WO1996038146A1 - Water soluble camptothecin analogs - Google Patents

Water soluble camptothecin analogs Download PDF

Info

Publication number
WO1996038146A1
WO1996038146A1 PCT/US1996/008283 US9608283W WO9638146A1 WO 1996038146 A1 WO1996038146 A1 WO 1996038146A1 US 9608283 W US9608283 W US 9608283W WO 9638146 A1 WO9638146 A1 WO 9638146A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
animal
pharmaceutically acceptable
carrier
Prior art date
Application number
PCT/US1996/008283
Other languages
French (fr)
Inventor
David A. Berges
John J. Taggart
Original Assignee
Smithkline Beecham Corporation
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 Smithkline Beecham Corporation filed Critical Smithkline Beecham Corporation
Priority to EP96916929A priority Critical patent/EP0871447A4/en
Priority to JP8536757A priority patent/JPH11506453A/en
Publication of WO1996038146A1 publication Critical patent/WO1996038146A1/en

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
    • C07D491/22Heterocyclic 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 four or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to a water soluble camptothecin analog which is particularly useful as an antineoplastic agent, pharmaceutical compositions thereof, and methods of treatment of cancer in animals, including human beings, in need thereof comprising inhibition of the growth of tumor cells sensitive to such an analog.
  • the structure of the DNA helix within eukaryotic cells imposes certain topological problems that the cellular apparatus must solve in order to use its genetic material as a template.
  • the separation of the DNA strands is fundamental to cellular processes such as DNA replication and transcription. Since eukaryotic DNA is organized into chromatin by chromosomal proteins, the ends are constrained and the strands cannot unwind without the aid of enzymes that alter topology. It has long been recognized that the advancement of the transcription or replication complex along the DNA helix would be facilitated by a swivel point which would relieve the torsional strain generated during these processes.
  • Topoisomerases are enzymes that are capable fo altering DNA topology in eukaryotic cells. They are critical for important cellular functions and cell proliferation.
  • Topoisomerase I is a monomeric enzyme of approximately 100,000 molecular weight. The enzyme binds to DNA and introduces a transient single strand break, unwinds the double helix (or allows it to unwind), and subsequently reseals the break before dissociating from the DNA strand.
  • Topoisomerase II consists of two identical subunits of molecular weight 170,000. Topoisomerase II transiently breaks both strands of the helix and passes another double- strand segment through the break.
  • Camptothecin is a water-insoluble, cytotoxic alkaloid produced by Camptotheca accuminata trees indigenous to China and Nothapodytes foetida trees indigenous to India. Camptothecin and a few close congeners thereof are the only class of compounds known to inhibit topoisomerase I.
  • topoisomerase II Inhibition of topoisomerase II is the major target of important commercial oncolytic agents (e.g., etoposide, doxorubicin and mitoxantrone) as well as other oncolytic agents still undergoing development. Camptothecin and its known congeners have no effect on topoisomerase II and none of the known topoisomerase II inhibitors has any significant effect on topoisomerase I.
  • oncolytic agents e.g., etoposide, doxorubicin and mitoxantrone
  • Camptothecin and most of its analogs have not proven to be attractive for clinical drug development as cytolytic agents because of unacceptable dose limiting toxicity, unpredictable toxicity, poor aqueous solubility, unacceptable shelf life stability, and/or lack of clinical efficacy.
  • the present invention relates to pharmaceutical compositions of the compound of Formula I.
  • the present invention relates to methods of treatment of cancer in animals, including human beings, in need thereof comprising inhibition of the growth of tumor cells by administration of an effective amount of the compound of Formula I, alone or in combination with a carrier, diluent or excipient.
  • an effective amount means that amount of a compound or pharmaceutical composition of the present invention which, upon administration to an animal, including a human being, in need thereof for the treatment of cancer, provides a clinically desirable result in the treatment of such cancer as it is understood by one of ordinary skill in the antineoplastic treatment art, including, but not limited to, inhibition of the growth of tumor cells, remission, or cure.
  • compositions are prepared in a manner well-known to those of ordinary skill in the art.
  • the parent compound dissolved in a suitable solvent, is reacted with an excess of an organic or inorganic acid.
  • Representative acids are hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, acetic acid, maleic acid, succinic acid or methanesulfonic acid.
  • a chiral center or another form of an isomeric center is present in the compound of the present invention, all forms of such isomer or isomers are intended to be covered herein.
  • Such compound containing a chiral center may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well- known techniques and an individual enantiomer may be used alone.
  • the present invention provides a method of treatment of cancer in an animal, preferably a mammal, most preferably a human, in need of such treatment, comprising administering to such animal an effective amount of a compound of Formula I as described hereinabove, or a pharmaceutically acceptable salt thereof, alone or in combination with a carrier, excipient or diluent.
  • Chinese Hamster ovary cells are grown in Alpha MEM Medium with L- glutamine and nucleosides and containing 10% fetal bovine serum and 100 units per mL penicillin-streptomycin in 75 cm 2 canted neck tissue culture flasks. They are harvested from these flasks using 0.5% trypsin.
  • Microtiter plates (96-well, sterile, flat bottom) (Corning 25860) are seeded with 1.6xl0 3 wild-type (AUX-B1) Chinese Hamster ovary cells per well or 2xl0 3 multidrug resistant (CH R C5) Chinese Hamster ovary cells per well. The plates are incubated at 37°C, 5% CO 2 overnight to allow the cells to attach.
