Next Article in Journal
Association between Adverse Reactions and Humoral Immune Response No Longer Detectable after BNT162b2 Booster Vaccination
Previous Article in Journal
Knowledge of SARS-CoV-2 Epitopes and Population HLA Types Is Important in the Design of COVID-19 Vaccines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Insights into SARS-CoV-2 and Cancer Cross-Talk: Does a Novel Oncogenesis Driver Emerge?

by
Vasiliki Rapti
1,*,†,
Thomas Tsaganos
2,†,
Ioannis A. Vathiotis
1,
Nikolaos K. Syrigos
1,3,
Peifeng Li
4 and
Garyfallia Poulakou
1
1
3rd Department of Internal Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
2
1st Department of Internal Medicine, Alexandra General Hospital, 11528 Athens, Greece
3
Harvard School of Public Health, Boston, MA 02115, USA
4
Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Vaccines 2022, 10(10), 1607; https://doi.org/10.3390/vaccines10101607
Submission received: 31 August 2022 / Revised: 18 September 2022 / Accepted: 22 September 2022 / Published: 25 September 2022
(This article belongs to the Section Cancer Vaccines and Immunotherapy)

Abstract

:
Since the pandemic’s onset, a growing population of individuals has recovered from SARS-CoV-2 infection and its long-term effects in some of the convalescents are gradually being reported. Although the precise etiopathogenesis of post-acute COVID-19 sequelae (PACS) remains elusive, the mainly accepted rationale is that SARS-CoV-2 exerts long-lasting immunomodulatory effects, promotes chronic low-grade inflammation, and causes irreversible tissue damage. So far, several viruses have been causally linked to human oncogenesis, whereas chronic inflammation and immune escape are thought to be the leading oncogenic mechanisms. Excessive cytokine release, impaired T-cell responses, aberrant activation of regulatory signaling pathways (e.g., JAK-STAT, MAPK, NF-kB), and tissue damage, hallmarks of COVID-19 disease course, are also present in the tumor microenvironment. Therefore, the intersection of COVID-19 and cancer is partially recognized and the long-term effects of the virus on oncogenesis and cancer progression have not been explored yet. Herein, we present an up-to-date review of the current literature regarding COVID-19 and cancer cross-talk, as well as the oncogenic pathways stimulated by SARS-CoV-2.

1. Introduction

Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus that emerged in the city of Wuhan, China, at the end of 2019. Unlike SARS and Middle East respiratory syndrome (MERS) outbreaks that temporarily caused considerable global health consternation, SARS-CoV-2 continues to pose unprecedented challenges for healthcare systems and socioeconomic structures worldwide, since its precise pathophysiology is still poorly understood [1,2]. Lately, research has been focusing on the identification of the long-term consequences of COVID-19 on human health and the quality of life and its impact on pre-existing or emerging diseases, including cancer [3,4,5].
As expected, the systemic immunosuppressive status of cancer patients, whether caused by the disease itself or the anticancer treatment, confers an increased risk of COVID-19 compared to the general population [6,7,8]. Several studies point out that cancer patients experience a longer and severer COVID-19 disease course and higher mortality rates are also observed [6,9,10,11]. Notably, cancer type, staging, and therapeutics are determinants of SARS-CoV-2-associated morbidity and mortality [9,12,13,14].
COVID-19 and cancer present clinical and molecular similarities. Immune dysregulation, manifested either as aberrant cytokine release or impaired adaptive immune responses, tissue hypoxia and damage, oxidative stress, and signaling pathways manipulation have been recognized as key components of both oncogenesis and COVID-19 etiopathogenesis [6,7,15]. Although cancer rarely results from an isolated event, the aforementioned eventually raise concerns about SARS-CoV-2 potentiality to behave as an oncogenic virus, predispose or accelerate malignant transformation and tumor progression [4,5,6,7]. The present review aims to summarize current knowledge on the overlapping mechanisms and the molecular interplay between SARS-CoV-2 and cancer and highlight virus ability to reshape the human cancer landscape.

2. Viral Infections and Cancer

Viral infections are known to drive oncogenesis and account for an estimated 15 to 20% of cancer cases worldwide [16]. So far, seven human viruses have been implicated in oncogenesis, namely Epstein–Barr virus (EBV), hepatitis B virus (HBV), human T-lymphotropic virus 1 (HTLV-1), human papillomaviruses (HPVs), hepatitis C virus (HCV), human herpesvirus 8 (HHV-8) and Merkel cell polyomavirus (MCPyV) [16].
Viral oncology has been a major research focus and a plethora of mechanisms and pathways contributing to oncogenesis have been reported. [16,17,18]. Despite the diverse genomes and cellular tropisms, oncoviruses share common characteristics that allow them to act either as primary etiological agents behind certain cancer types or as accumulating factors that over a long period of time and in conjunction with environmental and host-cooperating events predispose the human body to cancer development and accelerate its progression [16,17]. Like other viruses, they induce epigenetic changes and usurp host signal transduction pathways associated with cell-cycle regulation and metabolism, in order to optimize the cellular conditions for viral replication and immune escape [16,17,19]. Hence, they convey cancer hallmarks on the host cells by triggering unlimited cell proliferation and replicative immortality, inhibiting apoptosis, exploiting the DNA damage response network (DDR), evading immune responses, stimulating tumor-intrinsic inflammation, and promoting angiogenesis, invasion, and metastasis [18,20].
Oncoviruses encode viral proteins that target tumor suppressors’ pathways and activate cellular oncoproteins [21,22]. The function of p53 and the retinoblastoma protein (pRB) pathway, which tightly regulate cell cycle progression and induce apoptosis, is perturbed by viral oncoproteins [21,22]. For instance, oncoproteins encoded by HPV and EBV can suppress the activity of both p53 and pRB and drive cells into the S phase of DNA synthesis, permitting viral access to host replication machinery. Likewise, HTLV-1 oncoproteins transactivator from X-gene region (Tax), basic zipper factor (HBZ) and HBV-encoded hepatitis B X-antigen (HBx) oncoproteins inactivate p53 and interrupt p53-mediated apoptosis [16,21,22]. Loss of p53 or/and pRB-induced apoptotic signaling permits viral proliferation and spread before host cell death, thus manipulating antiviral immune responses and establishing chronic, persistent inflammation [23]. All the above eventually lead to genomic instability and accumulation of mutations predisposing to oncogenesis [24]. Other host signaling pathways subverted by oncoproteins, which impel cells towards malignant transformation, include phosphoinositide-3 kinase–protein kinase–mammalian target of rapamycin (PI3K–AKT–mTOR), mitogen-activated protein kinase (MAPK), Janus kinase/signal transducers and activators of transcription (JAK/STAT), Notch, nuclear factor kappa B (NF-kB) and vascular endothelial growth factor (VEGF) signaling [16,17].
Induction of oxidative stress and interplay with DDR network are two other mechanisms employed by oncoviruses granting genomic instability. Persistent viral infection triggers chronic inflammatory responses and reactive oxygen species (ROS) production. For example, EBV nuclear antigen (EBNA)-1 upregulates the transcription of nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) oxidase resulting in ROS production, which, in turn, induces chromosomal aberrations and DNA double-strand breaks, and contributes to EBV-associated malignancy [25]. Oncoviruses are also capable of engaging DDR machinery to their advantage and establish a highly proliferative state [16]. The aforementioned gradually cause loss of genome integrity and mutations raise [26].
Furthermore, viral genome integration and epigenetic alterations have been linked to oncoviruses-mediated cancer development [17,19]. Both DNA and RNA viruses insert their viral genome into host cell DNA causing insertional mutagenesis and viral persistence in cells. The transformed cell enters an immortal state and acquires limitless replicative potential [21]. Simultaneously, insertional mutagenesis harbors tumor suppressor gene disruption and oncogenes overexpression [27]. Lastly, in order to regulate their life cycle, oncoviruses wield cellular gene expression through host DNA methylation, histone modification, chromatin remodeling, and viral-encoded non-coding RNAs (e.g., microRNAs, long non-coding RNAs, circular RNAs) and provoke host cell genome alterations [19,28].

3. Chronic Inflammation and Cancer

Chronic inflammation is considered to be one of the cardinal characteristics of tumor genesis, promotion, and progression, whereas it favors immune escape by suppressing immune surveillance and promoting the development of a tumor microenvironment (TME) that sustains cancer [29,30,31]. The potential link between inflammation and cancer was firstly proposed by Rudolf Virchow in the mid-19th century, based on the presence of inflammatory cells in tumor biopsies and the observation that tumors emerge at the sites of chronic inflammation [32]. Evidence suggests that tumor-related inflammation endows cancer cells with endless proliferative potential, resistance to growth inhibition and escape from programmed cell death, and enables angiogenesis, invasion, and metastasis [15]. Simultaneously, cancer treatment (e.g., surgery, chemotherapy, radiotherapy) can stimulate inflammatory responses and provoke tissue damage that may result in tumor recurrence and drug resistance [29].
Inflammation underlies a wide variety of processes essential for host defense against pathogens, tissue repair adaption to stress, and restoration of a homeostatic state [33]. In this context, acute inflammation has been proposed to enhance cancer immunosurveillance; it promotes the recruitment and the function of dendritic cells (DCs), which, in turn, stimulate anti-tumor inflammatory responses via diverse mechanisms, including cross-presenting tumor-associated antigens to cytotoxic T lymphocytes (CTLs) and recruitment of natural killer (NK) cells that sustain T-responses [34]. In case that acute inflammatory responses do not resolve properly or in time, chronic inflammation develops as a sequela. Within a dysregulated and persistent inflammatory state, cancer cells are able to subvert DCs function and a myriad of immunosuppressive cells (e.g., myeloid-derived suppressor cells [MDSCs], regulatory T cells [Tregs], regulatory B cells [Bregs], M2 tumor-associated macrophages [M2-TAMs], N2 tumor-associated neutrophils [N2-TANs] and T helpers 2 [Th2 cells]) is activated by cytokines, chemokines and inflammatory mediators secreted by tumor cells [29,31,35,36,37,38]. All the above eventually lead to oncogenes activation alongside tumor suppressor gene inactivation, induction of oxidative stress and DNA damage, stimulation of signaling pathways (e.g., NF-kB, K-RAS, etc.), and epigenetic alterations that reinforce cancer development [29,35,36].

Cytokine Release

In chronically inflamed tissue, upon exposure to a plethora of inflammatory mediators, tumor cells become capable of responding to these signals, thus acquiring a growth advantage [39]. So far, several cytokines have been suggested to play a pivotal role in oncogenesis [36,39]. Transforming growth factor-beta (TGF-β), a highly pleiotropic cytokine that mainly regulates wound healing and angiogenesis, and prevents uncontrolled inflammation progression, is present in TME and enhances tumor cell invasion and metastasis [40,41,42]. Tumor necrosis factor-alpha (TNF-a), a pro-inflammatory cytokine that exerts various effects (e.g., apoptosis, necrosis, angiogenesis, immune cell activation, differentiation, and cell migration) depending on the cellular context, represents a double-edged sword in tumor initiation and development, since both pro- and anti-tumoral activity has been reported [43]. Tumor necrosis factor receptor (TNFR) activation leads to the stimulation of multiple signal transduction pathways, including NF-kB signaling that generates negative regulators of apoptosis (e.g., caspase-8 (FLICE)-like inhibitory protein [FLIPL], B cell lymphoma 2 [Bcl-2], superoxide dismutase) [44,45].
Although interleukin signaling holds a key role in tumor-directed immune responses, upon an oncogenic event, it can nurture an environment that harbors cancer growth [46,47]. In brief, persistent and aberrant inflammatory responses to pathogen-associated molecular patterns (PAMPs) trigger NF-kB signaling cascade, thereby enabling inflammasomes activation and active interleukin-1β (IL-1β) release from fibroblasts, epithelial cells, DCs, monocytes and macrophages [48]; ΙL-1β activates VEGF and promotes oxidative stress and DNA damage via production of nitric oxide (ΝO) and ROS by the epithelial cells [46,49]. IL-1β alongside TGF-β regulate the differentiation of Th17 cells and IL-17 release. Under homeostatic conditions, IL-17 contributes to the wound-healing process by triggering NF-kΒ. However, during chronic inflammation, it may recruit and transactivate epidermal growth factor receptor (EGFR) in skin cells, permitting their expansion and migration, and provoke general tumor outgrowth [46,50]. Moreover, IL-33 expression has been found to be upregulated within TME; tumor-initiating cells facilitate IL-33 secretion, which consecutively recruits TAMs, MDSCs, and Tregs, therefore establishing a highly suppressive milieu for immune responses and contributing to further tumor spread [51]. In malignant cells and/or cells bearing oncogenic mutations, interleukins, such as IL-6, IL-11, and IL-22, stimulate STAT3 signaling that upholds cell proliferation, survival, stemness, epithelial–mesenchymal transition (EMT), and migration [46]; excessive STAT3 activation driven by IL-6 and IL-11 overexpression combined with oncogenic driver mutations mediate the development of solid tumors (e.g., lung, pancreatic, colon and gastric cancer) [46]. Among cytokines correlated with inflammation-induced oncogenesis, IL-6 predominantly regulates all cancer hallmarks and can be present in different stages of cancer development from promotion to metastasis [39,44,49,52,53]. IL-6 favors angiogenesis and tumor vascularization via VEGF secretion, inhibits oxidative stress and DNA damage related to cancer treatment by stimulating antioxidant and anti-apoptotic pathways, and contributes to cancer-related cachexia [46,49,53]. Furthermore, PI3K–AKT–mTOR signaling induced by IL-1 and IL-6 enhances further IL-1 and IL-6 production through metabolism shift towards glycolysis and the suppression of oxidative phosphorylation, thus aggravating inflammation-related carcinogenesis [46,54,55].

