Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 12772, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32728172

ABSTRACT

Folate receptor (FR)-targeted small molecule drug conjugates (SMDCs) have shown promising results in early stage clinical trials with microtubule destabilizing agents, such as vintafolide and EC1456. In our effort to develop FR-targeted SMDCs with varying mechanisms of action, we synthesized EC2629, a folate conjugate of a DNA crosslinking agent based on a novel DNA-alkylating moiety. This agent was found to be extremely potent with an in vitro IC50 ~ 100× lower than folate SMDCs constructed with various microtubule inhibitors. EC2629 treatment of nude mice bearing FR-positive KB human xenografts led to cures in 100% of the test animals with very low dose levels (300 nmol/kg) following a convenient once a week schedule. The observed activity was not accompanied by any noticeable weight loss (up to 20 weeks post end of dosing). Complete responses were also observed against FR-positive paclitaxel (KB-PR) and cisplatin (KB-CR) resistant models. When evaluated against FR-positive patient derived xenograft (PDX) models of ovarian (ST070), endometrial (ST040) and triple negative breast cancers (ST502, ST738), EC2629 showed significantly greater anti-tumor activity compared to their corresponding standard of care treatments. Taken together, these studies thus demonstrated that EC2629, with its distinct DNA reacting mechanism, may be useful in treating FR-positive tumors, including those that are classified as drug resistant.


Subject(s)
Antineoplastic Agents/pharmacology , Cross-Linking Reagents/pharmacology , DNA/chemistry , Endometrial Neoplasms/drug therapy , Folate Receptors, GPI-Anchored/chemistry , Ovarian Neoplasms/drug therapy , Triple Negative Breast Neoplasms/drug therapy , Alkylating Agents/chemistry , Animals , Cattle , Cisplatin/administration & dosage , Dogs , Drug Delivery Systems , Drug Design , Drug Evaluation, Preclinical , Female , Folic Acid/analogs & derivatives , Folic Acid/pharmacology , Humans , Inhibitory Concentration 50 , KB Cells , Ligands , Mice , Mice, Inbred C57BL , Mice, Nude , Paclitaxel/administration & dosage , Rats , Vinca Alkaloids/pharmacology , Xenograft Model Antitumor Assays
2.
Mol Pharm ; 16(9): 3985-3995, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31356752

ABSTRACT

Folate-based small molecule drug conjugates (SMDCs) are currently under development and have shown promising preclinical and clinical results against various cancers and polycystic kidney disease. Two requisites for response to a folate-based SMDC are (i) folate receptor alpha (FRα) protein is expressed in the diseased tissues, and (ii) FRα in those tissues is accessible and functionally competent to bind systemically administered SMDCs. Here we report on the development of a small molecule reporter conjugate (SMRC), called EC2220, which is composed of a folate ligand for FRα binding, a multilysine containing linker that can cross-link to FRα in the presence of formaldehyde fixation, and a small hapten (fluorescein) used for immunohistochemical detection. Data show that EC2220 produces a far greater IHC signal in FRα-positive tissues over that produced with EC17, a folate-fluorescein SMRC that is released from the formaldehyde-denatured FRα protein. Furthermore, the extent of the EC2220 IHC signal was proportional to the level of FRα expression. This EC2220-based assay was qualified both in vitro and in vivo using normal tissue, cancer tissue, and polycystic kidneys. Overall, EC2220 is a sensitive and effective reagent for evaluating functional and accessible receptor expression in vitro and in vivo.


Subject(s)
Folate Receptor 1/metabolism , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Polycystic Kidney Diseases/drug therapy , A549 Cells , Animals , Doxycycline/pharmacology , Fluorescein-5-isothiocyanate/chemistry , Fluorescein-5-isothiocyanate/metabolism , Folate Receptor 1/analysis , Folic Acid/analogs & derivatives , Folic Acid/chemistry , Folic Acid/metabolism , HeLa Cells , Humans , Lysine/analogs & derivatives , Lysine/chemistry , Lysine/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Neoplasms/metabolism , Oligopeptides/chemistry , Oligopeptides/metabolism , Polycystic Kidney Diseases/chemically induced , Polycystic Kidney Diseases/metabolism , Protein Kinase C/genetics , Tissue Distribution , Trityl Compounds/chemistry , Trityl Compounds/metabolism , Xenograft Model Antitumor Assays
3.
Bioconjug Chem ; 30(6): 1805-1813, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31075200

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a biomarker that is overexpressed on prostate cancer, and it is also present on the neovasculature within many non-prostate solid tumors. Herein, we report on the construction and biological testing of novel tubulysin B-containing therapeutic agents for the treatment of PSMA-expressing cancer. One of these compounds, EC1169, emerged as a lead candidate for preclinical development and phase 1 clinical testing. This water-soluble conjugate was shown to have high affinity for PSMA-positive cells. When tested in vitro, EC1169 was found to inhibit the growth of PSMA-positive cells, but it displayed no activity against PSMA-negative cells. Brief treatment of nude mice bearing PSMA-positive LNCaP human xenografts with EC1169 led to complete remissions and cures. Furthermore, this activity occurred in the absence of weight loss. In contrast, the nontargeted tubulysin B drug proved to be inactive against the LNCaP tumor model when administered at doses near to or greater than the maximum tolerated level. PSMA-negative KB tumors did not appreciably respond to EC1169 therapy, thereby confirming this compound's targeted specificity for PSMA-positive cells. Finally, treatment of LNCaP-bearing mice with docetaxel (the most active chemotherapeutic agent approved for late stage prostate cancer therapy) was found to produce only modest anti-tumor activity, and this outcome was also associated with severe weight loss. Taken together, these results strongly indicate that PSMA-positive tumors may be effectively treated using highly potent, PSMA-targeted small-molecule drug conjugates using regimens that do not cause undesirable side effects.


Subject(s)
Antigens, Surface/analysis , Antineoplastic Agents/therapeutic use , Glutamate Carboxypeptidase II/analysis , Oligopeptides/therapeutic use , Pipecolic Acids/therapeutic use , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Male , Mice, Nude , Oligopeptides/chemistry , Pipecolic Acids/chemistry , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
Sci Rep ; 8(1): 8943, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29895863

ABSTRACT

EC1456 is a folate-tubulysin conjugate constructed with an all-D enantiomeric spacer/linker configuration. When tested against folate receptor (FR)-positive cells, EC1456 demonstrated dose-responsive activity with an approximate 1000-fold level of specificity. Treatment of nude mice bearing FR-positive human xenografts (as large as 800 mm3) with non-toxic doses of EC1456 led to cures in 100% of the mice. Combinations of low dose EC1456 with standard of care agents such as platins, taxanes, topotecan and bevacizumab, safely and significantly augmented the growth inhibitory effects of these commonly used agents. When tested against FR-positive human tumor xenograft models having confirmed resistance to a folate-vinca alkaloid (vintafolide), cisplatin or paclitaxel, EC1456 was found to generate partial to curative responses. Taken together, these studies demonstrate that EC1456 has significant anti-proliferative activity against FR-positive tumors, including models which were anticancer drug resistant, thereby justifying a Phase 1 trial of this agent for the treatment of advanced human cancers.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Folic Acid/chemistry , Neoplasms/drug therapy , Oligopeptides/chemistry , Pipecolic Acids/chemistry , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/administration & dosage , Bevacizumab/administration & dosage , Carboplatin/administration & dosage , Cell Line, Tumor , Cisplatin/administration & dosage , Docetaxel/administration & dosage , Drug Evaluation, Preclinical/methods , Humans , Mice, Nude , Neoplasms/pathology , Paclitaxel/administration & dosage , Topotecan/administration & dosage , Tumor Burden/drug effects
5.
Bioconjug Chem ; 28(12): 2921-2931, 2017 Dec 20.
Article in English | MEDLINE | ID: mdl-29211454

ABSTRACT

Pyrrolobenzodiazepines (PBDs) and their dimers (bis-PBDs) have emerged as some of the most potent chemotherapeutic compounds, and are currently under development as novel payloads in antibody-drug conjugates (ADCs). However, when used as stand-alone therapeutics or as warheads for small molecule drug conjugates (SMDCs), dose-limiting toxicities are often observed. As an elegant solution to this inherent problem, we designed diazepine-ring-opened conjugated prodrugs lacking the imine moiety. Once the prodrug (pro-PBD) conjugate enters a targeted cell, cleavage of the linker system triggers the generation of a reactive intermediate possessing an aldehyde and aromatic amine. An intramolecular ring-closing reaction subsequently takes place as the aromatic amine adds to the aldehyde with the loss of water to give the imine and, as a result, the diazepine ring. In our pro-PBDs, we mask the aldehyde as a hydrolytically sensitive oxazolidine moiety which in turn is a part of a reductively labile self-immolative linker system. To prove the range of applications for this new class of latent DNA-alkylators, we designed and synthesized several novel latent warheads: pro-PBD dimers and hybrids of pro-PBD with other sequence-selective DNA minor groove binders. Preliminary preclinical pharmacology studies showed excellent biological activity and specificity.


Subject(s)
Benzodiazepines/chemistry , Benzodiazepines/metabolism , Drug Design , Molecular Targeted Therapy , Neoplasms/drug therapy , Prodrugs/chemical synthesis , Prodrugs/metabolism , Pyrroles/chemistry , Pyrroles/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzodiazepines/pharmacology , Benzodiazepines/therapeutic use , Chemistry Techniques, Synthetic , Humans , KB Cells , Neoplasms/pathology , Prodrugs/chemistry , Pyrroles/pharmacology , Pyrroles/therapeutic use
6.
Cancer Chemother Pharmacol ; 79(6): 1151-1160, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28451831

