1
|
Dennis M, Charlton M, Elam S, Wolfensohn S. Facility design and management. Strategies for high level biocontainment. Lab Anim 2024:236772241247104. [PMID: 39118535 DOI: 10.1177/00236772241247104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
UK Health Security Agency is required to investigate the pathogenesis of emerging or re-emerging infections and to test novel interventions, such as vaccines and therapeutics against these and other diseases, such as tuberculosis and Ebola, that have a significant impact on human health world-wide. Research into the causative agents (mainly BSL 3 and 4) using a wide range of animal species as pre-clinical models brings a number of challenges in terms of effective biocontainment to address human safety whilst optimising delivery of scientific objectives and the welfare of the animals. Here we describe the strategies used for high containment of species that include mice, ferrets, hamsters, rabbits and macaques that have been infected with high consequence pathogens. To ensure relevance of these models we frequently challenge by the aerosol route and monitor the development of disease and protective or therapeutic efficacy by methodologies similar to those used in the clinic. We have devised methods of sampling that can inform on pathogenesis and immune function that include lung lavage and medical imaging such as computed tomography and positron emission tomography-computed tomography. Imaging assists our assessment of progression to disease whilst providing refinement in application of early humane endpoints. We have developed directional flow containment systems that provide quantifiable operator protection whilst allowing group housing and a wide range of enrichment strategies appropriate for each species. Furthermore, we have demonstrated our improvements in animal welfare through use of a software-based Animal Welfare Assessment Grid that was developed with help of NC3Rs funding and enables us to quantify the lifetime experience of animals.
Collapse
Affiliation(s)
- Mike Dennis
- UK Health Security Agency Porton, Vaccine Evaluation Department, Salisbury, UK
| | - Michael Charlton
- UK Health Security Agency, High Containment and Biological Services, Salisbury, UK
| | - Samantha Elam
- UK Health Security Agency, High Security and Biological Services, Salisbury, UK
| | | |
Collapse
|
2
|
Tucker EW, Ruiz-Bedoya CA, Mota F, Erice C, Kim J, de Jesus P, Jahdav R, Bahr M, Flavahan K, Chen X, Peloquin CA, Freundlich JS, Jain SK. Linezolid does not improve bactericidal activity of rifampin-containing first-line regimens in animal models of TB meningitis. Int J Antimicrob Agents 2024; 63:107048. [PMID: 38061419 PMCID: PMC10841818 DOI: 10.1016/j.ijantimicag.2023.107048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/15/2023] [Accepted: 11/28/2023] [Indexed: 01/02/2024]
Abstract
Tuberculous meningitis (TB meningitis) is the most devastating form of tuberculosis (TB) and there is a critical need to optimize treatment. Linezolid is approved for multidrug resistant TB and has shown encouraging results in retrospective TB meningitis studies, with several clinical trials underway assessing its additive effects on high-dose (35 mg/kg/day) or standard-dose (10 mg/kg/day) rifampin-containing regimens. However, the efficacy of adjunctive linezolid to rifampin-containing first-line TB meningitis regimens and the tissue pharmacokinetics (PK) in the central nervous system (CNS) are not known. We therefore conducted cross-species studies in two mammalian (rabbits and mice) models of TB meningitis to test the efficacy of linezolid when added to the first-line TB regimen and measure detailed tissue PK (multicompartmental positron emission tomography [PET] imaging and mass spectrometry). Addition of linezolid did not improve the bactericidal activity of the high-dose rifampin-containing regimen in either animal model. Moreover, the addition of linezolid to standard-dose rifampin in mice also did not improve its efficacy. Linezolid penetration (tissue/plasma) into the CNS was compartmentalized with lower than previously reported brain and cerebrospinal fluid (CSF) penetration, which decreased further two weeks after initiation of treatment. These results provide important data regarding the addition of linezolid for the treatment of TB meningitis.
Collapse
Affiliation(s)
- Elizabeth W Tucker
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Camilo A Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Filipa Mota
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clara Erice
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Kim
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patricia de Jesus
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ravindra Jahdav
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Melissa Bahr
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kelly Flavahan
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xueyi Chen
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles A Peloquin
- Infectious Disease Pharmacokinetics Laboratory, Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL, USA
| | - Joel S Freundlich
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Hammoud DA, Clifford Lane H, Jain SK. Molecular Imaging of Infections: Advancing the Search for the Hidden Enemy. J Infect Dis 2023; 228:S233-S236. [PMID: 37788496 PMCID: PMC10547366 DOI: 10.1093/infdis/jiad079] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023] Open
Abstract
Even before the coronavirus disease 2019 pandemic, infections were a major threat to human health, as the third leading cause of death and the leading cause of morbidity among all human diseases. Although conventional imaging studies are routinely used for patients with infections, they provide structural or anatomic information only. Molecular imaging technologies enable noninvasive visualization of molecular processes at the cellular level within intact living subjects, including patients, and hold great potential for infections. We hope that this supplement will spur interest in the field and establish new collaborations to develop and translate novel molecular imaging approaches to the clinic.
Collapse
Affiliation(s)
- Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, NIH Clinical Center, Bethesda, Maryland, USA
| | - H Clifford Lane
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Singh SB, Bhandari S, Siwakoti S, Bhatta R, Raynor WY, Werner TJ, Alavi A, Hess S, Revheim ME. Is Imaging Bacteria with PET a Realistic Option or an Illusion? Diagnostics (Basel) 2023; 13:diagnostics13071231. [PMID: 37046449 PMCID: PMC10093025 DOI: 10.3390/diagnostics13071231] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/14/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
The application of [18F]-fluorodeoxyglucose ([18F]FDG) as a radiotracer to detect sites of inflammation (either due to bacterial infection or primary inflammation) has led to exploring the role of PET in visualizing bacteria directly at sites of infection. However, the results from such efforts are controversial and inconclusive so far. We aimed to assess the limitations of PET as an effective modality in the diagnosis of bacterial infections. Inflammation due to bacterial infections can be visualized by using [18F]FDG-PET. However, the non-specificity of [18F]FDG makes it undesirable to visualize bacteria as the underlying cause of inflammation. Hence, more specific radiotracers that possibly bind to or accumulate in bacteria-specific receptors or enzymes are being explored. Several radiotracers, including 2-deoxy-2-[18F]fluorosorbitol ([18F]FDS), 6-[18F]-fluoromaltose, [11C]para-aminobenzoic acid ([11C]PABA), radiolabeled trimethoprim (11C-TMP) and its analog fluoropropyl-trimethoprim (18F-FPTMP), other radiolabeled sugars, and antimicrobial drugs have been used to image microorganisms. Unfortunately, no progress has been made in translating the results to routine human use; feasibility and other factors have constrained their success in clinical settings. In the current article, we discuss the limitations of direct bacterial visualization with PET tracers, but emphasize the important role of [18F]FDG-PET as the only option for detecting evidence of infection.
Collapse
Affiliation(s)
- Shashi B Singh
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Sadikshya Bhandari
- Kathmandu University School of Medical Sciences, Dhulikhel Hospital, Dhulikhel 45200, Nepal
| | - Shisir Siwakoti
- Kathmandu University School of Medical Sciences, Dhulikhel Hospital, Dhulikhel 45200, Nepal
| | - Rabi Bhatta
- Universal College of Medical Sciences, Bhairahawa 32900, Nepal
| | - William Y Raynor
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, 1 Robert Wood Johnson Place, MEB #404, New Brunswick, NJ 08901, USA
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Soren Hess
- Department of Radiology and Nuclear Medicine, Hospital Southwest Jutland, 6700 Esbjerg, Denmark
- Department of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, 5230 Odense, Denmark
| | - Mona-Elisabeth Revheim
- The Intervention Center, Division of Technology and Innovation, Oslo University Hospital, 0424 Oslo, Norway
- Division for Radiology and Nuclear Medicine, Oslo University Hospital, 0424 Oslo, Norway
- Norway and Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0315 Oslo, Norway
| |
Collapse
|
5
|
Foss CA, Ordonez AA, Naik R, Das D, Hall A, Wu Y, Dannals RF, Jain SK, Pomper MG, Horti AG. PET/CT imaging of CSF1R in a mouse model of tuberculosis. Eur J Nucl Med Mol Imaging 2022; 49:4088-4096. [PMID: 35713665 PMCID: PMC9922090 DOI: 10.1007/s00259-022-05862-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/03/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE Macrophages represent an essential means of sequestration and immune evasion for Mycobacterium tuberculosis. Pulmonary tuberculosis (TB) is characterized by dense collections of tissue-specific and recruited macrophages, both of which abundantly express CSF1R on their outer surface. 4-Cyano-N-(5-(1-(dimethylglycyl)piperidin-4-yl)-2',3',4',5'-tetrahydro-[1,1'-biphenyl]-2-yl)-1H-imidazole-2-carboxamide (JNJ-28312141) is a reported high affinity, CSF1R-selective antagonist. We report the radiosynthesis of 4-cyano-N-(5-(1-(N-methyl-N-([11C]methyl)glycyl)piperidin-4-yl)-2',3',4',5'-tetrahydro-[1,1'-biphenyl]-2-yl)-1H-imidazole-2-carboxamide ([11C]JNJ-28312141) and non-invasive detection of granulomatous and diffuse lesions in a mouse model of TB using positron emission tomography (PET). METHODS Nor-methyl-JNJ-28312141 precursor was radiolabeled with [11C]iodomethane to produce [11C]JNJ-28312141. PET/CT imaging was performed in the C3HeB/FeJ murine model of chronic pulmonary TB to co-localize radiotracer uptake with granulomatous lesions observed on CT. Additionally, CSF1R, Iba1 fluorescence immunohistochemistry was performed to co-localize CSF1R target with reactive macrophages in infected and healthy mice. RESULTS Radiosynthesis of [11C]JNJ-28312141 averaged a non-decay-corrected yield of 18.7 ± 2.1%, radiochemical purity of 99%, and specific activity averaging 658 ± 141 GBq/µmol at the end-of-synthesis. PET/CT imaging in healthy mice showed hepatobiliary [13.39-25.34% ID/g, percentage of injected dose per gram of tissue (ID/g)] and kidney uptake (12.35% ID/g) at 40-50 min post-injection. Infected mice showed focal pulmonary lesion uptake (5.58-12.49% ID/g), hepatobiliary uptake (15.30-40.50% ID/g), cervical node uptake, and renal uptake (11.66-29.33% ID/g). The ratio of infected lesioned lung/healthy lung uptake is 5.91:1, while the ratio of lesion uptake to adjacent infected radiolucent lung is 2.8:1. Pre-administration of 1 mg/kg of unlabeled JNJ-28312141 with [11C]JNJ-28312141 in infected animals resulted in substantial blockade. Fluorescence microscopy of infected and uninfected whole lung sections exclusively co-localized CSF1R staining with abundant Iba1 + macrophages. Healthy lung exhibited no CSF1R staining and very few Iba1 + macrophages. CONCLUSION [11C]JNJ-28312141 binds specifically to CSF1R + macrophages and delineates granulomatous foci of disease in a murine model of pulmonary TB.
Collapse
Affiliation(s)
- Catherine A Foss
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA.
