Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61.024
Filter
Add more filters

Publication year range
1.
Cell ; 185(10): 1694-1708.e19, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35447074

ABSTRACT

Immunotherapy is a promising treatment for triple-negative breast cancer (TNBC), but patients relapse, highlighting the need to understand the mechanisms of resistance. We discovered that in primary breast cancer, tumor cells that resist T cell attack are quiescent. Quiescent cancer cells (QCCs) form clusters with reduced immune infiltration. They also display superior tumorigenic capacity and higher expression of chemotherapy resistance and stemness genes. We adapted single-cell RNA-sequencing with precise spatial resolution to profile infiltrating cells inside and outside the QCC niche. This transcriptomic analysis revealed hypoxia-induced programs and identified more exhausted T cells, tumor-protective fibroblasts, and dysfunctional dendritic cells inside clusters of QCCs. This uncovered differential phenotypes in infiltrating cells based on their intra-tumor location. Thus, QCCs constitute immunotherapy-resistant reservoirs by orchestrating a local hypoxic immune-suppressive milieu that blocks T cell function. Eliminating QCCs holds the promise to counteract immunotherapy resistance and prevent disease recurrence in TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Immunosuppressive Agents/therapeutic use , Immunotherapy , Neoplasm Recurrence, Local , T-Lymphocytes/pathology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
2.
Cell ; 181(1): 63-80, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32243797

ABSTRACT

Autoimmune diseases are a result of the immune system being misdirected toward its host and have major and increasing unmet clinical needs. In general, present therapies are broadly acting and non-disease specific; consequently, they are associated with numerous side effects. Precise and early intervention strategies are urgently needed. We highlight the challenges, progress, and prospects in achieving these goals.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Immunosuppressive Agents/therapeutic use , Immunotherapy/methods , Humans
5.
Cell ; 155(5): 1178-87, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24267896

ABSTRACT

There are few substantive methods to measure the health of the immune system, and the connection between immune strength and the viral component of the microbiome is poorly understood. Organ transplant recipients are treated with posttransplant therapies that combine immunosuppressive and antiviral drugs, offering a window into the effects of immune modulation on the virome. We used sequencing of cell-free DNA in plasma to investigate drug-virome interactions in a cohort of organ transplant recipients (656 samples, 96 patients) and find that antivirals and immunosuppressants strongly affect the structure of the virome in plasma. We observe marked virome compositional dynamics at the onset of the therapy and find that the total viral load increases with immunosuppression, whereas the bacterial component of the microbiome remains largely unaffected. The data provide insight into the relationship between the human virome, the state of the immune system, and the effects of pharmacological treatment and offer a potential application of the virome state to predict immunocompetence.


Subject(s)
Antiviral Agents/therapeutic use , Blood/virology , Heart Transplantation , Immunosuppressive Agents/therapeutic use , Lung Transplantation , Viruses/isolation & purification , Adult , Antibiotic Prophylaxis , Blood/microbiology , Child , DNA/blood , DNA/genetics , Humans , Viruses/classification
6.
N Engl J Med ; 390(1): 9-19, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-37888915

ABSTRACT

BACKGROUND: Among patients with chronic limb-threatening ischemia (CLTI) and infrapopliteal artery disease, angioplasty has been associated with frequent reintervention and adverse limb outcomes from restenosis. The effect of the use of drug-eluting resorbable scaffolds on these outcomes remains unknown. METHODS: In this multicenter, randomized, controlled trial, 261 patients with CLTI and infrapopliteal artery disease were randomly assigned in a 2:1 ratio to receive treatment with an everolimus-eluting resorbable scaffold or angioplasty. The primary efficacy end point was freedom from the following events at 1 year: amputation above the ankle of the target limb, occlusion of the target vessel, clinically driven revascularization of the target lesion, and binary restenosis of the target lesion. The primary safety end point was freedom from major adverse limb events at 6 months and from perioperative death. RESULTS: The primary efficacy end point was observed (i.e., no events occurred) in 135 of 173 patients in the scaffold group and 48 of 88 patients in the angioplasty group (Kaplan-Meier estimate, 74% vs. 44%; absolute difference, 30 percentage points; 95% confidence interval [CI], 15 to 46; one-sided P<0.001 for superiority). The primary safety end point was observed in 165 of 170 patients in the scaffold group and 90 of 90 patients in the angioplasty group (absolute difference, -3 percentage points; 95% CI, -6 to 0; one-sided P<0.001 for noninferiority). Serious adverse events related to the index procedure occurred in 2% of the patients in the scaffold group and 3% of those in the angioplasty group. CONCLUSIONS: Among patients with CLTI due to infrapopliteal artery disease, the use of an everolimus-eluting resorbable scaffold was superior to angioplasty with respect to the primary efficacy end point. (Funded by Abbott; LIFE-BTK ClinicalTrials.gov number, NCT04227899.).


Subject(s)
Angioplasty , Blood Vessel Prosthesis Implantation , Chronic Limb-Threatening Ischemia , Drug-Eluting Stents , Peripheral Arterial Disease , Popliteal Artery , Humans , Absorbable Implants , Angioplasty/adverse effects , Angioplasty/methods , Angioplasty, Balloon/adverse effects , Angioplasty, Balloon/methods , Blood Vessel Prosthesis Implantation/methods , Chronic Disease , Chronic Limb-Threatening Ischemia/etiology , Chronic Limb-Threatening Ischemia/surgery , Everolimus/administration & dosage , Everolimus/adverse effects , Everolimus/therapeutic use , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Ischemia/drug therapy , Ischemia/etiology , Ischemia/surgery , Peripheral Arterial Disease/complications , Peripheral Arterial Disease/drug therapy , Peripheral Arterial Disease/surgery , Popliteal Artery/surgery , Tissue Scaffolds , Treatment Outcome
7.
Cell ; 150(6): 1096-9, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22980971

ABSTRACT

The 2012 Lasker-DeBakey Clinical Medical Research Award will be conferred on Thomas Starzl of the University of Pittsburgh School of Medicine in Pittsburgh, Pennsylvania, USA and Roy Calne of the University of Cambridge in Cambridge, UK. They are recognized for pioneering the development of liver transplantation, an intervention that saves 20,000 lives world-wide each year.


Subject(s)
Awards and Prizes , Liver Transplantation/history , History, 20th Century , Humans , Immunosuppressive Agents/therapeutic use , Liver/physiology , Liver Failure/therapy , Liver Transplantation/immunology , Tissue and Organ Procurement , United Kingdom , United States
8.
Lancet ; 403(10441): 2326-2338, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38642569

ABSTRACT

Systemic lupus erythematosus (SLE) is a multisystemic autoimmune disease characterised by the presence of autoantibodies towards nuclear antigens, immune complex deposition, and chronic inflammation at classic target organs such as skin, joints, and kidneys. Despite substantial advances in the diagnosis and management of SLE, the burden of disease remains high. It is important to appreciate the typical presentations and the diagnostic process to facilitate early referral and diagnosis for patients. In most patients, constitutional, mucocutaneous, and musculoskeletal symptoms represent the earliest complaints; these symptoms can include fatigue, lupus-specific rash, mouth ulcers, alopecia, joint pain, and myalgia. In this Seminar we will discuss a diagnostic approach to symptoms in light of the latest classification criteria, which include a systematic evaluation of clinical manifestations (weighted within each domain) and autoantibody profiles (such as anti-double-stranded DNA, anti-Sm, hypocomplementaemia, or antiphospholipid antibodies). Non-pharmacotherapy management is tailored to the individual, with specific lifestyle interventions and patient education to improve quality of life and medication (such as hydroxychloroquine or immunosuppressant) adherence. In the last decade, there have been a few major breakthroughs in approved treatments for SLE and lupus nephritis, such as belimumab, anifrolumab, and voclosporin. However the disease course remains variable and mortality unacceptably high. Access to these expensive medications has also been restricted across different regions of the world. Nonetheless, understanding of treatment goals and strategies has improved. We recognise that the main goal of treatment is the achievement of remission or low disease activity. Comorbidities due to both disease activity and treatment adverse effects, especially infections, osteoporosis, and cardiovascular disease, necessitate vigilant prevention and management strategies. Tailoring treatment options to achieve remission, while balancing treatment-related comorbidities, are priority areas of SLE management.


Subject(s)
Lupus Erythematosus, Systemic , Humans , Lupus Erythematosus, Systemic/therapy , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/complications , Immunosuppressive Agents/therapeutic use , Autoantibodies
9.
Lancet ; 403(10431): 1093-1108, 2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38402885

ABSTRACT

Behçet's syndrome is a rare, chronic multisystemic inflammatory disorder also known as the Silk Route disease due to its geographical distribution. Behçet's syndrome is a multifactorial disease and infectious, genetic, epigenetic, and immunological factors contribute to its pathogenesis. Its heterogeneous spectrum of clinical features include mucocutaneous, articular, ocular, vascular, neurological, and gastrointestinal manifestations that can present with a relapsing and remitting course. Differential diagnosis is often hampered by the non-specific clinical presentation and the absence of laboratory biomarkers or pathognomonic histological features. The therapeutic approach is tailored on the basis of patient-specific manifestations and relies on glucocorticoids, colchicine, and traditional and biological immunosuppressants. Despite progress in the knowledge and management of the disease, unmet needs in diagnostics, monitoring, prediction, and treatment personalisation challenge clinical practice, making Behçet's syndrome a complex disorder associated with an increased risk of morbidity.


Subject(s)
Behcet Syndrome , Humans , Behcet Syndrome/diagnosis , Behcet Syndrome/complications , Behcet Syndrome/drug therapy , Immunosuppressive Agents/therapeutic use , Glucocorticoids/therapeutic use , Recurrence , Diagnosis, Differential
10.
N Engl J Med ; 387(3): 237-247, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35857660

ABSTRACT

BACKGROUND: FLT180a (verbrinacogene setparvovec) is a liver-directed adeno-associated virus (AAV) gene therapy that uses a synthetic capsid and a gain-of-function protein to normalize factor IX levels in patients with hemophilia B. METHODS: In this multicenter, open-label, phase 1-2 trial, we assessed the safety and efficacy of varying doses of FLT180a in patients with severe or moderately severe hemophilia B (factor IX level, ≤2% of normal value). All the patients received glucocorticoids with or without tacrolimus for immunosuppression to decrease the risk of vector-related immune responses. After 26 weeks, patients were enrolled in a long-term follow-up study. The primary end points were safety and efficacy, as assessed by factor IX levels at week 26. RESULTS: Ten patients received one of four FLT180a doses of vector genomes (vg) per kilogram of body weight: 3.84×1011 vg, 6.40×1011 vg, 8.32×1011 vg, or 1.28×1012 vg. After receiving the infusion, all the patients had dose-dependent increases in factor IX levels. At a median follow-up of 27.2 months (range, 19.1 to 42.4), sustained factor IX activity was observed in all the patients except one, who resumed factor IX prophylaxis. As of the data-cutoff date (September 20, 2021), five patients had normal factor IX levels (range, 51 to 78%), three patients had levels from 23 to 43%, and one had a level of 260%. Of the reported adverse events, approximately 10% were related to FLT180a and 24% to immunosuppression. Increases in liver aminotransferase levels were the most common FLT180a-related adverse events. Late increases in aminotransferase levels occurred in patients who had received prolonged tacrolimus beyond the glucocorticoid taper. A serious adverse event of arteriovenous fistula thrombosis occurred in the patient with high factor IX levels. CONCLUSIONS: Sustained factor IX levels in the normal range were observed with low doses of FLT180a but necessitated immunosuppression with glucocorticoids with or without tacrolimus. (Funded by Freeline Therapeutics; ClinicalTrials.gov numbers, NCT03369444 and NCT03641703; EudraCT numbers, 2017-000852-24 and 2017-005080-40.).


Subject(s)
Dependovirus , Genetic Therapy , Glucocorticoids , Hemophilia B , Dependovirus/genetics , Factor IX/analysis , Factor IX/genetics , Follow-Up Studies , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/therapeutic use , Glucocorticoids/adverse effects , Glucocorticoids/therapeutic use , Hemophilia B/genetics , Hemophilia B/metabolism , Hemophilia B/therapy , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Tacrolimus/adverse effects , Tacrolimus/therapeutic use , Transaminases/analysis
11.
N Engl J Med ; 386(1): 11-23, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34986284

ABSTRACT

BACKGROUND: A single-group, phase 1-2 study indicated that eltrombopag improved the efficacy of standard immunosuppressive therapy that entailed horse antithymocyte globulin (ATG) plus cyclosporine in patients with severe aplastic anemia. METHODS: In this prospective, investigator-led, open-label, multicenter, randomized, phase 3 trial, we compared the efficacy and safety of horse ATG plus cyclosporine with or without eltrombopag as front-line therapy in previously untreated patients with severe aplastic anemia. The primary end point was a hematologic complete response at 3 months. RESULTS: Patients were assigned to receive immunosuppressive therapy (Group A, 101 patients) or immunosuppressive therapy plus eltrombopag (Group B, 96 patients). The percentage of patients who had a complete response at 3 months was 10% in Group A and 22% in Group B (odds ratio, 3.2; 95% confidence interval [CI], 1.3 to 7.8; P = 0.01). At 6 months, the overall response rate (the percentage of patients who had a complete or partial response) was 41% in Group A and 68% in Group B. The median times to the first response were 8.8 months (Group A) and 3.0 months (Group B). The incidence of severe adverse events was similar in the two groups. With a median follow-up of 24 months, a karyotypic abnormality that was classified as myelodysplastic syndrome developed in 1 patient (Group A) and 2 patients (Group B); event-free survival was 34% and 46%, respectively. Somatic mutations were detected in 29% (Group A) and 31% (Group Β) of the patients at baseline; these percentages increased to 66% and 55%, respectively, at 6 months, without affecting the hematologic response and 2-year outcome. CONCLUSIONS: The addition of eltrombopag to standard immunosuppressive therapy improved the rate, rapidity, and strength of hematologic response among previously untreated patients with severe aplastic anemia, without additional toxic effects. (Funded by Novartis and others; RACE ClinicalTrials.gov number, NCT02099747; EudraCT number, 2014-000363-40.).


Subject(s)
Anemia, Aplastic/therapy , Antilymphocyte Serum/therapeutic use , Benzoates/therapeutic use , Cyclosporine/therapeutic use , Hydrazines/therapeutic use , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Pyrazoles/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Anemia, Aplastic/drug therapy , Anemia, Aplastic/genetics , Antilymphocyte Serum/adverse effects , Benzoates/adverse effects , Cyclosporine/adverse effects , Drug Therapy, Combination , Female , Humans , Hydrazines/adverse effects , Immunosuppressive Agents/adverse effects , Male , Middle Aged , Progression-Free Survival , Prospective Studies , Pyrazoles/adverse effects , Receptors, Thrombopoietin/agonists , Remission Induction , Young Adult
12.
N Engl J Med ; 387(8): 704-714, 2022 08 25.
Article in English | MEDLINE | ID: mdl-36001711

ABSTRACT

BACKGROUND: The monoclonal antibody ublituximab enhances antibody-dependent cellular cytolysis and produces B-cell depletion. Ublituximab is being evaluated for the treatment of relapsing multiple sclerosis. METHODS: In two identical, phase 3, double-blind, double-dummy trials (ULTIMATE I and II), participants with relapsing multiple sclerosis were randomly assigned in a 1:1 ratio to receive intravenous ublituximab (150 mg on day 1, followed by 450 mg on day 15 and at weeks 24, 48, and 72) and oral placebo or oral teriflunomide (14 mg once daily) and intravenous placebo. The primary end point was the annualized relapse rate. Secondary end points included the number of gadolinium-enhancing lesions on magnetic resonance imaging (MRI) by 96 weeks and worsening of disability. RESULTS: A total of 549 participants were enrolled in the ULTIMATE I trial, and 545 were enrolled in the ULTIMATE II trial; the median follow-up was 95 weeks. In the ULTIMATE I trial, the annualized relapse rate was 0.08 with ublituximab and 0.19 with teriflunomide (rate ratio, 0.41; 95% confidence interval [CI], 0.27 to 0.62; P<0.001); in the ULTIMATE II trial, the annualized relapse rate was 0.09 and 0.18, respectively (rate ratio, 0.51; 95% CI, 0.33 to 0.78; P = 0.002). The mean number of gadolinium-enhancing lesions was 0.02 in the ublituximab group and 0.49 in the teriflunomide group (rate ratio, 0.03; 95% CI, 0.02 to 0.06; P<0.001) in the ULTIMATE I trial and 0.01 and 0.25, respectively (rate ratio, 0.04; 95% CI, 0.02 to 0.06; P<0.001), in the ULTIMATE II trial. In the pooled analysis of the two trials, 5.2% of the participants in the ublituximab group and 5.9% in the teriflunomide group had worsening of disability at 12 weeks (hazard ratio, 0.84; 95% CI, 0.50 to 1.41; P = 0.51). Infusion-related reactions occurred in 47.7% of the participants in the ublituximab group. Serious infections occurred in 5.0% in the ublituximab group and in 2.9% in the teriflunomide group. CONCLUSIONS: Among participants with relapsing multiple sclerosis, ublituximab resulted in lower annualized relapse rates and fewer brain lesions on MRI than teriflunomide over a period of 96 weeks but did not result in a significantly lower risk of worsening of disability. Ublituximab was associated with infusion-related reactions. (Funded by TG Therapeutics; ULTIMATE I and II ClinicalTrials.gov numbers, NCT03277261 and NCT03277248.).


Subject(s)
Antibodies, Monoclonal , Multiple Sclerosis, Relapsing-Remitting , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies, Monoclonal/therapeutic use , Crotonates , Double-Blind Method , Gadolinium/therapeutic use , Humans , Hydroxybutyrates , Immunosuppressive Agents/therapeutic use , Magnetic Resonance Imaging , Multiple Sclerosis/complications , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Multiple Sclerosis, Relapsing-Remitting/complications , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/pathology , Nitriles , Toluidines
13.
J Virol ; 98(7): e0084624, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38899900

ABSTRACT

Chronic hepatitis E mostly occurs in organ transplant recipients and can lead to rapid liver fibrosis and cirrhosis. Previous studies found that the development of chronic hepatitis E virus (HEV) infection is linked to the type of immunosuppressant used. Animal models are crucial for the study of pathogenesis of chronic hepatitis E. We previously established a stable chronic HEV infection rabbit model using cyclosporine A (CsA), a calcineurin inhibitor (CNI)-based immunosuppressant. However, the immunosuppression strategy and timing may be optimized, and how different types of immunosuppressants affect the establishment of chronic HEV infection in this model is still unknown. Here, we showed that chronic HEV infection can be established in 100% of rabbits when CsA treatment was started at HEV challenge or even 4 weeks after. Tacrolimus or prednisolone treatment alone also contributed to chronic HEV infection, resulting in 100% and 77.8% chronicity rates, respectively, while mycophenolate mofetil (MMF) only led to a 28.6% chronicity rate. Chronic HEV infection was accompanied with a persistent activation of innate immune response evidenced by transcriptome analysis. The suppressed adaptive immune response evidenced by low expression of genes related to cytotoxicity (like perforin and FasL) and low anti-HEV seroconversion rates may play important roles in causing chronic HEV infection. By analyzing HEV antigen concentrations with different infection outcomes, we also found that HEV antigen levels could indicate chronic HEV infection development. This study optimized the immunosuppression strategies for establishing chronic HEV infection in rabbits and highlighted the potential association between the development of chronic HEV infection and immunosuppressants.IMPORTANCEOrgan transplant recipients are at high risk of chronic hepatitis E and generally receive a CNI-based immunosuppression regimen containing CNI (tacrolimus or CsA), MMF, and/or corticosteroids. Previously, we established stable chronic HEV infection in a rabbit model by using CsA before HEV challenge. In this study, we further optimized the immunosuppression strategies for establishing chronic HEV infection in rabbits. Chronic HEV infection can also be established when CsA treatment was started at the same time or even 4 weeks after HEV challenge, clearly indicating the risk of progression to chronic infection under these circumstances and the necessity of HEV screening for both the recipient and the donor preoperatively. CsA, tacrolimus, or prednisolone instead of MMF significantly contributed to chronic HEV infection. HEV antigen in acute infection phase indicates the development of chronic infection. Our results have important implications for understanding the potential association between chronic HEV infection and immunosuppressants.


Subject(s)
Cyclosporine , Disease Models, Animal , Hepatitis E virus , Hepatitis E , Immunosuppression Therapy , Immunosuppressive Agents , Tacrolimus , Animals , Rabbits , Hepatitis E/immunology , Hepatitis E/virology , Hepatitis E/drug therapy , Hepatitis E virus/immunology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Cyclosporine/pharmacology , Cyclosporine/therapeutic use , Tacrolimus/pharmacology , Tacrolimus/therapeutic use , Prednisolone/therapeutic use , Prednisolone/pharmacology , Male , Immunity, Innate/drug effects , Mycophenolic Acid/pharmacology , Hepatitis, Chronic/drug therapy , Hepatitis, Chronic/immunology , Hepatitis, Chronic/virology , Chronic Disease , Calcineurin Inhibitors/pharmacology , Calcineurin Inhibitors/therapeutic use
14.
Nat Immunol ; 14(2): 106-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23334822

ABSTRACT

Therapies that target molecular pathways do not provide uniform benefits for all patients at present. New transformative therapies for autoimmune and inflammatory diseases require greater molecular understanding of patient subsets and the ability to personalize targeted therapies for each subset.


Subject(s)
Autoimmune Diseases/therapy , Inflammation/therapy , Molecular Targeted Therapy , Neoplasms/therapy , Precision Medicine , Antineoplastic Agents/economics , Antineoplastic Agents/therapeutic use , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Genetic Heterogeneity , Humans , Immunosuppressive Agents/economics , Immunosuppressive Agents/therapeutic use , Inflammation/genetics , Inflammation/immunology , Neoplasms/genetics , Neoplasms/immunology
15.
Blood ; 142(12): 1037-1046, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37471585

ABSTRACT

Acute graft-versus-host disease (aGVHD) is a major life-threatening complication after allogeneic hematopoietic cell transplant. Traditional standard prophylaxis for aGVHD has included a calcineurin inhibitor plus an antimetabolite, whereas treatment has relied mainly on corticosteroids, followed by multiple nonstandard second-line options. In the past decade, this basic framework has been reshaped by approval of antithymocyte globulin products, the emergence of posttransplant cyclophosphamide, and recent pivotal trials studying abatacept and vedolizumab for GVHD prophylaxis, whereas ruxolitinib was approved for corticosteroid-refractory aGVHD treatment. Because of this progress, routine acute GVHD prophylaxis and treatment practices are starting to shift, and results of ongoing trials are eagerly awaited. Here, we review recent developments in aGVHD prevention and therapy, along with ongoing and future planned clinical trials in this space, outlining what future goals should be and the limitations of current clinical trial designs and end points.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Immunosuppressive Agents/therapeutic use , Transplantation, Homologous/adverse effects , Cyclophosphamide/therapeutic use , Adrenal Cortex Hormones/therapeutic use
16.
Am J Respir Crit Care Med ; 209(2): 137-152, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-37772985

ABSTRACT

Background: Interstitial lung disease (ILD) is a significant cause of morbidity and mortality in patients with systemic sclerosis (SSc). To date, clinical practice guidelines regarding treatment for patients with SSc-ILD are primarily consensus based. Methods: An international expert guideline committee composed of 24 individuals with expertise in rheumatology, SSc, pulmonology, ILD, or methodology, and with personal experience with SSc-ILD, discussed systematic reviews of the published evidence assessed using the Grading of Recommendations, Assessment, Development, and Evaluation approach. Predetermined conflict-of-interest management strategies were applied, and recommendations were made for or against specific treatment interventions exclusively by the nonconflicted panelists. The confidence in effect estimates, importance of outcomes studied, balance of desirable and undesirable consequences of treatment, cost, feasibility, acceptability of the intervention, and implications for health equity were all considered in making the recommendations. This was in accordance with the American Thoracic Society guideline development process, which is in compliance with the Institute of Medicine standards for trustworthy guidelines. Results: For treatment of patients with SSc-ILD, the committee: 1) recommends the use of mycophenolate; 2) recommends further research into the safety and efficacy of (a) pirfenidone and (b) the combination of pirfenidone plus mycophenolate; and 3) suggests the use of (a) cyclophosphamide, (b) rituximab, (c) tocilizumab, (d) nintedanib, and (e) the combination of nintedanib plus mycophenolate. Conclusions: The recommendations herein provide an evidence-based clinical practice guideline for the treatment of patients with SSc-ILD and are intended to serve as the basis for informed and shared decision making by clinicians and patients.


Subject(s)
Lung Diseases, Interstitial , Scleroderma, Systemic , Humans , United States , Immunosuppressive Agents/therapeutic use , Lung Diseases, Interstitial/drug therapy , Lung Diseases, Interstitial/etiology , Cyclophosphamide/therapeutic use , Rituximab/therapeutic use , Scleroderma, Systemic/complications , Lung
17.
Proc Natl Acad Sci U S A ; 119(43): e2205417119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36256820

ABSTRACT

Antigen-specific therapies hold promise for treating autoimmune diseases such as multiple sclerosis while avoiding the deleterious side effects of systemic immune suppression due to delivering the disease-specific antigen as part of the treatment. In this study, an antigen-specific dual-sized microparticle (dMP) treatment reversed hind limb paralysis when administered in mice with advanced experimental autoimmune encephalomyelitis (EAE). Treatment reduced central nervous system (CNS) immune cell infiltration, demyelination, and inflammatory cytokine levels. Mechanistic insights using single-cell RNA sequencing showed that treatment impacted the MHC II antigen presentation pathway in dendritic cells, macrophages, B cells, and microglia, not only in the draining lymph nodes but also strikingly in the spinal cord. CD74 and cathepsin S were among the common genes down-regulated in most antigen presenting cell (APC) clusters, with B cells also having numerous MHC II genes reduced. Efficacy of the treatment diminished when B cells were absent, suggesting their impact in this therapy, in concert with other immune populations. Activation and inflammation were reduced in both APCs and T cells. This promising antigen-specific therapeutic approach advantageously engaged essential components of both innate and adaptive autoimmune responses and capably reversed paralysis in advanced EAE without the use of a broad immunosuppressant.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Mice , Antigens , Cytokines , Mice, Inbred C57BL , Paralysis , Cathepsins , Immunosuppressive Agents/therapeutic use
18.
Proc Natl Acad Sci U S A ; 119(31): e2205042119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35881799

ABSTRACT

Dimethyl fumarate (DMF) is an immunomodulatory treatment for multiple sclerosis (MS). Despite its wide clinical use, the mechanisms underlying clinical response are not understood. This study aimed to reveal immune markers of therapeutic response to DMF treatment in MS. For this purpose, we prospectively collected peripheral blood mononuclear cells (PBMCs) from a highly characterized cohort of 44 individuals with MS before and at 12 and 48 wk of DMF treatment. Single cells were profiled using high-dimensional mass cytometry. To capture the heterogeneity of different immune subsets, we adopted a bioinformatic multipanel approach that allowed cell population-cluster assignment of more than 50 different parameters, including lineage and activation markers as well as chemokine receptors and cytokines. Data were further analyzed in a semiunbiased fashion implementing a supervised representation learning approach to capture subtle longitudinal immune changes characteristic for therapy response. With this approach, we identified a population of memory T helper cells expressing high levels of neuroinflammatory cytokines (granulocyte-macrophage colony-stimulating factor [GM-CSF], interferon γ [IFNγ]) as well as CXCR3, whose abundance correlated with treatment response. Using spectral flow cytometry, we confirmed these findings in a second cohort of patients. Serum neurofilament light-chain levels confirmed the correlation of this immune cell signature with axonal damage. The identified cell population is expanded in peripheral blood under natalizumab treatment, substantiating a specific role in treatment response. We propose that depletion of GM-CSF-, IFNγ-, and CXCR3-expressing T helper cells is the main mechanism of action of DMF and allows monitoring of treatment response.


Subject(s)
Biomarkers, Pharmacological , Cytokines , Dimethyl Fumarate , Immunosuppressive Agents , Multiple Sclerosis , T-Lymphocytes, Helper-Inducer , Biomarkers, Pharmacological/metabolism , Cytokines/metabolism , Dimethyl Fumarate/pharmacology , Dimethyl Fumarate/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Interferon-gamma/metabolism , Lymphocyte Depletion , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Single-Cell Analysis , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology
19.
Proc Natl Acad Sci U S A ; 119(16): e2117716119, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35412894

ABSTRACT

As a critical sphingolipid metabolite, sphingosine-1-phosphate (S1P) plays an essential role in immune and vascular systems. There are five S1P receptors, designated as S1PR1 to S1PR5, encoded in the human genome, and their activities are governed by endogenous S1P, lipid-like S1P mimics, or nonlipid-like therapeutic molecules. Among S1PRs, S1PR1 stands out due to its nonredundant functions, such as the egress of T and B cells from the thymus and secondary lymphoid tissues, making it a potential therapeutic target. However, the structural basis of S1PR1 activation and regulation by various agonists remains unclear. Here, we report four atomic resolution cryo-electron microscopy (cryo-EM) structures of Gi-coupled human S1PR1 complexes: bound to endogenous agonist d18:1 S1P, benchmark lipid-like S1P mimic phosphorylated Fingolimod [(S)-FTY720-P], or nonlipid-like therapeutic molecule CBP-307 in two binding modes. Our results revealed the similarities and differences of activation of S1PR1 through distinct ligands binding to the amphiphilic orthosteric pocket. We also proposed a two-step "shallow to deep" transition process of CBP-307 for S1PR1 activation. Both binding modes of CBP-307 could activate S1PR1, but from shallow to deep transition may trigger the rotation of the N-terminal helix of Gαi and further stabilize the complex by increasing the Gαi interaction with the cell membrane. We combine with extensive biochemical analysis and molecular dynamic simulations to suggest key steps of S1P binding and receptor activation. The above results decipher the common feature of the S1PR1 agonist recognition and activation mechanism and will firmly promote the development of therapeutics targeting S1PRs.


Subject(s)
Sphingosine 1 Phosphate Receptor Modulators , Sphingosine-1-Phosphate Receptors , Colitis, Ulcerative/drug therapy , Cryoelectron Microscopy , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Humans , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis/drug therapy , Organophosphates/chemistry , Organophosphates/pharmacology , Organophosphates/therapeutic use , Protein Binding , Protein Conformation, alpha-Helical , Sphingosine/analogs & derivatives , Sphingosine/chemistry , Sphingosine/pharmacology , Sphingosine/therapeutic use , Sphingosine 1 Phosphate Receptor Modulators/chemistry , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use , Sphingosine-1-Phosphate Receptors/agonists , Sphingosine-1-Phosphate Receptors/chemistry
20.
Eur Heart J ; 45(30): 2697-2726, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-38923509

ABSTRACT

Cardiac sarcoidosis (CS) is a form of inflammatory cardiomyopathy associated with significant clinical complications such as high-degree atrioventricular block, ventricular tachycardia, and heart failure as well as sudden cardiac death. It is therefore important to provide an expert consensus statement summarizing the role of different available diagnostic tools and emphasizing the importance of a multidisciplinary approach. By integrating clinical information and the results of diagnostic tests, an accurate, validated, and timely diagnosis can be made, while alternative diagnoses can be reasonably excluded. This clinical expert consensus statement reviews the evidence on the management of different CS manifestations and provides advice to practicing clinicians in the field on the role of immunosuppression and the treatment of cardiac complications based on limited published data and the experience of international CS experts. The monitoring and risk stratification of patients with CS is also covered, while controversies and future research needs are explored.


Subject(s)
Cardiomyopathies , Sarcoidosis , Humans , Sarcoidosis/diagnosis , Sarcoidosis/therapy , Sarcoidosis/complications , Cardiomyopathies/diagnosis , Cardiomyopathies/therapy , Immunosuppressive Agents/therapeutic use , Death, Sudden, Cardiac/prevention & control , Death, Sudden, Cardiac/etiology
SELECTION OF CITATIONS
SEARCH DETAIL