Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Imaging Biol ; 22(2): 377-383, 2020 04.
Article in English | MEDLINE | ID: mdl-31292915

ABSTRACT

PURPOSE: Neuroendocrine tumors (NETs) have reasonably high 5-year survival rates when diagnosed at an early stage but are significantly more lethal when discovered only after metastasis. Although several imaging modalities such as computed tomography (CT), positron emission tomography, and magnetic resonance imaging can detect neuroendocrine tumors, their high false positive rates suggest that more specific diagnostic tests are required. Targeted imaging agents such as Octreoscan® have met some of this need for improved specificity, but their inability to image poorly differentiated NETs suggests that improved NET imaging agents are still needed. Because neurokinin 1 receptors (NK1Rs) are widely over-expressed in neuroendocrine tumors, but show limited expression in healthy tissues, we have undertaken to develop an NK1R-targeted imaging agent for improved diagnosis and staging of neuroendocrine tumors. PROCEDURE: A small molecule NK1R antagonist was conjugated via a flexible spacer to a Tc-99m chelating peptide. After complexation with Tc-99m, binding of the conjugate to human embryonic kidney (HEK293) cells transfected with the human NK1R was evaluated as a function of radioimaging agent concentration. In vivo imaging of HEK293-NK1R tumor xenografts in mice was also performed by single-photon emission computed tomography/computed tomography (γ-SPECT/CT), and the distribution of the conjugate in various tissues was quantified by tissue resection and γ-counting. RESULTS: NK1R-targeted Tc-99m-based radioimaging agent displayed excellent affinity (Kd = 16.8 nM) and specificity for HEK293-NK1R tumor xenograft. SPECT/CT analysis of tumor-bearing mice demonstrated significant tumor uptake and high tumor to background ratio as early as 2 h post injection. CONCLUSION: The excellent tumor contrast afforded by our NK1R-targeted radioimaging agent exhibits properties that could improve early diagnosis and staging of many neuroendocrine tumors.


Subject(s)
Neuroendocrine Tumors/diagnostic imaging , Receptors, Neurokinin-1/chemistry , Technetium/chemistry , Animals , Chelating Agents/chemistry , False Positive Reactions , Female , HEK293 Cells , Humans , Ligands , Mice , Mice, Nude , Neoplasm Transplantation , Peptides/chemistry , Single Photon Emission Computed Tomography Computed Tomography , Somatostatin/analogs & derivatives , Tomography, X-Ray Computed
2.
Cancer Res ; 79(2): 387-396, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30482775

ABSTRACT

Most solid tumors are comprised of multiple clones that express orthogonal antigens, suggesting that novel strategies must be developed in order to adapt chimeric antigen receptor (CAR) T-cell therapies to treat heterogeneous solid tumors. Here, we utilized a cocktail of low-molecular-weight bispecific adapters, each comprised of fluorescein linked to a different tumor-specific ligand, to bridge between an antifluorescein CAR on the engineered T cell and a unique antigen on the cancer cell. This formation of an immunologic synapse between the CAR T cell and cancer cell enabled use of a single antifluorescein CAR T cell to eradicate a diversity of antigenically different solid tumors implanted concurrently in NSG mice. Based on these data, we suggest that a carefully designed cocktail of bispecific adapters in combination with antifluorescein CAR T cells can overcome tumor antigen escape mechanisms that lead to disease recurrence following many CAR T-cell therapies. SIGNIFICANCE: A cocktail of tumor-targeted bispecific adapters greatly augments CAR T-cell therapies against heterogeneous tumors, highlighting its potential for broader applicability against cancers where standard CAR T-cell therapy has failed.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/therapy , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Animals , Antigens, Neoplasm/immunology , Cell Engineering/methods , Cell Line, Tumor , Epitopes , Female , HEK293 Cells , Humans , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Xenograft Model Antitumor Assays
3.
Bioconjug Chem ; 29(4): 1047-1059, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29446616

