Submitted:
15 July 2025
Posted:
16 July 2025
You are already at the latest version
Abstract
Keywords:
Introduction
Methods
- (1)
- Article must use an analytical technique to show direct or proxy evidence for spike protein or vaccinal mRNA.
- (2)
- Sample must be taken from a vaccinated individual.
- (1)
- Spike protein is attributed to SARS-CoV-2 infection by the study authors.
Results
Analytical Techniques
Tissue/Organ Spike/mRNA Persistency
Characterizing the Decay Curves
Spike as a Potential Biomarker
Proposed Persistence Mechanisms
Discussion
References
- M. T. J. Halma, J. Rose, and T. Lawrie, “The Novelty of mRNA Viral Vaccines and Potential Harms: A Scoping Review,” J, vol. 6, no. 2, Art. no. 2, Jun. 2023. [CrossRef]
- M. Alberer et al., “Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial,” The Lancet, vol. 390, no. 10101, pp. 1511–1520, Sep. 2017. [CrossRef]
- R. A. Feldman et al., “mRNA vaccines against H10N8 and H7N9 influenza viruses of pandemic potential are immunogenic and well tolerated in healthy adults in phase 1 randomized clinical trials,” Vaccine, vol. 37, no. 25, pp. 3326–3334, May 2019. [CrossRef]
- on behalf of the iHIVARNA consortium et al., “iHIVARNA phase IIa, a randomized, placebo-controlled, double-blinded trial to evaluate the safety and immunogenicity of iHIVARNA-01 in chronically HIV-infected patients under stable combined antiretroviral therapy,” Trials, vol. 20, no. 1, p. 361, Dec. 2019. [CrossRef]
- C. Aldrich et al., “Proof-of-concept of a low-dose unmodified mRNA-based rabies vaccine formulated with lipid nanoparticles in human volunteers: A phase 1 trial,” Vaccine, vol. 39, no. 8, pp. 1310–1318, Feb. 2021. [CrossRef]
- K. Bahl et al., “Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza Viruses,” Mol. Ther., vol. 25, no. 6, pp. 1316–1327, Jun. 2017. [CrossRef]
- J. M. Jacobson et al., “Dendritic Cell Immunotherapy for HIV-1 Infection Using Autologous HIV-1 RNA: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial,” vol. 72, no. 1, 2016. [CrossRef]
- R. T. Gandhi et al., “Immunization of HIV-1-Infected Persons With Autologous Dendritic Cells Transfected With mRNA Encoding HIV-1 Gag and Nef: Results of a Randomized, Placebo-Controlled Clinical Trial,” vol. 71, no. 3, 2016. [CrossRef]
- L. Leal et al., “Phase I clinical trial of an intranodally administered mRNA-based therapeutic vaccine against HIV-1 infection,” AIDS, vol. 32, no. 17, pp. 2533–2545, Nov. 2018. [CrossRef]
- L. G. Boros, A. M. Kyriakopoulos, C. Brogna, M. Piscopo, P. A. McCullough, and S. Seneff, “Long-lasting, biochemically modified mRNA, and its frameshifted recombinant spike proteins in human tissues and circulation after COVID-19 vaccination,” Pharmacol. Res. Perspect., vol. 12, no. 3, p. e1218, 2024. [CrossRef]
- I. Pateev, K. Seregina, R. Ivanov, and V. Reshetnikov, “Biodistribution of RNA Vaccines and of Their Products: Evidence from Human and Animal Studies,” Biomedicines, vol. 12, no. 1, p. 59, Dec. 2023. [CrossRef]
- C. Brogna et al., “Detection of recombinant Spike protein in the blood of individuals vaccinated against SARS-CoV-2: Possible molecular mechanisms,” PROTEOMICS – Clin. Appl., vol. 17, no. 6, p. 2300048, Nov. 2023. [CrossRef]
- B. König and J. O. Kirchner, “Methodological Considerations Regarding the Quantification of DNA Impurities in the COVID-19 mRNA Vaccine Comirnaty®,” Methods Protoc., vol. 7, no. 3, Art. no. 3, Jun. 2024. [CrossRef]
- D. Speicher, J. Rose, L. Gutschi, D. Wiseman, and K. McKernan, Speicher DJ et al, DNA fragments detected in COVID-19 vaccines in Canada. DNA fragments detected in monovalent and bivalent. 2023.
