COVID Vaccines

FDA’s Failure to Safeguard the Public Resulted in Irreparable Erosion of Trust

Should the FDA have eased mRNA vaccine regulations during the pandemic and what are the safety consequences of regulatory delays?
Lightspring/Shutterstock
0:00
This is part 6 in Promise or Peril: COVID-19 mRNA Vaccine Issues Series

The pandemic has ended, but the introduction of the COVID-19 vaccines, which use mRNA technology, signifies the start of a new era in modern medicine. The lagging regulatory framework that the FDA cobbled together specifically for mRNA vaccine approval has set the stage for adverse events related to genetic therapies using this new technology. In this series, we reveal emerging concerns about mRNA injections related to the lipid nanoparticles, spike protein, and vaccine contamination as public documents are released.

In Part 6, we consider the ramifications of the “lagging” regulatory framework for COVID-19 vaccines. In June 2020, the U.S. Food and Drug Administration (FDA) set up special emergency guidance for the manufacturers making the COVID-19 vaccines; this special guidance expires on Nov. 7.
The FDA approval process is intended to safeguard human health when introducing new products. As established previously (in Part 1), mRNA is gene therapy yet the FDA has excluded vaccines from the normal testing requirements imposed on gene therapy products. The regulatory framework isn’t just “lagging,” the FDA guidance appears to have been derelict in its duty to properly test these new products.

FDA Negligence or Intentional Bypass?

Although ribonucleic acid (RNA) therapeutics have been in development for a decade, when messenger RNA (mRNA) technology was proposed for COVID-19 vaccines, a new set of regulatory rules were put in place to expedite production. The new rules allowed manufacturers to submit test results on similar products in lieu of submitting tests on the precise vaccine components.

Did this relaxation of the rules have unforeseen consequences? Likely, yes.

Gene-based therapies offer the promise of personalized therapy to cure difficult conditions such as cancer, hereditary conditions, and autoimmune and infectious diseases. Theoretically, one can replace, inactivate, or modify the disease-causing gene by delivering DNA or RNA fragments directly to affected cells. It’s the platform upon which the mRNA vaccines were built.
However, several concerning findings about mRNA injections have emerged in the literature and in scientific circles which merit urgent consideration. These were the focus of Parts 2, 3, and 4 in our series. In this piece, we broadly discuss why mRNA gene therapy was developed and the rules that such products are supposed to follow.

Gene Therapy Technology

Delivering treatment by direct injection can be very complicated. In the case of proteins, they must meet size limitations, be folded properly, and must be stabilized so they can reach their intended target.

In contrast, delivering genetic instructions to manufacture a lifesaving protein or to turn off a gene that isn’t working properly offers great promise for parents of children with a hereditary disorder.

Once inside the cell, a broken gene is replaced or a missing protein is produced, such as insulin among patients with Type 1 diabetes. The FDA approved the first RNA-based therapy to treat peripheral nerve disease in 2018.  This is a beautiful model for delivering on-demand biologics needed by the body to function properly.

Delivering a snippet of genetic code via mRNA appears to be an attractive idea. The protein doesn’t need to be packaged perfectly because the human cells take over the production process.

Gene therapy was initially focused on DNA technology because RNA was too delicate. However, the DNA-based therapeutics did not work well in human trials. There was also the concern about integration into the human genome. Researchers then shifted to synthetic RNA-based therapeutics, which set the stage for the development of vaccines based on the new mRNA technology.

The Shift to RNA-Based Technology

The quickly disintegrating RNA appeared to offer some safety advantages over DNA, chiefly that RNA would stay in the cytoplasm and wouldn’t enter the nucleus, and therefore, would be in the body only transiently. The mRNA platform used to deliver the SARS-CoV-2 vaccine relied on these advantages. However, as we noted in Part 4, the vaccine mRNA does stay in the body after vaccination for at least 60 days.
The in vitro-transcribed mRNA had the first proof-of-concept animal study in 1990. Since then, numerous studies have been conducted to overcome challenges related to the fragility of mRNA and the immune system’s tendency to attack foreign mRNA. In other words, if the mRNA is destroyed by the body before it can create new proteins, the vaccine is useless.
Vaccine technology is shifting toward this approach because it offers the advantage of stimulating not only antibodies, as the influenza (flu) shot does, but also cellular immunity (T-cells). 
“Genetic immunization may promote superior adaptive immunity by activating both humoral and cell-mediated responses, and has manufacturing advantages over traditional vaccines,” according to a 2020 review by Bloom, et al. Both arms of the immune system are very important: antibodies block viruses and bacteria from entering cells while T-cells target and destroy infected cells.
Traditional flu shots work moderately well because they depend on antibodies to do the hard work of stopping the influenza virus from gaining entry into cells. However, without a good T-cell response, the body isn’t as well prepared to attack cells that are already infected.

