at least one of R[-R5 are selected from the group consisting of 2-haloalkyl, 2- alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2- 15 heteroalkylsulfonyloxyalkyl; or

[0161] (ii) at least one of R1-R5 is selected from the group consisting of 2-haloalkyl, 2-Ci- Ce alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-

heteroalkylsulfonyloxyalkyl; and at least one of NR2R3 and NR4R5 is

[0162] (iii) each NR2R3 and NR4R5 are

20 [0163] In one aspect, the present invention provides phosphoramidate alkylator prodrugs of formula (I):

38 wherein

[0164] Rj is a -[C(Zl)2-Y3]v-[C(=0)-O]q-[C(Z1)2-Z2-Y4]u-Z3 or -[C(Zi)2-Y3]v-(S(=O)2)q-[C(Zi)2-Z2-Y4]u-Z3) wherein each v, q, and u independently is 0 or 1; 5 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 al., US Patent No. 5,622,936;

[0165] each of R2-R5 independently is hydrogen, hydroxyl, amino, Cj-Q alkyl, Ci-Ce heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, C|-C6alkoxy, Ci-Ce alkylamino, Cj-C^ 10 dialkylamino, aryl and heteroaiyl, CrC6 acyl, Cj-Cgheteroacyl, aroyl, or heteroaroyl; or together any two of R1-R5 form a C3-C10 heterocycle;

[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- 15 heteroalkylsulfonyloxyalkyl;

[0168] (ii) at least one of R2-R5 is selected from the group consisting of 2-haloalkyl, 2-Ci- Ci alkylsulfonyloxyalkyl, 2-heteroalkylsulfonyloxyalkyl, 2-arylsulfonyloxyalkyl, and 2-

\ΛΛΤΝ I heteroalkylsulfonyloxyalkyl; and at least one of NR2R3 and NR4R5 is or

[0169] (iii) each NR2R3 and NR4R5 are ; and

20 [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:

39 Phosphoramidate alylator prodrugs

O o

%/*■

/ —N

Κ» Phosphoramidate alkylator cytotoxins

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

X4

wherein X4 and Z3 are defined as above.

5 [0173] In another embodiment, the present invention provides the compounds:

40 2011202075 05 May 2011 Phosphoramidate alkylator wherein [0175] In [0174]

one

embodiment,

R

The prodrug In 2

-

another R5

following

alkylator

are

to

defined

yield

embodiment,

prodrug

scheme is

the

-(N-CHzCHzXjh.

as

corresponding

in

exemplifies

formula

the

present

(Π).

hypoxic phosphoramidate 41

invention

reduction

provides

alkylator.

of

the

the

compounds:

phosphoramidate active

alkylator

J 2011202075 05 May 2011 10 5 (IV) Phosphoramidate [0178] alkylator wherein [0177] [0176]

-(VII)

R2

In prodrug The In

one - another

R5

alkylator following

embodiment, are

to

defined

yield embodiment, prodrug - NR4R5 nr

scheme 2

r the 3 N

as \II/

(IV)

the corresponding

in

hypoxic exemplifies

present formula

the N(R

^ present

reduction

invention

(II).

hypoxic phosphoramidate 42

invention

provides

reduction

provides (V)

the

alkylator.

of

compounds

the

the NR4R

compounds: nr

phosphoramidate 2 s r 3

of

the active

formulas

0 alkylator p I» \

NR4R5

(VI) (VII)

wherein each R9 independently is hydrogen, deuterium, aryl, heteroaryl, Cj-Q alkyl, Ci-Ce heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, Ci-Ci acyl, Cj-Ciheteroacyl, aroyl, heteroaroyl, 5 Ci-Ce alkoxycarbonyl, Ci-Ce alkylaminocarbonyl, di CrCe alkylaminocarbonyl, or C1-C6 alkoxy, or together two R9 groups form a heterocycle; each Rio is hydrogen, Ci-Ce alkyl, Cr Cg heteroalkyl, C3-C8 cycloalkyl, heterocyclyl aroyl or heteroaroyl, or together two Rio groups form a heterocycle;

[0179] Rn is independently is hydrogen, deuterium, aryl, heteroaryl, Ci-Cg alkyl, Cj-Ce 10 heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, Cj-Ci acyl, Ci-Ceheteroacyl, aroyl, heteroaroyl, Ci-Ce alkoxycarbonyl, Cj-Ce alkylaminocarbonyl, di Ci-Ce alkylaminocarbonyl, or C1-C6 alkoxy, or together two R9 groups form a heterocycle with the proviso that when Rn is Ci-Cg

alkyl or Ci-C6 heteroalkyl then Rn excludes χ4 ; or together two Ri i groups form a heterocycle;

15 [0180] X4 is Cl, Br, alkylsulfonyloxy, heteroalkylsulfonyloxy, arylsulfonyloxy, or heteroalkylsulfonyloxy; and

[0181] Trigger T is [QZ^-Y3]v-(C(=O)-O)q-[C(Z1)2-Z2-Y4]u-[C(Z1)2]2-[C(Z1)=C(ZI)]g-Z3.

[0182] In a related embodiment, in formulas (TV) - (VII), each R9 is independently hydrogen, deuterium, C1-C3 alkyl, CpCi heteroalkyl, C3-C« cycloalkyl, heterocyclyl, aryl or 20 heteroaryl. In another embodiment, each R9 is independently hydrogen, deuterium, or Ci-Ce alkyl. In another related embodiment, each R9 is independently methyl, ethyl, propyl, isopropyl, isobutyl, tertiary butyl, or cyclopropyl.

43 ) 2011202075 05 May 2011 10 5 [0186] wherein [0185] heteroaromatic wherein [0184] [0183]

X4 In one In In In

one

another and another one

of

embodiment, moiety R

Rio embodiment, jo

is is embodiment, embodiment,

-(CH2) defined

capable

e the

-Intercalator

the as

of

present

in

the present

the intercalating

formula

present present

invention

invention

wherein (VIII)

44 (IV).

invention invention

between

provides

an provides

Intercalator

provides provides

a

nucleic

the

a

compound

compound

the the

acid is

compound: compound: an

base

aromatic

of

of

pair.

formula

formula

or

(IV)

(VIII):

2011202075 05 May 2011 15 10 5

methyl, (X-A), and wherein [0189] (X): wherein [0188] (IX): and wherein [0187]

each

N(R

(X-B) cyclopropyl,

each

In

Ri each 10 In each In )

one 1

2

one one

is is and R9

R9

R9

independently selected embodiment,

βηύχ^ίηιβηζ independently

embodiment,

independently (X-C): is

methoxy, independently

from

the

NH hydrogen,

the and the

is

is

present 2 hydrogen,

present

hydrogen, , present hydroxyl; hydrogen, < NHMe, / \ xh

methyl, invention

invention invention

NMe

methyl,

or methyl, (X) 45 (ix) methyl, I °\

j

together

2 ethyl, Trigger ,

Trigger

NEt rA provides

provides

provides

ethyl,

ethyl, . ethyl,

2 propyl, ,

Λ two

propyl,

propyl, ,

propyl,

the

Ri

the the

isopropyl,

j

compounds

compound form compound

isopropyl,

isopropyl, isopropyl,

a

heterocycle.

benzyl,

of of of

or

or or

the

the the

cyclopropyl;

cyclopropyl.

cyclopropyl; substituted

formula formula formulas

2011202075 05 May 20 10 5

(XI)-(XV): [0190] (TV), wherein

(VI)

X In

2 and

one and (X

4 (VII). H

X4, embodiment, 2 X R CH 4 n'

are 2 C) 2

defined N

χ xA

0 Ϊ 11

the

as ” N(CH

present 2 in

formula 2 CH \

2 invention X4)2 2 - (X-A) 46

(I), X 2

and

provides

Rio,

and

the

Rn

compounds

are Me Me

defined NO NO

of NO 2 2

, as

and the 2

in

formula formulas

I

2011202075 05 May 20 10

is isopropyl. in heterocycloalkylsulfonyloxy, is propyl, wherein

Cl, compounds

Br,

cyclopropyl,

each

alkylsulfonyloxy,

(ΧΠΙ) (XI) In

one Rn

of (XV)

N(R

formulas

independently embodiment, io

methoxy, )2

(XII),

heteroalkylsulfonyloxy,

arylsulfonyloxy,

and

a

is

(XIV), compound

hydrogen, hydroxyl; and

and

of (XV),

47 (XIV) and methyl

or

formula

heteroarylsulfonyloxy. X|,

when

cycloalkylsulfonyloxy,

(ΧΠ) or X

2 substituted (X) ,

X4 and

excludes is

Z3

Cl

are

or

methyl,

defined Br

a

compound then

In

benzyl,

one Rn as

above;

embodiment, excludes

wherein

isopropyl,

and

X4 Z3

2011202075 05 May 2011 20 15 10

5

NR CH wherein (XVI) prodrug of [0193] oxidatively prodrug cyclopropyl. [0192] present XII, embodiment, or (XIV), [0191]

2 12

the XV CH2(-X6-CH2CH2)f,

wherein XIV,

invention

include

K

or

invention compared wherein In In

In

is (XV) or

metabolizing one

one

Ci-Qalkylene one

XV each

In the

compounds

embodiment,

provide embodiment wherein general,

embodiment,

Rn

wherein present

excludes R12 to

is known

is

C3-C8

or

a independently

a each proteins invention

pharmacokinetically

R CH cyclopropyl

or the 5,

h phosphoramidate

the

the

2 cycloalkyl. 7, Cj-C6 Ri excludes the

(-X6-CH2)f compound:

8,9,

i present present

in present is

provides

heteroalkylene. hydrogen. the 10,

group propyl defined

cell,

invention invention 13,

invention

In wherein (XVI)

48 another phosphoramidate particularly

14,15, can

alkylator

or

improved as Examples

isopropyl. be

above.

provides e provides

provides

In

19, more is embodiment,

one 1-10,

prodrugs. 23,24,25,26,

in phopsphoramidate

of stable

embodiment

the

a

the

In compounds f a

phopsphoramidate

is

alkylator

compound

another liver

compounds

0-3, than

the

the

and an cycloalkyl

32,

embodiment,

prodrugs

K prodrug alkyl of

of

Xe

is 34,

formula

of

alkylator formula

(C(R is

and group

the

O,

compounds alkylator is of

I2 36. formula S,

)2)

ΧΠ, formulas

to the (XII),

or

c

In ,

XIV,

one

2011202075 05 May 2011 10 5

compounds [0196] compound or wherein or wherein [0195] (XVII-(XVIII) [0194]

heteroarylsulfonyloxy. heteroarylsulfonyloxy;

e e In In

In

is

is of

one

of

one

one 0-4, 0-4,

formula

formulas

embodiment,

embodiment, embodiment,

and X4

is X4

(XIX)

Cl

described is

or Χό In Cl,

Br, wherein

a is the

the

Br, the related

O, alkylsulfonyloxy,

present

present present herein.

alkylsulfonyloxy, S,

or e embodiment,

is

NR12

invention

invention 1 Ha invention .

(XIX) See 49

wherein

EXAMPLE

heteroalkylsulfonyloxy,

provides

provides

provides the heteroalkylsulfonyloxy,

R12

present

is

the

defined the the section (XVIII)

invention

compound

compound compounds

as for

above.

examples

provides

arylsulfonyloxy,

of of

arylsulfonyloxy, of

formula

formula

the

of a formula

(XX): (XIX):

2011202075 05 May 2011 5

[0198] provides heteroalkylsulfonyloxy, [0197] analog wherein

includes

R In a In

g

compound

one

is one

glucose

embodiment, mono, embodiment,

of or

di

arylsulfonyloxy,

formula a and

glucose

the

the tri

saccharides. present

XX present

analog;

wherein

invention invention or

e

heteroarylsulfonyloxy.

(XX) is

50

In e

0-4,

is

a

1 related provides

provides . and

X4

embodiment,

is

the

the Cl,

compounds:

compounds: Br,

As alkylsulfonyloxy,

used the

present

herein,

invention

a

glucose

2011202075 05 May 2011 10 5

hydrolyzed [0201] alkylator wherein [0200] (XXI) wherein [0199]

Yi In

R In conjugate: In

9

enzymatically one

is another and one

S

embodiment,

or X4 embodiment,

O;

are

βπύ^ΐπιεηζ

and

defined

to Trigger

produce

such

the

as

the present in

T an

is formula

present oxime-phosphoramidate

defined

invention (XXI) 51

VI. invention

as Br

Trigger in

provides formula Br.

provides

(I). the

alkylator

the compound

oxime-phosphoramidate

conjugate

of

the

formula can -

be

.) 2011202075 05 May 2011 20 15 10 5

pharmacophores, [0209] an [0208] 4C(Z at 2-heteroalkylsulfonyloxyalkyl, [0207] heteroalkylsulfonyloxyalkyl, 4C(Z [0206] with independently heteroaryl; [0205] hydrogen, wherein (XXII): [0203] [0204] [0202]

least individual

the

I 1

) ) one 2 2

-Y3]v-[C(=0)-O] proviso -Y3]Y-(S(=O)2) each each (iii) (i) (ii) Ri each In

hydroxyl,

or

of

at another -

at isomer

each

t

Z NR2R3 together

R5, least R1-R5

is

least independently

R independently

that a 2 a

/ki Yi,

NR

Trigger pharmaceutically

two

one aspect,

or CpCe in and

and 2 and

R r R2

q a formula -[C(Z of 3 3

of

racemic

q Ri

NR and and -[C(Z Y2

R1-R5 T

R1-R5 alkyl,

the *-R

f

selected 2-arylsulfonyloxyalkyl, 2 K

are

U'K 1 1 is NR R2* 1 *R3*

)2-Z2-Y4]u-[C(Z

is 2-arylsulfonyloxyalkyl, present 5

1 (XXII): *

1 )2-Z2-Y4]u-[C(Z

defined

C,

and 2 CpCealkoxy, or

and independently or *R form R

' i

. is

4 S, 1

non-racemic

acceptable 2;

3 from R vwn

Ri *

or ^(zo invention i both a *-R

*-R

as

P; heterocycle;

'\l;or the

: in 5 t

s ) 52 wv*N * r

*

-K 1 formula group are

)2]

is

(ΧΧΠ) Ci-C salt,

1

is

)2] provides mixture z — 2-haloalkyl,

-[C(Z 2-haloalkyl,

selected z

-[-C(Z

solvate, 6 (Z0 or consisting

alkylamino,

; or

(I); or

and 2 1 t )=C(Z

)

each r

z heteroalkylsulfonyloxyalkyl;

of 2-heteroalkylsulfonyloxyalkyl; / the 1

>C(Z from Ν R

I isomers,

hydrate, 4 . \ΙΙ/

‘ compounds 2-Cj-Ce

R1-R5

2-alkylsulfonyloxyalkyl, I

)] of Y2 P x- the g

1

-Z Ci-Cedialkylamino, )] \

g

3 ζ

group and

-Z · bioisosteres, -; or R Ri* Ν

I alkylsulfonyloxyalkyl,

3 3 \

*

a

and R

prodrug of

i R consisting *-R or

2 the * 3

* formula

thereof.

of

aryl, or

2.

and

I I I I I I

2011202075 05 May 2011 20 15 10 5

heteroalkylsulfonyloxyalkyl, 4C(Z,) 4C(Z with [0218] Trigger heteroaryl; hydrogen, wherein [0217] [0216] (ΧΧΙΠ): bonded ofY [0215] [0213] [0214] arylene, [0212] [0211] [0210]

the 4,Υί, 1

)

2 2

T -Y3]v-(S(=O)2) proviso -Y3]v-[C(=O)-O] to

(i) or each Ri

In each each

each K each selected and

hydroxyl,

the

or

heteroarylene, at is - another

together

Ye least R

R1-R5

same selected Y4/Y5, R9 m r

s that independently ,

has independently is

Yi from

two

independently carbon aspect, Cj-Ce , in and

to and and

R2

q

from formula

-[C(Z of be the

q Ri*-R

and -[C(Zi)2-Z2-Y4]

Y

Y

R2-R5

(C(R O, alkyl,

group atom 2 the 6

the 2-arylsulfonyloxyalkyl,

are

independently l S, R )2-Z2-Y4]u-[C(Z

is 5

present 9 2

(ΧΧΠΙ) * is )2) group or

*

defined and

Ci-Ce or 0

independently consisting form

1-4; n Ci-Ce NR7. or

; adjacent

and R2*-Rs*

1 consisting

invention ; alkoxy, a

as (Y alkyl u heterocycle; -[C(Z

(XXIII) in 5 is

-(C(R9)2) carbon of 53

1 Trigger

formula O, are )2]

or

C|-Ce 1 is )2]

z provides of S, heteroalkyl,

-[C(Z 2-haloalkyl,

selected z

NR -[-C(Z Ci-C

atoms or

m alkylamino,

or (I); -Y4-(C(R9)2)in-Y6)n

2 7 1 )=C(Z ,

each 6 heteroalkyl-sulfonyloxyalkyl; or

alkylene,

the I are

)=C(Z from

a

bond;

or R1-R5* compounds cycloalkyl 2-alkylsulfonyloxyalkyl, 1 )]

together the g

1 -Z3-; )] Cj-Cedialkylamino,

CrCe

g

with group -Z3and

independently

the or when

heteroalkylene,

of

wherein consisting

heterocyclyl;

the proviso

covalently

formula

n

is is that

of

aryl, a

1-8; 2-

one and

[0219] (ii) at least one of R2-R5 and R2*-Rs* is 2-haloalkyl, 2-Ci-Cealkylsulfonyloxyalkyl, 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 NR^Rs*

I 5 together are both ^1; 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] L2 is

10 wherein X is defined as above.

[0223] In another embodiment, the present invention provides the compound of the formula (XXIV):

f3 V4 r4* r3* I ii I 0 < r2^ (ZO2)rK-(ZO2)r-^N\H//N\ I °\ 0 Trigger ^Trigger

(XXTV)

15 wherein R2, R3, R4, R2*, R3*, R4*, Z, K and Trigger are as defined in Formula (ΧΧΠ).

[0224] In another embodiment, the present invention provides the compounds of formula (XXTV) having the structure of formula (XXV) or (XXVI):

54 (XXV) (XXVI)

[0225] In another embodiment, the present invention provides the compound of the formula (XXVI):

(XXVI)

wherein Xb X2, X4, and e are defined as in formula (XXV).

[0226] In another aspect, the present invention provides the compound of the formula (XXVII):

Trigger^^W^^Tngg^ . 10

(XXVII)

wherein R2-Rs, r, k, Yi, and Trigger T are defined as in formula (XXIV).

[0227] In one embodimenL the the present invention provides a compound of Formula:

55 2011202075 05 May 2011 5 consisting [0228] wherein Li

is

CH

T 2 In ,

of: CHMe, is

another

L-Z3;

C(Me)

embodiment, 2 ,

CH 2 OCH2,

the

present

(CH 2 )

56 3 invention ,

CH 2 S(CH

provides 2 )2,

CH

2 a S(CH

Z3

selected 2 ) 3 ,

from

the

group [0229] In another embodiment, the present invention provides a moiety having the formula:

R3

Rs °x selected from the group consisting of:

57 2011202075 05 May 20 α 58 2011202075 05 May 20 OP(=O)(OH)2. methyl, wherein consisting [0230]

or each In

ethyl; of:

another

Zj,

Within

R R 7 7,

is

embodiment, and

methyl,

this

Re

embodiment,

is

trifluoroethyl, defined

the

present

as

above.

59 ethyl, invention

Within

propyl,

provides

this

and

embodiment,

cyclohexyl;

a

T

selected

Zj

and

from is

Re hydrogen,

the is

OH

group

or

2011202075 05 May 2011 5 and wherein or embodiment, wherein [0231]

methanesulfonyloxy.

Z3

is

T each In

is

another

defined

R9 R9

is is

hydrogen, hydrogen

embodiment,

as

above

or

methyl, or

Ci-Cg

more

the

present

ethyl, particularly alkyl

60 and isopropyl,

invention

each

T

is X4

or L-Z3 provides

is

isobutyl; X

halo 4

wherein

or a

compound R and su

L HO i S(=O)2O-

X4 CH

is 2 ,

chloro,

CHMe, of

formula: .

In

bromo, another CMe2,

2011202075 05 May 2011 15 5

determining described synthesis phosphoramidate of pharmacokinetic resonance plasma, phosphoramidate the tumor wherein and [0232]

phosphoramidate

likes,.

deuterated

tissue

can X4

of

in In methods However,

dosage,

is a the be

one as

octadeuterated-compound halo

determined

phosphoramidate their EXAMPLE

aspect or

alkylator alkylators

or

and pharmacodynamic

frequency alkylators

non-deuterated the

R

SU the such

presence |S(=O)2O.

prodrugs. more present

section. and deuterated

and/or of

their efficiently

dosing, alkylator

of

invention In or

such Pharmacokinetic

phosphoramidate prodrugs

another properties hydrogenated

analogs 25

and

deuterated

prodrugs

and compared

61 provides similar

embodiment,

are

octadeuterated can

of

equally

the be of

administration analogs analogs,

to deuterated and

useful formula alkylator phosphoramidate

their

or cytotoxic

X4

pharmacodynamic in

in corresponding such isofosfamide

is vivo,

determining

prodrugs

phosphoramidate Cl

as

related

with or for

compounds

Br.

example

alkylators respect

is

parameters. Such

alkylator used

information

deuterated to

in in 25,36

alkylators

and/or hypoxic blood

is

The

and

2011202075 05 May 2011 invention selective [0233]

groupings In include:

another

of group

the

compounds of

embodiments,

of

the

the

62 EXAMPLES.

present

invention

Examples

provides

of

compounds

the

individual

of

the

and

2011202075 05 May 2011 63 2011202075 05 May 2011 64 r 2011

Cl May

05

2011202075

65 Γ i 2011

May

05

2011202075

66 2011202075 05 May 2011 15 10

146 145 141 140

67 147 2011202075 05 May 20 normoxic as

Z3

and

shows tisue.

hypoxic

tumor

specific

toxicity 68

while

being

much

less

toxic

to

healthy,

2011202075 05 May 20 10 5

phosphoramidate alkylator benzenesulfonyloxy, (VII), wherein [0236] phosphoramidate alkylator [0235]

and

X4

prodrug In la prodrug In

ionized

one is one

OH defined

embodiment, embodiment,

alkylators

which

alkylator which forms

or

as

para-toluenesulfonyloxy.

upon thereof. upon in

formula

the bioreduction the bioreduction Cl

present In present

(I), a

related

and

invention invention

releases

releases R9, OH 69 lb embodiment,

Rio,

provides

provides the the and

corresponding corresponding

Rn,

X4

>

and

a a are

novel is novel

Cl, defined

phosphoramidate Br, phosphoramidate

novel novel

methanesulfonyloxy,

as

in

or or OH OH ii

If formulas

OH known known

lc

(IV)-

and ionized forms thereof; wherein N(R)0)2 is selected from the group consisting of NH2, NHMe, NMe2, NEt2,

each Ri i is independently hydrogen, Me, ethyl, cyclopropyl, isopropyl, propyl, benzyl, 5 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 10 alkylator of formula le (each Rn is hydrogen, see Wiessler et al., US Pat. No. 5,622,936; PCT application No. US05/03370 entitled “Anti Cancer Therapies”, US Pat. Appl. No. 60/638995 entitled “Glufosfamide Combination Therapy” and Attorney docket No. 021305- OO59OOUS filed on 11 May 2005 entitled “Glufosfamide Combination Therapy”). Telcyta™ which is being evaluated in the clinic for cancer treatment, releases 1 f (Rosen et al., Clin 15 Cancer Res. 2004, 10(11):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 20 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 phosphoramidate alkylator prodrugs of the present invention do not produce acroleiiy by in vivo metabolism. Examples of toxic by products resulting from metabolism of the prodrugs of the invention include chloro, bromo, alkylsulfonyloxy, 25 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 30 ifosfamide metabolism.

70 2011202075 05 May 2011 30 25 20 15 10 5

formula: [C(Z [0243] [CiZ^HCi^j-OHqZjh-^-Y^H; modified [0242] -[CXZ^Jz-tcizo^zoj-Zj [CfZ^-Zz^] [C(Z,) [CCZ.h-YaHCiOj-OXCiZ^h-ZjH.od; a [C(z [C(Z [C(Z [CCZjji-YJ-fCCZ^-ZrYd Z [C(Z alkylator- hypoxic hypoxic [0241] alkylator above. phosphoramidate bonded bioreductive Alk-Triggermod a different [0240] alkylator [0239]

bystander 3

phosphoramidate .mod> I I 1 I ) 1 ) h-Y3]-[C(Zj) ) 2 )

2 2 2 2

-Y

-Y -Y to -Y3]-[C(Zi) -Y

In

to

In In tumor enough

Z

3 Upon 3 3 yield of In from The prodrug 3 ]-H;

T ]-(C(=O)-O)-[C(Z ]-(C(=O)-o)-[C(z 3 one ]-(C(=O)-O)-[C(Z

the one

another .

M effect

the one

group

or

phosphoramidate phosphoramidate

-[C(Z embodiment, the a zone reduction

embodiment, present upon

Aik- embodiment, to modified

upon alkylator

and

2 2 phosphoramidate

activate embodiment, ]z-[C(Z ] forms alkylator

z which I

-Z ) reduction T

2 is

]

M 3 activation invention z

.