4. Immune Escape and Cancer

Cancer immunoediting serves as a conceptual framework that integrates the immune system’s dual role in tumor suppression and promotion [56,57]. It is a dynamic, multivariable process, in which robust immunological responses are produced in response to diverse tumor stimuli [58,59,60]. Immune cells (e.g., CTLs, NK cells, NKT cells, γδ T cells, and DCs) translocate in tumor sites and secrete various cytotoxic effectors, such as anti-angiogenic interferon-gamma (IFN-γ) and IL-4, perforin and granzyme [59,60]. Simultaneously, CD8+ T cells, NKs, and DCs recognize and lyse stromal or/and tumor cells through multiple mechanisms, including major histocompatibility complex (MHC)-, perforin- and tumor-necrosis factor-related apoptosis-inducing ligand (TRAIL)-dependent manner [60]. However, tumor cells apply a variety of strategies in order to debilitate cancer immunosurveillance, of which immunoselection (the selection of non-immunogenic tumor-cell variants) and immunosubvertion (the active suppression of innate and adaptive cytolytic effectors) are the most well documented [60,61,62].
Mostly present during the early stages of carcinogenesis, immunoselection is linked to antigen-processing and presenting machinery loss and desensitization to immune effectors, such as IFNs [59,60,62]. For example, downregulation or loss of human leukocyte antigen (HLA) class I molecules expression, as seen in melanomas, lung, and epithelial-cell cancers, outstrips immunological restraints and harbors immune escape [60,63,64]. Simultaneously, tumor cells interfere with molecules related to antigen presentation, such as transporter associated with antigen processing-1 (TAP-1), low-molecular-mass protein-2 (LMP2), lipoma preferred partner 7 (LPP7), and tapasin, the expression of which is deregulated during cancer development, thereby evading elimination [60,62,65]. Moreover, blockage of the granzyme B/perforin pathway through overexpression of the serine-protease inhibitor PI9, mutation of death receptors, methylation of the gene encoded caspase-8, and upregulation of FLIP are other mechanisms employed by tumor cells towards inhibition of CTL-mediated target-cell lysis [60,66,67].
Tumor cells can orchestrate a highly immunosuppressive microenvironment and immunosubvertion is thought to accompany advancing tumor growth [60,61,62]. For instance, tumor-associated myeloid cells can suppress T-cell function by overproducing NO and displaying increased arginase-1 activity [68]. Additionally, cancer cells impede CTL-mediated toxicity by releasing immunosuppressive factors, such as IL-10, TGF-β, programmed death ligand 1 (PD-L1), and prostaglandin E2 (PGE2), which further reduce immunogenicity [59,62,69]. IL-10, derived by monocytes, M2 cells, Th2 cells, mast cells, Tregs, MDSCs, and mesenchymal stem cells (MSCs), activates STAT3-mediated suppressors of cytokine signaling 3 (SOCS3) expression, which, in turn, inhibits Th1 cell proliferation and alters Th1/Th2 differentiation [59,70,71]. PD-L1, abundantly expressed in numerous human cancers, induces T cell exhaustion by stimulating PD-1 signaling [72,73]. Additionally, interdigitating DCs and tumor cells overexpress indoleamine 2,3-dioxygenase (IDO) that is known to prevent immune-mediated rejection of the tumor cells, block CD8+ T cells proliferation, and favor CD4+ apoptosis [74,75]. TAMs, CD4+ T cells, CD25+ T cells, Tregs, and MDSCs are other tumor-infiltrating immunosuppressive cells that dominate in the TME and sustain tumor progression [59]. For example, M2-TAMs are angiogenesis promoters and CTL response suppressors and have been associated with poor prognosis in patients diagnosed with breast or ovarian cancer [76]. Similarly, MDSCs abolish adaptive immune responses through induction of Treg activity and M2 differentiation, deactivation of local T cells by cytotoxic oxidizing molecules secretion, interference with T cell migration, and blockage of NKs activation [59,77,78]. Finally, within immunosuppressive TME, tumor-specific CD8+ T cells are activated at the stage of initiation of tumor growth and gradually lose their cytolytic function at the later stages, hence promoting progressive tolerance to the tumor [60,79]. Likewise, during tumor expansion, tumor-specific CD4+ T cells lose their anti-tumor activity, whereas an increase in the number of Treg cells is observed [80,81].

5. Gut Microbiota and Cancer

Gut microbiota is comprised of a heterogeneous population of commensal microorganisms (e.g., bacteria, fungi, archaea, and viruses) that colonize the intestinal tract system [82]. It performs multiple functions, including nutrient abortion, toxin metabolism, host immune system regulation, and prevention of resident pathogens expansion, whereas it is dynamically changed by internal (e.g., age, stress), dietary or environmental factors, as well as by infectious agents or antibiotic consumption [82,83,84]. The gut microbial balance is essential for homeostasis maintenance and any deviation from it, a condition known as dysbiosis, can have detrimental consequences for human health [85]. For instance, impaired gut microbiota has been associated with a plethora of human pathologies, such as inflammatory bowel disease, obesity, diabetes mellitus type II, non-alcoholic liver disease, cardio-metabolic diseases, cancer, and malnutrition [85,86].
In the context of cancer, ample evidence suggests the duality in the role of gut microbiota, since it can be either tumor-promoter or tumor-suppressor [87,88]. In fact, the relationship between microbiome and cancer is more complex than assumed; the microbiome may influence cancer progression and, in turn, cancer development may cause microbiome alterations [89,90].
Production of putative oncometabolites, perturbation of DDR network, and immune system manipulation are the leading mechanisms mediated by impaired gut microbiota towards cancer genesis and progression [89,91,92]. Within the dysbiotic gut, pathogenic bacteria produce and release a wide variety of protein virulence factors, some of which are protein toxins, that have a pro-carcinogenetic activity; they either induce DNA single- and double-strand breaks within the host’s epithelial cells or regulate crucial cellular proliferative pathways, thereby promoting a transient cell cycle arrest [92,93,94,95]. Failure to properly respond to DNA damage favors genomic instability and acquisition of malignant traits in predisposed cells [92,94,96]. Moreover, dysbiotic gut bacteria interfere with the DDR network. For example, in the case of S. flexneri and H. pylori, the secretion of Shigella virulence effector VirA and cytotoxin-associated-gene A (CagA), respectively, subverts the tumor suppressor function of p53, enhancing mutations raise during the DNA damage response in the infected cells [97,98,99]. Several pathogenic bacteria, such as H. pylori, B. fragilis, and E. faecalis, are also able to generate oxidative stress and consequently drive cell-autonomous genetic mutation [100,101].
Lastly, an impaired gut microbiome can drive inflammation-induced cancer through inflammation enhancement or inhibition of host immune responses [102]. Dysbiosis promotes the transcription of chemokine C-C chemokine ligand 5 (CCL5), which recruits a variety of innate and adaptive immune cells in the intestine, thus inducing the local activation of the IL-6 pathway and resulting in intestinal epithelial cell reprogramming [91,103]. As a consequence of local inflammation, toll-like receptors (TLRs) are upregulated by lipopolysaccharide (LPS) and other microbial products and stimulate cytokine release and the activation of signaling pathways (e.g., NF-kB, JAK/STAT3, c-Jun N-terminal kinase [JNK]) that are known to fuel tumor growth [91,104]. Moreover, commensal bacteria and their bioproducts induce inflammasomes activation and inflammasome-mediated IL-18 secretion, which ensure intestinal tissue remodeling and gastrointestinal barrier maintenance. However, within dysbiotic gut, IL-18 production is reduced due to the deficiency of inflammasomes components, which, in turn, causes larger commensal bacteria penetration and inflammation exacerbation and triggers tumor formation [89]. Finally, gut bacteria can inhibit immune effectors that prevent tumorgenesis [89]. For example, the Fap2 protein encoded by F. nucleatum interacts with T cell immunoreceptor with immunoglobulin and ITM domain (TIGIT), an inhibitory receptor present on NKs and various T cells, and impedes the NK-mediated tumor cell cytotoxicity [105].

6. Cytokine Release and Long-COVID

Cytokines play a crucial role in the evolution of infections, both microbial and viral, and determine the duration and severity of infections. Particularly for COVID-19, the course and outcome of the disease are determined by viral (mutational) and host (e.g., age, sex, comorbidities, and immunological) factors [106,107]. In SARS-CoV-2 infection, the innate and adaptive immune systems are activated following binding of the virus to the angiotensin-converting enzyme 2 (ACE2) protein of alveolar epithelial cells and subsequent production and interaction of chemokines, colony-stimulating factors, interferons, interleukins, and TNF-a. As vascular permeability increases, COVID-19 is developed [108,109]. When the immune response is dysregulated, it contributes to disease pathogenesis, as in the case of a “cytokine storm” [110,111], or hypercytokinemia, which is characterized by (a) perpetuated activation of lymphocytes and macrophages causing immune dysregulation, (b) large secretions of cytokines caused by such perpetuated activation, and (c) overwhelming systemic inflammation and multi-organ failure with high mortality [112,113]. The most important inflammatory mediators released by immune cells during the “cytokine storm” are IFN-α, IFN-γ, IL-1β, IL-6, IL-12, IL-18, IL-33, TNF-a, and TGF-β and they are associated with different clinical features of COVID-19 [106,114]. Indeed, cytokine storms correlate with the severity and progression of COVID-19 and can result in serious complications such as acute respiratory distress syndrome (ARDS) and multiple organ failure, which are the leading causes of death from the disease [115,116].
Shortly after the beginning of the COVID-19 pandemic, a phenomenon known as “long-COVID,” or post-acute sequelae of SARS-CoV-2 (PASC), appeared to medical professionals [117,118,119,120,121,122,123]. While COVID-19 symptoms are resolved in 1–4 weeks in the majority of patients, some patients complain of symptoms lasting longer than 28 days. These symptoms include fatigue, shortness of breath, headache (or “brain fog”), loss of smell (anosmia), sleep disturbances, fevers, gastrointestinal symptoms, anxiety, and depression and can persist for months and range from mild to disabling [124]. Even though a study on patients with pneumonia secondary to COVID-19 infection suggests that persistent symptoms may be attributable to “biopsychosocial” effects of COVID-19 (mainly due to the absence of residual radiographic abnormalities) [125], a number of studies have disclosed abnormal cardiopulmonary exercise testing [126], and most studies attribute the range of PASC symptoms to a number of differing pathological traits of the virus [127].
In the study by Queiroz et al., among patients in the post-COVID-19 group, subjects with PASC had higher levels of IL-17 and IL-2 and lower levels of IL-10, IL-6, and IL-4 than subjects without sequelae (i.e., without PASC symptoms/signs) [128]. The fact that pro-inflammatory cytokines were increased in the PASC group underlines the association of PASC with residual inflammation and inflammatory organ damage [129,130]. The higher levels of IL-10 and IL-4 in patients not suffering from PASC suggest better control of the inflammatory process due to increased levels of anti-inflammatory cytokines in these individuals. Furthermore, the significantly higher levels of IL-17 and IL-2 and lower levels of IL-4 and IL-10 in individuals with PASC suggest a possible “molecular signature” for PASC characterized by a Th17 inflammatory profile with a reduced anti-inflammatory response mediated by IL-4 and IL-10. In addition, since cytokines are prevalent in circulation, they are present in many different organ systems, and this explains the variety of symptoms associated with PASC. Nonetheless, since SARS-CoV-2 shows tropism for the nervous system and neurological manifestations (e.g., anosmia, ageusia, headache, stroke, Guillain–Barré syndrome, seizure, and encephalopathy) are observed in the majority of patients with acute COVID-19, microvascular inflammation in cells of the nervous system during acute infection may trigger mild symptoms of the disease [131], which may persist even after the infection has resolved.
In the study by Schultheiss et al., PASC appeared in up to 60% of the patients up to 24 months post-infection; elevated levels of IL-1β, IL-6, and TNF-a were detected in those patients with their most probable source of secretion being the monocytes and macrophages of the lung [132]. Similar results were observed in the study by Peluso et al.: higher levels of IL-6 and IFN-γ-induced protein 10 (IP-10) during early recovery from COVID-19 were associated with subsequent development of PASC; in addition, among patients with PASC, IL-6 showed a trend towards from early to late recovery [133]. Similar findings were noted in the study by Acosta-Ampudia et al., where a pro-inflammatory state was observed in patients with PASC characterized by up-regulated IFN-α, TNF-a, granulocyte colony-stimulating factor (G-CSF), IL-17A, IL-6, IL-1β, and IL-13, whereas IP-10 was decreased [134]. In yet another study, several cytokines from interferon I and III classes were highly elevated and stayed up for over 8 months [135].
The differences in the results among the previous studies could be attributed to methodological differences, e.g., different time-points of cytokine determination, population size, and characteristics (e.g., country, severity of acute COVID-19). Nonetheless, the paramount finding is a persistent and diffuse proinflammatory state and a dysregulated cellular immune response, which is compatible with two of the currently most discussed hypotheses on the immune pathogenesis of PASC: (a) ongoing immune responses against persisting virus or viral antigens and/or (b) chronic reprogramming of immune cells [128,132,133,134,135].

7. T-Cell Response and Long-COVID

The immune system is broadly divided into the innate immune system and the adaptive immune system. The adaptive immune system is important for the control of most viral infections. The three fundamental components of the adaptive immune system are B cells (the source of antibodies), CD4+ T cells (they possess a range of helper and effector functionalities), and CD8+ T cells (they kill infected cells). Adaptive immune responses are slow due to the intrinsic requirement of selecting and expanding virus-specific cells from the large pools of naive B cells and T cells: they take 6–10 days after priming to generate sufficient cells to control a viral infection, due to the inherent time demands for extensive proliferation and differentiation of naive cells into effector cells. Once sufficient populations of effector T cells (Th cells and CTLs) and effector B cells (antibody-secreting cells, known as plasmablasts and plasma cells) have proliferated and differentiated, they often work together to rapidly and specifically clear infected cells and circulating virions [136]. In a SARS-CoV-2 infection, the virus is particularly effective at avoiding or delaying triggering intracellular innate immune responses associated with type I and type III IFNs [137,138,139,140]. Without those responses, the virus initially replicates unabated and the adaptive immune responses are delayed, so that an asymptomatic infection or clinically mild disease follows; this is because the T cell and antibody responses occur relatively quickly and control the infection. Whenever impaired and delayed type I and type III IFN responses occur, the virus replicates heavily in the upper respiratory tract and lungs (due to failure of adaptive immune response priming) and a very high risk of severe or fatal COVID-19 ensues [141,142,143,144,145].
In PASC patients, the following findings have been observed: increases in antigen-specific CD4+ T cell responses to the SARS-CoV-2 S protein, antigen-specific activation in the circulating T follicular helper cells, and populations of CD8+ T cells (mainly attributed to their effector subpopulation). All these data show a prolonged T cell response magnitude [146], which eventually leads to exhaustion of T cells, as indicated by the increased expression of exhaustion markers, namely PD-1-expressing T lymphocytes [147,148,149]. Interestingly, T cell exhaustion can be reversed and T cell function can be restored by PD-1 blockade, which in turn ex vivo increases the CD4+ T cell-mediated response to SARS-CoV-2 spike and nucleocapsid peptides [147]. Another feature of PASC is the lower and more rapidly waning N-specific CD8+ T cell responses (IFNγ-/CD107a+ and IFNγ+); this lower frequency of degranulating virus-specific CD8+ T cells in individuals with PASC could be attributed either to the decreased functional capacity of these cells or dysfunction of the immune response [150]. Another feature of PASC is the significantly increased level of Tregs (CD4+ CD25+ CD127 low): this can be explained in the light of the increase in the PD-1-expressing T lymphocytes, indicating failing attempts of the immune system to control the persistent immune response [149]. Towards a better understanding of the pathophysiology of PASC, it has been suggested that T-cell subsets exhibit different dynamics, depending on the severity of the initial infection and the time since then; in severe convalescents, there is a tendency towards an exhausted/senescent state of CD4+ and CD8+ T cells and perturbances in CD4+ Tregs 3 months post-infection, a remodeling that is clearly visible at 6 months post-infection. In addition, CD8+ T cells exhibit a high proportion of CD57+ terminal effector cells, together with a significant decrease in the naive cell population, augmented granzyme B and IFN-γ production, and unresolved inflammation 6 months post-infection. On the contrary, mild convalescents showed increased naive Tregs, and decreased central memory and effector memory CD4+ Treg subsets [151].
Apart from circulating T and B cells, there are tissue-resident memory cells that reside in the peripheral non-lymphoid tissue [152,153]; those cells provide immediate and superior immunity against viral reinfections [154]. However, dysregulated lung-resident T cell responses may cause chronic lung inflammation and fibrosis after respiratory viral infection [155,156]. In line with those findings, Cheon et al. have found that exuberant CD8+ T cell responses may be connected to the development of chronic lung sequelae after the resolution of acute COVID-19 infection in aged individuals [157].