FDA Avoided ‘Gene Therapy’ Testing Requirements

The regulatory framework keeping watch over gene therapies was established by the same agency charged with oversight of vaccines and other biologics.

In the United States, gene-based therapies and vaccines both are regulated by the FDA’s Center for Biologics Evaluation and Research (CBER).

Although CBER monitors both gene therapies and vaccines, the regulatory framework for DNA and RNA “therapeutics” is different than for “vaccines.” Guidance from the FDA excludes vaccines from the biodistribution studies otherwise required for gene therapy products.
A 2022 review by Vervaeke, et al. stated, “The success of the messenger RNA-based COVID-19 vaccines of Moderna and Pfizer/BioNTech marks the beginning of a new chapter in modern medicine. However, the rapid rise of mRNA therapeutics has resulted in a regulatory framework that is somewhat lagging.”

While the regulatory pathways share similar characteristics, RNA therapeutics are required to go through pharmacokinetic testing to see what happens to the product once it’s in the body. Oddly, as we discussed in Part 1, the mRNA vaccines weren’t required to go through human biodistribution testing and relied solely on animal studies. Further, these animal biodistribution studies only looked at the lipid nanoparticle (LNP) capsule, not the mRNA cargo as the studies used a luciferase-encoding mRNA surrogate to take the place of a spike protein-encoding mRNA.

With new technologies, it is important to study the actual components. Using a surrogate instead of the spike protein is a problem because it prevented the FDA from understanding the precise effects of the spike protein on the body. Additionally, the animal models did not examine the biodistribution of the spike protein or its subunit (S1) in the body, yet we were told it breaks down quickly.

The European Medicines Agency (EMA), which is the European Union’s equivalent to the FDA, refers to the World Health Organization for vaccine regulations. The FDA, on the other hand, created its own guidance in response to the pandemic.

The EMA and the FDA both require biodistribution studies before an RNA gene therapy goes through human trials. These studies can test the components individually. In the case of the COVID-19 mRNA vaccines, this would include components such as the LNP capsule, the spike protein, and the combined final product.

However, both the EMA and the FDA don’t consider the mRNA vaccines to be “gene therapy” products, and therefore, they avoided the requirement for biodistribution studies (Vervaeke, et al. Table 1). While the FDA didn’t align the regulatory oversight for mRNA vaccines with the gene therapy rules, Moderna, which is funded by the National Institutes of Health, stated in a June 2020 Securities and Exchange Commission filing, “Currently, mRNA is considered a gene therapy product by the FDA.”

If mRNA is considered gene therapy by the FDA, the mRNA vaccines should have been put through the same safety testing as all other gene therapy products. How did they avoid this testing?

The risk-benefit analysis for a therapeutic used to treat someone with a serious terminal illness should be very different from that of a therapeutic targeting a large healthy population. The FDA should not have used the pandemic as a reason to abdicate its responsibility for stringent safety oversight and regulation of mRNA vaccines.

The FDA’s Failure to Protect the Public

Human studies could have been conducted, even in the context of the pandemic. For example, the rat models used to test biodistribution on the mRNA products began in July 2020 and were completed two months later. Human studies are typically completed very early in the drug development process and are required for an FDA investigational new drug application. Only six volunteers are needed for such a study, and it could have been completed quickly. In this research, for example, six volunteers participated in a biodistribution study that was completed in 24 hours.

Given the FDA’s authority in shaping health policies, despite not engaging directly in patient treatment as doctors do, every policy or action they undertake carries substantial implications for the collective well-being of individuals nationwide or even globally.