ABSTRACT

PURPOSE: EC0305 represents a folate-tubulysin B construct capable of specifically eradicating folate receptor (FR)-positive subcutaneous tumors from mice (Leamon et al., Cancer Res 68:9839-9844, 8). Herein we report on the use of multiple polar carbohydrate segments (e.g. 1-amino-1-deoxy-glucitolyl-γ-glutamate) placed in-between the folate and tubulysin B moieties of EC0305 creating a new conjugate, herein referred to as EC0531, with more desirable biological properties. METHODS: The synthesis of EC0531 and its tritium-labeled counterpart are described. EC0531's affinity for FR binding and specific cytotoxic activity was assessed using standard in vitro assays. Human tumor xenografts were used to directly compare EC0305 and EC0531's antitumor activity. Finally, bile duct cannulated, female Sprague-Dawley rats were used to compare hepatobiliary clearance of these two targeted chemotherapeutic agents. RESULTS: EC0531 tightly binds to the FR with an affinity about half that of folic acid. It was found to specifically inhibit the growth of FR+ cells (IC50 of ~2 nM) in a dose-dependent manner. Using 3H-labeled compounds, more than a 12-fold higher amount of tubulysin was measured in a FR + human tumor xenograft compared to the unconjugated drug, a finding that explains, in part, why EC0531 displays curative activity, whereas the unconjugated tubulysin agent is essentially inactive. EC0531 was found to produce greater FR-specific anti-tumor activity at lower dose levels than EC0305; furthermore, EC0531's maximum tolerated dose level was significantly higher than that of EC0305, likely because EC0531's saccharopeptidic-based spacer allows for ~sixfold reduction in hepatic clearance. CONCLUSIONS: These data provide additional evidence that the therapeutic range of targeted small-molecule drug conjugates can be favorably increased using molecular spacers constructed with 1-amino-1-deoxy-glucitolyl-γ-glutamate residues.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Folate Receptor 1/drug effects , Folic Acid/analogs & derivatives , Oligopeptides/chemistry , Oligopeptides/pharmacology , Pipecolic Acids/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Bile/metabolism , Dose-Response Relationship, Drug , Drug Delivery Systems , Female , Folic Acid/metabolism , Folic Acid/pharmacology , Humans , Isotope Labeling , Liver/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 15(8): 1998-2008, 2016 08.
Article in English | MEDLINE | ID: mdl-27256377

ABSTRACT

Targeting surface receptors overexpressed on cancer cells is one way to specifically treat cancer versus normal cells. Vintafolide (EC145), which consists of folate linked to a cytotoxic small molecule, desacetylvinblastine hydrazide (DAVLBH), takes advantage of the overexpression of folate receptor (FR) on cancer cells. Once bound to FR, vintafolide enters the cell by endocytosis, and the reducing environment of the endosome cleaves the linker, releasing DAVLBH to destabilize microtubules. Vintafolide has shown efficacy and improved tolerability compared with DAVLBH in FR-positive preclinical models. As the first FR-targeting drug to reach the clinic, vintafolide has achieved favorable responses in phase II clinical trials in FR-positive ovarian and lung cancer. However, some FR-positive patients in these clinical trials do not respond to vintafolide. We sought to identify potential biomarkers of resistance to aid in the future development of this and other FR-targeting drugs. Here, we confirm that high P-glycoprotein (P-gp) expression was the strongest predictor of resistance to DAVLBH in a panel of 359 cancer cell lines. Furthermore, targeted delivery of DAVLBH via the FR, as in vintafolide, fails to overcome P-gp-mediated efflux of DAVLBH in both in vitro and in vivo preclinical models. Therefore, we suggest that patients whose tumors express high levels of P-gp be excluded from future clinical trials for vintafolide as well as other FR-targeted therapeutics bearing a P-gp substrate. Mol Cancer Ther; 15(8); 1998-2008. ©2016 AACR.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Folic Acid/analogs & derivatives , Gene Expression , Vinca Alkaloids/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cluster Analysis , Computational Biology/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Folate Receptors, GPI-Anchored/antagonists & inhibitors , Folic Acid/pharmacology , Gene Expression Profiling , Humans , Mice , Platinum/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Xenograft Model Antitumor Assays
8.
J Pharm Biomed Anal ; 122: 148-56, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26855286

ABSTRACT

EC1456 is a novel folate-targeted small molecule drug conjugate of tubulysin B hydrazide being developed as an anticancer agent for patients with advanced solid tumors expressing the folate receptor. To try and correlate circulating systemic levels of EC1456 and its metabolites to tumor concentrations and potentially develop a PK/PD model, a sensitive bioanalytical method was developed and validated for the quantitation of the analytes in KB tumor homogenates. The method involved homogenizing tumors with buffer containing N-maleoyl-ß-alanine, mannitol and acetic acid, precipitation of the homogenate with acetone followed by heating at 55°C for 1h to convert tubulysin B hydrazide to its corresponding hydrazone. The extracts were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The method demonstrated good inter-day (3 runs, n=18) accuracy (-2.3% to 7.3%) and precision (1.7% to 10.3%) for all three analytes. Stability was established for three freeze-thaw cycles, 4h on the bench-top on ice, 20h in the autosampler at 8°C and for at least 46days frozen at -70°C. This method was successfully used to determine concentration of EC1456 and its metabolites tubulysin B hydrazide and tubulysin B in tumor homogenates in preliminary experiments with KB tumor bearing mice dosed intravenously with EC1456.


