Next-Generation Sequencing for Infectious Disease Diagnostics in Pediatric Patients with Malignancies or After Hematopoietic Cell Transplantation: A Systematic Review
Abstract
1. Introduction
2. Overview of Next-Generation Sequencing Approaches in Infectious Disease Diagnostics
2.1. Metagenomic Next-Generation Sequencing
2.2. Targeted Next-Generation Sequencing (tNGS)
2.3. Whole-Genome Sequencing (WGS)
2.4. RNA Sequencing (RNA-Seq, Metatranscriptomics)
3. Materials and Methods
3.1. Study Design
3.2. Eligibility Criteria
- Population: Pediatric patients (aged 0–18 years) with oncological diseases, including those undergoing hematopoietic cell transplantation (HCT), both autologous and allogeneic.
- Intervention: Use of NGS technologies (e.g., metagenomic NGS, targeted NGS, whole-genome sequencing, 16S/18S/ITS rRNA sequencing) for the diagnosis of infections.
- Outcomes: Diagnostic performance of NGS (e.g., pathogen identification, diagnostic yield, turnaround time), clinical relevance (e.g., impact on antimicrobial therapy), and/or patient outcomes.
- Study type: Original studies including prospective and retrospective cohorts, case series, or clinical trials.
- Language: Articles published in English.
- Timeframe: Studies published between January 2010 and April 2025.
- Studies exclusively involving adult populations or lacking separate pediatric analysis.
- Focusing solely on tumor genomics or microbiome composition without an infectious disease context.
- Reviews, editorials, conference abstracts, and commentaries.
3.3. Information Sources and Search Strategy
3.4. Study Selection
3.5. Data Extraction
- Basic study information (author, year, study type);
- Patient population (e.g., pediatric cancer patients, HCT recipients);
- Type of clinical sample used for NGS (e.g., blood, bronchoalveolar lavage (BAL), cerebrospinal fluid (CSF) BAL, CSF);
- NGS approach (e.g., mNGS, WGS, 16S/18S sequencing);
- Type of infection investigated (e.g., bacterial, viral, fungal);
- Pathogens detected;
- Diagnostic performance (e.g., sensitivity, specificity, positivity rate);
- Turnaround time for NGS results;
- Comparison with conventional diagnostic methods;
- Clinical impact (e.g., changes in antimicrobial treatment, diagnosis confirmation);
- Detection of antimicrobial resistance (if reported);
- Reported limitations of NGS use;
- Additional relevant notes (e.g., cost, feasibility).
3.6. Risk of Bias Assessment
3.7. Data Synthesis
4. Results
4.1. Characteristics of Included Studies
4.2. Comparison of Diagnostic Performance Between NGS and Conventional Methods
4.3. Influence on Clinical Management
4.4. NGS Methodologies and Sample Types
4.5. Detection of Antimicrobial Resistance (AMR) and Surveillance Applications
4.6. Limitations of Evidence
5. Discussion
5.1. Pathogen Detection Sequencing
5.2. Host-Response, Microbiome, and Host-Genome Sequencing
5.3. Overall Interpretation and Limitations
5.4. Future Directions
6. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
ADV | adenovirus |
AUC | area under the curve |
BAL | bronchoalveolar lavage |
BALF | bronchoalveolar lavage fluid |
BKV | BK virus |
BSI | bloodstream infection |
CDI | Clostridioides difficile infection |
cfDNA | cell-free DNA |
CMV | cytomegalovirus |
CNS | central nervous system |
CVC | central venous catheter |
DEGs | differentially expressed genes |
EBV | Epstein–Barr virus |
EIA | enzyme immunoassay |
FIND | Foundation for Innovative New Diagnostics |
FN | febrile neutropenia |
FUO | fever of unknown origin |
GI | gastrointestinal |
GM | galactomannan |
G-test | β-D-glucan test |
GVHD | graft-versus-host disease |
HAdV | human adenovirus |
HCT | hematopoietic cell transplantation |
HHV | human herpesvirus |
HPgV-1 | human pegivirus-1 |
HRV | human rhinovirus |
ICU | intensive care unit |
ICT | immunochromatographic test |
IL-6 | interleukin-6 |
JBI | Joanna Briggs Institute |
mNGS | metagenomic next-generation sequencing |
NGS | next-generation sequencing |
NPV | negative predictive value |
PCR | polymerase chain reaction |
PPV | positive predictive value |
PRISMA | Preferred Reporting Items for Systematic Reviews and Meta-Analyses |
qPCR | quantitative polymerase chain reaction |
qRT-PCR | quantitative reverse transcription PCR |
RNA-seq | RNA sequencing |
RTI | respiratory tract infection |
SARS-CoV-2 | severe acute respiratory syndrome coronavirus 2 |
TPD | traditional pathogen detection |
UTI | urinary tract infection |
VZV | varicella-zoster virus |
WGS | whole-genome sequencing |
References
- Getzenberg, R.B.; Hijano, D.R.; Hakim, H.; Dallas, R.H.; Ferrolino, J.A.; de Cardenas, J.B.; Garner, C.D.; Tang, L.; Su, Y.; Wolf, J.; et al. Rothia mucilaginosa Infections in Pediatric Cancer Patients. J. Pediatr. Infect. Dis. Soc. 2020, 10, 341–344. [Google Scholar] [CrossRef] [PubMed]
- Norris, L.B.; Kablaoui, F.; Brilhart, M.K.; Bookstaver, P.B. Systematic review of antimicrobial lock therapy for prevention of central-line-associated bloodstream infections in adult and pediatric cancer patients. Int. J. Antimicrob. Agents 2017, 50, 308–317. [Google Scholar] [CrossRef] [PubMed]
- Soudani, N.; Caniza, M.A.; Assaf-Casals, A.; Shaker, R.; Lteif, M.; Su, Y.; Tang, L.; Akel, I.; Muwakkit, S.; Chmaisse, A.; et al. Prevalence and characteristics of acute respiratory virus infections in pediatric cancer patients. J. Med. Virol. 2019, 91, 1191–1201. [Google Scholar] [CrossRef]
