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










Publication year range
1.
Sci Rep ; 13(1): 4748, 2023 03 23.
Article in English | MEDLINE | ID: mdl-36959353

ABSTRACT

Fabry disease is caused by a deficiency of α-galactosidase A (GLA) leading to the lysosomal accumulation of globotriaosylceramide (Gb3) and other glycosphingolipids. Fabry patients experience significant damage to the heart, kidney, and blood vessels that can be fatal. Here we apply directed evolution to generate more stable GLA variants as potential next generation treatments for Fabry disease. GLAv05 and GLAv09 were identified after screening more than 12,000 GLA variants through 8 rounds of directed evolution. Both GLAv05 and GLAv09 exhibit increased stability at both lysosomal and blood pH, stability to serum, and elevated enzyme activity in treated Fabry fibroblasts (19-fold) and GLA-/- podocytes (10-fold). GLAv05 and GLAv09 show improved pharmacokinetics in mouse and non-human primates. In a Fabry mouse model, the optimized variants showed prolonged half-lives in serum and relevant tissues, and a decrease of accumulated Gb3 in heart and kidney. To explore the possibility of diminishing the immunogenic potential of rhGLA, amino acid residues in sequences predicted to bind MHC II were targeted in late rounds of GLAv09 directed evolution. An MHC II-associated peptide proteomics assay confirmed a reduction in displayed peptides for GLAv09. Collectively, our findings highlight the promise of using directed evolution to generate enzyme variants for more effective treatment of lysosomal storage diseases.


Subject(s)
Fabry Disease , Humans , Mice , Animals , Fabry Disease/drug therapy , Fabry Disease/genetics , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism , Kidney/metabolism , Disease Models, Animal , Fibroblasts/metabolism
2.
Neuro Oncol ; 23(10): 1723-1735, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34216463

ABSTRACT

BACKGROUND: High-grade meningioma is an aggressive type of brain cancer that is often recalcitrant to surgery and radiotherapy, leading to poor overall survival. Currently, there are no FDA-approved drugs for meningioma, highlighting the need for new therapeutic options, but development is challenging due to the lack of predictive preclinical models. METHODS: To leverage the known overexpression of procaspase-3 in meningioma, PAC-1, a blood-brain barrier penetrant procaspase-3 activator, was evaluated for its ability to induce apoptosis in meningioma cells. To enhance the effects of PAC-1, combinations with either hydroxyurea or temozolomide were explored in cell culture. Both combinations were further investigated in small groups of canine meningioma patients and assessed by MRI, and the novel apoptosis tracer, [18F]C-SNAT4, was evaluated in patients treated with PAC-1 + HU. RESULTS: In meningioma cell lines in culture, PAC-1 + HU are synergistic while PAC-1 + TMZ show additive-to-synergistic effects. In canine meningioma patients, PAC-1 + HU led to stabilization of disease and no change in apoptosis within the tumor, whereas PAC-1 + TMZ reduced tumor burden in all three canine patients treated. CONCLUSIONS: Our results suggest PAC-1 + TMZ as a potentially efficacious combination for the treatment of human meningioma, and also demonstrate the utility of including pet dogs with meningioma as a means to assess anticancer strategies for this common brain tumor.


Subject(s)
Meningeal Neoplasms , Meningioma , Animals , Apoptosis , Caspase 3 , Cell Culture Techniques , Cell Line, Tumor , Dogs , Humans , Hydroxyurea/pharmacology , Meningeal Neoplasms/drug therapy , Meningeal Neoplasms/veterinary , Meningioma/drug therapy , Meningioma/veterinary , Temozolomide/pharmacology
3.
Curr Opin Chem Biol ; 64: 38-47, 2021 10.
Article in English | MEDLINE | ID: mdl-33933937

ABSTRACT

Therapeutic proteins alleviate disease pathology by supplementing missing or defective native proteins, sequestering superfluous proteins, or by acting through designed non-natural mechanisms. Although therapeutic proteins often have the same amino acid sequence as their native counterpart, their maturation paths from expression to the site of physiological activity are inherently different, and optimizing protein sequences for properties that 100s of millions of years of evolution did not need to address presents an opportunity to develop better biological treatments. Because therapeutic proteins are inherently non-natural entities, optimization for their desired function should be considered analogous to that of small molecule drug candidates, which are optimized through expansive combinatorial variation by the medicinal chemist. Here, we review recent successes and challenges of protein engineering for optimized therapeutic efficacy.