-[C(Z n

termed conjugate. of

,o<

a phosphoramidate

J

I can

or )=C(Z the -mod -[CiZjhjaCiZ^ZOj-Zj^od; ; modified the

the

1 Trigger prodrug 1 1

)

) ) the some 2 1 2 2

)=C(Z prodrug

-Z

activate of ] ] bioreductive wherein alkylator bystander alkylator z a z

the

-[C(Z -[C(z

2 to present I

in modified the -Y )]-Z

other

diffuse hypoxic alkylator T

In 4

T

phosphoramidate 1 bioreductive can ] )]-Z3- m 3

1 I

-[C(Z includes . one )=C(Z compounds )=c(z these

mod Z is

derivative

invention modified can yield 3

[C(Zi>2-Y3]-[C(Zi) effect selected .

;

mO m phosphoramidate

embodiment or alkylator

71 od; group tissue

1

d prodrugs. undergo ) being

1 I

penetrate 2 )]-Z3.

)]-Z3H„od; is

a ]

and

the 2

allows modified bioreduced -[C(Z

group

phosphoramidate (Z

by

provides from:

of protected formed following m

o 3 prodrug

d degradation incorporating ) the

1 alkylator ;

)=C(Z

within a into

the

(Z modified

invention phosphoramidate

3 upon

T ). tumor

or a 2

alkylator

and

-Z 1 m such linkers compound )]-Z the

When

otherwise

is 2 prodrug,

-Y4]-H; the

the

selected Trigger

3 to

phosphoramidate zones . as

mod

but

a

reduction alkylator

yield

reduction phosphoramidate

(L) phosphoramidate linker

prodrug. ;

reside

and having which which the reduced

T either from:

alkylator (L) is

linker

nearby prodrug. of modified

of

are demonstrates

as For

the the

Z

or the

described 3

(L) not

example, can

prodrug the

to be

-[C(Z1)2-Y3]-(C(=O)-O)-[C(Z1)2-Z2-Y4]-[C(Z1)2]z-[C(Zi)=C(ZJ)]-; -[C(Z,)2-Y3]-[C(ZI)2-Z2-Y4]-[C(Z1)2]z-[C(ZI)=C(Z1)]-; -[C(Z1)2-Y3]-[C(Z1)2]z-[C(Z1)=C(Zl)]-;-[C(Z1)2-Y3]-[C(Z1)2]z-;

-[C(Z1)2-Y3]-(C(=O)-O)-[C(Z1)2]z-[C(Z1)=C(Zi)]-;-[C(Z1)2-Y3]-(C(=O)-O)-C(Z1)2-; 5 -[C(Zj)2-Y3]-(C(=O)-O) -[QZOJrtQZO^iZj)]-; and -[C(Zi)2-Z2-Y4] -[CiZihktCiZO^Zj)]-; -[C(Z1)2]z-[C(Z1)=C(ZI)J- and -[C(Zt)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 alkylator is separated

from Tm in less than 0.1 second. In another embodiment, the phosphoramidate alkylator is

10 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.

15 [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 pm from the site of activation or reduction; or 20 to 100 pm 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 20 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 25 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 pm 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 30 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

72 2011202075 05 May 20 30 25 20 15 10 5

prodrug, phosphoramidate die least prodrugs from the and [0248] phosphoramidate prodrugs efficiently concentrations molecular determining vasculature. prodrug second, with by HAP hypoxic monolayer [0247] aerobic equilibrating number reactivity [0246] effect). activated number spheroid.

phosphoramidate hypoxic knowing the

ten a a ’

s

modification

half

cell

can

between cells and

the

which of The of In that fraction Anti that

In

drug

of kill weights The

different

factors; life

one

membrane culture be

selectivity

cytoxicity

one the up In

the

the

assessment or are gas can

cancers tumor

combined

hypoxic one to upon ofless

activation to

embodiment,

bystander are hypoxic

embodiment,

activated

safer

alkylator

of 1.0

alkylator and reach leave one

that versus

embodiment, the able

at the

levels.

alkylator to activation cells.

therefore

the million-fold

than than drugs

of permeability and

ability

impede the 10.0 spheroid hypoxic tumor

to

fraction of

with the

of site

intact

effect prodrug drug

hypoxic used target site 0.1 the

For the

The

seconds, hypoxic can

phosphoramidate

the

of the prodrugs of other zones

second; corresponding

effectiveness diffusion such change from in

example,

in

spheroids.

cancer

effect

bind are compound attachment can present the

the and

present the

a releases safer..

cytotoxicity

chemotherapeutic

of

hypoxia described

as present that where be

entire activated the and to cell

the of

the

between bioreduction

cells are modulated

tissue

can and invention

expected

for the invention between

in

prodrug

In ratio

spheroid.

converted it The the

to

of

which invention

away of a

73 be not one compound activated

was

phosphoramidate

penetrate hypoxia in prodrug surrounding alkylator

the

alkylator/cytotoxic

a

of up HAP of 0.01

reach greater compound

embodiment, activated

the Trigger from cell the provides and 10.0 by

to it provides

releases

to was

The can into 150

varying

agents aerobic

phosphoramidate provides

prodrug

kill in

the (HAP) activated

to 0.10

the

into and alone detail

therapeutically

used activated; -

expected

the 100.0

T of phosphoramidate

the 200 and

vasculature.

phosphoramidate

can

its

the second, a the (activation that

each

and corresponding can by the

phosphoramidate

in as vasculature the

plays

rate kill phosphoramidate alkylators

μΜ

deepest phosphoramidate therefore seconds.

phosphoramidate

Examples

nitroreductases; either a

concentration

hypoxic

be of increased

agent).

monotherapy.

cell nearby of and

away

between

an the compared

cellular

kill target

important the of alkylator regions

effective monotherapies. Some

be from formed

and/or In compartments. the cells

can ability 35

alkylator safety alkylator evaluated

either 0.1 only alkylator

alkylation,

and

methods

the

be to of alkylator (bystander of

to

alkylator

prodrug. the have

in

alkylator The cells role

alkylator predicted of O

the the 37. results

event,

1.0 2 vivo

the

of impact

in

outer

to

on high in The

for

the

(at

a

2011202075 05 May 2011 30 25 20 15 10 5

prodrug or ability prodrugs cytotoxicity IC50, LC10, cytotoxic alkylator proliferation recognize without released anoxic [0251] corresponding fold, yields prodrug, or example, phosphoramidate [0250] fold and least alkylator. alkylator, phosphoramidate resulting hypoxic [0249]

LC99

LC

can IC90, less

and about IC90

jo of

a

the or is

oxygen

phosphoramidate of

from show

of

prodrug the

To tissues any than

typically activity In larger hypoxic

In in that for

LC50, or in higher

the This

the

ratio

10

one its prodnig determine one

assay

LC90, a phosphoramidate

a

the the cytotoxicity

up corresponding phosphoramidate

manner

fold phosphoramidate present removal

(anoxia) than;

masking LC?o, version, by

embodiment,

(HCR)

of as to is

compounds

cytotoxic

alkylator.

alkyl conditions,

is about

or

less

virtue its

measured the about

relative expressed

that

IC99

or

hypoxic that invention. if

ator.

phosphoramidate cytotoxic

and

and

or

compounds LC99 a 100 effect

determined

of alkylator

a or

of phosphoramidate

masks with

activity

million

the

the can

Typically, to phosphoramidate to LC99. a

under cells

the in determined

phosphoramidate alkylator

by

can cell healthy

about

concomitant

activation very alkylator be alkylator a

activity

Trigger or The the

clonogenic

are

prodrug vary

of a selective fold The hypoxic

reduces provided

measure little based IC50; 10,000-fold,

the larger exposed

normoxic the

ratio or and that

alkylator than

or T corresponding with

in

or oxygen

less phosphoramidate

and that

on

is

alkylator modified 74 conditions. can the or normoxia can

toxicity the reduction

of

survival herein of

covalently the

alkylator IC90 an cytotoxic to subsequent

the

cytotoxicity is

cytotoxic HCR depend be alkylator the

cells.

the

IC corresponding

at (hypoxia). prodrug less

cytotoxicity or linked

hypoxia 5

least

and include

o,

drug prodrug

phosphoramidate of LC90.

experiment

and active of IC90,

The

prodrug on phosphoramidate

the activity. bonded greater

the

about prodrug

activity

to either release hypoxia is the

HCR

phosphoramidate

as alkylator or

as selective a bioreductive about as

One

is

Trigger

cytotoxic measured

LC50 phosphoramidate

of

a

10-fold

selectively can phosphoramidate the with

to

cytotoxic

of determined In is or a as of

the

is 100

phosphoramidate

hypoxic

expressed be the of one generation

measured

skill

called prodrug air

cytotoxicity

T phosphoramidate

alkylator 1 1 fold

to

phosphoramidate

activity in version, (normoxic) nM, for μΜ

alkylator. in group about

a agent active

hypoxia to

the example tumor

manner

or

based

the alkylator

will

about as in

of greater. art

(Z 1,000,000- of

that

alkylator

the than IC50, IC10

an under

alkylator,

the of show 3

the killing

will ), on

As anti or

that

10,000 is

the

by

or

the

IC90,

IC99

one

­ at

2011202075 05 May 2011 30 25 20 15 10 5

phosphoramidate prodrug for [0256] formula present measure Such about provides prodrug, [0255] alkylators, invention 1,000,00; further adjacent provides folds than phosphoramidate normoxic prodrug [0254] such present hypoxic [0253] section). related invention [0252]

i.v.

phosphoramidate the

as, more

0.5-0.6%

or

invention away

having embodiment, XV invention

the

In having corresponding for under to The phosphoramidate cytotoxicity

Tumors a i.p. In In

provides 10-10,00; In

toxicity. has

phosphoramidate 5-1,000,00; one than the

lipophilicity wherein one example, a

another

injection. from logP

related a

vasculature, a a

oxygen embodiment,

hypoxic hypoxic the

embodiment,

log? variety

alkylator provides

have alkylator

has

of the phosphoramidate

In and corresponding

each related

a the

of embodiment,

the a greater vasculature

another alkylator

phopsphramidate a

10-10,00; normoxic concentrations. hypoxic Another

25-5,000 0.1

of

gradient toxicity cytotoxicity or phosphoramidate

compounds

Ri

prodrug a alkylator

oxygen prodrug the to

nM

phosphoramidate embodiment,

i alkylator

the

than is

the 0.5% embodiment,

hydrophilicity

to H prodrugs

cytotoxicity example

present which

and of

toxicity. folds present

and

0. 50 and normoxic having concentrations. the

having

in prodrugs oxygen

alkylator

of 25-5,000 In μΜ as

prodrug

near tissues

phosphoramidate can

more

is

0.1

one described invention alkylator

invention of

and can the 5

a In

alkylator be

hypoxic the nM

to

logP concentration the such

embodiment,

75 of

toxicity. toxic another

phosphoramidate

generate

be about easily of alkylator a

prodrugs

1,000,000 having folds necrotic

0.1 to HCR present

the hydrophilic,

between prodrugs

prodrug 50

in provides than

nM

provides In

toxicity

prodrug prodrug. 150 formulated more

Examples embodiment, μΜ of

hypoxic one phosphoramidate

to prodrug the core. invention

that

25

μΜ alkylator

folds

and

5 0 the that toxic embodiment,

of are

corresponding to

μΜ a which

and

can

away, of

a

In

the phosphoramidate such a present

100,000 In

phosphoramidate more compounds can toxicity

alkylator

HCR the

29,30

one as having than and generate 4

one present

provides

prodrug

such

an vary is as the present and

embodiment, than a

the

embodiment, of

10

a aqueous

invention

HCR

and present (see

which

prodrug

lower

as

a from

10 the to

alkylators, corresponding

prodrug

invention the IogP phosphoramidate

prodrug, those

of

31. 24,25 invention to 10,000 a

EXAMPLE

of present

corresponding

the 10%, in 100,000. is

50 invention less formulation

alkylator having

provides 25 exemplified tissues alkylator

present

of

to

and the

folds

than the

in to can under

invention

5 the 10,000

has ­

present tissues 5,000

36.

In more 0.

a

a

a

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).

5

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

10 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 15 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 20 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

76 2011202075 05 May 20 10

phosphoramidate phosphoramidate wherein [0259] known the L-Z

product. 3 ,

wherein

phosphoramidate R In 9

is one

as Such

Z3

embodiment, defined

alkylators: alkylator

is: a

Mitsunobu

above.

alkylator

prodrug

the

type

present

prodrugs comprising

reaction

invention o

or 77

is

reacting derivatives,

particularly

provides

each

Alk-Trigger,

suitable a

of

method

novel

for

or of

synthesis

known

wherein synthesizing

Trigger

of

novel

a

is

or

)

2011202075 05 May 2011 10 5

comprising heteroalkylsulfonyloxyalkyl; alkylsulfonyloxyalkyl, compound wherein [0260] respectively, with Trigger

a

(i) Trigger-OH,

X4, wherein

In at

reacting of

one least R5,

formula:

R7, embodiment,

R2-R5 two

(a) and a

of trisubstituted

2.-heteroalkylsulfonyloxyalkyl, a

are Rg

R novel r

are defined R

5 the

as or are

defined known present Trigger

selected

R5R4N. phosphine, as R5R4N.

in

formula phosphoramidate invention as

Ο

II from

in 78 OH O P^ OTrigger || O P^

||

I formula

μ

and

the /

(I) , ,NR

nr a provides

/ group with

dialkyl

2 2 2-arylsulfonyioxyalkyl,

^Rg r (I). R 3 3

the

X ^R consisting alkylator

4

azodicarboxylate a

proviso

method 9

HO Trigger-. of

that of

to

formula: —

2-haloalkyl,

synthesize ο Ρ II

Ο

to N(R and —

yield O

P 1 11 2- o

— ) a

2 2-

N(R 10 ) 2 2011202075 05 May 2011 10 5

is [0262] is (b) (c) heteroalkylsulfonyloxyalkyl; [0261] alkylsulfonyloxyalkyl,

selected selected

a a

dialkyl Trigger-OH

In In (iii) (ii)

another one from

from at

NR2R3

azodicarboxylate

least embodiment,

the the

wherein embodiment,

one

group and group

of 2-heteroalkylsulfonyloxyaIkyl, NR4R5

R1-R5 consisting

consisting Trigger

the

and

to

both

the compound

is yield

at

is selected

group

RsR^^JI^NRzRg R5R4N

together least

ofi defined of:

the

one of

compound

of from

formula: are~' 79 as

of formula: OTrigger O OH O || I

in

NR2R3 the

N Formula ^; NR

group

of

2 2-arylsulfonyloxyalkyl,

R and

formula: 3

consisting

(I), NR4R5

a

trisubstituted

is

of

2-haloalkyl, or

phosphine, and

2-

2-CrCe

and 2011202075 05 May 20 Cl 80 Γ 2011

May

05

Br 2011202075

Cl

[0263] In another embodiment, the reaction includes a solvent such as THF, dioxane, a Cj- C6 alkyl acetate, chloroform, dichloromethane, acetonitrile and the like. In another βπύκχΐΗηεηζ each substituent in the trisubstituted phosphine is independently selected from a 5 Ci-Q alkyl, CrCe heteroalkyl, C3-Cg cycloalkyl, heterocyclyl, aryl, heteroaryl, and Ci-Cg alkoxy substituent. In another embodiment, Trigger T- is

wherein Xb X2, Z], and Z2 are defined as in formula (I).

10 [0264] In another embodiment, the present invention provides a method to synthesize a phosphoramidate alkylator prodrug comprising

81 [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 Ri i is independently hydrogen, cycloprpyl, methyl, ethyl, benzyl, or methoxy; 5 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

10 (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:

82 (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

Π R\1

5

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 Rj2 is H, Ci-Ce alkyl, Ci-Ce heteroalkyl, C3-Cs cycloalkyl, heterocyclyl, aryl, or heteroaryl. In 10 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 trisubstituted phosphines are commercially available, for example, from 15 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

83 2011202075 05 May 2011 10

haloethyl-N-(Ri3)ammonium {0271] yield hydrogen, phosphoramidate [0270] [0269] compound [0268]

a

dichlorophosphoramidate

(b) (a) In

In Ci-Ci

wherein

one

reacting refluxing another

embodiment,

alkyl,

alkylator

the

embodiment, the

POClj

Ci-Cgheteroalkyl,

Trigger

dichlorophosphoramidate

prodrug

salt, with the

selected

intermediate;

present

wherein a the

comprising N-2-haloethyl-N-(Ri3)ammonium

present

from

invention C3-C8

R13

84

invention

the is

the

cycloalkyl,

hydrogen,

group

intermediate steps provides

provides

consisting of:

Ci-Ce heterocyclyl,

a

method

in

the

alkyl,

step of:

method

to salt,

(a)

CpCeheteroalkyl,

synthesize aryl,

with

wherein

of

heteroaryl,

making a

N-2-

R a

n

a is

to

2011202075 05 May 2011 25 20 15 10 5

phosphoramidate cycloalkyl, (Ri3)ammonium to [0278] phosphoramidate [0277] diisopropylamide. [0275] diglyme, is before [0276] separated tetrahydrofuran diisopropylethylamine. employed embodiment, from In and separated Trigger-OH [0274] monochlorophosphoramidate [0273] Cj-Cg [0272]

yield lithium,

another

then

the

cycloalkyl, subjecting

a

0>)

(a) In In diethylether, rest distilling dichlorophosphate In In (c)

from from

in sodium, heterocyclyl,

embodiment,

one one

reacting reacting one one

and reacting step

of the

the

the

embodiment, embodiment, (THF) the salt,

embodiment,

embodiment,

alkylator a

base alkylator

it (b)

heterocyclyl,

base the rest In or rest to reaction

in the

wherein

include

the

one potassium

or the dichlorophosphoramidate employed

or

of a of

in

In

aryl, dichlorophosphate

the

THF. solvent monochlorophosphoramidate

dioxane. reaction the embodiment, the

a prodrug. prodrug one

solvent intermediate; mixture separation

trialkyl

heteroaryl, the reaction the R13 reaction the intermediate; the

embodiment,

aryl,

hexaallcyldisilazide;

in

about present monochlorophosphoramidate

is phosphoramidate dichlorophosphoramidate

in

comprising step hydrogen, to

by heteroaryl, amines, step

mixture yield mixture

is 1

the flash

(b)

and invention equivalent

performed

(c). and

solvent

intermediate is and the

85

the

a such column a

In by Ci-Ce before base

the tertiary phosphoramidate

under and

solvent one

flash

as,

steps provides employed

alkylator

in each

by

a

sodium

alkyl,

embodiment, chromatography

triethyl subjecting

base

a reduced

intermediate

column amine. first

solvent employed of: in

of

in Ci-Q step

intermediate POCI3, removing

a prodrug

or

amine in a

method

intermediate pressure.

chromatography Suitable solvent

potassium (a) to

step it

alkylator heteroalkyl,

to in yield

the with

a

obtained

or

(c)

the step Trigger-OH, of

to excess

on base

to

tertiary step is

the a synthesize of reaction

silica

yield N-2-haloethyl-N- (b)-is

hydride; dimethoxyethane, prodrug.

step

useful

of

(c) in

POCI3 C3-Cg

step

gel.

a

step amines is (a) on

in

separated in and

or silica (b)

is

a step In

(b) in

step

lithium

a

one is vacuo

with

base

(b).

gel (c)

2011202075 05 May 2011 20 15 10

5

schematically [0286] acetate. embodiment, prodrug. [0282] [0285] intermediate; [0284] to phosphoramidate [0283] heterocyclic wherein [0281] the another (0280] (0279]

yield

temperature

a

In Various embodiment, X4 In (a) (c) (b)

In monochlorophosphamide In In

another

one

=

one reacting oxidizing

reacting another one phopsphoramidate

the below:

and Br

embodiment,

embodiment,

of embodiment, solvent l-N-alkyl-2-aminoimidazole-5-carboxylate or

alkylator

embodiment,

step Cl;

embodiment,

PCI3 the

step the

e (a).

used monochlorophosphamide =

intermediate with

(b) prodrug 1-3

the

in the is steps

a

ankylator

step performed Trigger-OH N,N-di(2-haloethyl)ammonium

base

present the

derivative; comprising (a)

(c)

present used

in and

is

invention step prodrugs

dimethoxyethane,

in (b) at

86

is

invention

step a (b)

are

step the temperature

to

(b) performed derivative

steps of provides

(c) yield

the is

is provides

triethylamine. of:

present the

between

a can

diglyme, with phosphoramidate at

method

temperatures

be a salt invention

Trigger-OH method

synthesized

and 20-100°C

to or In

synthesize a a

another

for

base C1-C6 as

below

shown synthesizing

to

higher alkylator in as

alkyl yield

a described

a solvent 0°C.

below:

than

an

In

/ 2011202075 05 May 2011 25 20 15 10 5

prodrug resistant provided EXAMPLE phosphoramidate present “ [0292] patient alkylator are [0291] present Illa. Nos. al., methods [0290] Appl. bioreductive N-alkyl-2-amino-5-hydroxymethylimidazole below. 1122; [0289] [0288] [0287] Hypoxia NHRCH

provided PCT

Methods 2004/254103

Pub. Denny

in

of invention invention

to

Cancer by 2 prodrug Appl.

In of

Examples The Synthesis The need Activated COOMe

the

these

No.

one section. the the

by

group

et present synthetic of 1

of

Pub.

-N-alkyl-2-aminoimidazole-5-carboxylates present present WO the embodiment,

al., therapy

alkylating Treatment

alkylator has employed therapy are

and

references of

of Z US

anti-Cancer Nos. 04/009667,

3 provided the invention

. bioreductive methods

2005/043244, invention invention methods Pat.

with

formula

WO thereof

prodrug agents.

in Nos.

alkylatin the

Borch 04/85421 treating

in

to or

Agents and

are

present by 5,750,782; can further selected is

synthesize one

Alkylating employed groups

selected

provided

administering Hypoxia

et be and

g that cancer

” al.,

agents

adapted and ;

detail invention

Borch deGroot

from

is and supra.

from

WO 5,780,585;

phosphoramidate 87

known.

derivatives in in activated according

agents phosphoramidate

in can (I)

treating further et from

04/85361; H+ the the

In

et — to al., provides lead

(XXVII).

al., one

“ the can compounds Known

the EXAMPLES

(supra).

produgs

detail

to 5,872,129;

to

cancer

2001, employed patient embodimenL

kill references

NH development can the

and a

phosphoramidate cancer 2 in

method

methods CN be alkylator Current

In

according

US

Lin the a alkylator

reduced

exemplified one phosphoramidate

and in

” Pat.

EXAMPLES et

Matteucci cells section

the of embodiment,

al.,

the

Med. 6,251,933;

provided

Appl. of prodrugs

treating

present

prodrugs,

to in to phosphoramidate US cancers

the yield the below. Chem.

in Pat. entitled.

et alkylators

more methods

the

cancer by invention al.,

of section

various

Davis Appl. that

the

alkylator 8: and the the

PCT

rapidly 1093-

are

in

et Pub.