- Kalbitz, S.; Ermisch, J.; Kellner, N.; Nickel, O.; Borte, S.; Marx, K.; Lübbert, C. Metagenomic next-generation sequencing as a diagnostic tool in the clinical routine of an infectious diseases department: A retrospective cohort study. Infection 2024, 52, 1595–1600. [Google Scholar] [CrossRef]
- Simner, P.J.; Miller, S.; Carroll, K.C. Understanding the Promises and Hurdles of Metagenomic Next-Generation Sequencing as a Diagnostic Tool for Infectious Diseases. Clin. Infect. Dis. 2018, 66, 778–788. [Google Scholar] [CrossRef]
- Schlaberg, R.; Chiu, C.Y.; Miller, S.; Procop, G.W.; Weinstock, G.; the Professional Practice Committee Committee on Laboratory Practices of the American Society for Microbiology; the Microbiology Resource Committee of the College of American Pathologists. Validation of Metagenomic Next-Generation Sequencing Tests for Universal Pathogen Detection. Arch. Pathol. Lab. Med. 2017, 141, 776–786. [Google Scholar] [CrossRef]
- Gu, W.; Miller, S.; Chiu, C.Y. Clinical Metagenomic Next-Generation Sequencing for Pathogen Detection. Annu. Rev. Pathol. Mech. Dis. 2019, 14, 319–338. [Google Scholar] [CrossRef]
- Liu, Y.; Ma, Y. Clinical applications of metagenomics next-generation sequencing in infectious diseases. J. Zhejiang Univ. Sci. B 2024, 25, 471–484. [Google Scholar] [CrossRef] [PubMed]
- Gan, M.; Zhang, Y.; Yan, G.; Wang, Y.; Lu, G.; Wu, B.; Chen, W.; Zhou, W. Antimicrobial resistance prediction by clinical metagenomics in pediatric severe pneumonia patients. Ann. Clin. Microbiol. Antimicrob. 2024, 23, 33. [Google Scholar] [CrossRef] [PubMed]
- Lehman, A.C.; Goren, L.R.; Evans, M.D.; Toles, O.; Drozdov, D.; Andrews, S.L.; McAllister, S.C.; Thielen, B.K. Clinical Performance of Plasma Metagenomic Sequencing in Immunocompromised Pediatric Patients. J. Pediatr. Infect. Dis. Soc. 2024, 13, 276–281. [Google Scholar] [CrossRef]
- Qin, D. Next-generation sequencing and its clinical application. Cancer Biol. Med. 2019, 16, 4–10. [Google Scholar] [CrossRef]
- Behjati, S.; Tarpey, P.S. What is next generation sequencing? Arch. Dis. Child. Educ. Pract. Ed. 2013, 98, 236–238. [Google Scholar] [CrossRef]
- Chiu, C.Y.; Miller, S.A. Clinical metagenomics. Nat. Rev. Genet. 2019, 20, 341–355. [Google Scholar] [CrossRef] [PubMed]
- Chen, Q.; Yi, J.; Liu, Y.; Yang, C.; Sun, Y.; Du, J.; Liu, Y.; Gu, D.; Liu, H.; Xu, Y.; et al. Clinical diagnostic value of targeted next-generation sequencing for infectious diseases (Review). Mol. Med. Rep. 2024, 30, 153. [Google Scholar] [CrossRef] [PubMed]
- Spatz, S.; Afonso, C.L. Non-Targeted RNA Sequencing: Towards the Development of Universal Clinical Diagnosis Methods for Human and Veterinary Infectious Diseases. Vet. Sci. 2024, 11, 239. [Google Scholar] [CrossRef] [PubMed]
- Köser, C.U.; Ellington, M.J.; Peacock, S.J. Whole-genome sequencing to control antimicrobial resistance. Trends Genet. 2014, 30, 401–407. [Google Scholar] [CrossRef]
- Timsit, S.; Armand-Lefèvre, L.; Le Goff, J.; Salmona, M. The clinical and epidemiological impacts of whole genomic sequencing on bacterial and virological agents. Infect. Dis. Now. 2024, 54, 104844. [Google Scholar] [CrossRef]
- Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
- Ouzzani, M.; Hammady, H.; Fedorowicz, Z.; Elmagarmid, A. Rayyan—A web and mobile app for systematic reviews. Syst. Rev. 2016, 5, 210. [Google Scholar] [CrossRef]
- Haddaway, N.R.; Page, M.J.; Pritchard, C.C.; McGuinness, L.A. PRISMA2020: An R package and Shiny app for producing PRISMA 2020-compliant flow diagrams, with interactivity for optimised digital transparency and Open Synthesis. Campbell Syst. Rev. 2022, 18, e1230. [Google Scholar] [CrossRef]
- Armstrong, A.E.; Rossoff, J.; Hollemon, D.; Hong, D.K.; Muller, W.J.; Chaudhury, S. Cell-free DNA next-generation sequencing successfully detects infectious pathogens in pediatric oncology and hematopoietic stem cell transplant patients at risk for invasive fungal disease. Pediatr. Blood Cancer 2019, 66, e27734. [Google Scholar] [CrossRef] [PubMed]
- Shen, H.; Shen, D.; Song, H.; Wu, X.; Xu, C.; Su, G.; Liu, C.; Zhang, J. Clinical assessment of the utility of metagenomic next-generation sequencing in pediatric patients of hematology department. Int. J. Lab. Hematol. 2021, 43, 244–249. [Google Scholar] [CrossRef]
- Horiba, K.; Torii, Y.; Okumura, T.; Takeuchi, S.; Suzuki, T.; Kawada, J.-I.; Muramatsu, H.; Takahashi, Y.; Ogi, T.; Ito, Y. Next-Generation Sequencing to Detect Pathogens in Pediatric Febrile Neutropenia: A Single-Center Retrospective Study of 112 Cases. Open Forum Infect. Dis. 2021, 8, ofab223. [Google Scholar] [CrossRef]
- Zhang, P.; Zhang, Z.-H.; Liang, J.; Shen, D.-Y.; Li, J.; Wang, D.; Jin, F.-F.; Song, H.; Zhang, J.-Y.; Xu, W.-Q.; et al. Metagenomic next-generation sequencing for the diagnosis of fever of unknown origin in pediatric patients with hematological malignancy. Clin. Chim. Acta 2022, 537, 133–139. [Google Scholar] [CrossRef]
- Qu, Y.; Ding, W.; Liu, S.; Wang, X.; Wang, P.; Liu, H.; Xia, H.; Chen, Y.; Jiang, H. Metagenomic Next-Generation Sequencing vs. Traditional Pathogen Detection in the Diagnosis of Infection After Allogeneic Hematopoietic Stem Cell Transplantation in Children. Front. Microbiol. 2022, 13, 868160. [Google Scholar] [CrossRef]
- Wang, D.; Wang, W.; Ding, Y.; Tang, M.; Zhang, L.; Chen, J.; You, H. Metagenomic Next-Generation Sequencing Successfully Detects Pulmonary Infectious Pathogens in Children With Hematologic Malignancy. Front. Cell. Infect. Microbiol. 2022, 12, 899028. [Google Scholar] [CrossRef]