8. Tissue Damage and Long-COVID

The recent coronavirus outbreaks (SARS epidemic of 2003 and MERS outbreak of 2012) have demonstrated that persistent respiratory symptoms and radiographic abnormalities continue beyond the period of acute illness. In a considerable number of these patients post viral sequelae in the form of lung fibrosis have been reported. Given the genetic homology between SARS-CoV-2 with both SARS and MERS and the increased infected population worldwide during the current pandemic, it can be assumed that an increased likelihood of post-COVID-19 long-term pulmonary complications, and particular lung fibrosis, is expected [158]. In COVID-19 survivors, more than one-third describe respiratory symptoms in the context of PASC; of those greater at risk are females, who had severe acute COVID-19 disease and/or required invasive or noninvasive ventilation.
Pulmonary fibrosis can occur in the setting of maladaptive resolution of lung injury or exaggeration of the reparative process [159]. Fibroblasts respond to alveolar injury and secrete the extracellular matrix; during acute infection and the post-COVID period they are overreactive due to the upregulation of inflammatory cytokines and this results in distortion and remodeling of the pulmonary architecture. Monocyte-derived alveolar macrophages (MoAMs) are profibrotic and stimulate and form reciprocal circuits with fibroblasts [160]; the maladaptive repair of the alveolar injury results in a positive feedback loop that results in filling the alveolar interstitium and alveoli with matrix proteins and fibroblasts. In this setting, PASC-pulmonary fibrosis may develop following a prolonged phenotype of a slowly unfolding, spatially limited inflammatory alveolitis [161]. The prolonged exposure of profibrotic monocytes to elevated levels of cytokines results in intense interaction with fibroblasts to promote fibrotic repair processes. Altogether there is concern that the protracted nature of acute COVID-19, persistence of high levels of cytokines, monocyte/macrophage and T-cell circuits stimulating potential circuits between monocytes and fibroblasts, coupled with the extended duration of mechanical ventilation, could combine to promote a milieu in the alveoli with an increased likelihood of PASC-pulmonary fibrosis [134]. SARS-CoV-2 can also impose lung tissue damage by impairing microcirculation with a number of mechanisms [162]: capillary pericytes are damaged in COVID-19, thus hindering lung repair and neo-angiogenesis; endothelial cells (EC) become apoptotic and protrude in the lumen; capillary endothelium glycocalyces’ are shed; capillaries are obstructed by circulating neutrophils, by microthrombi formation or by fibrin-rich amyloids formation which are resistant to fibrinolysis [163,164].
Blood microcirculation disturbances during COVID-19 and subsequent tissue damage that may contribute to PASC can also affect other organs [162]. In the heart, endothelial infection results in EC swelling in small vessels and scattered necrosis of individual myocytes. In the brain, infection of subcortical white matter microvessel endothelium is associated with hyper-acute, microscopic ischemic lesions, and older ischemic and hemorrhagic microscopic lesions. In the skin, EC infection is associated with endothelial swelling and in some patients with thrombosis and fibrinoid necrosis in surrounding tissue. Keeping in mind that erythrocyte diameters are bigger than the capillary lumen, endothelial damage is likely to disturb capillary flow patterns in all affected organs, resulting in extreme shunting of oxygenated blood through the shortest capillary pathways. Thus, it has been proposed that COVID-19 is an endothelial disease and PASC may have its origins in subsequent tissue hypoxia [162,165].
Tissue damage has been disclosed in many organs of convalescent people, even in young ones, mostly free of risk factors for severe COVID-19: more than half of them had at least one radiographic abnormality of the lungs, heart, liver, pancreas, kidneys, or spleen and these abnormalities could persist for at least 2–3 month after hospital discharge [166]. Although not a pure tissue damage, gut microbiome disruption (i.e., gut dysbiosis) has been observed among patients with COVID-19 and persists even after disease resolution. Gut dysbiosis is also correlated with increased COVID-19 severity and inflammatory biomarkers and prolonged SARS-CoV-2 [167]. It has also been postulated that the gut microbiome modulates the neurotransmitter circuitries in the gut and brain via the microbiota gut–brain axis.

9. Gut Microbiota and Long-COVID

COVID-19 is an infection that primarily affects the human respiratory tract. Nonetheless, around 5–33% of COVID-19 patients have gastrointestinal (GI) symptoms, including diarrhea, nausea, and vomiting [114,168,169], and SARS-CoV-2 has been detected in stool samples and anal swabs [170,171]. These findings are in line with the enhanced expression of ACE2 in the GI tract, rendering the digestive tract an extra-pulmonary site for SARS-CoV-2 infection [172]. Since the GI tract is the largest immune organ in humans, playing critical roles in combating infections of pathogens [173], it can be assumed that the trillions of microorganisms inside the gut of humans are regulating host immunity [174]. A number of studies have disclosed that the gut microbiome ecology is broadly altered in patients with COVID-19 and that the gut microbiome configurations are associated with immune responses and disease presentations in COVID-19, in both the short term and long term [167,175,176,177,178], which in return influence the human host’s health.

9.1. Compositional Changes of the Gut Bacterial Microbiome

In the study by Zuo T et al. [179], it was shown that during acute COVID-19 the gut bacterial microbiome of patients was characterized by significant depletion of beneficial commensals and enrichment of opportunistic pathogens in the gut compared with healthy controls [177]. Interestingly, these alterations in the bacterial microbiome ecology persisted even after the clearance of SARS-CoV-2 from the respiratory tract [177]. A similar pattern of gut microbiome dysbiosis in COVID-19 patients was disclosed in the studies by Tang et al. and Gu et al., where the (beneficial) butyrate-producing bacteria were significantly decreased in patients with COVID-19 with a corresponding abundance of the common opportunistic pathogens (e.g., Enterobacteriaceae, Enterococcus, Streptococcus, etc.) [180,181].
The human digestive tract is also affected in the long term by COVID-19, rendering it one of the systems affected during PASC [182]: prolonged shedding of viral RNA in stool specimens up to 42 days and the presence of SARS-CoV-2 virus in the gut epithelium up to 90 days after disease resolution [183,184]. Concordantly, long-lasting gut microbiome dysbiosis is also consistently observed in subjects who recovered from COVID-19 [167,177,185,186,187,188], implying that the gut microbiome is closely linked to host health in a post-COVID-19 age [189]. In a six-month follow-up study on the gut microbiome of patients with COVID-19, significant decreases in the richness of the gut microbiome were observed across the acute, convalescence, and post-convalescence phases of COVID-19 [185]. In addition, COVID-19 patients had a significantly reduced gut bacterial diversity [181,190]. It should be noted that microbial diversity is a critical determinant of microbial ecosystem stability [191] because stable ecosystems provide colonization resistance to opportunistic pathogens [192]. Therefore, the reduction in gut microbiota diversity and richness may somewhat contribute to the expansion of opportunistic bacteria and have a long-term impact on patients with COVID-19 [193], especially in the light of the persistence of certain perturbations even after disease resolution [177,181] and the presence of different PASC symptoms (including respiratory and neuropsychiatric symptoms) [194,195].

9.2. The Gut Mycobiome in COVID-19

The human gut also harbors a large number of fungi, known as the gut mycobiome. The gut fungi have been demonstrated to be causally implicated in microbiome assembly, ecology, and immune development [196,197]. In the study of Zuo et al., patients with COVID-19 also presented with alterations in the gut mycobiome, characterized by enrichment of Candida albicans and highly heterogeneous mycobiome configurations [176]. The abundance of opportunistic fungal pathogens, Candida albicans, Candida auris, and Aspergillus lineages, was increased in the feces of COVID-19 patients during the disease course [176,198]. Fungal pathogens associated with pneumonia and respiratory symptoms, Aspergillus flavus and Aspergillus niger, were detected in the fecal samples from a subset of patients with COVID-19, even after disease resolution [176]. Unstable gut mycobiomes and prolonged dysbiosis persisted in a significant proportion (~30%) of COVID-19 patients [176]. These data suggest a gut mycobiome dysbiosis in COVID-19 and its relationship with a systemic dysregulation of host immunity.
Overall, such fungal bloom in the gut of patients with COVID-19 is likely a result of SARS-CoV-2 infection. Secondary fungal infection or co-infection in patients with COVID-19 during the pandemic was frequently observed [199,200,201]. Candida albicans has been shown to impair gut microbiome assembly, including gut microbiome reassembly after disruption by antibiotics and inflammation [202,203]. This detrimental effect of Candida albicans and Aspergillus lineages in the microbiome reassembly has been demonstrated in a fecal microbiota transplantation (FMT) study in Clostridium difficile infection, where fungi impair colonization of donor bacteria into recipients [203]. These studies suggest a crucial role of gut fungi in the gut microbiome ecology, revealing the centrality of simple microbial–microbial interactions in shaping host-associated microbiota. Moreover, gut colonization by Candida albicans can aggravate inflammation in the gut and non-gut tissues [204,205]. Therefore, the opportunistic expansion of certain fungi in COVID-19 patients potentially has a deleterious role on gut microbiome assembly, where a persistent gut microbiome dysbiosis is consistently seen even after disease resolution and hospital discharge.

9.3. The Gut Virome in COVID-19

In addition to bacteria and fungi, the human gut also harbors an immense diversity of viruses collectively known as the gut virome [206,207]. The virome consists of both RNA and DNA viruses that chronically infect their eukaryotic (humans, animals, and plants) and prokaryotic hosts (bacteria) [207]. The gut virome serves to modulate the ecology of the co-resident gut bacterial microbiota as well as the immunity of the mammalian host [206].
Even in the absence of GI symptoms and after respiratory clearance of SARS-CoV-2, an active presence of SARS-CoV-2 in the fecal RNA virome was found in 47% of patients with COVID-19 [208]. Meanwhile, more eukaryotic viruses were enriched in the feces of COVID-19 patients [175], as a result of SARS-CoV-2 infection. The eukaryotic viruses may harness the immune dysfunction of the host after SARS-CoV-2 infection to expand [207]. Thus, the gut virome in COVID-19 showed more stress-, inflammation-, and virulence-associated gene coding capacities [175]. Phages associated with opportunistic bacteria were also enriched in the gut virome and their increase was proportional to the increase in their host bacteria as a result of SARS-CoV-2 infection [180,186]; the co-expansion of phage and their respective bacteria has also been reported in gut inflammation [209,210].
Gut inflammation per se is able to boost bacteriophage transfer between bacteria [211]. Therefore, the alterations in the ecology of gut virome, particularly in the bacteriophage community, are at least partly caused by the alterations of the bacterial microbiome under the influence of SARS-CoV-2 infection and the subsequent immune dysfunction. Similarly, the gut virome dysbiosis persisted along with the dysbiosis of the gut bacterial microbiome, even after disease resolution of COVID-19 [175]. Thus, the strong correlation between the composition of virome and bacterial microbiome observed in COVID-19 patients was no surprise [212].
Along with the phenotypic changes in the host in SARS-CoV-2 infection, the gut microbiome (consisting of the bacterial microbiome, mycobiome, and virome) is broadly altered in COVID-19. Moreover, subsequent blooms of opportunistic bacteria, fungi, and viruses under the circumstances of SARS-CoV-2 infection and quiescent/overt gut inflammation in COVID-19 result in decreases in microbiome diversity. In conjunction with the impaired host immunity, such alterations may hinder the reassembly of the gut microbiome post-COVID-19, resulting in a significant weakening of the intricate microbiome ecological network in a steady state and contributing to the development of PASC.

10. Oncogenic Pathways and SARS-CoV-2

Growing evidence suggests that SARS-CoV-2 can stimulate oncogenic pathways, hence raising concerns about its potentiality to reshape the human cancer landscape. Interplay with the DDR network, aberrant cytokine release, impaired T-cell responses, manipulation of regulatory signaling pathways, oxidative stress, and tissue hypoxia are the main mechanisms employed by SARS-CoV-2 that are suspected to contribute to carcinogenesis as a COVID-19 sequela (Figure 1) [213,214,215].
As discussed above, viruses interfere with the DDR network by recruiting DNA damage proteins to viral replication centers and regulating apoptosis via repair pathways suppression. As such, SARS-CoV-2 non-structural protein 1 (NSP1) has been shown to interact with all four members of DNA polymerase alpha (Pol a), an essential complex that is involved in the initiation of DNA replication and couples cycle cell progression to DRR [216]. Like other RNA viruses, in order to be replicated and escape immune surveillance, SARS-CoV-2 manipulates directly or indirectly mitochondrial metabolism, a key component of cellular homeostasis maintenance and cancer pathophysiology, by interfering with mitochondrial proteins [217]. Gordon et al. reported the interaction of SARS-CoV-2 ORF9c, a membrane-associated protein enabling immune evasion, with electron transport chain (ETC) components, suggesting a possible role as an oxidative phosphorylation regulator [216]. It is worth mentioning that ETC induces oxidative stress, as well. SARS-CoV-2 NSP8 and NSP5 have been proposed by Tutuncuoglou et al. to interact with histone methyltransferase NSD2 and HDAC2, respectively, and the latter have been linked to several oncogenic pathways [213,218]. For example, both of them are involved in NF-kB activation by proinflammatory cytokines, whereas NSD2 overexpression results in oncogenic RAS-driven transcription in lung cancer cells, and HDAC2 is a coactivator of the tumor suppressor p53 [219,220]. Lastly, the SARS-CoV NSP3 and NSP15 have been implicated in the degradation of p53 and pRb, respectively, and the SARS-CoV-2 S2 subunit has been demonstrated in silico to interact with p53 and BRCA 1/2, granting genomic instability [221,222,223]
Immune dysregulation, a hallmark of COVID-19 course and severity, is mainly orchestrated by cytokines and chemokines (e.g., IL-6, IL-1β, IL-8, IL-18, TNF-a) that are identified as tumorigenesis drivers [35]. Additionally, multiple signaling pathways (e.g., IL-6/JAK/STAT, NF-kΒ, IFN-Ι), which are prominent in oncogenesis, have been proved to contribute to COVID-19 etiopathogenesis, as well [214]. For instance, aberrant IL-6/JAK/STAT-3 signaling potentiates inflammatory responses in COVID-19, therefore aggravating disease severity; in the context of cancer, it attenuates anti-tumor T-cell mediated responses and favors tumor growth, survival, invasiveness, and or/metastasis [224]. Furthermore, the NF-kB pathway has been documented to be hyper-activated in moderately or critically ill COVID-19 patients and it is characterized as a crosstalk mediator between inflammation and cancer [225]. The NF-kB pathway is capable of remodeling the immune landscape to benefit tumor proliferation, inhibiting apoptosis and attracting angiogenesis [225]. Impaired IFN signaling, which underlies severe COVID-19, employs pro-tumoral properties and it is assumed as a key mechanism in tumor proliferation [226]. IFNs have been reported to trigger the NF-kB pathway, protect cells against apoptotic stimuli and foment angiogenesis [226,227,228]. Interestingly, IFNs harbor immunoevasion by decreasing sensitivity to NK cells and downregulating tumor-associated antigen presentation and contribute to drug resistance via IFN-related DNA damage-resistant signature (IRDS) induction [226,229].
It is no surprise that SARS-CoV-2 and tumor cells interfere with similar signaling pathways to their advantage. mTOR pathway displays fundamental functions (e.g., proliferation, metabolism, protein synthesis, autophagy, and apoptosis) and it is perturbated in cancer [230]. SARS-CoV-2 is also known to exploit mTOR signaling and at least seven targets have been identified so far [231,232]. The Notch pathway, a highly conserved signaling pathway that controls multiple cell differentiation processes, is an attractive target in COVID-19 and its overexpression enhances viral entry and the manifestation of inflammatory, coagulopathic and hypoxic events [233]. In addition, it is one of the most commonly activated signaling pathways in different cancer types and emerging data highlight its contribution to invasion, tumor heterogeneity, angiogenesis, or tumor cell dormancy within solid cancer tissues [234,235]. The p38 MAPK pathway allows cells to interpret and respond to various signals and stimuli, such as DNA damaging genotoxic agents, inflammatory cytokines, oxidative stress, and heat shock [236]. In COVID-19, it has been stated as disproportionately upregulated as a result of ACE2 activity loss upon viral entry or/and direct viral activation of p38 MAPK; the activated p38 MAPK pathway facilitates viral entry via ACE2 endocytosis and predisposes for pathological processes, such as inflammation and thrombosis [237]. Dysregulated p38 MAPK signaling is implicated in a wide range of cancers and it oversees the expression and the deposition of pro-angiogenic and pro-tumorigenic factors aiding in carcinoma growth, metastasis, and treatment resistance [236,238].
Persistent tissue hypoxia, derived by hyperinflammation or SARS-CoV-2-induced ACE2 depletion, promotes oxidative stress and genomic instability. Within predisposed cells, the proteomic and genomic changes may initiate cell cycle arrest and apoptosis evasion, while they facilitate tumor overgrowth, invasion, and metastasis [239,240]. In the case of SARS-CoV-2-mediated oxidative stress, NO overproduction alongside bradykinin degradation stimulates EGFR signaling, which is commonly upregulated in diverse carcinoma types [231].

11. Conclusions

Long COVID or PASC is an emerging condition that has attracted public interest, and scientists are gradually piecing together the puzzle of the lingering disorder that affects some of the convalescents. Although little is known about its precise etiopathogenesis, it seems that common pathogenic mechanisms intervene in PASC occurrence and oncogenesis, as depicted in Figure 1. So far, SARS-CoV-2 has not been proven to integrate into the host genome and act as a traditional oncovirus. Nevertheless, by exploiting host immunity and stimulating signaling and oncogenic pathways, it raises concerns about his ability to establish an oncogenic microenvironment. Simultaneously, since COVID-19 vaccination has been suggested to reduce the likelihood of PASC, of interest is its potential involvement in the suppression of the oncogenic pathways triggered by SARS-CoV-2. Future studies designed to further explore the intersection between SARS-CoV-2 and cancer and the disposition of the patients experiencing long-COVID for cancer are needed.