The FDA is the gatekeeper of human safety. Its ultimate responsibility is safeguarding people’s health and wellness while remaining independent of pharmaceutical company influence. Yet the FDA still does not hold mRNA vaccines to the same standards as gene therapy products. The “Nonclinical Biodistribution Considerations for Gene Therapy Products Guidance for Industry,“ dated May 2023 states, ”This guidance does not apply to prophylactic vaccines.” Why?
The “revolving door” phenomenon—when people leave the FDA to work for a large pharmaceutical company—should be strictly prohibited, as this will jeopardize the mission of the FDA. This research letter by Prasad and co-authors found that 57 percent of drug reviewers who left the FDA went to work for or provided consulting to the pharmaceutical industry.

The transition from being a regulator to advising companies, or the other way around, would unavoidably raise conflict of interest concerns, creating doubts about whether someone could act in the public interest. Especially when evaluating novel biotechnology, they might be easily biased by the economic return rather than adhering to the ethical principle “first do no harm.”

Not every novel technology is safe and without risks. A technology with the potential to affect the human genome should be handled with a higher degree of caution due to the serious nature of the associated risks.

The FDA should strictly follow normal pharmacokinetic, biodistribution, and safety data that is required for a new drug registration.

The FDA should also follow the ethical principles that guide the regulation of a new drug intended for use in an otherwise healthy population. As a broad example, placebo controls should be biologically inert (i.e., saline) so that adverse events can be identified and their frequencies estimated more accurately. As seen with the HPV vaccine studies, a “pseudo” placebo was used, which disguised the vaccine’s safety issues.

The Erosion of Public Trust

The promise of mRNA vaccines to save lives was based on several factors. One was the improved safety profile of using RNA instead of DNA. Another was the novelty of quickly encoding an antigen (the spike protein) as an immune target. Finally, the mRNA and the lipid nanoparticle delivery capsule proved to be highly immunogenic, inducing a vigorous immune response even in older adults, making them an effective adjuvant.

However, by intentionally creating two different pathways for RNA therapeutics and vaccines to expedite distribution, regulatory oversight was seriously undermined, resulting in the erosion of public trust in our regulatory agencies. Many of the adverse events now being detected would likely have been anticipated had the same human biodistribution studies and adequate safety and toxicological studies been performed as required for drugs.

Given the exigent nature of the pandemic, the urgent production of a lifesaving vaccine for those at high risk may have been considered reasonable had public health officials not simultaneously, and emphatically, urged a one-size-fits-all vaccination for the entire population. Yet these studies have still not been completed, inciting a catastrophic loss of confidence in our regulatory agencies.

We call for a reinvigorated FDA that diligently fulfills its paramount responsibilities in safeguarding the health and safety of the population. This includes upholding unwavering ethical principles rooted in human well-being for perpetuity.