Subject(s)
Protein Engineering , Proteins , Amino Acid Sequence , Models, Molecular , Proteins/chemistry
4.
Nat Chem ; 12(10): 906-913, 2020 10.
Article in English | MEDLINE | ID: mdl-32868886

ABSTRACT

Drug candidates that form covalent linkages with their target proteins have been underexplored compared with the conventional counterparts that modulate biological function by reversibly binding to proteins, in part due to concerns about off-target reactivity. However, toxicity linked to off-target reactivity can be minimized by using latent electrophiles that only become activated towards covalent bond formation on binding a specific protein. Here we study sulfuramidimidoyl fluorides, a class of weak electrophiles that undergo sulfur(VI) fluoride exchange chemistry. We show that equilibrium binding of a sulfuramidimidoyl fluoride to a protein can allow nucleophilic attack by a specific amino acid side chain, which leads to conjugate formation. We incubated small molecules, each bearing a sulfuramidimidoyl fluoride electrophile, with human cell lysate, and the protein conjugates formed were identified by affinity chromatography-mass spectrometry. This inverse drug discovery approach identified a compound that covalently binds to and irreversibly inhibits the activity of poly(ADP-ribose) polymerase 1, an important anticancer target in living cells.


Subject(s)
Drug Discovery , Fluorides/chemistry , Sulfhydryl Compounds/chemistry , Sulfur/chemistry , Chromatography, Affinity , HEK293 Cells , Humans , Mass Spectrometry , Molecular Structure , Sulfhydryl Compounds/chemical synthesis
5.
Oncotarget ; 8(46): 80124-80138, 2017 Oct 06.
Article in English | MEDLINE | ID: mdl-29113289

ABSTRACT

PURPOSE: Glioblastoma is a deadly brain cancer with a median survival time of ∼15 months. Ionizing radiation plus the DNA alkylator temozolomide (TMZ) is the current standard therapy. PAC-1, a procaspase-3 activating small molecule, is blood-brain barrier penetrant and has previously demonstrated ability to synergize with diverse pro-apoptotic chemotherapeutics. We studied if PAC-1 could enhance the activity of TMZ, and whether addition of PAC-1 to standard treatment would be feasible in spontaneous canine malignant gliomas. EXPERIMENTAL DESIGN: Using cell lines and online gene expression data, we identified procaspase-3 as a potential molecular target for most glioblastomas. We investigated PAC-1 as a single agent and in combination with TMZ against glioma cells in culture and in orthotopic rodent models of glioma. Three dogs with spontaneous gliomas were treated with an analogous human glioblastoma treatment protocol, with concurrent PAC-1. RESULTS: Procaspase-3 is expressed in gliomas, with higher gene expression correlating with increased tumor grade and decreased prognosis. PAC-1 is cytotoxic to glioma cells in culture and active in orthotopic rodent glioma models. PAC-1 added to TMZ treatments in cell culture increases apoptotic death, and the combination significantly increases survival in orthotopic glioma models. Addition of PAC-1 to TMZ and radiation was well-tolerated in 3 out of 3 pet dogs with spontaneous glioma, and partial to complete tumor reductions were observed. CONCLUSIONS: Procaspase-3 is a clinically relevant target for treatment of glioblastoma. Synergistic activity of PAC-1/TMZ in rodent models and the demonstration of feasibility of the combined regime in canine patients suggest potential for PAC-1 in the treatment of glioblastoma.

6.
ACS Cent Sci ; 2(8): 545-59, 2016 Aug 24.
Article in English | MEDLINE | ID: mdl-27610416

ABSTRACT

Conventional chemotherapeutics remain essential treatments for most cancers, but their combination with other anticancer drugs (including targeted therapeutics) is often complicated by unpredictable synergies and multiplicative toxicities. As cytotoxic anticancer chemotherapeutics generally function through induction of apoptosis, we hypothesized that a molecularly targeted small molecule capable of facilitating a central and defining step in the apoptotic cascade, the activation of procaspase-3 to caspase-3, would broadly and predictably enhance activity of cytotoxic drugs. Here we show that procaspase-activating compound 1 (PAC-1) enhances cancer cell death induced by 15 different FDA-approved chemotherapeutics, across many cancer types and chemotherapeutic targets. In particular, the promising combination of PAC-1 and doxorubicin induces a synergistic reduction in tumor burden and enhances survival in murine tumor models of osteosarcoma and lymphoma. This PAC-1/doxorubicin combination was evaluated in 10 pet dogs with naturally occurring metastatic osteosarcoma or lymphoma, eliciting a biologic response in 3 of 6 osteosarcoma patients and 4 of 4 lymphoma patients. Importantly, in both mice and dogs, coadministration of PAC-1 with doxorubicin resulted in no additional toxicity. On the basis of the mode of action of PAC-1 and the high expression of procaspase-3 in many cancers, these results suggest the combination of PAC-1 with cytotoxic anticancer drugs as a potent and general strategy to enhance therapeutic response.

7.
Cell Rep ; 13(9): 2027-36, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655912

ABSTRACT

Apoptosis is generally believed to be a process that requires several hours, in contrast to non-programmed forms of cell death that can occur in minutes. Our findings challenge the time-consuming nature of apoptosis as we describe the discovery and characterization of a small molecule, named Raptinal, which initiates intrinsic pathway caspase-dependent apoptosis within minutes in multiple cell lines. Comparison to a mechanistically diverse panel of apoptotic stimuli reveals that Raptinal-induced apoptosis proceeds with unparalleled speed. The rapid phenotype enabled identification of the critical roles of mitochondrial voltage-dependent anion channel function, mitochondrial membrane potential/coupled respiration, and mitochondrial complex I, III, and IV function for apoptosis induction. Use of Raptinal in whole organisms demonstrates its utility for studying apoptosis in vivo for a variety of applications. Overall, rapid inducers of apoptosis are powerful tools that will be used in a variety of settings to generate further insight into the apoptotic machinery.


Subject(s)
Apoptosis , Cyclopentanes/chemistry , Fluorenes/chemistry , Animals , Apoptosis/drug effects , Caspase 3/chemistry , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Complement C8/deficiency , Complement C8/genetics , Cyclopentanes/pharmacokinetics , Cyclopentanes/toxicity , Embryo, Nonmammalian/metabolism , Fas-Associated Death Domain Protein/deficiency , Fas-Associated Death Domain Protein/genetics , Fluorenes/pharmacokinetics , Fluorenes/toxicity , Half-Life , Humans , Jurkat Cells , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Zebrafish
8.
J Med Chem ; 58(9): 4046-65, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25856364

ABSTRACT

Procaspase-activating compound 1 (PAC-1) is an o-hydroxy-N-acylhydrazone that induces apoptosis in cancer cells by chelation of labile inhibitory zinc from procaspase-3. PAC-1 has been assessed in a wide variety of cell culture experiments and in vivo models of cancer, with promising results, and a phase 1 clinical trial in cancer patients has been initiated (NCT02355535). For certain applications, however, the in vivo half-life of PAC-1 could be limiting. Thus, with the goal of developing a compound with enhanced metabolic stability, a series of PAC-1 analogues were designed containing modifications that systematically block sites of metabolic vulnerability. Evaluation of the library of compounds identified four potentially superior candidates with comparable anticancer activity in cell culture, enhanced metabolic stability in liver microsomes, and improved tolerability in mice. In head-to-head experiments with PAC-1, pharmacokinetic evaluation in mice demonstrated extended elimination half-lives and greater area under the curve values for each of the four compounds, suggesting them as promising candidates for further development.


Subject(s)
Antineoplastic Agents/chemistry , Hydrazones/chemistry , Piperazines/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Area Under Curve , Cell Line, Tumor , Dogs , Drug Screening Assays, Antitumor , Half-Life , Humans , Hydrazones/pharmacokinetics , Hydrazones/pharmacology , Mice , Microsomes, Liver/metabolism , Piperazines/pharmacokinetics , Piperazines/pharmacology , Rats , Small Molecule Libraries , Structure-Activity Relationship
9.
J Am Chem Soc ; 136(4): 1312-9, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-24383395

