Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Natl Cancer Inst ; 112(10): 1021-1029, 2020 10 01.
Article in English | MEDLINE | ID: mdl-31922567

ABSTRACT

BACKGROUND: The proportion of tumors of various histologies that may respond to drugs targeted to molecular alterations is unknown. NCI-MATCH, a collaboration between ECOG-ACRIN Cancer Research Group and the National Cancer Institute, was initiated to find efficacy signals by matching patients with refractory malignancies to treatment targeted to potential tumor molecular drivers regardless of cancer histology. METHODS: Trial development required assumptions about molecular target prevalence, accrual rates, treatment eligibility, and enrollment rates as well as consideration of logistical requirements. Central tumor profiling was performed with an investigational next-generation DNA-targeted sequencing assay of alterations in 143 genes, and protein expression of protein expression of phosphatase and tensin homolog, mutL homolog 1, mutS homolog 2, and RB transcriptional corepressor 1. Treatments were allocated with a validated computational platform (MATCHBOX). A preplanned interim analysis evaluated assumptions and feasibility in this novel trial. RESULTS: At interim analysis, accrual was robust, tumor biopsies were safe (<1% severe events), and profiling success was 87.3%. Actionable molecular alteration frequency met expectations, but assignment and enrollment lagged due to histology exclusions and mismatch of resources to demand. To address this lag, we revised estimates of mutation frequencies, increased screening sample size, added treatments, and improved assay throughput and efficiency (93.9% completion and 14-day turnaround). CONCLUSIONS: The experiences in the design and implementation of the NCI-MATCH trial suggest that profiling from fresh tumor biopsies and assigning treatment can be performed efficiently in a large national network trial. The success of such trials necessitates a broad screening approach and many treatment options easily accessible to patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Biopsy , Clinical Trial Protocols as Topic , Clinical Trials, Phase II as Topic , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Molecular Targeted Therapy , Neoplasms/pathology , Precision Medicine , Young Adult
2.
J Clin Oncol ; 38(1): 20-28, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31532724

ABSTRACT

PURPOSE: To assess the efficacy of pembrolizumab in patients with advanced relapsed or refractory mycosis fungoides (MF) or Sézary syndrome (SS). PATIENTS AND METHODS: CITN-10 is a single-arm, multicenter phase II trial of 24 patients with advanced MF or SS. Patients were treated with pembrolizumab 2 mg/kg every 3 weeks for up to 24 months. The primary end point was overall response rate by consensus global response criteria. RESULTS: Patients had advanced-stage disease (23 of 24 with stage IIB to IV MF/SS) and were heavily pretreated with a median of four prior systemic therapies. The overall response rate was 38% with two complete responses and seven partial responses. Of the nine responding patients, six had 90% or more improvement in skin disease by modified Severity Weighted Assessment Tool, and eight had ongoing responses at last follow-up. The median duration of response was not reached, with a median response follow-up time of 58 weeks. Immune-related adverse events led to treatment discontinuation in four patients. A transient worsening of erythroderma and pruritus occurred in 53% of patients with SS. This cutaneous flare reaction did not result in treatment discontinuation for any patient. The flare reaction correlated with high PD-1 expression on Sézary cells but did not associate with subsequent clinical responses or lack of response. Treatment responses did not correlate with expression of PD-L1, total mutation burden, or an interferon-γ gene expression signature. CONCLUSION: Pembrolizumab demonstrated significant antitumor activity with durable responses and a favorable safety profile in patients with advanced MF/SS.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents, Immunological/administration & dosage , Mycosis Fungoides/drug therapy , Sezary Syndrome/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/immunology , Biomarkers, Tumor/metabolism , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Mycosis Fungoides/immunology , Mycosis Fungoides/metabolism , Mycosis Fungoides/pathology , Neoplasm Staging , Recurrence , Sezary Syndrome/immunology , Sezary Syndrome/metabolism , Sezary Syndrome/pathology , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
3.
J Oncol Pract ; : JOP1800092, 2018 Oct 04.
Article in English | MEDLINE | ID: mdl-30285529

ABSTRACT

PURPOSE:: Research biopsy specimens collected in clinical trials often present requirements beyond those of tumor biopsy specimens collected for diagnostic purposes. Research biopsies underpin hypothesis-driven drug development, pharmacodynamic assessment of molecularly targeted anticancer agents, and, increasingly, genomic assessment for precision medicine; insufficient biopsy specimen quality or quantity therefore compromises the scientific value of a study and the resources devoted to it, as well as each patient's contribution to and potential benefit from a clinical trial. METHODS:: To improve research biopsy specimen quality, we consulted with other translational oncology teams and reviewed current best practices. RESULTS:: Among the recommendations were improving communication between oncologists and interventional radiologists, providing feedback on specimen sufficiency, increasing academic recognition and financial support for the time investment required by radiologists to collect and preserve research biopsy specimens, and improving real-time assessment of tissue quality. CONCLUSION:: Implementing these recommendations at the National Cancer Institute's Developmental Therapeutics Clinic has demonstrably improved the quality of biopsy specimens collected; more widespread dissemination of these recommendations beyond large clinical cancer centers is possible and will be of value to the community in improving clinical research and, ultimately, patient care.

4.
J Clin Pharmacol ; 57 Suppl 10: S67-S77, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28921647

ABSTRACT

The National Institutes of Health Clinical Center (NIH CC) is the largest hospital in the United States devoted entirely to clinical research, with a highly diverse spectrum of patients. Patient safety and clinical quality are major goals of the hospital, and therapy is often complicated by multiple cotherapies and comorbidities. To this end, we implemented a pharmacogenomics program in 2 phases. In the first phase, we implemented genotyping for HLA-A and HLA-B gene variations with clinical decision support (CDS) for abacavir, carbamazepine, and allopurinol. In the second phase, we implemented genotyping for drug-metabolizing enzymes and transporters: SLCO1B1 for CDS of simvastatin and TPMT for CDS of mercaptopurine, azathioprine, and thioguanine. The purpose of this review is to describe the implementation process, which involves clinical, laboratory, informatics, and policy decisions pertinent to the NIH CC.


Subject(s)
Biomedical Research/organization & administration , National Institutes of Health (U.S.)/organization & administration , Pharmacogenetics/methods , Decision Support Systems, Clinical , Genotype , Humans , Medical Informatics , Organizational Policy , United States
5.
J Mol Diagn ; 19(2): 313-327, 2017 03.
Article in English | MEDLINE | ID: mdl-28188106

ABSTRACT

The National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH) trial is a national signal-finding precision medicine study that relies on genomic assays to screen and enroll patients with relapsed or refractory cancer after standard treatments. We report the analytical validation processes for the next-generation sequencing (NGS) assay that was tailored for regulatory compliant use in the trial. The Oncomine Cancer Panel assay and the Personal Genome Machine were used in four networked laboratories accredited for the Clinical Laboratory Improvement Amendments. Using formalin-fixed paraffin-embedded clinical specimens and cell lines, we found that the assay achieved overall sensitivity of 96.98% for 265 known mutations and 99.99% specificity. High reproducibility in detecting all reportable variants was observed, with a 99.99% mean interoperator pairwise concordance across the four laboratories. The limit of detection for each variant type was 2.8% for single-nucleotide variants, 10.5% for insertion/deletions, 6.8% for large insertion/deletions (gap ≥4 bp), and four copies for gene amplification. The assay system from biopsy collection through reporting was tested and found to be fully fit for purpose. Our results indicate that the NCI-MATCH NGS assay met the criteria for the intended clinical use and that high reproducibility of a complex NGS assay is achievable across multiple clinical laboratories. Our validation approaches can serve as a template for development and validation of other NGS assays for precision medicine.


Subject(s)
High-Throughput Nucleotide Sequencing , Neoplasms/diagnosis , Neoplasms/genetics , Clinical Trials as Topic , Computational Biology/methods , Genetic Variation , Genomics/methods , Genomics/standards , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Quality Assurance, Health Care , Quality Control , Reproducibility of Results , Sensitivity and Specificity , Workflow
6.
Ann Lab Med ; 36(6): 513-20, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27578503

ABSTRACT

Characterized reference materials (RMs) are needed for clinical laboratory test development and validation, quality control procedures, and proficiency testing to assure their quality. In this article, we review the development and characterization of RMs for clinical molecular genetic tests. We describe various types of RMs and how to access and utilize them, especially focusing on the Genetic Testing Reference Materials Coordination Program (Get-RM) and the Genome in a Bottle (GIAB) Consortium. This review also reinforces the need for collaborative efforts in the clinical genetic testing community to develop additional RMs.


Subject(s)
Genetic Testing/standards , High-Throughput Nucleotide Sequencing/standards , Humans , Public Relations , Quality Control , Reference Values , Sequence Analysis, DNA/standards
7.
Antimicrob Agents Chemother ; 60(10): 5640-8, 2016 10.
Article in English | MEDLINE | ID: mdl-27401576

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) causes large-scale epidemics of acute bacterial skin and skin structure infections (ABSSSI) within communities across the United States. Animal models that reproduce ABSSSI as they occur in humans are urgently needed to test new therapeutic strategies. Alpha-toxin plays a critical role in a variety of staphylococcal infection models in mice, but its role in the pathogenesis of ABSSSI remains to be elucidated in rabbits, which are similar to humans in their susceptibility to S. aureus superantigens and certain bicomponent pore-forming leukocidins. We report here a new rabbit model of ABSSSI and show that those infected with a mutant deficient in expression of alpha-toxin (Δhla) developed a small dermonecrotic lesion, whereas those infected with isogenic USA300 MRSA wild-type or complemented Δhla strains developed ABSSSI that mimic the severe infections that occur in humans, including the large central dermonecrotic core surrounded by erythema, induration, and marked subcutaneous hemorrhage. More importantly, immunoprophylaxis with MEDI4893*, an anti-alpha-toxin human monoclonal antibody, significantly reduced the severity of disease caused by a USA300 wild-type strain to that caused by the Δhla mutant, indicating that this toxin could be completely neutralized during infection. Thus, this study illustrates a potential high standard for the development of new immunotherapeutic agents in which a toxin-neutralizing antibody provides protection to the same degree achieved with a toxin gene knockout. When MEDI4893* was administered as adjunctive therapy with a subtherapeutic dose of linezolid, the combination was significantly more efficacious than either agent alone in reducing the severity of ABSSSI.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bacterial Toxins/immunology , Hemolysin Proteins/immunology , Skin Diseases, Bacterial/microbiology , Staphylococcal Skin Infections/drug therapy , Animals , Antibodies, Monoclonal, Humanized , Bacterial Toxins/genetics , Broadly Neutralizing Antibodies , Disease Models, Animal , Hemolysin Proteins/genetics , Humans , Linezolid/blood , Linezolid/pharmacology , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Pre-Exposure Prophylaxis/methods , Rabbits , Skin Diseases, Bacterial/immunology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology
8.
J Mol Diagn ; 18(1): 51-67, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26602013

ABSTRACT

Robust and analytically validated assays are essential for clinical studies. We outline an analytical validation study of a targeted next-generation sequencing mutation-detection assay used for patient selection in the National Cancer Institute Molecular Profiling-Based Assignment of Cancer Therapy (NCI-MPACT) trial (NCT01827384). Using DNA samples from normal or tumor cell lines and xenografts with known variants, we assessed the sensitivity, specificity, and reproducibility of the NCI-MPACT assay in five variant types: single-nucleotide variants (SNVs), SNVs at homopolymeric (HP) regions (≥3 identical bases), small insertions/deletions (indels), large indels (gap ≥4 bp), and indels at HP regions. The assay achieved sensitivities of 100% for 64 SNVs, nine SNVs at HP regions, and 11 large indels, 83.33% for six indels, and 93.33% for 15 indels at HP regions. Zero false positives (100% specificity) were found in 380 actionable mutation loci in 96 runs of haplotype map cells. Reproducibility analysis showed 96.3% to 100% intraoperator and 98.1% to 100% interoperator mean concordance in detected variants and 100% reproducibility in treatment selection. To date, 38 tumors have been screened, 34 passed preanalytical quality control, and 18 had actionable mutations for treatment assignment. The NCI-MPACT assay is well suited for its intended investigational use and can serve as a template for developing next-generation sequencing assays for other cancer clinical trial applications.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Molecular Diagnostic Techniques/methods , Mutation/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Base Sequence , Biopsy, Large-Core Needle , Cell Line, Tumor , Humans , Patient Selection , Pilot Projects , Plasmids/genetics , Sequence Analysis, DNA
9.
Antimicrob Agents Chemother ; 59(1): 299-309, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25348518

ABSTRACT

Alpha-toxin (AT) is a major virulence determinant in Staphylococcus aureus skin and soft tissue infection models. We previously demonstrated that prophylactic administration of 2A3, an AT-neutralizing monoclonal antibody (MAb), prevents S. aureus disease in a mouse dermonecrosis model by neutralizing AT-mediated tissue necrosis and immune evasion. In the present study, MEDI4893*, an affinity-optimized version of 2A3, was characterized for therapeutic activity in the dermonecrosis model as a single agent and in combination with two frontline antibiotics, vancomycin and linezolid. MEDI4893* postinfection therapy was found to exhibit a therapeutic treatment window similar to that for linezolid but longer than that for vancomycin. Additionally, when combined with either vancomycin or linezolid, MEDI4893* resulted in reduced tissue damage, increased neutrophil and macrophage infiltration and abscess formation, and accelerated healing relative to those with the antibiotic monotherapies. These data suggest that AT neutralization with a potent MAb holds promise for both prophylaxis and adjunctive therapy with antibiotics and may be a valuable addition to currently available options for the treatment of S. aureus skin and soft tissue infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bacterial Toxins/immunology , Hemolysin Proteins/immunology , Staphylococcal Skin Infections/drug therapy , Animals , Anti-Bacterial Agents/pharmacokinetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Broadly Neutralizing Antibodies , Disease Models, Animal , Drug Therapy, Combination , Female , Linezolid/pharmacokinetics , Linezolid/pharmacology , Mice, Inbred BALB C , Necrosis/drug therapy , Necrosis/microbiology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Vancomycin/pharmacokinetics , Vancomycin/pharmacology
10.
Sci Transl Med ; 6(262): 262ra155, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25391481

ABSTRACT

Widespread drug resistance due to empiric use of broad-spectrum antibiotics has stimulated development of bacteria-specific strategies for prophylaxis and therapy based on modern monoclonal antibody (mAb) technologies. However, single-mechanism mAb approaches have not provided adequate protective activity in the clinic. We constructed multifunctional bispecific antibodies, each conferring three mechanisms of action against the bacterial pathogen Pseudomonas aeruginosa by targeting the serotype-independent type III secretion system (injectisome) virulence factor PcrV and persistence factor Psl exopolysaccharide. A new bispecific antibody platform, BiS4, exhibited superior synergistic protection against P. aeruginosa-induced murine pneumonia compared to parent mAb combinations or other available bispecific antibody structures. BiS4αPa was protective in several mouse infection models against disparate P. aeruginosa strains and unexpectedly further synergized with multiple antibiotic classes even against drug-resistant clinical isolates. In addition to resulting in a multimechanistic clinical candidate (MEDI3902) for the prevention or treatment of P. aeruginosa infections, these antibody studies suggest that multifunctional antibody approaches may be a promising platform for targeting other antibiotic-resistant bacterial pathogens.


