Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(22): 15443-15456, 2022 11 24.
Article in English | MEDLINE | ID: mdl-36331527

ABSTRACT

Overexpression of the receptor tyrosine kinase EphA2 is invariably associated with poor prognosis and development of aggressive metastatic cancers. Guided by our recently solved X-ray structure of the complex between an agonistic peptide and EphA2-LBD, we report on a novel agent, targefrin, that binds to EphA2-LBD with a 21 nM dissociation constant by isothermal titration calorimetry and presents an IC50 value of 10.8 nM in a biochemical assay. In cell-based assays, a dimeric version of the agent is as effective as the natural dimeric ligands (ephrinA1-Fc) in inducing cellular receptor internalization and degradation in several pancreatic cancer cell lines. When conjugated with chemotherapy, the agents can effectively deliver paclitaxel to pancreatic cancers in a mouse xenograft study. Given the pivotal role of EphA2 in tumor progression, we are confident that the agents reported could be further developed into innovative EphA2-targeting therapeutics.


Subject(s)
Peptides , Receptor, EphA2 , Animals , Humans , Mice , Cell Line , Ligands , Peptides/pharmacology , Receptor Protein-Tyrosine Kinases , Receptor, EphA2/drug effects , Receptor, EphA2/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
2.
J Med Chem ; 64(21): 16147-16158, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34705456

ABSTRACT

We have recently reported on Lys-covalent agents that, based on aryl-sulfonyl fluorides, were designed to target binding site Lys 311 in the X-linked inhibitor of apoptosis protein (XIAP). Similar to XIAP, melanoma-IAP (ML-IAP), a less well-characterized IAP family protein, also presents a lysine residue (Lys 135), which is in a position equivalent to that of Lys 311 of XIAP. On the contrary, two other members of the IAP family, namely, cellular-IAPs (cIAP1 and cIAP2), present a glutamic acid residue in that position. Hence, in the present work, we describe the derivation and characterization of the very first potent ML-IAP Lys-covalent inhibitor with cellular activity. The agent can be used as a pharmacological tool to further validate ML-IAP as a drug target and eventually for the development of ML-IAP-targeted therapeutics.


Subject(s)
Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Lysine/chemistry , Melanoma/pathology , Cell Line, Tumor , Humans , Inhibitor of Apoptosis Proteins/chemistry
3.
Molecules ; 26(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204178

ABSTRACT

We recently reported on a potent synthetic agent, 135H11, that selectively targets the receptor tyrosine kinase, EphA2. While 135H11 possesses a relatively high binding affinity for the ligand-binding domain of EphA2 (Kd~130 nM), receptor activation in the cell required the synthesis of dimeric versions of such agent (namely 135H12). This was expected given that the natural ephrin ligands also need to be dimerized or clustered to elicit agonistic activity in cell. In the present report we investigated whether the agonistic activity of 135H11 could be enhanced by biotin conjugation followed by complex formation with streptavidin. Therefore, we measured the agonistic EphA2 activity of 135H11-biotin (147B5) at various agent/streptavidin ratios, side by side with 135H12, and a scrambled version of 147B5 in pancreatic- and breast-cancer cell lines. The (147B5)n-streptavidin complexes (when n = 2, 3, 4, but not when n = 1) induced a strong receptor degradation effect in both cell lines compared to 135H12 or the (scrambled-147B5)4-streptavidin complex as a control, indicating that multimerization of the targeting agent resulted in an increased ability to cause receptor clustering and internalization. Subsequently, we prepared an Alexa-Fluor-streptavidin conjugate to demonstrate that (147B5)4-AF-streptavidin, but not the scrambled equivalent complex, concentrates in pancreatic and breast cancers in orthotopic nude-mouse models. Hence, we conclude that these novel targeting agents, with proper derivatization with imaging reagents or chemotherapy, can be used as diagnostics, and/or to deliver chemotherapy selectively to EphA2-expressing tumors.


