1
|
Polavarapu N, Doty M, Dobrovolny HM. Exploring the treatment of SARS-CoV-2 with modified vesicular stomatitis virus. J Theor Biol 2024; 595:111959. [PMID: 39366462 DOI: 10.1016/j.jtbi.2024.111959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/13/2024] [Accepted: 09/28/2024] [Indexed: 10/06/2024]
Abstract
SARS-CoV-2 caused a global pandemic and is now an endemic virus that will require continued antiviral and vaccine development. A possible new treatment modality was recently suggested that would use vesicular stomatitis virus (VSV) modified to express the ACE2 receptor. Since the modified VSV expresses the cell surface receptor that is used by the SARS-CoV-2 spike protein, the thought is that SARS-CoV-2 virions would bind to the modified VSV and thus be neutralized. Additionally, since SARS-CoV-2 infected cells also express the spike protein, the modified VSV could potentially infect these cells, allowing for its own replication, but also potentially interfering with replication of SARS-CoV-2. This idea has not yet been tested experimentally, but we can investigate the feasibility of this possible treatment theoretically. In this manuscript, we develop a mathematical model of this suggested treatment and explore conditions under which it might be effective. We find that treatment with modified VSV does little to change the SARS-CoV-2 time course except when the treatment is applied at the onset of the SARS-CoV-2 infection at very high doses. In this case, VSV reduces the peak SARS-CoV-2 viral load, but lengthens the duration of the SARS-CoV-2 infection. Thus, we find that modified VSV treatment is unlikely to be effective largely because it does not prevent infection of cells by SARS-CoV-2.
Collapse
Affiliation(s)
- Nishnath Polavarapu
- Department of Physics & Astronomy, Texas Christian University, Fort Worth, TX, United States
| | - Madison Doty
- Burnett School of Medicine at TCU, Fort Worth, TX, USA
| | - Hana M Dobrovolny
- Department of Physics & Astronomy, Texas Christian University, Fort Worth, TX, United States.
| |
Collapse
|
2
|
Dogra P, Shinglot V, Ruiz-Ramírez J, Cave J, Butner JD, Schiavone C, Duda DG, Kaseb AO, Chung C, Koay EJ, Cristini V, Ozpolat B, Calin GA, Wang Z. Translational modeling-based evidence for enhanced efficacy of standard-of-care drugs in combination with anti-microRNA-155 in non-small-cell lung cancer. Mol Cancer 2024; 23:156. [PMID: 39095771 PMCID: PMC11295620 DOI: 10.1186/s12943-024-02060-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Elevated microRNA-155 (miR-155) expression in non-small-cell lung cancer (NSCLC) promotes cisplatin resistance and negatively impacts treatment outcomes. However, miR-155 can also boost anti-tumor immunity by suppressing PD-L1 expression. Therapeutic targeting of miR-155 through its antagonist, anti-miR-155, has proven challenging due to its dual molecular effects. METHODS We developed a multiscale mechanistic model, calibrated with in vivo data and then extrapolated to humans, to investigate the therapeutic effects of nanoparticle-delivered anti-miR-155 in NSCLC, alone or in combination with standard-of-care drugs. RESULTS Model simulations and analyses of the clinical scenario revealed that monotherapy with anti-miR-155 at a dose of 2.5 mg/kg administered once every three weeks has substantial anti-cancer activity. It led to a median progression-free survival (PFS) of 6.7 months, which compared favorably to cisplatin and immune checkpoint inhibitors. Further, we explored the combinations of anti-miR-155 with standard-of-care drugs, and found strongly synergistic two- and three-drug combinations. A three-drug combination of anti-miR-155, cisplatin, and pembrolizumab resulted in a median PFS of 13.1 months, while a two-drug combination of anti-miR-155 and cisplatin resulted in a median PFS of 11.3 months, which emerged as a more practical option due to its simple design and cost-effectiveness. Our analyses also provided valuable insights into unfavorable dose ratios for drug combinations, highlighting the need for optimizing dose regimens to prevent antagonistic effects. CONCLUSIONS This work bridges the gap between preclinical development and clinical translation of anti-miR-155 and unravels the potential of anti-miR-155 combination therapies in NSCLC.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA.
| | - Vrushaly Shinglot
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | | | - Joseph Cave
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Joseph D Butner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carmine Schiavone
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
| | - Dan G Duda
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ahmed O Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
- Department of Imaging Physics, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Bulent Ozpolat
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA.
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
3
|
Ling-Hu T, Simons LM, Dean TJ, Rios-Guzman E, Caputo MT, Alisoltani A, Qi C, Malczynski M, Blanke T, Jennings LJ, Ison MG, Achenbach CJ, Larkin PM, Kaul KL, Lorenzo-Redondo R, Ozer EA, Hultquist JF. Integration of individualized and population-level molecular epidemiology data to model COVID-19 outcomes. Cell Rep Med 2024; 5:101361. [PMID: 38232695 PMCID: PMC10829796 DOI: 10.1016/j.xcrm.2023.101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 08/07/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with enhanced transmissibility and immune escape have emerged periodically throughout the coronavirus disease 2019 (COVID-19) pandemic, but the impact of these variants on disease severity has remained unclear. In this single-center, retrospective cohort study, we examined the association between SARS-CoV-2 clade and patient outcome over a two-year period in Chicago, Illinois. Between March 2020 and March 2022, 14,252 residual diagnostic specimens were collected from SARS-CoV-2-positive inpatients and outpatients alongside linked clinical and demographic metadata, of which 2,114 were processed for viral whole-genome sequencing. When controlling for patient demographics and vaccination status, several viral clades were associated with risk for hospitalization, but this association was negated by the inclusion of population-level confounders, including case count, sampling bias, and shifting standards of care. These data highlight the importance of integrating non-virological factors into disease severity and outcome models for the accurate assessment of patient risk.
Collapse
Affiliation(s)
- Ted Ling-Hu
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Lacy M Simons
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Taylor J Dean
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Estefany Rios-Guzman
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Matthew T Caputo
- Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Arghavan Alisoltani
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Chao Qi
- Clinical Microbiology Laboratory, Department of Pathology, Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Michael Malczynski
- Clinical Microbiology Laboratory, Department of Pathology, Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Timothy Blanke
- Diagnostic Molecular Biology Laboratory, Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Lawrence J Jennings
- Clinical Microbiology Laboratory, Department of Pathology, Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Michael G Ison
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chad J Achenbach
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Paige M Larkin
- Department of Molecular Microbiology, Northshore University HealthSystem, Evanston, IL 60201, USA
| | - Karen L Kaul
- Department of Pathology, Northshore University HealthSystem, Evanston, IL 60201, USA
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Egon A Ozer
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Judd F Hultquist
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA.
| |
Collapse
|
4
|
Godwin PO, Polsonetti B, Caron MF, Oppelt TF. Remdesivir for the Treatment of COVID-19: A Narrative Review. Infect Dis Ther 2024; 13:1-19. [PMID: 38193988 PMCID: PMC10828241 DOI: 10.1007/s40121-023-00900-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
Despite the wide availability of effective vaccines, COVID-19 continues to be an infectious disease of global importance. Remdesivir is a broad-spectrum antiviral and was the first US Food and Drug Administration-approved treatment for COVID-19. In clinical guidelines, remdesivir is currently the only recommended antiviral for use in hospitalized patients with COVID-19, with or without a supplemental oxygen requirement. It is also recommended for nonhospitalized patients with COVID-19 and hospitalized patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection who are at high risk of progression to severe disease. This narrative review explores the evidence for remdesivir across various clinical outcomes and evolution of clinical guidelines through a survey over time of randomized controlled trials, observational studies, and meta-analyses. Remdesivir, compared to standard of care, appears to improve survival and disease progression in a variety of patient populations with COVID-19 across a spectrum of disease severity and SARS-CoV-2 variant periods. Remdesivir also appears to improve time to clinical recovery, increase rate of recovery, and reduce time on supplemental oxygen and readmission rates. More recent large, real-world studies further support the early use of remdesivir in a range of patient populations, including those with immunocompromising conditions.
Collapse
Affiliation(s)
- Patrick O Godwin
- Department of Medicine, Division of Academic Internal Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | | | | | | |
Collapse
|
5
|
Staquicini DI, Cardó-Vila M, Rotolo JA, Staquicini FI, Tang FHF, Smith TL, Ganju A, Schiavone C, Dogra P, Wang Z, Cristini V, Giordano RJ, Ozawa MG, Driessen WHP, Proneth B, Souza GR, Brinker LM, Noureddine A, Snider AJ, Canals D, Gelovani JG, Petrache I, Tuder RM, Obeid LM, Hannun YA, Kolesnick RN, Brinker CJ, Pasqualini R, Arap W. Ceramide as an endothelial cell surface receptor and a lung-specific lipid vascular target for circulating ligands. Proc Natl Acad Sci U S A 2023; 120:e2220269120. [PMID: 37579172 PMCID: PMC10450669 DOI: 10.1073/pnas.2220269120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/21/2023] [Indexed: 08/16/2023] Open
Abstract
The vascular endothelium from individual organs is functionally specialized, and it displays a unique set of accessible molecular targets. These serve as endothelial cell receptors to affinity ligands. To date, all identified vascular receptors have been proteins. Here, we show that an endothelial lung-homing peptide (CGSPGWVRC) interacts with C16-ceramide, a bioactive sphingolipid that mediates several biological functions. Upon binding to cell surfaces, CGSPGWVRC triggers ceramide-rich platform formation, activates acid sphingomyelinase and ceramide production, without the associated downstream apoptotic signaling. We also show that the lung selectivity of CGSPGWVRC homing peptide is dependent on ceramide production in vivo. Finally, we demonstrate two potential applications for this lipid vascular targeting system: i) as a bioinorganic hydrogel for pulmonary imaging and ii) as a ligand-directed lung immunization tool against COVID-19. Thus, C16-ceramide is a unique example of a lipid-based receptor system in the lung vascular endothelium targeted in vivo by circulating ligands such as CGSPGWVRC.
Collapse
Affiliation(s)
- Daniela I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ07103
| | - Marina Cardó-Vila
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ85724
- Department of Otolaryngology-Head and Neck Surgery, University of Arizona, Tucson, AZ85724
| | - Jimmy A. Rotolo
- Department of Molecular Pharmacology, Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, NY10021
| | - Fernanda I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ07103
| | - Fenny H. F. Tang
- Rutgers Cancer Institute of New Jersey, Newark, NJ07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ07103
| | - Tracey L. Smith
- Rutgers Cancer Institute of New Jersey, Newark, NJ07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ07103
| | - Aditya Ganju
- Department of Molecular Pharmacology, Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, NY10021
| | - Carmine Schiavone
- Department of Medicine, Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX77030
| | - Prashant Dogra
- Department of Medicine, Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX77030
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY10065
| | - Zhihui Wang
- Department of Medicine, Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX77030
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY10065
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX77030
| | - Vittorio Cristini
- Department of Medicine, Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX77030
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX77030
- Department of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, Houston, TX77030
- Physiology, Biophysics and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY10065
| | - Ricardo J. Giordano
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP05508, Brazil
| | - Michael G. Ozawa
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Wouter H. P. Driessen
- David H. Koch Center and Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX77030
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Zentrum Muenchen, Muenchen, Neuherberg85764, Germany
| | - Glauco R. Souza
- David H. Koch Center and Department of Genitourinary Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX77030
| | - Lina M. Brinker
- Department of Chemical and Biological Engineering, Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, NM87131
| | - Achraf Noureddine
- Department of Chemical and Biological Engineering, Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, NM87131
| | - Ashley J. Snider
- Stony Brook Cancer Center, Stony Brook University Hospital and Department of Medicine, Renaissance School of Medicine, Stony Brook University, Brook for Brookhaven, Suffolk County, NY11794
| | - Daniel Canals
- Stony Brook Cancer Center, Stony Brook University Hospital and Department of Medicine, Renaissance School of Medicine, Stony Brook University, Brook for Brookhaven, Suffolk County, NY11794
| | - Juri G. Gelovani
- Office of the Provost, United Arab Emirates University, Al Ain, Abu Dhabi15551, UAE
| | - Irina Petrache
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO80206
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Rubin M. Tuder
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Lina M. Obeid
- Stony Brook Cancer Center, Stony Brook University Hospital and Department of Medicine, Renaissance School of Medicine, Stony Brook University, Brook for Brookhaven, Suffolk County, NY11794
| | - Yusuf A. Hannun
- Stony Brook Cancer Center, Stony Brook University Hospital and Department of Medicine, Renaissance School of Medicine, Stony Brook University, Brook for Brookhaven, Suffolk County, NY11794
- Stony Brook Cancer Center, Stony Brook University Hospital and Departments of Biochemistry and Pathology, Renaissance School of Medicine, Stony Brook University, Brookhaven, NY11794
| | - Richard N. Kolesnick
- Department of Molecular Pharmacology, Laboratory of Signal Transduction, Memorial Sloan-Kettering Cancer Center, New York, NY10021
| | - C. Jeffrey Brinker
- Department of Chemical and Biological Engineering, Center for Micro-Engineered Materials, University of New Mexico, Albuquerque, NM87131
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ07101
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ07103
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ07101
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ07103
| |
Collapse
|
6
|
Dogra P, Schiavone C, Wang Z, Ruiz-Ramírez J, Caserta S, Staquicini DI, Markosian C, Wang J, Sostman HD, Pasqualini R, Arap W, Cristini V. A modeling-based approach to optimize COVID-19 vaccine dosing schedules for improved protection. JCI Insight 2023; 8:e169860. [PMID: 37227783 PMCID: PMC10371350 DOI: 10.1172/jci.insight.169860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/23/2023] [Indexed: 05/27/2023] Open
Abstract
While the development of different vaccines slowed the dissemination of SARS-CoV-2, the occurrence of breakthrough infections has continued to fuel the COVID-19 pandemic. To secure at least partial protection in the majority of the population through 1 dose of a COVID-19 vaccine, delayed administration of boosters has been implemented in many countries. However, waning immunity and emergence of new variants of SARS-CoV-2 suggest that such measures may induce breakthrough infections due to intermittent lapses in protection. Optimizing vaccine dosing schedules to ensure prolonged continuity in protection could thus help control the pandemic. We developed a mechanistic model of immune response to vaccines as an in silico tool for dosing schedule optimization. The model was calibrated with clinical data sets of acquired immunity to COVID-19 mRNA vaccines in healthy and immunocompromised participants and showed robust validation by accurately predicting neutralizing antibody kinetics in response to multiple doses of COVID-19 mRNA vaccines. Importantly, by estimating population vulnerability to breakthrough infections, we predicted tailored vaccination dosing schedules to minimize breakthrough infections, especially for immunocompromised individuals. We identified that the optimal vaccination schedules vary from CDC-recommended dosing, suggesting that the model is a valuable tool to optimize vaccine efficacy outcomes during future outbreaks.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
| | - Carmine Schiavone
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
| | - Zhihui Wang
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
| | - Javier Ruiz-Ramírez
- Centro de Ciencias de la Salud, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Sergio Caserta
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Daniela I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Christopher Markosian
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jin Wang
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Surgery, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - H. Dirk Sostman
- Weill Cornell Medicine, New York, New York, USA
- Houston Methodist Research Institute, Houston, Texas, USA
- Houston Methodist Academic Institute, Houston, Texas, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, USA
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, Texas, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
7
|
Sarkar S, Mishra A, Periasamy S, Dyett B, Dogra P, Ball AS, Yeo LY, White JF, Wang Z, Cristini V, Jagannath C, Khan A, Soni SK, Drummond CJ, Conn CE. Prospective Subunit Nanovaccine against Mycobacterium tuberculosis Infection─Cubosome Lipid Nanocarriers of Cord Factor, Trehalose 6,6' Dimycolate. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37262346 DOI: 10.1021/acsami.3c04063] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
An improved vaccine is urgently needed to replace the now more than 100-year-old Bacillus Calmette-Guérin (BCG) vaccine against tuberculosis (TB) disease, which represents a significant burden on global public health. Mycolic acid, or cord factor trehalose 6,6' dimycolate (TDM), a lipid component abundant in the cell wall of the pathogen Mycobacterium tuberculosis (MTB), has been shown to have strong immunostimulatory activity but remains underexplored due to its high toxicity and poor solubility. Herein, we employed a novel strategy to encapsulate TDM within a cubosome lipid nanocarrier as a potential subunit nanovaccine candidate against TB. This strategy not only increased the solubility and reduced the toxicity of TDM but also elicited a protective immune response to control MTB growth in macrophages. Both pre-treatment and concurrent treatment of the TDM encapsulated in lipid monoolein (MO) cubosomes (MO-TDM) (1 mol %) induced a strong proinflammatory cytokine response in MTB-infected macrophages, due to epigenetic changes at the promoters of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) in comparison to the untreated control. Furthermore, treatment with MO-TDM (1 mol %) cubosomes significantly improved antigen processing and presentation capabilities of MTB-infected macrophages to CD4 T cells. The ability of MO-TDM (1 mol %) cubosomes to induce a robust innate and adaptive response in vitro was further supported by a mathematical modeling study predicting the vaccine efficacy in vivo. Overall, these results indicate a strong immunostimulatory effect of TDM when delivered through the lipid nanocarrier, suggesting its potential as a novel TB vaccine.
