Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters











Publication year range
1.
Cell Rep Med ; 2(10): 100422, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34755134

ABSTRACT

Provoked by sterile/nonsterile insults, prolonged monocyte mobilization and uncontrolled monocyte/macrophage activation can pose imminent or impending harm to the affected organs. Curiously, folate receptor beta (FRß), with subnanomolar affinity for the vitamin folic acid (FA), is upregulated during immune activation in hematopoietic cells of the myeloid lineage. This phenomenon has inspired a strong interest in exploring FRß-directed diagnostics/therapeutics. Previously, we have reported that FA-targeted aminopterin (AMT) therapy can modulate macrophage function and effectively treat animal models of inflammation. Our current investigation of a lead compound (EC2319) leads to discovery of a highly FR-specific mechanism of action independent of the root causes against inflammatory monocytes. We further show that EC2319 suppresses interleukin-6/interleukin-1ß release by FRß+ monocytes in a triple co-culture leukemic model of cytokine release syndrome with anti-CD19 chimeric antigen receptor T cells. Because of its chemical stability and metabolically activated linker, EC2319 demonstrates favorable pharmacokinetic characteristics and cross-species translatability to support future pre-clinical and clinical development.


Subject(s)
Aminopterin/pharmacology , Cytokine Release Syndrome/prevention & control , Folate Receptor 2/genetics , Folic Acid Antagonists/pharmacology , Folic Acid/metabolism , Macrophages/drug effects , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , CHO Cells , Cricetulus , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Female , Folate Receptor 1/antagonists & inhibitors , Folate Receptor 1/genetics , Folate Receptor 1/immunology , Folate Receptor 2/antagonists & inhibitors , Folate Receptor 2/immunology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Models, Biological , Monocytes/drug effects , Monocytes/immunology , Monocytes/pathology , RAW 264.7 Cells , Rats , Rats, Inbred Lew , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
2.
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
3.
Bioorg Med Chem Lett ; 30(7): 126987, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32029324

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 and synthesized a diazepine-ring-opened bis-PBD prodrug (pro-PBD-PBD) folate conjugate lacking the one of the two imine moieties found in the corresponding free bis-PBD. Upon entering a targeted cell, cleavage of the linker system, including the hydrolysis of an oxazolidine moiety, results in the formation of a reactive intermediate which possesses a newly formed aldehyde as well as an aromatic amine. A fast and spontaneous 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, thereby delivering the bis-PBD to the targeted cell. The in vitro and in vivo activity of this conjugate has been evaluated on folate receptor positive KB cells. Sub-nanomolar activity with good specificity and high cure rates with minimal toxicity have been observed.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Benzodiazepines/therapeutic use , Folate Receptors, GPI-Anchored/metabolism , Neoplasms/drug therapy , Prodrugs/therapeutic use , Pyrroles/therapeutic use , Animals , Antibiotics, Antineoplastic/chemical synthesis , Antibiotics, Antineoplastic/pharmacology , Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacology , Drug Design , Female , HeLa Cells , Humans , Mice, Nude , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Xenograft Model Antitumor Assays
4.
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
5.
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
6.
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
7.
Oncotarget ; 9(97): 37042-37053, 2018 Dec 11.
Article in English | MEDLINE | ID: mdl-30651934

ABSTRACT

PURPOSE: The purpose was to determine the safety and antitumor activity of a folate-tubulysin conjugate (EC0531) in a relevant preclinical animal model, dogs with naturally-occurring invasive urothelial carcinoma (iUC). Canine iUC is an aggressive cancer with high folate receptor (FR) expression similar to that in certain forms of human cancer. EXPERIMENTAL DESIGN: A 3+3 dose escalation study of EC0531 (starting dose 0.2 mg/kg given intravenously at two-week intervals) was performed in dogs with iUC expressing high levels of FRs (>50% positive tumor cells). Pharmacokinetic (PK) analysis was performed, and the maximum tolerated dose (MTD) was determined. The dose cohort at the MTD was expanded to determine antitumor activity. RESULTS: The MTD of EC0531 was 0.26 mg/kg every two weeks, with grade 3-4 neutropenia and gastrointestinal toxicity observed at higher doses. Treatment at the MTD was well tolerated. Clinical benefit was found in 20 of 28 dogs (71%), including three dogs with partial remission and 17 dogs with stable disease. Plasma EC0531 concentrations in the dogs far exceeded those required to inhibit proliferation of FR-expressing cell in vitro. Unlike human neutrophils, canine neutrophils were found to express FRs, which contributes to the neutropenia at higher doses of EC0531 in dogs. CONCLUSION: EC0531 was well tolerated and had good antitumor activity in dogs with iUC. It is likely that humans will tolerate higher, potentially more effective doses of folate-tubulysin without myelotoxicity because of the absence of FRs on human neutrophils. The results clearly justify the evaluation of folate-tubulysin in human clinical trials.

8.
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
9.
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
10.
Mol Med ; 21: 584-96, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26181632

ABSTRACT

Folate receptor (FR)-ß has been identified as a promising target for antimacrophage and antiinflammatory therapies. In the present study, we investigated EC0565, a folic acid-derivative of everolimus, as a FR-specific inhibitor of the mammalian target of rapamycin (mTOR). Because of its amphiphilic nature, EC0565 was first evaluated for water solubility, critical micelle formation, stability in culture and FR-binding specificity. Using FR-expressing macrophages, the effect of EC0565 on mTOR signaling and cellular proliferation was studied. The pharmacokinetics, metabolism and bioavailability of EC0565 were studied in normal rats. The in vivo activity of EC0565 was assessed in rats with adjuvant arthritis, a "macrophage-rich" model with close resemblance to rheumatoid arthritis. EC0565 forms micellar aggregates in physiological buffers and demonstrates good water solubility as well as strong multivalent FR-binding capacity. EC0565 inhibited mTOR signaling in rat macrophages at nanomolar concentrations and induced G0/G1 cell cycle arrest in serum-starved RAW264.7 cells. Subcutaneously administered EC0565 in rats displayed good bioavailability and a relatively long half-life (~12 h). When given at 250 nmol/kg, EC0565 selectively inhibited proliferating cell nuclear antigen expression in thioglycollate-stimulated rat peritoneal cells. With limited dosing regimens, the antiarthritic activity of EC0565 was found superior to that of etanercept, everolimus and a nontargeted everolimus analog. The in vivo activity of EC0565 was also comparable to that of a folate-targeted aminopterin. Folate-targeted mTOR inhibition may be an effective way of suppressing activated macrophages in sites of inflammation, especially in nutrient-deprived conditions, such as in the arthritic joints. Further investigation and improvement upon the physical and biochemical properties of EC0565 are warranted.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Everolimus/analogs & derivatives , Everolimus/administration & dosage , Folic Acid/analogs & derivatives , Folic Acid/administration & dosage , Inflammation/drug therapy , TOR Serine-Threonine Kinases/genetics , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Arthritis, Experimental/drug therapy , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Cell Proliferation/drug effects , Everolimus/chemistry , Folate Receptor 2/genetics , Folate Receptor 2/metabolism , Folic Acid/chemistry , Humans , Inflammation/genetics , Inflammation/pathology , Rats , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors
11.
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
12.
Clin Immunol ; 150(1): 64-77, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24333534

ABSTRACT

EC0746 is a rationally designed anti-inflammatory drug conjugate consisting of a modified folic acid-based ligand linked to a γ-hydrazide analog of aminopterin. In this report, EC0746's effectiveness was evaluated against experimental retinal S-antigen (PDSAg) induced autoimmune uveitis (EAU) and myelin-basic-protein induced autoimmune encephalomyelitis (EAE). In both models, functional FR-ß was detected on activated macrophages in local (retinal or central-nervous-system, respectively) and systemic (peritoneal cavity) sites of inflammation. In myelin-rich regions of EAE rats, an increased uptake of (99m)Tc-EC20 (etarfolatide; a FR-specific radioimaging agent) was also observed. EC0746 treatment at disease onset suppressed the clinical severity of both EAU and EAE, and it strongly attenuated progressive histopathological changes in the affected organs. In all parameters assessed, EC0746 activity was completely blocked by a benign folate competitor, suggesting that these therapeutic outcomes were specifically FR-ß mediated. EC0746 may emerge as a useful macrophage-modulating agent for treating inflammatory episodes of organ-specific autoimmunity.


Subject(s)
Aminopterin/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Folic Acid Antagonists/therapeutic use , Uveitis/drug therapy , Aminopterin/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Brain/immunology , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Folate Receptor 2/immunology , Folic Acid Antagonists/pharmacology , Macrophages/drug effects , Macrophages/immunology , Rats , Rats, Inbred Lew , Spinal Cord/drug effects , Spinal Cord/immunology , Uveitis/immunology , Uveitis/pathology
13.
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
14.
J Am Soc Nephrol ; 23(10): 1674-81, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22859856

ABSTRACT

Activation of the mammalian target of rapamycin (mTOR) signaling pathway is aberrant in autosomal-dominant polycystic kidney disease (ADPKD). The mTOR inhibitors, such as rapamycin, ameliorate PKD in rodent models, but clinical trials have not shown benefit, possibly as a result of low tissue concentrations of rapamycin at clinically tolerable doses. To overcome this limitation, we synthesized a folate-conjugated form of rapamycin (FC-rapa) that is taken up by folate receptor-mediated endocytosis and cleaved intracellularly to reconstitute the active drug. We found that renal cyst-lining cells highly express the folate receptor in ADPKD and mouse models. In vitro, FC-rapa inhibited mTOR activity in a dose- and folate receptor-dependent manner. Treatment of a PKD mouse model with FC-rapa inhibited mTOR in the target tissue, strongly attenuated proliferation and growth of renal cysts and preserved renal function. Furthermore, FC-rapa inhibited mTOR activity in the kidney but not in other organs. In summary, these results suggest that targeting the kidney using FC-rapa may overcome the significant side effects and lack of renal efficacy observed in clinical trials with mTOR inhibitors in ADPKD.


Subject(s)
Polycystic Kidney, Autosomal Dominant/drug therapy , Sirolimus/therapeutic use , Animals , Cell Line , Disease Models, Animal , Endocytosis , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/analogs & derivatives , Folic Acid/therapeutic use , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Mice , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Signal Transduction/drug effects , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/metabolism
15.
Bioconjug Chem ; 23(7): 1357-69, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22667324

ABSTRACT

The folate receptor (FR) is a potentially useful biological target for the management of many human cancers. This membrane protein binds extracellular folates with very high affinity and, through an endocytic process, physically delivers them inside the cell for biological consumption. There are now many examples of how this physiological system can be exploited for the targeted delivery of biologically active molecules to cancer. In fact, strong preclinical as well as emerging clinical evidence exists showing how FR-positive cancers can be (i) anatomically identified using folate conjugates of radiodiagnostic imaging agents and (ii) effectively treated with companion folate-targeted chemotherapies. While the biological results are compelling, it is of equal importance to understand the conjugation chemistries that were developed to produce these active molecules. Therefore, this review will focus on the methods utilized to construct folate-based small-molecule drug conjugates (SMDCs), with particular attention focused on modular design, hydrophilic spacers, and self-immolative linkers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Delivery Systems , Folic Acid/chemistry , Neoplasms/drug therapy , Protein Engineering , Animals , Antineoplastic Combined Chemotherapy Protocols/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/metabolism , Folic Acid/pharmacokinetics , Humans , Models, Biological , Neoplasms/metabolism
16.
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
17.
Nucl Med Biol ; 38(5): 715-23, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21718947

ABSTRACT

INTRODUCTION: Use of folic acid radioconjugates for folate receptor (FR) targeting is a promising strategy for imaging purposes as well as for potential therapy of cancer and inflammatory diseases due to the frequent FR overexpression found on cancer cells and activated macrophages. Herein, we report on preclinical results using a novel DOTA-Bz-EDA-folate conjugate radiolabeled with [(67)Ga]-gallium. METHODS: DOTA-Bz-EDA-folate was prepared by conjugation of ethylenediamine-(γ)-folate with 2-(p-isothiocyanobenzyl)-DOTA. Radiolabeling was carried out with (67)GaCl(3) according to standard procedures. Biodistribution studies of the tracer were performed in mice bearing FR-positive KB tumor xenografts. The effects on radiofolate biodistribution with coadministered renal uptake-blocking amino acids, diuretic agents, antifolates as well as different routes of administration were likewise investigated. Supportive imaging studies were performed using a small-animal single photon emission computed tomography (SPECT)/CT scanner. RESULTS: (67)Ga-DOTA-Bz-EDA-folate showed a high and specific accumulation in tumors (6.30%±0.75% ID/g, 1 h pi and 6.08%±0.89% ID/g, 4 h pi). Nonspecific radioactivity uptake in nontargeted tissues was negligible, but significant accumulation was found in FR-positive kidneys, which resulted in unfavorably low tumor-to-kidney ratios (<0.1). Coadministered amino acids or diuretics did not effectively reduce renal accumulation; in contrast, predosed pemetrexed did significantly reduce kidney uptake (<29% of control values). The SPECT/CT studies confirmed the excellent tumor-to-background contrast of (67)Ga-radiofolate and the favorable reduction in kidney uptake (with improved imaging quality) resulting from pemetrexed administration. CONCLUSION: Conventional methods to reduce kidney uptake of radiofolates fail. However, the novel (67)Ga-radiolabeled DOTA-Bz-EDA-folate can effectively be used to image FR-positive cancer and potentially inflammatory diseases. Due to its rapid blood clearance properties, this tracer is also a promising candidate for positron emission tomography imaging if radiolabeled with the short-lived [(68)Ga]-gallium radionuclide.