  • XTT Eight mg of XTT (SIGMA X-4251) is dissolved in 100 ⁇ L of DMSO which is then added to 3.9 mL of phosphate buffered saline without cations (PBS). Phenazine methosulfate (SIGMA P-9625) is dissolved in PBS to a concentration of 3 mg/mL and 20 ⁇ L of this is added to the XTT solution. Fifty ⁇ L of this XTT/PMS solution is added to each well of the microtiter plate and the plates are incubated for 90 minutes at 37°C, 5% CO 2 (until the OD 4 5 0 --I.O). The plate is then read on a UV Max plate reader, using wells without cells (i.e., containing only 200 ⁇ L of medium and 50 ⁇ L of XTT/PMS solution) as a background control.
  • PBS phosphate buffered saline without cations
  • the compound of Formula I is prepared by the method described in Scheme 1.
  • the hydroxy group of 10-hydroxycamptothecin 1, which is readily available from camptothecin by the process described in US Patent No. 5,004,758, is protected as an ester, for example as a propionate ester 2, by reaction with an acylating agent such as propionic anhydride in the presence of a base such as pyridine.
  • compositions prepared from the compound of Formula I have both a human and a veterinary utility, and comprise an excipient, diluent, or carrier which is acceptable for the intended pharmaceutical end use and the inventive compound.
  • the carrier may be a liquid, or spray, or may be formulated in a solid, non- degradeable or degradeable form for insertion in the rumen. Selected excipients and carriers may be employed to prepare compositions acceptable or adaptable for human use.
  • compositions of the present invention may be contained in one embodiment, such as in a single pill, capsule, or pre- measured intravenous dose or pre-filled syringe for injection.
  • the composition will be prepared in individual dose forms where one unit, such as a pill, will contain a sub-optimal dose but the user will be instructed to take two or more unit doses per treatment.
  • the composition When the composition is presented as a cream, it will contain a discrete amount of drug and the user will apply some amount of the cream one or more times until the disease is in remission or has been effectively treated. Concentrates for later dilution by the end user may also be prepared, for instance for intravenous (TV) formulations and multi-dose injectable formulations.
  • TV intravenous
  • Excipients, diluents, or carriers contemplated for use in these compositions are generally known in the pharmaceutical formulary arts. Reference to useful materials can be found in well-known compilations such as Remington's Pharmaceutical Sciences. Mack Publishing Co., Easton, Pa.
  • compositions and the pharmaceutical excipient, diluent or carrier will, of course, depend upon the intended route of administration, for example whether by intravenous and intramuscular injection, parenterally, topically, orally, or by inhalation.
  • the pharmaceutical composition will be in the form of a sterile injectable liquid such as an ampule or an aqueous or nonaqueous liquid suspension.
  • the pharmaceutical composition will be in the form of a cream, ointment, liniment, lotion, paste, spray or drops suitable for administration to the skin, eye, ear, nose or genitalia.
  • the pharmaceutical composition will be in the form of a tablet, capsule, powder, pellet, troche, lozenge, syrup, liquid, or emulsion.
  • the pharmaceutical excipient, diluent or carrier employed may be either a solid or liquid.
  • suitable pharmaceutical carriers or diluents include: for aqueous systems, water; for non-aqueous systems: ethanol, glycerin, propylene glycol, olive oil, corn oil, cottonseed oil, peanut oil, sesame oil, liquid paraffins, and mixtures thereof with water; for solid systems: lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid, kaolin and mannitol; and for aerosol systems: dichlorodifluoromethane, chlorotrifluoroethane and compressed carbon dioxide.
  • a wide variety of pharmaceutical forms can be employed.
  • the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form or in the form of a troche or lozenge.
  • the amount of solid carrier will vary widely but preferably will be from about 25 mg to about 1 gram.
  • the preparation will be in the form of a syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in an ampule or vial or nonaqueous liquid suspension.
  • a pharmaceutically acceptable salt of the compound of Formula I is dissolved in an aqueous solution of an organic or inorganic acid or base.
  • the compound of Formula I may be dissolved in a suitable co-solvent or combinations thereof.
  • suitable cosolvents include, but are not limited to, alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin and the like in concentrations ranging from 0-60% of the total volume.
  • the actual preferred dosages of the compound used in the compositions and methods of treatmentof the present invention will vary according to the particular complex being used, the particular composition formulated, the mode of administration and the particular site, host and tumor type being treated.
  • Optimal dosages for a specific pathological condition in a particular patient may ascertained by those of ordinary skill in the antineoplastic art using conventional dosage determination tests in view of the above experimental data.
  • the dose of the compound of Formula I generally employed is from about 2 to about 50 mg m ⁇ of body surface area per day for one to five days, preferably repeated about every fourth week for four courses of treatment.
  • the dose generally employed is about 0.5 mg/m ⁇ /day for 5 to 21 days.
  • the dose generally employed is about 20 to about 150 mg/m ⁇ of body surface area per day for one to five days, with courses of treatment repeated at appropriate intervals.
  • a parenteral pharmaceutical composition of this invention suitable for administration by injection 100 mg of a water soluble salt of a compound of Formula I is mixed with 10 ml of 0.9% sterile saline, and the mixture is incorporated into a dosage unit form suitable for administration by injection.
  • an oral pharmaceutical composition of this invention 100 mg of a compound of Formula I is mixed with 750 mg of lactose, and the mixture is incorporated into an oral dosage unit form, such as a hard gelatin capsule, which is suitable for oral administration.

Abstract

The present invention provides a water soluble camptothecin analog of Formula (I), which is particularly useful as an antineoplastic agent; pharmaceutical compositions thereof; and a method of treating cancer in an animal in need thereof, including human beings, comprising inhibition of the growth of tumor cells in said animal by administration of an effective amount of a compound of Formula (I).

