CA2273497A1 - Delivery and activation through liposome incorporation of diaminocyclohexane platinum(ii) complexes - Google Patents

Delivery and activation through liposome incorporation of diaminocyclohexane platinum(ii) complexes Download PDF

Info

Publication number
CA2273497A1
CA2273497A1 CA002273497A CA2273497A CA2273497A1 CA 2273497 A1 CA2273497 A1 CA 2273497A1 CA 002273497 A CA002273497 A CA 002273497A CA 2273497 A CA2273497 A CA 2273497A CA 2273497 A1 CA2273497 A1 CA 2273497A1
Authority
CA
Canada
Prior art keywords
liposome
complex
composition
platinum
nddp
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.)
Abandoned
Application number
CA002273497A
Other languages
French (fr)
Inventor
Roman Perez-Soler
Abdul R. Khokhar
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.)
University of Texas System
Original Assignee
Roman Perez-Soler
Abdul R. Khokhar
The Board Of Regents Of The University Of Texas System
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 Roman Perez-Soler, Abdul R. Khokhar, The Board Of Regents Of The University Of Texas System filed Critical Roman Perez-Soler
Publication of CA2273497A1 publication Critical patent/CA2273497A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Abstract

Antitumor compositions are disclosed, as well as methods of preparing the compositions and methods of using them to inhibit tumor growth in mammals. The invention can take advantage of intraliposomal conversion of a platinum complex having formula (I) where R1 is diaminocycloalkyl, and R2 and R3 each have formula (a) where R4, R5, and R6 are each independently hydrocarbon moieties having from 1 to about 10 carbon atoms, into a complex having formula (II): R1-Pt-X2 where X is halogen.

Description

DELIVERY AND ACTIVATION THROUGH LIPOSOME INCORPORATION
OF DIAMINOCYCLOHEXANE PLATINUM(II) COMPLEXES
BACKGROUND OF THE INVENTION
The present invention relates to methods and compositions for the treatment of cancer.
One drug that has proven effective in the treatment of certain tumors is cisplatin (cis-dichlorodiamine-platinum(II)). However, cisplatin has certain disadvantages.
For example, its use in some circumstances is limited by its toxicity to the patient, especially its nephrotoxicity. As another example, tumors sometimes develop resistance to cisplatin.
~ o In an effort to overcome the disadvantages of cisplatin, researchers have synthesized and tested various other platinum complexes as potential antitumor agents. One such compound is dichloro( 1,2-diaminocyclohexane) platinum(II) (referred to in the remainder of this patent as "DACH-Pt-C12"). However, this compound has very low solubility in water, making it impractical for formulation and administration in aqueous solution.
Further, i s although various platinum complexes have been formulated in liposomes in the past, a liposornal formulation of DACH-Pt-C12 has not been developed because that complex is insoluble in most organic solvents. Although it has good solubility in dimethylformamide, that solvent has a very high boiling point. therefore making it impossible or impractical to prepare a liposomal formulation of DACI-I-Pt-C1, using standard evaporation methods.
2o Other platinum-based antitumor drugs, such as cis-bis-neodecanoato-trans-R,R-1,2-diaminocyclohexane platinum (II) (NDDP) have been prepared and tested as antitumor agents. However, a need still exists for improved antitumor drug formulations that have good antitumor activity, low toxicity to non-cancerous cells in a patient, and desirable storage characteristics. -2s SUMMARY OF THE INVENTION
The present invention relates to a liposomal antitumor composition and to methods of using the composition to inhibit tumor growth in mammals. The invention also concerns methods of preparing the antitumor composition.
SUBSTITUTE SHEET (RULE 26) WO 98/24416 ~ PCT/US97/22359 _2_ The present invention can take advantage of intraliposomal conversion of a platinum complex having the formula RI-Pt/
R3 (I) s into a complex having the formula R~-Pt-Xz (II) io where X is halogen. This makes it possible to prepare liposomal formulations of the complex (II), which had not been practical previously due to its low solubility in water and most organic solvents.
In the above complexes, R, is diaminocycloalkyl, preferably a 1,2-diaminocycloalkyl group having from about 3 to about 6 carbon atoms, most preferably 1,2-i ~ diaminocyclohexane. R, and R3 each have the formula where R4, R5, and R6 are each independently hydrocarbon moieties having from 1 to about 10 2o carbon atoms, preferably alkyl having from 1 to about 6 carbon atoms, most preferably alkyl --having from 1 to about 3 carbon atoms. R2 and R3 can be the same but do not have to be the same. Likewise, R4, R;, and R6 can be the same but do not have to be the same.
X is most preferably chlorine.
SUBSTITUTE SHEET (RULE 26) .._ ~_ _._~____....T.. __~._~_ __ One aspect of the present invention is a liposomal antitumor composition that comprises the complex (II) entrapped in a liposome. In a particular embodiment of the invention, the liposome comprises an acidic phospholipid, for example dimyristoyl phosphatidyl glycerol. Without being bound by theory, it is believed that the presence of the acidic phospholipid in the liposome enhances or accelerates the conversion of the complex (I) to the complex (II).
Another aspect of the present invention is a method of inhibiting tumor growth. The method comprises administering to a mammal a composition that comprises the complex (II) entrapped in a liposome. with the platinum complex being present in an amount effective to io inhibit tumor growth.
Another aspect of the present invention is a method of preparing the antitumor composition. This method comprises the step of adjusting the pH of a composition that comprises the platinum complex (I) entrapped in a liposome, whereby the pH is made somewhat acidic, preferably between about 2 and about 6.5, resulting in the conversion of the ~ s complex (I) into the complex (II). The resulting composition can then be administered to a patient.
In one particular embodiment of this method, the complex (I) is converted to the complex (II) within the liposome. This allows a liposomal formulation of the complex (I) to be manufactured and stored. and then shortly before administration to a patient, the liposomal Zo formulation of complex (I) can be converted to a liposomal formulation of complex (II) in situ by simply adding an acidic solution to the formulation. Optionally, after a predetermined time has passed since the addition of the acidic solution, the pH can be readjusted, preferably to at least about 7, in order to stop the conversion of complex (I) to (II).
A broader aspect of the invention concerns a method of delivering a biologically zs active chemical moiety internally to a mammal. The biologically active moiety can be, for example, an antitumor agent. The method comprises (a) providing an aqueous formulation of a prodrug of the biologically active moiety, the prodrug being entrapped in a liposome, and the prodrug further being capable of forming the biologically active moiety upon exposure to a solution having an acidic pH; (b) reducing the pH to an acidic level, thereby converting the SUBSTITUTE SHEET (RULE 26) prodrug to the biologically active compound; and (c) administering the aqueous formulation to a mammal. Administration of the liposomal formulation to the mammal can suitably be done after the conversion of the prodrug, but it might also be done before conversion, such that the conversion would then occur in vivo, for example due to acidic components (e.g., s acidic phospholipids) of the Iiposome.
The present invention has a number of advantages over prior art platinum antitumor formulations and methods, including better antitumor activity, greater potency, and reduced toxicity to non-cancerous cells of the patient. Further, the compositions of the present invention permit the formulation in liposomes and the delivery of platinum complexes that io could not be so formulated in the past.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. Chemical structure of NDDP, B 10, and L 10.
Figure 2. Effect of structure of Pt-complex, DMPG content, and aqueous solution on the intraliposomal stability and cytotoxicity of NDDP ( ~ ), B I 0 ( O ), and L 10 ( ~ ). Fig.
~s 2A -- DMPC:DMPG=7:3, saline; Fig. 2B -- DMPC:DMPG=7:3, PBS; Fig. 2C --DMPC:DMPG=3:7, saline; Fig. 2D -- DMPC:DMPG=3:7, PBS. IC;o values are in g/ml.
Figure 3. Effect of lipid composition on the intraliposomal stability of NDDP.
0.9%
NaCI aqueous solution (starting pI I=7.0) was used as reconstituting solution of liposomes and the final pH was checked at 6 h after liposome preparation.
20 ° Figure 4. ~9'Pt NMR of Iiposomal NDDP suspension. Fig. 4A -- ~9'Pt NMR of Sample 1 in chloroform prepared by extraction with CHCl3 from liposomal NDDP
suspension reconstituted in saline and kept for 6 h at room temperature. Fig.
4B -- ~9'Pt NMR of Sample 2 in CH30H prepared by reconstitution of liposomal NDDP in saline for 6 h, lyophilization of water for 2 d, and redissolution of mixtures in CH30H. Fig.
4C -- ~95Pt is NMR of Sample 3 in CH30H prepared by evaporation of t-butanol and redissolution of mixtures in CH30H.
Figure 5. Fig. SA -- IH-~H correlated spectroscopy of DACH-Pt-C1, in DMF-d~.
Fig.
5B --'3C NMR of DACH-Pt-C12 in DMF-d~.
SUBSTITUTE SHEET (RULE 26) _.__ ___ _ _.____ ___ T _ _ ._.