◊ References

Addgene. Molecular Biology Reference. https://www.addgene.org/mol-bio-reference/#introduction
Alana F Ogata, Chi-An Cheng, Michaël Desjardins, Yasmeen Senussi, Amy C Sherman, Megan Powell, Lewis Novack, Salena Von, Xiaofang Li, Lindsey R Baden, David R Walt, Circulating Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Vaccine Antigen Detected in the Plasma of mRNA-1273 Vaccine Recipients, Clinical Infectious Diseases, Volume 74, Issue 4, 15 February 2022, Pages 715–718, https://doi.org/10.1093/cid/ciab465
Aldén M, Olofsson Falla F, Yang D, Barghouth M, Luan C, Rasmussen M, De Marinis Y. Intracellular Reverse Transcription of Pfizer BioNTech COVID-19 mRNA Vaccine BNT162b2 In Vitro in Human Liver Cell Line. Curr Issues Mol Biol. 2022 Feb 25;44(3):1115-1126. doi: 10.3390/cimb44030073. PMID: 35723296; PMCID: PMC8946961. https://pubmed.ncbi.nlm.nih.gov/35723296/
Allergic Reactions Including Anaphylaxis After Receipt of the First Dose of Pfizer-BioNTech COVID-19 Vaccine — United States, December 14–23, 2020. MMWR Morb Mortal Wkly Rep 2021;70:46–51. DOI: http://dx.doi.org/10.15585/mmwr.mm7002e1
Anderson EJ, Rouphael NG, Widge AT, et al. Safety and Immunogenicity of SARS-CoV-2 mRNA-1273 Vaccine in Older Adults N Engl J Med 2020; 383:2427-2438  https://www.nejm.org/doi/full/10.1056/NEJMoa2028436
Anderson S. CBER Plans for Monitoring COVID-19 Vaccine Safety and Effectiveness. https://stacks.cdc.gov/view/cdc/97349 October 20, 2020. Accessed 3/20/23.
Angeli F, Spanevello A, Reboldi G, Visca D, Verdecchia P. SARS-CoV-2 vaccines: Lights and shadows. Eur J Intern Med. 2021 Jun;88:1-8. doi: 10.1016/j.ejim.2021.04.019. Epub 2021 Apr 30. PMID: 33966930; PMCID: PMC8084611. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8084611/#bib0043
Baker, A. T., Boyd, R. J., Sarkar, D., Teijeira-Crespo, A., Chan, C. K., Bates, E., Waraich, K., Vant, J., Wilson, E., Truong, C. D., Lipka-Lloyd, M., Fromme, P., Vermaas, J., Williams, D., Machiesky, L., Heurich, M., Nagalo, B. M., Coughlan, L., Umlauf, S., Chiu, P. L., … Borad, M. J. (2021). ChAdOx1 interacts with CAR and PF4 with implications for thrombosis with thrombocytopenia syndrome. Science Advances. 7(49), eabl8213. https://doi.org/10.1126/sciadv.abl8213
Baumeier C, Aleshcheva G, Harms D, Gross U, Hamm C, Assmus B, Westenfeld R, Kelm M, Rammos S, Wenzel P, Münzel T, Elsässer A, Gailani M, Perings C, Bourakkadi A, Flesch M, Kempf T, Bauersachs J, Escher F, Schultheiss H-P. Intramyocardial Inflammation after COVID-19 Vaccination: An Endomyocardial Biopsy-Proven Case Series. International Journal of Molecular Sciences. 2022; 23(13):6940. https://doi.org/10.3390/ijms23136940
Bloom, K., van den Berg, F. & Arbuthnot, P. Self-amplifying RNA vaccines for infectious diseases. Gene Ther 28, 117–129 (2021). https://doi.org/10.1038/s41434-020-00204-y.
Chauhan, H., Mohapatra, S., Munt, D.J. et al. Physical-Chemical Characterization and Formulation Considerations for Solid Lipid Nanoparticles. AAPS PharmSciTech 17, 640–651 (2016). https://doi.org/10.1208/s12249-015-0394-x
Chui CSL, Fan M, Wan EYF, et al. Thromboembolic events and hemorrhagic stroke after mRNA (BNT162b2) and inactivated (CoronaVac) COVID-19 vaccination: A self-controlled case series study. Lancet. 2022;(50). https://www.thelancet.com/journals/eclinm/article/PIIS2589-5370(22)00234-6/fulltext
Dag Berild J, Bergstad Larsen V, Myrup Thiesson E, et al. Analysis of Thromboembolic and Thrombocytopenic Events After the AZD1222, BNT162b2, and MRNA-1273 COVID-19 Vaccines in 3 Nordic Countries. JAMA Netw Open. 2022;5(6):e2217375. doi:10.1001/jamanetworkopen.2022.17375 https://jamanetwork.com/journals/jamanetworkopen/fullarticle/2793348
daSilva RL. Viral-associated thrombotic microangiopathies. Hematology/Oncology and Stem Cell Therapy. 2011:4(2):51-59. https://www.sciencedirect.com/science/article/pii/S165838761150038X
De A, Ko YT. Why mRNA-ionizable LNPs formulations are so short-lived: causes and way-out. Expert Opin Drug Deliv. 2023 Feb;20(2):175-187. doi: 10.1080/17425247.2023.2162876. Epub 2023 Jan 1. PMID: 36588456. https://pubmed.ncbi.nlm.nih.gov/36588456/
Ehaideb, S.N., Abdullah, M.L., Abuyassin, B. et al. Evidence of a wide gap between COVID-19 in humans and animal models: a systematic review. Crit Care 24, 594 (2020). https://doi.org/10.1186/s13054-020-03304-8
Faizullin D, Valiullina Y, Salnikov V, Zuev Y. Direct interaction of fibrinogen with lipid microparticles modulates clotting kinetics and clot structure. Nanomedicine. 2020 Jan;23:102098. doi: 10.1016/j.nano.2019.102098. Epub 2019 Oct 23. PMID: 31655206. https://pubmed.ncbi.nlm.nih.gov/31655206/
FDA. Considerations for Human Radiolabeled Mass Balance Studies - Guidance for Industry. https://www.fda.gov/media/158178/download May, 2022.
