Randomized double-blind clinical study in patients with COVID-19 to evaluate the safety and efficacy of a phytomedicine (P2Et)

Claudia Urueña, Ricardo Ballesteros-Ramírez, Alejandra Gomez-Cadena, Alfonso Barreto, Karol Prieto, Sandra Quijano, Pablo Aschner, Carlos Martínez, Maria I Zapata-Cardona, Hajar El-Ahanidi, Camilla Jandus, Lizdany Florez-Alvarez, Maria Teresa Rugeles, Wildeman Zapata-Builes, Angel Alberto Garcia, Susana Fiorentino, Claudia Urueña, Ricardo Ballesteros-Ramírez, Alejandra Gomez-Cadena, Alfonso Barreto, Karol Prieto, Sandra Quijano, Pablo Aschner, Carlos Martínez, Maria I Zapata-Cardona, Hajar El-Ahanidi, Camilla Jandus, Lizdany Florez-Alvarez, Maria Teresa Rugeles, Wildeman Zapata-Builes, Angel Alberto Garcia, Susana Fiorentino

Abstract

Background: It has been proposed that polyphenols can be used in the development of new therapies against COVID-19, given their ability to interfere with the adsorption and entrance processes of the virus, thus disrupting viral replication. Seeds from Caesalpinia spinosa, have been traditionally used for the treatment of inflammatory pathologies and respiratory diseases. Our team has obtained an extract called P2Et, rich in polyphenols derived from gallic acid with significant antioxidant activity, and the ability to induce complete autophagy in tumor cells and reduce the systemic inflammatory response in animal models.

Methods: In this work, a phase II multicenter randomized double-blind clinical trial on COVID-19 patients was designed to evaluate the impact of the P2Et treatment on the clinical outcome and the immunological parameters related to the evolution of the disease. The Trial was registered with the number No. NCT04410510*. A complementary study in an animal model of lung fibrosis was carried out to evaluate in situ lung changes after P2Et in vivo administration. The ability of P2Et to inhibit the viral load of murine and human coronaviruses in cellular models was also evaluated.

Results: Patients treated with P2Et were discharged on average after 7.4 days of admission vs. 9.6 days in the placebo group. Although a decrease in proinflammatory cytokines such as G-CSF, IL-15, IL-12, IL-6, IP10, MCP-1, MCP-2 and IL-18 was observed in both groups, P2Et decreased to a greater extent G-CSF, IL-6 and IL-18 among others, which are related to lower recovery of patients in the long term. The frequency of T lymphocytes (LT) CD3+, LT double negative (CD3+CD4-CD8-), NK cells increased in the P2Et group where the population of eosinophils was also significantly reduced. In the murine bleomycin model, P2Et also reduced lung inflammation and fibrosis. P2Et was able to reduce the viral replication of murine and human coronaviruses in vitro, showing its dual antiviral and anti-inflammatory role, key in disease control.

Conclusions: Taken together these results suggest that P2Et could be consider as a good co-adjuvant in the treatment of COVID-19.

Clinical trail registration: https://ichgcp.net/clinical-trials-registry/NCT04410510, identifier: NCT04410510.

Keywords: COVID-19; complementary and alternative medicine; immunomodulation; phytomedicine; polymolecular drugs; polyphenols.

Conflict of interest statement

Authors SF and CU are inventors of a granted patent related to P2Et. Authors SF, CU, and RB-R are partners of the DreemBio company who was a licensee of related patents. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Urueña, Ballesteros-Ramírez, Gomez-Cadena, Barreto, Prieto, Quijano, Aschner, Martínez, Zapata-Cardona, El-Ahanidi, Jandus, Florez-Alvarez, Rugeles, Zapata-Builes, Garcia and Fiorentino.