- Department of Pediatrics, Center for Infection and Inflammation Imaging Research, Baltimore, MD, USA.
| | - Alvaro A Ordonez
- Department of Pediatrics, Center for Infection and Inflammation Imaging Research, Baltimore, MD, USA
| | - Ravi Naik
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Deepankar Das
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew Hall
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Yunkou Wu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Robert F Dannals
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Sanjay K Jain
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
- Department of Pediatrics, Center for Infection and Inflammation Imaging Research, Baltimore, MD, USA
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew G Horti
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
6
|
Woong Yoo S, Young Kwon S, Kang SR, Min JJ. Molecular imaging approaches to facilitate bacteria-mediated cancer therapy. Adv Drug Deliv Rev 2022; 187:114366. [PMID: 35654213 DOI: 10.1016/j.addr.2022.114366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 12/14/2022]
Abstract
Bacteria-mediated cancer therapy is a potential therapeutic strategy for cancer that has unique properties, including broad tumor-targeting ability, various administration routes, the flexibility of delivery, and facilitating the host's immune responses. The molecular imaging of bacteria-mediated cancer therapy allows the therapeutically injected bacteria to be visualized and confirms the accurate delivery of the therapeutic bacteria to the target lesion. Several hurdles make bacteria-specific imaging challenging, including the need to discriminate therapeutic bacterial infection from inflammation or other pathologic lesions. To realize the full potential of bacteria-specific imaging, it is necessary to develop bacteria-specific targets that can be associated with an imaging assay. This review describes the current status of bacterial imaging techniques together with the advantages and disadvantages of several imaging modalities. Also, we describe potential targets for bacterial-specific imaging and related applications.
Collapse
Affiliation(s)
- Su Woong Yoo
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Hwasun, Jeonnam, Korea
| | - Seong Young Kwon
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Hwasun, Jeonnam, Korea; Department of Nuclear Medicine, Chonnam National University Medical School, Hwasun, Jeonnam, Korea
| | - Sae-Ryung Kang
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Hwasun, Jeonnam, Korea
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Hwasun, Jeonnam, Korea; Department of Nuclear Medicine, Chonnam National University Medical School, Hwasun, Jeonnam, Korea.
| |
Collapse
|
7
|
Vorster M, Sathekge MM. Positron Emission Tomography (PET) Imaging in Tuberculosis. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00097-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
8
|
More S, Marakalala MJ, Sathekge M. Tuberculosis: Role of Nuclear Medicine and Molecular Imaging With Potential Impact of Neutrophil-Specific Tracers. Front Med (Lausanne) 2021; 8:758636. [PMID: 34957144 PMCID: PMC8703031 DOI: 10.3389/fmed.2021.758636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/03/2021] [Indexed: 01/02/2023] Open
Abstract
With Tuberculosis (TB) affecting millions of people worldwide, novel imaging modalities and tools, particularly nuclear medicine and molecular imaging, have grown with greater interest to assess the biology of the tuberculous granuloma and evolution thereof. Much early work has been performed at the pre-clinical level using gamma single photon emission computed tomography (SPECT) agents exploiting certain characteristics of Mycobacterium tuberculosis (MTb). Both antituberculous SPECT and positron emission tomography (PET) agents have been utilised to characterise MTb. Other PET tracers have been utilised to help to characterise the biology of MTb (including Gallium-68-labelled radiopharmaceuticals). Of all the tracers, 2-[18F]FDG has been studied extensively over the last two decades in many aspects of the treatment paradigm of TB: at diagnosis, staging, response assessment, restaging, and in potentially predicting the outcome of patients with latent TB infection. Its lower specificity in being able to distinguish different inflammatory cell types in the granuloma has garnered interest in reviewing more specific agents that can portend prognostic implications in the management of MTb. With the neutrophil being a cell type that portends this poorer prognosis, imaging this cell type may be able to answer more accurately questions relating to the tuberculous granuloma transmissivity and may help in characterising patients who may be at risk of developing active TB. The formyl peptide receptor 1(FPR1) expressed by neutrophils is a key marker in this process and is a potential target to characterise these areas. The pre-clinical work regarding the role of radiolabelled N-cinnamoyl –F-(D) L – F – (D) –L F (cFLFLF) (which is an antagonist for FPR1) using Technetium 99m-labelled conjugates and more recently radiolabelled with Gallium-68 and Copper 64 is discussed. It is the hope that further work with this tracer may accelerate its potential to be utilised in responding to many of the current diagnostic dilemmas and challenges in TB management, thereby making the tracer a translatable option in routine clinical care.
Collapse
Affiliation(s)
- Stuart More
- Division of Nuclear Medicine, Department of Radiation Medicine, University of Cape Town, Cape Town, South Africa
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Nuclear Medicine Research Infrastructure, Steve Biko Academic Hospital, Pretoria, South Africa
- *Correspondence: Stuart More
| | - Mohlopheni J. Marakalala
- Africa Health Research Institute, Durban, South Africa
- Division of Infection and Immunity, University College London, London, United Kingdom
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Michael Sathekge
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Nuclear Medicine Research Infrastructure, Steve Biko Academic Hospital, Pretoria, South Africa
| |
Collapse
|
9
|
Skaar EP. Imaging Infection Across Scales of Size: From Whole Animals to Single Molecules. Annu Rev Microbiol 2021; 75:407-426. [PMID: 34343016 DOI: 10.1146/annurev-micro-041521-121457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Infectious diseases are a leading cause of global morbidity and mortality, and the threat of infectious diseases to human health is steadily increasing as new diseases emerge, existing diseases reemerge, and antimicrobial resistance expands. The application of imaging technology to the study of infection biology has the potential to uncover new factors that are critical to the outcome of host-pathogen interactions and to lead to innovations in diagnosis and treatment of infectious diseases. This article reviews current and future opportunities for the application of imaging to the study of infectious diseases, with a particular focus on the power of imaging objects across a broad range of sizes to expand the utility of these approaches. Expected final online publication date for the Annual Review of Microbiology, Volume 75 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Eric P Skaar
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA;
| |
Collapse
|
10
|
Ordonez AA, Tucker EW, Anderson CJ, Carter CL, Ganatra S, Kaushal D, Kramnik I, Lin PL, Madigan CA, Mendez S, Rao J, Savic RM, Tobin DM, Walzl G, Wilkinson RJ, Lacourciere KA, Via LE, Jain SK. Visualizing the dynamics of tuberculosis pathology using molecular imaging. J Clin Invest 2021; 131:145107. [PMID: 33645551 PMCID: PMC7919721 DOI: 10.1172/jci145107] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Nearly 140 years after Robert Koch discovered Mycobacterium tuberculosis, tuberculosis (TB) remains a global threat and a deadly human pathogen. M. tuberculosis is notable for complex host-pathogen interactions that lead to poorly understood disease states ranging from latent infection to active disease. Additionally, multiple pathologies with a distinct local milieu (bacterial burden, antibiotic exposure, and host response) can coexist simultaneously within the same subject and change independently over time. Current tools cannot optimally measure these distinct pathologies or the spatiotemporal changes. Next-generation molecular imaging affords unparalleled opportunities to visualize infection by providing holistic, 3D spatial characterization and noninvasive, temporal monitoring within the same subject. This rapidly evolving technology could powerfully augment TB research by advancing fundamental knowledge and accelerating the development of novel diagnostics, biomarkers, and therapeutics.
Collapse
Affiliation(s)
- Alvaro A. Ordonez
- Center for Infection and Inflammation Imaging Research
- Center for Tuberculosis Research
- Department of Pediatrics, and
| | - Elizabeth W. Tucker
- Center for Infection and Inflammation Imaging Research
- Center for Tuberculosis Research
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Claire L. Carter
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, New Jersey, USA
| | - Shashank Ganatra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Igor Kramnik
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusets, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Philana L. Lin
- Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Cressida A. Madigan
- Department of Biological Sciences, UCSD, San Diego, La Jolla, California, USA
| | - Susana Mendez
- National Institute of Allergy and Infectious Diseases (NIAID), NIH, Rockville, Maryland, USA
| | - Jianghong Rao
- Molecular Imaging Program at Stanford, Department of Radiology and Chemistry, Stanford University, Stanford, California, USA
| | - Rada M. Savic
- Department of Bioengineering and Therapeutic Sciences, School of Pharmacy and Medicine, UCSF, San Francisco, California, USA
| | - David M. Tobin
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Gerhard Walzl
- SAMRC Centre for Tuberculosis Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Robert J. Wilkinson
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
- Wellcome Centre for Infectious Diseases Research in Africa and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- The Francis Crick Institute, London, United Kingdom
| | - Karen A. Lacourciere
- National Institute of Allergy and Infectious Diseases (NIAID), NIH, Rockville, Maryland, USA
| | - Laura E. Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, and Tuberculosis Imaging Program, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Sanjay K. Jain
- Center for Infection and Inflammation Imaging Research
- Center for Tuberculosis Research
- Department of Pediatrics, and
| |
Collapse
|
11
|
Jacobs AH, Schelhaas S, Viel T, Waerzeggers Y, Winkeler A, Zinnhardt B, Gelovani J. Imaging of Gene and Cell-Based Therapies: Basis and Clinical Trials. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00060-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
12
|
Ramakrishna C, Mendonca S, Ruegger PM, Kim JH, Borneman J, Cantin EM. Herpes simplex virus infection, Acyclovir and IVIG treatment all independently cause gut dysbiosis. PLoS One 2020; 15:e0237189. [PMID: 32760124 PMCID: PMC7410316 DOI: 10.1371/journal.pone.0237189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
Herpes simplex virus 1 (HSV) is a ubiquitous human virus resident in a majority of the global population as a latent infection. Acyclovir (ACV), is the standard of care drug used to treat primary and recurrent infections, supplemented in some patients with intravenous immunoglobulin (IVIG) treatment to suppress infection and deleterious inflammatory responses. As many diverse medications have recently been shown to change composition of the gut microbiome, we used Illumina 16S rRNA gene sequencing to determine the effects of ACV and IVIG on the gut bacterial community. We found that HSV, ACV and IVIG can all independently disrupt the gut bacterial community in a sex biased manner when given to uninfected C57BL/6 mice. Treatment of HSV infected mice with ACV or IVIG alone or together revealed complex interactions between these drugs and infection that caused pronounced sex biased dysbiosis. ACV reduced Bacteroidetes levels in male but not female mice, while levels of the Anti-inflammatory Clostridia (AIC) were reduced in female but not male mice, which is significant as these taxa are associated with protection against the development of graft versus host disease (GVHD) in hematopoietic stem cell transplant (HSCT) patients. Gut barrier dysfunction is associated with GVHD in HSCT patients and ACV also decreased Akkermansia muciniphila, which is important for maintaining gut barrier functionality. Cumulatively, our data suggest that long-term prophylactic ACV treatment of HSCT patients may contribute to GVHD and also potentially impact immune reconstitution. These data have important implications for other clinical settings, including HSV eye disease and genital infections, where ACV is given long-term.
