Next Article in Journal
Pneumocystis jirovecii Pneumonia Prophylaxis for Cancer Patients during Chemotherapy
Next Article in Special Issue
Survey of the Transcription Factor Responses of Mouse Lung Alveolar Macrophages to Pneumocystis murina
Previous Article in Journal
In vitro–in vivo Validation of Stimulatory Effect of Oat Ingredients on Lactobacilli
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pneumocystis Pneumonia: Immunity, Vaccines, and Treatments

by
Aaron D. Gingerich
1,2,
Karen A. Norris
1,2 and
Jarrod J. Mousa
1,2,*
1
Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
2
Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
*
Author to whom correspondence should be addressed.
Pathogens 2021, 10(2), 236; https://doi.org/10.3390/pathogens10020236
Submission received: 22 January 2021 / Revised: 11 February 2021 / Accepted: 16 February 2021 / Published: 19 February 2021

Abstract

:
For individuals who are immunocompromised, the opportunistic fungal pathogen Pneumocystis jirovecii is capable of causing life-threatening pneumonia as the causative agent of Pneumocystis pneumonia (PCP). PCP remains an acquired immunodeficiency disease (AIDS)-defining illness in the era of antiretroviral therapy. In addition, a rise in non-human immunodeficiency virus (HIV)-associated PCP has been observed due to increased usage of immunosuppressive and immunomodulating therapies. With the persistence of HIV-related PCP cases and associated morbidity and mortality, as well as difficult to diagnose non-HIV-related PCP cases, an improvement over current treatment and prevention standards is warranted. Current therapeutic strategies have primarily focused on the administration of trimethoprim-sulfamethoxazole, which is effective at disease prevention. However, current treatments are inadequate for treatment of PCP and prevention of PCP-related death, as evidenced by consistently high mortality rates for those hospitalized with PCP. There are no vaccines in clinical trials for the prevention of PCP, and significant obstacles exist that have slowed development, including host range specificity, and the inability to culture Pneumocystis spp. in vitro. In this review, we overview the immune response to Pneumocystis spp., and discuss current progress on novel vaccines and therapies currently in the preclinical and clinical pipeline.

1. Introduction

Pneumocystis pneumonia was first documented in the early 1900s, yet was first identified in humans following the conclusion of World War II in malnourished infants who had severe pneumonia [1]. Pneumocystis spp. were initially classified as protozoan parasites; however, it was determined in 1988 via RNA sequencing that Pneumocystis is an ascomycetous fungus [2]. Since 1988, five different species have been fully identified, including Pneumocystis carinii and Pneumocystis wakefieldiae in rats, Pneumocystis murina in mice, Pneumocystis oryctolagi in rabbits, and Pneumocystis jirovecii (formerly carinii) in humans [3]. P. jirovecii is the etiological agent of Pneumocystis pneumonia (PCP), a life-threatening pneumonia occurring primarily in immunocompromised individuals. PCP accounts for an estimated 10,000 hospitalizations [4] in the United States and >400,000 cases worldwide each year [5]. A sharp increase in cases of PCP was observed during the early years of the human immunodeficiency virus (HIV)/acquired immunodeficiency disease (AIDS) epidemic, as PCP was the most common defining opportunistic infection of individuals with HIV [6]. As antiretroviral therapy became more effective and widespread, the incidence of PCP decreased substantially; however, PCP remains a significant cause of mortality and morbidity for those with HIV [7]. The majority of new HIV infections, and thus HIV-related PCP cases, occur in developing countries, and an increase in the rate of PCP was observed from 2002 to 2010 [8].
PCP also has increased incidence in cancer patients undergoing immunosuppressive regimens [9], organ transplant recipients [10], and those on immunomodulating drugs [11,12,13,14]. Currently, percentages of PCP cases in HIV+ vs. HIV− populations are approximately 40% and 60%, respectively [14]. HIV-negative patients who develop PCP are at higher risk of death than those who are HIV+, with a mortality rate above 30% [15]. PCP can also progress more rapidly in HIV− patients and remains difficult to diagnose. HIV− patients typically have a more functional immune system than those who are HIV+, and thus, more severe PCP-associated inflammation [16]. In addition, PCP is correlated with the development and severity of chronic obstructive pulmonary disease (COPD) in patients with and without HIV infection [17,18,19,20,21,22,23,24].
One of the most challenging aspects in treating PCP is the presence of cholesterol in the cell membrane, which cannot be targeted by common antifungals such as amphotericin B and azoles [25]. Based on the high mortality rate with both HIV and non-HIV associated PCP [11,26], the need for novel therapeutic options and an effective vaccine remains. Currently, several drugs, vaccines, and even antibody therapies are being pursued. The host range specificity of Pneumocystis spp. has made it difficult to translate research in animal models into humans, and the inability to culture the organism in a lab setting has further impeded progress. In this review, we will summarize the immune response to PCP in the context of promising prevention and treatment options.

2. Development of PCP and the Immune Response

Pneumocystis spp. have a penchant to infect the lungs of immunocompromised individuals. Development of PCP has been hypothesized to be a reactivation event of a latent infection within a host when the immune system becomes compromised [27]. However, current thought is that person-to-person transmission is the likely culprit of new infections [28,29,30,31,32]. Based on microscopic examinations, Pneumocystis organisms attach to type I alveolar epithelium, which allows the fungus to transition from its small trophic form to the larger cystic form [33]. Pneumocystis is primarily an alveolar pathogen, but in very rare cases of severely immunocompromised individuals, it can disseminate from the lungs to other regions, including the central nervous system, bone marrow, lymph nodes, eyes, gastrointestinal tract, thyroid, liver, spleen, and kidney [34,35,36,37]. Adherence of Pneumocystis to alveoli is not the singular cause of diffuse alveolar damage, but rather, it is the host inflammatory response that can cause significant lung injury and impaired gas exchange, potentially leading to hypoxia and respiratory failure [38]. In the section below, we briefly summarize several aspects of the immune response to Pneumocystis infection based on immune cell type.

2.1. CD4+ T-Cells

CD4+ T-cells play a key role in the immune response to PCP by leading the recruitment of macrophages and monocytes to the lungs. In patients with HIV/AIDS, lower CD4+ T cell levels are correlated with a higher incidence of developing PCP [39]. Furthermore, depletion of CD4+ T cells in mice leads to the development of PCP [40], and in SCID mice, which lack a T and B lymphocyte response, spontaneous Pneumocystis infection is observed within three weeks [41]. SCID mice are able to salvage their ability to clear Pneumocystis infection effectively when reconstituted with CD4+ T cells from immunocompetent mice [42]. In addition, several studies have shown that proper CD4+ T-cell signaling is necessary for control of Pneumocystis infection. For example, mice deficient in both CD2 and CD28 (CD2/CD8) exhibit susceptibility to spontaneous P. carinii infection. In contrast, mice deficient in only CD2 or CD28 are not susceptible to spontaneous infection [43], although CD28-deficient mice can be infected when directly inoculated. However, CD28-deficient and CD2/CD28 deficient mice are able to clear infection, although such clearance is delayed. CD28-deficient mice have increased IFN-γ [43], which is important for limiting Pneumocystis-associated inflammation, but not in the resolution of infection [44,45,46,47]. In contrast, TNF-α is critical for clearance of PCP [44]. Supporting these observations, CD2/CD28-deficiency leads to reduced anti-Pneumocystis antibody titers. In addition, CD28-deficient mice have an increase in IL-10 levels [43], which is notable since IL-10-deficient mice have improved clearance of Pneumocystis infection [48]. Furthermore, the absence of IL-10 is correlated to improved Pneumocystis clearance and an increase in the presence of CD4+ T cells, CD8+ T-cells, and neutrophils in the lungs [48].

2.2. CD8+ T Cells

The role of CD8+ T cells in the clearance of Pneumocystis infection remains uncertain as evidence both for and against the role of this cell type has been demonstrated. CD4+ T cell-depleted mice modified to upregulate IFN-γ have improved clearance of Pneumocystis organisms, as well as an increase in recruited CD8+ T cells and NK cells to the lung [49]. CD8+ T cells derived from Pneumocystis-infected mice and stimulated with IFN-γ improve organism killing by macrophages [50]. Furthermore, administration of human IL-7 to CD4+ T cell-depleted mice results in an increase in IFN-γ-positive CD8+ T cell recruitment to the lungs, which is correlated to Pneumocystis clearance [51]. In a study assessing secondary immune responses to Pneumocystis, mice depleted of CD8+ T cells between first and second Pneumocystis challenge have higher fungal burden compared to CD4+ T cell-depleted mice [52], as previous work has shown that CD4+ T cells are not required for memory recall responses against Pneumocystis [53]. In contrast to these studies supporting the involvement of CD8+ T cells in Pneumocystis clearance, no difference in the organism lung burden is observed when both CD8+ and CD4+ T cells are depleted compared to CD4+ T cell-depleted mice, and reconstitution with sensitized CD8+ T cells leads to enhanced pulmonary injury [54].

2.3. Macrophages

Macrophages play a key role in clearing Pneumocystis infection, as alveolar macrophage depletion leads to an increase in Pneumocystis burden in the lungs of rats [55]. Alveolar macrophages are the primary responders to the organism, and once stimulated by IFN-γ from CD4+ T-cells, they are responsible for phagocytosis and killing of Pneumocystis organisms. In addition, alternatively activated macrophages are potent effector cells that can lead to robust organism killing [56,57]. However, Pneumocystis can subvert the effectiveness of macrophages by causing them to undergo apoptosis [58], as one study demonstrated that infected animal bronchoalveolar lavage (BAL) samples have high levels of polyamines, which induce apoptosis of alveolar macrophages [59]. Much like CD4+ T-cells, macrophages play a fundamental role in the immune response to Pneumocystis via proinflammatory cytokines and chemokines; however, while these are effective at organism elimination, the response can come at a cost of pulmonary injury [55].

2.4. Neutrophils

Neutrophils are predominantly involved in inflammation rather than clearance of Pneumocystis organisms. Elevated neutrophil counts are correlated to a decrease in pulmonary function in HIV+ patients with PCP [60,61]. Mouse studies analyzing neutrophil function in knockouts of the NADPH oxidase component gp91(phox), a double knockout of gp91(phox) and inducible nitric oxide synthase, and a knockout of CXCR2, as well as antibody-induced neutrophil depletion have been examined [62]. In each of these models, no difference in Pneumocystis organism burdens, respiratory rates, arterial oxygen partial pressures, and intra-alveolar albumin concentrations were observed. Overall, neutrophils appear to be a marker of lung damage, but are not involved in causing lung damage in the mouse model.

2.5. B-Cells

B-cells play an essential role in the immune response to PCP. Using mixed bone marrow chimeric mice that lack expression of MHC class II on B-cells, rendering them unable to present antigen to CD4+ T-cells, an increase in susceptibility to infection is observed [63]. The increased susceptibility is likely due to diminished antibody production against Pneumocystis, and a lack of B-cell antigen presentation [63]. Additionally, patients with hyper-IgM syndrome have increased susceptibility to PCP [64,65]. This phenomenon has been observed in the clinic, whereby a 5-month-old infant tested positive for PCP with no known previous medical conditions, and it was later determined that the infant suffered from hyper-IgM syndrome [66]. In addition, another study examining a cohort of patients with a CD40 deficiency that resulted in hyper-IgM syndrome had a PCP incidence rate of 27% [64].

2.6. Natural Killer Cells

The role of natural killer (NK) cells in Pneumocystis clearance is poorly understood. Low numbers of NK cells or impaired NK cell function in HIV+ individuals have been suggested as a correlate of increased incidence of PCP [67,68,69,70]. Depletion of CD4+ T cells in mice leads to a decrease in NK cells in lung tissue, and the NK cells remaining have decreased upregulation of NKp46 and production of IFN-γ [71]. An increase in fungal killing occurs when NK cells are combined with CD4+ T cells compared to each cell type alone [71]. Furthermore, adoptive transfer of memory CD4+ T cells is required for NK cell upregulation of activation maker NK group 2D, and production of IFN-γ, granzyme B, and perforin during Pneumocystis infection in mice [71].

3. Drug Treatments

3.1. Current Drugs in Use

A variety of drugs are currently available for the prevention and treatment of Pneumocystis infection (Figure 1). Trimethoprim-sulfamethoxazole (TMP-SMX) is the primary option for the prevention of PCP, and for treatment of mild to severe cases of PCP [72,73,74]. However, treatment side effects and increasing drug resistance are a major concern [75]. Adverse reactions are relatively frequent and can result in a number of conditions, including rashes, fever, gastrointestinal complications, cytopenia, marrow suppression, hyperkalemia, hepatoxicity, interstitial nephritis, aseptic meningitis, anaphylaxis, renal insufficiency, and pancreatitis [76]. Trimethoprim is a dihydrofolate reductase inhibitor, and sulfamethoxazole is a dihydropteroate synthetase inhibitor, and these drugs have a synergistic effect [77,78]. Trimethoprim binds to dihydrofolate reductase and inhibits the reduction of dihydrofolic acid to tetrahydrofolic acid, while sulfamethoxazole inhibits dihydropteroate synthase [79]. Both drugs interfere with the thymidine synthase pathway, which ultimately inhibits DNA synthesis of the organism [79]. Mutations to the dihydrofolate reductase (DHFR) and dihydropteroate synthase (DHPS) have been reported in P. jirovecii, possibly leading to treatment failure [80]. For patients with a sulfa-allergy, trimethoprim-dapsone can be used; however, in patients who have had serious reactions to TMP-SMX, dapsone is avoided as it can lead to fatal idiosyncratic dapsone-hypersensitivity syndrome [76]. Clindamycin-primaquine has been suggested for patients who have severe disease who are unable to take TMP-SMX [81]. Atovaquone can also be used in the treatment of mild cases of PCP, but is less effective than TMP-SMX in clinical trials [82]. Furthermore, drug resistance to atovaquone has begun to occur, as mutations in the cytochrome b gene have led to prophylactic failure of atovaquone [83].
While most drug treatments focus on targeting the organism, the use of sulfasalazine takes a different approach. Sulfasalazine is a disease-modifying antirheumatic drug used therapeutically in patients with rheumatoid arthritis [84]. While the mechanism of action of sulfasalazine has not been fully elucidated, its immunomodulatory and anti-inflammatory effects have been effective in treating PCP in mice by attenuating pulmonary inflammation and increasing fungal clearance [56]. The fungal clearance is driven by the ability of sulfasalazine to alter the T-cell response and drive macrophages to an M2 phenotype, leading to killing of Pneumocystis organisms [56,57,85]. In addition to these drugs, the anti-inflammatory properties of corticosteroids have improved the clinical outcomes of patients with moderate to severe PCP by limiting pulmonary damage [86]. However, such steroids must be used with care as non-HIV patients on corticosteroid regiments have an increased risk of developing PCP [87]. This observation has been supported in cancer patients receiving corticosteroids [88].

3.2. Drugs in Development

In addition to these current treatments already in use, newer drugs have been examined in preclinical mouse models and are now in clinical trials. Caspofungin, an echinocandin and competitive inhibitor of for 1,3-β-D-glucan synthesis, which is a major component of the fungal cell wall, is effective at reducing the cystic form of Pneumocystis and enhancing survival of mice [89]. However, the drug does not eliminate the trophic reservoir, which leads to cyst reformation following treatment withdrawal. Numerous reports of caspofungin alone and in combination with TMP-SMX have been reported, with most indicating efficacy in treatment [90]. Based on these data, two clinical trials are ongoing to determine the efficacy of caspofungin in non-HIV patients with PCP (NCT02603575, NCT03978559). Another echinocandin, rezafungin [91], has shown prophylactic efficacy against Pneumocystis in an immunosuppressed mouse model by a reduction in nuclei and asci counts [92]. In addition, rezafungin was shown to limit Pneumocystis reactivation following cessation of therapy in the same mouse model [93]. A current phase III clinical trial is ongoing to determine the efficacy of rezafungin in allogenic blood and marrow transplant patients (NCT04368559).
Due to the immunocompromised state of individuals who develop PCP, utilization of passive immunization with antibodies is a viable potential therapeutic option. The first attempt at determining the effectiveness of antibody treatment demonstrated that the mAb M5E12 conferred partial protection against PCP in drug-induced immunosuppressive mouse and ferret animal models [94]. Mice born to Pneumocystis-immunized dams have higher antibody titers compared to mice born from naïve dams, and this increase in antibody levels is correlated with enhanced Pneumocystis clearance in challenged mice [95]. The use of hyperimmune serum in both mouse and rat models was shown to be protective in two independent studies reinforcing the effectiveness of antibodies against PCP [96,97]. A promising avenue of treatment is the use of combination therapy to treat PCP. It was recently shown that a cocktail of antibody and sulfasalazine leads to a dramatic improvement in the severity of PCP in a mouse model compared to single therapy methods; however, the mechanism by which this occurs needs further investigation [98].

4. Vaccine Development against PCP

4.1. Vaccination with Whole Organisms

Some of the first attempts at developing vaccines for Pneumocystis utilized inactivated whole organisms [53,99,100]. Since Pneumocystis is an opportunistic pathogen, the animal models used must undergo immunosuppression following vaccination for efficient infection. Pneumocystis has a very specific host range, demonstrated by a study immunizing mice with either mouse- or ferret-derived Pneumocystis. Mice immunized with mouse-derived Pneumocystis demonstrated a robust protective effect upon intratracheal infection with Pneumocystis, yet mice immunized with ferret-derived Pneumocystis were not protected [101]. Another study utilizing non-viable organisms along with cholera toxin fraction B administered intranasally was found to be protective and generated robust IgA and IgG antibody titers [99]. These results are promising for utilizing whole organisms in vaccinations; however, a major hurdle in this approach is the inability to grow Pneumocystis in vitro, limiting this approach from being effective on a large scale. Overall, while immunization with whole Pneumocystis organisms has been shown to be quite effective in animal models [53,99,100], the growth limitation has led to the exploration of subunit-based vaccines [102].