Description

WATER SOLUBLE CAMPTOTHECIN ANALOGS
FIELD OF THE INVENTION
The present invention relates to a water soluble camptothecin analog which is particularly useful as an antineoplastic agent, pharmaceutical compositions thereof, and methods of treatment of cancer in animals, including human beings, in need thereof comprising inhibition of the growth of tumor cells sensitive to such an analog.
BACKGROUND OF THE INVENTION
The structure of the DNA helix within eukaryotic cells imposes certain topological problems that the cellular apparatus must solve in order to use its genetic material as a template. The separation of the DNA strands is fundamental to cellular processes such as DNA replication and transcription. Since eukaryotic DNA is organized into chromatin by chromosomal proteins, the ends are constrained and the strands cannot unwind without the aid of enzymes that alter topology. It has long been recognized that the advancement of the transcription or replication complex along the DNA helix would be facilitated by a swivel point which would relieve the torsional strain generated during these processes. Topoisomerases are enzymes that are capable fo altering DNA topology in eukaryotic cells. They are critical for important cellular functions and cell proliferation.
There are two classes of topoisomerases in eukaryotic cells, type I and type π. Topoisomerase I is a monomeric enzyme of approximately 100,000 molecular weight. The enzyme binds to DNA and introduces a transient single strand break, unwinds the double helix (or allows it to unwind), and subsequently reseals the break before dissociating from the DNA strand.
Topoisomerase II consists of two identical subunits of molecular weight 170,000. Topoisomerase II transiently breaks both strands of the helix and passes another double- strand segment through the break.
Camptothecin is a water-insoluble, cytotoxic alkaloid produced by Camptotheca accuminata trees indigenous to China and Nothapodytes foetida trees indigenous to India. Camptothecin and a few close congeners thereof are the only class of compounds known to inhibit topoisomerase I.
Inhibition of topoisomerase II is the major target of important commercial oncolytic agents (e.g., etoposide, doxorubicin and mitoxantrone) as well as other oncolytic agents still undergoing development. Camptothecin and its known congeners have no effect on topoisomerase II and none of the known topoisomerase II inhibitors has any significant effect on topoisomerase I.
Camptothecin and most of its analogs have not proven to be attractive for clinical drug development as cytolytic agents because of unacceptable dose limiting toxicity, unpredictable toxicity, poor aqueous solubility, unacceptable shelf life stability, and/or lack of clinical efficacy.
Figure imgf000004_0001
(S)-Camptothecin
However, water soluble camptothecin analogs having efficacy as topoisomerase I inhibitor antineoplastic agents are known. U.S. Patent No. 5,004,758, issued to Boehm, et al. on April 2, 1991, the specification of which is incorporated herein by reference, discloses water soluble camptothecin analogs, preferably topotecan (9- dimethylaminomethyl-10-hydroxycamptothecin), preferably (S)-topotecan, of formula:
Figure imgf000004_0002
(S)-Topotecan
most preferably as the hydrochloride salt. In clinical tests, topotecan has demonstrated efficacy against several solid tumor cancers, particularly ovarian cancer and non-small cell lung carcinoma in humans.
Masuda, et al, J. Clin. Oncology. 1992, 10, 1225-1229 describes CPT-11 ((S)- [1 ,4'-bipiperidine]- 1 '-carboxylic acid,4, 1 l-diethyl-3,4, 12, 14-tetrahydro-4-hydroxy-3, 14- dioxo-lH-pyrano[3',4':6,7]indolizino[l,2- >]quinolin-9-yl ester). However, efforts to develop CPT-11 as an antineoplastic agent have been hampered by an adverse toxicity profile.
Wall, etal, J. Med. Chem.. 1993, 36, 2689-2700 describes 9-aminocamptothecin ((S)-10-amino-4-ethyl-4-hydroxy-lH-pyrano[3',4,:6,7]indolizino[l,2-fc]quinoline- 3,14(4H,12H)-dione). However, this compound possesses limited water solubility which has posed formulation and bioavailability problems in its development as an antineoplastic agent.
There is a need for new topoisomerase I inhibiting agents which avoid the undesirable features described above. The compounds of the present invention satisfy such need.
SUMMARY OF THE INVENTION
In a first aspect, the present invention provides a compound of Formula I:
Figure imgf000005_0001
I known as S-9-ethyl-2,3-dihydro-4,9-dihydroxy-2-methyl-lH,12H- benzo[ι/]pyrano[3',4':6,7]indolizino[ 1 ,2-c][2,6]naphthyridine- 10, 13(H, 15H)-dione, and pharmaceutically acceptable salts thereof.
In another aspect, the present invention relates to pharmaceutical compositions of the compound of Formula I.
In yet another aspect, the present invention relates to methods of treatment of cancer in animals, including human beings, in need thereof comprising inhibition of the growth of tumor cells by administration of an effective amount of the compound of Formula I, alone or in combination with a carrier, diluent or excipient. DETAILED DESCRIPTION OF THE INVENTION
The term "effective amount" means that amount of a compound or pharmaceutical composition of the present invention which, upon administration to an animal, including a human being, in need thereof for the treatment of cancer, provides a clinically desirable result in the treatment of such cancer as it is understood by one of ordinary skill in the antineoplastic treatment art, including, but not limited to, inhibition of the growth of tumor cells, remission, or cure.