Collapse
Affiliation(s)
- Chandran Ramakrishna
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Stacee Mendonca
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, California, United States of America
| | - Paul M. Ruegger
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, United States of America
| | - Jane Hannah Kim
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, United States of America
| | - James Borneman
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, United States of America
- * E-mail: (EMC); (JB)
| | - Edouard M. Cantin
- Department of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, California, United States of America
- * E-mail: (EMC); (JB)
| |
Collapse
|
13
|
Foss CA, Kulik L, Ordonez AA, Jain SK, Michael Holers V, Thurman JM, Pomper MG. SPECT/CT Imaging of Mycobacterium tuberculosis Infection with [ 125I]anti-C3d mAb. Mol Imaging Biol 2020; 21:473-481. [PMID: 29998399 DOI: 10.1007/s11307-018-1228-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Diagnosis and therapeutic monitoring of chronic bacterial infection requires methods to detect and localize sites of infection accurately. Complement C3 activation fragments are generated and covalently bound to selective bacterial pathogens during the immune response and can serve as biomarkers of ongoing bacterial infection. We have developed several probes for detecting tissue-bound C3 deposits, including a monoclonal antibody (mAb 3d29) that recognizes the tissue-bound terminal processing fragments iC3b and C3d but does not recognize native circulating C3 or tissue-bound C3b. PROCEDURES To determine whether mAb 3d29 could be used to detect chronic Mycobacterium tuberculosis infection non-invasively, aerosol-infected female C3HeB/FeJ mice were injected with [125I]3d29 mAb and either imaged using single-photon emission computed tomography (SPECT)/X-ray computed tomography (CT) imaging at 24 and 48 h after radiotracer injection or being subjected to biodistribution analysis. RESULTS Discrete lesions were detected by SPECT/CT imaging in the lungs and spleens of infected mice, consistent with the location of granulomas in the infected animals as detected by CT. Low-level signal was seen in the spleens of uninfected mice and no signal was seen in the lungs of healthy mice. Immunofluorescence microscopy revealed that 3d29 in the lungs of infected mice co-localized with aggregates of macrophages (detected with anti-CD68 antibodies). 3d29 was detected in the cytoplasm of macrophages, consistent with the location of internalized M. tuberculosis. 3d29 was also present within alveolar epithelial cells, indicating that it detected M. tuberculosis phagocytosed by other CD68-positive cells. Healthy controls showed very little retention of fluorescent or radiolabeled antibody across tissues. Radiolabeled 3d29 compared with radiolabeled isotype control showed a 3.5:1 ratio of increased uptake in infected lungs, indicating specific uptake by 3d29. CONCLUSION 3d29 can be used to detect and localize areas of infection with M. tuberculosis non-invasively by 24 h after radiotracer injection and with high contrast.
Collapse
Affiliation(s)
- Catherine A Foss
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, 1550 Orleans St. CRB2 493, Baltimore, MD, 21228, USA. .,Center for Infection and Inflammation Imaging Research, Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21228, USA.
| | - Liudmila Kulik
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21228, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21228, USA
| | - V Michael Holers
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Joshua M Thurman
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, 1550 Orleans St. CRB2 493, Baltimore, MD, 21228, USA.,Center for Infection and Inflammation Imaging Research, Department of Pediatrics, Johns Hopkins University, Baltimore, MD, 21228, USA
| |
Collapse
|
14
|
Rapid Tuberculosis Diagnosis Using Reporter Enzyme Fluorescence. J Clin Microbiol 2019; 57:JCM.01462-19. [PMID: 31511338 PMCID: PMC6879286 DOI: 10.1128/jcm.01462-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022] Open
Abstract
Tuberculosis is the most frequent cause of death in humans from a single infectious agent. Due to low numbers of bacteria present in sputum during early infection, diagnosis does not usually occur until >3 to 4 months after symptoms develop. We created a new more sensitive diagnostic that can be carried out in 10 min with no processing or technical expertise. Tuberculosis is the most frequent cause of death in humans from a single infectious agent. Due to low numbers of bacteria present in sputum during early infection, diagnosis does not usually occur until >3 to 4 months after symptoms develop. We created a new more sensitive diagnostic that can be carried out in 10 min with no processing or technical expertise. This assay utilizes the Mycobacterium tuberculosis-specific biomarker BlaC in reporter enzyme fluorescence (REF) that has been optimized for clinical samples, designated REFtb, along with a more specific fluorogenic substrate, CDG-3. We report the first evaluation of clinical specimens with REFtb assays in comparison to the gold standards for tuberculosis diagnosis, culture and smear microscopy. REFtb assays allowed diagnosis of 160 patients from 16 different countries with a sensitivity of 89% for smear-positive, culture-positive samples and 88% for smear-negative, culture-positive samples with a specificity of 82%. The negative predictive value of REFtb for tuberculosis infection is 93%, and the positive predictive value is 79%. Overall, these data point toward the need for larger accuracy studies by third parties using a commercially available REFtb kit to determine whether incorporation of REFtb into the clinical toolbox for suspected tuberculosis patients would improve case identification. If results similar to our own can be obtained by all diagnostic laboratories, REFtb would allow proper treatment of more than 85% of patients that would be missed during their initial visit to a clinic using current diagnostic strategies, reducing the potential for further spread of disease.
Collapse
|
15
|
Abstract
Molecular imaging is an emerging technology that enables the noninvasive visualization, characterization, and quantification of molecular events within living subjects. Positron emission tomography (PET) is a clinically available molecular imaging tool with significant potential to study pathogenesis of infections in humans. Molecular imaging is an emerging technology that enables the noninvasive visualization, characterization, and quantification of molecular events within living subjects. Positron emission tomography (PET) is a clinically available molecular imaging tool with significant potential to study pathogenesis of infections in humans. PET enables dynamic assessment of infectious processes within the same subject with high temporal and spatial resolution and obviates the need for invasive tissue sampling, which is difficult in patients and generally limited to a single time point, even in animal models. This review presents current state-of-the-art concepts on the application of molecular imaging for infectious diseases and details how PET imaging can facilitate novel insights into infectious processes, ongoing development of pathogen-specific imaging, and simultaneous in situ measurements of intralesional antimicrobial pharmacokinetics in multiple compartments, including privileged sites. Finally, the potential clinical applications of this promising technology are also discussed.
Collapse
|
16
|
Rajamani S, Kuszpit K, Scarff JM, Lundh L, Khan M, Brown J, Stafford R, Cazares LH, Panchal RG, Bocan T. Bioengineering of bacterial pathogens for noninvasive imaging and in vivo evaluation of therapeutics. Sci Rep 2018; 8:12618. [PMID: 30135466 PMCID: PMC6105664 DOI: 10.1038/s41598-018-30806-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/06/2018] [Indexed: 12/19/2022] Open
Abstract
Critical bacterial pathogens of public health and biodefense concerns were engineered to constitutively express Escherichia coli enzyme thymidine kinase (TK) that allows for noninvasive nuclear imaging via phosphorylation and entrapment of radiolabeled nucleoside analog 1-(2′deoxy-2′-fluoro-β-D-arabinofuranosyl)-5-iodouracil (FIAU). Expression of functional TK was established using a nucleoside analog Zidovudine that impeded the growth of tk-engineered bacteria. Significantly, no observable growth differences were detected for FIAU. High resolution mass spectrometry with Pseudomonas aeruginosa PAO1 and its tk variant (PAO1TK) confirmed FIAU phosphorylation and retention only in PAO1TK. In vitro gamma counting with wild-type PAO1, Acinetobacter baumannii and Burkholderia pseudomallei Bp82 and their tk derivatives with [18F]FIAU further confirmed that tk variants selectively incorporated the radiotracer, albeit with varying efficiencies. In vitro [18F]FIAU labeling coupled with in vivo Positron Emission Tomography/Computed Tomography (PET/CT) imaging of PAO1 and PAO1TK confirmed that only PAO1TK can be imaged in mice at sensitivities ≥107 bacteria per infection site. This was further verified by administering [18F]FIAU to animals infected with PAO1 and PAO1TK. Utility of tk-engineered P. aeruginosa in noninvasive PET/CT imaging for bacterial therapeutic evaluation in animals was demonstrated employing antibiotic ciprofloxacin, underscoring the immediate use of PAO1TK and potentially other engineered pathogens for evaluating experimental therapeutics.
Collapse
Affiliation(s)
- Sathish Rajamani
- Molecular & Translational Sciences Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, United States.
| | - Kyle Kuszpit
- Molecular & Translational Sciences Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, United States
| | - Jennifer M Scarff
- Molecular & Translational Sciences Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, United States
| | - Linnea Lundh
- Molecular & Translational Sciences Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, United States
| | - Maisha Khan
- Molecular & Translational Sciences Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, United States
| | - Jennifer Brown
- Molecular & Translational Sciences Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, United States
| | - Robert Stafford
- Molecular & Translational Sciences Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, United States
| | - Lisa H Cazares
- Molecular & Translational Sciences Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, United States
| | - Rekha G Panchal
- Molecular & Translational Sciences Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, United States
| | - Thomas Bocan
- Molecular & Translational Sciences Division, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD, 21702, United States.
| |
Collapse
|
17
|
Malherbe ST, Kleynhans L, Walzl G. The potential of imaging tools as correlates of infection and disease for new TB vaccine development. Semin Immunol 2018; 39:73-80. [PMID: 29914653 DOI: 10.1016/j.smim.2018.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 06/07/2018] [Indexed: 12/17/2022]
Abstract
The development of an improved vaccine to stimulate an effective response against Mycobacterium tuberculosis (MTB) infection and disease will be a major breakthrough in the fight against TB. A lack of tools to adequately track the progression or resolution of events in TB pathogenesis that occur at bacterial loads below the threshold for culture in human samples seriously hampers vaccine development and evaluation. In this review we discuss recent studies that use new imaging applications, modalities and analysis techniques to provide insight into the dynamic processes of MTB infection and disease that are challenging to monitor. These include early infection, the spectrum of latency and subclinical disease, the paucibacillary state induced by treatment, and events leading to recurrence, including relapse.
Collapse
Affiliation(s)
- Stephanus T Malherbe
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Léanie Kleynhans
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
18
|
Abstract
Infectious diseases are a major threat to humanity, and it is imperative that we develop imaging tools that aid in their study, facilitate diagnosis, and guide treatment. The alarming rise of highly virulent and multi-drug-resistant pathogens, their rapid spread leading to frequent global pandemics, fears of bioterrorism, and continued life-threatening nosocomial infections in hospitals remain as major challenges to health care in the USA and worldwide. Early diagnosis and rapid monitoring are essential for appropriate management and control of infections. Tomographic molecular imaging enables rapid, noninvasive visualization, localization, and monitoring of molecular processes deep within the body and offers several advantages over traditional tools used for the study of infectious diseases. Noninvasive, longitudinal assessments could streamline animal studies, allow unique insights into disease pathogenesis, and expedite clinical translation of new therapeutics. Since molecular imaging is already in common use in the clinic, it could also become a valuable tool for clinical studies, for patient care, for public health, and for enabling precision medicine for infectious diseases.
Collapse
|
19
|
Tucker EW, Pokkali S, Zhang Z, DeMarco VP, Klunk M, Smith ES, Ordonez AA, Penet MF, Bhujwalla Z, Jain SK, Kannan S. Microglia activation in a pediatric rabbit model of tuberculous meningitis. Dis Model Mech 2017; 9:1497-1506. [PMID: 27935825 PMCID: PMC5200899 DOI: 10.1242/dmm.027326] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/08/2016] [Indexed: 01/17/2023] Open
Abstract
Central nervous system (CNS) tuberculosis (TB) is the most severe form of extra-pulmonary TB and disproportionately affects young children where the developing brain has a unique host response. New Zealand white rabbits were infected with Mycobacterium tuberculosis via subarachnoid inoculation at postnatal day 4-8 and evaluated until 4-6 weeks post-infection. Control and infected rabbit kits were assessed for the development of neurological deficits, bacterial burden, and postmortem microbiologic and pathologic changes. The presence of meningitis and tuberculomas was demonstrated histologically and by in vivo magnetic resonance imaging (MRI). The extent of microglial activation was quantified by in vitro immunohistochemistry as well as non-invasive in vivo imaging of activated microglia/macrophages with positron emission tomography (PET). Subarachnoid infection induced characteristic leptomeningeal and perivascular inflammation and TB lesions with central necrosis, a cellular rim and numerous bacilli on pathologic examination. Meningeal and rim enhancement was visible on MRI. An intense microglial activation was noted in M. tuberculosis-infected animals in the white matter and around the TB lesions, as evidenced by a significant increase in uptake of the tracer 124I-DPA-713, which is specific for activated microglia/macrophages, and confirmed by quantification of Iba-1 immunohistochemistry. Neurobehavioral analyses demonstrated signs similar to those noted in children with delayed maturation and development of neurological deficits resulting in significantly worse composite behavior scores in M. tuberculosis-infected animals. We have established a rabbit model that mimics features of TB meningitis in young children. This model could provide a platform for evaluating novel therapies, including host-directed therapies, against TB meningitis relevant to a young child's developing brain.
