Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Oncotarget ; 14: 1-13, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36634212

ABSTRACT

Overexpression of CD74, a type II transmembrane glycoprotein involved in MHC class II antigen presentation, has been reported in many B-cell non-Hodgkin lymphomas (NHLs) and in multiple myeloma (MM). STRO-001 is a site-specific, predominantly single-species antibody-drug conjugate (ADC) that targets CD74 and has demonstrated efficacy in xenograft models of MM and tolerability in non-human primates. Here we report results of preclinical studies designed to elucidate the potential role of STRO-001 in B-cell NHL. STRO-001 displayed nanomolar and sub-nanomolar cytotoxicity in 88% (15/17) of cancer cell lines tested. STRO-001 showed potent cytotoxicity on proliferating B cells while limited cytotoxicity was observed on naïve human B cells. A linear dose-response relationship was demonstrated in vivo for DLBCL models SU-DHL-6 and U2932. Tumor regression was induced at doses less than 5 mg/kg, while maximal activity with complete cures were observed starting at 10 mg/kg. In MCL Mino and Jeko-1 xenografts, STRO-001 starting at 3 mg/kg significantly prolonged survival or induced tumor regression, respectively, leading to tumor eradication in both models. In summary, high CD74 expression levels in tumors, nanomolar cellular potency, and significant anti-tumor in DLBCL and MCL xenograft models support the ongoing clinical study of STRO-001 in patients with B-cell NHL.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Lymphoma, Non-Hodgkin , Multiple Myeloma , Animals , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Multiple Myeloma/pathology , Lymphoma, Non-Hodgkin/drug therapy , Cell Line, Tumor
2.
Mol Cancer Ther ; 22(2): 155-167, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36459691

ABSTRACT

STRO-002 is a novel homogeneous folate receptor alpha (FolRα) targeting antibody-drug conjugate (ADC) currently being investigated in the clinic as a treatment for ovarian and endometrial cancers. Here, we describe the discovery, optimization, and antitumor properties of STRO-002. STRO-002 was generated by conjugation of a novel cleavable 3-aminophenyl hemiasterlin linker-warhead (SC239) to the nonnatural amino acid para-azidomethyl-L-phenylalanine incorporated at specific positions within a high affinity anti-FolRα antibody using Sutro's XpressCF+, which resulted in a homogeneous ADC with a drug-antibody ratio (DAR) of 4. STRO-002 binds to FolRα with high affinity, internalizes rapidly into target positive cells, and releases the tubulin-targeting cytotoxin 3-aminophenyl hemiasterlin (SC209). SC209 has reduced potential for drug efflux via P-glycoprotein 1 drug pump compared with other tubulin-targeting payloads. While STRO-002 lacks nonspecific cytotoxicity toward FolRα-negative cell lines, bystander killing of target negative cells was observed when cocultured with target positive cells. STRO-002 is stable in circulation with no change in DAR for up to 21 days and has a half-life of 6.4 days in mice. A single dose of STRO-002 induced significant tumor growth inhibition in FolRα-expressing xenograft models and patient-derived xenograft models. In addition, combination treatment with carboplatin or Avastin further increased STRO-002 efficacy in xenograft models. The potent and specific preclinical efficacy of STRO-002 supports clinical development of STRO-002 for treating patients with FolRα-expressing cancers, including ovarian, endometrial, and non-small cell lung cancer. Phase I dose escalation for STRO-002 is in progress in ovarian cancer and endometrial cancer patients (NCT03748186 and NCT05200364).


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Endometrial Neoplasms , Immunoconjugates , Lung Neoplasms , Female , Humans , Animals , Mice , Immunoconjugates/chemistry , Tubulin/metabolism , Folate Receptor 1 , Antineoplastic Agents/pharmacology , Endometrial Neoplasms/drug therapy , Cell Line, Tumor , Xenograft Model Antitumor Assays
3.
Front Immunol ; 13: 899617, 2022.
Article in English | MEDLINE | ID: mdl-35720389

ABSTRACT

COVID-19 emergency use authorizations and approvals for vaccines were achieved in record time. However, there remains a need to develop additional safe, effective, easy-to-produce, and inexpensive prevention to reduce the risk of acquiring SARS-CoV-2 infection. This need is due to difficulties in vaccine manufacturing and distribution, vaccine hesitancy, and, critically, the increased prevalence of SARS-CoV-2 variants with greater contagiousness or reduced sensitivity to immunity. Antibodies from eggs of hens (immunoglobulin Y; IgY) that were administered the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein were developed for use as nasal drops to capture the virus on the nasal mucosa. Although initially raised against the 2019 novel coronavirus index strain (2019-nCoV), these anti-SARS-CoV-2 RBD IgY surprisingly had indistinguishable enzyme-linked immunosorbent assay binding against variants of concern that have emerged, including Alpha (B.1.1.7), Beta (B.1.351), Delta (B.1.617.2), and Omicron (B.1.1.529). This is different from sera of immunized or convalescent patients. Culture neutralization titers against available Alpha, Beta, and Delta were also indistinguishable from the index SARS-CoV-2 strain. Efforts to develop these IgY for clinical use demonstrated that the intranasal anti-SARS-CoV-2 RBD IgY preparation showed no binding (cross-reactivity) to a variety of human tissues and had an excellent safety profile in rats following 28-day intranasal delivery of the formulated IgY. A double-blind, randomized, placebo-controlled phase 1 study evaluating single-ascending and multiple doses of anti-SARS-CoV-2 RBD IgY administered intranasally for 14 days in 48 healthy adults also demonstrated an excellent safety and tolerability profile, and no evidence of systemic absorption. As these antiviral IgY have broad selectivity against many variants of concern, are fast to produce, and are a low-cost product, their use as prophylaxis to reduce SARS-CoV-2 viral transmission warrants further evaluation. Clinical Trial Registration: https://www.clinicaltrials.gov/ct2/show/NCT04567810, identifier NCT04567810.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Viral , COVID-19/prevention & control , Chickens , Female , Humans , Immunoglobulins , Rats , Spike Glycoprotein, Coronavirus
4.
J Gen Virol ; 102(7)2021 07.
Article in English | MEDLINE | ID: mdl-34236957

ABSTRACT

Mosquito-borne flaviviruses are significant contributors to the arboviral disease burdens both in Australia and globally. While routine arbovirus surveillance remains a vital exercise to identify known flaviviruses in mosquito populations, novel or divergent and emerging species can be missed by these traditional methods. The MAVRIC (monoclonal antibodies to viral RNA intermediates in cells) system is an ELISA-based method for broad-spectrum isolation of positive-sense and double-stranded RNA (dsRNA) viruses based on detection of dsRNA in infected cells. While the MAVRIC ELISA has successfully been used to detect known and novel flaviviruses in Australian mosquitoes, we previously reported that dsRNA could not be detected in dengue virus-infected cells using this method. In this study we identified additional flaviviruses which evade detection of dsRNA by the MAVRIC ELISA. Utilising chimeric flaviviruses we demonstrated that this outcome may be dictated by the non-structural proteins and/or untranslated regions of the flaviviral genome. In addition, we report a modified fixation method that enables improved detection of flavivirus dsRNA and inactivation of non-enveloped viruses from mosquito populations using the MAVRIC system. This study demonstrates the utility of anti-dsRNA monoclonal antibodies for identifying viral replication in insect and vertebrate cell systems and highlights a unique characteristic of flavivirus replication.


Subject(s)
Culicidae/virology , Flavivirus/isolation & purification , Flavivirus/physiology , RNA, Double-Stranded/analysis , RNA, Viral/analysis , Aedes/virology , Animals , Antibodies, Monoclonal , Australia , Cell Line , Dengue Virus/genetics , Dengue Virus/isolation & purification , Dengue Virus/physiology , Enzyme-Linked Immunosorbent Assay , Flavivirus/genetics , RNA, Double-Stranded/immunology , RNA, Viral/immunology , Viral Envelope Proteins/analysis , Viral Envelope Proteins/metabolism , Viral Nonstructural Proteins/analysis , Viral Nonstructural Proteins/metabolism , Virus Replication
5.
PLoS One ; 14(9): e0216013, 2019.
Article in English | MEDLINE | ID: mdl-31513666

ABSTRACT

Prions cause transmissible infectious diseases in humans and animals and have been found to be transmissible by blood transfusion even in the presymptomatic stage. However, the concentration of prions in body fluids such as blood and urine is extremely low; therefore, direct diagnostic tests on such specimens often yield false-negative results. Quantitative preanalytical prion enrichment may significantly improve the sensitivity of prion assays by concentrating trace amounts of prions from large volumes of body fluids. Here, we show that beads conjugated to positively charged peptoids not only captured PrP aggregates from plasma of prion-infected hamsters, but also adsorbed prion infectivity in both the symptomatic and preclinical stages of the disease. Bead absorbed prion infectivity efficiently transmitted disease to transgenic indicator mice. We found that the readout of the peptoid-based misfolded protein assay (MPA) correlates closely with prion infectivity in vivo, thereby validating the MPA as a simple, quantitative, and sensitive surrogate indicator of the presence of prions. The reliable and sensitive detection of prions in plasma will enable a wide variety of applications in basic prion research and diagnostics.


Subject(s)
Biological Assay/methods , Microspheres , Peptoids , Prion Diseases/blood , Prion Diseases/diagnosis , Prions/blood , Animals , Biomarkers , Cerebral Cortex/metabolism , Cricetinae , Disease Models, Animal , Mice , Peptoids/chemistry , Prion Diseases/mortality , Sensitivity and Specificity
6.
Oncotarget ; 9(102): 37700-37714, 2018 Dec 28.
Article in English | MEDLINE | ID: mdl-30701025

ABSTRACT

STRO-001 is a site-specific, predominantly single-species, fully human, aglycosylated anti-CD74 antibody-drug conjugate incorporating a non-cleavable linker-maytansinoid warhead with a drug-antibody ratio of 2 which was produced by a novel cell-free antibody synthesis platform. We examined the potential pharmacodynamics and anti-tumor effects of STRO-001 in multiple myeloma (MM). CD74 expression was assessed in MM cell lines and primary bone marrow (BM) MM biopsies. CD74 mRNA was detectable in CD138+ enriched plasma cells from 100% (892/892) of patients with newly diagnosed MM. Immunohistochemistry confirmed CD74 expression in 35/36 BM biopsies from patients with newly diagnosed and relapsed/refractory MM. Cytotoxicity assays demonstrated nanomolar STRO-001 potency in 4/6 MM cell lines. In ARP-1 and MM.1S tumor-bearing mice, repeat STRO-001 dosing provided significant antitumor activity with eradication of malignant hCD138+ BM plasma cells and prolonged survival. In a luciferase-expressing MM.1S xenograft model, dose-dependent STRO-001 efficacy was confirmed using bioluminescent imaging and BM tumor burden quantification. Consistent with the intended pharmacodynamic effect, STRO-001 induced dose-responsive, reversible B-cell and monocyte depletion in cynomolgus monkeys, up to a maximum tolerated 10 mg/kg, with no evidence of off-target toxicity. Collectively, these data suggest that STRO-001 is a promising therapeutic agent for the treatment of MM.

7.
Blood ; 130(21): 2326-2338, 2017 11 23.
Article in English | MEDLINE | ID: mdl-28978570

ABSTRACT

A number of studies have recently demonstrated that super-enhancers, which are large cluster of enhancers typically marked by a high level of acetylation of histone H3 lysine 27 and mediator bindings, are frequently associated with genes that control and define cell identity during normal development. Super-enhancers are also often enriched at cancer genes in various malignancies. The identification of such enhancers would pinpoint critical factors that directly contribute to pathogenesis. In this study, we performed enhancer profiling using primary leukemia samples from adult T-cell leukemia/lymphoma (ATL), which is a genetically heterogeneous intractable cancer. Super-enhancers were enriched at genes involved in the T-cell activation pathway, including IL2RA/CD25, CD30, and FYN, in both ATL and normal mature T cells, which reflected the origin of the leukemic cells. Super-enhancers were found at several known cancer gene loci, including CCR4, PIK3R1, and TP73, in multiple ATL samples, but not in normal mature T cells, which implicated those genes in ATL pathogenesis. A small-molecule CDK7 inhibitor, THZ1, efficiently inhibited cell growth, induced apoptosis, and downregulated the expression of super-enhancer-associated genes in ATL cells. Furthermore, enhancer profiling combined with gene expression analysis identified a previously uncharacterized gene, TIAM2, that was associated with super-enhancers in all ATL samples, but not in normal T cells. Knockdown of TIAM2 induced apoptosis in ATL cell lines, whereas overexpression of this gene promoted cell growth. Our study provides a novel strategy for identifying critical cancer genes.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Profiling , Genes, Neoplasm , Leukemia-Lymphoma, Adult T-Cell/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Regulation, Leukemic/drug effects , Genetic Association Studies , Humans , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/pathology , Lymphocyte Activation/genetics , Phenylenediamines/pharmacology , Phenylenediamines/therapeutic use , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA Polymerase II/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Cyclin-Dependent Kinase-Activating Kinase
8.
Sci Rep ; 7(1): 3026, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28596531

ABSTRACT

Amber codon suppression for the insertion of non-natural amino acids (nnAAs) is limited by competition with release factor 1 (RF1). Here we describe the genome engineering of a RF1 mutant strain that enhances suppression efficiency during cell-free protein synthesis, without significantly impacting cell growth during biomass production. Specifically, an out membrane protease (OmpT) cleavage site was engineered into the switch loop of RF1, which enables its conditional inactivation during cell lysis. This facilitates extract production without additional processing steps, resulting in a scaleable extract production process. The RF1 mutant extract allows nnAA incorporation at previously intractable sites of an IgG1 and at multiple sites in the same polypeptide chain. Conjugation of cytotoxic agents to these nnAAs, yields homogeneous antibody drug conjugates (ADCs) that can be optimized for conjugation site, drug to antibody ratio (DAR) and linker-warheads designed for efficient tumor killing. This platform provides the means to generate therapeutic ADCs inaccessible by other methods that are efficient in their cytotoxin delivery to tumor with reduced dose-limiting toxicities and thus have the potential for better clinical impact.


Subject(s)
Amino Acids/chemistry , Immunoconjugates , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Protein Engineering , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, Liquid , Codon, Terminator , Drug Stability , Humans , Immunoconjugates/chemistry , Immunoconjugates/isolation & purification , Immunoconjugates/metabolism , Immunoconjugates/pharmacology , Immunoglobulin G/chemistry , Immunoglobulin G/pharmacology , Mass Spectrometry , Models, Molecular , Mutation , Peptide Termination Factors/metabolism , Protein Binding , Protein Conformation , Structure-Activity Relationship , Trastuzumab/chemistry , Trastuzumab/pharmacology
9.
Genes Dev ; 31(23-24): 2343-2360, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29326336

ABSTRACT

The oncogenic transcription factor TAL1/SCL induces an aberrant transcriptional program in T-cell acute lymphoblastic leukemia (T-ALL) cells. However, the critical factors that are directly activated by TAL1 and contribute to T-ALL pathogenesis are largely unknown. Here, we identified AT-rich interactive domain 5B (ARID5B) as a collaborating oncogenic factor involved in the transcriptional program in T-ALL. ARID5B expression is down-regulated at the double-negative 2-4 stages in normal thymocytes, while it is induced by the TAL1 complex in human T-ALL cells. The enhancer located 135 kb upstream of the ARID5B gene locus is activated under a superenhancer in T-ALL cells but not in normal T cells. Notably, ARID5B-bound regions are associated predominantly with active transcription. ARID5B and TAL1 frequently co-occupy target genes and coordinately control their expression. ARID5B positively regulates the expression of TAL1 and its regulatory partners. ARID5B also activates the expression of the oncogene MYC Importantly, ARID5B is required for the survival and growth of T-ALL cells, and forced expression of ARID5B in immature thymocytes results in thymus retention, differentiation arrest, radioresistance, and tumor formation in zebrafish. Our results indicate that ARID5B reinforces the oncogenic transcriptional program by positively regulating the TAL1-induced regulatory circuit and MYC in T-ALL, thereby contributing to T-cell leukemogenesis.


Subject(s)
Carcinogenesis/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Survival/genetics , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Profiling , Genes, myc/genetics , HEK293 Cells , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Protein Binding , Protein Domains/genetics , Thymocytes/metabolism , Thymus Gland/growth & development , Transcription Factors/genetics , Transcriptional Activation/genetics , Zebrafish
10.
PLoS Negl Trop Dis ; 9(3): e0003629, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25799391

ABSTRACT

Mosquito-borne viruses encompass a range of virus families, comprising a number of significant human pathogens (e.g., dengue viruses, West Nile virus, Chikungunya virus). Virulent strains of these viruses are continually evolving and expanding their geographic range, thus rapid and sensitive screening assays are required to detect emerging viruses and monitor their prevalence and spread in mosquito populations. Double-stranded RNA (dsRNA) is produced during the replication of many of these viruses as either an intermediate in RNA replication (e.g., flaviviruses, togaviruses) or the double-stranded RNA genome (e.g., reoviruses). Detection and discovery of novel viruses from field and clinical samples usually relies on recognition of antigens or nucleotide sequences conserved within a virus genus or family. However, due to the wide antigenic and genetic variation within and between viral families, many novel or divergent species can be overlooked by these approaches. We have developed two monoclonal antibodies (mAbs) which show co-localised staining with proteins involved in viral RNA replication in immunofluorescence assay (IFA), suggesting specific reactivity to viral dsRNA. By assessing binding against a panel of synthetic dsRNA molecules, we have shown that these mAbs recognise dsRNA greater than 30 base pairs in length in a sequence-independent manner. IFA and enzyme-linked immunosorbent assay (ELISA) were employed to demonstrate detection of a panel of RNA viruses from several families, in a range of cell types. These mAbs, termed monoclonal antibodies to viral RNA intermediates in cells (MAVRIC), have now been incorporated into a high-throughput, economical ELISA-based screening system for the detection and discovery of viruses from mosquito populations. Our results have demonstrated that this simple system enables the efficient detection and isolation of a range of known and novel viruses in cells inoculated with field-caught mosquito samples, and represents a rapid, sequence-independent, and cost-effective approach to virus discovery.


