Possible effects of sirolimus treatment on the long‑term efficacy of COVID‑19 vaccination in patients with β‑thalassemia: A theoretical perspective

  • Authors:
    • Matteo Zurlo
    • Francesco Nicoli
    • Monica Borgatti
    • Alessia Finotti
    • Roberto Gambari
  • View Affiliations

  • Published online on: January 19, 2022     https://doi.org/10.3892/ijmm.2022.5088
  • Article Number: 33
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The pandemic caused by the severe acute respiratory syndrome coronavirus (SARS‑CoV‑2), responsible for coronavirus disease 2019 (COVID‑19) has posed a major challenge for global health. In order to successfully combat SARS‑CoV‑2, the development of effective COVID‑19 vaccines is crucial. In this context, recent studies have highlighted a high COVID‑19 mortality rate in patients affected by β‑thalassemia, probably due to their co‑existent immune deficiencies. In addition to a role in the severity of SARS‑CoV‑2 infection and in the mortality rate of COVID‑19‑infected patients with thalassemia, immunosuppression is expected to deeply affect the effectivity of anti‑COVID‑19 vaccines. In the context of the interplay between thalassemia‑associated immunosuppression and the effectiveness of COVID‑19 vaccines, the employment of immunomodulatory molecules is hypothesized. For instance, short‑term treatment with mammalian target of rapamycin inhibitors (such as everolimus and sirolimus) has been found to improve responses to influenza vaccination in adults, with benefits possibly persisting for a year following treatment. Recently, sirolimus has been considered for the therapy of hemoglobinopathies (including β‑thalassemia). Sirolimus induces the expression of fetal hemoglobin (and this may contribute to the amelioration of the clinical parameters of patients with β‑thalassemia) and induces autophagy (thereby reducing the excessive levels of α‑globin). It may also finally contribute to the mobilization of erythroid cells from the bone marrow (thereby reducing anemia). In the present study, the authors present the hypothesis that sirolimus treatment, in addition to its beneficial effects on erythroid‑related parameters, may play a crucial role in sustaining the effects of COVID‑19 vaccination in patients with β‑thalassemia. This hypothesis is based on several publications demonstrating the effects of sirolimus treatment on the immune system.

Introduction

The dramatic pandemic caused by the severe acute respiratory syndrome coronavirus (SARS-CoV-2), responsible for coronavirus disease 2019 (COVID-19) has posed a major new challenge for human health worldwide (1,2). The rapidly increasing amounts of research on COVID-19 has allowed for the understanding of several aspects of the pathophysiology of SARS-CoV-2, including the key steps of infection, the hyper-inflammatory state (termed ‘cytokine storm’) leading to acute respiratory distress syndrome (ARDS), and the fact that severe forms of this disease are more frequently observed in elderly patients, particularly when associated with underlying comorbidities, such as hypertension, diabetes, obesity, ischemic heart disease (IHD) and chronic obstructive pulmonary disease (COPD) (2,3). Moreover, the mortality rate caused by COVID-19 has been found to increase exponentially with age, even considering the very high variability in the reported mortality rates by studies employed on very different testing strategies and therapeutic interventions (46).

Recent research has highlighted a high COVID-19 mortality rate in patients with β-thalassemia (7), probably due to co-existent immune deficiencies (8). Immune dysfunctions characterizing patients with thalassemia include changes in lymphocyte subsets, such as the accumulation of suppressor T-cells and the reduced proliferative capacity and numbers of T-helper cells, as well as the defective activity of natural killer (NK) cells. Similarly, an altered humoral immunity has been found in patients with β-thalassemia (9). Subjects presenting with similar immune system defects (i.e., the elderly) exhibit a marked susceptibility to severe COVID-19-related symptoms (46,10). This suggests that β-thalassemia-associated immunosuppression should be actively targeted to protect these patients.

In addition to a role in the severity of SARS-CoV-2 infection and in the mortality of the COVID-19-infected patients with thalassemia, immunosuppression is expected to greatly affect the effectiveness of anti-COVID-19 vaccines. This is a key issue, since it is widely accepted that the development of effective COVID-19 vaccination is crucial in order to successfully combat SARS-CoV-2 (1113). In this context, the need for prospective immunosurveillance studies in order to estimate the duration of immunity is of utmost importance and impact (1114). Effective and long-lasting COVID-19 vaccination will require interventions that generate potent humoral and cellular responses against SARS-CoV-2 antigens (12,13). In this respect, one of the unanswered issues regarding COVID-19 vaccination is the length of time this approach will protect the vaccinated population from infection by SARS-CoV-2 and from the development of severe COVID-19-associated symptoms (12,13).

In the context of the interplay between thalassemia-associated immunosuppression and the effectiveness of COVID-19 vaccines, the employment of immunomodulatory molecules has been considered. For instance, short-term treatment with mammalian target of rapamycin (mTOR) inhibitors (such as everolimus and sirolimus) has been found to improve responses to influenza vaccination in adults, with benefits possibly persisting for a year following treatment (15,16). Such drugs suppress excess inflammation, while also improving innate immunity.

Sirolimus has been considered for the therapy of hemoglobinopathies (for example β-thalassemia and sickle-cell disease) (1722). Two clinical trials based on the employment of sirolimus for thalassemia have been activated, NCT03877809 (A Personalized Medicine Approach for β-thalassemia Transfusion Dependent Patients: Testing Sirolimus in a First Pilot Clinical Trial) and NCT04247750 (Treatment of β-thalassemia Patients with Rapamycin: From Pre-clinical Research to a Clinical Trial), both using low doses of sirolimus for a 12-month period. The rationale of these two trials is that sirolimus may be of interest for use in β-thalassemia, since it induces the expression of fetal hemoglobin (and this may contribute to ameliorate the clinical parameters of these patients), induces autophagy (thereby reducing the excess of α-globin) and, finally, may contribute to mobilization of erythroid cell from the bone marrow (thereby reducing anemia). In addition to these positive effects on the hematopoietic system, sirolimus may improve the immune system of these patients. This may be a crucial issue, particularly considering that the majority of patients with β-thalassemia who are currently being vaccinated against COVID-19 are in the 45–60 age category. Moreover, it should be underlined that sirolimus and sirolimus analogs (such as everolimus) are extensively used in routine therapy and in clinical studies for the treatment of other diseases, such as renal, cardiac and liver transplantation (2326), systemic lupus erythematosus (27), lymphangioleiomyomatosis (28), tuberous sclerosis complex (29), recurrent meningioma (30), pancreatic neuroendocrine tumors (31), advanced differentiated thyroid cancers (32), advanced breast cancer (33), diffuse large B-cell lymphomas (34), metastatic renal cell carcinoma (35).

The hypothesis

The working hypothesis is that use of sirolimus can sustain the effectiveness of COVID-19 vaccination in patients with β-thalassemia (Fig. 1) (36). This hypothesis is based on several publications demonstrating the effects of sirolimus on the immune system, as well as on the growing interest for immunomodulators functioning through metabolic manipulation (37). For instance, Amiel et al (38) demonstrated that sirolimus promoted dendritic cell (DC) activation and enhanced therapeutic autologous vaccination in mice. These findings define mTOR as a molecular target for augmenting DC survival and activation, and document a novel pharmacologic approach for enhancing the efficacy of therapeutic autologous DC vaccination. In addition, Araki et al (39) proposed that sirolimus improved both the quantity and quality of memory CD8+ T-cells induced by viral infection and vaccination, demonstrating that mTOR is also a major regulator of memory CD8+ T-cell differentiation. These discoveries have implications for the development of novel vaccine regimens, and sirolimus can thus have potential for use in improving vaccine efficacy. Notably, the timing of treatment may be of utmost importance. Indeed, mTOR activity is required for B- and T-cell priming, thus arguing against a concomitant use of sirolimus together with vaccination (40,41). Conversely, mTOR inhibition may be crucial for the maintenance of memory lymphocytes (39,42), thus potentially prolonging vaccine immunogenicity. It was thus hypothesized that sirolimus may be tested for possible administration during the early memory phases; i.e., 30–60 days following vaccination.

The information on the possible effects of sirolimus on vaccines is also of great interest considering that sirolimus is extensively used in routine therapy and in clinical studies for other diseases. In addition, the campaign for COVID-19 vaccination is ongoing and will include patients presently being treated with sirolimus.

This hypothesis is sustained by preliminary results obtained in the concluded NCT03877809 trial, indicating that treatment with sirolimus did not lead to a major alteration of the immunophenotype. In particular, the in vivo treatment of patients with β-thalassemia with a daily administration of 0.5 mg of sirolimus does not affect the CD8+ T-lymphocyte population over the period of 180 days of therapy. This conclusion was achieved by flow cytometric analysis of peripheral blood mononuclear cells (PBMCs) aimed at assessing the preservation of different immune cell subsets (B-cells, regulatory and conventional CD4+ T-cells, CD8+ T-cells and monocytes) (unpublished data).

Possible experimental evaluation of the hypothesis

In order to verify whether sirolimus treatment affects COVID-19 vaccine immune memory maintenance, two study groups are required, both undergoing COVID-19 vaccination: The first vaccinated cohort can be treated daily with 0.5-2 mg sirolimus for 6 months, that will generate a blood concentration ranging between 2-2.5 ng/ml in these patients. The second (control) vaccinated cohort will not be treated with sirolimus. Blood sampling for PBMCs and plasma isolation can be performed in both cohorts, at 1–2 months after the second dose of the vaccine and before commencing treatment with sirolimus; the subsequent samplings will be performed after 90, 180 and 360 days.

Concerning general/non-specific sirolimus-mediated effects on immune-cell subsets, longitudinal immunophenotypic analyses of PBMCs are necessary to assess any eventual fluctuation in the proportion of myeloid and lymphoid cells as well as of specific T- and B-lymphocyte subsets (naive, memory and suppressor cells). This will allow the study of possible modulations of immune cells in the cohort of patients being treated with sirolimus after vaccination compared to those who are only vaccinated, with particular interest in some subpopulations that may be altered in patients with thalassemia and/or be positively modulated by sirolimus (for example NK cells, DCs and memory CD8+ T-lymphocytes).

To assess whether sirolimus improves vaccine-specific memory cellular immunity, the frequency of SARS-CoV-2-specific CD4+ and CD8+ memory T-cells should be measured at different time points. In this respect, several approaches have been envisaged, spanning from the use of tetramers [for defined human leukocyte antigen (HLA)-haplotypes] to the use of pools of overlapping peptides covering different HLA alleles (43,44) or of specific epitopes restricted for specific HLAs (45). As it may be speculated that sirolimus may exert effects on all the memory T-cell compartments, the assessment of recall responses towards other previous encountered antigens could also be performed, e.g., through the use of HLA class I- and II-presented peptide pools containing various antigenic stimuli (Epstein-Barr virus, cytomegalovirus, tetanus and flu). This approach could help to elucidate whether sirolimus limits the normal decline in function that is expected in the passage of time.

To evaluate whether the intake of sirolimus improves vaccine-specific memory humoral immunity, the titers of binding and/or neutralizing specific antibodies against the SARS-CoV-2 spike protein induced by vaccination should be measured longitudinally. This analysis, in addition to verifying the immunomodulatory effect of sirolimus, may provide an estimate of how effective COVID-19 vaccines are over time.

Patients with thalassemia exhibit several immune alterations which may severely affect vaccine effectiveness or in general, the susceptibly to antigenic challenges. Thus, beyond the effects of sirolimus on COVID-19 vaccine responsiveness, its use may be envisaged to restore these immunological abnormalities. For instance, DC maturation is reduced in patients with thalassemia. Therefore, its induction from monocytes in patients before and after treatment could be assessed. In addition, patients with thalassemia present with an increased number and functionality of suppressor T-cells and reduced T-helper cell proliferation. Thus, taking advantage of the proposed research, it can be eventually estimated whether sirolimus can restore the balance and functionality within the T-cell subsets.

Additionally, to ensure the safety profile of sirolimus treatment and since the patient's inflammatory state may influence the vaccine response (46), a qualitative-quantitative analysis of the cytokines involved in adaptive immunity and inflammation in the plasma of patients treated or not with sirolimus may be appropriate.

Conclusions and future perspectives

The expected outcomes of the research activity finalized to examine the effects of in vivo treatment with sirolimus in patients with β-thalassemia vaccinated against SARS-CoV-2 are several and crucial. A first point is related to the detection of SARS-CoV-2-specific IgG in the plasma of treated patients and the evaluation of the effects of sirolimus. A second point is the quantification and determinations of the biological activity of SARS-CoV-2-specific memory T-cells in patients with β-thalassemia vaccinated against COVID-19. This point is crucial since molecules potentiating vaccination may be of great interest in a pandemic period in which the administration of a third dose of anti-SARS-CoV-2 vaccines is ongoing in several countries. In this respect, the overall effects of sirolimus treatment on the immune defects of patients with β-thalassemia is a key factor for determining whether this treatment may be proposed.

It should be noted that the impact of this research activity is not limited the sirolimus-treated patients with β-thalassemia. In fact, considering that mTOR inhibitors, such as sirolimus are employed in the treatment of a large variety of pathologies (2336), the number of patients taking advantages in the case this hypothesis will be confirmed, is relevant. Moreover, it can be envisaged that sirolimus treatment could be used to improve COVID-19 vaccine responses in other populations as well, such as the elderly where similar approaches have already been shown to improve the efficacy of the flu vaccine (47,48). On the other hand, it should be emphasized that the importance of testing geroprotective drugs (such as sirolimus) will extend far beyond the COVID-19 pandemic to improve overall health resilience of aged populations (46,49,50).

Finally, mTOR inhibitors (such as sirolimus and metformin) are at present undergoing clinical trials as anti-COVID-19 drugs (NCT04461340 and NCT04510194) (51), also considering their effects on activation of autophagy, that is deeply down-regulated during SARS-CoV-2 infection (52). In consideration of the multiple biochemical and cellular effects of sirolimus against SARS-CoV2, it can be considered as a repurposed drug for anti-COVID-19 therapy (53).

In conclusion, the proposed approaches may lead to the development of protocols for sustaining the effects of COVID-19 vaccines in fragile subjects. This is a major issue in the management of patients with COVID-19 in the future and in planning mass immunization strategies finalized in reaching the herd immunity in short period of time.

Acknowledgements

The authors would like to thank Dr Maria Rita Gamberini (Day Hospital Thalassemia, Arcispedale S. Anna, Ferrara) for her support and helpful discussions.

Funding

The present study was supported by the Wellcome Trust (United Kingdom, Innovator Award 208872/Z/17/Z), by AIFA (Agenzia Italiana del Farmaco, Italy, AIFA-2016-02364887) and by the MUR-FISR COVID-miRNAPNA Project (FISR2020IP_04128).

Availability of data and materials

Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.

Authors' contributions

MZ, RG and FN were involved in the conceptualization of the study. AF, MB and RG were involved in the writing and preparation of the original draft, and in the writing, reviewing and editing of the study. MZ and FN were involved in the processing of the figure. RG and AF supervised the study. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

COVID-19

coronavirus disease 2019

SARS-CoV-2

severe acute respiratory syndrome coronavirus 2

mTOR

mammalian target of rapamycin

ARDS

acute respiratory distress syndrome

IHD

ischemic heart disease

COPD

chronic obstructive pulmonary disease

NK cells

natural killer cells

DC

dendritic cell

PBMC

peripheral blood mononuclear cell

HLA

human leukocyte antigen

APC

antigen-presenting cell

References

1 

Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, et al: Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 395:497–506. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, Wang T, Zhang X, Chen H, Yu H, et al: Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 130:2620–2629. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Mahmudpour M, Roozbeh J, Keshavarz M, Farrokhi S and Nabipour I: COVID-19 cytokine storm: The anger of inflammation. Cytokine. 133:1551512020. View Article : Google Scholar : PubMed/NCBI

4 

Nicoli F, Solis-Soto MT, Paudel D, Marconi P, Gavioli R, Appay V and Caputo A: Age-related decline of de novo T cell responsiveness as a cause of COVID-19 severity. Geroscience. 42:1015–1019. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Nikolich-Zugich J, Knox KS, Rios CT, Natt B, Bhattacharya D and Fain MJ: SARS-CoV-2 and COVID-19 in older adults: What we may expect regarding pathogenesis, immune responses, and outcomes. Geroscience. 42:505–514. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Genebat M, Tarancón-Díez L, de Pablo-Bernal R, Calderón A, Muñoz-Fernández MÁ and Leal M: Coronavirus disease (COVID-19): A Perspective from Immunosenescence. Aging Dis. 12:3–6. 2021. View Article : Google Scholar : PubMed/NCBI

7 

Zafari M, Rad MTS, Mohseni F and Nikbakht N: β-Thalassemia Major and Coronavirus-19, mortality and morbidity: A systematic review study. Hemoglobin. 45:1–4. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Farmakis D, Giakoumis A, Polymeropoulos E and Aessopos A: Pathogenic aspects of immune deficiency associated with beta-thalassemia. Med Sci Monit. 9:RA19–RA22. 2003.PubMed/NCBI

9 

Ghaffari J, Abediankenari S and Nasehi M: Thalassemia and immune system dysfunction-review article. Int J Curr Res. 3:105–108. 2011.

10 

Blagosklonny MV: From causes of aging to death from COVID-19. Aging (Albany NY). 12:10004–10021. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Kaur SP and Gupta V: COVID-19 vaccine: A comprehensive status report. Virus Res. 288:1981142020. View Article : Google Scholar : PubMed/NCBI

12 

Chakraborty C, Sharma AR, Bhattacharya M, Sharma G, Saha RP and Lee SS: Ongoing clinical trials of vaccines to fight against COVID-19 pandemic. Immune Netw. 21:e52021. View Article : Google Scholar : PubMed/NCBI

13 

Dorrington MG and Bowdish DM: Immunosenescence and novel vaccination strategies for the elderly. Front Immunol. 4:1712013. View Article : Google Scholar : PubMed/NCBI

14 

Tsatsakis A, Vakonaki E, Tzatzarakis M, Flamourakis M, Nikolouzakis TK, Poulas K, Papazoglou G, Hatzidaki E, Papanikolaou NC, Drakoulis N, et al: Immune response (IgG) following full inoculation with BNT162b2 COVID-19 mRNA among healthcare professionals. Int J Mol Med. 48:2002021. View Article : Google Scholar : PubMed/NCBI

15 

Mannick JB, Del Giudice G, Lattanzi M, Valiante NM, Praestgaard J, Huang B, Lonetto MA, Maecker HT, Kovarik J, Carson S, et al: mTOR inhibition improves immune function in the elderly. Sci Transl Med. 6:268ra1792014. View Article : Google Scholar : PubMed/NCBI

16 

Mannick JB, Morris M, Hockey HP, Roma G, Beibel M, Kulmatycki K, Watkins M, Shavlakadze T, Zhou W, Quinn D, et al: TORC1 inhibition enhances immune function and reduces infections in the elderly. Sci Transl Med. 10:eaaq15642018. View Article : Google Scholar : PubMed/NCBI

17 

Mischiati C, Sereni A, Lampronti I, Bianchi N, Borgatti M, Prus E, Fibach E and Gambari R: Rapamycin-mediated induction of gamma-globin mRNA accumulation in human erythroid cells. Br J Haematol. 126:612–621. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Fibach E, Bianchi N, Borgatti M, Zuccato C, Finotti A, Lampronti I, Prus E, Mischiati C and Gambari R: Effects of rapamycin on accumulation of alpha-, beta- and gamma-globin mRNAs in erythroid precursor cells from beta-thalassaemia patients. Eur J Haematol. 77:437–441. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Zuccato C, Bianchi N, Borgatti M, Lampronti I, Massei F, Favre C and Gambari R: Everolimus is a potent inducer of erythroid differentiation and gamma-globin gene expression in human erythroid cells. Acta Haematol. 117:168–176. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Pecoraro A, Troia A, Calzolari R, Scazzone C, Rigano P, Martorana A, Sacco M, Maggio A and Di Marzo R: Efficacy of rapamycin as inducer of HbF in primary erythroid cultures from sickle cell disease and beta-thalassemia patients. Hemoglobin. 39:225–229. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Gaudre N, Cougoul P, Bartolucci P, Dörr G, Bura-Riviere A, Kamar N and Del Bello A: Improved fetal hemoglobin with mTOR inhibitor-based immunosuppression in a kidney transplant recipient with sickle cell disease. Am J Transplant. 17:2212–2214. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Al-Khatti AA and Alkhunaizi AM: Additive effect of Sirolimus and hydroxycarbamide on fetal haemoglobin level in kidney transplant patients with sickle cell disease. Br J Haematol. 185:959–961. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Kahan BD: Sirolimus: A new agent for clinical renal transplantation. Transplant Proc. 29:48–50. 1997. View Article : Google Scholar : PubMed/NCBI

24 

Hernández D, Martínez D, Gutiérrez E, López V, Gutiérrez C, García P, Cobelo C, Cabello M, Burgos D, Sola E and González-Molina M: Clinical evidence on the use of anti-mTOR drugs in renal transplantation. Nefrologia. 31:27–34. 2011.PubMed/NCBI

25 

Schaffer SA and Ross HJ: Everolimus: Efficacy and safety in cardiac transplantation. Expert Opin Drug Saf. 9:843–854. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Tang CY, Shen A, Wei XF, Li QD, Liu R, Deng HJ, Wu YZ and Wu ZJ: Everolimus in de novo liver transplant recipients: A systematic review. Hepatobiliary Pancreat Dis Int. 14:461–469. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Ji L, Xie W and Zhang Z: Efficacy and safety of Sirolimus in patients with systemic lupus erythematosus: A systematic review and meta-analysis. Semin Arthritis Rheum. 50:1073–1080. 2020. View Article : Google Scholar : PubMed/NCBI

28 

Wang Q, Luo M, Xiang B, Chen S and Ji Y: The efficacy and safety of pharmacological treatments for lymphangioleiomyomatosis. Respir Res. 21:552020. View Article : Google Scholar : PubMed/NCBI

29 

Sasongko TH, Ismail NF and Zabidi-Hussin Z: Rapamycin and rapalogs for tuberous sclerosis complex. Cochrane Database Syst Rev. 7:CD0112722016.PubMed/NCBI

30 

Graillon T, Sanson M, Campello C, Idbaih A, Peyre M, Peyrière H, Basset N, Autran D, Roche C, Kalamarides M, et al: Everolimus and octreotide for patients with recurrent meningioma: Results from the phase II CEVOREM trial. Clin Cancer Res. 26:552–557. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Gallo M, Malandrino P, Fanciulli G, Rota F, Faggiano A and Colao A; NIKE Group, : Everolimus as first line therapy for pancreatic neuroendocrine tumours: Current knowledge and future perspectives. J Cancer Res Clin Oncol. 143:1209–1224. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Manohar PM, Beesley LJ, Taylor JM, Hesseltine E, Haymart MR, Esfandiari NH, Hanauer DA and Worden FP: Retrospective study of sirolimus and cyclophosphamide in patients with advanced differentiated thyroid cancers. J Thyroid Disord Ther. 4:1882015. View Article : Google Scholar : PubMed/NCBI

33 

Hortobagyi GN: Everolimus plus exemestane for the treatment of advanced breast cancer: A review of subanalyses from BOLERO-2. Neoplasia. 17:279–288. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Merli M, Ferrario A, Maffioli M, Arcaini L and Passamonti F: Everolimus in diffuse large B-cell lymphomas. Future Oncol. 11:373–383. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grünwald V, Thompson JA, Figlin RA, Hollaender N, et al: Phase 3 trial of Everolimus for metastatic renal cell carcinoma: Final results and analysis of prognostic factors. Cancer. 116:4256–4265. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Kaech SM and Cui W: Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol. 12:749–761. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Nicoli F, Paul S and Appay V: Harnessing the induction of CD8+T-cell responses through metabolic regulation by pathogen-recognition-receptor triggering in antigen presenting cells. Front Immunol. 9:23722018. View Article : Google Scholar : PubMed/NCBI

38 

Amiel E, Everts B, Freitas TC, King IL, Curtis JD, Pearce EL and Pearce EJ: Inhibition of mechanistic target of Rapamycin promotes dendritic cell activation and enhances therapeutic autologous vaccination in mice. J Immunol. 189:2151–2158. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Araki K, Youngblood B and Ahmed R: The role of mTOR in memory CD8 T-cell differentiation. Immunol Rev. 235:234–243. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Nicoli F, Papagno L, Frere JJ, Cabral-Piccin MP, Clave E, Gostick E, Toubert A, Price DA, Caputo A and Appay V: Naïve CD8+ T-cells engage a versatile metabolic program upon activation in humans and differ energetically from memory CD8+ T-Cells. Front Immunol. 9:27362018. View Article : Google Scholar : PubMed/NCBI

41 

Zhang S, Pruitt M, Tran D, Du Bois W, Zhang K, Patel R, Hoover S, Simpson RM, Simmons J, Gary J, et al: B cell-specific deficiencies in mTOR limit humoral immune responses. J Immunol. 191:1692–1703. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Nicoli F: Angry, Hungry T-Cells: How Are T-Cell responses induced in low nutrient conditions? Immunometabolism. 2:e2000042020.

43 

Grifoni A, Weiskopf D, Ramirez SI, Mateus J, Dan JM, Moderbacher CR, Rawlings SA, Sutherland A, Premkumar L, Jadi RS, et al: Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. 181:1489–1501, e15. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Schulien I, Kemming J, Oberhardt V, Wild K, Seidel LM, Sagar Killmer S, Daul F, Salvat Lago M, Decker A, et al: Characterization of pre-existing and induced SARS-CoV-2-specific CD8+ T cells. Nat Med. 27:78–85. 2021. View Article : Google Scholar : PubMed/NCBI

45 

Gallerani E, Proietto D, Dallan B, Campagnaro M, Pacifico S, Albanese V, Marzola E, Marconi P, Caputo A, Appay V, et al: Impaired Priming of SARS-CoV-2-specific naive CD8+ T cells in older subjects. Front Immunol. 12:6930542021. View Article : Google Scholar : PubMed/NCBI

46 

Frasca D and Blomberg BB: Inflammaging decreases adaptive and innate immune responses in mice and humans. Biogerontology. 17:7–19. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Keating R, Hertz T, Wehenkel M, Harris TL, Edwards BA, McClaren JL, Brown SA, Surman S, Wilson ZS, Bradley P, et al: The kinase mTOR modulates the antibody response to provide cross-protective immunity to lethal infection with influenza virus. Nat Immunol. 14:1266–1276. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Cohen J: Infectious disease. Immune suppressant unexpectedly boosts flu vaccine. Science. 342:4132013. View Article : Google Scholar : PubMed/NCBI

49 

Blasi F, Gramegna A, Sotgiu G, Saderi L, Voza A, Aliberti S and Amati F: SARS-CoV-2 vaccines: A critical perspective through efficacy data and barriers to herd immunity. Respir Med. 180:1063552021. View Article : Google Scholar : PubMed/NCBI

50 

Cunningham AL, McIntyre P, Subbarao K, Booy R and Levin MJ: Vaccines for older adults. BMJ. 372:n1882021. View Article : Google Scholar : PubMed/NCBI

51 

Nitulescu GM, Paunescu H, Moschos SA, Petrakis D, Nitulescu G, Ion GND, Spandidos DA, Nikolouzakis TK, Drakoulis N and Tsatsakis A: Comprehensive analysis of drugs to treat SARS-CoV-2 infection: Mechanistic insights into current COVID-19 therapies (Review). Int J Mol Med. 46:467–488. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Gassen NC, Papies J, Bajaj T, Emanuel J, Dethloff F, Chua RL, Trimpert J, Heinemann N, Niemeyer C, Weege F, et al: SARS-CoV-2-mediated dysregulation of metabolism and autophagy uncovers host-targeting antivirals. Nat Commun. 12:38182021. View Article : Google Scholar : PubMed/NCBI

53 

Patocka J, Kuca K, Oleksak P, Nepovimova E, Valis M, Novotny M and Klimova B: Rapamycin: Drug repurposing in SARS-CoV-2 infection. Pharmaceuticals (Basel). 14:2172021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2022
Volume 49 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zurlo M, Nicoli F, Borgatti M, Finotti A and Gambari R: Possible effects of sirolimus treatment on the long‑term efficacy of COVID‑19 vaccination in patients with β‑thalassemia: A theoretical perspective. Int J Mol Med 49: 33, 2022
APA
Zurlo, M., Nicoli, F., Borgatti, M., Finotti, A., & Gambari, R. (2022). Possible effects of sirolimus treatment on the long‑term efficacy of COVID‑19 vaccination in patients with β‑thalassemia: A theoretical perspective. International Journal of Molecular Medicine, 49, 33. https://doi.org/10.3892/ijmm.2022.5088
MLA
Zurlo, M., Nicoli, F., Borgatti, M., Finotti, A., Gambari, R."Possible effects of sirolimus treatment on the long‑term efficacy of COVID‑19 vaccination in patients with β‑thalassemia: A theoretical perspective". International Journal of Molecular Medicine 49.3 (2022): 33.
Chicago
Zurlo, M., Nicoli, F., Borgatti, M., Finotti, A., Gambari, R."Possible effects of sirolimus treatment on the long‑term efficacy of COVID‑19 vaccination in patients with β‑thalassemia: A theoretical perspective". International Journal of Molecular Medicine 49, no. 3 (2022): 33. https://doi.org/10.3892/ijmm.2022.5088