Collapse
Affiliation(s)
- Elizabeth W Tucker
- Department of Anesthesiology and Critical Care Medicine, Division of Pediatric Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Anesthesiology and Critical Care Medicine, Division of Critical Care Medicine, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701, USA
| | - Supriya Pokkali
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zhi Zhang
- Department of Anesthesiology and Critical Care Medicine, Division of Pediatric Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Vincent P DeMarco
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mariah Klunk
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth S Smith
- Department of Anesthesiology and Critical Care Medicine, Division of Pediatric Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Marie-France Penet
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zaver Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA .,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Division of Pediatric Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA .,Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
20
|
Nooshabadi F, Yang HJ, Cheng Y, Durkee MS, Xie H, Rao J, Cirillo JD, Maitland KC. Intravital excitation increases detection sensitivity for pulmonary tuberculosis by whole-body imaging with β-lactamase reporter enzyme fluorescence. JOURNAL OF BIOPHOTONICS 2017; 10:821-829. [PMID: 27753271 PMCID: PMC5703064 DOI: 10.1002/jbio.201600132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/08/2016] [Accepted: 09/18/2016] [Indexed: 05/08/2023]
Abstract
Tuberculosis is a pulmonary disease with an especially high mortality rate in immuno-compromised populations, specifically children and HIV positive individuals. The causative agent, Mycobacterium tuberculosis (Mtb), is a very slow growing and difficult organism to work with, making both diagnosis and development of effective treatments cumbersome. We utilize a fiber-optic fluorescence microendoscope integrated with a whole-body imaging system for in vivo Mtb detection. The system exploits an endogenous enzyme of Mtb (β-lactamase, or BlaC) using a BlaC-specific NIR fluorogenic substrate. In the presence of BlaC, this substrate is cleaved and becomes fluorescent. Using intravital illumination of the lung to excite this probe, sensitivity of the optical system increases over trans- and epi-illumination methods of whole-body fluorescence imaging. We demonstrate that integration of these imaging technologies with BlaC-specific fluorescent reporter probe improves the level of detection to ∼100 colony forming units, a 100× increase in sensitivity in comparison to epi-illumination and a 10× increase in sensitivity in comparison to previous work in intravital excitation of tdTomato-expressing Mtb. This lower detection threshold enables the study of early stage bacterial infections with clinical strains of Mtb and longitudinal studies of disease pathogenesis and therapeutic efficacy with multiple time points in a single animal.
Collapse
Affiliation(s)
- Fatemeh Nooshabadi
- Biomedical Engineering Department, 3120 TAMU, Texas A&M University, College Station, TX 77843, United States
| | - Hee-Jeong Yang
- Microbial Pathogenesis and Immunology Department, Texas A&M University Health Science Center, Bryan, Texas 77807, United States
| | - Yunfeng Cheng
- Radiology Department, Stanford University, Stanford, CA 94304, United States
| | - Madeleine S. Durkee
- Biomedical Engineering Department, 3120 TAMU, Texas A&M University, College Station, TX 77843, United States
| | - Hexin Xie
- Radiology Department, Stanford University, Stanford, CA 94304, United States
| | - Jianghong Rao
- Radiology Department, Stanford University, Stanford, CA 94304, United States
| | - Jeffrey D. Cirillo
- Microbial Pathogenesis and Immunology Department, Texas A&M University Health Science Center, Bryan, Texas 77807, United States
| | - Kristen C. Maitland
- Biomedical Engineering Department, 3120 TAMU, Texas A&M University, College Station, TX 77843, United States
| |
Collapse
|
21
|
Nooshabadi F, Yang HJ, Cheng Y, Durkee MS, Xie H, Rao J, Cirillo JD, Maitland KC. Intravital excitation increases detection sensitivity for pulmonary tuberculosis by whole-body imaging with β-lactamase reporter enzyme fluorescence. JOURNAL OF BIOPHOTONICS 2017; 10:821-829. [PMID: 27753271 DOI: 10.1002/jbio.v10.6-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/08/2016] [Accepted: 09/18/2016] [Indexed: 05/22/2023]
Abstract
Tuberculosis is a pulmonary disease with an especially high mortality rate in immuno-compromised populations, specifically children and HIV positive individuals. The causative agent, Mycobacterium tuberculosis (Mtb), is a very slow growing and difficult organism to work with, making both diagnosis and development of effective treatments cumbersome. We utilize a fiber-optic fluorescence microendoscope integrated with a whole-body imaging system for in vivo Mtb detection. The system exploits an endogenous enzyme of Mtb (β-lactamase, or BlaC) using a BlaC-specific NIR fluorogenic substrate. In the presence of BlaC, this substrate is cleaved and becomes fluorescent. Using intravital illumination of the lung to excite this probe, sensitivity of the optical system increases over trans- and epi-illumination methods of whole-body fluorescence imaging. We demonstrate that integration of these imaging technologies with BlaC-specific fluorescent reporter probe improves the level of detection to ∼100 colony forming units, a 100× increase in sensitivity in comparison to epi-illumination and a 10× increase in sensitivity in comparison to previous work in intravital excitation of tdTomato-expressing Mtb. This lower detection threshold enables the study of early stage bacterial infections with clinical strains of Mtb and longitudinal studies of disease pathogenesis and therapeutic efficacy with multiple time points in a single animal.
Collapse
Affiliation(s)
- Fatemeh Nooshabadi
- Biomedical Engineering Department, 3120 TAMU, Texas A&M University, College Station, TX, 77843, United States
| | - Hee-Jeong Yang
- Microbial Pathogenesis and Immunology Department, Texas A&M University Health Science Center, Bryan, TX, 77807, United States
| | - Yunfeng Cheng
- Radiology Department, Stanford University, Stanford, CA, 94304, United States
| | - Madeleine S Durkee
- Biomedical Engineering Department, 3120 TAMU, Texas A&M University, College Station, TX, 77843, United States
| | - Hexin Xie
- Radiology Department, Stanford University, Stanford, CA, 94304, United States
| | - Jianghong Rao
- Radiology Department, Stanford University, Stanford, CA, 94304, United States
| | - Jeffrey D Cirillo
- Microbial Pathogenesis and Immunology Department, Texas A&M University Health Science Center, Bryan, TX, 77807, United States
| | - Kristen C Maitland
- Biomedical Engineering Department, 3120 TAMU, Texas A&M University, College Station, TX, 77843, United States
| |
Collapse
|
22
|
Jain SK. Introduction. IMAGING INFECTIONS 2017. [PMCID: PMC7122386 DOI: 10.1007/978-3-319-54592-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Infectious diseases are a major cause of morbidity and mortality worldwide and in the USA. Overall costs and morbidity are expected to continue to rise due to increasing rates of drug-resistant pathogens, use of invasive techniques such as implants, as well as immunosuppressive and cancer therapies. Tomographic molecular imaging techniques enable rapid visualization and monitoring of molecular processes noninvasively and promise unparalleled opportunities for field of infectious diseases. These technologies are an emerging field of research, overcome several fundamental limitations of current tools, and could have a broad impact on both basic research and patient care. Beyond diagnosis and monitoring disease, these technologies could also provide a uniform cross-species platform for animal studies, allow unique insights into understanding disease pathogenesis, and expedite bench-to-bedside translation of new therapeutics. Finally, since molecular imaging is readily available for humans, validated tracers could also become valuable tools for clinical applications and for enabling personalized medicine for infectious diseases.
Collapse
Affiliation(s)
- Sanjay K. Jain
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland USA
| |
Collapse
|
23
|
Rundell SR, Wagar ZL, Meints LM, Olson CD, O'Neill MK, Piligian BF, Poston AW, Hood RJ, Woodruff PJ, Swarts BM. Deoxyfluoro-d-trehalose (FDTre) analogues as potential PET probes for imaging mycobacterial infection. Org Biomol Chem 2016; 14:8598-609. [PMID: 27560008 DOI: 10.1039/c6ob01734g] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mycobacterium tuberculosis, the etiological agent of human tuberculosis, requires the non-mammalian disaccharide trehalose for growth and virulence. Recently, detectable trehalose analogues have gained attention as probes for studying trehalose metabolism and as potential diagnostic imaging agents for mycobacterial infections. Of particular interest are deoxy-[(18)F]fluoro-d-trehalose ((18)F-FDTre) analogues, which have been suggested as possible positron emission tomography (PET) probes for in vivo imaging of M. tuberculosis infection. Here, we report progress toward this objective, including the synthesis and conformational analysis of four non-radioactive deoxy-[(19)F]fluoro-d-trehalose ((19)F-FDTre) analogues, as well as evaluation of their uptake by M. smegmatis. The rapid synthesis and purification of several (19)F-FDTre analogues was accomplished in high yield using a one-step chemoenzymatic method. Conformational analysis of the (19)F-FDTre analogues using NMR and molecular modeling methods showed that fluorine substitution had a negligible effect on the conformation of the native disaccharide, suggesting that fluorinated analogues may be successfully recognized and processed by trehalose metabolic machinery in mycobacteria. To test this hypothesis and to evaluate a possible route for delivery of FDTre probes specifically to mycobacteria, we showed that (19)F-FDTre analogues are actively imported into M. smegmatis via the trehalose-specific transporter SugABC-LpqY. Finally, to demonstrate the applicability of these results to the efficient preparation and use of short-lived (18)F-FDTre PET radiotracers, we carried out (19)F-FDTre synthesis, purification, and administration to M. smegmatis in 1 hour.
Collapse
Affiliation(s)
- Sarah R Rundell
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ordonez AA, DeMarco VP, Klunk MH, Pokkali S, Jain SK. Imaging Chronic Tuberculous Lesions Using Sodium [(18)F]Fluoride Positron Emission Tomography in Mice. Mol Imaging Biol 2016; 17:609-14. [PMID: 25750032 DOI: 10.1007/s11307-015-0836-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
PURPOSE Calcification is a hallmark of chronic tuberculosis (TB) in humans, often noted years to decades (after the initial infection) on chest radiography, but not visualized well with traditional positron emission tomography (PET). We hypothesized that sodium [(18)F]fluoride (Na[(18)F]F) PET could be used to detect microcalcifications in a chronically Mycobacterium tuberculosis-infected murine model. PROCEDURES C3HeB/FeJ mice, which develop necrotic and hypoxic TB lesions, were aerosol-infected with M. tuberculosis and imaged with Na[(18)F]F PET. RESULTS Pulmonary TB lesions from chronically infected mice demonstrated significantly higher Na[(18)F]F uptake compared with acutely infected or uninfected animals (P < 0.01), while no differences were noted in the blood or bone compartments (P > 0.08). Ex vivo biodistribution studies confirmed the imaging findings, and tissue histology demonstrated microcalcifications in TB lesions from chronically infected mice, which has not been demonstrated previously in a murine model. CONCLUSION Na[(18)F]F PET can be used for the detection of chronic TB lesions and could prove to be a useful noninvasive biomarker for TB studies.