Subject(s)
Antibodies, Monoclonal/analysis , Culicidae/virology , RNA, Double-Stranded/isolation & purification , RNA, Viral/isolation & purification , Virus Replication/immunology , Animals , Dengue Virus/genetics , Dengue Virus/immunology , Enzyme-Linked Immunosorbent Assay , Flavivirus/genetics , Flavivirus/immunology , Fluorescent Antibody Technique , Humans , RNA Virus Infections/metabolism , RNA Viruses/genetics , RNA Viruses/immunology , RNA, Double-Stranded/genetics , RNA, Viral/genetics , Virus Replication/genetics , West Nile virus/genetics , West Nile virus/immunology
11.
MAbs ; 6(3): 671-8, 2014.
Article in English | MEDLINE | ID: mdl-24517929

ABSTRACT

Antibodies are well-established as therapeutics, and the preclinical and clinical pipeline of these important biologics is growing rapidly. Consequently, there is considerable interest in technologies to engineer and manufacture them. Mammalian cell culture is commonly used for production because eukaryotic expression systems have evolved complex and efficient chaperone systems for the folding of antibodies. However, given the ease and manipulability of bacteria, antibody discovery efforts often employ bacterial expression systems despite their limitations in generating high titers of functional antibody. Open-Cell Free Synthesis (OCFS) is a coupled transcription-translation system that has the advantages of prokaryotic systems while achieving high titers of antibody expression. Due to the open nature of OCFS, it is easily modified by chemical or protein additives to improve the folding of select proteins. As such, we undertook a protein additive screen to identify chaperone proteins that improve the folding and assembly of trastuzumab in OCFS. From the screen, we identified the disulfide isomerase DsbC and the prolyl isomerase FkpA as important positive effectors of IgG folding. These periplasmic chaperones function synergistically for the folding and assembly of IgG, and, when present in sufficient quantities, gram per liter IgG titers can be produced. This technological advancement allows the rapid development and manufacturing of immunoglobulin proteins and pushes OCFS to the forefront of production technologies for biologics.


Subject(s)
Bacteria/genetics , Bacteria/metabolism , Immunoglobulins/biosynthesis , Immunoglobulins/genetics , Molecular Chaperones/metabolism , Antibodies, Monoclonal, Humanized/biosynthesis , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Biotechnology , Cell-Free System , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulins/chemistry , Molecular Chaperones/genetics , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Engineering , Protein Folding , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Trastuzumab
12.
Bioconjug Chem ; 25(2): 351-61, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-24437342

ABSTRACT

Antibody-drug conjugates (ADCs) are a targeted chemotherapeutic currently at the cutting edge of oncology medicine. These hybrid molecules consist of a tumor antigen-specific antibody coupled to a chemotherapeutic small molecule. Through targeted delivery of potent cytotoxins, ADCs exhibit improved therapeutic index and enhanced efficacy relative to traditional chemotherapies and monoclonal antibody therapies. The currently FDA-approved ADCs, Kadcyla (Immunogen/Roche) and Adcetris (Seattle Genetics), are produced by conjugation to surface-exposed lysines, or partial disulfide reduction and conjugation to free cysteines, respectively. These stochastic modes of conjugation lead to heterogeneous drug products with varied numbers of drugs conjugated across several possible sites. As a consequence, the field has limited understanding of the relationships between the site and extent of drug loading and ADC attributes such as efficacy, safety, pharmacokinetics, and immunogenicity. A robust platform for rapid production of ADCs with defined and uniform sites of drug conjugation would enable such studies. We have established a cell-free protein expression system for production of antibody drug conjugates through site-specific incorporation of the optimized non-natural amino acid, para-azidomethyl-l-phenylalanine (pAMF). By using our cell-free protein synthesis platform to directly screen a library of aaRS variants, we have discovered a novel variant of the Methanococcus jannaschii tyrosyl tRNA synthetase (TyrRS), with a high activity and specificity toward pAMF. We demonstrate that site-specific incorporation of pAMF facilitates near complete conjugation of a DBCO-PEG-monomethyl auristatin (DBCO-PEG-MMAF) drug to the tumor-specific, Her2-binding IgG Trastuzumab using strain-promoted azide-alkyne cycloaddition (SPAAC) copper-free click chemistry. The resultant ADCs proved highly potent in in vitro cell cytotoxicity assays.