Author Contributions

Conceptualization, V.R., T.T. and G.P.; writing—original draft preparation, V.R. and T.T.; writing—review and editing, V.R., T.T., I.A.V., N.K.S., P.L. and G.P.; supervision, G.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Guarner, J. Three Emerging Coronaviruses in Two Decades. Am. J. Clin. Pathol. 2020, 153, 420–421. [Google Scholar] [CrossRef] [PubMed]
  2. Lopez-Leon, S.; Wegman-Ostrosky, T.; Perelman, C.; Sepulveda, R.; Rebolledo, P.A.; Cuapio, A.; Villapol, S. More than 50 long-term effects of COVID-19: A systematic review and meta-analysis. Sci. Rep. 2021, 11, 16144. [Google Scholar] [CrossRef] [PubMed]
  3. Yelin, D.; Wirtheim, E.; Vetter, P.; Kalil, A.C.; Bruchfeld, J.; Runold, M.; Guaraldi, G.; Mussini, C.; Gudiol, C.; Pujol, M.; et al. Long-term consequences of COVID-19: Research needs. Lancet Infect. Dis. 2020, 20, 1115–1117. [Google Scholar] [CrossRef]
  4. Stingi, A.; Cirillo, L. SARS-CoV-2 infection and cancer: Evidence for and against a role of SARS-CoV-2 in cancer onset. BioEssays 2021, 43, e2000289. [Google Scholar] [CrossRef]
  5. Alpalhão, M.; Ferreira, J.A.; Filipe, P. Persistent SARS-CoV-2 infection and the risk for cancer. Med. Hypotheses 2020, 143, 109882. [Google Scholar] [CrossRef]
  6. Bora, V.R.; Patel, B.M. The Deadly Duo of COVID-19 and Cancer! Front. Mol. Biosci. 2021, 8, 643004. [Google Scholar] [CrossRef]
  7. Zong, Z.; Wei, Y.; Ren, J.; Zhang, L.; Zhou, F. The intersection of COVID-19 and cancer: Signaling pathways and treatment implications. Mol. Cancer 2021, 20, 76. [Google Scholar] [CrossRef]
  8. Al-Quteimat, O.M.; Amer, A.M. The Impact of the COVID-19 Pandemic on Cancer Patients. Am. J. Clin. Oncol. 2020, 43, 452–455. [Google Scholar] [CrossRef]
  9. Robilotti, E.V.; Babady, N.E.; Mead, P.A.; Rolling, T.; Perez-Johnston, R.; Bernardes, M.; Bogler, Y.; Caldararo, M.; Figueroa, C.J.; Glickman, M.S.; et al. Determinants of COVID-19 disease severity in patients with cancer. Nat. Med. 2020, 26, 1218–1223. [Google Scholar] [CrossRef]
  10. Rugge, M.; Zorzi, M.; Guzzinati, S. SARS-CoV-2 infection in the Italian Veneto region: Adverse outcomes in patients with cancer. Nat. Cancer 2020, 1, 784–788. [Google Scholar] [CrossRef]
  11. Russell, B.; Moss, C.L.; Shah, V.; Ko, T.K.; Palmer, K.; Sylva, R.; George, G.; Monroy-Iglesias, M.J.; Patten, P.; Ceesay, M.M.; et al. Risk of COVID-19 death in cancer patients: An analysis from Guy’s Cancer Centre and King’s College Hospital in London. Br. J. Cancer 2021, 125, 939–947. [Google Scholar] [CrossRef] [PubMed]
  12. Passamonti, F.; Cattaneo, C.; Arcaini, L.; Bruna, R.; Cavo, M.; Merli, F.; Angelucci, E.; Krampera, M.; Cairoli, R.; Della Porta, M.G.; et al. Clinical characteristics and risk factors associated with COVID-19 severity in patients with haematological malignancies in Italy: A retrospective, multicentre, cohort study. Lancet Haematol. 2020, 7, e737–e745. [Google Scholar] [CrossRef]
  13. Luo, J.; Rizvi, H.; Preeshagul, I.R.; Egger, J.V.; Hoyos, D.; Bandlamudi, C.; McCarthy, C.G.; Falcon, C.J.; Schoenfeld, A.J.; Arbour, K.C.; et al. COVID-19 in patients with lung cancer. Ann. Oncol. 2020, 31, 1386–1396. [Google Scholar] [CrossRef] [PubMed]
  14. Derosa, L.; Melenotte, C.; Griscelli, F.; Gachot, B.; Marabelle, A.; Kroemer, G.; Zitvogel, L. The immuno-oncological challenge of COVID-19. Nat. Cancer 2020, 1, 946–964. [Google Scholar] [CrossRef]
  15. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  16. Krump, N.A.; You, J. Molecular mechanisms of viral oncogenesis in humans. Nat. Rev. Microbiol. 2018, 16, 684–698. [Google Scholar] [PubMed]
  17. Kellogg, C.; Kouznetsova, V.L.; Tsigelny, I.F. Implications of viral infection in cancer development. Biochim. Biophys. Acta Rev. Cancer 2021, 1876, 188622. [Google Scholar] [CrossRef]
  18. Tempera, I.; Lieberman, P.M. Oncogenic Viruses as Entropic Drivers of Cancer Evolution. Front. Virol. 2021, 1, 753366. [Google Scholar] [CrossRef]
  19. Pietropaolo, V.; Prezioso, C.; Moens, U. Role of Virus-Induced Host Cell Epigenetic Changes in Cancer. Int. J. Mol. Sci. 2021, 22, 8346. [Google Scholar] [CrossRef]
  20. Mesri, E.A.; Feitelson, M.A.; Munger, K. Human viral oncogenesis: A cancer hallmarks analysis. Cell Host Microbe 2014, 15, 266–282. [Google Scholar] [CrossRef] [Green Version]
  21. Akram, N.; Imran, M.; Noreen, M.; Ahmed, F.; Atif, M.; Fatima, Z.; Bilal Waqar, A. Oncogenic Role of Tumor Viruses in Humans. Viral Immunol. 2017, 30, 20–27. [Google Scholar] [CrossRef] [PubMed]
  22. McLaughlin-Drubin, M.E.; Munger, K. Viruses associated with human cancer. Biochim. Biophys. Acta 2008, 1782, 127–150. [Google Scholar] [CrossRef] [PubMed]
  23. Morales-Sánchez, A.; Fuentes-Pananá, E.M. Human viruses and cancer. Viruses 2014, 6, 4047–4079. [Google Scholar] [CrossRef] [PubMed]
  24. Moore, P.S.; Chang, Y. Why do viruses cause cancer? Highlights of the first century of human tumour virology. Nat. Rev. Cancer 2010, 10, 878–889. [Google Scholar] [CrossRef]
  25. Gruhne, B.; Sompallae, R.; Marescotti, D.; Kamranvar, S.A.; Gastaldello, S.; Masucci, M.G. The Epstein-Barr virus nuclear antigen-1 promotes genomic instability via induction of reactive oxygen species. Proc. Natl. Acad. Sci. USA 2009, 106, 2313–2318. [Google Scholar] [CrossRef] [PubMed]
  26. Weitzman, M.D.; Weitzman, J.B. What’s the damage? The impact of pathogens on pathways that maintain host genome integrity. Cell Host Microbe 2014, 15, 283–294. [Google Scholar] [CrossRef]
  27. Desfarges, S.; Ciuffi, A. Viral Integration and Consequences on Host Gene Expression. In Viruses: Essential Agents of Life; Witzany, G., Ed.; Springer: Dordrecht, The Netherlands, 2012; pp. 147–175. [Google Scholar]
  28. Soliman, S.H.A.; Orlacchio, A.; Verginelli, F. Viral Manipulation of the Host Epigenome as a Driver of Virus-Induced Oncogenesis. Microorganisms 2021, 9, 1179. [Google Scholar] [CrossRef]
  29. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef]
  30. Colotta, F.; Allavena, P.; Sica, A.; Garlanda, C.; Mantovani, A. Cancer-related inflammation, the seventh hallmark of cancer: Links to genetic instability. Carcinogenesis 2009, 30, 1073–1081. [Google Scholar] [CrossRef]
  31. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef]
  32. Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
  33. Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef] [PubMed]
  34. Chan, C.W.; Housseau, F. The ‘kiss of death’ by dendritic cells to cancer cells. Cell Death Differ. 2008, 15, 58–69. [Google Scholar] [CrossRef] [PubMed]
  35. Zhao, H.; Wu, L.; Yan, G.; Chen, Y.; Zhou, M.; Wu, Y.; Li, Y. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduct. Target. Ther. 2021, 6, 263. [Google Scholar] [CrossRef]
  36. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef]
  37. DeVito, N.C.; Plebanek, M.P.; Theivanthiran, B.; Hanks, B.A. Role of Tumor-Mediated Dendritic Cell Tolerization in Immune Evasion. Front. Immunol. 2019, 10, 2876. [Google Scholar] [CrossRef]
  38. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
  39. Piotrowski, I.; Kulcenty, K.; Suchorska, W. Interplay between inflammation and cancer. Rep. Pract. Oncol. Radiother. 2020, 25, 422–427. [Google Scholar] [CrossRef]
  40. Prud’homme, G.J. Pathobiology of transforming growth factor beta in cancer, fibrosis and immunologic disease, and therapeutic considerations. Lab. Investig. 2007, 87, 1077–1091. [Google Scholar] [CrossRef]
  41. Yang, L.; Huang, J.; Ren, X.; Gorska, A.E.; Chytil, A.; Aakre, M.; Carbone, D.P.; Matrisian, L.M.; Richmond, A.; Lin, P.C.; et al. Abrogation of TGF beta signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell 2008, 13, 23–35. [Google Scholar] [CrossRef] [Green Version]
  42. Massagué, J. TGFbeta in Cancer. Cell 2008, 134, 215–230. [Google Scholar] [CrossRef] [PubMed]
  43. Wajant, H. The role of TNF in cancer. Results Probl. Cell Differ. 2009, 49, 1–15. [Google Scholar] [PubMed]
  44. Grivennikov, S.I.; Karin, M. Inflammatory cytokines in cancer: Tumour necrosis factor and interleukin 6 take the stage. Ann. Rheum. Dis. 2011, 70, i104–i108. [Google Scholar] [CrossRef] [PubMed]
  45. Balkwill, F. Tumour necrosis factor and cancer. Nat. Rev. Cancer 2009, 9, 361–371. [Google Scholar] [CrossRef] [PubMed]
  46. Briukhovetska, D.; Dörr, J.; Endres, S.; Libby, P.; Dinarello, C.A.; Kobold, S. Interleukins in cancer: From biology to therapy. Nat. Rev. Cancer 2021, 21, 481–499. [Google Scholar] [CrossRef]
  47. Mantovani, A.; Dinarello, C.A.; Molgora, M.; Garlanda, C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity 2019, 50, 778–795. [Google Scholar] [CrossRef] [PubMed]
  48. Lamkanfi, M.; Dixit, V.M. Mechanisms and functions of inflammasomes. Cell 2014, 157, 1013–1022. [Google Scholar] [CrossRef]
  49. Qian, S.; Golubnitschaja, O.; Zhan, X. Chronic inflammation: Key player and biomarker-set to predict and prevent cancer development and progression based on individualized patient profiles. EPMA J. 2019, 10, 365–381. [Google Scholar] [CrossRef]
  50. Chen, X.; Cai, G.; Liu, C.; Zhao, J.; Gu, C.; Wu, L.; Hamilton, T.A.; Zhang, C.J.; Ko, J.; Zhu, L.; et al. IL-17R-EGFR axis links wound healing to tumorigenesis in Lrig1+ stem cells. J. Exp. Med. 2019, 216, 195–214. [Google Scholar] [CrossRef]
  51. Choi, M.R.; Sosman, J.A.; Zhang, B. The Janus Face of IL-33 Signaling in Tumor Development and Immune Escape. Cancers 2021, 13, 3281. [Google Scholar] [CrossRef]
  52. Hirano, T. IL-6 in inflammation, autoimmunity and cancer. Int. Immunol. 2021, 33, 127–148. [Google Scholar] [CrossRef] [PubMed]
  53. Kumari, N.; Dwarakanath, B.S.; Das, A.; Bhatt, A.N. Role of interleukin-6 in cancer progression and therapeutic resistance. Tumour Biol. 2016, 37, 11553–11572. [Google Scholar] [CrossRef] [PubMed]
  54. Biswas, S.K. Metabolic Reprogramming of Immune Cells in Cancer Progression. Immunity 2015, 43, 435–449. [Google Scholar] [CrossRef]
  55. Tannahill, G.M.; Curtis, A.M.; Adamik, J.; Palsson-McDermott, E.M.; McGettrick, A.F.; Goel, G.; Frezza, C.; Bernard, N.J.; Kelly, B.; Foley, N.H.; et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 2013, 496, 238–242. [Google Scholar] [CrossRef]
  56. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef] [PubMed]
  57. Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef]
  58. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The three es of cancer immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360. [Google Scholar] [CrossRef]
  59. Lin, C.F.; Lin, C.M.; Lee, K.Y.; Wu, S.Y.; Feng, P.H.; Chen, K.Y.; Chuang, H.C.; Chen, C.L.; Wang, Y.C.; Tseng, P.C.; et al. Escape from IFN-γ-dependent immunosurveillance in tumorigenesis. J. Biomed. Sci. 2017, 24, 10. [Google Scholar] [CrossRef]
  60. Zitvogel, L.; Tesniere, A.; Kroemer, G. Cancer despite immunosurveillance: Immunoselection and immunosubversion. Nat. Rev. Immunol. 2006, 6, 715–727. [Google Scholar] [CrossRef]
  61. Kim, S.K.; Cho, S.W. The Evasion Mechanisms of Cancer Immunity and Drug Intervention in the Tumor Microenvironment. Front. Pharmacol. 2022, 13, 868695. [Google Scholar] [CrossRef]
  62. Beatty, G.L.; Gladney, W.L. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res. 2015, 21, 687–692. [Google Scholar] [CrossRef]
  63. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef]
  64. So, T.; Takenoyama, M.; Mizukami, M.; Ichiki, Y.; Sugaya, M.; Hanagiri, T.; Sugio, K.; Yasumoto, K. Haplotype loss of HLA class I antigen as an escape mechanism from immune attack in lung cancer. Cancer Res. 2005, 65, 5945–5952. [Google Scholar] [CrossRef] [PubMed]
  65. Atkins, D.; Breuckmann, A.; Schmahl, G.E.; Binner, P.; Ferrone, S.; Krummenauer, F.; Störkel, S.; Seliger, B. MHC class I antigen processing pathway defects, ras mutations and disease stage in colorectal carcinoma. Int. J. Cancer 2004, 109, 265–273. [Google Scholar] [CrossRef] [PubMed]
  66. Ochsenbein, A.F. Immunological ignorance of solid tumors. Semin. Immunopathol. 2005, 27, 19–35. [Google Scholar] [CrossRef] [PubMed]
  67. Medema, J.P.; de Jong, J.; Peltenburg, L.T.; Verdegaal, E.M.; Gorter, A.; Bres, S.A.; Franken, K.L.; Hahne, M.; Albar, J.P.; Melief, C.J.; et al. Blockade of the granzyme B/perforin pathway through overexpression of the serine protease inhibitor PI-9/SPI-6 constitutes a mechanism for immune escape by tumors. Proc. Natl. Acad. Sci. USA 2001, 98, 11515–11520. [Google Scholar] [CrossRef]
  68. Bronte, V.; Zanovello, P. Regulation of immune responses by L-arginine metabolism. Nat. Rev. Immunol. 2005, 5, 641–654. [Google Scholar] [CrossRef]
  69. Wu, A.A.; Drake, V.; Huang, H.S.; Chiu, S.; Zheng, L. Reprogramming the tumor microenvironment: Tumor-induced immunosuppressive factors paralyze T cells. Oncoimmunology 2015, 4, e1016700. [Google Scholar] [CrossRef]
  70. Yu, C.R.; Mahdi, R.M.; Ebong, S.; Vistica, B.P.; Gery, I.; Egwuagu, C.E. Suppressor of cytokine signaling 3 regulates proliferation and activation of T-helper cells. J. Biol. Chem. 2003, 278, 29752–29759. [Google Scholar] [CrossRef]
  71. Egwuagu, C.E.; Yu, C.R.; Zhang, M.; Mahdi, R.M.; Kim, S.J.; Gery, I. Suppressors of cytokine signaling proteins are differentially expressed in Th1 and Th2 cells: Implications for Th cell lineage commitment and maintenance. J. Immunol. 2002, 168, 3181–3187. [Google Scholar] [CrossRef] [Green Version]
  72. Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef] [PubMed]
  73. Pauken, K.E.; Wherry, E.J. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. 2015, 36, 265–276. [Google Scholar] [CrossRef] [PubMed]