Collapse
Affiliation(s)
- Alvaro A Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University, 1550 Orleans Street, CRB-II, Rm 1.09, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Vincent P DeMarco
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University, 1550 Orleans Street, CRB-II, Rm 1.09, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Mariah H Klunk
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University, 1550 Orleans Street, CRB-II, Rm 1.09, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Supriya Pokkali
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University, 1550 Orleans Street, CRB-II, Rm 1.09, Baltimore, MD, USA
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University, 1550 Orleans Street, CRB-II, Rm 1.09, Baltimore, MD, USA.
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, MD, USA.
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
25
|
Abstract
The rise in multidrug resistant (MDR) bacteria has become a global crisis. Rapid and accurate diagnosis of infection will facilitate antibiotic stewardship and preserve our ability to treat and cure patients from bacterial infection. Direct in situ imaging of bacteria offers the prospect of accurately diagnosing disease and monitoring patient outcomes and response to treatment in real-time. There have been many recent advances in the field of optical imaging of infection; namely in specific probe and fluorophore design. This combined with the advances in imaging device technology render direct optical imaging of infection a feasible approach for accurate diagnosis in the clinic. Despite this, there are currently no licensed molecular probes for clinical optical imaging of infection. Here we report some of the most promising and interesting probes and approaches under development for this purpose, which have been evaluated in in vivo models within the laboratory setting.
Collapse
|
26
|
Sood S, Kaur S, Shrivastava R. A lacZ Reporter-Based Strategy for Rapid Expression Analysis and Target Validation of Mycobacterium tuberculosis Latent Infection Genes. Curr Microbiol 2015; 72:213-219. [PMID: 26597215 DOI: 10.1007/s00284-015-0942-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 10/01/2015] [Indexed: 11/25/2022]
Abstract
We report a novel lacZ fusion vector and demonstrate its utility for expression analysis of genes associated with Mycobacterium tuberculosis latent infection. The vector contains E. coli (oriE) and mycobacterial (oriM) origins of replication, a kanamycin resistance gene (Km(r)) as selection marker, and a lacZ reporter gene in fusion with MCS for cloning of upstream regulatory sequence of the desired genes. β-galactosidase activity of the vector was standardized for expression analysis under latent mycobacterial conditions using Phsp60, a constitutive mycobacterial promoter, utilizing Mycobacterium smegmatis as model organism. Validation of the vector was done by cloning and expression analysis of PhspX (alpha crystalline) and Picl (isocitrate lyase), promoters from two of the genes shown to be involved in M. tuberculosis persistence. Both genes showed appreciable levels of β-galactosidase expression under hypoxia-induced persistent conditions in comparison to their actively replicating state. Expression analysis of a set of hypothetical genes was also done, of which Rv0628c showed increased expression under persistent conditions. The reported fusion vector and the strategy can be effectively used for short listing and validation of drug targets deduced from various non-conclusive approaches such as bioinformatics and microarray analysis against latent/persistent form of mycobacterial infection.
Collapse
Affiliation(s)
- Shivani Sood
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India
| | - Satinder Kaur
- Division of Microbiology, Central Drug Research Institute, Lucknow, Uttar Pradesh, 226001, India
| | - Rahul Shrivastava
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173234, India.
| |
Collapse
|
27
|
Bocan TM, Panchal RG, Bavari S. Applications of in vivo imaging in the evaluation of the pathophysiology of viral and bacterial infections and in development of countermeasures to BSL3/4 pathogens. Mol Imaging Biol 2015; 17:4-17. [PMID: 25008802 PMCID: PMC4544652 DOI: 10.1007/s11307-014-0759-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
While preclinical and clinical imaging have been applied to drug discovery/development and characterization of disease pathology, few examples exist where imaging has been used to evaluate infectious agents or countermeasures to biosafety level (BSL)3/4 threat agents. Viruses engineered with reporter constructs, i.e., enzymes and receptors, which are amenable to detection by positron emission tomography (PET), single photon emission tomography (SPECT), or magnetic resonance imaging (MRI) have been used to evaluate the biodistribution of viruses containing specific therapeutic or gene transfer payloads. Bioluminescence and nuclear approaches involving engineered reporters, direct labeling of bacteria with radiotracers, or tracking bacteria through their constitutively expressed thymidine kinase have been utilized to characterize viral and bacterial pathogens post-infection. Most PET, SPECT, CT, or MRI approaches have focused on evaluating host responses to the pathogens such as inflammation, brain neurochemistry, and structural changes and on assessing the biodistribution of radiolabeled drugs. Imaging has the potential when applied preclinically to the development of countermeasures against BSL3/4 threat agents to address the following: (1) presence, biodistribution, and time course of infection in the presence or absence of drug; (2) binding of the therapeutic to the target; and (3) expression of a pharmacologic effect either related to drug mechanism, efficacy, or safety. Preclinical imaging could potentially provide real-time dynamic tools to characterize the pathogen and animal model and for developing countermeasures under the U.S. FDA Animal Rule provision with high confidence of success and clinical benefit.
Collapse
Affiliation(s)
- Thomas M Bocan
- Molecular and Translational Sciences, US Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Ft. Detrick, MD, 21702, USA,
| | | | | |
Collapse
|
28
|
Gumbo T, Lenaerts AJ, Hanna D, Romero K, Nuermberger E. Nonclinical Models for Antituberculosis Drug Development: A Landscape Analysis. J Infect Dis 2015; 211 Suppl 3:S83-95. [DOI: 10.1093/infdis/jiv183] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
29
|
The Complex Mechanism of Antimycobacterial Action of 5-Fluorouracil. ACTA ACUST UNITED AC 2015; 22:63-75. [DOI: 10.1016/j.chembiol.2014.11.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/30/2014] [Accepted: 11/03/2014] [Indexed: 11/17/2022]
|
30
|
Radioiodinated DPA-713 imaging correlates with bactericidal activity of tuberculosis treatments in mice. Antimicrob Agents Chemother 2014; 59:642-9. [PMID: 25403669 DOI: 10.1128/aac.04180-14] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Current tools for monitoring response to tuberculosis treatments have several limitations. Noninvasive biomarkers could accelerate tuberculosis drug development and clinical studies, but to date little progress has been made in developing new imaging technologies for this application. In this study, we developed pulmonary single-photon emission computed tomography (SPECT) using radioiodinated DPA-713 to serially monitor the activity of tuberculosis treatments in live mice, which develop necrotic granulomas and cavitary lesions. C3HeB/FeJ mice were aerosol infected with Mycobacterium tuberculosis and administered either a standard or a highly active bedaquiline-containing drug regimen. Serial (125)I-DPA-713 SPECT imaging was compared with (18)F-fluorodeoxyglucose ((18)F-FDG) positron emission tomography (PET) and standard microbiology. Ex vivo studies were performed to characterize and correlate DPA-713 imaging with cellular and cytokine responses. Pulmonary (125)I-DPA-713 SPECT, but not (18)F-FDG PET, was able to correctly identify the bactericidal activities of the two tuberculosis treatments as early as 4 weeks after the start of treatment (P < 0.03). DPA-713 readily penetrated the fibrotic rims of necrotic and cavitary lesions. A time-dependent decrease in both tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ) levels was observed with treatments, with (125)I-DPA-713 SPECT correlating best with tissue TNF-α levels (ρ = 0.94; P < 0.01). (124)I-DPA-713 was also evaluated as a PET probe and demonstrated a 4.0-fold-higher signal intensity in the infected tuberculous lesions than uninfected controls (P = 0.03). These studies provide proof of concept for application of a novel noninvasive imaging biomarker to monitor tuberculosis treatments, with the potential for application for humans.
Collapse
|
31
|
Johnson DH, Via LE, Kim P, Laddy D, Lau CY, Weinstein EA, Jain S. Nuclear imaging: a powerful novel approach for tuberculosis. Nucl Med Biol 2014; 41:777-84. [PMID: 25195017 DOI: 10.1016/j.nucmedbio.2014.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/16/2014] [Accepted: 08/03/2014] [Indexed: 02/06/2023]
Abstract
Nearly 20 years after the World Health Organization declared tuberculosis (TB) a global public health emergency, TB still remains a major global threat with 8.6 million new cases and 1.3 million deaths annually. Mycobacterium tuberculosis adapts to a quiescent physiological state, and is notable for complex interaction with the host, producing poorly-understood disease states ranging from latent infection to fully active disease. Of the approximately 2.5 billion people latently infected with M. tuberculosis, many will develop reactivation disease (relapse), years after the initial infection. While progress has been made on some fronts, the alarming spread of multidrug-resistant, extensively drug-resistant, and more recently totally-drug resistant strains is of grave concern. New tools are urgently needed for rapidly diagnosing TB, monitoring TB treatments and to allow unique insights into disease pathogenesis. Nuclear bioimaging is a powerful, noninvasive tool that can rapidly provide three-dimensional views of disease processes deep within the body and conduct noninvasive longitudinal assessments of the same patient. In this review, we discuss the application of nuclear bioimaging to TB, including the current state of the field, considerations for radioprobe development, study of TB drug pharmacokinetics in infected tissues, and areas of research and clinical needs that could be addressed by nuclear bioimaging. These technologies are an emerging field of research, overcome several fundamental limitations of current tools, and will have a broad impact on both basic research and patient care. Beyond diagnosis and monitoring disease, these technologies will also allow unique insights into understanding disease pathogenesis; and expedite bench-to-bedside translation of new therapeutics. Finally, since molecular imaging is readily available for humans, validated tracers will become valuable tools for clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sanjay Jain
- Center for Infection and Inflammation Imaging Research, Center for Tuberculosis Research and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
32
|
Murawski AM, Gurbani S, Harper JS, Klunk M, Younes L, Jain SK, Jedynak BM. Imaging the evolution of reactivation pulmonary tuberculosis in mice using 18F-FDG PET. J Nucl Med 2014; 55:1726-9. [PMID: 25082854 DOI: 10.2967/jnumed.114.144634] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Latent tuberculosis infection affects one third of the world's population and can reactivate (relapse) decades later. However, current technologies, dependent on postmortem analyses, cannot follow the temporal evolution of disease. METHODS C3HeB/FeJ mice, which develop necrotic and hypoxic tuberculosis lesions, were aerosol-infected with Mycobacterium tuberculosis. PET and CT were used to serially image the same cohort of infected mice through pretreatment, tuberculosis treatment, and subsequent development of relapse. RESULTS A novel diffeomorphic registration was successfully used to monitor the spatial evolution of individual pulmonary lesions. Although most lesions during relapse developed in the same regions as those noted during pretreatment, several lesions also arose de novo within regions with no prior lesions. CONCLUSION This study presents a novel model that simulates infection and reactivation disease as seen in humans and could prove valuable to study tuberculosis pathogenesis and evaluate novel therapeutics.