FDA. Development and Licensure of Vaccines to Prevent COVID-19. https://www.fda.gov/media/139638/download
FDA. S12 Nonclinical Biodistribution Considerations for Gene Therapy Products Guidance for Industry. https://www.fda.gov/media/167605/download
FDA-CBER-2021-5683-0013962 approved on: 09-Nov-2020. A Tissue Distribution Study of a [3H]-Labeled Lipid Nanoparticle-mRNA Formulation Containing ALC-0315 and ALC-0159 Following Intramuscular Administration in Wistar Han Rats. FINAL REPORT Test Facility Study No. 185350 Sponsor Reference No. ALC-NC-0552 https://phmpt.org/wp-content/uploads/2022/03/125742_S1_M4_4223_185350.pdf
Fertig TE, Chitoiu L, Marta DS, Ionescu VS, Cismasiu VB, Radu E, Angheluta G, Dobre M, Serbanescu A, Hinescu ME, Gherghiceanu M. Vaccine mRNA Can Be Detected in Blood at 15 Days Post-Vaccination. Biomedicines. 2022 Jun 28;10(7):1538. doi: 10.3390/biomedicines10071538. PMID: 35884842; PMCID: PMC9313234. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9313234/
Grobbelaar LM et al. SARS-CoV-2 spike protein S1 induces fibrin(ogen) resistant to fibrinolysis: implications for microclot formation in COVID-19   Biosci Rep (2021) 41 (8): BSR20210611. https://doi.org/10.1042/BSR20210611
Hou, X., Zaks, T., Langer, R. et al. Lipid nanoparticles for mRNA delivery. Nat Rev Mater 6, 1078–1094 (2021). https://doi.org/10.1038/s41578-021-00358-0
Iaboni A, Jindal N, Betschel SD, Song C. Second-dose mRNA COVID-19 vaccine safety in patients with immediate reactions after the first dose: A case series. Journal of Allergy and Clinical Immunology: Global. 2022;1(3): 172-174. https://www.sciencedirect.com/science/article/pii/S2772829322000200
Let’s talk about lipid nanoparticles. Nat Rev Mater 6, 99 (2021). https://www.nature.com/articles/s41578-021-00281-4
Li, JX., Wang, YH., Bair, H. et al. Risk assessment of retinal vascular occlusion after COVID-19 vaccination. npj Vaccines 8, 64 (2023). https://doi.org/10.1038/s41541-023-00661-7
Michieletto, D., Lusic, M., Marenduzzo, D. et al. Physical principles of retroviral integration in the human genome. Nat Commun 10, 575 (2019). https://doi.org/10.1038/s41467-019-08333-8
Moghimi, S. M., & Simberg, D. (2022). Pro-inflammatory concerns with lipid nanoparticles. Molecular therapy: The Journal of the American Society of Gene Therapy, 30(6), 2109–2110. https://doi.org/10.1016/j.ymthe.2022.04.011
Nyström S, Hammarström P. Amyloidogenesis of SARS-CoV-2 Spike Protein. J. Am. Chem. Soc. 2022, 144, 20, 8945–8950. https://pubs.acs.org/doi/10.1021/jacs.2c03925
Ogata AF, Maley AM, Wu C, et al. Ultra-Sensitive Serial Profiling of SARS-CoV-2 Antigens and Antibodies in Plasma to Understand Disease Progression in COVID-19 Patients with Severe Disease, Clinical Chemistry, Volume 66, Issue 12, December 2020, Pages 1562–1572, https://doi.org/10.1093/clinchem/hvaa213
Ohlson J. Plasmid manufacture is the bottleneck of the genetic medicine revolution. Drug Discov Today. 2020 Oct 16;25(11):1891–3. doi: 10.1016/j.drudis.2020.09.040. Epub ahead of print. PMID: 33075470; PMCID: PMC7564888. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7564888/
Perico L, Marina Morigi M, Galbusera M, et al.  SARS-CoV-2 Spike Protein 1 Activates Microvascular Endothelial Cells and Complement System Leading to Platelet Aggregation. Front. Immunol. 2022 https://www.frontiersin.org/articles/10.3389/fimmu.2022.827146/full
Qin, S., Tang, X., Chen, Y. et al. mRNA-based therapeutics: powerful and versatile tools to combat diseases. Sig Transduct Target Ther 7, 166 (2022). https://doi.org/10.1038/s41392-022-01007-w
Rafati A, Pasebani Y, Jameie M, et al. Association of SARS-CoV-2 Vaccination or Infection With Bell Palsy: A Systematic Review and Meta-analysis. JAMA Otolaryngol Head Neck Surg. Published online April 27, 2023. doi:10.1001/jamaoto.2023.0160 https://jamanetwork.com/journals/jamaotolaryngology/fullarticle/2804297
Rhea EM, Logsdon AF, Hansen KM, Williams LM, Reed MJ, Baumann KK, Holden SJ, Raber J, Banks WA, Erickson MA. The S1 protein of SARS-CoV-2 crosses the blood-brain barrier in mice. Nat Neurosci. 2021 Mar;24(3):368-378. doi: 10.1038/s41593-020-00771-8. Epub 2020 Dec 16. PMID: 33328624; PMCID: PMC8793077. https://pubmed.ncbi.nlm.nih.gov/33328624/
Röltgen K, Nielsen SCA, Silva O. Immune imprinting, breadth of variant recognition, and germinal center response in human SARS-CoV-2 infection and vaccination. Cell 2022;185(6):1025-1040. https://www.cell.com/cell/fulltext/S0092-8674(22)00076-9
Rosenblum HG,  Gee J, Liu R, et al. Safety of mRNA vaccines administered during the initial 6 months of the US COVID-19 vaccination programme: an observational study of reports to the Vaccine Adverse Event Reporting System and v-safe. Lancet Infectious Diseases. 2022;22(6):802-812. https://doi.org/10.1016/S1473-3099(22)00054-8

Schmeling, M, Manniche, V, Hansen, PR. Batch-dependent safety of the BNT162b2 mRNA COVID-19 vaccine. Eur J Clin Invest. 2023; 00:e13998. doi:10.1111/eci.13998

Srinivasan M, Thangaraj SR, Arzoun H. Gene Therapy - Can it Cure Type 1 Diabetes? Cureus. 2021 Dec 19;13(12):e20516. doi: 10.7759/cureus.20516. PMID: 35004071; PMCID: PMC8723777. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8723777/
Trougakos IP, Terpos E, Alexopoulos H, et al. Adverse effects of COVID-19 mRNA vaccines: the spike hypothesis. Cell 2022;28(7): P542-554. https://www.cell.com/trends/molecular-medicine/fulltext/S1471-4914(22)00103-4
Vervaeke P, Borgos SE, Sanders NN, Combes F. Regulatory guidelines and preclinical tools to study the biodistribution of RNA therapeutics. Adv Drug Deliv Rev. 2022 May;184:114236. doi: 10.1016/j.addr.2022.114236. Epub 2022 Mar 26. PMID: 35351470; PMCID: PMC8957368. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8957368/

Wong DWL, Klinkhammer BM, Djudjaj S, Villwock S, Timm MC, Buhl EM, Wucherpfennig S, Cacchi C, Braunschweig T, Knüchel-Clarke R, Jonigk D, Werlein C, Bülow RD, Dahl E, von Stillfried S, Boor P. Multisystemic Cellular Tropism of SARS-CoV-2 in Autopsies of COVID-19 Patients. Cells. 2021 Jul 27;10(8):1900. doi: 10.3390/cells10081900. PMID: 34440669; PMCID: PMC8394956.

Yonker LM, Swank Z, Bartsch YC, et al. Circulating Spike Protein Detected in Post–COVID–19 mRNA Vaccine Myocarditis. Circulation. 2023:147(11).  https://www.ahajournals.org/doi/10.1161/CIRCULATIONAHA.122.061025
Zheng Y, Zhao J, Li J, Guo Z, Sheng J, Ye X, Jin G, Wang C, Chai W, Yan J, Liu D, Liang X. SARS-CoV-2 spike protein causes blood coagulation and thrombosis by competitive binding to heparan sulfate. Int J Biol Macromol. 2021 Dec 15;193(Pt B):1124-1129. doi: 10.1016/j.ijbiomac.2021.10.112. Epub 2021 Oct 29. PMID: 34743814; PMCID: PMC8553634. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8553634/
Views expressed in this article are the opinions of the author and do not necessarily reflect the views of The Epoch Times. Epoch Health welcomes professional discussion and friendly debate. To submit an opinion piece, please follow these guidelines and submit through our form here.