Trained Immunity-Based Vaccine in B Cell Hematological Malignancies With Recurrent Infections: A New Therapeutic Approach

Juliana Ochoa-Grullón, Celina Benavente Cuesta, Ataúlfo González Fernández, Gustavo Cordero Torres, Cristina Pérez López, Ascensión Peña Cortijo, Laura Conejero Hall, Marta Mateo Morales, Antonia Rodríguez de la Peña, Carmen M Díez-Rivero, Edgard Rodríguez de Frías, Kissy Guevara-Hoyer, Miguel Fernández-Arquero, Silvia Sánchez-Ramón, Juliana Ochoa-Grullón, Celina Benavente Cuesta, Ataúlfo González Fernández, Gustavo Cordero Torres, Cristina Pérez López, Ascensión Peña Cortijo, Laura Conejero Hall, Marta Mateo Morales, Antonia Rodríguez de la Peña, Carmen M Díez-Rivero, Edgard Rodríguez de Frías, Kissy Guevara-Hoyer, Miguel Fernández-Arquero, Silvia Sánchez-Ramón

Abstract

Infectious complications are a major cause of morbidity and mortality in B-cell hematological malignancies (HM). Prophylaxis for recurrent infections in HM patients with antibody deficiency consists of first-line antibiotics and when unsuccessful, gammaglobulin replacement therapy (IgRT). Recent knowledge of trained immunity-based vaccines (TIbV), such as the sublingual polybacterial formulation MV130, has shown a promising strategy in the management of patients with recurrent infections. We sought to determine the clinical benefit of MV130 in a cohort of HM patients with recurrent respiratory tract infections (RRTIs) who underwent immunization with MV130 for 3 months. Clinical information included the frequency of infections, antibiotic use, number of visits to the GP and hospitalizations previous and after MV130 immunotherapy. Improvement on infection rate was classified as: clear (>60% reduction of infection), partial (26%-60%) and low (≤25%) improvement. Fifteen HM patients (aged 42 to 80 years; nine females) were included in the study. All patients reduced their infection rate. Analysis of paired data revealed that the median (range, min - max) of respiratory infectious rate significantly decreased from 4.0 (8.0-3.0) to 2.0 (4.0-0.0) (p<0.001) at 12 months of MV130. A clear clinical improvement was observed in 53% (n = 8) of patients, partial improvement in 40% (n = 6) and low improvement in 7% (n = 1). These data correlated with a decrease on antibiotic consumption from 3.0 (8.0-1.0) to 1.0 (2.0-0.0) (p = 0.002) during 12 months after initiation of treatment with MV130. The number of infectious-related GP or emergency room visits declined from 4.0 (8.0-2.0) to 2.0 (3.0-0.0) (p<0.001), in parallel with a reduction in hospital admissions due to infections (p = 0.032). Regarding safety, no adverse events were observed. On the other hand, immunological assessment of serum IgA and IgG levels demonstrated an increase in specific antibodies to MV130-contained bacteria following MV130 immunotherapy. In conclusion, MV130 may add clinical benefit reducing the rate of infections and enhancing humoral immune responses in these vulnerable patients.

Keywords: IgA; MV130; hematological malignancies; prophylaxis; recurrent respiratory tract infections; trained immunity-based vaccines.

Conflict of interest statement

LC and CD-R belong to the Immunotek R+D Department. 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 © 2021 Ochoa-Grullón, Benavente Cuesta, González Fernández, Cordero Torres, Pérez López, Peña Cortijo, Conejero Hall, Mateo Morales, Rodríguez de la Peña, Díez-Rivero, Rodríguez de Frías, Guevara-Hoyer, Fernández-Arquero and Sánchez-Ramón.

Figures

Figure 1
Figure 1
Flow chart of the study including enrolment, medical intervention, therapy and follow-up, and analysis. RRTIs, recurrent respiratory tract infection; IT, immunotherapy; GP, general practitioner; ER, emergency room.
Figure 2
Figure 2
Prophylaxis with MV130 reduces the incidence of respiratory infection rate (%) in patients with B cell hematological malignancies. (A, B) Number of respiratory tract infections one year before immunotherapy with MV130 (Pre) and in the 12 months after initiation of immunotherapy (Post) (A-tabulated, B-graph). (A, C) Percentage of improvement (reduction of infections) (A-tabulated, C-pie chart). Improvement was classified as: clear (>60% reduction of infectious episodes), partial (26-60% reduction of infectious episodes) and low (<25% reduction of infectious episodes). A total of 15 patients were included. Normal distribution was evaluated using Shapiro-Wilk test, and P value was calculated comparing infectious episodes pre and post immunotherapy with MV130 using Wilcoxon signed rank test.
Figure 3
Figure 3
MV130 cuts down on antibiotic and healthcare resource consumption. (A) Antibiotic consumption, (B) unscheduled medical visits, and (C) hospitalizations during the year before (Pre) and after (Post) the initiation of immunotherapy with MV130. Data from n=12 (A) n=15 (B, C) patients are displayed, Normal distribution was evaluated using Shapiro-Wilk test, and P values were calculated using Wilcoxon signed rank test. GP, general practitioner; ER, emergency room.
Figure 4
Figure 4
Prophylaxis with MV130 increases serum specific IgA antibody production. Serum IgA specific antibodies against (A) MV130 (bacterial mixture), (B) S. epidermidis, (C) S. aureus, (D) S. pneumoniae, (E) H. influenzae, (F) M. catarrhalis and (G) K. pneumoniae were quantified from patients before immunotherapy with MV130 (Pre), and 4 to 41 months after initiation of immunotherapy with MV130 (Post). Levels of antibodies were determined by ELISA. Sera samples from a total of 13 patients were included. Normal distribution was evaluated using Shapiro-Wilk test, and P values were calculated using Wilcoxon signed rank test.

References

    1. Jolles S, Chapel H, Litzman J. When to initiate immunoglobulin replacement therapy (IGRT) in antibody deficiency: a practical approach: When to initiate IgG therapy in antibody deficiency. Clin Exp Immunol (2017) 188:333–41. 10.1111/cei.12915
    1. Wingard JR, Hsu J, Hiemenz JW. Hematopoietic Stem Cell Transplantation: An Overview of Infection Risks and Epidemiology. Hematol/Oncol Clinics North America (2011) 25:101–16. 10.1016/j.hoc.2010.11.008
    1. Cho S-Y, Lee H-J, Lee D-G. Infectious complications after hematopoietic stem cell transplantation: current status and future perspectives in Korea. Korean J Internal Med (2018) 33:256–76. 10.3904/kjim.2018.036
    1. Prestinaci F, Pezzotti P, Pantosti A. Antimicrobial resistance: a global multifaceted phenomenon. Pathog Global Health (2015) 109:309–18. 10.1179/2047773215Y.0000000030
    1. Chokshi A, Sifri Z, Cennimo D, Horng H. Global contributors to antibiotic resistance. J Global Infect Dis (2019) 11:36. 10.4103/jgid.jgid_110_18
    1. Dinh A, Davido B, Duran C, Bouchand F, Bernard L. Antibiotic prophylaxis approaches for urinary tract infections. Lancet Infect Dis (2018) 18:1065. 10.1016/S1473-3099(18)30501-2
    1. Taplitz RA, Kennedy EB, Bow EJ, Crews J, Gleason C, Hawley DK, et al. . Antimicrobial Prophylaxis for Adult Patients With Cancer-Related Immunosuppression: ASCO and IDSA Clinical Practice Guideline Update. J Clin Oncol (2018) 36:3043–54. 10.1200/JCO.18.00374
    1. Munita JM, Arias CA. Mechanisms of Antibiotic Resistance. Microbiol Spectr (2016) 4:1–24. 10.1128/microbiolspec.VMBF-0016-2015
    1. Aslam B, Wang W, Arshad MI, Khurshid M, Muzammil S, Rasool MH, et al. . Antibiotic resistance: a rundown of a global crisis. Infect Drug Resist (2018) 11:1645–58. 10.2147/IDR.S173867
    1. O’Neill J. Antimicrobial resistance: tackling a crisis for the health and wealth of nations. Rev Antimicrob Resist (2016) 2–80.
    1. Sanchez-Ramon S, Diego R, Dieli-Crimi R, Subiza J-L. Extending the Clinical Horizons of Mucosal Bacterial Vaccines: Current Evidence and Future Prospects. Curr Drug Targets (2014) 15:1132–43. 10.2174/1389450115666141020160705
    1. Stevens WBC, Netea MG, Kater AP, van der Velden WJFM. ‘Trained immunity: consequences for lymphoid malignancies. Haematologica (2016) 101:1460–8. 10.3324/haematol.2016.149252
    1. Netea MG, Domínguez-Andrés J, Barreiro LB, Chavakis T, Divangahi M, Fuchs E, et al. . Defining trained immunity and its role in health and disease. Nat Rev Immunol (2020) 20:375–88. 10.1038/s41577-020-0285-6
    1. Mourits VP, Wijkmans JC, Joosten LA, Netea MG. Trained immunity as a novel therapeutic strategy. Curr Opin Pharmacol (2018) 41:52–8. 10.1016/j.coph.2018.04.007
    1. Sánchez-Ramón S, Conejero L, Netea MG, Sancho D, Palomares Ó, Subiza JL. Trained Immunity-Based Vaccines: A New Paradigm for the Development of Broad-Spectrum Anti-infectious Formulations. Front Immunol (2018) 9:1–11. 10.3389/fimmu.2018.02936
    1. Rozy A, Chorostowska-Wynimko J. Bacterial immunostimulants–mechanism of action and clinical application in respiratory diseases. Pneumonol Alergol Pol (2008) 76:353–9.
    1. De Benedetto F, Sevieri G. Prevention of respiratory tract infections with bacterial lysate OM-85 bronchomunal in children and adults: a state of the art. Multidiscip Respir Med (2013) 8:1–9. 10.1186/2049-6958-8-33
    1. Collet JP, Shapiro S, Ernst P, Renzi P, Ducruet T, Robinson A, et al. . Effects of an Immunostimulating Agent on Acute Exacerbations and Hospitalizations in Patients with Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med (1997) 156:1719–24. 10.1164/ajrccm.156.6.9612096
    1. Alecsandru D, Valor L, Sánchez-Ramón S, Gil J, Carbone J, Navarro J, et al. . Sublingual therapeutic immunization with a polyvalent bacterial preparation in patients with recurrent respiratory infections: immunomodulatory effect on antigen-specific memory CD4+ T cells and impact on clinical outcome: Sublingual immunization in patients with recurrent respiratory infections. Clin Exp Immunol (2011) 164:100–7. 10.1111/j.1365-2249.2011.04320.x
    1. Steurer-Stey C, Bachmann LM, Steurer J, Tramèr MR. Oral Purified Bacterial Extracts in Chronic Bronchitis and COPD. Chest (2004) 126:1645–55. 10.1378/chest.126.5.1645
    1. García González L-A, Arrutia Díez F. Mucosal bacterial immunotherapy with MV130 highly reduces the need of tonsillectomy in adults with recurrent tonsillitis. Hum Vaccines Immunother (2019) 15:2150–3. 10.1080/21645515.2019.1581537
    1. Conejero Hall L, Nieto García A, Brandi P, Nieto Cid M, Mazón Á, Cueto FJ, et al. . Bacterial immunotherapy in children with wheezing attacks: clinical impact and mechanism of action. Eur Respir J (2019) 54(Suppl.63):PA4998. 10.1183/13993003.congress-2019.PA4998
    1. Cirauqui C, Benito-Villalvilla C, Sánchez-Ramón S, Sirvent S, Diez-Rivero CM, Conejero L, et al. . Human dendritic cells activated with MV130 induce Th1, Th17 and IL-10 responses via RIPK2 and MyD88 signalling pathways. Eur J Immunol (2018) 48:180–93. 10.1002/eji.201747024
    1. Molero-Abraham M, Sanchez-Trincado JL, Gomez-Perosanz M, Torres-Gomez A, Subiza JL, Lafuente EM, et al. . Human Oral Epithelial Cells Impair Bacteria-Mediated Maturation of Dendritic Cells and Render T Cells Unresponsive to Stimulation. Front Immunol (2019) 10:1–14. 10.3389/fimmu.2019.01434
    1. Sánchez Ramón S, Manzanares M, Candelas G. Vacunas antiinfecciosas de mucosas en la profilaxis de infecciones recurrentes: más allá de las vacunas convencionales. Reumatología Clínica (2020) 16:49–55. 10.1016/j.reuma.2018.10.012
    1. Guevara-Hoyer K, Saz-Leal P, Diez-Rivero CM, Ochoa-Grullón J, Fernández-Arquero M, Pérez de Diego R, et al. . Trained Immunity Based-Vaccines as a Prophylactic Strategy in Common Variable Immunodeficiency. A Proof of Concept Study. Biomedicines (2020) 8:203. 10.3390/biomedicines8070203
    1. Tejera-Alhambra M, Palomares O, de Diego R, Diaz-Lezcano I, Sanchez-Ramon S. New Biological Insights in the Immunomodulatory Effects of Mucosal Polybacterial Vaccines in Clinical Practice. Curr Pharm Design (2016) 22:6283–93. 10.2174/1381612822666160829143129
    1. Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, et al. . The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood (2016) 127:2375–90. 10.1182/blood-2016-01-643569
    1. Tangcharoensathien V, Sattayawutthipong W, Kanjanapimai S, Kanpravidth W, Brown R, Sommanustweechai A. Antimicrobial resistance: from global agenda to national strategic plan, Thailand. Bull World Health Organ (2017) 95:599–603. 10.2471/BLT.16.179648
    1. Esposito S, Soto-Martinez ME, Feleszko W, Jones MH, Shen K-L, Schaad UB. Nonspecific immunomodulators for recurrent respiratory tract infections, wheezing and asthma in children: a systematic review of mechanistic and clinical evidence. Curr Opin Allergy Clin Immunol (2018) 18:198–209. 10.1097/ACI.0000000000000433
    1. Del-Rio-Navarro BE, Espinosa-Rosales FJ, Flenady V, Sienra-Monge JJ. Cochrane Review: Immunostimulants for preventing respiratory tract infection in children. Evidence-Based Child Health: A Cochrane Rev J (2012) 7:629–717. 10.1002/ebch.1833
    1. Moreno-Fierros L, García-Silva I, Rosales-Mendoza S. Development of SARS-CoV-2 vaccines: should we focus on mucosal immunity? Expert Opin Biol Ther (2020) 20:831–6. 10.1080/14712598.2020.1767062
    1. Clancy RL, Dunkley M. Acute Exacerbations in COPD and Their Control with Oral Immunization with Non-Typeable Haemophilus Influenzae. Front Immunol (2011) 2:1–6. 10.3389/fimmu.2011.00007
    1. Pasquali C, Salami O, Taneja M, Gollwitzer ES, Trompette A, Pattaroni C, et al. . Enhanced Mucosal Antibody Production and Protection against Respiratory Infections Following an Orally Administered Bacterial Extract. Front Med (2014) 1:1–9. 10.3389/fmed.2014.00041
    1. Cheng S-C, Quintin J, Cramer RA, Shepardson KM, Saeed S, Kumar V, et al. . mTOR- and HIF-1 -mediated aerobic glycolysis as metabolic basis for trained immunity. Science (2014) 345:1250684–1250684. 10.1126/science.1250684
    1. Bulut O, Kilic G, Domínguez-Andrés J, Netea MG. Overcoming immune dysfunction in the elderly: trained immunity as a novel approach. Int Immunol (2020) 32(12):741–53. 10.1093/intimm/dxaa052

Source: PubMed

3
Subscribe