4.2. Subunit Vaccines

Below, we summarize the protein candidates that have been examined as vaccine antigens in Pneumocystis subunit vaccines (Table 1) [99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128].
gpA (Msg): The major surface glycoprotein (Msg or glycoprotein A (gpA)) is an important B and T cell target following immunization with whole organisms [103,104]. However, gpA has enormous amino acid diversity, which is likely a result of evolutionary immune evasion [104,105]. Mice vaccinated with gpA demonstrate a robust specific antibody response, but are not protected from infection [102]. An additional study in the rat model showed that animals vaccinated with gpA saw results with variable levels of protection [106]. It was further demonstrated that immunization with gpA leads to cross-reactive antibody responses to gpA variants, but not cross-reactive T-cells, suggesting variation in gpA by Pneumocystis is used for T-cell escape [107]. Humans generate antibodies to gpA, and this protein is predominantly used in Pneumocystis serological studies, as antibodies to gpA increase following PCP [108,109,110,111]. Due to the genetic variation in gpA, the protein is not actively being pursued as a vaccine candidate.
p55: A 55 kDa protein antigen, termed p55, resides inside the Pneumocystis cell wall, and both natural and recombinant p55 proteins can stimulate a protective immune response [112,113]. p55 protein from rat-derived Pneumocystis antigen has >80% sequence homology with nonhuman primate (NHP) and human variants of the protein [113,114]. A p55-based multi-epitope antigen incorporating 12 B-cell epitopes elicits both cell and humoral immune responses in rats, and vaccination with this antigen followed by dexamethasone immunosuppression to induce Pneumocystis infection results in reduced fungal burden [114]. In another study utilizing methylprednisone for rat immunosuppression, immunization with p55 led to a decrease in organism burden, improved histology scores, and reduced inflammation, yet only partial protection against PCP was observed [112]. In a T-cell independent approach, an adenovirus 5 vector encoding murine CD40L was used to modify dendritic cells to express CD40L, and these cells were then pulsed with Pneumocystis organisms ex vivo. Mice immunosuppressed with an anti-CD4 monoclonal antibody were then administered the modified dendritic cells. This immunization lowered the organism burden in challenged mice, and antibodies generated as a result of vaccination induced opsonic killing via a macrophage-mediated mechanism [115]. The antibody response generated by this vaccination led to p55-specific IgG [115].
A12 (Pca1): The A12 antigen was discovered using a cross-reactive mouse monoclonal antibody, 4F11, that is cross-reactive against multiple Pneumocystis spp. [116]. The 142-AA A12 polypeptide elicits a protective response as a vaccine antigen, and facilitates a decrease in Pneumocystis organism burden and lung inflammation in a mouse model [117]. Following identification of the full-length protein containing the A12 sequence, the entire molecule was renamed Pneumocystis cross-reactive antigen 1 (Pca1). Active immunization with the N-terminal half of Pca1 is protective in a mouse model. Interestingly, although sharing limited sequence identity with homologs with other Pneumocystis spp., Pca1 is able to induce antibodies that are able to cross react with P. jirovecii [118].
Kexin: Kexin is a serine protease that shares sequence homology with kexin proteins from other fungal pathogens, and was initially identified by probing a cDNA library with monoclonal antibodies [119]. Intranasally administered anti-kexin monoclonal antibodies are protective in an SCID mouse model of PCP [120]. Several studies have investigated the utility of kexin as a candidate vaccine. DNA vaccination of anti-CD4 antibody depleted mice with full length kexin and mouse CD40L (to induce a CD4-independent response) results in anti-pneumocystis antibody titers in CD4+ T cell-depleted mice [115]. This immunization lowers the organism burden in challenged mice, and antibodies generated as a result of vaccination induce opsonic killing via a macrophage-mediated mechanism [115]. A subunit recombinant protein vaccine consisting of a 90 amino acid fragment of Kexin (KEX1) has also been examined as a potential candidate. In a NHP model of HIV and PCP, rhesus macaques vaccinated with KEX1 followed by simian-human immunodeficiency virus (SHIV) infection and Pneumocystis challenge by exposure to Pneumocystis-colonized animals are protected from developing PCP up to 36 weeks after immunosuppression [121]. In a separate model using methylprednisone-immunosuppressed rhesus macaques as a model for drug-induced immunosuppression, KEX1 vaccination prior to immunosuppression produces a robust and enduring antibody response that continues throughout immunosuppression, and boosting with KEX1 during the immunosuppression phase induces recall of memory responses against KEX1 [122].
KEX1 has also been examined as a serological marker for the risk of developing PCP. Most adults have been exposed to P. jirovecii, based on studies demonstrating children are seropositive by the age of four [123,124,125]. In a retrospective study of AIDS patients comparing those who developed PCP and those who developed a non-PCP AIDS-defining illness, those who developed PCP were more likely to have low KEX1 IgG prior to PCP diagnosis [126]. It was concluded that antibody levels to KEX1 are predictive of HIV patients developing PCP, as those with elevated antibody levels specific to KEX1 had a reduced risk of infection [126]. This observation was further confirmed in a nonhuman primate model of HIV and Pneumocystis coinfection, whereby higher baseline plasma levels of KEX1-IgG were correlated with decreased incidence of Pneumocystis colonization [127].
SPD1: Utilizing a chemical labeling technique to identify surface proteins as potential vaccine candidates led to the antigen SPD1 being identified in P. murina [128]. SPD1 has high sequence identity with homologs from P. jirovecii. Mice immunized with SPD1 develop IgG antibody responses and an increase in memory B-cells following Pneumocystis challenge. In addition, serum from rhesus macaques exposed to Pneumocystis reacts to SPD1. Vaccination of mice with N-terminal and C-terminal fragments of SPD1 shows no effect in CD4+ T cell-depleted mice at 1-week post vaccination. However, a decrease in organism burden is observed at 4 weeks for the C-terminal fragment and at 6 weeks for both fragments. These data suggest SPD1 is expressed on the surface of Pneumocystis organisms later during the infection time course, and/or there is a lag time in the development of functional antibodies to SPD1.

5. Conclusions

PCP remains a significant public health concern. At the advent of the HIV epidemic, the incidence of PCP increased rapidly, and as treatments to control HIV infection gained widespread use and high efficacy, the incidence of HIV patients diagnosed with PCP decreased. However, PCP remains an AIDS-defining illness with a consistently high mortality rate [129]. In addition, cases of non-HIV-associated PCP are now more prominent due to increased usage of immunosuppressive and immunomodulating drugs [13,130,131]. While drugs to prevent the development of PCP, primarily TMP-SMX, have been generally successful, major concerns remain in those patients who are diagnosed with PCP. Clinical trials for additional antifungal drugs are currently ongoing, and antibodies have shown promise in preclinical models, especially when combined with other drugs. An ideal course of action would be development of a vaccine for prevention of PCP, which could be selectively administered to those at high risk of HIV infection, and those individuals who have planned procedures or drug regimens that will lead to immunosuppressive states. Several vaccine candidates have shown promise in preclinical models, primarily in mouse models, and the KEX1 candidate has proven effective in immunosuppressed NHPs in both HIV-associated and drug-induced immunosuppression. Since Pneumocystis species have restricted host specificity, those vaccine candidates that have high conservation in P. jirovecii would presumably have the highest chance of success, yet it is unclear if such vaccine candidates are able to protect against P. jirovecii. However, the inability to grow Pneumocystis in vitro and the lack of a genetically tractable system limits the ability to examine if vaccine candidates that have shown efficacy in preclinical models have the same function in P. jirovecii. Recent advancements in single-cell sequencing and immune profiling may assist these efforts by identifying genes expressed at each stage of the Pneumocystis life cycle, and identifying additional monoclonal antibody candidates and T cell epitopes for therapy design and vaccine development. Overall, vaccine and therapeutic development has made promising advances in recent years, although significant hurdles remain to advance preclinical studies to human clinical trials.

Funding

This work was supported by National Institutes of Health grants 1K01OD026569 (J.J.M.) and 1R01AI148365 (K.A.N.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

J.J.M. declares no conflicts of interest. K.A.N. has a financial interest in a company and is co-inventor of inventions related to some research reported in the enclosed paper. There is an approved plan in place with the University of Georgia for managing any potential conflict.

References

  1. Chabe, M.; Aliouat-Denis, C.M.; Delhaes, L.; El Moukhtar, A.; Viscogliosi, E.; Dei-Cas, E. Pneumocystis: From a doubtful unique entity to a group of highly diversified fungal species. FEMS Yeast Res. 2011, 11, 2–17. [Google Scholar] [CrossRef] [Green Version]
  2. Edman, J.C.; Kovacs, J.A.; Masur, H.; Santi, D.V.; Elwood, H.J.; Sogin, M.L. Ribosomal RNA sequence shows Pneumocystis carinii to be a member of the fungi. Nature 1988, 334, 519–522. [Google Scholar] [CrossRef]
  3. Aliouat-Denis, C.M.; Chabe, M.; Demanche, C.; Aliouat el, M.; Viscogliosi, E.; Guillot, J.; Delhaes, L.; Dei-Cas, E. Pneumocystis species, co-evolution and pathogenic power. Infect. Genet. Evol. 2008, 8, 708–726. [Google Scholar] [CrossRef] [PubMed]
  4. Benedict, K.; Jackson, B.R.; Chiller, T.; Beer, K.D. Estimation of Direct Healthcare Costs of Fungal Diseases in the United States. Clin. Infect. Dis. 2019, 68, 1791–1797. [Google Scholar] [CrossRef] [Green Version]
  5. Brown, G.D.; Denning, D.W.; Gow, N.A.R.; Levitz, S.M.; Netea, M.G.; White, T.C. Hidden Killers: Human Fungal Infections. Sci. Transl. Med. 2012, 4, 165rv113. [Google Scholar] [CrossRef] [Green Version]
  6. Morris, A.; Lundgren, J.D.; Masur, H.; Walzer, P.D.; Hanson, D.L.; Frederick, T.; Huang, L.; Beard, C.B.; Kaplan, J.E. Current epidemiology of Pneumocystis pneumonia. Emerg. Infect. Dis. 2004, 10, 1713–1720. [Google Scholar] [CrossRef]
  7. Miller, R.F.; Huang, L.; Walzer, P.D. Pneumocystis pneumonia associated with human immunodeficiency virus. Clin. Chest Med. 2013, 34, 229–241. [Google Scholar] [CrossRef] [Green Version]
  8. De Armas Rodríguez, Y.; Wissmann, G.; Müller, A.L.; Pederiva, M.A.; Brum, M.C.; Brackmann, R.L.; Capó de Paz, V.; Calderón, E.J. Pneumocystis jirovecii pneumonia in developing countries. Parasite 2011, 18, 219–228. [Google Scholar] [CrossRef] [Green Version]
  9. Varthalitis, I.; Aoun, M.; Daneau, D.; Meunier, F. Pneumocystis carinii pneumonia in patients with cancer. An increasing incidence. Cancer 1993, 71, 481–485. [Google Scholar] [CrossRef]
  10. Gordon, S.M.; LaRosa, S.P.; Kalmadi; Arroliga, A.C.; Avery, R.K.; Truesdell-LaRosa; Longworth, D.L. Should Prophylaxis for Pneumocystis carinii Pneumonia in Solid Organ Transplant Recipients Ever Be Discontinued? Clin. Infect. Dis. 1999, 28, 240–246. [Google Scholar] [CrossRef] [Green Version]
  11. Liu, Y.; Su, L.; Jiang, S.-J.; Qu, H. Risk factors for mortality from pneumocystis carinii pneumonia (PCP) in non-HIV patients: A meta-analysis. Oncotarget 2017, 8, 59729–59739. [Google Scholar] [CrossRef] [Green Version]
  12. Arend, S.M.; Kroon, F.P.; van’t Wout, J.W. Pneumocystis carinii Pneumonia in Patients without AIDS, 1980 through 1993: An Analysis of 78 Cases. Arch. Intern. Med. 1995, 155, 2436–2441. [Google Scholar] [CrossRef]
  13. Burghi, G.; Biard, L.; Roux, A.; Valade, S.; Robert-Gangneux, F.; Hamane, S.; Maubon, D.; Debourgogne, A.; Le Gal, S.; Dalle, F.; et al. Characteristics and outcome according to underlying disease in non-AIDS patients with acute respiratory failure due to Pneumocystis pneumonia. Eur. J. Clin. Microbiol. Infect. Dis. 2021. [Google Scholar] [CrossRef] [PubMed]
  14. Roux, A.; Canet, E.; Valade, S.; Gangneux-Robert, F.; Hamane, S.; Lafabrie, A.; Maubon, D.; Debourgogne, A.; Le Gal, S.; Dalle, F.; et al. Pneumocystis jirovecii pneumonia in patients with or without AIDS, France. Emerg. Infect. Dis. 2014, 20, 1490–1497. [Google Scholar] [CrossRef]
  15. Roblot, F.; Godet, C.; Le Moal, G.; Garo, B.; Faouzi Souala, M.; Dary, M.; De Gentile, L.; Gandji, J.A.; Guimard, Y.; Lacroix, C.; et al. Analysis of underlying diseases and prognosis factors associated with Pneumocystis carinii pneumonia in immunocompromised HIV-negative patients. Eur. J. Clin. Microbiol. Infect. Dis. 2002, 21, 523–531. [Google Scholar] [CrossRef]
  16. Sokulska, M.; Kicia, M.; Wesołowska, M.; Hendrich, A.B. Pneumocystis jirovecii—From a commensal to pathogen: Clinical and diagnostic review. Parasitol. Res. 2015, 114, 3577–3585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Norris, K.A.; Morris, A. Pneumocystis infection and the pathogenesis of chronic obstructive pulmonary disease. Immunol. Res. 2011, 50, 175–180. [Google Scholar] [CrossRef] [Green Version]
  18. George, M.P.; Kannass, M.; Huang, L.; Sciurba, F.C.; Morris, A. Respiratory symptoms and airway obstruction in HIV-infected subjects in the HAART era. PLoS ONE 2009, 4, e6328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Gingo, M.R.; George, M.P.; Kessinger, C.J.; Lucht, L.; Rissler, B.; Weinman, R.; Slivka, W.A.; McMahon, D.K.; Wenzel, S.E.; Sciurba, F.C.; et al. Pulmonary function abnormalities in HIV-infected patients during the current antiretroviral therapy era. Am. J. Respir. Crit. Care Med. 2010, 182, 790–796. [Google Scholar] [CrossRef] [Green Version]
  20. Morris, A.; Alexander, T.; Radhi, S.; Lucht, L.; Sciurba, F.C.; Kolls, J.K.; Srivastava, R.; Steele, C.; Norris, K.A. Airway obstruction is increased in pneumocystis-colonized human immunodeficiency virus-infected outpatients. J. Clin. Microbiol. 2009, 47, 3773–3776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Norris, K.A.; Morris, A.; Patil, S.; Fernandes, E. Pneumocystis colonization, airway inflammation, and pulmonary function decline in acquired immunodeficiency syndrome. Immunol. Res. 2006, 36, 175–187. [Google Scholar] [CrossRef]
  22. Morris, A.M.; Huang, L.; Bacchetti, P.; Turner, J.; Hopewell, P.C.; Wallace, J.M.; Kvale, P.A.; Rosen, M.J.; Glassroth, J.; Reichman, L.B.; et al. Permanent declines in pulmonary function following pneumonia in human immunodeficiency virus-infected persons. The Pulmonary Complications of HIV Infection Study Group. Am. J. Respir. Crit. Care Med. 2000, 162, 612–616. [Google Scholar] [CrossRef]
  23. Morris, A.; Sciurba, F.C.; Lebedeva, I.P.; Githaiga, A.; Elliott, W.M.; Hogg, J.C.; Huang, L.; Norris, K.A. Association of chronic obstructive pulmonary disease severity and Pneumocystis colonization. Am. J. Respir. Crit. Care Med. 2004, 170, 408–413. [Google Scholar] [CrossRef] [PubMed]
  24. Morris, A.; Netravali, M.; Kling, H.M.; Shipley, T.; Ross, T.; Sciurba, F.C.; Norris, K.A. Relationship of pneumocystis antibody response to severity of chronic obstructive pulmonary disease. Clin. Infect. Dis. 2008, 47, e64–e68. [Google Scholar] [CrossRef] [Green Version]
  25. Kaneshiro, E.S.; Ellis, J.E.; Jayasimhulu, K.; Beach, D.H. Evidence for the presence of “metabolic sterols” in Pneumocystis: Identification and initial characterization of Pneumocystis carinii sterols. J. Eukaryot. Microbiol. 1994, 41, 78–85. [Google Scholar] [CrossRef] [PubMed]
  26. Fei, M.W.; Kim, E.J.; Sant, C.A.; Jarlsberg, L.G.; Davis, J.L.; Swartzman, A.; Huang, L. Predicting mortality from HIV-associated Pneumocystis pneumonia at illness presentation: An observational cohort study. Thorax 2009, 64, 1070–1076. [Google Scholar] [CrossRef] [Green Version]
  27. Pifer, L.L.; Hughes, W.T.; Stagno, S.; Woods, D. Pneumocystis carinii infection: Evidence for high prevalence in normal and immunosuppressed children. Pediatrics 1978, 61, 35–41. [Google Scholar] [PubMed]
  28. Morris, A.; Beard, C.B.; Huang, L. Update on the epidemiology and transmission of Pneumocystis carinii. Microbes. Infect. 2002, 4, 95–103. [Google Scholar] [CrossRef]
  29. Beck, J.M. Pneumocystis carinii and geographic clustering: Evidence for transmission of infection. Am. J. Respir. Crit. Care Med. 2000, 162, 1605–1606. [Google Scholar] [CrossRef]
  30. Chen, W.; Gigliotti, F.; Harmsen, A.G. Latency is not an inevitable outcome of infection with Pneumocystis carinii. Infect. Immun. 1993, 61, 5406–5409. [Google Scholar] [CrossRef] [Green Version]
  31. Vargas, S.L.; Hughes, W.T.; Wakefield, A.E.; Oz, H.S. Limited Persistence in and Subsequent Elimination of Pneumocystis carinii from the Lungs after P. carinii Pneumonia. J. Infect. Dis. 1995, 172, 506–510. [Google Scholar] [CrossRef] [PubMed]
  32. Beard, C.B.; Carter, J.L.; Keely, S.P.; Huang, L.; Pieniazek, N.J.; Moura, I.N.; Roberts, J.M.; Hightower, A.W.; Bens, M.S.; Freeman, A.R.; et al. Genetic variation in Pneumocystis carinii isolates from different geographic regions: Implications for transmission. Emerg. Infect. Dis. 2000, 6, 265–272. [Google Scholar] [CrossRef]
  33. Dumoulin, A.; Mazars, E.; Seguy, N.; Gargallo-Viola, D.; Vargas, S.; Cailliez, J.C.; Aliouat, E.M.; Wakefield, A.E.; Dei-Cas, E. Transmission of Pneumocystis carinii disease from immunocompetent contacts of infected hosts to susceptible hosts. Eur. J. Clin. Microbiol. Infect. Dis. 2000, 19, 671–678. [Google Scholar] [CrossRef]
  34. Witt, K.; Nielsen, T.N.; Junge, J. Dissemination of Pneumocystis carinii in Patients with AIDS. Scand. J. Infect. Dis. 1991, 23, 691–695. [Google Scholar] [CrossRef]
  35. Karam, M.B.; Mosadegh, L. Extra-pulmonary Pneumocystis jiroveci infection: A case report. Braz. J. Infect. Dis. 2014, 18, 681–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Kim, B.; Kim, J.; Paik, S.S.; Pai, H. Atypical Presentation of Pneumocystis jirovecii Infection in HIV Infected Patients: Three Different Manifestations. J. Korean Med. Sci. 2018, 33, e115. [Google Scholar] [CrossRef]
  37. Sharma, N.; Ahlawat, R.S.; Singh, H.; Sharma, C.; Anuradha, S. Pneumocystis jirovecii infection of bilateral adrenal glands in an immunocompetent adult: A case report. J. R. Coll. Physicians Edinb. 2019, 49, 222–224. [Google Scholar] [CrossRef] [PubMed]
  38. Limper, A.H.; Offord, K.P.; Smith, T.F.; Martin, W.J., 2nd. Pneumocystis carinii pneumonia. Differences in lung parasite number and inflammation in patients with and without AIDS. Am. Rev. Respir. Dis. 1989, 140, 1204–1209. [Google Scholar] [CrossRef]
  39. Phair, J.; Munoz, A.; Detels, R.; Kaslow, R.; Rinaldo, C.; Saah, A. The risk of Pneumocystis carinii pneumonia among men infected with human immunodeficiency virus type 1. Multicenter AIDS Cohort Study Group. N. Engl. J. Med. 1990, 322, 161–165. [Google Scholar] [CrossRef]
  40. Shellito, J.; Suzara, V.V.; Blumenfeld, W.; Beck, J.M.; Steger, H.J.; Ermak, T.H. A new model of Pneumocystis carinii infection in mice selectively depleted of helper T lymphocytes. J. Clin. Investig. 1990, 85, 1686–1693. [Google Scholar] [CrossRef] [Green Version]
  41. Roths, J.B.; Marshall, J.D.; Allen, R.D.; Carlson, G.A.; Sidman, C.L. Spontaneous Pneumocystis carinii pneumonia in immunodeficient mutant scid mice. Natural history and pathobiology. Am. J. Pathol. 1990, 136, 1173–1186. [Google Scholar]
  42. Harmsen, A.G.; Stankiewicz, M. Requirement for CD4+ cells in resistance to Pneumocystis carinii pneumonia in mice. J. Exp. Med. 1990, 172, 937–945. [Google Scholar] [CrossRef] [Green Version]
  43. Beck, J.M.; Blackmon, M.B.; Rose, C.M.; Kimzey, S.L.; Preston, A.M.; Green, J.M. T cell costimulatory molecule function determines susceptibility to infection with Pneumocystis carinii in mice. J. Immunol. 2003, 171, 1969–1977. [Google Scholar] [CrossRef]
  44. Chen, W.; Havell, E.A.; Harmsen, A.G. Importance of endogenous tumor necrosis factor alpha and gamma interferon in host resistance against Pneumocystis carinii infection. Infect. Immun. 1992, 60, 1279–1284. [Google Scholar] [CrossRef] [Green Version]
  45. Meissner, N.N.; Swain, S.; Tighe, M.; Harmsen, A.; Harmsen, A. Role of Type I IFNs in Pulmonary Complications of Pneumocystis murina Infection. J. Immunol. 2005, 174, 5462–5471. [Google Scholar] [CrossRef]
  46. Meissner, N.; Swain, S.; McInnerney, K.; Han, S.; Harmsen, A.G. Type-I IFN Signaling Suppresses an Excessive IFN-γ Response and Thus Prevents Lung Damage and Chronic Inflammation During Pneumocystis (PC) Clearance in CD4 T Cell-Competent Mice. Am. J. Pathol. 2010, 176, 2806–2818. [Google Scholar] [CrossRef] [PubMed]
  47. Garvy, B.A.; Ezekowitz, R.A.; Harmsen, A.G. Role of gamma interferon in the host immune and inflammatory responses to Pneumocystis carinii infection. Infect. Immun. 1997, 65, 373–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Qureshi, M.H.; Harmsen, A.G.; Garvy, B.A. IL-10 Modulates Host Responses and Lung Damage Induced by Pneumocystis carinii Infection. J. Immunol. 2003, 170, 1002–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Kolls, J.K.; Habetz, S.; Shean, M.K.; Vazquez, C.; Brown, J.A.; Lei, D.; Schwarzenberger, P.; Ye, P.; Nelson, S.; Summer, W.R.; et al. IFN-gamma and CD8+ T cells restore host defenses against Pneumocystis carinii in mice depleted of CD4+ T cells. J. Immunol. 1999, 162, 2890–2894. [Google Scholar]
  50. Mc Allister, F.; Steele, C.; Zheng, M.; Young, E.; Shellito, J.E.; Marrero, L.; Kolls, J.K. T Cytotoxic-1 CD8+ T Cells Are Effector Cells against Pneumocystis in Mice. J. Immunol. 2004, 172, 1132–1138. [Google Scholar] [CrossRef]
  51. Ruan, S.; Samuelson, D.R.; Assouline, B.; Morre, M.; Shellito, J.E. Treatment with Interleukin-7 Restores Host Defense against Pneumocystis in CD4+ T-Lymphocyte-Depleted Mice. Infect. Immun. 2016, 84, 108–119. [Google Scholar] [CrossRef] [Green Version]
  52. de la Rua, N.M.; Samuelson, D.R.; Charles, T.P.; Welsh, D.A.; Shellito, J.E. CD4+ T-Cell-Independent Secondary Immune Responses to Pneumocystis Pneumonia. Front. Immunol. 2016, 7. [Google Scholar] [CrossRef] [Green Version]
  53. Harmsen, A.G.; Chen, W.; Gigliotti, F. Active immunity to Pneumocystis carinii reinfection in T-cell-depleted mice. Infect. Immun. 1995, 63, 2391–2395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Gigliotti, F.; Crow, E.L.; Bhagwat, S.P.; Wright, T.W. Sensitized CD8+ T cells fail to control organism burden but accelerate the onset of lung injury during Pneumocystis carinii pneumonia. Infect. Immun. 2006, 74, 6310–6316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Limper, A.H.; Hoyte, J.S.; Standing, J.E. The role of alveolar macrophages in Pneumocystis carinii degradation and clearance from the lung. J. Clin. Investig. 1997, 99, 2110–2117. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, J.; Gigliotti, F.; Bhagwat, S.P.; George, T.C.; Wright, T.W. Immune modulation with sulfasalazine attenuates immunopathogenesis but enhances macrophage-mediated fungal clearance during Pneumocystis pneumonia. PLoS Pathog. 2010, 6, e1001058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Nelson, M.P.; Christmann, B.S.; Werner, J.L.; Metz, A.E.; Trevor, J.L.; Lowell, C.A.; Steele, C. IL-33 and M2a alveolar macrophages promote lung defense against the atypical fungal pathogen Pneumocystis murina. J. Immunol. 2011, 186, 2372–2381. [Google Scholar] [CrossRef] [Green Version]
  58. Deckman, J.M.; Kurkjian, C.J.; McGillis, J.P.; Cory, T.J.; Birket, S.E.; Schutzman, L.M.; Murphy, B.S.; Garvy, B.A.; Feola, D.J. Pneumocystis infection alters the activation state of pulmonary macrophages. Immunobiology 2017, 222, 188–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Lasbury, M.E.; Merali, S.; Durant, P.J.; Tschang, D.; Ray, C.A.; Lee, C.H. Polyamine-mediated apoptosis of alveolar macrophages during Pneumocystis pneumonia. J. Biol. Chem. 2007, 282, 11009–11020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Jensen, B.N.; Lisse, I.M.; Gerstoft, J.; Borgeskov, S.; Skinhøj, P. Cellular profiles in bronchoalveolar lavage fluid of HIV-infected patients with pulmonary symptoms: Relation to diagnosis and prognosis. Aids 1991, 5, 527–533. [Google Scholar] [CrossRef]
  61. Sadaghdar, H.; Huang, Z.B.; Eden, E. Correlation of bronchoalveolar lavage findings to severity of Pneumocystis carinii pneumonia in AIDS. Evidence for the development of high-permeability pulmonary edema. Chest 1992, 102, 63–69. [Google Scholar] [CrossRef] [PubMed]
  62. Swain, S.D.; Wright, T.W.; Degel, P.M.; Gigliotti, F.; Harmsen, A.G. Neither neutrophils nor reactive oxygen species contribute to tissue damage during Pneumocystis pneumonia in mice. Infect. Immun. 2004, 72, 5722–5732. [Google Scholar] [CrossRef] [Green Version]
  63. Lund, F.E.; Hollifield, M.; Schuer, K.; Lines, J.L.; Randall, T.D.; Garvy, B.A. B cells are required for generation of protective effector and memory CD4 cells in response to Pneumocystis lung infection. J. Immunol. 2006, 176, 6147–6154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Al-Saud, B.K.; Al-Sum, Z.; Alassiri, H.; Al-Ghonaium, A.; Al-Muhsen, S.; Al-Dhekri, H.; Arnaout, R.; Alsmadi, O.; Borrero, E.; Abu-Staiteh, A.; et al. Clinical, immunological, and molecular characterization of hyper-IgM syndrome due to CD40 deficiency in eleven patients. J. Clin. Immunol. 2013, 33, 1325–1335. [Google Scholar] [CrossRef]
  65. Tsai, H.Y.; Yu, H.H.; Chien, Y.H.; Chu, K.H.; Lau, Y.L.; Lee, J.H.; Wang, L.C.; Chiang, B.L.; Yang, Y.H. X-linked hyper-IgM syndrome with CD40LG mutation: Two case reports and literature review in Taiwanese patients. J. Microbiol. Immunol. Infect. 2015, 48, 113–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Kim, D.; Shin, J.A.; Han, S.B.; Chung, N.G.; Jeong, D.C. Pneumocystis jirovecii pneumonia as an initial manifestation of hyper-IgM syndrome in an infant: A case report. Medicine 2019, 98, e14559. [Google Scholar] [CrossRef] [PubMed]
  67. Bonagura, V.R.; Cunningham-Rundles, S.; Edwards, B.L.; Ilowite, N.T.; Wedgwood, J.F.; Valacer, D.J. Common variable hypogammaglobulinemia, recurrent Pneumocystis carinii pneumonia on intravenous γ-globulin therapy, and natural killer deficiency. Clin. Immunol. Immunopathol. 1989, 51, 216–231. [Google Scholar] [CrossRef]
  68. Bonagura, V.R.; Cunningham-Rundles, S.L.; Schuval, S. Dysfunction of natural killer cells in human immunodeficiency virus-infected children with or without Pneumocystis carinii pneumonia. J. Pediatr. 1992, 121, 195–201. [Google Scholar] [CrossRef]
  69. Duncan, R.A.; von Reyn, C.F.; Alliegro, G.M.; Toossi, Z.; Sugar, A.M.; Levitz, S.M. Idiopathic CD4+ T-Lymphocytopenia—Four Patients with Opportunistic Infections and No Evidence of HIV Infection. N. Engl. J. Med. 1993, 328, 393–398. [Google Scholar] [CrossRef]
  70. Guzman, J.; Wang, Y.M.; Teschler, H.; Kienast, K.; Brockmeyer, N.; Costabel, U. Phenotypic analysis of bronchoalveolar lavage lymphocytes from acquired immunodeficiency patients with and without Pneumocystis carinii pneumonia. Acta Cytol. 1992, 36, 900–904. [Google Scholar]
  71. Kelly, M.N.; Zheng, M.; Ruan, S.; Kolls, J.; D’Souza, A.; Shellito, J.E. Memory CD4+ T Cells Are Required for Optimal NK Cell Effector Functions against the Opportunistic Fungal Pathogen Pneumocystis murina. J. Immunol. 2013, 190, 285–295. [Google Scholar] [CrossRef] [Green Version]
  72. Catherinot, E.; Lanternier, F.; Bougnoux, M.E.; Lecuit, M.; Couderc, L.J.; Lortholary, O. Pneumocystis jirovecii Pneumonia. Infect. Dis. Clin. N. Am. 2010, 24, 107–138. [Google Scholar] [CrossRef]
  73. Thomas, C.F.; Limper, A.H. Pneumocystis Pneumonia. N. Engl. J. Med. 2004, 350, 2487–2498. [Google Scholar] [CrossRef]
  74. Hardak, E.; Brook, O.; Yigla, M. Radiological features of Pneumocystis jirovecii Pneumonia in immunocompromised patients with and without AIDS. Lung 2010, 188, 159–163. [Google Scholar] [CrossRef]
  75. Khodavaisy, S.; Mortaz, E.; Mohammadi, F.; Aliyali, M.; Fakhim, H.; Badali, H. Pneumocystis jirovecii colonization in Chronic Obstructive Pulmonary Disease (COPD). Curr. Med. Mycol. 2015, 1, 42–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. White, P.L.; Price, J.S.; Backx, M. Therapy and Management of Pneumocystis jirovecii Infection. J. Fungi 2018, 4, 127. [Google Scholar] [CrossRef] [Green Version]
  77. Krajicek, B.J.; Thomas, C.F., Jr.; Limper, A.H. Pneumocystis pneumonia: Current concepts in pathogenesis, diagnosis, and treatment. Clin. Chest Med. 2009, 30, 265–278. [Google Scholar] [CrossRef] [PubMed]
  78. Tasaka, S. Recent Advances in the Diagnosis and Management of Pneumocystis Pneumonia. Tuberc. Respir. Dis. 2020, 83, 132–140. [Google Scholar] [CrossRef] [Green Version]
  79. Brogden, R.N.; Carmine, A.A.; Heel, R.C.; Speight, T.M.; Avery, G.S. Trimethoprim: A Review of its Antibacterial Activity, Pharmacokinetics and Therapeutic Use in Urinary Tract Infections. Drugs 1982, 23, 405–430. [Google Scholar] [CrossRef] [PubMed]
  80. Suarez, I.; Roderus, L.; van Gumpel, E.; Jung, N.; Lehmann, C.; Fatkenheuer, G.; Hartmann, P.; Plum, G.; Rybniker, J. Low prevalence of DHFR and DHPS mutations in Pneumocystis jirovecii strains obtained from a German cohort. Infection 2017, 45, 341–347. [Google Scholar] [CrossRef]
  81. Safrin, S.; Finkelstein, D.M.; Feinberg, J.; Frame, P.; Simpson, G.; Wu, A.; Cheung, T.; Soeiro, R.; Hojczyk, P.; Black, J.R. Comparison of three regimens for treatment of mild to moderate Pneumocystis carinii pneumonia in patients with AIDS. A double-blind, randomized, trial of oral trimethoprim-sulfamethoxazole, dapsone-trimethoprim, and clindamycin-primaquine. ACTG 108 Study Group. Ann. Intern. Med. 1996, 124, 792–802. [Google Scholar] [CrossRef]
  82. Martin, S.I.; Fishman, J.A. Pneumocystis pneumonia in solid organ transplantation. Am. J. Transpl. 2013, 13 (Suppl. 4), 272–279. [Google Scholar] [CrossRef]
  83. Argy, N.; Le Gal, S.; Coppee, R.; Song, Z.; Vindrios, W.; Massias, L.; Kao, W.C.; Hunte, C.; Yazdanpanah, Y.; Lucet, J.C.; et al. Pneumocystis Cytochrome b Mutants Associated With Atovaquone Prophylaxis Failure as the Cause of Pneumocystis Infection Outbreak Among Heart Transplant Recipients. Clin. Infect. Dis. 2018, 67, 913–919. [Google Scholar] [CrossRef] [PubMed]
  84. Plosker, G.L.; Croom, K.F. Sulfasalazine: A review of its use in the management of rheumatoid arthritis. Drugs 2005, 65, 1825–1849. [Google Scholar] [CrossRef]
  85. Nunokawa, T.; Yokogawa, N.; Shimada, K.; Sugii, S. Effect of sulfasalazine use on the presence of Pneumocystis organisms in the lung among patients with rheumatoid arthritis: A test-negative design case-control study with PCR tests. Mod. Rheumatol. 2019, 29, 436–440. [Google Scholar] [CrossRef]
  86. Bozzette, S.A.; Sattler, F.R.; Chiu, J.; Wu, A.W.; Gluckstein, D.; Kemper, C.; Bartok, A.; Niosi, J.; Abramson, I.; Coffman, J.; et al. A controlled trial of early adjunctive treatment with corticosteroids for Pneumocystis carinii pneumonia in the acquired immunodeficiency syndrome. California Collaborative Treatment Group. N. Engl. J. Med. 1990, 323, 1451–1457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Yale, S.H.; Limper, A.H. Pneumocystis carinii pneumonia in patients without acquired immunodeficiency syndrome: Associated illness and prior corticosteroid therapy. Mayo Clin. Proc. 1996, 71, 5–13. [Google Scholar] [CrossRef]