- O. Andries, S. Mc Cafferty, S. C. De Smedt, R. Weiss, N. N. Sanders, and T. Kitada, “N1-methylpseudouridine-incorporated mRNA outperforms pseudouridine-incorporated mRNA by providing enhanced protein expression and reduced immunogenicity in mammalian cell lines and mice,” J. Controlled Release, vol. 217, pp. 337–344, Nov. 2015. [CrossRef]
- C. Leong, L. Jin, D. Kim, J. Kim, Y. Y. Teo, and T.-H. Ho, “Assessing the impact of novelty and conformity on hesitancy towards COVID-19 vaccines using mRNA technology,” Commun. Med., vol. 2, no. 1, pp. 1–6, May 2022. [CrossRef]
- H. Zhang et al., “Fluorescence-Based Quantification of Messenger RNA and Plasmid DNA Decay Kinetics in Extracellular Biological Fluids and Cell Extracts,” Adv. Biosyst., vol. 4, no. 5, p. 2000057, 2020. [CrossRef]
- J. B. Ulmer, P. W. Mason, A. Geall, and C. W. Mandl, “RNA-based vaccines,” Vaccine, vol. 30, no. 30, pp. 4414–4418, Jun. 2012. [CrossRef]
- F. P. Polack et al., “Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine,” N. Engl. J. Med., vol. 383, no. 27, pp. 2603–2615, Dec. 2020. [CrossRef]
- L. R. Baden et al., “Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine,” N. Engl. J. Med., vol. 384, no. 5, pp. 403–416, Feb. 2021. [CrossRef]
- “Authorizations of Emergency Use of Two Biological Products During the COVID-19 Pandemic; Availability,” Federal Register. Accessed: Jun. 02, 2025. [Online]. Available: https://www.federalregister.gov/documents/2021/01/19/2021-01022/authorizations-of-emergency-use-of-two-biological-products-during-the-covid-19-pandemic-availability.
- M. T. J. Halma, C. Plothe, P. Marik, and T. A. Lawrie, “Strategies for the Management of Spike Protein-Related Pathology,” Microorganisms, vol. 11, no. 5, Art. no. 5, May 2023. [CrossRef]
- B. K. Patterson et al., “Detection of S1 spike protein in CD16+ monocytes up to 245 days in SARS-CoV-2-negative post-COVID-19 vaccine syndrome (PCVS) individuals,” Hum. Vaccines Immunother., vol. 21, no. 1, p. 2494934, Dec. 2025. [CrossRef]
- L. Huang et al., “Human body-fluid proteome: quantitative profiling and computational prediction,” Brief. Bioinform., vol. 22, no. 1, pp. 315–333, Jan. 2021. [CrossRef]
- A. J. Krauson, F. V. C. Casimero, Z. Siddiquee, and J. R. Stone, “Duration of SARS-CoV-2 mRNA vaccine persistence and factors associated with cardiac involvement in recently vaccinated patients,” Npj Vaccines, vol. 8, no. 1, p. 141, Sep. 2023. [CrossRef]
- L. M. Yonker et al., “Circulating Spike Protein Detected in Post–COVID-19 mRNA Vaccine Myocarditis,” Circulation, vol. 147, no. 11, pp. 867–876, Mar. 2023. [CrossRef]
- S. J. Kent et al., “Blood Distribution of SARS-CoV-2 Lipid Nanoparticle mRNA Vaccine in Humans,” ACS Nano, vol. 18, no. 39, pp. 27077–27089, Oct. 2024. [CrossRef]
- N. Hanna et al., “Detection of Messenger RNA COVID-19 Vaccines in Human Breast Milk,” JAMA Pediatr., vol. 176, no. 12, p. 1268, Dec. 2022. [CrossRef]
- B. Bhattacharjee et al., “Immunological and Antigenic Signatures Associated with Chronic Illnesses after COVID-19 Vaccination,” Feb. 25, 2025, medRxiv. [CrossRef]
- T. E. Fertig et al., “Vaccine mRNA Can Be Detected in Blood at 15 Days Post-Vaccination,” Biomedicines, vol. 10, no. 7, p. 1538, Jun. 2022. [CrossRef]
- J. A. S. Castruita et al., “SARS-CoV -2 spike mRNA vaccine sequences circulate in blood up to 28 days after COVID -19 vaccination,” APMIS, vol. 131, no. 3, pp. 128–132, Mar. 2023. [CrossRef]
- K. Röltgen et al., “Immune imprinting, breadth of variant recognition, and germinal center response in human SARS-CoV-2 infection and vaccination,” Cell, vol. 185, no. 6, pp. 1025-1040.e14, Mar. 2022. [CrossRef]
- J. M. Low et al., “Codominant IgG and IgA expression with minimal vaccine mRNA in milk of BNT162b2 vaccinees,” Npj Vaccines, vol. 6, no. 1, p. 105, Aug. 2021. [CrossRef]
- “National Center for Biotechnology Information.” Accessed: May 25, 2025. [Online]. Available: https://www.ncbi.nlm.nih.gov/.
