Imaging sensitive and drug-resistant bacterial infection with [11C]-trimethoprim

Iris K Lee, Daniel A Jacome, Joshua K Cho, Vincent Tu, Anthony J Young, Tiffany Dominguez, Justin D Northrup, Jean M Etersque, Hsiaoju S Lee, Andrew Ruff, Ouniol Aklilu, Kyle Bittinger, Laurel J Glaser, Daniel Dorgan, Denis Hadjiliadis, Rahul M Kohli, Robert H Mach, David A Mankoff, Robert K Doot, Mark A Sellmyer, Iris K Lee, Daniel A Jacome, Joshua K Cho, Vincent Tu, Anthony J Young, Tiffany Dominguez, Justin D Northrup, Jean M Etersque, Hsiaoju S Lee, Andrew Ruff, Ouniol Aklilu, Kyle Bittinger, Laurel J Glaser, Daniel Dorgan, Denis Hadjiliadis, Rahul M Kohli, Robert H Mach, David A Mankoff, Robert K Doot, Mark A Sellmyer

Abstract

BACKGROUNDSeveral molecular imaging strategies can identify bacterial infections in humans. PET affords the potential for sensitive infection detection deep within the body. Among PET-based approaches, antibiotic-based radiotracers, which often target key bacterial-specific enzymes, have considerable promise. One question for antibiotic radiotracers is whether antimicrobial resistance (AMR) reduces specific accumulation within bacteria, diminishing the predictive value of the diagnostic test.METHODSUsing a PET radiotracer based on the antibiotic trimethoprim (TMP), [11C]-TMP, we performed in vitro uptake studies in susceptible and drug-resistant bacterial strains and whole-genome sequencing (WGS) in selected strains to identify TMP resistance mechanisms. Next, we queried the NCBI database of annotated bacterial genomes for WT and resistant dihydrofolate reductase (DHFR) genes. Finally, we initiated a first-in-human protocol of [11C]-TMP in patients infected with both TMP-sensitive and TMP-resistant organisms to demonstrate the clinical feasibility of the tool.RESULTSWe observed robust [11C]-TMP uptake in our panel of TMP-sensitive and -resistant bacteria, noting relatively variable and decreased uptake in a few strains of P. aeruginosa and E. coli. WGS showed that the vast majority of clinically relevant bacteria harbor a WT copy of DHFR, targetable by [11C]-TMP, and that despite the AMR, these strains should be "imageable." Clinical imaging of patients with [11C]-TMP demonstrated focal radiotracer uptake in areas of infectious lesions.CONCLUSIONThis work highlights an approach to imaging bacterial infection in patients, which could affect our understanding of bacterial pathogenesis as well as our ability to better diagnose infections and monitor response to therapy.TRIAL REGISTRATIONClinicalTrials.gov NCT03424525.FUNDINGInstitute for Translational Medicine and Therapeutics, Burroughs Wellcome Fund, NIH Office of the Director Early Independence Award (DP5-OD26386), and University of Pennsylvania NIH T32 Radiology Research Training Grant (5T32EB004311-12).

Keywords: Bacterial infections; Diagnostic imaging; Infectious disease.

Figures

Figure 1. Structure of [ 11 C]-TMP…
Figure 1. Structure of [11C]-TMP and in vitro TMP dose-response assays of different bacterial strains.
(A) Structures of trimethoprim (TMP) and [11C]-TMP. (B) TMP dose-response assay on bacterial strains. OD600 measurement was taken following a 6-hour incubation of different bacterial strains with TMP. The susceptibility or resistance of a bacterial strain to TMP is color-coded based on the IC50 and minimum inhibitory concentration (MIC). Blue indicates susceptible bacteria, and red indicates resistant bacteria. n = 3; data represent mean ± SD. (C) Representative [11C]-TMP uptake in bacterial cultures after a 30-minute incubation at 37°C. n = 3–5; data represent mean ± SD. The experiment was repeated a total of 2–3 times for biological replicates.
Figure 2. Bioinformatic analysis of clinically relevant…
Figure 2. Bioinformatic analysis of clinically relevant bacteria.
(A) Proportion of clinically relevant bacterial strains from the NCBI RefSeq database, with the indicated number of DHFR genes per genome. (B) Resistance characterization of DHFR genes in relevant bacterial strains from the NCBI RefSeq database. Each dot represents a strain.
Figure 3. CONSORT diagram of the […
Figure 3. CONSORT diagram of the [11C]-TMP study.
*Laboratory test results may have been collected from the medical record if they were completed within 30 days of screening; in these cases, they were not repeated for the purposes of this study. Refusal of labs did not preclude a patient from the study. **Surgical or systemic therapy was started if clinically indicated at the judgment of treating physicians. ***For patients who received systemic antibiotic therapy, this may have been within 1 week of therapy cessation. For patients who received surgical management, this may have occurred within 3–6 weeks after surgery, as clinical appropriate. Given that some patients are on chronic antibiotics, this scan may have occurred after completion of alternative or more intensive antibiotic therapy. $In some cases, and at the discretion of the investigator, the patient may have been scanned while on i.v. antibiotics.
Figure 4. Biodistribution of [ 18 F]-FDG…
Figure 4. Biodistribution of [18F]-FDG versus [11C]-TMP in a patient with lung cancer and biodistribution in a patient with underlying chronic lung disease.
(A) The image on the left shows a 64-year-old man with known lung adenocarcinoma who underwent a [18F]-FDG (549 MBq) and then a [11C]-TMP (563 MBq) PET/CT 2 days later. The [18F]-FDG image was acquired starting 71 minutes after injection. Whole-body maximum intensity projection (MIP) images demonstrate the difference in biodistribution of the tracers. In the lungs, [18F]-FDG is taken up both by metabolically active tumor and inflammatory cells, whereas [11C]-TMP is not. The image on the right shows a comparison MIP image of a patient with cystic fibrosis and chronic lung infections who underwent a [11C]-TMP PET/CT (780 MBq). The image was acquired starting 78 minutes after injection. PET images are scaled at 0–7 g/mL SUV. (B) [11C]-TMP imaging of a 44-year-old woman with several foci of infection in the chest (red arrows). Other sites of signal include the liver, the kidneys, red bone marrow, and the stomach. PET images are scaled at 0–7 g/mL SUV.
Figure 5. Time activity curves and bacterial…
Figure 5. Time activity curves and bacterial heterogeneity.
(A and B) [11C]-TMP PET/CT images of a 44-year-old woman with an acute exacerbation of cystic fibrosis (A) before and (B) after treatment with 2 weeks of i.v. ceftriaxone. Regions of interest were drawn around 2 separate pulmonary airspace opacities. In addition, reference regions were also drawn around a reference lymph node and paraspinal musculature and within the aorta. Comparing PET/CT images before and after treatment, the visible changes in relative [11C]-TMP uptake in lesion 1 compared with lesion 2 demonstrate the bacterial heterogeneity and an apparent new infection with S. aureus based on sputum cultures. The patient received 487 MBq and 780 MBq of [11C]-TMP at the first and second time points, respectively. PET images are scaled at 0–5 g/mL SUV and CT images are scaled at –1,024 to +300 HU.
Figure 6. Acute exacerbation of cystic fibrosis.
Figure 6. Acute exacerbation of cystic fibrosis.
(A) A 64-year-old man with known lung adenocarcinoma underwent a [18F]-FDG (549 MBq) and then a [11C]-TMP (563 MBq) PET/CT 2 days later. The [18F]-FDG image was acquired starting 71 minutes after injections. Whole-body maximum intensity projection (MIP) images demonstrate the difference in biodistribution of the tracers. In the lungs, [18F]-FDG is taken up both by metabolically active tumor and inflammatory cells, whereas [11C]-TMP is not. (B) A comparison MIP image of a 44-year-old woman with cystic fibrosis and chronic lung infections, who underwent a [11C]-TMP PET/CT (780 MBq). The image was acquired starting 78 minutes after injection. The PET images show several foci of infection in the chest (red arrows). Other sites of signal include the liver, the kidneys, red bone marrow, and the stomach. PET images are scalled 0-7 g/mL SUV.
Figure 7. Biopsy proven discitis osteomyelitis treated…
Figure 7. Biopsy proven discitis osteomyelitis treated with antibiotics.
A 55-year-old man with clinically suspected lumbar discitis osteomyelitis was scanned with [11C]-TMP PET/CT at the initiation of empiric antibiotic therapy and after 6 more weeks of targeted antibiotic therapy. (A) Time line of patient disease sequelae and imaging. (B) Axial PET/CT images show a clear site of asymmetric [11C]-TMP uptake in the left L4–L5 facet at the start of therapy and lack of uptake after 6 weeks of i.v. treatment. Facet biopsy of the left L4–L5 facet grew methicillin-sensitive S. aureus. Note: the patient received different doses of [11C]-TMP, 129 MBq at the first time point and 672 MBq at the second time point; thus, the image quality was noisier at the first time point. PET images are scaled at 0–5 SUVmax. (C) The temporal morphologic sequelae of discitis osteomyelitis are demonstrated by sagittal CT images before, during, and after treatment. (D) In contrast to the PET/CT images, the gadolinium-enhanced MRI images of the patient at 10 weeks after therapy continue to demonstrate marrow replacement and contrast enhancement, findings that are nonspecific for active infection versus continued inflammation.

References

    1. Vinjamuri S, et al. Comparison of 99mTc infecton imaging with radiolabelled white-cell imaging in the evaluation of bacterial infection. Lancet. 1996;347(8996):233–235. doi: 10.1016/S0140-6736(96)90407-9.
    1. Tucker AT, et al. Discovery of next-generation antimicrobials through bacterial self-screening of surface-displayed peptide libraries. Cell. 2018;172(3):618–628. doi: 10.1016/j.cell.2017.12.009.
    1. Heuker M, et al. Preclinical studies and prospective clinical applications for bacteria-targeted imaging: the future is bright. Clin Transl Imaging. 2016;4:253–264. doi: 10.1007/s40336-016-0190-y.
    1. Palestro CJ, et al. Diagnostic imaging tests and microbial infections. Cell Microbiol. 2007;9(10):2323–2333. doi: 10.1111/j.1462-5822.2007.01013.x.
    1. Gowrishankar G, et al. Specific imaging of bacterial infection using 6’’-(18)F-fluoromaltotriose: a second-generation PET tracer targeting the maltodextrin transporter in bacteria. J Nucl Med. 2017;58(10):1679–1684. doi: 10.2967/jnumed.117.191452.
    1. Mutch C, et al. [ 11 C]Para-aminobenzoic acid: a positron emission tomography tracer targeting bacteria-specific metabolism. ACS Infect Dis. 2018;4(7):1067–1072. doi: 10.1021/acsinfecdis.8b00061.
    1. Neumann KD, et al. Imaging active infection in vivo using D-amino acid derived PET radiotracers. Sci Rep. 2017;7(1):7903. doi: 10.1038/s41598-017-08415-x.
    1. Zhang Z, et al. Positron emission tomography imaging with 2-[(18)F]F- p-aminobenzoic acid detects Staphylococcus aureus infections and monitors drug response. ACS Infect Dis. 2018;4(11):1635–1644. doi: 10.1021/acsinfecdis.8b00182.
    1. Weinstein EA, et al. Imaging Enterobacteriaceae infection in vivo with 18F-fluorodeoxysorbitol positron emission tomography. Sci Transl Med. 2014;6(259):259ra146.
    1. Ordonez AA, et al. Imaging Enterobacterales infections in patients using pathogen-specific positron emission tomography. Sci Transl Med. 2021;13(589):eabe9805. doi: 10.1126/scitranslmed.abe9805.
    1. Ordonez AA, et al. 11C-para-aminobenzoic acid PET imaging of S. aureus and MRSA infection in preclinical models and humans. JCI Insight. 2022;7(1):154117.
    1. Sellmyer MA, et al. Bacterial infection imaging with [(18)F]fluoropropyl-trimethoprim. Proc Natl Acad Sci U S A. 2017;114(31):8372–8377. doi: 10.1073/pnas.1703109114.
    1. Sellmyer MA, et al. Quantitative PET reporter gene imaging with [(11)C]trimethoprim. Mol Ther. 2017;25(1):120–126. doi: 10.1016/j.ymthe.2016.10.018.
    1. Baccanari DP, Kuyper LF. Basis of selectivity of antibacterial diaminopyrimidines. J Chemother. 1993;5(6):393–399. doi: 10.1080/1120009X.1993.11741086.
    1. Ordonez AA, et al. Molecular imaging of bacterial infections: overcoming the barriers to clinical translation. Sci Transl Med. 2019;11(508):eaax8251. doi: 10.1126/scitranslmed.aax8251.
    1. Huovinen P, et al. Trimethoprim and sulfonamide resistance. Antimicrob Agents Chemother. 1995;39(2):279–289. doi: 10.1128/AAC.39.2.279.
    1. Bankevich A, et al. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J Comput Biol. 2012;19(5):455–477. doi: 10.1089/cmb.2012.0021.
    1. Parks DH, et al. CheckM: assessing the quality of microbial genomes recovered from isolates, single cells, and metagenomes. Genome Res. 2015;25(7):1043–1055. doi: 10.1101/gr.186072.114.
    1. Lombardo MN N GD, et al. Crystal structures of trimethoprim-resistant DfrA1 rationalize potent inhibition by propargyl-linked antifolates. ACS Infect Dis. 2016;2(2):149–156. doi: 10.1021/acsinfecdis.5b00129.
    1. Heaslet H, et al. Structural comparison of chromosomal and exogenous dihydrofolate reductase from Staphylococcus aureus in complex with the potent inhibitor trimethoprim. Proteins. 2009;76(3):706–717. doi: 10.1002/prot.22383.
    1. Lamb KM, et al. Crystal structures of Klebsiella pneumoniae dihydrofolate reductase bound to propargyl-linked antifolates reveal features for potency and selectivity. Antimicrob Agents Chemother. 2014;58(12):7484–7491. doi: 10.1128/AAC.03555-14.
    1. Aeschlimann JR. The role of multidrug efflux pumps in the antibiotic resistance of Pseudomonas aeruginosa and other gram-negative bacteria. Insights from the Society of Infectious Diseases Pharmacists. Pharmacotherapy. 2003;23(7):916–924. doi: 10.1592/phco.23.7.916.32722.
    1. Webber MA, Piddock LJ. The importance of efflux pumps in bacterial antibiotic resistance. J Antimicrob Chemother. 2003;51(1):9–11. doi: 10.1093/jac/dkg050.
    1. Kohler T, et al. Multidrug efflux in intrinsic resistance to trimethoprim and sulfamethoxazole in Pseudomonas aeruginosa. Antimicrob Agents Chemother. 1996;40(10):2288–2290. doi: 10.1128/AAC.40.10.2288.
    1. Faltyn M, et al. Evolution and nomenclature of the trimethoprim resistant dihydrofolate (dfr) reductases [preprint]. Posted on May 10, 2019.
    1. Weiner-Lastinger LM, et al. Antimicrobial-resistant pathogens associated with adult healthcare-associated infections: summary of data reported to the National Healthcare Safety Network, 2015-2017. Infect Control Hosp Epidemiol. 2020;41(1):1–18. doi: 10.1017/ice.2019.296.
    1. Rubin RH, Swartz MN. Trimethoprim-sulfamethoxazole. N Engl J Med. 1980;303(8):426–432. doi: 10.1056/NEJM198008213030804.
    1. Gleckman R, et al. Trimethoprim: mechanisms of action, antimicrobial activity, bacterial resistance, pharmacokinetics, adverse reactions, and therapeutic indications. Pharmacotherapy. 1981;1(1):14–20. doi: 10.1002/j.1875-9114.1981.tb03548.x.
    1. Phetsang W, et al. Fluorescent trimethoprim conjugate probes to assess drug accumulation in wild type and mutant Escherichia coli. ACS Infect Dis. 2016;2(10):688–701. doi: 10.1021/acsinfecdis.6b00080.
    1. Cai H, et al. Development of a liquid chromatography/mass spectrometry-based drug accumulation assay in Pseudomonas aeruginosa. Anal Biochem. 2009;385(2):321–325. doi: 10.1016/j.ab.2008.10.041.
    1. Davis TD, et al. General platform for systematic quantitative evaluation of small-molecule permeability in bacteria. ACS Chem Biol. 2014;9(11):2535–2544. doi: 10.1021/cb5003015.
    1. Combs MP, et al. Lung microbiota predict chronic rejection in healthy lung transplant recipients: a prospective cohort study. Lancet Respir Med. 2021;9(6):601–612. doi: 10.1016/S2213-2600(20)30405-7.
    1. Demiroglu H, et al. Radiosynthesis and biodistribution of Tc-99m-trimethoprim: a novel radiolabeled antibiotic for bacterial infection imaging using experimental animals. Kafkas Univ Vet Fak Derg. 2018;24(3):393–400.
    1. Rahmim A, Zaidi H. PET versus SPECT: strengths, limitations and challenges. Nucl Med Commun. 2008;29(3):193–207. doi: 10.1097/MNM.0b013e3282f3a515.
    1. Rashid U, et al. Design, synthesis, antibacterial activity and docking study of some new trimethoprim derivatives. Bioorg Med Chem Lett. 2016;26(23):5749–5753. doi: 10.1016/j.bmcl.2016.10.051.
    1. Bolger AM, et al. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):2114–2120. doi: 10.1093/bioinformatics/btu170.
    1. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 2011;17(1):10–12. doi: 10.14806/ej.17.1.200.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):1754–1760. doi: 10.1093/bioinformatics/btp324.
    1. Eren AM, et al. Anvi’o: an advanced analysis and visualization platform for ‘omics data. PeerJ. 2015;3:e1319. doi: 10.7717/peerj.1319.
    1. Hyatt D, et al. Prodigal: prokaryotic gene recognition and translation initiation site identification. BMC Bioinformatics. 2010;11:119. doi: 10.1186/1471-2105-11-119.
    1. McArthur AG, et al. The comprehensive antibiotic resistance database. Antimicrob Agents Chemother. 2013;57(7):3348–3357. doi: 10.1128/AAC.00419-13.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9(4):357–359. doi: 10.1038/nmeth.1923.
    1. Kolthammer JA, et al. Performance evaluation of the Ingenuity TF PET/CT scanner with a focus on high count-rate conditions. Phys Med Biol. 2014;59(14):3843–3859. doi: 10.1088/0031-9155/59/14/3843.

Source: PubMed

3
Sottoscrivi