Subject(s)
Antibodies, Bacterial/therapeutic use , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Pseudomonas Infections/therapy , Pseudomonas aeruginosa/immunology , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/chemistry , Antibodies, Bispecific/chemistry , Antibodies, Monoclonal/chemistry , Antigens, Bacterial/immunology , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Bacterial , Humans , Mice , Molecular Conformation , Phagocytosis , Pseudomonas Infections/immunology
11.
PLoS One ; 8(10): e75103, 2013.
Article in English | MEDLINE | ID: mdl-24098366

ABSTRACT

An optimal host response against Staphylococcus aureus skin and soft tissue infections (SSTI) is dependent on IL-1ß and IL-17 mediated abscess formation. Alpha toxin (AT), an essential virulence factor for SSTI, has been reported to damage tissue integrity; however its effect on the immune response has not been investigated. Here, we demonstrate that infection with USA300 AT isogenic mutant (Δhla), or passive immunization with an AT neutralizing mAb, 2A3, 24 h prior to infection with wild type USA300 (WT), resulted in dermonecrotic lesion size reduction, and robust neutrophil infiltration. Infiltration correlates with increase in proinflammatory cytokines and chemokines, as well as enhanced bacterial clearance relative to immunization with a negative control mAb. In addition, infection with Δhla, or with WT +2A3, resulted in an early influx of innate IL-17(+)γδT cells and a more rapid induction of an adaptive immune response as measured by Th1 and Th17 cell recruitment at the site of infection. These results are the first direct evidence of a role for AT in subverting the innate and adaptive immune responses during a S. aureus SSTI. Further, these effects of AT can be overcome with a high affinity anti-AT mAb resulting in a reduction in disease severity.


Subject(s)
Adaptive Immunity , Bacterial Toxins/metabolism , Immunity, Innate , Skin/microbiology , Skin/pathology , Staphylococcus aureus/metabolism , Animals , Antibodies, Neutralizing/immunology , Bacterial Toxins/immunology , Chemokines/metabolism , Disease Models, Animal , Female , Interleukin-17/metabolism , Mice , Mice, Inbred BALB C , Necrosis/immunology , Skin/immunology , Soft Tissue Infections/immunology , Soft Tissue Infections/microbiology , Staphylococcus aureus/physiology , Th1 Cells/immunology , Th1 Cells/metabolism
12.
J Allergy Clin Immunol ; 132(5): 1086-1096.e5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23866823

ABSTRACT

BACKGROUND: Many asthmatic patients exhibit sputum eosinophilia associated with exacerbations. Benralizumab targets eosinophils by binding IL-5 receptor α, inducing apoptosis through antibody-dependent cell-mediated cytotoxicity. OBJECTIVES: We sought to evaluate the safety of benralizumab in adults with eosinophilic asthma and its effects on eosinophil counts in airway mucosal/submucosal biopsy specimens, sputum, bone marrow, and peripheral blood. METHODS: In this multicenter, double-blind, placebo-controlled phase I study, 13 subjects were randomized to single-dose intravenous placebo or 1 mg/kg benralizumab (day 0; cohort 1), and 14 subjects were randomized to 3 monthly subcutaneous doses of placebo or 100 or 200 mg of benralizumab (days 0, 28, and 56; cohort 2). Cohorts 1 and 2 were consecutive. RESULTS: The incidence of adverse events was similar between groups. No serious adverse events related to benralizumab occurred. In cohort 1 intravenous benralizumab produced a median decrease from baseline of 61.9% in airway mucosal eosinophil counts (day 28; placebo: +19.6%; P = .28), as well as an 18.7% decrease (day 21) in sputum and a 100% decrease (day 28) in blood counts. Eosinophils were not detectable in bone marrow of benralizumab-treated subjects (day 28, n = 4). In cohort 2 subcutaneous benralizumab demonstrated a combined (100 + 200 mg) median reduction of 95.8% in airway eosinophil counts (day 84; placebo, 46.7%; P = .06), as well as an 89.9% decrease (day 28) in sputum and a 100% decrease (day 84) in blood counts. CONCLUSION: Single-dose intravenous and multiple-dose subcutaneous benralizumab reduced eosinophil counts in airway mucosa/submucosa and sputum and suppressed eosinophil counts in bone marrow and peripheral blood. The safety profile supports further development. Additional studies are needed to assess the clinical benefit in asthmatic patients.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/drug therapy , Eosinophils/drug effects , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Sputum/cytology , Adult , Anti-Asthmatic Agents/administration & dosage , Anti-Asthmatic Agents/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Basophils , Bone Marrow/pathology , Eosinophils/immunology , Female , Humans , Leukocyte Count , Male , Middle Aged , Neutrophils , Receptors, Interleukin-5/antagonists & inhibitors , Sputum/immunology , Treatment Outcome , Young Adult
13.
Genes Chromosomes Cancer ; 52(1): 99-106, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22996177

ABSTRACT

Langerhans cell histiocytosis (LCH) is a well-known but rare disease that may occur at any age with markedly variable clinical features: self-regressive, localized, multiorgan, aggressive, or fatal outcome. Congenital LCH is rare and often clinically benign. While LCH is characterized by a clonal proliferation of Langerhans cells, its etiology is unknown. Although BRAF V600E mutations were recently identified as a recurrent genetic alteration in LCH cases, the clinical significance of this mutation within the heterogeneous spectrum of LCH is also currently unknown. We studied a cutaneous, benign form of congenital LCH that occurred in a newborn male, without recurrence for 8 years. Histopathologically, the skin lesion excised after birth showed the typical cytologic and immunophenotypic features of LCH. Sequencing analysis of Exon 15 of the BRAF gene revealed the V600D mutation, with an allelic abundance of 25-30%, corresponding to the LCH cells being hemizygous for the mutant allele. BRAF V600E-specific polymerase chain reaction was negative. Our report is the first to identify the rare, variant BRAF V600D mutation in LCH, and provides support for constitutively activated BRAF oncogene-induced cell senescence as a mechanism of regression in congenital, benign LCH. Further, our clinicopathologic findings provide proof for the first time that the V600D mutation can also occur in the absence of ultraviolet light, and can occur in a clinically benign proliferation, similar to the V600E mutation. Additional clinicopathologic studies in larger numbers of LCH patients may be valuable to ascertain the pathophysiologic role of BRAF mutations in LCH.


Subject(s)
Exons , Histiocytosis, Langerhans-Cell/congenital , Histiocytosis, Langerhans-Cell/genetics , Point Mutation , Proto-Oncogene Proteins B-raf/genetics , Base Sequence , Histiocytosis, Langerhans-Cell/pathology , Histiocytosis, Langerhans-Cell/surgery , Humans , Immunohistochemistry , Infant, Newborn , Male , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Analysis, DNA , Skin Diseases/congenital , Skin Diseases/genetics , Skin Diseases/pathology , Skin Diseases/surgery
14.
J Bacteriol ; 185(4): 1208-17, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12562790

ABSTRACT

The group A Streptococcus (GAS) sof gene encodes the serum opacity factor protein, which is capable of opacifying mammalian sera and binding at least two host proteins, fibronectin and fibrinogen. The sof gene exists in approximately 50% of clinical isolates, and there is a classical association of so-called nephritogenic strains with the opacity factor-positive phenotype. In both a type emm49 strain and a type emm12 strain, the sequences upstream of the 5' end of sof and downstream of the putative terminator were determined to be nearly identical to a region in the M type 1 genome approximately 10 kb upstream of the emm1 gene. This close genetic linkage is likely reflected in the strict correlation of opacity factor phenotype with specific emm genotypes. A new fibronectin-binding protein gene, sfbX, was discovered immediately downstream of sof in emm12 and emm49 strains and in several other sof-positive strains. The sof and sfbX genes were found to be expressed on the same transcription unit, which was correlated with the putative promoter and rho-independant terminator sequences that flank these two genes. The sfbX genes from different emm types are predicted to encode approximately 650-residue surface-bound proteins sharing 89 to 92% sequence identity. SfbX residues approximately 1 to 480 are not highly similar to those of other known proteins, with the closest match being the Staphylococcus aureus coagulase protein. The remaining portions of these proteins (residues 481 to 650) contain four putative fibronectin-binding repeats highly similar to those of other streptococcal fibronectin-binding proteins and a potential LP(X)SG cell wall anchor motif. Targeted in-frame allelic-exchange mutagenesis, complementation, and heterologous-expression studies found that serum opacification is encoded by sof alone and that sfbX encodes a fibronectin-binding function. A recombinant SfbX protein was found to bind immobilized fibronectin and to partially inhibit GAS adherence to fibronectin. The sfbX gene was found to be present only in sof-positive strains, and together these genes could influence the spectrum of tissues colonized by sof-positive GAS.


Subject(s)
Bacterial Proteins/genetics , Carrier Proteins/genetics , Fibronectins/metabolism , Operon , Peptide Hydrolases/genetics , Streptococcus pyogenes/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Chromosome Mapping , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Sequence Analysis, DNA , Streptococcus pyogenes/metabolism , Transcription, Genetic
15.
Lancet ; 359(9301): 124-9, 2002 Jan 12.
Article in English | MEDLINE | ID: mdl-11809255

ABSTRACT

BACKGROUND: We encountered three patients with severe necrotising soft tissue infections due to beta-haemolytic group G streptococcus. Due to strong clinical similarities with invasive infections produced by group A streptococcus, we investigated a potential link of shared beta-haemolytic phenotype to disease pathogenesis. METHODS: Hybridisation, DNA sequencing, targeted mutagenesis, and complementation studies were used to establish the genetic basis for group G streptococcus beta-haemolytic activity. The requirement of group G streptococcus beta-haemolysin in producing necrotising infection was examined in mice. FINDINGS: Each patient had an underlying medical condition. beta-haemolytic group G streptococcus was the sole microbial isolate from debrided necrotic tissue. The group G streptococcus chromosome contained a homologue of the nine-gene group A streptococcus sag operon encoding the beta-haemolysin streptolysin S (SLS). Targeted mutagenesis of the putative SLS structural gene sagA in group G streptococcus eliminated beta-haemolytic activity. Mice injected subcutaneously with wild-type group A streptococcus or group G streptococcus developed an inflammatory lesion with high bacterial counts, marked neutrophil infiltration, and histopathological evidence of diffuse tissue necrosis. These changes were not found in mice injected with the isogenic group A streptococcus or group G streptococcus SLS-negative mutants. INTERPRETATION: In patients with underlying medical conditions, beta-haemolytic group G streptococcus can produce necrotising soft tissue infections resembling those produced by group A streptococcus. The beta-haemolytic phenotype of group G streptococcus is produced by the exotoxin SLS, encoded by a functional homologue of the nine-gene group A streptococcus sag operon. SLS expression contributes to the pathogenesis of streptococcal necrotising soft tissue infection.


Subject(s)
Bacterial Proteins , Fasciitis, Necrotizing/microbiology , Streptococcus pyogenes/classification , Streptococcus pyogenes/genetics , Streptococcus/classification , Streptococcus/genetics , Streptolysins/antagonists & inhibitors , Streptolysins/physiology , Animals , Anti-Bacterial Agents/therapeutic use , Combined Modality Therapy , DNA Transposable Elements/genetics , DNA, Bacterial/genetics , Debridement , Disease Models, Animal , Electrophoresis, Gel, Pulsed-Field , Fasciitis, Necrotizing/pathology , Fasciitis, Necrotizing/therapy , Humans , Male , Mice , Middle Aged , Mutagenesis/genetics , Operon/genetics , Phenotype , Recombination, Genetic/genetics , Serotyping , Streptococcus/pathogenicity , Streptococcus pyogenes/pathogenicity , Transformation, Bacterial/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...