Collapse
Affiliation(s)
- Sampa Sarkar
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Selvakannan Periasamy
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Brendan Dyett
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York 10021, United States
| | - Andrew S Ball
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Leslie Y Yeo
- School of Engineering, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Jacinta F White
- The Commonwealth Scientific and Industrial Research Organisation, Clayton 3169, Victoria, Australia
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York 10021, United States
- Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Imaging Physics, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, United States
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, New York 10021, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Sarvesh K Soni
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Calum J Drummond
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Charlotte E Conn
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| |
Collapse
|
8
|
Xu J, Carruthers J, Finnie T, Hall I. Simplified within-host and Dose-response Models of SARS-CoV-2. J Theor Biol 2023; 565:111447. [PMID: 36898624 PMCID: PMC9993737 DOI: 10.1016/j.jtbi.2023.111447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/12/2023]
Abstract
Understanding the mechanistic dynamics of transmission is key to designing more targeted and effective interventions to limit the spread of infectious diseases. A well-described within-host model allows explicit simulation of how infectiousness changes over time at an individual level. This can then be coupled with dose-response models to investigate the impact of timing on transmission. We collected and compared a range of within-host models used in previous studies and identified a minimally-complex model that provides suitable within-host dynamics while keeping a reduced number of parameters to allow inference and limit unidentifiability issues. Furthermore, non-dimensionalised models were developed to further overcome the uncertainty in estimates of the size of the susceptible cell population, a common problem in many of these approaches. We will discuss these models, and their fit to data from the human challenge study (see Killingley et al. (2022)) for SARS-CoV-2 and the model selection results, which has been performed using ABC-SMC. The parameter posteriors have then used to simulate viral-load based infectiousness profiles via a range of dose-response models, which illustrate the large variability of the periods of infection window observed for COVID-19.
Collapse
Affiliation(s)
- Jingsi Xu
- Department of Mathematics, University of Manchester, United Kingdom.
| | | | - Thomas Finnie
- PHAGE Joint Modelling Team, UK Health Security Agency, United Kingdom
| | - Ian Hall
- Department of Mathematics, University of Manchester, United Kingdom; PHAGE Joint Modelling Team, UK Health Security Agency, United Kingdom.
| |
Collapse
|
9
|
Beahm DR, Deng Y, DeAngelo TM, Sarpeshkar R. Drug Cocktail Formulation via Circuit Design. IEEE TRANSACTIONS ON MOLECULAR, BIOLOGICAL, AND MULTI-SCALE COMMUNICATIONS 2023; 9:28-48. [PMID: 37397625 PMCID: PMC10312325 DOI: 10.1109/tmbmc.2023.3246928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Electronic circuits intuitively visualize and quantitatively simulate biological systems with nonlinear differential equations that exhibit complicated dynamics. Drug cocktail therapies are a powerful tool against diseases that exhibit such dynamics. We show that just six key states, which are represented in a feedback circuit, enable drug-cocktail formulation: 1) healthy cell number; 2) infected cell number; 3) extracellular pathogen number; 4) intracellular pathogenic molecule number; 5) innate immune system strength; and 6) adaptive immune system strength. To enable drug cocktail formulation, the model represents the effects of the drugs in the circuit. For example, a nonlinear feedback circuit model fits measured clinical data, represents cytokine storm and adaptive autoimmune behavior, and accounts for age, sex, and variant effects for SARS-CoV-2 with few free parameters. The latter circuit model provided three quantitative insights on the optimal timing and dosage of drug components in a cocktail: 1) antipathogenic drugs should be given early in the infection, but immunosuppressant timing involves a tradeoff between controlling pathogen load and mitigating inflammation; 2) both within and across-class combinations of drugs have synergistic effects; 3) if they are administered sufficiently early in the infection, anti-pathogenic drugs are more effective at mitigating autoimmune behavior than immunosuppressant drugs.
Collapse
Affiliation(s)
| | - Yijie Deng
- Thayer School or Engineering, Dartmouth College, Hanover, NH 03755 USA
| | - Thomas M DeAngelo
- Thayer School or Engineering, Dartmouth College, Hanover, NH 03755 USA
| | - Rahul Sarpeshkar
- Departments of Engineering, Physics, Microbiology & Immunobiology, and Molecular & Systems Biology, Dartmouth College, Hanover, NH 03755 USA
| |
Collapse
|
10
|
Dogra P, Schiavone C, Wang Z, Ruiz-Ramírez J, Caserta S, Staquicini DI, Markosian C, Wang J, Sostman HD, Pasqualini R, Arap W, Cristini V. A modeling-based approach to optimize COVID-19 vaccine dosing schedules for improved protection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2022.09.14.22279959. [PMID: 36415468 PMCID: PMC9681049 DOI: 10.1101/2022.09.14.22279959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
While the development of different vaccines has slowed the dissemination of SARS-CoV-2, the occurrence of breakthrough infections continues to fuel the pandemic. As a strategy to secure at least partial protection, with a single dose of a given COVID-19 vaccine to maximum possible fraction of the population, delayed administration of subsequent doses (or boosters) has been implemented in many countries. However, waning immunity and emergence of new variants of SARS-CoV-2 suggest that such measures may jeopardize the attainment of herd immunity due to intermittent lapses in protection. Optimizing vaccine dosing schedules could thus make the difference between periodic occurrence of breakthrough infections or effective control of the pandemic. To this end, we have developed a mechanistic mathematical model of adaptive immune response to vaccines and demonstrated its applicability to COVID-19 mRNA vaccines as a proof-of-concept for future outbreaks. The model was thoroughly calibrated against multiple clinical datasets involving immune response to SARS-CoV-2 infection and mRNA vaccines in healthy and immunocompromised subjects (cancer patients undergoing therapy); the model showed robust clinical validation by accurately predicting neutralizing antibody kinetics, a correlate of vaccine-induced protection, in response to multiple doses of mRNA vaccines. Importantly, we estimated population vulnerability to breakthrough infections and predicted tailored vaccination dosing schedules to maximize protection and thus minimize breakthrough infections, based on the immune status of a sub-population. We have identified a critical waiting window for cancer patients (or, immunocompromised subjects) to allow recovery of the immune system (particularly CD4+ T-cells) for effective differentiation of B-cells to produce neutralizing antibodies and thus achieve optimal vaccine efficacy against variants of concern, especially between the first and second doses. Also, we have obtained optimized dosing schedules for subsequent doses in healthy and immunocompromised subjects, which vary from the CDC-recommended schedules, to minimize breakthrough infections. The developed modeling tool is based on generalized adaptive immune response to antigens and can thus be leveraged to guide vaccine dosing schedules during future outbreaks.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Carmine Schiavone
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
| | - Zhihui Wang
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Javier Ruiz-Ramírez
- Centro de Ciencias de la Salud, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Sergio Caserta
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Daniela I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Christopher Markosian
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jin Wang
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - H. Dirk Sostman
- Weill Cornell Medicine, New York, NY, USA
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Academic Institute, Houston, TX, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
- Department of Imaging Physics, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
11
|
Dedifferentiation-mediated stem cell niche maintenance in early-stage ductal carcinoma in situ progression: insights from a multiscale modeling study. Cell Death Dis 2022; 13:485. [PMID: 35597788 PMCID: PMC9124196 DOI: 10.1038/s41419-022-04939-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 12/14/2022]
Abstract
We present a multiscale agent-based model of ductal carcinoma in situ (DCIS) to study how key phenotypic and signaling pathways are involved in the early stages of disease progression. The model includes a phenotypic hierarchy, and key endocrine and paracrine signaling pathways, and simulates cancer ductal growth in a 3D lattice-free domain. In particular, by considering stochastic cell dedifferentiation plasticity, the model allows for study of how dedifferentiation to a more stem-like phenotype plays key roles in the maintenance of cancer stem cell populations and disease progression. Through extensive parameter perturbation studies, we have quantified and ranked how DCIS is sensitive to perturbations in several key mechanisms that are instrumental to early disease development. Our studies reveal that long-term maintenance of multipotent stem-like cell niches within the tumor are dependent on cell dedifferentiation plasticity, and that disease progression will become arrested due to dilution of the multipotent stem-like population in the absence of dedifferentiation. We have identified dedifferentiation rates necessary to maintain biologically relevant multipotent cell populations, and also explored quantitative relationships between dedifferentiation rates and disease progression rates, which may potentially help to optimize the efficacy of emerging anti-cancer stem cell therapeutics.