Subject(s)
Chromatography, High Pressure Liquid/methods , Folic Acid/metabolism , Neoplasms/metabolism , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Small Molecule Libraries/chemistry , Tandem Mass Spectrometry/methods , Animals , Female , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/metabolism , Pipecolic Acids/metabolism , Small Molecule Libraries/pharmacology
9.
Drug Resist Updat ; 17(4-6): 89-95, 2014.
Article in English | MEDLINE | ID: mdl-25457975

ABSTRACT

Conventional cancer treatment modalities have several limitations including lack of sufficient efficacy, serious untoward toxicity, as well as innate and acquired drug resistance. In contrast, targeted imaging agents can identify patients with receptors overexpressed on the surface of cancer cells, thus allowing appropriate selection of patients for personalized treatment with a desirable targeted therapeutic. The folate receptor (FR) has been identified as a new molecularly targeted entity, which is highly overexpressed on the surface of a spectrum of solid tumor cells, including ovarian, kidney, lung, brain, endometrial, colorectal, pancreatic, gastric, prostate, testicular, bladder, head and neck, breast, and non-small cell lung cancer. Folic acid conjugation is a novel approach for targeting FR-expressing tissues for personalized treatment. With the development of FRα-targeted therapies comes a concomitant prerequisite for reliable methods for the quantification of FRα tissue expression. Therefore, attaching a radioactive probe to folic acid to target diseased tissue has become a novel and powerful imaging technique. Currently available diagnostic tools frequently require invasive surgical biopsy. In contrast, the noninvasive single-photon emission computed tomography-based companion imaging agent, (99m)Tc-etarfolatide ((99m)Tc-EC20), is in development for use as a companion diagnostic with the FRα-targeted folate conjugate, vintafolide (EC145), to identify patients whose tumors express FRα. Vintafolide is a folic acid conjugate of Vinca alkaloid (desacetylvinblastine hydrazide) that targets FRα-expressing tumors, thereby disrupting microtubule polymerization. (99m)Tc-etarfolatide is taken up by FR-positive tumors and allows for noninvasive, whole-body monitoring of FRα expression status throughout treatment. The combination of vintafolide plus etarfolatide has been evaluated in three Phase 2 studies for the treatment of various solid tumors, including ovarian, endometrial, peritoneal, and platinum-resistant ovarian cancer, as well as lung cancer. Patients with FR-positive tumors, as identified by etarfolatide uptake, have had better clinical outcomes than patients with FR-negative tumors, indicating the potential of etarfolatide as a companion biomarker for predicting vintafolide response. Targeted therapies combined with a reliable companion diagnostic test represent a novel approach toward efficient personalized medicine for malignant and nonmalignant disorders. Furthermore, the recent availability of the crystal structures of FRα and FRß in complex with folates and antifolates forms a realistic basis for the rational design and implementation of novel FR-targeted drugs for the treatment of cancer and inflammatory disorders.


Subject(s)
Folic Acid Antagonists/pharmacology , Folic Acid Antagonists/therapeutic use , Folic Acid Transporters/metabolism , Folic Acid/metabolism , Neoplasms/drug therapy , Animals , Humans , Molecular Targeted Therapy , Precision Medicine/methods
10.
Bioconjug Chem ; 25(3): 560-8, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24564229

ABSTRACT

Vintafolide is a potent folate-targeted vinca alkaloid small molecule drug conjugate (SMDC) that has shown promising results in multiple clinical oncology studies. Structurally, vintafolide consists of 4 essential modules: (1) folic acid, (2) a hydrophilic peptide spacer, (3) a disulfide-containing, self-immolative linker, and (4) the cytotoxic drug, desacetylvinblastine hydrazide (DAVLBH). Here, we report a structure-activity study evaluating the biological impact of (i) substituting DAVLBH within the vintafolide molecule with other vinca alkaloid analogues such as vincristine, vindesine, vinflunine, or vinorelbine; (ii) substituting the naturally (S)-configured Asp-Arg-Asp-Asp-Cys peptide with alternative hydrophilic spacers of varied composition; and (iii) varying the composition of the linker module. A series of vinca alkaloid-containing SMDCs were synthesized and purified by HPLC and LCMS. The SMDCs were screened in vitro against folate receptor (FR)-positive cells, and anti-tumor activity was tested against well-established subcutaneous FR-positive tumor xenografts. The cytotoxic and anti-tumor activity was directly compared to that produced by vintafolide. Among all the folate vinca alkaloid SMDCs tested, DAVLBH-containing SMDCs were active, while those constructed with vincristine, vindesine, or vinorelbine analogues failed to produce meaningful biological activity. Within the DAVLBH series, having a bioreleasable, self-immolative linker system was found to be critical for activity since multiple analogues constructed with thioether-based linkers all failed to produce meaningful activity both in vitro and in vivo. Substitutions of some or all of the natural amino acids within vintafolide's hydrophilic spacer module did not significantly change the in vitro or in vivo potency of the SMDCs. Vintafolide remains one of the most potent folate-vinca alkaloid SMDCs produced to date, and continued clinical development is warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Folic Acid/pharmacology , Neoplasms, Experimental/drug therapy , Vinca Alkaloids/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Folic Acid/chemistry , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Conformation , Neoplasms, Experimental/pathology , Stereoisomerism , Structure-Activity Relationship , Vinca Alkaloids/chemistry
11.
Clin Cancer Res ; 20(8): 2104-14, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24429878

ABSTRACT

PURPOSE: When evaluated in patients with ovarian and other cancer, vintafolide (EC145), a potent folate-targeted vinca alkaloid conjugate, displayed a toxicity profile that seemed to be nonoverlapping with many standard-of-care cancer therapeutics. It was, therefore, hypothesized that combining vintafolide with certain approved anticancer drugs may afford greater therapeutic efficacy compared with single-agent therapy. To explore this concept, vintafolide was evaluated in combination with pegylated liposomal doxorubicin (PLD; DOXIL), cisplatin, carboplatin, paclitaxel, docetaxel, topotecan, and irinotecan against folate receptor (FR)-positive models. EXPERIMENTAL DESIGN: FR-expressing KB, M109, IGROV, and L1210 cells were first exposed to graded concentrations of vintafolide, either alone or in combination with doxorubicin (active ingredient in PLD), and isobologram plots and combination index values generated. The vintafolide combinations were also studied in mice bearing various FR-expressing tumors. RESULTS: Vintafolide displayed strong synergistic activity against KB cells when combined with doxorubicin, and no less-than-additive effects resulted when tested against M109, IGROV, and L1210 cells. In contrast, when either desacetylvinblastine hydrazide (DAVLBH; the vinca alkaloid moiety in vintafolide) or vindesine (the vinca alkaloid most structurally similar to DAVLBH) were tested in combination with doxorubicin, less-than-additive antitumor effects were observed. In vivo, all vintafolide drug combinations produced far greater antitumor effect (complete responses and cures) compared with the single agents alone, without significant increase in overall toxicity. Importantly, these benefits were not observed with combinations of PLD and DAVLBH or vindesine. CONCLUSIONS: On the basis of these encouraging preclinical results, clinical studies to evaluate vintafolide drug combination therapies are now under way.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms, Experimental/drug therapy , Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Carboplatin/administration & dosage , Carboplatin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Docetaxel , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Folic Acid/administration & dosage , Folic Acid/analogs & derivatives , Folic Acid/pharmacology , HeLa Cells , Humans , Irinotecan , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Nude , Neoplasms/pathology , Neoplasms, Experimental/pathology , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Taxoids/administration & dosage , Taxoids/pharmacology , Topotecan/administration & dosage , Topotecan/pharmacology , Treatment Outcome , Vinca Alkaloids/administration & dosage , Vinca Alkaloids/pharmacology
12.
Cancer Res ; 73(2): 875-84, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23204225

ABSTRACT

Folate receptors (FR) may be of use for targeted delivery of cytotoxic drugs in invasive urothelial carcinoma (iUC), for which improved therapy is needed. FR expression and function in iUC were explored and the antitumor activity and toxicity of a folate-targeted vinblastine conjugate were evaluated in dogs with naturally occurring iUC, an excellent model for human iUC. FR immunohistochemistry was carried out on iUC and normal human and dog bladder tissues together with nuclear scintigraphy in dogs to monitor iUC folate uptake. Dose escalation of a folate-targeted vinblastine compound, EC0905, was conducted in dogs with biopsy-confirmed, FR-positive iUC. FRs were detected by immunohistochemistry (PU17) in most primary iUC and many nodal and lung metastases from dogs, and scintigraphy confirmed folate uptake in both primary and metastatic lesions. The maximum tolerated dose of EC0905 in dogs was 0.25 mg/kg IV weekly, with neutropenia at higher doses. Tumor responses included partial remission (≥ 50% reduction in tumor volume) in five dogs and stable disease (<50% change in tumor volume) in four dogs. Immunoreactivity to PU17 was similar in humans (78% of primary iUC, 80% of nodal metastases). Less immunoreactivity to mab343 (22% of cases) occurred. FR-ß was noted in 21% of human iUC cases. Our findings suggest folate-targeted therapy holds considerable promise for treating iUC, where FR-ß may be important in addition to FR-α.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Folate Receptors, GPI-Anchored/antagonists & inhibitors , Urinary Bladder Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Disease Models, Animal , Dogs , Female , Folic Acid/analogs & derivatives , Folic Acid/therapeutic use , Folic Acid/toxicity , Humans , Male , Maximum Tolerated Dose , Mice , Mice, Nude , Middle Aged , Pilot Projects , Vinblastine/analogs & derivatives , Vinblastine/therapeutic use , Vinblastine/toxicity
13.
Bioorg Med Chem Lett ; 21(22): 6778-81, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21974954

