J Hematol
Journal of Hematology, ISSN 1927-1212 print, 1927-1220 online, Open Access
Article copyright, the authors; Journal compilation copyright, J Hematol and Elmer Press Inc
Journal website https://www.thejh.org

Short Communication

Volume 12, Number 4, August 2023, pages 170-175


Local and Systemic Immunity During Five Vaccinations Against SARS-CoV-2 in Zanubrutinib-Treated Patients With Chronic Lymphocytic Leukemia

Maria Anderssona, b, i, Jinghua Wuc, i, David Wullimannc, Yu Gaoc, Mikael Abergd, Sandra Muschiole, f, Katie Healye, Sabrina Naudg, Gordana Bogdanovice, f, Marzia Palmaa, b, Hakan Mellstedta, Puran Chenc, Hans-Gustaf Ljunggrenc, Lotta Hanssona, b, Margaret Sallberg Cheng, Marcus Buggertc, Hanna M. Ingelman-Sundberga, h, i, j, Anders Osterborga, b, i

aDepartment of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
bDepartment of Hematology, Karolinska University Hospital Solna, Stockholm, Sweden
cCenter for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
dDepartment of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
eDepartment of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
fDepartment of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
gDepartment of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
hDepartment of Oncology, Karolinska University Hospital Solna, Stockholm, Sweden
iThese authors contributed equally to this article.
jCorresponding Author: Hanna Muren Ingelman-Sundberg, Department of Oncology, Karolinska University Hospital Solna, SE-171 76 Stockholm, Sweden

Manuscript submitted May 14, 2023, accepted July 14, 2023, published online August 8, 2023
Short title: COVID-19 Vaccine and Zanubrutinib Treatment
doi: https://doi.org/10.14740/jh1140

Abstract▴Top 

Background: Patients with chronic lymphocytic leukemia (CLL) are vulnerable to coronavirus disease 2019 (COVID-19) and are at risk of inferior response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination, especially if treated with the first-generation Bruton’s tyrosine kinase inhibitor (BTKi) ibrutinib. We aimed to evaluate the impact of the third-generation BTKi, zanubrutinib, on systemic and mucosal response to SARS-CoV-2 vaccination.

Methods: Nine patients with CLL with ongoing zanubrutinib therapy were included and donated blood and saliva during SARS-CoV-2 vaccination, before vaccine doses 3 and 5 and 2 - 3 weeks after doses 3, 4, and 5. Ibrutinib-treated control patients (n = 7) and healthy aged-matched controls (n = 7) gave blood 2 - 3 weeks after vaccine dose 5. We quantified reactivity and neutralization capacity of SARS-CoV-2-specific IgG and IgA antibodies (Abs) in both serum and saliva, and reactivity of T cells activated with viral peptides.

Results: Both zanubrutinib- and ibrutinib-treated patients had significantly, up to 1,000-fold, lower total spike-specific Ab levels after dose 5 compared to healthy controls (P < 0.01). Spike-IgG levels in serum from zanubrutinib-treated patients correlated well to neutralization capacity (r = 0.68; P < 0.0001) and were thus functional. Mucosal immunity (specific IgA in serum and saliva) was practically absent in zanubrutinib-treated patients even after five vaccine doses, whereas healthy controls had significantly higher levels (tested in serum after vaccine dose 5) (P < 0.05). In contrast, T-cell reactivity against SARS-CoV-2 peptides was equally high in zanubrutinib- and ibrutinib-treated patients as in healthy control donors.

Conclusions: In our small cohort of zanubrutinib-treated CLL patients, we conclude that up to five doses of SARS-CoV-2 vaccination induced no detectable IgA mucosal immunity, which likely will impair the primary barrier defence against the infection. Systemic IgG responses were also impaired, whereas T-cell responses were normal. Further and larger studies are needed to evaluate the impact of these findings on disease protection.

Keywords: CLL; Zanubrutinib; SARS-CoV-2; Immunity; Vaccination

Introduction▴Top 

Immunocompromised patients with chronic lymphocytic leukemia (CLL) are vulnerable to coronavirus disease 2019 (COVID-19) and exhibit variable responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccination [1, 2]. Antibody (Ab) titers are notably low in patients receiving the first-generation Bruton’s tyrosine kinase inhibitor (BTKi) ibrutinib [2, 3], and their vaccine-induced T-cell responses are weaker than in most other patient groups with immunodeficiency [4]. Zanubrutinib, a third-generation BTKi, offers improved progression-free survival compared to ibrutinib [5] and possesses increased selectivity to BTK with fewer off-target effects [6], including on interleukin-2-inducible T-cell kinase (ITK). This suggests that zanubrutinib’s immunological imprint on the outcome of vaccination might differ from earlier generations of BTKi. We here examined systemic and mucosal immunity during repeated (up to 5) SARS-CoV-2 vaccinations in a cohort of patients with CLL on long-term zanubrutinib therapy. Results were compared with patients on first-generation BTKi therapy (ibrutinib) and healthy age-matched donors that were tested after vaccine dose 5.

Materials and Methods▴Top 

Patients with CLL receiving zanubrutinib in the clinical trials BGB-3111-304 [7] and BGB-3111-305 [5] at one single site (Karolinska University Hospital) without a history of previous COVID-19 tested negative for anti-nucleocapsid Abs, who had received at least two vaccine doses against SARS-CoV-2, were included. Patients who developed COVID-19 during the study period were withdrawn from further analyses from that time point. The study was approved by the Swedish Ethical Review Authority [8]. The study was conducted in compliance with the ethical standards of the responsible institution on human subjects as well as with the Helsinki Declaration.

Zanubrutinib-treated patients donated blood and saliva samples before vaccine doses 3 and 5 and 2 - 3 weeks after doses 3, 4, and 5. Ibrutinib-treated control patients (n = 7) (median age 67 years, range 67 - 82) and healthy aged-matched controls (n = 7) (median age 71 years, range 64 - 88) gave blood after dose 5. Quantitative measurements of antibodies against the spike-receptor-binding domain (RBD) and the nucleocapsid were performed on serum samples (Elecsys®, Roche Diagnostics), and further analyses of anti-spike IgA, IgG, and ACE inhibition capacity were performed on both serum and saliva samples (V-PLEX, Meso Scale Diagnostics). Peripheral blood mononuclear cells (PBMCs) were used for T-cell response analysis using an activation-induced marker (AIM) assay as described [4]. A complete description of all methods, mainly as described earlier [9], is provided in the Supplementary Material 1 (www.thejh.org).

Results▴Top 

Patient characteristics and vaccination details are shown in Table 1. Nine zanubrutinib-treated patients with a median age of 75 years (range 52 - 83) were included. Six received zanubrutinib as first-line treatment and three as salvage therapy > 2.5 years since their last treatment. The median time from zanubrutinib initiation to the first vaccination was 28 months (range 4 - 34). All had stable partial remission during the vaccination period. One patient experienced a SARS-CoV-2 breakthrough infection before dose 4 and 1 before dose 5 and was subsequently excluded from further analyses.

Table 1.
Click to view
Table 1. Clinical Characteristics at Start of Vaccination Against SARS-CoV-2 and Timepoints of Vaccination and Tests
 

All zanubrutinib-treated patients (9/9) were seronegative prior to dose 3 (total spike-Ab < 0.8 U/mL). When compared to baseline, significant increases of total titers were noted after vaccine doses 3, 4, and 5 (P < 0.05), albeit 44% (4/9) were still seronegative after dose 3, 50% (4/8) after dose 4 and 43% (3/7) after dose 5 (Fig. 1a). Similar results were found for serum tested against both anti-wt- (Wuhan ancestral strain) and anti-Omicron spike IgG (data not shown). Total Ab levels after dose 5 were significantly lower in the zanubrutinib cohort than in age-matched healthy donors (median 2.7 U/mL vs. 21,648 U/mL; P < 0.01; Fig. 1b), with comparable results for anti-spike IgG against wt and Omicron (P < 0.01 and P < 0.05). Also, the anti-spike total Ab levels in the ibrutinib-treated cohort were significantly lower compared to healthy donors (median 2,148 U/mL; range 0 - 3,331; Fig. 1b). Two zanubrutinib-treated patients mounted Ab levels in parity with the healthy controls, but no individual factors (for example, age, gender, duration of zanubrutinib treatment, or type of vaccine) that might explain this were deviant compared to the other patients (data not shown).


Click for large image
Figure 1. Serological immunity in zanubrutinib- and ibrutinib-treated patients with CLL after SARS-CoV-2 vaccination. Levels of SARS-CoV-2 Abs against spike-receptor-binding domain (RBD) were quantified in serum samples from zanubrutinib treated patients pre- and post-vaccine dose 3, post-dose 4, and pre- and post-dose 5 (a), and in addition post-dose 5 in PBMC from seven ibrutinib-treated patients and seven age- and gender-matched healthy donors (b). The median level of reactivity and number of samples are indicated above each time point. Dashed line represents positive threshold of 0.8 U/mL and the upper limit of detection is 25,000 U/mL. Error bars represent the median (red line) and interquartile range where applicable. Non-parametric Kruskal-Wallis test was used to assess differences between all-time points and Mann-Whitney test for comparison between patient groups and healthy controls. CLL: chronic lymphocytic leukemia; PBMC: peripheral blood mononuclear cell.

The serum inhibition of angiotensin-converting enzyme 2 (ACE-2) binding to wt spike strongly correlated with the level of wt spike-specific IgG for all tested sera (r = 0.82; P < 0.0001) and for sera from zanubrutinib-treated patients when tested separately (r = 0.68; P < 0.0001). Saliva analysis showed a low but significant increase of IgG against wt spike in zanubrutinib-treated patients (P < 0.05), which strongly correlated with spike-specific serum IgG (r = 0.77; P < 0.0001). In contrast to IgG, anti-spike IgA levels remained unchanged in serum or saliva in zanubrutinib-treated patients (data not shown). However, healthy donors tested after dose 5 exhibited elevated serum IgA levels (saliva was not available) against both wt- and Omicron spike compared to patients (both P < 0.05).

Additionally, we measured SARS-CoV-2-specific T-cell responses. There was no significant change in wt- or Omicron-specific CD4+ T-cell frequencies after three or more vaccine doses (Fig. 2a), but a significant increase of Omicron-specific CD8+ T cells after dose 3 (P < 0.05), with a similar trend for wt-specific CD8+ T cells (P = 0.050, Fig. 2b). Doses 4 and 5 did not result in additional increases. In contrast to the serology results, spike-specific T-cell responses after vaccine dose 5 were similar between zanubrutinib-treated patients and healthy controls for both CD4+ (Fig. 2c) and CD8+ (Fig. 2d) T cells, with comparable levels observed in the ibrutinib cohort (Fig. 2c, d).


Click for large image
Figure 2. SARS-CoV-2 specific T-cell responses in zanubrutinib- and ibrutinib-treated patients with CLL after SARS-CoV-2 vaccination. Antigen specific CD4+ (CD69+CD154+) and CD8+ (CD69+CD137+) memory T cells were measured after SARS-CoV-2 wild-type and Omicron BA.1 peptide stimulation in PBMC samples from zanubrutinib-treated patients pre- and post-vaccine dose 3, post-dose 4, and pre- and post-dose 5 (a, b), and in addition post-dose 5 in PBMC from seven ibrutinib-treated patients and seven age- and gender-matched healthy donors (c, d). The median level of reactivity and number of samples are indicated above each time point. Omicron-specific CD8+ T cells increased significantly (P < 0.05) after dose 3, and there was a similar trend for wt-specific CD8+ T cells (P = 0.050). No significant differences were noticed between the different groups of patients, or between patients and healthy controls, post-dose 5. Background is subtracted using DMSO negative control of AIM+ CD4+/CD8+ among all patients. A positive response was defined with a cut-off level of 0.05% (dashed lines). Error bars represent the median (red line) and interquartile range where applicable. Non-parametric Kruskal-Wallis test was used to assess differences between all-time points and Mann-Whitney test for comparison between CLL patients and healthy controls. ns: P > 0.05, not statistically significant. CLL: chronic lymphocytic leukemia; PBMC: peripheral blood mononuclear cell.
Discussion▴Top 

Patients with CLL remain at risk of severe COVID-19 even during the Omicron era [10, 11] and often display limited responsiveness to vaccination against SARS-CoV-2, with the lowest seroconversion rates if receiving CD20 monoclonal Ab-containing therapy or BTKi [2]. Even though multiple vaccinations may lead to seroconversion in more patients, some continue to remain seronegative or to have very low Ab titers [12]. Limited information is available on the impact on serological response under a strong BTK inhibitor such as zanubrutinib. Recently, a seroconversion rate of 77% was reported after three vaccine doses in zanubrutinib-treated patients with CLL or Waldenstrom’s macroglobulinemia using an alternative antibody detection method (even though the levels were not specified in U/mL) [13]. In the zanubrutinib-treated patients included in the present study, the median Ab level after five vaccine doses as determined by the standardized Elecsys® assay was only 2.7 U/mL, which was 1,000-fold lower than in age-matched healthy donors, and 43% of tested patients were still seronegative. There was considerable inter-individual response variation, and enlarged studies are needed to explore if there may be a relation to factors such as type of vaccine, age, or duration of zanubrutinib therapy. The serum neutralization capacity correlated very well to serum IgG levels, suggesting functionality of the Abs but also lower protection in individuals with low responses. The numerically higher Ab levels found in our cohort of ibrutinib-treated patients should be viewed with caution; enlarged studies of both BTKi are warranted. Local virus-specific IgA in mucosa probably plays a key role in protection against SARS-CoV-2 infection [14, 15], as it has a dimeric form with enhanced neutralizing capacity [16]. The fact that spike-specific IgA failed to increase either in serum or saliva after repeated subcutaneous vaccination in zanubrutinib-treated patients is, therefore, worrisome.

The extent to which seronegative patients with CLL can mount sufficient T-cell responses is currently unclear [2, 12]. The present study showed that seronegative patients with CLL on long-term zanubrutinib treatment could generate T-cell responses comparable to healthy individuals (tested after dose 5 only, thus, time kinetics could not be compared). This aligns with findings in patients with primary immunodeficiency (PID) who developed T-cell responses upon mRNA vaccination despite lacking B cells [4]. There are conflicting reports on whether additional vaccinations may boost T-cell responses. For instance, a fourth dose did so in older healthy individuals [17] as well as in patients with HIV [17], whereas a plateau was reached after the second dose in another report on healthy individuals [18]. Our small cohort size precluded definitive conclusions about T-cell kinetics, although we found a significant boost in CD8+ T-cell reactivity after dose 3, with no further increase after doses 4 and 5. Also in the three-dose zanubrutinib vaccine report by Nguyen et al, T-cell reactivities were increased by a third dose [13]. A third-dose boost was also reported in other subsets of patients with CLL [19, 20], whereas in lymphoma patients, a Th1, but not CD8+ T cell response, was reported by dose 3 [21].

The major limitations of our study are the small number of included patients and inter-individual data variation, limiting generalized conclusions from the observations. The risk of applying multiple analyses on small materials should also be kept in mind. Another limitation was the absence of saliva and longitudinally sampled serum from healthy donors. Finally, using exact cell counting techniques would have improved the flow cytometry analyses. Nevertheless, to our knowledge, this is the first study of systemic and mucosal vaccine responses in patients with ongoing third-generation BTKi therapy.

In summary, our study on a limited number of patients suggested that many but not all zanubrutinib-treated patients demonstrated a very low ability to generate spike-specific Abs in serum and almost no spike-specific Abs in saliva. Importantly, however, their capacity to generate spike-specific T cells was normal, reaching levels similar to age-matched healthy donors when tested after five vaccine doses. Further research is needed to understand how this impaired vaccine immunity affects the risk of severe SARS-CoV-2 infection and whether differences exist between various BTKi types.

Supplementary Material▴Top 

Suppl 1. Complete description of all methods.

Acknowledgments

We thank all patients and controls who donated blood for this study and A. Fahlen, K. Heimersson, and L. Relander for technical assistance.

Financial Disclosure

This study was supported by grants from the SciLifeLab National COVID-19 Research Program, the Swedish Research Council, Region Stockholm, the Swedish Cancer Society, the Cancer Society in Stockholm, the Cancer and Allergy Foundation, the Swedish Blood Cancer Foundation, Beigene Ltd, Center for Innovative Medicine (CIMED) and Karolinska Institutet.

Conflict of Interest

The authors declare no competing financial interests. MB is a consultant for Oxford Immunotech. AO and MP received research funding from Beigene LDT.

Informed Consent

Written informed consent was obtained before sampling.

Author Contributions

HMIS, MA, AO, GB, MSC, HGL, HM, and MB contributed to the conceptualization, funding acquisition, and discussion of data. JW, DW, YG, KH, SN, PC, MA, and SM performed experiments and analyzed data. MA, MP, HMIS, LH, and AO recruited study participants, conducted the management of participants during the study, and analyzed data. MA, HMIS, KH, DW, MSC, AO, HGL, and MB wrote the original draft of the manuscript. All authors reviewed and edited revisions of the manuscript and had final responsibility for the decision to submit for publication.

Data Availability

The data supporting the findings of this study are available from the corresponding author upon reasonable request.


References▴Top 
  1. Pagano L, Salmanton-Garcia J, Marchesi F, Blennow O, Gomes da Silva M, Glenthoj A, van Doesum J, et al. Breakthrough COVID-19 in vaccinated patients with hematologic malignancies: results from the EPICOVIDEHA survey. Blood. 2022;140(26):2773-2787.
    doi pubmed pmc
  2. Shadman M, Liu C, Eakle K, Hiew HJ, Biondo JML, Ghia P, Mato AR. COVID-19 vaccination response and its practical application in patients with chronic lymphocytic leukemia. Hemasphere. 2023;7(1):e811.
    doi pubmed pmc
  3. Bergman P, Blennow O, Hansson L, Mielke S, Nowak P, Chen P, Soderdahl G, et al. Safety and efficacy of the mRNA BNT162b2 vaccine against SARS-CoV-2 in five groups of immunocompromised patients and healthy controls in a prospective open-label clinical trial. EBioMedicine. 2021;74:103705.
    doi pubmed pmc
  4. Gao Y, Cai C, Wullimann D, Niessl J, Rivera-Ballesteros O, Chen P, Lange J, et al. Immunodeficiency syndromes differentially impact the functional profile of SARS-CoV-2-specific T cells elicited by mRNA vaccination. Immunity. 2022;55(9):1732-1746.e1735.
    doi pubmed pmc
  5. Brown JR, Eichhorst B, Hillmen P, Jurczak W, Kazmierczak M, Lamanna N, O'Brien SM, et al. Zanubrutinib or ibrutinib in relapsed or refractory chronic lymphocytic leukemia. N Engl J Med. 2023;388(4):319-332.
    doi pubmed
  6. Estupinan HY, Berglof A, Zain R, Smith CIE. Comparative analysis of BTK inhibitors and mechanisms underlying adverse effects. Front Cell Dev Biol. 2021;9:630942.
    doi pubmed pmc
  7. Tam CS, Brown JR, Kahl BS, Ghia P, Giannopoulos K, Jurczak W, Simkovic M, et al. Zanubrutinib versus bendamustine and rituximab in untreated chronic lymphocytic leukaemia and small lymphocytic lymphoma (SEQUOIA): a randomised, controlled, phase 3 trial. Lancet Oncol. 2022;23(8):1031-1043.
    doi pubmed
  8. www.etikprovningsmyndigheten.se.
  9. Blixt L, Gao Y, Wullimann D, Muren Ingelman-Sundberg H, Muschiol S, Healy K, Bogdanovic G, et al. Hybrid immunity in immunocompromised patients with CLL after SARS-CoV-2 infection followed by booster mRNA vaccination. Blood. 2022;140(22):2403-2407.
    doi pubmed pmc
  10. Niemann CU, da Cunha-Bang C, Helleberg M, Ostrowski SR, Brieghel C. Patients with CLL have a lower risk of death from COVID-19 in the Omicron era. Blood. 2022;140(5):445-450.
    doi pubmed pmc
  11. Mikulska M, Testi D, Russo C, Balletto E, Sepulcri C, Bussini L, Dentone C, et al. Outcome of early treatment of SARS-CoV-2 infection in patients with haematological disorders. Br J Haematol. 2023;201(4):628-639.
    doi pubmed
  12. Shen Y, Freeman JA, Holland J, Naidu K, Solterbeck A, Van Bilsen N, Downe P, et al. Multiple COVID-19 vaccine doses in CLL and MBL improve immune responses with progressive and high seroconversion. Blood. 2022;140(25):2709-2721.
    doi pubmed pmc
  13. Nguyen THO, Lim C, Lasica M, Whitechurch A, Tennakoon S, Saunders NR, Allen LF, et al. Prospective comprehensive profiling of immune responses to COVID-19 vaccination in patients on zanubrutinib therapy. EJHaem. 2023;4(1):216-220.
    doi pubmed pmc
  14. Havervall S, Marking U, Svensson J, Greilert-Norin N, Bacchus P, Nilsson P, Hober S, et al. Anti-spike mucosal IgA protection against SARS-CoV-2 omicron infection. N Engl J Med. 2022;387(14):1333-1336.
    doi pubmed pmc
  15. Russell MW, Mestecky J. Mucosal immunity: The missing link in comprehending SARS-CoV-2 infection and transmission. Front Immunol. 2022;13:957107.
    doi pubmed pmc
  16. Wang Z, Lorenzi JCC, Muecksch F, Finkin S, Viant C, Gaebler C, Cipolla M, et al. Enhanced SARS-CoV-2 neutralization by dimeric IgA. Sci Transl Med. 2021;13(577):eabf1555.
    doi pubmed pmc
  17. Saiag E, Alcalay Y, Marudi O, Orr-Urtreger A, Hagin D. Cellular and humoral immune response to the fourth Pfizer-BioNTech COVID-19 vaccine dose in individuals aged 60 years and older. Vaccine. 2023;41(4):914-921.
    doi pubmed pmc
  18. Pusnik J, Monzon-Posadas WO, Zorn J, Peters K, Baum M, Proksch H, Schluter CB, et al. SARS-CoV-2 humoral and cellular immunity following different combinations of vaccination and breakthrough infection. Nat Commun. 2023;14(1):572.
    doi pubmed pmc
  19. Hernandez SPA, Hersby DS, Munk KK, Tamhane T, Trubach D, Tagliamonte M, Buonaguro L, et al. Three doses of BNT162b2 COVID-19 mRNA vaccine establish long-lasting CD8(+) T cell immunity in CLL and MDS patients. Front Immunol. 2022;13:1035344.
    doi pubmed pmc
  20. Lee HK, Hoechstetter MA, Buchner M, Pham TT, Huh JW, Muller K, Zange S, et al. Analysis of immune responses in patients with CLL after heterologous COVID-19 vaccination. Blood Adv. 2023;7(10):2214-2227.
    doi pubmed pmc
  21. Gressens SB, Wiedemann A, Dechenaud M, Dupuis J, Gallien S, Melica G, Haioun C, et al. Anti-SARS-CoV-2 cellular response after 2 and 3 doses of BNT162b2 mRNA vaccine in lymphoma patients receiving anti-CD20 antibodies. Vaccine. 2023;41(9):1550-1553.
    doi pubmed pmc


This article is distributed under the terms of the Creative Commons Attribution Non-Commercial 4.0 International License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.


Journal of Hematology is published by Elmer Press Inc.

 

Browse  Journals  

     

Journal of Clinical Medicine Research

Journal of Endocrinology and Metabolism

Journal of Clinical Gynecology and Obstetrics

World Journal of Oncology

Gastroenterology Research

Journal of Hematology

Journal of Medical Cases

Journal of Current Surgery

Clinical Infection and Immunity

Cardiology Research

World Journal of Nephrology and Urology

Cellular and Molecular Medicine Research

Journal of Neurology Research

International Journal of Clinical Pediatrics

 

 

 

 

 

Journal of Hematology, bimonthly, ISSN 1927-1212 (print), 1927-1220 (online), published by Elmer Press Inc.                            
The content of this site is intended for health care professionals.
This is an open-access journal distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International License, which permits unrestricted
non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Creative Commons Attribution license (Attribution-NonCommercial 4.0 International CC BY-NC 4.0)



This journal follows the International Committee of Medical Journal Editors (ICMJE) recommendations for manuscripts submitted to biomedical journals,
the Committee on Publication Ethics (COPE) guidelines, and the Principles of Transparency and Best Practice in Scholarly Publishing.

website: www.thejh.org    editorial contact: editor@thejh.org
Address: 9225 Leslie Street, Suite 201, Richmond Hill, Ontario, L4B 3H6, Canada
 

© Elmer Press Inc. All Rights Reserved.


Disclaimer: The views and opinions expressed in the published articles are those of the authors and do not necessarily reflect the views or opinions of the editors and Elmer Press Inc. This website is provided for medical research and informational purposes only and does not constitute any medical advice or professional services. The information provided in this journal should not be used for diagnosis and treatment, those seeking medical advice should always consult with a licensed physician.