Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26.325
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 40: 121-141, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35007128

ABSTRACT

Invasive fungal diseases are rare in individuals with intact immunity. This, together with the fact that there are only a few species that account for most mycotic diseases, implies a remarkable natural resistance to pathogenic fungi. Mammalian immunity to fungi rests on two pillars, powerful immune mechanisms and elevated temperatures that create a thermal restriction zone for most fungal species. Conditions associated with increased susceptibility generally reflect major disturbances of immune function involving both the cellular and humoral innate and adaptive arms, which implies considerable redundancy in host defense mechanisms against fungi. In general, tissue fungal invasion is controlled through either neutrophil or granulomatous inflammation, depending on the fungal species. Neutrophils are critical against Candida spp. and Aspergillus spp. while macrophages are essential for controlling mycoses due to Cryptococcus spp., Histoplasma spp., and other fungi. The increasing number of immunocompromised patients together with climate change could significantly increase the prevalence of fungal diseases.


Subject(s)
Mycoses , Animals , Fungi , Humans , Immunity, Innate , Immunocompromised Host , Macrophages , Mammals
2.
Annu Rev Immunol ; 36: 157-191, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29237128

ABSTRACT

In the last few decades, the AIDS pandemic and the significant advances in the medical management of individuals with neoplastic and inflammatory conditions have resulted in a dramatic increase in the population of immunosuppressed patients with opportunistic, life-threatening fungal infections. The parallel development of clinically relevant mouse models of fungal disease and the discovery and characterization of several inborn errors of immune-related genes that underlie inherited human susceptibility to opportunistic mycoses have significantly expanded our understanding of the innate and adaptive immune mechanisms that protect against ubiquitous fungal exposures. This review synthesizes immunological knowledge derived from basic mouse studies and from human cohorts and provides an overview of mammalian antifungal host defenses that show promise for informing therapeutic and vaccination strategies for vulnerable patients.


Subject(s)
Host-Pathogen Interactions/immunology , Mycoses/immunology , Mycoses/microbiology , Acquired Immunodeficiency Syndrome/complications , Acquired Immunodeficiency Syndrome/immunology , Adaptive Immunity , Animals , Disease Susceptibility , Fungal Vaccines/immunology , Fungi/immunology , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunity, Innate , Immunocompromised Host , Immunotherapy , Mycoses/prevention & control , Mycoses/therapy , Signal Transduction
3.
Annu Rev Immunol ; 33: 787-821, 2015.
Article in English | MEDLINE | ID: mdl-25706097

ABSTRACT

Epstein-Barr virus (EBV) is usually acquired silently early in life and carried thereafter as an asymptomatic infection of the B lymphoid system. However, many circumstances disturb the delicate EBV-host balance and cause the virus to display its pathogenic potential. Thus, primary infection in adolescence can manifest as infectious mononucleosis (IM), as a fatal illness that magnifies the immunopathology of IM in boys with the X-linked lymphoproliferative disease trait, and as a chronic active disease leading to life-threatening hemophagocytosis in rare cases of T or natural killer (NK) cell infection. Patients with primary immunodeficiencies affecting the NK and/or T cell systems, as well as immunosuppressed transplant recipients, handle EBV infections poorly, and many are at increased risk of virus-driven B-lymphoproliferative disease. By contrast, a range of other EBV-positive malignancies of lymphoid or epithelial origin arise in individuals with seemingly intact immune systems through mechanisms that remain to be understood.


Subject(s)
Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Adaptive Immunity , Animals , Carrier State , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/physiology , Humans , Immunity, Innate , Immunocompromised Host , Immunologic Deficiency Syndromes/etiology , Lymphoproliferative Disorders/etiology
4.
Cell ; 184(10): 2605-2617.e18, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33831372

ABSTRACT

Many individuals mount nearly identical antibody responses to SARS-CoV-2. To gain insight into how the viral spike (S) protein receptor-binding domain (RBD) might evolve in response to common antibody responses, we studied mutations occurring during virus evolution in a persistently infected immunocompromised individual. We use antibody Fab/RBD structures to predict, and pseudotypes to confirm, that mutations found in late-stage evolved S variants confer resistance to a common class of SARS-CoV-2 neutralizing antibodies we isolated from a healthy COVID-19 convalescent donor. Resistance extends to the polyclonal serum immunoglobulins of four out of four healthy convalescent donors we tested and to monoclonal antibodies in clinical use. We further show that affinity maturation is unimportant for wild-type virus neutralization but is critical to neutralization breadth. Because the mutations we studied foreshadowed emerging variants that are now circulating across the globe, our results have implications to the long-term efficacy of S-directed countermeasures.