- Wang, D.; Lai, M.; Song, H.; Zhang, J.-Y.; Zhao, F.-Y.; Liang, J.; Xu, W.-Q.; Tang, Y.-M.; Xu, X.-J. Integration of Interleukin-6 Improves the Diagnostic Precision of Metagenomic Next-Generation Sequencing for Infection in Immunocompromised Children. Front. Microbiol. 2022, 13, 819467. [Google Scholar] [CrossRef]
- Fu, Y.; Zhu, X.; Cao, P.; Shen, C.; Qian, X.; Miao, H.; Yu, Y.; Wang, H.; Zhai, X. Metagenomic Next-Generation Sequencing in the Diagnosis of Infectious Fever During Myelosuppression Among Pediatric Patients with Hematological and Neoplastic Diseases. Infect. Drug Resist. 2022, 15, 5425–5434. [Google Scholar] [CrossRef] [PubMed]
- Guo, F.; Kang, L.; Zhang, L. mNGS for identifying pathogens in febrile neutropenic children with hematological diseases. Int. J. Infect. Dis. 2022, 116, 85–90. [Google Scholar] [CrossRef]
- Wu, Q.; Wu, Y.; Zhao, Y.; Zhang, Y.; Cao, J.; Wu, D.; Zhou, J.; Chen, F. Adenovirus infection diagnosed by metagenomic next-generation sequencing after haploidentical hematopoietic stem cell transplantation: A multicenter study in China. Transpl. Infect. Dis. 2023, 25, e14054. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Zhou, D.; Xia, H.; Wang, J.; Yang, H.; Xu, L.; Huang, K.; Fang, J. Metagenomic next-generation sequencing for detection of pathogens in children with hematological diseases complicated with infection. Mol. Cell. Probes 2023, 67, 101889. [Google Scholar] [CrossRef]
- Xu, X.; Zheng, Y.; Zhang, X.; Zhang, C.; Gai, W.; Yang, Z. Utility of Metagenomic Next-Generation Sequencing for Diagnosis of Infectious Diseases in Critically Ill Immunocompromised Pediatric Patients. Infect. Drug Resist. 2024, 17, 3579–3591. [Google Scholar] [CrossRef]
- Wu, J.; Song, W.; Yan, H.; Luo, C.; Hu, W.; Xie, L.; Shen, N.; Cao, Q.; Mo, X.; An, K.; et al. Metagenomic next-generation sequencing in detecting pathogens in pediatric oncology patients with suspected bloodstream infections. Pediatr. Res. 2024, 95, 843–851. [Google Scholar] [CrossRef]
- Abraham, A.; Green, A.; Ferrolino, J.; Flerlage, T.; Gowen, A.; Allison, K.J.; Suliman, A.Y.; Bhakta, N.; McArthur, J.; Srinivasan, S.; et al. Utility and Safety of Bronchoalveolar Lavage for Diagnosis and Management of Respiratory Illnesses in Immunocompromised Children. J. Pediatr. Infect. Dis. Soc. 2025, 14, piaf015. [Google Scholar] [CrossRef] [PubMed]
- da Silva, A.S.; de Campos, G.M.; Altizani, G.M.; de Carvalho, E.; Barros, A.C.; Cella, E.; Kashima, S.; Sampaio, S.C.; Elias, M.C.; Giovanetti, M.; et al. Utilizing Viral Metagenomics to Characterize Pathogenic and Commensal Viruses in Pediatric Patients with Febrile Neutropenia. Viruses 2025, 17, 345. [Google Scholar] [CrossRef] [PubMed]
- Jalal, D.; Elzayat, M.G.; Diab, A.A.; El-Shqanqery, H.E.; Samir, O.; Bakry, U.; Hassan, R.; Elanany, M.; Shalaby, L.; Sayed, A.A.; et al. Deciphering Multidrug-Resistant Acinetobacter baumannii from a Pediatric Cancer Hospital in Egypt. mSphere 2021, 6, e00725-21. [Google Scholar] [CrossRef]
- Fattouh, R.; Stapleton, P.J.; Eshaghi, A.; Thomas, A.D.; Science, M.E.; Schechter, T.; Streitenberger, L.; Hubacek, P.; Yau, Y.C.W.; Brown, M.; et al. A Prolonged Outbreak of Human Adenovirus A31 (HAdV-A31) Infection on a Pediatric Hematopoietic Stem Cell Transplantation Ward with Whole Genome Sequencing Evidence of International Linkages. J. Clin. Microbiol. 2022, 60, e00665-22. [Google Scholar] [CrossRef]
- Putri, N.D.; Johar, E.; Dewi, Y.P.; Indrasari, N.D.; Wulandari, D.; Pasaribu, M.M.B.; Sari, T.T.; Cakti, F.P.; Jasin, M.R.; Tartila, T.; et al. Whole-Genome Sequencing of SARS-CoV-2 Infection in a Cluster of Immunocompromised Children in Indonesia. Front. Med. 2022, 9, 835998. [Google Scholar] [CrossRef] [PubMed]
- Hakim, H.; Glasgow, H.L.; Brazelton, J.N.; Gilliam, C.H.; Richards, L.; Hayden, R.T. A prospective bacterial whole-genome-sequencing-based surveillance programme for comprehensive early detection of healthcare-associated infection transmission in paediatric oncology patients. J. Hosp. Infect. 2024, 143, 53–63. [Google Scholar] [CrossRef]
- Jansen, S.A.; Nijhuis, W.; Leavis, H.L.; Riezebos-Brilman, A.; Lindemans, C.A.; Schuurman, R. Broad Virus Detection and Variant Discovery in Fecal Samples of Hematopoietic Transplant Recipients Using Targeted Sequence Capture Metagenomics. Front. Microbiol. 2020, 11, 560179. [Google Scholar] [CrossRef]
- Ludowyke, N.; Phumiphanjarphak, W.; Apiwattanakul, N.; Manopwisedjaroen, S.; Pakakasama, S.; Sensorn, I.; Pasomsub, E.; Chantratita, W.; Hongeng, S.; Aiewsakun, P.; et al. Target Enrichment Metagenomics Reveals Human Pegivirus-1 in Pediatric Hematopoietic Stem Cell Transplantation Recipients. Viruses 2022, 14, 796. [Google Scholar] [CrossRef]
- Haeusler, G.M.; Garnham, A.L.; Li-Wai-Suen, C.S.; Clark, J.E.; Babl, F.E.; Allaway, Z.; A Slavin, M.; Mechinaud, F.; Smyth, G.K.; Phillips, B.; et al. Blood transcriptomics identifies immune signatures indicative of infectious complications in childhood cancer patients with febrile neutropenia. Clin. Transl. Immunol. 2022, 11, e1383. [Google Scholar] [CrossRef]
- Ghaffari, K.; Falahati, V.; Motallebirad, T.; Safarabadi, M.; Tashakor, A.H.; Azadi, D. Microbiological and Molecular Study of Paranasal Sinus Infections of Children with Malignancy and Unknown Origin Fever in Markazi Province, Iran. Curr. Ther. Res. 2024, 100, 100745. [Google Scholar] [CrossRef] [PubMed]
- New Tool Indicates Woeful Diagnostic Readiness for Pandemic Prevention, Preparedness and Response. Available online: https://www.finddx.org/publications-and-statements/press-release/new-tool-indicates-woeful-diagnostic-readiness-for-pandemic-prevention-preparedness-and-response/ (accessed on 17 July 2025).