WO 98/24416 ~ PCT/US97/22359 Figure 6. Sample preparation for NMR tracking experiment of liposomal NDDP.
Fig. 6A -- with lipids in chloroform solutions. Fig. 6B -- with lipids in dry powder.
DESCRIPTION OF SPECIFIC EMBODIMENTS
Cis-bis-neodecanoato-trans-R,R-1,2-diaminocyclohexane platinum (II) (NDDP) is a lipophilic platinum complex (Pt-complex) that can be formulated in a liposomal carrier.
Various details about the making and use of NDDP and other platinum complexes are disclosed in U.S. patents 5,041,581, 5,1 I 7,022, 5,186,940, 5,178,876, and 5,384,127. Those patents are incorporated here by reference.
Prior studies have suggested that NDDP is a pro-drug that exerts its biological activity io through activation within the iiposome bilayers. In order to understand the kinetics of the intraliposomal degradation/activation of different liposomal Pt-complexes, we studied the effects that the structure of the Pt-complex, pH, temperature, lipid composition, content of acidic phospholipids, liposome size, and presence of residual chloroform have on the stability, in vitro cytotoxicity, and in vivo antitumor activity of different liposomal i s Pt-complex preparations. The fol lowing factors were found to enhance the intraliposomal degradation/activation of Pt-complexes: i ) the size and spatial configuration of the Pt-complex, 2) an acidic pH, 3) a high temperature, 4) the presence and amount of acidic phospholipids, and 5) the presence of residual chloroform. Liposome size did not affect the intraliposomal stability of different Pt-complexes.
zo A good inverse relationship between the extent of drug degradation and in vitro cytotoxicity and between the extent of drug degradation and in vivo antitumor potency was observed, thus confirming that the biological activity of these complexes is exerted through the intraliposomal formation of certain active intermediate(s). The only active intermediate that could be identified was cis-bis-dichloro-trans-R,R-1,2-diaminocyclohexane platinum (II) zs whose structure was confirmed by 'H, 13C, and ~95Pt nuclear magnetic resonance (NMR) spectroscopy.
We have previously developed liposomal formulations of lipophilic Pt-complexes for in vivo administration [5,6] and have studied their chemical and biological properties. (1,4,7]
The general structure of the preferred Pt-complexes used is [DACH-Pt-Rz], where DACH is SUBSTITUTE SHEET (RULE 26) trans-R,R-1,2-diaminocyclohexane and R is a lipophilic carboxylato group. The Pt-complex is thought to intercalate between the phospholipid molecules of the lipid bilayers of the liposomes. The most remarkable characteristic of these complexes is that they are not cross-resistant with cisplatin, both in vitro and in vivo. [ 1,5] The leading formulation, s liposomal cis-bis-neodecanoato-DACH-platinum (II) (NDDP) uses large liposomes composed of dimyristoylphosphatidyl choline (DMPC) and dimyristoylphosphatidyl glycerol (DMPG) at a 7:3 molar ratio and is now in clinical trials. Interestingly, liposomal-NDDP
must undergo a chemical degradation/activation process into an active intermediates) within the Iiposomes shortly after iiposome preparation in order to exert its antitumor activity. [7]
io We have previously reported that this chemical reaction depends on the content of DMPG in the lipid bilayer, and based on this finding, we hypothesized that a DMPG-Pt complex might be one of the active intermediates. We have also reported that the structure of the Pt-complex has an effect on the intraliposomal drug stability: the compounds with linear and short carboxylato leaving groups are more stable and less potent than the compounds with i a branched or longer linear leaving groups. [8] A full characterization of the active intermediates) as well as the different factors that influence the degradation/activation process is mandatory for the development of one of these agents as a pharmaceutical product.
For that purpose, we selected NDDP (highly branched structure) and two isomers, B 10 (minimally branched structure) and L 10 (linear structure) (Figure I ) and studied the 2o relationship between their biological activity and their intraliposomal stability. We examined the effect of pH, temperature, lipid composition, liposome size, and presence of residual chloroform on the degradation of the Pt-complexes, and attempted to identify the active intermediates) by tracking experiments using 3 ~ P and ~ 95Pt NMR spectra. Our results indicate that the degradation/activation of these Pt-complexes is greatly dependent on the pH
2~ of the suspension, and that DACH-dichloroplatinum (DACH-Pt-C12) is the only intermediate that can be identified. thus suggesting that these Pt-complexes are prodrugs of DACH-Pt-Cl, when incorporated in liposomes suspended in saline.
SUBSTITUTE SHEET (RULE 26) _ _ ~__.....__~......_T_.