Salts may be made from the compound of the present invention by reaction with its basic nitrogen. Particularly preferred are the pharmaceutically acceptable salts of the instant compound. These latter salts are those which are acceptable in their application to a pharmaceutical use. By that it is meant that the salt will retain the biological activity of the parent compound and the salt will not have untoward or deleterious effects in its application and use in treating diseases.
Pharmaceutically acceptable salts are prepared in a manner well-known to those of ordinary skill in the art. The parent compound, dissolved in a suitable solvent, is reacted with an excess of an organic or inorganic acid. Representative acids are hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, acetic acid, maleic acid, succinic acid or methanesulfonic acid.
Here and throughout this application, the ring system of the compounds of the present invention is numbered according to Formula H
Figure imgf000006_0001
π
If a chiral center or another form of an isomeric center is present in the compound of the present invention, all forms of such isomer or isomers are intended to be covered herein. Such compound containing a chiral center may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well- known techniques and an individual enantiomer may be used alone.
The present invention provides a compound, and pharmaceutically acceptable salts thereof, which exhibits antineoplastic activity, said compound having the structure represented by Formula I hereinabove.
No unacceptable toxicological effects are expected when the compound of the present invention is administered in accordance with the present invention.
The present invention provides a method of treatment of cancer in an animal, preferably a mammal, most preferably a human, in need of such treatment, comprising administering to such animal an effective amount of a compound of Formula I as described hereinabove, or a pharmaceutically acceptable salt thereof, alone or in combination with a carrier, excipient or diluent.
The monohydrochloride salt of the compound of Formula I is the preferred embodiment of the present invention.
The in vitro assays used to test the compound of the present invention for antitumor activity are well-known. A generalized description of these assays follows.
CHO Microtiter Cytotoxicity Assay
Chinese Hamster ovary cells are grown in Alpha MEM Medium with L- glutamine and nucleosides and containing 10% fetal bovine serum and 100 units per mL penicillin-streptomycin in 75 cm2 canted neck tissue culture flasks. They are harvested from these flasks using 0.5% trypsin. Microtiter plates (96-well, sterile, flat bottom) (Corning 25860) are seeded with 1.6xl03 wild-type (AUX-B1) Chinese Hamster ovary cells per well or 2xl03 multidrug resistant (CHRC5) Chinese Hamster ovary cells per well. The plates are incubated at 37°C, 5% CO2 overnight to allow the cells to attach. The outside wells of each plate are not used, due to evaporation during the incubation time. They are filled with medium and used as blanks. The next day, the medium is aspirated from the wells and 180 μL of fresh medium is added to each well. Compounds are diluted from stock solution in DMSO into fresh medium to a 10X concentration containing 2% DMSO. Twenty μL of this is then added to the 180 μL of fresh medium in the wells. The plates are then incubated for another 3 days at 37°C, 5% CO2. Eight mg of XTT (SIGMA X-4251) is dissolved in 100 μL of DMSO which is then added to 3.9 mL of phosphate buffered saline without cations (PBS). Phenazine methosulfate (SIGMA P-9625) is dissolved in PBS to a concentration of 3 mg/mL and 20 μL of this is added to the XTT solution. Fifty μL of this XTT/PMS solution is added to each well of the microtiter plate and the plates are incubated for 90 minutes at 37°C, 5% CO2 (until the OD450--I.O). The plate is then read on a UV Max plate reader, using wells without cells (i.e., containing only 200 μL of medium and 50 μL of XTT/PMS solution) as a background control.
The cytotoxicity and efficacy of the compound of the present invention was also tested in vivo using the well-known P388 mouse tumor model.
Table I provides a comparison of the cytotoxicity and efficacy in mouse tumor models of the compound of Formula I with the known compounds topotecan and camptothecin. These results demonstrate that the compound of Formula I possesses biological activity comparable to topotecan.
Table I
Cytotoxicity IC50 (μM) Efficacy in Mouse Tumor Models
Wildtyp Multidrug % Inc. in > Lifespan/Dose (mg/kg)
SB No. (AUX-B1) Resistant P388 P388
Topotecan 0.79 2.3 0.03 156/14.4
Camptothecin 0.015 0.035 0.012
Formula I 0.052 0.84 0.024 111/40 ip
The compound of Formula I is prepared by the method described in Scheme 1. The hydroxy group of 10-hydroxycamptothecin 1, which is readily available from camptothecin by the process described in US Patent No. 5,004,758, is protected as an ester, for example as a propionate ester 2, by reaction with an acylating agent such as propionic anhydride in the presence of a base such as pyridine. Treatment of 2 in N,N- dimethylformamide with a free-radical generating reagent such as benzoyl peroxide in the presence of an acid such as trifluoroacetic acid followed by chromatography on silica gel with methanol in the solvent produces formamide 3 which is deformylated by heating with a strong acid such as hydrochloric acid in methanol. The resulting amine 4) is then treated with formaldehyde in aqueous acetic acid to effect a Pictet-Spengler cyclization giving the compound of Formula I.
Figure imgf000009_0001
a) (CH CH2CO)2O, pyridine, DMF;b) (C6H5CO)2θ2, TFA, DMF, 85°C and then CH3OH, silica gel; c) 12 N HCI in CH3OH (1:19), reflux; d) HCHO, CH3COOH, H2O, 70°C.
Scheme 1