Subject(s)
Receptor, EphA2/agonists , Receptor, EphA2/chemistry , Animals , Binding Sites/physiology , Biotin/chemistry , Biotin/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Ligands , Mice , Pancreatic Neoplasms/metabolism , Protein Binding/physiology , Receptor, EphA2/metabolism , Streptavidin/chemistry , Streptavidin/metabolism
4.
J Med Chem ; 64(8): 4903-4912, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33797903

ABSTRACT

Modulating disease-relevant protein-protein interactions (PPIs) using pharmacological tools is a critical step toward the design of novel therapeutic strategies. Over the years, however, targeting PPIs has proven a very challenging task owing to the large interfacial areas. Our recent efforts identified possible novel routes for the design of potent and selective inhibitors of PPIs using a structure-based design of covalent inhibitors targeting Lys residues. In this present study, we report on the design, synthesis, and characterizations of the first Lys-covalent BH3 peptide that has a remarkable affinity and selectivity for hMcl-1 over the closely related hBfl-1 protein. Our structural studies, aided by X-ray crystallography, provide atomic-level details of the inhibitor interactions that can be used to further translate these discoveries into novel generation, Lys-covalent pro-apoptotic agents.


Subject(s)
Drug Design , Lysine/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Peptide Fragments/chemistry , Proto-Oncogene Proteins/chemistry , A549 Cells , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Kinetics , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/metabolism , Molecular Dynamics Simulation , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Protein Binding , Proto-Oncogene Proteins/chemical synthesis , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation/drug effects
5.
Cancers (Basel) ; 12(10)2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33023262

ABSTRACT

The EphA2 tyrosine kinase receptor is highly expressed in several types of solid tumors. In our recent studies, we targeted EphA2 in pancreatic cancer with agonistic agents and demonstrated that suppression of EphA2 significantly reduced cancer-cell migration in cell-based assays. In the present study, we focused on targeting EphA2 in prostate cancer. While not all prostate cancers express EphA2, we showed that enzalutamide induced EphA2 expression in prostate cancer cells and in a patient-derived xenograft (PDX) animal model, which provides further impetus to target EphA2 in prostate cancer. Western blot studies showed that agonistic dimeric synthetic (135H12) and natural (ephrinA1-Fc) ligands effectively degraded EphA2 receptor in the prostate cancer cell line PC-3. The agents also delayed cell migration of prostate cancer (PC-3) cells, while an in vivo PC-3 orthotopic metastatic nude-mouse model also revealed that administration of ephrinA1-Fc or 135H12 strongly reduced metastases. The present study further validates EphA2 as an important target in metastatic prostate cancer treatment. Our results should incentivize further efforts aimed at developing potent and effective EphA2 synthetic agonistic agents for the treatment of EphA2-driven aggressive metastatic tumors including prostate, pancreatic, and breast cancer.

6.
ChemMedChem ; 15(22): 2176-2184, 2020 11 18.
Article in English | MEDLINE | ID: mdl-32790900

ABSTRACT

Recently we reported on aryl-fluorosulfates as possible stable and effective electrophiles for the design of lysine covalent, cell permeable antagonists of protein-protein interactions (PPIs). Here we revisit the use of aryl-sulfonyl fluorides as Lys-targeting moieties, incorporating these electrophiles in XIAP (X-linked inhibitor of apoptosis protein) targeting agents. We evaluated stability in buffer and reactivity with Lys311 of XIAP of various aryl-sulfonyl fluorides using biochemical and biophysical approaches, including displacement assays, mass spectrometry, SDS gel electrophoresis, and denaturation thermal shift measurements. To assess whether these modified electrophilic "warheads" can also react with Tyr, we repeated these evaluations with a Lys311Tyr XIAP mutant. Using a direct cellular assay, we could demonstrate that selected agents are cell permeable and interact covalently with their intended target in cell. These results suggest that certain substituted aryl-sulfonyl fluorides can be useful Lys- or Tyr-targeting electrophiles for the design of covalent pharmacological tools or even future therapeutics targeting protein-protein interactions.