Collapse
|
12
|
Markosian C, Staquicini DI, Dogra P, Dodero-Rojas E, Lubin JH, Tang FH, Smith TL, Contessoto VG, Libutti SK, Wang Z, Cristini V, Khare SD, Whitford PC, Burley SK, Onuchic JN, Pasqualini R, Arap W. Genetic and Structural Analysis of SARS-CoV-2 Spike Protein for Universal Epitope Selection. Mol Biol Evol 2022; 39:msac091. [PMID: 35511693 PMCID: PMC9129195 DOI: 10.1093/molbev/msac091] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Evaluation of immunogenic epitopes for universal vaccine development in the face of ongoing SARS-CoV-2 evolution remains a challenge. Herein, we investigate the genetic and structural conservation of an immunogenically relevant epitope (C662-C671) of spike (S) protein across SARS-CoV-2 variants to determine its potential utility as a broad-spectrum vaccine candidate against coronavirus diseases. Comparative sequence analysis, structural assessment, and molecular dynamics simulations of C662-C671 epitope were performed. Mathematical tools were employed to determine its mutational cost. We found that the amino acid sequence of C662-C671 epitope is entirely conserved across the observed major variants of SARS-CoV-2 in addition to SARS-CoV. Its conformation and accessibility are predicted to be conserved, even in the highly mutated Omicron variant. Costly mutational rate in the context of energy expenditure in genome replication and translation can explain this strict conservation. These observations may herald an approach to developing vaccine candidates for universal protection against emergent variants of coronavirus.
Collapse
Affiliation(s)
- Christopher Markosian
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Daniela I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10022, USA
| | - Esteban Dodero-Rojas
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA
| | - Joseph H. Lubin
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Fenny H.F. Tang
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Tracey L. Smith
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | | | - Steven K. Libutti
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10022, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY 10022, USA
| | - Sagar D. Khare
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Paul C. Whitford
- Department of Physics and Center for Theoretical Biological Physics, Northeastern University, Boston, MA 02115, USA
| | - Stephen K. Burley
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- RCSB Protein Data Bank, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- RCSB Protein Data Bank, San Diego Supercomputer Center, and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92067, USA
| | - José N. Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA
- Department of Biosciences, Rice University, Houston, TX 77005, USA
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- Department of Physics and Astronomy, Rice University, Houston, TX 77005, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07101, USA
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
13
|
SARS-CoV-2 Dynamics in the Mucus Layer of the Human Upper Respiratory Tract Based on Host–Cell Dynamics. SUSTAINABILITY 2022. [DOI: 10.3390/su14073896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A thorough understanding of the inhalation dynamics of infectious aerosols indoors and infection dynamics within the host by inhaled viruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) plays an important role in the assessment and control of infection risks indoors. Here, by combining computational fluid–particle dynamics (CFPD) and host–cell dynamics (HCD), SARS-CoV-2 infection dynamics in the mucus layer of the human upper airway were studied. To reproduce the diffusive and convective transport of the virus in the nasal cavity–nasopharynx by mucociliary motion, a three-dimensional (3D)-shell model with a mucus layer was developed. The initial virus concentrations for HCD calculation were estimated based on the deposition distribution of droplets with representative sizes analyzed by CFPD. To develop a new HCD model, the target-cell-limited model was integrated with the convection–diffusion equation. Additionally, the sensitivity of the infection rate β to the infection dynamics was systematically investigated. The results showed that the time series of SARS-CoV-2 concentration in the mucus layer strongly depended on diffusion, convection, and β. Although the SARS-CoV-2 dynamics obtained here have not been verified by corresponding clinical data, they can preliminarily reveal its transmission mode in the upper airway, which will contribute to the prevention and treatment of coronavirus disease 2019.
Collapse
|
14
|
Dogra P, Ramírez JR, Butner JD, Peláez MJ, Chung C, Hooda-Nehra A, Pasqualini R, Arap W, Cristini V, Calin GA, Ozpolat B, Wang Z. Translational Modeling Identifies Synergy between Nanoparticle-Delivered miRNA-22 and Standard-of-Care Drugs in Triple-Negative Breast Cancer. Pharm Res 2022; 39:511-528. [PMID: 35294699 PMCID: PMC8986735 DOI: 10.1007/s11095-022-03176-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/21/2022] [Indexed: 12/29/2022]
Abstract
Purpose Downregulation of miRNA-22 in triple-negative breast cancer (TNBC) is associated with upregulation of eukaryotic elongation 2 factor kinase (eEF2K) protein, which regulates tumor growth, chemoresistance, and tumor immunosurveillance. Moreover, exogenous administration of miRNA-22, loaded in nanoparticles to prevent degradation and improve tumor delivery (termed miRNA-22 nanotherapy), to suppress eEF2K production has shown potential as an investigational therapeutic agent in vivo. Methods To evaluate the translational potential of miRNA-22 nanotherapy, we developed a multiscale mechanistic model, calibrated to published in vivo data and extrapolated to the human scale, to describe and quantify the pharmacokinetics and pharmacodynamics of miRNA-22 in virtual patient populations. Results Our analysis revealed the dose-response relationship, suggested optimal treatment frequency for miRNA-22 nanotherapy, and highlighted key determinants of therapy response, from which combination with immune checkpoint inhibitors was identified as a candidate strategy for improving treatment outcomes. More importantly, drug synergy was identified between miRNA-22 and standard-of-care drugs against TNBC, providing a basis for rational therapeutic combinations for improved response Conclusions The present study highlights the translational potential of miRNA-22 nanotherapy for TNBC in combination with standard-of-care drugs. Supplementary Information The online version contains supplementary material available at 10.1007/s11095-022-03176-3.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, 10065, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA
| | - Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA
| | - Maria J Peláez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Anupama Hooda-Nehra
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, 07101, USA
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, 07101, USA
- Department of Radiation Oncology, Division of Cancer Biology, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, New Jersey, 07101, USA
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA
- Department of Imaging Physics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77230, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, New York, 10065, USA
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, 77030, USA.
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, 10065, USA.
| |
Collapse
|
15
|
Howell R, Clarke MA, Reuschl AK, Chen T, Abbott-Imboden S, Singer M, Lowe DM, Bennett CL, Chain B, Jolly C, Fisher J. Executable network of SARS-CoV-2-host interaction predicts drug combination treatments. NPJ Digit Med 2022; 5:18. [PMID: 35165389 PMCID: PMC8844383 DOI: 10.1038/s41746-022-00561-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 01/07/2022] [Indexed: 12/15/2022] Open
Abstract
The COVID-19 pandemic has pushed healthcare systems globally to a breaking point. The urgent need for effective and affordable COVID-19 treatments calls for repurposing combinations of approved drugs. The challenge is to identify which combinations are likely to be most effective and at what stages of the disease. Here, we present the first disease-stage executable signalling network model of SARS-CoV-2-host interactions used to predict effective repurposed drug combinations for treating early- and late stage severe disease. Using our executable model, we performed in silico screening of 9870 pairs of 140 potential targets and have identified nine new drug combinations. Camostat and Apilimod were predicted to be the most promising combination in effectively supressing viral replication in the early stages of severe disease and were validated experimentally in human Caco-2 cells. Our study further demonstrates the power of executable mechanistic modelling to enable rapid pre-clinical evaluation of combination therapies tailored to disease progression. It also presents a novel resource and expandable model system that can respond to further needs in the pandemic.
Collapse
Affiliation(s)
- Rowan Howell
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Matthew A Clarke
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Ann-Kathrin Reuschl
- Division of Infection and Immunity, University College London, London, WC1E 6BT, UK
| | - Tianyi Chen
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Sean Abbott-Imboden
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, WC1E 6BT, UK
| | - David M Lowe
- Institute of Immunity and Transplantation, University College London, London, NW3 2PF, UK
| | - Clare L Bennett
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
- Institute of Immunity and Transplantation, University College London, London, NW3 2PF, UK
| | - Benjamin Chain
- Division of Infection and Immunity, University College London, London, WC1E 6BT, UK
- Department of Computer Science, Gower Street, University College London, London, WC1E 6BT, UK
| | - Clare Jolly
- Division of Infection and Immunity, University College London, London, WC1E 6BT, UK.
| | - Jasmin Fisher
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
16
|
Challenger JD, Foo CY, Wu Y, Yan AWC, Marjaneh MM, Liew F, Thwaites RS, Okell LC, Cunnington AJ. Modelling upper respiratory viral load dynamics of SARS-CoV-2. BMC Med 2022; 20:25. [PMID: 35022051 PMCID: PMC8755404 DOI: 10.1186/s12916-021-02220-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 12/15/2021] [Indexed: 02/09/2023] Open
Abstract
Relationships between viral load, severity of illness, and transmissibility of virus are fundamental to understanding pathogenesis and devising better therapeutic and prevention strategies for COVID-19. Here we present within-host modelling of viral load dynamics observed in the upper respiratory tract (URT), drawing upon 2172 serial measurements from 605 subjects, collected from 17 different studies. We developed a mechanistic model to describe viral load dynamics and host response and contrast this with simpler mixed-effects regression analysis of peak viral load and its subsequent decline. We observed wide variation in URT viral load between individuals, over 5 orders of magnitude, at any given point in time since symptom onset. This variation was not explained by age, sex, or severity of illness, and these variables were not associated with the modelled early or late phases of immune-mediated control of viral load. We explored the application of the mechanistic model to identify measured immune responses associated with the control of the viral load. Neutralising antibodies correlated strongly with modelled immune-mediated control of viral load amongst subjects who produced neutralising antibodies. Our models can be used to identify host and viral factors which control URT viral load dynamics, informing future treatment and transmission blocking interventions.
Collapse
Affiliation(s)
- Joseph D Challenger
- Medical Research Council Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK.
| | - Cher Y Foo
- School of Medicine, Imperial College London, London, UK
| | - Yue Wu
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Ada W C Yan
- Department of Infectious Disease, Imperial College London, London, UK
| | - Mahdi Moradi Marjaneh
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | - Felicity Liew
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Lucy C Okell
- Medical Research Council Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Aubrey J Cunnington
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK.,Centre for Paediatrics and Child Health, Imperial College London, London, UK
| |
Collapse
|
17
|
Cama VF, Marín-Prida J, Acosta-Rivero N, Acosta EF, Díaz LO, Casadesús AV, Fernández-Marrero B, Gilva-Rodríguez N, Cremata-García D, Cervantes-Llanos M, Piniella-Matamoros B, Sánchez D, Del Rosario-Cruz L, Borrajero I, Díaz A, González Y, Pentón-Arias E, Montero-González T, Guillen-Nieto G, Pentón-Rol G. The microglial NLRP3 inflammasome is involved in human SARS-CoV-2 cerebral pathogenicity: A report of three post-mortem cases. J Neuroimmunol 2021; 361:577728. [PMID: 34619427 PMCID: PMC8480138 DOI: 10.1016/j.jneuroim.2021.577728] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/18/2021] [Accepted: 09/26/2021] [Indexed: 12/21/2022]
Abstract
We herein report, by using confocal immunofluorescence, the colocalization of the SARS-CoV-2 nucleocapsid within neurons, astrocytes, oligodendrocytes and microglia in three deceased COVID-19 cases, of between 78 and 85 years of age at death. The viral nucleocapsid was detected together with its ACE2 cell entry receptor, as well as the NLRP3 inflammasome in cerebral cortical tissues. It is noteworthy that NLRP3 was colocalized with CD68 + macrophages in the brain and lung of the deceased, suggesting the critical role of this type of inflammasome in SARS-CoV-2 lesions of the nervous system/lungs and supporting its potential role as a therapeutic target.
Collapse
Affiliation(s)
- Viviana Falcón Cama
- Center for Genetic Engineering and Biotechnology (CIGB), Ave. 31 e/158 y 190, Cubanacán, Playa, PO Box 6162, Havana, Cuba; Latin American School of Medicine, Carretera Panamericana Km 3 1/2, Playa, Havana, 11600, Cuba..
| | - Javier Marín-Prida
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, Ave. 23 e/214 y 222, La Lisa, PO Box: 430, Havana, Cuba.
| | - Nelson Acosta-Rivero
- Center of Protein Research, Department of Biochemistry, Faculty of Biology, University of Havana, Havana, Cuba.
| | - Emilio F Acosta
- Center for Advanced Studies of Cuba, Havana, Cuba; Latin American School of Medicine, Carretera Panamericana Km 3 1/2, Playa, Havana, 11600, Cuba..
| | - Leonardo Oramas Díaz
- Center for Genetic Engineering and Biotechnology (CIGB), Ave. 31 e/158 y 190, Cubanacán, Playa, PO Box 6162, Havana, Cuba.
| | - Ana V Casadesús
- Direction of Immunology and Immunotherapy, Center of Molecular Immunology, Havana, Cuba.
| | | | - Nathalie Gilva-Rodríguez
- Center for Genetic Engineering and Biotechnology (CIGB), Ave. 31 e/158 y 190, Cubanacán, Playa, PO Box 6162, Havana, Cuba.
| | - Daina Cremata-García
- Center for Genetic Engineering and Biotechnology (CIGB), Ave. 31 e/158 y 190, Cubanacán, Playa, PO Box 6162, Havana, Cuba.
| | - Majel Cervantes-Llanos
- Center for Genetic Engineering and Biotechnology (CIGB), Ave. 31 e/158 y 190, Cubanacán, Playa, PO Box 6162, Havana, Cuba.
| | - Beatriz Piniella-Matamoros
- Center for Genetic Engineering and Biotechnology (CIGB), Ave. 31 e/158 y 190, Cubanacán, Playa, PO Box 6162, Havana, Cuba.
| | | | - Leticia Del Rosario-Cruz
- Hospital Militar Central "Dr. Luis Díaz Soto", Avenida Monumental km 2, Habana delEste, Havana, Cuba.
| | - Israel Borrajero
- Hospital Clínico Quirúrgico "Hermanos Ameijeiras", Havana, Cuba.
| | | | | | - Eduardo Pentón-Arias
- Center for Genetic Engineering and Biotechnology (CIGB), Ave. 31 e/158 y 190, Cubanacán, Playa, PO Box 6162, Havana, Cuba; Latin American School of Medicine, Carretera Panamericana Km 3 1/2, Playa, Havana, 11600, Cuba..
| | - Teresita Montero-González
- Hospital Militar Central "Dr. Luis Díaz Soto", Avenida Monumental km 2, Habana delEste, Havana, Cuba.
| | - Gerardo Guillen-Nieto
- Center for Genetic Engineering and Biotechnology (CIGB), Ave. 31 e/158 y 190, Cubanacán, Playa, PO Box 6162, Havana, Cuba; Latin American School of Medicine, Carretera Panamericana Km 3 1/2, Playa, Havana, 11600, Cuba..
| | - Giselle Pentón-Rol
- Center for Genetic Engineering and Biotechnology (CIGB), Ave. 31 e/158 y 190, Cubanacán, Playa, PO Box 6162, Havana, Cuba; Latin American School of Medicine, Carretera Panamericana Km 3 1/2, Playa, Havana, 11600, Cuba..
| |
Collapse
|
18
|
Su WQ, Yu XJ, Zhou CM. SARS-CoV-2 ORF3a Induces Incomplete Autophagy via the Unfolded Protein Response. Viruses 2021; 13:v13122467. [PMID: 34960736 PMCID: PMC8706696 DOI: 10.3390/v13122467] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/21/2022] Open
Abstract
In the past year and a half, SARS-CoV-2 has caused 240 million confirmed cases and 5 million deaths worldwide. Autophagy is a conserved process that either promotes or inhibits viral infections. Although coronaviruses are known to utilize the transport of autophagy-dependent vesicles for the viral life cycle, the underlying autophagy-inducing mechanisms remain largely unexplored. Using several autophagy-deficient cell lines and autophagy inhibitors, we demonstrated that SARS-CoV-2 ORF3a was able to induce incomplete autophagy in a FIP200/Beclin-1-dependent manner. Moreover, ORF3a was involved in the induction of the UPR (unfolded protein response), while the IRE1 and ATF6 pathways, but not the PERK pathway, were responsible for mediating the ORF3a-induced autophagy. These results identify the role of the UPR pathway in the ORF3a-induced classical autophagy process, which may provide us with a better understanding of SARS-CoV-2 and suggest new therapeutic modalities in the treatment of COVID-19.
Collapse
|
19
|
Rodriguez T, Dobrovolny HM. Estimation of viral kinetics model parameters in young and aged SARS-CoV-2 infected macaques. ROYAL SOCIETY OPEN SCIENCE 2021; 8:202345. [PMID: 34804559 PMCID: PMC8595996 DOI: 10.1098/rsos.202345] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 10/25/2021] [Indexed: 06/13/2023]
Abstract
The SARS-CoV-2 virus disproportionately causes serious illness and death in older individuals. In order to have the greatest impact in decreasing the human toll caused by the virus, antiviral treatment should be targeted to older patients. For this, we need a better understanding of the differences in viral dynamics between SARS-CoV-2 infection in younger and older adults. In this study, we use previously published averaged viral titre measurements from the nose and throat of SARS-CoV-2 infection in young and aged cynomolgus macaques to parametrize a viral kinetics model. We find that all viral kinetics parameters differ between young and aged macaques in the nasal passages, but that there are fewer differences in parameter estimates from the throat. We further use our parametrized model to study the antiviral treatment of young and aged animals, finding that early antiviral treatment is more likely to lead to a lengthening of the infection in aged animals, but not in young animals.
Collapse
Affiliation(s)
- Thalia Rodriguez
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, USA
| | - Hana M. Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, USA
| |
Collapse
|
20
|
Dogra P, Ramirez JR, Butner JD, Pelaez MJ, Cristini V, Wang Z. A Multiscale Model to Identify Limiting Factors in Nanoparticle-Based miRNA Delivery for Tumor Inhibition . ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:4230-4233. [PMID: 34892157 PMCID: PMC8712117 DOI: 10.1109/embc46164.2021.9630862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
MicroRNA-based gene therapy for cancer treatment via nanoparticles (NPs) requires navigation of multiple physical and physiological barriers in order to efficiently deliver the miRNAs to the cancer cell cytoplasm. We here present a mathematical model to investigate the variability associated with tumor, NP, and miRNA characteristics, and identify the limiting factors in miRNA delivery to tumors. Through global parameter analysis, the miRNA release rate from NPs and NP degradability were found to have the most significant impact on cytosolic accumulation of miRNAs. These NP properties can be fine-tuned in order to optimize the delivery system for enhancing the efficacy of miRNA-based therapy.Clinical Relevance-Understanding the effect of nanoparticle, tumor, and miRNA characteristics in governing the efficacy of miRNA-based cancer therapy will support its clinical translation.
Collapse
|
21
|
Immunology and controlling of coronaviruses; the current enemy for humanity: A review. Int J Biol Macromol 2021; 193:1532-1540. [PMID: 34732305 PMCID: PMC8557928 DOI: 10.1016/j.ijbiomac.2021.10.216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/30/2021] [Accepted: 10/28/2021] [Indexed: 12/17/2022]
Abstract
The Severe Acute Respiratory Syndrome-related Coronavirus 2 (COVID-19 or SARS-CoV-2) epidemic is professed as world disaster producing a worrying increasing mortality, particularly amongst vulnerable humans worldwide. Whether COVID-19 has a strong ability for acceptable genetic flexibility that amended for breaking immune responses quickly, it is critical to understand the adaptation mechanism between viruses and hosts that allows individuals to follow viral development. This can contribute to finding the appropriate treatment to combat the epidemic. However, the present information about viral adaptation mechanisms in hosts is still insufficient, and future investigations may reveal the unknown. Mutations and genetic variations are naturally occurring; however, the current knowledge about their mechanism and pathways still has many secrets. The present review also provides insights into the immune system, immunological memory, and the development of the COVID-19 vaccine. Other fighting methods against COVID-19 are also highlighted. The potential of antibodies, natural metabolites, and current suggest vaccines were applied to the face of this new threat.
Collapse
|
22
|
Campos M, Sempere JM, Galán JC, Moya A, Llorens C, de-Los-Angeles C, Baquero-Artigao F, Cantón R, Baquero F. Simulating the impact of non-pharmaceutical interventions limiting transmission in COVID-19 epidemics using a membrane computing model. ACTA ACUST UNITED AC 2021; 2:uqab011. [PMID: 34642663 PMCID: PMC8499911 DOI: 10.1093/femsml/uqab011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/03/2021] [Indexed: 01/08/2023]
Abstract
Epidemics caused by microbial organisms are part of the natural phenomena of increasing biological complexity. The heterogeneity and constant variability of hosts, in terms of age, immunological status, family structure, lifestyle, work activities, social and leisure habits, daily division of time and other demographic characteristics make it extremely difficult to predict the evolution of epidemics. Such prediction is, however, critical for implementing intervention measures in due time and with appropriate intensity. General conclusions should be precluded, given that local parameters dominate the flow of local epidemics. Membrane computing models allows us to reproduce the objects (viruses and hosts) and their interactions (stochastic but also with defined probabilities) with an unprecedented level of detail. Our LOIMOS model helps reproduce the demographics and social aspects of a hypothetical town of 10 320 inhabitants in an average European country where COVID-19 is imported from the outside. The above-mentioned characteristics of hosts and their lifestyle are minutely considered. For the data in the Hospital and the ICU we took advantage of the observations at the Nursery Intensive Care Unit of the Consortium University General Hospital, Valencia, Spain (included as author). The dynamics of the epidemics are reproduced and include the effects on viral transmission of innate and acquired immunity at various ages. The model predicts the consequences of delaying the adoption of non-pharmaceutical interventions (between 15 and 45 days after the first reported cases) and the effect of those interventions on infection and mortality rates (reducing transmission by 20, 50 and 80%) in immunological response groups. The lockdown for the elderly population as a single intervention appears to be effective. This modeling exercise exemplifies the application of membrane computing for designing appropriate multilateral interventions in epidemic situations.
Collapse
Affiliation(s)
- M Campos
- Department of Microbiology, Ramón y Cajal University Hospital, M-607, km 9,1 28034 Madrid, Spain
| | - J M Sempere
- Valencian Research Institute for Artificial Intelligence (VRAIN), Universitat Politècnica de Valencia, Camí de Vera s/n, 46022 Valencia, Spain
| | - J C Galán
- Department of Microbiology, Ramón y Cajal University Hospital, M-607, km 9,1 28034 Madrid, Spain
| | - A Moya
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública, M-607, km 9,1. 28034 Madrid, Spain
| | - C Llorens
- Biotechvana, Valencia, CEEI Building, Valencia Technological Park., C. agustín Escardino 9, 46980, Paterna, Valencia, Spain
| | - C de-Los-Angeles
- Nursery Unit, Intensive Care Unit and Pain Therapy, Consortium University General Hospital (CHGUV)., Av. Tres Cruces 2, 46014 Valencia, Spain
| | - F Baquero-Artigao
- Department of Infectious Diseases and Tropical Pediatrics, La Paz University Hospital., Av. Monforte de Lemos 2D, 28029 Madrid, Spain
| | - R Cantón
- Department of Microbiology, Ramón y Cajal University Hospital, M-607, km 9,1 28034 Madrid, Spain
| | - F Baquero
- Department of Microbiology, Ramón y Cajal University Hospital, M-607, km 9,1 28034 Madrid, Spain
| |
Collapse
|
23
|
Zarnitsyna VI, Gianlupi JF, Hagar A, Sego TJ, Glazier JA. Advancing therapies for viral infections using mechanistic computational models of the dynamic interplay between the virus and host immune response. Curr Opin Virol 2021; 50:103-109. [PMID: 34450519 PMCID: PMC8384423 DOI: 10.1016/j.coviro.2021.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/17/2022]
Abstract
The COVID-19 pandemic has highlighted a need for improved frameworks for drug discovery, repurposing, clinical trial design and therapy optimization and personalization. Mechanistic computational models can play an important role in developing these frameworks. We discuss how mechanistic models, which consider viral entry, replication in target cells, viral spread in the body, immune response, and the complex factors involved in tissue and organ damage and recovery, can clarify the mechanisms of humoral and cellular immune responses to the virus, viral distribution and replication in tissues, the origins of pathogenesis and patient-to-patient heterogeneity in responses. These models are already improving our understanding of the mechanisms of action of antivirals and immune modulators. We discuss how closer collaboration between the experimentalists, clinicians and modelers could result in more predictive models which may guide therapies for viral infections, improving survival and leading to faster and more complete recovery.
Collapse
Affiliation(s)
- Veronika I Zarnitsyna
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Juliano Ferrari Gianlupi
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA; Biocomplexity Institute, Indiana University, Bloomington, IN, USA
| | - Amit Hagar
- Department of History and Philosophy of Science, Indiana University, Bloomington, IN, USA
| | - T J Sego
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA; Biocomplexity Institute, Indiana University, Bloomington, IN, USA
| | - James A Glazier
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA; Biocomplexity Institute, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
24
|
Vaidya NK, Bloomquist A, Perelson AS. Modeling Within-Host Dynamics of SARS-CoV-2 Infection: A Case Study in Ferrets. Viruses 2021; 13:1635. [PMID: 34452499 PMCID: PMC8402735 DOI: 10.3390/v13081635] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/27/2021] [Accepted: 08/09/2021] [Indexed: 12/26/2022] Open
Abstract
The pre-clinical development of antiviral agents involves experimental trials in animals and ferrets as an animal model for the study of SARS-CoV-2. Here, we used mathematical models and experimental data to characterize the within-host infection dynamics of SARS-CoV-2 in ferrets. We also performed a global sensitivity analysis of model parameters impacting the characteristics of the viral infection. We provide estimates of the viral dynamic parameters in ferrets, such as the infection rate, the virus production rate, the infectious virus proportion, the infected cell death rate, the virus clearance rate, as well as other related characteristics, including the basic reproduction number, pre-peak infectious viral growth rate, post-peak infectious viral decay rate, pre-peak infectious viral doubling time, post-peak infectious virus half-life, and the target cell loss in the respiratory tract. These parameters and indices are not significantly different between animals infected with viral strains isolated from the environment and isolated from human hosts, indicating a potential for transmission from fomites. While the infection period in ferrets is relatively short, the similarity observed between our results and previous results in humans supports that ferrets can be an appropriate animal model for SARS-CoV-2 dynamics-related studies, and our estimates provide helpful information for such studies.
Collapse
Affiliation(s)
- Naveen K. Vaidya
- Department of Mathematics and Statistics, San Diego State University, San Diego, CA 92182, USA;
- Computational Science Research Center, San Diego State University, San Diego, CA 92182, USA
- Viral Information Institute, San Diego State University, San Diego, CA 92182, USA
| | - Angelica Bloomquist
- Department of Mathematics and Statistics, San Diego State University, San Diego, CA 92182, USA;
- Computational Science Research Center, San Diego State University, San Diego, CA 92182, USA
- Viral Information Institute, San Diego State University, San Diego, CA 92182, USA
| | - Alan S. Perelson
- Los Alamos National Laboratory, Theoretical Biology and Biophysics Group, Los Alamos, NM 87545, USA;
| |
Collapse
|
25
|
Badu K, Oyebola K, Zahouli JZB, Fagbamigbe AF, de Souza DK, Dukhi N, Amankwaa EF, Tolba MF, Sylverken AA, Mosi L, Mante PK, Matoke-Muhia D, Goonoo N. SARS-CoV-2 Viral Shedding and Transmission Dynamics: Implications of WHO COVID-19 Discharge Guidelines. Front Med (Lausanne) 2021; 8:648660. [PMID: 34239886 PMCID: PMC8259580 DOI: 10.3389/fmed.2021.648660] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/14/2021] [Indexed: 12/20/2022] Open
Abstract
The evolving nature of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has necessitated periodic revisions of COVID-19 patient treatment and discharge guidelines. Since the identification of the first COVID-19 cases in November 2019, the World Health Organization (WHO) has played a crucial role in tackling the country-level pandemic preparedness and patient management protocols. Among others, the WHO provided a guideline on the clinical management of COVID-19 patients according to which patients can be released from isolation centers on the 10th day following clinical symptom manifestation, with a minimum of 72 additional hours following the resolution of symptoms. However, emerging direct evidence indicating the possibility of viral shedding 14 days after the onset of symptoms called for evaluation of the current WHO discharge recommendations. In this review article, we carried out comprehensive literature analysis of viral shedding with specific focus on the duration of viral shedding and infectivity in asymptomatic and symptomatic (mild, moderate, and severe forms) COVID-19 patients. Our literature search indicates that even though, there are specific instances where the current protocols may not be applicable ( such as in immune-compromised patients there is no strong evidence to contradict the current WHO discharge criteria.
Collapse
Affiliation(s)
- Kingsley Badu
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Kolapo Oyebola
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Biochemistry and Nutrition Department, Nigerian Institute of Medical Research, Lagos, Nigeria
- Department of Zoology, Faculty of Science, University of Lagos, Lagos, Nigeria
| | - Julien Z. B. Zahouli
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, Abidjan, Côte d'Ivoire
- Centre d'Entomologie Médicale et Vétérinaire, Université Alassane Ouattara, Bouaké, Côte d'Ivoire
| | - Adeniyi Francis Fagbamigbe
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Epidemiology and Medical Statistics, Faculty of Public Health, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Division of Population and Behavioral Sciences, School of Medicine, St. Andrews University, St. Andrews, United Kingdom
| | - Dziedzom K. de Souza
- African Academy of Sciences Affiliates, Nairobi, Kenya
- College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Natisha Dukhi
- African Academy of Sciences Affiliates, Nairobi, Kenya
- College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
- Human and Social Capabilities Division, Human Sciences Research Council, Cape Town, South Africa
| | - Ebenezer F. Amankwaa
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Geography and Resource Development, University of Ghana, Accra, Ghana
| | - Mai F. Tolba
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- The Center of Drug Discovery Research and Development, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Egypt
| | - Augustina A. Sylverken
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Lydia Mosi
- African Academy of Sciences Affiliates, Nairobi, Kenya
- West African Centre for Cell Biology of Infectious Diseases, University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Accra, Ghana
| | - Priscilla Kolibea Mante
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Department of Pharmacology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Damaris Matoke-Muhia
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, Nairobi, Kenya
| | - Nowsheen Goonoo
- African Academy of Sciences Affiliates, Nairobi, Kenya
- Biomaterials, Drug Delivery and Nanotechnology Unit, Center for Biomedical and Biomaterials Research (CBBR), University of Mauritius, Reduit, Mauritius
| |
Collapse
|
26
|
Dogra P, Koay EJ, Wang Z, Vahidy FS, Ferrari M, Pasqualini R, Arap W, Boom ML, Dirk Sostman H, Cristini V. Is the worst of the COVID-19 global pandemic yet to come? Application of financial mathematics as candidate predictive tools. Transl Psychiatry 2021; 11:299. [PMID: 34016952 PMCID: PMC8134815 DOI: 10.1038/s41398-021-01429-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/21/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
The Elliott Wave principle is a time-honored, oft-used method for predicting variations in the financial markets. It is based on the notion that human emotions drive financial decisions. In the fight against the COVID-19 global pandemic, human emotions are similarly decisive, for instance in that they determine one's willingness to be vaccinated, and/or to follow preventive measures including the personal wearing of masks, the application of social distancing protocols, and frequent handwashing. On this basis, we postulated that the Elliott Wave Principle may similarly be used to predict the future evolution of the COVID-19 pandemic. We demonstrated that this method reproduces the data pattern for various countries and the world (daily new cases). Potential scenarios were then extrapolated, from the best-case corresponding to a rapid, full vaccination of the population, to the utterly disastrous case of slow vaccination, and poor adherence to preventive protocols.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Eugene J Koay
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Farhaan S Vahidy
- Center for Outcomes Research, Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Neurological Institute, Houston Methodist, Houston, TX, USA
| | - Mauro Ferrari
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
- Dompé X-Therapeutics, San Mateo, CA, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
- Department of Radiation Oncology, Division of Cancer Biology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Marc L Boom
- Department of Medicine, Houston Methodist, Houston, TX, USA
| | - H Dirk Sostman
- Weill Cornell Medicine, New York, NY, USA
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Academic Institute, Houston, TX, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
27
|
Kriegova E, Fillerova R, Raska M, Manakova J, Dihel M, Janca O, Sauer P, Klimkova M, Strakova P, Kvapil P. Excellent option for mass testing during the SARS-CoV-2 pandemic: painless self-collection and direct RT-qPCR. Virol J 2021; 18:95. [PMID: 33947425 PMCID: PMC8094981 DOI: 10.1186/s12985-021-01567-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/29/2021] [Indexed: 11/10/2022] Open
Abstract
The early identification of asymptomatic yet infectious cases is vital to curb the 2019 coronavirus (COVID-19) pandemic and to control the disease in the post-pandemic era. In this paper, we propose a fast, inexpensive and high-throughput approach using painless nasal-swab self-collection followed by direct RT-qPCR for the sensitive PCR detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This approach was validated in a large prospective cohort study of 1038 subjects, analysed simultaneously using (1) nasopharyngeal swabs obtained with the assistance of healthcare personnel and analysed by classic two-step RT-qPCR on RNA isolates and (2) nasal swabs obtained by self-collection and analysed with direct RT-qPCR. Of these subjects, 28.6% tested positive for SARS-CoV-2 using nasopharyngeal swab sampling. Our direct RT-qPCR approach for self-collected nasal swabs performed well with results similar to those of the two-step RT-qPCR on RNA isolates, achieving 0.99 positive and 0.98 negative predictive values (cycle threshold [Ct] < 37). Our research also reports on grey-zone viraemia, including samples with near-cut-off Ct values (Ct ≥ 37). In all investigated subjects (n = 20) with grey-zone viraemia, the ultra-small viral load disappeared within hours or days with no symptoms. Overall, this study underscores the importance of painless nasal-swab self-collection and direct RT-qPCR for mass testing during the SARS-CoV-2 pandemic and in the post-pandemic era.
Collapse
Affiliation(s)
- Eva Kriegova
- Department of Immunology, OLGEN, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hnevotinska 3, 77900 Olomouc, Czech Republic
| | - Regina Fillerova
- Department of Immunology, OLGEN, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hnevotinska 3, 77900 Olomouc, Czech Republic
| | - Milan Raska
- Department of Immunology, OLGEN, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hnevotinska 3, 77900 Olomouc, Czech Republic
| | - Jirina Manakova
- Department of Immunology, OLGEN, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hnevotinska 3, 77900 Olomouc, Czech Republic
| | - Martin Dihel
- Department of Immunology, OLGEN, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hnevotinska 3, 77900 Olomouc, Czech Republic
| | - Ondrej Janca
- Department of Immunology, OLGEN, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Hnevotinska 3, 77900 Olomouc, Czech Republic
| | - Pavel Sauer
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | | | | | - Petr Kvapil
- Institute of Applied Biotechnologies a.s., Prague, Czech Republic
| |
Collapse
|
28
|
Dogra P, Koay EJ, Wang Z, Vahidy FS, Ferrari M, Pasqualini R, Arap W, Boom ML, Sostman HD, Cristini V. Do Pandemics Obey the Elliott Wave Principle of Financial Markets? MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.21.21250273. [PMID: 33501456 PMCID: PMC7836128 DOI: 10.1101/2021.01.21.21250273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The Elliott Wave principle is a time-honored, oft-used method for predicting variations in the financial markets. It is based on the notion that human emotions drive financial decisions. In the fight against COVID-19, human emotions are similarly decisive, for instance in that they determine one's willingness to be vaccinated, and/or to follow preventive measures including the wearing of masks, the application of social distancing protocols, and frequent handwashing. On this basis, we postulated that the Elliott Wave Principle may similarly be used to predict the future evolution of the COVID-19 pandemic. We demonstrated that this method reproduces the data pattern especially well for USA (daily new cases). Potential scenarios were then extrapolated, from the best-case corresponding to a rapid, full vaccination of the population, to the utterly disastrous case of slow vaccination, and poor adherence to preventive protocols.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Eugene J. Koay
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| | - Farhaan S. Vahidy
- Center for Outcomes Research, Houston Methodist Research Institute, Houston, Texas
- Houston Methodist Neurological Institute, Houston Methodist, Houston, Texas
| | - Mauro Ferrari
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
- Dompé X-Therapeutics, San Mateo, California, USA
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
- Department of Radiation Oncology, Division of Cancer Biology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Marc L. Boom
- Department of Medicine, Houston Methodist, Houston, Texas
| | - H. Dirk Sostman
- Weill Cornell Medicine, New York, NY, USA
- Houston Methodist Research Institute, Houston, TX, USA
- Houston Methodist Academic Institute, Houston, TX, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
29
|
Baquero F, Campos M, Llorens C, Sempere JM. P systems in the time of COVID-19. JOURNAL OF MEMBRANE COMPUTING 2021; 3. [PMCID: PMC8555730 DOI: 10.1007/s41965-021-00083-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
In this paper, we present LOIMOS, which is an epidemiological scenario simulator developed in the context of the fight against the pandemic caused by coronavirus SARS-CoV-2 on a global scale. LOIMOS has been fully developed under the paradigm of membrane computing using transition P systems with communication rules, active membranes and a stochastic simulator engine. In this paper we detail the main components of the system and we report some examples of epidemiological scenarios evaluated with LOIMOS.
Collapse
Affiliation(s)
- Fernando Baquero
- Department of Microbiology, Ramón y Cajal Hospital, IRYCIS, Madrid, Spain
| | - Marcelino Campos
- Department of Microbiology, Ramón y Cajal Hospital, IRYCIS, Madrid, Spain
- VRAIN, Universitat Politècnica de València, Valencia, Spain
| | | | | |
Collapse
|