The present invention provides pharmaceutical compositions prepared from the compound of Formula I. These compositions have both a human and a veterinary utility, and comprise an excipient, diluent, or carrier which is acceptable for the intended pharmaceutical end use and the inventive compound. For example, if a veterinary use is intended, the carrier may be a liquid, or spray, or may be formulated in a solid, non- degradeable or degradeable form for insertion in the rumen. Selected excipients and carriers may be employed to prepare compositions acceptable or adaptable for human use.
An effective amount of one or more pharmaceutical compositions of the present invention may be contained in one embodiment, such as in a single pill, capsule, or pre- measured intravenous dose or pre-filled syringe for injection. Alternatively, as is frequently the case, the composition will be prepared in individual dose forms where one unit, such as a pill, will contain a sub-optimal dose but the user will be instructed to take two or more unit doses per treatment. When the composition is presented as a cream, it will contain a discrete amount of drug and the user will apply some amount of the cream one or more times until the disease is in remission or has been effectively treated. Concentrates for later dilution by the end user may also be prepared, for instance for intravenous (TV) formulations and multi-dose injectable formulations.
Excipients, diluents, or carriers contemplated for use in these compositions are generally known in the pharmaceutical formulary arts. Reference to useful materials can be found in well-known compilations such as Remington's Pharmaceutical Sciences. Mack Publishing Co., Easton, Pa.
The nature of the composition and the pharmaceutical excipient, diluent or carrier will, of course, depend upon the intended route of administration, for example whether by intravenous and intramuscular injection, parenterally, topically, orally, or by inhalation.
For parenteral administration the pharmaceutical composition will be in the form of a sterile injectable liquid such as an ampule or an aqueous or nonaqueous liquid suspension.
For topical administration the pharmaceutical composition will be in the form of a cream, ointment, liniment, lotion, paste, spray or drops suitable for administration to the skin, eye, ear, nose or genitalia.
For oral administration the pharmaceutical composition will be in the form of a tablet, capsule, powder, pellet, troche, lozenge, syrup, liquid, or emulsion.
The pharmaceutical excipient, diluent or carrier employed may be either a solid or liquid. When the pharmaceutical composition is employed in the form of a solution or suspension, examples of appropriate pharmaceutical carriers or diluents include: for aqueous systems, water; for non-aqueous systems: ethanol, glycerin, propylene glycol, olive oil, corn oil, cottonseed oil, peanut oil, sesame oil, liquid paraffins, and mixtures thereof with water; for solid systems: lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid, kaolin and mannitol; and for aerosol systems: dichlorodifluoromethane, chlorotrifluoroethane and compressed carbon dioxide. Also, in addition to the pharmaceutical carrier or diluent, the instant compositions may include other ingredients such as stabilizers, antioxidants, preservatives, lubricants, suspending agents, viscosity modifiers and the like, provided that the additional ingredients do not have a detrimental effect on the therapeutic action of the instant compositions. Similarly, the carrier or diluent may include time delay material well known to the art, such as glyceryl monostearate or glyceryl distearate alone or with a wax, ethylcellulose, hydroxypropylmethylcellulose, methylmethacrylate and the like.
A wide variety of pharmaceutical forms can be employed. Thus, if a solid carrier is used, the preparation can be tableted, placed in a hard gelatin capsule in powder or pellet form or in the form of a troche or lozenge. The amount of solid carrier will vary widely but preferably will be from about 25 mg to about 1 gram. If a liquid carrier is used, the preparation will be in the form of a syrup, emulsion, soft gelatin capsule, sterile injectable solution or suspension in an ampule or vial or nonaqueous liquid suspension. To obtain a stable water soluble dose form, a pharmaceutically acceptable salt of the compound of Formula I is dissolved in an aqueous solution of an organic or inorganic acid or base. If a soluble salt form is not available, the compound of Formula I may be dissolved in a suitable co-solvent or combinations thereof. Examples of such suitable cosolvents include, but are not limited to, alcohol, propylene glycol, polyethylene glycol 300, polysorbate 80, glycerin and the like in concentrations ranging from 0-60% of the total volume.
It will be appreciated that the actual preferred dosages of the compound used in the compositions and methods of treatmentof the present invention will vary according to the particular complex being used, the particular composition formulated, the mode of administration and the particular site, host and tumor type being treated. Optimal dosages for a specific pathological condition in a particular patient may ascertained by those of ordinary skill in the antineoplastic art using conventional dosage determination tests in view of the above experimental data. For parenteral administration, the dose of the compound of Formula I generally employed is from about 2 to about 50 mg m^ of body surface area per day for one to five days, preferably repeated about every fourth week for four courses of treatment. For continuous intravenous administration, the dose generally employed is about 0.5 mg/m^/day for 5 to 21 days. For oral administration, the dose generally employed is about 20 to about 150 mg/m^ of body surface area per day for one to five days, with courses of treatment repeated at appropriate intervals. EXAMPLES