- Wilson, M.R.; Sample, H.A.; Zorn, K.C.; Arevalo, S.; Yu, G.; Neuhaus, J.; Federman, S.; Stryke, D.; Briggs, B.; Langelier, C.; et al. Clinical Metagenomic Sequencing for Diagnosis of Meningitis and Encephalitis. N. Engl. J. Med. 2019, 380, 2327–2340. [Google Scholar] [CrossRef] [PubMed]
- Rodino, K.G.; Simner, P.J. Status check: Next-generation sequencing for infectious-disease diagnostics. J. Clin. Investig. 2024, 134, e178003. [Google Scholar] [CrossRef] [PubMed]
- Sardzikova, S.; Andrijkova, K.; Svec, P.; Beke, G.; Klucar, L.; Minarik, G.; Bielik, V.; Kolenova, A.; Soltys, K. Gut diversity and the resistome as biomarkers of febrile neutropenia outcome in paediatric oncology patients undergoing hematopoietic stem cell transplantation. Sci. Rep. 2024, 14, 5504. [Google Scholar] [CrossRef]
- Heston, S.M.; Young, R.R.; Jenkins, K.; Martin, P.L.; Stokhuyzen, A.; Ward, D.V.; Bhattarai, S.K.; Bucci, V.; Arshad, M.; Chao, N.J.; et al. The effects of antibiotic exposures on the gut resistome during hematopoietic cell transplantation in children. Gut Microbes 2024, 16, 2333748. [Google Scholar] [CrossRef]
- Krumbholz, M.; Dolnik, A.; Sträng, E.; Ghete, T.; Skambraks, S.; Hutter, S.; Simonis, A.; Stegelmann, F.; Suttorp, M.; Horn, A.H.; et al. A high proportion of germline variants in pediatric chronic myeloid leukemia. Mol. Cancer 2024, 23, 206. [Google Scholar] [CrossRef]
- von Hardenberg, S.; Klefenz, I.; Steinemann, D.; Di Donato, N.; Baumann, U.; Auber, B.; Klemann, C. Current genetic diagnostics in inborn errors of immunity. Front. Pediatr. 2024, 12, 1279112. [Google Scholar] [CrossRef]
Clinical Impact | Comparison with Conventional Methods | Diagnostic Performance | Type of Infection/Pathogens Detected | NGS Approach | Sample Type | Patients Population | Study Type | Author |
---|---|---|---|---|---|---|---|---|
cfDNA NGS identified fungal pathogens in some high-risk patients but was not available in real-time and did not influence treatment decisions. Despite negative NGS results, 72% of patients without proven IFD received ≥1 week of antifungal therapy. | NGS showed good concordance with invasive fungal diagnostics (lung, pancreatic, scalp), detecting pathogens at the species level. Missed some tissue-only findings (e.g., Rhizopus oryzae), but identified P. jirovecii not found by conventional methods. | NGS identified fungal pathogens in 7 of 40 high-risk patients and matched conventional diagnostics in 4 of 6 proven IFD cases. Missed detections attributed to limited cfDNA release or uncertain clinical relevance. | Polymicrobial infections detected, including fungal (e.g., Aspergillus fumigatus, Candida spp., Rhizopus delemar, Pneumocystis jirovecii), viral (e.g., CMV, BK virus, HHV-6A, VZV), and bacterial pathogens (e.g., Escherichia coli, Pseudomonas spp., Enterococcus spp., Streptococcus mitis, Helicobacter pylori). | cfDNA NGS | Blood samples. | 40 pediatric hematology, oncology, and stem cell transplant patients at risk for invasive fungal disease (IFD). | Prospective observational cohort study | Armstrong et al., 2019 [21] |
mNGS guided antimicrobial adjustment in 55/70 (78.6%) patients, leading to clinical improvement. In 21.4%, results were irrelevant or missed likely pathogens (e.g., P. jirovecii, A. fumigatus), highlighting both utility and limitations in real-world use. | mNGS detected all pathogens found by conventional tests (5/70; 7.1%) and identified additional clinically relevant microbes in cases with negative routine diagnostics. Enabled overlapping detection across sample types (e.g., blood + swab in 14/34 cases). | mNGS showed a high detection rate (84.6%; 88/104 samples), identifying pathogens in both plasma and respiratory samples, including polymicrobial and rare infections. Enabled pathogen detection even when routine tests were negative. | Bacterial, viral, and fungal pathogens detected by mNGS, including Pseudomonas aeruginosa, Klebsiella pneumoniae, Staphylococcus aureus, Candida albicans, EBV, CMV, HSV-1, HHV-7, parvovirus B19, adenovirus, rhinovirus, and polyomaviruses. | mNGS | Plasma (n = 62), throat swabs (n = 34), bone marrow (n = 4), bronchoalveolar fluid (BALF) (n = 4); total 104 samples. | 70 febrile pediatric patients with hematological disorders; mostly immunocompromised due to chemotherapy, HCT, or underlying disease. | Prospective observational diagnostic study | Shen et al., 2021 [22] |
NGS findings suggest that translocation of oral, skin, and gut microbiota may contribute to FN pathogenesis in neutropenic patients. Potential to reveal hidden sources of infection when cultures are negative. | NGS confirmed all culture-positive FN cases and identified additional pathogens in culture-negative cases, demonstrating complementary value to conventional diagnostics. | NGS detected pathogens in 5/10 FN patients with positive cultures, 15/87 (17%) culture-negative FN cases, and 3/8 neutropenic enterocolitis patients. Showed added value in cases with negative standard diagnostics. | Putative bacterial pathogens detected in 15/87 culture-negative FN cases; confirmed all 5 culture-positive FN cases. DNA viruses (e.g., CMV, HHV-6B, EBV, TTV) in 19 patients; Malassezia restricta in 1 case. Findings suggest flora translocation. | mNGS | Plasma/serum samples. | Plasma/serum samples of 112 pediatric patients with FN and 10 patients with neutropenia without fever. | Retrospective diagnostic cohort study | Horiba et al., 2021 [23] |
mNGS revealed a high rate of co-infections (35.7%), emphasizing the need for careful clinical correlation. Frequent detection of viruses not matching symptoms highlighted the importance of integrating mNGS with host-response markers and clinical judgment. | Outperformed conventional methods in FUO by detecting pathogens in cases missed by standard tests; added microbiological value in 27.9% of patients. | mNGS identified pathogens in 76.2% (112/147) of FUO cases with prior negative conventional tests. Pathogens were considered causative in 44.6%, and clinical resolution followed therapy adjustment in 27.9%. | Bacterial, viral, and fungal pathogens detected: most common were P. aeruginosa, K. pneumoniae, Acinetobacter baumannii, CMV, HHV-1, parvovirus B19, A. fumigatus, and Candida parapsilosis. | mNGS | Blood samples. | 147 pediatric patients with hematological malignancies undergoing fever of unknown origin (FUO) (after chemotherapy or HCT). | Retrospective observational diagnostic study | Zhang et al., 2022 [24] |