1- a

as

2011202075 05 May 2011 30 25 20 15 10 5

treating one phosphoramidate other as provides compounds combination administering , , with present prodrugs [0295] alkylators drug [0294] alone supra). treatment one resistant other for pathway, Alkyltransferase, [0293] transported and dividing slower

a

embodiment, second cancer

embodiment,

can

the anti-cancer having

therapies. or

cancer invention growing

produce alkylators

a in

In cancer or In which , The

and/or by method prodrug

resistance

line combination ,

of one one higher

drugs substantially with

one

as

prodrugs

the by

therapy embodiment,

are embodiment, cells

hypoxic

a

agents. Slowly

the

alkylator

the cells

or provides other administering present

glutathione, the

first such

of , is cyclophosphamide, oxygen

not

more

mismatch

present

treating administered to and present

resistant

line

anti-cancer cross-resistant of as

the

alkylators with dividing

In

cancer

strains , no invention alkylators

mechlorethamine the

prodrug phosphoramidate

containing therapy compounds one

, invention nephrotoxicity.

a another invention the present the

glutathione

repair metastatic

to

embodiment,

as region.

and

present

cancer present

such

(for

with a

of agents.

the

alone

by

thir

are

proteins,

anticancer invention the cancer

with

example, provides ifosfamide, ,

compounds alkylators. administering provides of carboplatin. d

The killed cells

and invention

invention invention

fine or cancer

the transferases,

In known

and

in

alkylators 88 latter region . In

the

in present

another therapy

and by combination

are

one a see, the melphalan, agent.

a by present

method the

method

Increased cells decreased glufosfamide,

alkylators. provides of

provides effective

is

hypoxic

administering as embodiment Hardman

prodrugs

the

invention administered the embodiment,

the ,

compared prodrugs the

In survive

invention

compounds

present

of

compounds

one of nucleotide

are

with

a phosphoramidate cancer treating

in activity permeation

treating

method

In et embodime^

of

treating

postulated the alone

which

al.,

mechlorethamine, to another other the invention

the

as streptozocin,

provides

in treatment zone the the

pages

cancer phosphoramidate of

a present

of a

combination excision or of of

first

anti-cancer cancer are

cancers cancer present guanine-O

the

in

treating the act of embodiment,

to

not

1393 combination

alone fine present actively by

a as present a

be

invention. by

method resistant

alkylators cells

repair

cross-resistant

administering a

invention

resistant

responsible

therapy bendamustin,

alkylators

and source

cancer or 6

agents. with -

invention in

in 1433,

the of

to

of a the

by with to In

In

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 5 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 10 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. In 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 15 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 20 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 cell carcinoma, squamous cell carcinoma of both ulcerating and papillary type, metastatic 25 skin carcinoma, osteosarcoma, Ewing's sarcoma, veticulum cell sarcoma, myeloma, giant cell tumor, small-cell lung tumor, gallstones, islet cell tumor, primary brain tumor, acute and chronic lymphocytic and granulocytic tumors, hairy-cell tumor, adenoma, hyperplasia, medullary carcinoma, pheochromocytoma, mucosal neuronms, intestinal ganglioneuromas, hyperplastic comeal nerve tumor, marfanoid habitus tumor, Wilm's tumor, seminoma, 30 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

89 2011202075 05 May 2011 30 25 20 15 10 5

including been thiotepa, regression agents, cisplatin) [0300] (CAVP-1 (CAV); variety phosphoramidate can chemotherapy death. States, of [0299] combination 22 often illustrative Attorney and invention types entirety head multiforma, hypercalcemia, limited cancers [0298]

illustrative December

be

PCT alkylating

and involves of

used and most Current

of

to

herin containing

cisplatin, cancers Likewise, the

6). Lung treatment Pat. Docket The

neck combination are of lung

as purposes

as

Modest

of combinations

leukemias, ovarian therapies

discussed provided

cancers Pub. by 2005, phosphoramidate

with

a the

cancer cancer, protocols who agents,

cancer,

renal

single

that

reference No.

alkylator and and

simultaneous

No. several have significant or

die

and survival

below.

cancer. can

cell

021305-005900PC;

containing

cisplatin without affects

carboplatin.

as including ovarian chemotherapy agent

WO in especially

within

further lymphomas,

PCT

been

for be provided tumor, the

consisting

different prodrug, Tidmarsh

2005/076888. treated

Those the

or

benefits

The more

following Pat.

reported

radiotherapy areas

cancer, 1

(VP-16); below, in

alkylator or treatment

year polycythermia cyclophosphamide, hypoxic

non-small most Appl.

combination

by

successive than of

with

Current cytotoxic

of examples

malignant

et of

of regimens from

the

skill

pancreatic

a

hypoxic for

active al., references,

cyclophosphamide, diagnosis, entitled 100,000 phosphoramidate the

and

prodrug

methods

regions and

of

non-small

in Several combination

PCT or

combination phosphoramidate

cell

90

cyclophosphamide, lung

the

drugs drugs US administration surgery. of

with have melanomas,

tissue.

vera,

“ Pat. lung

males

combination

art cancer, Glufosfamide

Pat. amenable can

cancer

described making of each

have in

existing will

been

ifosfamide, Appl. cell

adenocarcinoma, cancer, particularly these

App.

the Such and A

chemotherapy

of

therapies appreciate and involve produced lung

phosphoramidate reported alkylator treatment

it

which No.

50,000 cancers No. to and

cancers herein. combination the of breast prostate

therapies alkylator treatment cancer.

PCT/US2005/0473

combination a 60/760,599

epidermoid leading melphalan,

doxorubicin

the variety is be

for

females for

at

prodrug are that cancer,

incorporated

of include used Thus, integration

least cancer.

ovarian

small

glioblastoma (etoposide

ovarian discussed prodrugs

are

cancer with

cause and of

in therapies.

temporary in

in

also

colon anti-cancer

and alkylator

carcinomas. can

the chlorambucil, but cell therapy

a and the the Examples

cancer

of chemotherapy cancer

described. 60/719,787 treatment

be

of are

of lung

in

description cancer for

United cancer, VP-16 plus

14

used

the its not ”

A

prodrug

, include filed

cancer,

have

in of

of

on

2011202075 05 May 2011 30 25 20 15 10 5

prodrug limited body-directed (VDEPT), be meeting [0303] colon combination benefit, likelihood alone Agents chemotherapy United agent(s) invention [0302] treating and , cancer, have has and replace described methods [0301] six cisplatin

used

to been 5-, is

been or cancer

eight

the

to

States in for

therapy including

5-fluorouracil the an prostate in currently In

Cancer the or reported for

Cancer

is various “

of second

herein reported the combination agent

gene-directed one

unmet cycles. carboplatin used

with using

treating reducing foregoing

and enzyme

in treatment

” version and

cancer

(BDEPT). of patients or

of is

in the

most treatment known is

used.

to need

a

melphalan,

the used

for cisplatin. in the such The

likewise prodrug,

consist ovarian compounds

prodrug measurable is addition

large use

treatment common prostate of in for in with

of the compounds

as combinations, with approaches enzyme

the which combination

in prostate

better a

with

most The primarily

bowel

other

single cancer a

can late

treatment

Combination

advanced therapy cancer and to

is

cause

estramustine general

a

methods.

and the prodrug offer treatment effective

stage tumor the

phosphoramidate drugs, hydroxyurea.

is agent cancer

in

and

to agent(s)

methods the most characterized ”

of

which of

significant

with

disease

cancer (ADEPT), either methods,

colorectal methods uses masses hypoxic

cancer but second or

therapy

include

treatment

common chemotherapy

methods in cyclophosphamide 91

is a

of currently phosphate to

described

existing therapy

phosphoramidate following associated

a death by replace most

The tissue.

a

described ” phosphoramidate

therapeutic “ the

cancer

phosphoramidate (GDEPT), 50 virus-directed by

alkylator malignancy for for

present

alkylators common in percent

hypoxic including used. such

this

an plus this herein, men

Several

relapse

has is with

agent herein

combination disease. also

disease.

prednimustine

invention

proven prodrug benefit above

and or

only regions. at

cause

and

estramustine

in

alkylator but used chemotherapy after more. or

3-

enzyme “ provide

men

bacteria-directed

alkylator

in

age the a to

not

alkylator to and

5-Fluorouracil of

to hormonal of

15 addition

4-week

provides

be

methods

in Using 55, cancer

limited

treat While

the to therapy,

potential

prodrug

prodrug the prodrug of

20

and and present

prodrug

only prostate phosphate,

United

prodrug

percent 5-FU intervals

to death protocols

cisplatin, to this treatment.

methods

for

the either

marginal

as “ for therapy forms

anti

cancer treating is in

enzyme

in

States are

useful can

­

the for to

not and for

.

/ 2011202075 05 May 2011 30 25 20 15 10 5

ankyiosis a on multisystem are multiple another psoriasis, elevated, by In administered vasculitis disorders, diabetes, [0305] duodenum, periodontal adhesions, syndrome, neoplastic disease, hemolytic bronchitis hypersensitivity demyelinating tube hyperproliferative atrophic consisting abnormally cancer [0304] compound

one a the administered

prosthesis

diseases,

cellular embodiment,

hyperproliferative embodiment,

sclerosis, multiple

gastritis, of scaly In

ulcerative multiple n of

vasculitis

anemia, vasculitis, and

primary of

stenosis

another joints

some small disease, increased of

chronic, the to

allergic

hyperproliferation giant

implanted lesions. the neuropathies, chronic

treat

central to vasculitis

sclerosis,

bowel,

embodiments

benign affected.

a

present sclerosis, disease

prevent

inflammatory

or sclerosing aspect, colitis, disease cell

the associated polycystic

relapsing, a

the pancreatitis,

angiitis

restenosis, rate

hyperproliferative

obstructive

nervous In hyperpriliferative arteritis,

hyperproliferative

and in

prostatic

diseases

invention.

another treated or

(Henoch-Schonlein ulcerative the myocarditis, a

In

characterized a rheumatoid

hyperproliferative dermatomyositis,

colon), amount

subject and

another

cholangitis,

present

with kidney inflammatory

of system,

graft of neutropenia, scleritis,

granulomatosis

according pemphigus

embodiment, the hyperplasia, airway characterized

keratinocytes

congenital

silicosis

by proctitis, of

In

invention, embodiment, rejection, invention disease,

cellular myositis, and another arthritis, coating

disease

by

disease disease, scleroderma, rheumatoid

diseases

to Wegener's progressive

92

disease and

vulgaris,

inflammatory

purpura),

eczema vasculitis the polyarteritis

deficiencies bullous

disease

proliferation).

the the hypersensitivity provides

embodiment, by

a selected

restenosis, (Churg-Strauss other treated

which nasal

chronic compound present

cellular

hyperproliferative the prosthesis treated

that

arthritis,

primary including

pemphigoid, resulting compounds granulomatosis. forms polyps, strictures

irritant builds

demyelination associated

is from a can

method sinusitis,

method psoriasis, of nodosa, is hyperproliferation

bowel and

of lead

of rheumatoid

the the

with

the

up In glomerulonephritis, nasolacrimal serum dermatitis,

the

pneumoconiosis, benign

pneumonitis, from atopic

disease), of

hyperproliferative

one

complement

on is to group

disease,

of of with

a polyangitis present Crohn's bile

coeliac

selected

destruction composition the a

sickness, embodiment, treatment cellular the disease

disease

dermatitis, in prostatic

ducts,

connective skin consisting

present arthritis,

the asbestosis,

inflammatory

Kawasaki's

invention disease,

disease,

duct from

to brain. proliferation treated

(e.g.,

strictures

characterized system,

surgical overlap

of

form

and

hyperplasia.

diseases, invention type

containing non eustachean the

a

psoriasis,

the

of an tissue chronic disease

In

is asthma,

is

group ­

I

(of

2011202075 05 May 2011 30 25 20 15 10 5

physician. (e.g., agent the product amount among administration, [0308] type prodrug and will gut. intramuscular, administration oral must formulations, only hypoxic modes a can using pharmaceutical Illb. [0307] [0306] treated abnormally

phosphoramidate

cancer,

corresponding be

recognize routes, of

molecular If

ready-for-injection released Fonnulations, be

a

other

such cancer effected of

of comprising is

phosphoramidate prior

rehydrated region The

Administration A

administration, benign the localization

intraduodenal

phosphoramidate

activation and factors.

amount to phosphoramidate a by

being

that or intravascular

the

weighfl phosphoramidate by of

formulations its thereby

a prostatic a

a

formulation phosphoramidate severity

any

a

entry formulation that

alkylator or modes

tumor.

treated, phosphoramidate

of

diluted,

is

of method

solubility dose,

a

formulations block

into not

of

the formulations, phosphoramidate alkylator routes,

hyperplasia,

of of

Many a or

the

desired,

prodrug

cancer, for the phosphoramidate will administration, alkylator

the

respectively,

urine infusion),

of that alkylator that phosphoramidate

administration

large

parenteral alkylator and

cancer, the

be cancer

prodrug

enables results

alkylator flow. the dependent

then

phosphoramidate can hypoxic but

alkylator

intestine

prodrug a and

rate

prodrug disease

topical, be drugs and

also the

prodrug

in

delivery described alkylator prior dosages injection

formulated

93 of dosages

absorption the practitioner

prodrug, lyophilized and

alkylator

will are or administration, on prodrug to

in

and to

age, contained

alkylator

a colon. normoxic

the

administered which can

injection. typically subject that of

prodrug

(including rectal herein. weight,

subject

the and alkylator be

for

may

prodrug can of

The

can prostate or

formulated prodrugs

prodrug the a routes. such

in

be

concentrated cytotoxicity),

Described

phosphoramidate be be

administered,

employ and being actual In the

discretion the used

intravenous,

activated formulated

by prodrug

administration, addition for

dose condition epithelial

disposition

Those

intravenous selected

to

treated, the when route

for

a the

administered route in

treatment

will

administration

of

to

by

formulations of

this site of treating the

and

as

the of cells these

the for

bacteria skill subcutaneous,

administration of depend

of section cytotoxic of

alkylator the

prescribing thus injection, severity including

action,

the grow in

of

patient, cancers

the

the

and prodrug

cancer in on

are

that

the art

the by the of the

and not

2011202075 05 May 2011 30 25 20 15 10 5

of over phosphoramidate range [0312] embodiment, administered dose [0311] administered treatement (combination) daily, 0.5 about invention or about invention single to 35 treatement throughout side central kg [0310] adequate, human, some a [0309]

a

patient

divided about

mg/kg/day, mg/kg/day body phosphoramidate

two

in effect;

of

0.25

either 2 instances,

or

In the

catheters

to about this In

weekly

10 weight, divided

wherein In

one In for for

while

about

of doses. range to another

of

the mg/kg/day; larger

one

as

one would

on a

once

treatement cancer about treatement

embodiment, cancer

5 with

dose in a about

day

once

to cycles. embodiment,

in dosage monotherapy

embodiment, 8 of alkylator (PICC or single doses.

In an doses a mg/kg/day;

about

other embodiment,

standard

amount 2.5 by about in about

week.

and

one and effective alkylator 0.5 every

the

infusion

mg/kg/day;

or about

line)

levels

of can of other In cases 30 embodiment, to other

3 range 0.1

prodrug divided

cancer three cancer one a to about to

mg/kg; of also

larger and

to

the 0.5 about

about the hyperproliferative (compound below dosage

about prodrug still

hyperproliferative

care

for embodiment, of

about

for days

be

portable effective to 20

present

and

and

doses.

about is larger an

about

dose

about therapies.

divided

about 0.05 20

treatment 2

mg/kg/day; the

administered

is hour

for 35 other to other the

is mg/kg;

typically

is

lower

0.5 about

to not 0.25 doses mg/kg/day two

invention In 10 intravenous of effective

8

dose

administered or

about

hyperproliferative mg/kg/day, into hyperproliferative

94

one to

to the

greater the

continuously weeks. of to

In

limit 4 about about about

can

of

present several about

embodimenζ

mg/kg/day; effective about human diseases

one diseases in 7

once a

be dose provides g/day,

the

of compound in

than

6 8 embodiment,

15

bag In

employed

0.5 the single to mg/kg/day 1

range

a and

small patients, invention intermittently mg/kg; of another mg/kg/day

week 500 is about

is dose about and to

aforesaid

using a

in

about in

a and

compound

about

or of

mg/kg

doses

the method pump. the diseases diseases

the

of for of

0.2 10

or

divided about

without embodiment, about a the

a

dose range the

alone)

is 1 range

peripherally mg/kg;

four the 12.5

15

compound

to to

range for

administered and patient maximum

present

mg/kg/day;

0.001 of

(less about

about effective 2

is is is

administration weeks. mg/kg

of

as doses.

of

about mg/kg/day causing

or cancer administered in in

can

or the described

about

frequently); in the the

weight 5 to

2.5

8 invention

be

conjunction present a

mg/kg/day of of

mg/kg 0.25

about

inserted

For daily range

In range dose dose treatment g/day.

any

more

the 0.1 the

about for one

a and, to in

present earlier to for harmful O.lg dose in

70

5 of is of

about single

i.v.

than

for days

about In the a

0.5

kg

in

per in

2011202075 05 May 20 20 15 10 5

’ HED standard animal

the humans animal prodrug prodrug prodrug [03131 another In prodrug In mg/kg phosphoramidate embodiment, of of accordingly,

another another a

about Baboon Marmoset Squirrel Monkeys Rabbit Dog Mini-pig Micro-pig Ferret Mouse Guinea Rat Hamster Animal table

phosphoramidate =

b dose a animal to can

To

embodiment, ranges, and is is is is For below:

1 Guidance about

about pig about embodiment, embodiment, mg about about

be convert monkey

by 13

other example,

a

the

converted dose of phosphoramidate HED

500 human

25 25 1.25

10

a therapeutically

alkylator mammalian

phosphoramidate

animal concerning in mg/kg

mg/kg mg/kg mg/kg convertion

the mg/kg

mg/kg alkylator cynomolgus,

equivalent

the the

to therapeutically

dose

to to to of the prodrug

therapeutically therapeutically

to

x about about about the

animals.

corresponding administration (animal

about

prodrug/kg factor. in

alkylator effective | body

dose mg/kgto

50 250 is rhesus, 150

Human alkylator

12.5 administered

mg/kg

weight

weight

A

For (HED) mg/kg mg/kg

effective

therapeutically prodrug dose mg/kg

of effective HED effective species

or Equivalent 95

human of

patient can

prodrug/kg

of

in

stumptail. can of of is

body

the kg/human (assumes of

a the the

dose also is

be daily not in

body

patient dose dose administered

equivalent body body weight.

weight

calculated a

be

of

listed Dose daily

dose

effective

provided weight a of of of

a weight weight

phosphoramidate 12.3 weight 5.3 6.2 3.1 3.1 4.6 1.8 5.3 6.2 7.4 to 1.8 1.4 1.1

patient a a 60

dose or of (HED)

of In phosphoramidate phosphoramidate be

kg

for the dose

a one from

of

treated.

in of of phosphoramidate

in

dose

human) in by

weights the patient weight

a embodiment, conversion the the the

(HED) kg)

the and daily

patient determined

patient patient range

0J3

formula: In from

in to to

outside dose

.

as

another

alkylator mg/kg, be about

to

of

described

studies alkylator to alkylator to factor treated.

in be about

be be a

500

for the

the alkylator treated.

divide treated. treated.

8

an range in

5

mg

In

in

2011202075 05 May 2011 30 25 20 1 10 5 5

continued prodrug [0316] alternatively, at weeks within prodrug six weeks [0315] within six prodrug time. in dose), administration used. administration will administrations. administered smaller dose administration). administration phosphoramidate related days, time. one [0314]

least

any

consecutive consecutive

be embodiment,

can

or For within Administration within every a Typically, a event,

one

apparent

embodiments,

is is doses

six-month is six-month In at Administration

be To

for

administered example, administered administered

month least one

administered other

multiple achieve at a a a twice through

once phosphoramidate

twelve-month is selected twelve-month least weeks, embodiment, weeks,

10

to For not daily

alkylator or a day,

period, period,

one

consecutive daily, whether phosphoramidate

every

one,

at limited therapeutic example,

once days,

administration

the

or, administration

or, or

least

of at by

at at schedules

two,

less or once

least alternatively, least skill third course alternatively, least or, or,

the

every prodrug typically

two, divided administration to

the

period. period.

alternatively,

alternatively, frequently,

three, practitioner

the in daily,

one 3 10 2 day, days.

alkylator

administration effectiveness,

days of three, days other the

days dose other

administration

is

a

or into

four,

for is art or

In alkylator day In for Depending usually per per

once

day

per for four, can

for the for

than

one of one at

three

at

in

(including

five prodrug week week

and

at least

cancer

month, for at is (qod) at

daily for be least

one

a embodiment

96 embodiment,

five

consecutive

least

daily least least administered

week

or

smaller divided the prodrug the at at

of

for three for

embodiment

3

dose at is least or least on

treatment

per

convenience the 5 therapeutically is

consecutive optionally two,

two,

(including,

particularly least at

at can

at consecutive by the repeatedly

consecutive

least therapeutically

day least can doses two, least two, into

is

infusion three, be three,

dose, six

daily a administered

but be a

appropiate two, phosphoramidate two multiple two,

three,

three, six

phosphoramidate that, months and

administered a

at

four, can formulation, four,

as

days phosphoramidate administration consecutive

of

smaller

convenienL administered

least

three,

three,

with days,

administered

as

noted,

four,

four, effective the include days,

five five

used will times in

20

effective a patient,

in

four,

at four,

a

daily five pump five

doses

or or and a

days

be 6-month some least herein,

divided multiple to

in at at

and

daily

months, employed. or

or five and five

often

for the

alkylator

least least

over multiple per

or and the alkylator

Ί can thrice

at at instances,

dose

route

consecutive

or intravenous a

patient. or “ least

dose least alkylator

month,

entire

daily period.

period

daily for also six a six

at at

period is or, daily.

of

least least at

weeks weeks of six six

” be

In

daily least

a but

of for In

of

It

2011202075 05 May 20 30 25 20 15 10 5

concentration administration the one consecutive phosphoramidate [0319] a the wherein patient of prodrug phosphoramidate quided multiple-round period [0318] twice, more two period some administrations, and can phosphoramidate administration. be for five [0317]

phosphoramidate

patient

present

cancer administered month,

several be

such to to

times,

embodiments,

or

ranging ten weight) ranging by a ten

the in dminis In

In

three three Consistent weight,

in this

administrations, invention,

the consecutive

at

optimizing

days, days,

one administration

sometimes

the

curve least

can

ranges

disclosure.

or

tered to from from for schedules

aspect, patient For

and a

a alkylator

alkylator more ten

10 typically alkylator be phosphoramidate

a

10 alkylator week, (AUC)

specified example, one a once with consecutive one

such

selected or

the stated

days phosphoramidate

a

times days, “

with

five phosphoramidate

administering to to dose

Illustrative

a administration for two, daily

prodrug

of prodrug

over per 25 more month,

prodrug several several

(e.g.,

prodrug

to

a or

with administration

a

to

minimum in to

and

no-treatment month, three

ten phosphoramidate

for

for the

achieve

some often

150 days, 1

a

frequency

day two ” weeks

at include weeks mg a

course

can no-treatment alkylator

refers therapeutically or for

week, mg least

and

embodiments, three

at a treatments months, to five

be

alkylator

number 3 regimens a therapeutically of

least

between

between 1

alkylator maximal

consecutive

three at to

of administered

those

or a to

(with g to will

97

of least phosphoramidate

prodrug (i) treatment

of

for ten 3 ten

three

a

to days a administering

of be

described alkylator

prodrug phosphoramidate (with a

several

can administrations phosphoramidate

of 20 administrations,

each ten, each

phosphoramidate days

prodrug sustained apparent effective

other a months,

per

days be in

days, phosphoramidate or a

multiple-day multiple-day effective

in

within accordance

The repeated

phosphoramidate

week, weeks

at is

prodrug per multiple anticancer herein,

least treatment administered

5

to

theoretically dose area six

month. consecutive

a

alkylator

the a

at or phosphoramidate

specified months,

dose five

qod

once,

least

under regimens

such more. or has skilled

alkylator

alkylator “ with

rounds alkylator

treatment.

treatment. agents,

five

to

can regimen

or

a

3

twice, as

ten prodrug

alkylator the therapeutic

regimen

days the every

Thus,

or

to be

practicioner optimal administration alkylator days, time

consecutive

plasma for

a

ten prodrug

present prodrug repeated a of

year per

prodrug) or according

other a period,

a 7 Other

Similarly, per

three

patient

prodrug of week alkylator

dosing or

prodrug)

effect

methods a kg

day per

longer.

once,

or

of for days,

can

to kg of for

on in

2011202075 05 May 2011 30 25 20 15 10 5

prodrugs will prodrug remain [0322] present 30 remain another 20-80% prodrugs embodiment, (update prodrug method [0321] alkylator require between In compounds invention alkylator invention, of observed, with prodrug, determine regimen [0320] for

minutes.

one the

be any

a

present

lower. greater embodiment, phosphoramidate greater

invention

an embodiment, with of of single unchanged

can

In of

will 10 In that prodrug as prodrug. In

can

the

or infusion

administration, and efficacy.

the a

another to measured of one In increase

human related

for the remain

result

invention, have than invention than

30 administration. the present another then

embodiment, the

present

which

minutes. having

present

In 80% time

80% when a

in

embodiment, resumed. example embodiment, convenience

the

As

up the in the

one by

a

invention

embodiment,

vivo

when maximal

is

to compounds

employed in unchanged unchanged

its invention maximum alkylator present a AUC,

incubated

embodiment, and invention understood

20% longer treatment

A therapeutically

half

and

short

incubated the the A

while unchanged

upon invention

life

higher of exposure data the pharmacokinetics

half the kind

prodrug

provides for

half tolerated

with include when the

of can

examples of that present

bioreductive

minimizing

in the if life. reduction/activation

the of

between

patient,

the life

C with have

available) the

mouse is

cancer ma treatment incubated

present

can

provides

of effective when

phosphoramidate A longer

x of phosphoramidate 1,25,

art

98

dose invention

will mouse

the a short

the be of

in for without liver

1

that

incubated tumor

phosphoramidate

suspended contribute

vivo the

and (MTD) phosphoramidate

to invention

than protein group other

half

of of

phosphoramidate with

liver dose

300 microsomal

is maximal

36.

the half cancer

provides

that treated

cells departing life

cancer

of minutes.

mouse

microsomal

and for

phosphoramidate

The for

in

life

with

required the

of

to can temporarily

to

alkylator that

a can alkylator undesirable 30

the

with plasma toxicity

of higher hypoxic

phosphoramidate a

therapeutic

phosphoramidate have liver mouse

minutes. phosphoramidate

protein

prodrug. between determine

from phosphoramidate

In alkylator

the alkylator

for

one

microsomal alkylator a protein

the prodrug concentration

prodrugs while

the

phosphoramidate in

liver tumor a

if for

MLM phosphoramidate embodiment,

patient vivo

3

In toxicity scope

drugs,

alkylator

the

to prodrugs

30

microsomal

prodrug the another for

zone

half of prodrugs 10

minutes. dose,

stability

that

AUC

of 30

alkylator

side

the alkylator protein treatment minutes.

alkylator

is

life

form the

alkylator minutes

(C

remain

prodrug present

the can

effects of

will

ma of the

that

the of x)

a In for

a

2011202075 05 May 2011 30 25 20 15 10 5

a formulations magnesium binding various tablets ingredients organic symptoms administration conventional dosage [0325] as or formulation, form phosphoramidate [0324] defined indirectly phosphoramidate be prior longer plasma were phosphoramidate

[0323] clearance Various diffuse similar

a emulsion;

temporarily

suppository.

suitable measured treatment.

containing

contains forms disintegrants,

agents solvents.