In the following synthetic examples, temperature is in degrees Centigrade (°C). Unless otherwise indicated, all of the starting materials were obtained from commercial sources. Without further elaboration, it is believed that one skilled in the art can, using the preceding description, utilize the present invention to its fullest extent. These Examples are given to illustrate the invention, not to limit its scope. Reference is made to the claims for what is reserved to the inventors hereunder.
EXAMPLE 1
Preparation of (SV9-Ethyl-2.3-dihyro-4.9-dihvdroxy-2-methyl-lH.12H- benzor /1pyranor3'.4':6.71indolizinor 1 ,2-cir2.61naρhthridine- 10.13C9H.15HVdione monohydrochloride monohydroacetate monohydrate
a) (SV 10-Propanoyloxycamptothecin
(S)-lO-Ηydroxycamptothecin (3.93 g, 0.0108 mol) in dry DMF (100 mL) and dry pyridine ( 8.5 mL) was treated in one portion with propanoic anhydride (1.48 g, 0.0114 mol). The solution was stirred for several hours. More propanoic anhydride (0.370 g, 0.00285 mol) was added, and the solution was stirred for an additional 5 h. The reaction mixture was evaporated to dryness under reduced pressure, and the residue was partitioned between methylene chloride and water. The layers were separated, and the aqueous phase was re-extracted with methylene chloride. The combined organic layer was dried (sodium sulfate) and filtered, and the filtrate was concentrated to afford a yellow-ecru solid. This solid was triturated with methanol to afford the title compound as a tan-ecru solid (3.89 g, 86%). !H NMR (400 MHz, CDCI3), δ 8.40 (s, IH), 8.20 (d, J=9.2, IH), 7.71 (d, J=3.3, IH), 7.56 (dd, J=9.2, J=2.3, IH), 5.68 (d, J=16.3, IH), 5.31 (d, J=16.3 Hz, IH), 5.30 (s, 2H), 3.86 (s, 2H), 2.71 (q, J=7.5 Hz, 2H), 1.93 (m, 2H), 1.33 (t, J=7.4 Hz, 3H).
b) (S)-7-N-Formyl-N-methylaminomethyl- 10-hvdroxycamptothecin
A stirred suspension of (S)-lO-propanoyloxycamptothecin (2.00 g, 0.00476 mol) and dry DMF (100 mL) was treated with trifluoroacetic acid (0.92 mL, 0.012 mol). The resulting clear solution was then treated with benzoyl peroxide (1.15 g, 0.00476 mol). The solution was warmed to 85°C, stirred for 6 h, and evaporated to a dark amber residue which was chromatographed on silica gel (gradient from methylene chloride to 96.5:3.5 methylene chloride:methanol). Fractions containing the desired product were pooled, evaporated to dryness, and sonicated with methanol (6 mL) The resulting solid was collected and dried in vacuo to afford the title compound as a tan solid (64 mg, 3.1%). *H NMR (400 MHz, CDCI3 + CD3OD) δ 8.18 (s, IH), 8.08 (d, J=9.2, IH), 7.65 (s, IH), 7.37-7.46 (m, 2H), 5.67 (d, J=16, IH), 5.31 (s, 2H), 5.30 (d, J=16, IH), 5.07 (s, 2H), 2.87 (s, 3H), 1.93 (m, 2H), 1.03 (t, J=7.3 Hz, 3H).
c) ( S ι-7-Methylaminomethyl- 10-hydroxycamptothecin hydrochloride
(S)-7-N-Formyl-N-methylaminomethyl-10-hydroxycamptothecin (32 mg, 0.0735 mmol) was suspended in 5% HCI in methanol (5 mL). The mixture was heated at just below reflux temperature for 1 h and then cooled in ice, and a solid was collected, washed sparingly with cold methanol, and dried in vacuo to afford 20.2 mg of the title compound. A second crop (5 mg, 84% total yield) was obtained by allowing the filtrate to stand overnight at room temperature. *H NMR (400 MHz, CDCI3 + CD3OD) δ 8.16 (d, J=9.1 Hz, IH), 7.68 (s, IH), 7.55 (dd, J =9.1 Hz and J=2.0 Hz, IH), 7.49 (t, J= 2.0 Hz, IH), 5.63 (d, J=16.3 Hz, IH), 5.48 (s, 2H), 5.35 (d, J=16.3 Hz, IH), 4.71-4.80 (m, 2H), 2.85 (s, 3H), 1.97 (m, 2H), 1.03 (t, J=7.4 Hz, 3H).
d) fSV9-Ethyl-2.3-dihyro-4.9-dihvdroxy-2-rnethyl-lH.12H- benzor//lρyranor3'.4':6.71indolizinor 1.2-cir2.61naphthridine- 10.13( 9H.15HVdione monohydrochloride monohyrdoacetate monohydrate
(S)- 7-Methylaminomethyl-lO-hydroxycamptothecinohydrochloride (20 mg, 0.0416 mmol) in acetic acid (4 mL) and water (1 mL) was treated with 37% formaldehyde solution (72 μL). The solution was stirred at 75_ C for 5 h and evaporated to dryness. The residue was taken up in water and subjected to MPLC (Partisil 40 ODS- 3 using a gradient from:water with 0.1% acetic acid to 7:3 water-methanol with 0.1% acetic acid). The effluent was monitored at 254 nm, and product-containing fractions were pooled, concentrated in vacuo to 3 mL and treated with 10 μL of 0.1 N ΗC1 solution. This solution was lyophilized to give a canary colored solid (15.8 mg, 68%) Η NMR (400 MHz, CDCI3 + CD3OD) δ 7.85-7.95 (m, IH), 7.63 (s, IH), 7.30-7.45 (m, IH), 5.65 (d, J=16 Hz, IH), 5.30 (d, J=16 Hz, IH), 5.18 (s, 2H), 3.97 (s, 2H), 3.35 (s, 2H), 2.72 (s, 3H), 2.05 (s, 3H), 1.85-1.98 (m, 2H), 1.03 (t, J=7.3 Hz, 3H); MS (electrospray ionization) m/e 420 [M+H]+; Anal.: (C23H21N3O50HCI0C2H4O20H2O) calcd.: C, 56.23; H, 5.29; N, 7.87. found: C, 55.97; H, 4.97; N, 7.72. EXAMPLE 2 Parenteral Composition
To prepare a parenteral pharmaceutical composition of this invention suitable for administration by injection, 100 mg of a water soluble salt of a compound of Formula I is mixed with 10 ml of 0.9% sterile saline, and the mixture is incorporated into a dosage unit form suitable for administration by injection.
EXAMPLE 3 Oral Composition
To prepare an oral pharmaceutical composition of this invention, 100 mg of a compound of Formula I is mixed with 750 mg of lactose, and the mixture is incorporated into an oral dosage unit form, such as a hard gelatin capsule, which is suitable for oral administration.
Although the above specification and Examples fully describe the present invention, particularly the preferred embodiments thereof, it is understood that the present invention is not limited to these particular disclosed embodiments. Thus, the present invention includes all embodiments coming within the scope of the following claims.

Claims

We claim:
1. A compound of Formula I:
Figure imgf000015_0001
known as S-9-ethyl-2,3-dihydro-4,9-dihydroxy-2-methyl-lH,12H- benzo[ι/']pyrano[3,,4': 6,7]indolizino[ 1 ,2-c] [2,6]naphthyridine- 10, 13(H, 15H)-dione, and pharmaceutically acceptable salts thereof.
2. A pharmaceutical composition comprising a compound of Formula I:
Figure imgf000015_0002
known as S-9-ethyl-2,3-dihydro-4,9-dihydroxy2-methyl-lH,12H- benzo[ι/]pyrano[3'.4':6,7]indolizino[ 1 ,2-c] [2,6]naphthyridine-10, 13(H, 15H)-dione, or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable carrier, diluent or excipient. 3. A method of treating cancer in an animal in need thereof comprising inhibition of the growth of tumor cells in said animal by administration of an effective amount of a compound of Formula I:
Figure imgf000016_0001
I known as S-9-ethyl-2,
3-dihydro-4,9-dihydroxy-2-methyl-lH,12H- benzo[ /]pyrano[3,,4,:6,7]indolizino[l,2-c][2,6]naphthyridine-10,13(H,15H)-dione, or a pharmaceutically acceptable salt thereof, alone or in combination with a carrier, diluent or excipient.
4. A method of treating cancer according to Claim 3 wherein said animal is a human being.
5. A formulation comprising a compound of Claim 1 in admixture with a carrier, diluent, or excipient.
PCT/US1996/008283 1995-05-31 1996-05-30 Water soluble camptothecin analogs WO1996038146A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP96916929A EP0871447A4 (en) 1995-05-31 1996-05-30 Water soluble camptothecin analogs
JP8536757A JPH11506453A (en) 1995-05-31 1996-05-30 Water-soluble camptothecin analog

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/454,793 US5663177A (en) 1995-05-31 1995-05-31 Water soluble camptothecin analogs
US08/454,793 1995-05-31

Publications (1)

Publication Number Publication Date
WO1996038146A1 true WO1996038146A1 (en) 1996-12-05

Family

ID=23806114

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/008283 WO1996038146A1 (en) 1995-05-31 1996-05-30 Water soluble camptothecin analogs

Country Status (4)

Country Link
US (1) US5663177A (en)
EP (1) EP0871447A4 (en)
JP (1) JPH11506453A (en)
WO (1) WO1996038146A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6486320B2 (en) 2000-09-15 2002-11-26 Aventis Pharma S.A. Preparation of camptothecin and of its derivatives
US6503889B2 (en) 2000-02-28 2003-01-07 Aventis Pharma S.A. Composition comprising camptothecin and a pyrimidine derivative for the treatment of cancer
US6545010B2 (en) 2000-03-17 2003-04-08 Aventis Pharma S.A. Composition comprising camptothecin or a camptothecin derivative and a platin derivative for the treatment of cancer
US6548488B2 (en) 2000-03-17 2003-04-15 Aventis Pharma S.A. Composition comprising camptothecin or a camptothecin derivative and an alkylating agent for the treatment of cancer
US6562834B2 (en) 2000-10-27 2003-05-13 Aventis Pharma S. A. Combination comprising camptothecin and a stilbene derivative for the treatment of cancer
WO2003045952A2 (en) * 2001-11-30 2003-06-05 Chugai Seiyaku Kabushiki Kaisha Condensed camptothecins as antitumor agents
WO2007113687A2 (en) 2006-03-30 2007-10-11 Diatos S.A. Camptothecin-cell penetrating peptide conjugates and pharmaceutical compositions containing the same
US7910593B2 (en) 2004-04-09 2011-03-22 Chugai Seiyaku Kabushiki Kaisha Water-soluble prodrugs
US8022047B2 (en) 2005-08-22 2011-09-20 Chugai Seiyaku Kabushiki Kaisha Combination anticancer agents
CN106588946A (en) * 2017-01-25 2017-04-26 郑州大学 10-HCPT (10-hydroxycamptothecine) derivative, synthesis method and application thereof
US9763968B2 (en) 2004-12-22 2017-09-19 Rutgers, The State University Of New Jersey Hydrogel formulation for dermal and ocular delivery
US9999596B2 (en) 2004-12-22 2018-06-19 Rutgers, The State University Of New Jersey Controlled release hydrogels
WO2023216956A1 (en) * 2022-05-13 2023-11-16 四川科伦博泰生物医药股份有限公司 Camptothecin compound, preparation method therefor and use thereof

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6011042A (en) * 1997-10-10 2000-01-04 Enzon, Inc. Acyl polymeric derivatives of aromatic hydroxyl-containing compounds
WO2020200880A1 (en) 2019-03-29 2020-10-08 Medimmune Limited Compounds and conjugates thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4939255A (en) * 1987-06-24 1990-07-03 Daiichi Pharmaceutical Co., Ltd. Hexa-cyclic camptothecin derivatives

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5004758A (en) * 1987-12-01 1991-04-02 Smithkline Beecham Corporation Water soluble camptothecin analogs useful for inhibiting the growth of animal tumor cells
JPH0615547B2 (en) * 1988-01-20 1994-03-02 株式会社ヤクルト本社 Novel camptothecin derivative
HU213136B (en) * 1990-08-14 1997-02-28 Kyorin Seiyaku Kk Process for producing fluoroethyl camptothecin derivatives and pharmaceutical compositions containing them
JP3008226B2 (en) * 1991-01-16 2000-02-14 第一製薬株式会社 Hexacyclic compounds
IS4152A (en) * 1993-04-29 1994-10-30 Glaxo Inc. Water-soluble Camptothecin derivatives and their method of production

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4939255A (en) * 1987-06-24 1990-07-03 Daiichi Pharmaceutical Co., Ltd. Hexa-cyclic camptothecin derivatives
US5061795A (en) * 1987-06-24 1991-10-29 Daiichi Pharmaceutical Co., Ltd. Hexa-cyclic compound

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JOUR. MED. CHEM., 1993, Vol. 36, WALL et al., "Plant Antitumor Agents. 30, 1a,b Synthesis and Structure of Novel CAMPTOTHECIN Analogs", pages 2689-2700. *
JOURNAL OF CLINICAL ONCOLOGY, Vol. 10, MASUDA et al., "CPT-11: A New Derivative of CAMPTOTHECIN for the Treatment of Refractory or Relapsed Small-Cell Lung Cancer", pages 1225-1229. *
See also references of EP0871447A4 *