mNGS supported antibiotic guidance through faster and broader pathogen detection, despite false negatives/positives. Interpretation was challenged by sample type (e.g., BALF contamination), delayed processing, and absence of RNA sequencing, limiting viral detection. | Compared to culture and RT-PCR, mNGS detected significantly more pathogens, especially fungi and viruses. mNGS outperformed conventional methods in BALF, blood, and CSF samples in terms of sensitivity, and enabled detection of mixed and atypical infections. | Higher sensitivity of mNGS vs. conventional tests (89.7% vs. 21.8%), especially for fungal and viral infections; slightly lower specificity (78.5% vs. 92.9%). | mNGS identified pathogens in 91.7% BAL, 85.7% blood, and 73.3% CSF samples; common pathogens: CMV, P. jirovecii, P. aeruginosa, K. pneumoniae, HHV-6B, Aspergillus, Mucor; mixed infections in 6 BAL and 5 blood cases. | mNGS | BALF (n = 54), blood (n = 32), and cerebrospinal fluid (CSF) (n = 15) samples. | 101 pediatric recipients after allo-HCT. | Retrospective observational diagnostic study | Qu et al., 2022 [25] |
mNGS supported timely and targeted antibiotic adjustments in cases not covered by empirical therapy, potentially improving outcomes. It also enabled de-escalation in selected cases. Authors recommend early BAL mNGS in children with hematologic malignancy and pulmonary infection. | Conventional tests included culture, serology (e.g., RSV, CMV, EBV), and fungal antigen assays (GM/G-test). mNGS detected mixed infections in 30.9% vs. 7.3% by conventional methods, especially bacterial–viral and fungal–viral co-infections. | mNGS alone had a higher positivity rate (87.3%) than conventional methods (34.5%, p < 0.001); it detected bacteria in 31%, viruses in 45.5%, and fungi in 34.5%. Mixed infections identified in 31% of cases. Combined with conventional tests, etiology was established in 91%. | Pulmonary infections with bacterial (e.g., Streptococcus pneumoniae, Haemophilus influenzae, S. aureus), viral (e.g., CMV, RSV, HPIV3, EBV), and fungal pathogens (e.g., P. jirovecii, A. fumigatus, R. oryzae) detected by mNGS in BALF. | mNGS | BALF samples. | 55 children with hematologic malignancies and suspected pulmonary infections undergoing bronchoscopy. | Retrospective cohort study | Wang et al., 2022 [26] |
mNGS improved etiologic diagnosis in FUO cases and enabled more targeted antimicrobial decisions. Integration with IL-6 enhanced clinical interpretation of ambiguous or mixed results, supporting precision treatment in immunocompromised children. | mNGS detected pathogens in 59.7% of cases missed by conventional methods, while only 3.1% were positive by conventional tests alone. Agreement was limited (27.1%), with differing results in 37% of concordant positives. mNGS had higher positivity rates across all sample types, especially NPS (94.3% vs. 18.9%) and BALF (96.7% vs. 36.7%). | mNGS was positive in 86.8% of cases vs. 30.2% with conventional methods; 59.7% of infections were detected by mNGS only. In 71.4% of mNGS-positive cases, pathogens were considered clinically relevant. IL-6 ≥ 390 pg/mL improved diagnostic precision for bacterial infections in mixed results. | mNGS detected bacteria (e.g., Haemophilus parainfluenzae, P. aeruginosa, K. pneumoniae), viruses (e.g., CMV, EBV, HHV-7, HSV-1, parvovirus B19), and fungi (e.g., P. jirovecii i, A. fumigatus, C. parapsilosis, Fusarium spp.). Co-infections (bacteria/viruses/fungi) were frequent; >50% of positives involved ≥2 pathogens. | mNGS | Blood (n = 157), nasopharyngeal swabs (n = 53), BALF (n = 30), sputum (n = 6), pus (n = 5), hydrothorax/ascites (n = 4), CSF (n = 3). | 258 febrile pediatric patients with leukemia, lymphoma, other malignancies, or HCT; infections included RTI/pneumonia, FUO, bloodstream infection (BSI), abdominal, CNS, GI, oral, and soft tissue infections. | Retrospective observational study | Wang et al., 2022 [27] |
Early use of mNGS (<48 h) in febrile, immunocompromised children was associated with shorter fever duration, lower anti-infective and hospitalization costs. High sensitivity in myelosuppressed patients enabled faster etiologic diagnosis and better-targeted treatment. | mNGS yielded significantly higher positivity than conventional methods (83.3% vs. 17.7%, p < 0.05); detected pathogens in 80 vs. 17 events, respectively. | mNGS showed higher sensitivity (91.8%) and NPV (56.3%) than conventional methods (17.7% and 11.4%, respectively), with similar specificity (81.8%). PPV was 97.5% for mNGS vs. 88.2% for conventional testing. | Detected bloodstream infections (76%), pneumonia (44.8%), and UTI (2.1%). Most common pathogens: P. aeruginosa (20.5%), K. pneumoniae (8.7%), CMV (21.3%), and Candida spp. (12.6%). | DNA-based mNGS (Illumina platform) | Blood, CSF, BALF, sputum, urine, and tissue samples; total n = 127 (including 107 blood, 6 CSF, 2 BALF, 3 sputum, 7 urine, 2 tissue). | 70 febrile pediatric patients (median age 5 y) with malignancies or hematologic disorders, including ALL, AML, NHL, LCH, aplastic anemia, RB, and Evans syndrome; all underwent mNGS testing. | Retrospective observational study | Fu et al., 2022 [28] |
mNGS results influenced clinical management in ~35% of cases, leading to initiation of targeted therapy in 39 patients and de-escalation in 3. Findings support mNGS as a complementary tool, particularly in culture-negative FN, though interpretation requires caution. | Outperformed conventional methods in detection rate (63.2% vs. 42.5%, p < 0.001); better at identifying mixed and rare infections. | mNGS showed higher sensitivity (90.9%) but lower specificity (12%) vs. TPD (sensitivity 24.7%, specificity 100%). mNGS positivity rate was 85.7% vs. 38.8% for TPD (p = 0.000); AUC for mNGS was 48.5%. | mNGS detected 70 pathogenic strains in 42 FN cases, including 25 mixed infections. Predominant pathogens included Aspergillus spp. and Gram-negative bacteria. Aspergillus was detected in 19 cases (often G-test/GM-test negative), and 13/20 Gram-negative bacteria were culture-negative but mNGS-positive. | mNGS | Plasma (n = 49) and BALF (n = 12). | 49 children with febrile neutropenia after chemotherapy; majority with leukemia (ALL: 65.3%, AML: 30.6%); 2 cases with HLH. | Retrospective cohort study | Guo et al., 2022 [29] |
