Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nanomedicine ; 30: 102275, 2020 11.
Article in English | MEDLINE | ID: mdl-32750494

ABSTRACT

CPX-351, a liposomal encapsulation of cytarabine and daunorubicin at a synergistic 5:1 molar ratio, is indicated for adults with newly diagnosed, therapy-related acute myeloid leukemia or acute myeloid leukemia with myelodysplasia-related changes. In preclinical species, this article demonstrated (1) similar release of cytarabine and daunorubicin by CPX-351 in plasma; (2) similar patterns of metabolism of cytarabine and daunorubicin following administration of CPX-351 versus non-liposomal cytarabine/daunorubicin combination; (3) prolonged tissue exposure to CPX-351; (4) dramatically different tissue distribution of cytarabine and daunorubicin following administration of CPX-351 versus non-liposomal combination (tissue:plasma ratios generally <1 versus >1, respectively); and (5) dramatically lower unbound plasma and tissue concentrations of cytarabine and daunorubicin following administration of CPX-351 versus non-liposomal combination. Together, these results provide insight into the safety profile of CPX-351, as well as mechanisms that drive the improved efficacy observed for CPX-351 versus the conventional 7 + 3 cytarabine/daunorubicin regimen in clinical studies.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cytarabine/pharmacokinetics , Daunorubicin/pharmacokinetics , Animals , Area Under Curve , Bile/chemistry , Chromatography, High Pressure Liquid/methods , Cytarabine/blood , Cytarabine/urine , Daunorubicin/blood , Daunorubicin/urine , Dogs , Drug Combinations , Feces/chemistry , Female , Half-Life , Limit of Detection , Male , Mice , Rats , Spectrometry, Fluorescence/methods , Spectrophotometry, Ultraviolet/methods , Tissue Distribution
2.
Int J Nanomedicine ; 14: 3819-3830, 2019.
Article in English | MEDLINE | ID: mdl-31213803

ABSTRACT

Combination regimens are a standard of care for many cancers. However, components of such regimens are typically first developed individually and subsequently combined using strategies to minimize toxicity. Little or no consideration is given to strategies that potentially maximize efficacy. In contrast, CPX-351 (Vyxeos®) is a dual-drug liposomal encapsulation of cytarabine and daunorubicin that was rationally designed to improve efficacy over the traditional 7+3 cytarabine/daunorubicin chemotherapy regimen for patients with acute myeloid leukemia (AML). The notable clinical efficacy of CPX-351 is achieved through maintenance of a synergistic 5:1 molar ratio of cytarabine and daunorubicin within the liposome after intravenous injection. The CPX-351 liposome, which is formulated to contain bilayers of distearoylphosphatidylcholine, distearoylphosphatidylglycerol, and cholesterol at a 7:2:1 molar ratio and remains in a gel phase at body temperature, provides stability without polyethylene glycol, controlled release of cytarabine and daunorubicin, limited systemic drug distribution, and preferential internalization within malignant myeloblasts in the bone marrow via active uptake of liposomes into cytoplasmic vacuoles. Thus, the CPX-351 liposome protects cytarabine and daunorubicin from metabolism and elimination, while overcoming pharmacokinetic differences between the two agents. In clinical studies, these liposome properties markedly increased the elimination half-life of CPX-351 versus free cytarabine and daunorubicin and maintained a synergistic drug ratio for over 24 hrs after administration. Preferential uptake of liposomes by leukemia cells suggests that relatively large amounts of cytarabine and daunorubicin enter malignant cells via liposomes, potentially bypassing P-glycoprotein-based efflux pumps, which are important mediators of chemotherapy resistance, and contribute to the rapid clearance of leukemia cells from the circulation and bone marrow. These pharmacologic advantages, a direct consequence of properties of the encapsulating liposome, may explain the efficacy of CPX-351 in patients with newly diagnosed high-risk/secondary AML and the reduced drug exposure in off-target tissues that contribute to a manageable safety profile.


Subject(s)
Cytarabine/pharmacokinetics , Daunorubicin/pharmacokinetics , Drug Compounding , Endocytosis , Neoplasms/metabolism , Daunorubicin/administration & dosage , Daunorubicin/pharmacology , Endocytosis/drug effects , Humans , Liposomes , Tissue Distribution/drug effects
3.
Leuk Res ; 53: 39-49, 2017 02.
Article in English | MEDLINE | ID: mdl-28013106