Figures

Figure 1
Figure 1
Screening, Randomization, and Outcomes. COVID-19 patients were included in the study. 1,042 patients were screened and 91 patients underwent randomization. 47 patients received placebo and 44 patients received P2Et extract. A blood sample was taken from each patient before and after treatment to assess safety parameters, clinical parameters, immune innate cell populations by flow cytometry and cytokines for a MAGPIX® System. This figure was created using BioRender (https://Biorender.com/).
Figure 2
Figure 2
Absolute counts of Innate lymphocytes cells in the peripheral blood of COVID-19 patients. (A) Absolute counts of total Innate lymphocytes cells (ILCs) in COVID-19 patients (moderate and severe) treated with placebo or P2Et. Healthy donors (HD) were included. (B) Absolute counts of Innate type 2 lymphocytes cells (ILC2s) in COVID-19 patients (moderate and severe) treated with placebo or P2Et. HD were included. (C) Absolute counts of Natural killer Bright cells (NKBr) in COVID-19 patients (moderate and severe) treated with placebo or P2Et. HD were included. (D) Absolute counts of Natural killer Dim cells (NKDim) in COVID-19 patients (moderate and severe) treated with placebo or P2Et. HD were included. Data are represented as the mean ± SEM. The p values between Placebo and P2Et group or within groups were calculated using a Mann-Whitney test. *p < 0.05, **p < 0.01. The p values between COVID-19 patients and HD were calculated using a Mann-Whitney test. ♦p < 0.05, ♦♦p < 0.01.
Figure 3
Figure 3
P2Et treatment modulate of inflammatory response in COVID-19 patients. Samples from placebo, P2Et and Healthy donors (HD) groups were analyzed by Milliplex to identify the quantity of cytokines present in the serum. (A) IL-5, (B) IL-6, (C) G.CSF, (D) IL-10, (E). IL-18, (F) IFN-γ, (G) MCP-2, and (H) MCP-1 cytokines. Each dot represent one human sample. The p values between placebo and P2Et group or within groups were calculated using a Wilconxon test. *p < 0.05, **p < 0.01, ***p < 0.001.
Figure 4
Figure 4
P2Et exhibit antiviral activity against SARS-CoV-2 with a concentration-depending effect. (A) Vero E6 viability after 48 h of P2Et treatment was evaluated by MTT assay (6.25-−200 μg/mL). Data were presented as Mean ± SEM. The viability percentages of the treated cell were calculated based on untreated control. Two independent experiments with three replicates each were performed (n = 6). (B) The SARS-CoV-2 titer (PFU/mL) was reduced in Vero E6 supernatants after pre-post treatment with P2Et (n = 2). CQ (100 μM) was used as a positive control of viral inhibition. Data are represented as the mean ± SEM. The p values were calculated using a Mann-Whitney test, **p < 0.01, ***p < 0.001. Representative plaques of each treatment condition are shown.
Figure 5
Figure 5
P2Et treatment reduces ILC2 the pro-fibrotic factors in the lung of bleomycine challenged mice. (A) Lungs from naïve animals were used as negative controls compared and to bleomycine challenged animals treated either with PBS or P2Et. Absolut counts from lung ex vivo samples are shown. (B) Frequency of PD1 and CD69 positive cells among the ILC2 subset is presented. (C) Part of the lung cell suspension was used for an overnight re-stimulation with IL-33 and IL-25 in vitro. Intracellular cytokine staining was performed, and the frequencies of IL-5 and IL-13 positive ILC2 are shown. (D) From the ex-vivo lung cell suspension eosinophils and neutrophils were identified, and absolute numbers are represented. (E) Samples from all groups were analyzed by LegendPlex to identify the quantity of cytokines present in the serum of each animal. (F) Lung tissue was analyzed by qPCR to check the mRNA expression of pro-fibrotic factors. All graphs are presented as box and whiskers graphs showing the min and max. The middle line is plotted at the median. Each dot represent one mouse. The p values were calculated using a Kruskall-Wallis test, *p < 0.05, **p ≤ 0.01, ***p < 0.001, ****p < 0.001.
Figure 6
Figure 6
Proposed model for the P2Et effect in COVID-19. COVID-19 patients (moderate or severe) were treated with P2Et or Placebo. P2Et treatment (upper left) decreases ex vivo viral replication as well as proinflammatory cytokines such as IL-5, IL-6, IL-10, IL-18, G-CSF, and increases IFN-γ in peripheral blood. A decrease in eosinophils and an increase in other cell populations such as LT CD3+CD4-CD8-, NKBr, NKDim, and ILC2s are observed. The upper right image shows the effect of P2Et treatment in a mouse model of bleomycin-induced pulmonary fibrosis. In the lung, it decreases CD45+ cells, total ILC, ILC-2, eosinophils, and neutrophils. In peripheral blood it increases the production of IL-22. In summary, P2Et treatment regulates the inflammation induced by SARS-CoV-2 infection, modulating factors related to long-term clinical manifestations. The lower left image shows the effect of Placebo treatment in patients with COVID-19. A slight decrease in IL-6 and IL-10 is observed in moderate patients but an increase in IL-10 in severe patients. For the other cytokines (IL-5, IL-18, G-CSF, and IFN-γ) no significant changes are observed. On the contrary, in this group of patients, an increase in eosinophils and a slight increase in NKDim cells, as well as a decrease in LT CD3+CD4−CD8−, NKBr and ILC2s, are observed. In the PBS mouse model (negative control) there is evidence of an increase in CD45+ cells, total ILCs, ILC-2s, eosinophils, and neutrophils as well as a slight increase in markers related to pulmonary fibrosis in the lung that show the exaggerated inflammation. Image created using BioRender (https://Biorender.com/).

References

    1. Dutra RC, Campos MM, Santos ARS, Calixto JB. Medicinal plants in Brazil: Pharmacological studies, drug discovery, challenges and perspectives. Pharmacol Res. (2016) 112:4–29. 10.1016/j.phrs.2016.01.021
    1. Brendler T, Al-Harrasi A, Bauer R, Gafner S, Hardy ML, Heinrich M, et al. . Botanical drugs and supplements affecting the immune response in the time of COVID-19: implications for research and clinical practice. Phyther Res. (2021) 35:3013–31. 10.1002/ptr.7008
    1. Nadeem A, Chhabra SK, Masood A, Raj HG. Increased oxidative stress and altered levels of antioxidants in asthma. J Allergy Clin Immunol. (2003) 111:72–8. 10.1067/mai.2003.17
    1. Aruoma OI, Spencer JPE, Rossi R, Aeschbach R, Khan A, Mahmood N, et al. . An evaluation of the antioxidant and antiviral action of extracts of rosemary and Provencal herbs. Food Chem Toxicol. (1996) 34:449–56. 10.1016/0278-6915(96)00004-X
    1. Muhammad Y, Kani YA, Iliya S, Muhammad JB, Binji A, El-Fulaty Ahmad A, et al. . Deficiency of antioxidants and increased oxidative stress in COVID-19 patients: a cross-sectional comparative study in Jigawa, Northwestern Nigeria. SAGE Open Med. (2021) 9:2050312121991246. 10.1177/2050312121991246
    1. da Silva Antonio A, Wiedemann LSM, Veiga-Junior VF. Natural products' role against COVID-19. RSC Adv. (2020) 10:23379–93. 10.1039/D0RA03774E
    1. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. . Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. (2020) 395:497–506. 10.1016/S0140-6736(20)30183-5
    1. Stebbing J, Phelan A, Griffin I, Tucker C, Oechsle O, Smith D, et al. . COVID-19: combining antiviral and anti-inflammatory treatments. Lancet Infect Dis. (2020) 20:400–2. 10.1016/S1473-3099(20)30132-8
    1. Guo W, Kong E, Meydani M. Dietary polyphenols, inflammation, and cancer. Nutr Cancer. (2009) 61:807–10. 10.1080/01635580903285098
    1. Prieto K, Cao Y, Mohamed E, Trillo-Tinoco J, Sierra RA, Urueña C, et al. . Polyphenol-rich extract induces apoptosis with immunogenic markers in melanoma cells through the ER stress-associated kinase PERK. Cell death Discov. (2019) 5:1–15. 10.1038/s41420-019-0214-2
    1. Sandoval TA, Urueña CP, Llano M, Gómez-Cadena A, Hernández JF, Sequeda LG, et al. . Standardized extract from Caesalpinia spinosa is cytotoxic over cancer stem cells and enhance anticancer activity of doxorubicin. Am J Chin Med. (2016) 44:1693–717. 10.1142/S0192415X16500956
    1. Limanaqi F, Busceti CL, Biagioni F, Lazzeri G, Forte M, Schiavon S, et al. . Cell clearing systems as targets of polyphenols in viral infections: potential implications for COVID-19 pathogenesis. Antioxidants. (2020) 9:1105. 10.3390/antiox9111105
    1. Urueña C, Mancipe J, Hernandez J, Castañeda D, Pombo L, Gomez A, et al. . Gallotannin-rich Caesalpinia spinosa fraction decreases the primary tumor and factors associated with poor prognosis in a murine breast cancer model. BMC Complement Altern Med. (2013) 13:74. 10.1186/1472-6882-13-74
    1. Organización Panamericana de la Salud . Buenas prácticas clínicas: documento de las Américas. IV Conferencia Panamericana para la Armonización de la Reglamentación Farmacéutica. Organización Panamericana de la salud. Oficina Regional de la Organización Mundial de la salud. (2005)
    1. World Medical Association . World medical association declaration of helsinki: ethical principles for medical research involving human subjects. JAMA. (2013) 310:2191–4. 10.1001/jama.2013.281053
    1. Rubenfeld GD, Thompson T, Ferguson ND, Caldwell E, Fan E, Camporota L, et al. . Acute respiratory distress syndrome. The Berlin Def JAMA. (2012) 307:2526–33. 10.1001/jama.2012.5669
    1. Olson G, Davis AM. Diagnosis and treatment of adults with community-acquired pneumonia. JAMA. (2020) 323:885–6. 10.1001/jama.2019.21118
    1. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, et al. . The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA. (2016) 315:801–10. 10.1001/jama.2016.0287
    1. Saavedra Trujillo CH. Consenso colombiano de atención, diagnóstico y manejo de la infección por SARS-CoV-2/COVID-19 en establecimientos de atención de la salud. Recomendaciones basadas en consenso de expertos e informadas en la evidencia. Infectio. (2020) 24:186–261. 10.22354/in.v24i3.852
    1. Ballesteros-Ramírez R, Durán MI, Fiorentino S. Genotoxicity and mutagenicity assessment of a standardized extract (P2Et) obtained from Caesalpinia spinosa. Toxicol Rep. (2021) 8:258–63. 10.1016/j.toxrep.2020.12.024
    1. Castañeda DM, Pombo LM, Urueña CP, Hernandez JF, Fiorentino S. A gallotannin-rich fraction from Caesalpinia spinosa (Molina) Kuntze displays cytotoxic activity and raises sensitivity to doxorubicin in a leukemia cell line. BMC Complement Altern Med. (2012) 12:38. 10.1186/1472-6882-12-38
    1. Administration F and D . Botanical Drug Development Guidance for Industry. U.S Food and Drug Administration (FDA) (2016).
    1. International conference on harmonization of technical requirements for the registration of pharmaceuticals for human use . M3 (R2): Nonclinical safety studies for the conduct of human clinical trials and marketing authorization for pharmaceuticals. International conference on harmonization of technical requirements for the registration of pharmaceuticals for human use. European Medicines Agency. Science Medicines Health; (2009).
    1. Duran MI, Ballesteros-Ramírez R, Tellez A, Torregrosa L, Olejua PA, Galvis S, et al. . Safety evaluation in healthy colombian volunteers of P2Et extract obtained from caesalpinia spinosa: design 3+ 3 phase i clinical trial. Evidence-Based Complement Altern Med. (2022) 1–11. 10.1155/2022/7943001
    1. National Cancer Institute . National Institutes of Health Common Terminology Criteria for Adverse Events (CTCAE). National Cancer Institute. Division of Cancer Treatment & Diagnosis; (2009).
    1. Zapata-Cardona MI, Flórez-Álvarez L, Zapata-Builes W, Guerra-Sandoval AL, Guerra-Almonacid CM, Hincapié-García J, et al. . Atorvastatin Effectively Inhibits Ancestral and Two Emerging Variants of SARS-CoV-2 in vitro. Front Microbiol. (2022) 13:721103. 10.3389/fmicb.2022.721103
    1. Marín-Palma D, Tabares-Guevara JH, Zapata-Cardona MI, Flórez-Álvarez L, Yepes LM, Rugeles MT, et al. . Curcumin Inhibits invitro SARS-CoV-2 infection in vero E6 cells through multiple antiviral mechanisms. Molecules. (2021) 26:6900. 10.3390/molecules26226900
    1. Van Dongen JJM, Lhermitte L, Böttcher S, Almeida J, Van Der Velden VHJ, Flores-Montero J, et al. . EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes. Leukemia. (2012) 26:1908–75. 10.1038/leu.2012.120
    1. Smadja DM, Mentzer SJ, Fontenay M, Laffan MA, Ackermann M, Helms J, et al. . COVID-19 is a systemic vascular hemopathy: insight for mechanistic and clinical aspects. Angiogenesis. (2021) 24:755–88. 10.1007/s10456-021-09805-6
    1. Choi HM, Moon SY, Yang HI, Kim KS. Understanding viral infection mechanisms and patient symptoms for the development of COVID-19 therapeutics. Int J Mol Sci. (2021) 22:1737. 10.3390/ijms22041737
    1. Jain VK, Iyengar KP, Ish P. Elucidating causes of COVID-19 infection and related deaths after vaccination. Diabetes Metab Syndr Clin Res Rev. (2021) 15:102212. 10.1016/j.dsx.2021.102212
    1. Levy E, Delvin E, Marcil V, Spahis S. Can phytotherapy with polyphenols serve as a powerful approach for the prevention and therapy tool of novel coronavirus disease 2019 (COVID-19)? Am J Physiol Metab. (2020) 319:E689–708. 10.1152/ajpendo.00298.2020
    1. Neeland MR, Bannister S, Clifford V, Dohle K, Mulholland K, Sutton P, et al. . Innate cell profiles during the acute and convalescent phase of SARS-CoV-2 infection in children. Nat Commun. (2021) 12:1–5. 10.1038/s41467-021-21414-x
    1. Liao M, Liu Y, Yuan J, Wen Y, Xu G, Zhao J, et al. . Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat Med. (2020) 26:842–4. 10.1038/s41591-020-0901-9
    1. Huang W, Li M, Luo G, Wu X, Su B, Zhao L, et al. . The inflammatory factors associated with disease severity to predict COVID-19 progression. J Immunol. (2021) 206:1597–608. 10.4049/jimmunol.2001327
    1. Lim YX, Ng YL, Tam JP, Liu DX. Human coronaviruses: a review of virus–host interactions. Diseases. (2016) 4:26. 10.3390/diseases4030026
    1. Udwadia ZF, Koul PA, Richeldi L. Post-COVID lung fibrosis: the tsunami that will follow the earthquake. Lung India Off Organ Indian Chest Soc. (2021) 38:S41. 10.4103/lungindia.lungindia_818_20
    1. Taylor S, Huang Y, Mallett G, Stathopoulou C, Felizardo TC, Sun M-A, et al. . PD-1 regulates KLRG1+ group 2 innate lymphoid cells. J Exp Med. (2017) 214:1663–78. 10.1084/jem.20161653
    1. Kouro T, Takatsu K. IL-5-and eosinophil-mediated inflammation: from discovery to therapy. Int Immunol. (2009) 21:1303–9. 10.1093/intimm/dxp102
    1. Gounni AS, Nutku E, Koussih L, Aris F, Louahed J, Levitt RC, et al. . IL-9 expression by human eosinophils: regulation by IL-1β and TNF-α. J Allergy Clin Immunol. (2000) 106:460–6. 10.1067/mai.2000.109172
    1. Long H, Liao W, Wang L, Lu Q. A player and coordinator: the versatile roles of eosinophils in the immune system. Transfus Med Hemotherapy. (2016) 43:96–108. 10.1159/000445215
    1. Zahran AM, Zahran ZAM, Mady YH, Mahran EEMO, Rashad A, Makboul A, et al. . Differential alterations in peripheral lymphocyte subsets in COVID-19 patients: upregulation of double-positive and double-negative T cells. Multidiscip Respir Med. (2021) 16:758. 10.4081/mrm.2021.758
    1. Wu Z, Zheng Y, Sheng J, Han Y, Yang Y, Pan H, et al. . CD3+ CD4-CD8-(Double-Negative) T Cells in Inflammation, Immune Disorders and Cancer. Front Immunol. (2022) 13:816005. 10.3389/fimmu.2022.816005
    1. Neyt K, GeurtsvanKessel CH, Lambrecht BN. Double-negative T resident memory cells of the lung react to influenza virus infection via CD11chi dendritic cells. Mucosal Immunol. (2016) 9:999–1014. 10.1038/mi.2015.91
    1. van Daal MT, Folkerts G, Garssen J, Braber S. Pharmacological modulation of immune responses by nutritional components. Pharmacol Rev. (2021) 73:1369–403. 10.1124/pharmrev.120.000063
    1. Kawai K, Tsuno NH, Kitayama J, Sunami E, Takahashi K, Nagawa H. Catechin inhibits adhesion and migration of peripheral blood B cells by blocking CD11b. Immunopharmacol Immunotoxicol. (2011) 33:391–7. 10.3109/08923973.2010.522195

Source: PubMed

3
订阅