ABSTRACT

Tubuylsins are extremely potent cytotoxic agents which inhibit tubulin polymerization and lead to cell cycle arrest and apoptosis. Tubulysins have been isolated from fermentation mixtures and have been chemically synthesized; however, these efforts have been hampered by poor yields and arduous purifications. In contrast, treatment of a mixture of natural tubulysins A, B, C, G, and I, obtained from a fermentation batch with trifluoroacetic acid results in the formation of a single N-acyliminium ion. Subsequent addition of butyric, isopentyl, or acetic acid results in the formation of tubulysin B, A, or I, respectively, as a single species. New tubulysin analogs can be formed upon treatment of the acyliminium ion with other nucleophiles such as alcohols, thiols, and nitriles, resulting in corresponding N-acyl-N,O-acetals, N-acyl-N,S-thioacetals, and N,N'-diacyl-aminals. Carbon-carbon bond formation is also possible with a modification of this protocol. The cytotoxicies of the natural tubulysins and tubulysin analogs synthesized by this method were evaluated on KB cells.


Subject(s)
Biological Products/chemical synthesis , Chemistry Techniques, Synthetic/methods , Myxococcales/chemistry , Oligopeptides/chemical synthesis , Pipecolic Acids/chemical synthesis , Tubulin Modulators/chemical synthesis , Acetic Acid/chemistry , Biological Products/chemistry , Chemistry Techniques, Synthetic/economics , Imines/chemistry , Ions/chemistry , Oligopeptides/chemistry , Pipecolic Acids/chemistry , Tubulin Modulators/chemistry
14.
J Pharmacol Exp Ther ; 336(2): 336-43, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20978169

ABSTRACT

During a phase I trial of EC145 (a folate-targeted vinca alkaloid conjugate), constipation was identified as the dose-limiting toxicity, probably from a nonfolate receptor-related liver clearance process capable of releasing unconjugated vinca alkaloid from EC145 and shuttling it to the bile. Here, we report on the selective placement of novel carbohydrate segments (1-amino-1-deoxy-glucitolyl-γ-glutamate) spaced in-between the folate and vinca alkaloid moieties of EC145, which yielded a new agent (EC0489) that is equipotent but less toxic than EC145. Whereas both compounds could cure tumor-bearing mice reproducibly, EC0489 differed from EC145 with i) a shorter elimination half-life, ii) approximately 70% decrease in bile clearance, iii) a 4-fold increase in urinary excretion, and iv) improved tolerability in rodents. This combination of improvements justified the clinical evaluation of EC0489 where currently administered dose levels have exceeded the maximal tolerated dose of EC145 by approximately 70%, thereby reflecting the translational benefits to this new approach.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Folic Acid/analogs & derivatives , Liver/metabolism , Vinca Alkaloids/pharmacokinetics , Animals , Antineoplastic Agents/toxicity , Dose-Response Relationship, Drug , Drug Discovery , Female , Folate Receptors, GPI-Anchored/physiology , Folic Acid/pharmacokinetics , Folic Acid/toxicity , Kidney/metabolism , Male , Metabolic Clearance Rate , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Vinca Alkaloids/toxicity
15.
Mol Pharm ; 7(2): 597-604, 2010 Apr 05.
Article in English | MEDLINE | ID: mdl-20199053

ABSTRACT

Administration of certain antifolates before radiofolate application has previously proven to have a positive effect on undesired kidney uptake of radiofolates in mice bearing human tumor xenografts. The aims of this study were to (i) test the effects of the antifolates, pemetrexed and CB3717, on tissue distribution of the clinically investigated radiofolate, (99m)Tc-EC20, and (ii) to determine if pemetrexed's kidney-selective blocking effect also functions in mice bearing syngeneic tumors. Relative binding affinities of pemetrexed and CB3717 were determined in folate receptor (FR)-positive KB cells at 0 and 37 degrees C using (3)H-folic acid. In vivo studies were performed in nude mice with KB tumor xenografts (A) and in Balb/c mice bearing FR-positive M109 tumor grafts (B). (99m)Tc-EC20 was prepared via a kit formulation. The antifolates pemetrexed and CB3717 (20 mumol/kg body weight) were administered intravenously 1 h before injection of (99m)Tc-EC20 (67 nmol/kg body weight). Similar to previously published data we found that FR-binding affinities of pemetrexed and CB3717 at 0 degrees C were in the same range as that of folic acid. Interestingly, experiments performed at 37 degrees C showed that pemetrexed has a nearly approximately 700-fold lower FR-affinity than CB3717. Tissue distribution of (99m)Tc-EC20 was largely comparable in both animal models (A and B). Radiofolate accumulation was found in FR-positive tumors (A, 8.92 +/- 2.14% ID/g; B, 15.02 +/- 0.95% ID/g) and FR-positive kidneys (A, 59.10 +/- 8.03% ID/g; B, 69.44 +/- 4.66% ID/g, 4 h p.i.). Preinjection of pemetrexed resulted in a significant decrease of (99m)Tc-EC20 uptake in kidney (A, 18.80 +/- 2.73% ID/g; B, 15.27 +/- 2.64% ID/g; 4 h p.i), whereas uptake in the tumors was unaltered. However, administration of the CB3717 resulted in a reduction of (99m)Tc-EC20 uptake in both the kidney and tumor (<1% ID/g, 4 h p.i.). We have thus demonstrated that pemetrexed effectively reduces kidney uptake of radiofolates not only in xenografted mice but also in a syngeneic tumor mouse model, thereby indicating that the kidney-specific blocking effect is not based on differences between human and murine FRs that are expressed in xenografts and kidneys, respectively. This effect was not observed with the antifolate, CB3717, which targets the FR selectively in contrast to pemetrexed that is predominantly transported into cells through carrier systems.