Early HAdV detection by mNGS may enable timely antiviral treatment and improve outcomes. Highlights the value of mNGS in diagnosing adenovirus infections in immunocompromised pediatric patients post-haplo-HCT. | mNGS offers a comprehensive diagnostic approach and may detect infections missed by conventional methods; no direct comparison or statistical analysis reported. | mNGS successfully identified HAdV infections post-haplo-HCT; no formal sensitivity or specificity metrics reported. | Systemic human adenovirus (HAdV) infection involving blood, urine, CSF, and lungs; clinical manifestations included ADV hepatitis and encephalitis. | mNGS | Blood, CSF, urine, and BALF samples. | 7 patients with adenovirus infection after haploidentical HCT (from cohort of 976); 6 with acute leukemia, 1 with aplastic anemia; 5 male, 2 female. | Retrospective multicenter study | Wu et al., 2023 [30] |
mNGS guided treatment adjustments (e.g., linezolid initiation, antifungal addition, antibiotic de-escalation), with most patients improving clinically. Enabled early differentiation between infectious and non-infectious fevers. | mNGS showed higher and earlier pathogen detection than culture, identifying organisms missed by conventional methods. In one case, K. pneumoniae was confirmed by culture three days after mNGS detection. | mNGS showed higher positivity for bacteria and fungi (57.2%) compared to culture (12.5%, p < 0.01); detected bacteria (n = 27), fungi (n = 12), viruses (n = 7), and mixed infections (n = 16). No fungi were detected by culture. | Infections included sepsis, respiratory tract infections, and febrile neutropenia with suspected bloodstream infection. mNGS identified pathogens in 85/96 specimens (88.5%), including simple bacterial (43.5%), fungal (19.4%), viral (11.3%) and mixed infections (25.8%). Frequent pathogens: K. pneumoniae, P. aeruginosa, A. baumannii, E. coli, Yersinia pneumoniae, Aspergillus flavus, and human herpesviruses. | mNGS | 96 specimens: plasma (n = 71), CSF (n = 11), sputum (n = 8), BALF (n = 2), hydrothorax, urine, liver biopsy, and abscess fluid (n = 1 each). | 67 pediatric patients with hematological diseases, mostly ALL (52.2%) and AML (14.9%); 28.4% post-HCT; 39 males. | Retrospective observational study | Zhang et al., 2023 [31] |
mNGS influenced clinical diagnosis in 91.7% and led to treatment modifications in 95.8% of immunocompromised patients. The study highlights the value of mNGS for rapid, comprehensive pathogen detection in critically ill immunocompromised children, where infections may be atypical or otherwise undiagnosed. | mNGS showed superior pathogen detection compared to conventional microbiological testing (CMT), with a significantly higher positivity rate (76.5% vs. 55.5%). mNGS identified additional pathogens missed by CMT, especially in mixed infections and among immunocompromised patients. In some cases, mNGS provided the only microbiologic evidence of infection. | mNGS positivity rate: 76.5%, significantly higher than conventional testing (55.5%, p = 0.0006). Positive percent agreement was higher in immunocompromised patients (95.2%) vs. immunocompetent (77.8%). | Detected Gram-positive and Gram-negative bacteria (e.g., S. pneumoniae, K. pneumoniae), Herpesviridae viruses (CMV, EBV, HSV), respiratory viruses (RSV, HPIV, HRV), and fungi including P. jirovecii and C.albicans. Co-infections were common in immunocompromised children. | DNA mNGS (BGI platform) | 119 clinical samples, including BALF, CSF, blood, stool, peritoneal fluid, pleural fluid, pus, sputum, and swabs; 48 samples from immunocompromised patients. | 48 immunocompromised pediatric patients (avg. age 88 mo), mainly with leukemia (52.1%) and solid tumors (20.8%). Most common symptoms: fever (84.9%), cough (29.4%), convulsions (2.1%). Total of 119 samples analyzed. | Retrospective cohort study | Xu et al., 2024 [32] |
mNGS guided treatment changes (54.3%), improved diagnostic confidence, reduced unnecessary antibiotics, and enabled precise therapy in pediatric cancer patients with suspected BSI. | mNGS significantly outperformed conventional microbiological tests (culture, PCR for EBV/CMV, GM/G tests) in sensitivity, clinical agreement, and pathogen coverage—especially for viruses. While both methods were positive in 25.4% of samples, only 15.8% showed identical pathogen identification. mNGS detected 94.5% of clinician-confirmed pathogens, including 100% of viruses, and was the sole method to detect 75.2% of them. | mNGS outperformed conventional tests in sensitivity (89.8% vs. 32.5%) and clinical agreement (76.3% vs. 51.3%). | The overall positive detection rate of mNGS regardless of clinical relevance was 69.2% (155/224). mNGS demonstrated higher sensitivity (89.8%) compared to conventional tests (32.5%, p < 0.001); higher clinical agreement (76.3% vs. 51.3%, p < 0.001). | mNGS: metaDNA-seq (n = 223), metaRNA-seq (n = 1), and both DNA/RNA sequencing (n = 8). | 224 blood samples analyzed. | 195 pediatric oncology patients with suspected bloodstream infections (BSI). | Retrospective observational study | Wu et al., 2024 [33] |
Plasma mNGS influenced clinical management in 13% of cases (14/104), including new or earlier diagnoses in 8 cases. De-escalation of therapy occurred in 28% of cases with positive clinical impact. However, incidental or non-pathogenic organisms were frequently detected, and mNGS led to negative impacts in 4 cases. The study emphasizes the need for cautious interpretation and further prospective validation. | Plasma mNGS showed low concordance with conventional diagnostics. It detected additional organisms not found by standard tests, but many were judged clinically irrelevant or non-causative. Positive percent agreement with conventional diagnostics was 50%; negative agreement was 44%. In some cases, mNGS uniquely identified pathogens missed by other methods, but interpretation was limited by frequent detection of background or non-pathogenic organisms. | Overall agreement between plasma mNGS and conventional diagnostics was 47%. Among confirmed infections, positive percent agreement was 50%; negative agreement in non-infectious cases was 44%. In total, 63.8% of mNGS results identified at least one organism; 33% were concordant with final diagnosis; in 8.5% mNGS uniquely identified the causative pathogen. | Plasma mNGS detected a broad range of organisms, including bacteria, viruses, and fungi., e.g., P. jirovecii, S. aureus, E. coli, P. aeruginosa, CMV, EBV, and Candida spp. Some detected organisms (e.g., Torque teno virus, Anelloviridae) were of unclear clinical significance. | mNGS | 104 plasma samples analyzed via mNGS; repeat testing performed in 22 patients (2–4 tests per patient). | 71 immunocompromised pediatric patients, including those with hematologic and solid tumors, HCT, and/or solid organ transplantation; tested for indications such as fever, pulmonary syndrome, sepsis, deep-seated, CNS, or musculoskeletal infections. | Retrospective observational study | Lehman et al., 2024 [10] |