ABSTRACT

Combination anticancer therapy typically consists of drugs that target different biochemical pathways or those that act on different targets in the same pathway. Here we demonstrate a new concept in combination therapy, that of enzyme activation with two compounds that hit the same biological target, but through different mechanisms. Combinations of procaspase-3 activators PAC-1 and 1541B show considerable synergy in activating procaspase-3 in vitro, stimulate rapid and dramatic maturation of procaspase-3 in multiple cancer cell lines, and powerfully induce caspase-dependent apoptotic death to a degree well exceeding the additive effect. In addition, the combination of PAC-1 and 1541B effectively reduces tumor burden in a murine lymphoma model at dosages for which the compounds alone have minimal or no effect. These data suggest the potential of PAC-1/1541B combinations for the treatment of cancer and, more broadly, demonstrate that differentially acting enzyme activators can potently synergize to give a significantly heightened biological effect.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Caspase 3/metabolism , Hydrazones/pharmacology , Lymphoma/drug therapy , Piperazines/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , HL-60 Cells , Humans , Hydrazones/chemistry , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Molecular Structure , Piperazines/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
10.
ACS Comb Sci ; 14(1): 44-50, 2012 Jan 09.
Article in English | MEDLINE | ID: mdl-22007686

ABSTRACT

Procaspase-Activating Compound 1 (PAC-1) is an ortho-hydroxy N-acyl hydrazone that enhances the enzymatic activity of procaspase-3 in vitro and induces apoptosis in cancer cells. An analogue of PAC-1, called S-PAC-1, was evaluated in a veterinary clinical trial in pet dogs with lymphoma and found to have considerable potential as an anticancer agent. With the goal of identifying more potent compounds in this promising class of experimental therapeutics, a combinatorial library based on PAC-1 was created, and the compounds were evaluated for their ability to induce death of cancer cells in culture. For library construction, 31 hydrazides were condensed in parallel with 27 aldehydes to create 837 PAC-1 analogues, with an average purity of 91%. The compounds were evaluated for their ability to induce apoptosis in cancer cells, and through this work, six compounds were discovered to be substantially more potent than PAC-1 and S-PAC-1. These six hits were further evaluated for their ability to relieve zinc-mediated inhibition of procaspase-3 in vitro. In general, the newly identified hit compounds are two- to four-fold more potent than PAC-1 and S-PAC-1 in cell culture, and thus have promise as experimental therapeutics for treatment of the many cancers that have elevated expression levels of procaspase-3.


Subject(s)
Antineoplastic Agents/chemical synthesis , Chemistry Techniques, Synthetic/methods , Hydrazones/chemistry , Piperazines/chemistry , Small Molecule Libraries/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Caspase 3/biosynthesis , Caspase Inhibitors , Cell Culture Techniques , Cell Survival/drug effects , Drug Discovery , Drug Screening Assays, Antitumor , Enzyme Induction , Flow Cytometry , Humans , Molecular Structure , Small Molecule Libraries/chemistry , U937 Cells
11.
Nat Protoc ; 5(2): 294-302, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20134429

ABSTRACT

This protocol describes the gram-scale solution-phase synthesis of the colorimetric caspase-3/7 substrate Ac-DEVD-pNA. The caspase enzymes are integral to cellular inflammation and apoptotic cascades, and are commonly studied by cell biologists, medicinal chemists and chemical biologists. In particular, the assessment of caspase enzymatic activity is a standard method to evaluate cell death pathways and new apoptosis-modulating agents. Caspase enzymatic activity can be conveniently monitored with peptidic chromogenic or fluorogenic substrates, with certain peptide sequences imparting selectivity for certain caspases. The synthesis of these peptide substrates is typically carried out by solid-phase synthesis, a method that is not ideal for production of the gram quantities needed for high-throughput screening. Described herein is a facile method for the synthesis of the Ac-DEVD-pNA caspase-3/7 substrate using solution-phase peptide synthesis. This protocol, involving iterative PyBOP-mediated couplings and Fmoc deprotections, is rapid (about 5 d), operationally simple and can be used to generate over 1 g of product at a fraction of the cost of the commercial substrate.


Subject(s)
Anilides/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Oligopeptides/metabolism , Anilides/chemical synthesis , Aniline Compounds/metabolism , Apoptosis/drug effects , Enzyme Activation , Humans , Hydrolysis , Inflammation/enzymology , Inflammation/pathology , Oligopeptides/chemical synthesis , Peptides/chemical synthesis , Solutions , Solvents , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...