Skip to main content

HYPOTHESIS AND THEORY article

Front. Immunol., 23 February 2022
Sec. Immunological Tolerance and Regulation
This article is part of the Research Topic Soluble Immune Checkpoints: Novel Physiological Immunomodulators View all 6 articles

Soluble Immune Checkpoints Are Dysregulated in COVID-19 and Heavy Alcohol Users With HIV Infection

Wei Li&#x;Wei Li1‡Fahim Syed&#x;Fahim Syed1‡Richard YuRichard Yu2Jing YangJing Yang1Ying Xia,&#x;Ying Xia1,3†Ryan F. RelichRyan F. Relich4Patrick M. RussellPatrick M. Russell4Shanxiang ZhangShanxiang Zhang4Mandana KhaliliMandana Khalili5Laurence HuangLaurence Huang5Melissa A. KacenaMelissa A. Kacena6Xiaoqun Zheng,Xiaoqun Zheng3,7Qigui Yu*Qigui Yu1*
  • 1Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
  • 2Department of Internal Medicine, School of Medicine, University of Nevada, Reno, NV, United States
  • 3School of Laboratory Medicine, Wenzhou Medical University, Wenzhou, China
  • 4Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
  • 5Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
  • 6Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
  • 7Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China

Immune checkpoints (ICPs) consist of paired receptor-ligand molecules that exert inhibitory or stimulatory effects on immune defense, surveillance, regulation, and self-tolerance. ICPs exist in both membrane and soluble forms in vivo and in vitro. Imbalances between inhibitory and stimulatory membrane-bound ICPs (mICPs) in malignant cells and immune cells in the tumor immune microenvironment (TIME) have been well documented. Blockades of inhibitory mICPs have emerged as an immense breakthrough in cancer therapeutics. However, the origin, structure, production regulation, and biological significance of soluble ICPs (sICPs) in health and disease largely remains elusive. Soluble ICPs can be generated through either alternative mRNA splicing and secretion or protease-mediated shedding from mICPs. Since sICPs are found in the bloodstream, they likely form a circulating immune regulatory system. In fact, there is increasing evidence that sICPs exhibit biological functions including (1) regulation of antibacterial immunity, (2) interaction with their mICP compartments to positively or negatively regulate immune responses, and (3) competition with their mICP compartments for binding to the ICP blocking antibodies, thereby reducing the efficacy of ICP blockade therapies. Here, we summarize current data of sICPs in cancer and infectious diseases. We particularly focus on sICPs in COVID-19 and HIV infection as they are the two ongoing global pandemics and have created the world’s most serious public health challenges. A “storm” of sICPs occurs in the peripheral circulation of COVID-19 patients and is associated with the severity of COVID-19. Similarly, sICPs are highly dysregulated in people living with HIV (PLHIV) and some sICPs remain dysregulated in PLHIV on antiretroviral therapy (ART), indicating these sICPs may serve as biomarkers of incomplete immune reconstitution in PLHIV on ART. We reveal that HIV infection in the setting of alcohol misuse exacerbates sICP dysregulation as PLHIV with heavy alcohol consumption have significantly elevated plasma levels of many sICPs. Thus, both stimulatory and inhibitory sICPs are present in the bloodstream of healthy people and their balance can be disrupted under pathophysiological conditions such as cancer, COVID-19, HIV infection, and alcohol misuse. There is an urgent need to study the role of sICPs in immune regulation in health and disease.

Introduction

Immune checkpoints (ICPs) consist of paired receptor-ligand molecules that exert inhibitory, stimulatory, or dual effects on immune regulation, surveillance, defense, and self-tolerance (15). Under normal circumstances, ICPs regulate the magnitude, breadth, and duration of the immune responses against infection and malignancy while protecting tissues from excessive insult. However, in pathological situations such as malignancy and persistent infection, the balance between ICP stimulatory and inhibitory functions becomes dysregulated (17). Malignancy can dysregulate the expression of ICPs on the surface of immune cells to evade or subvert the immune response, leading to insufficiency or failure of anti-tumor immune attacks. The up-regulated expression of inhibitory ICPs, including CTLA-4, LAG-3, PD-1, PD-L1, PD-L2, and TIM-3 has been found on the surface of both CD4 and CD8 T cells in cancer patients (6, 810). These important findings have laid the foundation for the clinical development of ICP blockade therapies, which abrogate ICP inhibitory signals, restoring and enhancing the anti-tumor activity of cytotoxic T lymphocytes (CTLs) (3, 5, 11, 12). Since 2011, ICP blockers targeting CTLA-4, PD-1, and PD-L1 have yielded unprecedented responses in a portion of cancer patients, leading to eight FDA-approved ICP blocking antibodies against these ICPs, including one anti-CTLA-4 antibody (Ipilimumab), four anti-PD-1 antibodies (Nivolumab, Pembrolizumab, Cemiplimab, and Dostarlimab), and three anti-PD-L1 antibodies (Avelumab, Durvalumab, and Atezolizumab), for treating several types of cancer such as melanoma, lung cancer, bladder cancer, liver cancer, kidney cancer, Merkel cell carcinoma, squamous cell skin cancer, endometrial cancer, mesothelioma, colorectal cancer, esophageal cancer, gastric cancer, head and neck cancer, lymphoma, breast cancer, and cervical cancer (5, 1316).

Similar to malignant cells, several pathogens, including HIV (the human immunodeficiency virus), HBV (hepatitis B virus), TB (tuberculosis), and malaria have been demonstrated to dysregulate ICPs to limit host-protective CTLs (6, 7, 17, 18). For example, in antiretroviral therapy (ART)-naïve people living with HIV (PLHIV), there is upregulated expression of multiple inhibitory ICPs including CTLA-4, PD-1, TIM-3, and LAG-3 on total and HIV-specific CD4 and CD8 T cells (68, 1822), which is associated with an accelerated decline in the number of CD4 T cells in PLHIV (8, 23). Following ART, expression of these ICPs on the surface of T cells declines, but remains elevated when compared with healthy controls (810). Importantly, expression of inhibitory ICPs can be used as surrogate immunological biomarkers of ART effectiveness. In ART-treated PLHIV, PD-1 expression on CD8 T cells has been associated with impaired reconstitution of CD4 T cells and a shorter time to viral rebound after stopping ART (24, 25). In addition, CD4 T cells expressing high levels of PD-1, LAG-3, and TIGIT alone or in combination are enriched for integrated HIV DNA during ART (26, 27). Furthermore, most CD4 T cells expressing at least one of these ICPs, carry inducible and replication-competent HIV genomes (26). Thus, CD4 T cells expressing inhibitory ICPs such as PD-1 and LAG-3 contribute to HIV persistence during ART. In vitro and ex vivo studies have shown that blocking antibodies against either PD-1, CTLA-4, LAG-3, or TIM-3 can significantly restore the proliferative capacities and functions of T cells and B cells from PLHIV on ART (22). Currently, there are several clinical trials in PLHIV with and without cancer using the FDA-approved ICP blockers (6, 22, 2832). These clinical trials have yielded mixed results ranging from little therapeutic benefit to significant expansions of HIV-specific CD4 and CD8 T cells in a subset of participants (2833), indicating ICP blockade for the treatment of HIV infection needs further study.

ICP molecules exist in both membrane and soluble forms in vivo and in vitro (3445). Similar to membrane-bound ICPs (mICPs), soluble ICPs (sICPs) are also present in normal physiological conditions and highly dysregulated in patients with cancer, viral infections, or alcohol-associated liver disease (ALD) (3446). Soluble ICPs can be generated through either secretion of protein isoforms encoded by alternative mRNA splicing that lack transmembrane domains or protease-mediated shedding from mICPs by actions of matrix metalloproteinases (MMPs) (45, 47). Although the detailed structure, production regulation, function, and clinical relevance of sICPs largely remain unknown, many sICPs exist in native forms that exhibit biological functions such as regulation of antibacterial immunity (43, 47). Since sICPs are paired receptor-ligand molecules and circulate in the bloodstream, they likely form a circulating immune regulatory system. In addition, increasing evidence has shown that sICPs interact with their mICP compartments to positively or negatively regulate immune responses (44). Furthermore, sICPs can compete with their mICP compartments for binding to the ICP blocking antibodies, thereby interrupting the efficacy of ICP blockade therapies. Thus, there is an urgent need to study the role of sICPs in immune regulation in health and disease. Here, we summarize current data of sICPs in cancer and infectious diseases. We particularly focus on sICPs in COVID-19 and HIV infection as they are the two ongoing global pandemics and have created the world’s most serious public health and development challenges.

Soluble Immune Checkpoints in the Peripheral Circulation of Healthy People

As described in our recent report (46), we used a multiplex immunoassay (the Human Immuno-Oncology Checkpoint Protein Panel, MilliporeSigma, Burlington, MA) to simultaneously quantify plasma concentrations of 16 sICPs (sBTLA, sCD27, sCD28, sCD40, sCD80/B7-1, sCD86/B7-2, sCTLA-4, sGITR, sGITRL, sHVEM, sICOS, sLAG-3, sPD-1, sPD-L1, sTIM-3, and sTLR-2) in healthy blood donors. Plasma levels of two additional ICPs (sCD160 and sLIGHT) were quantified using the Human CD160 Matched ELISA Antibody Pair Set (Sino Biological, Beijing, China) and the Human LIGHT Duoset ELISA Kit (R&D Systems, Minneapolis, MN), respectively. We found that, except for sGITR and sLIGHT, which were only detected in 30-50% of plasma samples, all other 16 sICPs were steadily detected in plasma samples from all healthy blood donors (Table 1). These sICPs could be generated through either alternative mRNA splicing and secretion or protease-mediated shedding from mICPs by actions of MMPs (45, 47). Several studies have demonstrated that sICPs are present in the peripheral blood as the native polypeptide products of their genes and have biological functions. For example, CTLA-4, also known as CD152, is a member of the immunoglobulin (Ig) gene superfamily (48). CTLA4 is constitutively expressed in regulatory T cells but can be upregulated in conventional T cells after activation (48, 49). CTLA-4 and CD28 are homologous receptors that share a pair of ligands (B7-1 and B7-2) expressed on the surface of antigen-presenting cells (APCs), but mediate opposing functions in T-cell activation (49). CTLA-4 interacts with its ligands to inhibit T-cell responses (49), while CD28 acts as a major co-stimulatory receptor in promoting full activation of T cells in response to T cell receptor (TCR) engagement (50). As shown in Table 1, plasma levels of sCTLA-4 in healthy individuals were detected at a median concentration of 31 pg/mL with the interquartile ranges of 12 – 81 pg/mL (n=39), which are higher than the detection limit (9.3 pg/mL) of the multiplex immunoassay. These results argue against a previous report showing that circulating CTLA-4 was undetectable in healthy volunteers using an enzyme immunoassay (EIA) (45). This is because the EIA has limited detection sensitivity (≥ 4 ng/mL) (45), which is insufficiently sensitive for the detection of sCTLA-4 concentrations in healthy people. Immunoprecipitation and Western blotting analyses of serum sCTLA-4 revealed a polypeptide consistent with the predicted size (23 kDa) from an alternative transcript of the CTLA-4 gene, suggesting sCTLA-4 is present as a native molecule rather than a product of proteolytic digestion or shedding of mCTLA-4 (45). Functional studies have shown that sCTLA-4 immunoreactivity can be blocked by B7.1 (also known as CD80), one of its known ligands. This supports the notion that sCTLA-4 is present as a soluble functional molecule. Thus, sCTLA-4 likely has important immunoregulatory functions, which is similar to soluble cytokine receptors such as soluble forms of TNF receptor, IL-2α receptor, IL-4 receptor, and IL-7 receptor that exist in the biological fluids and regulate cytokine activity in vitro and in vivo (5156).