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6664242B2 (en) 2000-02-28 2003-12-16 Aventis Pharma S.A. Composition and method comprising CPT-11 and a pyrimidine derivative for the treatment of cancer
US6503889B2 (en) 2000-02-28 2003-01-07 Aventis Pharma S.A. Composition comprising camptothecin and a pyrimidine derivative for the treatment of cancer
US6545010B2 (en) 2000-03-17 2003-04-08 Aventis Pharma S.A. Composition comprising camptothecin or a camptothecin derivative and a platin derivative for the treatment of cancer
US6548488B2 (en) 2000-03-17 2003-04-15 Aventis Pharma S.A. Composition comprising camptothecin or a camptothecin derivative and an alkylating agent for the treatment of cancer
US6960596B2 (en) 2000-03-17 2005-11-01 Aventis Pharma S.A. Composition comprising camptothecin or a comptothecin derivative and a platin derivative for the treatment of cancer
US6486320B2 (en) 2000-09-15 2002-11-26 Aventis Pharma S.A. Preparation of camptothecin and of its derivatives
US6562834B2 (en) 2000-10-27 2003-05-13 Aventis Pharma S. A. Combination comprising camptothecin and a stilbene derivative for the treatment of cancer
US6825194B2 (en) 2001-11-30 2004-11-30 Chugai Seiyaku Kabushiki Kaisha Hexacyclic compounds
WO2003045952A3 (en) * 2001-11-30 2003-10-16 Hoffmann La Roche Condensed camptothecins as antitumor agents
WO2003045952A2 (en) * 2001-11-30 2003-06-05 Chugai Seiyaku Kabushiki Kaisha Condensed camptothecins as antitumor agents
US7109195B2 (en) 2001-11-30 2006-09-19 Chugai Seiyaku Kabushiki Kaisha Hexacyclic compounds and their therapeutic use
US7910593B2 (en) 2004-04-09 2011-03-22 Chugai Seiyaku Kabushiki Kaisha Water-soluble prodrugs
US9763968B2 (en) 2004-12-22 2017-09-19 Rutgers, The State University Of New Jersey Hydrogel formulation for dermal and ocular delivery
US9999596B2 (en) 2004-12-22 2018-06-19 Rutgers, The State University Of New Jersey Controlled release hydrogels
US8022047B2 (en) 2005-08-22 2011-09-20 Chugai Seiyaku Kabushiki Kaisha Combination anticancer agents
WO2007113687A2 (en) 2006-03-30 2007-10-11 Diatos S.A. Camptothecin-cell penetrating peptide conjugates and pharmaceutical compositions containing the same
US8410045B2 (en) 2006-03-30 2013-04-02 Drais Pharmaceuticals, Inc. Camptothecin-peptide conjugates and pharmaceutical compositions containing the same
CN106588946A (en) * 2017-01-25 2017-04-26 郑州大学 10-HCPT (10-hydroxycamptothecine) derivative, synthesis method and application thereof
WO2023216956A1 (en) * 2022-05-13 2023-11-16 四川科伦博泰生物医药股份有限公司 Camptothecin compound, preparation method therefor and use thereof

Also Published As

Publication number Publication date
JPH11506453A (en) 1999-06-08
EP0871447A4 (en) 1999-06-23
US5663177A (en) 1997-09-02
EP0871447A1 (en) 1998-10-21

Similar Documents

Publication Publication Date Title
KR930009357B1 (en) Water soluble campothecin analogs and the preparation method thereof
KR100702085B1 (en) Camptothecin derivatives having antitumor activity
US5663177A (en) Water soluble camptothecin analogs
JP5890806B2 (en) N-substituted indenoisoquinoline and its synthesis
KR100484045B1 (en) Benzonaphthyridines as bronchial therapeutics
JP2978850B2 (en) 2-aminobenzazepine derivatives
CA2700824A1 (en) Heterocyclic compounds as crth2 receptor antagonists
US5670500A (en) Water soluble camptothecin analogs
FI67686B (en) PROCEDURE FOR THE FRAMEWORK OF THERAPEUTIC ANIMAL PRODUCTS
JP2001506270A (en) Prodrug form of camptothecin and novel homologs, and their use as pharmaceuticals
EP0555347A1 (en) SUBSTITUTED INDOLIZINO 1,2-b]QUINOLINONES
PT87604B (en) PROCESS FOR THE PREPARATION OF ANTI-ALLERGIC AND ANTI-INFLAMMATORY DI-HYDROPYRIDINIC AGENTS
WO1993016698A1 (en) SUBSTITUTED FURO[3',4':6,7]INDOLIZINO[1,2-b]QUINOLINONES
BRPI0707089A2 (en) camptothecin derivatives and their uses
KR100557804B1 (en) Indolomorphinane Derivatives and Remedies/Preventives for Cerebral Disorders
RU2561118C2 (en) Camptothecin derivatives, possessing antitumour activity
US6046209A (en) Water soluble camptothecin analogs
CZ2000711A3 (en) Optically pure camptothecin analogs, synthesis intermediates and process for preparing thereof
KR20050107756A (en) 7-imino derivatives of camptothecin having antitumor activity
ES2401563T3 (en) Methylenedioxybenzo [I] phenanthridine derivatives used to treat cancer
JP2008528552A (en) New indolopyridines, benzofuranopyridines and benzothienopyridines
US20100256177A1 (en) Flavopereirine derivatives for cancer therapy
CA2792613C (en) Phenoxy thiophene sulfonamides and their use as inhibitors of glucuronidase
RU2200163C2 (en) Carbocyclic analogs of 20(s)-camptothecin, methods of their synthesis, pharmaceutical composition, method of treatment
AU2005247032A1 (en) New benzo [b]pyrano[3,2-h]acridin-7-one cinnamate compounds, a process for their preparation and pharmaceutical compositions containing them

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref country code: JP

Ref document number: 1996 536757

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1996916929

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1996916929

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1996916929

Country of ref document: EP