Subject(s)
Drug Design , Lysine/pharmacology , Permeability/drug effects , Sulfinic Acids/pharmacology , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , HEK293 Cells , Humans , Lysine/chemistry , Molecular Structure , Protein Binding/drug effects , Sulfinic Acids/chemical synthesis , Sulfinic Acids/chemistry
7.
Pharmaceuticals (Basel) ; 13(5)2020 May 10.
Article in English | MEDLINE | ID: mdl-32397624

ABSTRACT

Recently, we reported on potent EphA2 targeting compounds and demonstrated that dimeric versions of such agents can exhibit remarkably increased agonistic activity in cellular assays compared to the monomers. Here we further characterize the activity of dimeric compounds at the structural, biochemical, and cellular level. In particular, we propose a structural model for the mechanism of receptor activation by dimeric agents and characterize the effect of most potent compounds in inducing EphA2 activation and degradation in a pancreatic cancer cell line. These cellular studies indicate that the pro-migratory effects induced by the receptor can be reversed in EphA2 knockout cells, by treatment with either a dimeric natural ligand (ephrinA1-Fc), or by our synthetic agonistic dimers. Based on these data we conclude that the proposed agents hold great potential as possible therapeutics in combination with standard of care, where these could help suppressing a major driver for cell migration and tumor metastases. Finally, we also found that, similar to ephrinA1-Fc, dimeric agents cause a sustained internalization of the EphA2 receptor, hence, with proper derivatizations, these could also be used to deliver chemotherapy selectively to pancreatic tumors.

8.
Molecules ; 25(10)2020 05 22.
Article in English | MEDLINE | ID: mdl-32455942

ABSTRACT

Processing of certain viral proteins and bacterial toxins by host serine proteases is a frequent and critical step in virulence. The coronavirus spike glycoprotein contains three (S1, S2, and S2') cleavage sites that are processed by human host proteases. The exact nature of these cleavage sites, and their respective processing proteases, can determine whether the virus can cross species and the level of pathogenicity. Recent comparisons of the genomes of the highly pathogenic SARS-CoV2 and MERS-CoV, with less pathogenic strains (e.g., Bat-RaTG13, the bat homologue of SARS-CoV2) identified possible mutations in the receptor binding domain and in the S1 and S2' cleavage sites of their spike glycoprotein. However, there remains some confusion on the relative roles of the possible serine proteases involved for priming. Using anthrax toxin as a model system, we show that in vivo inhibition of priming by pan-active serine protease inhibitors can be effective at suppressing toxicity. Hence, our studies should encourage further efforts in developing either pan-serine protease inhibitors or inhibitor cocktails to target SARS-CoV2 and potentially ward off future pandemics that could develop because of additional mutations in the S-protein priming sequence in coronaviruses.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Serine Proteases/metabolism , Serine Proteinase Inhibitors/pharmacology , Spike Glycoprotein, Coronavirus/metabolism , Animals , Antigens, Bacterial/toxicity , Antiviral Agents/therapeutic use , Bacterial Toxins/toxicity , Betacoronavirus/pathogenicity , Binding Sites , COVID-19 , Drug Delivery Systems , Female , Furin/pharmacology , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Pandemics , RAW 264.7 Cells , SARS-CoV-2 , Serine Proteinase Inhibitors/therapeutic use , Spike Glycoprotein, Coronavirus/chemistry
9.
Chem Biol Drug Des ; 95(4): 412-426, 2020 04.
Article in English | MEDLINE | ID: mdl-31898401

ABSTRACT

Recently, it was reported that tetrapeptides cyclized via lactam bond between the amino terminus and a glutamic residue in position 4 (termed here N-lock) can nucleate helix formation in longer peptides. We applied such strategy to derive N-locked covalent BH3 peptides that were designed to selectively target the anti-apoptotic protein Bfl-1. The resulting agents were soluble in aqueous buffer and displayed a remarkable (low nanomolar) affinity for Bfl-1 and cellular activity. The crystal structure of the complex between such N-locked covalent peptide and Bfl-1 provided insights on the geometry of the N-locking strategy and of the covalent bond between the agent and Bfl-1.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Forkhead Transcription Factors/chemistry , Nerve Tissue Proteins/chemistry , Peptide Fragments/chemistry , Proto-Oncogene Proteins/chemistry , Amino Acid Sequence , Apoptosis Regulatory Proteins/pharmacokinetics , Crystallization , Forkhead Transcription Factors/metabolism , Humans , Models, Molecular , Nerve Tissue Proteins/metabolism , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Proto-Oncogene Proteins/metabolism
10.
J Med Chem ; 62(20): 9188-9200, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31550155

ABSTRACT

We have recently investigated the reactivity of aryl-fluorosulfates as warheads to form covalent adducts with Lys, Tyr, and His residues. However, the rate of reaction of aryl-fluorosulfates seemed relatively slow, putting into question their effectiveness to form covalent adducts in cell. Unlike the previously reported agents that targeted a relatively remote Lys residue with respect to the target's binding site, the current agents were designed to more directly juxtapose an aryl-fluorosulfate with a Lys residue that is located within the binding pocket of the BIR3 domain of X-linked inhibitor of apoptosis protein (XIAP). We found that such new agents can effectively and rapidly form a covalent adduct with XIAP-BIR3 in vitro and in cell, approaching the rate of reaction, cellular permeability, and stability that are similar to what attained by acrylamides when targeting Cys residues. Our studies further validate aryl-fluorosulfates as valuable Lys-targeting electrophiles, for the design of inhibitors of both enzymes and protein-protein interactions.


Subject(s)
Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Lysine/chemistry , Sulfates/chemistry , Apoptosis/drug effects , Binding Sites , Cell Membrane Permeability/drug effects , Drug Design , Drug Stability , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Lysine/metabolism , Molecular Docking Simulation , Permeability/drug effects , Protein Binding , Protein Interaction Maps/drug effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sulfates/metabolism , Sulfates/pharmacology , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
11.
J Med Chem ; 62(11): 5616-5627, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31095386

ABSTRACT

We have recently reported a series of Lys-covalent agents targeting the BIR3 domain of the X-linked inhibitor of apoptosis protein (XIAP) using a benzamide-sulfonyl fluoride warhead. Using XIAP as a model system, we further investigated a variety of additional warheads that can be easily incorporated into binding peptides and analyzed their ability to form covalent adducts with lysine and other amino acids, including tyrosine, histidine, serine, and threonine, using biochemical and biophysical assays. Moreover, we tested aqueous, plasma stability, cell permeability, and cellular efficacy of the most effective agents. These studies identified aryl-fluoro sulfates as likely the most suitable electrophiles to effectively form covalent adducts with Lys, Tyr, and His residues, given that these agents were cell permeable and stable in aqueous buffer and in plasma. Our studies contain a number of general findings that open new possible avenues for the design of potent covalent protein-protein interaction antagonists.


Subject(s)
Benzamides/pharmacology , X-Linked Inhibitor of Apoptosis Protein/chemistry , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Benzamides/chemistry , Benzamides/metabolism , Cell Line, Tumor , Drug Design , Humans , Mice , Models, Molecular , Permeability , Protein Binding/drug effects , Protein Conformation , Water/chemistry
12.
ACS Chem Biol ; 13(9): 2633-2644, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30110533

ABSTRACT

EphA2 overexpression is invariably associated with poor prognosis and development of aggressive metastatic cancers in pancreatic, prostate, lung, ovarian, and breast cancers and melanoma. Recent efforts from our laboratories identified a number of agonistic peptides targeting the ligand-binding domain of the EphA2 receptor. The individual agents, however, were still relatively weak in affinities (micromolar range) that precluded detailed structural studies on the mode of action. Using a systematic optimization of the 12-mer peptide mimetic 123B9, we were able to first derive an agent that displayed a submicromolar affinity for the receptor. This agent enabled cocrystallization with the EphA2 ligand-binding domain providing for the first time the structural basis for their agonistic mechanism of action. In addition, the atomic coordinates of the complex enabled rapid iterations of structure-based optimizations that resulted in a novel agonistic agent, named 135H11, with a nanomolar affinity for the receptor, as demonstrated by in vitro binding assays (isothermal titration calorimetry measurements), and a biochemical displacement assay. As we have recently demonstrated, the cellular activity of these agents is further increased by synthesizing dimeric versions of the compounds. Hence, we report that a dimeric version of 135H11 is extremely effective at low nanomolar concentrations to induce cellular receptor activation, internalization, and inhibition of cell migration in a pancreatic cancer cell line. Given the pivotal role of EphA2 in tumor growth, angiogenesis, drug resistance, and metastasis, these agents, and the associated structural studies, provide significant advancements in the field for the development of novel EphA2-targeting therapeutics or diagnostics.


Subject(s)
Drug Design , Peptides/chemistry , Peptides/pharmacology , Receptor, EphA2/agonists , Amino Acid Sequence , Binding Sites/drug effects , Cell Line, Tumor , Crystallography, X-Ray , HEK293 Cells , Humans , Ligands , Molecular Docking Simulation , Receptor, EphA2/chemistry , Receptor, EphA2/metabolism
13.
J Med Chem ; 61(14): 6350-6363, 2018 Jul 26.
Article in English | MEDLINE | ID: mdl-29940121

ABSTRACT

Recently we reported that rapid determination of enthalpy of binding can be achieved for a large number of congeneric agents or in combinatorial libraries fairly efficiently. We show that using a thermodynamic Craig plot can be very useful in dissecting the enthalpy and entropy contribution of different substituents on a common scaffold, in order to design potent, selective, or pan-active compounds. In our implementation, the approach identified a critical Lys residue in the BIR3 domain of XIAP. We report for the first time that it is possible to target such residue covalently to attain potent and selective agents. Preliminary cellular studies in various models of leukemia, multiple myeloma, and pancreatic cancers suggest that the derived agents possess a potentially intriguing pattern of activity, especially for cell lines that are resistant to the pan-IAP antagonist and clinical candidate LCL161.


Subject(s)
Drug Design , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Cell Line , Humans , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/metabolism , Molecular Docking Simulation , Protein Conformation , Thermodynamics , X-Linked Inhibitor of Apoptosis Protein/chemistry , X-Linked Inhibitor of Apoptosis Protein/metabolism
14.
J Med Chem ; 61(5): 2052-2061, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29470068

ABSTRACT

EphA2 overexpression has been associated with metastasis in multiple cancer types, including melanomas and ovarian, prostate, lung, and breast cancers. We have recently proposed the development of peptide-drug conjugates (PDCs) using agonistic EphA2-targeting agents, such as the YSA peptide or its optimized version, 123B9. Although our studies indicated that YSA- and 123B9-drug conjugates can selectively deliver cytotoxic drugs to cancer cells in vivo, the relatively low cellular agonistic activities (i.e., the high micromolar concentrations required) of the agents toward the EphA2 receptor remained a limiting factor to the further development of these PDCs in the clinic. Here, we report that a dimeric version of 123B9 can induce receptor activation at nanomolar concentrations. Furthermore, we demonstrated that the conjugation of dimeric 123B9 with paclitaxel is very effective at targeting circulating tumor cells and inhibiting lung metastasis in breast-cancer models. These studies represent an important step toward the development of effective EphA2-targeting PDCs.


Subject(s)
Breast Neoplasms/drug therapy , Drug Delivery Systems/methods , Neoplasm Metastasis/drug therapy , Neoplastic Cells, Circulating/drug effects , Receptor, EphA2/agonists , Animals , Antineoplastic Agents/administration & dosage , Humans , Mice , Paclitaxel/administration & dosage , Peptides/therapeutic use
15.
Proc Natl Acad Sci U S A ; 114(48): E10399-E10408, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29133417

ABSTRACT

Helminths trigger multiple immunomodulatory pathways that can protect from sepsis. Human resistin (hRetn) is an immune cell-derived protein that is highly elevated in helminth infection and sepsis. However, the function of hRetn in sepsis, or whether hRetn influences helminth protection against sepsis, is unknown. Employing hRetn-expressing transgenic mice (hRETNTg+) and recombinant hRetn, we identify a therapeutic function for hRetn in lipopolysaccharide (LPS)-induced septic shock. hRetn promoted helminth-induced immunomodulation, with increased survival of Nippostrongylus brasiliensis (Nb)-infected hRETNTg+ mice after a fatal LPS dose compared with naive mice or Nb-infected hRETNTg- mice. Employing immunoprecipitation assays, hRETNTg+Tlr4-/- mice, and human immune cell culture, we demonstrate that hRetn binds the LPS receptor Toll-like receptor 4 (TLR4) through its N terminal and modulates STAT3 and TBK1 signaling, triggering a switch from proinflammatory to anti-inflammatory responses. Further, we generate hRetn N-terminal peptides that are able to block LPS proinflammatory function. Together, our studies identify a critical role for hRetn in blocking LPS function with important clinical significance in helminth-induced immunomodulation and sepsis.


Subject(s)
Lipopolysaccharides/metabolism , Resistin/immunology , Shock, Septic/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Animals , Biological Therapy/methods , Disease Models, Animal , Female , Gram-Negative Bacteria/immunology , Gram-Negative Bacteria/metabolism , Humans , Lipopolysaccharide Receptors/immunology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nippostrongylus/immunology , Protective Agents , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , STAT3 Transcription Factor/metabolism , Shock, Septic/microbiology , Shock, Septic/therapy , Signal Transduction/immunology
16.
ACS Chem Biol ; 12(2): 444-455, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28026162

ABSTRACT

Upregulation of antiapoptotic Bcl-2 proteins in certain tumors confers cancer cell resistance to chemotherapy or radiations. Members of the antiapoptotic Bcl-2 proteins, including Bcl-2, Mcl-1, Bcl-xL, Bcl-w, and Bfl-1, inhibit apoptosis by selectively binding to conserved α-helical regions, named BH3 domains, of pro-apoptotic proteins such as Bim, tBid, Bad, or NOXA. Five antiapoptotic proteins have been identified that interact with various selectivity with BH3 containing pro-apoptotic counterparts. Cancer cells present various and variable levels of these proteins, making the design of effective apoptosis based therapeutics challenging. Recently, BH3 profiling was introduced as a method to classify cancer cells based on their ability to resist apoptosis following exposure to selected BH3 peptides. However, these studies were based on binding affinities measured with model BH3 peptides and Bcl-2-proteins taken from mouse sequences. While the majority of these interactions are conserved between mice and humans, we found surprisingly that human NOXA binds to human Bfl-1 potently and covalently via conserved Cys residues, with over 2 orders of magnitude increased affinity over hMcl-1. Our data suggest that some assumptions of the original BH3 profiling need to be revisited and that perhaps further targeting efforts should be redirected toward Bfl-1, for which no suitable specific inhibitors or pharmacological tools have been reported. In this regard, we also describe the initial design and characterizations of novel covalent BH3-based agents that potently target Bfl-1. These molecules could provide a novel platform on which to design effective Bfl-1 targeting therapeutics.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Minor Histocompatibility Antigens/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Sequence , Apoptosis , Cell Line, Tumor , Humans
17.
Bioconjug Chem ; 27(12): 2911-2922, 2016 Dec 21.
Article in English | MEDLINE | ID: mdl-27809498

ABSTRACT

Gold nanocages (AuNCs) have been shown to be a useful tool for harnessing imaging and hyperthermia therapy of cancer, thanks to their unique optical properties, low toxicity, and facile surface functionalization. Herein, we use AuNCs for selective targeting of prostate cancer cells (PC3) via specific interaction between neuropeptide Y (NPY) receptor and three different NPY analogs conjugated to AuNCs. Localized surface plasmon resonance band of the nanoconjugates was set around 800 nm, which is appropriate for in vivo applications. Long-term stability of nanoconjugates in different media was confirmed by UV-vis and DLS studies. Active NPY receptor targeting was observed by confocal microscopy showing time-dependent AuNCs cellular uptake. Activation of ERK1/2 pathway was evaluated by Western blot to confirm the receptor-mediated specific interaction with PC3. Cellular uptake kinetics were compared as a function of peptide structure. Cytotoxicity of nanoconjugates was evaluated by MTS and Annexin V assays, confirming their safety within the concentration range explored. Hyperthermia studies were carried out irradiating the cells, previously incubated with AuNCs, with a pulsed laser at 800 nm wavelength, showing a heating enhancement ranging from 6 to 35 °C above the culture temperature dependent on the irradiation power (between 1.6 and 12.7 W/cm2). Only cells treated with AuNCs underwent morphological alterations in the cytoskeleton structure upon laser irradiation, leading to membrane blebbing and loss of microvilli associated with cell migration. This effect is promising in view of possible inhibition of proliferation and invasion of cancer cells. In summary, our Au-peptide NCs proved to be an efficient theranostic nanosystem for targeted detection and activatable killing of prostate cancer cells.