- “Thermo Fisher Scientific - US.” Accessed: May 25, 2025. [Online]. Available: https://www.thermofisher.com/us/en/home.html.
- “Leading Life Science Research & Clinical Diagnostics | Bio-Rad.” Accessed: May 25, 2025. [Online]. Available: https://www.bio-rad.com/.
- CDC, “About OneLabTM,” CDC Laboratory Training. Accessed: May 25, 2025. [Online]. Available: https://www.cdc.gov/lab-training/php/onelab/index.html.
- “Ultra-Sensitive Biomarker Detection,” Quanterix. Accessed: May 25, 2025. [Online]. Available: https://www.quanterix.com/.
- Z. Rong et al., “Persistence of spike protein at the skull-meninges-brain axis may contribute to the neurological sequelae of COVID-19,” Cell Host Microbe, vol. 32, no. 12, pp. 2112-2130.e10, Dec. 2024. [CrossRef]
- J. S. Turner et al., “SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses,” Nature, vol. 596, no. 7870, pp. 109–113, Aug. 2021. [CrossRef]
- E. Taus et al., “Persistent memory despite rapid contraction of circulating T Cell responses to SARS-CoV-2 mRNA vaccination,” Front. Immunol., vol. 14, p. 1100594, Feb. 2023. [CrossRef]
- N. Ota et al., “Expression of SARS-CoV-2 spike protein in cerebral Arteries: Implications for hemorrhagic stroke Post-mRNA vaccination,” J. Clin. Neurosci., vol. 136, p. 111223, Jun. 2025. [CrossRef]
- L. Martin-Navarro, C. De Andrea, B. Sangro, and J. Argemi, “In situ detection of vaccine mRNA in the cytoplasm of hepatocytes during COVID-19 vaccine-related hepatitis,” J. Hepatol., vol. 78, no. 1, pp. e20–e22, Jan. 2023. [CrossRef]
- M. Mörz, “A Case Report: Multifocal Necrotizing Encephalitis and Myocarditis after BNT162b2 mRNA Vaccination against COVID-19,” Vaccines, vol. 10, no. 10, p. 1651, Oct. 2022. [CrossRef]
- Y. Yoshimura et al., “An autopsy case of COVID-19-like acute respiratory distress syndrome after mRNA-1273 SARS-CoV-2 vaccination.,” Int. J. Infect. Dis., vol. 121, pp. 98–101, Aug. 2022. [CrossRef]
- C. M. Nyein, Z. H. S. Liew, W.-Q. Leow, P. S. J. Yeong, and G. H. Ho, “Severe de novo liver injury after Moderna vaccination – not always autoimmune hepatitis,” J. Hepatol., vol. 77, no. 2, pp. 556–558, Aug. 2022. [CrossRef]
- E. Magen et al., “Clinical and Molecular Characterization of a Rare Case of BNT162b2 mRNA COVID-19 Vaccine-Associated Myositis,” Vaccines, vol. 10, no. 7, p. 1135, Jul. 2022. [CrossRef]
- A. M. Fleming and C. J. Burrows, “Nanopore sequencing for N1-methylpseudouridine in RNA reveals sequence-dependent discrimination of the modified nucleotide triphosphate during transcription,” Nucleic Acids Res., vol. 51, no. 4, pp. 1914–1926, Feb. 2023. [CrossRef]
- W. Li et al., “Stability of SARS-CoV-2-Encoded Proteins and Their Antibody Levels Correlate with Interleukin 6 in COVID-19 Patients,” mSystems, vol. 7, no. 3, pp. e00058-22, May 2022. [CrossRef]
- M. Lu et al., “SARS-CoV-2 prefusion spike protein stabilized by six rather than two prolines is more potent for inducing antibodies that neutralize viral variants of concern,” Proc. Natl. Acad. Sci., vol. 119, no. 35, p. e2110105119, Aug. 2022. [CrossRef]
- Y. V. Khramtsov et al., “Intracellular Degradation of SARS-CoV-2 N-Protein Caused by Modular Nanotransporters Containing Anti-N-Protein Monobody and a Sequence That Recruits the Keap1 E3 Ligase,” Pharmaceutics, vol. 16, no. 1, Art. no. 1, Jan. 2024. [CrossRef]
- M. T. J. Halma, P. E. Marik, and Y. M. Saleeby, “Exploring autophagy in treating SARS-CoV-2 spike protein-related pathology,” Endocr. Metab. Sci., vol. 14, p. 100163, Mar. 2024. [CrossRef]
- P. I. Parry et al., “‘Spikeopathy’: COVID-19 Spike Protein Is Pathogenic, from Both Virus and Vaccine mRNA,” Biomedicines, vol. 11, no. 8, Art. no. 8, Aug. 2023. [CrossRef]