TABLE 1
www.frontiersin.org

Table 1 Plasma levels of sICPs in healthy adults.

In contrast to sCTLA-4, several sICPs including sHVEM (also known as TNFRSF14 or CD270), sCD160, sLAG-3, and sTIM-3 were present at high concentrations (Table 1) in healthy people. These are potent inhibitory ICPs. HVEM was initially identified as the receptor of herpes simplex virus 1 (HSV-1) through binding to the HSV-1 glycoprotein D (gD) (57). Since then, HVEM has been identified as a co-signaling molecular switch through interacting with BTLA (also known as CD272), CD160, and LIGHT (58). In addition, HVEM can bind to SALM5 (synaptic adhesion-like molecule 5) to regulate neuroinflammation (59). We have recently reported that recombinant sHVEM affects TNF-α and IFN-γ production by anti-CD3/anti-CD28-stimulated T cells from healthy volunteers (46), indicating sHVEM may act as a circulating immune regulator. Like sCTLA-4 and sHVEM, other sICPs such as sLAG-3, sTIM-3, sPD-1, and sPD-L1 are biologically active and participate in immune regulation (40, 6062). Thus, the majority (if not all) of ICPs have soluble forms that are detectable in the peripheral blood of healthy individuals. Different sICPs are likely produced at different levels and at distinct checkpoints to fine-tune immune homeostasis in health, although their origin, production regulation, and biological function are yet to be discovered. Of note, these data related to plasma sICPs were obtained from adults with a median age of 42 years (26-52) and 64% were from males (46). Future study is needed to investigate whether sICP levels and functions in healthy people are affected by age, gender, and race.

Soluble Immune Checkpoints in Cancer

ICPs act as gatekeepers for immune responses and play a central role in immune homeostasis that is maintained by a precise balance between stimulatory and inhibitory ICPs on the surface of effector and regulatory immune cells. The immune homeostasis is a tightly regulated network which fails during tumor development due to an imbalance between inhibitory and stimulatory ICPs. Indeed, high levels of inhibitory ICPs on the surface of tumor cells is a hallmark of the tumor immune microenvironment (TIME) that is infiltrated with many types of innate and adaptive immune cells (63). Increased inhibitory ICPs are responsible for tumor immune escape and thereby have become major targets for cancer immunotherapy (5, 1315).

While mICPs have been extensively studied in cancer immunity and cancer immunotherapy, the origin, production regulation, and biological significance of sICPs largely remains elusive. Due to their function in both positive and negative immune regulation, sICPs and their levels change in the peripheral blood, which may affect the development, prognosis, and treatment of cancer. Studies have shown that plasma or serum levels of sICPs can serve as biomarkers and/or predictors of cancer patient outcomes or therapeutic responses (44, 64). Plasma or serum levels of numerous sICPs including sPD-L1, sPD-1, sLAG-3, sTIM-3, sCTLA-4, sHVEM, sCD80, sCD86, sCD27, sCD40, and sBTLA are highly elevated in patients with various types of tumors and serve as prognostic markers for solid tumors such as non-small cell lung cancer, gastric cancer, colon cancer, and cervical cancer (43, 61, 6568). Soluble ICPs are also biologically active in cancer patients. Studies have revealed that plasma/serum levels of sCD40L are highly elevated in patients with lung cancer and undifferentiated nasopharyngeal carcinoma (69, 70). The elevated sCD40L in cancer patients is likely derived from activated platelets rather than T cells, because cancer patients have significant platelet activation, but inadequate T-cell activation (7173). A functional study has shown that the upregulated sCD40L seen in cancer patients exerts an immunosuppressive effect through enhancement of MDSC (myeloid-derived suppressor cell)-mediated suppression of T cell proliferation and IFN-γ production, expansion of regulatory T cells (Treg), and enrichment of PD-1+ T cells (74). On the other hand, sPD-1 is likely generated through mRNA splicing and secretion, as four PD-1 splice variants have been identified (75). In vitro and in vivo studies have shown sPD-1 is able to bind its membrane-bound ligands (mPD-L1 and mPD-L2) to block mPD-1/mPD-L1/mPD-L2 interaction, thereby restoring T cell immunity (61). Indeed, local delivery of sPD-1 in the tumor microenvironment through adeno-associated virus-mediated delivery vector induces antitumor immunity through improving T cell function (76, 77). The origin, production regulation, function, and biological significance of sICPs in tumors have been systematically reviewed (44, 61, 78, 79). Collectively, the levels of sICPs in the peripheral circulation in cancer patient are frequently altered, which likely has clinical significances. That said, a better understanding of the underlying mechanisms of the sICP network could lay the foundation for the development of new strategies for treating cancers with immunotherapies.

Soluble Immune Checkpoints in Patients With COVID-19

Two recent studies, reported by Kong Y et al. (80) and Avendano-Ortiz J et al. (81), have demonstrated that a “storm” of sICPs occurs in COVID-19 patients and is associated with the severity of COVID-19 (80, 81). The Kong study quantified 14 sICPs including sBTLA, sCTLA-4, sGITR, sHVEM, sIDO, sLAG-3, sPD-1, sPD-L1, sPD-L2, sTIM-3, sCD27, sCD28, sCD80, and s4-1BB in the serum samples from patients with asymptomatic, mild/moderate, and severe/critical COVID-19 using the ProcartaPlex Human ImmunoOncology Checkpoint Panel (Invitrogen, Carlsbad, CA) (80), while the Avendano-Ortiz study quantified 9 sICPs including sCD25, sCD86, sCTLA-4, Galectin-9, sLAG-3, sPD-1, sPD-L1, sTim-3, and s4-1BB using the LEGENDplex HU Immune Checkpoint Panel 1 (BioLegend, San Diego, CA) (81). After merging the overlapping 6 sICPs that were detected in both studies, a total of 17 sICPs including sBTLA, sCTLA-4, sGalectin-9, sGITR, sHVEM, sIDO, sLAG-3, sPD-1, sPD-L1, sPD-L2, sTIM-3, sCD25, sCD27, sCD28, sCD80, sCD86, and s4-1BB were studied in the serum or plasma samples from COVID-19 patients (80, 81). The Kong study showed that, except for sPD-L2, each of the other 13 sICPs was significantly higher in the severe/critical group than in the mild/moderate and asymptomatic groups (80). On the other hand, the Avendano-Ortiz study showed that plasma levels of sCD25, sCD86, Galectin-9, sPD-1, sPD-L1, and sTim-3, but not sLAG-3, sCTLA-4, and s4-1BB, were significantly higher in the severe/critical group than in the mild/moderate groups (81). Therefore, both studies demonstrated that the serum or plasma levels of sPD-1, sPD-L1, and sTIM-3 were significantly higher in the severe/critical group than in the mild/moderate and asymptomatic groups, but their data conflicted regarding the serum or plasma levels of sLAG-3, sCTLA-4, and s4-1BB between healthy controls and COVID-19 patients (81). The Kong study also showed that serum levels of 11 sICPs (sGITR, s4-1BB, sTIM-3, sCD27, sLAG-3, sPD-1, sCD28, sCTLA-4, sBTLA, sHVEM, and sCD80) were persistently higher in severe/critical patients than in mild/moderate cases during hospitalization. In addition, the levels of 8 sICPs (sIDO, sGITR, s4-1BB, sTIM-3, sCD27, sLAG-3, sPD-1, and sCD28) were negatively correlated with absolute counts of CD4 and CD8 T cells. The Avendano-Ortiz study also demonstrated that plasma levels of sCD25, sTIM-3, Galectin-9, and sPD-L1, but not sCD86, showed a negative correlation with the absolute lymphocyte count (ALC). These results suggest that sICPs are dysregulated in COVID-19 and sICP dysregulation may be linked to COVID-19 lymphopenia, an abnormal reduction in lymphocyte numbers. Lymphopenia is a prominent clinical feature of COVID-19 patients and has been associated with the severity of COVID-19 (8287). Indeed, non-survivors of COVID-19 have a significantly lower lymphocyte count than survivors (83, 88). The absolute cell counts of lymphoid lineage cells, including T cells, B cells, and NK cells, are abnormally reduced with a more pronounced decrease in CD8 T cells (8991). In contrast, myeloid lineage cells such as neutrophils are highly increased in the blood of patients with severe COVID-19 (84), which is noted as a major clinical feature of severe COVID-19 (92). The mechanisms of COVID-19 lymphopenia remain unclear, although several hypotheses are proposed including a cytokine storm impact (9395), direct infection of immune cells (96), overaggressive T cell responses (97), and lymphocyte infiltration and sequestration in the lungs (88). However, these hypotheses have been challenged, because (1) most COVID-19 patients do not have remarkably high levels of inflammatory cytokines, as only 4% of critically ill COVID-19 patients develop cytokine storm symptom (CSS) and anti-CSS medications have no benefit for most COVID-19 patients (84, 98102), (2) direct viral infection is an unlikely cause of immune cell loss (103), as infectious SARS-CoV-2 has not been successfully isolated from peripheral blood cells in COVID-19 patients (95), (3) the overall magnitude of the T cell response in COVID-19 patients is either insufficient or excessive remains debated (97), as T cell responses are insufficient in some COVID-19 patients, but excessive in others (97), and (4) post-mortem biopsies from COVID-19 patients with marked lymphopenia reveal prominent infiltration of neutrophils, but neither T cells nor B cells, in the lungs (104107). Thus, studies are urgently needed to determine the cause and impact of the commonly observed lymphopenia in patients with severe COVID-19, and whether dysregulated sICPs are associated with the pathogenesis of COVID-19 lymphopenia.

We also used a multiplex immunoassay (the Human Immuno-Oncology Checkpoint Protein Panel, MilliporeSigma, Burlington, MA) to simultaneously quantify the concentrations of 16 sICPs in plasma samples from healthy controls (n=23) and patients with asymptomatic (n=15) or hospitalized (severe/critical) COVID-19 (n=24). Among these 16 sICPs, 4 (sCD40, sGITRL, sICOS, and sTLR-2) were not previously studied in COVID-19 patients, while 12 were already tested in the two studies by Kong and Avendano-Ortiz et al. In our studies, the healthy control subjects were matched with the COVID-19 patients in terms of age, sex, and race. We found that plasma levels of the majority of 16 sICPs were significantly higher in COVID-19 patients when compared with healthy controls (data not shown). The 4 sICPs (sCD40, sGITRL, sICOS, and sTLR-2) that had not previously been studied in COVID-19 were significantly higher in the plasma from patients with asymptomatic or severe/critical COVID-19 when compared with healthy controls (Figure 1A). Plasma levels of sGITRL, sICOS, and sTLR-2, but not sCD40 were further elevated in severe/critical COVID-19 patients than in asymptomatic cases (Figure 1A). We also found that sCTLA-4 and sLAG-3, 2 sICPs that were studied by Kong and Avendano-Ortiz et al. with conflicting results, were elevated in plasma from patients with severe/critical COVID-19 when compared with healthy controls (Figure 1B), which is in agreement with Kong’s results.

FIGURE 1
www.frontiersin.org

Figure 1 Plasma levels of sICPs were highly elevated in COVID-19. (A) Scatter plots demonstrating the plasma levels of 4 sICPs that were not previously studied in COVID-19. (B) Scatter plots demonstrating the plasma levels of 2 sICPs in COVID-19 that were previously reported with different results. Kruskal-Wallis test with Dunn’s corrections for pairwise comparisons among hospitalized (severe/critical) COVID-19 patients (Hosp), SARS-CoV-2-infected individuals without symptoms (Asym), and healthy controls (HCs). Red lines represent the mean and the standard error of the mean. ns, no significant; *p < 0.05; **p < 0.01; ***p < 0.001.

Taken together, a storm of sICPs occurs in the peripheral circulation of COVID-19 patients and is associated with the disease severity. The circulating sICP levels on admission appear to be better mortality predictors than inflammatory cytokines and chemokines (81), and thereby can potentially serve as biomarkers of COVID-19 progress and outcome. Given that some, if not all, sICPs are biological active, they may also serve as circulating immune regulators or pharmaceutical targets for COVID-19 therapy. To this end, mechanistic studies and large-scale, cross-sectional and longitudinal studies are needed to investigate the origin, production regulation, and clinical significance of sICPs in patients with COVID-19.

Soluble Immune Checkpoints in People Living With HIV (PLHIV)

Chronic immune activation and exhaustion are important features of persistent viral infections such as infection with HIV (108111). Indeed, immune exhaustion represents a barrier to effective and specific immunity against HIV infection (111, 112). Chronic immune activation and exhaustion are at least in part attributed to the dysregulation of ICPs (112). In addition to mICPs that have been demonstrated to play a critical role in immune homeostasis in PLHIV (610, 1823), sICPs may also be dysregulated in PLHIV and thereby contribute to immune exhaustion in PLHIV. A recent study used ELISA to quantify plasma levels of sPD-L1 in PLHIV and healthy controls and found that plasma levels of sPD-L1 were significantly elevated in PLHIV and remained high despite control of HIV infection by ART (113). In addition, PLHIV on ART with virological failure had the highest plasma levels of sPD-L1 (113). Thus, sPD-L1 in the peripheral blood represents a potential biomarker of immune exhaustion and virological failure in PLHIV.

Here, we simultaneously quantified plasma levels of 16 sICPs from 23 healthy controls, 46 PLHIV who were ART-naïve, and 65 PLHIV who were on ART using a multiplex immunoassay as detailed above. These three groups of study subjects were matched in terms of demographic parameters including age, gender, and race. As shown in Figure 2, except GITR which was only detected in 34.8% (8/23) of healthy controls, each of the other 15 sICPs was detectable in the plasma samples from healthy controls, indicating that they could play biological roles in immune homeostasis under physiologic conditions. Compared to healthy controls, ART-naïve PLHIV had significantly higher plasma levels of the 15 sICPs tested. Specifically, only sTIM-3 was not elevated in ART-naïve PLHIV. In comparison to healthy controls, PLHIV on ART only had higher plasma levels of 3 sICPs (sCD40, sCTLA-4, and sHVEM) (Figure 2). These findings appear to indicate that ART effectively, but not completely, restores ICP homeostasis. Among these 3 sICPs that remained at higher levels in the peripheral blood of PLHIV on ART, sHVEM and sCD40 were not affected by ART, while sCTLA-4 was dramatically reduced, but did not return to a normal level (Figure 2). Both CD40 (also known as TNFRSF5) and HVEM (also known as TNFRSF14) are members of the tumor necrosis factor receptor superfamily. CD40 is a co-stimulatory molecule that is mainly expressed on the surface of APCs such as dendritic cells, monocytes/macrophages, and B cells. CD40 is required for APC activation via binding to CD154 (also known as CD40 ligand or CD40L) on T cells. CD40-CD40L interaction leads to the initiation of bidirectional intracellular signaling in both CD40+ APCs and CD40L+ T cells, resulting in APC activation and T cell responses (114116). Dysregulation of CD40/CD40L expression and interactions contributes to the severity in numerous diseases such as HIV infection (117119), cancer (114, 120), and autoimmune disorders (121123). However, the role of sCD40 in the peripheral circulation of PLHIV largely remains elusive. We found that ART-naïve PLHIV had excessive production of sCD40, which was minimally affected by ART, suggesting that circulating sCD40 may represent an indicator of dysregulation of APC and T cell function that is a hallmark of HIV-associated deficiency in cell-mediated immunity.

FIGURE 2
www.frontiersin.org

Figure 2 Plasma levels of sICPs were highly dysregulated in PLHIV. Scatter plots demonstrating the plasma levels of sICPs in HCs (healthy controls), ART-naïve PLHIV, and PLHIV on ART. Kruskal-Wallis test with Dunn’s corrections for pairwise comparisons among PLHIV on ART, ART-naïve PLHIV, and HCs. Red lines represent the mean and the standard error of the mean. ns, no significant; *p < 0.05; **p < 0.01; ***p < 0.001. HC, healthy control; ART-, people living with HIV (PLHIV) who were not treated with antiretroviral therapy (ART); and ART+, people living with HIV (PLHIV) who were treated with antiretroviral therapy (ART).

Similar to sCD40, sHVEM was also excessively produced in PLHIV and was barely affected by ART (Figure 2). HVEM serves as a shared receptor/ligand for stimulatory and inhibitory ligands/receptors, including LIGHT, BTLA, and CD160 that are expressed on both hematopoietic and non-hematopoietic cells. HVEM acts as a bifunctional ligand/receptor that exhibits costimulatory signals upon binding to LIGHT and co-inhibitory signals upon binding to BTLA or CD160 (58, 124). Due to its role of bifunctional ligand/receptor, HVEM serves as a molecular switch between stimulatory and inhibitory signaling (125, 126), thereby playing a unique role in immune homeostasis. Currently, the regulation and function of sCD27, sHVEM, and other sICPs in HIV immunopathogenesis remains unclear. Thus, studies are needed to elucidate the mechanisms of action for the sICP axis and the interplay between sICPs and mICPs in HIV infection.

Soluble Immune Checkpoints in Heavy Alcohol Users With HIV Infection

Alcohol misuse and HIV infection are both major health issues worldwide. Globally, more than 2 billion people consume alcohol on a regular basis, and approximately 76 million suffer from alcohol-related disorders (127, 128). Long-term heavy alcohol users develop a spectrum of ALD, ranging from AH, fibrosis/cirrhosis, to hepatocellular carcinoma (HCC) (129). Alcohol overconsumption contributes to 5.1% of the global burden of diseases and causes approximately 3.3 million deaths every year (130, 131). HIV is the causative agent of AIDS and has claimed over 36 million lives with an estimated 38 million PLHIV worldwide at the end of 2020 (132). Alcohol overconsumption is common among PLHIV and adversely influences the health outcomes by increasing HIV-associated comorbidities such as liver disease, cardiovascular disease, pulmonary disease, bone disease, and cancer (133135). It is well known that alcohol overconsumption and HIV infection independently damage the gastrointestinal (GI) tract mucosal barrier, leading to a leaky gut that allows microbial translocation and accumulation of microbial components such as lipopolysaccharide (LPS) in the blood. Specifically, alcohol disrupts gap junction integrity of gut mucosal epithelial cells, leading to increased GI permeability and translocation of microbial components such as LPS from the GI tract into the blood and liver (136138). Alcohol-induced microbial translocation has been considered a major driver of chronic immune activation and inflammation in AH patients (139142). In PLHIV, microbial translocation is a cause of chronic immune activation and inflammation, which is a hallmark of progressive HIV infection and a stronger predictor of disease outcome compared to plasma viral load (143, 144). We therefore hypothesize that alcohol overconsumption and HIV infection exacerbate microbial translocation, immune dysregulation, and inflammation, thereby accelerating disease progression of HIV infection and ALD. To test this hypothesis, we established a cohort of heavy alcohol users with and without HIV infection. We analyzed and compared the profiles of sICPs in the peripheral blood in heavy drinkers without overt ALD (HDC) versus PLHIV on ART (HIV) versus PLHIV on ART who were heavy drinkers, but did not have ALD (HDC + HIV). We found that plasma levels of all 16 sICPs were similar between HDC and HIV groups (Table 2). Fourteen out of sixteen sICPs examined were dramatically elevated in the peripheral blood in HDC+HIV when compared with either HDC or HIV (Table 2). sCD27 and sTIM-3 were not elevated in HDC + HIV compared with either HDC or HIV (Table 2). These results indicate that sICPs were highly dysregulated in HDC + HIV even though these individuals had no clinical evidence of overt ALD and their liver enzyme parameters including the circulating levels of AST and ALT and AST : ALT ratio were similar to healthy individuals (data not shown). Previous studies from our group and others have demonstrated that chronic excessive drinking leads to immune abnormalities in heavy drinkers even when there are no obvious signs of clinical liver disease (142, 146148). HDCs have increased bacterial translocation as they have higher serum levels of LPS and markers of monocyte/macrophage activation (sCD14 and sCD163) than non-excessive drinkers (147). In addition, mucosal-associated invariant T (MAIT) cells in the peripheral blood are significantly decreased in HDCs compared to healthy controls (148). Moreover, HDCs have increased levels of MAIT activation-associated cytokines such as IL-18 and IL-12 (148). MAIT cells are innate-like lymphocytes that are highly enriched in liver, mucosa, and peripheral blood, and play a protective role in antimicrobial immunity (149, 150). These results not only highlight the presence of immune dysregulation in HDCs even when there are no obvious signs of ALD but also suggest that altered sICPs in the peripheral blood can potentially be used as surrogate biomarkers of immune dysregulation in HDCs. As we previously reported, plasma levels of sHVEM were highly dysregulated in heavy alcohol users with ALD, specifically alcoholic hepatitis (AH), when compared with heavy alcohol users without AH (46). Plasma levels of upregulated sHVEM in AH patients remained high for 6 months of complete alcohol abstinence (46), indicating sHVEM might serve as a prognostic marker for AH. We also found that sHVEM-his, consisting of the soluble extracellular domain of human mHVEM linked to a polyhistidine tag at the C-terminus, significantly inhibited TCR-induced TNF-α production by both CD4 T cells and CD8 T cells from AH patients and healthy controls (46), indicating sHVEM plays an inhibitory role in HVEM axis-mediated TNF-α production. Therefore, sHVEM levels in the peripheral blood may serve a biomarker of immune abnormalities that are exacerbated in heavy alcohol users with HIV infection. Future study is needed to investigate the mechanisms underlying exacerbated sICP dysregulation in heavy drinkers with HIV infection.

TABLE 2
www.frontiersin.org

Table 2 Plasma levels of sICPs in heavy alcohol users with HIV infection.

Current Progress and Future Perspectives of ICP Blockade in COVID-19 and PLHIV

As described above, there are eight FDA-approved ICP blocking antibodies against CTLA-4 (Ipilimumab), PD-1 (Nivolumab, Pembrolizumab, Cemiplimab, and Dostarlimab), or PD-L1 (Avelumab, Durvalumab, and Atezolizumab) for cancer immunotherapy (5, 1316). These ICP blocking antibodies are also being explored as vital components of the complex therapeutic strategies for treating cancer patients with COVID-19 or HIV infection (16, 22, 81, 151154), and appear to be generally tolerable (16, 155). Further studies are needed to investigate the potential risks and benefits of ICP blockade therapy in COVID-19 patients and PLHIV who have no cancer. Indeed, numerous clinical trials are currently investigating the efficacy and safety of ICP blocking antibodies as an add-on immunotherapy to the standard of care for the treatment of COVID-19 patients and PLHIV (Table 3). One of these clinical trials tests Abatacept as an immune modulator for treating COVID-19 (NCT04593940). Abatacept is a fusion protein consisting of the extracellular domain of CTLA-4 fused to the Fc region of the immunoglobulin IgG (CTLA-4Fc), thereby representing a soluble form of CTLA-4 (sCTLA-4). This is the only clinical trial studying sICPs (sCTLA-4) for treating patients with moderate or severe COVID-19, while others test ICP blocking antibodies to block mICP. Abatacept was developed by Bristol-Myers Squibb and received an approval from the FDA in 2005 to treat autoimmune diseases such as rheumatoid arthritis (RA), psoriatic arthritis, and juvenile idiopathic arthritis (156159). CTLA-4 is considered the “leader” of inhibitory ICPs as it potently inhibits T cell responses at the initial stage of T cell activation (160). CTLA-4 is a CD28 homolog with much higher binding affinity for CD80 (B7-1) and CD86 (B7-2) on APCs than CD28 (160), thereby competing with CD28 for binding of CD80/CD86. CD28 is highly and constitutively expressed on resting and activated T cells, while CTLA-4 expression is induced in response to TCR engagement (161). As CD28 is one of the most important co-stimulatory receptors necessary for full T cell activation, CTLA-4 functions by scavenging CD80/86 ligands away from CD28, thereby down-regulating T cell immunity. It is expected that sCTLA-4Fc ameliorates inflammation and exacerbated T cell responses in COVID-19 patients. Thus, sICPs are promising targets or therapeutic agents for treating infectious diseases such as COVID-19 and HIV infection in addition to serving as surrogate immunological biomarkers and/or predictors of disease severity and progression. However, ICP blockade therapy for infectious diseases is still in the early stages and a lot more research needs to be done. A plethora of ICPs have been discovered, which may exert inhibitory, co-stimulatory, or dual activities. Each of these ICPs possesses unique properties that set it apart from the others. In addition, these ICPs are differentially regulated in different diseases. Their distinct biological properties and functional profiles should be taken into account in therapeutic strategies that aim to enhance immune responses or inhibit inflammation to combat infectious diseases.

TABLE 3
www.frontiersin.org

Table 3 Registered clinical trials studying ICPs for treating COVID-19 and HIV infection.

Conclusion

ICP molecules exist in both membrane and soluble forms in vivo and in vitro. Imbalance between inhibitory and stimulatory mICPs in malignant cells and immune cells in tumor microenvironment has been well documented and blockade of inhibitory mICPs has emerged as an immense breakthrough in cancer therapeutics. While mICPs have been extensively studied, their soluble compartments have not been adequately studied. sICPs can be generated through either alternative mRNA splicing and secretion or protease-mediated shedding from mICPs. However, the cellular resource, structure, production regulation, and biological significance of sICPs in health and disease largely remain elusive. Here, we summarize current data of sICPs in cancer and infectious diseases. A storm of sICPs occurs in the peripheral circulation of COVID-19 patients and is associated with the severity of COVID-19. Similarly, sICPs are highly dysregulated in PLHIV and some sICPs remain dysregulated in PLHIV on ART, indicating these sICPs may serve as biomarkers of incomplete immune reconstitution for PLHIV on ART. Strikingly, HIV infection and alcohol misuse dramatically exacerbate sICP dysregulation. Thus, both stimulatory and inhibitory sICPs are present in the bloodstream of healthy people and their balance can be disrupted under pathophysiological conditions such as cancer, COVID-19, HIV infection, and alcohol misuse. Further studies are needed to investigate whether sICPs act as critical circulating immune regulators in health and disease and whether sICPs can be used as pharmaceutical targets or agents for infectious disease therapy.

Ethics Statement

This study was performed with the approval of the Institutional Review Board at Indiana University School of Medicine and University of California San Francisco. Blood samples were drawn after each participant provided a written informed consent form.

Author Contributions

WL, FS, JY, and YX performed experiments. WL, FS, RY, RR, PR, SZ, MK, and LH provided clinical samples, discussed results, and contributed to manuscript writing. MK and XZ discussed results and contributed to manuscript writing. QY provided funding, designed experiments, supervised lab, coordinated clinical sample collection, and wrote manuscript. All authors contributed to the article and approved the submitted version.

Funding

This work was supported by the National Institute on Alcohol Abuse and Alcoholism (grant number UH2AA026218 to QY); the Bill and Melinda Gates Foundation (grant numbers 53183 and OPP1035237 to QY); the Indiana Biobank and the Indiana Clinical and Translational Sciences Institute funded by the NIH (grant number UL1TR002529); and the National Center for Advancing Translational Sciences, Clinical and Translational Sciences Award. MK was also in part supported by the National Institute of Alcohol Abuse and Alcoholism (grant number K24AA022523).

Author Disclaimer

The content of this article is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health (NIH) or the Bill and Melinda Gates Foundation.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Glossary

www.frontiersin.org

References

1. Sanmamed MF, Chen L. A Paradigm Shift in Cancer Immunotherapy: From Enhancement to Normalization. Cell (2018) 175:313–26. doi: 10.1016/j.cell.2018.09.035

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Chen L, Flies DB. Molecular Mechanisms of T Cell Co-Stimulation and Co-Inhibition. Nat Rev Immunol (2013) 13:227–42. doi: 10.1038/nri3405

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Sharma P, Allison JP. The Future of Immune Checkpoint Therapy. Science (2015) 348:56–61. doi: 10.1126/science.aaa8172

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced Expression of PD-1, A Novel Member of the Immunoglobulin Gene Superfamily, Upon Programmed Cell Death. EMBO J (1992) 11:3887–95. doi: 10.1002/j.1460-2075.1992.tb05481.x

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Wei SC, Duffy CR, Allison JP. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov (2018) 8:1069–86. doi: 10.1158/2159-8290.CD-18-0367

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Wykes MN, Lewin SR. Immune Checkpoint Blockade in Infectious Diseases. Nat Rev Immunol (2018) 18:91–104. doi: 10.1038/nri.2017.112

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Dyck L, Mills KHG. Immune Checkpoints and Their Inhibition in Cancer and Infectious Diseases. Eur J Immunol (2017) 47:765–79. doi: 10.1002/eji.201646875

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Trautmann L, Janbazian L, Chomont N, Said EA, Gimmig S, Bessette B, et al. Upregulation of PD-1 Expression on HIV-Specific CD8+ T Cells Leads to Reversible Immune Dysfunction. Nat Med (2006) 12:1198–202. doi: 10.1038/nm1482

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Kaufmann DE, Kavanagh DG, Pereyra F, Zaunders JJ, Mackey EW, Miura T, et al. Upregulation of CTLA-4 by HIV-Specific CD4+ T Cells Correlates With Disease Progression and Defines a Reversible Immune Dysfunction. Nat Immunol (2007) 8:1246–54. doi: 10.1038/ni1515

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Chew GM, Fujita T, Webb GM, Burwitz BJ, Wu HL, Reed JS, et al. TIGIT Marks Exhausted T Cells, Correlates With Disease Progression, and Serves as a Target for Immune Restoration in HIV and SIV Infection. PloS Pathog (2016) 12:e1005349. doi: 10.1371/journal.ppat.1005349

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Topalian SL, Drake CG, Pardoll DM. Immune Checkpoint Blockade: A Common Denominator Approach to Cancer Therapy. Cancer Cell (2015) 27:450–61. doi: 10.1016/j.ccell.2015.03.001

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Pardoll DM. The Blockade of Immune Checkpoints in Cancer Immunotherapy. Nat Rev Cancer (2012) 12:252–64. doi: 10.1038/nrc3239

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Hargadon KM, Johnson CE, Williams CJ. Immune Checkpoint Blockade Therapy for Cancer: An Overview of FDA-Approved Immune Checkpoint Inhibitors. Int Immunopharmacol (2018) 62:29–39. doi: 10.1016/j.intimp.2018.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Berghoff AS, Venur VA, Preusser M, Ahluwalia MS. Immune Checkpoint Inhibitors in Brain Metastases: From Biology to Treatment. Am Soc Clin Oncol Educ Book (2016) 35:e116–122. doi: 10.1200/EDBK_100005

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Vaddepally RK, Kharel P, Pandey R, Garje R, Chandra AB. Review of Indications of FDA-Approved Immune Checkpoint Inhibitors Per NCCN Guidelines With the Level of Evidence. Cancers (Basel) (2020) 12(3). doi: 10.3390/cancers12030738

CrossRef Full Text | Google Scholar

16. Castelli V, Lombardi A, Palomba E, Bozzi G, Ungaro R, Alagna L, et al. Immune Checkpoint Inhibitors in People Living With HIV/AIDS: Facts and Controversies. Cells (2021) 10(9). doi: 10.3390/cells10092227

CrossRef Full Text | Google Scholar

17. Naran K, Nundalall T, Chetty S, Barth S. Principles of Immunotherapy: Implications for Treatment Strategies in Cancer and Infectious Diseases. Front Microbiol (2018) 9:3158. doi: 10.3389/fmicb.2018.03158

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Rao M, Valentini D, Dodoo E, Zumla A, Maeurer M. Anti-PD-1/PD-L1 Therapy for Infectious Diseases: Learning From the Cancer Paradigm. Int J Infect Dis (2017) 56:221–8. doi: 10.1016/j.ijid.2017.01.028

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Petrovas C, Casazza JP, Brenchley JM, Price DA, Gostick E, Adams WC, et al. PD-1 Is a Regulator of Virus-Specific CD8+ T Cell Survival in HIV Infection. J Exp Med (2006) 203:2281–92. doi: 10.1084/jem.20061496

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, et al. PD-1 Expression on HIV-Specific T Cells Is Associated With T-Cell Exhaustion and Disease Progression. Nature (2006) 443:350–4. doi: 10.1038/nature05115

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Velu V, Shetty RD, Larsson M, Shankar EM. Role of PD-1 Co-Inhibitory Pathway in HIV Infection and Potential Therapeutic Options. Retrovirology (2015) 12:14. doi: 10.1186/s12977-015-0144-x

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Sperk M, Domselaar RV, Neogi U. Immune Checkpoints as the Immune System Regulators and Potential Biomarkers in HIV-1 Infection. Int J Mol Sci (2018) 19(7). doi: 10.3390/ijms19072000

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Hoffmann M, Pantazis N, Martin GE, Hickling S, Hurst J, Meyerowitz J, et al. Exhaustion of Activated CD8 T Cells Predicts Disease Progression in Primary HIV-1 Infection. PloS Pathog (2016) 12:e1005661. doi: 10.1371/journal.ppat.1005661

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Shive CL, Clagett B, McCausland MR, Mudd JC, Funderburg NT, Freeman ML, et al. Inflammation Perturbs the IL-7 Axis, Promoting Senescence and Exhaustion That Broadly Characterize Immune Failure in Treated HIV Infection. J Acquir Immune Defic Syndr (2016) 71:483–92. doi: 10.1097/QAI.0000000000000913

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Hurst J, Hoffmann M, Pace M, Williams JP, Thornhill J, Hamlyn E, et al. Immunological Biomarkers Predict HIV-1 Viral Rebound After Treatment Interruption. Nat Commun (2015) 6:8495. doi: 10.1038/ncomms9495

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Fromentin R, Bakeman W, Lawani MB, Khoury G, Hartogensis W, DaFonseca S, et al. CD4+ T Cells Expressing PD-1, TIGIT and LAG-3 Contribute to HIV Persistence During ART. PloS Pathog (2016) 12:e1005761. doi: 10.1371/journal.ppat.1005761

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, et al. HIV Reservoir Size and Persistence Are Driven by T Cell Survival and Homeostatic Proliferation. Nat Med (2009) 15:893–900. doi: 10.1038/nm.1972

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Wightman F, Solomon A, Kumar SS, Urriola N, Gallagher K, Hiener B, et al. Effect of Ipilimumab on the HIV Reservoir in an HIV-Infected Individual With Metastatic Melanoma. AIDS (2015) 29:504–6. doi: 10.1097/QAD.0000000000000562

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Le Garff G, Samri A, Lambert-Niclot S, Even S, Lavole A, Cadranel J, et al. Transient HIV-Specific T Cells Increase and Inflammation in an HIV-Infected Patient Treated With Nivolumab. AIDS (2017) 31:1048–51. doi: 10.1097/QAD.0000000000001429

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Guihot A, Marcelin AG, Massiani MA, Samri A, Soulie C, Autran B, et al. Drastic Decrease of the HIV Reservoir in a Patient Treated With Nivolumab for Lung Cancer. Ann Oncol (2018) 29:517–8. doi: 10.1093/annonc/mdx696

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Polizzotto MN, Chen G, Tressler RL, Godfrey C. Leveraging Cancer Therapeutics for the HIV Cure Agenda: Current Status and Future Directions. Drugs (2015) 75:1447–59. doi: 10.1007/s40265-015-0426-6

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Davar D, Wilson M, Pruckner C, Kirkwood JM. PD-1 Blockade in Advanced Melanoma in Patients With Hepatitis C and/or HIV. Case Rep Oncol Med (2015) 2015:737389. doi: 10.1155/2015/737389

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Gay CL, Bosch RJ, Ritz J, Hataye JM, Aga E, Tressler RL, et al. Clinical Trial of the Anti-PD-L1 Antibody BMS-936559 in HIV-1 Infected Participants on Suppressive Antiretroviral Therapy. J Infect Dis (2017) 215:1725–33. doi: 10.1093/infdis/jix191

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Riva A, Chokshi S. Immune Checkpoint Receptors: Homeostatic Regulators of Immunity. Hepatol Int (2018) 12:223–36. doi: 10.1007/s12072-018-9867-9

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Li YM, Shi YY, Li Y, Yan L, Tang JT, Bai YJ, et al. Soluble Tim-3 and Gal-9 Are Associated With Renal Allograft Dysfunction in Kidney Transplant Recipients: A Cross-Sectional Study. Int Immunopharmacol (2018) 55:330–5. doi: 10.1016/j.intimp.2018.01.008

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Chen D, Peng W, Jiang H, Yang H, Wu J, Wang H, et al. Noninvasive Detection of Acute Renal Allograft Rejection by Measurement of Soluble Tim-3 in Urine. Mol Med Rep (2017) 16:915–21. doi: 10.3892/mmr.2017.6670

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Dahal LN, Basu N, Youssef H, Khanolkar RC, Barker RN, Erwig LP, et al. Immunoregulatory Soluble CTLA-4 Modifies Effector T-Cell Responses in Systemic Lupus Erythematosus. Arthritis Res Ther (2016) 18:180. doi: 10.1186/s13075-016-1075-1

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Ren F, Li J, Jiang X, Xiao K, Zhang D, Zhao Z, et al. Plasma Soluble Tim-3 Emerges as an Inhibitor in Sepsis: Sepsis Contrary to Membrane Tim-3 on Monocytes. Tissue Antigens (2015) 86:325–32. doi: 10.1111/tan.12653

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Simone R, Pesce G, Antola P, Rumbullaku M, Bagnasco M, Bizzaro N, et al. The Soluble Form of CTLA-4 From Serum of Patients With Autoimmune Diseases Regulates T-Cell Responses. BioMed Res Int (2014) 2014:215763. doi: 10.1155/2014/215763

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Casati C, Camisaschi C, Rini F, Arienti F, Rivoltini L, Triebel F, et al. Soluble Human LAG-3 Molecule Amplifies the In Vitro Generation of Type 1 Tumor-Specific Immunity. Cancer Res (2006) 66:4450–60. doi: 10.1158/0008-5472.CAN-05-2728

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Haile ST, Horn LA, Ostrand-Rosenberg S. A Soluble Form of CD80 Enhances Antitumor Immunity by Neutralizing Programmed Death Ligand-1 and Simultaneously Providing Costimulation. Cancer Immunol Res (2014) 2:610–5. doi: 10.1158/2326-6066.CIR-13-0204

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Zhu X, Lang J. Soluble PD-1 and PD-L1: Predictive and Prognostic Significance in Cancer. Oncotarget (2017) 8:97671–82. doi: 10.18632/oncotarget.18311

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Heo SK, Ju SA, Kim GY, Park SM, Back SH, Park NH, et al. The Presence of High Level Soluble Herpes Virus Entry Mediator in Sera of Gastric Cancer Patients. Exp Mol Med (2012) 44:149–58. doi: 10.3858/emm.2012.44.2.010

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Gu D, Ao X, Yang Y, Chen Z, Xu X. Soluble Immune Checkpoints in Cancer: Production, Function and Biological Significance. J Immunother Cancer (2018) 6:132. doi: 10.1186/s40425-018-0449-0

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Oaks MK, Hallett KM. Cutting Edge: A Soluble Form of CTLA-4 in Patients With Autoimmune Thyroid Disease. J Immunol (2000) 164:5015–8. doi: 10.4049/jimmunol.164.10.5015

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Li W, Xia Y, Yang J, Guo H, Sun G, Sanyal AJ, et al. Immune Checkpoint Axes Are Dysregulated in Patients With Alcoholic Hepatitis. Hepatol Commun (2020) 4:588–605. doi: 10.1002/hep4.1475

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Oaks MK, Hallett KM, Penwell RT, Stauber EC, Warren SJ, Tector AJ. A Native Soluble Form of CTLA-4. Cell Immunol (2000) 201:144–53. doi: 10.1006/cimm.2000.1649

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Brunet JF, Denizot F, Luciani MF, Roux-Dosseto M, Suzan M, Mattei MG, et al. A New Member of the Immunoglobulin Superfamily–CTLA-4. Nature (1987) 328:267–70. doi: 10.1038/328267a0

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Rowshanravan B, Halliday N, Sansom DM. CTLA-4: A Moving Target in Immunotherapy. Blood (2018) 131:58–67. doi: 10.1182/blood-2017-06-741033

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Esensten JH, Helou YA, Chopra G, Weiss A, Bluestone JA. CD28 Costimulation: From Mechanism to Therapy. Immunity (2016) 44:973–88. doi: 10.1016/j.immuni.2016.04.020

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Schall TJ, Lewis M, Koller KJ, Lee A, Rice GC, Wong GH, et al. Molecular Cloning and Expression of a Receptor for Human Tumor Necrosis Factor. Cell (1990) 61:361–70. doi: 10.1016/0092-8674(90)90816-W

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Josimovic-Alasevic O, Herrmann T, Diamantstein T. Demonstration of Two Distinct Forms of Released Low-Affinity Type Interleukin 2 Receptors. Eur J Immunol (1988) 18:1855–7. doi: 10.1002/eji.1830181133

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Mosley B, Beckmann MP, March CJ, Idzerda RL, Gimpel SD, VandenBos T, et al. The Murine Interleukin-4 Receptor: Molecular Cloning and Characterization of Secreted and Membrane Bound Forms. Cell (1989) 59:335–48. doi: 10.1016/0092-8674(89)90295-X

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Goodwin RG, Friend D, Ziegler SF, Jerzy R, Falk BA, Gimpel S, et al. Cloning of the Human and Murine Interleukin-7 Receptors: Demonstration of a Soluble Form and Homology to a New Receptor Superfamily. Cell (1990) 60:941–51. doi: 10.1016/0092-8674(90)90342-C

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Fernandez-Botran R. Soluble Cytokine Receptors: Their Role in Immunoregulation. FASEB J (1991) 5:2567–74. doi: 10.1096/fasebj.5.11.1868981

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Levine SJ. Mechanisms of Soluble Cytokine Receptor Generation. J Immunol (2004) 173:5343–8. doi: 10.4049/jimmunol.173.9.5343

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Montgomery RI, Warner MS, Lum BJ, Spear PG. Herpes Simplex Virus-1 Entry Into Cells Mediated by a Novel Member of the TNF/NGF Receptor Family. Cell (1996) 87:427–36. doi: 10.1016/S0092-8674(00)81363-X

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Rodriguez-Barbosa JI, Schneider P, Weigert A, Lee KM, Kim TJ, Perez-Simon JA, et al. HVEM, a Cosignaling Molecular Switch, and Its Interactions With BTLA, CD160 and LIGHT. Cell Mol Immunol (2019) 16:679–82. doi: 10.1038/s41423-019-0241-1

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Zhu Y, Yao S, Augustine MM, Xu H, Wang J, Sun J, et al. Neuron-Specific SALM5 Limits Inflammation in the CNS via Its Interaction With HVEM. Sci Adv (2016) 2:e1500637. doi: 10.1126/sciadv.1500637

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Riva A, Palma E, Devshi D, Corrigall D, Adams H, Heaton N, et al. Soluble TIM3 and Its Ligands Galectin-9 and CEACAM1 Are in Disequilibrium During Alcohol-Related Liver Disease and Promote Impairment of Anti-Bacterial Immunity. Front Physiol (2021) 12:632502. doi: 10.3389/fphys.2021.632502

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Khan M, Zhao Z, Arooj S, Fu Y, Liao G. Soluble PD-1: Predictive, Prognostic, and Therapeutic Value for Cancer Immunotherapy. Front Immunol (2020) 11:587460. doi: 10.3389/fimmu.2020.587460

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Ng KW, Attig J, Young GR, Ottina E, Papamichos SI, Kotsianidis I, et al. Soluble PD-L1 Generated by Endogenous Retroelement Exaptation Is a Receptor Antagonist. Elife (2019) 8. doi: 10.7554/eLife.50256

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the Tumor Immune Microenvironment (TIME) for Effective Therapy. Nat Med (2018) 24:541–50. doi: 10.1038/s41591-018-0014-x

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Dong MP, Enomoto M, Thuy LTT, Hai H, Hieu VN, Hoang DV, et al. Clinical Significance of Circulating Soluble Immune Checkpoint Proteins in Sorafenib-Treated Patients With Advanced Hepatocellular Carcinoma. Sci Rep (2020) 10:3392. doi: 10.1038/s41598-020-60440-5

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Cho I, Lee H, Yoon SE, Ryu KJ, Ko YH, Kim WS, et al. Serum Levels of Soluble Programmed Death-Ligand 1 (sPD-L1) in Patients With Primary Central Nervous System Diffuse Large B-Cell Lymphoma. BMC Cancer (2020) 20:120. doi: 10.1186/s12885-020-6612-2

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Li N, Jilisihan B, Wang W, Tang Y, Keyoumu S. Soluble LAG3 Acts as a Potential Prognostic Marker of Gastric Cancer and Its Positive Correlation With CD8+T Cell Frequency and Secretion of IL-12 and INF-Gamma in Peripheral Blood. Cancer biomark (2018) 23:341–51. doi: 10.3233/CBM-181278

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Li F, Jilisihan B, Wang W, Tang Y, Keyoumu S. Highly Elevated Soluble Tim-3 Levels Correlate With Increased Hepatocellular Carcinoma Risk and Poor Survival of Hepatocellular Carcinoma Patients in Chronic Hepatitis B Virus Infection. Cancer Manag Res (2018) 10:941–51. doi: 10.2147/CMAR.S162478

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Simone R, Tenca C, Fais F, Luciani M, De Rossi G, Pesce G, et al. A Soluble Form of CTLA-4 Is Present in Paediatric Patients With Acute Lymphoblastic Leukaemia and Correlates With CD1d+ Expression. PloS One (2012) 7:e44654. doi: 10.1371/journal.pone.0044654

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Roselli M, Mineo TC, Basili S, Martini F, Mariotti S, Aloe S, et al. Soluble CD40 Ligand Plasma Levels in Lung Cancer. Clin Cancer Res (2004) 10:610–4. doi: 10.1158/1078-0432.CCR-0348-03

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Caggiari L, Guidoboni M, Vaccher E, Barzan L, Franchin G, Gloghini A, et al. High Serum Levels of Soluble CD40-L in Patients With Undifferentiated Nasopharyngeal Carcinoma: Pathogenic and Clinical Relevance. Infect Agent Cancer (2007) 2:5. doi: 10.1186/1750-9378-2-5

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Henn V, Slupsky JR, Grafe M, Anagnostopoulos I, Forster R, Muller-Berghaus G, et al. CD40 Ligand on Activated Platelets Triggers an Inflammatory Reaction of Endothelial Cells. Nature (1998) 391:591–4. doi: 10.1038/35393

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Osada J, Rusak M, Kamocki Z, Dabrowska MI, Kedra B. Platelet Activation in Patients With Advanced Gastric Cancer. Neoplasma (2010) 57:145–50. doi: 10.4149/neo_2010_02_145

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Mizoguchi H, O'Shea JJ, Longo DL, Loeffler CM, McVicar DW, Ochoa AC. Alterations in Signal Transduction Molecules in T Lymphocytes From Tumor-Bearing Mice. Science (1992) 258:1795–8. doi: 10.1126/science.1465616

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Huang J, Jochems C, Talaie T, Anderson A, Jales A, Tsang KY, et al. Elevated Serum Soluble CD40 Ligand in Cancer Patients May Play an Immunosuppressive Role. Blood (2012) 120:3030–8. doi: 10.1182/blood-2012-05-427799

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Nielsen C, Ohm-Laursen L, Barington T, Husby S, Lillevang ST. Alternative Splice Variants of the Human PD-1 Gene. Cell Immunol (2005) 235:109–16. doi: 10.1016/j.cellimm.2005.07.007

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Elhag OA, Hu XJ, Wen-Ying Z, Li X, Yuan YZ, Deng LF, et al. Reconstructed Adeno-Associated Virus With the Extracellular Domain of Murine PD-1 Induces Antitumor Immunity. Asian Pac J Cancer Prev (2012) 13:4031–6. doi: 10.7314/APJCP.2012.13.8.4031

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Tan Z, Chiu MS, Yan CW, Man K, Chen Z. Eliminating Mesothelioma by AAV-Vectored, PD1-Based Vaccination in the Tumor Microenvironment. Mol Ther Oncolytics (2021) 20:373–86. doi: 10.1016/j.omto.2021.01.010

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Chakrabarti R, Kapse B, Mukherjee G. Soluble Immune Checkpoint Molecules: Serum Markers for Cancer Diagnosis and Prognosis. Cancer Rep (Hoboken) (2019) 2:e1160. doi: 10.1002/cnr2.1160

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Fritz JM, Lenardo MJ. Development of Immune Checkpoint Therapy for Cancer. J Exp Med (2019) 216:1244–54. doi: 10.1084/jem.20182395

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Kong Y, Wang Y, Wu X, Han J, Li G, Hua M, et al. Storm of Soluble Immune Checkpoints Associated With Disease Severity of COVID-19. Signal Transduct Target Ther (2020) 5:192. doi: 10.1038/s41392-020-00308-2

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Avendano-Ortiz J, Lozano-Rodriguez R, Martin-Quiros A, Terron V, Maroun-Eid C, Montalban-Hernandez K, et al. The Immune Checkpoints Storm in COVID-19: Role as Severity Markers at Emergency Department Admission. Clin Transl Med (2021) 11:e573. doi: 10.1002/ctm2.573

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, et al. Clinical Course and Outcomes of Critically Ill Patients With SARS-CoV-2 Pneumonia in Wuhan, China: A Single-Centered, Retrospective, Observational Study. Lancet Respir Med (2020) 8:475–81. doi: 10.1016/S2213-2600(20)30079-5

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical Predictors of Mortality Due to COVID-19 Based on an Analysis of Data of 150 Patients From Wuhan, China. Intensive Care Med (2020) 46:846–8. doi: 10.1007/s00134-020-05991-x

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical Features of Patients Infected With 2019 Novel Coronavirus in Wuhan, China. Lancet (2020) 395:497–506. doi: 10.1016/S0140-6736(20)30183-5

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Giamarellos-Bourboulis EJ, Netea MG, Rovina N, Akinosoglou K, Antoniadou A, Antonakos N, et al. Complex Immune Dysregulation in COVID-19 Patients With Severe Respiratory Failure. Cell Host Microbe (2020) 27:992–1000 e1003. doi: 10.1016/j.chom.2020.04.009

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Tan L, Wang Q, Zhang D, Ding J, Huang Q, Tang YQ, et al. Lymphopenia Predicts Disease Severity of COVID-19: A Descriptive and Predictive Study. Signal Transduct Target Ther (2020) 5:33. doi: 10.1038/s41392-020-0148-4

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, et al. Clinical and Immunological Features of Severe and Moderate Coronavirus Disease 2019. J Clin Invest (2020) 130:2620–9. doi: 10.1172/JCI137244

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Huang I, Pranata R. Lymphopenia in Severe Coronavirus Disease-2019 (COVID-19): Systematic Review and Meta-Analysis. J Intensive Care (2020) 8:36. doi: 10.1186/s40560-020-00453-4

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Mathew D, Giles JR, Baxter AE, Oldridge DA, Greenplate AR, Wu JE, et al. Deep Immune Profiling of COVID-19 Patients Reveals Distinct Immunotypes With Therapeutic Implications. Science (2020) 369.

PubMed Abstract | Google Scholar

90. Urra JM, Cabrera CM, Porras L, Rodenas I. Selective CD8 Cell Reduction by SARS-CoV-2 Is Associated With a Worse Prognosis and Systemic Inflammation in COVID-19 Patients. Clin Immunol (2020) 217:108486. doi: 10.1016/j.clim.2020.108486

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Tavakolpour S, Rakhshandehroo T, Wei EX, Rashidian M. Lymphopenia During the COVID-19 Infection: What It Shows and What Can Be Learned. Immunol Lett (2020) 225:31–2. doi: 10.1016/j.imlet.2020.06.013

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Reusch N, De Domenico E, Bonaguro L, Schulte-Schrepping J, Bassler K, Schultze JL, et al. Neutrophils in COVID-19. Front Immunol (2021) 12:652470. doi: 10.3389/fimmu.2021.652470

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, et al. Reduction and Functional Exhaustion of T Cells in Patients With Coronavirus Disease 2019 (COVID-19). Front Immunol (2020) 11:827. doi: 10.3389/fimmu.2020.00827

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Wan S, Yi Q, Fan S, Lv J, Zhang X, Guo L, et al. Relationships Among Lymphocyte Subsets, Cytokines, and the Pulmonary Inflammation Index in Coronavirus (COVID-19) Infected Patients. Br J Haematol (2020) 189:428–37. doi: 10.1111/bjh.16659

CrossRef Full Text | Google Scholar

95. Vabret N, Britton GJ, Gruber C, Hegde S, Kim J, Kuksin M, et al. Immunology of COVID-19: Current State of the Science. Immunity (2020) 52:910–41. doi: 10.1016/j.immuni.2020.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Pontelli MC, Castro IA, Martins RB, Veras FP, Serra L, Nascimento DC, et al. Infection of Human Lymphomononuclear Cells by SARS-CoV-2. bioRxiv (2020). doi: 10.1101/2020.07.28.225912

CrossRef Full Text | Google Scholar

97. Chen Z, John Wherry E. T Cell Responses in Patients With COVID-19. Nat Rev Immunol (2020) 20:529–36. doi: 10.1038/s41577-020-0402-6

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Mudd PA, Crawford JC, Turner JS, Souquette A, Reynolds D, Bender D, et al. Distinct Inflammatory Profiles Distinguish COVID-19 From Influenza With Limited Contributions From Cytokine Storm. Sci Adv (2020) 6. doi: 10.1126/sciadv.abe3024

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Scherger S, Henao-Martinez A, Franco-Paredes C, Shapiro L. Rethinking Interleukin-6 Blockade for Treatment of COVID-19. Med Hypotheses (2020) 144:110053. doi: 10.1016/j.mehy.2020.110053

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Kox M, Waalders NJB, Kooistra EJ, Gerretsen J, Pickkers P. Cytokine Levels in Critically Ill Patients With COVID-19 and Other Conditions. JAMA (2020). doi: 10.1001/jama.2020.17052

CrossRef Full Text | Google Scholar

101. Syed F, Li W, Relich RF, Russell PM, Zhang S, Zimmerman MK, et al. Excessive Matrix Metalloproteinase-1 and Hyperactivation of Endothelial Cells Occurred in COVID-19 Patients and Were Associated With the Severity of COVID-19. J Infect Dis (2021). doi: 10.1101/2021.01.19.21250115

CrossRef Full Text | Google Scholar

102. Sinha P, Matthay MA, Calfee CS. Is a "Cytokine Storm" Relevant to COVID-19? JAMA Intern Med (2020) 180:1152–4. doi: 10.1001/jamainternmed.2020.3313

PubMed Abstract | CrossRef Full Text | Google Scholar

103. de Candia P, Prattichizzo F, Garavelli S, Matarese G. T Cells: Warriors of SARS-CoV-2 Infection. Trends Immunol (2021) 42:18–30. doi: 10.1016/j.it.2020.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Tian S, Xiong Y, Liu H, Niu L, Guo J, Liao M, et al. Pathological Study of the 2019 Novel Coronavirus Disease (COVID-19) Through Postmortem Core Biopsies. Mod Pathol (2020) 33:1007–14. doi: 10.1038/s41379-020-0536-x

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Liao M, Liu Y, Yuan J, Wen Y, Xu G, Zhao J, et al. Single-Cell Landscape of Bronchoalveolar Immune Cells in Patients With COVID-19. Nat Med (2020) 26:842–4. doi: 10.1038/s41591-020-0901-9

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Wichmann D, Sperhake JP, Lutgehetmann M, Steurer S, Edler C, Heinemann A, et al. Autopsy Findings and Venous Thromboembolism in Patients With COVID-19: A Prospective Cohort Study. Ann Intern Med (2020) 173:268–77. doi: 10.7326/M20-2003

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Chua RL, Lukassen S, Trump S, Hennig BP, Wendisch D, Pott F, et al. COVID-19 Severity Correlates With Airway Epithelium-Immune Cell Interactions Identified by Single-Cell Analysis. Nat Biotechnol (2020) 38:970–9. doi: 10.1038/s41587-020-0602-4

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Papagno L, Spina CA, Marchant A, Salio M, Rufer N, Little S, et al. Immune Activation and CD8+ T-Cell Differentiation Towards Senescence in HIV-1 Infection. PloS Biol (2004) 2:E20. doi: 10.1371/journal.pbio.0020020

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Kahan SM, Wherry EJ, Zajac AJ. T Cell Exhaustion During Persistent Viral Infections. Virology (2015) 479-480:180–93. doi: 10.1016/j.virol.2014.12.033

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Saeidi A, Zandi K, Cheok YY, Saeidi H, Wong WF, Lee CYQ, et al. T-Cell Exhaustion in Chronic Infections: Reversing the State of Exhaustion and Reinvigorating Optimal Protective Immune Responses. Front Immunol (2018) 9:2569. doi: 10.3389/fimmu.2018.02569

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Fenwick C, Joo V, Jacquier P, Noto A, Banga R, Perreau M, et al. T-Cell Exhaustion in HIV Infection. Immunol Rev (2019) 292:149–63. doi: 10.1111/imr.12823

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Mylvaganam G, Yanez AG, Maus M, Walker BD. Toward T Cell-Mediated Control or Elimination of HIV Reservoirs: Lessons From Cancer Immunology. Front Immunol (2019) 10:2109. doi: 10.3389/fimmu.2019.02109

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Avendano-Ortiz J, Rubio-Garrido M, Lozano-Rodriguez R, Del Romero J, Rodriguez C, Moreno S, et al. Soluble PD-L1: A Potential Immune Marker for HIV-1 Infection and Virological Failure. Medicine (Baltimore) (2020) 99:e20065. doi: 10.1097/MD.0000000000020065

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Ara A, Ahmed KA, Xiang J. Multiple Effects of CD40-CD40L Axis in Immunity Against Infection and Cancer. Immunotargets Ther (2018) 7:55–61. doi: 10.2147/ITT.S163614

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Elgueta R, Benson MJ, de Vries VC, Wasiuk A, Guo Y, Noelle RJ. Molecular Mechanism and Function of CD40/CD40L Engagement in the Immune System. Immunol Rev (2009) 229:152–72. doi: 10.1111/j.1600-065X.2009.00782.x

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Daub S, Lutgens E, Munzel T, Daiber A. CD40/CD40L and Related Signaling Pathways in Cardiovascular Health and Disease-The Pros and Cons for Cardioprotection. Int J Mol Sci (2020) 21(22). doi: 10.3390/ijms21228533

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Kornbluth RS. The Emerging Role of CD40 Ligand in HIV Infection. J Leukoc Biol (2000) 68:373–82.

PubMed Abstract | Google Scholar

118. Chougnet C. Role of CD40 Ligand Dysregulation in HIV-Associated Dysfunction of Antigen-Presenting Cells. J Leukoc Biol (2003) 74:702–9. doi: 10.1189/jlb.0403171

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Garcia-Bates TM, Palma ML, Shen C, Gambotto A, Macatangay BJC, Ferris RL, et al. Contrasting Roles of the PD-1 Signaling Pathway in Dendritic Cell-Mediated Induction and Regulation of HIV-1-Specific Effector T Cell Functions. J Virol (2019) 93(5). doi: 10.1128/JVI.02035-18

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Vonderheide RH. CD40 Agonist Antibodies in Cancer Immunotherapy. Annu Rev Med (2020) 71:47–58. doi: 10.1146/annurev-med-062518-045435

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Peters AL, Stunz LL, Bishop GA. CD40 and Autoimmunity: The Dark Side of a Great Activator. Semin Immunol (2009) 21:293–300. doi: 10.1016/j.smim.2009.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Karnell JL, Rieder SA, Ettinger R, Kolbeck R. Targeting the CD40-CD40L Pathway in Autoimmune Diseases: Humoral Immunity and Beyond. Adv Drug Deliv Rev (2019) 141:92–103. doi: 10.1016/j.addr.2018.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Aarts S, Seijkens TTP, van Dorst KJF, Dijkstra CD, Kooij G, Lutgens E. The CD40-CD40L Dyad in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis. Front Immunol (2017) 8:1791. doi: 10.3389/fimmu.2017.01791

PubMed Abstract | CrossRef Full Text | Google Scholar

124. del Rio ML, Lucas CL, Buhler L, Rayat G, Rodriguez-Barbosa JI. HVEM/LIGHT/BTLA/CD160 Cosignaling Pathways as Targets for Immune Regulation. J Leukoc Biol (2010) 87:223–35. doi: 10.1189/jlb.0809590

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Cai G, Freeman GJ. The CD160, BTLA, LIGHT/HVEM Pathway: A Bidirectional Switch Regulating T-Cell Activation. Immunol Rev (2009) 229:244–58. doi: 10.1111/j.1600-065X.2009.00783.x

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Cai G, Anumanthan A, Brown JA, Greenfield EA, Zhu B, Freeman GJ. CD160 Inhibits Activation of Human CD4+ T Cells Through Interaction With Herpesvirus Entry Mediator. Nat Immunol (2008) 9:176–85. doi: 10.1038/ni1554

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Gowing LR, Ali RL, Allsop S, Marsden J, Turf EE, West R, et al. Global Statistics on Addictive Behaviours: 2014 Status Report. Addiction (2015) 110:904–19. doi: 10.1111/add.12899

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Gilmore W, Chikritzhs T, Stockwell T, Jernigan D, Naimi T, Gilmore I. Alcohol: Taking a Population Perspective. Nat Rev Gastroenterol Hepatol (2016) 13:426–34. doi: 10.1038/nrgastro.2016.70

PubMed Abstract | CrossRef Full Text | Google Scholar

129. O'Shea RS, Dasarathy S, McCullough AJ. Practice Guideline Committee of the American Association for the Study of Liver, D. & Practice Parameters Committee of the American College of, G. Alcoholic Liver Disease. Hepatology (2010) 51:307–28. doi: 10.1002/hep.23258

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Mitchell J, Dunlop A. Alcohol Consumption: Monitoring, Regulation and Impact on Public Health. Public Health Res Pract (2016) 26(4). doi: 10.17061/phrp2641640.

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Wigg S, Stafford LD. Health Warnings on Alcoholic Beverages: Perceptions of the Health Risks and Intentions Towards Alcohol Consumption. PloS One (2016) 11:e0153027. doi: 10.1371/journal.pone.0153027

PubMed Abstract | CrossRef Full Text | Google Scholar

132. WHO. HIV Data and Statistics. In: HIV/AIDS Fact Sheets (2021).

Google Scholar

133. Justice A, Sullivan L, Fiellin D, Veterans Aging Cohort Study Project T. HIV/AIDS, Comorbidity, and Alcohol: Can We Make a Difference? Alcohol Res Health (2010) 33:258–66.

PubMed Abstract | Google Scholar

134. Meyerhoff DJ. Effects of Alcohol and HIV Infection on the Central Nervous System. Alcohol Res Health (2001) 25(4):288–98.

PubMed Abstract | Google Scholar

135. Monnig MA. Immune Activation and Neuroinflammation in Alcohol Use and HIV Infection: Evidence for Shared Mechanisms. Am J Drug Alcohol Abuse (2017) 43:7–23. doi: 10.1080/00952990.2016.1211667

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Elamin E, Masclee A, Dekker J, Jonkers D. Ethanol Disrupts Intestinal Epithelial Tight Junction Integrity Through Intracellular Calcium-Mediated Rho/ROCK Activation. Am J Physiol Gastrointest Liver Physiol (2014) 306:G677–685. doi: 10.1152/ajpgi.00236.2013

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Bode C, Bode JC. Activation of the Innate Immune System and Alcoholic Liver Disease: Effects of Ethanol Per Se or Enhanced Intestinal Translocation of Bacterial Toxins Induced by Ethanol? Alcohol Clin Exp Res (2005) 29:166S–71S. doi: 10.1097/01.alc.0000189280.19073.28

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Gao B, Bataller R. Alcoholic Liver Disease: Pathogenesis and New Therapeutic Targets. Gastroenterology (2011) 141:1572–85. doi: 10.1053/j.gastro.2011.09.002

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Pasala S, Barr T, Messaoudi I. Impact of Alcohol Abuse on the Adaptive Immune System. Alcohol Res (2015) 37(2):185–97.

PubMed Abstract | Google Scholar

140. Gao B, Seki E, Brenner DA, Friedman S, Cohen JI, Nagy L, et al. Innate Immunity in Alcoholic Liver Disease. Am J Physiol Gastrointest Liver Physiol (2011) 300:G516–525. doi: 10.1152/ajpgi.00537.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Miller AM, Horiguchi N, Jeong WI, Radaeva S, Gao B. Molecular Mechanisms of Alcoholic Liver Disease: Innate Immunity and Cytokines. Alcohol Clin Exp Res (2011) 35:787–93. doi: 10.1111/j.1530-0277.2010.01399.x

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Li W, Amet T, Xing Y, Yang D, Liangpunsakul S, Puri P, et al. Alcohol Abstinence Ameliorates the Dysregulated Immune Profiles in Patients With Alcoholic Hepatitis: A Prospective Observational Study. Hepatology (2017) 66:575–90. doi: 10.1002/hep.29242

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial Translocation Is a Cause of Systemic Immune Activation in Chronic HIV Infection. Nat Med (2006) 12:1365–71. doi: 10.1038/nm1511

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Dinh DM, Volpe GE, Duffalo C, Bhalchandra S, Tai AK, Kane AV, et al. Intestinal Microbiota, Microbial Translocation, and Systemic Inflammation in Chronic HIV Infection. J Infect Dis (2015) 211:19–27. doi: 10.1093/infdis/jiu409

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Liangpunsakul S, Puri P, Shah VH, Kamath P, Sanyal A, Urban T, et al. Effects of Age, Sex, Body Weight, and Quantity of Alcohol Consumption on Occurrence and Severity of Alcoholic Hepatitis. Clin Gastroenterol Hepatol (2016) 14:1831–8.e1833. doi: 10.1016/j.cgh.2016.05.041

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Puri P, Liangpunsakul S, Christensen JE, Shah VH, Kamath PS, Gores GJ, et al. The Circulating Microbiome Signature and Inferred Functional Metagenomics in Alcoholic Hepatitis. Hepatology (2018) 67:1284–302. doi: 10.1002/hep.29623

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Liangpunsakul S, Toh E, Ross RA, Heathers LE, Chandler K, Oshodi A, et al. Quantity of Alcohol Drinking Positively Correlates With Serum Levels of Endotoxin and Markers of Monocyte Activation. Sci Rep (2017) 7:4462. doi: 10.1038/s41598-017-04669-7

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Li W, Lin EL, Liangpunsakul S, Lan J, Chalasani S, Rane S, et al. Alcohol Abstinence Does Not Fully Reverse Abnormalities of Mucosal-Associated Invariant T Cells in the Blood of Patients With Alcoholic Hepatitis. Clin Transl Gastroenterol (2019) 10:e00052. doi: 10.14309/ctg.0000000000000052

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Wong EB, Ndung'u T, Kasprowicz VO. The Role of Mucosal-Associated Invariant T Cells in Infectious Diseases. Immunology (2017) 150:45–54. doi: 10.1111/imm.12673

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Kurioka A, Walker LJ, Klenerman P, Willberg CB. MAIT Cells: New Guardians of the Liver. Clin Transl Immunol (2016) 5:e98. doi: 10.1038/cti.2016.51

CrossRef Full Text | Google Scholar

151. Vivarelli S, Falzone L, Torino F, Scandurra G, Russo G, Bordonaro R, et al. Immune-Checkpoint Inhibitors From Cancer to COVID19: A Promising Avenue for the Treatment of Patients With COVID19 (Review). Int J Oncol (2021) 58:145–57. doi: 10.3892/ijo.2020.5159

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Vivarelli S, Falzone L, Grillo CM, Scandurra G, Torino F, Libra M. Cancer Management During COVID-19 Pandemic: Is Immune Checkpoint Inhibitors-Based Immunotherapy Harmful or Beneficial? Cancers (Basel) (2020) 12(8). doi: 10.3390/cancers12082237

CrossRef Full Text | Google Scholar

153. Gambichler T, Reuther J, Scheel CH, Becker JC. On the Use of Immune Checkpoint Inhibitors in Patients With Viral Infections Including COVID-19. J Immunother Cancer (2020) 8(2). doi: 10.1136/jitc-2020-001145

CrossRef Full Text | Google Scholar

154. Cook MR, Kim C. Safety and Efficacy of Immune Checkpoint Inhibitor Therapy in Patients With HIV Infection and Advanced-Stage Cancer: A Systematic Review. JAMA Oncol (2019) 5:1049–54. doi: 10.1001/jamaoncol.2018.6737

PubMed Abstract | CrossRef Full Text | Google Scholar

155. Pezeshki PS, Rezaei N. Immune Checkpoint Inhibition in COVID-19: Risks and Benefits. Expert Opin Biol Ther (2021) 21:1173–9. doi: 10.1080/14712598.2021.1887131

PubMed Abstract | CrossRef Full Text | Google Scholar

156. Ozen G, Pedro S, Schumacher R, Simon TA, Michaud K. Safety of Abatacept Compared With Other Biologic and Conventional Synthetic Disease-Modifying Antirheumatic Drugs in Patients With Rheumatoid Arthritis: Data From an Observational Study. Arthritis Res Ther (2019) 21:141. doi: 10.1186/s13075-019-1921-z

PubMed Abstract | CrossRef Full Text | Google Scholar

157. Kuemmerle-Deschner JB, Benseler S. Abatacept in Difficult-to-Treat Juvenile Idiopathic Arthritis. Biologics (2008) 2(4):865–74. doi: 10.2147/btt.s3355.

PubMed Abstract | CrossRef Full Text | Google Scholar

158. Hara R, Umebayashi H, Takei S, Okamoto N, Iwata N, Yamasaki Y, et al. Intravenous Abatacept in Japanese Patients With Polyarticular-Course Juvenile Idiopathic Arthritis: Results From a Phase III Open-Label Study. Pediatr Rheumatol Online J (2019) 17:17. doi: 10.1186/s12969-019-0319-4

PubMed Abstract | CrossRef Full Text | Google Scholar

159. Noisette A, Hochberg MC. Abatacept for the Treatment of Adults With Psoriatic Arthritis: Patient Selection and Perspectives. Psoriasis (Auckl) (2018) 8:31–9. doi: 10.2147/PTT.S146076

PubMed Abstract | CrossRef Full Text | Google Scholar

160. Buchbinder EI, Desai A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am J Clin Oncol (2016) 39:98–106. doi: 10.1097/COC.0000000000000239

PubMed Abstract | CrossRef Full Text | Google Scholar

161. Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 Coreceptor Expression and Signal Transduction. Immunol Rev (2009) 229:12–26. doi: 10.1111/j.1600-065X.2009.00770.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: immune checkpoint, soluble immune checkpoint, COVID-19, HIV, heavy alcohol user, alcohol-associated liver disease (ALD), inflammation

Citation: Li W, Syed F, Yu R, Yang J, Xia Y, Relich RF, Russell PM, Zhang S, Khalili M, Huang L, Kacena MA, Zheng X and Yu Q (2022) Soluble Immune Checkpoints Are Dysregulated in COVID-19 and Heavy Alcohol Users With HIV Infection. Front. Immunol. 13:833310. doi: 10.3389/fimmu.2022.833310

Received: 11 December 2021; Accepted: 28 January 2022;
Published: 23 February 2022.

Edited by:

Antonio Riva, Foundation for Liver Research, United Kingdom

Reviewed by:

Luca Falzone, G. Pascale National Cancer Institute Foundation (IRCCS), Italy
Robert Siggins, Louisiana State University, United States

Copyright © 2022 Li, Syed, Yu, Yang, Xia, Relich, Russell, Zhang, Khalili, Huang, Kacena, Zheng and Yu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Qigui Yu, andyu@iupui.edu

Present address: Ying Xia, Department of Chemistry and Molecular Biology, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.