Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
ACS Infect Dis ; 10(5): 1780-1792, 2024 May 10.
Article En | MEDLINE | ID: mdl-38651692

The recent COVID-19 pandemic underscored the limitations of currently available direct-acting antiviral treatments against acute respiratory RNA-viral infections and stimulated major research initiatives targeting anticoronavirus agents. Two novel nsp5 protease (MPro) inhibitors have been approved, nirmatrelvir and ensitrelvir, along with two existing nucleos(t)ide analogues repurposed as nsp12 polymerase inhibitors, remdesivir and molnupiravir, but a need still exists for therapies with improved potency and systemic exposure with oral dosing, better metabolic stability, and reduced resistance and toxicity risks. Herein, we summarize our research toward identifying nsp12 inhibitors that led to nucleoside analogues 10e and 10n, which showed favorable pan-coronavirus activity in cell-infection screens, were metabolized to active triphosphate nucleotides in cell-incubation studies, and demonstrated target (nsp12) engagement in biochemical assays.


Antiviral Agents , COVID-19 Drug Treatment , Nucleosides , SARS-CoV-2 , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , SARS-CoV-2/drug effects , Humans , Nucleosides/pharmacology , Nucleosides/chemistry , Animals , Drug Discovery , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Chlorocebus aethiops , Vero Cells , COVID-19/virology , Coronavirus RNA-Dependent RNA Polymerase
2.
Viruses ; 16(3)2024 02 21.
Article En | MEDLINE | ID: mdl-38543689

HBV RNA destabilizers are a class of small-molecule compounds that target the noncanonical poly(A) RNA polymerases PAPD5 and PAPD7, resulting in HBV RNA degradation and the suppression of viral proteins including the hepatitis B surface antigen (HBsAg). AB-161 is a next-generation HBV RNA destabilizer with potent antiviral activity, inhibiting HBsAg expressed from cccDNA and integrated HBV DNA in HBV cell-based models. AB-161 exhibits broad HBV genotype coverage, maintains activity against variants resistant to nucleoside analogs, and shows additive effects on HBV replication when combined with other classes of HBV inhibitors. In AAV-HBV-transduced mice, the dose-dependent reduction of HBsAg correlated with concentrations of AB-161 in the liver reaching above its effective concentration mediating 90% inhibition (EC90), compared to concentrations in plasma which were substantially below its EC90, indicating that high liver exposure drives antiviral activities. In preclinical 13-week safety studies, minor non-adverse delays in sensory nerve conductance velocity were noted in the high-dose groups in rats and dogs. However, all nerve conduction metrics remained within physiologically normal ranges, with no neurobehavioral or histopathological findings. Despite the improved neurotoxicity profile, microscopic findings associated with male reproductive toxicity were detected in dogs, which subsequently led to the discontinuation of AB-161's clinical development.


Coordination Complexes , Hepatitis B virus , Hepatitis B, Chronic , Naphthalenesulfonates , Male , Mice , Rats , Animals , Dogs , Hepatitis B virus/physiology , Hepatitis B Surface Antigens/genetics , RNA, Viral , RNA, Messenger , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , DNA, Viral/genetics , Hepatitis B, Chronic/drug therapy , DNA, Circular
3.
Bioorg Med Chem Lett ; 94: 129456, 2023 10 01.
Article En | MEDLINE | ID: mdl-37633618

Disruption of the HBV capsid assembly process through small-molecule interaction with HBV core protein is a validated target for the suppression of hepatitis B viral replication and the development of new antivirals. Through combination of key structural features associated with two distinct series of capsid assembly modulators, a novel aminochroman-based chemotype was identified. Optimization of anti-HBV potency through generation of SAR in addition to further core modifications provided a series of related functionalized aminoindanes. Key compounds demonstrated excellent cellular potency in addition to favorable ADME and pharmacokinetic profiles and were shown to be highly efficacious in a mouse model of HBV replication. Aminoindane derivative AB-506 was subsequently advanced into clinical development.


Antiviral Agents , Capsid Proteins , Capsid , Animals , Mice , Antiviral Agents/pharmacology , Disease Models, Animal , Structure-Activity Relationship , Hepatitis B virus/drug effects , Hepatitis B virus/metabolism
4.
Antiviral Res ; 197: 105211, 2022 01.
Article En | MEDLINE | ID: mdl-34826506

AB-506, a small-molecule inhibitor targeting the HBV core protein, inhibits viral replication in vitro (HepAD38 cells: EC50 of 0.077 µM, CC50 > 25 µM) and in vivo (HBV mouse model: ∼3.0 log10 reductions in serum HBV DNA compared to the vehicle control). Binding of AB-506 to HBV core protein accelerates capsid assembly and inhibits HBV pgRNA encapsidation. Furthermore, AB-506 blocks cccDNA establishment in HBV-infected HepG2-hNTCP-C4 cells and primary human hepatocytes, leading to inhibition of viral RNA, HBsAg, and HBeAg production (EC50 from 0.64 µM to 1.92 µM). AB-506 demonstrated activity across HBV genotypes A-H and maintains antiviral activity against nucleos(t)ide analog-resistant variants in vitro. Evaluation of AB-506 against a panel of core variants showed that T33N/Q substitutions results in >200-fold increase in EC50 values, while L30F, L37Q, and I105T substitutions showed an 8 to 20-fold increase in EC50 values in comparison to the wild-type. In vitro combinations of AB-506 with NAs or an RNAi agent were additive to moderately synergistic. AB-506 exhibits good oral bioavailability, systemic exposure, and higher liver to plasma ratios in rodents, a pharmacokinetic profile supporting clinical development for chronic hepatitis B.


Antiviral Agents/pharmacology , Hepatitis B virus/drug effects , Viral Core Proteins/antagonists & inhibitors , Virus Replication/drug effects , Animals , Antiviral Agents/pharmacokinetics , Cells, Cultured , Drug Evaluation, Preclinical , Female , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Mice , Rats , Virus Assembly/drug effects
5.
Article En | MEDLINE | ID: mdl-29555628

AB-423 is a member of the sulfamoylbenzamide (SBA) class of hepatitis B virus (HBV) capsid inhibitors in phase 1 clinical trials. In cell culture models, AB-423 showed potent inhibition of HBV replication (50% effective concentration [EC50] = 0.08 to 0.27 µM; EC90 = 0.33 to 1.32 µM) with no significant cytotoxicity (50% cytotoxic concentration > 10 µM). Addition of 40% human serum resulted in a 5-fold increase in the EC50s. AB-423 inhibited HBV genotypes A through D and nucleos(t)ide-resistant variants in vitro Treatment of HepDES19 cells with AB-423 resulted in capsid particles devoid of encapsidated pregenomic RNA and relaxed circular DNA (rcDNA), indicating that it is a class II capsid inhibitor. In a de novo infection model, AB-423 prevented the conversion of encapsidated rcDNA to covalently closed circular DNA, presumably by interfering with the capsid uncoating process. Molecular docking of AB-423 into crystal structures of heteroaryldihydropyrimidines and an SBA and biochemical studies suggest that AB-423 likely also binds to the dimer-dimer interface of core protein. In vitro dual combination studies with AB-423 and anti-HBV agents, such as nucleos(t)ide analogs, RNA interference agents, or interferon alpha, resulted in additive to synergistic antiviral activity. Pharmacokinetic studies with AB-423 in CD-1 mice showed significant systemic exposures and higher levels of accumulation in the liver. A 7-day twice-daily administration of AB-423 in a hydrodynamic injection mouse model of HBV infection resulted in a dose-dependent reduction in serum HBV DNA levels, and combination with entecavir or ARB-1467 resulted in a trend toward antiviral activity greater than that of either agent alone, consistent with the results of the in vitro combination studies. The overall preclinical profile of AB-423 supports its further evaluation for safety, pharmacokinetics, and antiviral activity in patients with chronic hepatitis B.


Antiviral Agents/pharmacology , Capsid/metabolism , Hepatitis B virus/drug effects , Hepatitis B/drug therapy , Virus Assembly/drug effects , Animals , Binding Sites , Cell Line, Tumor , DNA, Circular/metabolism , DNA, Viral/blood , DNA, Viral/metabolism , Female , Guanine/analogs & derivatives , Guanine/pharmacology , Hepatitis B virus/growth & development , Humans , Mice , Molecular Docking Simulation , Protein Binding , RNA, Viral/genetics
6.
Pediatr Blood Cancer ; 62(1): 65-71, 2015 Jan.
Article En | MEDLINE | ID: mdl-25203866

BACKGROUND: CPX-351, a liposomal formulation of cytarabine and daunorubicin co-encapsulated at an optimized synergistic 5:1 molar ratio, has demonstrated improved clinical outcomes over conventional cytarabine/daunorubicin treatment in a randomized phase 2 trial in patients with AML as well as superior efficacy against preclinical leukemia models when compared to the free drugs in combination. PROCEDURES: Given the promising phase 2 data, limited toxicities observed, and the known clinical activities of cytarabine/daunorubicin, we assessed the efficacy of CPX-351 against a panel of childhood ALL xenograft models. Plasma pharmacokinetics of cytarabine and daunorubicin following CPX-351 treatment were determined by HPLC in order to correlate efficacy with drug exposure. RESULTS: CPX-351, at a dose of 5 units/kg (corresponding to 5 mg/kg cytarabine and 2.2 mg/kg daunorubicin), was highly efficacious against all xenografts tested, inducing complete responses in four B-lineage xenografts and partial response in one T-lineage xenograft. These therapeutic responses were achieved with CPX-351 doses that provided drug exposures (based on Cmax and AUC) comparable to those observed in patients with AML. CONCLUSIONS: These results suggest that CPX-351 may be a promising chemotherapeutic to be utilized in the treatment of ALL and support its testing in pediatric patients with leukemia.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Child , Child, Preschool , Cytarabine/administration & dosage , Daunorubicin/administration & dosage , Female , Humans , Liposomes , Male , Maximum Tolerated Dose , Mice , Mice, Inbred NOD , Mice, SCID , Pediatrics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tissue Distribution , Treatment Outcome , Xenograft Model Antitumor Assays
7.
Exp Hematol ; 39(7): 741-50, 2011 Jul.
Article En | MEDLINE | ID: mdl-21530609

OBJECTIVE: To evaluate the possibility of improved selective killing of acute myeloid leukemia (AML) cells with CPX-351 (a liposomal formulation of cytarabine and daunorubicin). CPX-351 and the same molar ratio of free drugs were compared for cytotoxicity against colony-forming cells (CFCs) and subpopulations of cells enriched for primitive progenitors from AML patients and normal granulocyte colony-stimulating factor-mobilized peripheral blood (PB) and bone marrow (BM) donors. MATERIALS AND METHODS: AML blasts (n = 13) and normal PB and BM cells (n = 7) were incubated for 24 hours in various concentrations of CPX-351 or free drugs before plating in CFC assay or staining with anti-CD34 and anti-CD38 antibodies, Annexin-V, and propidium iodide followed by fluorescence-activated cell sorting analysis. High performance liquid chromatography was used to measure intracellular daunorubicin accumulation. RESULTS: AML blasts and progenitors from patients who achieved complete remission were more sensitive to both CPX-351 and free drugs than the same cells from patients with chemotherapy refractory leukemia. However, AML CFCs and CD34(+)CD38(-) AML blasts (enriched for candidate leukemia stem cells) from the same patient showed similar sensitivity to the liposomal or free drug formulations. In contrast, CFCs and CD34(+)CD38(-) cells from normal PB and BM were fivefold more sensitive to the free drugs than to CPX-351. Consistent with these observations, preferential intracellular accumulation of CPX-351 in AML over normal cells was observed, while there was little difference in drug uptake between AML and normal cells with the free drug cocktail. CONCLUSIONS: CPX-351, as compared to free cytarabine:daunorubicin, shows enhanced selective in vitro cytotoxicity for AML rather than normal progenitors.


Bone Marrow Cells/drug effects , Cytarabine/pharmacology , Daunorubicin/pharmacology , Leukocytes, Mononuclear/drug effects , Stem Cells/drug effects , Acute Disease , Bone Marrow Cells/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Colony-Forming Units Assay , Dose-Response Relationship, Drug , Drug Combinations , Drug Compounding , Drug Synergism , Flow Cytometry , Humans , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Leukocytes, Mononuclear/metabolism , Liposomes , Stem Cells/metabolism , Tumor Cells, Cultured
8.
Leuk Lymphoma ; 51(8): 1536-42, 2010 Aug.
Article En | MEDLINE | ID: mdl-20528246

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.


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
9.
Leuk Res ; 34(9): 1214-23, 2010 Sep.
Article En | MEDLINE | ID: mdl-20138667

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.


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
10.
Methods Mol Biol ; 596: 291-323, 2010.
Article En | MEDLINE | ID: mdl-19949929

A newly identified form of multidrug resistance (MDR) in tumor cells is presented, pertaining to the commonly encountered resistance of cancer cells to anticancer drug combinations at discrete drug:drug ratios. In vitro studies have revealed that whether anticancer drug combinations interact synergistically or antagonistically can depend on the ratio of the combined agents. Failure to control drug ratios in vivo due to uncoordinated pharmacokinetics could therefore lead to drug resistance if tumor cells are exposed to antagonistic drug ratios. Consequently, the most efficacious drug combination may not occur at the typically employed maximum tolerated doses of the combined drugs if this leads to antagonistic ratios in vivo after administration and resistance to therapeutic effects of the drug combination. Our approach to systematically screen a wide range of drug ratios and concentrations and encapsulate the drug combination in a liposomal delivery vehicle at identified synergistic ratios represents a means to mitigate this drug ratio-dependent MDR mechanism. The in vivo efficacy of the improved agents (CombiPlex formulations) is demonstrated and contrasted with the decreased efficacy when drug combinations are exposed to tumor cells in vivo at antagonistic ratios.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Antagonism , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/physiology , Neoplasms , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Combinations , Drug Delivery Systems , Drug Synergism , Humans , Liposomes/chemistry , Liposomes/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism
11.
Mol Cancer Ther ; 8(8): 2266-75, 2009 Aug.
Article En | MEDLINE | ID: mdl-19671743

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.


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.
J Med Chem ; 51(11): 3288-96, 2008 Jun 12.
Article En | MEDLINE | ID: mdl-18465845

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.


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
13.
Oncol Res ; 16(8): 361-74, 2007.
Article En | MEDLINE | ID: mdl-17913044

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.


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
14.
Mol Cancer Ther ; 5(7): 1854-63, 2006 Jul.
Article En | MEDLINE | ID: mdl-16891472

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.


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
15.
J Pharmacol Exp Ther ; 312(3): 1020-6, 2005 Mar.
Article En | MEDLINE | ID: mdl-15525796

Polyethylene glycol (PEG) is used widely in the pharmaceutical industry to improve the pharmacokinetics and reduce the immunogenicity of therapeutic and diagnostic agents. The incorporation of lipid-conjugated PEG into liposomal drug delivery systems greatly enhances the circulation times of liposomes by providing a protective, steric barrier against interactions with plasma proteins and cells. Here we report that liposome compositions containing PEG-lipid derivatives and encapsulated antisense oligodeoxynucleotide (ODN) or plasmid DNA elicit a strong immune response that results in the rapid blood clearance of subsequent doses in mice. The magnitude of this response is sufficient to induce significant morbidity and, in some instances, mortality. This effect has been observed in several strains of mice and was independent of sequence motifs, such as immunostimulatory CpG motifs. The ODN-to-lipid ratio and ODN dose was also determined to be important, with abrogation of the response occurring at a ratio between 0.04 and 0.08 (w/w). Rapid elimination of liposome-encapsulated ODN from blood depends on the presence of PEG-lipid in the membrane because the use of nonpegylated liposomes or liposomes containing rapidly exchangeable PEG-lipid also abrogated the response. These studies have important implications for the evaluation and therapeutic use of liposomal formulations of nucleic acid, as well as the potential development of liposomal vaccines.


DNA/administration & dosage , Lipids/administration & dosage , Liposomes/immunology , Liposomes/pharmacokinetics , Oligodeoxyribonucleotides, Antisense/administration & dosage , Polyethylene Glycols/administration & dosage , Animals , Drug Carriers , Female , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Plasmids
16.
Clin Cancer Res ; 10(2): 728-38, 2004 Jan 15.
Article En | MEDLINE | ID: mdl-14760096

PURPOSE: There is an opportunity to augment the therapeutic potential of drug combinations through use of drug delivery technology. This report summarizes data obtained using a novel liposomal formulation with coencapsulated doxorubicin and vincristine. The rationale for selecting these drugs is due in part to the fact that liposomal formulations of doxorubicin and vincristine are being separately evaluated as components of drug combinations. EXPERIMENTAL DESIGN: Doxorubicin and vincristine were coencapsulated into liposomes using two distinct methods of drug loading. A manganese-based drug loading procedure, which relies on drug complexation with a transition metal, was used to encapsulate doxorubicin. Subsequently the ionophore A23187 was added to induce formation of a pH gradient, which promoted vincristine encapsulation. RESULTS: Plasma elimination studies in mice indicated that the drug:drug ratio before injection [4:1 doxorubicin:vincristine (wt:wt ratio)] changed to 20:1 at the 24-h time point, indicative of more rapid release of vincristine from the liposomes than doxorubicin. Efficacy studies completed in MDA MB-435/LCC6 tumor-bearing mice suggested that at the maximum tolerated dose, the coencapsulated formulation was therapeutically no better than liposomal vincristine. This result was explained in part by in vitro cytotoxicity studies evaluating doxorubicin and vincristine combinations analyzed using the Chou and Talalay median effect principle. These data clearly indicated that simultaneous addition of vincristine and doxorubicin resulted in pronounced antagonism. CONCLUSION: These results emphasize that in vitro drug combination screens can be used to predict whether a coformulated drug combination will act in an antagonistic or synergistic manner.


Doxorubicin/pharmacology , Liposomes/metabolism , Vincristine/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/pathology , Calcimycin/pharmacology , Cell Line, Tumor , Coloring Agents/pharmacology , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Ionophores/pharmacology , Ions , Lipid Metabolism , Manganese/pharmacology , Manganese Compounds/pharmacology , Mice , Mice, SCID , Neoplasm Transplantation , Protons , Sulfates/pharmacology , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors
...