_7_ MATERIALS AND METHODS
Preparation of liposomal Pt-complex.
NDDP, B 10, and L I 0 were synthesized as previously described [ 1,2) and recrystallized in acetone. DMPC, DMPG, dioleyl phosphatidyl choline (DOPC), dioleyl s phosphatidyl glycerol (DOPG), phosphatidic acid (PA), phosphatidyl ethanolamine (PE), and phosphatidyl serine (PS) were purchased from Avanti Polar Lipids (Alabaster, AL).
Multilamellar vesicles containing Pt-complexes were prepared by the lyophilization method using the lipids in chloroform solutions or in dry powder.
Method 1: With chloroform solutions of lipids [1 ), lipids were mixed at the desired io molar ratio. and the chloroform was removed in a rotary evaporator. To the dried lipid film, t-butanol solutions of Pt-complex were added and shaken at 40°C for 10 min. The solutions were then frozen in a dry-ice-acetone bath, and t-butanol was removed by lyophilization overnight to give lyophilized preliposomal powder.
Method 2: With lipids in dry powder, lipids were mixed and dissolved in i s t-butanol: water ( 10:1 /v:v). To this solution, t-butanol solutions of Pt-complex were added, and the rest of the procedure was the same as described above.
Saline or PBS was added (1 ml/mg of Pt-complex) to reconstitute the lyophilized preliposomai powder. and the suspension was hand-shaken for 10 min to obtain large-size liposomes. Small-size liposomes were prepared by sonication of large-size liposomes for 1 2o min with an ultrasonic cell disrupter (Laboratory Supplies Co., New York, NY). The size distribution of the different liposomal preparations was determined with a Nicomp Submicron Particle Sizer Model 370 (Nicomp Particle Sizing Systems, Santa Barbara, CA).
Intraliposomal stability.
Stability of the different Pt-complexes incorporated in liposomes was determined as zs described previously [8] by comparing the HPLC profiles as a function of time. In brief, aliquoted samples of liposome suspension were diluted (7x) with methanol at 0, 2, 6, and 24 h after liposome preparation, and each sample was then monitored by HPLC using chromega-8 bond column (4.6mm x 25cm, 8 Tm: ES Industries, NJ) and 10% water-methanol as eluant.
The flow rate was I ml/min, and the complexes were detected by UV at 224 nm wavelength.
SUBSTITUTE SHEET (RULE 26) _g_ Biological activity.
In vitro. The in vitro cytotoxicity of liposomal Pt-complexes against A2780 human ovarian carcinoma cells was assessed by the MTT dye reduction assay. In brief, A2780 cells were seeded in 96-well plates, allowed to attach overnight, and then exposed to various s concentrations of drugs for 20 h. After washing the cells with PBS, fresh medium was added for 52 hours and the cell survival fractions determined by MTT assay.
In vivo. The in vivo antitumor activity of liposomal Pt-complexes was assessed against intraperitoneal L 1210 mouse leukemia. Groups of 6-8 mice weighing 18-20 g were inoculated with 106 cells (0.2 ml, i.p.) on day 0, and treatment (25, 50, 100 and 1 SO mg/kg) i o was started on day 1 (0.15-0.5 ml, i.p.). The results were expressed as the median survival of treated animals divided by the median survival of control animals x 100 (%
T/C).
Identification of active intermediates.
To characterize the active intermediates) in the reaction cascade of L-NDDP
reconstituted in saline (0.9% NaCI), tracking experiments using ~9'Pt NMR
spectra in i s combination with 3 ~ P NMR were performed. The procedures for the preparation of the samples is summarized in Fig. 6. Samples 1-3 were prepared using the lipids DMPC and DMPG purchased in chloroform solution, while samples 4-8 were prepared using lipids in dry powder. In samples 1-3, the chloroform was initially evaporated in a rotavapor. The lipid f lm was dissolved in t-butanol containing the NDDP in solution. An aliquot of this solution 2o was kept at 40°C for 6 h and then lyophilized and extracted with methanol (sample 3). The remaining was lyophilized immediately, thus resulting in a preliposomal powder, which was reconstituted with saline to produce the liposome suspension. The liposome suspension was kept at room temperature for 6 hours. The Pt compounds and lipids were then extracted with chloroform (sample 1 ) or the sample was lyophilized to eliminate the water and the powder zs dissolved in methanol (sample 2) (see Fig. 6). Samples 4, 5, and 6 were prepared by complete evaporation of solvents after keeping the samples at 40°C for 6 h, and redissolving them in methanol. Samples 7 and 8 were prepared by lyophiIization of water for 1-2 d and redissolution in methanol. All samples were prechecked by HPLC before tracking with SUBSTITUTE SHEET (RULE 26) _~ .~_._. ___.~....._ ...T.. ..._._.~r___._ .