Collapse
Affiliation(s)
- Allison M Murawski
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University, Baltimore, Maryland Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland Department of Pediatrics, Johns Hopkins University, Baltimore, Maryland
| | - Saumya Gurbani
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, Maryland and Center for Imaging Science, Johns Hopkins University, Baltimore, Maryland
| | - Jamie S Harper
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University, Baltimore, Maryland Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland Department of Pediatrics, Johns Hopkins University, Baltimore, Maryland
| | - Mariah Klunk
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University, Baltimore, Maryland Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland Department of Pediatrics, Johns Hopkins University, Baltimore, Maryland
| | - Laurent Younes
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, Maryland and Center for Imaging Science, Johns Hopkins University, Baltimore, Maryland
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University, Baltimore, Maryland Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland Department of Pediatrics, Johns Hopkins University, Baltimore, Maryland
| | - Bruno M Jedynak
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, Maryland and Center for Imaging Science, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
33
|
Scanga CA, Lopresti BJ, Tomko J, Frye LJ, Coleman TM, Fillmore D, Carney JP, Lin PL, Flynn JL, Gardner CL, Sun C, Klimstra WB, Ryman KD, Reed DS, Fisher DJ, Cole KS. In vivo imaging in an ABSL-3 regional biocontainment laboratory. Pathog Dis 2014; 71:207-12. [PMID: 24838691 DOI: 10.1111/2049-632x.12186] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 05/02/2014] [Indexed: 11/27/2022] Open
Abstract
The Regional Biocontainment Laboratory (RBL) at the University of Pittsburgh is a state-of-the-art ABSL-3 facility that supports research on highly pathogenic viruses and bacteria. Recent advances in radiologic imaging provide several noninvasive, in vivo imaging modalities that can be used to longitudinally monitor animals following experimental infection or vaccination. The University of Pittsburgh RBL provides digital radiography, bioluminescence imaging, and PET/CT. Operating these platforms in an ABSL-3 poses unique challenges. This review will discuss the development and refinement of these imaging platforms in high containment, emphasizing specific challenges and how they were overcome.
Collapse
Affiliation(s)
- Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Dutta NK, Illei PB, Jain SK, Karakousis PC. Characterization of a novel necrotic granuloma model of latent tuberculosis infection and reactivation in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2045-55. [PMID: 24815353 DOI: 10.1016/j.ajpath.2014.03.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/20/2014] [Accepted: 03/18/2014] [Indexed: 01/09/2023]
Abstract
We sought to develop and characterize a novel paucibacillary model in mice, which develops necrotic lung granulomas after infection with Mycobacterium tuberculosis. Six weeks after aerosol immunization with recombinant Mycobacterium bovis bacillus Calmette-Guerin overexpressing the 30-kDa antigen, C3HeB/FeJ mice were aerosol infected with M. tuberculosis H37Rv. Six weeks later, mice were treated with one of three standard regimens for latent tuberculosis infection or tumor necrosis factor (TNF)-neutralizing antibody. Mouse lungs were analyzed by histological features, positron emission tomography/computed tomography, whole-genome microarrays, and RT-PCR. Lungs and sera were studied by multiplex enzyme-linked immunosorbent assays. Paucibacillary infection was established, recapitulating the sterilizing activities of human latent tuberculosis infection regimens. TNF neutralization led to increased lung bacillary load, disrupted granuloma architecture with expanded necrotic foci and reduced tissue hypoxia, and accelerated animal mortality. TNF-neutralized mouse lungs and sera showed significant up-regulation of interferon γ, IL-1β, IL-6, IL-10, chemokine ligands 2 and 3, and matrix metalloproteinase genes. Clinical and microbiological reactivation of paucibacillary infection by TNF neutralization was associated with reduced hypoxia in lung granulomas and induction of matrix metalloproteinases and proinflammatory cytokines. This model may be useful for screening the sterilizing activity of novel anti-tuberculosis drugs, and identifying mycobacterial regulatory and metabolic pathways required for bacillary growth restriction and reactivation.
Collapse
Affiliation(s)
- Noton K Dutta
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Peter B Illei
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sanjay K Jain
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Petros C Karakousis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.
| |
Collapse
|
35
|
Foss CA, Harper JS, Wang H, Pomper MG, Jain SK. Noninvasive molecular imaging of tuberculosis-associated inflammation with radioiodinated DPA-713. J Infect Dis 2013; 208:2067-74. [PMID: 23901092 DOI: 10.1093/infdis/jit331] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Increased expression of translocator protein (TSPO) is a feature of microglial and macrophage activation. Since activated macrophages are key components of tuberculosis-associated inflammation, we evaluated radioiodinated DPA-713, a synthetic ligand of TSPO, for in vivo imaging of host response. METHODS Mice were infected with aerosolized Mycobacterium tuberculosis and evaluated using whole-body [(125)I]iodo-DPA-713 single-photon emission computed tomography (SPECT). Ex vivo biodistribution and correlative immunofluorescence studies were also performed. RESULTS [(125)I]Iodo-DPA-713 SPECT imaging clearly delineated tuberculosis-associated pulmonary inflammation in live animals. Biodistribution studies confirmed radiotracer specificity for inflamed pulmonary tissues. Immunofluorescence studies demonstrated that TSPO is highly expressed in CD68(+) macrophages and phagocytic cells within tuberculosis lesions and that [(125)I]DPA-713 specifically accumulates within these cells. Coadministration of excess unlabelled DPA-713 abrogated both the SPECT and ex vivo fluorescence signals. Lesion-specific signal-to-noise ratios were significantly higher with [(125)I]iodo-DPA-713 SPECT (4.06 ± 0.52) versus [(18)F]fluorodeoxyglucose (FDG) positron emission tomography (PET) (2.00 ± 0.28) performed in the same mice (P = .004). CONCLUSIONS [(125)I]Iodo-DPA-713 accumulates specifically in tuberculosis-associated inflammatory lesions by selective retention within macrophages and phagocytic cells. [(125)I]Iodo-DPA-713 SPECT provides higher lesion-specific signal-to-noise ratios than [(18)F]FDG PET and may prove to be a more specific biomarker to monitor tuberculosis in situ.
Collapse
|
36
|
Bagci U, Foster B, Miller-Jaster K, Luna B, Dey B, Bishai WR, Jonsson CB, Jain S, Mollura DJ. A computational pipeline for quantification of pulmonary infections in small animal models using serial PET-CT imaging. EJNMMI Res 2013; 3:55. [PMID: 23879987 PMCID: PMC3734217 DOI: 10.1186/2191-219x-3-55] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 07/06/2013] [Indexed: 12/19/2022] Open
Abstract
Background Infectious diseases are the second leading cause of death worldwide. In order to better understand and treat them, an accurate evaluation using multi-modal imaging techniques for anatomical and functional characterizations is needed. For non-invasive imaging techniques such as computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET), there have been many engineering improvements that have significantly enhanced the resolution and contrast of the images, but there are still insufficient computational algorithms available for researchers to use when accurately quantifying imaging data from anatomical structures and functional biological processes. Since the development of such tools may potentially translate basic research into the clinic, this study focuses on the development of a quantitative and qualitative image analysis platform that provides a computational radiology perspective for pulmonary infections in small animal models. Specifically, we designed (a) a fast and robust automated and semi-automated image analysis platform and a quantification tool that can facilitate accurate diagnostic measurements of pulmonary lesions as well as volumetric measurements of anatomical structures, and incorporated (b) an image registration pipeline to our proposed framework for volumetric comparison of serial scans. This is an important investigational tool for small animal infectious disease models that can help advance researchers’ understanding of infectious diseases. Methods We tested the utility of our proposed methodology by using sequentially acquired CT and PET images of rabbit, ferret, and mouse models with respiratory infections of Mycobacterium tuberculosis (TB), H1N1 flu virus, and an aerosolized respiratory pathogen (necrotic TB) for a total of 92, 44, and 24 scans for the respective studies with half of the scans from CT and the other half from PET. Institutional Administrative Panel on Laboratory Animal Care approvals were obtained prior to conducting this research. First, the proposed computational framework registered PET and CT images to provide spatial correspondences between images. Second, the lungs from the CT scans were segmented using an interactive region growing (IRG) segmentation algorithm with mathematical morphology operations to avoid false positive (FP) uptake in PET images. Finally, we segmented significant radiotracer uptake from the PET images in lung regions determined from CT and computed metabolic volumes of the significant uptake. All segmentation processes were compared with expert radiologists’ delineations (ground truths). Metabolic and gross volume of lesions were automatically computed with the segmentation processes using PET and CT images, and percentage changes in those volumes over time were calculated. (Continued on next page)(Continued from previous page) Standardized uptake value (SUV) analysis from PET images was conducted as a complementary quantitative metric for disease severity assessment. Thus, severity and extent of pulmonary lesions were examined through both PET and CT images using the aforementioned quantification metrics outputted from the proposed framework. Results Each animal study was evaluated within the same subject class, and all steps of the proposed methodology were evaluated separately. We quantified the accuracy of the proposed algorithm with respect to the state-of-the-art segmentation algorithms. For evaluation of the segmentation results, dice similarity coefficient (DSC) as an overlap measure and Haussdorf distance as a shape dissimilarity measure were used. Significant correlations regarding the estimated lesion volumes were obtained both in CT and PET images with respect to the ground truths (R2=0.8922,p<0.01 and R2=0.8664,p<0.01, respectively). The segmentation accuracy (DSC (%)) was 93.4±4.5% for normal lung CT scans and 86.0±7.1% for pathological lung CT scans. Experiments showed excellent agreements (all above 85%) with expert evaluations for both structural and functional imaging modalities. Apart from quantitative analysis of each animal, we also qualitatively showed how metabolic volumes were changing over time by examining serial PET/CT scans. Evaluation of the registration processes was based on precisely defined anatomical landmark points by expert clinicians. An average of 2.66, 3.93, and 2.52 mm errors was found in rabbit, ferret, and mouse data (all within the resolution limits), respectively. Quantitative results obtained from the proposed methodology were visually related to the progress and severity of the pulmonary infections as verified by the participating radiologists. Moreover, we demonstrated that lesions due to the infections were metabolically active and appeared multi-focal in nature, and we observed similar patterns in the CT images as well. Consolidation and ground glass opacity were the main abnormal imaging patterns and consistently appeared in all CT images. We also found that the gross and metabolic lesion volume percentage follow the same trend as the SUV-based evaluation in the longitudinal analysis. Conclusions We explored the feasibility of using PET and CT imaging modalities in three distinct small animal models for two diverse pulmonary infections. We concluded from the clinical findings, derived from the proposed computational pipeline, that PET-CT imaging is an invaluable hybrid modality for tracking pulmonary infections longitudinally in small animals and has great potential to become routinely used in clinics. Our proposed methodology showed that automated computed-aided lesion detection and quantification of pulmonary infections in small animal models are efficient and accurate as compared to the clinical standard of manual and semi-automated approaches. Automated analysis of images in pre-clinical applications can increase the efficiency and quality of pre-clinical findings that ultimately inform downstream experimental design in human clinical studies; this innovation will allow researchers and clinicians to more effectively allocate study resources with respect to research demands without compromising accuracy.
Collapse
Affiliation(s)
- Ulas Bagci
- Center for Infectious Disease Imaging, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Jelicks LA, Lisanti MP, Machado FS, Weiss LM, Tanowitz HB, Desruisseaux MS. Imaging of small-animal models of infectious diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2012. [PMID: 23201133 DOI: 10.1016/j.ajpath.2012.09.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Infectious diseases are the second leading cause of death worldwide. Noninvasive small-animal imaging has become an important research tool for preclinical studies of infectious diseases. Imaging studies permit enhanced information through longitudinal studies of the same animal during the infection. Herein, we briefly review recent studies of animal models of infectious disease that have used imaging modalities.
Collapse
Affiliation(s)
- Linda A Jelicks
- Department of Physiology and Biophysics and the Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Noninvasive determination of 2-[18F]-fluoroisonicotinic acid hydrazide pharmacokinetics by positron emission tomography in Mycobacterium tuberculosis-infected mice. Antimicrob Agents Chemother 2012; 56:6284-90. [PMID: 23006755 DOI: 10.1128/aac.01644-12] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Tuberculosis (TB) is a global pandemic requiring sustained therapy to facilitate curing and to prevent the emergence of drug resistance. There are few adequate tools to evaluate drug dynamics within infected tissues in vivo. In this report, we evaluated a fluorinated analog of isoniazid (INH), 2-[(18)F]fluoroisonicotinic acid hydrazide (2-[(18)F]-INH), as a probe for imaging Mycobacterium tuberculosis-infected mice by dynamic positron emission tomography (PET). We developed a tail vein catheter system to safely deliver drugs to M. tuberculosis aerosol-infected mice inside sealed biocontainment devices. Imaging was rapid and noninvasive, and it could simultaneously visualize multiple tissues. Dynamic PET imaging demonstrated that 2-[(18)F]-INH was extensively distributed and rapidly accumulated at the sites of infection, including necrotic pulmonary TB lesions. Compared to uninfected animals, M. tuberculosis-infected mice had a significantly higher PET signal within the lungs (P < 0.05) despite similar PET activity in the liver (P > 0.85), suggesting that 2-[(18)F]-INH accumulated at the site of the pulmonary infection. Furthermore, our data indicated that similar to INH, 2-[(18)F]-INH required specific activation and accumulated within the bacterium. Pathogen-specific metabolism makes positron-emitting INH analogs attractive candidates for development into imaging probes with the potential to both detect bacteria and yield pharmacokinetic data in situ. Since PET imaging is currently used clinically, this approach could be translated from preclinical studies to use in humans.
Collapse
|
39
|
Cronin M, Stanton RM, Francis KP, Tangney M. Bacterial vectors for imaging and cancer gene therapy: a review. Cancer Gene Ther 2012; 19:731-40. [PMID: 22996740 DOI: 10.1038/cgt.2012.59] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The significant burden of resistance to conventional anticancer treatments in patients with advanced disease has prompted the need to explore alternative therapeutic strategies. The challenge for oncology researchers is to identify a therapy which is selective for tumors with limited toxicity to normal tissue. Engineered bacteria have the unique potential to overcome traditional therapies' limitations by specifically targeting tumors. It has been shown that bacteria are naturally capable of homing to tumors when systemically administered resulting in high levels of replication locally, either external to (non-invasive species) or within tumor cells (pathogens). Pre-clinical and clinical investigations involving bacterial vectors require relevant means of monitoring vector trafficking and levels over time, and development of bacterial-specific real-time imaging modalities are key for successful development of clinical bacterial gene delivery. This review discusses the currently available imaging technologies and the progress to date exploiting these for monitoring of bacterial gene delivery in vivo.
Collapse
Affiliation(s)
- M Cronin
- Cork Cancer Research Centre, BioSciences Institute, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
40
|
Dorward DA, Lucas CD, Rossi AG, Haslett C, Dhaliwal K. Imaging inflammation: molecular strategies to visualize key components of the inflammatory cascade, from initiation to resolution. Pharmacol Ther 2012; 135:182-99. [PMID: 22627270 DOI: 10.1016/j.pharmthera.2012.05.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 05/07/2012] [Indexed: 12/19/2022]
Abstract
Dysregulation of inflammation is central to the pathogenesis of innumerable human diseases. Understanding and tracking the critical events in inflammation are crucial for disease monitoring and pharmacological drug discovery and development. Recent progress in molecular imaging has provided novel insights into spatial associations, molecular events and temporal sequelae in the inflammatory process. While remaining a burgeoning field in pre-clinical research, increasing application in man affords researchers the opportunity to study disease pathogenesis in humans in situ thereby revolutionizing conventional understanding of pathophysiology and potential therapeutic targets. This review provides a description of commonly used molecular imaging modalities, including optical, radionuclide and magnetic resonance imaging, and details key advances and translational opportunities in imaging inflammation from initiation to resolution.
Collapse
Affiliation(s)
- D A Dorward
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
| | | | | | | | | |
Collapse
|
41
|
Jonsson CB, Camp JV, Wu A, Zheng H, Kraenzle JL, Biller AE, Vanover CD, Chu YK, Ng CK, Proctor M, Sherwood L, Steffen MC, Mollura DJ. Molecular imaging reveals a progressive pulmonary inflammation in lower airways in ferrets infected with 2009 H1N1 pandemic influenza virus. PLoS One 2012; 7:e40094. [PMID: 22911695 PMCID: PMC3401186 DOI: 10.1371/journal.pone.0040094] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 05/31/2012] [Indexed: 12/16/2022] Open
Abstract
Molecular imaging has gained attention as a possible approach for the study of the progression of inflammation and disease dynamics. Herein we used [(18)F]-2-deoxy-2-fluoro-D-glucose ([(18)F]-FDG) as a radiotracer for PET imaging coupled with CT (FDG-PET/CT) to gain insight into the spatiotemporal progression of the inflammatory response of ferrets infected with a clinical isolate of a pandemic influenza virus, H1N1 (H1N1pdm). The thoracic regions of mock- and H1N1pdm-infected ferrets were imaged prior to infection and at 1, 2, 3 and 6 days post-infection (DPI). On 1 DPI, FDG-PET/CT imaging revealed areas of consolidation in the right caudal lobe which corresponded with elevated [(18)F]-FDG uptake (maximum standardized uptake values (SUVMax), 4.7-7.0). By days 2 and 3, consolidation (CT) and inflammation ([(18)F]-FDG) appeared in the left caudal lobe. By 6 DPI, CT images showed extensive areas of patchy ground-glass opacities (GGO) and consolidations with the largest lesions having high SUVMax (6.0-7.6). Viral shedding and replication were detected in most nasal, throat and rectal swabs and nasal turbinates and lungs on 1, 2 and 3 DPI, but not on day 7, respectively. In conclusion, molecular imaging of infected ferrets revealed a progressive consolidation on CT with corresponding [(18)F]-FDG uptake. Strong positive correlations were measured between SUVMax and bronchiolitis-related pathologic scoring (Spearman's ρ = 0.75). Importantly, the extensive areas of patchy GGO and consolidation seen on CT in the ferret model at 6 DPI are similar to that reported for human H1N1pdm infections. In summary, these first molecular imaging studies of lower respiratory infection with H1N1pdm show that FDG-PET can give insight into the spatiotemporal progression of the inflammation in real-time.
Collapse
Affiliation(s)
- Colleen B Jonsson
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Pullambhatla M, Tessier J, Beck G, Jedynak B, Wurthner JU, Pomper MG. [(125)I]FIAU imaging in a preclinical model of lung infection: quantification of bacterial load. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2012; 2:260-270. [PMID: 23133816 PMCID: PMC3477740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/11/2012] [Indexed: 06/01/2023]
Abstract
2'-Fluoro-2'-deoxy-1β-D-arabinofuranosyl-5-[(125)I]iodouracil ([(125)I]FIAU), a substrate for the thymidine kinase (TK) present in most bacteria, has been used as an imaging agent for single photon emission computed tomography (SPECT) in an experimental model of lung infection. Using SPECT-CT we show that [(125)I]FIAU is specific for bacterial infection rather than sterile inflammation. We report [(125)I]FIAU lung uptake values of 1.26 ± 0.20 percent injected dose per gram (%ID/g) in normal controls, 1.69 ± 0.32 %ID/g in lung inflammation and up to 7.14 ± 1.09 %ID/g in lung infection in ex vivo biodistribution studies at 24 h after intranasal administration of bacteria. Images of [(125)I]FIAU signal within lung can be used to estimate the number of bacteria present, with a limit of detection of 10(9) colony forming units per mL on the X-SPECT scanner. [(125)I]FIAU-Based bacterial imaging may be useful in preclinical models to facilitate the development of new antibiotics, particularly in cases where a corresponding human trial is planned.
Collapse
Affiliation(s)
- Mrudula Pullambhatla
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins UniversityMacclesfield, UK
| | | | - Graham Beck
- Department of Applied Mathematics and Statistics, Johns Hopkins UniversityBaltimore, Maryland, USA
| | - Bruno Jedynak
- Department of Applied Mathematics and Statistics, Johns Hopkins UniversityBaltimore, Maryland, USA
| | | | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins UniversityMacclesfield, UK
| |
Collapse
|
43
|
Rifapentine is not more active than rifampin against chronic tuberculosis in guinea pigs. Antimicrob Agents Chemother 2012; 56:3726-31. [PMID: 22547623 DOI: 10.1128/aac.00500-12] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Rifamycins are key sterilizing drugs in the current treatment of active tuberculosis (TB). Daily dosing of rifapentine (P), a potent rifamycin with high intracellular accumulation, in place of rifampin (R) in the standard antitubercular regimen significantly shortens the duration of treatment needed to prevent relapse in a murine model of active TB. We undertook the current study to compare directly the activities of human-equivalent doses of P and R in a guinea pig model of chronic TB, in which bacilli are predominantly extracellular within human-like necrotic granulomas. Hartley strain guinea pigs were aerosol infected with ~200 bacilli of Mycobacterium tuberculosis H37Rv, and treatment given 5 days/week was initiated 6 weeks later. R at 100 mg/kg of body weight and P at 100 mg/kg were given orally alone or in combination with isoniazid (H) at 60 mg/kg and pyrazinamide (Z) at 300 mg/kg. Culture-positive relapse was assessed in subgroups of guinea pigs after completion of 1 and 2 months of treatment. Human-equivalent doses of R and P showed equivalent bactericidal activity when used alone and in combination therapy. In guinea pigs treated with rifampin, isoniazid, and pyrazinamide (RHZ) or PHZ, microbiological relapse occurred in the lungs of 8/10 animals treated for 1 month and in 0/10 animals treated for 2 months. Substitution of P for R in the standard antitubercular regimen did not shorten the time to cure in this guinea pig model of chronic TB. Data from ongoing clinical trials comparing the activity of these two drugs are awaited to determine the relevance of the guinea pig TB model in preclinical drug screening.
Collapse
|
44
|
Harper J, Skerry C, Davis SL, Tasneen R, Weir M, Kramnik I, Bishai WR, Pomper MG, Nuermberger EL, Jain SK. Mouse model of necrotic tuberculosis granulomas develops hypoxic lesions. J Infect Dis 2012; 205:595-602. [PMID: 22198962 PMCID: PMC3266133 DOI: 10.1093/infdis/jir786] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 09/23/2011] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Preclinical evaluation of tuberculosis drugs is generally limited to mice. However, necrosis and hypoxia, key features of human tuberculosis lesions, are lacking in conventional mouse strains. METHODS We used C3HeB/FeJ mice, which develop necrotic lesions in response to Mycobacterium tuberculosis infection. Positron emission tomography in live infected animals, postmortem pimonidazole immunohistochemistry, and bacterial gene expression analyses were used to assess whether tuberculosis lesions in C3HeB/FeJ are hypoxic. Efficacy of combination drug treatment, including PA-824, active against M. tuberculosis under hypoxic conditions, was also evaluated. RESULTS Tuberculosis lesions in C3HeB/FeJ (but not BALB/c) were found to be hypoxic and associated with up-regulation of known hypoxia-associated bacterial genes (P < .001). Contrary to sustained activity reported elsewhere in BALB/c mice, moxifloxacin and pyrazinamide (MZ) combination was not bactericidal beyond 3 weeks in C3HeB/FeJ. Although PA-824 added significant activity, the novel combination of PA-824 and MZ was less effective than the standard first-line regimen in C3HeB/FeJ. CONCLUSIONS We demonstrate that tuberculosis lesions in C3HeB/FeJ are hypoxic. Activities of some key tuberculosis drug regimens in development are represented differently in C3HeB/FeJ versus BALB/c mice. Because C3HeB/FeJ display key features of human tuberculosis, this strain warrants evaluation as a more pathologically relevant model for preclinical studies.
Collapse
Affiliation(s)
- Jamie Harper
- Department of Medicine, Center for Infection and Inflammation Imaging Research
- Department of Medicine, Center for Tuberculosis Research
- Department of Pediatrics
| | - Ciaran Skerry
- Department of Medicine, Center for Tuberculosis Research
- Department of Pediatrics
| | - Stephanie L. Davis
- Department of Medicine, Center for Infection and Inflammation Imaging Research
- Department of Medicine, Center for Tuberculosis Research
- Department of Pediatrics
| | - Rokeya Tasneen
- Department of Medicine, Center for Tuberculosis Research
| | - Mariah Weir
- Department of Medicine, Center for Infection and Inflammation Imaging Research
- Department of Medicine, Center for Tuberculosis Research
- Department of Pediatrics
| | - Igor Kramnik
- National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Massachusetts
| | | | - Martin G. Pomper
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Sanjay K. Jain
- Department of Medicine, Center for Infection and Inflammation Imaging Research
- Department of Medicine, Center for Tuberculosis Research
- Department of Pediatrics
| |
Collapse
|
45
|
Jang SJ, Lee YJ, Lim S, Kim KI, Lee KC, An GI, Lee TS, Cheon GJ, Lim SM, Kang JH. Imaging of a localized bacterial infection with endogenous thymidine kinase using radioisotope-labeled nucleosides. Int J Med Microbiol 2012; 302:101-7. [PMID: 22264560 DOI: 10.1016/j.ijmm.2011.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 09/29/2011] [Accepted: 11/06/2011] [Indexed: 02/05/2023] Open
Abstract
The importance of noninvasive imaging methods to bacterial infections is widely recognized. To obtain bacterial infection imaging with radioisotope-labeled nucleosides, bacterial thymidine kinase (tk) activities of Salmonella typhimurium with [(125)I]5-iodo-1-(2'-fluoro-2'-deoxy-β-d-arabinofuranosyl)uracil ([(125)I]FIAU) or 3'-deoxy-3'-[(18)F]fluorothymidine ([(18)F]FLT) were measured. The infection model in BALB/c mice was imaged with [(125)I]FIAU or [(18)F]FLT using small-animal Single Photon Emission Computed Tomography (SPECT) or Positron Emission Tomography (PET), respectively. The accumulated radioactivity of [(125)I]FIAU or [(18)F]FLT in the two strains showed a linearly increased pattern with increasing incubation time or bacterial numbers. The image clearly demonstrated a high uptake of [(125)I]FIAU and [(18)F]FLT in the bacterial infection site. [(18)F]FLT uptake in the infection site of was 7.286±2.405, whereas that in the uninfected site was 0.519±0.561. The relative activity ratio of the infected region in relation to the uninfected region was 2.98 at 4h after an injection with [(125)I]FIAU determined by biodistribution data. In conclusion, the bacterial tk activity was confirmed by the cellular uptake and imaging with [(125)I]FIAU or [(18)F]FLT. Therefore, a localized bacterial infection in living mice can be monitored using radioisotope-labeled nucleosides with a nuclear medicine imaging modality.
Collapse
Affiliation(s)
- Su Jin Jang
- Molecular Imaging Research Center, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zhang T, Li SY, Nuermberger EL. Autoluminescent Mycobacterium tuberculosis for rapid, real-time, non-invasive assessment of drug and vaccine efficacy. PLoS One 2012; 7:e29774. [PMID: 22253776 PMCID: PMC3256174 DOI: 10.1371/journal.pone.0029774] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Accepted: 12/04/2011] [Indexed: 11/19/2022] Open
Abstract
Preclinical efforts to discover and develop new drugs and vaccines for tuberculosis are hampered by the reliance on colony-forming unit (CFU) counts as primary outcomes for in vivo efficacy studies and the slow growth of Mycobacterium tuberculosis. The utility of bioluminescent M. tuberculosis reporter strains for real-time in vitro and ex vivo assessment of drug and vaccine activity has been demonstrated but a simple, non-invasive, real-time surrogate marker to replace CFU counts for real-time evaluation of drug and vaccine efficacy in vivo has not been described. We describe the development of a fully virulent and stable autoluminescent strain of M. tuberculosis and proof-of-concept experiments demonstrating its utility for in vivo bioluminescence imaging to assess the efficacy of new drugs and vaccines for tuberculosis in a mouse model. Relative light unit (RLU) counts paralleled CFU counts during the active phase of bacterial growth, with a lower limit of detection of approximately 106 CFU in live, anesthetized mice. Experiments distinguishing active from inactive anti-tuberculosis drugs and bacteriostatic drug effects from bactericidal effects were completed in less than 5 days. The ability of a recombinant BCG vaccine to limit bacterial growth was demonstrated within 3 weeks. Use of this autoluminescent reporter strain has the potential to drastically reduce the time, effort, animals and costs consumed in the evaluation of drug activity in vitro and the in vivo assessment of drug and vaccine efficacy.
Collapse
Affiliation(s)
- Tianyu Zhang
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, People's Republic of China
| | - Si-Yang Li
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Eric L. Nuermberger
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
47
|
Bray M, Lawler J, Paragas J, Jahrling PB, Mollura DJ. Molecular imaging of influenza and other emerging respiratory viral infections. J Infect Dis 2011; 203:1348-59. [PMID: 21422476 PMCID: PMC3080905 DOI: 10.1093/infdis/jir038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Research on the pathogenesis and therapy of influenza and other emerging respiratory viral infections would be aided by methods that directly visualize pathophysiologic processes in patients and laboratory animals. At present, imaging of diseases, such as swine-origin H1N1 influenza, is largely restricted to chest radiograph and computed tomography (CT), which can detect pulmonary structural changes in severely ill patients but are more limited in characterizing the early stages of illness, differentiating inflammation from infection or tracking immune responses. In contrast, imaging modalities, such as positron emission tomography, single photon emission CT, magnetic resonance imaging, and bioluminescence imaging, which have become useful tools for investigating the pathogenesis of a range of disease processes, could be used to advance in vivo studies of respiratory viral infections in patients and animals. Molecular techniques might also be used to identify novel biomarkers of disease progression and to evaluate new therapies.
Collapse
Affiliation(s)
- Mike Bray
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | |
Collapse
|
48
|
Abstract
An organism requires a range of biomolecules for its growth. By definition, these are essential molecules which constitute the basic metabolic requirements of an organism. A small organic molecule with chemical similarity to that of an essential metabolite may bind to the enzyme that catalyzes its production and inhibit it, likely resulting in the stasis or death of the organism. Here, we report a high-throughput approach for identifying essential metabolites of an organism using genetic and biochemical approaches and then implement computational approaches to identify metabolite mimics. We generated and genotyped 5,126 Mycobacterium tuberculosis mutants and performed a statistical analysis to determine putative essential genes. The essential molecules of M. tuberculosis were classified as products of enzymes that are encoded by genes in this list. Although incomplete, as many enzymes of M. tuberculosis have yet to be identified and characterized, this is the first report of a large number of essential molecules of the organism. We identified essential metabolites of three distinct metabolic pathways in M. tuberculosis and selected molecules with chemical similarity using cheminformatics strategies that illustrate a variety of different pharmacophores. Our approach is aimed at systematic identification of essential molecules and their mimics as a blueprint for development of effective chemical probes of M. tuberculosis metabolism, with the ultimate goal of seeking drugs that can kill this pathogen. As an illustration of this approach, we report that compounds JFD01307SC and l-methionine-S-sulfoximine, which share chemical similarity with an essential molecule of M. tuberculosis, inhibited the growth of this organism at micromolar concentrations.
Collapse
|
49
|
Peterson RA, Gabrielson KL, Allan Johnson G, Pomper MG, Coatney RW, Winkelmann CT. Continuing education course #1: non-invasive imaging as a problem-solving tool and translational biomarker strategy in toxicologic pathology. Toxicol Pathol 2010; 39:267-72. [PMID: 21147931 DOI: 10.1177/0192623310390392] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The continuing education course "Non-Invasive Imaging as a Problem-Solving Tool and Translational Biomarker Strategy in Toxicologic Pathology" provided a thorough overview of commonly used imaging modalities and the logistics required for integration of small animal imaging into toxicologic pathology. Non-invasive imaging (NIN) is gaining acceptance as an important modality in toxicologic pathology. This technology allows nonterminal, time-course evaluation of functional and morphologic endpoints and can be used to translate biomarkers between preclinical animal models and human patients. NIN can support drug development as well as basic research in academic or industrial environments. An initial overview of theoretical principles was followed by focused presentations on magnetic resonance imaging (MRI)/magnetic resonance microscopy (MRM), positron emission tomography (PET)/single proton emission computed tomography (SPECT), ultrasonography (US, primarily focused on echocardiography), optical (bioluminescent) imaging, and computed tomography (CT). The choice of imaging modality will depend on the research question and the needed resolution.
Collapse
Affiliation(s)
- Richard A Peterson
- GlaxoSmithKline Safety Assessment, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Using bioluminescence to monitor treatment response in real time in mice with Mycobacterium ulcerans infection. Antimicrob Agents Chemother 2010; 55:56-61. [PMID: 21078940 DOI: 10.1128/aac.01260-10] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium ulcerans causes Buruli ulcer, a potentially disabling ulcerative skin disease. Only recently was antimicrobial therapy proven effective. Treatment for 2 months with rifampin plus streptomycin was first proposed after experiments in the mouse footpad model demonstrated bactericidal activity. This treatment is now considered the treatment of choice, although larger ulcers may require adjunctive surgery. Shorter, oral regimens are desired, but evaluating drug activity in mice is hampered by the very slow growth of M. ulcerans, which takes 3 months to produce countable colonies. We created a recombinant bioluminescent M. ulcerans strain expressing luxAB from Vibrio harveyi for real-time evaluation of antimicrobial effects in vivo. Mouse footpads were injected with wild-type M. ulcerans 1059 (WtMu) or the recombinant bioluminescent strain (rMu). Two weeks later, mice received rifampin plus streptomycin, kanamycin alone (to which rMu is resistant), or streptomycin alone for 4 weeks and were observed for footpad swelling (preventive model). Untreated controls and kanamycin-treated rMu-infected mice received rifampin plus streptomycin for 4 weeks after developing footpad swelling (curative model). Compared to WtMu, rMu exhibited similar growth and virulence in vivo and similar drug susceptibility. A good correlation was observed between luminescence (measured as relative light units) and number of viable bacteria (measured by CFU) in footpad homogenates. Proof of concept was also shown for serial real-time evaluation of drug activity in live mice. These results indicate the potential of bioluminescence as a real-time surrogate marker for viable bacteria in mouse footpads to accelerate the identification of new treatments for Buruli ulcer.
Collapse
|