Subject(s)
Amino Acids/chemistry , Immunoconjugates/chemistry , Cell Line , Cell-Free System , Chromatography, Liquid , High-Throughput Screening Assays , Humans , Immunoconjugates/pharmacology , Tandem Mass Spectrometry
13.
PLoS One ; 8(2): e56534, 2013.
Article in English | MEDLINE | ID: mdl-23460804

ABSTRACT

Recent reports of a novel group of flaviviruses that replicate only in mosquitoes and appear to spread through insect populations via vertical transmission have emerged from around the globe. To date, there is no information on the presence or prevalence of these insect-specific flaviviruses (ISFs) in Australian mosquito species. To assess whether such viruses occur locally, we used reverse transcription-polymerase chain reaction (RT-PCR) and flavivirus universal primers that are specific to the NS5 gene to detect these viruses in mosquito pools collected from the Northern Territory. Of 94 pools of mosquitoes, 13 were RT-PCR positive, and of these, 6 flavivirus isolates were obtained by inoculation of mosquito cell culture. Sequence analysis of the NS5 gene revealed that these isolates are genetically and phylogenetically similar to ISFs reported from other parts of the world. The entire coding region of one isolate (designated 56) was sequenced and shown to have approximately 63.7% nucleotide identity and 66.6% amino acid identity with its closest known relative (Nakiwogo virus) indicating that the prototype Australian ISF represents a new species. All isolates were obtained from Coquillettidia xanthogaster mosquitoes. The new virus is tentatively named Palm Creek virus (PCV) after its place of isolation. We also demonstrated that prior infection of cultured mosquito cells with PCV suppressed subsequent replication of the medically significant West Nile and Murray Valley encephalitis viruses by 10-43 fold (1 to 1.63 log) at 48 hr post-infection, suggesting that superinfection exclusion can occur between ISFs and vertebrate-infecting flaviviruses despite their high level of genetic diversity. We also generated several monoclonal antibodies (mAbs) that are specific to the NS1 protein of PCV, and these represent the first ISF-specific mAbs reported to date.


Subject(s)
Culicidae/virology , Encephalitis Virus, Murray Valley/physiology , Flavivirus Infections/virology , Flavivirus/physiology , Host Specificity/physiology , Virus Replication/physiology , West Nile virus/physiology , Amino Acids/genetics , Animals , Antibodies, Monoclonal/immunology , Antigens, Viral/immunology , Cell Line , Coinfection/virology , Flavivirus/genetics , Flavivirus/growth & development , Flavivirus/isolation & purification , Genome, Viral/genetics , Mice , Northern Territory , Nucleotides/genetics , Phylogeny , Recombinant Proteins/immunology , Sequence Analysis, DNA , Species Specificity , Viral Nonstructural Proteins/immunology , Virion/ultrastructure
14.
J Biol Chem ; 287(23): 18872-87, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22493452

ABSTRACT

Luminescent conjugated polymers (LCPs) interact with ordered protein aggregates and sensitively detect amyloids of many different proteins, suggesting that they may possess antiprion properties. Here, we show that a variety of anionic, cationic, and zwitterionic LCPs reduced the infectivity of prion-containing brain homogenates and of prion-infected cerebellar organotypic cultured slices and decreased the amount of scrapie isoform of PrP(C) (PrP(Sc)) oligomers that could be captured in an avidity assay. Paradoxically, treatment enhanced the resistance of PrP(Sc) to proteolysis, triggered the compaction, and enhanced the resistance to proteolysis of recombinant mouse PrP(23-231) fibers. These results suggest that LCPs act as antiprion agents by transitioning PrP aggregates into structures with reduced frangibility. Moreover, ELISA on cerebellar organotypic cultured slices and in vitro conversion assays with mouse PrP(23-231) indicated that poly(thiophene-3-acetic acid) may additionally interfere with the generation of PrP(Sc) by stabilizing the conformation of PrP(C) or of a transition intermediate. Therefore, LCPs represent a novel class of antiprion agents whose mode of action appears to rely on hyperstabilization, rather than destabilization, of PrP(Sc) deposits.


Subject(s)
Cerebellum/metabolism , Peptide Fragments/metabolism , Polymers/pharmacology , PrPSc Proteins/metabolism , Prions/metabolism , Proteolysis/drug effects , Thiophenes/pharmacology , Animals , Cerebellum/pathology , Mice , PrPSc Proteins/pathogenicity , Prions/pathogenicity , Protein Stability/drug effects , Protein Structure, Tertiary
15.
Biochemistry ; 50(20): 4322-9, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21539296

ABSTRACT

Diseases associated with the misfolding of endogenous proteins, such as Alzheimer's disease and type II diabetes, are becoming increasingly prevalent. The pathophysiology of these diseases is not totally understood, but mounting evidence suggests that the misfolded protein aggregates themselves may be toxic to cells and serve as key mediators of cell death. As such, an assay that can detect aggregates in a sensitive and selective fashion could provide the basis for early detection of disease, before cellular damage occurs. Here we report the evolution of a reagent that can selectively capture diverse misfolded proteins by interacting with a common supramolecular feature of protein aggregates. By coupling this enrichment tool with protein specific immunoassays, diverse misfolded proteins and sub-femtomole amounts of oligomeric aggregates can be detected in complex biological matrices. We anticipate that this near-universal approach for quantitative misfolded protein detection will become a useful research tool for better understanding amyloidogenic protein pathology as well as serve as the basis for early detection of misfolded protein diseases.


Subject(s)
Amyloid/chemistry , Protein Folding , Proteostasis Deficiencies/diagnosis , Amyloid/metabolism , Early Diagnosis , Humans , Indicators and Reagents/chemistry , Indicators and Reagents/metabolism , Molecular Weight , Peptoids/chemistry , Peptoids/metabolism , Protein Multimerization , Protein Structure, Secondary
16.
PLoS One ; 5(2): e9316, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20195363

ABSTRACT

BACKGROUND: Prion diseases are fatal neurodegenerative disorders characterized by misfolding and aggregation of the normal prion protein PrP(C). Little is known about the details of the structural rearrangement of physiological PrP(C) into a still-elusive disease-associated conformation termed PrP(Sc). Increasing evidence suggests that the amino-terminal octapeptide sequences of PrP (huPrP, residues 59-89), though not essential, play a role in modulating prion replication and disease presentation. METHODOLOGY/PRINCIPAL FINDINGS: Here, we report that trypsin digestion of PrP(Sc) from variant and sporadic human CJD results in a disease-specific trypsin-resistant PrP(Sc) fragment including amino acids approximately 49-231, thus preserving important epitopes such as the octapeptide domain for biochemical examination. Our immunodetection analyses reveal that several epitopes buried in this region of PrP(Sc) are exposed in PrP(C). CONCLUSIONS/SIGNIFICANCE: We conclude that the octapeptide region undergoes a previously unrecognized conformational transition in the formation of PrP(Sc). This phenomenon may be relevant to the mechanism by which the amino terminus of PrP(C) participates in PrP(Sc) conversion, and may also be exploited for diagnostic purposes.


Subject(s)
Creutzfeldt-Jakob Syndrome/metabolism , Epitopes/metabolism , PrPC Proteins/metabolism , PrPSc Proteins/metabolism , Blotting, Western , Epitopes/chemistry , Epitopes/genetics , Humans , Molecular Weight , Oligopeptides/chemistry , Oligopeptides/metabolism , PrPC Proteins/chemistry , PrPC Proteins/genetics , PrPSc Proteins/chemistry , PrPSc Proteins/genetics , Protein Conformation , Protein Folding , Repetitive Sequences, Amino Acid , Trypsin/metabolism
17.
PLoS One ; 5(12): e15725, 2010 Dec 30.
Article in English | MEDLINE | ID: mdl-21209907

ABSTRACT

Alzheimer's Disease (AD) is the most prevalent form of dementia worldwide, yet the development of therapeutics has been hampered by the absence of suitable biomarkers to diagnose the disease in its early stages prior to the formation of amyloid plaques and the occurrence of irreversible neuronal damage. Since oligomeric Aß species have been implicated in the pathophysiology of AD, we reasoned that they may correlate with the onset of disease. As such, we have developed a novel misfolded protein assay for the detection of soluble oligomers composed of Aß x-40 and x-42 peptide (hereafter Aß40 and Aß42) from cerebrospinal fluid (CSF). Preliminary validation of this assay with 36 clinical samples demonstrated the presence of aggregated Aß40 in the CSF of AD patients. Together with measurements of total Aß42, diagnostic sensitivity and specificity greater than 95% and 90%, respectively, were achieved. Although larger sample populations will be needed to confirm this diagnostic sensitivity, our studies demonstrate a sensitive method of detecting circulating Aß40 oligomers from AD CSF and suggest that these oligomers could be a powerful new biomarker for the early detection of AD.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Biomarkers/metabolism , Gene Expression Regulation , Peptide Fragments/metabolism , Aged , Amyloid beta-Peptides/cerebrospinal fluid , Brain/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Epitopes/chemistry , Humans , Middle Aged , Models, Chemical , Molecular Conformation , Peptide Fragments/cerebrospinal fluid , Peptides/chemistry , Protein Conformation , Sensitivity and Specificity
18.
Nat Struct Mol Biol ; 15(12): 1255-62, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19011634

ABSTRACT

Folding within the crowded cellular milieu often requires assistance from molecular chaperones that prevent inappropriate interactions leading to aggregation and toxicity. The contribution of individual chaperones to folding the proteome remains elusive. Here we demonstrate that the eukaryotic chaperonin TRiC/CCT (TCP1-ring complex or chaperonin containing TCP1) has broad binding specificity in vitro, similar to the prokaryotic chaperonin GroEL. However, in vivo, TRiC substrate selection is not based solely on intrinsic determinants; instead, specificity is dictated by factors present during protein biogenesis. The identification of cellular substrates revealed that TRiC interacts with folding intermediates of a subset of structurally and functionally diverse polypeptides. Bioinformatics analysis revealed an enrichment in multidomain proteins and regions of beta-strand propensity that are predicted to be slow folding and aggregation prone. Thus, TRiC may have evolved to protect complex protein topologies within its central cavity during biosynthesis and folding.


Subject(s)
Chaperonins/metabolism , Cells, Cultured , Chaperonin Containing TCP-1 , Electrophoresis, Gel, Two-Dimensional , Fibroblasts/metabolism , Humans , Kinetics , Models, Biological , Protein Binding , Protein Structure, Secondary , Proteins/chemistry , Proteins/isolation & purification , Substrate Specificity
19.
Proc Natl Acad Sci U S A ; 104(28): 11551-6, 2007 Jul 10.
Article in English | MEDLINE | ID: mdl-17601775

ABSTRACT

On our initial discovery that prion protein (PrP)-derived peptides were capable of capturing the pathogenic prion protein (PrP(Sc)), we have been interested in how these peptides interact with PrP(Sc). After screening peptides from the entire human PrP sequence, we found two peptides (PrP(19-30) and PrP(100-111)) capable of binding full-length PrP(Sc) in plasma, a medium containing a complex mixture of other proteins including a vast excess of the normal prion protein (PrP(C)). The limit of detection for captured PrP(Sc) was calculated to be 8 amol from a approximately 10(5)-fold dilution of 10% (wt/vol) human variant Creutzfeldt-Jakob disease brain homogenate, with >3,800-fold binding specificity to PrP(Sc) over PrP(C). Through extensive analyses, we show that positively charged amino acids play an important, but not exclusive, role in the interaction between the peptides and PrP(Sc). Neither hydrophobic nor polar interactions appear to correlate with binding activity. The peptide-PrP(Sc) interaction was not sequence-specific, but amino acid composition affected binding. Binding occurs through a conformational domain that is only present in PrP(Sc), is species-independent, and is not affected by proteinase K digestion. These and other findings suggest a mechanism by which cationic domains of PrP(C) may play a role in the recruitment of PrP(C) to PrP(Sc).


Subject(s)
Peptide Fragments/chemical synthesis , PrPC Proteins/chemical synthesis , PrPSc Proteins/chemical synthesis , Antibodies/metabolism , Humans , Immunoglobulin G/metabolism , Microspheres , Peptide Fragments/blood , Peptide Fragments/immunology , PrPC Proteins/blood , PrPC Proteins/classification , PrPC Proteins/immunology , PrPSc Proteins/blood , PrPSc Proteins/classification , PrPSc Proteins/immunology , Prion Diseases/diagnosis , Prion Diseases/immunology , Protein Binding
20.
Cell ; 124(1): 75-88, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16413483

ABSTRACT

Molecular chaperones assist the folding of newly translated and stress-denatured proteins. In prokaryotes, overlapping sets of chaperones mediate both processes. In contrast, we find that eukaryotes evolved distinct chaperone networks to carry out these functions. Genomic and functional analyses indicate that in addition to stress-inducible chaperones that protect the cellular proteome from stress, eukaryotes contain a stress-repressed chaperone network that is dedicated to protein biogenesis. These stress-repressed chaperones are transcriptionally, functionally, and physically linked to the translational apparatus and associate with nascent polypeptides emerging from the ribosome. Consistent with a function in de novo protein folding, impairment of the translation-linked chaperone network renders cells sensitive to misfolding in the context of protein synthesis but not in the context of environmental stress. The emergence of a translation-linked chaperone network likely underlies the elaborate cotranslational folding process necessary for the evolution of larger multidomain proteins characteristic of eukaryotic cells.


Subject(s)
Eukaryotic Cells/physiology , Gene Expression Profiling , Molecular Chaperones/classification , Molecular Chaperones/physiology , Adenosine Triphosphatases , Cytosol/chemistry , Cytosol/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Eukaryotic Cells/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/physiology , Molecular Chaperones/genetics , Oligonucleotide Array Sequence Analysis , Protein Folding , Ribosomes/physiology , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Sensitivity and Specificity , Systems Biology/methods , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...