Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Control Release ; 296: 81-92, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30639692

ABSTRACT

Auristatins are a class of highly cytotoxic tubulin-disrupting peptides, which have shown limited therapeutic effect as free agents in clinical trials. In our continuing effort to develop acid-sensitive albumin-binding anticancer drugs exploiting circulating serum albumin as the drug carrier, we investigated the highly toxic drug payload auristatin E to assess whether the corresponding albumin-binding prodrugs were a viable option for achieving significant and concomitant tolerable antitumor activity. To achieve our goal, we developed a new aromatic maleimide-bearing linker (Sulf07) which enhanced both water solubility and stability of the prodrugs. In this study, we describe two auristatin E-based albumin-binding drugs, AE-Keto-Sulf07 and AE-Ester-Sulf07, which were designed to release the active compound at the tumor site in a pH-dependent manner. These prodrugs incorporate an acid-sensitive hydrazone bond, formed by the reaction of a carbonyl-containing auristatin E derivative with the hydrazide group of the water-solubilizing maleimide-bearing linker Sulf07. A panel of patient- and cell-derived human tumor xenograft models (melanoma A375, ovarian carcinoma A2780, non-small-cell lung cancer LXFA737 and LXFE937, and head and neck squamous cell carcinomas) were screened with starting tumor volumes in the range of either 130-150 mm3 (small tumors) or 270-380 mm3 (large tumors). Both albumin-binding prodrugs showed compelling anticancer efficacy compared to the parent drug auristatin E, inducing statistically significant long-term partial and/or complete tumor regressions. AE-Keto-Sulf07 displayed very good antitumor response over a wide dose range, 3.0-6.5 mg/kg (5-8 injections, biweekly). AE-Ester-Sulf07 was highly efficacious between 1.9 and 2.4 mg/kg (8 injections, biweekly) or at 3.8 mg/kg (4 injections, weekly), but caused cumulative skin irritation due to scratching and biting. In contrast at its MTD, auristatin E (0.3 mg/kg, 8 injections, biweekly) was only marginally active. In summary, AE-Keto-Sulf07 and AE-Ester-Sulf07 are novel acid-sensitive albumin-binding prodrugs demonstrating tumor regressions in all of the evaluated human tumor xenograft models thus supporting the stratagem that albumin can be used as an effective drug carrier for the highly potent class of auristatins.


Subject(s)
Aminobenzoates/administration & dosage , Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Oligopeptides/administration & dosage , Prodrugs/administration & dosage , Serum Albumin/metabolism , Aminobenzoates/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Liberation , Female , Humans , Hydrogen-Ion Concentration , Mice, Nude , Models, Molecular , Neoplasms/metabolism , Oligopeptides/chemistry , Prodrugs/chemistry , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
2.
J Med Chem ; 55(17): 7502-15, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22882004

ABSTRACT

Bone metastases are a frequent cause of morbidity in cancer patients. The present palliative therapeutic options are chemotherapy, hormone therapy, and the administration of bisphosphonates. The affinity between bisphosphonates and the apatite structure of bone metastases is strong. Thus, we designed two low-molecular-weight and water-soluble prodrugs which incorporate a bisphosphonate group as a bone targeting ligand, doxorubicin as the anticancer agent, and either an acid-sensitive bond (1) or a cathepsin B cleavable bond (3) for ensuring an effective release of doxorubicin at the site of action. Cleavage studies of both prodrugs showed a fast release of doxorubicin but sufficient stability over several hours in human plasma. Effective binding of prodrug 1 and 3 was demonstrated with hydroxyapatite and with native bone. In orientating toxicity studies in nude mice, the MTD of 1 was 3-fold higher compared to conventional doxorubicin, whereas 3 showed essentially the same MTD as doxorubicin.