Subject(s)
Folic Acid Antagonists/pharmacology , Folic Acid/analogs & derivatives , Glutamates/pharmacology , Guanine/analogs & derivatives , Oligopeptides/metabolism , Quinazolines/pharmacology , Technetium/chemistry , Animals , Cell Line, Tumor , Female , Folic Acid/metabolism , Folic Acid/pharmacology , Guanine/pharmacology , Humans , Kidney/drug effects , Kidney/metabolism , Mice , Mice, Nude , Organotechnetium Compounds/chemistry , Pemetrexed , Xenograft Model Antitumor Assays
16.
Mol Pharm ; 6(5): 1518-25, 2009.
Article in English | MEDLINE | ID: mdl-19630399

ABSTRACT

Herein we report on the potencies of 4 related folate-conjugated tubulysins constructed with either tubulysin B hydrazide (EC0305), tubulysin A hydrazide (EC0510), the N,O-acetal derivative of natural tubulysins (EC0317) or a tubulysin B ester (EC0302). Our results confirmed that EC0305 is the most favorable conjugate of the group due to its potent antitumor activity [100% cures at 1 micromol/kg, three times a week (TIW) for 2 weeks] and its favorably low toxicity profile. In contrast, the natural tubulysin B drug proved to be inactive against a human nasopharyngeal tumor model when administered at doses near to or greater than the maximum tolerated dose (MTD). When tested against more chemoresistant folate receptor expressing M109 and 4T1-cl2 tumors, EC0305 displayed superior antitumor activity over a previously disclosed folate conjugate of desacetylvinblastine monohydrazide (EC145). These studies demonstrate that EC0305 has significant antiproliferative activity against FR expressing tumors, including those which are generally more chemoresistant, and that EC0305 should be considered for development as a candidate for the treatment of advanced FR-expressing human cancers.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Folic Acid/analogs & derivatives , Oligopeptides/chemistry , Oligopeptides/pharmacology , Animals , Antineoplastic Agents/toxicity , Blood Proteins/metabolism , Carrier Proteins/metabolism , Cattle , Cell Line, Tumor , Dogs , Female , Folate Receptors, GPI-Anchored , Folic Acid/chemistry , Folic Acid/pharmacology , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/toxicity , Protein Binding , Rabbits , Rats , Receptors, Cell Surface/metabolism , Structure-Activity Relationship
17.
Cancer Res ; 68(23): 9839-44, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047164

ABSTRACT

The membrane-bound high-affinity folate receptor (FR) is highly expressed on a wide range of primary and metastatic human cancers, such as those originating in ovary, lung, breast, endometrium, kidney, and brain. Because folate-linked conjugates bind to and become internalized within FR-expressing cells (similar to that of free folic acid), we explored the possibility of using the folate ligand to target a potent, semisynthetic analogue of the microtubule inhibitor tubulysin B to FR-enriched tumors. When tested in vitro, a novel folate conjugate, herein referred to as EC0305, was found to specifically inhibit the growth of a panel of FR-positive cell lines (IC50 range, 1-10 nmol/L) in a dose-dependent manner, whereas cells lacking FR expression were unaffected. The potency of EC0305 was also confirmed against a human KB xenograft-nu/nu mouse cancer model. Here, a brief three times per week, 2-week regimen yielded remarkable antitumor activity (100% tumor-free animals) without causing significant weight loss or major organ tissue degeneration. In contrast, antitumor activity was completely abolished in EC0305-treated animals that were co-dosed with an excess of a nontoxic folate-containing analogue, thereby confirming that the antitumor effect of this agent was mediated by FRs. The advantage provided by folate conjugation was further proved by the untargeted free drug, which was found to be completely inactive at both tolerable and highly toxic dose levels. Collectively, these results show that this potent antiproliferative tubulysin compound can be specifically delivered to FR-positive tumors to provide substantial therapeutic benefit using well-tolerable dosing regimens.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Oligopeptides/pharmacology , Receptors, Cell Surface/metabolism , Animals , Antineoplastic Agents/metabolism , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Ethylenediamines/pharmacology , Female , Fluorescein/pharmacology , Folate Receptors, GPI-Anchored , Folic Acid/metabolism , Folic Acid/pharmacology , Humans , KB Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/metabolism , Substrate Specificity , Xenograft Model Antitumor Assays
18.
J Pharmacol Exp Ther ; 327(3): 918-25, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18791065

ABSTRACT

Herein, we present a detailed analysis on the effects of feeding laboratory mice both high and low folic acid (folate)-containing diets as related to associated changes in serum and red blood cell (RBC) folate levels, tissue-derived folate receptor levels, and the ability of folate-drug conjugates to bind and effectuate activity against folate receptor (FR)-positive tumor xenografts. Our data show that serum and RBC folate concentrations sharply drop immediately after mice are switched to low folate diets; however, both parameters reach steady-state, "human-like" levels after 6 weeks. Interestingly, tissue-related folate binding capacities were also lowered during the dietary modulation period, whereas the net uptake of a radiolabeled folate conjugate was simultaneously increased 2.6- and 5-fold in FR-positive kidney and tumor tissue, respectively. Finally, the performances of several clinically and preclinically relevant folate-drug conjugates were evaluated against tumors in mice that were fed high or low folate diets. Except when administered at a dose level 6-fold less than that required to saturate endogenous FRs, no significant loss of antitumor activity was observed. From these findings, we conclude that lowering the dietary intake of folates in mice has little impact on the biological activity of repetitively dosed folate-targeted agents but that low folate diet regimens will reduce serum and RBC folate levels down to levels that more closely approximate the normal human ranges.


Subject(s)
Carrier Proteins/analysis , Folic Acid/administration & dosage , Neoplasms/drug therapy , Receptors, Cell Surface/analysis , Animals , Carrier Proteins/drug effects , Diet , Drug Delivery Systems , Folate Receptors, GPI-Anchored , Folic Acid/blood , Folic Acid/pharmacology , Humans , Mice , Receptors, Cell Surface/drug effects , Transplantation, Heterologous , Xenograft Model Antitumor Assays
19.
Mol Pharm ; 4(5): 659-67, 2007.
Article in English | MEDLINE | ID: mdl-17874843

ABSTRACT

We have designed a new type of tumor-targeted agent by tethering two different drug molecules, with distinct biological mechanisms of action, to the same ligand. This compound, named EC0225, represents the "first in class" multidrug, folate receptor (FR)-targeted agent to be disclosed. It was constructed with a single folate molecule, extended by a hydrophilic peptide-based spacer, which was in turn attached to mitomycin and Vinca alkaloid units via two separate disulfide-containing linkers. EC0225 produced potent, dose-responsive activity in vitro, and curative activity was observed against FR-positive syngeneic and xenograft tumors following the administration of well-tolerated dosing regimens. Multiple complete responses and cures were also noted when EC0225 was used to treat mice initially bearing tumors as large as 750 mm (3) in volume. Overall, EC0225's impressive preclinical activity allowed for its selection as a development candidate and for the start of Phase 1 clinical trials, which began in March of 2007, for the treatment of advanced malignancies.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical , Folic Acid/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents/toxicity , Cell Line, Tumor , Disease Models, Animal , Female , Folic Acid/chemistry , Folic Acid/pharmacology , Folic Acid/toxicity , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasms/pathology , Substrate Specificity , Xenograft Model Antitumor Assays
20.
Cancer Res ; 67(13): 6376-82, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17616697

ABSTRACT

EC131, a new folate receptor (FR)-targeted drug conjugate, was prepared by covalently attaching the vitamin folic acid (FA) to a potent microtubule-inhibiting agent, maytansinoid DM1, via an intramolecular disulfide bond. When tested on cells in culture, EC131 was found to retain high affinity for FR-positive cells and to provide FR-specific cytotoxicity with an IC(50) in the low nanomolar range. The activity of EC131 was completely blocked in the presence of an excess of free FA, and no activity was detected against FR-negative cells. When evaluated against s.c. FR-positive M109 tumors in BALB/c mice, EC131 showed marked antitumor efficacy. Furthermore, this therapeutic effect occurred in the apparent absence of weight loss or noticeable organ tissue degeneration. In contrast, no significant antitumor activity was observed in EC131-treated animals that were codosed with an excess of FA, thus demonstrating the targeted specificity of the in vivo activity. EC131 also showed marked antitumor activity against FR-positive human KB tumors, but not against FR-negative A549 tumors, in nude mice with no evidence of systemic toxicity during or after the therapy. In contrast, therapy with the free maytansinoid drug (in the form of DM1-S-Me) proved not to be effective against the KB model when administered at its maximum tolerated dose (MTD). Taken together, these results indicate that EC131 is a highly potent agent capable of producing therapeutic benefit in murine tumor models at sub-MTD levels.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Folic Acid/pharmacology , Maytansine/pharmacology , Plant Extracts/pharmacology , Receptors, Cell Surface/metabolism , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor , Folate Receptors, GPI-Anchored , Folic Acid/chemistry , Folic Acid/metabolism , Humans , KB Cells , Maximum Tolerated Dose , Maytansine/chemistry , Maytenus/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...