Limited clinical impact. In 8.5% of cases, mNGS findings alone led to final diagnosis. More commonly provided confirmatory or supplementary data rather than guiding initial diagnosis or therapy. Delayed turnaround (~9 days) reduced utility in acute management. | BAL mNGS showed lower clinical concordance (13.9%) compared to other studies (e.g., 75.6% in adult ICU). Often performed after negative conventional tests, with delayed turnaround. Provided unique diagnostic information in 8.5% of cases. Detected organism types varied by host condition and antimicrobial timing, offering added value in selected subgroups. | BAL mNGS increased diagnostic yield, particularly for viral pathogens. Of 36 tests, 63.8% identified ≥1 organism, but only 13.9% (8/36) were concordant with final ARI diagnosis. Approximately 50% of mNGS results provided additional diagnostic information beyond conventional methods. | Suspected pulmonary infections. mNGS on BAL identified bacterial, viral, and fungal pathogens including P. aeruginosa, Enterobacter cloacae, Enterococcus faecium, CMV, P. jirovecii, yeasts, and molds. | mNGS | 137 BALF samples collected from 108 patients. Among these, 36 underwent mNGS testing. | 108 immunocompromised patients, including hematologic/solid malignancies, aplastic anemia, sickle cell disease with asplenia, and post-HCT. | Single-center retrospective case series | Abraham et al., 2025 [34] |
Viral metagenomics improved characterization of viral diversity in FN, identifying clinically relevant viruses, though direct impact on treatment decisions was unclear. Viral composition differed more by sample type than FN diagnosis. Highlights need for further research on clinical utility of viral mNGS in FN management. | Viral metagenomics detected a broader spectrum of viruses than routine PCR. qPCR confirmed key findings (e.g., herpesviruses, polyomaviruses), while nested PCR and Sanger sequencing were used for adenovirus typing. mNGS enabled detection of co-infections and uncommon viruses not routinely tested for. | Viral mNGS demonstrated higher detection rates than standard PCR, including co-infections and rare viruses. Of 1.42 billion post-trimming reads, 21.2% were classified, with 12% of those viral. FN plasma samples had higher viral read counts than swabs. | Viral infections in FN patients: Herpesviridae, Anelloviridae, Adenoviridae, Polyomaviridae; SARS-CoV-2 also detected. | Viral mNGS with Kraken2-based taxonomic classification and qPCR confirmation | Blood and oropharyngeal samples (paired). | 15 pediatric patients presenting with febrile neutropenia at admission. Control group: 15 pediatric oncology patients undergoing treatment or in remission. | Case–control study | Sarana da Silva et al., 2025 [35] |
Clinical Impact | Comparison with Conventional Methods | Diagnostic Performance | Type of Infection/Pathogens Detected | NGS Approach | Sample Type | Patients Population | Study Type | Author |
---|---|---|---|---|---|---|---|---|
WGS enabled detailed analysis of resistance and virulence genes in A. baumannii isolates, supporting infection control efforts and informing future treatment strategies through improved understanding of bacterial transmission. | WGS enabled detailed resistance gene profiling and clonal lineage assignment not achievable with conventional phenotypic or culture-based methods. | Not applicable—study focused on WGS for resistance profiling, not diagnostic detection. | Multidrug-resistant A. baumannii. | WGS | Blood (n = 16), central venous port blood (n = 6), BALF samples (n = 6), wound, tissue, and pleural fluid samples (n = 1 each). | 27 infected pediatric cancer patients with different types of malignancies. | Retrospective genomic surveillance study | Jalal et al., 2021 [36] |
WGS findings informed enhanced infection control strategies and supported recommendations for routine genomic surveillance in HCT settings. Enabled detailed outbreak mapping and identification of international HAdV-A31 dissemination. | WGS offered higher resolution than conventional methods, revealing nosocomial transmission and international linkages undetected by standard epidemiology. | WGS enabled high-resolution phylogenetic analysis; 17/20 HAdV-A31 isolates clustered closely (0–8 mutations), indicating outbreak. No formal sensitivity/specificity reported. | Human adenovirus detected in 57 episodes (86% blood PCR positive); WGS identified HAdV-A31 (outbreak strain), HAdV-C1, and HAdV-C2. No evidence of recent transmission based on phylogenetic analysis. | WGS with Illumina MiSeq | Stool culture isolates (n = 15), urine culture (n = 2), direct WGS from urine (n = 2) and stool (n = 1). | 55 pediatric HCT recipients, with leukemia/lymphoma (43%), solid tumors (24%), immunodeficiency (16%), hematologic disorders (13%), and Hurler syndrome (4%). | Retrospective case series | Fattouh et al., 2022 [37] |
WGS confirmed nosocomial SARS-CoV-2 transmission in immunocompromised pediatric patients, supporting recommendations for enhanced infection control and routine genomic surveillance in hospital settings. | WGS provided higher resolution than RT-PCR, enabling identification of transmission links and viral mutations not detectable by standard methods. | WGS revealed high genomic similarity among SARS-CoV-2 isolates, indicating nosocomial transmission. No formal sensitivity or specificity reported. | Viral infection: SARS-CoV-2 (lineage B.1.470). | WGS | Nasopharyngeal swabs. | 5 immunocompromised children (AML/Ewing sarcoma, age 1–14), mostly male. | Retrospective case series | Putri et al., 2022 [38] |
WGS enabled targeted infection prevention and control investigations and may have prevented further transmission. None of the clusters would have been identified without WGS. This was the first broad, prospective WGS-based bacterial surveillance study in immunocompromised pediatric patients over multiple years. | Conventional surveillance failed to detect any of the 18 WGS-identified transmission clusters. Standard methods relied on clinical suspicion and basic epidemiology, missing silent or indirect transmission events revealed by genomic relatedness. | WGS identified 18 multi-patient transmission clusters among 1497 isolates (1.2% of total), spanning 9 bacterial species. Genomic data enabled detection of transmission with as few as 0–20 allelic differences (cgMLST), enhancing resolution beyond conventional typing. | Healthcare-associated infections, including bloodstream, wound, and catheter-related infections. Detected pathogens included S. aureus, E. coli, K. pneumoniae, P. aeruginosa, E. cloacae, Enterococcus faecalis, and Pseudomonas putida. | WGS with core genome multilocus sequence typing (cgMLST) | Clinical diagnostic specimens (inpatient and outpatient), including blood, respiratory tract, urine, skin/soft tissue, and sterile sites; species with ≥3 isolates per year included CoNS excluded. | 1025 patients; 1497 bacterial isolates (16 species) obtained from clinical diagnostic specimens as part of a genomic surveillance program. | Prospective observational cohort study | Hakim et al., 2024 [39] |