Subject(s)
Bone Neoplasms/secondary , Cathepsin B/metabolism , Diphosphonates/therapeutic use , Doxorubicin/therapeutic use , Durapatite/metabolism , Prodrugs/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone and Bones/metabolism , Diphosphonates/metabolism , Diphosphonates/pharmacology , Doxorubicin/metabolism , Doxorubicin/pharmacology , Humans , Hydrogen-Ion Concentration , Prodrugs/metabolism , Prodrugs/pharmacology
3.
Breast Cancer Res Treat ; 134(1): 117-29, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22228402

ABSTRACT

The P-glycoprotein (P-gp) is a 170-kDa protein that acts as an energy dependent, transmembrane efflux pump and is encoded by the MDR1 gene. It has been shown to be responsible for multidrug resistance (MDR) in a defined subpopulation of breast cancer patients and thus represents a molecular target for circumventing MDR in this tumor indication. MDR modulators have been developed and demonstrated high selectivity for P-gp with inhibitory activities in the low nanomolar range. Although some objective responses were achieved in clinical trials, combination therapy with these MDR modulators, such as Ca2+ antagonists caused unacceptable toxicity. Targeting P-gp inhibitors to the tumor site is a mean to increase their therapeutic index, and in this context binding of tailor-made prodrugs to circulating albumin is an established technology to reduce the toxicity and enhance the efficacy of anticancer drugs. In this study, we consequently developed an acid-sensitive albumin-binding prodrug of the P-gp inhibitor zosuquidar (LY335979) in a two-step synthesis using a maleimide hydrazone linker system established in our laboratory that first introduces acetylbenzoic acid at the HO-group of zosuquidar followed by derivatization with 6-maleimidocaproyl hydrazide to form the acid-sensitive hydrazone bond. The maleimide group enables the prodrug to bind rapidly and selectively to the cysteine-34 position of endogenous albumin after intravenous administration. HPLC analysis demonstrated rapid albumin binding of the zosuquidar prodrug as well as the quantitative release of the acetylbenzoic ester derivative of zosuquidar at pH 5.0. Subsequently, its ability to circumvent MDR was tested in two doxorubicin-resistant breast carcinoma cell lines (MCF-7/ADR and MT-3/ADR). The MDR status of these cell lines can be reversed by zosuquidar which was confirmed in a rhodamine 123 assay using fluorescence microscopy and FACS analysis. Furthermore, zosuquidar as well its acid-sensitive albumin conjugate re-sensitized cells to doxorubicin as well as to an albumin-binding prodrug of doxorubicin, i.e., the 6-maleimidocaproyl hydrazone derivative of doxorubicin, achieving IC50 values in the same order of magnitude as the parental cell lines. Thus, a novel formulation of zosuquidar has been developed that could have the potential to improve the toxicity issues and tumor targeting properties of the original compound.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Dibenzocycloheptenes/pharmacology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Hydrazones/pharmacology , Prodrugs/pharmacology , Quinolines/pharmacology , Cell Line, Tumor/drug effects , Drug Carriers/pharmacology , Drug Design , Drug Stability , Drug Synergism , Female , Fluorescent Dyes/metabolism , Humans , Hydrolysis , Inhibitory Concentration 50 , Rhodamine 123/metabolism , Serum Albumin/pharmacology
4.
Eur J Cancer ; 48(13): 2054-65, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21937219

ABSTRACT

Two new water-soluble folate receptor-targeted drug conjugates that contain the highly active doxorubicin derivative N-(5,5-diacetoxybut-1-yl)doxorubicin were designed and evaluated for their biological activity against folate receptor positive tumours. The prodrugs were designed to contain an acid-sensitive hydrazone bond KO019 or in addition a disulphide bond KO013 in order to elucidate the importance of the pre-determined breaking point for their in vitro and in vivo properties. Fluorescence microscopy studies confirmed higher uptake of the prodrugs in folate receptor positive KB cells than in the folate receptor negative A549 lung cancer cells. In subsequent in vivo studies in the folate receptor positive KB xenograft model, KO019 was as active as the free drug but significantly less toxic when dosed at twice the dose of the free drug whereas KO013 showed no anticancer efficacy. As an explanation, we could show by HPLC that the prodrug KO013 that additionally contains a disulphide bond undergoes rapid disulphide exchange in murine plasma in the order of 40% after 5h at 37°C in contrast to KO019 which was essentially stable after a 5h incubation.


Subject(s)
Doxorubicin/analogs & derivatives , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/analogs & derivatives , Prodrugs/pharmacology , Pterins/pharmacology , Vinca Alkaloids/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Evaluation, Preclinical , Folate Receptors, GPI-Anchored/antagonists & inhibitors , Folic Acid/pharmacology , Mice , Mice, Nude , Prodrugs/chemistry , Xenograft Model Antitumor Assays
5.
Eur J Cancer ; 46(18): 3434-44, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20933385

ABSTRACT

In developed countries, prostate cancer is the third most common cause of death from cancer in men. Unfortunately, whilst accumulating clinical data have suggested that taxanes may prolong the survival in a subset of men with prostate carcinoma, the dose and duration of administration of these drugs are limited by their significant systemic toxicities due to a lack of tumour selectivity. In an attempt to improve both the water solubility and tumour-targeting properties of paclitaxel (Taxol®), we set out to develop a water soluble paclitaxel prodrug that is activated specifically by prostate-specific antigen (PSA) which is almost exclusively expressed in prostate tissue and prostate carcinoma making it an ideal molecular target for prodrug strategies. Using albumin as a drug carrier, we describe a novel albumin-binding prodrug of paclitaxel, EMC-Arg-Ser-Ser-Tyr-Tyr-Ser-Leu-PABC-paclitaxel [EMC: ε-maleimidocaproyl; PABC: p-aminobenzyloxycarbonyl] that was synthesised by reacting EMC-Arg-Ser-Ser-Tyr-Tyr-Ser-OH with H-Leu-PABC-paclitaxel. This prodrug was water soluble and was bound to endogenous and exogenous albumin. Moreover, incubation studies with PSA demonstrated that the albumin-bound form of the prodrug was cleaved rapidly at the P1-P1' scissile bond releasing the paclitaxel-dipeptide H-Ser-Leu-PABC-paclitaxel. Last but not least, due to the incorporation of a PABC self-eliminating linker, this dipeptide was rapidly degraded to liberate paclitaxel as a final cleavage product within a few hours in prostate tumour tissue homogenates. Of note was that the albumin-bound form of the prodrug was approximately three-fold more active in killing PSA-positive LNCaP cells than paclitaxel. In addition, orientating toxicity studies in mice showed that the maximum tolerated dose of the novel paclitaxel prodrug was twice that of conventional paclitaxel. When tested in vivo in an orthotopic mouse model of human prostate cancer using luciferase-transduced LNCaP LLN cells, both paclitaxel and the new paclitaxel prodrug showed distinct antitumour efficacy on the primary tumour and metastases that was significantly better than the effect of doxorubicin which was used as a comparison and showed no antitumour efficacy. The new paclitaxel prodrug (3 × 24 mg paclitaxel equivalents) showed comparable antitumour activity on the primary tumour to paclitaxel at its maximum-tolerated dose (3 × 12mg/kg), reduced circulating PSA levels and demonstrated a better antitumour effect on lung metastases but not on bone metastases.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Paclitaxel/therapeutic use , Prodrugs/therapeutic use , Prostate-Specific Antigen/physiology , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/pharmacokinetics , Chromatography, High Pressure Liquid , Drug Evaluation , Humans , Luminescence , Male , Maximum Tolerated Dose , Mice , Mice, SCID , Neoplasm Transplantation , Paclitaxel/chemical synthesis , Paclitaxel/pharmacokinetics , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Prostatic Neoplasms/metabolism , Tumor Cells, Cultured
6.
Bioconjug Chem ; 20(2): 390-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19199576

ABSTRACT

The aim of this work was to develop a new bifunctional maleimide linker for the development of dual-acting prodrugs that incorporate two pharmaceutically different anticancer agents independently bound by enzymatically cleavable substrates. The linker consists of a carboxyl group in one arm and an activated 1,6-self-immolative para-aminobenzyloxycarbonyl spacer together with a cathepsin B cleavable dipeptide Phe-Lys in the other. Aided with this linker, we have prepared a thiol-binding prodrug that contains the anticancer drugs doxorubicin and paclitaxel. Bound to the cysteine-34 position of albumin, it was cleaved efficiently by cathepsin B releasing the free drugs.


Subject(s)
Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism , Drug Design , Prodrugs/chemistry , Proteins/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cathepsin B/metabolism , Cross-Linking Reagents/chemical synthesis , Doxorubicin/chemistry , Doxorubicin/metabolism , Humans , Maleimides/chemistry , Paclitaxel/chemistry , Paclitaxel/metabolism , Prodrugs/chemical synthesis , Prodrugs/metabolism , Prodrugs/pharmacology , Serum Albumin/metabolism
7.
Cancer Chemother Pharmacol ; 64(2): 413-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19229536

ABSTRACT

PURPOSE: This study was designed to evaluate the in vitro and in vivo antitumor activity of an albumin-binding prodrug of doxorubicin 1 which incorporates a maleimide moiety and a para-aminobenzyloxycarbonyl (PABC) spacer coupled to the dipeptide Phe-Lys that is cleaved by cathepsin B. METHODS: Cleavage of the albumin conjugate was studied with cathepsin B and in homogenates of MDA-MB 435 tumors. For in vivo studies, nude mice were injected with (a) glucose buffer, (b) doxorubicin (2 x 8 mg/kg, i.v, on days 10 and 17), or (c) compound 1 (3 x 24 mg/kg doxorubicin equivalent, on days 10, 17 and 24). RESULTS: Prodrug 1 once bound to albumin was effectively cleaved by cathepsin B and in tumor homogenates releasing doxorubicin. A cytotoxicity assay of the albumin conjugate of 1 in two human tumor cell lines showed that doxorubicin was ~6 times more active than the conjugate. In contrast, in an in vivo study, the prodrug exhibited superior antitumor activity (T/C 15%) compared to doxorubicin (T/C 49%) in an equitoxic comparison. CONCLUSIONS: The cathepsin B cleavable spacer Phe-Lys-PABC incorporated in an albumin-binding prodrug is an effective way to increase the therapeutic index of doxorubicin.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Breast Neoplasms/drug therapy , Cathepsin B/metabolism , Doxorubicin/therapeutic use , Prodrugs/metabolism , Serum Albumin/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Dipeptides/metabolism , Female , Humans , In Vitro Techniques , Maleimides/metabolism , Mice , Mice, Nude , Prodrugs/chemical synthesis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Bioorg Med Chem Lett ; 19(3): 995-1000, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19131246

ABSTRACT

We have developed a novel dual-acting maleimide-bearing prodrug that incorporates the anticancer agent doxorubicin and an inhibitor of the P-glycoprotein efflux pump that is over-expressed in multidrug resistant tumor cells. Additionally, the prodrug contains a 1,6-self-immolative spacer coupled to the dipeptide Phe-Lys that acts as a substrate for cathepsin B. The prodrug, once bound through its maleimide moiety to the cysteine-34 group of human serum albumin, was cleaved by cathepsin B and in tumor homogenates demonstrating a release of the anticancer agent doxorubicin and the inhibitor.


Subject(s)
Cathepsin B/chemistry , Chemistry, Pharmaceutical/methods , Drug Resistance, Multiple , Prodrugs/chemical synthesis , ATP Binding Cassette Transporter, Subfamily B/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cysteine/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Design , Humans , Lysine/chemistry , Peptides/chemistry , Phenylalanine/chemistry , Prodrugs/pharmacology , Serum Albumin/chemistry
9.
Nat Immunol ; 9(7): 761-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18516040

ABSTRACT

Toll-like receptor 2 (TLR2) initiates inflammation in response to bacterial lipopeptide (BLP). However, the molecular mechanisms enabling the detection of BLP by TLR2 are unknown. Here we investigated the interaction of BLP with human serum proteins and identified vitronectin as a BLP-recognition molecule. Vitronectin and its receptor, integrin beta(3), were required for BLP-induced TLR2-mediated activation of human monocytes. Furthermore, monocytes from patients with Glanzmann thrombasthenia, which lack integrin beta(3), were completely unresponsive to BLP. In addition, integrin beta(3) formed a complex with TLR2 and this complex dissociated after BLP stimulation. Notably, vitronectin and integrin beta(3) coordinated responses to other TLR2 agonists such as lipoteichoic acid and zymosan. Our findings show that vitronectin and integrin beta(3) contribute to the initiation of TLR2 responses.


Subject(s)
Bacterial Proteins/immunology , Integrin beta3/immunology , Monocytes/immunology , Toll-Like Receptor 2/immunology , Vitronectin/immunology , Cell Differentiation/immunology , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunoprecipitation , Integrin beta3/metabolism , Lymphocyte Activation/immunology , Monocytes/cytology , Reverse Transcriptase Polymerase Chain Reaction , Thrombasthenia/immunology , Thrombasthenia/metabolism , Toll-Like Receptor 2/metabolism , Vitronectin/metabolism
10.
Expert Opin Investig Drugs ; 16(7): 1037-58, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17594188

ABSTRACT

Coupling of low molecular weight anticancer drugs to antibodies, serum proteins or polymers through a cleavable linker has been an effective method for improving the therapeutic index of cytotoxic established agents. Modern drug-antibody conjugates that have recently entered clinical trials have primarily used highly potent drugs such as calicheamicin or maytansins. Gemtuzumab ozogamicin, a conjugate of calicheamicin and an anti-CD33 humanized antibody, is the first drug-antibody conjugate to receive market approval. Drug conjugates that have undergone clinical assessment include N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer conjugates with doxorubicin, camptothecin, paclitaxel and Pt(II) complexes, poly(ethylene glycol) conjugates with camptothecin and paclitaxel, polyglutamate conjugates with paclitaxel and camptothecin, a methotrexate-albumin conjugate and an albumin-binding doxorubicin prodrug. This review summarizes the Phase I-III studies that have been performed with these macromolecular prodrugs.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Carriers/therapeutic use , Drugs, Investigational , Neoplasms/drug therapy , Prodrugs/therapeutic use , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Drug Carriers/pharmacology , Drug Delivery Systems , Female , Humans , Male , Neoplasms/pathology , Prodrugs/pharmacology , Sensitivity and Specificity
11.
Biochim Biophys Acta ; 1758(4): 552-61, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16681993

ABSTRACT

Liposomes equipped with cellular uptake-mediating peptidic vector compounds have attracted much attention as target-specific drug delivery systems. Aside from the development of the target recognition motif itself, vector coupling to liposomes while conserving the active conformation constitutes an important element in carrier development. To elucidate the most efficient way for adsorptive peptide binding to liposomes, we synthesized and characterized two-domain peptides comprising a cationic sequence derived from the binding domain of apolipoprotein E (apoE) for the low-density lipoprotein receptor and different lipid-binding motifs, that is, an amphipathic helix, a transmembrane helix, single fatty acids or two palmitoyl chains. Peptide properties considered relevant for peptide-liposome complexes to initiate an endocytotic cellular uptake such as lipid binding, helicity, stability of anchorage, bilayer-disturbing activity, and toxicity showed that the dipalmitoyl derivative was the most suitable to associate the apoE peptide to the surface of liposomes. The peptide showed pronounced lipid affinity and was stably anchored within the lipid bilayer on a time scale of at least 30 min. The helicity of about 40% in the lipid-bound state and the location of the amphipathic helix on the liposomal surface provided the prerequisites for interaction of the complex with the cell surface-located receptor. The concentration of the dipalmitoylated peptide to permeabilize neutral lipid bilayers (lipid concentration 25 microM) was 0.06 microM and a 2 microM concentration reduced cell viability to about 80%. Efficient internalization of liposomes bearing about 180 peptide derivatives on the surface into brain capillary endothelial cells was monitored by confocal laser scanning microscopy. The concept of complexation using dipalmitoylated peptides may offer an efficient substitute to covalent vector coupling and a prospective way to optimize the capacity of liposomes as drug delivery systems also for different targets.


Subject(s)
Apolipoproteins E/metabolism , Drug Carriers , Liposomes , Palmitates , Peptide Fragments/chemistry , Amino Acid Sequence , Binding Sites , Circular Dichroism , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Transport , Receptors, LDL/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...