- I. P. Trougakos et al., “Adverse effects of COVID-19 mRNA vaccines: the spike hypothesis,” Trends Mol. Med., vol. 28, no. 7, pp. 542–554, Jul. 2022. [CrossRef]
- M. Cosentino and F. Marino, “Understanding the Pharmacology of COVID-19 mRNA Vaccines: Playing Dice with the Spike?,” Int. J. Mol. Sci., vol. 23, no. 18, p. 10881, Sep. 2022. [CrossRef]
- E. Avolio et al., “The SARS-CoV-2 Spike protein disrupts human cardiac pericytes function through CD147 receptor-mediated signalling: a potential non-infective mechanism of COVID-19 microvascular disease,” Clin. Sci. Lond. Engl. 1979, vol. 135, no. 24, pp. 2667–2689, Dec. 2021. [CrossRef]
- A. Buoninfante, A. Andeweg, G. Genov, and M. Cavaleri, “Myocarditis associated with COVID-19 vaccination,” Npj Vaccines, vol. 9, no. 1, pp. 1–8, Jun. 2024. [CrossRef]
- J. Fraiman et al., “Serious adverse events of special interest following mRNA COVID-19 vaccination in randomized trials in adults,” Vaccine, vol. 40, no. 40, pp. 5798–5805, Sep. 2022. [CrossRef]
- A. K. Mundorf et al., “Clinical and Diagnostic Features of Post-Acute COVID-19 Vaccination Syndrome (PACVS),” Vaccines, vol. 12, no. 7, Art. no. 7, Jul. 2024. [CrossRef]
- B. Platschek and F. Boege, “The Post-Acute COVID-19-Vaccination Syndrome in the Light of Pharmacovigilance,” Vaccines, vol. 12, no. 12, Art. no. 12, Dec. 2024. [CrossRef]
- F. Scholkmann and C.-A. May, “COVID-19, post-acute COVID-19 syndrome (PACS, ‘long COVID’) and post-COVID-19 vaccination syndrome (PCVS, ‘post-COVIDvac-syndrome’): Similarities and differences,” Pathol. - Res. Pract., vol. 246, p. 154497, Jun. 2023. [CrossRef]
- H. M. Krumholz et al., “Post-Vaccination Syndrome: A Descriptive Analysis of Reported Symptoms and Patient Experiences After Covid-19 Immunization,” medRxiv, p. 2023.11.09.23298266, Nov. 2023. [CrossRef]
- B. Chen, B. Julg, S. Mohandas, S. B. Bradfute, and RECOVER Mechanistic Pathways Task Force, “Viral persistence, reactivation, and mechanisms of long COVID,” eLife, vol. 12, p. e86015, May 2023. [CrossRef]
- T. C. Theoharides, “Could SARS-CoV-2 Spike Protein Be Responsible for Long-COVID Syndrome?,” Mol. Neurobiol., vol. 59, no. 3, pp. 1850–1861, Mar. 2022. [CrossRef]
- N. Hulscher, P. A. McCullough, and D. E. Marotta, “Strategic deactivation of mRNA COVID-19 vaccines: New applications for siRNA therapy and RIBOTACs,” J. Gene Med., vol. 26, no. 9, p. e3733, 2024. [CrossRef]
- P. A. McCullough, C. Wynn, and B. C. Procter, “Clinical rationale for SARS-CoV-2 base spike protein detoxification in post COVID-19 and vaccine injury syndromes,” J Am Physicians Surg, vol. 28, pp. 90–3, 2023. [CrossRef]
- “Spike Protein Behavior.” Accessed: Oct. 01, 2022. [Online]. Available: https://www.science.org/content/blog-post/spike-protein-behavior.
- “Synthetic construct HCV1146 Moderna (mRNA-1273) SARS-CoV-2 vaccine sequence.” Sep. 28, 2021. Accessed: May 16, 2025. [Online]. Available: http://www.ncbi.nlm.nih.gov/nuccore/OK120841.1.
- “Wayback Machine.” Accessed: May 16, 2025. [Online]. Available: https://web.archive.org/web/20210105162941/https://mednet-communities.net/inn/db/media/docs/11889.doc.
- C. Wang and H. Liu, “Factors influencing degradation kinetics of mRNAs and half-lives of microRNAs, circRNAs, lncRNAs in blood in vitro using quantitative PCR,” Sci. Rep., vol. 12, no. 1, p. 7259, May 2022. [CrossRef]
- G. Chandramouly et al., “Polθ reverse transcribes RNA and promotes RNA-templated DNA repair,” Sci. Adv., vol. 7, no. 24, p. eabf1771, Jun. 2021. [CrossRef]
- L. G. Boros, T. P. J. Krüger, T. Letoha, J. A. Tuszynski, P. D. Dorfsman, and J. C. Lech, “To stabilize or not to stabilize RNA - that is still the question,” Science Advances. Accessed: Jan. 01, 7AD. [Online]. Available: https://advances.sciencemag.org/content/7/24/eabf1771.
- C. R. Beck, J. L. Garcia-Perez, R. M. Badge, and J. V. Moran, “LINE-1 Elements in Structural Variation and Disease,” Annu. Rev. Genomics Hum. Genet., vol. 12, no. Volume 12, 2011, pp. 187–215, Sep. 2011. [CrossRef]
- T. J. Wang, A. Kim, and K. Kim, “A rapid detection method of replication-competent plasmid DNA from COVID-19 mRNA vaccines for quality control,” J. High Sch. Sci., vol. 8, no. 4, pp. 427–439, Dec. 2024. [CrossRef]
- K. McKernan, Y. Helbert, L. T. Kane, and S. McLaughlin, “Sequencing of bivalent Moderna and Pfizer mRNA vaccines reveals nanogram to microgram quantities of expression vector dsDNA per dose,” OSF Prepr. April, vol. 10, 2023. [CrossRef]
- K. Chatterjee et al., “The archaeal COG1901/DUF358 SPOUT-methyltransferase members, together with pseudouridine synthase Pus10, catalyze the formation of 1-methylpseudouridine at position 54 of tRNA,” RNA, vol. 18, no. 3, pp. 421–433, Mar. 2012. [CrossRef]
- F. Sharifi and Y. Ye, “Identification and classification of reverse transcriptases in bacterial genomes and metagenomes,” Nucleic Acids Res., vol. 50, no. 5, p. e29, Dec. 2021. [CrossRef]
- B. K. Patterson et al., “Persistence of SARS CoV-2 S1 Protein in CD16+ Monocytes in Post-Acute Sequelae of COVID-19 (PASC) up to 15 Months Post-Infection,” Front. Immunol., vol. 12, 2022, Accessed: Oct. 05, 2022. [Online]. Available: https://www.frontiersin.org/articles/10.3389/fimmu.2021.746021.
- C. Li et al., “Intravenous Injection of Coronavirus Disease 2019 (COVID-19) mRNA Vaccine Can Induce Acute Myopericarditis in Mouse Model,” Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am., vol. 74, no. 11, pp. 1933–1950, Aug. 2021. [CrossRef]
- T. E. Mulroney et al., “N1-methylpseudouridylation of mRNA causes +1 ribosomal frameshifting,” Nature, vol. 625, no. 7993, Art. no. 7993, Jan. 2024. [CrossRef]
- N. Chaudhary, D. Weissman, and K. A. Whitehead, “mRNA vaccines for infectious diseases: principles, delivery and clinical translation,” Nat. Rev. Drug Discov., vol. 20, no. 11, pp. 817–838, Nov. 2021. [CrossRef]
- N. Pardi, M. J. Hogan, F. W. Porter, and D. Weissman, “mRNA vaccines — a new era in vaccinology,” Nat. Rev. Drug Discov., vol. 17, no. 4, Art. no. 4, Apr. 2018. [CrossRef]
- M. T. J. Halma and J. Guetzkow, “Public Health Needs the Public Trust: A Pandemic Retrospective,” BioMed, vol. 3, no. 2, Art. no. 2, Jun. 2023. [CrossRef]
- Y. Dong and D. G. Anderson, “Opportunities and Challenges in mRNA Therapeutics,” Acc. Chem. Res., vol. 55, no. 1, pp. 1–1, Jan. 2022. [CrossRef]



| Analytical Technique | Substance Found | Population | Intervention | Outcome measures | Max Duration Vaccine-Test | Reference |
|---|---|---|---|---|---|---|
| RFFIT, IgG and IgM ELISA, ICS | RABV-G-specific IgG/IgM; T cells (IFN-γ, TNF-α, IL-2, CD107a) |
Human: 101 adults (18–40 y), rabies-naïve, BMI: 18–32 kg/m² Groups: needle-syringe (n=42; 18 ID, 24 IM), needle-free (n=59; 46 ID, 13 IM) |
Vaccine: CV7201 (mRNA-RABV-G) Packaging: lyophilized, protamine-complexed mRNA Mod: protamine-stabilized Antigen: RABV-G Route: ID/IM via needle-syringe or needle-free injector Doses: 80–640 µg, 3 doses Schedule: days 0, 7, 28 or 0, 28, 56 Booster: 80 µg at 1 y (n=14) |
Neutralizing titers (RFFIT ≥0.5 IU/mL): ID (needle-free): GMT 0.83 IU/mL (95% CI: 0.51–1.34); IM: GMT 0.44 IU/mL (95% CI: 0.10–1.90); Responders: 71% ID, 46% IM Cytokines: ↑ at day 42; polyfunctionality ↑ (p<0.0001) |
1 year | [2] |
| HAI, MN, ELISPOT | Antibody responses (HAI, MN titers); RNA not directly measured |
Human: 201 adults (18–64 y) Groups: 25–400 µg IM (n=23–30), 25–50 µg ID (n=16–23), placebo (n=35) |
Vaccine: H10N8 mRNA or H7N9 mRNA Packaging: LNP Mod: not specified Antigen: H10N8 or H7N9 HA Route: IM or ID Doses: IM: 10–400 µg; ID: 25–50 µg, 2 doses Schedule: days 1 and 21 Booster: 25 or 50 µg H7N9 at 6 months (n=53) |
HAI titers: H10N8 100 µg IM: 100% ≥1:40, GMT 68.8; H7N9 25 µg IM: 96.3% ≥1:40, GMT 87.0 MN: H10N8: 87.0% ≥1:20, GMT 38.8; H7N9: 100% ≥1:20, GMT ≥1280 (post-booster, 6 months) |
1 y | [3] |
| ELISPOT, ICS, pVL PCR, ultrasensitive viral PCR | HIV-1 RNA (cell-associated RNA); no RNA measured in vivo |
Human: 70 HIV-1 adults (≥18 y), Stable cART ≥3 y, Nadir CD4⁺ ≥350 cells/µL, current CD4⁺ ≥450 cells/µL, HIV RNA <50 copies/mL Groups: iHIVARNA-01 (n=40), TriMix (n=15), placebo (n=15) |
Vaccine: iHIVARNA-01 (HTI-TriMix mRNA) Packaging: naked mRNA (no LNP) Mod: not specified Antigen: HTI Adjuvant: TriMix (CD40L, caTLR4, CD70) Route: intranodal inguinal Doses: not specified, 3 doses Schedule: weeks 0, 2, 4 |
HTI-specific T cell (ELISPOT): weeks 0, 6, 18, ↑ CD4⁺/CD8⁺ T cell (ELISPOT, ICS): weeks 0, 6, 30, ↑ CD8⁺ suppression (in vitro): weeks 6, 18, ↑ VL rebound, % with <50 copies/mL: weeks 18, 30, partial control Viral reservoir: proviral DNA, RNA: : weeks 0, 6, 18, no significant change Viral escape, host mRNA expression: weeks 0, 6, 18, no significant change |
30 weeks | [4] |
| RFFIT, ELISA | RABV-G-specific IgG |
Human: 55 adults (18–40 y), rabies-naive, BMI: 18–32 kg/m² Groups: 1 µg (n=16), 2 µg (n=16), 5 µg (n=10), Rabipur control (n=11) |
Vaccine: CV7202 (mRNA-RABV-G) Packaging: LNP Mod: unmodified mRNA Antigen: RABV-G Route: IM, needle-syringe Doses: 1–5 µg, 1–2 doses Schedule: days 1, 29 Control: Rabipur (≥2.5 IU/mL; days 1, 8, 29) |
Neutralizing titers (RFFIT ≥0.5 IU/mL, days 1–57): 1 µg: 63–100%, GMT 4.8 IU/mL; 2 µg: 83–100%, GMT 4.2 IU/mL; 5 µg (1 dose): 22%; Rabipur: 100%, GMT 13.5 IU/mL IgG (day 43): 1 µg: 34,186 U/mL; 2 µg: 20,707 U/mL; Rabipur: 33,373 U/mL |
57 days (interim); long-term follow-up to 2 y | [5] |
| HAI, MN, ELISA (IgG1/IgG2a), ELISPOT, TCID50, bDNA, Western blot | Vaccine mRNA; HA protein (Western blot) |
Mouse: BALB/c, F, 5–8 weeks (n not specified) Ferret: M, 13–15 weeks (n not specified) NHP: Cynomolgus, 2–4 y (n=1/group) Human: 31 adults (23 vaccine, 8 placebo) |
Vaccine: H10N8 or H7N9 mRNA Packaging: LNP (50:10:38.5:1.5 ionizable lipid:DSPC:cholesterol:PEG-lipid ratio), 80–100 nm, >90% encapsulation Mod: pseudouridine, 5-methylcytidine, cap-1 Antigen: HA Route: IM Doses: Mouse: not specified, single dose; Ferret: 100 µg H10N8 mRNA or 50 µg H7N9 mRNA, 2 doses; NHPs: 6mg H10N8 mRNA, single dose; Human: not specified, 2 doses Schedule: days 1 and 21 (2 doses) |
NHP tissue mRNA conc. (ng/mL): Muscle: 5,680; Lymph nodes: 2,120; Spleen: 86.9; Liver: 47.2; Plasma: 5.47; Heart: 0.799; Brain: 0.429; Lung: 1.82 HAI titers (ferret): ↑ at days 21-49 (p < 0.0001) Viral load (TCID₅₀, lung – mouse, ferret): ↓ post-vaccination |
Mouse: 11 days; Ferret: 49 days; Human: 43 days (interim); Titers stable 1 y (mouse) |
[6] |