ABSTRACT

Although peptides, antibodies/antibody fragments, siRNAs, antisense DNAs, enzymes, and aptamers are all under development as possible therapeutic agents, the breadth of their applications has been severely compromised by their inability to reach intracellular targets. Thus, while macromolecules can often enter cells by receptor-mediated endocytosis, their missions frequently fail due to an inability to escape their entrapping endosomes. In this paper, we describe a general method for promoting release of any biologic material from any entrapping endosome. The strategy relies on the fact that all nascent endosomes contain extracellular (Na+-enriched) medium, but are surrounded by intracellular (K+-enriched) fluid in the cytoplasm. Osmotic swelling and rupture of endosomes will therefore be facilitated if the flow of K+ down its concentration gradient from the cytosol into the endosome can be facilitated without allowing downhill flow of Na+ from the endosome into the cytosol. While any K+ selective ionophore can promote the K+ specific influx, the ideal K+ ionophore will also exchange influxed K+ for an osmotically inactive proton (H+) in order to prevent buildup of an electrical potential that would rapidly halt K+ influx. The only ionophore that catalyzes this exchange of K+ for H+ efficiently is nigericin. We demonstrate here that ligand-targeted delivery of nigericin into endosomes that contain an otherwise impermeable fluorescent dye can augment release of the dye into the cell cytosol via swelling/bursting of the entrapping endosomes. We further show that nigericin-facilitated escape of a folate-targeted luciferase siRNA conjugate from its entrapping endosomes promotes rapid suppression of the intended luciferase reporter gene. Taken together, we propose that ionophore-catalyzed entry of K+ into endosomal compartments can promote the release of otherwise impermeable contents from their encapsulating endosomes.


Subject(s)
Endosomes/drug effects , Hydrogen/metabolism , Ionophores/pharmacology , Nigericin/pharmacology , Potassium/metabolism , Animals , Cell Line, Tumor , Cytosol/metabolism , Endocytosis , Endosomes/metabolism , Fluorescent Dyes/metabolism , Humans , Mice , Osmosis , RAW 264.7 Cells , RNA, Small Interfering/metabolism , Sodium/metabolism
4.
Bioconjug Chem ; 29(4): 1319-1326, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29466853

ABSTRACT

Neurokinin 1 receptor (NK1R) is expressed in gliomas and neuroendocrine malignancies and represents a promising target for molecular imaging and targeted radionuclide therapy. The goal of this study was to synthesize and evaluate a novel NK1R ligand (NK1R-NOTA) for targeting NK1R-expressing tumors. Using a carboxymethyl moiety linked to L-733060 as a starting reagent, NK1R-NOTA was synthesized in a three-step reaction and then labeled with 64Cu (or 67Ga for in vitro studies) in the presence of CH3COONH4 buffer. The radioligand affinity and cellular uptake were evaluated with NK1R-transduced HEK293 cells (HEK293-NK1R) and NK1R nontransduced HEK293 cells (HEK293-WT) and their xenografts. Radiolabeled NK1R-NOTA was obtained with a radiochemical purity of >95% and specific activities of >7.0 GBq/µmol for 64Cu and >5.0 GBq/µmol for 67Ga. Both 64Cu- and 67Ga-labeled NK1R-NOTA demonstrated high levels of uptake in HEK293-NK1R cells, whereas co-incubation with an excess of NK1R ligand L-733060 reduced the level of uptake by 90%. Positron emission tomography (PET) imaging showed that [64Cu]NK1R-NOTA had a accumulated rapidly in HEK293-NK1R xenografts and a 10-fold lower level of uptake in HEK293-WT xenografts. Radioactivity was cleared by gastrointestinal tract and urinary systems. Biodistribution studies confirmed that the tumor-to-organ ratios were ≥5 for all studied organs at 1 h p.i., except kidneys, liver, and intestine, and that the tumor-to-intestine and tumor-to-kidney ratios were also improved 4 and 20 h post-injection. [64Cu]NK1R-NOTA is a promising ligand for PET imaging of NK1R-expressing tumor xenografts. Delayed imaging with [64Cu]NK1R-NOTA improves image contrast because of the continuous clearance of radioactivity from normal organs.


Subject(s)
Copper Radioisotopes/chemistry , Gallium Radioisotopes/chemistry , Heterocyclic Compounds/chemistry , Neoplasms/diagnostic imaging , Neurokinin-1 Receptor Antagonists/chemistry , Receptors, Neurokinin-1/analysis , Animals , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds, 1-Ring , Male , Mice, Nude , Neurokinin-1 Receptor Antagonists/chemical synthesis , Positron-Emission Tomography/methods
5.
Mol Pharm ; 14(11): 3859-3865, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28969417

ABSTRACT

The neurokinin-1 receptor (NK1R) plays a significant role in the progression and metastasis of several neuroendocrine tumors. Due to its upregulation in these cancers, NK1R constitutes an attractive receptor for development of ligand-targeted imaging and therapeutic agents. In this report, we present the design and synthesis of an NK1R targeting ligand conjugated to the chemotherapeutic agent, tubulysin B hydrazide (TubBH), via a self-immolative linker. We then explore the ability of this low molecular weight tubulysin conjugate to kill NK1R overexpressing cancer cells both in vitro and in vivo without killing receptor negative healthy cells. Because similar studies in mice bearing NK1-negative tumors reveal no therapeutic impact, we conclude that our NK1R targeting ligand is specific for NK1R-expressing cells. Taken together, the data suggest a possible new approach for the treatment of NK1R-positive neuroendocrine cancers.


Subject(s)
Neuroendocrine Tumors/metabolism , Receptors, Neurokinin-1/metabolism , Animals , HEK293 Cells , Humans , Mice , Molecular Structure , Neuroendocrine Tumors/genetics , Oligopeptides/chemistry , Pipecolic Acids/chemistry , Receptors, Neurokinin-1/genetics , Substance P/metabolism
6.
Int J Oncol ; 50(5): 1482-1490, 2017 May.
Article in English | MEDLINE | ID: mdl-28393217

ABSTRACT

Metastatic melanoma is the most aggressive of all skin cancers and is associated with poor prognosis owing to lack of effective treatments. 25-epi Ritterostatin GN1N is a novel antitumor agent with yet undefined mechanisms of action. We sought to delineate the antitumor mechanisms of 25-epi Ritterostatin GN1N in melanoma cells to determine the potential of this compound as a treatment for melanoma. Activation of the endoplasmic reticulum (ER) stress protein glucose-regulated protein 78 (GRP78) has been associated with increased melanoma progression, oncogenic signaling, drug resistance, and suppression of cell death. We found that 25-epi Ritterostatin GN1N induced cell death in melanoma cells at nanomolar concentrations, and this cell death was characterized by inhibition of GRP78 expression, increased expression of the ER stress marker CHOP, loss of mitochondrial membrane potential, and lipidation of the autophagy marker protein LC3B. Importantly, normal melanocytes exhibited limited sensitivity to 25-epi Ritterostatin GN1N. Subsequent in vivo results demonstrated that 25-epi Ritterostatin GN1N reduced melanoma growth in mouse tumor xenografts and did not affect body weight, suggesting minimal toxicity. In summary, our findings indicate that 25-epi Ritterostatin GN1N causes ER stress and massive autophagy, leading to collapse of mitochondrial membrane potential and cell death in melanoma cells, with minimal effects in normal melanocytes. Thus, 25-epi Ritterostatin GN1N is a promising anticancer agent that warrants further investigation.


Subject(s)
Heat-Shock Proteins/genetics , Melanoma/drug therapy , Microtubule-Associated Proteins/biosynthesis , Spiro Compounds/administration & dosage , Steroids/administration & dosage , Transcription Factor CHOP/biosynthesis , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/biosynthesis , Humans , Melanoma/genetics , Melanoma/pathology , Mice , Microtubule-Associated Proteins/genetics , Transcription Factor CHOP/genetics , Xenograft Model Antitumor Assays
7.
Bioconjug Chem ; 27(9): 2157-65, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27529726

ABSTRACT

The neurokinin-1 receptor (NK1R) is implicated in the growth and metastasis of many tumors, including cancers of the brain (e.g., gliomas, glioblastomas, and astrocytomas), skin (e.g., melanomas), and neuroendocrine tissues (cancers of the breast, stomach, pancreas, larynx, and colon). Because overexpression of NK1R has been reported in most of these malignancies, we have undertaken designing an NK1R-targeted near-infrared (NIR) fluorescent dye for fluorescence-guided surgeries of these cancers. We demonstrate here that an NK1R-binding ligand linked to the NIR dye LS288 selectively accumulates in NK1R-expressing tumor xenografts with high affinity (Kd = 13 nM), allowing intraoperative imaging of these cancers in live mice. Because tumor accumulation is nearly quantitatively blocked by excess unlabeled ligand, and because NK1R-negative tumors and normal tissues display virtually no uptake, we conclude that the observed tumor retention is NK1R-mediated. Results on the synthesis, in vitro characterization, and animal testing of NK1R-targeted NIR dye are presented.


Subject(s)
Carcinoma, Neuroendocrine/surgery , Drug Design , Fluorescent Dyes/chemical synthesis , Infrared Rays , Optical Imaging , Receptors, Neurokinin-1/metabolism , Surgery, Computer-Assisted , Animals , Carcinoma, Neuroendocrine/diagnostic imaging , Carcinoma, Neuroendocrine/metabolism , Chemistry Techniques, Synthetic , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , HEK293 Cells , Humans , KB Cells , Male , Mice , Rhodamines/chemistry
8.
Biomacromolecules ; 17(3): 922-7, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26815158

ABSTRACT

Polymer-ligand conjugates are designed to bind proteins for applications as drugs, imaging agents, and transport scaffolds. In this work, we demonstrate a folic acid (FA)-triggered exosite binding of a generation five poly(amidoamine) (G5 PAMAM) dendrimer scaffold to bovine folate binding protein (bFBP). The protein exosite is a secondary binding site on the protein surface, separate from the FA binding pocket, to which the dendrimer binds. Exosite binding is required to achieve the greatly enhanced binding constants and protein structural change observed in this study. The G5Ac-COG-FA1.0 conjugate bound tightly to bFBP, was not displaced by a 28-fold excess of FA, and quenched roughly 80% of the initial fluorescence. Two-step binding kinetics were measured using the intrinsic fluorescence of the FBP tryptophan residues to give a KD in the low nanomolar range for formation of the initial G5Ac-COG-FA1.0/FBP* complex, and a slow conversion to the tight complex formed between the dendrimer and the FBP exosite. The extent of quenching was sensitive to the choice of FA-dendrimer linker chemistry. Direct amide conjugation of FA to G5-PAMAM resulted in roughly 50% fluorescence quenching of the FBP. The G5Ac-COG-FA, which has a longer linker containing a 1,2,3-triazole ring, exhibited an ∼80% fluorescence quenching. The binding of the G5Ac-COG-FA1.0 conjugate was compared to poly(ethylene glycol) (PEG) conjugates of FA (PEGn-FA). PEG2k-FA had a binding strength similar to that of FA, whereas other PEG conjugates with higher molecular weight showed weaker binding. However, no PEG conjugates gave an increased degree of total fluorescence quenching.


Subject(s)
Dendrimers/chemistry , Folate Receptors, GPI-Anchored/chemistry , Folic Acid/chemistry , Animals , Binding Sites , Cattle , Dendrimers/pharmacology , Drug Synergism , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/pharmacology , Polyethylene Glycols/chemistry , Protein Binding
9.
J Med Chem ; 58(7): 3094-103, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25822623

ABSTRACT

Prostate-specific membrane antigen (PSMA) is overexpressed in most prostate cancer cells while being present at low or undetectable levels in normal cells. This difference provides an opportunity to selectively deliver cytotoxic drugs to prostate cancer cells while sparing normal cells that lack PSMA, thus improving potencies and reducing toxicities. PSMA has high affinity for 2-[3-(1,3-dicarboxypropyl)ureido]pentanedioic acid (DUPA) (Ki = 8 nM). After binding to a DUPA-drug conjugate, PSMA internalizes, unloads the conjugate, and returns to the surface. In the present studies, an indenoisoquinoline topoisomerase I inhibitor was conjugated to DUPA via a peptide linker and a drug-release segment that facilitates intracellular cleavage to liberate the drug cargo. The DUPA-indenoisoquinoline conjugate exhibited an IC50 in the low nanomolar range in 22RV1 cell cultures and induced a complete cessation of tumor growth with no toxicity, as determined by loss of body weight and death of treated mice.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Glutarates/chemistry , Isoquinolines/chemistry , Kallikreins/metabolism , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacology , Urea/analogs & derivatives , Animals , Antineoplastic Agents/chemical synthesis , Chemistry Techniques, Synthetic , Drug Design , Humans , Inhibitory Concentration 50 , Male , Mice, Nude , Molecular Targeted Therapy , Prostatic Neoplasms/metabolism , Urea/chemistry , Xenograft Model Antitumor Assays
10.
Mol Pharm ; 11(5): 1696-706, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24725205

ABSTRACT

Multivalent conjugation of folic acid has been employed to target cells overexpressing folate receptors. Such polymer conjugates have been previously demonstrated to have high avidity to folate binding protein. However, the lack of a monovalent folic acid-polymer material has prevented a full binding analysis of these conjugates, as multivalent binding mechanisms and polymer-mass mechanisms are convoluted in samples with broad distributions of folic acid-to-dendrimer ratios. In this work, the synthesis of a monovalent folic acid-dendrimer conjugate allowed the elucidation of the mechanism for increased binding between the folic acid-polymer conjugate and a folate binding protein surface. The increased avidity is due to a folate-keyed interaction between the dendrimer and protein surfaces that fits into the general framework of slow-onset, tight-binding mechanisms of ligand/protein interactions.


Subject(s)
Dendrimers/chemistry , Folic Acid/chemistry , Carrier Proteins , Models, Theoretical , Protein Binding
11.
J Org Chem ; 78(18): 9085-92, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23899273

ABSTRACT

The convergent synthesis of 25-epi ritterostatin GN1N is described for the first time, starting from hecogenin acetate (HA). Stereoselective dihydroxylation employing the chiral ligand (DHQ)2PHAL was used as the key step to introduce the C25 epi-stereocenter on the north 1 segment. The title compound was obtained through a coupling reaction between the C3-keto-azide (cstat North 1) and North G.


Subject(s)
Spiro Compounds/chemical synthesis , Steroids/chemical synthesis , Crystallography, X-Ray , Ligands , Models, Molecular , Molecular Conformation , Spiro Compounds/chemistry , Steroids/chemistry
12.
Bioorg Med Chem Lett ; 22(12): 3978-82, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22608391

ABSTRACT

A multi-disciplinary approach was used to identify the first pharmacophore model for KCC2 blockers: several physico-chemical studies such as XRD and NMR were combined to molecular modelling techniques, SAR analysis and synthesis of constrained analogues in order to determine a minimal conformational space regrouping few potential bioactive conformations. These conformations were further compared to the conformational space of a different series of KCC2 blockers in order to identify the common pharmacophoric features. The synthesis of more potent analogues in this second series confirmed the usefulness of this KCC2 blocker pharmacophore model.


Subject(s)
Anticonvulsants/chemical synthesis , Potassium Channel Blockers/chemical synthesis , Proline/analogs & derivatives , Proline/chemical synthesis , Symporters/antagonists & inhibitors , Animals , Anticonvulsants/pharmacology , Cell Line, Tumor , Furosemide/pharmacology , High-Throughput Screening Assays , Humans , Ion Transport/drug effects , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Potassium Channel Blockers/pharmacology , Proline/pharmacology , Rats , Structure-Activity Relationship , Symporters/metabolism , X-Ray Diffraction , K Cl- Cotransporters
13.
Bioorg Med Chem Lett ; 20(8): 2542-5, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20299216

ABSTRACT

The discovery and optimization of a novel class of selective submicromolar KCC2 blockers is described. Details of synthesis and SAR are given together with ADME properties of selected compounds. A methylsulfone residue on the R(1) phenyl group improved the overall general profile of these prolinate derivatives.


Subject(s)
Proline/analogs & derivatives , Symporters/antagonists & inhibitors , Animals , Proline/chemistry , Proline/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship , K Cl- Cotransporters
SELECTION OF CITATIONS
SEARCH DETAIL
...