ABSTRACT

PURPOSE: Identify AML patients most likely to respond to CPX-351, a nano-scale liposome formulation containing cytarabine and daunorubicin co-encapsulated at a 5:1 molar ratio. METHODS: We examined the ex vivo cytotoxic activity of CPX-351 against leukemic cells isolated from 53 AML patients and an additional 127 samples including acute lymphoblastic leukemia, myelodysplastic syndrome/myeloproliferative neoplasms, or chronic lymphocytic leukemia/lymphoma. We assessed activity with respect to common molecular lesions and used flow cytometry to assess CPX-351 cellular uptake. RESULTS: AML specimen sensitivity to CPX-351 was similar across conventional risk groups. FLT3-ITD cases were five-fold more sensitive to CPX-351. CPX-351 was active across other indications with nearly all cases exhibiting IC50 values markedly lower than reported 72-h plasma drug concentration in patients receiving CPX-351. The range and distribution of CPX-351 IC50 values were comparable for AML, CLL, and ALL, whereas MDS/MPN cases were less sensitive. CPX-351 uptake analysis revealed a correlation between uptake of CPX-351 and cytotoxic potency. CONCLUSIONS: Our findings are consistent with clinical data, in which CPX-351 activity is retained in high-risk AML patients. Ex vivo analysis of cytotoxic potency may provide a means to identify specific AML subsets, such as FLT3-ITD, that benefit most from CPX-351 and warrant additional clinical evaluation.


Subject(s)
Blast Crisis/drug therapy , Cytarabine/administration & dosage , Daunorubicin/administration & dosage , Liposomes/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Antibiotics, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Humans , Inhibitory Concentration 50 , Mutation , Tumor Cells, Cultured , fms-Like Tyrosine Kinase 3/genetics
4.
J Drug Target ; 24(9): 797-804, 2016 11.
Article in English | MEDLINE | ID: mdl-27143215

ABSTRACT

Historically, the use of liposomes to enhance delivery of anticancer agents to cancer cells has focused primarily on solid tumors, which are characterized by rapid angiogenesis resulting in a poorly formed hypervasculature with abnormal vessel walls. The leaky vasculature in combination with poor lymphatic drainage has been demonstrated to lead to the accumulation of liposomes via the enhanced permeation and retention effect. However, only very limited information exists on the disposition of such delivery systems in the bone marrow compartment, the primary site of tumor cell origination and growth for many hematological malignancies. In this review we discuss the biological properties of anionic low-cholesterol liposome formulations and their potential for passively accumulating within the bone marrow and being selectively engulfed by leukemia cells compared to normal bone marrow cells. The therapeutic implications for preferential bone marrow delivery as well as the potential routes for the internalization of drug-encapsulated liposomes into cells in the absence of a targeting ligand are reviewed.


Subject(s)
Antineoplastic Agents/administration & dosage , Bone Marrow Cells/physiology , Cholesterol/chemistry , Drug Delivery Systems , Leukemia/drug therapy , Liposomes/chemistry , Animals , Humans
5.
J Control Release ; 172(2): 558-67, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-23664969

ABSTRACT

Pharmacokinetic modeling and simulation is a powerful tool for the prediction of drug concentrations in the absence of analytical techniques that allow for direct quantification. The present study applied this modeling approach to determine active drug release from a nanoparticle prodrug formulation. A comparative pharmacokinetic study of a nanoscale micellar docetaxel (DTX) prodrug, Procet 8, and commercial DTX formulation, Taxotere, was conducted in bile duct cannulated rats. The nanoscale (~40nm) size of the Procet 8 formulation resulted in confinement within the plasma space and high prodrug plasma concentrations. Ex vivo prodrug hydrolysis during plasma sample preparation resulted in unacceptable error that precluded direct measurement of DTX concentrations. Pharmacokinetic modeling of Taxotere and Procet 8 plasma concentrations, and their associated biliary metabolites, allowed for prediction of the DTX concentration profile and DTX bioavailability, and thereby evaluation of Procet 8 metabolism. Procet 8 plasma decay and in vitro plasma hydrolytic rates were identical, suggesting that systemic clearance of the prodrug was primarily metabolic. The Procet 8 and Taxotere plasma profiles, and associated docetaxel hydroxy-tert-butyl carbamate (HDTX) metabolite biliary excretion, were best fit by a two compartment model, with both linear and non-linear DTX clearance, and first order Procet 8 hydrolysis. The model estimated HDTX clearance rate agreed with in vitro literature values, supporting the predictability of the proposed model. Model simulation at the 10mg DTX equivalent/kg dose level predicted DTX formation rate-limited kinetics and a peak plasma DTX concentration of 39ng/mL at 4h for Procet 8, in comparison to 2826ng/mL for Taxotere. As a result of nonlinear DTX clearance, the DTX AUCinf for the Procet 8 formulation was predicted to be 2.6 times lower than Taxotere (775 vs. 2017h×ng/mL, respectively), resulting in an absolute bioavailability estimate of 38%. As DTX clearance in man is considered linear, this low bioavailability is likely species-dependent. These data support the use of pharmacokinetic modeling and simulation in cases of complex formulations, where analytical methods for direct measurement of free (released) drug concentrations are unavailable. Uses of such models may include interpretation of preclinical toxicology studies, selection of first in man dosing regimens, and PK/PD model development.


Subject(s)
Antineoplastic Agents/metabolism , Bile/metabolism , Nanoparticles/metabolism , Prodrugs/metabolism , Taxoids/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Computer Simulation , Docetaxel , Female , Hydrolysis , Kinetics , Male , Micelles , Models, Biological , Prodrugs/pharmacokinetics , Rats , Rats, Sprague-Dawley , Taxoids/pharmacokinetics
6.
J Drug Deliv ; 2011: 349206, 2011.
Article in English | MEDLINE | ID: mdl-21773044

ABSTRACT

Diblock copolymer nanoparticles encapsulating a paclitaxel prodrug, Propac 7, have been used to demonstrate the usefulness of a nonmetabolizable radioactive marker, cholesteryl hexadecyl ether (CHE), to evaluate nanoparticle formulation variables. Since CHE did not exchange out of the nanoparticles, the rate of clearance of the CHE could be used as an indicator of nanoparticle stability in vivo. We simultaneously monitored prodrug circulation and carrier circulation in the plasma and the retention of CHE relative to the retention of prodrug in the plasma was used to distinguish prodrug release from nanoparticle plasma clearance. Nanoparticles labelled with CHE were also used to evaluate accumulation of nanoparticles in the tumour. This marker has provided relevant data which we have applied to optimise our nanoparticle formulations.

7.
Chemother Res Pract ; 2011: 213848, 2011.
Article in English | MEDLINE | ID: mdl-22312548

ABSTRACT

The first liposomal formulation of cisplatin to be evaluated clinically was SPI-077. Although the formulation demonstrated enhanced cisplatin tumor accumulation in preclinical models it did not enhance clinical efficacy, possibly due to limited cisplatin release from the formulation localized within the tumor. We have examined a series of liposomal formulations to address the in vivo relationship between cisplatin release rate and formulation efficacy in the P388 murine leukemia model. The base formulation of phosphatidylcholine: phosphatidylglycerol: cholesterol was altered in the C18 and C16 phospholipid content to influence membrane fluidity and thereby impacting drug circulation lifetime and drug retention. Phase transition temperatures (T(m)) ranged from 42-55°C. The high T(m) formulations demonstrated enhanced drug retention properties accompanied by low antitumor activity while the lowest T(m) formulations released the drug too rapidly in the plasma, limiting drug delivery to the tumor which also resulted in low antitumor activity. A formulation composed of DSPC : DPPC : DSPG : Chol; (35 : 35 : 20 : 10) with an intermediate drug release rate and a cisplatin plasma half-life of 8.3 hours showed the greatest antitumor activity. This manuscript highlights the critical role that drug release rates play in the design of an optimized drug delivery vehicle.

8.
Expert Opin Drug Deliv ; 7(12): 1329-41, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21118030

ABSTRACT

IMPORTANCE OF THE FIELD: Drug combinations have been the standard of care in the treatment of cancer for > 50 years. Typically, combination chemotherapy uses agents with non-overlapping toxicities which are escalated to their maximum tolerated dose. However, emerging evidence indicates that this approach may not be providing optimal efficacy depending on the drug ratios to which the tumor is exposed. Combined drugs can be synergistic whereas other ratios of the same agents may be antagonistic or additive. AREAS COVERED IN THIS REVIEW: In this review, we examine the importance of drug ratios in cancer therapy. We describe how manipulation of the lipid membrane and internal buffer composition maintains synergistic ratios of irinotecan and floxuridine (CPX-1), daunorubicin and cytarabine (CPX-351) or cisplatin and irinotecan (CPX-571). For polymer-based nanoparticles, prodrug hydrophobicity was exploited to coordinate the release of gemcitabine and the more hydrophobic paclitaxel. We present preclinical data for liposomal drug combinations which demonstrate that the most efficacious formulation is not always the highest dose of both agents. WHAT THE READER WILL GAIN: An insight into the use of liposomes and polymer-based nanoparticles to deliver synergistic drug combinations to the tumor site and avoid antagonistic drug-drug interactions. TAKE HOME MESSAGE: The ability to control and maintain drug ratios in vivo through the use of nanoscale delivery vehicles results in a significant improvement in therapeutic activity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Delivery Systems , Nanoparticles , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dose-Response Relationship, Drug , Drug Synergism , Humans , Liposomes , Neoplasms/drug therapy
9.
Leuk Lymphoma ; 51(8): 1536-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20528246

ABSTRACT

CPX-351, a liposomal formulation co-encapsulating cytarabine (Cyt) and daunorubicin (Daun), has been developed, which delivers synergistic Cyt:Daun molar ratios to bone marrow. CPX-351 has demonstrated markedly superior anti-leukemic activity over free Cyt:Daun drug cocktails in preclinical models. Given the prolonged plasma lifetime of CPX-351, we examined the relationship between therapeutic efficacy and the frequency of treatment in the consolidation setting using a bone marrow-engrafting human leukemia xenograft model. Adding a day 1,3,5 consolidation treatment course for CPX-351 therapy improved the increase in lifespan (ILS) from 116% and no cures for a single induction course, to 268% plus a 33% cure rate for an induction plus consolidation course. In contrast, free Cyt:Daun cocktail treatment provided much lower ILS values with no cures. Administering CPX-351 as consolidation therapy starting on day 42 using a day 1,3, day 1,5, or day 1,7 schedule yielded ILS values of 154%, 185%, and 108%, respectively. The increased efficacy observed for the day 1,3 and day 1,5 consolidation schedules was associated with elevated bone marrow drug accumulation for the second doses. The enhanced efficacy obtained for intermediate dosing frequency in the consolidation setting suggests that the anti-leukemic activity of synergistic drug ratios is dependent on both duration of exposure and maintenance above a therapeutic threshold.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dosage Forms , Drug Synergism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Bone Marrow Cells/drug effects , Chemistry, Pharmaceutical , Cytarabine/administration & dosage , DNA-Binding Proteins/physiology , Daunorubicin/administration & dosage , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Flow Cytometry , Humans , Liposomes , Maximum Tolerated Dose , Mice , Survival Rate , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Leuk Res ; 34(9): 1214-23, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20138667

ABSTRACT

The objective of this study was to examine the pharmacodynamic basis for the potent preclinical and clinical anti-leukemic activity of CPX-351, a nano-scale liposome formulation of cytarabine and daunorubicin co-encapsulated at a synergistic 5:1 molar ratio. A bone marrow-engrafting CCRF-CEM leukemia model in Rag2-M mice was utilized to correlate the therapeutic and myelosuppressive properties of CPX-351 with bone marrow delivery and drug uptake in leukemia cells relative to normal bone marrow cell populations. When administered to mice bearing CCRF-CEM human leukemia xenografts, CPX-351 ablated bone marrow (BM) leukemic cells to below detectable levels for multiple weeks, whereas the free-drug cocktail only transiently suppressed leukemia growth. In contrast to the activity against leukemia cells, CPX-351 and free-drug cocktail induced similar myelosuppression in non-tumor-bearing BM. In leukemia-laden BM, drug concentrations were markedly elevated for CPX-351 over free-drug cocktail and the first dose of CPX-351, but not free-drug cocktail, potentiated BM drug accumulation for subsequent doses. Confocal fluorescence microscopy revealed that CPX-351 liposomes are taken up by CCRF-CEM cells and subsequently release drugs intracellularly. The improved in vivo efficacy of CPX-351 appears related to increased and prolonged exposure of synergistic cytarabine:daunorubicin ratios in BM, and the selective killing of leukemia may arise from direct liposome-leukemia cell interactions. These features may also have broader applicability in the treatment of other haematological malignancies.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bone Marrow/drug effects , Cytarabine/pharmacokinetics , Daunorubicin/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Chemistry, Pharmaceutical , Cytarabine/administration & dosage , Daunorubicin/administration & dosage , Humans , Mice , Transplantation, Heterologous
11.
Mol Cancer Ther ; 8(8): 2266-75, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19671743

ABSTRACT

Irinotecan and cisplatin are two established anticancer drugs, which together constitute an effective combination for treating small-cell lung cancer. We investigated whether the efficacy of this combination could be improved by controlling drug ratios following in vivo administration. Irinotecan and cisplatin combinations were evaluated systematically for drug ratio-dependent synergy in vitro using a panel of 20 tumor cell lines. In vitro screening informatics on drug ratio-dependent cytotoxicity identified a consistently antagonistic region between irinotecan/cisplatin molar ratios of 1:2 to 4:1, which was bordered by two synergistic regions. Liposomal co-formulations of these two agents were developed that exhibited plasma drug half-lives of approximately 6 hours and maintained a fixed drug ratio for more than 24 hours. Drug ratio-dependent antitumor activity was shown in vivo for these liposome formulations, and irinotecan/cisplatin ratios between 5:1 and 10:1 were identified as therapeutically optimal. The relationship between irinotecan/cisplatin ratio and in vivo efficacy was consistent with in vitro drug ratio dependency results. Superior antitumor activity was observed for the liposome-encapsulated 7:1 molar ratio of irinotecan/cisplatin (designated CPX-571) compared with the free-drug cocktail in all models tested. Further efficacy studies in a range of human tumor xenografts, including an irinotecan-resistant model, showed that both liposomal agents contributed to the overall efficacy in a manner consistent with in vivo synergy. These results show the ability of drug delivery technology to enhance the therapeutic activity of irinotecan/cisplatin combination treatment by maintaining synergistic ratios in vivo. CPX-571, a fixed-ratio formulation of irinotecan and cisplatin, is a promising candidate for clinical development.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Camptothecin/analogs & derivatives , Cisplatin/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacokinetics , Drug Synergism , Humans , Irinotecan , Mice , Mice, Nude
12.
Leuk Res ; 33(1): 129-39, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18676016

ABSTRACT

We demonstrate here that cytarabine and daunorubicin, a standard drug combination used in the treatment of leukaemia, exhibits drug ratio-dependent synergistic antitumor activity in vitro and in vivo. A cytarabine:daunorubicin molar ratio of 5:1 displayed the greatest degree of synergy and minimum antagonism in a panel of 15 tumor cell lines in vitro. Co-encapsulating cytarabine and daunorubicin inside liposomes maintained the synergistic drug ratio in plasma for 24h post-injection. Liposome-encapsulated cytarabine:daunorubicin combinations exhibited drug ratio-dependent in vivo efficacy with the 5:1 molar drug ratio (designated CPX-351) having the greatest therapeutic index, despite using sub-MTD daunorubicin doses. CPX-351 exhibited superior therapeutic activity compared to free-drug cocktails, with high proportions of long-term survivors, consistent with in vivo synergy. The therapeutic advantage of CPX-351 was associated with prolonged maintenance of synergistic drug ratios in bone marrow. These results indicate that in vitro informatics on cytarabine:daunorubicin cytotoxicity can be translated in vivo to optimize the efficacy of anticancer drug combinations by controlling the exposure of drug ratios with drug delivery vehicles.


Subject(s)
Antineoplastic Agents/pharmacology , Cytarabine/pharmacology , Daunorubicin/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Proliferation , Cytarabine/administration & dosage , Cytarabine/pharmacokinetics , Daunorubicin/administration & dosage , Daunorubicin/pharmacokinetics , Flow Cytometry , Humans , Liposomes , Mice
13.
J Med Chem ; 51(11): 3288-96, 2008 Jun 12.
Article in English | MEDLINE | ID: mdl-18465845

ABSTRACT

A series of paclitaxel prodrugs designed for formulation in lipophilic nanoparticles are described. The hydrophobicity of paclitaxel was increased by conjugating a succession of increasingly hydrophobic lipid anchors to the drug using succinate or diglycolate cross-linkers. The prodrugs were formulated in well defined block copolymer-stabilized nanoparticles. These nanoparticles were shown to have an elimination half-life of approximately 24 h in vivo. The rate at which the prodrug was released from the nanoparticles could be controlled by adjusting the hydrophobicity of the lipid anchor, resulting in release half-lives ranging from 1 to 24 h. The diglycolate and succinate cross-linked prodrugs were 1-2 orders of magnitude less potent than paclitaxel in vitro. Nanoparticle formulations of the succinate prodrugs showed no evidence of efficacy in HT29 human colorectal tumor xenograph models. Efficacy of diglycolate prodrug nanoparticles increased as the anchor hydrophobicity increased. Long circulating diglycolate prodrug nanoparticles provided significantly enhanced therapeutic activity over commercially formulated paclitaxel at the maximum tolerated dose.


Subject(s)
Paclitaxel/administration & dosage , Prodrugs/administration & dosage , Animals , Cell Line, Tumor , Drug Stability , Humans , Hydrophobic and Hydrophilic Interactions , Lipids/chemistry , Mice , Mice, Nude , Micelles , Nanoparticles , Neoplasm Transplantation , Paclitaxel/chemistry , Paclitaxel/pharmacology , Polyethylene Glycols , Polystyrenes , Prodrugs/chemistry , Prodrugs/pharmacology , Transplantation, Heterologous
14.
Pharm Res ; 25(7): 1702-13, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18320294

ABSTRACT

PURPOSE: The inter/intramolecular interactions between drugs (floxuridine, irinotecan) and excipients (copper gluconate, triethanolamine) in the dual-drug liposomal formulation CPX-1 were elucidated in order to identify the physicochemical properties that allow coordinated release of irinotecan and floxuridine and maintenance of the two agents at a fixed, synergistic 1:1 molar ratio. METHODS: Release of irinotecan and floxuridine from the liposomes was assessed using an in vitro-release assay. Fluorescence, Nuclear Magnetic Resonance spectroscopy (NMR) and UV-Vis were used to characterize the aggregation state of the drugs within the liposomes. RESULTS: Coordinated release of the drugs from liposomes was disrupted by removing copper gluconate. Approximately 45% of the total irinotecan was detectable in the copper-containing CPX-1 formulation by NMR, which decreased to 19% without copper present in the liposomal interior. Formation of higher order, NMR-silent aggregates was associated with slower and uncoordinated irinotecan release relative to floxuridine and loss of the synergistic drug/drug ratio. Solution spectroscopy and calorimetry revealed that while all formulation components were required to achieve the highest solubility of irinotecan, direct drug-excipient binding interactions were absent. CONCLUSIONS: Long-range interactions between irinotecan, floxuridine and excipients modulate the aggregation state of irinotecan, allowing for simultaneous release of both drugs from the liposomes.


Subject(s)
Antimetabolites, Antineoplastic/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Camptothecin/analogs & derivatives , Floxuridine/chemistry , Liposomes , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Calorimetry , Camptothecin/administration & dosage , Camptothecin/chemistry , Chemical Phenomena , Chemistry, Pharmaceutical , Chemistry, Physical , Cholesterol , Copper , Drug Carriers , Drug Compounding , Excipients , Floxuridine/administration & dosage , Gluconates , Irinotecan , Magnetic Resonance Spectroscopy , Microscopy, Electron, Transmission , Phosphatidylcholines , Phosphatidylglycerols , Solubility , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet
15.
Oncol Res ; 16(8): 361-74, 2007.
Article in English | MEDLINE | ID: mdl-17913044

ABSTRACT

Whether anticancer drug combinations act synergistically or antagonistically often depends on the ratio of the agents being combined. We show here that combinations of irinotecan and floxuridine exhibit drug ratio-dependent cytotoxicity in a broad panel of tumor cell lines in vitro where a 1:1 molar ratio consistently provided synergy and avoided antagonism. In vivo delivery of irinotecan and floxuridine coencapsulated inside liposomes at the synergistic 1:1 molar ratio (referred to as CPX-1) lead to greatly enhanced efficacy compared to the two drugs administered as a saline-based cocktail in a number of human xenograft and murine tumor models. When compared to liposomal irinotecan or liposomal floxuridine, the therapeutic activity of CPX-1 in vivo was not only superior to the individual liposomal agents, but the extent of tumor growth inhibition was greater than that predicted for combining the activities of the individual agents. In contrast, liposome delivery of irinotecan:floxuridine ratios shown to be antagonistic in vitro provided antitumor activity that was actually less than that achieved with liposomal irinotecan alone, indicative of in vivo antagonism. Synergistic antitumor activity observed for CPX-1 was associated with maintenance of the 1:1 irinotecan:floxuridine molar ratio in plasma and tumor tissue over 16-24 h. In contrast, injection of the drugs combined in saline resulted in irinotecan:floxuridine ratios that changed 10-fold within 1 h in plasma and sevenfold within 4 h in tumor tissue. These results indicate that substantial improvements in the efficacy of drug combinations may be achieved by maintaining in vitro-identified synergistic drug ratios after systemic administration using drug delivery vehicles.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Floxuridine/administration & dosage , Neoplasms/drug therapy , Animals , Camptothecin/administration & dosage , Camptothecin/blood , Camptothecin/pharmacokinetics , Cell Survival , Dose-Response Relationship, Drug , Drug Antagonism , Drug Combinations , Drug Compounding , Drug Evaluation, Preclinical , Drug Synergism , Female , Floxuridine/blood , Floxuridine/pharmacokinetics , Humans , Injections, Intravenous , Irinotecan , Liposomes , Mice , Neoplasms/metabolism , Pharmaceutical Vehicles , Survival Rate , Tissue Distribution , Tumor Burden , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Int J Pharm ; 337(1-2): 219-28, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17270372

ABSTRACT

A novel method for encapsulating irinotecan into liposomes containing copper gluconate buffered to pH 7.0 with triethanolamine (TEA) has recently been developed. In the present study, the mechanism dictating drug encapsulation and retention inside those liposomes was investigated. Spectroscopic analyses revealed that irinotecan interacted with copper gluconate/TEA in solution. Fourier transformed infrared (FT-IR) spectroscopy indicated a strengthening of the hydrogen bonds involving the hydroxyl groups when solutions of irinotecan and copper gluconate/TEA are mixed at a 1:1 molar ratio. The intensity of the circular dichroism (CD) signal of copper gluconate/TEA increased in the presence of equimolar amounts of irinotecan. The addition of irinotecan to liposomes containing copper gluconate/TEA at 50 degrees C induced a shift of the absorption bands from 370 nm to 378 nm as well as a 60% quenching of the drug fluorescence at 440 nm suggesting the occurrence of irinotecan self association. Irinotecan encapsulation was found to be kinetically and stoichiometrically correlated with the release of TEA from the liposomes. The results suggested that the encapsulation of irinotecan was mediated by TEA in association with copper gluconate, leading to a final drug complex that is retained inside the liposomes. A neutral antiport exchange loading mechanism between irinotecan and TEA is proposed.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Camptothecin/analogs & derivatives , Ethanolamines/chemistry , Gluconates/chemistry , Liposomes , Camptothecin/chemistry , Chemistry, Pharmaceutical , Circular Dichroism , Drug Compounding , Hydrogen Bonding , Hydrogen-Ion Concentration , Irinotecan , Kinetics , Solubility , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared , Technology, Pharmaceutical/methods , Temperature
17.
Biochim Biophys Acta ; 1768(3): 678-87, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17208196

ABSTRACT

A liposomal delivery system that coordinates the release of irinotecan and floxuridine in vivo has been developed. The encapsulation of floxuridine was achieved through passive entrapment while irinotecan was actively loaded using a novel copper gluconate/triethanolamine based procedure. Coordinating the release rates of both drugs was achieved by altering the cholesterol content of distearoylphosphatidylcholine (DSPC)/distearoylphosphatidylglycerol (DSPG) based formulations. The liposomal retention of floxuridine in plasma after intravenous injection was dramatically improved by decreasing the cholesterol content of the formulation below 20 mol%. In the case of irinotecan, the opposite trend was observed where increasing cholesterol content enhanced drug retention. Liposomes composed of DSPC/DSPG/Chol (7:2:1, mole ratio) containing co-encapsulated irinotecan and floxuridine at a 1:1 molar ratio exhibited matched leakage rates for the two agents so that the 1:1 ratio was maintained after intravenous administration to mice. The encapsulation of irinotecan was optimal when copper gluconate/triethanolamine (pH 7.4) was used as the intraliposomal buffer. The efficiency of irinotecan loading was approximately 80% with a starting drug to lipid molar ratio of 0.1/1. Leakage of floxuridine from the liposomes during irinotecan loading at 50 degrees C complicated the ability to readily achieve the target 1:1 irinotecan/floxuridine ratio inside the formulation. As a result, a procedure for the simultaneous encapsulation of irinotecan and floxuridine was developed. This co-encapsulation method has the advantage over sequential loading in that extrusion can be performed in the absence of chemotherapeutic agents and the drug/drug ratios in the final formulation can be more precisely controlled.


Subject(s)
Antimetabolites, Antineoplastic/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Camptothecin/analogs & derivatives , Floxuridine/chemistry , Liposomes , Animals , Antimetabolites, Antineoplastic/blood , Antimetabolites, Antineoplastic/pharmacokinetics , Antineoplastic Agents, Phytogenic/blood , Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/blood , Camptothecin/chemistry , Camptothecin/pharmacokinetics , Cholesterol/chemistry , Drug Combinations , Drug Compounding , Drug Delivery Systems , Female , Floxuridine/blood , Floxuridine/pharmacokinetics , Hydrogen-Ion Concentration , Irinotecan , Mice , Mice, Inbred BALB C
18.
Eur J Pharm Biopharm ; 65(3): 289-99, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17123800

ABSTRACT

The purpose of these studies was to design an intravenous drug formulation consisting of two active agents having synergistic in vitro activity. Specifically, we describe a novel drug combination consisting of a cytotoxic agent (vinorelbine) with an apoptosis-inducing lipid (phosphatidylserine, PS). In vitro cytotoxicity screening of PS and vinorelbine, alone and in combination, against human MDA435/LCC6 breast cancer and H460 lung cancer cells was used to identify the molar ratio of these two agents required for synergistic activity. PS and vinorelbine were co-formulated in a lipid-based system at the synergistic molar ratio and the pharmacokinetic and antitumor characteristics of the combination assessed in mice bearing H460 tumors. The cytotoxicity of the lipid, and the synergy between the lipid and vinorelbine, were specific to PS; these effects were not observed using control lipids. A novel formulation of PS, incorporated as a membrane component in liposomes, and encapsulating vinorelbine using a pH gradient based loading method was developed. The PS to vinorelbine ratio in this formulation was 1/1, a ratio that produced synergistic in vitro cytotoxicity over a broad concentration range. The vinorelbine and PS dual-agent treatment significantly delayed the growth of subcutaneous human H460 xenograft tumors in Rag2M mice compared to the same dose of free vinorelbine given alone or given as a cocktail of the free vinorelbine simultaneously with empty PS-containing liposomes. These studies demonstrate the potential to develop clinically relevant drug combinations identified using in vitro drug-drug interactions combined with lipid-based delivery systems to co-formulate drugs at their synergistic ratios.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lung Neoplasms/drug therapy , Phosphatidylserines/administration & dosage , Vinblastine/analogs & derivatives , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Drug Compounding , Drug Design , Drug Synergism , Female , Humans , Injections, Intravenous , Liposomes , Lung Neoplasms/pathology , Mice , Phosphatidylserines/chemistry , Phosphatidylserines/pharmacokinetics , Technology, Pharmaceutical , Vinblastine/administration & dosage , Vinblastine/chemistry , Vinblastine/pharmacokinetics , Vinorelbine , Xenograft Model Antitumor Assays
19.
Mol Cancer Ther ; 5(7): 1854-63, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16891472

ABSTRACT

Anticancer drug combinations can act synergistically or antagonistically against tumor cells in vitro depending on the ratios of the individual agents comprising the combination. The importance of drug ratios in vivo, however, has heretofore not been investigated, and combination chemotherapy treatment regimens continue to be developed based on the maximum tolerated dose of the individual agents. We systematically examined three different drug combinations representing a range of anticancer drug classes with distinct molecular mechanisms (irinotecan/floxuridine, cytarabine/daunorubicin, and cisplatin/daunorubicin) for drug ratio-dependent synergy. In each case, synergistic interactions were observed in vitro at certain drug/drug molar ratio ranges (1:1, 5:1, and 10:1, respectively), whereas other ratios were additive or antagonistic. We were able to maintain fixed drug ratios in plasma of mice for 24 hours after i.v. injection for all three combinations by controlling and overcoming the inherent dissimilar pharmacokinetics of individual drugs through encapsulation in liposomal carrier systems. The liposomes not only maintained drug ratios in the plasma after injection, but also delivered the formulated drug ratio directly to tumor tissue. In vivo maintenance of drug ratios shown to be synergistic in vitro provided increased efficacy in preclinical tumor models, whereas attenuated antitumor activity was observed when antagonistic drug ratios were maintained. Fixing synergistic drug ratios in pharmaceutical carriers provides an avenue by which anticancer drug combinations can be optimized prospectively for maximum therapeutic activity during preclinical development and differs from current practice in which dosing regimens are developed empirically in late-stage clinical trials based on tolerability.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/blood , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/blood , Cytarabine/administration & dosage , Cytarabine/blood , Daunorubicin/administration & dosage , Daunorubicin/blood , Dose-Response Relationship, Drug , Drug Synergism , Floxuridine/administration & dosage , Floxuridine/blood , Humans , Irinotecan , Liposomes , Mice , Xenograft Model Antitumor Assays
20.
J Control Release ; 105(1-2): 89-105, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15878792

ABSTRACT

Idarubicin has been successfully encapsulated in cholesterol-free liposomes, however, little is known about how the rate of drug release from circulating liposomes influences therapeutic activity. The studies described herein assess the attributes of a liposome formulation required to significantly increase the plasma levels of idarubicin and further establish whether increases in the circulation longevity of the drug mediate improved antitumor activity. Pharmacokinetic assessments of 6 different 3[H]-labelled liposome formulations were compared to free idarubicin. The highest idarubicin plasma concentrations were observed with DSPC/DSPE-PEG2000 liposomes formulated with 2 mol% DSPE-PEG2000 and 150 mM (iso-osmotic) internal citrate concentration. It was shown that increased levels of PEG-lipid incorporation augmented IDA release and the optimal liposomal formulation needed to be prepared under iso-osmotic conditions. For efficacy studies in a murine leukemia model, groups of 12-14 mice were treated i.v. with saline or equivalent doses (1, 2, 3 mg/kg) of free or liposomal IDA. Liposomal treatment groups exhibited a higher % increase in life span (ILS) as compared to equivalent doses of free drug. Efficacy studies completed in two drug resistant models, P388/ADR and MDA435LCC6/MDR1, demonstrated that neither the free nor liposomal formulation of idarubicin was therapeutically active. Encapsulation of IDA in liposomes increased antitumor activity in an IDA sensitive model, however, the significant increase in plasma drug levels was not sufficient to overcome multidrug resistance.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/therapeutic use , Idarubicin/administration & dosage , Idarubicin/therapeutic use , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Area Under Curve , Buffers , Cell Line, Tumor , Cell Survival/drug effects , Cholesterol , Drug Carriers , Drug Compounding , Drug Resistance, Neoplasm , Humans , Hydrogen-Ion Concentration , Idarubicin/pharmacokinetics , Liposomes , Mice , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Pharmaceutical Vehicles , Polyethylene Glycols , Tetrazolium Salts , Thiazoles , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...