| Flow cytometry, RT-PCR, ELISA, cytokine multiplex | HIV-1 RNA (Gag, Nef, Rev, Vpr mRNA in DCs) |
Human: 54 HIV-1 adults (37 vaccine, 17 placebo), median CD4 count 450 cells/mm³ |
Vaccine: AGS-004 (DCs loaded with HIV Gag, Nef, Rev, Vpr RNA) Packaging: autologous DCs Mod: none Antigen: autologous HIV-1 Gag, Nef, Rev, Vpr Route: ID Dosage: 1.2 × 10⁷ DCs/mL, 4 doses Schedule: every 4 weeks (q4w) |
CTL (CD28⁺/CD45RA⁻): 69% at week 8, 83% at week 18, 92% at week 26 (vs 25% placebo) VL (week 11/12 ATI): 4.39 vs 4.47 log₁₀ HIV RNA (p=0.73) HIV DNA: no difference at weeks 8, 18, 28 |
28 weeks | [7] |
| ELISPOT, CFSE proliferation, ICS | HIV-1 Gag and Nef mRNA (in DCs) |
Human: 15 HIV-1 adults (median age: 47 y), median CD4 count 556 cells/mm³ |
Vaccine: DCs loaded with HIV-1 Gag/Nef mRNA Packaging: autologous DCs Mod: Kozak sequence, codon-optimized, lysosomal targeting Antigen: Gag, Nef (Clade B, 2001) Route: ID Dosage: 5–15M DCs, 4 doses Schedule: weeks 0, 2, 6, 10 |
CD4 to Nef: 2.3-fold ↑ from baseline (95% CI: 1.04–5.04); 6.3-fold ↑ vs placebo (95% CI: 1.6–24.6), p=0.009 CD4 to Gag: 2.5-fold ↑ from baseline (95% CI: 1.17–5.21), p=0.054 CD8 to Nef: 4.8-fold ↑ vs placebo (95% CI: 0.9–24.7), p=0.062 ELISPOT: no ↑ |
48 weeks; Peak effect: week 14 |
[8] |
| Analytical Technique | Substance Found | Fluid/Tissue | Vaccine | Sensitivity | Specificity | Concentration Observed | Duration Since Last Vaccination | Max Duration Observed | Reference |
|---|---|---|---|---|---|---|---|---|---|
| RT-qPCR, IHC | mRNA | Axillary LN (lymph) | BNT162b2; mRNA-1273 (HR2) | LOD: BNT162b2: 6 ssRNA copies/reaction; mRNA-1273: 11 ssRNA copies/reaction | Vaccine-specific sequences; none detected in unvaccinated or uninjected tissues | ~1–30 copies/ng total RNA (lymph); heart values not numerically reported | Detected primarily within 30 days | 30 days | [25] |
| SIMOA, Flow cytometry, ELISA | Spike protein (free and total) | Plasma (adolescents/young adults) | BNT162b2; mRNA-1273 | SIMOA LOD not stated | Free spike detected only in myocarditis cases; not in vaccinated controls | Free spike (myocarditis): 33.9 ± 22.4 pg/mL | Up to 24 days | 24 days | [26] |
| Flow cytometry, LC-MS | S1 subunit, S2 subunit, Mutant S1 peptides | CD16⁺ monocytes (PB) | mRNA-1273, Janssen, AstraZeneca | >99% sort purity; high-resolution peptide matching | S1 only in symptomatic group (p < 0.0007); cytokines matched PASC | S1 CD16⁺ monocytes: 92% of symptomatic (11/12); not in 9/10 controls | Range: 38–245 days; mean: 105 days | 245 days | [23] |
| LC–SACI–MS | Spike protein (vaccine-specific) | Blood (dried spot) | BNT162b2; mRNA-1273 | Not reported | 100% specificity: not found in unvaccinated controls | Detected in 50% of vaccinated subjects | 69–187 days | 187 days | [12] |
| ddPCR | mRNA (intact and degraded) | Plasma | mRNA-1273 bivalent | LLOQ: 0.001 ng/mL | ddPCR primers specific to vaccine mRNA; no WT S-gene amplification | Peak: 0.529 ng/mL (731 copies/μL); Range (4h): 6.5–112 copies/μL; 37% had 0.001–0.01 ng/mL at day 14–15 | 0–15 days | 15 days | (29) |
| LC-MS | SM-102 (ionizable lipid) | Plasma | mRNA-1273 bivalent | LLOQ: 0.01 ng/mL | R² = 0.9852 correlation to SM-102 concentration | Peak median: 3.22 ng/mL; Range (4h): 0.39–8.39 ng/mL; ≥0.12 ng/mL at day 7 | 0–7 days | 7 days | [27] |
| RT-qPCR, EV isolation | mRNA | Breast milk | BNT162b2; mRNA-1273 | Detection limit: 1 pg/mL | Trace mRNA in 3/11 participants | 1.3–11.7 pg/mL | 1 to 45 hours | 45 hours | [28] |
| SPEAR | Spike protein | Plasma | BNT162b2; mRNA-1273 | LLOD: 1.81 fM; LLOQ: 8.24 fM | Detected in 36% of controls; 43% of PACVS patients | Mean: 17 pM vs 10 fM (controls) | 26 to 709 days | 709 days | [29] |
| SPEAR | S1 subunit | Plasma | BNT162b2; mRNA-1273 | LLOD: 5.64 fM | Detected in 32% of controls; 36% of PACVS patients | Mean: 60 pM vs 3 fM (controls) | 26 to 709 days | 709 days | [29] |
| qPCR (codon-optimized), TEM | mRNA | Plasma and blood | BNT162b2 | LoQ: 10 copies/reaction (~5.26% CV); Efficiency 1.92–2.14; R²: 0.991–0.999 | No amplification of SARS-CoV-2 RNA or human genome | 50–300 copies/µg input RNA (plasma) | 1–15 days | 15 days | [30] |
| RNA-seq | mRNA | Plasma | BNT162b2; mRNA-1273 | Not stated | Unknown | Detected, not quantified | Up to 28 days | 28 days | [31] |
| RT-qPCR | mRNA | LN germinal centers | BNT162b2 | Not reported | BNT162b2-specific sequence targeted | Detected, not quantified | Up to 60 days | 60 days | [32] |
| RT-qPCR | mRNA | Breast milk | BNT162b2 | Not quantified; standard curve with ≥6 replicates | BNT162b2-specific sequence targeted | Max: 2 ng/mL in few samples | Up to 6 weeks post 2nd dose | 7 days post 2nd dose | [33] |
| Analytical Technique | Substance Found | Tissue/Organ | Vaccine | Sensitivity | Concentration Observed / Signal | Duration Sampled | Max Duration Observed | Reference |
|---|---|---|---|---|---|---|---|---|
| RT-qPCR, IHC | mRNA | Axillary LN (ALN), LV, RV, mediastinal LN, liver, spleen | BNT162b2; mRNA-1273 (HR2 domain) | LOD: BNT162b2: 6 ssRNA copies/reaction; mRNA-1273: 11 ssRNA copies/reaction | ~1–30 copies/ng total RNA (ALN); heart tissue values not explicitly reported | 1–154 days | ALN: 26 days; LV: 12 days; RV: 19 days | [25] |
| IHC | Spike protein | Brain (frontal cortex, nucleus ruber, capillary endothelium, microglia, astrocytes); heart (LV, endothelial cells); vasculature (aorta, iliac, basal cerebral arteries) | BNT162b2; ChAdOx1 nCov-19 | Not reported (described as “abundant deposits” in affected tissues) | Spike protein observed in brain, heart, and vessels; N protein absent | 3 weeks post 3rd dose | 3 weeks | [44] |
| IHC | Spike protein | Lungs | mRNA-1273 | Not reported | Spike protein not detected | 18 days (autopsy) | Negative at 18 days | [45] |
| FISH | mRNA | Liver | BNT162b2 | Probes targeted spike mRNA sequence; cross-reactivity with viral mRNA possible | Qualitative detection of cytoplasmic mRNA in hepatocytes | 12 days | 12 days | [43] |
| IHC, ISH | Spike protein; mRNA | Cerebral arteries | BNT162b2; mRNA-1273 | Spike: 43.8% (7/16 cases); mRNA: detected in 3/3 tested cases | Spike in vessel intima/smooth muscle (7/16); vaccine mRNA confirmed in 3 cases via FISH | Months after vaccination | Up to 17 months | [42] |
| IHC | Spike protein | Liver parenchyma | mRNA-1273 | Not reported | Qualitative detection of spike protein in liver tissue | 5 weeks | 5 weeks | [46] |
| RNA-seq | mRNA fragments | Right deltoid and quadriceps muscles | BNT162b2 | 75 bp vaccine-specific fragment aligned to spike mRNA | Qualitative detection of mRNA fragments | 4 weeks | 4 weeks | [47] |
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 (http://creativecommons.org/licenses/by/4.0/).