Clinical Impact | Comparison with Conventional Methods | Diagnostic Performance | Type of Infection/Pathogens Detected | NGS Approach | Sample Type | Patients Population | Study Type | Author |
---|---|---|---|---|---|---|---|---|
NGS enabled detection of novel and rare viruses (e.g., AstV VA3) not captured by PCR, supporting its potential relevance in HCT patients. Asymptomatic enteric viral presence may predispose to gastrointestinal GVHD and worse outcomes. | ViroCap matched all PCR-confirmed detections and identified viruses missed by PCR, EIA, and ICT, suggesting broader detection range than conventional assays. | ViroCap confirmed viruses detected by clinical PCR (e.g., ADV, NoV) and additionally identified other clinically relevant viruses (e.g., BKV, HRV, HHV-7) missed by routine testing. | Viral infections; detected pathogens included adenoviruses (A, C, AAV), norovirus, BKV, HRV (B, C), KI virus, HHV-7, astrovirus VA3, and alphatorquevirus. | Targeted NGS using ViroCap (hybrid-capture panel covering 34 viral families and 337 species of DNA/RNA viruses). | Stool samples | 11 clinical pediatric HCT recipients with gastrointestinal symptoms, suspected of GVHD | Retrospective diagnostic study | Jansen et al., 2020 [40] |
Study highlights need to screen HCT patients and donors for HPgV-1 to reduce transmission risk. First-time full-genome characterization of HPgV-1 in this cohort; phylogenetic and intra-host variation analyses provided insight into viral diversity. | TE-mNGS enabled detection and full genome characterization of HPgV-1, which was not achievable by conventional methods. | HPgV-1 detected in 3/14 patients (21.4%) by TE-mNGS; findings confirmed by qRT-PCR. No formal sensitivity or specificity reported. | HPgV-1 detected in 3 patients (day 0 and day 3 samples); CMV detected in 1 patient. | Target enrichment metagenomic NGS (TE-mNGS) using Illumina MiSeq; applied to detect viruses beyond routine qRT-PCR coverage. | Plasma samples from pediatric HCT recipients analyzed to detect human pegivirus-1 | 14 pediatric patients post-HCT, 13 with febrile neutropenia | Retrospective case series | Ludowyke et al., 2022 [41] |
Clinical Impact | Comparison with Conventional Methods | Diagnostic Performance | Type of Infection/Pathogens Detected | NGS Approach | Sample Type | Patients Population | Study Type | Author |
---|---|---|---|---|---|---|---|---|
Host transcriptomic profiles distinguished unexplained fever from true infections, suggesting many FUO episodes may not reflect occult infection. Gene expression signatures identifying bacteremia could support targeted antibiotic use in FN patients. | Conventional microbiological tests were often inconclusive; transcriptomic profiling provided discriminatory host-response signatures even in culture-negative episodes. | Transcriptomic analysis distinguished bacteremia and non-bloodstream MDI from unexplained fever via distinct gene expression profiles (e.g., 1206 DEGs in bacteremia vs. unexplained fever); limited discrimination between bacterial and viral MDI. | Bacteremia, non-bloodstream MDI (bacterial/viral), CDI, and unexplained fever. Transcriptomic profiles indicated host responses to pathogens including S. aureus, E. coli, Mycobacterium tuberculosis, Salmonella, and Leishmania. | RNA-seq | Peripheral blood mononuclear cell (PBMC) samples | 64 pediatric patients with solid tumors or leukemia on active treatment; 80 FN episodes included for transcriptomic analysis | Prospective multicenter observational cohort study | Haeusler et at, 2022 [42] |
Findings highlight the value of sinus evaluation and molecular diagnostics (e.g., 16S rRNA) in immunocompromised pediatric patients with FUO, supporting targeted therapy. Emphasizes need for expanded diagnostics in resource-limited settings. | Combined phenotypic and molecular methods improved pathogen identification compared to culture alone; enhanced diagnostic precision in sinus infections. | Culture positivity rate was 40% (36/90); molecular methods enabled precise species-level identification of bacterial pathogens. | Paranasal sinus infections (sinusitis); 36 bacterial isolates (40%), including P. aeruginosa, Streptococcus agalactiae, S. aureus, E. coli, K. pneumoniae, A. baumannii, Nocardia spp., S. pneumoniae, E. faecium. | 16S rRNA gene sequencing | Paranasal sinus samples (n = 90), analyzed using phenotypic and molecular methods. | 90 febrile pediatric patients with malignancy and FUO; underlying diseases: ALL (52.2%), Burkitt’s lymphoma (18.9%), aplastic anemia (14.5%), osteosarcoma (7.8%), medulloblastoma (6.7%) | Retrospective observational study | Ghaffari et al., 2024 [43] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Jabłońska, A.; Sadkowski, A.; Richert-Przygońska, M.; Styczyński, J. Next-Generation Sequencing for Infectious Disease Diagnostics in Pediatric Patients with Malignancies or After Hematopoietic Cell Transplantation: A Systematic Review. J. Clin. Med. 2025, 14, 6444. https://doi.org/10.3390/jcm14186444
Jabłońska A, Sadkowski A, Richert-Przygońska M, Styczyński J. Next-Generation Sequencing for Infectious Disease Diagnostics in Pediatric Patients with Malignancies or After Hematopoietic Cell Transplantation: A Systematic Review. Journal of Clinical Medicine. 2025; 14(18):6444. https://doi.org/10.3390/jcm14186444
Chicago/Turabian StyleJabłońska, Anna, Aleksander Sadkowski, Monika Richert-Przygońska, and Jan Styczyński. 2025. "Next-Generation Sequencing for Infectious Disease Diagnostics in Pediatric Patients with Malignancies or After Hematopoietic Cell Transplantation: A Systematic Review" Journal of Clinical Medicine 14, no. 18: 6444. https://doi.org/10.3390/jcm14186444
APA StyleJabłońska, A., Sadkowski, A., Richert-Przygońska, M., & Styczyński, J. (2025). Next-Generation Sequencing for Infectious Disease Diagnostics in Pediatric Patients with Malignancies or After Hematopoietic Cell Transplantation: A Systematic Review. Journal of Clinical Medicine, 14(18), 6444. https://doi.org/10.3390/jcm14186444