and stability, not

type pharmacokinetic Suitable A In

by and (CL),

such stearate,

formulation

only

reach

re-treatment

of

solution, for pharmaceutical

can cessation suitable for

such normal (drug

a

stopped

and significant

as topical

area

phosphoramidate by In

oral be

and The A various pharmaceutical

alkylators alkylator other alkylator-

flavorings,

as any

specific formulation listed sodium holiday) employed

under such

Cmax

administration pharmaceutical for and organ sucrose,

of

administration as event, parts of

regimens, single excipients,

symptoms).

parameters in suspension;

severe as

a of

concentrations released curve

released lauryl

phosphoramidate days T clearance

the carrier

starch, phosphoramidate of can

m

binders,

as in gelatin

administration the conjugate. of EXAMPLES

toxicity soft be symptoms carriers but (AUC),

sulfate, alkylator

a

the

tumor

or therefrom

alginic ended phosphoramidate as

therefrom.

such

individualized such

and compositions

for of and Therefore, excipient. as excipients, dose a

tablet, a is

an

include

mouse

hard parenteral

as of or

and acacia. as phosphoramidate at

The observed

acid,

prodrug

are can

ointment 99

citric

a other

the volume

section

alkylator

(which phosphoramidate talc filled of alkylator

observed. capsule, phosphoramidate be

liver

time and

inert

precise an

and acid tumors

Additionally, can

adjusted

can,

by

injection intermittent described

gelatin

during certain

of (see or microsomal

when can

the alkylator

diluents be

can drug prodrug

distribution prodrugs if pill cream;

dosages

used be like. in

also desired, The

be

the

to

capsules. repeat powder, holidays

alkylator complex the measured

as

employed

reflect herein. Hardman to period or

Thus first

prodrug

and can, alkylator case lubricating a dosing

prepare

alkylator- fillers, and of sterile

stability

administration, contain

for under organ

the

for

of sustained

for patient that

silicates, prodrug of will

Preferred Solid or

rectal

a

period present

example,

can

together temporary et oral water

solution,

metastatic prodrug the determined are

steady of typically

(MLM additional al.,

T

agents

be

compositions

tolerance

toxicity administration, m tablet based administration

or supra). release can and and

in conjugate

invention materials,

a state

with

be unit or stability), suspension

dosing

such

be

various with halting forms

on include disease.

a in no

(Vss), of

a

as

the

can can of

of of

a

2011202075 05 May 2011 3 25 20 15

10 0 5

[0329] prodrug Hie. alkylator other generated and in the drug-factory to prodrug leading containing the the Company, disclosure. polyethylene [0328] of phosphoramidate suitably When [0327] desired, ethanol, can therefore, [0326]

the normal

active

tumor

hypoxic present

start Combination be

anticancer

normoxic

aqueous

combined to can

In

can buffered, to propylene emulsifying prodrug,

The drug Methods

Exemplary

from can tissues.

a

include

Philadelphia, accordance increasing

divide. invention

be

higher For region

diffuse to

methods glycols, reduce

are

co-administered

a produce

suspensions region agents examples,

phosphoramidate

therapies The with

as alkylator

known, if

lactose

of

concentration of At glycol, from

desired.

described

parenteral toxic

agents preparing

are

populations a propylene

use

this

with of various or of

tumor.

within

Pa.,

effective

the cancer the cytoxins or

or of point, side-effects see

glycerin,

prodrug

the or or

milk

will the hypoxic 17 tumor

Remington

sweetening in suspending elixirs

a methods in Once th various administration

prodrug treatment tumor glycol

such of be the

Edition

combination sugar of

in alkylator administered

the in are or apparent,

rapidly killing

following released

cells are

cells sterile

arising combinations phosphoramidate an

pharmaceutical or destroyed, and

of

to desired (1984).

dextrose

alkylator s or agents, employing

and

the

can

generate

Pharmaceutical high the prodrug 100 dividing

aqueous flavoring

in

to due

forms invention,

with kill

section. be most

in

the those molecular for

together

killed a to

combination the solutions.

for

other hypoxic hypoxic

of oral the

thereof. normal include difficult

cells. solutions,

a

skilled

cancer agents,

killing this compositions phosphoramidate

phosphoramidate

by

administration, alkylator a anti-cancer

with

phosphoramidate invention weight The the Sciences,

cell

region region solutions

in

cells to Such adjacent coloring

phosphoramidate

with diluents hypoxic for

kill this toxicity.

within polyethylene in

dosage example,

can a

cancer

art the

with phosphoramidate agents adjacent or Mack

or

normoxic

matters in become

those such phosphoramidate the

region

suspensions alkylator

the

alkylator a view After

forms

cells

specific Publishing prodrug

(

alkylator

“ tumor, aqueous

as

known, anticancer regions

or

of acts

normoxic cancer water, glycols. growing

alkylators cancer

can dyes

this prodrug

within

amount as relative therein

be

of

or

a and, cell

a

cells by

in

of

if

2011202075 05 May 2011 30 25 20 15 10 5

phosphoramidate and continued to example administered other phosphoramidate [0332] administration with administration alkylator another embodiment, region, [0331] the the of agent palliation sensitivity survival diminishment prodrug [0330] agent Co-administration cancer administration agent

administration

cells

treatment regions known

a cancer

to alone,

phosphoramidate

).

.

therapies, the

anticancer otherwise

combination In

be or and

prodrug As The Without after or

of

phosphoramidate anti-cancer one of other adminstered

therapy.

used

contemporaneously stabilization e.g., the with

a cancer

co-administration

tumors

of of

of the

phosphoramidate can embodiment,

of anticancer alkylator

beneficial extent alkylator the the

herein, such

and greater

resistent a intending

cessation agent

provides

in the

phosphoramidate

cells Agent Agent, In

therapies that Agent some

agents as,

Agent,

other of

alkylator to

(also

a alleviation

of

to are for prodrug

disease, prodrug phosphoramidate the

to

agent (i.e.,

therapeutic

cases

a of

(i.e., to the the

are alkylator

a

embodiments, the not in example better

referred

patient and

be phosphoramidate administration as

disease of

anticancer general administered

the

alkylator

with

anticancer the has prodrug efficiently provide described bound

treatment a is delay is

therapeutic course phosphoramidate or

initiation first

a administered

alkylator or the

prodrugs

to co-administration amelioration synergistic results.

state,

targets

allowing by or herein

initiation administered

one is prodrug

agent,

of

slowing

any

in

agent 101 a

killed continued with alkylator

partial phosphoramidate as of

or the EXAMPLE of

prodrug

the

result of particular as,

part the more alkylator

the

allowing a other an

or

effect the by

is

after rapidly of phosphoramidate “

of of Agent.

Agent

or otherwise anticancer of other first

the

than the prodrug invention alkylator of

disease throughout one

prior complete

therapy). the

is

the

on of

several

anticancer other administered mechanism prodrug continued

” dividing cancer section. lower administration or

same a ) initiation induction

In

to when phosphoramidate

more

progression, is one refractory

administration cancer prodrug alkylator

agent

target

advantages “ remission, course In doses

therapy), co-administered

the

is

embodiment, a

symptoms cells another

agent

during

phosphoramidate

first

alkylator

or

to of

or course the therapy.

prior of

increases of completion be

cancer

effect, in

prodrug

to

administered

of the hypoxic alone.

therapy. used

the

and amelioration, part prolonged embodiment, treatment.

over the

to

of anticancer

of of

prodrug

alkylator normoxic

treatment such cell

See of anticancer for

a

the the

known is the ”

cells the

with death. first treatment of

for

In Agent, cancer, co

the

one prior ­ is While

in

a

2011202075 05 May 2011 25 20 15 10 5

an therapies. radiotherapy, alkylator is [0335] days a administered viewed alkylator administration [0334] receptor receptor superoxide. with present agent tissue superoxide with period completed inhibitors, (0333]

phosphoramidate

co-administered effective

during

a oxygen

is

from

chemoprotective of

as

invention ” ”

a

prodrug, In Anticancer prodrug,

In

from generated administration thiol administration

a prior

amount one

in each the the

phosphoramidate In in an to

noimoxic

of

reacting or

toxic another version any yield presence appropriate

to cycle.

the

provides a a each with

alkylator

the of phosphoramidate

or disulfide.

from drug

anti-cancer effects superoxide

one

all

administration

cycle the of

embodiment,

with In agent

tissues

of

of of of therapy a

the or

one Agent a phosphoramidate NHOH(+)

the

a oxygen,

surgery the of

prodrug

phosphoramidate proteins

more of

second method

or In

alkylator

aspect anticancer Agent.

can multiple administration

agent(s). a one and

today according

chemoprotectant chemotherapeutic

lead

procedure,

the

is drug. Z embodiment,

the

alkylator and of

of of 3

For administered

.

prodrug typically radical

to the

the Superoxide treating the cycles

nucleic drugs.

In

unwanted certain A

second invention, 102 to

chemoprotectant

alkylator the

phosphoramidate

alkylator

prodrug

a (as

or anion of

schedule administration

cancer context In involves

acid

any

treatment well drugs, the

drug. one

is

on Chemoprotective

agents, side formed

combination (see prodrug chemoprotectant

a a

is as

a prodrug

using

embodiment,

phosphoramidate cytotoxin

of

such

daily administered repeated effects. a multiple

below).

administering complete

with

an

upon

can

the

as

and

basis can alkylator effective

administered

certain the react a

In

and

the at rounds, prevent of be phosphoramidate

for one

other each set the

such

initiated reduction agents

the

with in can

at

topoisomerase

of prodrug amount embodiment,

chemoprotective

combination a

production

round. alkylator

least Agent;

“ or additional

reduce phosphoramidate

cycles) the Michael-

protect

in “

cycles, and

two combination of

Michael-

of

can

in

Z can

prodrug or

3 general, healthy

be of ” reacts

the with more

be of

2011202075 05 May 20 30 25 20 15 10 5

, 2-ethylhydrazide, interferon-alpha, elfomithine, , , , 6-, tubercidin, olivomycin, melphalan, , , azaserine, carmustine, estramustine, triethylenethiophosphoramide, phosphoramidate , analogs improsulfan, section. administration. with phosphoramidate administered administering be of [0338] [0337] agent, [0336]

the administered

some

additional

the

thereof The Chemotherapeutic

When

,

two

bestrabucil,

ubenimex, dideoxyuridine, time

novembichin,

chlorozotocin, peplomycin, elliptinium imatinib,

piposulfan, pulmozyme,

mitolactol, via ifosfamide, mopidamol, Agents

daunomycin,

the agents

(refrence

between a See interferon-beta,

at

therapies, , the

alkylator phosphoramidate alkylator agents thiamiprine,

the

for same

can , can zinostatin, cactinomycin,

bisantrene, same

acetate,

example

,

benzodepa,

, simultaneously administrations.

apps), gefitinib, be phenesterine,

be

nitracrine, aceglatone, or

,

prodrug prodrug a , 6-diazo-5-oxo-l-norleucine,

agents

time administered

administered phosphoramidate different

trimethylolomelamine, thioguanine, razoxane,

,

glufosfamide,

interferon-gamma,

,

combination or

carboplatin,

mechlorethamine,

that

of alkylator

is

can

carubicin,

, ,

porfiromycin,

aclacinomycins,

administered aldophosphamide routes. the

prednimustine, can

,

be and

sizofiran,

enocitabine,

invention as One simultaneously flutamide,

administered denopterin, be triethylenephosphoramide,

ancitabine,

103

the sequentially prodrug

therapies

gemcitibine, used defofamide, alkylator of carzinophilin,

same

phenamet,

skill 2-deoxy-D-glucose,

,

with spirogermanium, in interleukin-2, puromycin, include,

gallium is

mechlorethamine

or

combination chlorambucil, , in actinomycin ,

pteropterin, ,

as used

prodrug mycophenolic

glycoside,

separately. an different

the

and

or described

, erlotinib,

additional

,

but can

art in

nitrate, chromomycin, possible

combination

dacaibazine, streptonigrin, will

are be and

lentinan, formulations

5-fluorouracil,

aminolevulinic

with

trimetrexate, F(l), 6-azauridine, administered

not

understand in

For chlomaphazine, uracil hydroxyurea, additional meturedepa, ,

chemotherapeutic

time

the acid,

oxide

limited

the podophyllinic

example,

diaziquone, anthramycin,

EXAMPLE

, mustard, periods

a

nogalamycin, with , mannomustine,

hydrochloride, streptozocin,

and

therapy to,

methods tamoxifen,

,

sequentially

caimofur, one

, uredepa, and

if

busulfan,

acid, between can

a

or

acid,

be can

more

of

L-

) CM Ο 2011202075 05 May 30 25 20 15 10 5

tetraazabicyclo-[8, methoxyestradiol pleurotin PD98059 (SNP) KF58333; AAG), shock (lS,4S,10S,21R)-7-[(Z)-ethylidene]-4,21-diisopropyl-2-oxa-I2,I3-dithia-5,8,20,23- kinase suitable whose phosphoramidate alpha anti-cancer factor prodrug [0341] an carboplatin. antiangeogenisis with treated similar [0340] alkylator combination oxoplatin. used , erlotonib, (0339]

anti-cancer

an

include

(HIF1 protein and

or inhibitors; expression and

with

therapeutics. for antiangeogenisis

with

PX-478; EGFR

In In (2'-amino-3-inethoxyflavone); prodrug. In

genistein; other ,

other

use agent

a) another another one cyclophosphamide, an

In with platinum 90 another

or agent

in antiangeogenisis

one receptor. NO

version,

(Hsp90)

geldanamycin

to

or inhibitor, 7,

that LY294002; alkylator or this quinoxaline an

6]-tricos-16-ene-3, In

inhibit

version, 2-ME2), version embodiment,

donors; indanone; that activity

antiangeogenisis

In

chemotherapeutic acts, one tenuazonic

version derivatives,

one

a acts, inhibitor inhibitors

EGFR

version

phosphoramidate

a either prodrug the

of

protein version a is

vincristines,

rapamycin;

either phosphoramidate of

these method 1 staurosporin; increased analogs,

,4-dioxides;

and inhibitor

the inhibitors

directly acid,

a

of

and including

phosphoramidate of

such

or directly combination of methods vincristine.

the

6,9,19,

the inhibitor , subsequently is enzyme, the

agent,

and combination

PX-12

as used upon inhibitors

histone or , invention and

combination

suitable

geldanamycin,

indirectly, sodium 104 alkylator protein radicicol

or 22-pentanone but

and

subsequently to including increased

indirectly,

including (1-methyipropyl such alkylator

not treat Other

deacetylase methods

compositions

,

2,2 such

for

butyrate include

kinase- treated

limited

as

alkylator

treatment breast prodrug and ’

,2"-trichlorotriethylamine, to chemotherapeutic coadministration

treatment a

as but HEFla

prodrug glucose inhibit but

to radicicol

of

17-allylamino-geldanamycin (FR901228,

novobiocin,

1 with treated

gefitinib

to cancer. not

inhibit

(NaB);

treatment not (MEK-1) inhibitors

described

prodrug cis

discodeimolide, methods, levels.

described

limited hypoxia-inducible

2-imidazolyl a limited

transporter methods,

is platinum, phosphoramidate

with

derivatives

the administered sodium and

HIFla

using inhibitors is

epidermal panzem depsipeptide); such

to

a

herein with

to a erlotonib.

agents administered phosphoramidate

herein

Cisplatin,

subject a A

carboplatin, nitropurruside

as

or

subject an vastin a inhibitors disulfide);

such

can

[(E)-

VEGF,

and (2-

that include

urethan, such with growth

is

be factor

and

alkylator

as and is can treated

used

heat

as

an with

(17- and

be P

1 13

in

2011202075 05 May 20 30 25 20 15 10 5

"Evaluation Chemotherapy, Selection example, “ cancer variety Agents of agent anti-angiogenic treat mimics [0344] combination Naprosyn), [0343] (Clinoril), (Orudis, (Voltaren), lavendustin VEGF, compositions inhibitors tecogalan, group anti-angiogenic derivatives 67(8):947-954, ; [0342] synergistically

microtubule

cancer. F

selected

drugs. consisting

of

described

Lr and batimastat,

In Oruvail), the of In

In synergistic

for tolmetin 8 thalidomide,

of addition

etodolac aspirin, New

addition, of glycolytic

A,

another / In

reference with Those

purposes described

In

from

any

U assembly, ” Williams another medroxyprogesterone

incorporated agent and

agent,

with

Agents Vivo above,

of iji

an

ketoralac of

captopril,

(Tolectin), and the

U of angiostatin, YC-1, to version,

(Lodine), anti-angiogenic

combinations because

the

a and

Tumor

b Vendetti, inhibitors

skill

the including

version, of group phosphoramidate

acetaminophen herein thrombospondin,

the

X and for foregoing;

to

the another

combination a

ci

in

treat present (Toradol), Clinical

compound

a herein Wilkins,

cartilage Models pyruvic combination

b consisting

the

rofecoxib fenoprofen phosphoramidate include

a

an tii

"Relevance

like but

cancer.

phosphoramidate

art anti-cancer

tf of agent

by methods coumarins;

not

Trial," compound halopyruvates, can JI.

acetate,

for a acid Baltimore,

derived Cox-2 reference, oxaprozin

phosphoramidate (Tylenol). of described

limited of

(Vioxx), readily that Predicting alkylator

TNP-470,

(Nalfon),

a therapies plays

alkylators,

Pharmacological phosphoramidate 105 of

and

inhibits recombinant inhibitors agent,

inhibitor,

Transplantable

barbituric

and to

alkylator

an determine

compositions

and (Daypro), 1975, in ibuprofen

prodrug anti-angiogenic

alkylator

important

indomethacin and Clinical provided including Biochem. including a

or its

phosphoramidate Cisplatin,

and

like

Avastin. otherwise genistein, derivatives.

alkylator

prodrug and

human

as the

nabumetone Simpson (Advil), Activity prodrug

Basis

by

role alkylator described thiobarbituric Pharmacol., bromopyruvate, but

Animal-Tumor

anti-cancer described

Carboplatin, the Other

platelet

(Indocin), is not

in antagonizes endostatin, prodrug agents

of

present

administered naproxen

angiogenesis, is Herren for

limited Cancer (Celebrex),

prodrug useful

administered alkylator herein.

(Relafen),

Anticancer

herein selected factor

drugs

15 and

methods

acid ketoprofen et

and to

interleukin, angiogenesis

Apr Systems (Aleve, the can

al., other

with 4, provides For

a that analogs; prodrug

inhibitors with

cytotoxic diclofenac

from

action

be Taxol, 2001, pyruvate 1985, sulindac

Drugs,"

the with act and anti used

an

to

the

­

of

a

to the an

in

2011202075 05 May 2011 30 25 20 15 10 5

therapies later methods minus alkylator the [0348] a d administered paclitaxel, [0347] and can cancer absence delivered embodiments, phosphoramidate continued indicative to Differences synergy" the [0346] combination methods Proc. another describe [0345]

imini phosphoramidate

the standard

involve, sum for approved)

net shing

initiation Am.

treatment,

a

by

described of anticancer The of

prodrug)

These

longer methods When of While log can at

throughout of , Assoc.

a more the

unwanted dosages described a

in

for phosphoramidate

administration in therapeutic dose

of be lower

the drugs cell single

methods

accordance

of

a period example, tumor

synergy

defined

effectively

wherein phosphoramidate

alkylator

or Cancer drug herein to regimen

therapy

alkylator kill agent.

dose,

employed

are aid at

doxorubicin, Agents

the herein,

side

cell

of of co-administered lower

less can in

is synergy. as

can

course

greater time administering Res. and there

Two

with

effects kill

prodrug not the of with net

of

prodrug, killing be

than

therefore at

doses

in

the

optionally

alkylator a by together determination 26: required for

log

used the optimal drugs

is

phosphoramidate one the of

than

two those

the synergy

of

those 330, cisplatin,

of

other

treatment alkylator cancer

will,

(than methods

embodiment,

to the

the

optimal tumor can drugs ten-fold

allow

improve

each

with standard or for prodrug with an

Agents

other

at for drug

additional be currently

between maximum

cells

anti-cancer least therapeutic

produces

106 cell or longer

of a prodrug said

the a of

described with

dose

or therapy. phosphoramidate

(one phosphoramidate

carboplatin,

which

whether or

( patient

kill in administration. physician dosages.

drugs to

i.e.,

the

alkylator

the

stopping

some of

used),

periods, a log) possess anti-cancer by

phosphoramidate tolerated is

present a the when is other

drug,

benefit

When and

significantly herein. the used

outcomes

are incorporated two

embodiments,

most thus

to optimum

administration

at prodrug growth

used drug than considered therapeutic

including with drugs

invention treat employed

a

doses. in ameliorating active

agent(s)

lower

alkylator

alkylator Such would or without accordance

over another

cancer

act

of drugs.

in better

combination

single herein

The

than

“ synergistically. but

cancer

accordance alkylator

currently

conclusively

provides low be

synergy be in is

a

prodrug with

anti-cancer

prodrug

will not dosed "degree In administered combination tumor the approved phosphoramidate

dose Agent some

by

with some

cell

limited

case typically existing

reference,

if prodrug

” practiced a

using

cell

regimen the

or

therapies as

a method

will of with

in

all

well dose

kill agent, the to

either be

(or

of with the prior be

and

than

as

of

a

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 5 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, N.J 10 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, , antibiotics, aromatase inhibitors, bisphosphonates, cyclo-oxygenase inhibitors, estrogen receptor modulators, folate antagonists, inorganic aresenates, microtubule inhibitors, modifiers, 15 , nucleoside analogs, osteoclast inhibitors, platinum containing compounds, , topoisomerase 1 inhibitors, topoisomerase 2 inhibitors, and tyrosine kinase inhibitors. In accordance with the methods described herein, a phosphoramidate 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 20 addition, a phosphoramidate 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), tositumomab and ,3II Tositumomab (Bexxar),

rituximab (Rituxan).

25 [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), glufosfemide, melphalan, L-PAM (Alkeran), dacarbazine (DTIC-Dome), and temozolamide (Temodar). In accordance with the methods described herein a phosphoramidate alkylator prodrug is co-administered with an 30 alkylator to treat cancer. In one version, the cancer is chronic myelogenous leukemia, multiple myeloma, or anaplastic astrocytoma.

107 [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,

5 ifosfamide, glufosfamide, mechlorethamine, melphalan, chlorambucil, dacarbazine, temozolomide, carmustin'e, 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 al., (supra).

10 [0353] In one embodiment, the present invention provides a method of treating cancer by coadministering a phosphoramidate alkylator prodrug with at least the alkylator Cyclophosphamide, in the treatment of Stages ID and IV malignant lymphomas, multiple myeloma, leukemia, mycosis fungoides, neuroblastoma, ovarian adenocarcinoma, retinoblastoma, and carcinoma of the breast Cyclophosphamide is administered for 15 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 20 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 treatment regimen using at least the alkylator Mechlorethamine. For example, 25 Mechlorethamine is used in the combination chemotherapy regimen MOPP (mechlorethamine, Oncovin (vincristine), procarbazine, and prednisone) in patients with Hodgkin’s disease and administered by intravenous bolus administration is doses 6mg/m2 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 30 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

108 π 2011 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 May

al., supra). 05 [0356] In one embodiment, the present invention provides a method of treating cancer by

5 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 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,

2011202075 10 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 15 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 ethylenimine is Triethylenemelamine or Thiotepa.

[0358] Thiotepa can be used to treat adenocarcinomas of the breast, ovary, and bladder, 20 malignant lymphomas, branchiogenic 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 Monographs on the Evaluation of Carcinogenic Risk of Chemicals to 25 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.

30 [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 . Melphalan

109 ) 2011202075 05 May 20 30 25 20 15 10 5

regimen Doxorubicin is bladder, cancer. of platinum regimen administering sarcoma. can Dacarbazine. [0363] administering to myelomas, [0362] gastrointestinal alkylator regimen administering before regimen combination leukemia [0361] administering example, is [0360] developed treat

used

childhood. treat used

be

chronic

used to

bone

pancreatic

Combination to using head using

using using

coordination In In

treat

is In Γη

In non-Hodgkin and treat

by

malignant

one Carmustine. to one

one one to marrow another

with

lyphocytic

and

a at Cisplatin

Dacarbazine a chronic Salmedix a at

advanced treat a

at at treat

multiple phosphoramidate

phosphoramidate

tumors. embodiment, phosphoramidate phosphoramidate embodiment,

embodiment, embodiment, least least

least least neck,

Cyclophosphamide islet

malignant

ovarian

embodiment,

transplantation.

chemotherapy

a

a astrocytomas, complex myelogenous a

the

cell

’ platinum alone

endometrium,

s myolema Inc. testicular leukemia In

Carmustine lymphoma, alkylator

is another carcinoma.

carcinoma. can

used

the or the the gliomas. the

alkylator alkylator.

with

be

present coordination

alkylator present

alkylator present

present alkylator

alkylator and cancer; and the to

embodiment,

Busulfan. used alkylator. of leukemia. metastatic

treat can cyclophosphamide

chronis

triazene small to primary can

Cisplatin

treat

is to

invention be invention

invention invention

and malignant be

In

produg

produg Cisplatin. produg produg treat

used cell 110

one

lymphocytic administered patients

with

complex Busulfan alkylator

In macroblobulinemia. High tumors

hematological with carcinoma

the another embodiment, to

of

of

provides of of provides

one provides provides treat

nitrosourea doses melanoma,

the

Bleomycin, the

the the Cisplatin with

of

of alkylator. is is

is

invention

Hodgkin invention invention the embodiment, invention

Paclitaxel, Temozolomide.

used

leukemia,

used

of orally. acute of

a

a a a brain,

busulfan

method the method method method

malignancies, the

to

can

to Hodgkin

is myelogenous

Etoposide, lung, ’

treat

In Chlorambucil s

treat triazene

Streptozocin with

with

with melanoma,

with be Bendamustine, disease,

and one Cyclophosphamide,

of of

of of

can used

the

chronic

and advanced

a

a a

a

treating multiple

treating treating embodiment, ’ treating

cancer

cancer s cancer

cancer be nitrosourea

Temozolomide

alkylator

disease, some

to

lymphomas,

and

such used

treat

leukemia and granulocytic

which

is

treatment

treatment cancer treatment Vinblastine treatment

cancer

cancer cancer neoplasms myeloma. ovarian

as,

in used

cancer and

is

for

is the

by to adult by by

by

used

or

of

2011202075 05 May 2011 30 25 20 15 10 5

but lymphocytic [0367] herein cancer. and limited IJS05/008161. [0366] described doses). prior methods three intervals; bronchogenic dose carcinoma, been soft acute naphthacenedione, antibiotic lyxo-hexopyranosyl)oxy]-8-glycoloyl-7,8,9, breast methods Kaposi are [0365] [0364]

are

daunomycin

not tissue

in to successive used

lymphoblastic

a

not

cancer. In to

’ the limited phosphoramidate and Cyclic s

Aromatase Antibiotics

described

described As one dactinomycin, herein weekly Anthracyclines

sarcoma,

isolated limited

successfully and

(Idamycin), range

thyroid

continuing leukemia,

one

version, carcinoma,

to

bone

Anthracycline to,

(Cerubidine, days are

treat

example of

intravenous

to

from doxorubicin

herein,

prostate more

carcinoma, herein

inhibitors

30-75 provided sarcomas, useful anastrozole leukemia,

repeated

cancer.

the other

after to

cultures actinomycin

commonly

useful

alkylator

and

produce cancer the a a

mg/m in

cancer,

phosphoramidate phosphoramidate leukemias, the

DanuoXome). by cytotoxin In the injection compound gastric useful

every breast lymphomas acute (Adriamycin,

in

administration

2 one of (Arimidex)

the

is practice

as

(Valstar), regression

the

prodrug Streptomycespeucetius bladder a

known reference version, a in myeloblastic

four carcinoma. cancer carcinoma, D

practice single

at

the prodrugs and (Cosmegen),

(8S,1OS)-10-[(3-Amino-2,3,6-trideoxy-alpha.-L-

doses weeks. of

practice

of

cancer, is

as

Kaposi and 111

selected 1 and

the intravenous

the In Doxil,

in 0-tetrahydro-6,8,

alkylator both co-administered alkylator

Matteuci doxorubicin,

of of

accordance of

disseminated

methods

letroazole cancer ovarian useful In

Doxorubicin

the

doxorubicin

leukemia, 20

Hodgkin ’ metastatic

of

s accordance Rubex),

from methods bleomycin sarcoma. mg/m

the

prodrug

prodrug

et is in

injection carcinoma, described

methods

acute al.,

the the var.

(Ellence). (Femara). 2 with is

; and Wilm's mitoxantrone

or neoplastic

a carcinoma

PCT practice described group 11 at with is

caesius. with cytotoxic

is is

nonlymphocytic

30

(Blenoxane),

non-Hodgkin the -trihydroxy- such typically

co-administered co-administered

administered

described

mg/m Patent herein

an

methods tumor,

transitional consisting the

In

In doses

antibiotic of

accordance herein accordance conditions methods

Doxorubicin of 2

the anthracycline

Aplication administered include doses (Novantrone),

neuroblastoma,

the (or

methods herein 1

described

daunorubicin,

-methoxy-5, types,

include

ovary, of at

at

cell on leukemia, to

of

lower acute but 2

with

starting

such treat

1-day with each include with the

bladder

No.

has and are

but

an in

as

the of

the

not

a

12-

.) 2011202075 05 May 2011 30 25 20 15 10 5

intramuscular courses such of treatment of drug methylamino]benzoyl]-L-glutamic treat herein breast [0372] are [0371] is administered accordance include cancerous oxygenase include described [0370] cancer. the biphosphonate described include methods [0369] aromatase [0368]

mycosis patients breast

not

group

courses that cancer.

cancer. a

of limited

but but phosphoramidate but

cancer

As has Folate of Estrogen described Cyclo-oxygenase Bisphosphonate

therapy.

herein consisting herein with

condition

fungoides. inhibitor

inhibitor

with are

are are advanced one

repeated injections

been In with

to

not inhibitor chorioadenoma not one not antagonists or

example,

the a a

used

an

the

phosphoramidate receptor phosphoramidate

limited For limited limited herein

version,

to methods to

known

of

estrogen as

stages

treatment Methotrexate

treat treat of leukemias, in

multiple

needed to

doses the alkylator the inhibitors

a

to

to treat to

inhibitors

modulators useful

cancer. cancer. as phosphoramidate

the

of

tamoxifen described treatment celecoxib compound zoledronate

receptor

familial

and

malignant destruens

with of

cancer is cancer. myeloma,

in administered

twice acid,

15

tremetrexate. prodrug

In useful is

In alkylator alkylator rest the

useful to

administered

one one adenomatous modulator

is useful

herein of

(Celebrex). commonly

(Nolvadex)

30 practice

In weekly N-[4-[[(2,4-diamino-6-pteridinyl)methyl

period

and osteosarcoma. (Zometa). lymphoma

gestational

in bone version, version, one

mg

is in 112

the hydatiform

prodrug alkylator in prodrug

co-administered a

the

are

version,

to

of phosphoramidate

intramuscular metastases

the of practice In

to practice prevent

one

administered

known

the

the the

as and

In accordance practice treat polyposis.

In and choriocarcinoma

is is

follows.

accordance or

prodrug cancer methods cancer

accordance the

fulvestrant

co-administered

co-administered

mole. in of more cancer.

the as of

cancer from

the

the

of

methotrexate, the

occurrence

is is injections with

weeks the

is treatment

methods with

For

It daily described

breast

colon methods solid

alkylator In co-administered

is

with is

methods

with (Faslodex).

a

choriocarcinoma,

also one the a

folate

for

interposed

tumors, cancer cancer

and cancer. the

the

methods

version,

described

are

useful with with

a or of

described herein

methods five-day in prodrug

is

antagonist methods described

reoccurrence advanced administered

selected the

an or

a a

or

in In cyclo

a

between include the prostate treatment described

pre-

the with herein

is course, herein

cancer ­

co herein

from cases to

an ­

but

of

in

2011202075 05 May 2011 30 25 20 15 10 5

in prodrug with with sarcoma, oncolytic treatment obtained the phosphoramidate cancer, cancer. Application methods Anti binding 008400US methods (Velban), interferes treat but herein [0374] alkylator trimetrexate) methotrexate trimethoxyphenyl)-amino]methyl]-2,4-quinazolinediamine methods [0373] mg doses

weekly

compound

are

vincristine or, the

Cancer cancer.

of

a

Kaposi

alternatively, is not methods In

anticancer

phosphoramidate from rhabdomyosarcoma,

of intravenous described

agents 30 of

described Microtubule

Inorganic prodrug. not

paclitaxel with

one

and B

limited the

No. acute

mg/m Agents

or

In administered

is administered the 22-oxo-vincaleukoblastine,

version, s the 021305-004520US). present administered

D in another one

PCT/US2005/042095; sarcoma,

described

alkylator

common

leukemia. 2

or

the assembly drugs to . herein

herein,

arsenates

and (Taxol, version,

For

a of doses arsenic

inhibitors

treatment

derivative invention the

25 antifolate Prodrugs

mycosis and

include and

alkylator

prodrug

mg prior periwinkle herein, a cancer of at

Paxene),

It or phosphoramidate at

the

prodrugs trioxide useful neuroblastoma, such

2

has metastatic

are such disassembly

mg/m

of (as to cancer

are

of

fungoides,

Thereof but

drug is also administered

a

doses Hodgkin's treatment is

prodrug used either, in phosphoramidate doses. ovarian provided In

co-administered (Trisenox).

2 are

plant

the

thereof

been

for US that accordance is

herein, (or cancer not

refractory also practice

children

and

Patent In

(Attorney can

(Vinca

of 113 shown is weekly cancer, at

with limited

and

one disease,

in which alkylator

) co-administered commonly lower

discodermolide be twice

of a the

(Navelbine),

Wilm's Applications

In version,

a “ co-administered

of

breast

rosea, with microtubule to and

microtubule

breast reference intramuscular acute

accordance to can

doses).

the

Ref weekly. be alkylator with lymphosarcoma,

vincristine

prodrug 1.4 the

useful methods be

tumor. or

Linn.) known Nos. promyelocytic

a

cancer,

a (commonly mg/m

methods used

phosphoramidate ovary

5-Methyl-6-[[(3,4,5- microtubule useful

Matteucci In docetaxel

or 021 entitled with prodrug

in

inhibitor

is with inhibitor, and Vincristine

accordance

2 as in

its

described

combination origin. non-small (Oncovin), co-administered injections for 305-008500US, with

in the vincristine,

an

derivatives. is

described the

adults. the

useful “ inorganic known reticulum-cell practice

is

a Tubulin ”

(Taxotere),

et

methods

leukemia

inhibitor

phosphoramidate practice is co-administered As such

al.,

herein

is

with any

of

cell

one in vinblastine

alkylator In

administered

as PCT

herein with

doses as is of

the

agent accordance Binding

arsenate lung

Tubulin

the

example, an described a

of

the

to (APL). include with 021305-

, Patent other

alkaloid the

treat

of

a

that

50

to

i 2011202075 05 May 20 30 ( 25 20

15 10 5

lymphocytic administered accordance floxuridine thioguanine, but recurrent to [0377] described not BiCNU, uptake organic [0376] alkylator the deoxyglucose increase incorporated cell the compound cancer. but alkylator treat limited microtubule simultaneously [0375]

treat

are limited therapeutic

compound rather, to

colorectal

not take of

cancer.

to

nitrite

Gliadel In Nucleoside 2-deoxyglucose

Nitrosoureas glioblastoma

prodrug prodrug

Modifiers herein (Leustatin, glucose.

Leucovorin

limited with

(FUDR), one administration to that up

leukemia 6-TG

with inhibitor. herein

procarbazine

and or 2-deoxyglucose N-hydroxyurea.

methods version,

In increases cancer.

with the Wafer), a

a

a

is

is one phosphoramidate to (Thioguanine), a

spermineNONOate,

nucleoside

methods

by useful co-administered phosphoramidate

co-administered fludarabine analogs mercaptopurine, or 2-CdA),

(CLL),

useful version, (Wellcovorin), multifoime.

reference),

within the In

provided

and uptake the

of

in accordance

(Matulane), cancer

useful

described

a estramustine ability in the hairy

gemcitabine

phosphoramidate

analog a the (see N-hydroxyurea the

or few (Fludara), practice

herein.

hydroxyurea

and cancer is

to practice cell in

the of alkylator

with days colon with

treat 6-MP which to alkylator the

with administration a

herein reference to leukemia, ,

cell

treat

of practice

is nitric In treat to a (Emcyt).

leukemia (Gemzar),

azacytidine cancer.

the

modifier of

the prostate (Purinethol), 114 is a to another

prodrug cancer.

a week

the

used

cancer, has (Hydrea), take

methods prodrug phosphoramidate methods oxide alkylator

Smith

adenocarcinoma methods

of

CCNU

been

In up

with

after

In together

cancer such

the

and

and of In or is

one

as accordance glucose, (Vidaza),

et

as

reported described N-hydroxyurea co-administered

methods one a

described

other

another initiation

cytarabine fluorouracil, the prodrug

version,

(CeeBU), al., nitric version,

described

described or

version, latter with

1999, glioblastoma, drugs

including oxide

pentostatin

to anti-cancer alkylator

described

is

administration

herein a with herein of of compounds the

enhance phosphoramidate

Cancer (Cytosar-U, carmustine administered herein

such

herein the treatment the

precursor,

5-FU modifier (Xeloda).

the at

with

pancreas, a cancer

include but

as

levels prodrug

phosphoramidate

methods is

herein Letters

include (Adrucil),

the including (Nipent), 5-fluorouracil agent

not one a

stimulate

with

is nitrosourea (BCNU, ability

is

DepoCyt), such reported

of

limited but

In

a

version

B-cell include

to

is 141:

2- but a

treat are

co

as

of

­ are

85,

the an not to

to a of

to

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 5 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 Sth, 7th, 9th, or 11th days. In poor risk patients or those who are not in an 10 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 phosphoramidate alkylator prodrug is co­ 15 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 20 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 phosphoramidate 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 25 but are not limited to pamidronate (Aredia). In accordance with the methods described herein a phosphoramidate alkylator prodrug is co-administered with an osteoclast inhibitor to treat cancer, ha one version, the cancer is osteolytic bone metastases of breast cancer, and one or more additional anti-cancer agents are also co-administered with a phosphoramidate alkylator prodrug.

30 [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 phosphoramidate alkylator prodrug is co-administered with a platinum compound to treat cancer. In one version, the cancer is metastatic testicular cancer,

115 2011202075 05 May 2011 30 25 20 15 10 5

treat phosphoramidate include etoposide administration administration version, prodrugs the phosphoramidate prodrug. useful [0353] cancer. accordance include from treatment metastatic administered limited administered can administered described of Cisplatin, administered accordance example, [0352] [0351]

doses

reference be

cancer. the

in

co-administered

but to

but As

the

of

Topoisomerase group the Topoisomerase Retinoids thereof

the As phosphate herein, of

, when ovarian

are noted 50-70 with with are cancer

transitional

palliative one In with Matteucci with with compound at

of of not

not

consisting

one these

used the

the

useful

example, a alkylator a a above, alkylator mg/m

a a a

cancer,

limited phosphoramidate is limited phosphoramidate

topoisomerase useful

ATRA

version,

topoisomerase phosphoramidate methods methods (Etopophos).

metastatic for doses

treatment

with in 2

et

cis-Diaminedichloroplatinum cell however, 2 advanced once 1

of

in the al.,

ovarian

to

inhibitors to prodrug inhibitors Platinol, prodrug. (Vesanoid), (or

to

the bladder the the APL,

etoposide,

described described practice

PCT

every

treat at

carcinoma platinum cancer practice of

lower

1 carcinoma,

in Kaposi In bladder

Patent 1 metastatic is

Blenoxane,

inhibitor cancer.

three

As cancer useful alkylator inhibitor useful alkylator one

accordance co-administered

alkylator of

is

herein

herein (Hycamtin) doses). VP-16

another of

the

version

a compound ’ Application to

s

cancer,

of

cancer the

in which in In

sarcoma, four methods

116 but

the

testicular and to the prodrug prodrug the a one a

methods (Vepesid),

prodrug.

and phosphoramidate

phosphoramidate One example,

treat

is

with

weeks. of ovary,

practice

practice

transitional

is selected is

version, (Panretin),

not

and

Velbam the

not or and administered

cancer. of and with No. (II),

the

either is

administered

more

methods and

described

colon,

the

amenable a co-administered

In

T-cell methods Platinol

teniposide,

of of

PCT/US2005/041959. phosphoramidate

commonly from the

a

ovarian present accordance

can

the topoisomerase precedes the additional

Topoisomerase cell and

cancer or

be the lymphoma. methods methods described

rectum, alkylator alkylator

bladder herein and in

(Camptostar). to

described

co-administered

invention

tumors,

group concurrently

intravenous surgery

VM-26

known is Adriamycin or

anti-cancer with a

include

follows,

described with described or cancer cancer.

consisting

herein, prodrug prodrug

2

and

alkylator

small

the herein

or

as (Vumon), inhibitors (Targretin). inhibitor are

cisplatin

radiotherapy.

cisplatin, for

methods

but

injections selected

In provided

therewith. or

In

As cell

can

agents

is is the herein herein

with a

both, one

are

of

one co co

lung be

to

and and

­ not ­

a

is co

In

in

­

Γ 2011 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, May

administration of a phosphoramidate alkylator prodrug either precedes or follows, or both,

05 administration of a topoisomerase 2 inhibitor but is not administered concurrently therewith.

5 [0354] Tyrosine kinase inhibitors useful in the practice of the methods described herein include but are not limited to imatinib (Gleevec). In accordance with the methods described herein a phosphoramidate alkylator prodrug is co-administered with a tyrosine kinase inhibitor to treat cancer. In one version, the cancer is CML or a metastatic or unresectable malignant gastrointestinal stromal tumor.

2011202075 10 [0355] Lonidamine analogs useful in the practice of the present invention are provided in the Matteucd et al. U.S. Pat. Appl. Nos. 11/346632; 60/764,427; 60/764,438; and applications entitled “Heterocyclic Lonidamine Analogs” (Attorney Docket No. 021305-007220US; 021305-007900US) and PCT Publication Nos. WO 2006/015191, WO 2006/015263 and WO 2006/01007 A2.. 15 [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-methyl-6-[[(3,4,5-trimethoxyphenyl)amino]-methyl]-2,4- quinazolinediamine or a pharmaceutically acceptable salt thereof, (8S,10S)-10-(3-amino- 20 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- 1,7-dihydro-6H-purine-6-thione or a pharmaceutically acceptable salt thereof; 22-oxo- vincaleukoblastine or a pharmaceutically acceptable salt thereof; 2-bis[(2- 25 chloroethyl)amino]tetrahydro-2H-l ,3,2-oxazaphosphorine, 2-oxide, or a pharmaceutically acceptable salt thereof; N-[4-[[(2,4-diamino-6-pteridinyl)methyl]-methylamino]benzoyi]-L- glutamic acid, or a pharmaceutically acceptable salt thereof; or cisdiamminedichloro­ platinum (II).

30 IV. EXAMPLES

117 [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

5 [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-nitroimidazole-5-methanol was purchased from Syngene, India. Non-commercially 10 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, POC13 and PCI3, and their mono and dichloro derivatives were performed employing anhydrous solvents 15 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 20 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 25 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 prodrtfg compounds were prepared in DMSO and stored in a freezer.

30 Example 1 Synthesis of Compound 23

118 2011202075 05 May 2011 15 10 5

by was spectroscopy ml) hour, [0362] temperature chloroethylamine (TEA, 78°C concentrated. aqueous [0361]

LC/MS

was distilled

POCI3 the

0.22

refluxed layer

A To reaction

and

suspension ml,

HCI

was was a out

to

was solution Compound NMR

1.54 be

under

raised added (135°C)

+ hydrochloride mixture

pure. extracted

POCI3 mmol). + spectroscopy

vacuum of of to in POCI

N-Methyl-2-chloroethylammonium over 5-nitrofurfuryl

room 23 one was

with

Temperature 3 was

portion

night.

quenched

as temperature

was

DCM separated Synthesis

light Cl-P-N to TEA THF

be added

After followed O Cl

11

I

yellow

and pure.

5i

alcohol with (-jci(+)H Example

was /

removing followed by

of the (rt), 119

water

oil Compound LiN(TMS)2 flash increased

TEA, by combined DME

Cl the (200 3

N^ and

a 2

and THF column dropwise reaction

by HN^z^CI

excess mg, analyzed — / DIEA,

TEA

the to

CI

organic 5 1.4

-30°C

organic

chromatography

POCI3

was THF

(1 chloride mmol) addition

by

ml, HCI

continued

in solution

*H

under layer 7 one

Π in

mmol). . and

(10 of

THF

hour

triethylamine was

vacuum

31 gm) was

P for O

II

(10 5

and

23

and O-P-L^CI NMR separated.

After

in dried one

/ Cl s O

ml) NH

POC1 analyzed then

product

more

H the

and at Cl

2-

3

-

(40

The

5i

2011202075 05 May 2011 15 10 5

preparation by mmol) methanol was one was dimethoxyethane organic chloride was [0363] [0365] [0364]

flash

hour, wanned dried added

was

layer

chromatography (0.62

To To

Compounds

in the over

at

slowly

of

DCM solution a

up -78°C. reaction was

solution compound_5. gm,

MgSO

to

(DME)

dried

yielded -20°C, 4.75 added

After of

4 of mixture

of

and

over mmol) 5i

8

yielding lithium and TV-methyl diluted

5 (1 and 5.

concentrated.

min, MgSO

gm, the

was 16 in

with reaction bis(trimethylsilyl)amide THF 4.75

were 5ii compound

diluted 4 2-nitroimidazole-5-methanol

and (2.9

ethyl mmol)

at synthesized

concentrated

-78°C mmol, mixture

Purification with

acetate

120

5ii and LiN(TMS) LiN(TMS)

DME ethyl diisopropylethylamine

DME

as 770

JV-Methyl-2-chloroethylammonium was

oil. employing and

mg) acetate

by to warmed 2

2 washed

yield

flash was (3.2

and

added

mmol, a the

chromatography

to with

residue

washed (0.5

rt. procedure

brine. and

After 3.2 g,

(DIEA,

which 3.2

the with ml,

stirring The mmol)

reaction used 1

brine.

was M 1.65

with organic

in for

in at separated

ml,

THF)

6-12% The

mixture rt the

for 9.5 layer

i I

2011202075 05 May 2011 15 10 5

phosphorylation white refluxed was Additional remove filtered to g, in employed dried dibromopropane wise [0367] [0366] H

200 rt, DMF 2

N^ coevaporated

poured at

with solid

mmol) HBr, 120

rt through

the

(38 (4

A To

and

in

Na °C

aqueous product h). 48% solution aqueous

0H

mL) into a the

2

the

was solution SO

The

a

next provided

(10

water a reaction 4 celite

+ with

added

and solution

35b

reaction portion of portion

g, reaction.

compound_35a of

ethanol concentrated (250 50

pad. was

to

ethanolamine

below. mixture mmol).

the (5 of

(about mixture mL), filtered The

ml) p-toluenesulfonyl

thrice, reaction Synthesis

filtrate

and was was

After 20

and to was

and ml), extracted

(5 removed heated yield

(6.03 mixture,

washed

heating was TsCI, g) Example cooled

following

and

of in

compound_35a 121 DMF concentrated mL,

Compound up aqueous K

the

2

with

by

to chloride CO with for followed to

100 3 reaction

rt,

distillation

3

120°C two

addition ethyl

acetone diluted

mmol) HBr

more

(19 35

by to (bath acetate.

mixture

(48%, as 130

POCI

dryness g, of with dropwise

and twice

hours, and yellow

100 a temperature). °C

large 3

K2CO3

water 50 the

The

was and mmol)

the to

ml)

oil reaction

addition

volume

organic yield

refluxed employed (20 reaction

35a

which (13.8 was

35 was

mL),

a distilled

K2CO3 mixture residue g,

of

added layer of was

for was

100

and acetone

in 1,3-

40

the was

cooled mmol) (27.6 drop

to

which

was h.

a

[0368] A suspension of compound_35b (1 g) in POC13 (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 5 invention employing the same procedure as provided in Example 2 employing for column chromatographic separation silica gel andl0-80% acetone/toluene as the eluent.

Example 4 Synthesis of Compound 7 10 [0369] Compound 7 was prepared by employing N-cyclopropyl-2-chloroethylammonium chloride as provided below

i) /THF/NaOH Cl

>-nh2

ii) HCI/dioxanc iii) 7j SOC12

Cl POCl3/reflux

>-NH2(+)CI(-)

7i Cl !__ / Ϊ>-ΝΗ2(+)α(-) Cl

D^n,o p-ci DIEA/THF Cl' 7ii Cl

A P—N C|- 1 LiN(TMS)2 . ( 7111 Cl

[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 15 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

122 2011202075 05 May 2011 30 25 20 15 10 5

flash (110 was layers into After added. solution dissolved 78°C g, with was minutes, (47% for yield) 7.5 cooled solution vacuo dried through 95% colorless solution [0374] [0373] [0372] [0371]

2.11

1.5 hours.

separated 25 refluxed

chromatography mg, dry

yield)

(MgSO 15 and of were

which

at

to hours ml mmol) After

a of

theoretical) of of minutes

ether 53-56°C and

N-methyl-2-nitroimidazole-5-methanol

To 7i 7ii 51% liquid

0°C DIEA in

short water

The lithium

HC1 sodium which dried

(3.00g,

dry (0.50

a

was 4 extracted and (6

15

)

by and solution yield) (100 were

reaction and path

which h), in THF

minutes, (0.545

and

the flash

over concentrated analyzed

(1 was g,

SOC1 dioxane

bis(trimethylsilyl)amide

hydroxide cooled,

ml), 19.2 evaporated

combined

of distillation mm 2.11 reaction

as extracted

over (2

was MgSO chromatography

analyzed

4

of pale g,

a

2 mixture

ml) mmol) filtered,

times

Hg) yellow

(6.50g, mmol) the a 4.22

silica

by (4.0M, analyzed and

solution

yellow under

4

intermediate mixture

to

(7g)

*H in and 3 with to mmol)

to

concentrated

was

apparatus

by yield using

was times oil

and dry and 54.9 NMR give give

in 18.3 argon.

concentrated

oil *H

which ethyl of added by

THF residual concentrated water N-cyclopropyl-2-chloroethylamine

an was

mmol) added an 0-50% NMR

an with which 7iii

ml, in LC/MS to

intermediate

orange

orange acetate

to

0-10% be The under slowly alcohol to was (172

73.2 (50 in ethyl

yield

to slowly to 123

pure. POC1 was was volatiles

of THF

reaction

analyzed ml). be yield

and mg,

mmol) to

hexane

argon. oily

oily (75 (76.8

acetate methanol

warmed

added analyzed

7ii pure. (3.7 and give 3

(1.6M, by 1H

0.538 (15 The

ml). a residue.

residue. as alcohol

removed the residue.

mg,

g,

syringe, mixture NMR

was a

in The a ml)

by

by

(30

reaction

yellow 36.6

clear,

resulting The

to mmol)

ethyl 0.306

0.489 in to

syringe.

LC-MS

added.

and reaction ml). rt,

be to (5.94 DCM

The mmol) The combined

The

stirred warmed was pale

in

be

acetate refluxed

oily 7iii

ml, mmol) mixture in

The

vacuo

residue

residue

pure.

oil g,

The

and residue

to cooled 2 The

yellow by

0.489 residue. mixture

in 42%

ml combined for yield distilled

to MS.

dry reaction

to 'H

hydrochloride was

to reaction organic under THF

was

2 give was

was

rt mmol) yield)

NMR yield to

was hours, THF oil compound_7

partially slowly,

was

-78°C

The

stirred

was

separated distilled 315 (3.6g, in nitrogen

triturated

mixture

organic 7i layer

(30

as mixture to

vacuo cooled

was

poured residue

added. mg (5.42g,

be a

and stirred

ml)

for 79% clear,

was

pure. (0.33

in

for

a by was 10 to a

-

Example 5 Synthesis of Compounds 6 and 15 .Cl Tv* TEA OOH θ2Ν·^Ν Cl PC13 + I __ . I DCM

6

[0375] To a suspension of bis(2-chloroethyl)ammonium chloride (1.43 g, 8.01 mmol) in 5 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 //-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 10 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 MgSO4 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 15 Compound 6 above.

TEA OOH HCI DCM

15

Example 6 Synthesis of Compounds 23, 26 and 36

Cl

I 20 J

124 [0377] To a solution of 7V-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 5 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.

Example 7 Synthesis of Compound 1

10

[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 (IM 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 15 (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, wanned 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 20 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 25 [0380] To a solution of 2-bromoethylammmonium bromide (19.4 g) in DCM (90 mL) at - 10°C was added a solution of POClj (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

125 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:

O II H HO-P-N^ Br

5 [0381] Isophosphoramide mustard

O II H HO-P-N I ^Cl

can be synthesized employing the method provided in Example 8, substituting 2- bromoethylammmonium bromide with 2-chloroethylammmonium chloride. Synthesis of Isophosphoramide mustard has been described (see for example Wiessler et al., supra).

10 [0382] The phosphoramidate alkylator toxin:

O II H HO-P-N^

was transformed into compounds 24 and 25, employing the method provided in Example 6 and the appropriate Trigger-OH.

Example 9

15 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 20 employed was HOP(=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

126 2011202075 05 May 2011 10 5

nitrothiophene-2-methanol; methanol, HOP(=O)(NHCH(CHMe HOP(=O)(NHCHMeCH2Cl) and analog analog 93 [0384] 105,

,

the 49

included

employed employed The the ifosfamide

l-N-methyl-5-nitroimidazole-2-methanol, ifosfamide

various

the

was is following

mustard

Trigger-OH the

the mustard

S

2 enantiomer S )CH2Cl)

analog

enantiomer Trigger-OH 2 ;

in analog

compounds

compounds

2 employed ;

and

of

employed

of

HOP(=O)(NHCHMeCH

compounds: in

HOP(^)(NHCH(CHMe2)CH

compounds

employed 127

is 38,41,51,

the

was

R

5-nitrofuran-2-methanol, the enantiomer l-N-methyl-2-nitroimidazole-5-

in 46

and R

the

-

enantiomer

48,

57 synthesis

the the

2 of Cl)2;

ifosfamide ifosfamide

in of of

2

compounds Compounds Cl)2.

5-

mustard mustard

43

37

-

-

45

2011202075 05 May 20 10 [0385]

The

following

compounds

were

according· 128

to

the

method

described

in

Example

6. 2011202075 05 May 2011 129 2011202075 05 May 20 130 68 2011202075 05 May 20 15

synthesis Examples

of

10

phosphoramidate

26 88

describes

the

alkylator

synthesis Synthesis 89

prodrugs Example

of

of

various 131

Compound

of

10

the

Trigger-OH

invention.

52i

compounds 90

employed

in

the

Γ 2011

PdCl2(dppf)

I KOAc, DMF May OH 52ii 52iii 05 [0386] A solution of compound 52ii (100 mg, 0.48 mmol), 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 5 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.

2011202075 [0387] Compounds 55i, 63i, 59i, 65i, and 68i were prepared in a similar manner as described

PdCl2(dppf)

KOAc, DMF 55i

PdCl2(dppf)

KOAc, DMF 63ii 52iii 63i

PdCl2(dppf)

KOAc, DMF ϊ 1°

PdCl2(dppf)

KOAc, DMF 52iii

Example 11

DIEA

132 [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

5 yield compound 68i.

[0389] Compound 69i was made similarly as depicted in the scheme below.

69i

Example 12

K2co3

Acetone

10 [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 15 yield compound 70i. [0391] Compound 51 i was made similarly as depicted in the scheme below.

K7CO,

HO

51iii

Example 13 20 (HAP triggers - check #s)

PdCl2(dppf)

KOAc,DMF

[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 ϊζ

133 2011202075 05 May 2011 20 Ι 10

5 5

phenyldichlorophosphate employing compound syringe under added DCM. rt was TEA 100%) was [0393] eluent followed to atmosphere

for yield PhO-P-CI

separated diluted

(0.75

1 hydrogen

platinum(IV)oxide

EA\Hex to

The h,

filter, o Cl a I

To

by yield poured residue

67iii. a ml,

combined

with and Mitsunobu

a Cui

the . o-s by

suspension

+

5.38 compound

(0-70%)

for

the

Compound

(8.5 O O EA,

filtrate into column II II which -s-cr'X/NKjHCl

— 0.5

mmol) reaction O 2 II

+

mg,

brine, washed organic

h.

O

type

to was concentr

(0.2 2 67i

chromatography (20 of N 0.043 67ii. The

yield

and

Synthesis

the 67iii

67i mixture reaction

separated

ml, N-y mg),

with

layers

reaction

stirring.

organic To

mmol) (472 compound

ated was 1.34 TT

the brine, a „,

were nH was solution

mg,

as

reacted under of mmol)

reaction

by

-20°C

TEA, mixture

and

layer described Example The

Compoundsl06 Example

THF, the

heated

2.69 PPh column on dried

TEA

58i. vacuum

134 organic DCM

reaction

silica separated, at tort with 3 of

0°Ctort , mmol) mixture

DIAD was -20°C,

with compound

at (0.27

15 chromatography

for 14

l-N-methyl2-nitroimidazole-5-methanol 60 gel

diluted

and layer

MgSO»

mixture the °C in

ml, employingas followed degassed,

and

DCM coevaperated and for synthesis

separated,

1.92

67ii

with

the

two and

107 was

[

x\^O (20

mmol),

aqueous (42

by

MeOH, hours.

concentrated. and

67

wanned

ml) on of the

mg)

eluent dried,

vigorously

silica Compound —

with

was

dropwise at o O

S II II

The in layer filtered —

rt,

up EA/hexane EtOH

and added toluene gel

under reaction

to pto extracted

EtOH employing concentrated The

rt, through addition

stirred (5 36. 2,

argon

stirred

to h

ml) residue

2 mixture

o O S·

II II yield (ΙΟ

with was

a of

­

at as

2011202075 05 May 20 15 10 5

was [0395] ml, to separated twice, with addition (phenylsulfonyl)ethylamine added [0394]

100%)

1.54 synthesized

water

POCI3 the

of mmol).

Compounds

To

to by combined

and TEA

yield

a

column (0.13

solution 108

POC the

using (1

The product

organic *3 ml, ml,

organic

chromatography

+ reaction 108-112, a

of 1.4 7

similar Ο

mmol).

2 5-nitro 106.

Ν-ζΚ/ mmol) layer

hydrochloride layers

temperature

shown method. Compound

furfuryl separated.

The at

were

-78°C, ΟΗ

on below reaction

dried, alcohol silica TEA,

(832 was

107: The -78 followed 135 :

was THE gel concentrated

warmed °c'

mg,

aqueous (200

employing

warmed

109 3.5

by mg,

up mmol)

-10°C

the layer

1 to

.4 to dropwise to

-10°C as

mmol)

tort rt,

was yield added

eluent

stirred

extracted

in a

in

to addition residue

1

THF acetone\toluene for it, h,

2- followed

1

with (10

h, which

of

quenched

ml)

TEA DCM

by

was was

(0.216

the (30

110 111

112 5 were synthesized employing the procedure described for the synthesis of compound 35 in Ts Ts Example 3 and substituting HO OH with

Example 16 Synthesis of Compounds 113-117

1, LiN(TMS)2 9 ^Cl THF, -78 °C o2n OH + Cl—Ρ-Νχ Cl ^CI 2, NH3, -20 °C 10 113ii li [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 warmed up to -20°C and NH3 bubbled through the 15 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

O2N OH 20 Compound 13 and substituting with the appropriate Trigger-OH as starting material.

136 Γ 2011

O °—ρ-ν

May νη2

117 05

Example 17 5 Synthesis of octadeutereated ifosfamide and Compound 64 (octadeuterated-Compound 25)

165oC 2011202075

DIAD, PPh3 „ O/C°2 d2cs Br

64iii

[0398] 48% HBr (60 mL) was added dropwise to diethanolamine at 0°C. The reaction mixture 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% 10 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 11.3 g of <74-2- bromoethamine hydrobromide (compound 64i). Compound 64i (19.5 mmol, 1.0 eq.) was added dropwise to a suspension of J4-2-bromoethamine hydrobromide (40.0 mmol, 2.05 eq.) 15 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 Na2SO4> and concentrated under reduced pressure to yield a residue which was separated by column chromatography on silica gel employing as eluent 20 Hexane/EA (100:70(v/v)) to yield 7.0 g of compound 64ii. PtCh (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 it, and diluted with MeOH until the

137 138

white solid in the reaction mixture dissolved. The diluted reaction mixture was filtered, the 2012

filtrate concentrated under reduced pressure to yield a residue which 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 Dec

eq), l-N-methyl-2-nitroimidazolemethanol (1.01 g, 1.1 eq.), and PPh3 (2.39 g, 1.5 eq.) in THF

06 (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.

Example 18 2011202075 Synthesis of Compound 2i

2ii 2iii [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 (0.7 mL), followed by the addition of compound 2iii (100 mg, 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).

Example 19 Synthesis of Compound 94i

Bn Bn OAc hno3 rX/0Ac o2n^ NaBH4 < Ac2O/AcOH OAc OAc MeOH 94iii 94ii [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 (12mL) 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

6554197 1 RTK 139

employing as eluent EA/Hexane (0-100%) to yield compound 94ii. Compound 94ii (600 mg, 2012

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 (lOOmg) was added once every Dec

hour thrice, the reaction mixture was stirred for 3.5 h, poured into water, extracted with EA, the

06 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 a

yellow solid.

Example 20 Synthesis of Compound 96i

2011202075 NC\

ac NaBH4 FT,° ------O2N^sJryZ OAc DMF OAc MeOH OH 96ii 96iii 96i [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 Example 19 and using THF instead of MeOH as solvent in the reaction.

Example 21 Synthesis of Compound 99i

NaBH4 OAc PdCl2(PPh3)2 ------1 Cul/TEA MeOH 99ii [0402] A mixture of 99ii (500 mg), PdC12(PPh3)2 (208 mg), and Cui (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

65S4I97 I RTK 2011202075 05 May 2011 25 20 15 10 5

yield combined K2CO3 more solution solution (88 oil residue h. which residue with and flask. 585.7 silica [0403] compound

After to

g, stirred

the 48

than

which

gel mmol,

1.07 was The

to washed which

g concentrated which flask,

the

Ethyl

column

a Synthesis ethyl of Synthesis 45°C.

dissolved reaction mol). at 99i pH

was reaction

1 grounded rt

-N-methyl-2-amino

was

NaH (286

was of formate for acetate with

added

The

The chromatography 8-9

triturated

about

cooled, mixture

of (60%

mg).

of

2

in

mixture

concentrated to and

reaction

x

l-N-methyl-2-amino

layers into l-N-methyl-2-amino

10% ethanol (500

1/5 25

14 extracted oil

its mL powder

its was

h. aqueous

twice mL)

suspension, cooled were

pH mixture

original

(400 Volatiles of

cooled

was

adjusted imidazole-5-carboxylic ethanol. employing with

reaction

dried

with prior

down, mL) HO

added

was volume hexane in

Example

EA Example Ac, over

were to 54 and

an

to

the The mixture reaction) stirred

imidazole-5-carboxylic

imidazole-5-carboxylic g, 140 to (5

H2NCN

ice-water

as 1 cone MgSC>4,

removed

white 1.35 sarcosin (500 x using

using

filtrate eluent

200

23

22

at HC1

was mol) mL)

90-1 precipitate contained a concentrated mL (45

filtered, bath, EA/Hexane rotary

was

using methyl (50 carefully

to added

00°C followed g, acid

mL) evaporated

yield 1.07 stirred,

evaporator a

ethyl

and for in ester rotary

slowly

was

and

mol),

a neutralized a

HC1

1.5 by

sticky

volatiles (0-100%)

acid 1-L acid

a ester. hydrochloride filtered stirred

gas evaporator 3 h

during and to

and

round-bottomed x to

ethyl ethyl at

yield light outlet 50

yield

a sodium the

at

removed

off

mL). temperature to

by

ester a

ester 1

brown resulting

a

10°C period

yield

and connected

a addition

thick to

clear

The

(82 acetate

yield the

for

to paste

brown of

g,

1.5

2 of not a

h,

CHO Ο

ONa

1.HC1, EtOH

2. NCNH2> AcONa

[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 5 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 MgSO47 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 10 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 15 the filtrate concentrated to yield a thick brown oil. The oil was added to 10% HO Ac 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 20 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-methyl-2-nitroimidazole-5-carboxylis acid ethyl ester

141 [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 5 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 eluent EA/hexane (30%) to yield 1-N- methyl-2-nitroimidazole-5-carboxylic acid ethyl ester (“nitro ester”) as a light brown solid 10 (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 15 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.

Example 25

Synthesis of l-N-methyl-2-nitroimidazole-5-carboxylis acid

/N 20 CO2Et CO2H

[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 25 MgSO4 and concentrated to yield l-N-methyl-2-nitroimidazole-5-carboxylis acid (“nitro acid”) as a light brown solid (32.2 g, 95%).

142 2011202075 05 May 2011 20 15 10 5

mmol) mmol) DME, gel reaction nitroimidazole-5-methanol washed stirred dry (67 acetate. less dropwise [0408] [0409]

(0

mg, ice-acetonitrile than

-100%

LiN(TMS)2 for was in To

with 0.32 mixture

A

0°C. addition anhydrous

1 a

mixture added

h

suspension THF. mmol)

acetone\toluene) followed The

Synthesis was

drop

of

was reaction bath

The

of

was THF

water concentrated

the

wise added by

of 119i combined (temperature

added

(360

nitro

the

of l-N-methyl-2-nitroimidazole-5-methanol as during mixture

to

1

an at

addition

-N-methyl-2-nitroimidazole-5-carboxylis

to mL) this and acid

α

Synthesis -78°C orange

yield

a THF

and

cooled LiNflMSh

the period was was (30.82 Examples

< DME

with

the

of

Compound reaction -20

portions

solid stirred Example warmed Example

sodium

of

reaction residue

of

g, °C). vigorous

143

Compound (25 1 180.23

28A-28V

while

h

mixture were Isobutyl

up

O while

borohydride g)

26 - 27 was °2N

2 mixture 119.

N

which to

stirring. mmol)

the evaporated ζ

x separated 0°C. maintaining ,υ

was

reaction ,N chloroformate 119 119

was

during ch The

and warmed

After (36 2

recrystallized oh

solid by triethylamine

to

mixture

g, a

chromatography yield a period 10 no (50

947

to temperature was

2 min,

rt. (37.8 acid mg,

a

mmol)

l-N-methyl-2-

was filtered

of After

compound

0.32

from

mL,

10 (140 cooled

and

min

1

mmol)

off

around 288 ethyl h,

mL,

on

and the and in

11

silica

285 a in

or 9i

I 2011 [0410] Compounds 134 to 155 were synthesized by employing the corresponding substituted phosphormamidate and hydroxy substituted Trigger (Trigger-OH), according to May

the procedures described in Examples 1-27 above. 05 Example 29A

5 [0411] The solubility of the following compounds is as listed below :

Compound Solubility (in saline at room temperature)

10 10 mg/mL

25 15 mg/mL 2011202075 73 10 mg/mL

155 <1 mg/mL

Example 29B

Antiproliferation Assay

[0412] To determine the effect of phosphoramidate alkylator prodrugs on cell proliferation, 10 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 15 tested with 20,000 cells/well/500pL medium: NCI-H460 cells (ATCC HTB-177, RPMI medium (Gibco Products, Invitrogen Corporation, Carlsbad, CA)), HT29 cells (ATCC HTB- 38, RPMImedium (Gibco)), MES-SA cells (ATCC CRL-1976, McCoy’s 5amedium (ATCC)), MES-SA/Dx5 cells ((ATCC CRD-1977), McCoy’s 5a medium (ATCC)), ACHN cells (ATCC CRL-1611, Minimum essential medium, Eagle (ATCC)), PC3 cells (ATCC 20 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 (200pL volume) in the treatment groups at concentrations varying from 100, 30,10, 3, 1, 0.3, 0.1, 0.03, to 0.01 μΜ. All test

144 ο CM £ compounds were serially diluted in complete medium with final DMSO concentrations less s 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 o incubated for 2 hours in standard tissue-culture incubators. Following the 2 hour treatment 5 with a test compound, the test compound was removed from each well, cells were washed with 500pL medium, and incubated for 3 days in 500pL 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 IC50 herein)) of test compounds was calculated and tabulated in Table X below.

2011202075 10 Table X: IC50 values (uM)

Compound H460 HT29 MES-SA MES- ACHN PC3 Anoxi a/Air Anoxia/Air Anoxia/ SA/Dx5 Anoxia/ Anoxia/Air Air Anoxia/Air Air P2 44/>100 1 0.4/72 50/>100 1 / >100 23 0.04/5 7.5/- 23 0.1/14 154 0.9/2 139 16/100 140 5/-65 2 8/>100 5 0.05 / 6 10/>100 22 0.7/16 3 >20/>100 142 >40/>100 4 40/>100 143 4.5/3.5 6 0.7/>100 22/>100 5/>100 144 7/>100 145 >100/ >100. 147 >100/ >100 7 0.14/25 7/>100 0.59/83 11 5.2/>100 12 1.7/>100 9 >10/>100 - 8 0.013/0.6 36 0.88/ 55/>100 5/>100 7.5/>100 >100 149 50/>100 15 0.08 /1 16 1.6/>100

145 2011202075 05 May 20 110 109 108 107 106 119 118 150 38 90 89 88 87 37 86 27 24 25 82 85 84 81 79 78 77 76 75 74 35 33 32 28 31 26 83 80 35 34 10 14 18 17 0.03 0.03 100/>100 0.3/>100 0.3/>100 0.003 8.5/>100 0.075/50 22/>100 0.06/2.8 0.09/3.5 0.01/3.4 0.33 83/>100 0.05 0.01/1.8 l.l/>100 0.7/100 0.05 0.15/86 0.15/20 9/>100 25/100 3.5/3.5 50/100 0.3 0.3 1.6/5.5 0.01/4 0.1 >100/ >100/ >100/ >100/ 3.4/9 1.4/3 0.33/ 0.36/ 9/26 >100 >100 8/46 >100 >100 >100 >100 -/21 1/9

/

1 / /

/ / /

/

0.53 0.02

21 / 13 35 3.7 0.3

55 40

1.8/>100 16/>100 0.8

/

57 0.13/10 1/100 0.1/2 >100 0.9/ 146

0.3/>100 0.1/0.8 0.2

/

62 0.6/>100 Ο co 39 0.2/5 91 -/>100 92 >100/ May

>100

05 41 -/7 42 0.5/9 93 0.1/3.8 94 0.3/2 95 -/2.7 96 0.1/0.1 120 0.3 / 50 121 0.04/1 122 0.04/1.3 43 2/60 2011202075 44 3/100 45 6/>100 46 5/>100 47 4/>100 48 -/>100 97 0.01 / 0.1 49 -/>100 50 0.1/3 98 0.1/2 51 3/7 52 15/20 53 3/10 99 0.1/1 100 0.5 / 35 54 1/60 55 5/12 56 0.5/10 123 100/>100 57 14/100 124 -/0 125 -/100 126 -/0 111 50/100 58 5/10 59 2/6 60 15/15 61 0.3/4 62 2/45 63 1/8 127 0.02 / 5 128 0.02/10 112 70/>100 103 0.02/2 113 1/100

147 2011202075 05 May 2011 10 5

antiproliferative were medium (HT29 and desired cells/well/500pL assay [0413] Compound

105 air. 129 114 133 132 131 117 116 115 104 130

70 69 68 67 66 65 72 71 incubated

23 as 1

cells)

oxygen

The previously (Gibco)) To

determine below. calculated 100/>100

48/>100 22/>100 71 concentrations for 0.3/100 0.1/0.1 activity <0.1/1 0.4/12 0.5/12 0.7/20 0.5/15 25/75 >100/ 1/100

0.5/5

2/65 medium 8/70 >100 1/80

or 3/3 />100 0.05

0.3 2

n described

HT29 hours 2

the

Antiproliferation

ICso of

Table in

oxygen these (ATCC in 0.1%

values glass

a (see

varying

Bactron 5

Y1

compounds

Oz

inserts

Example dependence

HTB-38, : (μΜ)

IC™

from 0.3%

II

Example

Assay values are in anaerobic 10

29). 24-well

anoxia, RPMI was 148 Oz tabulated

of

- (uM)

NCI-H460 Oxygen tested

phosphoramidate 30 0.6%

medium

plates 0.1%, chamber

6 in 7 in

in

Oz H460

Table

Dependence

a one 0.3%,

multi-well (Gibco)) cells

flushed

cells day l%Oz Y1

50

0.6%, 5 (ATCC

prior (H460 alkylator

were with

Alamar 1%,

to

HTB-177,

10%

cells)

gasses seeded testing.

10%

prodrugs,

Oz Blue-based

or

oxygen,

of at

Table The

the RPMI 20,000

Air the 100 cells 5

Y2

2011202075 05 May 2011 100 110 109 108 106 119 99 54 53 52 98 49 51 50 97 47 46 44 24 38 37 35 26 25 88 84 34 36 16 10 5 0.006 0.007 >100 0.03 0.05 0.01 0.03 0.5 0.3 0.3 0.1 0.4 0.3 0.5 0.3 0.3 0.1 1 0 1 1 10>100 >100 30 10 3 1 1 -0.5 6 3 1 149 >100 >100 0.01 0.85 0.5 100 0.5 30 0.2 0.7 10 45 10 60 50 45 25 60 10 10 5 7 3 3 3 5 6 5 1 >100 >100 40 60 10 10 5 >100 100 25 10 5 >100 >100 >100 >100 >100 >100 >100 0.02 100 100 100 60 0.5 35 20 10 25 >1 40 40 50 40 55 15 7 2 3 1 4 5

' 2011202075 05 May 2011 15 10 5

with in before of 200ppm (ATCC)). HTB-38, were cells clonogenic (0414] Compound

aluminum oxygenation

pre-calibrated

per 25 tested: 114 127 113 128 123 70 66 63 61 56 62 55

the

O

dish To RPMI

2 test A ) survival

NCI-H460

was determine

vessels solution in

and

between

medium 5mL

achieved n

2 >100

added 0.02 0.02 gasses 2

0.3 0.3 0.5

assay 2 2 (see 1 1 5 1

of of

the

cells

medium)

200ppm the

(Gibco)), Example directly

0.1% Table

prior was Clonogenic

by oxygen

test

(ATCC

exposing

perfonned.

to O

Y2: compound

and

2

to 2

experimentation.

PC3 dependence 33)

days

cells

IC 0.3% HTB-1 air

Assay

for 5 the cells o

were

prior Example

(2mL values

2

O Cells glass was

hours. 77,

2

(ATCC 150 -

achieved to Oxygen

of

volume). RPMI

made

0.6%

(μΜ) dishes were compound

phosphoramidate

31

25 For For

>100 >100 CRL-1435,

O 20 30 in plated 30 15 in medium 4 5 8 8

1 1

Dependence

by in the 2

the

complete HT29

Anoxia

a flushing

anaerobic

Bactron aluminum 1%O testing. in

(Gibco)), 60 cells

Ham

2 or

medium mm

the

alkylator

hypoxia Π The

chamber, ’

s

anaerobic vessels, 10% glass anaerobic

F12K

HT29 following

immediately

O

dishes

(less prodrugs, 2

medium

anoxia desired cells

chamber chamber

than

>100 (5x1 cell Air

(ATCC

or

>100 >100

levels a

100 lines 65 0 80 45 10 10 12

4 5 8 5

or

2011202075 05 May 2011 20 10 15 5

aluminum below. hypoxia 90% the checked number to 50 were Levels a and evacuation a exposure after

37°C series

Compound (Cell a 70(H460) 37 25 35 24 23 25 cells 25

drug

plated shaker

killing,

which stained (HT29) (H460) (H460) (H460) (H460) (H460) (PC3)

of water

of

Line) were of was using was

to oxygenation jig

five

for evacuations. in and

one drug,

with achieved 10% washed

bath that counted

a

clonogenic

an

rapid

flushing, 37 more

oxygen allowed the

survival) crystal on degree

evacuations

off a

glass evacuation (see

by

between shaking 0.07

The 0.2 0.5 0.3 0.3 N2 0.2 0.1 the the

violet

2 exposing electrode survival C

for

Example

dishes of

incubator

platform cells oxygen

monitoring

test

platform (0.25% 200

Table

and and

by

were compounds in

the (Anima,

ppm 33).

rinsing concentrations plastic

flushing flushings

(with Electrochemistry

for

glass

Y3:IC removed in

0.1%

Example (controls

and

The of

the 95%

0.4

water

both Petri Phoenixville, with dishes

151 remainder

air

90% 90 02 were was

with ethanol),

values were

from

gas medium.

dishes. bath 32 are

calculated

growth in

performed,

in 95%

and

flushed pre-warmed, achieved

the the

and (μΜ)

of

and

nitrogen liquid Ten

chamber PA)

medium

jigs) 0.6% the inhibitory The

0.25 colonies

0.6 and as

to 5 8 3 3

5 in 1

and

by phases.

was cells

to well). 02

14

a

tabulated

plus

varying

2 specially or and air

the days

shaken hour

were

concentration

aluminum containing

tight

jigs

5% gas After Following

later,

drug

then

the

carbon phases were

aluminum in

for modified

the

Table degree

the

exposure. trypsinized 5 Air

20 40 90 30 50 30

14 vessels

transferred more fifth 5 minutes,

dishes dioxide were

the (ICgo,

Y3

and jigs

than

and

to in

r~ 2011 [0415] To determine the electrochemical properties and reduction potentials of phosphoramidate alkylator prodrugs, cyclic voltammograms of these compounds were May

generated by Bioanalytical Systems, Inc. All experiments were conducted with glassy carbon

05 (3.0mm diameter) working electrodes, Ag/AgCl reference electrodes, and platinum wire auxiliary electrodes. Compounds were dissolved in ImL methanol to make final drug 5 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, 150pm bore capillary, 2011202075 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.

Table: Reduction Potentials (mV)

Compound 100 mV/sec 10,000 mV/sec 1 -596 -638 5 -606 -634

36 -609 -634 25 -594 -626

24 -568 -636

34 -584 -663

78 -704 -746

82 -428,-610 -414,-769

88 -559 -629

108 -614 -593

103 -638, -769 , -875 -756

2-NO2-Imidazole -634 -693

5-NC>2-Furan -487 -638

4-NO2-Benzene -712, -1106 -735, -1268

Example 33

152 Γ 2011

Clonogenic Survival Assay

May [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 05 into 60mm notched glass plates at a density of between 2.5 and 5 xlO5 cells per plate and

5 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

2011202075 10 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.

[0417] At the end of prodrug treatment, plates were removed from each vessel, and the 15 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 20 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] HT 29 and cell based clonogenic assays were performed in the same way as 25 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 μΜ, and by anti-proliferation assay performed by modifying a multi-well assay described by Hay et al., J. Med. Chem., 2003,46:169-82 30 employing H460, HT29, HCT116, and DX-5 cell lines and expressed as IC50 in μΜ (see Example 29). The ratio of IC50 or IC90 determined in normoxia and hypoxia is called hypoxia

153 2011202075 05 May 20 22 21 20 18 17 16 9 P22 15 14 8 7 6 4 2 prodrugs 12 5 P3 11 10 3 P2 cytotoxicity S2 SI 1 Cpd#

0.5 0.9 1.4 1 of logP

the ration

present 0.7 0.25 3.4 1 0.08 8.5 5.2 >10 1 1.6 4.5 0.01 0.14 0.7 1.7 4.5 40 >20 0.4 >40 100 7 8 44.0

(HCR) P Hypoxia

invention.

and 0.2 .572.0 0.35 0.25

C can

be 7.8 9 8.5 9 >100 >100 >100 >100 16 >100 1 100 0.6 25 3.5 >100 3.5 >100 >100 >100 >100 >100 >100.0 10

a

measure P Nonnoxia Table 154 35

75.0

of 1A C

the

hypoxia 23 26 3 8 9 >12 >50 60 >20 >1 >10 60 12 ca. >140 >5 >12 >2 >14 >3 >5 180 1 180 HCR

1 P

selective 175 200 40 C

cytotoxicity

of

the

ο CN & 23 0.04 0.2 5 10 125 50 s 24 0.01 4 400 in o 25 0.05 50 1000 26 0.1 35 350 m 27 2.5 100 40 r- o 31 83 >100 >1 CM o 32 50 100 2 CM 34 <0.01 1.8 >180 o 625 CM 35 0.075 50 36 -0.1 0.88 0.2 >100 >100 >110 >500

a

5 Table IB j Comp HT29 DX-5 HCT116 No. P C HCR P HCR P HCR H N H N P C H N H N 1 50 100 0.4 100 >2 >100 1 >100 >100 23 7.5 2 5 10 >100 >10 6 55 >100 >2 5 >100 >20 7 7(100) >100 >5(1) 0.6 83 140 36 55(35) >100 3 >2 7.5 >100 >13 5 >100 >20 25 16 >100 >6 1 >100 >100 0.9 >100 >100 34 0.8 57 70 0.13 10 77

P = Proliferation; C = Clonogenic; H = Hypoxia; N = Normoxic

Example 34

155 2011202075 05 May 2011 15 5

oxygen- determined twice, per (Guava, centrifuged, concentrations [0420] ΓμΜ

60mm 5 0.5 0.05 0.005 0.0005 0 μΜ 50 5 0.5 0.05 0.005 0 μΜ

and

Hayward, and Cells

dish. incubated

using

and concentration-dependent |

Nitrogen Air Nitrogen

Air Nitrogen Nitrogen Air Nitrogen Air Air Nitrogen Nitrogen Nitrogen Air Air Air Nitrogen Nitrogen Nitrogen Air Air Air Nitrogen Air for (H60,

After fixed

Guava

2 CA).

h

for PC3 Effect

under in 24

additional Cell The 75% h

and

attachment, | of

either

G 35 21 data 55 55 43 55 56 59 Cycle 52 55 60 54 Go/G! 47 33 42 23 07 58 30 58 50 38 56 14 59 Go/G, ethanol

HT29) Compound q ’ /G

demonstrate

i normoxia 22

reagent

were manner at h

cells

IS in least HT29 H460 38 33 PC3 12 13 12 S

12 11 12 6 13 10 13 31 12 S 14 15

19 11 12 11 11 12 18 25 fresh

seeded

(Guava,

156

were (air)

on for

in

cells

that cells cells

medium. multiple

Cell 24

or exposed | at

Compound G^A

65 40 28 59 59 28 42 26 30 G h 38 11 17

Hayward, 26 46 anoxia 32 44

32 33 32 34 31 28 33 32 g a Cycle

2

at 2 density /M /

m -20°C.

human Cells

to

(nitrogen). Distribution

Compound

of CA)

25

were Cell

cancer 1.0

induces

by

x cycle trypsinized,

flow 1

Cells cell 0

6 25

cell cells/3ml distribution

cytometry lines. at

were

cycle the

indicated washed

medium arrest

was

in

Ο CM

0 Air 50 14 36 Nitrogen 47 13 39

May 12 35 0.005 Air 52 Nitrogen 46 14 40 05 0.05 Air 50 15 35 Nitrogen 37 11 52 0.5 Air 48 14 37 Nitrogen 8 8 84 5 Air 47 13 39 Nitrogen 14 50 36 5 Air Nitrogen

2011202075 Example 35 Spheroid Model [0421] Two human cancer cell lines were used in these spheroid studies to determine the 5 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 10 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. 15 [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% 20 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.

157 Γ 2011

[0424] Spheroids were placed in a new growth medium and incubated for at least 1 h before beginning experiments. Spheroids between 500 and 600 pm were isolated by filtering May the spheroid culture through a series of sterile mesh filters of defined size. Between 10 and 05 20 spheroids were placed on a siliconized notched 60 mm Pyrex dish in 3 mL of medium

5 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 O2 (0% O2, 3% O2,10% O2 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 10 removed and the spheroids were washed before being completely digested with trypsin. 2011202075 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, 15 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 20 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 25 used both in monolayer based experments and in spheroids as tabulated below for H460 cells exposed for 2 h.

IC90 values for H460 cells exposed as monolayers or spheroids Drug Monolayer Spheroid N2 0.6% 02 Air 10% 02 Cisplatin 4.2μΜ 7.7μΜ 7.3 μΜ 8.0μΜ Daunorubicin 0.16μΜ 19μΜ Tirapazamine 14μΜ 27μΜ >100μΜ >200μΜ

158 2011202075 05 May 2011 15 10

lying another measurement cisplatin spheroids determine [0429] activated, [0428] [0427] [0426] IC90

cells for

Cisplatin Daunorubicin Cisplatin Compound Daunorubicin 97 Compound Compound Compound 35 70 24 25 25 Compound 36 Compound 1 Phosphoramidate

Similar agent

or H460 The A

as and their in

daunorubicin series

tabulated

the phosphoramidate

of that kill

cells results

ability +

clonogenic spheroid

of targets Compound 25 25 the

Ν2 0.2μΜ 0.35 n 0.1 0.07 1 1.25μΜ

Antiproliferation exposed phosphoramidate +

2

μΜ hypoxic

below: for to

Compound μΜ

μΜ μΜ alkylator penetrate

and aerobic the and

survival.

as

the efficacy

25

kill cells.

3μΜ 0.6% alkylator monolayers Monolayer 0.25 0.6 0.6% Monolayer

spreroids cancer

hypoxic prodrugs 25 into 2.3 IC50 12 14

μΜ

02 The

μΜ

The Assay of alkylator Ο

the

2 2.3 9 IC50

17 cells. Example

prodrug Compound (μΜ) results

results

cancer inner exposed

or

demonstrate - 159 Air 40μΜ

4μΜ 20 Air 75 (μΜ) 100

DNA spheroids

prodrugs

are μΜ μΜ lying

was

are μΜ 36 cells IC99 5.4

for

tabulated Mutant

25 tabulated

combined hypoxic

alone

2 were (μΜ)

to the h 29μΜ Spheroid were 10%

22 »100 »100μΜ 25.5 3 Spheroid 13 15 10% to phosporamidate

Repair μΜ ability

demonstrated and

the

μΜ μΜ μΜ below.

02

tested below:

cancer

μΜ

Ο simultaneously

combination, in 2 μΜ

to Cells combination

in

penetrate cells,

spheroids

in

prodrugs become

the

followed

into with

with HT29 to

the

for

inner

by 2

h. 2011202075 05 May 2011 20 15 10 5

25 well retested Dulbecco from results. present initially (ATCC antibiotics: exposed removed, [0430] [0433] and and [0432] [0431] Compound

under

the 36

as 25 36 ATCC.

under Only The with The Chinese

invention

specific 0^1860),

with Cells screened to

hypoxia.

’ and s

TJV135 Comopund EM9 AA8 IrslSF UV41 Cell AA8

following Modified following

the

cell

the Cell

anoxic The were the IrslSF UV41

AA8

hamster homologous line gene

clonogenic (Anoxia lines cells

2/>100

8/>100 with

cells for Line following

Since AA8 exposed Air)

UV135

or

2 defect:

Eagle table

table

(ATCC defective Nucleotide Nucleotide None Homologous Homologous Mutant Base an 25 assayed. h aerobic

ovary

under

UV41

anti-proliferation / under

lists lists

Medium excision to

(Wild assay cell cells recombination

pathway CRL-1859),

cells selected

hypoxic conditions

participates

in the anoxic the (Anoxia The 0.02 0.02 0.85 4/>100 lines 7/>100

n

(ATCC excision excision

(as EM9 homologous Air) type) 2

IC90 effect mutant recombination recombination

repair (Gibco) following

previously were

IC90 doses

or or

values /

CRL-1

of aerobic

and

aerobic

repair EM9 repair

assay

to tested repair in (μΜ)

160 exposure

supplemented of

specific both assayed

table recombination for (Anoxia 0.03/20

phosphoramidate 0.2/95

cells

867), described) and UV41

and

with conditions. Air) pathway, >300 conditions,

clonogenic Air the 20 17

lists

those

(ATCC

DNA of

IRS1SF by

2,500 nucleotide

/

XPG XRCC1 XRCC3 XPF (None) Gene

various the

proliferation

demonstrating

with to Compound repair

cell

or

CRL-1861),

the

were survival confirm defect (Anoxia cells.

2/>100 6/>100 UV135 3,000

10% cell

lines,

Air) test alkylator excision

pathways

sensitive

lines

All compound fetal cells/well/500pL

the as the for / 25

cell

measured

sensitivity

UV41 to proliferation pathway possibly selected

bovine repair prodrugs

were

Compounds to lines (Anoxia 2/>100 IRS1SF 0.3

Compound

Air) was

cells pathway

obtained serum / were

cells

mutated, by also 59

of were

/

IC50.

the

and

25

as

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 5 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.

10 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 15 test compound was exposed from one side (exposed surface, normoxic side) while the other side was temporarily closed off (far side, hypoxic side). When MCC’s 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 pm of tissue becomes depleted of oxygen. The extent of O2 depletion is greater with 5% than the 20% O2 gassed media; 20 incubation with 5% O2 reflects the in vivo situation most closely. Incubating MCCs with media at 0% O2 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. 25 [0435] MCC based experiments were carried out with media gassed with 0, 5 or 20% O2 for 45 minutes prior to and during incubation with the test compound. HCTI16 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 30 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

161 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) 5 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. 10 [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% (¾ demonstrated that Compound 25 penetrates effectively through relatively well oxygenated tissue. Compound 25 is capable of killing hypoxic cells located about 150pm from functional blood vessels. Approximately 3-fold reduction in exposure to Compound 25 to the hypoxic 15 side was observed under 0% O2 relative to the exposure under 5% O2 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%). Side 0%Ο2(μΜ) 5%Οζ(μΜ) 20%Ο2(μΜ) Hypoxic -1.1 0.7 2.6 Normoxic ~1.7 8.0 >10

20 Example 38 Metabolism of Compound 25 By Human and Mouse Microsomal Protein [0439] An in vitro assessment of metabolic stability of a phosphoramidate alkylator 25 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 (500pL, 5μΜ) was prepared by diluting a DMSO stock solution 100 fold in a water:methanol bridge solution, adding microsomal protein (1 mg/mL) in PBS/MgCl2, and enzymatic reactions initiated by adding an NADPH solution. 50 μΐ of the reaction mixture 30 was withdrawn at 0,10, 20, and 30 minutes after addition of the NADPH solution, the

162 2011202075 05 May 2011 5

26 34 25 23 6 7 No. 16 5 Comp. 1 proteins used study amount

as

compared

positive

of were 28 56 92 71 40 0 0 min) 5(20) 90 (%at30 (MLM) stability Metabolic

Compound Testosterone Nifedipine Testosterone Nifedipine 25 25

precipitated

HLM controls. Compound Compound (at 85 min) 85 (at 84 85 60 (85) 102 30 (% stability Plasma 100 100

min)

20 at 20

25 to

RLM by

250 250 The (mg/ml) MTD with

reversed

first

(Tables acetonitrile min) (6.7 0.11 0.15 0.08 0.26 1 (hr) Intravenous

1/2 study

phase

2A

27.5 7.8 compared Cmax 35 (pg/ml) 16.9

Metabolic Metabolic administration

and and HLM 127% RLM LC-MS/MS. 22% 65% 84% 163 0% 6%

Table Table

Table

2B) the 2.3 4.5 x (gg/ml AUC 12.5

hr)

clear

RLM 2A 2B

in

1 stability stability

mice

3.3

(Vkg) Vss supernatant 1.48 1.25

Nifedipine to

MLM

(% (% 368 /kg) 4.0 CL (ml/min 185

at at

(Table

30 30

MLM 137% RLM was

33% and 89% 2% 4% 6%

min) min) min) (11 0.18 0.16 0.25 (hr) 1 in Intraperitoneal 1/2

mice testosterone analyzed

1)

and 22.9 8.5 3 29.9 (pg/ml) Cmax

the

administration

for

second

were

3.5 the x (pg/mJ AUC 1.0

hr)

2011202075 05 May 2011 10 5

(mg/kg) determined [0441] were (mg/kg) 35 36 [0440] 150 Dose Drug 150 100 128 50 50 24 37 85 37 23 23 36 36 l l 5 5 a a “

a ·* determined ’

a Balb/c Various ... Various 90 56 Route (mg/kg)

i.v. i.p. i-P- i.P· in Dose 25 50 25 20 20 50 50 50 50 50 50 50 50

CD-I mice

in plasma Iv plasma 30%PEG/70% 30%PEG/70% 60 min) 85

CD-I Vivo Formulation mice Route i-P- i.p. i.p. i.v. i.p. i.p. i.v. i.v. i.p. i.v. i.v. i.p. ί·Ρ· Saline Saline Saline Saline

Iv

or pharmacokinetic Pharmacokinetics except mice 400

Vivo tumor Cremophore: CremophorerEthanol

except

25%PEG/75% 25%PEG/7S% 25%PEG/75% 25%PEG/75% 25%PEG/75% 25%PEG/75% 25%PEG/75% 25%PEG/75% where Pharmacokinetics

pharmacokinetic 0.24 Formulation (min) 5.00 5.00 Tmax 15.0

15.0 10% 10% 2.0 where

noted (1:2:7) (1:2:7) Saline 27.7

Ethanol parameters

PEG PEG

of Example Example 164

noted as (pg'mL)

Table Phosphoramidate Table

Saline Saline Saline Saline Saline Saline Saline Saline 27.5 22.9 Cmax 3.38 90.1 15.8 listed 10.8 : :

Saline Saline

as of parameters

4 3

40 39 listed Compound below of

phosphoramidate

1.27 (pg-h/mL)

(min) below 5.00 2.00 5.00 5.00 2.00 5.00 5.00 5.00 Tmax 5.00 5.00 5.00 15.0 15.0 AUC in 1239 325 438 784 307

of Table 77.4 Alkylator

25 Compound

in (pg/mL)

Table

4. 7.60 3.85 3.93 7.80 44.0 Cmax 3.00 35.0 29.9 27.7 8.50 15.0 12.6 16.9 0.18 Half-life (min)

95.2 58.7

11.0 Prodrugs ND alkylator 6.7

3.

25 44 h/mL) AUC

64.0 1439 89.1 57.4 (pg- 12.5 270 210 646 102 172 196 136 were

-

prodrugs

(%)

F* - - - - - 26.1

(min) Half 4.10 9.00 2.56 9.60 23.2 4.67 8.31 8.87 10.0 15.3 11.0 14.1 life -

­ 2011202075 05 May 20 25 20 15 10 5

performed midazolam, for analyzed formic suitable enzyme/substrate prepared test 25) CYPC2 inhibitor phosphate phosphate, 100 100 [0442]

25 50 50 50 50 CYP2C9,

were compound /zL mL

acid

Eight ‘ ‘

of Tumor 19, Bioavailabilitv incubation of

by for (such prepared ude

a in

dehydrogenase, cofactor

pH

quinidine P·

in i.p. i.p. i.p. i.v. testosterone pre-warmed

adding

the

duplicate. 0 reaction (S)-mephenytoin

mice water

Cytochrome

· 7.4, PK ranges as

metabolite

solution. furafylline

along with

30% 30%

at 2.6 solution 50

(400

for

37 10% wells

from

mL

mM H460 Saline Saline Saline Saline Saline

A

PEG/70% PEG/70%

and °C, with enzyme/substrate CYP2D6 mL

summary

of andl PEG

P450

Isoform forms A

0.0229

NADP+, with

to the nifedipine 2C19 tumor for

2C9 1A2 of 10% eight control

for inactivate

:3

reactions acetonitrile

CYP1A2, Inhibition 100

of

serial CYPC219,

and formic /zM wells

of the

6.6 /zL reaction

45.0 5.00 15.0 15.0

2.0 ketoconazole the for to

dilutions probe

Example of mM

the of

were 200 acid

Table solution. IC50

sulfaphenazole CYP3A4)

of

a 1 for

165

enzymes,

:3

solution

glucose-6-phosphate,

dextromethorphan

Control the /zM. with substrate

(400

terminated serial 0.20 2C19).

6.0 8.6 values 6.33 0.93 27.5 38.5

5 9.2

of

Metabolism 41

no

IC50( The mL

the A for

using

dilutions

containing

inhibitor

then zero

are

The test of (phenacetin CYP3A4). reactions 'mM)

for

by

acetonitrile listed

Compound

HPLC/MS/MS. adding compound time-point reactions

40.4 the

247 635 CYP2C9, 177 of

~10

of NI - NI 25 was

for below. Compound 50

addition

a were

100

suitable 0.8

The mM also for CYP2D6

were

for

control (such

U/mL mL initiated CYP1A2,

N-benzylnirvanol

concentrations prepared. potassium

2C1 of 4.43 25.7 7.91

10.1

prepared of positive

Each

25 50 as

of

and 9) reaction

Compound mL by

glucose-6- in

assay diclofenac

.After adding

water

of

control 13.6 and - - -

10%

was

of was

a

to

for

2011202075 05 May 2011 25 20 15 10 5

parameters metabolite(s) be mg/kg Dawley monitored parent high-performance administration whether hepatocytes [0444] collected from spectrometry 60 [0443] Determination

determined and

100

compound Compound Pharmacokinetic Compound 120

rats each

and to

by

are minutes

at 520 are

in (LC/MS/MS). potential compared comparing

in a of computed.

plasma determined concentration

Determination amu. beagle of

20

(Compound liquid 25. 25

the by Formed

mg/kg

is NI

The The metabolite 3A4 quenching In 3A4

are

3A4 to dogs incubated parameters

chromatography the =

Vivo

the

No

pharmacokinetics product

determined Compound

Testosterone following

in Midazolam peak Potential Nifedipine

and 2D6

product

25) significant of Mouse,

of Pharmacokinetics

10

cynomologus

heights with is and the with

ion

Rat,

of Mass μΜ. related

Potential single metabolites

the ion

Compound Rat, acetonitrile by spectra 25.

mouse,

Dog

Example inhibition Example

(HPLC) The

at Balance appearance

a spectrum

Dog Concentrations

to LC/MS/MS

each Example

intravenous

166 and of

reactions

Compound

of

rat, Metabolites 0.049 monkeys

0.03 0.21 0.10 Compound and

Monkey

time the

of 42 detected 43

in are

25

Study prior dog,

of

Compound Human

conjunction potential

and 44

identified of point the

are

monkey

to method potential

administration

in

following its

25.

parent

centrifugation

of stopped

of Rats 25

acquired. Hepatocytes metabolite(s)

Compound

The metabolites Compound >50 >50 >50 and

NI

by 25

and and with

compound metabolites

disappearance performing

its and single at

mean human

metabolite(s) tandem 0

its of (pre-incubation),

and 25

25

intravenous

in are Metabolite pharmacokinetic 5,20,

to cryopreserved

and Sprague

analysis mass over

subsequently

determine full

50

its of

scans time

will and the

(s) by

are

100 also

in 30,

2011202075 05 May 2011 30 25 20 15 10 5

using Compound 30 samples is three bladder images prostate [0447] contents), intestine marrow, lens), phosphorimaging (brown of plasma, (LSC). times 25 Storm CM carcasses 25. [0445] [0446]

determined

minutes the

as

At 3600

concentrations

human

a Harderian and

image

820

The

specified are single

and (and

are and

gland, cecum and Normal

Sprague-Dawley

are cryomicrotome. small concentrations

produced or

at 25) protein

then white),

the

ultrafiltrate contents) analysis

contents,

embedded 2500 using 860. intravenous

salivary and

and blood intestine

gland, equilibrated times,

and plates

x binding The analysed

adrenal ultrafiltration. contents, with

for

g. software. bile-cannulated

and

are liver,

heart, gland, one concentration

A

in along each

standards

(and of Compound

measured dose. rats plasma 75

2%

Collected

gland, in

rat

total lung, using

Quantitative

to

kidney Plasma animal. epididymis, pL mouse,

contents), seminal with

CMC, are per

37°C. Exposed

Blood radioactivity aliquot

lymph blood, are

administered LC/MS/MS. for Ultrafiltration time 14

by

(cortex,

25

sections C

Protein

frozen

Sprague-Dawley rat, of analyzed the vesicles, The

plasma,

image autoradiographic point

into

spleen, radioactivity of

node brain screens

dog, Whole calibration esophagus,

Centrifree® the

Example

Example into 167 a

medulla, Binding

is

are

are analysis. Centrifree® submaxillary), monkey

(cerebrum,

ultrafilitrate

for

urine, skeletal euthanized. a spinal The

Body a

are

is freeze-dried, single determined

block

concentration performed

ultrafiltrates scanned

curve.

46 eyeball

in of 45 feces

papilla

cord, Autoradiography rats and

apparatus muscle, Autoradioluminographs intravenous

select Compound standards and

cerebellum,

device

are human is

and

trachea, Blood

sectioned

using (Uveal pancreas, by and

spiked

by tissues mounted administered

are

skin,

liquid

of

are aliquoting entire in is

plasma for is a collected radioactivity.

25 dose centrifuged

triplicate.

tract, thyroid

with Molecular

analyzed centrifuged stomach including

subsequent medulla)

at scintillation

pituitary

and section),

40 of

the aqueous of

exposed plasma 14 1

pm and

at Compound 4C-Compound I.S.

C-Compound

(and and

All

specified

adipose Dynamics bone,

at in and gland,

urinary

large to

(deuterated Frozen calibration

37°C

quantified

a plasma humor,

counting obtain

spiked

digital Leica on

bone

for 25

rat

at

2011202075 05 May 2011 25 20 10 15 5

5% American pathogen-free reduced measured mg/kg mm weeks compared Compound growth (L xenograft PBS) supplemented cancer caliper only in [0448] Compound and [0449] [0451] [0450]

the

x CO2.

3

PEG

W 5FU (day

at

peritoneal

of

Group of employing 2 twice Administration compared

)

tumor a

Example

Female The /2, 4 6 5 2 The 3 age, (lOmL/kg

5FU daily twice was at 8), Type to

36 36

where 10

vehicle.

per each body HT-29

were

with conditions. monitored and (in at growth

mg/kg dose weekly. Culture

subcutaneous week. CB17/SCID a

to

saline)

a

group 47

daily 5FU

L weight

10% allowed

HT-29 each),

administration Treatment of

cells is demonstrates In

to (Group

of

20,

the resulted

Tumor Collection.

dose addition fetal

of a

60 20 (Group by Compound

were

Human

Group of greater 60, human length

10

to 200 + + externally 60 20

each bovine of mice

8) 5FU 5FU acclimatize mice space. or

(mg/kg) volume harvested

20, in

as 5,6 combinations 200

1),

and degree colon mouse colon

greater

(purchased

the of

tabulated 60, was

The

Compound serum.

36 and

mg/kg

When vehicle W

usefulness measuring

(V)

and

at carcinoma

administered carcinoma cell

is than 7, was

from

and for 20,

the was

200 respectively) Example

the

(Groups 168 Cells lines below.

at alone. recorded

Group 5FU dose 60,

from width culture of

determined tumors mg/kg 46.9 least 35.7 20.2 34.6 36 16.1

tumors of 60 were and were

% alone.

cell alone xenograft related

Charles

a

and Administration

of three 1 2, 47

Inhibition for

compound 200

and

given twice

line cultured grew

maintained 3 a

200

in and three (dissolved xenograft. and

mg/kg/day inoculated days, inhibition

by HT-29 two River,

to per 2-3

mg/kg mouse compared Group 4,

the weeks,

an 13.3

3.3 vs dimensions in

respectively),

and week. hours of - - -

following average RPMI Cambridge, was in

this

in of model. 8 of handled Tumor

of

at each a

vehicle

30%

Compound

after a

obtained 3

37°C to tumor Growth invention

combination

1640

x

a volume reduced

cyclodextrin using 10 group volumes a

equation: incubator under

dose alone 6 and

growth media MA),

cells/

of from

a

in

receiving

36

each of

of tumor

digital

(saline

treating

7-8

were

animal

100 10 the of V

with

in =

2011202075 05 May 20 20 15 10 5

peritoneal [0454] inhibition. in occasional American pathogen-free weeks Example Compound (day the Compound 5% cancer [0452] [0453]

other

procedure EtOH,

(n= 8), Group

lb* la* of employing Associated 4 9 Ί 6 2 8 5 3

groups each Female

10)

Example 47 7 age, 8

subcutaneous

Type

5% mortality,

25). 25

above.

group

were described

conditions. Not (2.5 Cremophor

as

Culture CB17/SCID The

Compound well. Compound Compound Compound Compound a Compound

48 with

Applicable mg/ml

allowed of NCI Treatment

body particularly Vehicle demonstrates

Saline Taxol Taxol Taxol mice

these space. Collection. in Overall, 200 H460,

in Human and weight Example

was 5FU to

10% + mice

anti-tumor 25 25 25 25 25 25

acclimatize (NA) 90%

5FU When human

administered

Compound in

PEG; and colon (purchased

the

the saline; 47 The Treatment

the tumor

colon usefulness

and group

administration

carcinoma Example effects, (mg/kg) cell

tumors

Dose

for NA NA NA NA 100 100 administration 25 50 25 inoculated 50 10 10

36 169 volumes lines carcinoma

for treated at from

protocol showed least there 38.7 58.2

grew three

of 48 were

cell Charles

a

with three

were was

compound at to weeks, route

line cultured varying xenograft

1 an

- (q2d (q2d (q2d (qld (qld (qld the

(q7d (q2d (q7d (q2d (qld some x days,

measured

River, NCI i.v.

average - 1

high 0

as i.p.)

6 x x x x x x

2 x x x x x

rates

degree and

23 cells/

H460 and

tabulated 3) 3) 3) 5) 5) 5) h - of 3)/week

3)/week 5)/week l)/week l)/week

mouse

Cambridge,

Regimen dose and

after /week /week /week

/week /week /week

this

harvested volume

handled of NA as

animal

was

Taxol

of

tumor of described invention administration

model.

x

x x x x x weight x x x x

x in Compound 2 2 2 2 obtained

2

2 2 2 2 2 2 of

the under

in (1 weeks weeks weeks weeks weeks

weeks weeks weeks weeks weeks weeks growth MA), according

100

the

mg/ml table

loss

in in

mm

from 7-8 treating

and below: 36

of in 3

to

but

the

2011202075 05 May 20 25 20 15 10 5

weeks model. particularly benefit when cancer doses. administered comparisons saline therapeutically indicate some [0458] every similar [0455] [0457] [0456]

degree vehicle

day

or

of

as

qld/qd Groups to degrees

Female that no

Example Using Results The

age, demonstrated

that dosing

lung treatment

Compound of

in

with Group results treated were provided

=

effective

la this the weight of CB17/SCID 9 7 4 6 3 8 5

cancer, every are provided

a and tumor 49

mouse

allowed analysis. decreasing

presented demonstrate mice

describes

were

Taxol lb,

loss day; employing by

25 dose alone growth

n

had to the

additional

is but added

to q2d =

mice HED

of

efficacious

5;

acclimatize standard or reached

in frequency not the

Group

= about inhibition

in

Table that

65.9 46.7 52.6 63.1 50.1 (purchased in

a conversion, 46 52 every usefulness large

combination

H460, order

benefit. %

all

2 2

a

chemotherapeutic X2

to Inhibition

volume enough second three in of

for to

and about non-small based 10 this Example

dosing 170

indicate

from at

of

Each

regimens Compound that

model

least

day; to a of with

8 on Group

compound vs

mg/kg/day, Charles

cause

combination 946 38.8 for 31.7 combination 12.5

tumor

q7d three - - - any - 49 lung Taxol™,

of

higher

mm

for

6

any lung vehicle = agent,

(q2d

25

carcinoma days,

River,

volume 3 dosing every .

mortality. of can

doses Groups cancer

for x wherein

this therapy, taxol.

therapy and

3) effects be

Cambridge, the

seventh

Compound measurement

/week

compared

invention administered handled

and xenograft treatment of

Overall

the

but was particularly 5

provides

x day. mice

daily

2 are

associated

under

MA), weeks in 25 to

the mouse of not receiving

treating on dose lower

provided at

cancer,

additional

results

used

day 7-8

a with

can

with

29

for

be

2011

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

May harvested as described in Example 47 above, and inoculated at 3 x 106 cells/ animal in the

05 peritoneal subcutaneous space. When the tumors grew to an average volume of 100 mm3

5 (day 8), each group of mice (ten per group) was administered for three weeks, as tabulated in I

the table below: Compound 25 (2.5mg/ml in 10% PEG; administration route - i.p.); Compound 24 (0.3,0.1mg/mI 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).

10 Treatment protocol 2011202075 No. of Dose Group Treatment Regimen Mice (mg/kg) 1 10 Vehicle* NA (ql d x 5d)/week x 3week 2 8 Taxol 10 (q2d x3)/week x 2week 3 8 Compound 24 3 (qld x 5d)/week x 3week Compound 24 1 (qld x 5d)/week x 3week 4 9 Taxol 10 (q2d x3)/week x 2week Compound 24 3 (qld x 5d)/week x 3week 5 8 Taxol 10 (q2d x3)/week x 2week Compound 25 25 (qld x 5d)/week x 3week 6 8 Taxol 10 (q2d x3)/week x 2week * - 50% PEG [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 ) 15 vehicle group and those animals were sacrificed.

Group % Inhibition vs Group 1 Group 2 3 39.9 - 4 16 -32.7 5 51.5 23.3 6 56.8 31.7 2 36.7 -

[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 20 by mild weight reductions, particularly in the Compound 25 + taxol group.

171 2011202075 05 May 2011 20 10 15 5

cancer, therapeutically Female lung therapeutically alone. was tumors age, cancer saline) above, conditions. [0461] Culture [0462] 10%PEG), *-

administered were cancer,

tumor

as and on grew alone CB17/SCID Collection. 7** 6** 4** 3* 2* 8** 5** Group 12** 11** 10** 1*

Using Example

demonstrated

allowed the

administration inoculated

Human

alone location

to

or days

effective effective an the in

4 9 9 # 9 9 9 9 9 9 8 8 8

for

to average or 50 combination

the mouse mice

colon The

mice

acclimatize

in flank; three

describes

at combination

employing combination

dose dose 3 cell

(purchased route carcinoma

to

x No volume No CDDP Compound CDDP Compound Test weeks, Compound 5-FU Compound CDDP Vehicle Compound 5-FU 5-FU Saline

**

10 lines HED

of of

treatment treatment - 6

the

- Article with

tumor cells/ 0.25 2

for

i.p.,

mg/kg/day, were

as conversion,

of a usefulness

(saline) therapy

at HT-29,

Taxol™,

Treatment with tabulated cell mg/kg/day,

from

100

anima] administrated

24 24 24 24 24 location least

cultured

line

Taxol™. mm

Charles

was three Example human

172

in N/A HT29 N/A

5 6 5 3 50 6 6 3 50 Dose 5 (mg/kg) 50 3 10 10 for

of and

in Compound

(day

peritoneum;

protocol the

scheduled; and

ml/kg

ml/kg a

for the the days,

compound Compound

River,

peritoneal

colon

was

8), 2

harvested the table

treatment h 50

each and

before treatment obtained

Cambridge, carcinoma below: 25 N/A iv, ip, iv, N/A ip, iv, ip, ip, ip, iv, iv, ip, iv, Routes, iv,

handled 5FU

Q3d group

can of once once (qld q3d (qld once q3d (qld (qld (qld q3d (qld q3d

24 subcutaneous

as 5-FU of

this

alone

=

can described

be

from Compound cancer, x

x x x of

Regimens x x x of x x x every

4 4 under 4 4

invention administered

or xenograft cancer, 5)/week 5)/week 5)/week 5)/week 5)/week 5)/week be

mice

MA),

(in

the cisplatin

administered

third

particularly

saline),

pathogen-free

American

(ten in

space. 7-8 particularly

x x 24

x x x x

Example

day in

mouse 2week 3 3 3 3week 3

per

weeks (CDDP;

week week week (in week treating

or at

group) When

a

CDDP Type

lung

at model.

of 47

a

in

the

r 2011

[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

May 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 05 5 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

Group % Inhibition vs Group 4 Group 6 7 44.1 - 8 42.1 - 2011202075 9 71.1 28.9 10 53.2 24.8 11 50.7 20.9 5 59.3 - 6 37.4 -

[0464] The results demonstrate that Compound 24 as monotherapy resulted in tumor 10 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.

15 [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 20 [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 25 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

173 2011202075 05 May 2011 30 25 20 15 10 5

therapy administration was tumors and Female Culture conditions. age, cancer lower particularly [0469] can therapeutically regimens demonstrate that delay 25. [0468] 25 times times treatment peritoneal [0467] CDDP 6

mg/kg

are

inoculated be administered were employed

with per doses. (Group

illustrated as

grew administered

(Groups is CB17/SCID Collection.

(Groups Example

Using These

demonstrated

allowed scheduled add week

was

subcutaneous a

Human lung

to daily significant

to 5)

route

initiated

at for

an

effective

the results for

6

the

or

cancer,

3 in for

2

and average to the dose

52

x the two

mouse and

colon the The - effects

mice with and

acclimatize three

10

describes 50 i.p., 7,

performed combination

in 6 employing of weeks Figure 3, dose

cell doses respectively). CDDP space. alone cells/ mg/kg

a

(purchased which carcinoma

respectively, 50 volume administrated weeks,

to

decreasing of

lines

related HED mg/kg,

CDDP

(Group

animal or of

1.

the (in daily When

groups for

in about Figure were

as with

of

a conversion, usefulness saline,

of combination

at decrease HT-29, Treatment

tabulated cell and

from in

100 dose.

in 4), frequency

the least each

a Results

cultured IV 4 this

of

Example 2 the saline

2

line 100 to mm Compound

tumors

IV). one h

shows 10 Charles

dose three These about model. peritoneal before human 174

mg/kg HT29

3 in mice

of

in

time), (day

Compound formulated for protocol

tumor and

a

of of the

with

days,

similar grew

52

8

data compound groups

River, received

CDDP

dosing Compound 8), colon was

mg/kg/day,

harvested with

table 25 Compound

5FU

volume

subcutaneous each and also to

at obtained

results

less

Cambridge,

an carcinoma below:

receiving

on version 25

for 100 or

handled

demonstrate vehicle group average

the of can frequent CDDP,

higher

as

and mg/kg

25

for

for this

25

described days from Compound

be

with of of

increase

the under at

100 50 (Group

invention xenograft space. administered

Compound volume doses mice

wherein

every MA),

50

the dosing

the mg/kg treatment

either mg/kg

that

mg/kg

pathogen-free combination

American

(ten

in

compared in 1), three When 7-8

both 25

of

3 Example compared

of the tumor

in

of mouse per CDDP

or

weeks 100

in in

Compound 25

of treating

Compound days daily dosing

6 at

the saline saline, group)

cancer,

mg/kg mm

a growth Type

to model.

at

for

of 47 dose

3 to

5 3 ,

5

or of

2011202075 05 May 2011 10 5

in ended. inhibition in cancer Compound [0471] growth sufficient Data 9 8 4 2 7 6 5 3 [0470] 10 1 (n Group

the saline,

=

are 9) combination

in

are

The based this at

The

of size Compound CDDP Compound CDDP Compound CDDP CDDP Compound Compound Compound Compound Compound CDDP Saline

25 tabulated 50mg/kg/day

tumor

Test model. results

body

and Group to

on 10

2 9 7 8 6 5 4 3 require

Article

tumor CDDP

groups;

growth weight

demonstrate below. These

25 25 25 25 25 25 25 25

that

volumes

and enhanced

the in and

effects Group

the 5 5 5 5 50 5 100 50 this 100 100 100 100 100 lOml/kg 100 24.2 36.2 44.2 51.8 50.7 48.1 31.3 30.1 28.2

mice (mg/kg) tumor Dose

mice

that %

model mgZkg/day at

were

1

the Inhibition recovered day

volume monotherapy be

effectiveness

of 25

accompanied sacrificed. Once q7dx2 Once q3dx5 Once (q2dx3)/week Once (qdx5)/week (q2d Q7dx2 Q3dx5 (qdx5)/week Once (qd

colon when 175

Group Administration with

was

x the vs 21.5 33.2 27.5 30.8

x

5)/week - - - - -

cancer, 3)/week

tumors lost determined a

2

administering variety

The of

body by

Compound

x x and results in x2week x2week

modest

x 2week 2week

of the weights

2week

Regimens that

as dose

vehicle

of described

body

treatment Compound

inhibition regimens 25

after

for weight group

the

treatment in

combination

treatment results of Example 25, had

loss,

tumor

formulated reached

more

of in

47. colon

of

so

r 2011 [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, May

particularly non-small cell lung cancer, alone or in combination with CDDP, wherein the

05 daily dose can be administered with a decreasing frequency of dosing for higher doses 5 compared to lower doses.

Example 53 [0473] 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

2011202075 10 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. 15 When the tumors grew to an average volume of 100 mm3, 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 20 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 25 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, 30 alone or in combination with CDDP.

Example 54

176 Γ 2011

(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

May 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 05 5 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 10 Compound 25 (formulated as a 15 mg/ml saline solution, administration route - ip.,

2011202075 administrated 2 h before CDDP on the days the combination therapy is scheduled and CDDP in saline, IV.

Treatment protocol

Group Test Article Dose Regimens Dose Concentration: (mg/kg) Dose Volume 1 Saline 10 Q7dx2 0 mg/mL : 10 mL/kg 2 CDDP 6 Q7dx2 0.6 mg/mL: 10 mL/kg 3 Compound 25 150 Q7dx2 15 mg/mL : 10 mL/kg 4 Compound 25 150 q7dx2 15 mg/mL: 10 mL/kg CDDP 6 q7d x 2 0.6 mg/mL: 10 mL/kg 5 Compound 25 150 Q7d x 2 10 mg/mL: 15 mL/kg 6 Saline 0.2 ml 200 pL -1 week x2* 0 mg/mL : 1 pL/hr 7 Compound 25 15 mg/ml 200 pL -1 week x2* 15 mg/mL: 1 pL/hr * - Alzet pump, 200 pL for 1 week x 2 (re-implant new pump at the end of one week). 15 [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.

20

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. 25 MiaPaca-2 tumor is a highly invasive, rapidly growing tumor that results in death within 20-

177 ) 2011202075 05 May 20 25 20 15 10 5

significantly administered stand gemcitabine cured. and treatment provided survival Group [0481] body survival. [0480] 5) [0479] powder. manufacturer 25 Compound Compound fluorescent 30

and

were days the

of images

4 2 6 3 resulted 5 Group 5 These 1 tumors *

care,

in compared

demonstrated formulated Four

Tumors modest Groups qd Gemcitabine and Group

untreated

protein.

24 25,

is = greater

with were

’ gemcitabine. results

within

s of

every of

were were

in Compound instructions. Gemcitabine Compound Gemcitabine Compound Gemcitabine Compound Compound Vehicle Compound

2

these reduction

3

greater

were

a 100%

significantly obtained to and

combination

than

in

administered considered day; demonstrate Mice a animals. Group

significantly tumors short was saline imaged 4

lethality

benefit that resulted qw

were

These in

obtained 24,

24 25 25 to 24 period 2.

=

and in

tumor The

remained

confirm

once or every

Five reduced

to animals administered

in

of

by

provided results that in gemcitabine/

i.p.,

tumor be

200 200 6 30 30 200 Dose 6 this lOml/kg

failed

Compound reduced

minor day size weekly commercially out

week. Treatment cured.

combination

as

effects. model

tumor at

treated of

30

(mg/kg) demonstrate but

cells tabulated to

zero effects

by

8

(Figure

until emit tumor no 178

No tumors

doses

Threshold of had

volume

Compound 25 In fluorescence

with additional

protocol

tumors

cancer fluorescence the

on

Group been

at below and growth treatment

qw (qd qw (qd 6). (qd (qd qw (qd of Schedule

in gemcitabine 30

end tumor that

vehicle

’ prepared

and

’ Group x x

x x x x transfected

mg/mg/day compared

x in x

Pharmaceuticals,

of

1, 5)/week 5)/week 5)/week 5)/week (8

tumor 3 3

5)/week effects

and 3 Group

25

prolonged

weeks weeks the

volume the mice/group).

weeks until with

combinations

control, 5 (Figure significantly

tumors study

regressed freshly reduction

2

on as

for for the for for Compound

to

for

with

were

and and

a survival. monotherapy 2

2 2 2 end at

survival.

single

2 7). gemcitabine,

grew

weeks weeks weeks weeks

according had which gemcitabine the weeks

considered

of rapidly Compounds Inc. in

prolonged

gene or

little

rapidly

the agent. animals

25

In time as gemcitabine/

Group

experiment

and

for contrast, a effect

after

to

with

dry

open

(Figure

to red

is

6 24 be

on

the

and

[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.

5 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 10 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 toxicides such as tubular necrosis and 15 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.

20 [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 25 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 30 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.

179 180

CLAIMS 2012

1. A compound having the formula: Dec

06

wherein

2011202075 L is selected from the group consisting of CH2, CHMe, CMe2,

(6890905_2):RTK 181 2012

Dec

06

2011202075 each R.9 is independently selected from the group consisting of hydrogen, aryl,

heteroaryl, C1-C4 alkyl, C1-C6 heteroalkyl, C3-C8 cycloalkyl, heterocyclyl, C1-C6 acyl,

C|-C6heteroacyl, aroyl, heteroaroyl, C1-C6 alkoxycarbonyl, C1-C6

alkylaminocarbonyl, di C1-C6 alkylaminocarbonyl and C1-C6 alkoxy;

each X4 is selected from the group consisting of halo, alkylsulfonyloxy,

heteroalkylsulfonyloxy, cycloalkylsulfonyloxy, heterocyclylsulfonyloxy,

arylsulfonyloxy and heteroarylsulfonyloxy;

or a pharmaceutically acceptable salt thereof.

2. The compound of claim 1 wherein Z3 is selected from the group consisting of:

(6890905_2):RTK 182 2012

Dec

06

2011202075 each Rg is independently selected from the group consisting of hydrogen, aryl,

heteroaryl, C1-C4 alkyl, Ci-Ce heteroalkyl, C3-C8 cycloalkyl and heterocyclyl.

3. The compound of claim 2 wherein Z3 is selected from the group consisting of:

and X4 is selected from the group consisting of Br and Cl.

4. A compound of claim 1 having a formula selected from the group consisting of:

(6890905_2):RTK 183 2012

Dec

06

2011202075

(6890905_2):RTK 184 2012

Dec

06

2011202075

0 /\^Br

Br 5

(6890905 2) RTK 185 2012

Dec

06

2011202075

(6890905_2):RTK 186

5. A pharmaceutical formulation comprising the compound of any one of claims 1

2012 to 4, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable

excipient, carrier, or diluent. Dec

06 6. The pharmaceutical formulation of claim 5, wherein the diluent is water,

ethanol, propylene glycol, glycerin, or combinations thereof.

7. A method of treating cancer comprising administering to a patient in need of

therapy thereof, the pharmaceutical formulation of claim 5 or 6.

8. Use of a compound according to claim 1 for the manufacture of a 2011202075 pharmaceutical formulation for treating cancer.

9. The method of claim 7 or the use of claim 8, wherein the cancer is selected from

the group consisting of a solid tumor cancer, a soft tissue sarcoma, a leukemia,

multiple myeloma, prostate cancer, pancreatic cancer, breast cancer, lung cancer,

colorectal cancer, head and neck cancer, ovarian cancer, brain cancer, an astrocytoma

and a renal cell tumor.

10. The method of claim 7 or the use of claim 8, wherein the compound is

11. The method of claim 7 or the use of claim 8, wherein the compound is

(6890905_2):RTK 187

Br 2012

Dec

06

and the cancer is colorectal cancer.

12. The method of claim 7 or the use of claim 8, wherein the compound is 2011202075

and the cancer is ovarian cancer.

13. The method of claim 7 or the use of claim 8, wherein the compound is

and the cancer is lung cancer.

14. The method of claim 7 or the use of claim 8, wherein the compound is

(6890905_2):RTK 188 2012

Dec

06

15. The method of claim 7 or the use of claim 8, wherein the compound is 2011202075

16. The method of claim 7 or the use of claim 8, wherein the compound is

17. The method of claim 7 or the use of claim 8, wherein said method or use further

comprises treating the cancer in combination with another anti-cancer agent.

18. The method or use of claim 11, wherein said method or use further comprises

treating the cancer in combination with daunorubicin or cisplatin.

(6890905_2)RTK 189

19. The method or use of claim 13, wherein said method or use further comprises

2012 treating the cancer in combination with daunorubicin, paclitaxel or cisplatin.

Dec 20. The method or use of claim 15, wherein said method or use further comprises

06 treating the cancer in combination with gemcitabine.

21. The method of claim 7 or the use of claim 8, wherein the compound is used in a

dosage amount of 0.001 to 0.1 g/kg/day in single or divided doses.

22. The method of claim 7 or the use of claim 8, wherein the compound is used in a

dosage amount of 0.1 to 35 mg/kg/day in single or divided doses. 2011202075

23. The method of claim 7 or the use of claim 8, wherein the compound is used in a

dosage amount of 0.5 to 15 mg/kg/day in single or divided doses.

24. A method to synthesize a compound of claim 1 comprising reacting a compound

having the formula Z3-L-OH, a trisubstituted phosphine, a compound of the formula:

O

and a dialkyl azodicarboxylate to yield the compound of claim 1.

Dated 5 December 2012

Threshold Pharmaceuticals, Inc.

Patent Attorneys for the Applicant/Nominated Person

SPRUSON & FERGUSON

(6890905_2): RTK Ο (Μ Figure 1. 1/7

Compound 25 (50mg/kg) Efficacy in H460 Xenograft May

05

2011202075

Day 2/7 ο Figure 2 (Μ Compound 25 Efficacy at 100 mg/kg in the H460 Xenograft &

*D Ο

Ο (Μ

0 i 2 3 < 5 6 7 8 9 10 II 12 13 14 15 « 17 II 19 3 21 22 a a 55 S Ώ 3 B 30 31 fey 2011202075 05 May 2011 Figure

3 Study

Day 3/7 — — — • ♦ — — Compound Compound Safina COOP

25 26

(IP) »

CDDP 2011202075 05 May 2011 Figure

4 4/7 5/7 Figure 5

IT)

Ο (Μ Ο (Μ

Ο (Μ Figure 6. 6/7 20

May

05

2011202075

0 2011202075 05 May 201 Figure t- > 3 E o 3 o E Φ

10000 10

7 1

0 1 0

0 0 0

- - 2

0 Study 7/7

3

Day 0 4

0 6

o 6

0