Subject(s)
Folic Acid/chemistry , Folic Acid/pharmacokinetics , Heterocyclic Compounds/chemistry , Isothiocyanates/chemistry , Uterine Cervical Neoplasms/metabolism , Animals , Biological Transport/drug effects , Female , Folic Acid/metabolism , Folic Acid Antagonists/administration & dosage , Folic Acid Antagonists/pharmacology , Gallium Radioisotopes , Glutamates/administration & dosage , Glutamates/pharmacology , Guanine/administration & dosage , Guanine/analogs & derivatives , Guanine/pharmacology , Half-Life , Humans , Hydrogen-Ion Concentration , Injections , KB Cells , Kidney/drug effects , Kidney/metabolism , Mice , Pemetrexed , Radioactive Tracers , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Urine/chemistry , Uterine Cervical Neoplasms/diagnostic imaging , Uterine Cervical Neoplasms/urine
18.
Arthritis Res Ther ; 13(2): R56, 2011 Apr 04.
Article in English | MEDLINE | ID: mdl-21463515

ABSTRACT

INTRODUCTION: Folate receptor (FR)-expressing macrophages have been shown to accumulate at sites of inflammation, where they promote development of inflammatory symptoms. To target such a macrophage population, we designed and evaluated the biologic activity of EC0746, a novel folic acid conjugate of the highly potent antifolate, aminopterin. METHODS: Using a FR-positive subclone of murine macrophage-derived RAW264.7 cells and rat thioglycollate-elicited macrophages, we studied the effect of EC0746 on dihydrofolate reductase activity, cell proliferation, and cellular response towards bacterial lipopolysaccharide as well as IFNγ activation. The EC0746 anti-inflammatory activity, pharmacokinetics, and toxicity were also evaluated in normal rats or in rats with adjuvant-induced arthritis; that is, a FR-positive macrophage model that closely resembles rheumatoid arthritis in humans. RESULTS: EC0746 suppresses the proliferation of RAW264.7 cells and prevents the ability of nonproliferating rat macrophages to respond to inflammatory stimuli. In the macrophage-rich rat arthritis model, brief treatment with subcutaneously administered EC0746 is shown to mediate an FR-specific anti-inflammatory response that is more potent than either orally administered methotrexate or subcutaneously delivered etanercept. More importantly, EC0746 therapy is also shown to be ~40-fold less toxic than unmodified aminopterin, with fewer bone marrow and gastrointestinal problems. CONCLUSIONS: EC0746 is the first high FR-binding dihydrofolate reductase inhibitor that demonstrates FR-specific anti-inflammatory activities both in vitro and in vivo. Our data reveal that a relatively toxic anti-inflammatory drug, such as aminopterin, can be targeted with folic acid to inflammatory macrophages and thereby relieve inflammatory symptoms with greatly reduced toxicity.


Subject(s)
Aminopterin/analogs & derivatives , Aminopterin/pharmacology , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Folic Acid Antagonists/pharmacology , Folic Acid/analogs & derivatives , Folic Acid/pharmacology , Aminopterin/chemical synthesis , Aminopterin/pharmacokinetics , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacokinetics , Female , Folate Receptors, GPI-Anchored/drug effects , Folic Acid/chemical synthesis , Folic Acid/pharmacokinetics , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/pharmacokinetics , Macrophages/drug effects , Mice , Rats , Rats, Inbred Lew
19.
Bioorg Med Chem Lett ; 21(4): 1202-5, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21236665

ABSTRACT

Efficient syntheses of folate receptor (FR) targeting conjugates of the anti-inflammatory, aminopterin hydrazide, are described. 2-{4-Benzoylamino}-5-oxo-5-{N'-[2-(pyridin-2-yldisulfanyl)-ethoxycarbonyl]-hydrazino}-pentanoic acid is synthesized from commercially available 4-[(2-amino-4-imino-3,4-dihydro-pteridin-6-yl-methyl)-amino]-benzoic acid. Conjugation of this novel, activated aminopterin hydrazide to folic acid through cysteine-terminating (C-terminus), peptide/carbohydrate spacers results in highly water soluble conjugates which allow for the release of free aminopterin hydrazide within the endosomes of targeted cells.


Subject(s)
Aminopterin/chemistry , Anti-Inflammatory Agents/chemistry , Folic Acid/analogs & derivatives , Aminopterin/chemical synthesis , Aminopterin/therapeutic use , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/therapeutic use , Drug Design , Folic Acid/chemical synthesis , Folic Acid/chemistry , Folic Acid/therapeutic use , Humans , Inflammation/drug therapy , Stereoisomerism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL