1
|
Moreira L, Guimarães NM, Santos RS, Loureiro JA, Pereira MDC, Azevedo NF. Oligonucleotide probes for imaging and diagnosis of bacterial infections. Crit Rev Biotechnol 2025; 45:128-147. [PMID: 38830823 DOI: 10.1080/07388551.2024.2344574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/17/2023] [Indexed: 06/05/2024]
Abstract
The rise of infectious diseases as a public health concern has necessitated the development of rapid and precise diagnostic methods. Imaging techniques like nuclear and optical imaging provide the ability to diagnose infectious diseases within the body, eliminating delays caused by sampling and pre-enrichments of clinical samples and offering spatial information that can aid in a more informed diagnosis. Traditional molecular probes are typically created to image infected tissue without accurately identifying the pathogen. In contrast, oligonucleotides can be tailored to target specific RNA sequences, allowing for the identification of pathogens, and even generating antibiotic susceptibility profiles by focusing on drug resistance genes. Despite the benefits that nucleic acid mimics (NAMs) have provided in terms of stabilizing oligonucleotides, the inadequate delivery of these relatively large molecules into the cytoplasm of bacteria remains a challenge for widespread use of this technology. This review summarizes the key advancements in the field of oligonucleotide probes for in vivo imaging, highlighting the most promising delivery systems described in the literature for developing optical imaging through in vivo hybridization.
Collapse
Affiliation(s)
- Luís Moreira
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Nuno Miguel Guimarães
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Rita Sobral Santos
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Joana Angélica Loureiro
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Maria do Carmo Pereira
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Nuno Filipe Azevedo
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| |
Collapse
|
2
|
Narh JK, Casillas-Vega NG, Zarate X. LL-37_Renalexin hybrid peptide exhibits antimicrobial activity at lower MICs than its counterpart single peptides. Appl Microbiol Biotechnol 2024; 108:126. [PMID: 38229302 PMCID: PMC10787891 DOI: 10.1007/s00253-023-12887-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/16/2023] [Accepted: 11/25/2023] [Indexed: 01/18/2024]
Abstract
An alarming global public health and economic peril has been the emergence of antibiotic resistance resulting from clinically relevant bacteria pathogens, including Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumonia, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species constantly exhibiting intrinsic and extrinsic resistance mechanisms against last-resort antibiotics like gentamycin, ciprofloxacin, tetracycline, colistin, and standard ampicillin prescription in clinical practices. The discovery and applications of antimicrobial peptides (AMPs) with antibacterial properties have been considered and proven as alternative antimicrobial agents to antibiotics. In this study, we have designed, produced, and purified a recombinant novel multifunctional hybrid antimicrobial peptide LL-37_Renalexin for the first time via the application of newly designed flexible GS peptide linker coupled with the use of our previously characterized small metal-binding proteins SmbP and CusF3H+ as carrier proteins that allow for an enhanced bacterial expression, using BL21(DE3) and SHuffle T7(DE3) Escherichia coli strains, and purification of the hybrid peptide via immobilized metal affinity chromatography. The purified tag-free LL-37_Renalexin hybrid peptide exhibited above 85% reduction in bacteria colony-forming units and broad-spectrum antimicrobial effects against Staphylococcus aureus, Escherichia coli, Methicillin-resistant Staphylococcus aureus (MRSA), and Klebsiella pneumoniae bacteria clinical isolates at a lower minimum inhibition concentration level (10-33 μM) as compared to its counterpart single-AMPs LL-37 and Renalexin (50-100 μM). KEY POINTS: • The hybrid antimicrobial peptide LL-37_Renalexin has been designed using a GS linker. • The peptide was expressed with the carrier proteins SmbP and CusF3H+. • The hybrid peptide shows antibacterial potency against clinical bacterial isolates.
Collapse
Affiliation(s)
- Julius Kwesi Narh
- Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Avenida Universidad s/n, Ciudad Universitaria, 66455, San Nicolas de los Garza, NL, Mexico
| | - Nestor G Casillas-Vega
- Departamento de Patologia Clinica, Hospital Universitario Dr. Jose Eleuterio Gonzalez, Universidad Autonoma de Nuevo Leon, 64460, Monterrey, NL, Mexico
| | - Xristo Zarate
- Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Avenida Universidad s/n, Ciudad Universitaria, 66455, San Nicolas de los Garza, NL, Mexico.
| |
Collapse
|
3
|
Weiss L, Mirloup A, Blondé L, Manko H, Peluso J, Bonnet D, Dziuba D, Karpenko J. Fluorescent Antimicrobial Peptides Based on Nile Red: Effect of Conjugation Site and Chemistry on Wash-Free Staining of Bacteria. Bioconjug Chem 2024; 35:1779-1787. [PMID: 39435864 DOI: 10.1021/acs.bioconjchem.4c00331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Fluorescent probes for bacterial detection can be obtained by conjugating antimicrobial peptides with fluorescent dyes. However, little is known about the effect of the conjugation site and linker chemistry on staining efficiency. We synthesized three conjugates of the antimicrobial peptide ubiquicidin with the environmentally sensitive fluorophore Nile Red that differed by the attachment site and the chemical composition of the linker. We showed that incorporating fluorophore as a minimalistic non-natural amino acid resulted in a superior probe compared with the typically used bioconjugation approaches. The new peptide-based probe named UNR-1 displayed red fluorescence and enabled robust wash-free staining of Gram-positive and Gram-negative bacteria. The probe exhibited selectivity over mammalian cells and enabled rapid fluorescence detection of bacteria by fluorescence microscopy and flow cytometry in an add-and-read format. Our results may foster the development of next-generation fluorescent AMPs for clinical laboratory diagnostics and medical imaging.
Collapse
Affiliation(s)
- Lucille Weiss
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200 CNRS/Université de Strasbourg, Institut du Médicament de Strasbourg, Strasbourg F-67000, France
| | - Antoine Mirloup
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200 CNRS/Université de Strasbourg, Institut du Médicament de Strasbourg, Strasbourg F-67000, France
| | - Léa Blondé
- Plate-forme eBioCyt - UPS 1401, Faculté de Pharmacie, Université de Strasbourg, Strasbourg F-67000, France
| | - Hanna Manko
- Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, UMR 7021 CNRS/Université de Strasbourg, Strasbourg F-67000, France
| | - Jean Peluso
- Plate-forme eBioCyt - UPS 1401, Faculté de Pharmacie, Université de Strasbourg, Strasbourg F-67000, France
| | - Dominique Bonnet
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200 CNRS/Université de Strasbourg, Institut du Médicament de Strasbourg, Strasbourg F-67000, France
| | - Dmytro Dziuba
- Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, UMR 7021 CNRS/Université de Strasbourg, Strasbourg F-67000, France
| | - Julie Karpenko
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200 CNRS/Université de Strasbourg, Institut du Médicament de Strasbourg, Strasbourg F-67000, France
| |
Collapse
|
4
|
Koatale P, Welling MM, Ndlovu H, Kgatle M, Mdanda S, Mdlophane A, Okem A, Takyi-Williams J, Sathekge MM, Ebenhan T. Insights into Peptidoglycan-Targeting Radiotracers for Imaging Bacterial Infections: Updates, Challenges, and Future Perspectives. ACS Infect Dis 2024; 10:270-286. [PMID: 38290525 PMCID: PMC10862554 DOI: 10.1021/acsinfecdis.3c00443] [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: 08/28/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 02/01/2024]
Abstract
The unique structural architecture of the peptidoglycan allows for the stratification of bacteria as either Gram-negative or Gram-positive, which makes bacterial cells distinguishable from mammalian cells. This classification has received attention as a potential target for diagnostic and therapeutic purposes. Bacteria's ability to metabolically integrate peptidoglycan precursors during cell wall biosynthesis and recycling offers an opportunity to target and image pathogens in their biological state. This Review explores the peptidoglycan biosynthesis for bacteria-specific targeting for infection imaging. Current and potential radiolabeled peptidoglycan precursors for bacterial infection imaging, their development status, and their performance in vitro and/or in vivo are highlighted. We conclude by providing our thoughts on how to shape this area of research for future clinical translation.
Collapse
Affiliation(s)
- Palesa
C. Koatale
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Mick M. Welling
- Interventional
Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Honest Ndlovu
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Mankgopo Kgatle
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Sipho Mdanda
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Amanda Mdlophane
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Ambrose Okem
- Department
of Anaesthesia, School of Clinical Medicine, University of Witwatersrand, 2050 Johannesburg, South Africa
| | - John Takyi-Williams
- Pharmacokinetic
and Mass Spectrometry Core, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mike M. Sathekge
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
| | - Thomas Ebenhan
- Department
of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South Africa
- Nuclear
Medicine Research Infrastructure (NuMeRI) NPC, 0001 Pretoria, South Africa
- DSI/NWU Pre-clinical
Drug Development Platform, North West University, 2520 Potchefstroom, South Africa
| |
Collapse
|
5
|
Abbasali Z, Pirestani M, Dalimi A, Badri M, Fasihi-Ramandi M. Anti-parasitic activity of a chimeric peptide Cecropin A (2-8)-Melittin (6-9) (CM11) against tachyzoites of Toxoplasma gondii and the BALB/c mouse model of acute toxoplasmosis. Mol Biochem Parasitol 2023; 255:111578. [PMID: 37348706 DOI: 10.1016/j.molbiopara.2023.111578] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/18/2023] [Accepted: 06/18/2023] [Indexed: 06/24/2023]
Abstract
Toxoplasmosis is a zoonotic disease that infects most animals, including humans. Pyrimethamine/sulfadiazine is the standard treatment for toxoplasmosis. Although this treatment has been successful, it is often associated with side effects that cannot be tolerated. Therefore, various compounds have been proposed as alternative treatments for toxoplasmosis. Antimicrobial peptides (AMPs) act on various pathogens, from viruses to protozoa. The purpose of the present study was to evaluate the effects of CM11 on in vitro and in vivo Toxoplasma gondii infection. For in vitro experiments, VERO cells were treated with different concentrations of CM11 (1-128 μg/ml) compared to sulfadiazine (SDZ) (0.78-100 μg/ml). MTT and lactate dehydrogenase (LDH) assays evaluated the cell viability and plasma membrane integrity. Then, the inhibitory concentration (IC50) values were determined for treating tachyzoites of T. gondii before or on cells previously infected. Annexin V-FITC/propidium iodide (PI) staining was used to distinguish viable and apoptotic cells. The effect of CM11, SDZ, and a combination of CM11 and SDZ was evaluated in the BALB/c mouse model of acute toxoplasmosis. CM11 was effective on tachyzoites of T. gondii and had a time and dose-dependent manner. The results of the MTT assay showed that the CC50 values of CM11 and SDZ were estimated at 17.4 µg/ml and 62.3 µg/ml after 24-h, respectively. The inhibitory concentration (IC50) of CM11 and SDZ on infected cells was estimated at 1.9 µg/ml and 1.4 µg/ml after 24-h, respectively. The highest rate of apoptosis (early and late) in high concentrations of SDZ and CM11 was determined for tachyzoites (2.13 % and 13.88 %), non-infected VERO cells (6.1 % and 19.76 %), and infected VERO cells (7.45 % and 29.9 %), respectively. Treating infected mice with CM11 and a combination of CM11 and SDZ had increased survival time. Based on the mentioned results, it can be concluded that CM11 has a beneficial effect on tachyzoites of T. gondii in vitro. The result of the mouse model suggests that CM11, either alone or in combination with other chemotherapeutic agents, could be a potential therapeutic for toxoplasmosis. Hence, antimicrobial peptides could be applied as promising anti-toxoplasma agents for treating toxoplasmosis.
Collapse
Affiliation(s)
- Zahra Abbasali
- Department of Parasitology, Faculty of Medical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Majid Pirestani
- Department of Parasitology, Faculty of Medical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Abdolhossein Dalimi
- Department of Parasitology, Faculty of Medical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Milad Badri
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Viana de Freitas T, Karmakar U, Vasconcelos AG, Santos MA, Oliveira do Vale Lira B, Costa SR, Barbosa EA, Cardozo-Fh J, Correa R, Ribeiro DJS, Prates MV, Magalhães KG, Soller Ramada MH, Roberto de Souza Almeida Leite J, Bloch C, Lima de Oliveira A, Vendrell M, Brand GD. Release of immunomodulatory peptides at bacterial membrane interfaces as a novel strategy to fight microorganisms. J Biol Chem 2023; 299:103056. [PMID: 36822328 PMCID: PMC10074799 DOI: 10.1016/j.jbc.2023.103056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 02/23/2023] Open
Abstract
Cationic and amphiphilic peptides can be used as homing devices to accumulate conjugated antibiotics to bacteria-enriched sites and promote efficient microbial killing. However, just as important as tackling bacterial infections, is the modulation of the immune response in this complex microenvironment. In the present report, we designed a peptide chimaera called Chim2, formed by a membrane-active module, an enzyme hydrolysis site and a formyl peptide receptor 2 (FPR2) agonist. This molecule was designed to adsorb onto bacterial membranes, promote their lysis, and upon hydrolysis by local enzymes, release the FPR2 agonist sequence for activation and recruitment of immune cells. We synthesized the isolated peptide modules of Chim2 and characterized their biological activities independently and as a single polypeptide chain. We conducted antimicrobial assays, along with other tests aiming at the analyses of the cellular and immunological responses. In addition, assays using vesicles as models of eukaryotic and prokaryotic membranes were conducted and solution structures of Chim2 were generated by 1H NMR. Chim2 is antimicrobial, adsorbs preferentially to negatively charged vesicles while adopting an α-helix structure and exposes its disorganized tail to the solvent, which facilitates hydrolysis by tryptase-like enzymes, allowing the release of the FPR2 agonist fragment. This fragment was shown to induce accumulation of the cellular activation marker, lipid bodies, in mouse macrophages and the release of immunomodulatory interleukins. In conclusion, these data demonstrate that peptides with antimicrobial and immunomodulatory activities can be considered for further development as drugs.
Collapse
Affiliation(s)
- Thiago Viana de Freitas
- Universidade de Brasília, Instituto de Química, Laboratório de Síntese e Análise de Biomoléculas, LSAB, Brasília, Distrito Federal, Brasil
| | - Utsa Karmakar
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Andreanne G Vasconcelos
- Universidade de Brasília, Faculdade de Medicina, Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Brasília, Distrito Federal, Brasil
| | - Michele A Santos
- Universidade de Brasília, Instituto de Química, Laboratório de Síntese e Análise de Biomoléculas, LSAB, Brasília, Distrito Federal, Brasil; Universidade de Brasília, Instituto de Química, Laboratório de Ressonância Magnética Nuclear, LRMN, Brasília, Distrito Federal, Brasil
| | - Bianca Oliveira do Vale Lira
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brasil; Programa de Pós-Graduação em Gerontologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brasil
| | - Samuel Ribeiro Costa
- Universidade de Brasília, Instituto de Química, Laboratório de Síntese e Análise de Biomoléculas, LSAB, Brasília, Distrito Federal, Brasil
| | - Eder Alves Barbosa
- Universidade de Brasília, Instituto de Química, Laboratório de Síntese e Análise de Biomoléculas, LSAB, Brasília, Distrito Federal, Brasil
| | - José Cardozo-Fh
- Laboratório de Espectrometria de Massa, LEM, Embrapa Recursos Genéticos e Biotecnologia, Brasília, Distrito Federal, Brasil
| | - Rafael Correa
- Universidade de Brasília, Instituto de Biologia, Laboratório de Imunologia e Inflamação, LIMI, Brasília, Distrito Federal, Brasil
| | - Dalila J S Ribeiro
- Universidade de Brasília, Instituto de Biologia, Laboratório de Imunologia e Inflamação, LIMI, Brasília, Distrito Federal, Brasil
| | - Maura Vianna Prates
- Laboratório de Espectrometria de Massa, LEM, Embrapa Recursos Genéticos e Biotecnologia, Brasília, Distrito Federal, Brasil
| | - Kelly G Magalhães
- Universidade de Brasília, Instituto de Biologia, Laboratório de Imunologia e Inflamação, LIMI, Brasília, Distrito Federal, Brasil
| | - Marcelo Henrique Soller Ramada
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brasil; Programa de Pós-Graduação em Gerontologia, Universidade Católica de Brasília, Brasília, Distrito Federal, Brasil
| | - José Roberto de Souza Almeida Leite
- Universidade de Brasília, Faculdade de Medicina, Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Brasília, Distrito Federal, Brasil
| | - Carlos Bloch
- Laboratório de Espectrometria de Massa, LEM, Embrapa Recursos Genéticos e Biotecnologia, Brasília, Distrito Federal, Brasil
| | - Aline Lima de Oliveira
- Universidade de Brasília, Instituto de Química, Laboratório de Ressonância Magnética Nuclear, LRMN, Brasília, Distrito Federal, Brasil
| | - Marc Vendrell
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Guilherme Dotto Brand
- Universidade de Brasília, Instituto de Química, Laboratório de Síntese e Análise de Biomoléculas, LSAB, Brasília, Distrito Federal, Brasil.
| |
Collapse
|
7
|
Terán M, Osorio J, Cardoso E, Tejería E, Paolino A, Reyes AL, Cecchetto G. [ 99m Tc]Tc-HYNIC-EcgDf21: A defensin short analogue with potential application in infection foci imaging. Chem Biol Drug Des 2023; 101:593-604. [PMID: 36138520 DOI: 10.1111/cbdd.14149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/24/2022] [Accepted: 09/19/2022] [Indexed: 11/30/2022]
Abstract
Opportunistic infections are a problem of great relevance in public health and the precise detection and localization of infection in the early stages of the disease is of great importance for patient management as well as cost containment. Our proposal seeks to contribute to developing a new agent that meets the needs of diagnosis and follow-up of fungal and bacterial infections, focused on the design of a radiotracer with the potential for recognition of hidden infection foci. Defensins are plant antimicrobial peptides that not only show activity against plant pathogens but also against human ones. A short analogue of EcgDf1 defensin, EcgDf21d (NH2 -ERFTGGHCRGFRRRCFCTKHC-COOH), was labelled through the formation of a 99m Tc-HYNIC complex which was assessed for physicochemical and biological behaviour both in vitro and in vivo. The [99m Tc]Tc-HYNIC-EcgDf21 labelling procedure rendered a single product with remarkably high RCP and stability in the labelling milieu. The Log p value indicated that [99m Tc]Tc-HYNIC-EcgDf21 has a hydrophilic behaviour, confirmed by the biodistribution profiles. The optimal uptake value was obtained for Candida albicans infection model reaching a lesion/muscle ratio of 3, this correlates with in vitro binding studies, and the lesion can be definitely observed in the scintigraphic images.
Collapse
Affiliation(s)
- Mariella Terán
- Área de Radioquímica, Departamento Estrella Campos, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Jessica Osorio
- Área de Radioquímica, Departamento Estrella Campos, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Elena Cardoso
- Área de Radioquímica, Departamento Estrella Campos, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Emilia Tejería
- Área de Radioquímica, Departamento Estrella Campos, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| | - Andrea Paolino
- Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Ana Laura Reyes
- Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Gianna Cecchetto
- Área de Microbiología, Instituto de Química Biológica, Facultad de Ciencias-Departamento de Biociencias, Facultad de Química, Universidad de la República (Udelar), Montevideo, Uruguay
| |
Collapse
|
8
|
Signore A, Bentivoglio V, Varani M, Lauri C. Current Status of SPECT Radiopharmaceuticals for Specific Bacteria Imaging. Semin Nucl Med 2023; 53:142-151. [PMID: 36609002 DOI: 10.1053/j.semnuclmed.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Imaging infection still represents a challenge for researchers. Despite nuclear medicine (NM) offers valuable tools able to discriminate between infections and inflammation, there is an unmet clinical need to develop new strategies able to specifically target the causative pathogen, to select the best antimicrobial treatment for each patient and to accurately assess therapeutic efficacy. These aspects are commonly addressed by microbiology or histology but the diagnosis often relies on invasive procedures that are prone to contamination or sample bias and do not reflect the spatial heterogeneity of the infective process. Therefore, in the era of personalized medicine and treatment, a lot of efforts are in play to improve a personalized diagnosis. Molecular imaging is an ideal candidate for this purpose and, indeed, research is going fast to this direction aiming to find more selective and proper antimicrobial treatments and to overcome broad-spectrum antibiotic use, which still represents the major cause of bacterial drug-resistance. Several approaches for specifically image bacteria have been proposed and provided encouraging perspectives in preclinical studies. Nevertheless, the majority of these promising approaches are still confined in "bench stages" and crucial issues still need to be addressed before their translation in clinical practice. This review will focus on radiolabeled antibiotics for SPECT imaging of bacteria, their mechanisms of action, their potentiality and limitations for "bed-side" applications.
Collapse
Affiliation(s)
- Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, "Sapienza" University of Rome, Italy.
| | - Valeria Bentivoglio
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, "Sapienza" University of Rome, Italy
| | - Michela Varani
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, "Sapienza" University of Rome, Italy
| | - Chiara Lauri
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, "Sapienza" University of Rome, Italy
| |
Collapse
|
9
|
Marjanovic-Painter B, Kleynhans J, Zeevaart JR, Rohwer E, Ebenhan T. A decade of ubiquicidin development for PET imaging of infection: A systematic review. Nucl Med Biol 2023; 116-117:108307. [PMID: 36435145 DOI: 10.1016/j.nucmedbio.2022.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/25/2022] [Accepted: 11/07/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Ubiquicidin is a peptide fragment with selective binding to negatively charged bacterial cell membranes. Besides its earlier labelling with gamma emitting radionuclides, it has been labelled with Positron Emission Tomography (PET) radionuclides in the last decade for imaging infection and distinguishing infectious disease from sterile inflammation. This systematic review aims to evaluate the technology readiness level of PET based ubiquicidin radiopharmaceuticals. METHODS Two independent researchers reviewed all articles and abstracts pertaining ubiquicidin and PET imaging that are currently available. Scopus, Google Scholar and PubMed/Medline were used in the search. Upon completion of the literature search all articles and abstracts were evaluated and duplicates were excluded. All non-PET articles as well as review articles without new data were deemed ineligible. RESULTS From a total of 17 papers and 10 abstracts the studies were grouped into development, preclinical and clinical studies. Development was published in 15/17 (88%) publications and 6/10 (60%) abstracts, preclinical applications in 9/17 (53%) publications and 1/10 (10%) of abstracts. Finally, clinical studies made up 6/17 (35%) of full publications and 4/10 (40%) of the available abstracts. Development results were the most abundant. All the findings in the different areas of development of ubiquicidin as PET radiopharmaceutical are summarized in this paper. CONCLUSION Labelling procedures are generally uncomplicated and relatively fast and there are indications of adequate product stability. The production of PET radiopharmaceuticals based on UBI will therefore not be a barrier for clinical introduction of this technology. Systematization and unification of criteria for preclinical imaging and larger clinical trials are needed to ensure the translation of this radiopharmaceutical into the clinic. Therefore a conclusion with regards to the clinical relevance of ubiquicidin based PET is not yet possible.
Collapse
Affiliation(s)
| | - Janke Kleynhans
- Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
| | - Jan Rijn Zeevaart
- Radiochemistry, The South African Nuclear Energy Corporation, Pelindaba, South Africa; Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
| | - Egmont Rohwer
- Department of Chemistry, University of Pretoria, Pretoria, South Africa
| | - Thomas Ebenhan
- Radiochemistry, The South African Nuclear Energy Corporation, Pelindaba, South Africa; Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa; Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa.
| |
Collapse
|
10
|
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: 0.7] [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
|
11
|
Rima M, Rima M, Fajloun Z, Sabatier JM, Bechinger B, Naas T. Antimicrobial Peptides: A Potent Alternative to Antibiotics. Antibiotics (Basel) 2021; 10:1095. [PMID: 34572678 PMCID: PMC8466391 DOI: 10.3390/antibiotics10091095] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 01/07/2023] Open
Abstract
Antimicrobial peptides constitute one of the most promising alternatives to antibiotics since they could be used to treat bacterial infections, especially those caused by multidrug-resistant pathogens. Many antimicrobial peptides, with various activity spectra and mechanisms of actions, have been described. This review focuses on their use against ESKAPE bacteria, especially in biofilm treatments, their synergistic activity, and their application as prophylactic agents. Limitations and challenges restricting therapeutic applications are highlighted, and solutions for each challenge are evaluated to analyze whether antimicrobial peptides could replace antibiotics in the near future.
Collapse
Affiliation(s)
- Mariam Rima
- Team ReSIST, INSERM U1184, School of Medicine Université Paris-Saclay, 94270 Le Kremlin-Bicetre, France;
| | - Mohamad Rima
- Laboratory of Applied Biotechnology, Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon; (M.R.); (Z.F.)
| | - Ziad Fajloun
- Laboratory of Applied Biotechnology, Azm Center for Research in Biotechnology and Its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon; (M.R.); (Z.F.)
- Department of Biology, Faculty of Sciences III, Lebanese University, Tripoli 1300, Lebanon
| | - Jean-Marc Sabatier
- Institut de Neuro Physiopathologie, UMR7051, Aix-Marseille Université, Faculté de Pharmacie, 27 Boulevard Jean Moulin, 13005 Marseille, France
| | - Burkhard Bechinger
- Institut de Chimie de Strasbourg, CNRS, UMR7177, University of Strasbourg, 67008 Strasbourg, France;
- Institut Universitaire de France (IUF), 75005 Paris, France
| | - Thierry Naas
- Team ReSIST, INSERM U1184, School of Medicine Université Paris-Saclay, 94270 Le Kremlin-Bicetre, France;
- Bacteriology-Hygiene Unit, Assistance Publique/Hôpitaux de Paris, Bicêtre Hospital, 94270 Le Kremlin-Bicetre, France
- French National Reference Centre for Antibiotic Resistance: Carbapenemase-Producing Enterobacterales, 94270 Le Kremlin-Bicetre, France
| |
Collapse
|
12
|
Ankrah AO, Sathekge MM, Dierckx RAJO, Glaudemans AWJM. Radionuclide Imaging of Fungal Infections and Correlation with the Host Defense Response. J Fungi (Basel) 2021; 7:jof7060407. [PMID: 34067410 PMCID: PMC8224611 DOI: 10.3390/jof7060407] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/10/2021] [Accepted: 05/21/2021] [Indexed: 12/19/2022] Open
Abstract
The human response to invading fungi includes a series of events that detect, kill, or clear the fungi. If the metabolic host response is unable to eliminate the fungi, an infection ensues. Some of the host response’s metabolic events to fungi can be imaged with molecules labelled with radionuclides. Several important clinical applications have been found with radiolabelled biomolecules of inflammation. 18F-fluorodeoxyglucose is the tracer that has been most widely investigated in the host defence of fungi. This tracer has added value in the early detection of infection, in staging and visualising dissemination of infection, and in monitoring antifungal treatment. Radiolabelled antimicrobial peptides showed promising results, but large prospective studies in fungal infection are lacking. Other tracers have also been used in imaging events of the host response, such as the migration of white blood cells at sites of infection, nutritional immunity in iron metabolism, and radiolabelled monoclonal antibodies. Many tracers are still at the preclinical stage. Some tracers require further studies before translation into clinical use. The application of therapeutic radionuclides offers a very promising clinical application of these tracers in managing drug-resistant fungi.
Collapse
Affiliation(s)
- Alfred O. Ankrah
- National Centre for Radiotherapy Oncology and Nuclear Medicine, Korle Bu Teaching Hospital, Accra GA-222 7974, Ghana;
- Department of Nuclear Medicine, University of Pretoria, Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Medical Imaging Center, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Mike M. Sathekge
- Department of Nuclear Medicine, University of Pretoria, Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Rudi A. J. O. Dierckx
- Medical Imaging Center, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Andor W. J. M. Glaudemans
- Medical Imaging Center, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
- Correspondence:
| |
Collapse
|
13
|
Radiolabeled Complex of Antimicrobial Peptides UBI29-41 and UBI18-35 Labeled with 99mTc for Differential Diagnosis of Bone Infection of the Limbs. Bull Exp Biol Med 2021; 170:415-419. [PMID: 33713227 DOI: 10.1007/s10517-021-05078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Indexed: 10/21/2022]
Abstract
We studied an original radiolabeled complex of antimicrobial peptides UBI29-41 and UBI18-35, ubiquicidin derivatives, for distinguishing between bacterial and aseptic inflammation. For radiolabeling of the peptides with technetium-99m, a bifunctional chelating agent succinimide-1-yl 6-(bis(pyridin-2-ylmethyl)amino)hexanoate was used. The obtained complexes 99mТс-DPAH-UBI29-41 and 99mТс-DPAH-UBI18-35 had radiolabeling yield >80% and radiochemical purity >96%. Accumulation of the complexes in the focus of bacterial inflammation in bone structures and the absence of this complex in the site of aseptic inflammation was confirmed in a rat model of traumatic osteomyelitis by single-photon emission computed tomography.
Collapse
|
14
|
Pardoux É, Boturyn D, Roupioz Y. Antimicrobial Peptides as Probes in Biosensors Detecting Whole Bacteria: A Review. Molecules 2020; 25:E1998. [PMID: 32344585 PMCID: PMC7221689 DOI: 10.3390/molecules25081998] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/17/2022] Open
Abstract
Bacterial resistance is becoming a global issue due to its rapid growth. Potential new drugs as antimicrobial peptides (AMPs) are considered for several decades as promising candidates to circumvent this threat. Nonetheless, AMPs have also been used more recently in other settings such as molecular probes grafted on biosensors able to detect whole bacteria. Rapid, reliable and cost-efficient diagnostic tools for bacterial infection could prevent the spread of the pathogen from the earliest stages. Biosensors based on AMPs would enable easy monitoring of potentially infected samples, thanks to their powerful versatility and integrability in pre-existent settings. AMPs, which show a broad spectrum of interactions with bacterial membranes, can be tailored in order to design ubiquitous biosensors easily adaptable to clinical settings. This review aims to focus on the state of the art of AMPs used as the recognition elements of whole bacteria in label-free biosensors with a particular focus on the characteristics obtained in terms of threshold, volume of sample analysable and medium, in order to assess their workability in real-world applications.
Collapse
Affiliation(s)
- Éric Pardoux
- Univ. Grenoble Alpes, CNRS, CEA, IRIG, SyMMES, 38000 Grenoble, France;
- Univ. Grenoble Alpes, CNRS, DCM, 38000 Grenoble, France;
| | - Didier Boturyn
- Univ. Grenoble Alpes, CNRS, DCM, 38000 Grenoble, France;
| | - Yoann Roupioz
- Univ. Grenoble Alpes, CNRS, CEA, IRIG, SyMMES, 38000 Grenoble, France;
| |
Collapse
|
15
|
Silindir-Gunay M, Ozer AY. 99mTc-radiolabeled Levofloxacin and micelles as infection and inflammation imaging agents. J Drug Deliv Sci Technol 2020; 56:101571. [PMID: 32288835 PMCID: PMC7104933 DOI: 10.1016/j.jddst.2020.101571] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 01/01/2023]
Abstract
Easy and early detection of infection and inflammation is essential for early and effective treatment. In this study, PEGylated micelles were designed and both micelles and Levofloxacin were radiolabeled with 99mTcO4 - to develop potential radiotracers for detection of infection/inflammation. Radiolabeling efficiency, in vitro stability and bacterial binding of 99mTc-Levofloxacin and 99mTc-micelles were compared. The aim of this study is to formulate and compare 99mTc-Levofloxacin and 99mTc-micelles as infection and inflammation agents having different mechanisms for the accumulation at infection and inflammation site. PEGylated micelles were designed with a particle size of 80 ± 0.7 nm and proper characterization properties. High radiolabeling efficiency was achieved for 99mTc-Levofloxacin (96%) and 99mTc-micelles (87%). The radiolabeling efficiency was remained stable with some insignificant alterations for both radiotracers at 25 °C for 24 h. Although in vitro bacterial binding of 99mTc-levofloxacine was higher than 99mTc-micelles, 99mTc-micelles may also be evaluated potential agent due to long circulation and passive accumulation mechanisms at infection/inflammation site. Both radiopharmaceutical agents exhibit potential results in design, characterization, radiolabeling efficiency and in vitro bacterial binding point of view.
Collapse
Affiliation(s)
- Mine Silindir-Gunay
- Hacettepe University, Faculty of Pharmacy, Department of Radiopharmacy, 06100, Sıhhiye, Ankara, Turkey
| | - Asuman Yekta Ozer
- Hacettepe University, Faculty of Pharmacy, Department of Radiopharmacy, 06100, Sıhhiye, Ankara, Turkey
| |
Collapse
|
16
|
Giraudo C, Evangelista L, Fraia AS, Lupi A, Quaia E, Cecchin D, Casali M. Molecular Imaging of Pulmonary Inflammation and Infection. Int J Mol Sci 2020; 21:ijms21030894. [PMID: 32019142 PMCID: PMC7037834 DOI: 10.3390/ijms21030894] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
Infectious and inflammatory pulmonary diseases are a leading cause of morbidity and mortality worldwide. Although infrequently used in this setting, molecular imaging may significantly contribute to their diagnosis using techniques like single photon emission tomography (SPET), positron emission tomography (PET) with computed tomography (CT) or magnetic resonance imaging (MRI) with the support of specific or unspecific radiopharmaceutical agents. 18F-Fluorodeoxyglucose (18F-FDG), mostly applied in oncological imaging, can also detect cells actively involved in infectious and inflammatory conditions, even if with a low specificity. SPET with nonspecific (e.g., 67Gallium-citrate (67Ga citrate)) and specific tracers (e.g., white blood cells radiolabeled with 111Indium-oxine (111In) or 99mTechnetium (99mTc)) showed interesting results for many inflammatory lung diseases. However, 67Ga citrate is unfavorable by a radioprotection point of view while radiolabeled white blood cells scan implies complex laboratory settings and labeling procedures. Radiolabeled antibiotics (e.g., ciprofloxacin) have been recently tested, although they seem to be quite unspecific and cause antibiotic resistance. New radiolabeled agents like antimicrobic peptides, binding to bacterial cell membranes, seem very promising. Thus, the aim of this narrative review is to provide a comprehensive overview about techniques, including PET/MRI, and tracers that can guide the clinicians in the appropriate diagnostic pathway of infectious and inflammatory pulmonary diseases.
Collapse
Affiliation(s)
- Chiara Giraudo
- Department of Medicine-DIMED,Institute of Radiology, University of Padova, 35100 Padova, Italy; (A.S.F.); (A.L.); (E.Q.)
- Correspondence: ; Tel.: +39-049-821-2357; Fax: +39-049-821-1878
| | - Laura Evangelista
- Nuclear Medicine Unit, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy; (L.E.); (D.C.)
| | - Anna Sara Fraia
- Department of Medicine-DIMED,Institute of Radiology, University of Padova, 35100 Padova, Italy; (A.S.F.); (A.L.); (E.Q.)
| | - Amalia Lupi
- Department of Medicine-DIMED,Institute of Radiology, University of Padova, 35100 Padova, Italy; (A.S.F.); (A.L.); (E.Q.)
| | - Emilio Quaia
- Department of Medicine-DIMED,Institute of Radiology, University of Padova, 35100 Padova, Italy; (A.S.F.); (A.L.); (E.Q.)
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine-DIMED, University of Padova, 35128 Padova, Italy; (L.E.); (D.C.)
- Padova Neuroscience Center (PNC), University of Padova, 35131 Padova, Italy
| | - Massimiliano Casali
- Azienda Unità Sanitaria Locale–IRCCS di Reggio Emilia, 42121 Reggio Emilia, Italy;
| |
Collapse
|
17
|
Cho SY, Rowe SP, Jain SK, Schon LC, Yung RC, Nayfeh TA, Bingham CO, Foss CA, Nimmagadda S, Pomper MG. Evaluation of Musculoskeletal and Pulmonary Bacterial Infections With [ 124I]FIAU PET/CT. Mol Imaging 2020; 19:1536012120936876. [PMID: 32598214 PMCID: PMC7325456 DOI: 10.1177/1536012120936876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Imaging is limited in the evaluation of bacterial infection. Direct imaging of in situ bacteria holds promise for noninvasive diagnosis. We investigated the ability of a bacterial thymidine kinase inhibitor ([124I]FIAU) to image pulmonary and musculoskeletal infections. METHODS Thirty-three patients were prospectively accrued: 16 with suspected musculoskeletal infection, 14 with suspected pulmonary infection, and 3 with known rheumatoid arthritis without infection. Thirty-one patients were imaged with [124I]FIAU PET/CT and 28 with [18F]FDG PET/CT. Patient histories were reviewed by an experienced clinician with subspecialty training in infectious diseases and were determined to be positive, equivocal, or negative for infection. RESULTS Sensitivity, specificity, positive-predictive value, negative-predictive value, and accuracy of [124I]FIAU PET/CT for diagnosing infection were estimated as 7.7% to 25.0%, 0.0%, 50%, 0.0%, and 20.0% to 71.4% for musculoskeletal infections and incalculable-100.0%, 51.7% to 72.7%, 0.0% to 50.0%, 100.0%, and 57.1% to 78.6% for pulmonary infections, respectively. The parameters for [18F]FDG PET/CT were 75.0% to 92.3%, 0.0%, 23.1% to 92.3%, 0.0%, and 21.4% to 85.7%, respectively, for musculoskeletal infections and incalculable to 100.0%, 0.0%, 0.0% to 18.2%, incalculable, and 0.0% to 18.2% for pulmonary infections, respectively. CONCLUSIONS The high number of patients with equivocal clinical findings prevented definitive conclusions from being made regarding the diagnostic efficacy of [124I]FIAU. Future studies using microbiology to rigorously define infection in patients and PET radiotracers optimized for image quality are needed.
Collapse
Affiliation(s)
- Steve Y. Cho
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanjay K. Jain
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lew C. Schon
- Department of Orthopedic Surgery, MedStar Union Memorial Hospital, Baltimore, MD, USA
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rex C. Yung
- Division of Pulmonary Medicine and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Clifton O. Bingham
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Catherine A. Foss
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sridhar Nimmagadda
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Northrup JD, Mach RH, Sellmyer MA. Radiochemical Approaches to Imaging Bacterial Infections: Intracellular versus Extracellular Targets. Int J Mol Sci 2019; 20:E5808. [PMID: 31752318 PMCID: PMC6888724 DOI: 10.3390/ijms20225808] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 02/03/2023] Open
Abstract
The discovery of penicillin began the age of antibiotics, which was a turning point in human healthcare. However, to this day, microbial infections are still a concern throughout the world, and the rise of multidrug-resistant organisms is an increasing challenge. To combat this threat, diagnostic imaging tools could be used to verify the causative organism and curb inappropriate use of antimicrobial drugs. Nuclear imaging offers the sensitivity needed to detect small numbers of bacteria in situ. Among nuclear imaging tools, radiolabeled antibiotics traditionally have lacked the sensitivity or specificity necessary to diagnose bacterial infections accurately. One reason for the lack of success is that the antibiotics were often chelated to a radiometal. This was done without addressing the ramifications of how the radiolabeling would impact probe entry to the bacterial cell, or the mechanism of binding to an intracellular target. In this review, we approach bacterial infection imaging through the lens of bacterial specific molecular targets, their intracellular or extracellular location, and discuss radiochemistry strategies to guide future probe development.
Collapse
Affiliation(s)
- Justin D. Northrup
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.D.N.); (R.H.M.)
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert H. Mach
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.D.N.); (R.H.M.)
| | - Mark A. Sellmyer
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.D.N.); (R.H.M.)
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
19
|
|
20
|
Evaluating the potential of kit-based 68Ga-ubiquicidin formulation in diagnosis of infection. Nucl Med Commun 2019; 40:228-234. [DOI: 10.1097/mnm.0000000000000943] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Selectivity of Antimicrobial Peptides: A Complex Interplay of Multiple Equilibria. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1117:175-214. [DOI: 10.1007/978-981-13-3588-4_11] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
22
|
Production of the recombinant antimicrobial peptide UBI 18-35 in Escherichia coli. Protein Expr Purif 2018; 143:38-44. [DOI: 10.1016/j.pep.2017.10.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/18/2017] [Accepted: 10/18/2017] [Indexed: 11/21/2022]
|
23
|
Prospective of 68Ga Radionuclide Contribution to the Development of Imaging Agents for Infection and Inflammation. CONTRAST MEDIA & MOLECULAR IMAGING 2018. [PMID: 29531507 PMCID: PMC5817300 DOI: 10.1155/2018/9713691] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During the last decade, the utilization of 68Ga for the development of imaging agents has increased considerably with the leading position in the oncology. The imaging of infection and inflammation is lagging despite strong unmet medical needs. This review presents the potential routes for the development of 68Ga-based agents for the imaging and quantification of infection and inflammation in various diseases and connection of the diagnosis to the treatment for the individualized patient management.
Collapse
|
24
|
Censullo A, Vijayan T. Using Nuclear Medicine Imaging Wisely in Diagnosing Infectious Diseases. Open Forum Infect Dis 2017; 4:ofx011. [PMID: 28480283 PMCID: PMC5414026 DOI: 10.1093/ofid/ofx011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/23/2017] [Indexed: 02/06/2023] Open
Abstract
In recent years, there has been an increasing emphasis on efficient and accurate diagnostic testing, exemplified by the American Board of Internal Medicine’s “Choosing Wisely” campaign. Nuclear imaging studies can provide early and accurate diagnoses of many infectious disease syndromes, particularly in complex cases where the differential remains broad. This review paper offers clinicians a rational, evidence-based guide to approaching nuclear medicine tests, using an example case of methicillin-sensitive Staphylococcus aureus (MSSA) bacteremia in a patient with multiple potential sources. Fluorodeoxyglucose-positron emission tomography (FDG-PET) with computed tomography (CT) and sulfur colloid imaging with tagged white blood cell (WBC) scanning offer the most promise in facilitating rapid and accurate diagnoses of endovascular graft infections, vertebral osteomyelitis (V-OM), diabetic foot infections, and prosthetic joint infections (PJIs). However, radiologists at different institutions may have varying degrees of expertise with these modalities. Regardless, infectious disease consultants would benefit from knowing what nuclear medicine tests to order when considering patients with complex infectious disease syndromes.
Collapse
Affiliation(s)
- Andrea Censullo
- Division of Infectious Diseases, Cedars-Sinai Medical Center, University of California, Los Angeles
| | - Tara Vijayan
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles
| |
Collapse
|
25
|
Kniess T, Laube M, Wüst F, Pietzsch J. Technetium-99m based small molecule radiopharmaceuticals and radiotracers targeting inflammation and infection. Dalton Trans 2017; 46:14435-14451. [DOI: 10.1039/c7dt01735a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
99mTc-labeled antibiotics, antifungal drugs, antimicrobial peptides and COX-2 inhibitors are comprehensively reviewed.
Collapse
Affiliation(s)
- Torsten Kniess
- Helmholtz-Zentrum Dresden-Rossendorf
- Institute of Radiopharmaceutical Cancer Research
- 01328 Dresden
- Germany
| | - Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf
- Institute of Radiopharmaceutical Cancer Research
- 01328 Dresden
- Germany
| | - Frank Wüst
- University of Alberta
- Department of Oncology
- 11560 University Avenue
- Edmonton
- Canada
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf
- Institute of Radiopharmaceutical Cancer Research
- 01328 Dresden
- Germany
- Technische Universität Dresden
| |
Collapse
|
26
|
Dang Y, Li X, Zheng M, Liu H, Zhou X, Jin X. Development of a specific 99mTc-MAG3-mAb-WF-AF-1 for noninvasive detection of Aspergillus fumigatus. J Radioanal Nucl Chem 2016. [DOI: 10.1007/s10967-016-4802-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
27
|
Bhatt J, Mukherjee A, Korde A, Kumar M, Sarma HD, Dash A. Radiolabeling and Preliminary Evaluation of Ga-68 Labeled NODAGA-Ubiquicidin Fragments for Prospective Infection Imaging. Mol Imaging Biol 2016; 19:59-67. [DOI: 10.1007/s11307-016-0983-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
28
|
Fuscaldi LL, Dos Santos DM, Pinheiro NGS, Araújo RS, de Barros ALB, Resende JM, Fernandes SOA, de Lima ME, Cardoso VN. Synthesis and antimicrobial evaluation of two peptide LyeTx I derivatives modified with the chelating agent HYNIC for radiolabeling with technetium-99m. J Venom Anim Toxins Incl Trop Dis 2016; 22:16. [PMID: 27110232 PMCID: PMC4841036 DOI: 10.1186/s40409-016-0070-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/12/2016] [Indexed: 01/23/2023] Open
Abstract
Background Current diagnostic methods and imaging techniques are not able to differentiate septic and aseptic inflammation. Thus, reliable methods are sought to provide this distinction and scintigraphic imaging is an interesting option, since it is based on physiological changes. In this context, radiolabeled antimicrobial peptides have been investigated as they accumulate in infectious sites instead of aseptic inflammation. The peptide LyeTx I, from the venom of Lycosa erythrognatha, has potent antimicrobial activity. Therefore, this study aimed to synthesize LyeTx I derivatives with the chelating compound HYNIC, to evaluate their antimicrobial activity and to radiolabel them with 99mTc. Methods Two LyeTx I derivatives, HYNIC-LyeTx I (N-terminal modification) and LyeTx I-K-HYNIC (C-terminal modification), were synthesized by Fmoc strategy and purified by RP-HPLC. The purified products were assessed by RP-HPLC and MALDI-ToF-MS analysis. Microbiological assays were performed against S. aureus (ATCC® 6538) and E. coli (ATCC® 10536) in liquid medium to calculate the MIC. The radiolabeling procedure of LyeTx I-K-HYNIC with 99mTc was performed in the presence of co-ligands (tricine and EDDA) and reducing agent (SnCl2.2H2O), and standardized taking into account the amount of peptide, reducing agent, pH and heating. Radiochemical purity analysis was performed by thin-layer chromatography on silica gel strips and the radiolabeled compound was assessed by RP-HPLC and radioactivity measurement of the collected fractions. Data were analyzed by ANOVA, followed by Tukey test (p-values < 0.05). Results Both LyeTx I derivatives were suitably synthesized and purified, as shown by RP-HPLC and MALDI-ToF-MS analysis. The microbiological test showed that HYNIC-LyeTx I (N-terminal modification) did not inhibit bacterial growth, whereas LyeTx I-K-HYNIC (C-terminal modification) showed a MIC of 5.05 μmol.L−1 (S. aureus) and 10.10 μmol.L−1 (E. coli). Thus, only the latter was radiolabeled with 99mTc. The radiochemical purity analysis of LyeTx I-K-HYNIC-99mTc showed that the optimal radiolabeling conditions (10 μg of LyeTx I-K-HYNIC; 250 μg of SnCl2.2H2O; pH = 7; heating for 15 min) yielded a radiochemical purity of 87 ± 1 % (n = 3). However, RP-HPLC data suggested 99mTc transchelation from LyeTx I-K-HYNIC to the co-ligands (tricine and EDDA). Conclusions The binding of HYNIC to the N-terminal portion of LyeTx I seems to affect its activity against bacteria. Nevertheless, the radiolabeling of the C-terminal derivative, LyeTx I-K-HYNIC, must be better investigated to optimize the radiolabeled compound, in order to use it as a specific imaging agent to distinguish septic and aseptic inflammation.
Collapse
Affiliation(s)
- Leonardo Lima Fuscaldi
- Department of Clinical and Toxicological Analyses, School of Pharmacy, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901 Brazil
| | - Daniel Moreira Dos Santos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG Brazil
| | | | - Raquel Silva Araújo
- Department of Clinical and Toxicological Analyses, School of Pharmacy, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901 Brazil
| | - André Luís Branco de Barros
- Department of Clinical and Toxicological Analyses, School of Pharmacy, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901 Brazil
| | - Jarbas Magalhães Resende
- Department of Chemistry, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, MG Brazil
| | - Simone Odília Antunes Fernandes
- Department of Clinical and Toxicological Analyses, School of Pharmacy, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901 Brazil
| | - Maria Elena de Lima
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG Brazil
| | - Valbert Nascimento Cardoso
- Department of Clinical and Toxicological Analyses, School of Pharmacy, Federal University of Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, MG 31270-901 Brazil
| |
Collapse
|
29
|
Radiolabeling, biological evaluation and molecular docking of delafloxacin: a novel methicillin-resistant Staphylococcus aureus infection radiotracer. J Radioanal Nucl Chem 2015. [DOI: 10.1007/s10967-015-4586-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
30
|
The development of antimicrobial peptides as an approach to prevention of antibiotic resistance. ACTA ACUST UNITED AC 2015. [DOI: 10.1097/mrm.0000000000000032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
31
|
van Oosten M, Hahn M, Crane LMA, Pleijhuis RG, Francis KP, van Dijl JM, van Dam GM. Targeted imaging of bacterial infections: advances, hurdles and hopes. FEMS Microbiol Rev 2015; 39:892-916. [PMID: 26109599 DOI: 10.1093/femsre/fuv029] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2015] [Indexed: 02/06/2023] Open
Abstract
Bacterial infections represent an increasing problem in modern health care, in particular due to ageing populations and accumulating bacterial resistance to antibiotics. Diagnosis is rarely straightforward and consequently treatment is often delayed or indefinite. Therefore, novel tools that can be clinically implemented are urgently needed to accurately and swiftly diagnose infections. Especially, the direct imaging of infections is an attractive option. The challenge of specifically imaging bacterial infections in vivo can be met by targeting bacteria with an imaging agent. Here we review the current status of targeted imaging of bacterial infections, and we discuss advantages and disadvantages of the different approaches. Indeed, significant progress has been made in this field and the clinical implementation of targeted imaging of bacterial infections seems highly feasible. This was recently highlighted by the use of so-called smart activatable probes and a fluorescently labelled derivative of the antibiotic vancomycin. A major challenge remains the selection of the best imaging probes, and we therefore present a set of target selection criteria for clinical implementation of targeted bacterial imaging. Altogether, we conclude that the spectrum of potential applications for targeted bacterial imaging is enormous, ranging from fundamental research on infectious diseases to diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Marleen van Oosten
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands
| | - Markus Hahn
- Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands
| | - Lucia M A Crane
- Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands
| | - Rick G Pleijhuis
- Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands
| | | | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands
| | - Gooitzen M van Dam
- Department of Surgery, Division of Surgical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands
| |
Collapse
|
32
|
Chen H, Zhang M, Li B, Chen D, Dong X, Wang Y, Gu Y. Versatile antimicrobial peptide-based ZnO quantum dots for in vivo bacteria diagnosis and treatment with high specificity. Biomaterials 2015; 53:532-44. [DOI: 10.1016/j.biomaterials.2015.02.105] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/21/2015] [Accepted: 02/24/2015] [Indexed: 01/09/2023]
|
33
|
El-Kawy OA, Farah K. Radiocomplexation and biological evaluation of nemonoxacin in mice infected with multiresistant Staphylococcus aureus and penicillin-resistant Streptococci. J Radioanal Nucl Chem 2015. [DOI: 10.1007/s10967-015-4069-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
34
|
Synthesis, 68Ga-radiolabeling, and preliminary in vivo assessment of a depsipeptide-derived compound as a potential PET/CT infection imaging agent. BIOMED RESEARCH INTERNATIONAL 2015; 2015:284354. [PMID: 25699267 PMCID: PMC4324493 DOI: 10.1155/2015/284354] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 07/28/2014] [Accepted: 08/05/2014] [Indexed: 11/21/2022]
Abstract
Noninvasive imaging is a powerful tool for early diagnosis and monitoring of various disease processes, such as infections. An alarming shortage of infection-selective radiopharmaceuticals exists for overcoming the diagnostic limitations with unspecific tracers such as 67/68Ga-citrate or 18F-FDG. We report here TBIA101, an antimicrobial peptide derivative that was conjugated to DOTA and radiolabeled with 68Ga for a subsequent in vitro assessment and in vivo infection imaging using Escherichia coli-bearing mice by targeting bacterial lipopolysaccharides with PET/CT. Following DOTA-conjugation, the compound was verified for its cytotoxic and bacterial binding behaviour and compound stability, followed by 68Gallium-radiolabeling. µPET/CT using 68Ga-DOTA-TBIA101 was employed to detect muscular E. coli-infection in BALB/c mice, as warranted by the in vitro results. 68Ga-DOTA-TBIA101-PET detected E. coli-infected muscle tissue (SUV = 1.3–2.4) > noninfected thighs (P = 0.322) > forearm muscles (P = 0.092) > background (P = 0.021) in the same animal. Normalization of the infected thigh muscle to reference tissue showed a ratio of 3.0 ± 0.8 and a ratio of 2.3 ± 0.6 compared to the identical healthy tissue. The majority of the activity was cleared by renal excretion. The latter findings warrant further preclinical imaging studies of greater depth, as the DOTA-conjugation did not compromise the TBIA101's capacity as targeting vector.
Collapse
|
35
|
Eggleston H, Panizzi P. Molecular imaging of bacterial infections in vivo: the discrimination of infection from inflammation. INFORMATICS (MDPI) 2014; 1:72-99. [PMID: 26985401 PMCID: PMC4790455 DOI: 10.3390/informatics1010072] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Molecular imaging by definition is the visualization of molecular and cellular processes within a given system. The modalities and reagents described here represent a diverse array spanning both pre-clinical and clinical applications. Innovations in probe design and technologies would greatly benefit therapeutic outcomes by enhancing diagnostic accuracy and assessment of acute therapy. Opportunistic pathogens continue to pose a worldwide threat, despite advancements in treatment strategies, which highlights the continued need for improved diagnostics. In this review, we present a summary of the current clinical protocol for the imaging of a suspected infection, methods currently in development to optimize this imaging process, and finally, insight into endocarditis as a model of infectious disease in immediate need of improved diagnostic methods.
Collapse
Affiliation(s)
- Heather Eggleston
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849
| | - Peter Panizzi
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849
| |
Collapse
|
36
|
Abstract
Neuroinflammation plays a central role in a variety of neurological diseases, including stroke, multiple sclerosis, Alzheimer’s disease, and malignant CNS neoplasms, among many other. Different cell types and molecular mediators participate in a cascade of events in the brain that is ultimately aimed at control, regeneration and repair, but leads to damage of brain tissue under pathological conditions. Non-invasive molecular imaging of key players in the inflammation cascade holds promise for identification and quantification of the disease process before it is too late for effective therapeutic intervention. In this review, we focus on molecular imaging techniques that target inflammatory cells and molecules that are of interest in neuroinflammation, especially those with high translational potential. Over the past decade, a plethora of molecular imaging agents have been developed and tested in animal models of (neuro)inflammation, and a few have been translated from bench to bedside. The most promising imaging techniques to visualize neuroinflammation include MRI, positron emission tomography (PET), single photon emission computed tomography (SPECT), and optical imaging methods. These techniques enable us to image adhesion molecules to visualize endothelial cell activation, assess leukocyte functions such as oxidative stress, granule release, and phagocytosis, and label a variety of inflammatory cells for cell tracking experiments. In addition, several cell types and their activation can be specifically targeted in vivo, and consequences of neuroinflammation such as neuronal death and demyelination can be quantified. As we continue to make progress in utilizing molecular imaging technology to study and understand neuroinflammation, increasing efforts and investment should be made to bring more of these novel imaging agents from the “bench to bedside.”
Collapse
Affiliation(s)
- Benjamin Pulli
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| | - John W Chen
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| |
Collapse
|
37
|
Investigation of the antibacterial activity of a short cationic peptide against multidrug-resistant Klebsiella pneumoniae and Salmonella typhimurium strains and its cytotoxicity on eukaryotic cells. World J Microbiol Biotechnol 2013; 30:1533-40. [DOI: 10.1007/s11274-013-1575-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 12/02/2013] [Indexed: 12/14/2022]
|
38
|
Functional Imaging in Diagnostic of Orthopedic Implant-Associated Infections. Diagnostics (Basel) 2013; 3:356-71. [PMID: 26824928 PMCID: PMC4665528 DOI: 10.3390/diagnostics3040356] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/10/2013] [Accepted: 09/22/2013] [Indexed: 12/11/2022] Open
Abstract
Surgeries’ sterile conditions and perioperative antibiotic therapies decrease implant associated infections rates significantly. However, up to 10% of orthopedic devices still fail due to infections. An implant infection generates a high socio-economic burden. An early diagnosis of an infection would significantly improve patients’ outcomes. There are numerous clinical tests to diagnose infections. The “Gold Standard” is a microbiological culture, which requires an invasive sampling and lasts up to several weeks. None of the existing tests in clinics alone is sufficient for a conclusive diagnosis of an infection. Meanwhile, there are functional imaging modalities, which hold the promise of a non-invasive, quick, and specific infection diagnostic. This review focuses on orthopedic implant-associated infections, their pathogenicity, diagnosis and functional imaging.
Collapse
|