Subject(s)
Molecular Targeted Therapy/methods , Nanoparticles , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/therapy , Theranostic Nanomedicine/methods , Cell Line, Tumor , Drug Design , Gold , Humans , Lasers , Male , Microscopy, Confocal , Microscopy, Electron, Transmission , Nanoparticles/chemistry , Peptides/chemical synthesis , Peptides/chemistry , Prostatic Neoplasms/metabolism , Receptors, Neuropeptide Y/metabolism , Thermography/methods
18.
Pharmacol Res ; 111: 155-162, 2016 09.
Article in English | MEDLINE | ID: mdl-27293049

ABSTRACT

Tumor homing peptides (THPs) specific for a representative breast cancer cell line (MCF-7) were carefully selected basing on a phage-displayed peptide library freely available on the web, namely the "TumorHoPe: A Database of Tumor Homing Peptides". The selected THPs were synthesized and evaluated in terms of their affinity toward MCF-7 cells. Out of 5 tested THPs, 3 best-performing peptide sequences and 1 scrambled sequence were separately conjugated to spherical gold nanoparticles yielding stable nanoconjugates. THP nanoconjugates were examined for their ability to actively target MCF-7 cells in comparison to noncancerous 3T3-L1 fibroblast cells. These THP-gold nanoconjugates exhibited good selectivity and binding affinity by flow cytometry, and low cytotoxicity as assayed by cell death experiments. The uptake of targeted nanoconjugates by the breast cancer cells was confirmed by transmission electron microscopy analysis. This work demonstrates that it is possible to exploit the conjugation of short peptides selected from phage-displayed libraries to develop nanomaterials reliably endowed with tumor targeting potential irrespective of a specific knowledge of the target cell biology.


Subject(s)
Breast Neoplasms/metabolism , Cell Surface Display Techniques , Drug Carriers , Gold/chemistry , Metal Nanoparticles , Nanoconjugates , Peptide Library , Peptides/metabolism , 3T3-L1 Cells , Animals , Biological Transport , Breast Neoplasms/genetics , Breast Neoplasms/ultrastructure , Drug Compounding , Female , Humans , MCF-7 Cells , Mice , Microscopy, Electron, Transmission , Particle Size , Peptides/chemistry
20.
PLoS One ; 10(11): e0142509, 2015.
Article in English | MEDLINE | ID: mdl-26566224

ABSTRACT

Malaria is an infectious disease caused by Plasmodium parasites. It results in an annual death-toll of ~ 600,000. Resistance to all medications currently in use exists, and novel antimalarial drugs are urgently needed. Plasmepsin V (PmV) is an essential Plasmodium protease and a highly promising antimalarial target, which still lacks molecular characterization and drug-like inhibitors. PmV, cleaving the PExEl motif, is the key enzyme for PExEl-secretion, an indispensable parasitic process for virulence and infection. Here, we describe the accessibility of PmV catalytic pockets to inhibitors and propose a novel strategy for PmV inhibition. We also provide molecular and structural data suitable for future drug development. Using high-throughput platforms, we identified a novel scaffold that interferes with PmV in-vitro at picomolar ranges (~ 1,000-fold more active than available compounds). Via systematic replacement of P and P' regions, we assayed the physico-chemical requirements for PmV inhibition, achieving an unprecedented IC50 of ~20 pM. The hydroxyethylamine moiety, the hydrogen acceptor group in P2', the lipophilic groups upstream to P3, the arginine and other possible substitutions in position P3 proved to be critically important elements in achieving potent inhibition. In-silico analyses provided essential QSAR information and model validation. Our inhibitors act 'on-target', confirmed by cellular interference of PmV function and biochemical interaction with inhibitors. Our inhibitors are poorly performing against parasite growth, possibly due to poor stability of their peptidic component and trans-membrane permeability. The lowest IC50 for parasite growth inhibition was ~ 15 µM. Analysis of inhibitor internalization revealed important pharmacokinetic features for PExEl-based molecules. Our work disclosed novel pursuable drug design strategies for highly efficient PmV inhibition highlighting novel molecular elements necessary for picomolar activity against PmV. All the presented data are discussed in respect to human aspartic proteases and previously reported inhibitors, highlighting differences and proposing new strategies for drug development.


Subject(s)
Antimalarials/pharmacology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Protease Inhibitors/pharmacology , Protozoan Proteins/antagonists & inhibitors , Antimalarials/chemistry , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Catalytic Domain/drug effects , Humans , Malaria, Falciparum/parasitology , Molecular Docking Simulation , Plasmodium falciparum/chemistry , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development , Protease Inhibitors/chemistry , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...