NMR. Chemical shifts of the products are expressed in parts-per-million relative to Na,PtClb in ~95Pt and DMPC in 3'P NMR.
DA CH Pt-Cl, characterization.
Yellow precipitates from NMR samples were collected and redissolved in DMF-d~
to s characterize them with ~H and'3C NMR. ~H NMR (DMF-d~): 1.13-1.17 (m. 2H), 1.46-1.55 (m, 4H), 2.05-2.09 (broad, 2H) 2.55-2.59 (m, 2H), and 5.07 and 5.63 (broad s.
2 NH2) ppm.
~3C NMR (DMF-d7): 24.9 (C4, C5), 32.3 (C3, C6), and 64.1 (C1, C2) ppm. ~9'Pt NMR
(CHC13, CH;OH) 1950 ppm, (DMF) 2250 ppm (strong single peak). Elemental analysis:
Calc. C( 18.99). H(3.69), N(7.38), Pt(51.50); Found C( 18.58), H(3.72), N(7.40), Pt(51.30).
io All these data were confirmed by an authentic sample of DACI-1-Pt-C12.
RESULTS
Preparation of liposomal Pt-complexes.
NDDP and its two isomers, B 10 and L 10 were formulated in liposomes composed of combinations of various lipids including DMPC, DMPG, DOPC, DOPG, PA, PE, and PS.
i ~ The liposomes were formed by reconstituting preliposomal powders containing the Pt-complex and the lipids with unbuffered 0.9% NaCI aqueous solution (saline) or phosphate-buffered saline (PBS). The entrapment efficiency (%EE) of all liposomal formulations was >90% and was not significantly affected by the lipid composition, reconstitution solution, or NDDP isomer used. No crystals of free drug were observed in any 20 of these preparations within 24 h as assessed by optic microscopy. The median size of the multilamellar vesicles was 1-2 Tm in all preparations. The median size of the small-size liposomes prepared by ultrasonication of multilamellar vesicles was 50-100 nm.
Intraliposomal Stability of Pt-complexes.
1 ) Role of the spatial configuration of the Pt-complex and pH of the liposome - is suspension.
Figs. 2A-D show the stability of liposomal Pt-complex formulations using saline or PBS as the reconstitution solution and DMPC:DMPG ratios of 7:3 and 3:7. As observed previously [ 1 ]. the branched configuration of the leaving group of the Pt-complex and the content of DMPG in the lipid bilayers correlated with a higher rate of degradation of the SUBSTITUTE SHEET (RULE 26) Pt-complex. As a result, the complex with a linear leaving group L 10 was highly stable, while the highly branched NDDP was rather unstable, and the minimally branched B 10 had an intermediate stability. The use of PBS as the reconstitution solution resulted in a significantly higher stability of the Pt-complexes as compared with saline.
For example, 6 h a after liposome preparation, the percentages of intact NDDP in saline versus PBS were 43.7%
vs 82.1 %, whereas the percentages for B 10 were 85.0% vs 95.9%, and the percentages for L 10 93.1 % vs I 00%, respectively. The pH of the liposome suspension in saline decreased from 7.0 to 3.8-6.2 depending on the Pt-complex, whereas PBS held the pH of the solution to around 6.0-7.0 in all cases. These results indicate that: 1 ) an acidic pH
enhances the Eo intraliposomal degradation of the Pt-complexes, and 2) a good neutral buffer system can reduce or stop the intraliposomal degradation of the Pt-complexes. To confirm these results, we tested the intraliposomal stability of the Pt-complexes in strongly acidic (pH=3.0) or basic (pH=8.0) saline solutions prepared by adding 0.1 N HCI or NaOH aqueous solution to pH 7.0 saline. The pH 3.0 saline increased the degradation rate of all Pt-complexes, whereas the pH
i s 8.0 saline did not induce any significant Pt-complex degradation even at 24 h after liposome preparation. All formulations using a relative higher amount of DMPG
(DMPC:DMPG=3:7) displayed a higher rate of Pt-complex degradation in good correlation with the pH of the liposome suspension. because DMPG is an acidic phospholipid.
2) Role of temperature.
zo The intraliposomal stability of the Pt-complexes was checked at 40°C
and compared with results obtained at room temperature. Pt-complex degradation was temperature-dependent, with the degradation rates being about 30-70% higher at 40°C than at 25°C, depending on the Pt-complex tested.
3) Role of lipid composition.
zs Liposomal formulations of NDDP using DMPC:PA, PS:DMPG, and DMPC:PE at a 7:3 molar ratio, and DOPC:DOPG at 1:0, 7:3, 3:7, and 0:1 molar ratios were prepared and tested using saline as the reconstitution solution (Figure 3). The acidity of phospholipids (PA>PG>PS) and the relative DOPG content (DOPC:DOPG 0:1>3:7>7:3>I:0) enhanced the SUBSTITUTE SHEET (RULE 26) _ ~_.._ __. _..~_ __.~_T_. ._w_..._. _.~..~..~_ WO 98/24416 PCT/iJS97/22359 intraliposomal degradation of NDDP and a good correlation between Pt-complex degradation and acidic pH was again observed.
The same conclusion was drawn in studies with NDDP in liposomes with the same DMPC : DMPG molar ratio (7:3 ) but different NDDP aotal lipid ratios ( 1: 5, 1:10, 1:15, and s 1:30) and, therefore, different DMPG contents. Using a 1:5 or I :10 ratio, 85% of initial NDDP was present at 24 hours; in contrast, using a 1: I S or 1:30 ratio resulted in an enhanced NDDP degradation, with only 25% of the original NDDP remaining at 24 h, thus suggesting a correlation between extent of degradation and absolute amount of DMPG within the lipid biiayers.
~0 4) Role of liposome size.
Liposome size did not affect the intraliposomal stability of NDDP:
ultrasonication of the original suspension of multilamellar vesicles did not significantly change the stability of NDDP regardless of the lipid composition used.
Correlation between in vitro cytotoxicity and intraliposomal stability.
~s We studied the in vitro cytotoxicity of different liposomal Pt-complex preparations against A2780 cells with the MTT assay and correlated the results with the intraliposomal stability of the Pt-complex. Results are shown in Figs. 2A-D. The IC;o values correlated fairly well with drug stability: the more stable the Pt-complex, the less toxic or higher the IC;o. When about 20%, 50%, and 90% of the original Pt-complex remained at 6 h, the IC;~) 2o was approximately 3-5, 7-10, and 20-SO g/ml, respectively. These results indicate that the intraliposomal degradation of the Pt-complex is required to exert its cytotoxic effect and is, therefore, an intraliposomal activation step.
Identifrcation of active intermediates) of NDDP.
No new peaks corresponding to the degradation products are observed by the HPLC
z> method developed for NDDP, either because they elute with the phospholipids or they do not have UV absorbence. Attempts to separate any new peaks from the lipid peaks have been so far unsuccessful.
SUBSTITUTE SHEET (RULE 26) To keep track of the reaction cascade of liposomal NDDP, we tried to apply the NMR
tracking technique used by other researchers [2, 3, 9-11 ] to characterize the degraded/activated products of NDDP.
1) Formulations prepared using lipids in chloroform solution (samples 1, 2, and 3 s of Fig. 6A).
By ~9'Pt NMR of sample 1 (saline, chloroform extraction, Figure 4A) and sample (saline, lyophilization, Figure 4B), the NDDP peak was detected at 1750 ppm and a new peak corresponding to DACH-Pt-Clz was detected at 1950 ppm. Prolonging the reaction time, lowering the pH, increasing the temperature, and increasing the amount of DMPG
in the io liposomes enhanced the degradation/activation of NDDP, increasing the intensity of the peak at ~ 9'0 ppm. However, by 3 ~ P NMR, no new peaks were observed except those corresponding to DMPC (2 ppm) and DMPG (3 ppm), indicating that the new product shown by ~9sPt NMR
is not a DMPG-incorporating Pt-complex.
These spectra results were similar to those from sample 3 (Fig. 6A, t-butanol, i s lyophilization, Figure 4C), in which liposomal NDDP was not exposed to saline, thus indicating that the presence of CHC13 can also act as a donor of chloride to form DACH-Pt-C(z. After storing the yellowish NMR samples at 4°C overnight, yellow crystals slowly precipitated. The precipitates were filtered, dried, and the structure of the compound was proved to be DACH-Pt-Clz by ~H, CUSY (Figure SA),'3C (Figure SB), and ~y'Pt NMR.
zo A yellow precipitate was also observed 2-3 weeks after leaving the original liposomal NDDP
suspension at room temperature. NDDP in liposomes composed of only DMPC did not give any new peaks by either I9'Pt or 31 P NMR, which correlates with its completely preserved stability in the absence of DMPG.
2) Formulations prepared using lipids in dry powder (samples 4, 5, 6, 7, and 8).
zs We performed the same tracking experiment with formulations prepared using lipids in dry powder instead of chloroform solutions to eliminate the influence of the presence of _. residual CHC13 on the degradation of NDDP (Fig. 6). In samples 4 and 5 (solvent t-Butanol +
water and methanol, respectively) containing lipids and NDDP, no reactions occurred, whereas sample 6 (chloroform solution of lipids and NDDP) showed the presence of SUBSTITUTE SHEET (RULE 26) -IJ-DACH-Pt-Cl2 by ~ 9'Pt NMR. The results with these samples, which were incubated at 40°C
for 6 h, confirm that the presence of chloroform can induce the degradation of NDDP into DACH-Pt-C12. Sample 7 was prepared by reconstitution of preiiposomal NDDP
powder in saline of pH 6.5-7.0 for 6 h at room temperature. No significant degradation (<5%) was observed, whereas when reconstituted in acidic saline of pH 3.0-4.0 (sample 8), a 60-95%
degradation of NDDP occurred in I O min yielding DACH-Pt-Cl2 as determined by ~95Pt NMR and HPLC (Table 1 ).
SUBSTITUTE SHEET (RULE 26) Table 1 Effect of various pH values of saline on intraliposomal stability of NDDP prepared with lipids as dry powder.
pH of saline _________________________NDDP stability (%)_________________________ Time after reconstitution lOmin 1 h 2h 6h 3.0 43 -- 15 7 a 5.0 80 72 67 38 7.0 100 -- 95 92 Relationship between Pt-complex stability and in vivo antitumor activity.
1. Preparations containing residual chloroform ~s These studies were done with liposomal Pt-complex suspensions prepared using lipids dissolved in chloroform. In in vivo antitumor activity studies against L 1210 leukemia, an inverse relationship between drug stability and antitumor potency was observed (Table 2).
The DMPC:DMPG ratio was 7:3 for all preparations. The starting pH value of the saline and PBS was 7Ø The tumors were inoculated i.p. on day 0, followed by drug injection i.p. on ~o day 1.
SUBSTITUTE SHEET (RULE 26) __.__ __ ___.... __.r.. _.______ WO 98/24416 ~ ' PCT/US97/22359 Table 2 In vivo antitumor activity of liposomal Pt complexes against L 1210 leukemia.
Pt complex Dose of Pt complex (,ng~b) ____________-____%T/C_________________ s Reconstitution solution Saline PBS

100 Toxic 200 150 Toxic 150 to is 150 123 100 Cisplatin l0 142 The optimal doses of liposomal NDDP were 50 mg/kg in saline and 100 mg/kg in PBS (%T/C = ? 1 1 and 200, respectively). For liposomal B 10 and L 10, the optimal dose was 100 mg in saline (%T/C = 228 and i 78), but no significant antitumor activim was observed zo when both drugs were reconstituted in PBS. In conclusion, the most potent liposomal Pt-complex preparations are those with the lowest stability of the Pt-complex.
However, all formulations had similar antitumor activity when administered at the optimal dose.
2. Preparations not containing residual chloroform These studies were done with liposomal Pt-complex suspensions prepared with dry zs lipids. The relationship between NDDP stability and antitumor activity was again studied using the in vivo L 1210 leukemia model. Table 3 shows the results with formulations reconstituted with saline solutions of different pH and administered at different time points after reconstitution. (Values are the means of two separate experiments.) SUBSTITUTE SHEET (RULE 26) Table 3 Antitumor of liposomal activity NDDP against leukemia.

Drug pH of salineDose of Pt complex _________________%T/C_________________ ~mi~g) Time of drug administration 10 min 2 h 6h NDDP 3.0 12.5 157 0 50 Toxic NDDP 5.0 12.5 186 100 Tox is NDDP 7.0 25 186 Cisplatin 10 150 The optimal doses of liposomal NDDP reconstituted with salines of pH 3.0, 5.0, and 20 7.0 were 25, 50, and I 00 mg/kg, respectively. Therefore, the lower the pH, the higher the potency of the preparation in good correlation with the increased intraliposomal drug degradation/activation. At the optimal doses, the %T/C obtained were 2 I 4, 271, and 271, respectively. Delaying the time of drug administration increased the antitumor activity of the formulations, in good correlation with the increased drug activation with time. However, 2s antitumor activity did not correlate perfectly with the calculated amount of activated Pt-species formed intraliposomally during the activation process. For example, 100 mg/kg at pH 7.0 gave a similar %T/C value as 50 mg/kg at pH 5.0, although only 10% of NDDP at pH
7.0 ( 10 mg/kg) and about 70% at pH S.0 (3 5 mg/kg) are transformed into the active Pt-species under those conditions. Further in vivo activation must, therefore, occur to explain these 3o discrepancies.
SUBSTITUTE SHEET (RULE 26) _ _ _.__~ .. _ __~_._.~. __._.~ _.~.. ._ DISCUSSION
Our results indicate that NDDP and its isomers are prodrugs of DACH-Pt-C12 when entrapped in liposomes containing acidic phospholipids and in the presence of sodium chloride or residual chloroform as donors of chloride. The rate of transformation of NDDP
into DACH-Pt-C12 is directly related to the pH of the liposome suspension. The studies performed suggest that DMPG and other acidic phospholipids enhance the reaction by providing an acidic milieu within the liposome membranes. No evidence could be generated to support a direct reaction between NDDP and DMPG to form a DACH-Pt-DMPG
complex as one of the active intermediates of NDDP, as we had previously hypothesized, nor the io formation of DACH-Pt aquated species.
DACH-Pt-CIZ is the leading compound of the DACH family of Pt-complexes.
However, it was never developed because of a lack of solubility in water. We initially considered this compound for liposome entrapment but determined it to be an inappropriate drug for liposome formulation because it is insoluble in most organic solvents. DACH-Pt-Cl ~ s has only a good solubility in dimethylformamide (DMF), which has a very high boiling point and, therefore, can not be used to prepare liposomes using the standard evaporation methods and it is not soluble in any of the organic solvents used for the lyophilization methods. Our studies indicate that DACH-Pt-CI, can be generated within the Iiposome membranes under the conditions described and that the drug remains liposome-bound without leaking out and zo crystallizing for at least 24 h. In contrast, DACH-Pt-Cl2 precipitates quickly when formed from NDDP by the addition of HCI. These results constitute the first example of a liposome formulation in which the compound is synthesized in situ from an entrapped precursor and the liposornes prevent its spontaneous precipitation. The results are encouraging because they may suggest an avenue for the development of a much needed delivery system for this 2s very interesting compound.
A potential strategy is to use a two-step reconstitution procedure by which an acidic saline solution is used first to induce the fast transformation of >80% of NDDP into DACH-Pt-C12, followed after a predetermined period of time by the addition of a buffer solution to bring the pH to >7.0 and to stop the reaction.
SUBSTITUTE SHEET (RULE 26) WO 98!24416 PCT/US97/22359 Liposomes in accordance with the present invention can be prepared from various amphipathic substances including natural or synthetic phospholipids. Numerous suitable phospholipids are well known in the art. The liposomes of the present invention can be multilamellar, unilamellar, or have an undefined lameliar construction. A
pharmaceutical s composition comprising such liposomes can include a pharmaceutically acceptable carrier or diluent, as well as other pharmaceutically acceptable adjuvants.
Liposome compositions of the present invention can be used to inhibit the growth of tumor cells in mammals, particularly in humans. The compositions of the_present invention should be useful for treatment of various human malignancies, in particular any piatinum-io sensitive cancer, including ovarian, testicular, lung, head and neck, esophageal, and bladder tumors, sarcomas, lymphomas, and mesotheliomas. Methods of using the compositions of the present invention involve administering to a mammal an amount of the compositions effective to inhibit tumor growth. The administering step can suitably be parenteral and by intravenous, intraarterial, intramuscular, intralymphatic, intraperitoneal, subcutaneous, is intrapleural, or intrathecal injection, or by topical application or oral dosage. Such administration is preferably repeated on a timed schedule until tumor regression or disappearance has been achieved, and may be used in conjunction with other forms of tumor therapy such as surgery or chemotherapy with different agents. The dose administered of a composition in accordance with the present invention is preferably between approximately zo 100 and 750 mg/kg of body weight of the mammalian subject to which it is administered.
The preceding description of specific embodiments of the present invention is not intended to be a complete list of every possible embodiment of the invention.
Persons skilled in this field will recognize that modifications can be made to the specific embodiments - described here that would be within the scope of the present invention.
SUBSTITUTE SHEET (RULE 26) REFERENCES
2 The following references, to the extent that they provide exemplary procedural or 3 other details supplementary to those set forth herein, are specifically incorporated herein
4 by reference.
s 6 1. Han, L; ling, Y.-H.; Al-Baker, S.; Khokhar, A.R.; Perez-Soler, R.
Cellular pharmacology of liposomal cis-bis-neodecanoato- traps-R,R-1,2-s diaminocyclohexane- platinum(II) in A2780/S and A2780/PDD cells. Cancer 9 Res. 1993, 53, 4913-4919.
~0 2. Hopis, L. S.; Miller, A. V.; Amundsen, A. R.; Schurig, J.E.; Stern, E.W.
~ i cis-Diamineplatinum(II) complexes containing phosphono carboxylate ligands as iz antitumor agents. J. Med. Chem. 1990, 33, 105-111.
3. Ismail, I. M.; Sadler, P. J. ~9sPt- and ~sN-NMR studies of antitumor complexes.
is In: Platinum, gold, and other metal chemotherapeutic agents; Lippard, S. J.
(eds.) is American Chemical Society. 1983; pp 171-189.
i 6 4. Khokhar, A.R.; Al-Baker, S.; Brown, T.; Perez-Soler, R. Chemical and biological m studies on a series of lipid-soluble {traps-(R,R)- and -{S,S)-1,2-~ a diaminocyclohexane) platinum(II). J. Med. Chem. 1991, 34, 325-329.
i9 5. Perez-Soler, R.; Yang, L.Y.; Drewinko, B.; Lauterzstain, J.; Khokhar, A.R.
20 Increased cytotoxicity and reversal of resistance of cis-diamminedichloro z~ , platinum(II) with entrapment of cis-bis-neodecanoato- traps-R,R-1,2-zz diaminocyclohexaneplatinum(II) in multilamellar lipid vesicles. Cancer Res.
23 1988, 48, 4509-4512.
za 6. Perez-Soler, R.; Khokhar, A. R.; Lautersztain, J.; Al-Baker, S.;
Francis, K.;
zs Macias-Kiger, D.; Lopez-Berestein, B. Clinical development of liposomal z6 platinum. J. Liposome Res. 1990, 1, 437-449.
z~ 7. Perez-Soler, R.; Khokhar, A. R. Lipophilic cisplatin analogues entrapped in zs liposomes: role of intraliposomal drug activation in biological activity.
Cancer z9 Res. 1992, 52, 6341-6347.

I 8. Perez-Soler, R.; Han, L; Al-Baker, S.; Khokhar, A. R. Lipophilic platinum 2 complexes entrapped in liposomes: improved stability and preserved antitumor 3 activity with complexes containing linear alkyl carboxylato leaving groups.
a Cancer Chemother. Pharmacol. 1994, 33, 378-384.
s 9. Qu, Y.; Farrell, N. J. Effect of diamine linker on the chemistry of bis(platinum) 6 complexes. A comparison of the aqueous solution behavior of 1,4-butanediamine 7 and 2,5-dimethyl-2,5-hexanediamine complexes. J. Inorg. Biochem. 1990, 40, s 255-264.
9 10. Qu, Y.; Farrell, N. J. Interaction of bis(platinum) complexes with the to mononucleotide 5'-guanosine monophosphate. Effect of diamine linker and the i I nature of the bis(platinum) complex on product formation. J. Am. Chem.
Soc.
12 1991, 113, 4851-4857.
13 11. Slavin, L. L.; Bose, R. N. Phosphonato complexes of platinum(II):
kinetics of la formation and phosphorus-31 NMR characterization studies. J. Inorg.
Biochem.
Is 1990, 40, 339-347.

_. ___.....__.~~._.__.. ~_.~.___...._ __..

Claims (36)

CLAIMS:
1. A liposomal antitumor composition, comprising a platinum complex having the formula R1-Pt-X2 entrapped in a liposome, where R1 is diaminocycloalkyl and X is halogen.
2. The composition of claim 1, where R1 has from about 3 to about 6 carbon atoms.
3. The composition of claim 1, where R1 is 1,2-diaminocyclohexane.
4. The composition of claim 1, where X is chlorine.
5. The composition of claim 1, where the liposome comprises an acidic phospholipid.
6. The composition of claim 1, where the liposome comprises dimyristoyl phosphatidyl glycerol.
7. The composition of claim 1, where the platinum complex is intercalated between bilayers of the liposome.
8. A liposomal antitumor composition, comprising a platinum complex having the formula DACH-Pt-Cl2 intercalated between bilayers of a liposome, where DACH is diaminocyclohexane;
and where the liposome further comprises dimyristoyl phosphatidyl glycerol.
9. A method of inhibiting tumor growth, comprising:
administering to a mammal a composition that comprises a amount effective to inhibit tumor growth of a platinum complex having the formula R1-P t-X2 entrapped in a liposome, where R1 is diaminocycloalkyl and X is halogen.
10. The method of claim 9, where R1 has from about 3 to about 6 carbon atoms.
11. The method of claim 9, where R1 is 1,2-diaminocyclohexane.
12. The method of claim 9, where X is chlorine.
13. The method of claim 9, where the liposome comprises an acidic phospholipid.
14. The method of claim 9, where the liposome comprises dimyristoyl phosphatidyl glycerol.
15. The method of claim 9, where the complex is intercalated between bilayers of the liposome.
16. A method of inhibiting tumor growth, comprising:
administering to a mammal a composition that comprises a amount effective to inhibit tumor growth of a platinum complex having the formula DACH-P t-Cl2 intercalated between bilayers of a liposome, where DACH is diaminocyclohexane, and where the liposome further comprises dimyristoyl phosphatidyl glycerol.
17. A method of preparing an antitumor composition, comprising:
adjusting the pH of a composition that comprises a platinum complex having the formula entrapped in a liposome, where R1 is diaminocycloalkyl, and R2 and R3 each have the formula where R4, R5, and R6 are each independently hydrocarbon moieties having from 1 to about 10 carbon atoms, whereby the complex (I) is converted into a complex having the formula R1-P t-X2 (II) where R1 is diaminocycloalkyl and X is halogen.
18. The method of claim 17, where the pH is adjusted to between about 2 and about 6.5.
19. The method of claim 17. where R4, R5, and R6 are each independently alkyl having from 1 to about 6 carbon atoms.
20. The method of claim 17, where R4, R5, and R6 are each independently alkyl having from 1 to about 3 carbon atoms.
21. The method of claim 17, where the complex (I) is converted to the complex (II) within the liposome.
22. The method of claim 17, where the pH is adjusted by contacting the liposome with an acidic solution.
23. The method of claim 17. where the pH is adjusted by including an acidic phospholipid in the liposome.
24. The method of claim 17, where the liposome comprises dimyristoyl phosphatidyl glycerol.
25. The method of claim 17, where R2 and R3 are neodecanoato.
26. The method of claim 17, where R1 has from about 3 to about 6 carbon atoms.
25
28. The method of claim 17, where X is chlorine.
29. The method of claim 17, where the complex (I) is intercalated between bilayers of the liposome.
30. The method of claim 17, where the complex (II) is intercalated between bilayers of the liposome.
31. The method of claim 17, where the complex (I) is cis-bis-neodecanoato-trans-R,R-1,2-diaminocyclohexane platinum(II).
32. The method of claim 17, further comprising the step of subsequently readjusting the pH after a predetermined time to about 7.
33. A method of preparing an antitumor composition, comprising:
adjusting the pH of a composition that comprises cis-bis-neodecanoato-trans-R,R-1,2-diaminocyclohexane platinum (II) entrapped in a liposome, to a level less than 7, whereby the platinum complex is convened into dichlorodiamine platinum (II), and after a predetermined time, adjusting the pH to at least about 7.
34. A method of delivering a biologically active chemical moiety internally to a mammal, comprising:
providing an aqueous formulation of a prodrug of a biologically active moiety, the prodrug being entrapped in a liposome, the prodrug further being capable of forming the biologically active moiety upon exposure to a solution having an acidic pH;
reducing the pH to an acidic level, thereby converting the prodrug to the biologically active compound; and administering the aqueous formulation to a mammal.
35. The method of claim 34, where the biologically active moiety is an antitumor agent.
36. The method of claim 34, where the pH is reduced by including an acidic phospholipid in the liposome.
CA002273497A 1996-12-06 1997-12-05 Delivery and activation through liposome incorporation of diaminocyclohexane platinum(ii) complexes Abandoned CA2273497A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/764,095 1996-12-06
US08/764,095 US5843475A (en) 1996-12-06 1996-12-06 Delivery and activation through liposome incorporation of diaminocyclohexane platinum (II) complexes
PCT/US1997/022359 WO1998024416A1 (en) 1996-12-06 1997-12-05 Delivery and activation through liposome incorporation of diaminocyclohexane platinum(ii) complexes

Publications (1)

Publication Number Publication Date
CA2273497A1 true CA2273497A1 (en) 1998-06-11

Family

ID=25069669

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002273497A Abandoned CA2273497A1 (en) 1996-12-06 1997-12-05 Delivery and activation through liposome incorporation of diaminocyclohexane platinum(ii) complexes

Country Status (6)

Country Link
US (3) US5843475A (en)
EP (1) EP0956002A4 (en)
JP (1) JP2002513393A (en)
AU (1) AU736688B2 (en)
CA (1) CA2273497A1 (en)
WO (1) WO1998024416A1 (en)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5843475A (en) * 1996-12-06 1998-12-01 Board Of Regents, The University Of Texas System Delivery and activation through liposome incorporation of diaminocyclohexane platinum (II) complexes
IL131008A (en) * 1997-02-05 2003-06-24 Upjohn Co Lipid complexes and liposomes of highly insoluble platinum complex dicarboxylate
US6334895B1 (en) 1998-07-20 2002-01-01 The University Of Wyoming Research Corporation System for producing manufactured materials from coal combustion ash
EP1229934B1 (en) 1999-10-01 2014-03-05 Immunogen, Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
US6511676B1 (en) 1999-11-05 2003-01-28 Teni Boulikas Therapy for human cancers using cisplatin and other drugs or genes encapsulated into liposomes
TWI235066B (en) * 2001-10-03 2005-07-01 Celator Technologies Inc Liposome loading with metal ions
US20050214264A1 (en) * 2002-12-13 2005-09-29 Cosenza Lawrence W Sacromastigophoric therapeutic agent delivery system
WO2004098524A2 (en) * 2003-05-02 2004-11-18 Aronex Pharmaceuticals, Inc. Lipid platinum complexes and methods of use thereof
CA2526278A1 (en) * 2003-05-20 2004-12-02 Aronex Pharmaceuticals, Inc. Combination chemotherapy comprising a liposomal platinum complex
US20080026044A1 (en) * 2003-05-20 2008-01-31 Jonathan Lewis Combination Chemotherapy Comprising Capecitabine and a Liposomal Platinum Complex
EP1631301A4 (en) * 2003-05-20 2006-06-07 Aronex Pharmaceuticals Inc Combination chemotherapy comprising 5-fluorouracil or a derivative thereof and a liposomal platinum complex
US20080026045A1 (en) * 2003-05-20 2008-01-31 Jonathan Lewis Combination Chemotherapy Comprising a Gemcitabine and a Liposomal Platium Complex
US20050095283A1 (en) * 2003-09-16 2005-05-05 Aphios Corporation Compositions and methods for topically treating diseases
WO2006084248A2 (en) * 2005-02-04 2006-08-10 Antigenics, Inc. Compositions comprising a platinum complex, lipid, and surfactant
EP2090305A4 (en) * 2006-12-08 2012-12-26 Katayama Chemical Ind Co Ltd Liposome encapsulating ammine-platinum complex at high concentration, and method for production of the liposome
CA3176270A1 (en) * 2020-04-22 2021-10-28 Oncovolution, Llc Peptide platinum complexes and methods of use thereof

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1432563A (en) * 1972-04-10 1976-04-22 Rustenburg Platinum Mines Ltd Platinum- co-ordination compounds
JPS6041077B2 (en) * 1976-09-06 1985-09-13 喜徳 喜谷 Cis platinum(2) complex of 1,2-diaminocyclohexane isomer
JPS5829957B2 (en) * 1977-09-12 1983-06-25 喜徳 喜谷 Novel platinum complex
US4230631A (en) * 1977-10-19 1980-10-28 Johnson, Matthey & Co., Limited Platinum coordination compounds
US4203912A (en) * 1977-10-19 1980-05-20 Johnson, Matthey & Co., Limited Compositions containing platinum
JPS6034958B2 (en) * 1978-09-02 1985-08-12 喜徳 喜谷 New platinum complex
NL181434C (en) * 1980-01-03 1987-08-17 Tno PLATINUM (IV) DIAMINE COMPLEXES, AND THEIR PREPARATION AND APPLICATION.
NL8101026A (en) * 1981-03-03 1982-10-01 Tno PLATINADIAMINE COMPLEXES, A METHOD FOR PREPARING THE SAME, A METHOD FOR PREPARING A MEDICINAL PRODUCT USING SUCH PLATINADIAMINE COMPLEX FOR THE TREATMENT OF CANCER AND CONTAINED IN SUCH FORM.
EP0113508A1 (en) 1982-11-04 1984-07-18 Inco Research &amp; Development Center, Inc. Hydrophobic platinum compounds and their preparation
EP0130482B1 (en) * 1983-06-20 1988-12-28 Research Corporation Diaminocyclohexane platinum complexes, process for preparing same and pharmaceutical compositions containing same
US4758588A (en) * 1983-06-20 1988-07-19 Research Corporation Technologies Diaminocyclohexane platinum complexes
US4661516A (en) * 1983-06-20 1987-04-28 Research Corporation Diaminocyclohexane platinum complexes
DE3477788D1 (en) * 1983-08-05 1989-05-24 Yoshinori Kidani Cytostatic platinum complexes
DE3328667A1 (en) * 1983-08-09 1985-02-21 Bomoro Bocklenberg & Motte Gmbh & Co Kg, 5600 Wuppertal MOTOR VEHICLE DOOR LOCK, HOOD LOCK OD. DGL. WITH MOTORIC LOCKING AID
GB8328218D0 (en) * 1983-10-21 1983-11-23 Johnson Matthey Plc Oral compositions
DE3582961D1 (en) * 1984-06-27 1991-07-04 Johnson Matthey Plc PLATINO COORDINATION CONNECTIONS.
JPS6296A (en) 1985-03-06 1987-01-06 Sumitomo Pharmaceut Co Ltd Fat-soluble platinum(ii) complex and production thereof
US5384127A (en) * 1985-10-18 1995-01-24 Board Of Regents, The University Of Texas System Stable liposomal formulations of lipophilic platinum compounds
US5117022A (en) * 1985-10-18 1992-05-26 The Board Of Regents, The University Of Texas System Hydrophobic cis-platinum complexes efficiently incorporated into liposomes
US5041581A (en) * 1985-10-18 1991-08-20 The University Of Texas System Board Of Regents Hydrophobic cis-platinum complexes efficiently incorporated into liposomes
US4760157A (en) * 1986-01-31 1988-07-26 American Cyanamid Company (2,2,-bis(aminomethyl)-1,3-propanediol-N,N')platinum complexes
JPS62207283A (en) * 1986-03-07 1987-09-11 Yoshinori Kitani Novel platinum complex
US5011959A (en) * 1986-11-17 1991-04-30 The Board Of Regents, The University Of Texas System 1,2-diaminocyclohexane-platinum complexes with antitumor activity
JPS63203692A (en) * 1987-02-19 1988-08-23 Nippon Kayaku Co Ltd Novel platinum complex
US4956459A (en) * 1987-07-17 1990-09-11 Georgetown University Platinum compounds suitable for use as pharmaceuticals
CA1339034C (en) * 1988-08-22 1997-04-01 Paul A. Tremblay Platinum complexes of single isomer neoalkyl acids
US5041578A (en) * 1988-11-22 1991-08-20 Board Of Regents, The University Of Texas System Water soluble 1,2-diaminocyclohexane platinum (IV) complexes as antitumor agents
US4946954A (en) * 1989-01-17 1990-08-07 Georgetown University Platinum pharmaceutical agents
WO1994005282A1 (en) * 1992-09-04 1994-03-17 The Scripps Research Institute Water soluble taxol derivatives
US5843475A (en) * 1996-12-06 1998-12-01 Board Of Regents, The University Of Texas System Delivery and activation through liposome incorporation of diaminocyclohexane platinum (II) complexes

Also Published As

Publication number Publication date
AU736688B2 (en) 2001-08-02
US20010036470A1 (en) 2001-11-01
EP0956002A1 (en) 1999-11-17
WO1998024416A1 (en) 1998-06-11
AU7622598A (en) 1998-06-29
US6696079B2 (en) 2004-02-24
EP0956002A4 (en) 2002-09-11
US5843475A (en) 1998-12-01
JP2002513393A (en) 2002-05-08
US20040161456A1 (en) 2004-08-19
US7029696B2 (en) 2006-04-18

Similar Documents

Publication Publication Date Title
AU736688B2 (en) Delivery and activation through liposome incorporation of diaminocyclohexane platinum (II) complexes
EP0248036B1 (en) Hydrophobic cis-platinum complexes efficiently incorporated into liposomes
JP3415131B1 (en) Liposome preparation
EP0929293B1 (en) Liposomes containing a cisplatin compound
CA2462376C (en) Liposome loading with metal ions
WO1985000968A1 (en) Liposome delivery method for decreasing the toxicity of an antitumor drug
US20040022842A1 (en) Liposome preparations containing oxaliplatin
JP2006248978A (en) New liposome preparation
WO1990002131A1 (en) Hydrophobic cis-platinum complexes efficiently incorporated into liposomes
EP0358719B1 (en) Liposome compositions of anthracycline derivatives
US20090092662A1 (en) Liposome formulations of boronic acid compounds
JP2018530623A (en) Liposome composition co-encapsulating doxorubicin and mitomycin C prodrug
Perez-Soler et al. Lipophilic cisplatin analogues entrapped in liposomes: role of intraliposomal drug activation in biological activity
Perez-Soler et al. Lipophilic platinum complexes entrapped in liposomes: improved stability and preserved antitumor activity with complexes containing linear alkyl carboxylato leaving groups
US20220296520A1 (en) Liposome composition and preparation method thereof
CA1339034C (en) Platinum complexes of single isomer neoalkyl acids
Insook et al. Intraliposomal conversion of lipophilic cis-bis-carboxylato-trans-R, R-1, 2-diaminocyclohexane-platinum (II) complexes into cis-bis-dichloro-trans-R, R-1, 2-diaminocyclohexane-platinum (II)
AU593608B2 (en) Hydrophobic CIS-platinum complexes efficiently incorporated into liposomes
CA1330938C (en) Hydrophobic cis-platinum complexes efficiently incorporated into liposomes
Hamelers et al. Nanocapsules: a novel formulation technology for platinum-based anticancer drugs

Legal Events

Date Code Title Description
FZDE Discontinued