Subject(s)
Antibodies, Viral/immunology , COVID-19 , Evolution, Molecular , Immune Evasion/immunology , Immunocompromised Host , Immunoglobulin Fab Fragments/immunology , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Antibodies, Neutralizing , COVID-19/genetics , COVID-19/immunology , Female , HEK293 Cells , Humans , Male , Protein Domains , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
5.
Nat Immunol ; 19(3): 233-245, 2018 03.
Article in English | MEDLINE | ID: mdl-29358709

ABSTRACT

Malignancies can compromise innate immunity, but the mechanisms of this are largely unknown. Here we found that, via tumor-derived exosomes (TEXs), cancers were able to transfer activated epidermal growth factor receptor (EGFR) to host macrophages and thereby suppress innate antiviral immunity. Screening of the human kinome identified the kinase MEKK2 in macrophages as an effector of TEX-delivered EGFR that negatively regulated the antiviral immune response. In the context of experimental tumor implantation, MEKK2-deficient mice were more resistant to viral infection than were wild-type mice. Injection of TEXs into mice reduced innate immunity, increased viral load and increased morbidity in an EGFR- and MEKK2-dependent manner. MEKK2 phosphorylated IRF3, a transcription factor crucial for the production of type I interferons; this triggered poly-ubiquitination of IRF3 and blocked its dimerization, translocation to the nucleus and transcriptional activity after viral infection. These findings identify a mechanism by which cancer cells can dampen host innate immunity and potentially cause patients with cancer to become immunocompromised.


Subject(s)
ErbB Receptors/immunology , Exosomes/immunology , Immunity, Innate/immunology , Neoplasms/immunology , Virus Diseases/immunology , Adult , Animals , ErbB Receptors/metabolism , Exosomes/metabolism , Female , Humans , Immunocompromised Host/immunology , MAP Kinase Kinase Kinase 2/immunology , MAP Kinase Kinase Kinase 2/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged
6.
Nature ; 626(8001): 1094-1101, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38383783

ABSTRACT

Persistent SARS-CoV-2 infections may act as viral reservoirs that could seed future outbreaks1-5, give rise to highly divergent lineages6-8 and contribute to cases with post-acute COVID-19 sequelae (long COVID)9,10. However, the population prevalence of persistent infections, their viral load kinetics and evolutionary dynamics over the course of infections remain largely unknown. Here, using viral sequence data collected as part of a national infection survey, we identified 381 individuals with SARS-CoV-2 RNA at high titre persisting for at least 30 days, of which 54 had viral RNA persisting at least 60 days. We refer to these as 'persistent infections' as available evidence suggests that they represent ongoing viral replication, although the persistence of non-replicating RNA cannot be ruled out in all. Individuals with persistent infection had more than 50% higher odds of self-reporting long COVID than individuals with non-persistent infection. We estimate that 0.1-0.5% of infections may become persistent with typically rebounding high viral loads and last for at least 60 days. In some individuals, we identified many viral amino acid substitutions, indicating periods of strong positive selection, whereas others had no consensus change in the sequences for prolonged periods, consistent with weak selection. Substitutions included mutations that are lineage defining for SARS-CoV-2 variants, at target sites for monoclonal antibodies and/or are commonly found in immunocompromised people11-14. This work has profound implications for understanding and characterizing SARS-CoV-2 infection, epidemiology and evolution.


Subject(s)
COVID-19 , Health Surveys , Persistent Infection , SARS-CoV-2 , Humans , Amino Acid Substitution , Antibodies, Monoclonal/immunology , COVID-19/epidemiology , COVID-19/virology , Evolution, Molecular , Immunocompromised Host/immunology , Mutation , Persistent Infection/epidemiology , Persistent Infection/virology , Post-Acute COVID-19 Syndrome/epidemiology , Post-Acute COVID-19 Syndrome/virology , Prevalence , RNA, Viral/analysis , RNA, Viral/genetics , SARS-CoV-2/chemistry , SARS-CoV-2/classification , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Selection, Genetic , Self Report , Time Factors , Viral Load , Virus Replication
7.
CA Cancer J Clin ; 71(6): 505-526, 2021 11.
Article in English | MEDLINE | ID: mdl-34499351

ABSTRACT

Despite being highly preventable, cervical cancer is the fourth most common cancer and cause of cancer death in women globally. In low-income countries, cervical cancer is often the leading cause of cancer-related morbidity and mortality. Women living with human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome are at a particularly high risk of cervical cancer because of an impaired immune response to human papillomavirus, the obligate cause of virtually all cervical cancers. Globally, approximately 1 in 20 cervical cancers is attributable to HIV; in sub-Saharan Africa, approximately 1 in 5 cervical cancers is due to HIV. Here, the authors provide a critical appraisal of the evidence to date on the impact of HIV disease on cervical cancer risk, describe key methodologic issues, and frame the key outstanding research questions, especially as they apply to ongoing global efforts for prevention and control of cervical cancer. Expanded efforts to integrate HIV care with cervical cancer prevention and control, and vice versa, could assist the global effort to eliminate cervical cancer as a public health problem.


Subject(s)
HIV Infections/epidemiology , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/prevention & control , Early Detection of Cancer , Female , Humans , Immunocompromised Host , Papillomavirus Infections , Papillomavirus Vaccines , Precancerous Conditions/therapy , Primary Prevention , Secondary Prevention
8.
CA Cancer J Clin ; 71(6): 488-504, 2021 11.
Article in English | MEDLINE | ID: mdl-34546590

ABSTRACT

Infection is the second leading cause of death in patients with cancer. Loss of efficacy in antibiotics due to antibiotic resistance in bacteria is an urgent threat against the continuing success of cancer therapy. In this review, the authors focus on recent updates on the impact of antibiotic resistance in the cancer setting, particularly on the ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.). This review highlights the health and financial impact of antibiotic resistance in patients with cancer. Furthermore, the authors recommend measures to control the emergence of antibiotic resistance, highlighting the risk factors associated with cancer care. A lack of data in the etiology of infections, specifically in oncology patients in United States, is identified as a concern, and the authors advocate for a centralized and specialized surveillance system for patients with cancer to predict and prevent the emergence of antibiotic resistance. Finding better ways to predict, prevent, and treat antibiotic-resistant infections will have a major positive impact on the care of those with cancer.


Subject(s)
Drug Resistance, Multiple, Bacterial , Neoplasms/complications , Anti-Bacterial Agents/therapeutic use , Antimicrobial Stewardship , Humans , Immunocompromised Host , Opportunistic Infections/prevention & control , Prescription Drug Misuse/prevention & control
9.
Physiol Rev ; 100(2): 603-632, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31600121

ABSTRACT

Despite anti-retroviral therapy (ART), human immunodeficiency virus-1 (HIV)-related pulmonary disease continues to be a major cause of morbidity and mortality for people living with HIV (PLWH). The spectrum of lung diseases has changed from acute opportunistic infections resulting in death to chronic lung diseases for those with access to ART. Chronic immune activation and suppression can result in impairment of innate immunity and progressive loss of T cell and B cell functionality with aberrant cytokine and chemokine responses systemically as well as in the lung. HIV can be detected in the lungs of PLWH and has profound effects on cellular immune functions. In addition, HIV-related lung injury and disease can occur secondary to a number of mechanisms including altered pulmonary and systemic inflammatory pathways, viral persistence in the lung, oxidative stress with additive effects of smoke exposure, microbial translocation, and alterations in the lung and gut microbiome. Although ART has had profound effects on systemic viral suppression in HIV, the impact of ART on lung immunology still needs to be fully elucidated. Understanding of the mechanisms by which HIV-related lung diseases continue to occur is critical to the development of new preventive and therapeutic strategies to improve lung health in PLWH.


Subject(s)
AIDS-Related Opportunistic Infections/immunology , Asthma/immunology , HIV Infections/immunology , HIV/immunology , Hypertension, Pulmonary/immunology , Lung Neoplasms/immunology , Lung/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Respiratory Tract Infections/immunology , AIDS-Related Opportunistic Infections/drug therapy , AIDS-Related Opportunistic Infections/microbiology , AIDS-Related Opportunistic Infections/virology , Animals , Anti-HIV Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Asthma/virology , Disease Models, Animal , HIV/drug effects , HIV/pathogenicity , HIV Infections/drug therapy , HIV Infections/virology , Host-Pathogen Interactions , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/virology , Immunocompromised Host , Lung/drug effects , Lung/microbiology , Lung/virology , Lung Neoplasms/drug therapy , Lung Neoplasms/virology , Prognosis , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/virology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Risk Factors
10.
Trends Immunol ; 45(7): 511-522, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38890026

ABSTRACT

The effect of COVID-19 on the high number of immunocompromised people living with HIV-1 (PLWH), particularly in Africa, remains a critical concern. Here, we identify key areas that still require further investigation, by examining COVID-19 vaccine effectiveness, and understanding antibody responses in SARS-CoV-2 infection and vaccination in comparison with people without HIV-1 (PWOH). We also assess the potential impact of pre-existing immunity against endemic human coronaviruses on SARS-CoV-2 responses in these individuals. Lastly, we discuss the consequences of persistent infection in PLWH (or other immunocompromised individuals), including prolonged shedding, increased viral diversity within the host, and the implications on SARS-CoV-2 evolution in Africa.


Subject(s)
Antibodies, Viral , COVID-19 , HIV Infections , HIV-1 , Immunity, Humoral , SARS-CoV-2 , Humans , COVID-19/immunology , HIV Infections/immunology , HIV Infections/virology , SARS-CoV-2/immunology , HIV-1/immunology , Antibodies, Viral/immunology , COVID-19 Vaccines/immunology , Immunocompromised Host/immunology
11.
N Engl J Med ; 388(9): 804-812, 2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36856616

ABSTRACT

BACKGROUND: Immunosuppressed organ-transplant recipients have an increased incidence of, and mortality from, skin cancer. Nicotinamide (vitamin B3) enhances the repair of ultraviolet (UV) radiation-induced DNA damage, reduces the cutaneous immunosuppressive effects of UV radiation, and reduces the incidence of keratinocyte cancers (including squamous-cell and basal-cell carcinomas) and actinic keratoses among high-risk immunocompetent patients. Whether oral nicotinamide is useful for skin-cancer chemoprevention in organ-transplant recipients is unclear. METHODS: In this phase 3 trial, we randomly assigned, in a 1:1 ratio, organ-transplant recipients who had had at least two keratinocyte cancers in the past 5 years to receive 500 mg of nicotinamide or placebo twice daily for 12 months. Participants were examined for skin lesions by dermatologists at 3-month intervals for 12 months. The primary end point was the number of new keratinocyte cancers during the 12-month intervention period. Secondary end points included the numbers of squamous-cell and basal-cell carcinomas during the 12-month intervention period, the number of actinic keratoses until 6 months after randomization, safety, and quality of life. RESULTS: A total of 158 participants were enrolled, with 79 assigned to the nicotinamide group and 79 to the placebo group. The trial was stopped early owing to poor recruitment. At 12 months, there were 207 new keratinocyte cancers in the nicotinamide group and 210 in the placebo group (rate ratio, 1.0; 95% confidence interval, 0.8 to 1.3; P = 0.96). No significant between-group differences in squamous-cell and basal-cell carcinoma counts, actinic keratosis counts, or quality-of-life scores were observed. Adverse events and changes in blood or urine laboratory variables were similar in the two groups. CONCLUSIONS: In this 12-month, placebo-controlled trial, oral nicotinamide therapy did not lead to lower numbers of keratinocyte cancers or actinic keratoses in immunosuppressed solid-organ transplant recipients. (Funded by the National Health and Medical Research Council; ONTRANS Australian New Zealand Clinical Trials Registry number, ACTRN12617000599370.).


Subject(s)
Antineoplastic Agents , Niacinamide , Skin Neoplasms , Transplant Recipients , Humans , Australia , Carcinoma, Basal Cell/etiology , Carcinoma, Basal Cell/prevention & control , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/prevention & control , Chemoprevention , Keratosis, Actinic/etiology , Keratosis, Actinic/prevention & control , Niacinamide/administration & dosage , Niacinamide/therapeutic use , Quality of Life , Skin Neoplasms/etiology , Skin Neoplasms/prevention & control , Immunocompromised Host , Organ Transplantation/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Ultraviolet Rays/adverse effects
12.
Clin Microbiol Rev ; 37(2): e0009923, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38546225

ABSTRACT

SUMMARYAs Chagas disease remains prevalent in the Americas, it is important that healthcare professionals and researchers are aware of the screening, diagnosis, monitoring, and treatment recommendations for the populations of patients they care for and study. Management of Trypanosoma cruzi infection in immunocompromised hosts is challenging, particularly because, regardless of antitrypanosomal treatment status, immunocompromised patients with Chagas disease are at risk for T. cruzi reactivation, which can be lethal. Evidence-based practices to prevent and manage T. cruzi reactivation vary depending on the type of immunocompromise. Here, we review available data describing Chagas disease epidemiology, testing, and management practices for various populations of immunocompromised individuals, including people with HIV and patients undergoing solid organ and hematopoietic stem cell transplantation.


Subject(s)
Chagas Disease , Immunocompromised Host , Humans , Chagas Disease/diagnosis , Chagas Disease/epidemiology , Chagas Disease/immunology , Chagas Disease/therapy , Trypanosoma cruzi/immunology
13.
J Virol ; 98(9): e0090524, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39207133

ABSTRACT

Immunocompromised people are at high risk of prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and progression to severe coronavirus disease 2019 (COVID-19). However, the efficacy of late-onset direct-acting antiviral (DAA) therapy with therapeutics in clinical use and experimental drugs to mitigate persistent viral replication is unclear. In this study, we employed an immunocompromised mouse model, which supports prolonged replication of SARS-CoV-2 to explore late-onset treatment options. Tandem immuno-depletion of CD4+ and CD8+ T cells in C57BL/6 mice followed by infection with SARS-CoV-2 variant of concern (VOC) beta B.1.351 resulted in prolonged infection with virus replication for 5 weeks after inoculation. Early-onset treatment with nirmatrelvir/ritonavir (paxlovid) or molnupiravir was only moderately efficacious, whereas the experimental therapeutic 4'-fluorouridine (4'-FlU, EIDD-2749) significantly reduced virus load in the upper and lower respiratory compartments 4 days postinfection (dpi). All antivirals significantly lowered virus burden in a 7-day treatment regimen initiated 14 dpi, but paxlovid-treated animals experienced rebound virus replication in the upper respiratory tract 7 days after treatment end. Viral RNA was detectable 28 dpi in paxlovid-treated animals, albeit not in the molnupiravir or 4'-FlU groups, when treatment was initiated 14 dpi and continued for 14 days. Low-level virus replication continued 35 dpi in animals receiving vehicle but had ceased in all treatment groups. These data indicate that late-onset DAA therapy significantly shortens the duration of persistent virus replication in an immunocompromised host, which may have implications for clinical use of antiviral therapeutics to alleviate the risk of progression to severe disease in highly vulnerable patients. IMPORTANCE: Four years after the onset of the global coronavirus disease 2019 (COVID-19) pandemic, the immunocompromised are at greatest risk of developing life-threatening severe disease. However, specific treatment plans for this most vulnerable patient group have not yet been developed. Employing a CD4+ and CD8+ T cell-depleted immunocompromised mouse model of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, we explored therapeutic options of persistent infections with standard-of-care paxlovid, molnupiravir, and the experimental therapeutic 4'-fluorouridine (4'-FlU). Late-onset treatment initiated 14 days after infection was efficacious, but only 4'-FlU was rapidly sterilizing. No treatment-experienced viral variants with reduced susceptibility to the drugs emerged, albeit virus replication rebounded in animals of the paxlovid group after treatment end. This study supports the use of direct-acting antivirals (DAAs) for late-onset management of persistent SARS-CoV-2 infection in immunocompromised hosts. However, treatment courses likely require to be extended for maximal therapeutic benefit, calling for appropriately powered clinical trials to meet the specific needs of this patient group.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Disease Models, Animal , Immunocompromised Host , Mice, Inbred C57BL , SARS-CoV-2 , Viral Load , Virus Replication , Animals , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , SARS-CoV-2/immunology , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Mice , Virus Replication/drug effects , Viral Load/drug effects , COVID-19/virology , COVID-19/immunology , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Female , Humans , Ritonavir/therapeutic use , Cytidine/analogs & derivatives , Hydroxylamines
14.
CA Cancer J Clin ; 68(5): 340-355, 2018 09.
Article in English | MEDLINE | ID: mdl-29985544

ABSTRACT

Therapies in oncology have evolved rapidly over the last years. At the same pace, supportive care for patients receiving cancer therapy has also evolved, allowing patients to safely receive the newest advances in treatment in both an inpatient and outpatient basis. The recognition of the role of infection control and prevention (ICP) in the outcomes of patients living with cancer has been such that it is now a requirement for hospitals and involves multidisciplinary groups. Some unique aspects of ICP for patients with cancer that have gained momentum over the past few decades include catheter-related infections, multidrug-resistant organisms, community-acquired viral infections, and the impact of the health care environment on the horizontal transmission of organisms. Furthermore, as the potential for infections to cross international borders has increased, alertness for outbreaks or new infections that occur outside the area have become constant. As the future approaches, ICP in immunocompromised hosts will continue to integrate emerging disciplines, such as antibiotic stewardship and the microbiome, and new techniques for environmental cleaning and for controlling the spread of infections, such as whole-genome sequencing. CA Cancer J Clin 2018;000:000-000. © 2018 American Cancer Society.


Subject(s)
Cancer Care Facilities/standards , Immunocompromised Host , Infection Control/methods , Infection Control/standards , Neoplasms/immunology , Ambulatory Care/standards , Anti-Bacterial Agents/therapeutic use , Antifungal Agents/therapeutic use , Antimicrobial Stewardship , Catheter-Related Infections/prevention & control , Cross Infection/microbiology , Cross Infection/prevention & control , Diet , Drug Resistance, Multiple, Bacterial , Food Safety , Hand Disinfection , Humans , Patient Isolation , United States , Virus Diseases/prevention & control
15.
Nature ; 575(7783): 519-522, 2019 11.
Article in English | MEDLINE | ID: mdl-31666702

ABSTRACT

Immunosuppression increases the risk of cancers that are associated with viral infection1. In particular, the risk of squamous cell carcinoma of the skin-which has been associated with beta human papillomavirus (ß-HPV) infection-is increased by more than 100-fold in immunosuppressed patients2-4. Previous studies have not established a causative role for HPVs in driving the development of skin cancer. Here we show that T cell immunity against commensal papillomaviruses suppresses skin cancer in immunocompetent hosts, and the loss of this immunity-rather than the oncogenic effect of HPVs-causes the markedly increased risk of skin cancer in immunosuppressed patients. To investigate the effects of papillomavirus on carcinogen-driven skin cancer, we colonized several strains of immunocompetent mice with mouse papillomavirus type 1 (MmuPV1)5. Mice with natural immunity against MmuPV1 after colonization and acquired immunity through the transfer of T cells from immune mice or by MmuPV1 vaccination were protected against skin carcinogenesis induced by chemicals or by ultraviolet radiation in a manner dependent on CD8+ T cells. RNA and DNA in situ hybridization probes for 25 commensal ß-HPVs revealed a significant reduction in viral activity and load in human skin cancer compared with the adjacent healthy skin, suggesting a strong immune selection against virus-positive malignant cells. Consistently, E7 peptides from ß-HPVs activated CD8+ T cells from unaffected human skin. Our findings reveal a beneficial role for commensal viruses and establish a foundation for immune-based approaches that could block the development of skin cancer by boosting immunity against the commensal HPVs present in all of our skin.


Subject(s)
Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/prevention & control , Papillomaviridae/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Skin Neoplasms/prevention & control , Skin Neoplasms/virology , Symbiosis , Aged , Aged, 80 and over , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Female , Humans , Immunocompromised Host/immunology , Male , Mice , Middle Aged , Oncogenes , Papillomaviridae/genetics , Papillomaviridae/pathogenicity , RNA, Viral/analysis , RNA, Viral/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Ultraviolet Rays
16.
J Allergy Clin Immunol ; 153(6): 1655-1667, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38154666

ABSTRACT

BACKGROUND: Functional T-cell responses are essential for virus clearance and long-term protection after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, whereas certain clinical factors, such as older age and immunocompromise, are associated with worse outcome. OBJECTIVE: We sought to study the breadth and magnitude of T-cell responses in patients with coronavirus disease 2019 (COVID-19) and in individuals with inborn errors of immunity (IEIs) who had received COVID-19 mRNA vaccine. METHODS: Using high-throughput sequencing and bioinformatics tools to characterize the T-cell receptor ß repertoire signatures in 540 individuals after SARS-CoV-2 infection, 31 IEI recipients of COVID-19 mRNA vaccine, and healthy controls, we quantified HLA class I- and class II-restricted SARS-CoV-2-specific responses and also identified several HLA allele-clonotype motif associations in patients with COVID-19, including a subcohort of anti-type 1 interferon (IFN-1)-positive patients. RESULTS: Our analysis revealed that elderly patients with COVID-19 with critical disease manifested lower SARS-CoV-2 T-cell clonotype diversity as well as T-cell responses with reduced magnitude, whereas the SARS-CoV-2-specific clonotypes targeted a broad range of HLA class I- and class II-restricted epitopes across the viral proteome. The presence of anti-IFN-I antibodies was associated with certain HLA alleles. Finally, COVID-19 mRNA immunization induced an increase in the breadth of SARS-CoV-2-specific clonotypes in patients with IEIs, including those who had failed to seroconvert. CONCLUSIONS: Elderly individuals have impaired capacity to develop broad and sustained T-cell responses after SARS-CoV-2 infection. Genetic factors may play a role in the production of anti-IFN-1 antibodies. COVID-19 mRNA vaccines are effective in inducing T-cell responses in patients with IEIs.


Subject(s)
COVID-19 , Immunocompromised Host , SARS-CoV-2 , Humans , COVID-19/immunology , SARS-CoV-2/immunology , Male , Middle Aged , Female , Immunocompromised Host/immunology , Adult , Aged , T-Lymphocytes/immunology , COVID-19 Vaccines/immunology , Immunocompetence/immunology
17.
Clin Microbiol Rev ; 36(4): e0015922, 2023 12 20.
Article in English | MEDLINE | ID: mdl-37937988

ABSTRACT

Invasive fusariosis is a serious invasive fungal disease, affecting immunocompetent and, more frequently, immunocompromised patients. Localized disease is the typical clinical form in immunocompetent patients. Immunocompromised hosts at elevated risk of developing invasive fusariosis are patients with acute leukemia receiving chemotherapeutic regimens for remission induction, and those undergoing allogeneic hematopoietic cell transplant. In this setting, the infection is usually disseminated with positive blood cultures, multiple painful metastatic skin lesions, and lung involvement. Currently available antifungal agents have poor in vitro activity against Fusarium species, but a clear-cut correlation between in vitro activity and clinical effectiveness does not exist. The outcome of invasive fusariosis is largely dependent on the resolution of immunosuppression, especially neutrophil recovery in neutropenic patients.


Subject(s)
Fusariosis , Fusarium , Hematopoietic Stem Cell Transplantation , Humans , Fusariosis/drug therapy , Fusariosis/microbiology , Antifungal Agents/therapeutic use , Immunocompromised Host
18.
Clin Microbiol Rev ; 36(4): e0003323, 2023 12 20.
Article in English | MEDLINE | ID: mdl-37937980

ABSTRACT

Strongyloidiasis is a World Health Organization neglected tropical disease usually caused by Strongyloides stercoralis, a parasitic worm with a complex life cycle. Globally, 300-600 million people are infected through contact with fecally contaminated soil. An autoinfective component of the life cycle can lead to chronic infection that may be asymptomatic or cause long-term symptoms, including malnourishment in children. Low larval output can limit the sensitivity of detection in stool, with serology being effective but less sensitive in immunocompromise. Host immunosuppression can trigger catastrophic, fatal hyperinfection/dissemination, where large numbers of larvae pierce the bowel wall and disseminate throughout the organs. Stable disease is effectively treated by single-dose ivermectin, with disease in immunocompromised patients treated with multiple doses. Strategies for management include raising awareness, clarifying zoonotic potential, the development and use of effective diagnostic tests for epidemiological studies and individual diagnosis, and the implementation of treatment programs with research into therapeutic alternatives and medication safety.


Subject(s)
Strongyloides stercoralis , Strongyloidiasis , Animals , Child , Humans , Strongyloidiasis/diagnosis , Strongyloidiasis/drug therapy , Strongyloidiasis/parasitology , Ivermectin/pharmacology , Ivermectin/therapeutic use , Immunocompromised Host , Immunosuppression Therapy
19.
J Infect Dis ; 229(Supplement_2): S219-S228, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38243606

ABSTRACT

BACKGROUND: Pathology and Monkeypox virus (MPXV) tissue tropism in severe and fatal human mpox is not thoroughly described but can help elucidate the disease pathogenesis and the role of coinfections in immunocompromised patients. METHODS: We analyzed biopsy and autopsy tissues from 22 patients with severe or fatal outcomes to characterize pathology and viral antigen and DNA distribution in tissues by immunohistochemistry and in situ hybridization. Tissue-based testing for coinfections was also performed. RESULTS: Mucocutaneous lesions showed necrotizing and proliferative epithelial changes. Deceased patients with autopsy tissues evaluated had digestive tract lesions, and half had systemic tissue necrosis with thrombotic vasculopathy in lymphoid tissues, lung, or other solid organs. Half also had bronchopneumonia, and one-third had acute lung injury. All cases had MPXV antigen and DNA detected in tissues. Coinfections were identified in 5 of 16 (31%) biopsy and 4 of 6 (67%) autopsy cases. CONCLUSIONS: Severe mpox in immunocompromised patients is characterized by extensive viral infection of tissues and viremic dissemination that can progress despite available therapeutics. Digestive tract and lung involvement are common and associated with prominent histopathological and clinical manifestations. Coinfections may complicate mpox diagnosis and treatment. Significant viral DNA (likely correlating to infectious virus) in tissues necessitates enhanced biosafety measures in healthcare and autopsy settings.


Subject(s)
Coinfection , Mpox (monkeypox) , Humans , Monkeypox virus , Immunocompromised Host , Antigens, Viral , DNA, Viral
20.
J Infect Dis ; 229(4): 1041-1049, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-37956413

ABSTRACT

BACKGROUND: A severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection lasts longer in immunocompromised hosts than in immunocompetent patients. Prolonged infection is associated with a higher probability of selection for novel SARS-CoV-2 mutations, particularly in the spike protein, a critical target for vaccines and therapeutics. METHODS: From December 2020 to September 2022, respiratory samples from 444 immunocompromised patients and 234 health care workers positive for SARS-CoV-2, diagnosed at 2 hospitals in Paris, France, were analyzed using whole-genome sequencing using Nanopore technology. Custom scripts were developed to assess the SARS-CoV-2 genetic diversity between the 2 groups and within the host. RESULTS: Most infections were SARS-CoV-2 Delta or Omicron lineages. Viral genetic diversity was significantly higher in infections of immunocompromised patients than those of controls. Minor mutations were identified in viruses sequenced from immunocompromised individuals, which became signature mutations for newer SARS-CoV-2 variants as the epidemic progressed. Two patients were coinfected with Delta and Omicron variants. The follow-up of immunocompromised patients revealed that the SARS-CoV-2 genome evolution differed in the upper and lower respiratory tracts. CONCLUSIONS: This study found that SARS-CoV-2 infection in immunocompromised patients is associated with higher genetic diversity, which could lead to the emergence of new SARS-CoV-2 variants with possible immune evasion or different virulence characteristics.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Case-Control Studies , Retrospective Studies , SARS-CoV-2/genetics , Immunocompromised Host , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL