ArticlePDF Available

Abstract and Figures

A dysregulated immune response characterized by the hyperproduction of several pro-inflammatory cytokines (a.k.a. 'cytokine storm') plays a central role in the pathophysiology of severe coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this Perspective article we discuss the evidence for synergistic anti-inflammatory and immunomodulatory properties exerted by vitamin D and dipeptidyl peptidase-4 (DPP-4) inhibitors, the latter being a class of antihyperglycemic agents used for the treatment of Type 2 diabetes, which have also been reported as immunomodulators. Then, we provide the rationale for investigation of vitamin D and DPP-4 inhibitor combination therapy (VIDPP-4i) as an immunomodulation strategy to ratchet down the virulence of SARS-CoV-2, prevent disease progression and modulate the cytokine storm in COVID-19.
Content may be subject to copyright.
Perspective
For reprint orders, please contact: reprints@futuremedicine.com
Cytokine storm modulation in COVID-19: a
proposed role for vitamin D and DPP-4
inhibitor combination therapy (VIDPP-4i)
Marcelo Maia Pinheiro*,1 , Andrea Fabbri2& Marco Infante**,2,3,4
1Faculty of Medicine, UNIVAG University Center, V ´
arzea Grande, Mato Grosso, Brazil
2Department of Systems Medicine, Division of Endocrinology & Diabetes, Diabetes Research Institute Federation (DRIF), CTO
Hospital, University of Rome Tor Vergata, Rome, Italy
3UniCamillus, Saint Camillus International University of Health Sciences, Section of Endocrinology, Diabetes and Metabolism,
Rome, Italy
4Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientic Education and Research Network
(USERN), Rome, Italy
*Author for correspondence: marcelo.pinheiro@univag.edu.br
**Author for correspondence: marco.infante@unicamillus.org
A dysregulated immune response characterized by the hyperproduction of several pro-inammatory cy-
tokines (a.k.a. ‘cytokine storm’) plays a central role in the pathophysiology of severe coronavirus dis-
ease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In
this Perspective article we discuss the evidence for synergistic anti-inammatory and immunomodulatory
properties exerted by vitamin D and dipeptidyl peptidase-4 (DPP-4) inhibitors, the latter being a class
of antihyperglycemic agents used for the treatment of Type 2 diabetes, which have also been reported
as immunomodulators. Then, we provide the rationale for investigation of vitamin D and DPP-4 inhibitor
combination therapy (VIDPP-4i) as an immunomodulation strategy to ratchet down the virulence of SARS-
CoV-2, prevent disease progression and modulate the cytokine storm in COVID-19.
Tweetable abstract: Vitamin D and DPP-4 inhibitors exert anti-inammatory and immunomodulatory
properties. Vitamin D and DPP-4 inhibitor combination therapy (VIDPP-4i) may represent a valid therapeu-
tic approach to ratchet down the virulence of SARS-CoV-2 and modulate the cytokine storm in COVID-19.
Lay abstract: The so-called ‘cytokine storm’ that drives the hyperproduction of pro-inammatory medi-
ators, plays a central role in the pathophysiology of severe coronavirus disease 2019 (COVID-19) caused
by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Vitamin D has increasingly been
shown to play anti-inammatory and immunomodulatory properties beyond its role in the regulation
of bone homeostasis. Similarly, dipeptidyl peptidase-4 inhibitors (DPP-4i) – a class of antihyperglycemic
agents used for the treatment of Type 2 diabetes – have been reported as immunomodulators regard-
less of their glucose-lowering properties. We, therefore, discuss the role of vitamin D and DPP-4 inhibitor
combination therapy (VIDPP-4i) as a potential immunomodulation strategy to prevent the development
and/or halt the progression of the COVID-19-induced cytokine storm, particularly in patients with diabetes
and cardiovascular disease.
First draft submitted: 29 December 2020; Accepted for publication: 13 April 2021; Published online:
28 April 2021
Keywords: cardiovascular disease COVID-19 cytokine storm DPP-4 inhibitors drug repurposing
immunomodulation SARS-CoV-2 Type 2 diabetes VIDPP-4i vitamin D
Since the coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2
(SARS-CoV-2) was declared a pandemic on March 2020, there has been an unprecedented public health crisis
which has placed an enormous strain on healthcare systems worldwide. Older age, cardiovascular disease (CVD),
Immunotherapy (Epub ahead of print) ISSN 1750-743X10.2217/imt-2020-0349 C
2021 Future Medicine Ltd
Perspective Pinheiro, Fabbri & Infante
diabetes, obesity, malignancy, chronic lung disease, chronic kidney disease and chronic liver disease have emerged
as independent risk factors for adverse clinical outcomes and mortality related to COVID-19 [1–3].
A dysregulated immune response called a ‘cytokine storm’ (also known as ‘cytokine-release syndrome’) and
characterized by an excessive increase in circulating levels of several pro-inflammatory cytokines (such as IL-1,
IL-2, IL-6, IFN-γand TNF) has been shown to play a central role in the pathophysiology of the most severe cases
of COVID-19, leading to acute respiratory distress syndrome (ARDS), disseminated intravascular coagulation,
multiorgan failure and ultimately death [4,5]. A marked activation and exhaustion of CD4+and CD8+T cells
accompanied by a skewing of T-cell activation toward T-helper (Th) 17 functional phenotype have been reported in
patients with COVID-19 and may contribute to the excessive production of effector pro-inflammatory cytokines
such as IL-2, TNF and IFN-γ[4,6]. Patients with severe COVID-19 have higher serum concentration of IL-6, IL-10,
IL-2 and IFN-γ, higher numbers of neutrophils in the peripheral blood and reduced counts of T cells (particularly
CD8+T cells) compared with patients with mild disease [7]. This suggests that the numbers of neutrophils and
T cells, as well as the circulating levels of pro-inflammatory cytokines in the peripheral blood are dynamically
correlated with the severity of COVID-19. It is possible that the peripheral lymphopenia – observed especially in
patients with severe COVID-19 – reflects the overactivation and functional exhaustion of T cells, as well as the
recruitment of lymphocytes to the respiratory tract or adhesion to inflamed respiratory vascular endothelium [8].
Moreover, increased B-cell activation and proliferation have also been correlated with adverse outcomes in severe
cases of COVID-19 [9]. Emerging evidence also suggests that endothelial activation and dysfunction contribute
to COVID-19 pathogenesis by altering the integrity of vessel barrier, driving a procoagulant state, triggering
endothelial inflammation and mediating leukocyte infiltration [10,11].
Diverse SARS-CoV-2 vaccine types are becoming available in different countries and several repurposed drugs
have been used for the treatment of COVID-19 since the beginning of the pandemic [12]. Nevertheless, only a few
repurposed drugs have been shown to offer a certain degree of effectiveness in preliminary intervention trials [13].
In this Perspective article we propose vitamin D and dipeptidyl peptidase-4 (DPP-4) inhibitors as potential
candidates for prevention and modulation of cytokine storm in patients with COVID-19 based on the most recent
evidence. DPP-4 inhibitors (DPP-4i) represent a class of oral antihyperglycemic agents used for the treatment
of Type 2 diabetes (T2D) [14], which have also been reported to play anti-inflammatory and immunomodulatory
properties [15]. References for this manuscript were identified through searches of PubMed using the following terms:
‘vitamin D’, ‘cholecalciferol’, ‘calcifediol’, ‘calcidiol’, ‘dipeptidyl peptidase-4’, ‘DPP-4’ and ‘DPP-4 inhibitors’ in
combination with ‘COVID-19’, ‘ACE2’, ‘SARS-CoV-2’, ‘cytokine storm’ and ‘cytokine-release syndrome’. With
regard to the literature pertaining to COVID-19, no criteria for publication data were set and we included articles
published in English between 1 January 2020 and 28 March 2021. We also checked reference lists in relevant
articles and Google Scholar for additional references. Articles resulting from these searches and relevant references
cited in those articles were reviewed. The final reference list was generated on the basis of relevance to the topics
covered in this publication.
VitaminD&COVID-19
Over the last decade, a number of mechanistic studies demonstrated that vitamin D exerts anti-inflammatory
and immunomodulatory properties beyond its well-established role in the regulation of bone homeostasis [16,17].
These properties may occur in vivo upon the achievement of adequate serum 25-hydroxyvitamin D (25[OH]D)
levels, which amount to approximately 40–60 ng/ml [17]. The aforementioned properties are exerted by the bio-
logically active form of vitamin D, which is also referred to as 1,25-dihydroxyvitamin D3 or calcitriol. It has been
shown that vitamin D plays a central role in the regulation of innate and adaptive immune responses, promoting
antiviral effector mechanisms, reducing the expression of pro-inflammatory cytokines and inducing tolerogenic
responses [16–18]. In particular, calcitriol has been shown to: upregulate the transcription of antimicrobial peptides
(such as cathelicidin and defensin β2) in various human cell lines (myeloid cells, monocytes/macrophages, neu-
trophils and keratinocytes); promote the differentiation of monocytes/macrophages and enhance their chemotactic
and phagocytic capacity; inhibit the production of several pro-inflammatory cytokines (e.g., IL-6 and TNF-α)by
monocytes and macrophages; decrease macrophage antigen-presentation and T-cell stimulatory ability; elicit the
shift of macrophage polarization from the M1 pro-inflammatory phenotype (‘classically activated macrophages’) to-
wards the M2 anti-inflammatory phenotype (‘alternatively activated macrophages’); render the dendritic cells more
tolerogenic and reduce their antigen-presenting capacity; upregulate regulatory T cells; and favor the shift of T cells
from an ‘effector’pro-inflammatory phenotype toward a ‘regulatory’ anti-inflammatory phenotype by reducing Th1
10.2217/imt-2020-0349 Immunotherapy (Epub ahead of print) future science group
Vitamin D & DPP-4 inhibitors for treatment of COVID-19 Perspective
and Th17 cell differentiation and promoting Th2 cell differentiation [16,17]. Importantly, vitamin D receptor has
been identified in almost all immune cells [17], as well as in human airway epithelial cells [19]. The nearly ubiquitous
expression of vitamin D receptor enables vitamin D to exert its pleiotropic actions, including the regulation of
local ‘respiratory homeostasis’ by upregulating the expression of antimicrobial peptides and/or by directly affecting
the replication of respiratory viruses [19]. Vitamin D deficiency (defined as serum 25-hydroxyvitamin D levels
less than 20 ng/ml) is a global health issue afflicting more than one billion children and adults worldwide [20].
Since the beginning of COVID-19 pandemic, vitamin D deficiency has been suggested as an independent risk
factor for SARS-CoV-2 infection and hospitalization, development of cytokine storm and poor outcomes related to
COVID-19 [21–26]. Moreover, the overlap between risk factors for vitamin D deficiency and severe manifestations
of COVID-19 (such as Black or Asian ethnic origin, living at higher latitudes, older age and obesity) [27,28] led re-
searchers to consider vitamin D deficiency and COVID-19 as two related pandemics [29]. In a large US retrospective,
observational study involving more than 190,000 patients with SARS-CoV-2 results from all 50 states, vitamin D
deficiency has been associated with significantly higher SARS-CoV-2 positivity rates [30]. Interestingly, the decrease
in SARS-CoV-2 positivity rate associated with 25(OH)D levels appeared to plateau as values approached 55 ng/ml,
suggesting that additional benefits exist when 25(OH)D levels are higher than the cut-off value used to define
vitamin D sufficiency for bone health (30 ng/ml) [30].
Several observational studies have showed that hospitalized patients with COVID-19 exhibit a markedly high
prevalence of hypovitaminosis D, and that vitamin D deficiency is associated with a more advanced disease radiologic
stage, along with a significantly higher risk of noninvasive mechanical ventilation and in-hospital mortality [31–35].
It has also been shown that serum vitamin D levels are significantly lower in severe/critical COVID-19 cases
compared with mild-to-moderate cases [35]. We recently showed that a lower vitamin D status upon admission is
significantly and independently associated with an increased risk of COVID-19-related in-hospital mortality [36].
A systematic review and meta-analysis of observational studies conducted by Pereira et al. [37] confirmed a positive
association between vitamin D deficiency and COVID-19 severity. In this regard, it is worth mentioning that acute
illness and systemic inflammatory response can further lower circulating 25(OH)D levels [38,39], thus explaining,
at least in part, the high frequency of severe vitamin D deficiency observed in patients with infectious diseases,
including COVID-19 complicated by cytokine release syndrome.
Given the abovementioned anti-inflammatory and immunomodulatory properties of vitamin D, vitamin D
deficiency may exacerbate COVID-19 severity and mortality by triggering the hyperinflammatory state and the
cytokine storm associated with the most severe cases. Indeed, patients with COVID-19 and vitamin D deficiency
have been shown to exhibit significantly higher serum level of several inflammatory and coagulation biomarkers
such as C-reactive protein, IL-6, TNF-α, ferritin, fibrinogen and D-dimer [34,36,40]. Therefore, there has been a
growing interest in the use of vitamin D as an adjuvant anti-inflammatory and immunomodulatory agent aimed
to prevent SARS-CoV-2 infection and/or the progression of COVID-19 toward the severe stage of the disease
characterized by the development of cytokine release syndrome [21,41]. Pilot intervention studies investigating the
therapeutic role of vitamin D supplementation in COVID-19 yielded promising results. In particular, vitamin
D administration (at regular and high doses and in different formulations) has been proven safe and effective in
accelerating viral clearance, decreasing fibrinogen levels, reducing the severity of the disease as well as the need for
intensive care unit treatment among hospitalized patients (middle-aged and older adults) with COVID-19 [42–45].
Angiotensin-converting enzyme 2 (ACE2), the receptor used by SARS-CoV-2 for cellular entry [46], catalyzes
the cleavage of angiotensin II (Ang II) (a vasoconstrictor peptide) into angiotensin 1–7 (Ang-[1–7]) (a vasodilator
peptide), thus reducing blood pressure. Moreover, Ang-(1–7) induces nitric oxide synthase (NOS) and further
antagonizes Ang II activity via its Ang II type 1 receptor (AT1R) [47]. Following ACE2 receptor binding and
SARS-CoV-2 entry into host cells, there is a downregulation of ACE2, resulting in excessive accumulation and pro-
inflammatory activity of Ang II potentially accompanied by the development of lung injury, pneumonia, ARDS,
myocarditis and/or cardiac injury [48,49]. An analysis on gene expression data from cells in bronchoalveolar lavage
fluid from COVID-19 patients has also shown an atypical pattern of the renin-angiotensin system (RAS), which
is predicted to elevate bradykinin levels in multiple tissues and promote vasodilation, vascular permeability and
hypotension [50]. It has also been suggested that certain polymorphisms in coding, noncoding and regulatory sites of
the ACE2 gene may contribute to the worse clinical course of COVID-19 observed in males and in different regions
worldwide [51].Thus,ACE2 polymorphisms may partly facilitate or reduce the risk of SARS-CoV-2 infection and
adverse outcomes of COVID-19 by affecting the translation regulation and expression of ACE2.
future science group 10.2217/imt-2020-0349
Perspective Pinheiro, Fabbri & Infante
Importantly, vitamin D has been suggested as a negative endocrine modulator of the RAS [47,49]. Of note, vitamin
D may decrease the risk of SARS-CoV-2 infection and protect against the symptoms of COVID-19 by inhibiting
the synthesis of renin (a proteolytic enzyme and a positive regulator of Ang II) and by increasing ACE2 expression
and Ang-(1–7) production in the lung, thus preventing Ang II accumulation, decreasing the pro-inflammatory
activity of Ang II and reducing the risk of ARDS, myocarditis and cardiac injury [47,49].
Recently, it has also been suggested that polymorphisms of the vitamin D binding protein (DBP)genemay
influence circulating 25(OH)D levels and, as a consequence, COVID-19 pathophysiology (in terms of susceptibility
to and mortality from SARS-CoV-2 infection). DBP is a serum α-globulin encoded by the GC gene and represents
the major binding/transport protein of all vitamin D metabolites. Under physiological conditions, the majority of
vitamin D metabolites (85–90%) are tightly bound to DBP, whereas only 10–15% of the circulating vitamin D
is bound to albumin, and less than 1% represents the unbound (free) form of vitamin D. Besides mediating the
transport of vitamin D metabolites and regulating the access of such metabolites to cells and tissues, DBP exerts other
important functions such as fatty acid transport, actin scavenging, macrophage activation and chemotaxis [52,53].
Notably, a recent study revealed a significant association between the DBP1 allele frequency and a lower COVID-
19 prevalence and mortality, suggesting that DBP1 carriers may be less susceptible to SARS-CoV-2 infection and
mortality related to COVID-19 [54]. However, further research is needed to better elucidate the relationship between
DBP and its polymorphisms, vitamin D and COVID-19 pathophysiology.
Dipeptidyl peptidase-4 (DPP-4), DPP-4 inhibitors & COVID-19
Dipeptidyl peptidase-4 (DPP-4, also referred to as cluster of differentiation 26 or CD26) is a serine exopeptidase
existing in two forms: a plasma membrane-bound form (mDPP-4, consisting of a type II transmembrane homod-
imeric glycoprotein) and a soluble form (sDPP-4). The soluble form maintains its enzymatic (peptidase) activity
and is thought to be released from the membrane into the circulation. DPP-4/CD26 is expressed ubiquitously
in several cells and tissues, including, but not limited to, kidney, lung, endothelia, liver, intestine and immune
cells such as T cells, activated B cells, activated natural killer cells and myeloid cells [55]. DPP-4 has previously
been identified as a functional receptor for the spike protein of the Middle East respiratory syndrome coronavirus
(MERS-CoV) mediating the virus entry into host cells [56].
DPP-4 inhibitors (DPP-4i, also known as gliptins) have been widely used since 2006 as oral antihyperglycemic
agents for the treatment of T2D and have been proven effective in improving glucose control by preventing the
DPP-4-mediated cleavage and inactivation of incretin hormones, enhancing endogenous insulin secretion and
suppressing glucagon secretion [14]. DPP-4i include sitagliptin, saxagliptin, alogliptin, linagliptin and vildagliptin.
In April 2020, we published an article in CellR4 highlighting the possible therapeutic role of DPP-4 and DPP-4i
in COVID-19 pathophysiology [57].
Both SARS-CoV and SARS-CoV-2 use ACE2 as the primary receptor for viral entry into host cells [46]. However,
bioinformatics approaches combining human-virus protein interaction prediction, computational model-based
selective docking and protein-docking based on crystal structures suggest DPP-4 as a candidate binding target of
the receptor-binding S1 domain of the SARS-CoV-2 spike glycoprotein. In addition, the crucial binding residues of
DPP-4 are identical to those that are bound to the spike protein of MERS-CoV [58,59]. Since DPP-4 is ubiquitously
expressed in several cells and tissues other than lung and respiratory tract, it may therefore participate into the
direct SARS-CoV-2-mediated injury of such tissues. Thus, DPP-4 inhibition may have the potential to counteract
the DPP-4-mediated SARS-CoV-2 hijacking and virulence and to improve clinical outcomes of COVID-19 by
interfering with the interaction between SARS-CoV-2 and target host cells [60–62].
However, the main glucose-independent mechanisms that potentially account for beneficial effects of DPP-4i in
COVID-19 include the immunomodulatory, anti-inflammatory and antifibrotic properties exerted by these drugs,
which may represent a valid therapeutic tool to prevent or halt the progression toward the hyperinflammatory state
and cytokine storm associated with the most severe COVID-19 cases [63–65]. Indeed, a large number of studies
demonstrated that DPP-4/CD26 modulates both innate and adaptive immune responses [15,55]. DPP-4/CD26 is
expressed only on a fraction of resting T cells, while it becomes significantly upregulated upon T-cell activation [55].
DPP-4/CD26 on T-cell surface induces co-stimulatory effects on T-cell activation, resulting in increased production
of Th1 and pro-inflammatory cytokines TNF-α,IFN-γand IL-6 [66]. CD26-mediated co-stimulation of CD8+T
cells exerts a cytotoxic effect primarily via granzyme B, IFN-γ,TNF-αand Fas ligand [67]. In a study conducted by
Bengsch et al. [68], human Th17 cells producing type 17 cytokines (e.g., IL-17, IL-22, TNF) exhibited the highest
levels of enzymatically active DPP-4/CD26 compared with Th1, Th2 and regulatory T cells. Of note, the lowest
10.2217/imt-2020-0349 Immunotherapy (Epub ahead of print) future science group
Vitamin D & DPP-4 inhibitors for treatment of COVID-19 Perspective
CD26 expression levels were identified for IL-10-producing CD4+T cells and CD25hiCD127-FOXP3+regulatory
T cells, suggesting suppressive effects exerted by CD26 on such anti-inflammatory cells [68]. Upon activation,
approximately 50% of human B cells express DPP-4/CD26 and targeted suppression of DPP-4 activity decreases
B-cell activation and DNA synthesis in a dose-dependent manner [55]. Remarkably, a recent large retrospective
cohort study conducted on 774,198 patients with T2D showed that the use of DPP-4i is associated with lower risk
of autoimmune disorders [69].
DPP-4 inhibition has been shown to attenuate lipopolysaccharide-induced lung injury in murine models of
ARDS and to inhibit the release of TNF-α, IL-6 and IL-8 by human lung microvascular endothelial cells [70].
Soare et al. [64] recently found that pharmacological inhibition of DPP-4 promoted regression of bleomycin-
induced dermal thickness in murine models of systemic sclerosis, and DPP4-knockout mice were less susceptible
to bleomycin-induced dermal and pulmonary fibrosis.
Of note, two Italian retrospective observational studies recently showed that the use of DPP-4i at the time of
hospital admission in patients with T2D and COVID-19 was associated with reduced mortality, improved clinical
outcomes, lower need for noninvasive mechanical ventilation and greater number of hospital discharges [71,72].
In the study conducted by Solerte et al. [71], authors included 338 consecutive hospitalized T2D patients with
COVID-19 (169 were on standard of care, while 169 were on sitagliptin). Sitagliptin treatment at the time of
hospitalization was associated with reduced mortality (hazard ratio: 0.44 [95% CI: 0.29–0.66]; p = 0.0001), along
with an improvement in clinical outcomes (60 vs 38% of improved patients; p = 0.0001) and with a higher number
of hospital discharges (120 vs 89 of discharged patients; p = 0.0008) compared with patients receiving standard
of care, respectively [71]. The study conducted by Mirani et al. [72] was a case series involving 387 hospitalized
patients with COVID-19; 90 out of 387 participants (23.3%) had T2D. In diabetic patients, the use of DPP-4i
was significantly and independently associated with a lower risk of mortality (adjusted hazard ratio: 0.13; 95%
CI: 0.02–0.92; p = 0.042) [72]. A recent systematic review and meta-analysis of observational studies found that
in-hospital DPP4i use is associated with a significantly reduced COVID-19 mortality in diabetic patients [73].These
findings suggest that diabetic patients with COVID-19 taking DPP-4i may exhibit less severe COVID-19-related
pneumonia and end-organ complications.
Additionally, it has been suggested that DPP-4i may exert cardioprotective effects via various mechanisms involv-
ing insulin resistance, dysfunctional immunity, oxidative stress, apoptosis, dyslipidemia, adipose tissue dysfunction,
as well as antiapoptotic properties of these drugs in the heart and vasculature [74]. A number of studies (mostly
preclinical) have shown that DPP-4i can facilitate wound healing, improve endothelial function and prevent the de-
velopment and progression of atherosclerosis by reducing neutrophil recruitment and activity, attenuating vascular
oxidative stress and modulating monocyte/macrophage-mediated responses [15].
Therefore, Du et al. [75] have recently proposed DPP-4 inhibition as a potential therapeutic strategy aimed to
alleviate the cardiovascular injury (including arrhythmia, acute coronary syndrome and heart failure) caused either
directly by SARS-CoV-2 or indirectly by the COVID-19-induced cytokine storm. We previously conducted an in
vitro study to assess the effects of the DPP-4 inhibitor sitagliptin on human peripheral blood mononuclear cells from
healthy volunteers [76]. We demonstrated that sitagliptin is able to markedly reduce the expression of IL-6, IL-17
and IFN-γand to stimulate the differentiation of Th1 and Th17 cells into TGF-β1-secreting regulatory cells with
low CD26 expression [76]. A number of short-term randomized controlled trials demonstrated that sitagliptin exerts
anti-inflammatory properties in patients with T2D, resulting in increased expression of IL-10 (an anti-inflammatory
cytokine) and reduced expression of various markers of low-grade inflammation, pro-inflammatory cytokines and
cell adhesion molecules, such as C-reactive protein, IL-6, IL-18, TNF-α, serum amyloid A-low-density lipoprotein
(SAA-LDL) complex, secreted phospholipase-A2 (sPLA2), soluble intercellular adhesion molecule-1 (sICAM-1)
and E-selectin [77–79]. A recent Phase II clinical trial published in The New England Journal of Medicine showed that
sitagliptin in combination with a standard immunosuppressive regimen of tacrolimus and sirolimus resulted in a
low incidence of acute graft-versus-host disease by day 100 after myeloablative allogeneic hematopoietic stem-cell
transplantation [80].
The anti-inflammatory and immunomodulatory properties of DPP-4i may therefore represent a further advantage
in the prevention or management of cytokine storm in COVID-19. An alternative (but not mutually exclusive)
explanation for the protective effects of DPP-4i against SARS-CoV-2 infection and progression relies on the
hypothesis that pharmacological inhibition of DPP-4 may lead to a significant rise in circulating levels of the
soluble form of DPP-4 (sDPP-4) [81], as it has been demonstrated in mice [82]. The subsequent relative abundance
future science group 10.2217/imt-2020-0349
Perspective Pinheiro, Fabbri & Infante
of sDPP-4 could offer binding sites for SARS-CoV-2, thus preventing or limiting the attachment of the virus to
the membrane-bound DPP-4 (mDPP-4) on target host cells, such as pneumocytes, endothelial cells or other cells
relevant for viral spread and replication [81].
Proposed synergistic effects of vitamin D & DPP-4 inhibitors in COVID-19
Based on the aforementioned remarks, vitamin D and DPP-4i appear to exert synergistic anti-inflammatory and
immunomodulatory effects that can be leveraged to ratchet down the virulence of SARS-CoV-2 and prevent the
development and/or halt the progression of COVID-19-induced cytokine storm.
Our group [83–87] and other authors [88,89] previously showed that combination therapy with vitamin D and
DPP-4i (VIDPP-4i) has the potential ability to protect beta-cell function in autoimmune diabetes. We also reported
for the first time a remarkable clinical improvement after 3-year combination therapy with sitagliptin and vitamin
D3 in a patient with mononeuritis multiplex, an unusual form of peripheral neuropathy characterized by the
presence of perineuritis [90,91]. Recent in vitro and in vivo studies conducted in patients with T2D demonstrated
that VIDPP-4i is able to: reduce the expression of pro-inflammatory cytokines (IFN-γand IL-17); decrease the
proliferation of CD4+T cells and non-CD4+cells; increase the levels of the anti-inflammatory cytokine IL-4;
and upregulate the expression of IL-37 and FOXP3 (Forkhead box protein P3), which are well-known markers for
regulatory T cells [92–94].
Furthermore, a study conducted in a rat model of fructose/salt-induced insulin resistance showed that the
addition of vitamin D3 to the DPP-4i vildagliptin potentiated the renoprotective effects of vildagliptin, which
consisted of a series of renal anti-inflammatory, antifibrotic, antioxidant and anti-apoptotic actions [95].These
findings may have additional clinical implications for COVID-19. In fact, chronic kidney disease has emerged
as an independent risk factor for poor outcomes related to COVID-19 [1] and SARS-CoV-2 may cause kidney
injury through direct cytopathic effects and indirect mechanisms secondary to the cytokine storm and the systemic
involvement of the disease [96]. Overall, these findings suggest that vitamin D and DPP-4i administered together exert
anti-inflammatory and immunomodulatory actions to a greater extent than vitamin D or DPP-4i administered
alone. Moreover, VIDPP-4i may exert protective effects against endothelial dysfunction [97,98], which has been
shown to play an important role in COVID-19 pathophysiology [10].Figure 1 illustrates the potential protective
effects of VIDPP-4i against SARS-CoV-2 infection and COVID-19 progression to the hyperinflammatory state
and cytokine storm.
Conclusion
In conclusion, randomized controlled trials are warranted to determine whether VIDPP-4i represents an effective
therapeutic intervention for prevention of COVID-19 progression to severe stages and modulation of the cytokine
storm, which still represent unmet needs for reducing COVID-19-related hospitalization and mortality rates,
particularly in vulnerable and high-risk populations such as diabetic patients.
Future perspective
Given its proven favorable safety profile (even when administered in high doses) [99], its inexpensive cost and ease
of administration, vitamin D supplementation may be a simple therapeutic intervention to prevent disease pro-
gression and/or modulate the cytokine storm in COVID-19. On the other hand, DPP-4i exert anti-inflammatory,
immunomodulatory and antifibrotic actions beyond their well-known role as glucose-lowering drugs for treatment
of T2D. Importantly, DPP-4i are among the noninsulin glucose-lowering agents which have proven to be safe
and effective (alone or in combination with insulin therapy) for management of T2D in the hospital [100,101]
and outpatient settings [102], even in the context of COVID-19 [103]. This may facilitate the use of DPP-4i in
hospitalized patients with COVID-19, without posing additional risks for serious adverse events, including hypo-
glycemia [102]. Accordingly, practical recommendations for the management of diabetes in patients with COVID-19
do not suggest discontinuation of DPP-4i [104]. A recent systematic review and meta-analysis of the Cardiovascular
Outcomes Trials conducted by Grenet et al. [105] found that DPP-4i were not associated with either a decreased
or an increased risk of overall (non-COVID-19) respiratory tract infection compared with placebo, thus sup-
porting the practical recommendations for the management of diabetes during the COVID-19 pandemic. With
this regard, the use of VIDPP-4i may offer a simple and safe means to hamper the hyperinflammatory state and
cytokine storm in patients with COVID-19. In addition, the antihyperglycemic properties of DPP-4i, combined
with the anti-inflammatory actions of such compounds reported in preclinical and clinical studies of endothelial
10.2217/imt-2020-0349 Immunotherapy (Epub ahead of print) future science group
Vitamin D & DPP-4 inhibitors for treatment of COVID-19 Perspective
Expression of antimicrobial peptides (cathelicidin and defensin E2)
Dierentiation and chemotactic/phagocytic capacity of monocytes/macrophages
Shift of macrophages towards M2 phenotype
Pro-inammatory cytokines (lL-6, TNF-D)
Anti-inammatory cytokines (lL-4, lL-37)
Th1 and Th17 cell dierentiation, proliferation and activation
Macrophage and dendritic cell antigen-presentation and T-cell stimulatory ability
FOXP3 expression
Th2 cell dierentiation and activation
Renin synthesis
SARS-CoV-2-mediated downregulation of ACE2
ACE2 expression and Ang-(1–7) production
Ang ll accumulation
Anti-infective properties
Anti-inammatory properties
Tolerogenic and
immunomodulatory properties
Eects on RAS
VlDPP-4i
Vitamin D DPP-4i
Eects of DPP-4i on
DPP-4/SARS-CoV-2 interaction (?)
Anti-inammatory,
immunomodulatory and
anti-brotic properties
Pleiotropic actions of DPP-4i
SARS-CoV-2 Cytokine storm
SARS-CoV-2 infection and/or COVID-19
progression to the hyperinammatory
state and cytokine storm
Treg cell dierentiation and activation
DPP-4-mediated interaction between SARS-CoV-2
spike glycoprotein and target host cells
Circulating levels of sDPP-4
Binding sites for SARS-CoV-2
Binding of SARS-CoV-2 to mDPP-4 on target host cells
Pro-inammatory mediators and cell adhesion
molecules (lL-6, lL-8, lL-17, lL-18, lFN-J, TNF-D, CRP,
SAA-LDL complex, sPLA2, slCAM-1, E-selectin)
Anti-inflammatory cytokines (lL-4, lL-10, lL-37)
Th1 and Th17 cell differentiation
FOXP3 expression
TGF-E1-secreting regulatory T cell differentiation
Activation of cardiovascular and renal inflammatory,
fibrotic, oxidant and apoptotic pathways
Endothelial dysfunction and atherogenesis
Figure 1. Potential protective effects of VIDPP-4i against SARS-CoV-2 infection and COVID-19 progression to the hyperinammatory
state and cytokine storm. The left side of the gure shows the protective effects of vitamin D, whereas the right side of the gure
illustrates the protective effects of DPP-4 inhibitors (DPP-4i). Vitamin D and DPP-4i share many anti-inammatory and
immunomodulatory properties, resulting in synergistic actions of these compounds when they are co-administered. Vitamin D actions
refer to those exerted by the biologically active form of vitamin D, which is also known as 1,25-dihydroxyvitamin D3 or calcitriol.
ACE2: Angiotensin-converting enzyme 2; Ang-(1–7): Angiotensin 1–7; Ang II: Angiotensin II; COVID-19: Coronavirus disease 2019; CRP:
C-reactive protein; DPP-4i: DPP-4 inhibitors; FOXP3: Forkhead box protein P3; IFN-γ: Interferon-gamma; IL: Interleukin; M2: Alternatively
activated macrophages; mDPP-4: Membrane-bound DPP-4; RAS: Renin-angiotensin system; SAA-LDL complex: Serum amyloid
A-low-density lipoprotein complex; SARS-CoV-2: Severe acute respiratory syndrome coronavirus 2; sDPP-4: Soluble form of DPP-4;
sICAM-1: Soluble intercellular adhesion molecule-1; sPLA2: Secreted phospholipase-A2; TGF-β1: Transforming growth factor-beta 1; Th: T
helper cell; TNF-α: Tumor necrosis factor-alpha; Treg: Regulatory T cell; VIDPP-4i: Vitamin D and DPP-4 inhibitor combination therapy.
dysfunction and atherosclerosis, make VIDPP-4i combinatorial approach an intriguing theurapeutic strategy that
may be particularly advantageous for patients at high risk for adverse outcomes related to COVID-19, especially
for those with comorbid T2D and/or cardiovascular disease and atherosclerosis [106]. Nonetheless, DPP-4i have
been investigated in numerous preclinical models of inflammatory diseases, such as arthritis, inflammatory bowel
disease and multiple sclerosis [107]. Therefore, the wide repertoire of candidate DPP-4 substrates (e.g., cytokines,
chemokines and neuropeptides) and the immunomodulatory actions of DPP-4i suggest that DPP-4i have a broad
range of potential therapeutic applications. This may prompt the repurposing of DPP-4i as anti-inflammatory and
immunomodulatory agents for the management of different inflammatory diseases, including COVID-19-induced
cytokine storm [15,107].
Finally, vitamin D and DPP-4i may also play a role in the prevention of SARS-CoV-2 infection through
mechanisms independent of their effects on innate and adaptive immunity. Indeed, vitamin D and DPP-4i might
hamper different steps of the viral entry and viral infection cycle of SARS-CoV-2, specifically by interfering with
RAS and ACE/Ang II/AT1R axis, and by counteracting the SARS-CoV-2-mediated downregulation of ACE2
expression as well as the interaction between DPP-4 and the receptor-binding S1 domain of the SARS-CoV-2 spike
glycoprotein. However, these hypotheses need to be validated by robust mechanistic evidence.
future science group 10.2217/imt-2020-0349
Perspective Pinheiro, Fabbri & Infante
Executive summary
Coronavirus disease 2019-induced cytokine storm
A dysregulated immune response called ‘cytokine storm’ occurs in the most severe cases of coronavirus disease
2019 (COVID-19) and consists of an excessive production of pro-inammatory cytokines.
The cytokine storm can lead to acute respiratory distress syndrome, disseminated intravascular coagulation,
multiorgan failure and ultimately death.
Even though several repurposed drugs have been used for the treatment of COVID-19, only a few of them have
been shown to offer a certain degree of effectiveness in preliminary intervention trials.
Vitamin D & COVID-19
Vitamin D has been shown to exert anti-inammatory and immunomodulatory properties, promoting antiviral
effector mechanisms, reducing the expression of pro-inammatory cytokines and inducing tolerogenic responses.
Vitamin D has also been suggested to inhibit the synthesis of renin and to increase the expression of ACE2 and
angiotensin 1–7 in the lung, thus preventing angiotensin II (Ang II) accumulation and reducing the Ang II
pro-inammatory activity.
Observational studies found that a lower vitamin D status correlates with higher severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2) positivity rates in the general population, along with higher levels of
pro-inammatory cytokines and worse prognosis in hospitalized patients with COVID-19.
Preliminary ndings from pilot intervention studies suggest that vitamin D administration may be effective in
accelerating the viral clearance and reducing the disease severity in hospitalized patients with COVID-19.
Dipeptidyl peptidase-4 inhibitors & COVID-19
Dipeptidyl peptidase-4 (DPP-4) has been suggested as a candidate binding target of the receptor-binding S1
domain of the SARS-CoV-2 spike glycoprotein.
DPP-4 inhibitors (DPP-4i, a.k.a. gliptins) are a class of oral antihyperglycemic agents widely used for the treatment
of Type 2 diabetes.
DPP-4 inhibition may have the potential to interfere with the interaction between SARS-CoV-2 and target host
cells.
Emerging evidence suggests that DPP-4 inhibitors exhibit immunomodulatory, anti-inammatory and antibrotic
properties beyond their well-established role as antihyperglycemic agents.
Observational studies have recently showed that the use of DPP-4 inhibitors was associated with improved clinical
outcomes in hospitalized patients with Type 2 diabetes and COVID-19.
VIDPP-4i combination therapy & COVID-19
Based on emerging evidence coming from studies conducted in autoimmune diabetes, vitamin D and DPP-4
inhibitors appear to exert synergistic anti-inammatory and immunomodulatory effects.
According to the aforementioned remarks, vitamin D and DPP-4 inhibitor combination therapy (VIDPP-4i) may
represent a valid therapeutic approach to ratchet down the virulence of SARS-CoV-2, prevent disease progression
and modulate the cytokine storm in COVID-19, particularly in patients at high risk for COVID-19-related adverse
outcomes such as those with comorbid diabetes, cardiovascular disease and/or atherosclerosis.
VIDPP-4i may also play a role in the prevention of SARS-CoV-2 infection by interfering with renin-angiotensin
system and ACE/Ang II/AT1R axis, and counteracting the SARS-CoV-2-mediated downregulation of ACE2 as well
as the interaction between DPP-4 and the SARS-CoV-2 spike glycoprotein.
Randomized controlled trials are needed to establish whether VIDPP-4i represents a safe and effective
intervention for prevention of SARS-CoV-2 infection and COVID-19 progression to the cytokine storm.
Financial & competing interests disclosure
The authors have no relevant afliations or nancial involvement with any organization or entity with a nancial interest in or nan-
cial conict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria,
stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
No writing assistance was utilized in the production of this manuscript.
In memoriam
This article is dedicated to the memory of Miriam Pinheiro, sister of Dr Marcelo Maia Pinheiro and frontline nurse who lost her life
due to COVID-19 on 26 March 2021.
References
Papers of special note have been highlighted as: of interest; •• of considerable interest
1. Noor FM, Islam MM. Prevalence and associated risk factors of mortality among COVID-19 patients: a meta-analysis. J. Community
Health 45(6), 1270–1282 (2020).
10.2217/imt-2020-0349 Immunotherapy (Epub ahead of print) future science group
Vitamin D & DPP-4 inhibitors for treatment of COVID-19 Perspective
2. Roy S, Mazumder T, Banik S. The association of cardiovascular diseases and diabetes mellitus with COVID-19 (SARS-CoV-2) and their
possible mechanisms. SN. Compr. Clin. Med. 1–6 (2020) (Epub ahead of print).
3. Drucker DJ. Coronavirus infections and Type 2 diabetes-shared pathways with therapeutic implications. Endocr. Rev. 41(3), bnaa011
(2020).
•• Provides significant insights into the shared pathophysiology underlying coronavirus disease 2019 (COVID-19) and Type 2
diabetes.
4. Mangalmurti N, Hunter CA. Cytokine storms: understanding COVID-19. Immunity 53(1), 19–25 (2020).
•• Provides relevant information on the pathophysiology of COVID-19-related cytokine storm.
5. Ragab D, Salah Eldin H, Taeimah M, Khattab R, Salem R. The COVID-19 cytokine storm; what we know so far. Front. Immunol. 11,
1446 (2020).
6. De Biasi S, Meschiari M, Gibellini L et al. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with
COVID-19 pneumonia. Nat. Commun. 11(1), 3434 (2020).
7. LiuJ,LiS,LiangBet al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of
SARS-CoV-2 infected patients. EBioMedicine 55, 102763 (2020).
8. Chen Z, John Wherry E. T cell responses in patients with COVID-19. Nat. Rev. Immunol. 20(9), 529–536 (2020).
9. Yang L, Liu S, Liu J et al. COVID-19: immunopathogenesis and immunotherapeutics. Signal Transduct. Target. Ther. 5(1), 128 (2020).
10. Jin Y, Ji W, Yang H, Chen S, Zhang W, Duan G. Endothelial activation and dysfunction in COVID-19: from basic mechanisms to
potential therapeutic approaches. Signal Transduct. Target. Ther. 5(1), 293 (2020).
11. Sinha P, Matthay MA, Calfee CS. Is a “cytokine storm” relevant to COVID-19? JAMA Intern. Med. 180(9), 1152–1154 (2020).
12. Sultana J, Crisafulli S, Gabbay F, Lynn E, Shakir S, Trifir`
o G. Challenges for drug repurposing in the COVID-19 pandemic era. Front.
Pharmacol. 11, 588654 (2020).
13. Lenze EJ, Mattar C, Zorumski CF et al. Fluvoxamine vs placebo and clinical deterioration in outpatients with symptomatic COVID-19:
a randomized clinical trial. JAMA 324(22), 2292–2300 (2020).
14. Dicker D. DPP-4 inhibitors: impact on glycemic control and cardiovascular risk factors. Diabetes Care 34(Suppl. 2), S276–S278 (2011).
15. Yazbeck R, Jaenisch SE, Abbott CA. Dipeptidyl peptidase 4 inhibitors: applications in innate immunity? Biochem.
Pharmacol. 188, 114517 (2021) (Epub ahead of print).
16. Caprio M, Infante M, Calanchini M, Mammi C, Fabbri A. Vitamin D: not just the bone. Evidence for beneficial pleiotropic
extraskeletal effects. Eat. Weight Disord. 22(1), 27–41 (2017).
17. Fabbri A, Infante M, Ricordi C. Editorial - vitamin D status: a key modulator of innate immunity and natural defense from acute viral
respiratory infections. Eur. Rev. Med. Pharmacol. Sci. 24(7), 4048–4052 (2020).
18. Infante M, Ricordi C, Padilla N et al. The role of vitamin D and omega-3 PUFAs in islet transplantation. Nutrients 11(12), 2937 (2019).
19. Zdrenghea MT, Makrinioti H, Bagacean C, Bush A, Johnston SL, Stanciu LA. Vitamin D modulation of innate immune responses to
respiratory viral infections. Rev. Med. Virol. 27(1), (2017).
20. Holick MF. The vitamin D deficiency pandemic: approaches for diagnosis, treatment and prevention. Rev. Endocr. Metab. Disord. 18(2),
153–165 (2017).
21. Grant WB, Lahore H, McDonnell SL et al. Evidence that vitamin D supplementation could reduce risk of influenza and COVID-19
infections and deaths. Nutrients 12(4), 988 (2020).
22. Glinsky GV. Tripartite combination of candidate pandemic mitigation agents: vitamin D, quercetin, and estradiol manifest properties of
medicinal agents for targeted mitigation of the COVID-19 pandemic defined by genomics-guided tracing of SARS-CoV-2 targets in
human cells. Biomedicines 8(5), 129 (2020).
23. Bilezikian JP, Bikle D, Hewison M et al. Mechanisms in endocrinology: vitamin D and COVID-19. Eur. J. Endocrinol. 183(5),
R133–R147 (2020).
24. Ali N. Role of vitamin D in preventing of COVID-19 infection, progression and severity. J. Infect. Public Health 13(10), 1373–1380
(2020).
25. Meltzer DO, Best TJ, Zhang H, Vokes T, Arora V, Solway J. Association of vitamin D status and other clinical characteristics with
COVID-19 test results. JAMA Netw. Open 3(9), e2019722 (2020).
26. Merzon E, Tworowski D, Gorohovski A et al. Low plasma 25(OH) vitamin D level is associated with increased risk of COVID-19
infection: an Israeli population-based study. FEBS J. 287(17), 3693–3702 (2020).
27. Whittemore PB. COVID-19 fatalities, latitude, sunlight, and vitamin D. Am. J. Infect. Control 48(9), 1042–1044 (2020).
28. Martineau AR, Forouhi NG. Vitamin D for COVID-19: a case to answer? Lancet Diabetes Endocrinol. 8(9), 735–736 (2020).
29. Kara M, Ekiz T, Ricci V, Kara ¨
O, Chang KV, ¨
Ozc¸akar L. ‘Scientific strabismus’ or two related pandemics: coronavirus disease and
vitamin D deficiency. Br. J. Nutr. 124(7), 736–741 (2020).
future science group 10.2217/imt-2020-0349
Perspective Pinheiro, Fabbri & Infante
30. Kaufman HW, Niles JK, Kroll MH, Bi C, Holick MF. SARS-CoV-2 positivity rates associated with circulating 25-hydroxyvitamin D
levels. PLoS ONE 15(9), e0239252 (2020).
31. Carpagnano GE, Di Lecce V, Quaranta VN et al. Vitamin D deficiency as a predictor of poor prognosis in patients with acute
respiratory failure due to COVID-19. J. Endocrinol. Invest. 44(4), 765–771 (2020) (Epub ahead of print).
32. Radujkovic A, Hippchen T, Tiwari-Heckler S, Dreher S, Boxberger M, Merle U. Vitamin D deficiency and outcome of COVID-19
patients. Nutri ents 12(9), 2757 (2020).
33. De Smet D, De Smet K, Herroelen P, Gryspeerdt S, Martens GA. Serum 25(OH)D level on hospital admission associated with
COVID-19 stage and mortality. Am. J. Clin. Pathol. 155(3), 381–388 (2020).
34. Hern´
andez JL, Nan D, Fernandez-Ayala M et al. Vitamin D status in hospitalized patients with SARS-CoV-2 Infection. J. Clin.
Endocrinol. Metab.(2020) (Epub ahead of print) https://doi.org/doi:10.1210/clinem/dgaa733
35. Ye K, Tang F, Liao X et al. Does serum vitamin D level affect COVID-19 infection and its severity?-A case-control study. J. Am. Coll.
Nutr. 1–8 (2020) (Epub ahead of print).
36. Infante M, Buoso A, Pieri M et al. Low vitamin D status at admission as a risk factor for poor survival in hospitalized patients with
COVID-19: an Italian retrospective study. J. Am. Coll. Nutr. 1–16 (2021) (Epub ahead of print).
37. Pereira M, Dantas Damascena A, Galv˜
ao Azevedo LM, de Almeida Oliveira T, da Mota Santana J. Vitamin D deficiency aggravates
COVID-19: systematic review and meta-analysis. Crit. Rev. Food Sci. Nutr. 1–9 (2020) (Epub ahead of print).
38. French CB, McDonnell SL, Vieth R. 25-Hydroxyvitamin D variability within-person due to diurnal rhythm and illness: a case report. J.
Med. Case Rep. 13(1), 29 (2019).
39. Smolders J, van den Ouweland J, Geven C, Pickkers P, Kox M. Letter to the Editor: vitamin D deficiency in COVID-19: mixing up
cause and consequence. Metabolism 115, 154434 (2021).
40. Jain A, Chaurasia R, Sengar NS, Singh M, Mahor S, Narain S. Analysis of vitamin D level among asymptomatic and critically ill
COVID-19 patients and its correlation with inflammatory markers. Sci. Rep. 10(1), 20191 (2020).
Suggests the involvement of vitamin D deficiency in the hyperinflammatory state observed among patients with COVID-19.
41. Jakovac H. COVID-19 and vitamin D-is there a link and an opportunity for intervention? Am. J. Physiol. Endocrinol. Metab. 318(5),
E589 (2020).
42. Entrenas Castillo M, Entrenas Costa LM, Vaquero Barrios JM et al. “Effect of calcifediol treatment and best available therapy versus best
available therapy on intensive care unit admission and mortality among patients hospitalized for COVID-19: a pilot randomized clinical
study”. J. Steroid Biochem. Mol. Biol. 203, 105751 (2020).
Suggests a potential role of vitamin D (calcifediol) as an adjuvant treatment for patients with COVID-19.
43. Rastogi A, Bhansali A, Khare N et al. Short term, high-dose vitamin D supplementation for COVID-19 disease: a randomised,
placebo-controlled, study (SHADE study). Postgrad. Med. J. (2020) (Epub ahead of print)
http://dx.doi.org/doi:10.1136/postgradmedj-2020- 139065
44. Annweiler G, Corvaisier M, Gautier J et al. Vitamin D supplementation associated to better survival in hospitalized frail elderly
COVID-19 patients: the GERIA-COVID quasi-experimental study. Nutrients 12(11), 3377 (2020).
45. Annweiler C, Hanotte B, Grandin de l’Eprevier C, Sabatier JM, Lafaie L, C´
elarier T. Vitamin D and survival in COVID-19 patients: a
quasi-experimental study. J. Steroid Biochem. Mol. Biol. 204, 105771 (2020).
46. Bourgonje AR, Abdulle AE, Timens W et al. Angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2 and the pathophysiology of
coronavirus disease 2019 (COVID-19). J. Pathol. 251(3), 228–248 (2020).
47. Malek Mahdavi A. A brief review of interplay between vitamin D and angiotensin-converting enzyme 2: implications for a potential
treatment for COVID-19. Rev. Med. Virol. 30(5), e2119 (2020).
48. Hanff TC, Harhay MO, Brown TS, Cohen JB, Mohareb AM. Is there an association between COVID-19 mortality and the
renin-angiotensin system? A call for epidemiologic investigations. Clin. Infect. Dis. 71(15), 870–874 (2020).
49. Aygun H. Vitamin D can prevent COVID-19 infection-induced multiple organ damage. Naunyn Schmiedebergs Arch.
Pharmacol. 393(7), 1157–1160 (2020).
50. Garvin MR, Alvarez C, Miller JI et al. A mechanistic model and therapeutic interventions for COVID-19 involving a RAS-mediated
bradykinin storm. Elife 9, e59177 (2020).
51. Khayat AS, de Assumpc¸˜
ao PP, Meireles Khayat BC et al. ACE2 polymorphisms as potential players in COVID-19 outcome. PLoS
ONE 15(12), e0243887 (2020).
52. Speeckaert M, Huang G, Delanghe JR, Taes YE. Biological and clinical aspects of the vitamin D binding protein (Gc-globulin) and its
polymorphism. Clin. Chim. Acta 372(1–2), 33–42 (2006).
53. Speeckaert MM, Speeckeart R, Delanghe JR. Genetic polymorphisms, vitamin D binding protein and vitamin D deficiency in
COVID-19. Eur. Respir. J. 2004638 (2021) (Epub ahead of print).
54. Speeckaert MM, De Buyzere ML, Delanghe JR. Vitamin D binding protein polymorphism and COVID-19. J. Med. Virol. 93(2),
705–707 (2021).
10.2217/imt-2020-0349 Immunotherapy (Epub ahead of print) future science group
Vitamin D & DPP-4 inhibitors for treatment of COVID-19 Perspective
55. Klemann C, Wagner L, Stephan M, von H¨
orsten S. Cut to the chase: a review of CD26/dipeptidyl peptidase-4’s (DPP4) entanglement
in the immune system. Clin. Exp. Immunol. 185(1), 1–21 (2016).
56. Raj VS, Mou H, Smits SL et al. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC.
Nature 495(7440), 251–254 (2013).
57. Maia Pinheiro M, Monteiro Pinheiro J, Moura Maia Pinheiro F. Editorial – COVID-19 pandemic: is it time to learn about
DPP-4/CD26? CellR4 8, e2835 (2020).
58. Li Y, Zhang Z, Yang L et al. The MERS-CoV receptor DPP4 as a candidate binding target of the SARS-CoV-2 spike. iScience 23(6),
101160 (2020).
59. Vankadari N, Wilce JA. Emerging WuHan (COVID-19) coronavirus: glycan shield and structure prediction of spike glycoprotein and its
interaction with human CD26. Emerg. Microbes Infect. 9(1), 601–604 (2020).
60. Solerte SB, Di Sabatino A, Galli M, Fiorina P. Dipeptidyl peptidase-4 (DPP4) inhibition in COVID-19. Acta Diabetol. 57(7), 779–783
(2020).
61. Chen CF, Chien CH, Yang YP et al. Role of dipeptidyl peptidase-4 inhibitors in patients with diabetes infected with coronavirus-19. J.
Chin. Med. Assoc. 83(8), 710–711 (2020).
62. Bassendine MF, Bridge SH, McCaughan GW, Gorrell MD. COVID-19 and comorbidities: a role for dipeptidyl peptidase 4 (DPP4) in
disease severity? J. Diabetes 12(9), 649–658 (2020).
63. Strollo R, Pozzilli P. DPP4 inhibition: preventing SARS-CoV-2 infection and/or progression of COVID-19? Diabetes Metab. Res.
Rev. 36(8), e3330 (2020).
•• Provides insights into the potential mechanisms accounting for the role of DPP-4 inhibitors in the prevention of severe acute
respiratory syndrome coronavirus 2 infection and/or progression of COVID-19. Also discusses the anti-inflammatory and
antifibrotic properties exerted by these compounds.
64. Soare A, Gy¨
orfi HA, Matei AE et al. Dipeptidylpeptidase 4 as a marker of activated fibroblasts and a potential target for the treatment of
fibrosis in systemic sclerosis. Arthritis Rheumatol. 72(1), 137–149 (2020).
65. Iacobellis G. COVID-19 and diabetes: can DPP4 inhibition play a role? Diabetes Res. Clin. Pract. 162, 108125 (2020).
66. Pacheco R, Martinez-Navio JM, Lejeune M et al. CD26, adenosine deaminase, and adenosine receptors mediate costimulatory signals in
the immunological synapse. Proc. Natl Acad. Sci. USA 102(27), 9583–9588 (2005).
67. Hatano R, Ohnuma K, Yamamoto J, Dang NH, Morimoto C. CD26-mediated co-stimulation in human CD8(+) T cells provokes
effector function via pro-inflammatory cytokine production. Immunology 138(2), 165–172 (2013).
68. Bengsch B, Seigel B, Flecken T, Wolanski J, Blum HE, Thimme R. Human Th17 cells express high levels of enzymatically active
dipeptidylpeptidase IV (CD26). J. Immunol. 188(11), 5438–5447 (2012).
69. Chen YC, Chen TH, Sun CC et al. Dipeptidyl peptidase-4 inhibitors and the risks of autoimmune diseases in Type 2 diabetes mellitus
patients in Taiwan: a nationwide population-based cohort study. Acta Diabetol. 57(10), 1181–1192 (2020).
70. Kawasaki T, Chen W, Htwe YM, Tatsumi K, Dudek SM. DPP4 inhibition by sitagliptin attenuates LPS-induced lung injury in mice.
Am. J. Physiol. Lung Cell. Mol. Physiol. 315(5), L834–L845 (2018).
71. Solerte SB, D’Addio F, Trevisan R et al. Sitagliptin treatment at the time of hospitalization was associated with reduced mortality in
patients with Type 2 diabetes and COVID-19: a multicenter, case-control, retrospective, observational study. Diabetes Care 43(12),
2999–3006 (2020).
Suggests a potential therapeutic role of DPP-4 inhibitors in hospitalized patients with COVID-19 and Type 2 diabetes.
72. Mirani M, Favacchio G, Carrone F et al. Impact of comorbidities and glycemia at admission and dipeptidyl peptidase 4 inhibitors in
patients with Type 2 diabetes with COVID-19: a case series from an academic hospital in Lombardy, Italy. Diabetes Care 43(12),
3042–3049 (2020).
Suggests a potential therapeutic role of DPP-4 inhibitors in hospitalized patients with COVID-19 and Type 2 diabetes.
73. Pal R, Banerjee M, Mukherjee S, Singh Bhogal R, Kaur A, Bhadada SK. Dipeptidyl peptidase-4 inhibitor use and mortality in
COVID-19 patients with diabetes mellitus: an updated systematic review and meta-analysis. Ther. Adv. Endocrinol. Metab.
12, 2042018821996482 (2021) (Epub ahead of print).
74. Aroor AR, Sowers JR, Jia G, DeMarco VG. Pleiotropic effects of the dipeptidylpeptidase-4 inhibitors on the cardiovascular system. Am.
J. Physiol. Heart Circ. Physiol. 307(4), H477–H492 (2014).
75. Du H, Wang DW, Chen C. The potential effects of DPP-4 inhibitors on cardiovascular system in COVID-19 patients. J. Cell. Mol.
Med. 24(18), 10274–10278 (2020).
76. Pinheiro MM, Stoppa CL, Valduga CJ et al. Sitagliptin inhibit human lymphocytes proliferation and Th1/Th17 differentiation in vitro.
Eur. J. Pharm. Sci. 100, 17–24 (2017).
77. Makdissi A, Ghanim H, Vora M et al. Sitagliptin exerts an antinflammatory action. J. Clin. Endocrinol. Metab. 97(9), 3333–3341 (2012).
78. Satoh-Asahara N, Sasaki Y, Wada H et al. A dipeptidyl peptidase-4 inhibitor, sitagliptin, exerts anti-inflammatory effects in Type 2
diabetic patients. Metabolism 62(3), 347–351 (2013).
future science group 10.2217/imt-2020-0349
Perspective Pinheiro, Fabbri & Infante
79. Tremblay AJ, Lamarche B, Deacon CF, Weisnagel SJ, Couture P. Effects of sitagliptin therapy on markers of low-grade inflammation
and cell adhesion molecules in patients with type 2 diabetes. Metabolism 63(9), 1141–1148 (2014).
80. Farag SS, Abu Zaid M, Schwartz JE et al. Dipeptidyl peptidase 4 inhibition for prophylaxis of acute graft-versus-host disease. N. Engl. J.
Med. 384(1), 11–19 (2021).
81. Nauck MA, Meier JJ. Reduced COVID-19 mortality with sitagliptin treatment? Weighing the dissemination of potentially lifesaving
findings against the assurance of high scientific standards. Diabetes Care 43(12), 2906–2909 (2020).
82. Varin EM, Mulvihill EE, Beaudry JL et al. Circulating levels of soluble dipeptidyl peptidase-4 are dissociated from inflammation and
induced by enzymatic DPP4 inhibition. Cell. Metab. 29(2), 320–334.e5 (2019).
83. Pinheiro MM, Pinheiro FM, Torres MA. Four-year clinical remission of Type 1 diabetes mellitus in two patients treated with sitagliptin
and vitamin D3. Endocrinol. Diabetes Metab. Case Rep. 2016, 16–0099 (2016).
84. Pinheiro MM, Pinheiro FMM, Trabachin ML. Dipeptidyl peptidase-4 inhibitors (DPP-4i) combined with vitamin D3: an exploration
to treat new-onset Type 1 diabetes mellitus and latent autoimmune diabetes in adults in the future. Int. Immunopharmacol. 57, 11–17
(2018).
85. Pinheiro M, Pinheiro F. Prolonging the honeymoon phase in T1DM with sitagliptin plus vitamin D3. The Official Journal of ATTD
Advanced Technologies & Treatments for Diabetes Conference Madrid, Spain. Diabetes Technol. Ther. 22(S1), A1–A250 (2020).
86. Infante M, Ricordi C, Sanchez J et al. Influence of vitamin D on islet autoimmunity and beta-cell function in Type 1 diabetes.
Nutri ents 11(9), 2185 (2019).
87. Pinheiro MM, Pinheiro FMM, Diniz SN, Fabbri A, Infante M. Combination of vitamin D and dipeptidyl peptidase-4 inhibitors
(VIDPP-4i) as an immunomodulation therapy for autoimmune diabetes. Int. Immunopharmacol. 95, 107518 (2021).
88. Rapti E, Karras S, Grammatiki M et al. Combined treatment with sitagliptin and vitamin D in a patient with latent autoimmune
diabetes in adults. Endocrinol. Diabetes Metab. Case Rep. 2016, 150136 (2016).
89. Zhang Z, Yan X, Wu C et al. Adding vitamin D3 to the dipeptidyl peptidase-4 inhibitor saxagliptin has the potential to protect β-cell
function in LADA patients: a 1-year pilot study. Diabetes Metab. Res. Rev. 36(5), e3298 (2020).
90. Maia Pinheiro M, Moura Maia Pinheiro F, Pires Amaral Resende LL, Nogueira Diniz S, Fabbri A, Infante M. Improvement of pure
sensory mononeuritis multiplex and IgG1 deficiency with sitagliptin plus Vitamin D3. Eur. Rev. Med. Pharmacol. Sci. 24(15),
8151–8159 (2020).
91. Maia Pinheiro M, Maia Pinheiro FM, Amaral Resende LLP, Diniz SN, Fabbri A, Infante M. 36-month follow-up of a pure sensory
mononeuritis multiplex and IgG1 deficiency improved after treatment with sitagliptin and Vitamin D3. Eur. Rev. Med. Pharmacol.
Sci. 25(4), 1768–1769 (2021).
92. Mahabadi-Ashtiyani E, Sheikh V, Borzouei S, Salehi I, Alahgholi-Hajibehzad M. The increased T helper cells proliferation and
inflammatory responses in patients with Type 2 diabetes mellitus is suppressed by sitagliptin and vitamin D3 in vitro.Inflamm.
Res. 68(10), 857–866 (2019).
93. Telikani Z, Sheikh V, Zamani A et al. Effects of sitagliptin and vitamin D3 on T helper cell transcription factors and cytokine
production in clinical subgroups of Type 2 diabetes mellitus: highlights upregulation of FOXP3 and IL-37. Immunopharmacol.
Immunotoxicol. 41(2), 299–311 (2019).
94. Borzouei S, Sheikh V, Ghasemi M et al. Anti-Inflammatory effect of combined sitagliptin and vitamin D3 on cytokines profile in
patients with Type 2 diabetes mellitus. J. Interferon Cytokine Res. 39(5), 293–301 (2019).
95. Wahba NS, Abdel-Ghany RH, Ghareib SA, Abdel-Aal M, Alsemeh AE. Vitamin D3 potentiates the renoprotective effects of vildagliptin
in a rat model of fructose/salt-induced insulin resistance. Eur. J. Pharm. Sci. 144, 105196 (2020).
96. Fortarezza F, Pezzuto F. COVID-19 nephropathy: what could pathologist say? J. Nephropathol. 9(4), e32 (2020).
97. Caprio M, Mammi C, Rosano GM. Vitamin D: a novel player in endothelial function and dysfunction. Arch. Med. Sci. 8(1), 4–5 (2012).
98. Aini K, Fukuda D, Tanaka K et al. Vildagliptin, a DPP-4 Inhibitor, attenuates endothelial dysfunction and atherogenesis in nondiabetic
apolipoprotein E-deficient mice. Int. Hear t J. 60(6), 1421–1429 (2019).
99. Fassio A, Adami G, Rossini M et al. Pharmacokinetics of oral cholecalciferol in healthy subjects with vitamin D deficiency: a randomized
open-label study. Nutrients 12(6), 1553 (2020).
100. American Diabetes Association. 15. Diabetes care in the hospital: standards of medical care in diabetes – 2021. Diabetes
Care 44(Suppl. 1), S211–S220 (2021).
101. Lorenzo-Gonz´
alez C, Atienza-S´
anchez E, Reyes-Umpierrez D et al. Safety and efficacy of DPP4-inhibitors for management of
hospitalized general medicine and surgery patients with Type 2 diabetes. Endocr. Pract. 26(7), 722–728 (2020) (Epub ahead of print).
102. Ling J, Cheng P, Ge L et al. The efficacy and safety of dipeptidyl peptidase-4 inhibitors for Type 2 diabetes: a Bayesian network
meta-analysis of 58 randomized controlled trials. Acta Diabetol. 56(3), 249–272 (2019).
103. Roussel R, Darmon P, Pichelin M et al. Use of dipeptidyl peptidase-4 inhibitors and prognosis of COVID-19 in hospitalized patients
with Type 2 diabetes: a propensity score analysis from the CORONADO study. Diabetes Obes. Metab. (2021) (Epub ahead of print).
10.2217/imt-2020-0349 Immunotherapy (Epub ahead of print) future science group
Vitamin D & DPP-4 inhibitors for treatment of COVID-19 Perspective
104. Bornstein SR, Rubino F, Khunti K et al. Practical recommendations for the management of diabetes in patients with COVID-19. Lancet
Diabetes Endocrinol. 8(6), 546–550 (2020).
105. Grenet G, Mekhaldi S, Mainbourg S et al. DPP-4 inhibitors and respiratory infection: a systematic review and meta-analysis of the
cardiovascular outcomes trials. Diabetes Care 44(3), e36–e37 (2021).
106. de Almeida-Pititto B, Dualib PM, Zajdenverg L et al. Severity and mortality of COVID 19 in patients with diabetes, hypertension and
cardiovascular disease: a meta-analysis. Diabetol. Metab. Syndr. 12, 75 (2020).
107. Yazbeck R, Howarth GS, Abbott CA. Dipeptidyl peptidase inhibitors, an emerging drug class for inflammatory disease? Tre n d s
Pharmacol. Sci. 30(11), 600–607 (2009).
future science group 10.2217/imt-2020-0349
... 7 In addition, vitamin D plays a variety of roles, including regulating the transcription of antimicrobial peptides in different cell lines, contributing to the differentiation of monocytes and macrophages, and suppressing the production of proinflammatory cytokines. 8 Tang et al. demonstrated that normal vitamin D levels are associated with lower levels of IL-1 and IL-6. 9 The pathophysiology of SARS-CoV-2 infection includes the overproduction of several proinflammatory cytokines such as IL-1, TNF, IL-6, IL-12, IL-17, GM-CSF, and IFN-γ, which may trigger the cytokine storm. ...
... 2,8 Furthermore, vitamin D receptors have been identified in epithelial cells of the human respiratory tract, having the ability to regulate local respiratory homeostasis by upregulating the expression of antimicrobial peptides or by influencing viral replication. 8 Akbar et al. performed a meta-analysis with 14 studies and 999,179 participants, which showed that low serum vitamin D levels were associated with higher rates of SARS-CoV-2 infection, severe courses, and mortality. 10 According to Amos et al., vitamin D is involved in zinc homeostasis, which plays an important role in reducing coronavirus replication. ...
Article
Full-text available
Introduction During the COVID-19 pandemic, vitamin D was used along with vitamin C and zinc as a preventive and curative therapy against SARS-CoV-2 infection. Vitamin D toxicity, even if it is rare, occurs when serum concentrations exceed 150 ng/mL and is usually manifested by hypercalcemia phenomena. Case report We hereby report a case of two twin sisters who self-medicated with vitamin D in a dose of 4 × 4,000 IU/day for almost 10 months as a method of ‘protection’ against COVID-19, influenced by mass media advertising. The patients presented to the emergency department with hypervitaminosis D-related symptoms such as hypertension, headache, nausea, vomiting, and diffuse abdominal pain. Laboratory investigations revealed high levels of vitamin D and calcium. Conclusions Vitamin D toxicity can lead to difficulties in positive and differential diagnosis because of the multiple complications of hypercalcemia.
... Conversely, sufficient intracellular calcitriol availability transforms proinflammatory Th1 and Th17 cells into anti-inflammatory Th2 and Treg response cells [92,94]. This transformation reduces the release of inflammatory cytokines and enhances the expression of anti-inflammatory cytokines, mitigating the risk of cytokine storms and ARDS [100,103,104]. ...
Article
Full-text available
Introduction and aim: SARS-CoV-2 outbreaks occur cyclically, aligning with winter when vitamin D levels are lowest, except after new variant outbreaks. Adequate vitamin D is crucial for robust immune function. Hypovitaminosis diminishes immune responses, increasing susceptibility to viral infections. The manuscript explores the discrepancy between increased SARS-CoV-2 hospitalizations and lower mortality. Method: SARS-CoV-2 mutants, including Delta, BQ, and XBB Omicron lineages, developed immune evasion capabilities, reducing the effectiveness of COVID-19 vaccines and bivalent boosters. The failure of COVID-19 vaccines to prevent infections and spread to others, coupled with the immune evasion exhibited by mutant viruses, contributed to continued SARS-CoV-2 outbreaks. Interestingly, dominant new mutants, despite their increased transmissibility, have caused fewer deaths. This article scrutinizes the mentioned incongruity through an analysis of published data. Results: Achieving herd immunity and eradicating SARS-CoV-2 has proven elusive due to ongoing mutagenesis and immune evasion, leading to recurrent viral outbreaks. The failure to approve repurposed early therapies for COVID-19 by regulators and misinformation and weak strategies undertaken by leading health authorities exacerbated the situation. Repurposed agents, including vitamin D and ivermectin, have demonstrated high efficacy against SARS-CoV-2 from the beginning and remain unaffected by mutations. Despite their cost-effectiveness and widespread availability, regulatory approval for these generic agents in COVID-19 treatment is pending. Conclusion: Regulators hesitated to approve cost-effective, repurposed generic agents primarily to safeguard the temporary approval status of COVID-19 vaccines and anti-viral agents under Emergency Use Authorization, which persists. This reluctance overlooked the opportunity to implement an integrated approach with repurposed agents alongside COVID-19 vaccines, potentially reducing hospitalizations and fatalities and preventing outbreaks; this led to the failure to eradicate SARS-CoV-2 and becoming endemic. It is imperative that regulators now reconsider approving affordable generics for SARS-CoV-2 to effectively control future viral outbreaks. Non-technical Importance (Lay Abstract) Adequate vitamin D levels significantly bolster the human immune system-deficiency compromises immune responses and increases susceptibility, particularly to viruses. While new SARS-CoV-2 mutations like Omicron are less severe, they are more infectious and adept at evading immunity from vaccines; thus, they offer a limited spectrum of protection and duration. Primary COVID-19 vaccines have reduced disease severity but have failed to prevent viral spread, contributing to outbreaks. Booster doses had little effect on the virus but caused immune paresis, thus increasing susceptibility to infections. Regulators should consider approving generic, repurposed agents like vitamin D and ivermectin as adjunct therapies to address this challenge and better prepare for future pandemics. Proactively International Journal of Frontiers in Science and Technology Research, 2024, 06(02), 001-020 2 Integrating vitamin D supplementation to fortify the immune system can mitigate vital outbreaks, alleviate hospital burdens, and reduce healthcare costs.
... By inhibiting the degradation of these incretin hormones, DPP-4 inhibitors can enhance their activity, leading to improved glucose control [38]. Beyond its metabolic role, DPP-4 is also involved in immune modulation [39], T-cell activation, and migration [40]. DPP-4 inhibitors have shown potential therapeutic benefits beyond glycemic control, including anti-inflammatory effects [41] and cardiovascular benefits [42]. ...
... By inhibiting the degradation of these incretin hormones, DPP-4 inhibitors can enhance their activity, leading to improved glucose control [38]. Beyond its metabolic role, DPP-4 is also involved in immune modulation [39], T-cell activation, and migration [40]. DPP-4 inhibitors have shown potential therapeutic benefits beyond glycemic control, including anti-inflammatory effects [41] and cardiovascular benefits [42]. ...
Article
Full-text available
Citation: Hossain, A.; Rahman, M.E.; Faruqe, M.O.; Saif, A.; Suhi, S.; Zaman, R.; Hirad, A.H.; Matin, M.N.; Rabbee, M.F.; Baek, K.-H. Abstract: Diabetes, characterized by elevated blood sugar levels, poses significant health and economic risks, correlating with complications like cardiovascular disease, kidney failure, and blindness. Dipeptidyl peptidase-4 (DPP-4), also referred to as T-cell activation antigen CD26 (EC 3.4.14.5.), plays a crucial role in glucose metabolism and immune function. Inhibiting DPP-4 was anticipated as a potential new therapy for diabetes. Therefore, identification of plant-based natural inhibitors of DPP-4 would help in eradicating diabetes worldwide. Here, for the identification of the potential natural inhibitors of DPP-4, we developed a phytochemicals library consisting of over 6000 phyto-chemicals detected in 81 medicinal plants that exhibited anti-diabetic potency. The library has been docked against the target proteins, where isorhamnetin, Benzyl 5-Amino-5-deoxy-2,3-O-isopropyl-alpha-D-mannofuranoside (DTXSID90724586), and 5-Oxo-7-[4-(trifluoromethyl) phenyl]-4H,6H,7H-[1,2]thiazolo[4,5-b]pyridine 3-carboxylic acid (CHEMBL3446108) showed binding affinities of −8.5, −8.3, and −8.3 kcal/mol, respectively. These compounds exhibiting strong interactions with DPP-4 active sites (Glu205, Glu206, Tyr547, Trp629, Ser630, Tyr662, His740) were identified. ADME/T and bioactivity predictions affirmed their pharmacological safety. Density functional theory calculations assessed stability and reactivity, while molecular dynamics simulations demonstrated persistent stability. Analyzing parameters like RMSD, RG, RMSF, SASA, H-bonds, MM-PBSA, and FEL confirmed stable protein-ligand compound formation. Principal component analysis provided structural variation insights. Our findings suggest that those compounds might be possible candidates for developing novel inhibitors targeting DPP-4 for treating diabetes.
... Daneshkhah et al. [13] investigated the possible effect of vitamin D deficiency on the development of cytokine storm and subsequent mortality in COVID-19 patients, Life 2024, 14, 210 2 of 15 and they concluded that the possible role of vitamin D in modulating the rate of cytokine storm in COVID-19 should be further investigated. The overproduction of inflammatory cytokines during severe COVID-19 disease is involved in the pathogenesis of the cytokine storm and may lead to mortality [14]. Vitamin D, via its anti-inflammatory properties [15], may modulate the cytokine storm observed in severe COVID-19 disease [16]. ...
Article
Full-text available
The SARS-CoV-2 virus may cause severe infection, which is associated with diverse clinical manifestations. Vitamin D has immunomodulating properties and may enhance the body’s defense system against invading pathogenic organisms. The aim was to assess 25(OH)D3 levels in patients hospitalized for severe infection from the SARS-CoV-2 virus and explore the relationship between 25(OH)D3 and outcomes. In a group of 88 patients hospitalized for severe infection from the SARS-CoV-2 virus and a control group matched for age and sex, the levels of 25(OH)D3 were analyzed. Levels of 25(OH)D3 were 17.36 ± 8.80 ng/mL (mean ± SD) compared with 24.34 ± 10.34 ng/mL in patients with severe SARS-CoV-2 infection and the control group, respectively, p < 0.001 (Student’s t-test). 25(OH)D3 levels were significantly related to outcomes, i.e., survival as opposed to non-survival, as more patients with 25(OH)D3 deficiency (0–10 ng/mL) and insufficiency (10–20 ng/mL) had a fatal outcome as compared with those with vitamin D sufficiency (p < 0.001, chi-square test, p < 0.001, Fisher’s exact test). Levels of 25(OH)D3 were inversely related to C-reactive protein (CRP), ferritin, d-dimer, and fibrinogen levels (p < 0.001, linear regression analysis, beta coefficient of variation, −0.176, −0.160, −0.178, and −0.158, respectively). Vitamin D deficiency observed in severe SARS-CoV-2 infection was related to disease outcomes.
... [16,33,34] Nevertheless, the results have been equivocal. It has been documented that adequate circulating concentrations of 25-hydroxycalciferol (25(OH) D) and vitamin C may reduce the incidence of coronavirus-induced "cytokine storm", [35,36] which is strongly associated with lung injury and poor prognosis of the disease. [37] Currently, the large-scale administration of vitamin D supplements to populations at risk of COVID-19 is widely recommended. ...
Article
Full-text available
Background The coronavirus disease 2019 (COVID-19) pandemic elicited the general population to use various dietary supplements (DSs) and nutraceuticals as a protective means against the disease. The present study aimed to evaluate changes and certain determinants of DS intake during the COVID-19 lockdown among Iranian households. Methods This nationwide cross-sectional study was conducted from April 4 to April 25, 2020, during which Iran was in lockdown. To collect data, a web-based electronic self-administered questionnaire was created. The data were compared among provinces based on their food security situations. Results A total of 21,290 households were included in the analyses. Approximately 27% of the households were using DSs after the epidemic. The most common DSs used were vitamin D (42%) and vitamin C (20%), followed by multi-vitamin (16%), zinc (9%), omega-3 (6%), vitamin A (4%), and probiotics (3%). Logistic regression analysis revealed that DS intake was directly associated with the household income but inversely with household size and the food security status of the provinces. DS intake was positively associated with the presence of high-risk persons in the households, the education of the households' head, and the presence of a person with a history of COVID-19 within the household. Conclusions During epidemic lockdown, DS use was remarkably increased among the Iranian households. Apart from the debatable usefulness of DSs against COVID-19, the inverse association of DS use with a household's income and provincial food security well indicates inequity in accessibility to DS. Actions to improve the nutritional status of the under-privileged populations including targeted supplementation are strongly recommended.
... At 2 months from the T1D diagnosis, the patient had already entered the clinical remission phase, as indicated by an IDAA1c value of 5.66 (Supplementary Table 1 Figure 1). Therefore, we prescribed calcifediol (also known as 25-hydroxyvitamin D3 or calcidiol; brand name: Neodidro) at a dose of 0.266 mg every 20 days, in order to treat hypovitaminosis D and to exploit the anti-inflammatory and immunomodulatory properties of vitamin D [31][32][33][34][35][36][37]. ...
Article
Herein, we describe an unusually prolonged duration (31 months) of the clinical remission phase in a 22-year-old Italian man with new-onset type 1 diabetes. Shortly after the disease diagnosis, the patient was treated with calcifediol (also known as 25-hydroxyvitamin D3 or calcidiol), coupled with low-dose basal insulin, to correct hypovitaminosis D and to exploit the anti-inflammatory and immunomodulatory properties of vitamin D. During the follow-up period, the patient retained a substantial residual β-cell function and remained within the clinical remission phase, as evidenced by an insulin dose-adjusted glycated hemoglobin value <9. At 24 months, we detected a peculiar immunoregulatory profile of peripheral blood cells, which may explain the prolonged duration of the clinical remission sustained by calcifediol as add-on treatment to insulin.
... Vitamin D is known not only for its importance in calcium and phosphate metabolism but also for its biological actions in immunomodulation. It has been depicted that vitamin D plays a central role in regulating innate and adaptive immune responses, promoting the antiviral effect mechanism, reducing the expression of pro-inflammatory cytokines, and inducing a tolerance response [53][54][55][56] . This is because of the presence of the vitamin D receptor in most cell types, especially immune cells, including activated T and B lymphocytes and macrophages 57 . ...
Article
Full-text available
A few patients who have recovered from COVID-19 develop persistent or new symptoms that last for weeks or months; this is called "long COVID" or "post-COVID-19 syndrome." Over time, awareness of the short- and long-term consequences of COVID-19 has increased. The pulmonary consequences are now fairly well established, but little is known about the extrapulmonary system of COVID-19, particularly its effects on bones. Current evidence and reports indicate a direct relationship between SARS-CoV-2 infection and bone health, with SARS-CoV-2 having a significant negative effect on bone health. In this review, we analyzed the impact of SARS-CoV-2 infection on bone health and assessed the impact of COVID-19 on the diagnosis and treatment of osteoporosis.
... , is based on measurements obtained in two regions and might 85% of total VD levels, in Northern Bohemia, its effect reduces by pollution, physical stress, and unhealthy lifestyle[7, 9,10, 24,29, 31,38,39]. The nutritional role of VD levels in the Czech Republic, in general, and in Nothern Bohemia, in particular, is important compared to other European locations. ...
Article
Full-text available
Objective The COVID-19 epidemic resulted in a global crisis of public health. Therefore, the possibility of prevention, leading to reduced infection and/or an improved disease state, is the subject of intensive attention. The novelty of this study is the direct evaluation of vitamin D levels with the risk of COVID-19 infection. Background Currently, several nutraceuticals, including vitamin D, beta-glucan, and some minerals, are being studied for their role in stimulating immunity. Our study focused on the relationship between levels of vitamin D in immunodeficient patients and the risk of the development of COVID-19. Method In this study, patients were supplemented with vitamin D. Results In a group of 71 patients, we found that patients with vitamin D levels below 30 ng/ml had an increased risk of COVID-19 development and more severe disease progress. In patients with blood levels over 40 ng/ml, we consistently found high levels of protection against COVID-19 infection. Conclusion The most important finding is that vitamin D levels above 40 ng/ml result in the reduction of risks of serious clinical manifestation of COVID-19 infection.
Article
Full-text available
Background Few observational studies have shown a beneficial effect of dipeptidyl peptidase-4 inhibitors (DPP4i) in patients with coronavirus disease 2019 (COVID-19), although results are not consistent. The present systematic review and meta-analysis was undertaken to provide a precise summary of the effect of DPP4i use (preadmission or in-hospital) and mortality in COVID-19 patients with diabetes mellitus (DM). Methods PubMed and Google Scholar databases were systematically searched using appropriate keywords to 4 January 2021, to identify observational studies reporting mortality in COVID-19 patients with DM using DPP4i versus those not using DPP4i. Preadmission and in-hospital use of DPP4i were considered. Study quality was assessed using the Newcastle–Ottawa Scale. Unadjusted and adjusted pooled odds ratio (OR) with 95% confidence intervals (CIs) were calculated. Subgroup analysis was performed for studies reporting preadmission and in-hospital use of DPP4i. Results We identified nine observational studies of high quality pooling data retrieved from 7008 COVID-19 patients with DM. The pooled analysis of unadjusted and adjusted data did not show any significant association between DPP4i use and mortality in COVID-19 patients with DM. However, on subgroup analysis, we found that in-hospital (and not preadmission) DPP4i use was associated with reduced mortality (unadjusted OR 0.37, 95% CI 0.23, 0.58, p < 0.0001, I ² = 0% and adjusted OR 0.27, 95% CI 0.13, 0.55, p = 0.0003, I ² = 12%). Conclusions In-hospital use of DPP4i is associated with a significant reduction in COVID-19 mortality. Hence, it would be prudent to initiate or continue DPP4i in COVID-19 patients with DM if not contraindicated.
Article
Full-text available
Objective: Preliminary findings suggest a relationship between lower serum 25-hydroxyvitamin D [25(OH)D] levels and incidence and severity of COVID-19. The aim of this study was to evaluate the relationship between vitamin D status at admission and different markers of inflammation, coagulation, and sepsis in hospitalized patients with COVID-19. Method: We conducted a retrospective study on 137 consecutive patients with SARS-CoV-2 infection and available data on serum 25(OH)D levels, who were admitted to our Institution between March 1 and April 30, 2020. Patients were divided into two groups: survivors (n = 78; 57%) and non-survivors (n = 59; 43%). Results: At admission, all patients showed hypovitaminosis D. Median total serum 25(OH)D levels at admission were significantly higher in survivors than non-survivors (12 ng/mL vs 8 ng/mL; p < 0.01). Non-survivors exhibited significantly higher median levels of white blood cell (WBC) count, neutrophil-to-lymphocyte count ratio (NLR), high-sensitivity C-reactive protein (hsCRP), ferritin, interleukin 6 (IL-6), D-dimer, fibrinogen, and procalcitonin (PCT) compared to survivors at three different time points during hospitalization. In a multivariate analysis performed by a logistic regression model, serum 25(OH)D levels were significantly inversely associated with risk of COVID-19-related in-hospital mortality (odds ratio, 0.91; 95% confidence interval, 0.85-0.98; p = 0.01). According to receiver operating characteristic curve analysis, hsCRP, NLR, ferritin, and D-dimer were the best predictive biomarkers for poor prognosis of COVID-19, whereas IL-6, PCT, fibrinogen, 25(OH)D, WBC count, and tumor necrosis factor alpha (TNF-α) may serve as supportive biomarkers for worse clinical course of the disease. Conclusions: We found a markedly high prevalence (100%) of hypovitaminosis D in patients admitted to hospital with COVID-19, suggesting a possible role of low vitamin D status in increasing the risk of SARS-CoV-2 infection and subsequent hospitalization. The inverse association between serum 25(OH)D levels and risk of in-hospital mortality observed in our cohort suggests that a lower vitamin D status upon admission may represent a modifiable and independent risk factor for poor prognosis in COVID-19.
Article
Full-text available
Aims: Dipeptidyl Peptidase-4 (DPP-4), the target of oral antidiabetic drugs DDP-4 inhibitors, has been suggested to be involved in the pathogenesis of coronavirus infections, including COVID-19. It is unclear whether the routine use of DPP-4 inhibitors increases the severity of COVID-19 in people with type 2 diabetes (T2D). Our purpose was to investigate the association between routine use of DPP-4 inhibitors and the severity of COVID-19 infection in a large multicentric study. Material and methods: This study was a secondary analysis of the CORONADO study on 2449 patients with T2D hospitalized for COVID-19 in 68 French centres. The composite primary endpoint combined tracheal intubation for mechanical ventilation and death within 7 days of admission. Stabilized weights were computed for patients based on propensity score (DPP-4 inhibitors users vs non-users) and were used into multivariable logistic regression models to estimate Average Treatment effect in the Treated as Inverse Probability of Treatment Weighting (IPTW). Results: 596 participants were under DPP-4 inhibitors before admission to hospital (24.3%). The primary outcome occurred at similar rates in users and non-users of DPP-4 inhibitors (27.7% vs 28.6%, P=0.68). In propensity analysis, the IPTW-adjusted models showed no significant association between use of DPP-4 inhibitors and the primary outcome within day 7 (OR [95%CI]: 0.95 [0.77-1.17]) or day 28 (OR [95%CI]: 0.96 [0.78-1.17]). Similar neutral findings were found between use of DPP-4 inhibitors and the risk of tracheal intubation and death. Conclusions: These data support the safety of DPP-4 inhibitors for diabetes management during the COVID-19 pandemic and they should not be discontinued. This article is protected by copyright. All rights reserved.
Article
Full-text available
The clinical condition COVID-19, caused by SARS-CoV-2, was declared a pandemic by the WHO in March 2020. Currently, there are more than 5 million cases worldwide, and the pandemic has increased exponentially in many countries, with different incidences and death rates among regions/ethnicities and, intriguingly, between sexes. In addition to the many factors that can influence these discrepancies, we suggest a biological aspect, the genetic variation at the viral S protein receptor in human cells, ACE2 (angiotensin I-converting enzyme 2), which may contribute to the worse clinical outcome in males and in some regions worldwide. We performed exomics analysis in native and admixed South American populations, and we also conducted in silico genomics databank investigations in populations from other continents. Interestingly, at least ten polymorphisms in coding, noncoding and regulatory sites were found that can shed light on this issue and offer a plausible biological explanation for these epidemiological differences. In conclusion, there are ACE2 polymorphisms that could influence epidemiological discrepancies observed among ancestry and, moreover, between sexes.
Article
Type 1 diabetes (T1D) and latent autoimmune diabetes in adults (LADA) represent the most common types of autoimmune diabetes and are characterized by different age of onset, degrees of immune-mediated destruction of pancreatic beta cells and rates of disease progression towards insulin dependence. Several immunotherapies aimed to counteract autoimmune responses against beta cells and preserve beta-cell function are currently being investigated, particularly in T1D. Preliminary findings suggest a potential role of combination therapy with vitamin D and dipeptidyl peptidase-4 (DPP-4) inhibitors (VIDPP-4i) in preserving beta-cell function in autoimmune diabetes. This manuscript aims to provide a comprehensive overview of the immunomodulatory properties of vitamin D and DPP-4 inhibitors, as well as the rationale for investigation of their combined use as an immunomodulation therapy for autoimmune diabetes.
Article
Dipeptidyl peptidase (DPP)-4 inhibitors are a class of orally available, small molecule inhibitors that prolong the insulinotropic activity of the incretin hormone glucagon-like peptide-1 (GLP-1) and are highly effective for the treatment of Type-2 diabetes. DPP4 can also cleave several immunoregulatory peptides including chemokines. Emerging evidence continues to implicate DPP4 inhibitors as immunomodulators, with recent findings suggesting DPP4 inhibitors modify specific aspects of innate immunity. This review summarises recent insights into how DPP4 inhibitors could be implicated in endothelial, neutrophil and monocyte/macrophage mediated immunity. Additionally, this review highlights additional avenues of research with DPP4 inhibitors in the context of the COVID-19 pandemic.
Article
Background: Dipeptidyl peptidase 4 (DPP-4; also known as CD26), a transmembrane receptor expressed on T cells, has a costimulatory function in activating T cells. In a mouse model, down-regulation of CD26 prevented graft-versus-host disease (GVHD) but preserved graft-versus-tumor effects. Whether inhibition of DPP-4 with sitagliptin may prevent acute GVHD after allogeneic stem-cell transplantation is not known. Methods: We conducted a two-stage, phase 2 clinical trial to test whether sitagliptin plus tacrolimus and sirolimus would reduce the incidence of grade II to IV acute GVHD from 30% to no more than 15% by day 100. Patients received myeloablative conditioning followed by mobilized peripheral-blood stem-cell transplants. Sitagliptin was given orally at a dose of 600 mg every 12 hours starting the day before transplantation until day 14 after transplantation. Results: A total of 36 patients who could be evaluated, with a median age of 46 years (range, 20 to 59), received transplants from matched related or unrelated donors. Acute GVHD occurred in 2 of 36 patients by day 100; the incidence of grade II to IV GVHD was 5% (95% confidence interval [CI], 1 to 16), and the incidence of grade III or IV GVHD was 3% (95% CI, 0 to 12). Nonrelapse mortality was zero at 1 year. The 1-year cumulative incidences of relapse and chronic GVHD were 26% (95% CI, 13 to 41) and 37% (95% CI, 22 to 53), respectively. GVHD-free, relapse-free survival was 46% (95% CI, 29 to 62) at 1 year. Toxic effects were similar to those seen in patients undergoing allogeneic stem-cell transplantation. Conclusions: In this nonrandomized trial, sitagliptin in combination with tacrolimus and sirolimus resulted in a low incidence of grade II to IV acute GVHD by day 100 after myeloablative allogeneic hematopoietic stem-cell transplantation. (Funded by the National Heart, Lung, and Blood Institute; ClinicalTrials.gov number, NCT02683525.).