CA2647655C - Novel c-1 analogs of pancratistatin and 7-deoxypancratistatin and processes for their preparation - Google Patents

Novel c-1 analogs of pancratistatin and 7-deoxypancratistatin and processes for their preparation Download PDF

Info

Publication number
CA2647655C
CA2647655C CA2647655A CA2647655A CA2647655C CA 2647655 C CA2647655 C CA 2647655C CA 2647655 A CA2647655 A CA 2647655A CA 2647655 A CA2647655 A CA 2647655A CA 2647655 C CA2647655 C CA 2647655C
Authority
CA
Canada
Prior art keywords
compound
formula
nr2r3
groups
defined above
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CA2647655A
Other languages
French (fr)
Other versions
CA2647655A1 (en
Inventor
Tomas Hudlicky
Jonathan Collins
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Brock University
Original Assignee
Brock University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Brock University filed Critical Brock University
Priority to CA2647655A priority Critical patent/CA2647655C/en
Publication of CA2647655A1 publication Critical patent/CA2647655A1/en
Application granted granted Critical
Publication of CA2647655C publication Critical patent/CA2647655C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/056Ortho-condensed systems with two or more oxygen atoms as ring hetero atoms in the oxygen-containing ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/14Ortho-condensed systems

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present application relates to compounds of Formula I:
(See Formula I) to pharmaceutical compositions containing these compounds and to the use of these compounds in the treatment of cancer. The application also relates to processes for the preparation of these compounds and to the preparation of intermediates in this preparation.

Description

TITLE:

DEOXYPANCRATISTATIN AND PROCESSES FOR THEIR PREPARATION.
FIELD
The present application relates to novel C-1 analogs of pancratistatin and 7-deoxypancratistatin and to processes for their preparation. The application further relates to pharmaceutical compositions containing the novel analogs and to uses of the analogs.
BACKGROUND
Pacratistatin (1) and narciclasine (2) are natural products that are highly active against many cancer cell lines including murine P388 and lymphocytic leukemia;

human cancer cells pancreas BXPC-3, breast MCF-7, CNS SF-268, lung NCI-H460, colon KM20L2 and prostate DU145. Although the exact mode of action for pancratistatin remains unknown, narciclasine is believed to inhibit peptide bond formation in eukaryotic ribosomes. Lycoricidine (3) and 7-deoxypancratistatin (4) are significantly less active, than the corresponding C-7 hydroxylated compound.
The reduced activity maybe due to the absence of the hydrogen bonded donor acceptor pair in the phenanthridone functionality.
OH OH OH OH
HO 40 OH OH fib OH HO OH
<0 OH 0 7 OH <0 7 OH 0< w OH
0 NHo NH 0 NH 0 NH
2 SUMMARY
The present application describes novel C-1 substitution analogs of pancratistatin and 7-deoxypancratistatin.
Accordingly, one aspect of the present application includes a compound of the Formula I:
OH

O wOH
Co NH

Formula I
wherein:
R1 is selected from C(0)0R2, C(0)R2, C(0)NR2R3, CH=NR2, CH2NR2R3, CH2OR2, CH2R2, CH=CR2R3, NR2R3, NHC(0)R2, NHC(0)0R2, NHC(0)NR2R3, CH20C(0)NR2R3, CH2NHC(0)R2, CH2NHC(0)0R2, CH2CHC(0)NR2R3 and CH200(0)R2; and R2 and R3 are independently selected from H, C16alkyl, C2_6alkenyl, C3_ iocycloalkyl and C6_10ary1 said latter four groups being unsubstituted or substituted with one to 5 groups independently selected from halo, OH, OCi_ 4alkyl, OC(0)C1_6alkyl and nitro; and R4 is selected from H and OH;
or a pharmaceutically acceptable salt, solvate or prodrug thereof.
In another aspect of the application there is provided a pharmaceutical composition comprising one or more compounds of Formula I as defined above, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier.
3 In a further aspect of the application there is provided a use of one or more compounds of Formula I as defined above, or a pharmaceutically acceptable salt, solvate or prodrug thereof, to treat cancer.
In still a further aspect of the application there is provided a method of treating cancer comprising administering an effective amount of one or more compounds of Formula I as defined above, or a pharmaceutically acceptable salt, solvate or prodrug thereof, to a subject in need thereof.
In another aspect of the present application is a use of one or more compounds of Formula I, as defined above, or a pharmaceutically acceptable salt, solvate or prodrug thereof, as a medicament.
A further aspect of the present application is a process for the preparation of an intermediate of the Formula II:
0Pg H(0)C 0Pg 0 vip org 040 N- Pg wherein R4 is selected from H and 0Pg and each Pg may be the same or different and represent suitable protecting groups or any two adjacent Pg are joined to form a suitable cyclic protecting group;
the process comprising:
4 (i) reacting a compound of the Formula III with an aluminum acetylide derived from a compound of the Formula IV, followed by protection to form a compound of the Formula V, wherein R4 and each Pg is as defined above:
r el + < <0 0 . 0Pg _________________________________________ lir _ 0 R4 0 0Pg -K/L`'OPg PgKP'' R40P9 PgN
-0, III IV V .
, (ii) reducing the compound of Formula V to form a cis-alkene of the Formula VI, wherein R4 and each Pg is as defined above:
0 0 0Pg <o =
-- 0Pg 0Pg _ R4 0P9 --70 R4 el .iWo, 0Pg 0P9 0 Pg Pg1\i''' \-0 VI -V , (iii) reacting the compound of the Formula VI under solid-state, silica gel catalysis conditions to form a compound of the Formula VII, wherein R4 and each Pg is as defined above:
0Pg 0Pg -- dh 0Pg AbAh 0P9 40 õ..= 0Pg R4, l/1W 0Pg Nµµ
1 NPg 0 Pg -00- 0 \--0 \-0 VI VII .
, (iv) oxidatively cleaving the double bond in the compound of the Formula VII
to form an intermediate diketone of the Formula VIII which cyclizes to form a compound of the Formula IX, wherein R4 and each Pg is as defined above:
0Pg 0 0Pg 0Pg H(0)C i& opg 0Pg 0_ 0Pg R4 deW OP g _ip,, R4 si W
_ 0Pg -bi- (0 401 W_ 0Pg VP NPg NPg 0 Mpg VII ¨ ¨ VIII IX
5 , (v) oxidizing the compound of the Formula IX to form a compound of Formula II, wherein R4 and each Pg is as defined above:
0Pg 0Pg OHC 0Pg H(0)C 0Pg ( 0Pg _________ 0 lei 0 is W
el 30. 0Pg 0 NPg 0 NPg Ix II .
6 In a further aspect of the application there is included a process for preparing a compound of Formula I
OH

si 7 OH

wherein R1 is selected from C(0)0R2, C(0)R2, C(0)NR2R3, CH=NR2, CH2NR2R3, CH2OR2, CH2R2, NR2R3, NHC(0)R2, NHC(0)0R2, NHC(0)NR2R3, CH=CR2R3, CH200(0)NR2R3, CH2NHC(0)R2, CH2NHC(0)0R2, CH2CHC(0)NR2R3 and CH200(0)R2 ; and R2 and R3 are independently selected from H, C1_6alkyl, C2_6alkenyl, C3_ iocycloalkyl and C6_10aryl said latter four groups being unsubstituted or substituted with one to 5 groups independently selected from halo, OH, OCi-aalkyl, OC(0)C1_6alkyl and nitro, with the exception that R2 is not H when R1 is C(0)R2; and R4 is selected from H and OH, comprising:
(i) reacting a compound of the Formula II, wherein R4 is selected from H and 0Pg and each Pg may be the same or different and represent suitable protecting groups or any two adjacent Pg are joined to form a suitable cyclic protecting group under conditions to convert the aldehyde moiety to a group, other than C(0)H, selected from C(0)0R2, C(0)R2, C(0)NR2R3, CH=NR2, CH2NR2R3, CH2OR2, CH2R2, NR2R3, NHC(0)R2, NHC(0)0R2, NHC(0)NR2R3, CH=CR2R3,
7 CH200(0)NR2R3, CH2NHC(0)R2, CH2NHC(0)0R2, CH2CHC(0)NR2R3 and CH200(0)R2, wherein R2 and R3 are as defined above to form a compound of the Formula X wherein R1, R4 and each Pg are as defined above:
0Pg 0Pg oF
H(0)C di 0Pg R1 -g 0 40 v õ.g __ i,.
a i. 401 7 0Pg < ov 0 NPg 0 NPg II X .
, (ii) removing the Pg groups to form a compound of the Formula I wherein R1 and R4 are as defined above:
0Pg OH
R1 ib 0Pg R1 ib OH
si 7 ,. ____ vi.
<0 org 0 NH OH

0 .1Pg 0 X I .
Other features and advantages of the present application will become apparent from the following detailed description. It should be understood, however, that the scope of the claims should not be limited by the preferred embodiments and examples, but should be given the broadest interpretation consistent with the description as a whole.
8 BRIEF DESCRIPTION OF THE DRAWINGS
The application will now be described in greater detail with reference to the drawings in which:
Figure 1 is a scheme showing a process for the preparation of C-1 substituted analogues of 7-deoxypancratistatin analogues according to an embodiment of the present application;
Figure 2 is a graph showing the activity of the compounds 1(a), 1(c) and 1(d) in Jurkat cells.
Figure 3 is a graph showing the activity of the compound 1(b) at two concentrations (0.5 uM and 1.0 uM) in Jurkat cells (human leukemia cells).
DETAILED DESCRIPTION
(I) DEFINITIONS
The term "Ci_salkyl" as used herein means straight and/or branched chain, saturated alkyl groups containing from one to six carbon atoms and includes methyl, ethyl, propyl, isopropyl, n-butyl, s-butyl, isobutyl, t-butyl, 2,2-dimethylbutyl, n-pentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, n-hexyl and the like.
The term "C2_6alkenyl" as used herein means straight and/or branched chain, unsaturated alkyl groups containing from two to six carbon atoms and one to three double bonds, and includes (depending on the identity of n) vinyl, allyl, 2-methylprop-1-enyl, but-1-enyl, but-2-enyl, but-3-enyl, 2-methylbut-1-enyl, 2-methylpent-1-enyl, 4-methylpent-1-enyl, 4-methylpent-2-enyl, 2-methylpent-2-enyl, 4-methylpenta-1,3-dienyl, hexen-1-y1 and the like.
The term "C3_10cycloalkyl" as used herein a saturated carbocylic group containing from three to 10 carbon atoms and one or more rings and includes cyclopropyl, cyclobutyl, cyclopentyl, cyclodecyl, bicyclo[2.2.21octane, bicyclo[3.1.1]heptane, octahydro-1H-indene and the like.
9 The term "halo" as used herein refers to a halogen atom and includes F, Cl, Br and I.
In some cases the chemistries outlined herein may have to be modified, for instance by use of protecting groups, to prevent side reactions of reactive groups attached as substituents. This may be achieved by means of conventional protecting groups, for example as described in "Protective Groups in Organic Chemistry" McOmie, J.F.W. Ed., Plenum Press, 1973 and in Greene, T.W. and Wuts, P.G.M., "Protective Groups in Organic Synthesis", John Wiley & Sons, 3rd Edition, 1999.
The terms "protective group" or "protecting group" or "Pg" or the like as used herein refer to a chemical moiety which protects or masks a reactive portion of a molecule to prevent side reactions in those reactive portions of the molecule, while manipulating or reacting a different portion of the molecule. After the manipulation or reaction is complete, the protection group is removed under conditions that do not destroy or decompose the molecule. Many conventional protecting groups are known in the art for example as described in "Protective Groups in Organic Chemistry" McOmie, J.F.W. Ed., Plenum Press, 1973 and in Greene, T.W. and Wuts, P.G.M., "Protective Groups in Organic Synthesis", John Wiley & Sons, 3rd Edition, 1999. These may include but are not limited to Boc, Ts, Ms, TBDMS, TBDPS, Tf, Bn, allyl, Fmoc, C116acyl, silyl, acetal and the like.
The term "leaving group" as used herein refers to a group that is readily displaceable by a nucleophile, for example, under nucleophilic substitution reaction conditions. Examples of suitable leaving groups include, halo, Ms, Ts, Ns, Tf, Bn, C16acy1, C1_16alkyl, alkylsulphonyl and the like.

The term "suitable", as in for example, "suitable protecting group", "suitable leaving group" or "suitable reaction conditions" means that the selection of the particular group or conditions would depend on the specific synthetic manipulation to be performed and the identity of the molecule but the selection 5 would be well within the skill of a person trained in the art.
Boc as used herein refers to the group t-butyloxycarbonyl.
Ac as used herein refers to the group acetal.
Ts (tosyl) as used herein refers to the group p-toluenesulfonyl
10 Ms as used herein refers to the group methanesulfonyl TBDMS as used herein refers to the group t-butyldimethylsilyl.
TBDPS as used herein refers to the group t-butyldiphenylsilyl.
Tf as used herein refers to the group trifluoromethanesulfonyl.
Ns as used herein refers to the group naphthalene sulphonyl.
In all of the compounds disclosed herein, that is compounds of the Formulae I-X, one or more, including all, of the hydrogen atoms may be replaced with F. A
person skilled in the art would appreciate that only those hydrogens available for substitution by fluorine would be replaceable by fluorine.
The term "compound(s) of the application" or "intermediate compounds" used herein means compound(s) of Formulae I and ll as defined above, or any other novel compounds or intermediates defined above, stereoisomers thereof or pharmaceutically acceptable salts, solvates or prodrugs thereof, including mixtures thereof.
The compounds of the application all have at least one asymmetric centre.
Where the compounds according to the application possess more than one asymmetric centre, they may exist as diastereomers. It is to be understood that
11 all such isomers and mixtures thereof in any proportion are encompassed within the scope of the present application. It is to be understood that while the stereochemistry of the compounds of the application may be as provided for in any given compound listed herein, such compounds of the application may also contain certain amounts (e.g. less than 20%, preferably less than 10%, more preferably less than 5%) of compounds of the application having alternate stereochemistry.
The term "pharmaceutically acceptable" means compatible with the treatment of animals, in particular, humans.
The term "pharmaceutically acceptable salt" means an acid addition salt or base addition salt, which is suitable for or compatible with the treatment of patients.
The term "pharmaceutically acceptable acid addition salt" as used herein means any non-toxic organic or inorganic salt of any base compound of the application, or any of its intermediates. Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric and phosphoric acids, as well as metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate. Illustrative organic acids that form suitable salts include mono, di-, and tricarboxylic acids such as glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, benzoic, phenylacetic, cinnamic and salicylic acids, as well as sulfonic acids such as p-toluene sulfonic and methanesulfonic acids. Either the mono or di-acid salts can be formed, and such salts may exist in either a hydrated, solvated or substantially anhydrous form. In general, the acid addition salts of the compounds of the application are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms. The selection of the appropriate salt will be known to one skilled in the art.
Other
12 non-pharmaceutically acceptable salts, e.g. oxalates, may be used, for example, in the isolation of the compounds of the application, for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt. In embodiments of the application, the pharmaceutically acceptable acid addition salt is the hydrochloride salt, or the H3PO4 salt. The formation of a desired compound salt is achieved using standard techniques. For example, the neutral compound is treated with an acid or base in a suitable solvent and the formed salt is isolated by filtration, extraction or any other suitable method.
The term "pharmaceutically acceptable basic addition salt" as used herein means any non-toxic organic or inorganic base addition salt of any acid compound of the application, or any of its intermediates. Acidic compounds of the invention that may form a basic addition salt include, for example, where the group of R1 has an acidic hydrogen, for example when R1 is C(0)0H. Illustrative inorganic bases which form suitable salts include lithium, sodium, potassium, calcium, magnesium or barium hydroxide. Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as methylamine, trimethylamine and picoline or ammonia. The selection of the appropriate salt will be known to a person skilled in the art. Other non-pharmaceutically acceptable basic addition salts, may be used, for example, in the isolation of the compounds of the invention, for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
The term "solvate" as used herein means a compound of the application or a pharmaceutically acceptable salt of a compound of the application, wherein molecules of a suitable solvent are incorporated in the crystal lattice. A
suitable solvent is physiologically tolerable at the dosage administered. Examples of suitable solvents are ethanol, water and the like. When water is the solvent, the molecule is referred to as a "hydrate". The formation of solvates of the
13 compounds of the application will vary depending on the compound and the solvate. In general, solvates are formed by dissolving the compound in the appropriate solvent and isolating the solvate by cooling or using an antisolvent.
The solvate is typically dried or azeotroped under ambient conditions.
Compounds of the application includes prodrugs. In general, such prodrugs will be functional derivatives of a compound of the application, which are readily convertible in vivo into the compound from which it is notionally derived.
Prodrugs of the compounds of the application may be conventional esters formed with available hydroxy, or amino groups. For example, an available OH or nitrogen in a compound of the application may be acylated using an activated acid in the presence of a base, and optionally, in inert solvent (e.g. an acid chloride in pyridine). Some common esters which have been utilized as prodrugs are phenyl esters, aliphatic (C5-C24) esters, acyloxymethyl esters, carbamates and amino acid esters. In certain instances, the prodrugs of the compounds of the application are those in which one or more of the hydroxy groups in the compounds is masked as groups which can be converted to hydroxy groups in vivo. Conventional procedures for the selection and preparation of suitable prodrugs are described, for example, in "Design of Prodrugs" ed. H. Bundgaard, Elsevier, 1985.
Compounds of the application includes radiolabeled forms, for example, compounds of the application labeled by incorporation within the structure 3H
or 140 or a radioactive halogen such as 1251. A radiolabeled compound of the application may be prepared using standard methods known in the art. For example, tritium may be incorporated into a compound of the formula I using standard techniques, for example by hydrogenation of a suitable precursor to a compound of the application using tritium gas and a catalyst. Alternatively, a compound of the formula I containing radioactive iodo may be prepared from the
14 corresponding trialkyltin (suitably trimethyltin) derivative using standard iodination conditions, such as [1251] sodium iodide in the presence of chloramine-T in a suitable solvent, such as dimethylformamide. The trialkyltin compound may be prepared from the corresponding non-radioactive halo, suitably iodo, compound using standard palladium-catalyzed stannylation conditions, for example hexamethylditin in the presence of tetrakis(triphenylphosphine) palladium (0) in an inert solvent, such as dioxane, and at elevated temperatures, suitably 50-100 C.
The term "subject" as used herein includes all members of the animal kingdom including human. The subject is preferably a human.
The term "cancer" as used herein refers to a class of diseases or disorders characterized by uncontrolled division of cells and the ability of these cells to invade other tissues, either by direct growth into adjacent tissue through invasion or by implantation into distant sites by metastasis. Metastasis is defined as the stage in which cancer cells are transported through the bloodstream or lymphatic system. Examples of cancer that may be treated using the compounds of the invention include but are not limited to, prostate cancer, colon cancer, breast cancer, bladder cancer, lung cancer, ovarian cancer, endometrial cancer renal cancer and pancreatic cancer.
The term a "therapeutically effective amount", "effective amount" or a "sufficient amount" of a compound of the present invention is a quantity sufficient to, when administered to the subject, including a mammal, for example a human, effect beneficial or desired results, including clinical results, and, as such, an "effective amount" or synonym thereto depends upon the context in which it is being applied. In the context of disease, therapeutically effective amounts of the compounds of the present invention are used to treat, modulate, attenuate, reverse, or affect a disease or conditions for example, cancer in a subject.
An "effective amount" is intended to mean that amount of a compound that is sufficient to treat, prevent or inhibit such diseases or conditions. The amount of a given compound of the present invention that will correspond to such an amount 5 will vary depending upon various factors, such as the given drug or compound, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art. Also, as used herein, a "therapeutically effective amount" of a compound of the present 10 invention is an amount which prevents, inhibits, suppresses or reduces a disease or conditions for example, cancer as determined by clinical symptoms or the amount of cancer cells, in a subject as compared to a control. As defined herein, a therapeutically effective amount of a compound of the present invention may be readily determined by one of ordinary skill by routine methods known in the
15 art.
In an embodiment, a therapeutically effective amount of a compound of the present invention ranges from about 0.1 to about 40 mg/kg body weight, suitably about 1 to about 10 mg/kg body weight, and more suitably, from about 2 to about 5 mg/kg body weight. The skilled artisan will appreciate that certain factors may influence the dosage required to effectively treat a subject, or prevent a subject, suffering from a disease or condition for example cancer, and these factors include, but are not limited to, the severity of the disease or disorder, previous treatments, the general health and/or age of the subject and other diseases present.
As used herein, and as well understood in the art, "treatment" is an approach for obtaining beneficial or desired results, including clinical results.
Beneficial or desired clinical results can include, but are not limited to, alleviation or
16 amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e. not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment.
Moreover, a "treatment" or "prevention" regime of a subject with a therapeutically effective amount of the compound of the present invention may consist of a single administration, or alternatively comprise a series of applications. For example, the compound of the present invention may be administered at least once a week. However, in another embodiment, the compound may be administered to the subject from about one time per week to about once daily for a given treatment. In yet another embodiment the compound may be administered more than once daily up to 5 times per day. The length of the treatment period depends on a variety of factors, such as the severity of the disease, the age of the patient, the concentration and the activity of the compounds of the present invention, or a combination thereof. It will also be appreciated that the effective dosage of the compound used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required.
As used herein, "administered contemporaneously" means that two substances are administered to a subject such that they are both biologically active in the subject at the same time. The exact details of the administration will depend on the pharmacokinetics of the two substances in the presence of each other, and can include administering one substance within 24 hours of administration of the
17 other, if the pharmacokinetics are suitable. Designs of suitable dosing regimens are routine for one skilled in the art. In particular embodiments, two substances will be administered substantially simultaneously, i.e. within minutes of each other, or in a single composition that comprises both substances.
"Palliating" a disease or disorder means that the extent and/or undesirable clinical manifestations of a disorder or a disease state are lessened and/or time course of the progression is slowed or lengthened, as compared to not treating the disorder.
The term "prevention" or "prophylaxis", or synonym thereto, as used herein refers to a reduction in the risk or probability of a patient becoming afflicted with cancer or manifesting a symptom associated with cancer.
To "inhibit" or "suppress" or "reduce" a function or activity, is to reduce the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another condition. The terms "inhibitor" and "inhibition", in the context of the present application, are intended to have a broad meaning and encompass compounds of the application which directly or indirectly (e.g., via reactive intermediates, metabolites and the like) act on cancer.
The term "a cell" as used herein includes a plurality of cells. Administering a compound to a cell includes in vivo, ex vivo and in vitro treatment.
In understanding the scope of the present disclosure, the term "comprising"
and its derivatives, as used herein, are intended to be open ended terms that specify the presence of the stated features, elements, components, groups, integers, and/or steps, but do not exclude the presence of other unstated features,
18 elements, components, groups, integers and/or steps. The foregoing also applies to words having similar meanings such as the terms, "including", "having" and their derivatives. Finally, terms of degree such as "substantially", "about"
and "approximately" as used herein mean a reasonable amount of deviation of the modified term such that the end result is not significantly changed. These terms of degree should be construed as including a deviation of at least 5% of the modified term if this deviation would not negate the meaning of the word it modifies.
(II) COMPOUNDS
The present application includes compounds Formula I:
OH
OH

tC1H

wherein R1 is selected from C(0)0R2, C(0)R2, C(0)NR2R3, CH=NR2, CH2NR2R3, CH2OR2, CH2R2, NR2R3, NHC(0)R2, NHC(0)0R2, NHC(0)NR2R3, CH=CR2R3, CH200(0)NR2R3, CH2NHC(0)R2, CH2NHC(0)0R2, CH2CHC(0)NR2R3 and CH20C(0)R2; and R2 and R3 are independently selected from H, C1_6a1ky1, C2_6alkenyl, C3.
iocycloalkyl and C6_10aryl said latter four groups being unsubstituted or substituted with one to 5 groups independently selected from halo, OH, OCiAalkyl, OC(0)Ci_6alkyl and nitro; and R4 is selected from H and OH;
19 or a pharmaceutically acceptable salt, solvate or prodrug thereof.
In an embodiment of the application R1 is selected from C(0)0H, C(0)0Me, C(0)H, CH=NH and CH2NH2, CH2OH and CH200(0)CH3. In another embodiment of the application R4 is H.
(III) COMPOSITIONS AND USES/METHODS
As hereinbefore mentioned, novel compounds of the Formula I, and intermediates of the Formula II have been prepared. Accordingly, the present application includes all uses of the compounds of Formula I and the intermediates of Formula II including their use in therapeutic methods and compositions for treatment of cancer, their use in diagnostic assays and their use as research tools. In particular, the present application includes the use of a compound of Formula I as a medicament.
Another aspect of the application is a use of a compound of the application for treating cancer.
Another aspect of the application is a use of a compound of the application for the preparation of a medicament for treating cancer.
Another aspect of the application is a compound of the disclosure for use to treat cancer.
Also within the scope of the present application is a method of treating cancer comprising administering an effective amount of a compound of the application to a subject in need thereof.

The compounds of the application are suitably formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo. Accordingly, the present application further includes a pharmaceutical composition comprising a compound of the 5 application and a pharmaceutically acceptable carrier and/or diluent.
The compositions containing the compounds of the application can be prepared by known methods for the preparation of pharmaceutically acceptable compositions, which can be administered to subjects, such that an effective 10 quantity of the active substance is combined in a mixture with a pharmaceutically acceptable vehicle. Suitable vehicles are described, for example, in Remington's Pharmaceutical Sciences (2000 - 20th edition) and in The United States Pharmacopeia: The National Formulary (USP 24 NF19) published in 1999). On this basis, the compositions include, albeit not exclusively, solutions of the 15 substances in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered solutions with a suitable pH
and iso-osmotic with the physiological fluids.
The compounds of the application may be used in the form of the free base, in
20 the form of salts and/or solvates. All forms are within the scope of the application.
In accordance with the methods of the application, the described compounds, salts or solvates thereof may be administered to a patient in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art. The compositions of the application may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump or transdermal (topical) administration and the pharmaceutical compositions formulated accordingly.
Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal,
21 intrapulmonary, intrathecal, rectal and topical modes of administration.
Parenteral administration may be by continuous infusion over a selected period of time.
A compound of the application may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet. For oral therapeutic administration, the compound of the application may be incorporated with excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
A compound of the application may also be administered parenterally. Solutions of a compound of the application can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, DMSO and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. A

person skilled in the art would know how to prepare suitable formulations.
Conventional procedures and ingredients for the selection and preparation of suitable formulations are described, for example, in Remington's Pharmaceutical Sciences (2000 - 20th edition) and in The United States Pharmacopeia: The National Formulary (USP 24 NF19) published in 1999.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersion and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. Ampoules are convenient unit dosages.
22 Compositions for nasal administration may conveniently be formulated as aerosols, drops, gels and powders. Aerosol formulations typically comprise a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomizing device. Alternatively, the sealed container may be a unitary dispensing device such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve, which is intended for disposal after use. Where the dosage form comprises an aerosol dispenser, it will contain a propellant, which can be a compressed gas such as compressed air or an organic propellant such as fluorochlorohydrocarbon. The aerosol dosage forms can also take the form of a pump-atomizer.
Compositions suitable for buccal or sublingual administration include tablets, lozenges, and pastilles, wherein the active ingredient is formulated with a carrier such as sugar, acacia, tragacanth, or gelatin and glycerine. Compositions for rectal administration are conveniently in the form of suppositories containing a conventional suppository base such as cocoa butter.
Compositions for topical administration may include, for example, propylene glycol, isopropyl alcohol, mineral oil and glycerin. Preparations suitable for topical administration include liquid or semi-liquid preparations such as liniments, lotions, applicants, oil-in-water or water-in-oil emulsions such as creams, ointments or pastes; or solutions or suspensions such as drops. In addition to the aforementioned ingredients, the topical preparations may include one or more additional ingredients such as diluents, buffers, flavouring agents, binders, surface active agents, thickeners, lubricants, preservatives, e.g. methyl hydroxybenzoate (including anti-oxidants), emulsifying agents and the like.
23 Sustained or direct release compositions can be formulated, e.g. liposomes or those wherein the active compound is protected with differentially degradable coatings, such as by microencapsulation, multiple coatings, etc. It is also possible to freeze-dry the compounds of the formula I and use the lypolizates obtained, for example, for the preparation of products for injection.
The dosage administered will vary depending on the use and known factors such as the pharmacodynamic characteristics of the particular substance, and its mode and route of administration; age, health, and weight of the individual recipient; nature and extent of symptoms, kind of concurrent treatment, frequency of treatment, and the effect desired.
(IV) PROCESSES
The present application also includes a process for preparing a compound of Formula II
0Pg H(0)C 0Pg ut-'9 NPg wherein R4 is selected from H and 0Pg and each Pg may be the same or different and represent suitable protecting groups or any two adjacent Pg are joined to form a suitable cyclic protecting group;
the process comprising:
24 (i) reacting a compound of the Formula III with an aluminum acetylide derived from a compound of the Formula IV, followed by protection to form a compound of the Formula V, wherein R4 and each Pg is as defined above:
<0 0,, r --'71."'OPg 0 el 0Pg \r,..0Pg + <00 _________________________________________ IP-0 R4 0 0Pg -Pgr\i''' R4 0Pg PgN' III IV V =
, (ii) reducing the compound of Formula V to form a cis-alkene of the Formula VI, wherein R4 and each Pg is as defined above:
<o0 40 0Pg 0Pg 0Pg--_ .0::
N' 0 Rg PgN''' \-0 V VI =
, (iii) reacting the compound of the Formula VI under solid-state, silica gel catalysis conditions to form a compound of the Formula VII, wherein R4 and each Pg is as defined above:
0Pg 0Pg -- di 0P9 righdh 0P9 0Pg R4 APIM P 0Pg VP N Pg \-0 VI VII =
, (iv) oxidatively cleaving the double bond in the compound of the Formula VII
to form an intermediate diketone of the Formula VIII which cyclizes to form a compound of the Formula IX, wherein R4 and each Pg is as defined above:
0Pg 0 0Pg 0Pg 0Pg / - 0Pg H(0)C opg ilh R4 An" 0Pg R4 le WI
0Pg 0Pg NPg NPg 0 NPg VII VIII IX

(v) oxidizing the compound of the Formula IX to form a compound of Formula II, wherein R4 and each Pg is as defined above:
0Pg 0Pg OHC opg H(0)C 0Pg 0Pg 0 0Pg 0 NPg NPg In an embodiment of the application, the Pg group on the aziridine nitrogen in the 15 compound of Formula III is Ts. In another embodiment, the Pg groups on the oxygen atoms of the compound of Formula III are linked to form a cyclic acetal group in particular dimethyl acetal. In a further embodiment the protecting group (Pg) added in step (i) to the oxygen following ring opening of the epoxide is t-butyldimethylsilyl(TBDMS).
According to a specific embodiment, the process of the present disclosure is directed to the synthesis of a compound according to Formula II using the reaction conditions shown in Figure 1. According to this embodiment the aluminum acetylide of the compound of Formula IV described in (i) above is formed by the addition of nBuLi and dimethylaluminum chloride in toluene at reduced temperature, for example about -78 C. Reaction of the compound of Formula III with the aluminum acetylide of the compound of Formula IV is followed by protection of the hydroxyl product of the ring opening. In a particular embodiment the hydroxyl is protected by reaction with TBDMSOTf in the presence of base, for example Et3N.
In another embodiment of the process, in (ii), reduction of the alkyne to the cis alkene is achieved using BH3 in a suitable solvent at reduced temperature, for example about 0 C.
In another embodiment of the process, in (iii), the R2-OH ring opening of the compound of the Formula IV is mediated by copper trifluoromethanesulfonate (Cu (01-02).
In another embodiment of the process, in (iii), the cis alkene compound of Formula VI is adsorbed onto silica gel and heated without solvent. In a particular embodiment the reaction mixture is heated to 120 C for approximately 24 hours to provide a compound of Formula VII.
In a further embodiment of the process, in (iv), the compound of Formula VII
is converted to the intermediate VIII by ozonolysis under suitable reaction conditions, for example in Me2S and Me0H at reduced temperature, for example at about -78 C. In another embodiment, in (iv), the intermediate of Formula VIII is formed by a two step process comprising oxidation with osmium tetroxide (0s04) in the presence of N-methylmorpholine N-oxide (NMO), in CH2Cl2 to give a keto alcohol intermediate of the Formula XI, followed by reduction, for example with sodium borohydride, then periodate cleavage under suitable conditions to give the intermediate of Formula VIII.
OH 0Pg 0 dim& 0Pg R4 IRPW 0Pg NPg XI
In another embodiment of the process, in (v), oxidation of the compound of Formula IX to the compound of Formula ll is carried out using 2-iodoxybenzoic acid (IBX) under suitable conditions.
Compounds of the Formulae III and IV are prepared using methods known in the art [Schilling et al. Can J. Chem. 79:1659 (2001); Endoma, et al. Org. Process Res. Dev. 6:525 (2002)]

In another aspect of the application there is include a process for preparing a compound of Formula I
OH

wherein R1 is selected from C(0)0R2, C(0)R2, C(0)NR2R3, CH=NR2, CH2NR2R3, CH2OR2, CH2R2, NR2R3, NHC(0)R2, NHC(0)0R2, NHC(0)NR2R3, CH=CR2R3, CH200(0)NR2R3, CH2NHC(0)R2, CH2NHC(0)0R2, CH2CHC(0)NR2R3 and CH200(0)R2 ; and R2 and R3 are independently selected from H, C1_6alkyl, C2_6alkenyl, C3_ iocycloalkyl and C6_10aryl said latter four groups being unsubstituted or substituted with one to 5 groups independently selected from halo, OH, OCi-4alkyl, OC(0)C1_6alkyl and nitro, with the exception that R2 is not H when R1 is C(0)R2; and R4 isselected from H and OH, comprising:
(i) reacting a compound of the Formula II, wherein R4 is selected from H and 0Pg and each Pg may be the same or different and represent suitable protecting groups or any two adjacent Pg are joined to form a suitable cyclic protecting group under conditions to convert the aldehyde moiety to a group, other than C(0)H, selected from C(0)0R2, C(0)R2, C(0)NR2R3, CH=NR2, CH2NR2R3, CH2OR2, CH2R2, NR2R3, NHC(0)R2, NHC(0)0R2, NHC(0)NR2R3, CH=CR2R3, CH200(0)NR2R3, CH2NHC(0)R2, CH2NHC(0)0R2, CH2CHC(0)NR2R3 and CH200(0)R2, wherein R2 and R3 are as defined above to form a compound of the Formula X wherein R1, R4 and each Pg are as defined above:
0Pg 0Pg H(0)C 0Pg opg or r, g 0Pg 0 NPg 0 NPg II X
(ii) removing the Pg groups to form a compound of the Formula I wherein R1 and R4 are as defined above:
0Pg OH
R1 di OH

01-9 40 r"`
g <0 40 7 OH
0 NPg 0 NH

X
In another aspect of the application there is provided a process for preparing a compound of Formula I wherein R1 is COOH, wherein in (i) above the conditions comprise oxidizing the compound of Formula ll to form, after removal of the Pg groups in (ii), the compound of the Formula I. In a particular embodiment the conditions in step (i) comprise mCPBA Na2HPO4 in a suitable solvent at elevated temperature, for example, 40 C.
In another aspect of the application there is provided a process for preparing a compound of Formula I wherein R1 is C(0)0R2, wherein in (i) above the conditions comprise oxidizing the compound of the Formula II followed by alkylation with a compound of the Formula R2-LG, wherein LG is a suitable leaving group, to form, after removal of the Pg groups in (ii), the compound of the Formula I.

In another aspect of the application there is provided a process for preparing a compound of Formula I wherein R1 is CH=NR2 wherein in (i) above the conditions comprise reacting the compound of the Formula II with an amine of the Formula R2-NH2 to form, after removal of the Pg groups in (ii), the compound 10 of the Formula I.
In another aspect of the application there is provided a process for preparing a compound of Formula I wherein R1 is CH2NR2R3 wherein in step (i) above the conditions comprise reacting the compound of the Formula II with an amine, 15 followed by reduction and optional alkylation with a compound of the Formula R3-LG, wherein LG is a suitable leaving group, to form, after removal of the Pg groups in (ii), the compound of the Formula I.
In another aspect of the application there is provided a process for preparing a 20 compound of Formula I wherein R1 is CH=CR2R3 wherein in step (i) above the conditions comprise reacting the compound of the Formula II with a phosphonium ylide of the formula R2R3CH=PPh3 under Wittig reaction conditions to form, after removal of the Pg groups in (ii), the compound of Formula I.
25 In another aspect of the application there is provided a process for preparing a compound of Formula I wherein R1 is C(0)NR2R3, wherein in (i) above the conditions comprise oxidizing the compound of Formula II as described above followed by reaction with an amine of the formula NHR2R3 under amide bond forming conditions to form, after removal of the Pg groups in (ii), the compound of the Formula I.
In another aspect of the application there is provided a process for preparing a compound of Formula I wherein R1 is NR2R3, wherein in (i) above the conditions comprise oxidizing the compound of Formula ll as described above followed by subjecting the C(0)0H group to Curtius rearrangement to form a compound of Formula X, wherein R1 is NH2. This amine can be alkylated with various R2 and/or R3 groups. Alternatively, the C(0)0H groups can be converted to an amide (CONH2) and the amide subjected to Hoffman degradation to produce, after hydrolysis C-1 amine. These methods are known to those skilled in the art.
Also included in the present application is a process for preparing a compound of Formula I
OH
R1 al OH

0 fC11-1 wherein R1 is C(0)H and R4 is H or OH, comprising removing the Pg groups from a compound of Formula ll as defined above to form the compound of the Formula I.
(V) EXAMPLES
The following non-limiting examples are illustrative of the present application:
Examples 1-10 refer to compounds as shown in Figure 1.

Example 1: Preparation of (1S,2R,3R,4R,5S,6R)-3,4-(lsopropylidenedioxy)-5-[(tert-butyldimethylsily0oxy]-6-2-Benzo[1.3]dioxol-5-ylethynyl-(4'-methylphenylsulfonyl)-7-azabicyclo[4.1.0]heptane V(a) <o0 Cc/

Tsl\r' V(a) To a solution of acetylene 4 (2.74 g, 18.75 mmol) in 18 mL dry toluene was added at -78 C 8.33 mL of a solution of nBuLi in hexanes (2.25 M, 18.75 mmol).
The solution was stirred for 10 minutes before 18.75 mL of a solution of Me2AICI
(1.0M in CH2Cl2, 18.75 mmol) was added dropwise. The reaction flask was allowed to warm to room temperature and stir for 1h. The reaction flask was then cooled to -20 C and 18 mL of a solution of epoxide 3 (3.16 g, 9.38 mmol) in toluene was added dropwise over 20 min. The reaction was stirred at -20 C for 3.5 h before being place in an ice bath and allowed to slowly warm to room temperature and stir for 12h. The reaction was cooled in an ice bath and quenched with 1 M HCI. Ethyl acetate (200 mL) was added and the layers where separated. The aqueous phase was extracted 3 x 100 mL Et0Ac and the combined organic layers dried over Na2SO4. Concentration under reduced pressure and purification by flash column chromatography (hexanes:ethyl acetate, 7:1 to 4:1) afforded alcohol intermediate which was immediately subjected to protection protocol (2.01 g, 44%); [a]22D -113.05 (c 0.5, CHCI3);
Rf 0.30 (hexanes:ethyl acetate 2:1); IR (film) v3491, 2988, 1163 cm-1; 1H NMR
(300 MHz, CDCI3) 6: 7.78 (d, J = 8.1 Hz, 2H), 7.38 (d, J = 8.1 Hz, 2H), 6.91 (dd, J
=
8.2 Hz, 1.8 Hz 1H), 6.83 (d, J = 1.5 Hz, 1H), 6.73 (d, J = 7.9 Hz, 1H), 5.97 (s, 2H), 4.47 (d, J = 6.4 Hz, 1H), 4.22 (dd, J = 6.1, 4.4 Hz, 1H), 3.98 (m, 1H), 3.40 (d, J = 6.4 Hz, 1H), 3.24 (m, 2H), 3.06 (d, J = 9.6 Hz, 1H), 2.47 (s, 3H), 1.49 (s, 3H), 1.32 (s, 3H) ppm; 13C NMR (75 MHz, CDCI3) 6: 148.1, 147.5, 145.7, 134.2, 130.4, 128.1, 126.4, 116.2, 111.8, 110.3, 108.6, 101.5, 84.2, 83.8, 75.4, 70.1, 68.7, 42.3, 40.5, 31.1, 27.4, 25.2, 21.9 ppm; HRMS (FAB M+) calcd for 025H25N07S 484.1430, found 484.1428.
Alcohol intermediate (240mg, 0.49 mmol) was dissolved in 5 mL of CH2Cl2 and triethylamine (0.14 mL, 1.04 mmol) was added. The reaction flask was cooled to -78 C and t-butyldimethylsilytriflate (0.12 mL, 0.546 mmol) was added dropwise to the stirring solution. After stirring for 30 minutes at -78 0 the reaction was quenched with water and the two phases separated. The aqueous phase was extracted with CH2012 (2 x 15 mL) and the combined organic solution was washed sequentially with 5% citric acid (2 mL) and brine (2 mL) before drying over sodium sulfate. The solvent was removed under reduced pressure and the residue was purified by flash column chromatography (hexane:ethyl acetate, 9:1 to 2:1) affording V(a) (0.276g, 93%) as a colorless oil.; [a]24D +57.7 (c 0.5, CHC13); Rf 0.49 (hexanes:ethyl acetate, 2:1); IR (film) v 2953, 2929, 2892, 2856, 1599,1490 cm-1; 1H NMR (300 MHz, CDCI3) 6: 7.83 (d, J = 8.1 Hz, 2H), 7.38 (d, J
= 8.1 Hz, 2H), 6.94 (d, J = 8.1 Hz, 1H), 6.84 (s, 1H), 6.77 (d, J = 8.1 Hz, 1H), 5.99 (s, 2H), 4.45 (d, J = 5.1 Hz, 1H), 3.83 (m, 2H), 3.26 (m, 2H), 2.84 (d, J
= 7.5 Hz), 2.47 (s, 3H), 1.52 (s, 3H), 1.35 (s, 3H), 0.87 (s, 9H), 0.11 (s, 6H) ppm;

NMR (75 MHz, CDC13) 6: 147.8, 147.3, 134.7, 129.8, 127.9, 126.1, 111.6, 109.7, 108.4, 101.3, 86.3, 83.5, 71.7, 43.2, 39.53, 34.58, 27.9, 25.8, 25.79, 25.7, 21.7, 18.12, -4.4, -4.7 ppm; HRMS-E1 Calcd for 0301-136NO7SSi: 540.1481; Found, 540.1487; Anal. calcd for 0311-139NO7SSi C, 62.28; H, 6.58; found C, 62.22; H, 6.73 Example 2: 1S,2R,3R,4R,5S,6R)-3,4-(lsopropylidenedioxy)-5-gtert-butyldimethylsily0oxyl-6-2-Benzo[1,3]dioxol-5-ylethenyl-(4'-methylphenylsulfonyl)-7-azabicyclo[4.1.0]heptane VI(a).

OTBS
OZX
TsNrµ

\ ¨0 VI(a) To a 1.0 M solution of BH3.THF complex (2.5 mL, 2.5 mmol) was added cyclohexene (0.484 mL, 4.77 mmol) at 0 C. After 10 minutes a heavy precipitate was formed. The reaction mixture was kept at 0 C for 1 h before acetylene derivative V(a) (0.356 mg, 0.596 mmol) in 4.5 mL of THF was added.
The reaction mixture was stirred at 0 C until total consumption of starting material (2 h, TLC) before being quenched with 1 mL HOAc. 60 mL Et0Ac were added and the reaction mixture was washed with saturated aq. NaHCO3 (2 x 15 mL), H20 (2 x 15 mL), and brine (10 mL) before drying over Na2SO4 The solvent was removed under reduced pressure and the residue was purified by flash column chromatography (hexanes:ethyl acetate, 8:1) affording 0.271 g of VI(a) (76%).; [a123D -26.14 (c 1.0, CHCI3; Rf 0.35 (hexanes:ethyl acetate, 4:1); IR
(film) v 2986, 2930, 2894, 2856, 1598,1489 cm-1; 1H NMR (300 MHz, CDCI3) 6: 7.78 (d, J = 8.1 Hz, 2H), 7.29 (d, J = 8.1 Hz, 2H), 6.65 (m, 3H), 6.51 (d, J = 11.7 Hz, 1H), 5.97 (s, 2H), 5.54 (t, J = 11.3 Hz, 1H), 4.43 (d, J = 6, 1H), 3.85 (t, J
= 6.3, 1H), 3.61 (t, J = 7.2 Hz), 3.18 (d, J = 6.6, 1H), 2.91 (m, 2H), 2.44 (s, 3H), 1.52 (s, 3H), 1.33 (s, 3H), 0.79 (s, 9H), 0.02 (s, 3H), -0.04 (s, 3H) ppm; 13C NMR (150 MHz, CDCI3) 6: 147.5, 146.6, 144.6, 134.7, 132.0, 130.3, 129.8, 129.7, 129.5, 128.5, 127.9, 122.5, 109.35, 109.0, 108.1, 100.9, 83.2, 78.0, 72.6, 71.8, 43.7, 39.9, 39.5, 30.1, 27.8, 25.8, 25.79. 25.75, 25.72, 25.51, 23.7, 21.7, 18.1, -4.3, -4.7 ppm; HRMS-El Calcd for C311-141NO7SSi: 599.2373; Found, 599.2376; Anal.
calcd for 031 FI41 NO7SSi 0, 62.28; H, 6.58; found 0,61.30; H, 6.63 Example 3: N-[(1R,2aS,4aS,5S,5aR,12bR)-5-(tert-Butyl-dimethyl-silanyloxy)-3,3-dimethy1-1,2a,4a,5,5a,12b-hexahydro-phenanthro[2,3-d][1,3]dioxo1-1-yl]4-methyl-benzenesulfonamide VII(a) OTBS
sip 0, NHTs VII(a) A flame-dried 25-mL flask was charged with olefin 13 (336 mg, 0.561 mmol) and 10 silica gel which has been activate by heating under vacuum at 120 C
overnight (1.5 g). The starting materials were suspended in 10 mL freshly distilled methylene chloride and the solvent removed under reduced pressure. The silica gel supporting the absorbed reactants was heated externally at 120 C under nitrogen atmosphere for 24 h, after which time the silica gel was loaded onto 15 flash silica gel column and eluted with hexanes:ethyl acetate, 8:1 ¨ 5:1 to give 175 mg (52%) of olefin VII(a) as a clear and colorless oil.; [a]23D -123.7 (c 1.0, CHCI3); Rf 0.35 (hexanes:ethyl acetate, 2:1); IR (film) v3268, 2929, 2887, 2857, 1598,1503, 1485 cm-1; 1H NMR (600 MHz, CDCI3) 5: 7.43 (d, J = 7 Hz, 2H), 7.13 (d, J = 7 Hz, 2H), 6.49 (s, 2H), 6.34 (d, J = 8 Hz, 1H), 5.95 (s, 1H), 5.86 (s, 1H), 20 5.76 (d, J = 8 Hz, 1H), 4.51 (d, J = 7 Hz, 1H), 4.28 (m, 1H), 4.11 (m, 1H), 3.99 (m, 1H), 3.79 (m, 1H), 2.82 (m, 1H), 2.62 (dd, J= 11.1 Hz, J= 5.4 Hz, 1H), 2.40 (s, 3H), 1.43 (s, 3H), 1.33 (s, 3H), 0.89 (s, 9H), 0.11 (s, 3H), 0.07 (s, 3H) ppm;
130 NMR (150 MHz, CDC13) 6: 146.7, 145.9, 142.1, 138.9, 128.9, 128.6, 127.7, 126.8, 126.3, 126.2, 110.4, 109.2, 107.0, 79.0, 78.3, 70.3, 54.1, 42.5, 41.5, 38.9, 25 27.8, 26.3, 25.7, 25.3, 22.7, 21.5, 18.0, -5.0, -5.0 ppm; HRMS-El Calcd for C311-141N07SSi: 599.2373; Found, 599.2370; Anal. calcd for C311-141NO7SSi C, 62.07; H, 6.89; found C, 62.16; H, 6.94 Example 4: N-[(1 R,2aS,4aS,5S,5aS,12bR)-5-(tert-Butyl-dimethyl-silanyloxy)-6-hydroxy-3,3-dimethy1-7-oxo-1,2a,4a,5,5a,6,7,12b-octahydro-phenanthro[2,3-d][1,3]dioxo1-1-yl[4-methyl-benzenesulfonamide Xl(a) OH OTBS
0 0\/

NHTs 0 , \-0 To a solution of olefin VII(a) (0.240 mg, 0.4 mmol) in methylene chloride (10 mL) was added 4-methylmorpholine N-oxide (58 mg, 0.48 mmol). The reaction mixture was allowed to stir for 10 minutes before the introduction of a single crystal of osmium tetroxide and two drops of water. The reaction was stirred until total consumption of starting material (10 h) before being quenched with a saturated solution of saturated sodium bisulfite (6 mL). The two layers were separated and the aqueous phase was extracted with ethyl acetate (3 x 30 mL).
The organic phase was dried over sodium sulfate, filtered, and concentrated in vacuo to provide hydroxyketone Xl(a) as a white crystalline solid (0.227 g, 89%) that was used without further purification; Rf 0.42 (hexanes:ethyl acetate, 1:1);
mp >200 C; IR (film) v3478, 3263, 2929, 2857, 1670, 1614, 1504, 1482, 1444, 1386, 1330, 1252, 1218, 1156, 1075, 1039 cm-1; 1H NMR (600 MHz, CDCI3) 6:
7.54 (d, J= 7.8 Hz, 2H), 7.49, (s, 1H), 7.18 (d, J= 7.8 Hz, 2H), 6.70 (s, 1H), 6.07 (s, 1H), 6.00 (s, 1H), 4.79 (d, J = 8.7 Hz, 1H), 4.71 (m, 2H), 4.19 (m, 1H), 4.08 (m, 1H), 3.74 (m, 2H), 3.08 (dd, J = 10.2 Hz, J = 1.8 Hz, 1H), 2.45 (m, 1H), 2.41 (s, 3H), 1.36 (s, 3H), 1.31 (s, 3H), 0.87 (s, 9H), 0.12 (s, 3H), 0.07 (s, 3H) ppm;
130 NMR (150 MHz, CDCI3) 6: 196.6, 152.5, 147.9, 142.6, 140.5, 138.9, 129.1, 126.9, 124.7, 111.2, 109.6, 106.9, 102.1, 78.9, 78.7, 70.3, 65.9, 57.9, 49.4, 39.7, 27.9, 25.7, 21.5, 17.95, -5.1 ppm; HRMS-El Calcd for C27H32NO9SSi (M+-57):
574.1567, Found: 574.1572 Example 5: (3aS,3bR,10bR,11 R,12S,12aS)-12-(tert-Butyl-dimethyl-silanyloxy)-2, 2-dimethy1-5-oxo-4-(toluene-4-sulfony1)-3a,3b, 4,5,10b,11,12, 12a-octahydro-1, 3,7,9-tetraoxa-4-aza-dicyclopenta[a, h]phenanthrene-11-carbaldehyde II(a).

H V
(0 010 NIPIF \
0 N, Ts II(a) To a 10 mL round bottomed flask was added hydroxyl ketone Xl(a) (0.4 g, 0.63 mmol) and 6 mL of a 1:1 mixture of ethanol:dioxane. The reaction flask was cooled externally in an ice bath and NaBH4 (24 mg, 0.63 mmol) was added in one portion. The reaction was removed from the bath and allowed to warm to room temperature over 1 h. The reaction was quenched with 1 N HCI (4 mL) and separated. The aqueous phase was extracted with CH2Cl2 (3 x 20 mL) and the organic phase combined before drying over sodium sulfate. The crude mixture was concentrated in a 25 mL round bottomed flash and taken up in dioxane (8 mL). A stirring bar was added and the reaction was stirred while sodium periodate (0.332, 1.5 mmol) was added. The flask was covered to exclude light and H20 (15 drops) added. The reaction was stirred until total consumption of starting material (23h) as monitored by TLC. The reaction was quenched with H20 (10 mL) and separated. The aqueous phase was extracted with CH2Cl2 (3 x 50 mL) and the combined organic phases dried over sodium sulfate.
Concentration provided IX(a).

To a solution of hemi-aminal IX(a) (394 mg, 0.62 mmol) in N,N-Dimethylformamide (3 mL) was added 2-lodoxybenzoic acid (520 mg, 1.86 mmol). After total consumption of starting material (by TLC), the reaction mixture was diluted with diethyl ether (200 mL) and washed sequentially with saturated aqueous sodium bisulfite (10 mL), sodium bicarbonate (3 x 10 mL), H20 (10 x 1mL), and brine (10 mL). The organic phase was dried over magnesium sulfate, filtered and concentrated. The final product was isolated by column chromatography (hexanes:ethyl acetate, 4:1). Yield: 225 mg, 61%, white solid;
Rf 0.31 (hexanes:ethyl acetate, 4:1); mp >200 C, recrystallized from hexanes/ethyl acetate 4:1; [a]D21 + 31.67 (c 0.5, CHC13); IR (film) v 2929, 2857, 1725, 1689, 1619, 1505, 1484, 1386, 1361, 1287, 1255, 1220, 1172, 1077, 1036 cm-1; 1H NMR (600 MHz, CDCI3) 6: 9.49 (s, 1H), 8.3 (d, J = 8.2 Hz, 2H), 7.58 (s, 1H), 7.33 (d, J = 8.2 Hz, 2H), 7.28 (s, 1H), 6.55 (s, 1H), 6.04 (d, J = 5 Hz, 2H), 5.81 (dd, J = 8.4 Hz, J = 5.2 Hz, 1H), 4.79 (m, 1H), 4.50 (dd, J = 12.7 Hz, J
= 8.4 Hz, 1H), 4.27 (dd, J = 5.2 Hz, J = 2.7 Hz, 1H), 3.83 (dd, J = 12.6, J = 4.0 Hz, 1H), 3.31 (m, 1H), 2.45 (s, 3H), 1.42 (s, 3H), 1.32 (s, 1H), 0.99 (s, 9H), 0.26 (s, 3H), 0.25 (s, 3H) ppm; 130 NMR (150 MHz, CD013) 6: 196.2, 166.0, 153.0, 147.1, 143.9, 138.8, 137.0, 128.9, 128.8, 110.1, 109.4, 104.2, 102.2, 72.4, 66.6, 65.5, 55.6, 35.4, 31.0, 27.9, 26.9, 25.7, 22.7, 21.7, 18.1, 14.2, -4.7, -4.9 ppm;
HRMS-El Calcd for C301-136NO9SSi (M+-15): 614.1879, Found: 614.1870; Anal. calcd for C31 F139NO9SS C, 59.12; H, 6.24; found C, 59.31; H, 6.29.
Example 6: (3aS,3bR,10bR,11R,12S,12aS)-12-(tert-Butyl-dimethyl-silanyloxy)-2,2-dimethy1-5-oxo-4-(toluene-4-sulfony1)-3a,3b,4,5,10b,11,12,12a-octahydro-1,3,7,9-tetraoxa-4-aza-dicyclopenta[a,h]phenanthrene-11-carboxylic acid X(a) HO V
<0 N'Ts WI

X(a) To a solution of aldehyde IX(a) (144 mg, 0.229 mmol) in dry methylene chloride (5 mL) was added sodium phosphate dibasic (81 mg, 0.57 mmol). The suspension was stirred while 3-chloroperbenzoic acid (130 mg, 0.57 mmol) was added. The reaction flask was sealed and heated at 40 C overnight. The reaction mixture was diluted with methylene chloride (80 mL) and washed sequentially with saturated aqueous sodium bisulfite (10 mL), sodium bicarbonate (10 mL), and dried over sodium sulfate. The organic phase was filtered and concentrated in vacuo to provide carboxylic acid X(a) as a white crystalline solid (0.125g, 85%) that was used without further purification; ;
Rf 0.1 (hexanes/ethyl acetate, 1:1); mp >200 00; [a]p22 -35.09 (c 1.25, CHC13); IR
(KBr) v 3246, 2930, 2891, 2857, 1710, 1688, 1619, 1505, 1484, 1361, 1240, 1220, 1172, 1078, 1033 cm-1; 1H NMR (300 MHz, CDCI3) 6: 8.29 (d, J = 8.3 Hz, 2H), 7.53 (s, 1H), 7.32 (d, J = 8.3 Hz, 2H), 7.28 (s, 1H), 6.56 (s, 1H), 6.02 (d, J
= 3 Hz, 2H), 5.77 (dd, J = 8.30 Hz, J = 5.3 Hz, 1H), 4.85 (dd, J = 12.5 Hz, J =
8.4 Hz, 1H)), 4.84 (t, J = 4.7 Hz, 1H), 4.22 (dd J = 5.22, J = 2.8 Hz, 1H), 3.76 (dd, J =
12.4 Hz, J = 4.1 Hz, 1H), 3.38 (t, J = 3.5 Hz, 1H), 2.45 (s, 3H), 1.40 (s, 3H), 1.27 (s, 1H), 0.96 (s, 9H), 0.21 (s, 6H) ppm; 130 NMR (150 MHz, CDCI3) 6: 174.3, 166.2, 152.8, 146.9, 143.8, 138.9, 137.7, 129.0, 128.8, 122.4, 109.8, 109.2, 103.4, 102.1, 72.8, 68.2, 64.9, 48.0, 35.5, 27.4, 26.9, 25.7, 21.7, 18.0, -4.9, -5.0 ppm; HRMS-E1 Calcd for C271-130N010SSi (M+-57): 588.1359, Found: 588.1354;
Anal. calcd for 031H39NOloSSi C, 57.65; H, 6.09; found C, 58.01; H, 6.37 Example 7: (3aS, 3bR, 1 ObR,11 R,12S,12aS)-12-(tert-Butyl-d imethyl-silanyloxy)-2, 2-dimethy1-5-oxo-4-(toluene-4-sulfony1)-3a, 3b,4, 5,10b,11, 12, 12a-octahydro-1, 3,7, 9-tetraoxa-4-aza-dicyclopenta [a, h]phenanthrene-11-carboxylic acid methyl ester X(b) (0 0/\

,Ts WI

X(b) To a solution of carboxylic acid X(a) (45 mg, 0.069 mmol) in diethyl ether (3 mL) was added freshly prepared diazomethane solution in diethyl ether until the 10 persistence of yellow color and total consumption of starting material (by TLC).
The reaction was quenched with one drop of acetic acid followed by saturated sodium bicarbonate solution (1 mL), diluted with diethyl ether (30mL) and washed with saturated sodium bicarbonate solution (2 x 1 mL), dried over magnesium sulfate, filtered and concentrated. The crude reaction mixture was 15 passed through short silica plug using hexane/ethyl acetate 1:1 as eluent and concentrated to provide methyl ester X(b) that was used without further purification. Yield: 38mg, 83%, white crystalline solid; Rf 0.45 (hexanes:ethyl acetate, 1:1); mp >200 C; [a]D22 - 25.6809 (c 0.75, 0H013); IR (KBr) v 2986, 2953, 2931, 2896, 2858, 1739, 1692, 1620, 1598, 1505, 1485, 1361, 1289, 1264, 20 1173 cm-1; 1H NMR (300 MHz, CDCI3) 6: 8.30 (d, J = 8.4 Hz, 2H), 7.55 (s, 1H), 7.32 (d, J = 8.3 Hz, 2H), 6.58 (s, 1H), 6.02 (s, 2H), 5.78 (dd, J = 8.30 Hz, J
= 5.4 Hz, 1H), 4.9 (dd, J = 12.5 Hz, J = 8.3 Hz, 1H), 4.78 (t, J = 3.0 Hz, 1H), 4.24 (dd J
= 5.36 Hz, J = 2.9 Hz, 1H), 3.79 (dd, J = 12.4, J = 4.2 Hz, 1H), 3.56 (s, 3H), 3.40 (t, J = 3.7 Hz, 1H), 2.45 (s, 3H), 1.41 (s, 3H), 1.35 (s, 1H), 0.98 (s, 9H), 0.24 (s, 25 3H), 0.23 (s, 3H) ppm; 13C NMR (75 MHz, CDCI3) 6:169.4, 166.3, 152.8, 146.8, 143.7, 139.0, 138.2, 128.9, 128.8, 122.4, 109.8, 109.2, 103.5, 102.0, 72.9, 68.2, 65.2, 51.9, 48.1, 35.9, 27.5, 26.8, 25.7, 21.6, 18.0, -4.8, -4.9 ppm; HRMS-EI
Calcd for C28H32N010SS1 (M+-57): 602.1516, Found: 602.1516 Example 8: (3aS,3bR,10bR,11 R,12S,12aS)-12-(tert-Butyl-dimethyl-silanyloxy)-2,2-dimethy1-5-oxo-3a,3b,4,5,10b,11,12,12a-octahydro-1,3,7,9-tetraoxa-4-aza dicyclopenta[a,h]phenanthrene-11-carboxylic acid methyl ester X(c) Alk Ov (0 a/ \
NH

X(C) To a solution of X(b) (52 mg, 0.079 mmol) in dry THF (1 mL) at -50 C was added a 0.5 M solution of Na/naphthalene in DME until a green color persisted and total consumption of starting material was observed (by TLC). The solution was stirred for 10 minutes before the reaction was quenched with saturated aqueous ammonium chloride solution (1 mL). The reaction was warmed to room temperature and extracted with CH2Cl2 (6 x 15 mL). The combined organic phase was dried over sodium sulfate, filtered, and concentrated. The final product was isolated by column chromatography (hexanes:ethyl acetate, 5:1 to 2:1). Yield: 23 mg, 58%, clear oil; R10.28 (hexanes:ethyl acetate, 1:1);
[a]D22 ¨
14.51 (c 0.50, CHCI3); IR (film) v3320, 2952, 2930, 2895, 2857, 1743, 1669, 1619, 1504, 1484, 14601385, 1369, 1321, 1288, 1260, 1222 cm-1; 1H NMR (600 MHz, CDCI3) 6: 7.62 (s, 1H), 6.56 (s, 1H), 6.02 (s, 2H), 5.96 (s, 1H), 4.86 (t, J =
2.6, 1H), 4.41 (dd, J= 13.6 Hz, J= 8.2 Hz, 1H), 4.18 (dd, J= 8.25 Hz, J= 4.8 Hz, 1H), 4.11 (m, 1H), 3.66 (s, 3H), 3.40 (dd, J = 13.6 Hz, J = 3.7 Hz, 1H), 3.33 (m, 1H), 2.06 (s, 1H), 1.40 (s, 3H), 1.37 (s, 3H), 0.92 (s, 9H), 0.21 (s, 3H), 0.20 (s, 3H) ppm; 130 NMR (150 MHz, CDCI3) 6:169.6, 165.4, 151.4, 146.6, 135.4, 122.6, 110.5, 108.6, 103.3, 101.7, 69.2, 53.1, 51.9, 45.9, 33.4, 27.6, 26.5, 25.7, 17.9, -4.9, -5.0 ppm; HRMS-El Calcd for 025H35NO8Si (M+): 505.2132, Found:
505.2131 Example 9: (1 R, 2S, 3R,4 S, 4aR, 1 1 bR)-2, 3, 4-Trihydroxy-6-oxo-1 , 2, 3,4, 4a, 5,6,1 1 b-octahydro-f1,3]dioxolo[4,5-j]phenanthridine-1-carboxylic acid methyl ester 1(a) OH
(0 OH
NH

1(a) To a solution of the detosylated methyl ester X(c) (23 mg, 0.046 mmol) in methanol (2 mL) was added 3% HCI in methanol (0.5 mL). The reaction mixture was stirred until total consumption of starting material (3 days). The solvent was removed under reduced pressure and the residue was purified by flash column chromatography using a 30:1 to 20:1 gradient of methylene chloride:methanol as eluent to provide methyl ester 19 (11 mg, 69%) as a white crystalline solid.;
mp >200 C (methylene chloride: methanol); Rf 0.06 (methylene chloride/methanol, 20:1); [a]D22 + 24.53 (c 0.25,Me0H); IR (KBr) v3311, 2913, 1732, 1648, 1609, 1497, 1462, 1349, 1259, 1037 cm-1; 1H NMR (300 MHz, Me0D) 6: 7.33 (s, 1H), 6.59 (s, 1H), 5.93 (d, J = 3.7, 2H), 4.50 (t, J = 3.12, 1H), 4.21 (dd, J =
13.1 Hz, J
= 10.1 Hz, 1H), 3.86 (m, 1H), 3.79, (dd, J = 10.1, J = 3.0, 1H), 3.51 (s, 3H), 3.39 (m, 1H), 3.29 (dd, J = 13.1, J = 4.1, 1H) ppm; 130 NMR (75 MHz, Me0D) 6:
170.8, 166.4, 151.7, 146.4, 137.3, 121.7, 106.9, 103.7, 101.8, 72.2, 71.9, 70.9, 51.4, 50.6, 44.8, 35.4 ppm, HRMS-FAB: (m/z) (M + H): Calcd for 016F117N08:
352.1032, Found: 352.0941 Example 10: (1 R, 2S, 3R, 4S, 4aR, 11bR)-2, 3,4-Trihydroxy-6-oxo-1,2,3,4,4a,5,6,11b-octahydro-[1 ,3]dioxolo[4,5-llphenanthridine-1-carboxylic acid 1(b) HO gib OH
<0 OH

1(b) To a solution of 1(a) (6 mg, 0.017 mmol) in methanol (0.5 mL) was added LiOH
(1 mg, 1.5 mmol). The reaction mixture was heated at 45 C and stirred until total consumption of starting material (2 days) as monitored by TLC. The reaction was made slightly acidic with the addition of HCI (5 drops, 1M) and concentrated to provide acid 20 (5 mg, 95%) as a white crystalline solid.; mp >200 C; Rf 0.06 (methylene chloride:methanol, 4:1); IR (KBr) v 3412, 2920, 2115, 1641, 1505, 1471, 1409, 1462, 1363, 1267 cm-1; 1H NMR (300 MHz, Me0D) 6: 7.41 (s, 1H), 6.72 (s, 1H), 6.02 (d, J = 3.7, 2H), 4.64 (t, J = 3.12, 1H), 4.35 (dd, J =
13.1 Hz, J
= 10.1 Hz, 1H), 3.99 (m, 1H), 3.89, (dd, J = 10.1, J = 3.0, 1H), 3.45 (m, 1H), 3.38 (m, 1H) ppm; 130 NMR (75 MHz, Me0D) 6: 172.1, 166.4, 151.7, 146.4, 137.6, 121.7, 106.8, 103.8, 101.8, 72.4, 71.9, 71.1, 51.34, 45.03, 35.4 ppm.
Example 11: (3aS,3bR,10bR,11S,12S,12aS)-12-(tert-Butyl-dimethyl-silanyloxy)-11-hydroxymethy1-2,2-dimethy1-4-(toluene-4-sulfony1)-3b,4,10b,11,12,12a-hexahydro-3aH-1,3,7,9-tetraoxa-4-aza-dicyclopenta[a,h]phenanthren-5-one X(d) OH OTBS
Ov (0 o/N
0 Ts X(d) To a solution of II(a) (175 mg, 0.278 mmol) in Et0H/dioxane (1:1, 5 mL) at 0 C
was added NaBH4 (3 mg, 0.08 mmol). The reaction was allowed to warm to room temperature over 1.5 hours before being quenched with a solution of saturated NH4CI (1 mL). The Et0H/dioxane mixture was removed under reduced pressure and the aqueous residue was extracted with CH2Cl2 (3 x 25 mL). The organic phases were combined, dried over sodium sulfate, filtered, and concentrated to provide alcohol X(d) which was used without further purification. Yield: 150 mg, 85%, clear oil; ; Rf 0.44 (hexanes:ethyl acetate, 1:1); [a]D22 ¨ 47.72 (c 1.50, CHCI3); IR (film) v3547, 2986, 2932, 2586, 1692, 1616, 1594, 1508, 1481, 1360 cm-1; 1H NMR (300 MHz, CDCI3) 6: 8.28 (d, J = 8.3 Hz, 2H), 7.54 (s, 1H), 7.30 (d, J = 8.2 Hz, 2H), 6.77 (s, 1H), 6.04 (d, J = 1.6 Hz, 2H), 5.65 (dd, J = 8.8 Hz, J
= 5.6 Hz, 1H), 4.57 (d, J = 1.8 Hz, 1H), 4.32 (d, J = 4.6 Hz, 1H), 4.16 (dd J
=
12.8, J = 8.9 Hz, 1H), 3.78 (m 2H), 3.38 (dd, J = 11.3 Hz, J = 3.6 Hz, 1H), 2.55 (bs, 1H), 2.43 (s, 3H), 1.96 (bs, 1H), 1.43 (s, 3H), 1.35 (s, 3H), 0.96 (s, 9H), 0.20 (s, 6H) ppm; 13C NMR (75 MHz, 000I3) 6: 166.4, 153.1, 147.1, 143.7, 138.9, 137.0, 129.0, 128.7, 123.2, 109.1, 108.7, 104.9, 102.1, 73.1, 67.3, 64.8, 60.0, 46.9, 37.4, 28.1, 26.3, 25.8, 21.6, 18.0, -4.8, -4.9 ppm; HRMS-El Calcd for 031H41NO9SSi (M+-15): 616.2032, Found: 616.2032.
Example 12: Acetic acid (3aS, 3 bR,10bR,11 S, I 2S,12aS)-12-(tert-butyl-dimethyl-silanyloxy)-2, 2-d imethy1-5-oxo-4-(toluene-4-sulfony1)-3a, 3b, 4,5, 1 Ob, 11 , 12,12a-octahydro-1,3,7,9-tetraoxa-4-aza-dicyclopenta[a,h]phenanthren-11-ylmethyl ester X(e) OAc OTBS
Ov <0 0/\
0 Ts X(e) To a solution of X(d) (150 mg, 0.237 mmol) in dry CH2Cl2 (10 mL) was added 5 DMAP (1.5 mg, 0.012 mmol), followed by pyridine (0.1 mL, 1.187 mmol).
Ac20 (45 ,uL, 0.475 mmol) was added and the reaction was stirred for 1 hour before being quenched with saturated sodium bicarbonate (5 mL). The reaction was diluted with Et20 (75 mL) and separated. The aqueous layer was extracted with Et20 (2 x 75 mL) and the combined organic phases were washed with H20 (10 10 mL), brine (10 mL), dried over magnesium sulfate, filtered, and concentrated.
The final product was isolated by column chromatography using 5:1 mixture of hexanes: ethyl acetate as eluent. Yield: 128 mg, 81%, clear oil; Rf 0.51 (hexanes/ethyl acetate, 1:1); [a]p22 ¨ 41.081 (c 3.0, CHCI3); IR (film) v2988, 2952, 2930, 2858, 1742, 1694, 1619, 1598, 1505, 1485, 1395, 1362, 1254; 1H
15 NMR (600 MHz, CDCI3) 6: 8.29 (d, J = 8.3 Hz, 2H), 7.54 (s, 1H), 7.31 (d, J = 8.2 Hz, 2H), 6.84 (s, 1H), 6.03 (d, J = 12.6 Hz, 2H), 5.62 (dd, J = 8.7 Hz, J =
5.6 Hz, 1H), 4.50 (s, 1H), 4.31 (d, J= 5.3 Hz, 1H), 4.18 (t, J = 11.1 Hz, 1H), 3.97 (dd, J=
13.0 Hz, J = 8.8 Hz, 1H), 3.85 (dd, J = 11.0 Hz, J = 3.6 Hz, 1H), 3.80 (dd, J
=
13.0, J = 4.2, 1H), 2.7 (d, J = 5.2, 1H), 2.44 (s, 3H), 2.03 (s, 3H), 1.42 (s, 3H), 20 1.36 (s, 3H), 0.96 (s, 9H), 0.19 (s, 1H); 130 NMR (150 MHz, CDCI3) 6:170.7, 166.2, 153.2, 147.3, 143.8, 138.8, 136.2, 129.1, 128.7, 123.2, 108.9, 108.8, 105.0, 102.2, 78.4, 73.0, 66.3, 64.4, 60.8, 44.0, 37.0, 28.3, 26.2, 25.8, 25.78, 25.75, 25.6, 21.6, 20.8, 18.1, -4.8, -5.0; HRMS-El Calcd for 032H40NO10SSi (M-15): 658.2142, Found: 658.2152 Example 13: ((3aS,3bR,10bR,11 S,12S,12aR)-12-hydroxy-2,2-dimethy1-5-oxo-3a,3b,4, 5,10b,11,12,12a-octahydrobis[1, 3]dioxolo[4, 5-c:4', 5'-j]phenanthridin-11-yl)methyl acetate OH
OK
(OOH

To a solution of X(e) (137 mg, 0.203 mmol) in dry DME (5 mL) at -78 C was added a 0.5 M solution of Na/naphthalene in DME until a green color persisted and total consumption of starting material was observed (by TLC). The solution was stirred for 10 minutes before the reaction was quenched with saturated aqueous ammonium chloride solution (2 mL). The reaction was warmed to room temperature, concentrated to remove DME, and extracted with CH2Cl2 (3 x 40 mL). The combined organic phase was dried over sodium sulfate, filtered, and concentrated. The resulting crude acetate was taken up in THF (2.5 mL) and cooled to 000. TBAF (0.1 mL, 1M in THE) was added dropwise over 2 minutes.
The reaction was stirred until total consumption of starting material was observed (TLC) before the stirring bar was removed, silica (200 mg added), and the reaction concentrated to dryness. The final product was isolated by column chromatography using 1:1 mixture of hexanes: ethyl acetate as eluent. Yield:

mg, 74%, white solid; mp >200 C Rf 0.059 (hexanes/ethyl acetate, 1:1); [a]D22 ¨
38.301 (c 1.35, DMS0); IR (film) v3303, 2982, 2922, 2901, 2853, 1734, 1655, 1652, 1612, 1483, 1459, 1364, 1246, 1235, 1215; 1H NMR (300 MHz, DMSO) 6:
7.76 (s, 1H), 7.35 (s, 1H), 7.03 (s, 1H) 6.09 (d, J = 1.8, 2H), 5.48 (d, J =
4.2, 1H), 4.35, (s, 1H), 4.24(d, J= 5.3, 1H), 4.19 ¨ 4.10 (m, 3H), 3.46 (dd, J= 14.0 Hz, J=
8.2 Hz, 1H), 3.21 (dd, J = 13.9 Hz, J = 3.8 Hz, 1H), 2.80 (bs, 1H), 2.02 (s, 1H), 1.39 (s, 3H), 1.31 (s, 3H); 130 NMR (75 MHz, DMSO) 6:170.9, 163.9, 151.3, 146.7, 134.5, 124.2, 108.9, 107.6, 105.4, 102.2, 77.9, 77.2, 65.3, 61.2, 53.5, 34.7, 28.3, 26.4, 21.2; HRMS-E1 Calcd for 020H23N08 (Mt): 405.1424, Found:
405.1431 Example 14: ((1S,2S,3R,4S,4aR,11bR)-2 ,3,4-trihydroxy-6-oxo-1,2,3,4,4a ,5,6,11b-octahyd ro41,3]dioxolo[4,5-j]phenanthridin-1-yl)methyl acetate 1(c) )LO OH
Ai OH
(0 40/ OH
NH

1(c) To a solution of the acetate of Example 13 (21 mg, 0.052 mmol) in Me0H (1 mL) was added an HCI solution (3 % in Me0H, 3 mL). The reaction was stirred until total consumption of starting material as monitored by TLC (3 h) before being quenched to basic pH with saturated sodium bicarbonate solution. The crude reaction mixture was concentrated to dryness. The final product was isolated by column chromatography (methlene chloride: methanol, 5:1). Yield: 6 mg, 45%, white solid; mp >200 00; Rf 0.41 (methlene chloride: methanol, 5:1); [a]D22 97.32 (c 0.3, DMSO); 1H NMR (600 MHz, DMSO) 6:7.36 (s, 1H), 7.01 (s, 1H), 6.76, (s, 1H), 6.10, (s, 2H), 5.14, (bs, 3H), 4.38 (t, J = 10.7 Hz, 1H), 4.15 ¨ 4.10 (m, 2H), 3.84 (s, 1H), 3.70 (dd J = 9.8 Hz, J = 2.9 Hz, 1H), 3.50 (dd J = 13.2 Hz, J =
9.9 Hz, 1H), 3.27 (dd J = 13.3 Hz, J = 4.0 Hz, 1H), 2.69 (bs, 1H), 2.03 (s, 3H) ppm;
130 NMR (150 MHz, DMSO) 6: 171.0, 164.1, 151.3, 146.6, 135.3, 123.8, 107.5, 105.5, 102.2, 73.1, 71.3, 69.1, 61.9, 51.6, 36.9, 21.3 ppm; HRMS-FAB Calcd for 017H20N08 (M + 1): 366.0988, Found: 366.1088.
Example 15: (1 S,2S,3R,4S,4aR,1 1 b R)-2,3,4-trihydroxy-1-(hydroxymethyl)-1 ,2,3,4,4a,5-hexahydro-[1 ,3]dioxolo[4,5-ilphenanthridin-6(1 1 bH)-one 1(d) OH OH
OH
(0 Si OH
NH

1(d) To a solution of acetate 1(c) (25 mg, 0.062 mmol) at 0 C, in Me0H (5 mL) was added K2003 (40 mg, 0.62 mmol) and H20 (1 mL). The suspension was stirred until total consumption of starting material (TLC) before being quenched with HCI
(4 drops, 6N). The reaction was allowed to warm to room temperature and stir (4 h). The pH of the reaction was made basic with the addition of saturated sodium bicarbonate solution and the methanol removed under reduced pressure. The resulting aqueous phase was concentrated overnight on a freeze-dryer. The salts were triturated with Me0H (5 x 5 mL) and the Me0H washes collected and concentrated. The final product was isolated by column chromatography (methlene chloride: methanol, 5:1). Yield: 15 mg, 75%, white solid; mp >200 C;
Rf 0.20 (methlene chloride: methanol, 5:1); [a]D22 90.91 (c 0.25, DMSO); IR
(film) v3361, 2916, 1646, 1608, 1503, 1460, 1385, 1361, 1252; 1H NMR (600 MHz, DMSO) 6: 7.34 (s, 1H), 6.97 (s, 1H), 6.66, (s, 1H), 6.09, (d, J = 0.78, 2H), 5.04 ¨4.97, (m, 3H), 4.47 (dd J = 6.6 Hz, J = 3.8 Hz, 1H), 4.19 (s, 1H), 3.89 (q, J
= 7.86 Hz, 1H), 3.82 (s, 1H), 3.69 ¨ 3.64 (m, 1H), 3.42 (dd J = 13.2 Hz, J =
9.9 Hz, 1H), 3.39 ¨ 3.32 (m, 1H), 3.15, (dd J = 13.3 Hz, J = 4.5 Hz, 1H), 2.41 (s, 1H) ppm; 130 NMR (150 MHz, DMSO) 6: 164.2, 151.2, 146.3, 136.3, 123.7, 107.4, 105.6, 102.1, 73.3, 71.6, 69.7, 57.8, 51.8, 44.4, 37.3 ppm; HRMS-FAB Calcd for C15H18N07 (M + 1): 324.1085, Found: 324.1084.
Example 16: Anti-cancer activity The following cell lines and previously published research methods [Siedlakowski, et al. Cancer Biology and Therapy, 7:376-384 (2008); Kekre, et al.
Cancer Chemotherapy and Pharmacology, 56:29 (2005)] were used:
Cancer cell lines:
Human leukemia cells (Jurkat cells) Human neuroblastoma cells (shsy5y cells) Human Melanoma cells.
All these cells were obtained from ATCC, USA.
Normal non-cancerous cells:
Human peripheral nuclear blood cells (prepared from the blood donated by a helthy volunteer) Normal human fibroblasts (obtained from Corriel Cell repository, USA) Methods and assays:
Apoptosis characterization:
1. Morphology: Nuclear condensation as observed by bright Hoechst staining.
2. Annexin-V binding assay.
Results:
After correction with solvent control, it was observed that compound 1(c) induced apoptosis selectively in human cancer cell lines. Compound 1(d) also showed the apoptosis-inducing activity, but to a lower extent. The ED50 for 1(c) was 500 nM.
These compounds did not induce apoptosis in normal non-cancerous cells. (See Figures 2 and 3 for further results in Jurkat cells).

Claims (11)

THAT WHICH IS CLAIMED IS:
1. A compound of Formula I:
wherein R1 is selected from CH2NR2R3, CH2OR2, CH2R2, CH2OC(O)NR2R3, CH2NHC(O)R2, CH2NHC(O)OR2, CH2CHC(O)NR2R3 and CH2OC(O)R2 ; and R2 and R3 are independently selected from H, C1-6alkyl, C2-6alkenyl, C3-10cycloalkyl and C6-10aryl said latter four groups being unsubstituted or substituted with one to 5 groups independently selected from halo, OH, OC1-4alkyl, OC(O)C1-6alkyl and nitro; and R4 is selected from H and OH;
or a pharmaceutically acceptable salt and/or solvate thereof.
2. The compound according to claim 1, wherein R1 is selected from CH2NH2, CH2OH and CH2OC(O)CH3.
3. A pharmaceutical composition comprising one or more compounds of Formula I of claims 1 or 2, or a pharmaceutically acceptable salt and/or solvate thereof, and a pharmaceutically acceptable carrier.
4. A use of one or more compounds of Formula I of claims 1 or 2, or a pharmaceutically acceptable salt and/or solvate thereof, for treating cancer.
5. A process for preparing a compound of Formula II
wherein R4 is selected from H and OPg and each Pg may be the same or different and represent suitable protecting groups or any two adjacent Pg are joined to form a suitable cyclic protecting group;
the process comprising:
(i) reacting a compound of the Formula III with an aluminum acetylide derived from a compound of the Formula IV, followed by protection to form a compound of the Formula V, wherein R4 and each Pg is as defined above:
(ii) reducing the compound of Formula V to form a cis-alkene of the Formula VI, wherein R4 and each Pg is as defined above:
(iii) reacting the compound of the Formula VI under solid-state, silica gel catalysis conditions to form a compound of the Formula VII, wherein R4 and each Pg is as defined above:
(iv) oxidatively cleaving the non-aromatic double bond in the compound of the Formula VII to form an intermediate diketone of the Formula VIII which cyclizes to form a compound of the Formula IX, wherein R4 and each Pg is as defined above:
and (v) oxidizing the compound of the Formula IX to form a compound of Formula II, wherein R4 and each Pg is as defined above:

6. A process for preparing a compound of Formula I
wherein R1 is selected from C(O)OR2, C(O)R2, C(O)NR2R3, CH=NR2, CH2NR2R3, CH2OR2, CH2R2, NR2R3, NHC(O)R2, NHC(O)OR2, NHC(O)NR2R3, CH=CR2R3, CH2OC(O)NR2R3, CH2NHC(O)R2, CH2NHC(O)OR2, CH2CHC(O)NR2R3 and CH2OC(O)R2; and R2 and R3 are independently selected from H, C1-6alkyl, C2-6alkenyl, C3-10cycloalkyl and C6-10aryl said latter four groups being unsubstituted or substituted with one to 5 groups independently selected from halo, OH, OC1-4alkyl, OC(O)C1-6alkyl and nitro, with the exception that R2 is not H when R1 is C(O)R2; and R4 is selected from H and OH, comprising:
(i) reacting a compound of the Formula 11, wherein R4 is selected from H and OPg and each Pg may be the same or different and represent suitable protecting groups or any two adjacent Pg are joined to form a suitable cyclic protecting group under conditions to convert the aldehyde moiety in one or more steps to a group selected from C(O)OR2, C(O)R2, C(O)NR2R3, CH=NR2, CH2NR2R3, CH2OR2, CH2R2, NR2R3, NHC(O)R2, NHC(O)OR2, NHC(O)NR2R3, CH=CR2R3, CH2OC(O)NR2R3, CH2NHC(O)R2, CH2NHC(O)OR2, CH2CHC(O)NR2R3 and CH2OC(O)R2, wherein R2 and R3 are as defined above to form a compound of the Formula X wherein R1, R4 and each Pg are as defined above:
(ii) removing the Pg groups to form a compound of the Formula I wherein R1 and R4 are as defined above:
7. A process for preparing a compound of Formula I
i wherein R1 is C(O)H and R4 is H or OH, comprising removing the Pg groups from a compound of Formula II as defined in claim 5 to form the compound of the Formula I.
8. The compound of claim 1, wherein R1 is selected from CH2OR2 and CH2OC(O)R2.
9. The compound of claim 9, wherein R2 is selected from H, C1-6alkyl and C6-10aryl.
10. A compound of the Formula I(d):
or a pharmaceutically acceptable salt and/or solvate thereof.
11. A compound of the Formula I(c):
or a pharmaceutically acceptable salt and/or solvate thereof.
CA2647655A 2008-12-23 2008-12-23 Novel c-1 analogs of pancratistatin and 7-deoxypancratistatin and processes for their preparation Active CA2647655C (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA2647655A CA2647655C (en) 2008-12-23 2008-12-23 Novel c-1 analogs of pancratistatin and 7-deoxypancratistatin and processes for their preparation

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CA2647655A CA2647655C (en) 2008-12-23 2008-12-23 Novel c-1 analogs of pancratistatin and 7-deoxypancratistatin and processes for their preparation

Publications (2)

Publication Number Publication Date
CA2647655A1 CA2647655A1 (en) 2010-06-23
CA2647655C true CA2647655C (en) 2016-12-20

Family

ID=42283103

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2647655A Active CA2647655C (en) 2008-12-23 2008-12-23 Novel c-1 analogs of pancratistatin and 7-deoxypancratistatin and processes for their preparation

Country Status (1)

Country Link
CA (1) CA2647655C (en)

Also Published As

Publication number Publication date
CA2647655A1 (en) 2010-06-23

Similar Documents

Publication Publication Date Title
AU2003260002B2 (en) Synthesis of epothilones, intermediates thereto, analogues and uses thereof
EP0918776B1 (en) Total synthesis of antitumor acylfulvenes
EP1040119B1 (en) Hemiasterlin analogs
US20030176368A1 (en) Synthesis of epothilones, intermediates thereto and analogues thereof
CA2647655C (en) Novel c-1 analogs of pancratistatin and 7-deoxypancratistatin and processes for their preparation
US10000527B2 (en) 11-substituted bile acid derivatives, process for the preparation thereof and use of these compounds as medicaments
US8703792B2 (en) C-1 analogs of pancratistatin and 7-deoxypancratistatin and processes for their preparation
EP0154185B1 (en) Calciferol derivatives
EP1904485A2 (en) Synthesis and complete stereochemical assignment of psymberin/irciniastatin for anti-tumor use
Jackova et al. Total Synthesis and Bioactivity of ent-homospisulosine and N, O-diacetylhomoclavaminol A
EP0758316B1 (en) Borneol derivatives affecting tubulin polymerization and depolymerization
Gruszecka-Kowalik et al. Free-radical reactions of retronecine and heliotridine derivatives. The synthesis of (-)-supinidine
WO2004028452A2 (en) Method for preparing crambescidin core acid intermediates and their use for preparing crambescidin alkaloid analogs as therapeutic agents
WO1996025392A1 (en) Novel borneol derivatives, methods of manufacturing them, and their pharmaceutical use
WO2014100367A1 (en) Synthesis of fr901464 and analogs with antitumor activity
US9771325B2 (en) Tricyclic compounds and preparation thereof
JP2020533374A (en) Cyclic peptide antibiotic
TWI842251B (en) Antimalarial agents
Vlad et al. Asymmetric dihydroxylation of drim-7-en-11-ol: synthesis of diastereomerically pure driman-7α, 8α, 11-triol and its elaboration into novel chlorinated norlabdanic compounds
US20160002193A1 (en) Anticancer compounds and process for the preparation thereof
JP2000229977A (en) Tridecanolide derivative and anti-cancer agent
JP2004315373A (en) Compound having antitumor activity and neovascularization inhibitory activity and method for producing the same
US7153885B2 (en) Stolonoxides
CN115698018A (en) Meiyanmycin analogs and methods of use
US20040235939A1 (en) Synthesis of peloruside a and analogs thereof for use as antitumor agents

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20141030