  88. Sepkowitz, K.A.; Brown, A.E.; Telzak, E.E.; Gottlieb, S.; Armstrong, D. Pneumocystis carinii pneumonia among patients without AIDS at a cancer hospital. JAMA 1992, 267, 832–837. [Google Scholar] [CrossRef] [PubMed]
  89. Cushion, M.T.; Linke, M.J.; Ashbaugh, A.; Sesterhenn, T.; Collins, M.S.; Lynch, K.; Brubaker, R.; Walzer, P.D. Echinocandin treatment of pneumocystis pneumonia in rodent models depletes cysts leaving trophic burdens that cannot transmit the infection. PLoS ONE 2010, 5, e8524. [Google Scholar] [CrossRef]
  90. Nevez, G.; Le Gal, S. Caspofungin and Pneumocystis Pneumonia: It Is Time To Go Ahead. Antimicrob. Agents Chemother. 2019, 63. [Google Scholar] [CrossRef] [Green Version]
  91. Zhao, Y.; Perlin, D.S. Review of the Novel Echinocandin Antifungal Rezafungin: Animal Studies and Clinical Data. J Fungi 2020, 6, 192. [Google Scholar] [CrossRef] [PubMed]
  92. Cushion, M.; Ashbaugh, A. Rezafungin Prophylactic Efficacy in a Mouse Model of Pneumocystis Pneumonia. Biol. Blood Marrow Transplant. 2019, 25, S366. [Google Scholar] [CrossRef]
  93. Cushion, M.T.; Ashbaugh, A.; Ong, V. Rezafungin Prevention of Pneumocystis Pneumonia and Pneumocystis reactivation Using Different Doses and Durations of Prophylaxis in a Mouse Model. Blood 2019, 134, 3266. [Google Scholar] [CrossRef]
  94. Gigliotti, F.; Hughes, W.T. Passive immunoprophylaxis with specific monoclonal antibody confers partial protection against Pneumocystis carinii pneumonitis in animal models. J. Clin. Investig. 1988, 81, 1666–1668. [Google Scholar] [CrossRef] [PubMed]
  95. Empey, K.M.; Hollifield, M.; Schuer, K.; Gigliotti, F.; Garvy, B.A. Passive Immunization of Neonatal Mice against Pneumocystis carinii f. sp. muris Enhances Control of Infection without Stimulating Inflammation. Infect. Immun. 2004, 72, 6211–6220. [Google Scholar] [CrossRef] [Green Version]
  96. Bartlett, M.S.; Angus, W.C.; Shaw, M.M.; Durant, P.J.; Lee, C.H.; Pascale, J.M.; Smith, J.W. Antibody to Pneumocystis carinii protects rats and mice from developing pneumonia. Clin. Diagn. Lab. Immunol. 1998, 5, 74–77. [Google Scholar] [CrossRef] [Green Version]
  97. Roths, J.B.; Sidman, C.L. Single and combined humoral and cell-mediated immunotherapy of Pneumocystis carinii pneumonia in immunodeficient scid mice. Infect. Immun. 1993, 61, 1641–1649. [Google Scholar] [CrossRef] [Green Version]
  98. Hoy, Z.; Wright, T.W.; Elliott, M.; Malone, J.; Bhagwat, S.; Wang, J.; Gigliotti, F. Combination Immunotherapy with Passive Antibody and Sulfasalazine Accelerates Fungal Clearance and Promotes the Resolution of Pneumocystis-Associated Immunopathogenesis. Infect. Immun. 2020, 88. [Google Scholar] [CrossRef] [Green Version]
  99. Pascale, J.M.; Shaw, M.M.; Durant, P.J.; Amador, A.A.; Bartlett, M.S.; Smith, J.W.; Gregory, R.L.; McLaughlin, G.L. Intranasal Immunization Confers Protection against Murine Pneumocystis carinii Lung Infection. Infect. Immun. 1999, 67, 805–809. [Google Scholar] [CrossRef] [Green Version]
  100. Garvy, B.A.; Wiley, J.A.; Gigliotti, F.; Harmsen, A.G. Protection against Pneumocystis carinii pneumonia by antibodies generated from either T helper 1 or T helper 2 responses. Infect. Immun. 1997, 65, 5052–5056. [Google Scholar] [CrossRef] [Green Version]
  101. Gigliotti, F.; Harmsen, A.G. Pneumocystis carinii host origin defines the antibody specificity and protective response induced by immunization. J. Infect. Dis. 1997, 176, 1322–1326. [Google Scholar] [CrossRef] [Green Version]
  102. Gigliotti, F.; Wiley, J.A.; Harmsen, A.G. Immunization with Pneumocystis carinii gpA Is Immunogenic but Not Protective in a Mouse Model of P. carinii Pneumonia. Infect. Immun. 1998, 66, 3179. [Google Scholar] [CrossRef] [Green Version]
  103. Gigliotti, F.; McCool, T. Glycoprotein A is the immunodominant antigen of Pneumocystis carinii in mice following immunization. Parasitol. Res. 1996, 82, 90–91. [Google Scholar] [CrossRef]
  104. Graves, D.C.; McNabb, S.J.; Ivey, M.H.; Worley, M.A. Development and characterization of monoclonal antibodies to Pneumocystis carinii. Infect. Immun. 1986, 51, 125–133. [Google Scholar] [CrossRef] [Green Version]
  105. Theus, S.A.; Linke, M.J.; Andrews, R.P.; Walzer, P.D. Proliferative and cytokine responses to a major surface glycoprotein of Pneumocystis carinii. Infect. Immun. 1993, 61, 4703–4709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Theus, S.A.; Smulian, A.G.; Steele, P.; Linke, M.J.; Walzer, P.D. Immunization with the major surface glycoprotein of Pneumocystis carinii elicits a protective response. Vaccine 1998, 16, 1149–1157. [Google Scholar] [CrossRef]
  107. Bishop, L.R.; Helman, D.; Kovacs, J.A. Discordant antibody and cellular responses to Pneumocystis major surface glycoprotein variants in mice. BMC Immunol. 2012, 13, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Daly, K.R.; Koch, J.; Levin, L.; Walzer, P.D. Enzyme-linked immunosorbent assay and serologic responses to Pneumocystis jiroveci. Emerg. Infect. Dis. 2004, 10, 848–854. [Google Scholar] [CrossRef] [PubMed]
  109. Bishop, L.R.; Kovacs, J.A. Quantitation of anti-Pneumocystis jiroveci antibodies in healthy persons and immunocompromised patients. J. Infect. Dis. 2003, 187, 1844–1848. [Google Scholar] [CrossRef]
  110. Daly, K.; Koch, J.; Respaldiza, N.; de la Horra, C.; Montes-Cano, M.A.; Medrano, F.J.; Varela, J.M.; Calderon, E.J.; Walzer, P.D. Geographical variation in serological responses to recombinant Pneumocystis jirovecii major surface glycoprotein antigens. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2009, 15, 937–942. [Google Scholar] [CrossRef] [Green Version]
  111. Walzer, P.D.; Djawe, K.; Levin, L.; Daly, K.R.; Koch, J.; Kingsley, L.; Witt, M.; Golub, E.T.; Bream, J.H.; Taiwo, B.; et al. Long-term serologic responses to the Pneumocystis jirovecii major surface glycoprotein in HIV-positive individuals with and without P. jirovecii infection. J. Infect. Dis. 2009, 199, 1335–1344. [Google Scholar] [CrossRef] [Green Version]
  112. Smulian, A.G.; Sullivan, D.W.; Theus, S.A. Immunization with recombinant Pneumocystis carinii p55 antigen provides partial protection against infection: Characterization of epitope recognition associated with immunization. Microbes. Infect. 2000, 2, 127–136. [Google Scholar] [CrossRef]
  113. Theus, S.A.; Sullivan, D.W.; Walzer, P.D.; Smulian, A.G. Cellular responses to a 55-kilodalton recombinant Pneumocystis carinii antigen. Infect. Immun. 1994, 62, 3479–3484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Fan, H.; Guo, J.Y.; Ma, S.L.; Zhang, N.; An, C.L. Synthetic p55 tandem DNA vaccine against Pneumocystis carinii in rats. Microbiol. Immunol. 2016, 60, 397–406. [Google Scholar] [CrossRef] [Green Version]
  115. Zheng, M.; Shellito, J.E.; Marrero, L.; Zhong, Q.; Julian, S.; Ye, P.; Wallace, V.; Schwarzenberger, P.; Kolls, J.K. CD4+ T cell-independent vaccination against Pneumocystis carinii in mice. J. Clin. Investig. 2001, 108, 1469–1474. [Google Scholar] [CrossRef] [PubMed]
  116. Wells, J.; Gigliotti, F.; Simpson-Haidaris, P.J.; Haidaris, C.G. Epitope Mapping of a Protective Monoclonal Antibody against Pneumocystis carinii with Shared Reactivity to Streptococcus pneumoniae Surface Antigen PspA. Infect. Immun. 2004, 72, 1548–1556. [Google Scholar] [CrossRef] [Green Version]
  117. Wells, J.; Haidaris, C.G.; Wright, T.W.; Gigliotti, F. Active Immunization against Pneumocystis carinii with a Recombinant P. carinii Antigen. Infect. Immun. 2006, 74, 2446–2448. [Google Scholar] [CrossRef] [Green Version]
  118. Tesini, B.L.; Wright, T.W.; Malone, J.E.; Haidaris, C.G.; Harber, M.; Sant, A.J.; Nayak, J.L.; Gigliotti, F. Immunization with Pneumocystis Cross-Reactive Antigen 1 (Pca1) Protects Mice against Pneumocystis Pneumonia and Generates Antibody to Pneumocystis jirovecii. Infect. Immun. 2017, 85, e00850-16. [Google Scholar] [CrossRef] [Green Version]
  119. Lee, L.H.; Gigliotti, F.; Wright, T.W.; Simpson-Haidaris, P.J.; Weinberg, G.A.; Haidaris, C.G. Molecular characterization of KEX1, a kexin-like protease in mouse Pneumocystis carinii. Gene 2000, 242, 141–150. [Google Scholar] [CrossRef]
  120. Gigliotti, F.; Haidaris, C.G.; Wright, T.W.; Harmsen, A.G. Passive intranasal monoclonal antibody prophylaxis against murine Pneumocystis carinii pneumonia. Infect. Immun. 2002, 70, 1069–1074. [Google Scholar] [CrossRef] [Green Version]
  121. Kling, H.M.; Norris, K.A. Vaccine-Induced Immunogenicity and Protection Against Pneumocystis Pneumonia in a Nonhuman Primate Model of HIV and Pneumocystis Coinfection. J. Infect. Dis. 2016, 213, 1586–1595. [Google Scholar] [CrossRef] [Green Version]
  122. Cobos Jiménez, V.; Rabacal, W.; Rayens, E.; Norris, K.A. Immunization with Pneumocystis recombinant KEX1 induces robust and durable humoral responses in immunocompromised non-human primates. Hum. Vaccines Immunother. 2019, 15, 2075–2080. [Google Scholar] [CrossRef]
  123. Peglow, S.L.; Smulian, A.G.; Linke, M.J.; Pogue, C.L.; Nurre, S.; Crisler, J.; Phair, J.; Gold, J.W.; Armstrong, D.; Walzer, P.D. Serologic responses to Pneumocystis carinii antigens in health and disease. J. Infect. Dis. 1990, 161, 296–306. [Google Scholar] [CrossRef]
  124. Respaldiza, N.; Medrano, F.J.; Medrano, A.C.; Varela, J.M.; de la Horra, C.; Montes-Cano, M.; Ferrer, S.; Wichmann, I.; Gargallo-Viola, D.; Calderon, E.J. High seroprevalence of Pneumocystis infection in Spanish children. Clin. Microbiol. Infect. 2004, 10, 1029–1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Vargas, S.L.; Hughes, W.T.; Santolaya, M.E.; Ulloa, A.V.; Ponce, C.A.; Cabrera, C.E.; Cumsille, F.; Gigliotti, F. Search for primary infection by Pneumocystis carinii in a cohort of normal, healthy infants. Clin. Infect. Dis. 2001, 32, 855–861. [Google Scholar] [CrossRef] [Green Version]
  126. Gingo, M.R.; Lucht, L.; Daly, K.R.; Djawe, K.; Palella, F.J.; Abraham, A.G.; Bream, J.H.; Witt, M.D.; Kingsley, L.A.; Norris, K.A.; et al. Serologic responses to pneumocystis proteins in HIV patients with and without Pneumocystis jirovecii pneumonia. J. Acquir. Immune Defic. Syndr. 2011, 57, 190–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Kling, H.M.; Shipley, T.W.; Patil, S.P.; Kristoff, J.; Bryan, M.; Montelaro, R.C.; Morris, A.; Norris, K.A. Relationship of Pneumocystis jiroveci humoral immunity to prevention of colonization and chronic obstructive pulmonary disease in a primate model of HIV infection. Infect. Immun. 2010, 78, 4320–4330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Ruan, S.; Cai, Y.; Ramsay, A.J.; Welsh, D.A.; Norris, K.; Shellito, J.E. B cell and antibody responses in mice induced by a putative cell surface peptidase of Pneumocystis murina protect against experimental infection. Vaccine 2017, 35, 672–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Schmidt, J.J.; Lueck, C.; Ziesing, S.; Stoll, M.; Haller, H.; Gottlieb, J.; Eder, M.; Welte, T.; Hoeper, M.M.; Scherag, A.; et al. Clinical course, treatment and outcome of Pneumocystis pneumonia in immunocompromised adults: A retrospective analysis over 17 years. Crit. Care 2018, 22, 307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Matsumoto, T.; Fujita, M.; Hirano, R.; Sasaki, T.; Watanabe, K. Risk factors for pneumocystis pneumonia onset in HIV-negative patients treated with high-dose systemic corticosteroids. Infect. Dis. 2019, 51, 305–307. [Google Scholar] [CrossRef]
  131. Gonzalez Santiago, T.M.; Wetter, D.A.; Kalaaji, A.N.; Limper, A.H.; Lehman, J.S. Pneumocystis jiroveci pneumonia in patients treated with systemic immunosuppressive agents for dermatologic conditions: A systematic review with recommendations for prophylaxis. Int. J. Dermatol. 2016, 55, 823–830. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of drugs currently in use or in clinical trials for prevention and treatment of Pneumocystis infection.
Figure 1. Chemical structures of drugs currently in use or in clinical trials for prevention and treatment of Pneumocystis infection.
Pathogens 10 00236 g001
Table 1. Summary of preclinical vaccine studies for prevention of Pneumocystis infection.
Table 1. Summary of preclinical vaccine studies for prevention of Pneumocystis infection.
AntigenAnimal
Model
Dose
Route
Adjuvant
Immunosuppression MethodProtective?Efficacy Readout (Result)Ref
Inactivated
whole
organism
Murine106 trophs
IN
Cholera toxin B
CD4 T-cell depletion via mAbPartiali. Stained lung smears (no organisms present)
ii. PCR (positive for mitochondrial rRNA)
[99]
Murine107 organisms
IT
None
CD4 T-cell depletion via mAbPartiali. Stained lung smears (decreased organism burden)[100]
Murine107 nuclei
IT
None
CD4 T-cell depletion via mAbYesi. Stained lung smears (no organisms present)
ii. PCR (negative for glycoprotein A and mitochondrial rRNA)
[53]
Murine106 cysts
IT
None
CD4 T-cell depletion via mAbYesi. Stained lung smears (no organisms present)[101]
gpa (Msg)Murine10-10 µg
IT
Quil A
CD4 T-cell depletion via mAbNoi. Stained lung smears (organisms present)[102]
Rat1-100 µg
SubQ
Titermax
MethylprednisolonePartialii. Stained lung smears and sections (Decrease in organism burden)[109]
p55Rat100 µg
SubQ
Titermax
MethylprednisolonePartiali. Stained lung smears and (Decrease in organism burden)[112]
Rat100 µg
IM
None
DexamethasonePartiali. Stained lung sections (Decrease in organism burden)
ii. PCR (Positive for p55)
[114]
Murine104 Pc-pulsed DCs
IV
None
CD4 T-cell depletion via mAbPartiali. Stained lung sections (no organisms present)
ii. PCR (Positive for mitochondrial rRNA)
[115]
A12 (Pca1)Murine25 µg
SubQ
Titermax
CD4 T-cell depletion via mAbPartiali. PCR (10/14 mice negative for single-copy kex1 gene)
ii. PCR (Positive for gpA gene)
[117]
Murine100 µg
SubQ
Titermax
CD4 T-cell depletion via mAbPartial i. PCR (Positive for single-copy kex1 gene)
ii. PCR (negative for multicopy gpA gene)
[118]
SPD1Murine5 µg
SubQ
MF-59
CD4 T-cell depletion via mAbPartiali. PCR (reduction in mitochondrial rRNA copy number)[128]
KexinRhesus
macaques
50-100 µg
IM
Alum
Simian immunodeficiency virusPartialCriteria for PCP (1/6 animals developed PCP):
i. BAL smears (detection of organism in BAL fluid)
ii. PCR (PCR of organism DNA in BAL)
iii. Immunohistochemistry (organism detection in lungs)
[121]
IN—intranasal, IT—intratracheal, SubQ—subcutaneous, IM—intramuscular
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gingerich, A.D.; Norris, K.A.; Mousa, J.J. Pneumocystis Pneumonia: Immunity, Vaccines, and Treatments. Pathogens 2021, 10, 236. https://doi.org/10.3390/pathogens10020236

AMA Style

Gingerich AD, Norris KA, Mousa JJ. Pneumocystis Pneumonia: Immunity, Vaccines, and Treatments. Pathogens. 2021; 10(2):236. https://doi.org/10.3390/pathogens10020236

Chicago/Turabian Style

Gingerich, Aaron D., Karen A. Norris, and Jarrod J. Mousa. 2021. "Pneumocystis Pneumonia: Immunity, Vaccines, and Treatments" Pathogens 10, no. 2: 236. https://doi.org/10.3390/pathogens10020236

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