  74. Uyttenhove, C.; Pilotte, L.; Théate, I.; Stroobant, V.; Colau, D.; Parmentier, N.; Boon, T.; Van den Eynde, B.J. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat. Med. 2003, 9, 1269–1274. [Google Scholar] [CrossRef]
  75. Terness, P.; Bauer, T.M.; Röse, L.; Dufter, C.; Watzlik, A.; Simon, H.; Opelz, G. Inhibition of allogeneic T cell proliferation by indoleamine 2,3-dioxygenase-expressing dendritic cells: Mediation of suppression by tryptophan metabolites. J. Exp. Med. 2002, 196, 447–457. [Google Scholar] [CrossRef] [PubMed]
  76. Allavena, P.; Sica, A.; Solinas, G.; Porta, C.; Mantovani, A. The inflammatory micro-environment in tumor progression: The role of tumor-associated macrophages. Crit. Rev. Oncol. Hematol. 2008, 66, 1–9. [Google Scholar] [CrossRef] [PubMed]
  77. Khaled, Y.S.; Ammori, B.J.; Elkord, E. Myeloid-derived suppressor cells in cancer: Recent progress and prospects. Immunol. Cell Biol. 2013, 91, 493–502. [Google Scholar] [CrossRef] [PubMed]
  78. Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef]
  79. Huang, Y.; Obholzer, N.; Fayad, R.; Qiao, L. Turning on/off tumor-specific CTL response during progressive tumor growth. J. Immunol. 2005, 175, 3110–3116. [Google Scholar] [CrossRef]
  80. Zhou, G.; Lu, Z.; McCadden, J.D.; Levitsky, H.I.; Marson, A.L. Reciprocal changes in tumor antigenicity and antigen-specific T cell function during tumor progression. J. Exp. Med. 2004, 200, 1581–1592. [Google Scholar] [CrossRef]
  81. Sakaguchi, S. Naturally arising CD4+ regulatory T cells for immunologic self-tolerance and negative control of immune responses. Annu. Rev. Immunol. 2004, 22, 531–562. [Google Scholar] [CrossRef]
  82. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef] [PubMed]
  83. Durack, J.; Lynch, S.V. The gut microbiome: Relationships with disease and opportunities for therapy. J. Exp. Med. 2019, 216, 20–40. [Google Scholar] [CrossRef] [PubMed]
  84. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef]
  85. Carding, S.; Verbeke, K.; Vipond, D.T.; Corfe, B.M.; Owen, L.J. Dysbiosis of the gut microbiota in disease. Microb. Ecol. Health Dis. 2015, 26, 26191. [Google Scholar] [CrossRef] [PubMed]
  86. Fan, Y.; Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef]
  87. Fulbright, L.E.; Ellermann, M.; Arthur, J.C. The microbiome and the hallmarks of cancer. PLoS Pathog. 2017, 13, e1006480. [Google Scholar] [CrossRef]
  88. Gagnaire, A.; Nadel, B.; Raoult, D.; Neefjes, J.; Gorvel, J.P. Collateral damage: Insights into bacterial mechanisms that predispose host cells to cancer. Nat. Rev. Microbiol. 2017, 15, 109–128. [Google Scholar] [CrossRef]
  89. Vivarelli, S.; Salemi, R.; Candido, S.; Falzone, L.; Santagati, M.; Stefani, S.; Torino, F.; Banna, G.L.; Tonini, G.; Libra, M. Gut Microbiota and Cancer: From Pathogenesis to Therapy. Cancers 2019, 11, 38. [Google Scholar] [CrossRef]
  90. Zitvogel, L.; Daillère, R.; Roberti, M.P.; Routy, B.; Kroemer, G. Anticancer effects of the microbiome and its products. Nat. Rev. Microbiol. 2017, 15, 465–478. [Google Scholar] [CrossRef]
  91. Ge, Y.; Wang, X.; Guo, Y.; Yan, J.; Abuduwaili, A.; Aximujiang, K.; Yan, J.; Wu, M. Gut microbiota influence tumor development and Alter interactions with the human immune system. J. Exp. Clin. Cancer Res. 2021, 40, 42. [Google Scholar] [CrossRef]
  92. Bhatt, A.P.; Redinbo, M.R.; Bultman, S.J. The role of the microbiome in cancer development and therapy. CA Cancer J. Clin. 2017, 67, 326–344. [Google Scholar] [CrossRef] [PubMed]
  93. Fiorentini, C.; Carlini, F.; Germinario, E.A.P.; Maroccia, Z.; Travaglione, S.; Fabbri, A. Gut Microbiota and Colon Cancer: A Role for Bacterial Protein Toxins? Int. J. Mol. Sci. 2020, 21, 6201. [Google Scholar] [CrossRef] [PubMed]
  94. Frisan, T. Bacterial genotoxins: The long journey to the nucleus of mammalian cells. Biochim. Biophys. Acta 2016, 1858, 567–575. [Google Scholar] [CrossRef] [PubMed]
  95. Lara-Tejero, M.; Galán, J.E. A bacterial toxin that controls cell cycle progression as a deoxyribonuclease I-like protein. Science 2000, 290, 354–357. [Google Scholar] [CrossRef] [PubMed]
  96. Yao, Y.; Dai, W. Genomic Instability and Cancer. J. Carcinog. Mutagen. 2014, 5, 1000165. [Google Scholar] [PubMed]
  97. Wada, Y.; Takemura, K.; Tummala, P.; Uchida, K.; Kitagaki, K.; Furukawa, A.; Ishige, Y.; Ito, T.; Hara, Y.; Suzuki, T.; et al. Helicobacter pylori induces somatic mutations in TP53 via overexpression of CHAC1 in infected gastric epithelial cells. FEBS Open Bio 2018, 8, 671–679. [Google Scholar] [CrossRef]
  98. Bergounioux, J.; Elisee, R.; Prunier, A.L.; Donnadieu, F.; Sperandio, B.; Sansonetti, P.; Arbibe, L. Calpain activation by the Shigella flexneri effector VirA regulates key steps in the formation and life of the bacterium’s epithelial niche. Cell Host Microbe 2012, 11, 240–252. [Google Scholar] [CrossRef]
  99. Buti, L.; Spooner, E.; Van der Veen, A.G.; Rappuoli, R.; Covacci, A.; Ploegh, H.L. Helicobacter pylori cytotoxin-associated gene A (CagA) subverts the apoptosis-stimulating protein of p53 (ASPP2) tumor suppressor pathway of the host. Proc. Natl. Acad. Sci. USA 2011, 108, 9238–9243. [Google Scholar] [CrossRef]
  100. Chaturvedi, R.; Asim, M.; Romero-Gallo, J.; Barry, D.P.; Hoge, S.; de Sablet, T.; Delgado, A.G.; Wroblewski, L.E.; Piazuelo, M.B.; Yan, F.; et al. Spermine oxidase mediates the gastric cancer risk associated with Helicobacter pylori CagA. Gastroenterology 2011, 141, 1696–1708. [Google Scholar] [CrossRef]
  101. Goodwin, A.C.; Destefano Shields, C.E.; Wu, S.; Huso, D.L.; Wu, X.; Murray-Stewart, T.R.; Hacker-Prietz, A.; Rabizadeh, S.; Woster, P.M.; Sears, C.L.; et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc. Natl. Acad. Sci. USA 2011, 108, 15354–15359. [Google Scholar] [CrossRef] [Green Version]
  102. Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [PubMed]
  103. Hu, B.; Elinav, E.; Huber, S.; Strowig, T.; Hao, L.; Hafemann, A.; Jin, C.; Wunderlich, C.; Wunderlich, T.; Eisenbarth, S.C.; et al. Microbiota-induced activation of epithelial IL-6 signaling links inflammasome-driven inflammation with transmissible cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 9862–9867. [Google Scholar] [CrossRef] [PubMed]
  104. Caesar, R.; Tremaroli, V.; Kovatcheva-Datchary, P.; Cani, P.D.; Backhed, F. Crosstalk between gut microbiota and dietary lipids aggravates WAT inflammation through TLR signaling. Cell Metab. 2015, 22, 658–668. [Google Scholar] [CrossRef]
  105. Gur, C.; Ibrahim, Y.; Isaacson, B.; Yamin, R.; Abed, J.; Gamliel, M.; Enk, J.; Bar-On, Y.; Stanietsky-Kaynan, N.; Coppenhagen-Glazer, S.; et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity 2015, 42, 344–355. [Google Scholar] [CrossRef]
  106. Chang, S.H.; Minn, D.; Kim, S.W.; Kim, Y.K. Inflammatory Markers and Cytokines in Moderate and Critical Cases of COVID-19. Clin. Lab. 2021, 67. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, F.; Cao, J.; Yu, Y.; Ding, J.; Eshak, E.S.; Liu, K.; Mubarik, S.; Shi, F.; Wen, H.; Zeng, Z.; et al. Epidemiological characteristics of patients with severe COVID-19 infection in Wuhan, China: Evidence from a retrospective observational study. Int. J. Epidemiol. 2021, 49, 1940–1950. [Google Scholar] [CrossRef]
  108. da Silva Torres, M.K.; Bichara, C.D.A.; de Almeida, M.; Vallinoto, M.C.; Queiroz, M.A.F.; Vallinoto, I.; Dos Santos, E.J.M.; de Carvalho, C.A.M.; Vallinoto, A.C.R. The Complexity of SARS-CoV-2 Infection and the COVID-19 Pandemic. Front. Microbiol. 2022, 13, 789882. [Google Scholar] [CrossRef]
  109. Kaur, S.; Bansal, R.; Kollimuttathuillam, S.; Gowda, A.M.; Singh, B.; Mehta, D.; Maroules, M. The looming storm: Blood and cytokines in COVID-19. Blood Rev. 2021, 46, 100743. [Google Scholar] [CrossRef]
  110. Chen, Y.; Klein, S.L.; Garibaldi, B.T.; Li, H.; Wu, C.; Osevala, N.M.; Li, T.; Margolick, J.B.; Pawelec, G.; Leng, S.X. Aging in COVID-19: Vulnerability, immunity and intervention. Ageing Res. Rev. 2021, 65, 101205. [Google Scholar] [CrossRef]
  111. Java, A.; Apicelli, A.J.; Liszewski, M.K.; Coler-Reilly, A.; Atkinson, J.P.; Kim, A.H.; Kulkarni, H.S. The complement system in COVID-19: Friend and foe? JCI Insight 2020, 5, e140711. [Google Scholar] [CrossRef]
  112. Teijaro, J.R. Cytokine storms in infectious diseases. Semin. Immunopathol. 2017, 39, 501–503. [Google Scholar] [CrossRef] [PubMed]
  113. Chen, L.Y.C.; Quach, T.T.T. COVID-19 cytokine storm syndrome: A threshold concept. Lancet Microbe 2021, 2, e49–e50. [Google Scholar] [CrossRef]
  114. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef]
  115. Kunnumakkara, A.B.; Rana, V.; Parama, D.; Banik, K.; Girisa, S.; Henamayee, S.; Thakur, K.K.; Dutta, U.; Garodia, P.; Gupta, S.C.; et al. COVID-19, cytokines, inflammation, and spices: How are they related? Life Sci. 2021, 284, 119201. [Google Scholar] [CrossRef] [PubMed]
  116. Giamarellos-Bourboulis, E.J.; Netea, M.G.; Rovina, N.; Akinosoglou, K.; Antoniadou, A.; Antonakos, N.; Damoraki, G.; Gkavogianni, T.; Adami, M.E.; Katsaounou, P.; et al. Complex Immune Dysregulation in COVID-19 Patients with Severe Respiratory Failure. Cell Host Microbe 2020, 27, 992–1000.e3. [Google Scholar] [CrossRef]
  117. Logue, J.K.; Franko, N.M.; McCulloch, D.J.; McDonald, D.; Magedson, A.; Wolf, C.R.; Chu, H.Y. Sequelae in Adults at 6 Months After COVID-19 Infection. JAMA Netw. Open 2021, 4, e210830. [Google Scholar] [CrossRef]
  118. Al-Aly, Z.; Xie, Y.; Bowe, B. High-dimensional characterization of post-acute sequelae of COVID-19. Nature 2021, 594, 259–264. [Google Scholar] [CrossRef] [PubMed]
  119. Huang, C.; Huang, L.; Wang, Y.; Li, X.; Ren, L.; Gu, X.; Kang, L.; Guo, L.; Liu, M.; Zhou, X.; et al. 6-month consequences of COVID-19 in patients discharged from hospital: A cohort study. Lancet 2021, 397, 220–232. [Google Scholar] [CrossRef]
  120. Montefusco, L.; Ben Nasr, M.; D’Addio, F.; Loretelli, C.; Rossi, A.; Pastore, I.; Daniele, G.; Abdelsalam, A.; Maestroni, A.; Dell’Acqua, M.; et al. Acute and long-term disruption of glycometabolic control after SARS-CoV-2 infection. Nat. Metab. 2021, 3, 774–785. [Google Scholar] [CrossRef] [PubMed]
  121. Agarwala, P.; Salzman, S.H. Six-Minute Walk Test: Clinical Role, Technique, Coding, and Reimbursement. Chest 2020, 157, 603–611. [Google Scholar] [CrossRef]
  122. Blomberg, B.; Mohn, K.G.; Brokstad, K.A.; Zhou, F.; Linchausen, D.W.; Hansen, B.A.; Lartey, S.; Onyango, T.B.; Kuwelker, K.; Saevik, M.; et al. Long COVID in a prospective cohort of home-isolated patients. Nat. Med. 2021, 27, 1607–1613. [Google Scholar] [CrossRef] [PubMed]
  123. Nalbandian, A.; Sehgal, K.; Gupta, A.; Madhavan, M.V.; McGroder, C.; Stevens, J.S.; Cook, J.R.; Nordvig, A.S.; Shalev, D.; Sehrawat, T.S.; et al. Post-acute COVID-19 syndrome. Nat. Med. 2021, 27, 601–615. [Google Scholar] [CrossRef] [PubMed]
  124. Parums, D.V. Editorial: Long COVID, or Post-COVID Syndrome, and the Global Impact on Health Care. Med. Sci. Monit. 2021, 27, e933446. [Google Scholar] [CrossRef] [PubMed]
  125. Sykes, D.L.; Holdsworth, L.; Jawad, N.; Gunasekera, P.; Morice, A.H.; Crooks, M.G. Post-COVID-19 Symptom Burden: What is Long-COVID and How Should We Manage It? Lung 2021, 199, 113–119. [Google Scholar] [CrossRef] [PubMed]
  126. Naeije, R.; Caravita, S. Phenotyping long COVID. Eur. Respir. J. 2021, 58, 2101763. [Google Scholar] [CrossRef]
  127. Proal, A.D.; VanElzakker, M.B. Long COVID or Post-acute Sequelae of COVID-19 (PASC): An Overview of Biological Factors That May Contribute to Persistent Symptoms. Front. Microbiol. 2021, 12, 698169. [Google Scholar] [CrossRef]
  128. Queiroz, M.A.F.; Neves, P.; Lima, S.S.; Lopes, J.D.C.; Torres, M.; Vallinoto, I.; Bichara, C.D.A.; Dos Santos, E.F.; de Brito, M.; da Silva, A.L.S.; et al. Cytokine Profiles Associated With Acute COVID-19 and Long COVID-19 Syndrome. Front. Cell Infect. Microbiol. 2022, 12, 922422. [Google Scholar] [CrossRef]
  129. Moreno-Perez, O.; Merino, E.; Leon-Ramirez, J.M.; Andres, M.; Ramos, J.M.; Arenas-Jimenez, J.; Asensio, S.; Sanchez, R.; Ruiz-Torregrosa, P.; Galan, I.; et al. Post-acute COVID-19 syndrome. Incidence and risk factors: A Mediterranean cohort study. J. Infect. 2021, 82, 378–383. [Google Scholar] [CrossRef]
  130. Sapir, T.; Averch, Z.; Lerman, B.; Bodzin, A.; Fishman, Y.; Maitra, R. COVID-19 and the Immune Response: A Multi-Phasic Approach to the Treatment of COVID-19. Int. J. Mol. Sci. 2022, 23, 8606. [Google Scholar] [CrossRef]
  131. Lechner-Scott, J.; Levy, M.; Hawkes, C.; Yeh, A.; Giovannoni, G. Long COVID or post COVID-19 syndrome. Mult. Scler. Relat. Dis. 2021, 55, 103268. [Google Scholar] [CrossRef]
  132. Schultheiß, C.; Willscher, E.; Paschold, L.; Gottschick, C.; Klee, B.; Henkes, S.S.; Bosurgi, L.; Dutzmann, J.; Sedding, D.; Frese, T.; et al. The IL-1β, IL-6, and TNF cytokine triad is associated with post-acute sequelae of COVID-19. Cell Rep. Med. 2022, 3, 100663. [Google Scholar] [CrossRef]
  133. Peluso, M.J.; Lu, S.; Tang, A.F.; Durstenfeld, M.S.; Ho, H.E.; Goldberg, S.A.; Forman, C.A.; Munter, S.E.; Hoh, R.; Tai, V.; et al. Markers of Immune Activation and Inflammation in Individuals With Postacute Sequelae of Severe Acute Respiratory Syndrome Coronavirus 2 Infection. J. Infect. Dis. 2021, 224, 1839–1848. [Google Scholar] [CrossRef] [PubMed]
  134. Acosta-Ampudia, Y.; Monsalve, D.M.; Rojas, M.; Rodríguez, Y.; Zapata, E.; Ramírez-Santana, C.; Anaya, J.M. Persistent Autoimmune Activation and Proinflammatory State in Post-Coronavirus Disease 2019 Syndrome. J. Infect. Dis. 2022, 225, 2155–2162. [Google Scholar] [CrossRef] [PubMed]
  135. Phetsouphanh, C.; Darley, D.R.; Wilson, D.B.; Howe, A.; Munier, C.M.L.; Patel, S.K.; Juno, J.A.; Burrell, L.M.; Kent, S.J.; Dore, G.J.; et al. Immunological dysfunction persists for 8 months following initial mild-to-moderate SARS-CoV-2 infection. Nat. Immunol. 2022, 23, 210–216. [Google Scholar] [CrossRef] [PubMed]
  136. Sette, A.; Crotty, S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 2021, 184, 861–880. [Google Scholar] [CrossRef]
  137. Arunachalam, P.S.; Wimmers, F.; Mok, C.K.P.; Perera, R.; Scott, M.; Hagan, T.; Sigal, N.; Feng, Y.; Bristow, L.; Tak-Yin Tsang, O.; et al. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science 2020, 369, 1210–1220. [Google Scholar] [CrossRef]
  138. Bastard, P.; Rosen, L.B.; Zhang, Q.; Michailidis, E.; Hoffmann, H.H.; Zhang, Y.; Dorgham, K.; Philippot, Q.; Rosain, J.; Beziat, V.; et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 2020, 370, eabd4585. [Google Scholar] [CrossRef]
  139. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Moller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef]
  140. Laing, A.G.; Lorenc, A.; Del Molino Del Barrio, I.; Das, A.; Fish, M.; Monin, L.; Munoz-Ruiz, M.; McKenzie, D.R.; Hayday, T.S.; Francos-Quijorna, I.; et al. A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat. Med. 2020, 26, 1623–1635. [Google Scholar] [CrossRef]
  141. Li, S.; Jiang, L.; Li, X.; Lin, F.; Wang, Y.; Li, B.; Jiang, T.; An, W.; Liu, S.; Liu, H.; et al. Clinical and pathological investigation of patients with severe COVID-19. JCI Insight 2020, 5, e138070. [Google Scholar] [CrossRef]
  142. Schurink, B.; Roos, E.; Radonic, T.; Barbe, E.; Bouman, C.S.C.; de Boer, H.H.; de Bree, G.J.; Bulle, E.B.; Aronica, E.M.; Florquin, S.; et al. Viral presence and immunopathology in patients with lethal COVID-19: A prospective autopsy cohort study. Lancet Microbe 2020, 1, e290–e299. [Google Scholar] [CrossRef]
  143. Magleby, R.; Westblade, L.F.; Trzebucki, A.; Simon, M.S.; Rajan, M.; Park, J.; Goyal, P.; Safford, M.M.; Satlin, M.J. Impact of Severe Acute Respiratory Syndrome Coronavirus 2 Viral Load on Risk of Intubation and Mortality Among Hospitalized Patients With Coronavirus Disease 2019. Clin. Infect. Dis. 2021, 73, e4197–e4205. [Google Scholar] [CrossRef] [PubMed]
  144. Kuri-Cervantes, L.; Pampena, M.B.; Meng, W.; Rosenfeld, A.M.; Ittner, C.A.G.; Weisman, A.R.; Agyekum, R.S.; Mathew, D.; Baxter, A.E.; Vella, L.A.; et al. Comprehensive mapping of immune perturbations associated with severe COVID-19. Sci. Immunol. 2020, 5, eabd7114. [Google Scholar] [CrossRef] [PubMed]
  145. Galani, I.E.; Rovina, N.; Lampropoulou, V.; Triantafyllia, V.; Manioudaki, M.; Pavlos, E.; Koukaki, E.; Fragkou, P.C.; Panou, V.; Rapti, V.; et al. Untuned antiviral immunity in COVID-19 revealed by temporal type I/III interferon patterns and flu comparison. Nat. Immunol. 2021, 22, 32–40. [Google Scholar] [CrossRef]
  146. Files, J.K.; Sarkar, S.; Fram, T.R.; Boppana, S.; Sterrett, S.; Qin, K.; Bansal, A.; Long, D.M.; Sabbaj, S.; Kobie, J.J.; et al. Duration of post-COVID-19 symptoms is associated with sustained SARS-CoV-2-specific immune responses. JCI Insight 2021, 6, e151544. [Google Scholar] [CrossRef]
  147. Loretelli, C.; Abdelsalam, A.; D’Addio, F.; Ben Nasr, M.; Assi, E.; Usuelli, V.; Maestroni, A.; Seelam, A.J.; Ippolito, E.; Di Maggio, S.; et al. PD-1 blockade counteracts post-COVID-19 immune abnormalities and stimulates the anti-SARS-CoV-2 immune response. JCI Insight 2021, 6, e146701. [Google Scholar] [CrossRef]
  148. Glynne, P.; Tahmasebi, N.; Gant, V.; Gupta, R. Long COVID following mild SARS-CoV-2 infection: Characteristic T cell alterations and response to antihistamines. J. Investig. Med. 2022, 70, 61–67. [Google Scholar] [CrossRef]
  149. Galán, M.; Vigón, L.; Fuertes, D.; Murciano-Antón, M.A.; Casado-Fernández, G.; Domínguez-Mateos, S.; Mateos, E.; Ramos-Martín, F.; Planelles, V.; Torres, M.; et al. Persistent Overactive Cytotoxic Immune Response in a Spanish Cohort of Individuals With Long-COVID: Identification of Diagnostic Biomarkers. Front. Immunol. 2022, 13, 848886. [Google Scholar] [CrossRef]
  150. Peluso, M.J.; Deitchman, A.N.; Torres, L.; Iyer, N.S.; Munter, S.E.; Nixon, C.C.; Donatelli, J.; Thanh, C.; Takahashi, S.; Hakim, J.; et al. Long-term SARS-CoV-2-specific immune and inflammatory responses in individuals recovering from COVID-19 with and without post-acute symptoms. Cell Rep. 2021, 36, 109518. [Google Scholar] [CrossRef]
  151. Wiech, M.; Chroscicki, P.; Swatler, J.; Stepnik, D.; De Biasi, S.; Hampel, M.; Brewinska-Olchowik, M.; Maliszewska, A.; Sklinda, K.; Durlik, M.; et al. Remodeling of T Cell Dynamics During Long COVID Is Dependent on Severity of SARS-CoV-2 Infection. Front. Immunol. 2022, 13, 886431. [Google Scholar] [CrossRef]
  152. Jarjour, N.N.; Masopust, D.; Jameson, S.C. T cell memory: Understanding COVID-19. Immunity 2021, 54, 14–18. [Google Scholar] [CrossRef] [PubMed]
  153. Weisberg, S.P.; Ural, B.B.; Farber, D.L. Tissue-specific immunity for a changing world. Cell 2021, 184, 1517–1529. [Google Scholar] [CrossRef] [PubMed]
  154. Sasson, S.C.; Gordon, C.L.; Christo, S.N.; Klenerman, P.; Mackay, L.K. Local heroes or villains: Tissue-resident memory T cells in human health and disease. Cell. Mol. Immunol. 2020, 17, 113–122. [Google Scholar] [CrossRef] [PubMed]
  155. Wang, Z.; Wang, S.; Goplen, N.P.; Li, C.; Cheon, I.S.; Dai, Q.; Huang, S.; Shan, J.; Ma, C.; Ye, Z.; et al. PD-1hi CD8+ resident memory T cells balance immunity and fibrotic sequelae. Sci. Immunol. 2019, 4, eaaw1217. [Google Scholar] [CrossRef]
  156. Goplen, N.P.; Wu, Y.; Son, Y.M.; Li, C.; Wang, Z.; Cheon, I.S.; Jiang, L.; Zhu, B.; Ayasoufi, K.; Chini, E.N.; et al. Tissue-resident CD8(+) T cells drive age-associated chronic lung sequelae after viral pneumonia. Sci. Immunol. 2020, 5, eabc4557. [Google Scholar] [CrossRef]
  157. Cheon, I.S.; Li, C.; Son, Y.M.; Goplen, N.P.; Wu, Y.; Cassmann, T.; Wang, Z.; Wei, X.; Tang, J.; Li, Y.; et al. Immune signatures underlying post-acute COVID-19 lung sequelae. Sci. Immunol. 2021, 6, eabk1741. [Google Scholar] [CrossRef]
  158. Hu, B.; Guo, H.; Zhou, P.; Shi, Z.L. Characteristics of SARS-CoV-2 and COVID-19. Nat. Rev. Microbiol. 2021, 19, 141–154. [Google Scholar] [CrossRef]
  159. Burnham, E.L.; Janssen, W.J.; Riches, D.W.; Moss, M.; Downey, G.P. The fibroproliferative response in acute respiratory distress syndrome: Mechanisms and clinical significance. Eur. Respir. J. 2014, 43, 276–285. [Google Scholar] [CrossRef]
  160. Misharin, A.V.; Morales-Nebreda, L.; Reyfman, P.A.; Cuda, C.M.; Walter, J.M.; McQuattie-Pimentel, A.C.; Chen, C.I.; Anekalla, K.R.; Joshi, N.; Williams, K.J.N.; et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J. Exp. Med. 2017, 214, 2387–2404. [Google Scholar] [CrossRef]
  161. Budinger, G.R.S.; Misharin, A.V.; Ridge, K.M.; Singer, B.D.; Wunderink, R.G. Distinctive features of severe SARS-CoV-2 pneumonia. J. Clin. Investig. 2021, 131, e149412. [Google Scholar] [CrossRef]
  162. Østergaard, L. SARS-CoV-2 related microvascular damage and symptoms during and after COVID-19: Consequences of capillary transit-time changes, tissue hypoxia and inflammation. Physiol. Rep. 2021, 9, e14726. [Google Scholar] [CrossRef] [PubMed]
  163. Wang, C.; Yu, C.; Jing, H.; Wu, X.; Novakovic, V.A.; Xie, R.; Shi, J. Long COVID: The Nature of Thrombotic Sequelae Determines the Necessity of Early Anticoagulation. Front. Cell Infect. Microbiol. 2022, 12, 861703. [Google Scholar] [CrossRef] [PubMed]
  164. Kell, D.B.; Laubscher, G.J.; Pretorius, E. A central role for amyloid fibrin microclots in long COVID/PASC: Origins and therapeutic implications. Biochem. J. 2022, 479, 537–559. [Google Scholar] [CrossRef]
  165. Libby, P.; Luscher, T. COVID-19 is, in the end, an endothelial disease. Eur. Heart J. 2020, 41, 3038–3044. [Google Scholar] [CrossRef] [PubMed]
  166. Yong, S.J. Long COVID or post-COVID-19 syndrome: Putative pathophysiology, risk factors, and treatments. Infect. Dis. 2021, 53, 737–754. [Google Scholar] [CrossRef]
  167. Yeoh, Y.K.; Zuo, T.; Lui, G.C.; Zhang, F.; Liu, Q.; Li, A.Y.; Chung, A.C.; Cheung, C.P.; Tso, E.Y.; Fung, K.S.; et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut 2021, 70, 698–706. [Google Scholar] [CrossRef]
  168. Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, Y.; Wang, J.; Liu, Y.; Wei, Y.; et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020, 395, 507–513. [Google Scholar] [CrossRef]
  169. Liang, W.; Feng, Z.; Rao, S.; Xiao, C.; Xue, X.; Lin, Z.; Zhang, Q.; Qi, W. Diarrhoea may be underestimated: A missing link in 2019 novel coronavirus. Gut 2020, 69, 1141–1143. [Google Scholar] [CrossRef]
  170. Wolfel, R.; Corman, V.M.; Guggemos, W.; Seilmaier, M.; Zange, S.; Muller, M.A.; Niemeyer, D.; Jones, T.C.; Vollmar, P.; Rothe, C.; et al. Virological assessment of hospitalized patients with COVID-2019. Nature 2020, 581, 465–469. [Google Scholar] [CrossRef]
  171. Xu, Y.; Li, X.; Zhu, B.; Liang, H.; Fang, C.; Gong, Y.; Guo, Q.; Sun, X.; Zhao, D.; Shen, J.; et al. Characteristics of pediatric SARS-CoV-2 infection and potential evidence for persistent fecal viral shedding. Nat. Med. 2020, 26, 502–505. [Google Scholar] [CrossRef] [Green Version]
  172. Ng, S.C.; Tilg, H. COVID-19 and the gastrointestinal tract: More than meets the eye. Gut 2020, 69, 973–974. [Google Scholar] [CrossRef] [PubMed]
  173. Chassaing, B.; Kumar, M.; Baker, M.T.; Singh, V.; Vijay-Kumar, M. Mammalian gut immunity. Biomed. J. 2014, 37, 246–258. [Google Scholar] [PubMed]
  174. Zuo, T.; Kamm, M.A.; Colombel, J.F.; Ng, S.C. Urbanization and the gut microbiota in health and inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 440–452. [Google Scholar] [CrossRef]
  175. Zuo, T.; Liu, Q.; Zhang, F.; Yeoh, Y.K.; Wan, Y.; Zhan, H.; Lui, G.C.Y.; Chen, Z.; Li, A.Y.L.; Cheung, C.P.; et al. Temporal landscape of human gut RNA and DNA virome in SARS-CoV-2 infection and severity. Microbiome 2021, 9, 91. [Google Scholar] [CrossRef] [PubMed]
  176. Zuo, T.; Zhan, H.; Zhang, F.; Liu, Q.; Tso, E.Y.K.; Lui, G.C.Y.; Chen, N.; Li, A.; Lu, W.; Chan, F.K.L.; et al. Alterations in Fecal Fungal Microbiome of Patients With COVID-19 During Time of Hospitalization until Discharge. Gastroenterology 2020, 159, 1302–1310.e5. [Google Scholar] [CrossRef]
  177. Zuo, T.; Zhang, F.; Lui, G.C.Y.; Yeoh, Y.K.; Li, A.Y.L.; Zhan, H.; Wan, Y.; Chung, A.C.K.; Cheung, C.P.; Chen, N.; et al. Alterations in Gut Microbiota of Patients With COVID-19 During Time of Hospitalization. Gastroenterology 2020, 159, 944–955.e8. [Google Scholar] [CrossRef] [PubMed]
  178. Wang, B.; Zhang, L.; Wang, Y.; Dai, T.; Qin, Z.; Zhou, F.; Zhang, L. Alterations in microbiota of patients with covid-19: Potential mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 2022, 7, 143. [Google Scholar] [CrossRef]
  179. Zuo, T.; Wu, X.; Wen, W.; Lan, P. Gut Microbiome Alterations in COVID-19. Genom. Proteom. Bioinform. 2021, 19, 679–688. [Google Scholar] [CrossRef]
  180. Tang, L.; Gu, S.; Gong, Y.; Li, B.; Lu, H.; Li, Q.; Zhang, R.; Gao, X.; Wu, Z.; Zhang, J.; et al. Clinical Significance of the Correlation between Changes in the Major Intestinal Bacteria Species and COVID-19 Severity. Engineering 2020, 6, 1178–1184. [Google Scholar] [CrossRef]
  181. Gu, S.; Chen, Y.; Wu, Z.; Chen, Y.; Gao, H.; Lv, L.; Guo, F.; Zhang, X.; Luo, R.; Huang, C.; et al. Alterations of the Gut Microbiota in Patients With Coronavirus Disease 2019 or H1N1 Influenza. Clin. Infect. Dis. 2020, 71, 2669–2678. [Google Scholar] [CrossRef]
  182. Weng, J.; Li, Y.; Li, J.; Shen, L.; Zhu, L.; Liang, Y.; Lin, X.; Jiao, N.; Cheng, S.; Huang, Y.; et al. Gastrointestinal sequelae 90 days after discharge for COVID-19. Lancet Gastroenterol. Hepatol. 2021, 6, 344–346. [Google Scholar] [CrossRef]
  183. Gaebler, C.; Wang, Z.; Lorenzi, J.C.C.; Muecksch, F.; Finkin, S.; Tokuyama, M.; Cho, A.; Jankovic, M.; Schaefer-Babajew, D.; Oliveira, T.Y.; et al. Evolution of antibody immunity to SARS-CoV-2. Nature 2021, 591, 639–644. [Google Scholar] [CrossRef] [PubMed]
  184. Wang, Y.; Zhang, L.; Sang, L.; Ye, F.; Ruan, S.; Zhong, B.; Song, T.; Alshukairi, A.N.; Chen, R.; Zhang, Z.; et al. Kinetics of viral load and antibody response in relation to COVID-19 severity. J. Clin. Investig. 2020, 130, 5235–5244. [Google Scholar] [CrossRef] [PubMed]
  185. Chen, Y.; Gu, S.; Chen, Y.; Lu, H.; Shi, D.; Guo, J.; Wu, W.R.; Yang, Y.; Li, Y.; Xu, K.J.; et al. Six-month follow-up of gut microbiota richness in patients with COVID-19. Gut 2022, 71, 222–225. [Google Scholar] [CrossRef]
  186. Sokol, H.; Contreras, V.; Maisonnasse, P.; Desmons, A.; Delache, B.; Sencio, V.; Machelart, A.; Brisebarre, A.; Humbert, L.; Deryuter, L.; et al. SARS-CoV-2 infection in nonhuman primates alters the composition and functional activity of the gut microbiota. Gut Microbes 2021, 13, 1–19. [Google Scholar] [CrossRef] [PubMed]
  187. Liu, Q.; Mak, J.W.Y.; Su, Q.; Yeoh, Y.K.; Lui, G.C.; Ng, S.S.S.; Zhang, F.; Li, A.Y.L.; Lu, W.; Hui, D.S.; et al. Gut microbiota dynamics in a prospective cohort of patients with post-acute COVID-19 syndrome. Gut 2022, 71, 544–552. [Google Scholar] [CrossRef] [PubMed]
  188. Su, Q.; Lau, R.I.; Liu, Q.; Chan, F.K.L.; Ng, S.C. Post-acute COVID-19 syndrome and gut dysbiosis linger beyond 1 year after SARS-CoV-2 clearance. Gut, 2022; Online ahead of print. [Google Scholar] [CrossRef]
  189. Guo, Y.; Xie, J.P.; Deng, K.; Li, X.; Yuan, Y.; Xuan, Q.; Xie, J.; He, X.M.; Wang, Q.; Li, J.J.; et al. Prophylactic Effects of Bifidobacterium adolescentis on Anxiety and Depression-Like Phenotypes After Chronic Stress: A Role of the Gut Microbiota-Inflammation Axis. Front. Behav. Neurosci. 2019, 13, 126. [Google Scholar] [CrossRef]
  190. Ren, Z.; Wang, H.; Cui, G.; Lu, H.; Wang, L.; Luo, H.; Chen, X.; Ren, H.; Sun, R.; Liu, W.; et al. Alterations in the human oral and gut microbiomes and lipidomics in covid-19. Gut 2021, 70, 1253–1265. [Google Scholar] [CrossRef]
  191. Lahti, L.; Salojarvi, J.; Salonen, A.; Scheffer, M.; de Vos, W.M. Tipping elements in the human intestinal ecosystem. Nat. Commun. 2014, 5, 4344. [Google Scholar] [CrossRef] [Green Version]
  192. Buffie, C.G.; Pamer, E.G. Microbiota-mediated colonization resistance against intestinal pathogens. Nat. Rev. Immunol. 2013, 13, 790–801. [Google Scholar] [CrossRef]
  193. Finlay, B.B.; Amato, K.R.; Azad, M.; Blaser, M.J.; Bosch, T.C.G.; Chu, H.; Dominguez-Bello, M.G.; Ehrlich, S.D.; Elinav, E.; Geva-Zatorsky, N.; et al. The hygiene hypothesis, the COVID pandemic, and consequences for the human microbiome. Proc. Natl. Acad. Sci. USA 2021, 118, e2010217118. [Google Scholar] [CrossRef] [PubMed]
  194. Vestad, B.; Ueland, T.; Lerum, T.V.; Dahl, T.B.; Holm, K.; Barratt-Due, A.; Kåsine, T.; Dyrhol-Riise, A.M.; Stiksrud, B.; Tonby, K.; et al. Respiratory dysfunction three months after severe COVID-19 is associated with gut microbiota alterations. J. Intern. Med. 2022, 291, 801–812. [Google Scholar] [CrossRef] [PubMed]
  195. Nataf, S.; Pays, L. Molecular Insights into SARS-CoV2-Induced Alterations of the Gut/Brain Axis. Int. J. Mol. Sci. 2021, 22, 10440. [Google Scholar] [CrossRef] [PubMed]
  196. Santus, W.; Devlin, J.R.; Behnsen, J. Crossing Kingdoms: How the Mycobiota and Fungal-Bacterial Interactions Impact Host Health and Disease. Infect. Immun. 2021, 89, e00648-20. [Google Scholar] [CrossRef]
  197. van Tilburg Bernardes, E.; Pettersen, V.K.; Gutierrez, M.W.; Laforest-Lapointe, I.; Jendzjowsky, N.G.; Cavin, J.B.; Vicentini, F.A.; Keenan, C.M.; Ramay, H.R.; Samara, J.; et al. Intestinal fungi are causally implicated in microbiome assembly and immune development in mice. Nat. Commun. 2020, 11, 2577. [Google Scholar] [CrossRef] [PubMed]
  198. Lv, L.; Gu, S.; Jiang, H.; Yan, R.; Chen, Y.; Chen, Y.; Luo, R.; Huang, C.; Lu, H.; Zheng, B.; et al. Gut mycobiota alterations in patients with COVID-19 and H1N1 infections and their associations with clinical features. Commun. Biol. 2021, 4, 480. [Google Scholar] [CrossRef]
  199. Cox, M.J.; Loman, N.; Bogaert, D.; O’Grady, J. Co-infections: Potentially lethal and unexplored in COVID-19. Lancet Microbe 2020, 1, e11. [Google Scholar] [CrossRef]
  200. Peman, J.; Ruiz-Gaitan, A.; Garcia-Vidal, C.; Salavert, M.; Ramirez, P.; Puchades, F.; Garcia-Hita, M.; Alastruey-Izquierdo, A.; Quindos, G. Fungal co-infection in COVID-19 patients: Should we be concerned? Rev. Iberoam. Micol. 2020, 37, 41–46. [Google Scholar] [CrossRef]
  201. Song, G.; Liang, G.; Liu, W. Fungal Co-infections Associated with Global COVID-19 Pandemic: A Clinical and Diagnostic Perspective from China. Mycopathologia 2020, 185, 599–606. [Google Scholar] [CrossRef]
  202. Erb Downward, J.R.; Falkowski, N.R.; Mason, K.L.; Muraglia, R.; Huffnagle, G.B. Modulation of post-antibiotic bacterial community reassembly and host response by Candida albicans. Sci. Rep. 2013, 3, 2191. [Google Scholar] [CrossRef]
  203. Zuo, T.; Wong, S.H.; Cheung, C.P.; Lam, K.; Lui, R.; Cheung, K.; Zhang, F.; Tang, W.; Ching, J.Y.L.; Wu, J.C.Y.; et al. Gut fungal dysbiosis correlates with reduced efficacy of fecal microbiota transplantation in Clostridium difficile infection. Nat. Commun. 2018, 9, 3663. [Google Scholar] [CrossRef] [PubMed]
  204. Kumamoto, C.A. Inflammation and gastrointestinal Candida colonization. Curr. Opin. Microbiol. 2011, 14, 386–391. [Google Scholar] [CrossRef]
  205. Sonoyama, K.; Miki, A.; Sugita, R.; Goto, H.; Nakata, M.; Yamaguchi, N. Gut colonization by Candida albicans aggravates inflammation in the gut and extra-gut tissues in mice. Med. Mycol. 2011, 49, 237–247. [Google Scholar] [CrossRef] [PubMed]
  206. Neil, J.A.; Cadwell, K. The Intestinal Virome and Immunity. J. Immunol. 2018, 201, 1615–1624. [Google Scholar] [CrossRef] [PubMed]
  207. Virgin, H.W. The virome in mammalian physiology and disease. Cell 2014, 157, 142–150. [Google Scholar] [CrossRef] [PubMed]
  208. Zuo, T.; Liu, Q.; Zhang, F.; Lui, G.C.; Tso, E.Y.; Yeoh, Y.K.; Chen, Z.; Boon, S.S.; Chan, F.K.; Chan, P.K.; et al. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut 2021, 70, 276–284. [Google Scholar] [CrossRef]
  209. Norman, J.M.; Handley, S.A.; Baldridge, M.T.; Droit, L.; Liu, C.Y.; Keller, B.C.; Kambal, A.; Monaco, C.L.; Zhao, G.; Fleshner, P.; et al. Disease-specific alterations in the enteric virome in inflammatory bowel disease. Cell 2015, 160, 447–460. [Google Scholar] [CrossRef]
  210. Zuo, T.; Lu, X.J.; Zhang, Y.; Cheung, C.P.; Lam, S.; Zhang, F.; Tang, W.; Ching, J.Y.L.; Zhao, R.; Chan, P.K.S.; et al. Gut mucosal virome alterations in ulcerative colitis. Gut 2019, 68, 1169–1179. [Google Scholar] [CrossRef]
  211. Diard, M.; Bakkeren, E.; Cornuault, J.K.; Moor, K.; Hausmann, A.; Sellin, M.E.; Loverdo, C.; Aertsen, A.; Ackermann, M.; De Paepe, M.; et al. Inflammation boosts bacteriophage transfer between Salmonella spp. Science 2017, 355, 1211–1215. [Google Scholar] [CrossRef]
  212. Cao, J.; Wang, C.; Zhang, Y.; Lei, G.; Xu, K.; Zhao, N.; Lu, J.; Meng, F.; Yu, L.; Yan, J.; et al. Integrated gut virome and bacteriome dynamics in COVID-19 patients. Gut Microbes 2021, 13, 1–21. [Google Scholar] [CrossRef]
  213. Tutuncuoglu, B.; Cakir, M.; Batra, J.; Bouhaddou, M.; Eckhardt, M.; Gordon, D.E.; Krogan, N.J. The Landscape of Human Cancer Proteins Targeted by SARS-CoV-2. Cancer Discov. 2020, 10, 916–921. [Google Scholar] [CrossRef] [PubMed]
  214. Rahimmanesh, I.; Shariati, L.; Dana, N.; Esmaeili, Y.; Vaseghi, G.; Haghjooy Javanmard, S. Cancer Occurrence as the Upcoming Complications of COVID-19. Front. Mol. Biosci. 2022, 8, 813175. [Google Scholar] [CrossRef] [PubMed]
  215. Saini, G.; Aneja, R. Cancer as a prospective sequela of long COVID-19. BioEssays 2021, 43, e2000331. [Google Scholar] [CrossRef] [PubMed]
  216. Gordon, D.E.; Jang, G.M.; Bouhaddou, M.; Xu, J.; Obernier, K.; White, K.M.; O’Meara, M.J.; Rezelj, V.V.; Guo, J.Z.; Swaney, D.L.; et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature 2020, 583, 459–468. [Google Scholar] [CrossRef] [PubMed]
  217. Gatti, P.; Ilamathi, H.S.; Todkar, K.; Germain, M. Mitochondria Targeted Viral Replication and Survival Strategies-Prospective on SARS-CoV-2. Front. Pharmacol. 2020, 11, 578599. [Google Scholar] [CrossRef] [PubMed]
  218. Roy, D.M.; Walsh, L.A.; Chan, T.A. Driver mutations of cancer epigenomes. Protein Cell 2014, 5, 265–296. [Google Scholar] [CrossRef]
  219. García-Carpizo, V.; Sarmentero, J.; Han, B.; Graña, O.; Ruiz-Llorente, S.; Pisano, D.G.; Serrano, M.; Brooks, H.B.; Campbell, R.M.; Barrero, M.J. NSD2 contributes to oncogenic RAS-driven transcription in lung cancer cells through long-range epigenetic activation. Sci. Rep. 2016, 6, 32952. [Google Scholar] [CrossRef]
  220. Sun, D.; Yu, M.; Li, Y.; Xing, H.; Gao, Y.; Huang, Z.; Hao, W.; Lu, K.; Kong, C.; Shimozato, O.; et al. Histone deacetylase 2 is involved in DNA damage-mediated cell death of human osteosarcoma cells through stimulation of the ATM/p53 pathway. FEBS Open Bio 2019, 9, 478–489. [Google Scholar] [CrossRef]
  221. Ma-Lauer, Y.; Carbajo-Lozoya, J.; Hein, M.Y.; Müller, M.A.; Deng, W.; Lei, J.; Meyer, B.; Kusov, Y.; Von Brunn, B.; Bairad, D.R.; et al. p53 down-regulates SARS coronavirus replication and is targeted by the SARS-unique domain and PLpro via E3 ubiquitin ligase RCHY1. Proc. Natl. Acad. Sci. USA 2016, 113, E5192–E5201. [Google Scholar] [CrossRef]
  222. Bhardwaj, K.; Liu, P.; Leibowitz, J.L.; Kao, C.C. The coronavirus endoribonuclease Nsp15 interacts with retinoblastoma tumor suppressor protein. J. Virol. 2012, 86, 4294–4304. [Google Scholar] [CrossRef]
  223. Singh, N.; Bharara Singh, A. S2 subunit of SARS-nCoV-2 interacts with tumor suppressor protein p53 and BRCA: An in silico study. Trans. Oncol. 2020, 13, 100814. [Google Scholar] [CrossRef] [PubMed]
  224. Owen, K.L.; Brockwell, N.K.; Parker, B.S. JAK-STAT Signaling: A Double-Edged Sword of Immune Regulation and Cancer Progression. Cancers 2019, 11, 2002. [Google Scholar] [CrossRef] [PubMed]
  225. Xia, Y.; Shen, S.; Verma, I.M. NF-κB, an active player in human cancers. Cancer Immunol. Res. 2014, 2, 823–830. [Google Scholar] [CrossRef]
  226. Martin-Hijano, L.; Sainz, B., Jr. The Interactions Between Cancer Stem Cells and the Innate Interferon Signaling Pathway. Front. Immunol. 2020, 11, 526. [Google Scholar] [CrossRef] [PubMed]
  227. Yang, C.H.; Murti, A.; Pfeffer, S.R.; Basu, L.; Kim, J.G.; Pfeffer, L.M. IFNα/β promotes cell survival by activating NF-κB. Proc. Natl. Acad. Sci. USA 2000, 97, 13631–13636. [Google Scholar] [CrossRef] [PubMed]
  228. Gomez, D.; Reich, N.C. Stimulation of primary human endothelial cell proliferation by IFN. J. Immunol. 2003, 170, 5373–5381. [Google Scholar] [CrossRef]
  229. Beatty, G.L.; Paterson, Y. IFN-γ can promote tumor evasion of the immune system in vivo by down-regulating cellular levels of an endogenous tumor antigen. J. Immunol. 2000, 165, 5502–5508. [Google Scholar] [CrossRef]
  230. Zou, Z.; Tao, T.; Li, H.; Zhu, X. mTOR signaling pathway and mTOR inhibitors in cancer: Progress and challenges. Cell Biosci. 2020, 10, 31. [Google Scholar] [CrossRef]
  231. Farahani, M.; Niknam, Z.; Mohammadi Amirabad, L.; Amiri-Dashatan, N.; Koushki, M.; Nemati, M.; Danesh Pouya, F.; Rezaei-Tavirani, M.; Rasmi, Y.; Tayebi, L. Molecular pathways involved in COVID-19 and potential pathway-based therapeutic targets. Biomed. Pharmacother. 2022, 145, 112420. [Google Scholar] [CrossRef]
  232. Ramaiah, M.J. mTOR inhibition and p53 activation, microRNAs: The possible therapy against pandemic COVID-19. Gene Rep. 2020, 20, 100765. [Google Scholar] [CrossRef]
  233. Breikaa, R.M.; Lilly, B. The Notch Pathway: A Link Between COVID-19 Pathophysiology and Its Cardiovascular Complications. Front. Cardiovasc. Med. 2021, 8, 681948. [Google Scholar] [CrossRef] [PubMed]
  234. Zhou, B.; Lin, W.; Long, Y.; Yang, Y.; Zhang, H.; Wu, K.; Chu, Q. Notch signaling pathway: Architecture, disease, and therapeutics. Signal. Transduct. Target. Ther. 2022, 7, 95. [Google Scholar] [CrossRef]
  235. Misiorek, J.O.; Przybyszewska-Podstawka, A.; Kałafut, J.; Paziewska, B.; Rolle, K.; Rivero-Müller, A.; Nees, M. Context Matters: NOTCH Signatures and Pathway in Cancer Progression and Metastasis. Cells 2021, 10, 94. [Google Scholar] [CrossRef] [PubMed]
  236. Kudaravalli, S.; den Hollander, P.; Mani, S.A. Role of p38 MAP kinase in cancer stem cells and metastasis. Oncogene 2022, 41, 3177–3185. [Google Scholar] [CrossRef] [PubMed]
  237. Grimes, J.M.; Grimes, K.V. p38 MAPK inhibition: A promising therapeutic approach for COVID-19. J. Mol. Cell Cardiol. 2020, 144, 63–65. [Google Scholar] [CrossRef] [PubMed]
  238. Limoge, M.; Safina, A.; Truskinovsky, A.M.; Aljahdali, I.; Zonneville, J.; Gruevski, A.; Arteaga, C.L.; Bakin, A.V. Tumor p38MAPK signaling enhances breast carcinoma vascularization and growth by promoting expression and deposition of pro-tumorigenic factors. Oncotarget 2017, 8, 61969–61981. [Google Scholar] [CrossRef]
  239. Serebrovska, Z.O.; Chong, E.Y.; Serebrovska, T.V.; Tumanovska, L.V.; Xi, L. Hypoxia, HIF-1α, and COVID-19: From pathogenic factors to potential therapeutic targets. Acta Pharmacol. Sin. 2020, 41, 1539–1546. [Google Scholar] [CrossRef]
  240. Al Tameemi, W.; Dale, T.P.; Al-Jumaily, R.M.K.; Forsyth, N.R. Hypoxia-Modified Cancer Cell Metabolism. Front. Cell Dev. Biol. 2019, 7, 4. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The shared molecular mechanisms between SARS-CoV-2 and oncogenesis. The figure was generated using images assembled from BioRender (available online: https://biorender.com, accessed on 30 August 2022).
Figure 1. The shared molecular mechanisms between SARS-CoV-2 and oncogenesis. The figure was generated using images assembled from BioRender (available online: https://biorender.com, accessed on 30 August 2022).
Vaccines 10 01607 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rapti, V.; Tsaganos, T.; Vathiotis, I.A.; Syrigos, N.K.; Li, P.; Poulakou, G. New Insights into SARS-CoV-2 and Cancer Cross-Talk: Does a Novel Oncogenesis Driver Emerge? Vaccines 2022, 10, 1607. https://doi.org/10.3390/vaccines10101607

AMA Style

Rapti V, Tsaganos T, Vathiotis IA, Syrigos NK, Li P, Poulakou G. New Insights into SARS-CoV-2 and Cancer Cross-Talk: Does a Novel Oncogenesis Driver Emerge? Vaccines. 2022; 10(10):1607. https://doi.org/10.3390/vaccines10101607

Chicago/Turabian Style

Rapti, Vasiliki, Thomas Tsaganos, Ioannis A. Vathiotis, Nikolaos K. Syrigos, Peifeng Li, and Garyfallia Poulakou. 2022. "New Insights into SARS-CoV-2 and Cancer Cross-Talk: Does a Novel Oncogenesis Driver Emerge?" Vaccines 10, no. 10: 1607. https://doi.org/10.3390/vaccines10101607

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop